1
|
Cui X, Buonfiglio F, Pfeiffer N, Gericke A. Aging in Ocular Blood Vessels: Molecular Insights and the Role of Oxidative Stress. Biomedicines 2024; 12:817. [PMID: 38672172 PMCID: PMC11048681 DOI: 10.3390/biomedicines12040817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Acknowledged as a significant pathogenetic driver for numerous diseases, aging has become a focal point in addressing the profound changes associated with increasing human life expectancy, posing a critical concern for global public health. Emerging evidence suggests that factors influencing vascular aging extend their impact to choroidal and retinal blood vessels. The objective of this work is to provide a comprehensive overview of the impact of vascular aging on ocular blood vessels and related diseases. Additionally, this study aims to illuminate molecular insights contributing to vascular cell aging, with a particular emphasis on the choroid and retina. Moreover, innovative molecular targets operating within the domain of ocular vascular aging are presented and discussed.
Collapse
Affiliation(s)
- Xiuting Cui
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (N.P.)
| | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (N.P.)
| |
Collapse
|
2
|
Wang P, Konja D, Singh S, Zhang B, Wang Y. Endothelial Senescence: From Macro- to Micro-Vasculature and Its Implications on Cardiovascular Health. Int J Mol Sci 2024; 25:1978. [PMID: 38396653 PMCID: PMC10889199 DOI: 10.3390/ijms25041978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Endothelial cells line at the most inner layer of blood vessels. They act to control hemostasis, arterial tone/reactivity, wound healing, tissue oxygen, and nutrient supply. With age, endothelial cells become senescent, characterized by reduced regeneration capacity, inflammation, and abnormal secretory profile. Endothelial senescence represents one of the earliest features of arterial ageing and contributes to many age-related diseases. Compared to those in arteries and veins, endothelial cells of the microcirculation exhibit a greater extent of heterogeneity. Microcirculatory endothelial senescence leads to a declined capillary density, reduced angiogenic potentials, decreased blood flow, impaired barrier properties, and hypoperfusion in a tissue or organ-dependent manner. The heterogeneous phenotypes of microvascular endothelial cells in a particular vascular bed and across different tissues remain largely unknown. Accordingly, the mechanisms underlying macro- and micro-vascular endothelial senescence vary in different pathophysiological conditions, thus offering specific target(s) for therapeutic development of senolytic drugs.
Collapse
Affiliation(s)
- Peichun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sandeep Singh
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Beijia Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
3
|
Endo T, Kobayashi K, Matsumura T, Emori C, Ozawa M, Kawamoto S, Okuzaki D, Shimada K, Miyata H, Shimada K, Kodani M, Ishikawa-Yamauchi Y, Motooka D, Hara E, Ikawa M. Multiple ageing effects on testicular/epididymal germ cells lead to decreased male fertility in mice. Commun Biol 2024; 7:16. [PMID: 38177279 PMCID: PMC10766604 DOI: 10.1038/s42003-023-05685-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
In mammals, females undergo reproductive cessation with age, whereas male fertility gradually declines but persists almost throughout life. However, the detailed effects of ageing on germ cells during and after spermatogenesis, in the testis and epididymis, respectively, remain unclear. Here we comprehensively examined the in vivo male fertility and the overall organization of the testis and epididymis with age, focusing on spermatogenesis, and sperm function and fertility, in mice. We first found that in vivo male fertility decreased with age, which is independent of mating behaviors and testosterone levels. Second, overall sperm production in aged testes was decreased; about 20% of seminiferous tubules showed abnormalities such as germ cell depletion, sperm release failure, and perturbed germ cell associations, and the remaining 80% of tubules contained lower number of germ cells because of decreased proliferation of spermatogonia. Further, the spermatozoa in aged epididymides exhibited decreased total cell numbers, abnormal morphology/structure, decreased motility, and DNA damage, resulting in low fertilizing and developmental rates. We conclude that these multiple ageing effects on germ cells lead to decreased in vivo male fertility. Our present findings are useful to better understand the basic mechanism behind the ageing effect on male fertility in mammals including humans.
