1
|
Matuszewska J, Krawiec A, Radziemski A, Uruski P, Tykarski A, Mikuła-Pietrasik J, Książek K. Alterations of receptors and insulin-like growth factor binding proteins in senescent cells. Eur J Cell Biol 2024; 103:151438. [PMID: 38945074 DOI: 10.1016/j.ejcb.2024.151438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024] Open
Abstract
The knowledge about cellular senescence expands dynamically, providing more and more conclusive evidence of its triggers, mechanisms, and consequences. Senescence-associated secretory phenotype (SASP), one of the most important functional traits of senescent cells, is responsible for a large extent of their context-dependent activity. Both SASP's components and signaling pathways are well-defined. A literature review shows, however, that a relatively underinvestigated aspect of senescent cell autocrine and paracrine activity is the change in the production of proteins responsible for the reception and transmission of SASP signals, i.e., receptors and binding proteins. For this reason, we present in this article the current state of knowledge regarding senescence-associated changes in cellular receptors and insulin-like growth factor binding proteins. We also discuss the role of these alterations in senescence induction and maintenance, pro-cancerogenic effects of senescent cells, and aging-related structural and functional malfunctions.
Collapse
Affiliation(s)
- Julia Matuszewska
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Adrianna Krawiec
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Artur Radziemski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Paweł Uruski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Andrzej Tykarski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Justyna Mikuła-Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Krzysztof Książek
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland.
| |
Collapse
|
2
|
Dwivedi S, Chavan A, Paul AT. SET7, a lysine-specific methyl transferase: An intriguing epigenetic target to combat diabetic nephropathy. Drug Discov Today 2023; 28:103754. [PMID: 37648018 DOI: 10.1016/j.drudis.2023.103754] [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: 04/24/2023] [Revised: 07/14/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023]
Abstract
Diabetic nephropathy (DN) is a dreadful complication of diabetes that affects ∼50% of diabetics and is a leading cause of end-stage renal disease (ESRD). Studies have linked aberrant expression of lysine methyltransferases (KMTs) to the onset and progression of DN. SET7 is a KMT that methylates specific lysine residues of the histone and nonhistone proteins. It plays an important role in the transforming growth factor-β (TGF-β)-induced upregulation of extracellular matrix (ECM)-associated genes that are responsible for the inflammatory cascade observed in DN. Inhibiting SET7 has potential to attenuate renal disorders in animal studies. This review will focus on the role of SET7 in DN and its potential as a therapeutic target to combat DN.
Collapse
Affiliation(s)
- Samarth Dwivedi
- Natural Product Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (Pilani Campus), Pilani 333031, Rajasthan, India
| | - Atharva Chavan
- Natural Product Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (Pilani Campus), Pilani 333031, Rajasthan, India
| | - Atish T Paul
- Natural Product Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (Pilani Campus), Pilani 333031, Rajasthan, India.
| |
Collapse
|
3
|
Piletska E, Thompson D, Jones R, Cruz AG, Poblocka M, Canfarotta F, Norman R, Macip S, Jones DJL, Piletsky S. Snapshot imprinting as a tool for surface mapping and identification of novel biomarkers of senescent cells. NANOSCALE ADVANCES 2022; 4:5304-5311. [PMID: 36540121 PMCID: PMC9724690 DOI: 10.1039/d2na00424k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/11/2022] [Indexed: 06/17/2023]
Abstract
Cellular senescence has proved to be a strong contributor to ageing and age-related diseases, such as cancer and atherosclerosis. Therefore, the protein content of senescent cells is highly relevant to drug discovery, diagnostics and therapeutic applications. However, current technologies for the analysis of proteins are based on a combination of separation techniques and mass spectrometry, which require handling large sample sizes and a large volume of data and are time-consuming. This limits their application in personalised medicine. An easy, quick and inexpensive procedure is needed for qualitative and quantitative analysis of proteins expressed by a cell or tissue. Here, we describe the use of the "snapshot imprinting" approach for the identification of proteins differentially expressed by senescent cells. Molecularly imprinted polymer nanoparticles (MIPs) were formed in the presence of whole cells. Following trypsinolysis, protein epitopes protected by complex with MIPs were eluted from the nanoparticles and analysed by LC-MS/MS. In this work, "snapshot imprinting" was performed parallel to a standard proteomic "shaving approach", showing similar results. The analysis by "snapshot imprinting" identified three senescent-specific proteins: cell division cycle 7-related protein kinase, partitioning defective three homolog B and putative ATP-dependent RNA helicase DHX57, the abundance of which could potentially make them specific markers of senescence. Identifying biomarkers for the future elimination of senescent cells grants the potential for developing therapeutics for age-related diseases.
Collapse
Affiliation(s)
- Elena Piletska
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Dana Thompson
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Rebecca Jones
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Alvaro Garcia Cruz
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Marta Poblocka
- Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester Leicester LE1 7RH UK
| | - Francesco Canfarotta
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| | - Rachel Norman
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya 08018 Barcelona Spain
| | - Salvador Macip
- Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester Leicester LE1 7RH UK
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya 08018 Barcelona Spain
| | - Donald J L Jones
- Department of Cancer Studies, RKCSB, University of Leicester Leicester LE2 7LX UK
| | - Sergey Piletsky
- Chemistry Department, College of Science and Engineering, University of Leicester Leicester LE1 7RH UK
| |
Collapse
|
4
|
Xie Q, Lv H, Wang T, Sun J, Li Y, Niu Y, Xie W. Identifying Common Genes and Pathways Associated with Periodontitis and Aging by Bioinformatics Analysis. DISEASE MARKERS 2022; 2022:4199440. [PMID: 36438900 PMCID: PMC9691312 DOI: 10.1155/2022/4199440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/18/2022] [Accepted: 09/27/2022] [Indexed: 09/29/2023]
Abstract
BACKGROUND This work used bioinformatic analysis to identify the relationship between periodontitis (PD) and aging, which could lead to new treatments for periodontal disease in the elderly. METHOD Four microarray datasets were obtained from the Gene Expression Omnibus (GEO) database and analyzed in R language to identify differentially expressed genes (DEGs). The common DEGs of PD and aging were evaluated as key genes in this investigation by a Venn diagram. These common DEGs were analyzed through additional experiments and analysis, such as pathway analysis and enrichment analysis, and a network of protein-protein interactions (PPIs) was constructed. Cytoscape was used to visualize hub genes and critical modules based on the PPI network. Interaction of TF-genes and miRNAs with hub genes is identified. RESULT 84 common DEGs were found between PD and aging. Cytohubba was performed on the PPI network obtained from STRING tool, and the top 10 genes (MMP2, PDGFRB, CTGF, CD34, CXCL12, VIM, IL2RG, ACTA2, COL4A2, and TAGLN) were selected as hub genes. VIM may be a potential biomarker in the analysis of linked hub gene regulatory networks, and hsa-mir-21 and hsa-mir-125b are predicted to be associated in PD and aging. CONCLUSION This study investigated the key genes and pathways interactions between PD and aging, which may help reveal the correlation between PD and aging. The current research results are obtained by prediction, and follow-up biological experiments are required for further verification.
Collapse
Affiliation(s)
- Qi Xie
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
- Department of Stomatology, Harbin Children's Hospital, Harbin, Heilongjiang 150001, China
| | - Hongyu Lv
- Department of Prosthodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Tianqi Wang
- Department of Prosthodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Jingxuan Sun
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Yuekun Li
- Department of Prosthodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Yumei Niu
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Weili Xie
- Department of Prosthodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| |
Collapse
|
5
|
Connective Tissue Growth Factor in Idiopathic Pulmonary Fibrosis: Breaking the Bridge. Int J Mol Sci 2022; 23:ijms23116064. [PMID: 35682743 PMCID: PMC9181498 DOI: 10.3390/ijms23116064] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/23/2022] Open
Abstract
CTGF is upregulated in patients with idiopathic pulmonary fibrosis (IPF), characterized by the deposition of a pathological extracellular matrix (ECM). Additionally, many omics studies confirmed that aberrant cellular senescence-associated mitochondria dysfunction and metabolic reprogramming had been identified in different IPF lung cells (alveolar epithelial cells, alveolar endothelial cells, fibroblasts, and macrophages). Here, we reviewed the role of the CTGF in IPF lung cells to mediate anomalous senescence-related metabolic mechanisms that support the fibrotic environment in IPF.
Collapse
|
6
|
Lunin SM, Novoselova EG, Glushkova OV, Parfenyuk SB, Novoselova TV, Khrenov MO. Cell Senescence and Central Regulators of Immune Response. Int J Mol Sci 2022; 23:ijms23084109. [PMID: 35456927 PMCID: PMC9028919 DOI: 10.3390/ijms23084109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/13/2022] Open
Abstract
Pathways regulating cell senescence and cell cycle underlie many processes associated with ageing and age-related pathologies, and they also mediate cellular responses to exposure to stressors. Meanwhile, there are central mechanisms of the regulation of stress responses that induce/enhance or weaken the response of the whole organism, such as hormones of the hypothalamic-pituitary-adrenal (HPA) axis, sympathetic and parasympathetic systems, thymic hormones, and the pineal hormone melatonin. Although there are many analyses considering relationships between the HPA axis and organism ageing, we found no systematic analyses of relationships between the neuroendocrine regulators of stress and inflammation and intracellular mechanisms controlling cell cycle, senescence, and apoptosis. Here, we provide a review of the effects of neuroendocrine regulators on these mechanisms. Our analysis allowed us to postulate a multilevel system of central regulators involving neurotransmitters, glucocorticoids, melatonin, and the thymic hormones. This system finely regulates the cell cycle and metabolic/catabolic processes depending on the level of systemic stress, stage of stress response, and energy capabilities of the body, shifting the balance between cell cycle progression, cell cycle stopping, senescence, and apoptosis. These processes and levels of regulation should be considered when studying the mechanisms of ageing and the proliferation on the level of the whole organism.