Collapse
Affiliation(s)
- Tsutomu Endo
- Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Department of Experimental Animal Model for Human Disease, Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo, Japan.
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| | - Kiyonori Kobayashi
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Takafumi Matsumura
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Chihiro Emori
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Manabu Ozawa
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shimpei Kawamoto
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kentaro Shimada
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Mayo Kodani
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yu Ishikawa-Yamauchi
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Daisuke Motooka
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Eiji Hara
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masahito Ikawa
- Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
4
|
Ozawa M, Mori H, Endo T, Ishikawa-Yamauchi Y, Motooka D, Emori C, Ikawa M. Age-related decline in spermatogenic activity accompanied with endothelial cell senescence in male mice. iScience 2023; 26:108456. [PMID: 38077127 PMCID: PMC10700819 DOI: 10.1016/j.isci.2023.108456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/02/2023] [Accepted: 11/11/2023] [Indexed: 02/12/2024] Open
Abstract
Male fertility decreases with aging, with spermatogenic decline being one of its causes. Altered testis environment is suggested as a cause of the phenotype; however, the associated mechanisms remain unclear. Herein, we investigated the age-related changes in testicular somatic cells on spermatogenic activity. The number and proliferation of spermatogonia significantly reduced with aging in mice. Interestingly, senescence-associated β-galactosidase-positive cells appeared in testicular endothelial cell (EC) populations, but not in germ cell populations, with aging. Transcriptome analysis of ECs indicated that senescence occurred in the ECs of aged mice. Furthermore, the support capacity of ECs for spermatogonial proliferation significantly decreased with aging; however, the senolytic-induced removal of senescent cells from aged ECs restored their supporting capacity to a comparable level as that of young ECs. Our results suggest that the accumulation of senescent ECs in the testis is a potential factor contributing to the age-related decline in spermatogenic activity.
Collapse
Affiliation(s)
- Manabu Ozawa
- Laboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Hideto Mori
- Laboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tsutomu Endo
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yu Ishikawa-Yamauchi
- Laboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Daisuke Motooka
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Chihiro Emori
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Masahiro Ikawa
- Laboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
5
|
Teng RD, Yang CH, Chung CL, Sheu JR, Hsieh CY. Attenuation of indoxyl sulfate-induced cell damage by cinchonidine-a Cinchona alkaloid-through the downregulation of p53 signaling pathway by promoting MDM2 cytoplasmic-nuclear shuttling in endothelial cells. Life Sci 2023; 318:121477. [PMID: 36796718 DOI: 10.1016/j.lfs.2023.121477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
Renocardiac syndromes are a critical concern among patients with chronic kidney disease (CKD). High level of indoxyl sulfate (IS), a protein-bound uremic toxin, in plasma is known to promote the pathogenesis of cardiovascular diseases by impairing endothelial function. However, the therapeutic effects of the adsorbent of indole, a precursor of IS, on renocardiac syndromes is still debated. Therefore, novel therapeutic approaches should be developed to treat IS-associated endothelial dysfunction. In the present study, we have found that cinchonidine, a major Cinchona alkaloid, exhibited superior cell-protective effects among the 131 test compounds in IS-stimulated human umbilical vein endothelial cells (HUVECs). IS-induced cell death, cellular senescence, and impairment of tube formation in HUVECs were substantially reversed after treatment with cinchonidine. Despite the cinchonidine did not alter reactive oxygen species formation, cellular uptake of IS and OAT3 activity, RNA-Seq analysis showed that the cinchonidine treatment downregulated p53-modulated gene expression and substantially reversed IS-caused G0/G1 cell cycle arrest. Although the mRNA levels of p53 were not considerably downregulated by cinchonidine in IS-treated HUVECs, the treatment of cinchonidine promoted the degradation of p53 and the cytoplasmic-nuclear shuttling of MDM2. Cinchonidine exhibited cell-protective effects against the IS-induced cell death, cellular senescence, and impairment of vasculogenic activity in HUVECs through the downregulation of p53 signaling pathway. Collectively, cinchonidine may be a potential cell-protective agent to rescue IS-induced endothelial cell damage.
Collapse
Affiliation(s)
- Ruei-Dun Teng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Li Chung
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Ying Hsieh
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
6
|
Ohori M, Nakayama Y, Ogasawara-Shimizu M, Toyoshiba H, Nakanishi A, Aparicio S, Araki S. Gene regulatory network analysis defines transcriptome landscape with alternative splicing of human umbilical vein endothelial cells during replicative senescence. BMC Genomics 2021; 22:869. [PMID: 34856941 PMCID: PMC8641155 DOI: 10.1186/s12864-021-08185-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 11/18/2021] [Indexed: 01/10/2023] Open
Abstract
Background Endothelial cell senescence is the state of permanent cell cycle arrest and plays a critical role in the pathogenesis of age-related diseases. However, a comprehensive understanding of the gene regulatory network, including genome-wide alternative splicing machinery, involved in endothelial cell senescence is lacking. Results We thoroughly described the transcriptome landscape of replicative senescent human umbilical vein endothelial cells. Genes with high connectivity showing a monotonic expression increase or decrease with the culture period were defined as hub genes in the co-expression network. Computational network analysis of these genes led to the identification of canonical and non-canonical senescence pathways, such as E2F and SIRT2 signaling, which were down-regulated in lipid metabolism, and chromosome organization processes pathways. Additionally, we showed that endothelial cell senescence involves alternative splicing. Importantly, the first and last exon types of splicing, as observed in FLT1 and ACACA, were preferentially altered among the alternatively spliced genes during endothelial senescence. We further identified novel microexons in PRUNE2 and PSAP, each containing 9 nt, which were altered within the specific domain during endothelial senescence. Conclusions These findings unveil the comprehensive transcriptome pathway and novel signaling regulated by RNA processing, including gene expression and splicing, in replicative endothelial senescence. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08185-x.
Collapse
Affiliation(s)
- Momoko Ohori
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| | - Yusuke Nakayama
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.,Present address: Discovery Technology Research Laboratories, Tsukuba Research Institute, Ono Pharmaceutical Co., Ltd, 17-2 Wadai, 300-4247, Tsukuba, Ibaraki, Japan
| | - Mari Ogasawara-Shimizu
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Hiroyoshi Toyoshiba
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.,Present address: Life Science AI, FRONTEO Healthcare Inc., 2-12-23 Konan, Minato-ku, 108-0075, Tokyo, Japan
| | - Atsushi Nakanishi
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Samuel Aparicio
- Molecular Oncology, BC Cancer Agency, 675 W10th Avenue, V5Z 1L3, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, V6T 2B5, Vancouver, BC, Canada
| | - Shinsuke Araki
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| |
Collapse
|
7
|
Passage Number-Induced Replicative Senescence Modulates the Endothelial Cell Response to Protein-Bound Uremic Toxins. Toxins (Basel) 2021; 13:toxins13100738. [PMID: 34679030 PMCID: PMC8538293 DOI: 10.3390/toxins13100738] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
Endothelial aging may be induced early in pathological situations. The uremic toxins indoxyl sulfate (IS) and p-cresol (PC) accumulate in the plasma of chronic kidney disease (CKD) patients, causing accelerated endothelial aging, increased cardiovascular events and mortality. However, the mechanisms by which uremic toxins exert their deleterious effects on endothelial aging are not yet fully known. Thus, the aim of the present study is to determine the effects of IS and PC on endothelial damage and early senescence in cultured human umbilical vein endothelial cells (HUVECs). Hence, we establish an in vitro model of endothelial damage mediated by different passages of HUVECs and stimulated with different concentrations of IS and PC to evaluate functional effects on the vascular endothelium. We observe that cell passage-induced senescence is associated with apoptosis, ROS production and decreased endothelial proliferative capacity. Similarly, we observe that IS and PC cause premature aging in a dose-dependent manner, altering HUVECs' regenerative capacity, and decreasing their cell migration and potential to form vascular structures in vitro. In conclusion, IS and PC cause accelerated aging in HUVECs, thus contributing to endothelial dysfunction associated with CKD progression.