Collapse
|
7
|
Fu M, Peng D, Lan T, Wei Y, Wei X. Multifunctional regulatory protein connective tissue growth factor (CTGF): A potential therapeutic target for diverse diseases. Acta Pharm Sin B 2022; 12:1740-1760. [PMID: 35847511 PMCID: PMC9279711 DOI: 10.1016/j.apsb.2022.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Connective tissue growth factor (CTGF), a multifunctional protein of the CCN family, regulates cell proliferation, differentiation, adhesion, and a variety of other biological processes. It is involved in the disease-related pathways such as the Hippo pathway, p53 and nuclear factor kappa-B (NF-κB) pathways and thus contributes to the developments of inflammation, fibrosis, cancer and other diseases as a downstream effector. Therefore, CTGF might be a potential therapeutic target for treating various diseases. In recent years, the research on the potential of CTGF in the treatment of diseases has also been paid more attention. Several drugs targeting CTGF (monoclonal antibodies FG3149 and FG3019) are being assessed by clinical or preclinical trials and have shown promising outcomes. In this review, the cellular events regulated by CTGF, and the relationships between CTGF and pathogenesis of diseases are systematically summarized. In addition, we highlight the current researches, focusing on the preclinical and clinical trials concerned with CTGF as the therapeutic target.
Collapse
|
8
|
Marquez-Exposito L, Tejedor-Santamaria L, Valentijn FA, Tejera-Muñoz A, Rayego-Mateos S, Marchant V, Rodrigues-Diez RR, Rubio-Soto I, Knoppert SN, Ortiz A, Ramos AM, Goldschmeding R, Ruiz-Ortega M. Oxidative Stress and Cellular Senescence Are Involved in the Aging Kidney. Antioxidants (Basel) 2022; 11:301. [PMID: 35204184 PMCID: PMC8868560 DOI: 10.3390/antiox11020301] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) can be considered as a clinical model for premature aging. However, non-invasive biomarkers to detect early kidney damage and the onset of a senescent phenotype are lacking. Most of the preclinical senescence studies in aging have been done in very old mice. Furthermore, the precise characterization and over-time development of age-related senescence in the kidney remain unclear. To address these limitations, the age-related activation of cellular senescence-associated mechanisms and their correlation with early structural changes in the kidney were investigated in 3- to 18-month-old C57BL6 mice. Inflammatory cell infiltration was observed by 12 months, whereas tubular damage and collagen accumulation occurred later. Early activation of cellular-senescence-associated mechanisms was found in 12-month-old mice, characterized by activation of the DNA-damage-response (DDR), mainly in tubular cells; activation of the antioxidant NRF2 pathway; and klotho downregulation. However, induction of tubular-cell-cycle-arrest (CCA) and overexpression of renal senescent-associated secretory phenotype (SASP) components was only found in 18-month-old mice. In aging mice, both inflammation and oxidative stress (marked by elevated lipid peroxidation and NRF2 inactivation) remained increased. These findings support the hypothesis that prolonged DDR and CCA, loss of nephroprotective factors (klotho), and dysfunctional redox regulatory mechanisms (NRF2/antioxidant defense) can be early drivers of age-related kidney-damage progression.
Collapse
Affiliation(s)
- Laura Marquez-Exposito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Lucia Tejedor-Santamaria
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Floris A. Valentijn
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.G.)
| | - Antonio Tejera-Muñoz
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Raul R. Rodrigues-Diez
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
- Translational Immunology Laboratory, Health Research Institute of Asturias (ISPA), Av. Roma, s/n, 33011 Oviedo, Spain
| | - Irene Rubio-Soto
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Sebastiaan N. Knoppert
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.G.)
| | - Alberto Ortiz
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Adrian M. Ramos
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.G.)
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| |
Collapse
|
9
|
Roger I, Milara J, Belhadj N, Cortijo J. Senescence Alterations in Pulmonary Hypertension. Cells 2021; 10:3456. [PMID: 34943963 PMCID: PMC8700581 DOI: 10.3390/cells10123456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is the arrest of normal cell division and is commonly associated with aging. The interest in the role of cellular senescence in lung diseases derives from the observation of markers of senescence in chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (IPF), and pulmonary hypertension (PH). Accumulation of senescent cells and the senescence-associated secretory phenotype in the lung of aged patients may lead to mild persistent inflammation, which results in tissue damage. Oxidative stress due to environmental exposures such as cigarette smoke also promotes cellular senescence, together with additional forms of cellular stress such as mitochondrial dysfunction and endoplasmic reticulum stress. Growing recent evidence indicate that senescent cell phenotypes are observed in pulmonary artery smooth muscle cells and endothelial cells of patients with PH, contributing to pulmonary artery remodeling and PH development. In this review, we analyze the role of different senescence cell phenotypes contributing to the pulmonary artery remodeling process in different PH clinical entities. Different molecular pathway activation and cellular functions derived from senescence activation will be analyzed and discussed as promising targets to develop future senotherapies as promising treatments to attenuate pulmonary artery remodeling in PH.
Collapse
Affiliation(s)
- Inés Roger
- Centro de Investigación en Red Enfermedades Respiratorias CIBERES, Health Institute Carlos III, 28029 Valencia, Spain;
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Javier Milara
- Centro de Investigación en Red Enfermedades Respiratorias CIBERES, Health Institute Carlos III, 28029 Valencia, Spain;
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Pharmacy Unit, University General Hospital Consortium of Valencia, 46014 Valencia, Spain
| | - Nada Belhadj
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Julio Cortijo
- Centro de Investigación en Red Enfermedades Respiratorias CIBERES, Health Institute Carlos III, 28029 Valencia, Spain;
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Research and Teaching Unit, University General Hospital Consortium, 46014 Valencia, Spain
| |
Collapse
|
10
|
Mayro EL, Ritch R, Pasquale LR. Early-onset Exfoliation Syndrome: A Literature Synthesis. J Glaucoma 2021; 30:e164-e168. [PMID: 33449585 DOI: 10.1097/ijg.0000000000001784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/20/2020] [Indexed: 01/13/2023]
Abstract
PRECIS We conducted a literature review of younger patients with exfoliation syndrome (XFS) in an attempt to identify case similarities and better understand disease etiology. PURPOSE XFS that predisposes to secondary glaucoma is a strongly age-related condition. We performed a literature review of XFS and exfoliation glaucoma (XFG) in patients aged younger than 40 years to examine potential common characteristics and gain clues to its etiology. METHODS We conducted a broad literature search with appropriate keywords and manually extracted key demographic and ocular features on younger XFS and XFG cases. Articles that did not provide past ocular history on early-onset XFS/XFG were excluded. RESULTS We identified 12 cases of XFS and XFG in patients from 13 to 40 years old (8 females; 11 White; 5 from Iran). All had past ocular history remarkable for intraocular surgery for other glaucoma conditions (7 cases), other ocular diseases (3 cases), or ocular trauma (2 cases). CONCLUSIONS All reported early-onset XFS and XFG cases arise in the setting of events that produced a significant disruption of the blood-aqueous barrier. Understanding the metabolic alterations of aqueous humor from such cases could provide clues regarding how exfoliation material forms.
Collapse
Affiliation(s)
- Eileen L Mayro
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Robert Ritch
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY
| | - Louis R Pasquale
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY
| |
Collapse
|
11
|
α-Lipoic Acid Increases Collagen Synthesis and Deposition in Nondiabetic and Diabetic Rat Kidneys. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6669352. [PMID: 33777319 PMCID: PMC7979310 DOI: 10.1155/2021/6669352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/19/2021] [Accepted: 02/27/2021] [Indexed: 12/24/2022]
Abstract
α-Lipoic acid (ALA) is widely used as a nutritional supplement and therapeutic agent in diabetes management. Well-established antioxidant and hypoglycemic effects of ALA were considered to be particularly important in combating diabetic complications including renal injury. The present study evaluated the potential of ALA to affect profibrotic events in kidney that could alter its structure and functioning. ALA was administered intraperitoneally (10 mg/kg) to nondiabetic and streptozotocin-induced diabetic male Wistar rats for 4 and 8 weeks. The effects of ALA were assessed starting from structural/morphological alterations through changes that characterize profibrotic processes, to regulation of collagen gene expression in kidney. Here, we demonstrated that ALA improved systemic glucose and urea level, reduced formation of renal advanced glycation end products (AGEs), and maintained renal structural integrity in diabetic rats. However, profibrotic events provoked in diabetes were not alleviated by ALA since collagen synthesis/deposition and expression of transforming growth factor-β1 (TGF-β1) and α-smooth muscle actin (α-SMA) remained elevated in ALA-treated diabetic rats, especially after 8 weeks of diabetes onset. Moreover, 8 weeks treatment of nondiabetic rats with ALA led to the development of profibrotic features reflected in increased collagen synthesis/deposition. Besides the TGF-β1 downstream signaling, the additional mechanism underlying the upregulation of collagen IV in nondiabetic rats treated with ALA involves decreased DNA methylation of its promoter that could arise from increased Tet1 expression. These findings emphasize the therapeutic caution in the use of ALA, especially in patients with renal diabetic complication.
Collapse
|
12
|
Knoppert SN, Valentijn FA, Nguyen TQ, Goldschmeding R, Falke LL. Cellular Senescence and the Kidney: Potential Therapeutic Targets and Tools. Front Pharmacol 2019; 10:770. [PMID: 31354486 PMCID: PMC6639430 DOI: 10.3389/fphar.2019.00770] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasing health burden (affecting approximately 13.4% of the population). Currently, no curative treatment options are available and treatment is focused on limiting the disease progression. The accumulation of senescent cells has been implicated in the development of kidney fibrosis by limiting tissue rejuvenation and through the secretion of pro-fibrotic and pro-inflammatory mediators termed as the senescence-associated secretory phenotype. The clearance of senescent cells in aging models results in improved kidney function, which shows promise for the options of targeting senescent cells in CKD. There are several approaches for the development of “senotherapies”, the most rigorous of which is the elimination of senescent cells by the so-called senolytic drugs either newly developed or repurposed for off-target effects in terms of selectively inducing apoptosis in senescent cells. Several chemotherapeutics and checkpoint inhibitors currently used in daily oncological practice show senolytic properties. However, the applicability of such senolytic compounds for the treatment of renal diseases has hardly been investigated. A serious concern is that systemic side effects will limit the use of senolytics for kidney fibrosis. Specifically targeting senescent cells and/or targeted drug delivery to the kidney might circumvent these side effects. In this review, we discuss the connection between CKD and senescence, the pharmacological options for targeting senescent cells, and the means to specifically target the kidney.