Collapse
|
8
|
Lau S, Rangarajan R, Krüger-Genge A, Braune S, Küpper JH, Lendlein A, Jung F. Age-related morphology and function of human arterial endothelial cells. Clin Hemorheol Microcirc 2020; 74:93-107. [PMID: 31839605 DOI: 10.3233/ch-199238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endothelialization of cardiovascular implants is regarded as a promising strategy for long-term compatibility. While umbilical vein endothelial cells are typically applied in research, human arterial endothelial cells (HAEC) from elderly donors would be the obvious source for autologous cellularization strategies.In our approach, HAEC from 16 donors of varying age (16-63 years) were divided into two groups (<30 years and >30 years) and analyzed regarding morphology, viability, proliferation, function and senescence status.No age-related differences were found regarding morphology, viability, density, prostacyclin and nitrite secretion or collagen and laminin production. However, the metabolic activity was slightly decreased (p = 0.0374) and the membrane integrity marginally impaired (p = 0.0404) in cells from older donors. Two out of three senescence assays detected more senescence markers in cells from older donors.According to the assays applied here, HAEC from young and elderly donors up to the age of 63 years could be judged equally suitable for autologous cellularization strategies. However, this finding should be regarded with caution due to the extremely large variability between individual donors. Further studies comprising a larger sample size are necessary to investigate this issue more thoroughly.
Collapse
Affiliation(s)
- S Lau
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - R Rangarajan
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - A Krüger-Genge
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - S Braune
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - J-H Küpper
- Institute of Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - A Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - F Jung
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| |
Collapse
|
9
|
Kumar V, Agrawal R, Pandey A, Kopf S, Hoeffgen M, Kaymak S, Bandapalli OR, Gorbunova V, Seluanov A, Mall MA, Herzig S, Nawroth PP. Compromised DNA repair is responsible for diabetes-associated fibrosis. EMBO J 2020; 39:e103477. [PMID: 32338774 PMCID: PMC7265245 DOI: 10.15252/embj.2019103477] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/27/2020] [Accepted: 03/08/2020] [Indexed: 11/09/2022] Open
Abstract
Diabetes-associated organ fibrosis, marked by elevated cellular senescence, is a growing health concern. Intriguingly, the mechanism underlying this association remained unknown. Moreover, insulin alone can neither reverse organ fibrosis nor the associated secretory phenotype, favoring the exciting notion that thus far unknown mechanisms must be operative. Here, we show that experimental type 1 and type 2 diabetes impairs DNA repair, leading to senescence, inflammatory phenotypes, and ultimately fibrosis. Carbohydrates were found to trigger this cascade by decreasing the NAD+ /NADH ratio and NHEJ-repair in vitro and in diabetes mouse models. Restoring DNA repair by nuclear over-expression of phosphomimetic RAGE reduces DNA damage, inflammation, and fibrosis, thereby restoring organ function. Our study provides a novel conceptual framework for understanding diabetic fibrosis on the basis of persistent DNA damage signaling and points to unprecedented approaches to restore DNA repair capacity for resolution of fibrosis in patients with diabetes.
Collapse
Affiliation(s)
- Varun Kumar
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany.,European Molecular Biology Laboratory, Advanced Light Microscopy Facility, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Heidelberg, Germany
| | - Raman Agrawal
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Aparamita Pandey
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Stefan Kopf
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Heidelberg, Germany
| | - Manuel Hoeffgen
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Serap Kaymak
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Obul Reddy Bandapalli
- Hopp Children's Cancer Center, Heidelberg, Germany.,Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.,Department of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Stephan Herzig
- German Center for Diabetes Research (DZD), Heidelberg, Germany.,Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Germany.,Technical University Munich, Munich, Germany
| | - Peter P Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Heidelberg, Germany.,Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Germany
| |
Collapse
|
10
|
Madonna R, Pieragostino D, Cufaro MC, Doria V, Del Boccio P, Deidda M, Pierdomenico SD, Dessalvi CC, De Caterina R, Mercuro G. Ponatinib Induces Vascular Toxicity through the Notch-1 Signaling Pathway. J Clin Med 2020; 9:jcm9030820. [PMID: 32197359 PMCID: PMC7141219 DOI: 10.3390/jcm9030820] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/04/2020] [Accepted: 03/08/2020] [Indexed: 02/08/2023] Open
Abstract
Ponatinib, a third-generation tyrosine kinase inhibitor (TKI), is the only approved TKI that is effective against T315I mutations in patients with chronic myeloid leukemia (CML). Specific activation of Notch signaling in CML cells by ponatinib can be considered as the “on-target effect” on the tumor and represents a therapeutic approach for CML. Nevertheless, ponatinib-induced vascular toxicity remains a serious concern, with underlying mechanisms being poorly understood. We aimed to determine the mechanisms of ponatinib-induced vascular toxicity, defining associated signaling pathways and identifying potential rescue strategies. We exposed human umbilical endothelial cells (HUVECs) to ponatinib or vehicle in the presence or absence of the neutralizing factor anti-Notch-1 antibody for exposure times of 0–72 h. Label-free proteomics and network analysis showed that protein cargo of HUVECs treated with ponatinib triggered apoptosis and inhibited vasculature development. We validated the proteomic data showing the inhibition of matrigel tube formation, an up-regulation of cleaved caspase-3 and a downregulation of phosphorylated AKT and phosphorylated eNOS. We delineated the signaling of ponatinib-induced vascular toxicity, demonstrating that ponatinib inhibits endothelial survival, reduces angiogenesis and induces endothelial senescence and apoptosis via the Notch-1 pathway. Ponatinib induced endothelial toxicity in vitro. Hyperactivation of Notch-1 in the vessels can lead to abnormal vascular development and vascular dysfunction. By hyperactivating Notch-1 in the vessels, ponatinib exerts an “on-target off tumor effect”, which leads to deleterious effects and may explain the drug’s vasculotoxicity. Selective blockade of Notch-1 prevented ponatinib-induced vascular toxicity.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Institute of Cardiology, University of Pisa, 56124 Pisa, Italy;
- Department of Internal Medicine, Cardiology Division, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Damiana Pieragostino
- Department of Medical, Oral and Biotechnological Sciences, University ‘‘G. D’Annunzio’’ of Chieti-Pescara, 66100 Chieti, Italy;
- Analytical Biochemistry and Proteomics Laboratory, Center for Advanced Studies and Technology (CAST), University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
| | - Maria Concetta Cufaro
- Analytical Biochemistry and Proteomics Laboratory, Center for Advanced Studies and Technology (CAST), University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
- Department of Pharmacy, University ‘‘G. d’Annunzio’’ of Chieti-Pescara, 66100 Chieti, Italy
| | - Vanessa Doria
- Institute of Cardiology, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (S.D.P.)
| | - Piero Del Boccio
- Analytical Biochemistry and Proteomics Laboratory, Center for Advanced Studies and Technology (CAST), University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
- Department of Pharmacy, University ‘‘G. d’Annunzio’’ of Chieti-Pescara, 66100 Chieti, Italy
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy; (M.D.); (C.C.D.)
| | - Sante Donato Pierdomenico
- Institute of Cardiology, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (S.D.P.)
| | - Christian Cadeddu Dessalvi
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy; (M.D.); (C.C.D.)
| | - Raffaele De Caterina
- Institute of Cardiology, University of Pisa, 56124 Pisa, Italy;
- Correspondence: (R.D.C.); (G.M.)
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy; (M.D.); (C.C.D.)
- Correspondence: (R.D.C.); (G.M.)