Collapse
Affiliation(s)
- Sebastian N Knoppert
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Floris A Valentijn
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Internal Medicine, Diakonessenhuis, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
13
|
Zhang K, Ma Z, Wang W, Liu R, Zhang Y, Yuan M, Li G. Effect of doxycycline on chronic intermittent hypoxia-induced atrial remodeling in rats. Herz 2018; 45:668-675. [DOI: 10.1007/s00059-018-4768-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/17/2018] [Accepted: 10/29/2018] [Indexed: 12/26/2022]
|
14
|
Harper EI, Sheedy EF, Stack MS. With Great Age Comes Great Metastatic Ability: Ovarian Cancer and the Appeal of the Aging Peritoneal Microenvironment. Cancers (Basel) 2018; 10:E230. [PMID: 29996539 PMCID: PMC6070816 DOI: 10.3390/cancers10070230] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 12/22/2022] Open
Abstract
Age is one of the biggest risk factors for ovarian cancer. Older women have higher rates of diagnosis and death associated with the disease. In mouse models, it was shown that aged mice had greater tumor burden than their younger counterparts when intraperitoneally injected with ovarian tumor cells. While very few papers have been published looking at the direct link between ovarian cancer metastasis and age, there is a wealth of information on how age affects metastatic microenvironments. Mesothelial cells, the peritoneal extracellular matrix (ECM), fibroblasts, adipocytes and immune cells all exhibit distinct changes with age. The aged peritoneum hosts a higher number of senescent cells than its younger counterpart, in both the mesothelium and the stroma. These senescent cells promote an inflammatory profile and overexpress Matrix Metalloproteinases (MMPs), which remodel the ECM. The aged ECM is also modified by dysregulated collagen and laminin synthesis, increases in age-related crosslinking and increasing ovarian cancer invasion into the matrix. These changes contribute to a vastly different microenvironment in young and aged models for circulating ovarian cancer cells, creating a more welcoming “soil”.
Collapse
Affiliation(s)
- Elizabeth I Harper
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN 46617, USA.
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46617, USA.
- Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, IN 46617, USA.
| | - Emma F Sheedy
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46617, USA.
- Department of Mathematics, University of Notre Dame, South Bend, IN 46617, USA.
| | - M Sharon Stack
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN 46617, USA.
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46617, USA.
| |
Collapse
|
15
|
Ungvari Z, Valcarcel-Ares MN, Tarantini S, Yabluchanskiy A, Fülöp GA, Kiss T, Csiszar A. Connective tissue growth factor (CTGF) in age-related vascular pathologies. GeroScience 2017; 39:491-498. [PMID: 28875415 DOI: 10.1007/s11357-017-9995-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
Connective tissue growth factor (CTGF, also known as CCN2) is a matricellular protein expressed in the vascular wall, which regulates diverse cellular functions including cell adhesion, matrix production, structural remodeling, angiogenesis, and cell proliferation and differentiation. CTGF is principally regulated at the level of transcription and is induced by mechanical stresses and a number of cytokines and growth factors, including TGFβ. In this mini-review, the role of age-related dysregulation of CTGF signaling and its role in a range of macro- and microvascular pathologies, including pathogenesis of aorta aneurysms, atherogenesis, and diabetic retinopathy, are discussed. A potential role of CTGF and TGFβ in regulation and non-cell autonomous propagation of cellular senescence is also discussed.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Marta Noa Valcarcel-Ares
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gábor A Fülöp
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamas Kiss
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA.
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| |
Collapse
|
16
|
Chandrasekaran A, Idelchik MDPS, Melendez JA. Redox control of senescence and age-related disease. Redox Biol 2017; 11:91-102. [PMID: 27889642 PMCID: PMC5126126 DOI: 10.1016/j.redox.2016.11.005] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/10/2016] [Indexed: 12/17/2022] Open
Abstract
The signaling networks that drive the aging process, associated functional deterioration, and pathologies has captured the scientific community's attention for decades. While many theories exist to explain the aging process, the production of reactive oxygen species (ROS) provides a signaling link between engagement of cellular senescence and several age-associated pathologies. Cellular senescence has evolved to restrict tumor progression but the accompanying senescence-associated secretory phenotype (SASP) promotes pathogenic pathways. Here, we review known biological theories of aging and how ROS mechanistically control senescence and the aging process. We also describe the redox-regulated signaling networks controlling the SASP and its important role in driving age-related diseases. Finally, we discuss progress in designing therapeutic strategies that manipulate the cellular redox environment to restrict age-associated pathology.
Collapse
Affiliation(s)
- Akshaya Chandrasekaran
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, Albany, NY 12203, USA
| | | | - J Andrés Melendez
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, Albany, NY 12203, USA.
| |
Collapse
|
17
|
Jang JH, Chand HS, Bruse S, Doyle-Eisele M, Royer C, McDonald J, Qualls C, Klingelhutz AJ, Lin Y, Mallampalli R, Tesfaigzi Y, Nyunoya T. Connective Tissue Growth Factor Promotes Pulmonary Epithelial Cell Senescence and Is Associated with COPD Severity. COPD 2016; 14:228-237. [PMID: 28026993 DOI: 10.1080/15412555.2016.1262340] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The purpose of this study was to determine whether expression of connective tissue growth factor (CTGF) protein in chronic obstructive pulmonary disease (COPD) is consistent in humans and animal models of COPD and to investigate the role of this protein in lung epithelial cells. CTGF in lung epithelial cells of ex-smokers with COPD was compared with ex-smokers without COPD by immunofluorescence. A total of twenty C57Bl/6 mice and sixteen non-human primates (NHPs) were exposed to cigarette smoke (CS) for 4 weeks. Ten mice of these CS-exposed mice and eight of the CS-exposed NHPs were infected with H3N2 influenza A virus (IAV), while the remaining ten mice and eight NHPs were mock-infected with vehicle as control. Both mRNA and protein expression of CTGF in lung epithelial cells of mice and NHPs were determined. The effects of CTGF overexpression on cell proliferation, p16 protein, and senescence-associated β-galactosidase (SA-β-gal) activity were examined in cultured human bronchial epithelial cells (HBECs). In humans, CTGF expression increased with increasing COPD severity. We found that protein expression of CTGF was upregulated in lung epithelial cells in both mice and NHPs exposed to CS and infected with IAV compared to those exposed to CS only. When overexpressed in HBECs, CTGF accelerated cellular senescence accompanied by p16 accumulation. Both CTGF and p16 protein expression in lung epithelia are positively associated with the severity of COPD in ex-smokers. These findings show that CTGF is consistently expressed in epithelial cells of COPD lungs. By accelerating lung epithelial senescence, CTGF may block regeneration relative to epithelial cell loss and lead to emphysema.
Collapse
Affiliation(s)
- Jun-Ho Jang
- a Department of Medicine , University of Pittsburgh , Pittsburgh , PA , USA.,b VA Pittsburgh Healthcare System , Pittsburgh , PA , USA
| | - Hitendra S Chand
- c Department of Immunology , Herbert Wertheim College of Medicine, Florida International University Miami , Miami , FL , USA
| | | | - Melanie Doyle-Eisele
- e COPD Program, Lovelace Respiratory Research Institute , Albuquerque , NM , USA
| | - Christopher Royer
- e COPD Program, Lovelace Respiratory Research Institute , Albuquerque , NM , USA
| | - Jacob McDonald
- e COPD Program, Lovelace Respiratory Research Institute , Albuquerque , NM , USA
| | | | - Aloysius J Klingelhutz
- g Department of Microbiology , University of Iowa, Roy J. and Lucille A. Carver College of Medicine , Iowa City , IA , USA
| | - Yong Lin
- e COPD Program, Lovelace Respiratory Research Institute , Albuquerque , NM , USA
| | - Rama Mallampalli
- a Department of Medicine , University of Pittsburgh , Pittsburgh , PA , USA.,b VA Pittsburgh Healthcare System , Pittsburgh , PA , USA
| | - Yohannes Tesfaigzi
- e COPD Program, Lovelace Respiratory Research Institute , Albuquerque , NM , USA
| | - Toru Nyunoya
- a Department of Medicine , University of Pittsburgh , Pittsburgh , PA , USA.,b VA Pittsburgh Healthcare System , Pittsburgh , PA , USA
| |
Collapse
|
18
|
Serum from Varicose Patients Induces Senescence-Related Dysfunction of Vascular Endothelium Generating Local and Systemic Proinflammatory Conditions. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2069290. [PMID: 27994710 PMCID: PMC5141312 DOI: 10.1155/2016/2069290] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/25/2016] [Accepted: 11/01/2016] [Indexed: 11/17/2022]
Abstract
Although the role of endothelium in varicose vein development is indisputable, the effect of the pathology on biological properties of endothelial cells remains unclear. Here we examined if the presence of varicose veins affects senescence of endothelial cells (HUVECs) and, if so, what will be the local and systemic outcome of this effect. Experiments showed that HUVECs subjected to serum from varicose patients display improved proliferation, increased expression of senescence marker, SA-β-Gal, and increased generation of reactive oxygen species (ROS), as compared with serum from healthy donors. Both increased SA-β-Gal activity and ROS release were mediated by TGF-β1, the concentration of which in varicose serum was elevated and the activity of which in vitro was prevented using specific neutralizing antibody. Senescent HUVECs exposed to varicose serum generated increased amounts of ICAM-1, VCAM-1, P-selectin, uPA, PAI-1, and ET-1. Direct comparison of sera from varicose and healthy donors showed that pathological serum contained increased level of ICAM-1, VCAM-1, P-selectin, uPA, and ET-1. Calendar age of healthy subjects correlated positively with serum uPA and negatively with P-selectin. Age of varicose patients correlated positively with ICAM-1, VCAM-1, and ET-1. Collectively, our findings indicate that the presence of varicose veins causes a senescence-related dysfunction of vascular endothelium, which leads to the development of local and systemic proinflammatory environment.