| |
Collapse
|
11
|
Yi S, Lin K, Jiang T, Shao W, Huang C, Jiang B, Li Q, Lin D. NMR-based metabonomic analysis of HUVEC cells during replicative senescence. Aging (Albany NY) 2020; 12:3626-3646. [PMID: 32074082 PMCID: PMC7066908 DOI: 10.18632/aging.102834] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/27/2020] [Indexed: 01/18/2023]
Abstract
Cellular senescence is a physiological process reacting to stimuli, in which cells enter a state of irreversible growth arrest in response to adverse consequences associated with metabolic disorders. Molecular mechanisms underlying the progression of cellular senescence remain unclear. Here, we established a replicative senescence model of human umbilical vein endothelial cells (HUVEC) from passage 3 (P3) to 18 (P18), and performed biochemical characterizations and NMR-based metabolomic analyses. The cellular senescence degree advanced as the cells were sequentially passaged in vitro, and cellular metabolic profiles were gradually altered. Totally, 8, 16, 21 and 19 significant metabolites were primarily changed in the P6, P10, P14 and P18 cells compared with the P3 cells, respectively. These metabolites were mainly involved in 14 significantly altered metabolic pathways. Furthermore, we observed taurine retarded oxidative damage resulting from senescence. In the case of energy deficiency, HUVECs metabolized neutral amino acids to replenish energy, thus increased glutamine, aspartate and asparagine at the early stages of cellular senescence but decreased them at the later stages. Our results indicate that cellular replicative senescence is closely associated with promoted oxidative stress, impaired energy metabolism and blocked protein synthesis. This work may provide mechanistic understanding of the progression of cellular senescence.
Collapse
Affiliation(s)
- Shenghui Yi
- College of Chemistry and Chemical Engineering, Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen 361005, China.,Department of Medical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Kejiang Lin
- Department of Medical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Ting Jiang
- College of Chemistry and Chemical Engineering, Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen 361005, China
| | - Wei Shao
- College of Chemistry and Chemical Engineering, Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen 361005, China
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361024, China
| | - Bin Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Xiamen University, Xiamen 361102, China
| | - Qinxi Li
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Xiamen University, Xiamen 361102, China
| | - Donghai Lin
- College of Chemistry and Chemical Engineering, Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen 361005, China
| |
Collapse
|
12
|
The Effects of IL-1β on Astrocytes are Conveyed by Extracellular Vesicles and Influenced by Age. Neurochem Res 2020; 45:694-707. [DOI: 10.1007/s11064-019-02937-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/12/2019] [Accepted: 12/15/2019] [Indexed: 01/23/2023]
|
13
|
Zheng M, Chen Y, Park J, Song HC, Chen Y, Park JW, Joe Y, Chung HT. CO ameliorates endothelial senescence induced by 5-fluorouracil through SIRT1 activation. Arch Biochem Biophys 2019; 677:108185. [PMID: 31704100 DOI: 10.1016/j.abb.2019.108185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 01/25/2023]
Abstract
Endothelial senescence is the main risk factor that contributes to vascular dysfunction and the progression of vascular disease. Carbon monoxide (CO) plays an important role in preventing vascular dysfunction and in maintaining vascular physiology or homeostasis. The application of exogenous CO has been shown to confer protection in several models of cardiovascular injury or disease, including hypertension, atherosclerosis, balloon-catheter injury, and graft rejection. However, the mechanism by which CO prevents endothelial senescence has been largely unexplored. The aim of this study was to evaluate the effects of CO on endothelial senescence and to investigate the possible mechanisms underlying this process. We measured the levels of senescence-associated-β-galactosidase activity, senescence-associated secretory phenotype, reactive oxygen species (ROS) production, and stress granule in human umbilical vein endothelial cells and the WI-38 human diploid fibroblast cell line. We found that 5-fluorouracil (5FU)-induced ROS generation was inhibited by CO-releasing molecules (CORM)-A1 treatment, and endothelial senescence induced by 5FU was attenuated by CORM-A1 treatment. The SIRT1 inhibitor EX527 reversed the inhibitory effect of CO on the 5FU-induced endothelial senescence. Furthermore, SIRT1 deficiency abolished the stress granule formation by CO. Our results suggest that CO alleviates the endothelial senescence induced by 5FU through SIRT1 activation and may hence have therapeutic potential for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Min Zheng
- Department of Neurology, Affiliated Hospital of YanBian University, Yanji, 133000, China
| | - Yubing Chen
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Jeongmin Park
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Hyun-Chul Song
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Yingqing Chen
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, South Korea; Department of Pharmacology, Dalian University Medical College, Dalian, 116622, China
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Yeonsoo Joe
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Hun Taeg Chung
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, South Korea.