Collapse
|
19
|
Cr(VI) induces premature senescence through ROS-mediated p53 pathway in L-02 hepatocytes. Sci Rep 2016; 6:34578. [PMID: 27698449 PMCID: PMC5048307 DOI: 10.1038/srep34578] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/15/2016] [Indexed: 12/23/2022] Open
Abstract
Hexavalent Chromium [Cr(VI)], which can be found of various uses in industries such as metallurgy and textile dying, can cause a number of human disease including inflammation and cancer. Unlike previous research that focused on Cr(VI)-induced oxidative damage and apoptosis, this study placed emphasis on premature senescence that can be induced by low-dose and long-term Cr(VI) exposure. We found Cr(VI) induced premature senescence in L-02 hepatocytes, as confirmed by increase in senescence associated-β-galactosidase (SA-β-Gal) activity. Cr(VI) stabilized p53 through phosphorylation at Ser15 and increased expression of p53-transcriptional target p21. Mechanism study revealed Cr(VI) targeted and inhibited mitochondrial respiratory chain complex (MRCC) I and II to enhance reactive oxygen species (ROS) production. By applying antioxidant Trolox, we also confirmed that ROS mediated p53 activation. A tetracycline-inducible lentiviral expression system containing shRNA to p53 was used to knockout p53. We found p53 could inhibit pro-survival genes B-cell lymphoma-2 (Bcl-2), myeloid leukemia-1 (Mcl-1) and S phase related cell cycle proteins cyclin-dependent kinase 2 (CDK2), Cyclin E to induce premature senescence, and the functional role of ROS in Cr(VI)-induced premature senescence is depend on p53. The results suggest that Cr(VI) has a role in premature senescence by promoting ROS-dependent p53 activation in L-02 hepatocytes.
Collapse
|
20
|
Vasilieva OV, Golubtzova NN, Filippov FN, Gunin AG. Connective tissue growth factor (CTGF) in the human dermis through ontogenesis. Russ J Dev Biol 2016. [DOI: 10.1134/s1062360416020089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
21
|
Chang AS, Hathaway CK, Smithies O, Kakoki M. Transforming growth factor-β1 and diabetic nephropathy. Am J Physiol Renal Physiol 2015; 310:F689-F696. [PMID: 26719364 DOI: 10.1152/ajprenal.00502.2015] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/24/2015] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is established to be involved in the pathogenesis of diabetic nephropathy. The diabetic milieu enhances oxidative stress and induces the expression of TGF-β1. TGF-β1 promotes cell hypertrophy and extracellular matrix accumulation in the mesangium, which decreases glomerular filtration rate and leads to chronic renal failure. Recently, TGF-β1 has been demonstrated to regulate urinary albumin excretion by both increasing glomerular permeability and decreasing reabsorption in the proximal tubules. TGF-β1 also increases urinary excretion of water, electrolytes and glucose by suppressing tubular reabsorption in both normal and diabetic conditions. Although TGF-β1 exerts hypertrophic and fibrogenic effects in diabetic nephropathy, whether suppression of the function of TGF-β1 can be an option to prevent or treat the complication is still controversial. This is partly because adrenal production of mineralocorticoids could be augmented by the suppression of TGF-β1. However, differentiating the molecular mechanisms for glomerulosclerosis from those for the suppression of the effects of mineralocorticoids by TGF-β1 may assist in developing novel therapeutic strategies for diabetic nephropathy. In this review, we discuss recent findings on the role of TGF-β1 in diabetic nephropathy.
Collapse
Affiliation(s)
- Albert S Chang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Catherine K Hathaway
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Oliver Smithies
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Masao Kakoki
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
22
|
Slemmons KK, Crose LES, Rudzinski E, Bentley RC, Linardic CM. Role of the YAP Oncoprotein in Priming Ras-Driven Rhabdomyosarcoma. PLoS One 2015; 10:e0140781. [PMID: 26496700 PMCID: PMC4619859 DOI: 10.1371/journal.pone.0140781] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 09/30/2015] [Indexed: 12/21/2022] Open
Abstract
Rhabdomyosarcoma (RMS), a cancer characterized by features of skeletal muscle histogenesis, is the most common soft tissue sarcoma of childhood and adolescence. Survival for high-risk groups is less than 30% at 5 years. RMS also occurs during adulthood, with a lower incidence but higher mortality. Recently, mutational profiling has revealed a correlation between activating Ras mutations in the embryonal (eRMS) and pleomorphic (pRMS) histologic variants of RMS, and a poorer outcome for those patients. Independently, the YAP transcriptional coactivator, an oncoprotein kept in check by the Hippo tumor suppressor pathway, is upregulated in eRMS. Here we show that YAP promotes cell proliferation and antagonizes apoptosis and myogenic differentiation of human RMS cells bearing oncogenic Ras mutations in cell culture studies in vitro and in murine xenografts in vivo. Pharmacologic inhibition of YAP by the benzoporphyrin derivative verteporfin decreased cell proliferation and tumor growth in vivo. To interrogate the temporal contribution of YAP in eRMS tumorigenesis, we used a primary human cell-based genetic model of Ras-driven RMS. Constitutively active YAP functioned as an early genetic lesion, permitting bypass of senescence and priming myoblasts to tolerate subsequent expression of hTERT and oncogenic Ras, which were necessary and sufficient to generate murine xenograft tumors mimicking RMS in vivo. This work provides evidence for cooperation between YAP and oncogenic Ras in RMS tumorigenesis, laying the foundation for preclinical co-targeting of these pathways.
Collapse
Affiliation(s)
- Katherine K. Slemmons
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Lisa E. S. Crose
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Erin Rudzinski
- Department of Laboratories, Seattle Children’s Hospital, Seattle, Washington, United States of America
| | - Rex C. Bentley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Corinne M. Linardic
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
23
|
Takata T, Motoo Y, Tomosugi N. Effect of Saikokeishito, a Kampo medicine, on hydrogen peroxide-induced premature senescence of normal human dermal fibroblasts. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2015; 12:495-503. [PMID: 25412667 DOI: 10.1016/s2095-4964(14)60052-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Saikokeishito (TJ-10) is a Kampo (traditional Japanese herbal) medicine, clinically used for hundreds of years in East Asia. Among its various mechanisms elucidated so far, TJ-10 inhibits the production of transforming growth factor-β1 (TGF-β1) and development of pancreatic fibrosis in vivo. Oxidative damage of normal human dermal fibroblasts (NHDFs) in the corium is a cause of human dermal senescence. Our aim was to determine whether TJ-10 protects NHDFs from premature senescence by hydrogen peroxide (H₂O₂). METHODS Premature senescence was induced in NHDFs by 200 μmol/L H₂O₂ for 4 h. Cell viability and the expressions of p53, AMP-activated protein kinase α1 (AMPKα1), AMPKα2, and 14-3-3 protein sigma (14-3-3 σ) were measured in NHDFs treated with TJ-10 for 48 h before exposure to H₂O₂for 4 h. RESULTS Cell viability after treatment with 200 μmol/L H₂O₂ for 4 h was similar (about 80%) to after pre-treatment with TJ-10. Ascorbic acid as a control did not protect NHDFs from damage by 200 μmol/L H₂O₂. Treatment with 200 μmol/L H₂O₂tended to up-regulate p53 and to down-regulate SIRT1 and AMPKα1, but had no effect on AMPKα2 and 14-3-3 σ expression. Pretreatment with TJ-10 inhibited H₂O₂-induced up-regulation of p53 and enhanced AMPKα1 expression. CONCLUSION It is suggested that Saikokeishito has a protective effect on oxidative stress-induced senescence of NHDFs.
Collapse
Affiliation(s)
- Takanobu Takata
- Medical Research Institute of Kanazawa Medical University, Ishikawa, Japan
| | - Yoshiharu Motoo
- Medical Research Institute of Kanazawa Medical University, Ishikawa, Japan; E-mail:
| | - Naohisa Tomosugi
- Medical Research Institute of Kanazawa Medical University, Ishikawa, Japan
| |
Collapse
|
24
|
Crose LES, Galindo KA, Kephart JG, Chen C, Fitamant J, Bardeesy N, Bentley RC, Galindo RL, Chi JTA, Linardic CM. Alveolar rhabdomyosarcoma-associated PAX3-FOXO1 promotes tumorigenesis via Hippo pathway suppression. J Clin Invest 2013; 124:285-96. [PMID: 24334454 DOI: 10.1172/jci67087] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 10/10/2013] [Indexed: 02/06/2023] Open
Abstract
Alveolar rhabdomyosarcoma (aRMS) is an aggressive sarcoma of skeletal muscle characterized by expression of the paired box 3-forkhead box protein O1 (PAX3-FOXO1) fusion oncogene. Despite its discovery nearly two decades ago, the mechanisms by which PAX3-FOXO1 drives tumor development are not well characterized. Previously, we reported that PAX3-FOXO1 supports aRMS initiation by enabling bypass of cellular senescence checkpoints. We have now found that this bypass occurs in part through PAX3-FOXO1-mediated upregulation of RASSF4, a Ras-association domain family (RASSF) member. RASSF4 expression was upregulated in PAX3-FOXO1-positive aRMS cell lines and tumors. Enhanced RASSF4 expression promoted cell cycle progression, senescence evasion, and tumorigenesis through inhibition of the Hippo pathway tumor suppressor MST1. We also found that the downstream Hippo pathway target Yes-associated protein 1 (YAP), which is ordinarily restrained by Hippo signaling, was upregulated in RMS tumors. These data suggest that Hippo pathway dysfunction promotes RMS. This work provides evidence for Hippo pathway suppression in aRMS and demonstrates a progrowth role for RASSF4. Additionally, we identify a mechanism used by PAX3-FOXO1 to inhibit MST1 signaling and promote tumorigenesis in aRMS.
Collapse
|
25
|
Mikuła-Pietrasik J, Sosińska P, Janus J, Rubiś B, Brewińska-Olchowik M, Piwocka K, Książek K. Bystander senescence in human peritoneal mesothelium and fibroblasts is related to thrombospondin-1-dependent activation of transforming growth factor-β1. Int J Biochem Cell Biol 2013; 45:2087-96. [PMID: 23871936 DOI: 10.1016/j.biocel.2013.07.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 06/13/2013] [Accepted: 07/09/2013] [Indexed: 11/29/2022]
Abstract
Senescence bystander effect refers to a phenomenon in which senescent cells elicit the development of senescence phenotype in their nearby young counterparts. In this paper we examined the mechanism of senescence bystander effect triggered by senescent human peritoneal mesothelial cells (HPMCs) in proliferating HPMCs and peritoneal fibroblasts (HPFBs). The results showed that conditioned medium (CM) derived from senescent HPMCs elicited a senescence response (growth inhibition coupled with increased expression of senescence-associated β-galactosidase and accumulation of histone γ-H2A.X) in either early-passage HPMCs or HPFBs. Samples of CM from senescent HPMCs contained increased amounts of numerous soluble mediators of which only transforming growth factor-β1 (TGF-β1) was able to induce senescence phenotype in the both types of peritoneal cells, likely through an induction of reactive oxygen species (ROS) and p38 mitogen-activated protein kinase (MAPK). At the same time, senescent HPMCs released increased amounts of thrombospondin-1 (TSP-1), a major activator of TGF-β1. Significantly, TSP-1 itself was unable to induce senescence phenotype in HPMCs or in HPFBs. The experiments employing anti-TSP-1 antibodies and specific TSP-1 blocking peptide revealed that neutralization of TSP-1 in CM prevented TGF-β1-dependent development of senescence phenotype. Collectively, our findings indicate that senescent HPMCs exhibit senescence-promoting activity toward neighboring young cells (HPMCs and HPFBs), and this effect is, at least partly, related to TSP-1-dependent activation and further ROS- and p38 MAPK-related activity of TGF-β1.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Pathophysiology, Laboratory of Gerontology, Poznań University of Medical Sciences, Święcickiego 6 Str., 60-781 Poznań, Poland.
| | | | | | | | | | | | | |
Collapse
|
26
|
Tan JTM, McLennan SV, Williams PF, Rezaeizadeh A, Lo LWY, Bonner JG, Twigg SM. Connective tissue growth factor/CCN-2 is upregulated in epididymal and subcutaneous fat depots in a dietary-induced obesity model. Am J Physiol Endocrinol Metab 2013; 304:E1291-302. [PMID: 23571711 DOI: 10.1152/ajpendo.00654.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Connective tissue growth factor (CTGF), also known as CCN-2, is a cysteine-rich secreted protein that is involved in a range of biological processes, including regulation of cell growth and differentiation. Our previous in vitro studies have shown that CCN-2 inhibits adipocyte differentiation, although whether CCN-2 is regulated in vivo in adipogenesis is undetermined and was investigated in this study. C57BL/6 male mice were fed either standard laboratory chow (ND) or a diet high in fat (HFD; 45% fat) for 15 or 24 wk. HFD animals that gained >5 g in weight (termed HFD-fat) were insulin resistant and were compared with HFD-fed animals, which failed to gain weight (termed HFD-lean). HFD-fat mice had significantly increased CCN-2 mRNA levels in both the subcutaneous and epididymal fat pads, whereas CCN-2 mRNA was not induced in the epididymal site in HFD-lean mice. Also in HFD-fed animals, epididymal CCN-2 mRNA correlated positively with key genes involved in adipocyte differentiation, adiponectin and PPARγ (P < 0.001 and P < 0.002, respectively). Additionally, epididymal CCN-2 mRNA correlated positively with two markers of tissue turnover, PAI-1 in HFD-fat mice only and TIMP-1, but only in the HFD-lean mice. Collectively, these findings suggest that CCN-2 plays a role in adipocyte differentiation in vivo and thus in the pathogenesis of obesity linked with insulin resistance.
Collapse
Affiliation(s)
- Joanne T M Tan
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; and
| | | | | | | | | | | | | |
Collapse
|
27
|
Connective tissue growth factor regulates adipocyte differentiation of mesenchymal stromal cells and facilitates leukemia bone marrow engraftment. Blood 2013; 122:357-66. [PMID: 23741006 DOI: 10.1182/blood-2012-06-437988] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are a major component of the leukemia bone marrow (BM) microenvironment. Connective tissue growth factor (CTGF) is highly expressed in MSCs, but its role in the BM stroma is unknown. Therefore, we knocked down (KD) CTGF expression in human BM-derived MSCs by CTGF short hairpin RNA. CTGF KD MSCs exhibited fivefold lower proliferation compared with control MSCs and had markedly fewer S-phase cells. CTGF KD MSCs differentiated into adipocytes at a sixfold higher rate than controls in vitro and in vivo. To study the effect of CTGF on engraftment of leukemia cells into BM, an in vivo model of humanized extramedullary BM (EXM-BM) was developed in NOD/SCID/IL-2rg(null) mice. Transplanted Nalm-6 or Molm-13 human leukemia cells engrafted at a threefold higher rate in adipocyte-rich CTGF KD MSC-derived EXM-BM than in control EXM-BM. Leptin was found to be highly expressed in CTGF KD EXM-BM and in BM samples of patients with acute myeloid and acute lymphoblastic leukemia, whereas it was not expressed in normal controls. Given the established role of the leptin receptor in leukemia cells, the data suggest an important role of CTGF in MSC differentiation into adipocytes and of leptin in homing and progression of leukemia.
Collapse
|
28
|
Mesothelial cell: a multifaceted model of aging. Ageing Res Rev 2013; 12:595-604. [PMID: 23415666 DOI: 10.1016/j.arr.2013.01.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 01/13/2023]
Abstract
Human peritoneal mesothelial cells (HPMCs) dominate within the peritoneal cavity and thus play a central role in a variety of intraperitoneal processes, including the transport of water and solutes, inflammation, host response, angiogenesis, and extracellular matrix remodeling. In addition, they contribute to the development of abdominal adhesions, peritonitis, endometriosis, cancer cell metastases, and peritoneal dialysis complications. For less than a decade the primary cultures of omental HPMCs have also been used as an experimental tool in studies on cellular aging. This paper provides the first comprehensive overview of the current state of art on molecular mechanisms underlying HPMC senescence in vitro. Special attention is paid to the causes of the very fast dynamics of HPMC senescence, and in particular to the role of non-telomeric DNA damage, the autocrine activity of TGF-β1, and the causative effects of oxidative stress. In addition, some clinical manifestations of HPMC senescence will be discussed, including its interplay with organismal aging, peritoneal dialysis, and cancer progression.
Collapse
|
29
|
Li Y, Jian Z, Yang ZY, Chen L, Wang XF, Ma RY, Xiao YB. Increased Expression of Connective Tissue Growth Factor and Transforming Growth Factor-Beta-1 in Atrial Myocardium of Patients with Chronic Atrial Fibrillation. Cardiology 2013; 124:233-40. [DOI: 10.1159/000347126] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 01/11/2013] [Indexed: 01/28/2023]
|
30
|
Biological effects of cigarette smoke in cultured human retinal pigment epithelial cells. PLoS One 2012; 7:e48501. [PMID: 23155386 PMCID: PMC3498276 DOI: 10.1371/journal.pone.0048501] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 09/26/2012] [Indexed: 12/24/2022] Open
Abstract
The goal of the present study was to determine whether treatment with cigarette smoke extract (CSE) induces cell loss, cellular senescence, and extracellular matrix (ECM) synthesis in primary human retinal pigment epithelial (RPE) cells. Primary cultured human RPE cells were exposed to 2, 4, 8, and 12% of CSE concentration for 24 hours. Cell loss was detected by cell viability assay. Lipid peroxidation was assessed by loss of cis-parinaric acid (PNA) fluorescence. Senescence-associated ß-galactosidase (SA-ß-Gal) activity was detected by histochemical staining. Expression of apolipoprotein J (Apo J), connective tissue growth factor (CTGF), fibronectin, and laminin were examined by real-time PCR, western blot, or ELISA experiments. The results showed that exposure of cells to 12% of CSE concentration induced cell death, while treatment of cells with 2, 4, and 8% CSE increased lipid peroxidation. Exposure to 8% of CSE markedly increased the number of SA-ß-Gal positive cells to up to 82%, and the mRNA expression of Apo J, CTGF, and fibronectin by approximately 3–4 fold. Treatment with 8% of CSE also increased the protein expression of Apo J and CTGF and the secretion of fibronectin and laminin. Thus, treatment with CSE can induce cell loss, senescent changes, and ECM synthesis in primary human RPE cells. It may be speculated that cigarette smoke could be involved in cellular events in RPE cells as seen in age-related macular degeneration.
Collapse
|
31
|
Abdelmohsen K, Srikantan S, Kang MJ, Gorospe M. Regulation of senescence by microRNA biogenesis factors. Ageing Res Rev 2012; 11:491-500. [PMID: 22306790 DOI: 10.1016/j.arr.2012.01.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/07/2012] [Accepted: 01/19/2012] [Indexed: 12/16/2022]
Abstract
Senescence represents a state of indefinite growth arrest in cells that have reached the end of their replicative life span, have become damaged, or express aberrant levels of cancer-related proteins. While senescence is widely considered to represent a tumor-suppressive mechanism, the accumulation of senescent cells in tissues of older organisms is believed to underlie age-associated losses in physiologic function and age-related diseases. With the emergence of microRNAs (miRNAs) as a major class of molecular regulators of senescence, we review the transcriptional and post-transcriptional factors that control senescence-associated microRNA biosynthesis. Focusing on their enhancement or repression of senescence, we describe the transcription factors that govern the synthesis of primary (pri-)miRNAs, the proteins that control the nuclear processing of pri-miRNAs into precursor (pre-)miRNAs, including RNA editing enzymes, RNases, and RNA helicases, and the cytoplasmic proteins that affect the final processing of pre-miRNAs into mature miRNAs. We discuss how miRNA biogenesis proteins promote or inhibit senescence, and thus influence the senescent phenotype that affects normal tissue function and pathology.
Collapse
Affiliation(s)
- Kotb Abdelmohsen
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, MD 21224, USA.
| | | | | | | |
Collapse
|
32
|
Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-κB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal 2012; 24:835-45. [PMID: 22182507 DOI: 10.1016/j.cellsig.2011.12.006] [Citation(s) in RCA: 478] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 12/04/2011] [Indexed: 11/17/2022]
Abstract
The major hallmark of cellular senescence is an irreversible cell cycle arrest and thus it is a potent tumor suppressor mechanism. Genotoxic insults, e.g. oxidative stress, are important inducers of the senescent phenotype which is characterized by an accumulation of senescence-associated heterochromatic foci (SAHF) and DNA segments with chromatin alterations reinforcing senescence (DNA-SCARS). Interestingly, senescent cells secrete pro-inflammatory factors and thus the condition has been called the senescence-associated secretory phenotype (SASP). Emerging data has revealed that NF-κB signaling is the major signaling pathway which stimulates the appearance of SASP. It is known that DNA damage provokes NF-κB signaling via a variety of signaling complexes containing NEMO protein, an NF-κB essential modifier, as well as via the activation of signaling pathways of p38MAPK and RIG-1, retinoic acid inducible gene-1. Genomic instability evoked by cellular stress triggers epigenetic changes, e.g. release of HMGB1 proteins which are also potent enhancers of inflammatory responses. Moreover, environmental stress and chronic inflammation can stimulate p38MAPK and ceramide signaling and induce cellular senescence with pro-inflammatory responses. On the other hand, two cyclin-dependent kinase inhibitors, p16INK4a and p14ARF, are effective inhibitors of NF-κB signaling. We will review in detail the signaling pathways which activate NF-κB signaling and trigger SASP in senescent cells.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland.
| | | | | |
Collapse
|
33
|
Abstract
The characterization of functional CD8(+) inhibitory or regulatory T cells and their gene regulation remains a critical challenge in the field of tolerance and autoimmunity. Investigating the genes induced in regulatory cells and the regulatory networks and pathways that underlie mechanisms of immune resistance and prevent apoptosis in the CD8(+) T cell compartment are crucial to understanding tolerance mechanisms in systemic autoimmunity. Little is currently known about the genetic control that governs the ability of CD8(+) Ti or regulatory cells to suppress anti-DNA Ab production in B cells. Silencing genes with siRNA or shRNA and overexpression of genes with lentiviral cDNA transduction are established approaches to identifying and understanding the function of candidate genes in tolerance and immunity. Elucidation of interactions between genes and proteins, and their synergistic effects in establishing cell-cell cross talk, including receptor modulation/antagonism, are essential for delineating the roles of these cells. In this review, we will examine recent reports which describe the modulation of cells from lupus prone mice or lupus patients to confer anti-inflammatory and protective gene expression and novel associated phenotypes. We will highlight recent findings on the role of selected genes induced by peptide tolerance in CD8(+) Ti.
Collapse
|
34
|
Davalos AR, Coppe JP, Campisi J, Desprez PY. Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis Rev 2010; 29:273-83. [PMID: 20390322 PMCID: PMC2865636 DOI: 10.1007/s10555-010-9220-9] [Citation(s) in RCA: 463] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cellular senescence, which is associated with aging, is a process by which cells enter a state of permanent cell cycle arrest, therefore constituting a potent tumor suppressive mechanism. Recent studies show that, despite the beneficial effects of cellular senescence, senescent cells can also exert harmful effects on the tissue microenvironment. The most significant of these effects is the acquisition of a senescent-associated secretory phenotype (SASP), which entails a striking increase in the secretion of pro-inflammatory cytokines. Here, we summarize our knowledge of the SASP and the impact it has on tissue microenvironments and ability to stimulate tumor progression.
Collapse
Affiliation(s)
- Albert R. Davalos
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945 USA
| | - Jean-Philippe Coppe
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945 USA
| | - Judith Campisi
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945 USA
| | - Pierre-Yves Desprez
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945 USA
- California Pacific Medical Center, Research Institute, 475 Brannan Street, San Francisco, CA 94107 USA
| |
Collapse
|
35
|
Coppé JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2010; 5:99-118. [PMID: 20078217 DOI: 10.1146/annurev-pathol-121808-102144] [Citation(s) in RCA: 3465] [Impact Index Per Article: 231.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cellular senescence is a tumor-suppressive mechanism that permanently arrests cells at risk for malignant transformation. However, accumulating evidence shows that senescent cells can have deleterious effects on the tissue microenvironment. The most significant of these effects is the acquisition of a senescence-associated secretory phenotype (SASP) that turns senescent fibroblasts into proinflammatory cells that have the ability to promote tumor progression.
Collapse
Affiliation(s)
- Jean-Philippe Coppé
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | | | | | | |
Collapse
|
36
|
Westhoff JH, Schildhorn C, Jacobi C, Hömme M, Hartner A, Braun H, Kryzer C, Wang C, von Zglinicki T, Kränzlin B, Gretz N, Melk A. Telomere shortening reduces regenerative capacity after acute kidney injury. J Am Soc Nephrol 2009; 21:327-36. [PMID: 19959722 DOI: 10.1681/asn.2009010072] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Telomeres of most somatic cells progressively shorten, compromising the regenerative capacity of human tissues during aging and chronic diseases and after acute injury. Whether telomere shortening reduces renal regeneration after acute injury is unknown. Here, renal ischemia-reperfusion injury led to greater impairment of renal function and increased acute and chronic histopathologic damage in fourth-generation telomerase-deficient mice compared with both wild-type and first-generation telomerase-deficient mice. Critically short telomeres, increased expression of the cell-cycle inhibitor p21, and more apoptotic renal cells accompanied the pronounced damage in fourth-generation telomerase-deficient mice. These mice also demonstrated significantly reduced proliferative capacity in tubular, glomerular, and interstitial cells. These data suggest that critical telomere shortening in the kidney leads to increased senescence and apoptosis, thereby limiting regenerative capacity in response to injury.
Collapse
Affiliation(s)
- Jens H Westhoff
- Children's Hospital, Medical School Hannover, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kakoki M, Smithies O. The kallikrein-kinin system in health and in diseases of the kidney. Kidney Int 2009; 75:1019-30. [PMID: 19190676 DOI: 10.1038/ki.2008.647] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Since kallikrein was discovered as a vasodilatory substance in human urine, the kallikrein-kinin system (KKS) has been considered to play a physiological role in controlling blood pressure. Gene targeting experiments in mice in which the KKS has been inactivated to varying degrees have, however, questioned this role, because basal blood pressures are not altered. Rather, these experiments have shown that the KKS has a different and important role in preventing changes associated with normal senescence in mice, and in reducing the nephropathy and accelerated senescence-associated phenotypes induced in mice by diabetes. Other experiments have shown that the KKS suppresses mitochondrial respiration, partly by nitric oxide and prostaglandins, and that this suppression may be a key to understanding how the KKS influences senescence-related diseases. Here we review the logical progression and experimental data leading to these conclusions, and discuss their relevance to human conditions.
Collapse
Affiliation(s)
- Masao Kakoki
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525, USA.
| | | |
Collapse
|
38
|
Muller M. Cellular senescence: molecular mechanisms, in vivo significance, and redox considerations. Antioxid Redox Signal 2009; 11:59-98. [PMID: 18976161 DOI: 10.1089/ars.2008.2104] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cellular senescence is recognized as a critical cellular response to prolonged rounds of replication and environmental stresses. Its defining characteristics are arrested cell-cycle progression and the development of aberrant gene expression with proinflammatory behavior. Whereas the mechanistic events associated with senescence are generally well understood at the molecular level, the impact of senescence in vivo remains to be fully determined. In addition to the role of senescence as an antitumor mechanism, this review examines cellular senescence as a factor in organismal aging and age-related diseases, with particular emphasis on aberrant gene expression and abnormal paracrine signaling. Senescence as an emerging factor in tissue remodeling, wound repair, and infection is considered. In addition, the role of oxidative stress as a major mediator of senescence and the role of NAD(P)H oxidases and changes to intracellular GSH/GSSG status are reviewed. Recent findings indicate that senescence and the behavior of senescent cells are amenable to therapeutic intervention. As the in vivo significance of senescence becomes clearer, the challenge will be to modulate the adverse effects of senescence without increasing the risks of other diseases, such as cancer. The uncoupled relation between cell-cycle arrest and the senescent phenotype suggests that this is an achievable outcome.
Collapse
Affiliation(s)
- Michael Muller
- Centre for Education and Research on Ageing, ANZAC Research Institute, University of Sydney, Concord RG Hospital, Concord, Sydney, Australia.
| |
Collapse
|
39
|
Hypoxia/reoxygenation induces CTGF and PAI-1 in cultured human retinal pigment epithelium cells. Exp Eye Res 2008; 88:889-99. [PMID: 19118548 DOI: 10.1016/j.exer.2008.11.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Revised: 08/02/2008] [Accepted: 11/21/2008] [Indexed: 11/23/2022]
Abstract
Early age-related macular degeneration (AMD) is characterized by thickening of Bruch's membrane due to the accumulation of extracellular matrix (ECM). This finding could be related to hypoxia of the retinal pigment epithelium (RPE). In the present study, we investigated the effects of hypoxia and reoxygenation on the expression of connective tissue growth factor (CTGF), plasminogen activator inhibitor-1 (PAI-1), collagen type IV (Col IV) and fibronectin (Fn) in cultured human RPE cells. Cultured human RPE cells were kept for 12-36h under hypoxic conditions (1% O(2)). Reoxygenation was conducted for 24h. Hypoxia-mediated CTGF and PAI-1 expression were analyzed by using immunohistochemistry, Northern and Western blot analysis. Actinomycin D was added to examine whether hypoxia induces the transcription of CTGF and PAI-1 mRNA. Furthermore, cells were transfected with siRNA against hypoxia-inducible factor-1alpha (HIF-1alpha) and kept under hypoxic conditions. The effects of antioxidants on hypoxia/reoxygenation-mediated CTGF and PAI-1 expression were tested by real-time PCR analysis. Production of Col IV and Fn were investigated by real-time PCR and Western blot analysis. Both hypoxia and hypoxia/reoxygenation increased the expression of CTGF, PAI-1, Col IV and Fn. Actinomycin D prevented the new transcription of CTGF and PAI-1 mRNA by hypoxia. Using siRNA against HIF-1alpha, the hypoxia-mediated increase of CTGF and PAI-1 was inhibited. Antioxidants attenuated the reoxygenation-mediated increase of CTGF and PAI-1. The process of hypoxia/reoxygenation in the RPE may lead to an increase of ECM in the RPE and thus may contribute to the accumulation of ECM in Bruch's membrane.
Collapse
|
40
|
Tan JTM, McLennan SV, Song WW, Lo LWY, Bonner JG, Williams PF, Twigg SM. Connective tissue growth factor inhibits adipocyte differentiation. Am J Physiol Cell Physiol 2008; 295:C740-51. [PMID: 18596209 DOI: 10.1152/ajpcell.00333.2007] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Adipocyte differentiation is a key process implicated in the pathogenesis of obesity and insulin resistance. Its regulation is triggered by a cascade of transcription factors, including the CCAAT/enhancer binding proteins (C/EBPs) and peroxisome proliferator-activated receptor-gamma (PPARgamma). Growth factors such as transforming growth factor-beta1 (TGF-beta1) are known to inhibit adipocyte differentiation in vitro, via the C/EBP pathway, and in vivo, but whether a downstream mediator of TGF-beta1, connective tissue growth factor (CTGF), also known as CCN2, has a similar role is unknown. Mouse 3T3-L1 cells were differentiated into adipocytes by using standard methods, and effects and regulation of CTGF were studied. Intervention with recombinant human CTGF during differing stages of differentiation caused an inhibition in the development of the adipocyte phenotype, according to the gene expression of the differentiation markers adiponectin and PPARgamma, as well as suppression of lipid accumulation and expression of the lipogenic enzyme glycerol-3-phosphate dehydrogenase. Whereas CTGF gene expression promptly fell by 90% as 3T3-L1 preadipocytes differentiated into mature adipocytes, CTGF mRNA expression was induced by added TGF-beta1. CTGF applied to cells early in the course of differentiation inhibited total cell protein levels and nuclear localization of the beta-isoform of C/EBP (C/EBP-beta) and, subsequently, total cell C/EBP-alpha levels. CTGF also inhibited the adipocyte differentiation program in primary cultures of mouse preadipocytes. Expression of CTGF mRNA was twofold higher in the central fat depots of mice compared with subcutaneous fat, suggesting a potential role for CTGF in vivo. In summary, these data show that CTGF inhibits the adipocyte differentiation program.
Collapse
Affiliation(s)
- Joanne T M Tan
- Discipline of Medicine, University of Sydney, Sydney, NSW 2006, Australia
| | | | | | | | | | | | | |
Collapse
|
41
|
Borlon C, Weemaels G, Godard P, Debacq-Chainiaux F, Lemaire P, Deroanne C, Toussaint O. Expression profiling of senescent-associated genes in human dermis from young and old donors. Proof-of-concept study. Biogerontology 2008; 9:197-208. [PMID: 18270802 DOI: 10.1007/s10522-008-9127-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Accepted: 01/29/2008] [Indexed: 11/26/2022]
Abstract
It is often described that it is difficult to really discriminate the cause of intrinsic skin aging. The aim of this study was to compare the profiles of expression of senescence-associated genes in biopsies of dermis from young and old human donors. TGF-beta1 was up-regulated in the dermis of old donors as well as the TGF-beta1-regulated genes. The anti-oxidant enzymes Selenium-dependent Glutathione peroxidase and Glutatione S-Transferase Theta 1 were also up-regulated in old dermis as well as Tumor Necrosis Factor Receptor Superfamily 1A. None of these genes had altered expression level in skin fibroblasts embedded in a collagen matrix and exposed to sublethal doses of UVB, suggesting their involvement in intrinsic aging. This study represents a proof-of-concept of larger whole transcriptome studies where all avenues should be used to subtract changes in gene expression due to extrinsic aging from changes potentially due to intrinsic aging.
Collapse
Affiliation(s)
- Céline Borlon
- Research Unit on Cellular Biology (URBC), Department of Biology, Faculty of Sciences, University of Namur (FUNDP), Rue de Bruxelles, 61, 5000 Namur, Belgium
| | | | | | | | | | | | | |
Collapse
|
42
|
Winkler JL, Kedees MH, Teitelman G. Adenoviral-mediated inhibition of connective tissue growth factor in Rat-2 cells. Biochem Biophys Res Commun 2007; 358:209-14. [PMID: 17481581 DOI: 10.1016/j.bbrc.2007.04.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 04/16/2007] [Accepted: 04/16/2007] [Indexed: 10/23/2022]
Abstract
Connective tissue growth factor (CTGF) is a profibrotic factor shown to induce extracellular matrix production and angiogenesis, two processes involved in the development of diabetic retinopathy (DR). In this study we tested the effect of a recombinant adenovirus encoding for a CTGF antisense oligonucleotide (rAdASO) on the levels of transforming growth factor-beta (TGF-beta) induced expression of CTGF in Rat-2 fibroblasts. Using semi-quantitative RT-PCR, there was a 2-fold increase in CTGF message induced by TGF-beta. Western blot and immunocytochemical analyses revealed a significant increase in CTGF protein level. This upregulation of CTGF by TGF-beta was inhibited by infection with rAdASO. These findings indicate that infection of the Rat-2 cells with rAdASO was effective in decreasing TGF-beta-induced CTGF expression. These results indicate that this viral vector might have therapeutic potential to control elevated CTGF levels that occur in DR.
Collapse
Affiliation(s)
- Jennifer L Winkler
- Department of Anatomy and Cell Biology, State University of New York (SUNY) Downstate Medical Center, Room 2-94, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | | | | |
Collapse
|
43
|
Kakoki M, McGarrah RW, Kim HS, Smithies O. Bradykinin B1 and B2 receptors both have protective roles in renal ischemia/reperfusion injury. Proc Natl Acad Sci U S A 2007; 104:7576-81. [PMID: 17452647 PMCID: PMC1855073 DOI: 10.1073/pnas.0701617104] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
To explore the role of the kallikrein-kinin system in relation to ischemia/reperfusion injury in the kidney, we generated mice lacking both the bradykinin B1 and B2 receptor genes (B1RB2R-null, Bdkrb1-/-/Bdkrb2-/-) by deleting the genomic region encoding the two receptors. In 4-month-old mice, blood pressures were not significantly different among B1RB2R-null, B2R-null (Bdkrb2-/-), and WT mice. After 30 min of bilateral renal artery occlusion and 24 h of reperfusion, mortality rates, renal histological and functional changes, 8-hydroxy-2'-deoxyguanosine levels in total DNA, mtDNA deletions, and the number of TUNEL-positive cells in the kidneys increased progressively in the following order (from lowest to highest): WT, B2R-null, and B1RB2R-null mice. Increases in mRNA levels of TGF-beta1, connective tissue growth factor, and endothelin-1 after ischemia/reperfusion injury were also exaggerated in the same order (from lowest to highest): WT, B2R-null, and B1RB2R-null. Thus, both the B1 and B2 bradykinin receptors play an important role in reducing DNA damage, apoptosis, morphological and functional kidney changes, and mortality during renal ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Masao Kakoki
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525
- *To whom correspondence should be addressed. E-mail: or
| | - Robert W. McGarrah
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525
| | - Hyung-Suk Kim
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525
| | - Oliver Smithies
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525
- *To whom correspondence should be addressed. E-mail: or
| |
Collapse
|
44
|
Huang Z, Taylor L, Liu B, Yu J, Polgar P. Modulation by bradykinin of angiotensin type 1 receptor-evoked RhoA activation of connective tissue growth factor expression in human lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2006; 290:L1291-9. [PMID: 16684954 DOI: 10.1152/ajplung.00443.2005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mechanisms regulating the opposing physiological actions of bradykinin (BK) and angiotensin II (AngII) are not well understood. Here we investigate signaling interactions between these two effectors. Connective tissue growth factor (CTGF) expression in IMR-90, human lung fibroblasts, is used as the endpoint target. In these cells the BK B2 receptor (BKB2R) is expressed constitutively, while no binding of AngII is detected. An inducible expression system is used to insert AngII receptor 1 (AT1R) and to obtain a signal level in response to AngII at the magnitude of BK. AngII and BK activate G protein-coupled targets, arachidonate release from cellular phospholipid stores, and intracellular phosphatidylinositol turnover equally. Both activate ERK, JNK, and p38 equally. However, AngII activates, whereas BK inactivates, RhoA. AngII induces a rapid (1 h) CTGF mRNA expression. RhoA siRNA and RhoA activation inhibitor, Y-27632, markedly reduce the AngII effect. Simultaneous treatment with BK and AngII attenuates the AT1R action. Additionally, BK in the absence of AngII lowers CTGF mRNA expression below basal levels over a span of 4 h. An AT1R/BKB2R chimera lacking heterotrimeric G protein coupling continues to activate MAP kinases to the same extent as wild-type (WT) AT1R and BKB2R. However, the increase of CTGF mRNA expression by this mutant is low, almost identical with that obtained by the simultaneous treatment of the WT AT1R-expressing cells with BK and AngII. In this context the chimeric receptor displays the characteristics of both receptors. These data demonstrate that, in human lung fibroblasts, BK modulates the action of AngII through the small G protein RhoA, but in a Galphai/Galphaq-independent manner.
Collapse
Affiliation(s)
- Zhenhua Huang
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
45
|
Kakoki M, Kizer CM, Yi X, Takahashi N, Kim HS, Bagnell CR, Edgell CJS, Maeda N, Jennette JC, Smithies O. Senescence-associated phenotypes in Akita diabetic mice are enhanced by absence of bradykinin B2 receptors. J Clin Invest 2006; 116:1302-9. [PMID: 16604193 PMCID: PMC1430357 DOI: 10.1172/jci26958] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Accepted: 02/14/2006] [Indexed: 12/20/2022] Open
Abstract
We have previously reported that genetically increased angiotensin-converting enzyme levels, or absence of the bradykinin B2 receptor, increase kidney damage in diabetic mice. We demonstrate here that this is part of a more general phenomenon - diabetes and, to a lesser degree, absence of the B2 receptor, independently but also largely additively when combined, enhance senescence-associated phenotypes in multiple tissues. Thus, at 12 months of age, indicators of senescence (alopecia, skin atrophy, kyphosis, osteoporosis, testicular atrophy, lipofuscin accumulation in renal proximal tubule and testicular Leydig cells, and apoptosis in the testis and intestine) are virtually absent in WT mice, detectable in B2 receptor-null mice, clearly apparent in mice diabetic because of a dominant mutation (Akita) in the Ins2 gene, and most obvious in Akita diabetic plus B2 receptor-null mice. Renal expression of several genes that encode proteins associated with senescence and/or apoptosis (TGF-beta1, connective tissue growth factor, p53, alpha-synuclein, and forkhead box O1) increases in the same progression. Concomitant increases occur in 8-hydroxy-2'-deoxyguanosine, point mutations and deletions in kidney mitochondrial DNA, and thiobarbituric acid-reactive substances in plasma, together with decreases in the reduced form of glutathione in erythrocytes. Thus, absence of the bradykinin B2 receptor increases the oxidative stress, mitochondrial DNA damage, and many senescence-associated phenotypes already present in untreated Akita diabetic mice.
Collapse
Affiliation(s)
- Masao Kakoki
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7525, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zhu YH, Zhang CQ, Zhao YA, Chu YL, Sun CG. Construction and sequence analysis of connective tissue growth factor targeted recombinant plasmids by RNA interference. Shijie Huaren Xiaohua Zazhi 2005; 13:2880-2883. [DOI: 10.11569/wcjd.v13.i24.2880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct the recombinant plasmids expressing connective tissue growth factor (CTGF) short hairpin RNA (shRNA) by pEGFP plasmid vector.
METHODS: Three pairs of two DNA sequences containing small hairpin structure were designed and synthesized respectively, and then were formed into complementary chains by annealing. The obtained products were inserted into plasmid vector pEGFP containing U6 promoter. Then the recombinant plasmids were transformed into DH5α strain. Finally the plasmids that identified by restriction enzyme were used for sequence analysis.
RESULTS: The CTGF shRNA expression frames were successfully inserted into the plasmid vector pEGFP, and the shRNA coding sequences of the 3 obtained recombinant plasmids were consistent with the designed fragments. The cloned recombinants were identified and confirmed by endonuclease digestion and sequence analysis.
CONCLUSION: The recombinant plasmids of shRNA for CTGF are successfully constructed.
Collapse
|
47
|
Nørsett KG, Laegreid A, Langaas M, Wörlund S, Fossmark R, Waldum HL, Sandvik AK. Molecular characterization of rat gastric mucosal response to potent acid inhibition. Physiol Genomics 2005; 22:24-32. [PMID: 15827235 DOI: 10.1152/physiolgenomics.00245.2004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Potent acid inhibition with proton pump inhibitors (PPIs) is widely used in clinical medicine, especially for gastroesophageal reflux disease. PPIs cause profound changes in the intragastric environment with near-neutral pH and increase serum concentration of the gastric secretagogue hormone gastrin. Long-term hypergastrinemia increases mucosal thickness and enterochromaffin-like cell density in gastric corpus mucosa and results in development of gastric carcinoids in experimental animals. Our aim was to study responses to potent acid inhibition by characterizing genome-wide gene expression changes in gastric corpus mucosa in rats dosed with the PPI omeprazole. Nine rats received 400 micromol/kg omeprazole daily for 10 wk. Seven rats received vehicle only. Analysis of gastric corpus with microarrays representing 11,848 genes identified 134 genes with changed gene expression levels in omeprazole-dosed rats. Several of the identified genes were previously known to be affected by potent acid inhibition. Of the 62 genes with known functions that changed gene expression levels after PPI dosing, 27 are known to be involved in proliferation and apoptosis and immune, inflammatory, and stress responses. Our study indicates that microarray analysis can detect relevant gene expression changes in the complex gastric tissue, and that cellular processes involved in cell growth and defense responses are strongly affected by PPI dosing. Many genes are identified that were not previously known to be affected by inhibition of gastric acid secretion or that have unknown biological functions. Characterization of the roles of these genes may give new insight into molecular responses to treatment with PPIs.
Collapse
Affiliation(s)
- Kristin G Nørsett
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | | | | | | | | |
Collapse
|
48
|
Debacq-Chainiaux F, Borlon C, Pascal T, Royer V, Eliaers F, Ninane N, Carrard G, Friguet B, de Longueville F, Boffe S, Remacle J, Toussaint O. Repeated exposure of human skin fibroblasts to UVB at subcytotoxic level triggers premature senescence through the TGF-β1 signaling pathway. J Cell Sci 2005; 118:743-58. [PMID: 15671065 DOI: 10.1242/jcs.01651] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Premature senescence of human diploid fibroblasts (HDFs) can be induced by exposures to a variety of oxidative stress and DNA damaging agents. In this study we developed a robust model of UVB-induced premature senescence of skin HDFs. After a series of 10 subcytotoxic (non-proapoptotic) exposures to UVB at 250 mJ/cm2, the so-called biomarkers of senescence were markedly expressed: growth arrest, senescence-associated β-galactosidase activity, senescence-associated gene overexpression, deletion in mitochondrial DNA. A set of 44 stress- and senescence-associated genes were found to be differentially expressed in this model, among which clusterin/apolipoprotein J (apo J) and transforming growth factor-β1 (TGF-β1). Transfection of apo J cDNA provided protection against premature senescence-inducing doses of UVB and other stressful agents. Neutralizing antibodies against TGF-β1 or its receptor II (TβRII) sharply attenuated the senescence-associated features, suggesting a role for TGF-β1 in UVB-induced premature senescence. Both the latent and active forms of TGF-β1 were increased with time after the last UVB stress. Proteasome inhibition was ruled out as a potential mechanism of UVB-induced stress-induced premature senescence (SIPS). This model represents an alternative in vitro model in photoaging research for screening potential anti-photoaging compounds.
Collapse
Affiliation(s)
- Florence Debacq-Chainiaux
- Laboratory of Biochemistry and Cellular Biology, Department of Biology, University of Namur (FUNDP), Rue de Bruxelles, 61, 5000 Namur, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Rhyu DY, Yang Y, Ha H, Lee GT, Song JS, Uh ST, Lee HB. Role of Reactive Oxygen Species in TGF-β1-Induced Mitogen-Activated Protein Kinase Activation and Epithelial-Mesenchymal Transition in Renal Tubular Epithelial Cells. J Am Soc Nephrol 2005; 16:667-75. [PMID: 15677311 DOI: 10.1681/asn.2004050425] [Citation(s) in RCA: 425] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays an important role in renal tubulointerstitial fibrosis and TGF-beta1 is the key inducer of EMT. Phosphorylation of Smad proteins and/or mitogen-activated protein kinases (MAPK) is required for TGF-beta1-induced EMT. Because reactive oxygen species (ROS) are involved in TGF-beta1 signaling and are upstream signaling molecules to MAPK, this study examined the role of ROS in TGF-beta1-induced MAPK activation and EMT in rat proximal tubular epithelial cells. Growth-arrested and synchronized NRK-52E cells were stimulated with TGF-beta1 (0.2 to 20 ng/ml) or H(2)O(2) (1 to 500 microM) in the presence or absence of antioxidants (N-acetylcysteine or catalase), inhibitors of NADPH oxidase (diphenyleneiodonium and apocynin), mitochondrial electron transfer chain subunit I (rotenone), and MAPK (PD 98059, an MEK [MAP kinase/ERK kinase] inhibitor, or p38 MAPK inhibitor) for up to 96 h. TGF-beta1 increased dichlorofluorescein-sensitive cellular ROS, phosphorylated Smad 2, p38 MAPK, extracellular signal-regulated kinases (ERK)1/2, alpha-smooth muscle actin (alpha-SMA) expression, and fibronectin secretion and decreased E-cadherin expression. Antioxidants effectively inhibited TGF-beta1-induced cellular ROS, phosphorylation of Smad 2, p38 MAPK, and ERK, and EMT. H(2)O(2) reproduced all of the effects of TGF-beta1 with the exception of Smad 2 phosphorylation. Chemical inhibition of ERK but not p38 MAPK inhibited TGF-beta1-induced Smad 2 phosphorylation, and both MAPK inhibitors inhibited TGF-beta1- and H(2)O(2)-induced EMT. Diphenyleneiodonium, apocynin, and rotenone also significantly inhibited TGF-beta1-induced ROS. Thus, this data suggest that ROS play an important role in TGF-beta1-induced EMT primarily through activation of MAPK and subsequently through ERK-directed activation of Smad pathway in proximal tubular epithelial cells.
Collapse
Affiliation(s)
- Dong Young Rhyu
- Ewha Womans University College of Pharmacy, 11-1 Daehyun-dong, Sedaimun-gu, Seoul 120-750, Korea
| | | | | | | | | | | | | |
Collapse
|