| |
Collapse
|
14
|
Dong L, Gan L, Wang H, Cai W. Age-Related Impairment of Structure and Function of Iliac Artery Endothelium in Rats Is Improved by Elevated Fluid Shear Stress. Med Sci Monit 2019; 25:5127-5136. [PMID: 31291237 PMCID: PMC6637813 DOI: 10.12659/msm.916287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Background Aging plays an important role in endothelial dysfunction. Fluid shear stress (FSS) can activate endothelial cells (ECs). Herein, we tested the hypothesis that this endothelial impairment could be improved by elevated FSS (EFSS) in aged rats. Material/Methods EFSS was created through ligation of the unilateral common iliac artery in 20-−month-old rats, evaluated by measuring blood flow velocity with Doppler spectrum. The effect of FSS on aged ECs was examined by senescence-associated β-galactosidase (SA-β-Gal) staining, ultrastructural observation, and immunostaining and qPCR analysis of eNOS and SIRT1 expression on both the mRNA and protein levels. Results (1) FSS was significantly increased in the right common iliac artery (RCIA) in rats with the ligation of the left common iliac artery (LCIA). (2) SA-β-Gal staining was significantly attenuated by EFSS in the RCIA of aged rats. (3) Ultrastructural observation showed that ECs in the RCIA of normal aged rats became irregular and enlarged, with increasingly polypoid nuclei and fewer mitochondria, whereas ECs in the RCIA of aged rats with LCIA ligation became more prominent and contained more mitochondria. (4) eNOS and SIRT1 expression in the RCIA of aged rats with LCIA ligation was significantly upregulated compared with that in control group rats. Conclusions The present study for the first time shows that EFSS has the ability to improve age-related impairment of endothelial structure and functions.
Collapse
Affiliation(s)
- Liping Dong
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, Hunan, China (mainland).,Department of Anatomy, Histology, and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, Hunan, China (mainland)
| | - Liaoying Gan
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Hui Wang
- Department of Anatomy, School of Basic Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Weijun Cai
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
15
|
Maeda M, Tsuboi T, Hayashi T. An Inhibitor of Activated Blood Coagulation Factor X Shows Anti-Endothelial Senescence and Anti-Atherosclerotic Effects. J Vasc Res 2019; 56:181-190. [DOI: 10.1159/000499975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/28/2019] [Indexed: 11/19/2022] Open
|
16
|
Vo NTK, Moore LC, Spiteri KW, Hanner R, Wilkie MP, DeWitte-Orr SJ. Assessing off-target cytotoxicity of the field lampricide 3-trifluoromethyl-4-nitrophenol using novel lake sturgeon cell lines. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 162:536-545. [PMID: 30016760 DOI: 10.1016/j.ecoenv.2018.07.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/29/2018] [Accepted: 07/04/2018] [Indexed: 06/08/2023]
Abstract
Lampricides are currently being applied to streams and rivers to control the population of sea lamprey, an invasive species, in the Great Lakes. The most commonly used lampricide agent used in the field is 3-trifluoromethyl-4-nitrophenol (TFM), which targets larval sea lamprey in lamprey-infested rivers and streams. The specificity of TFM is due to the relative inability of sea lamprey to detoxify the agent relative to non-target fishes. There is increasing concern, however, about non-target effects on fishes, particularly threatened populations of juvenile lake sturgeon (LS; Acipenser fulvescens). There is therefore a need to develop models to better define lake sturgeon's response to TFM. Here we report the establishment of five LS cell lines derived from the liver, gill, skin and intestinal tract of juvenile LS and some of their cellular characteristics. All LS cell lines grew well at 25 °C in Leibovitz's (L)- 15 medium supplemented with 10% FBS. All cell lines demonstrated high senescence-associated β-galactosidase activity and varying levels of Periodic acid Schiff-positive polysaccharides, indicating substantial production of glycoproteins and mucosubstances by the cells. Comparative toxicity of TFM in the five LS cell lines was assessed by two fluorescent cell viability dyes, Alamar Blue and CFDA-AM, in conditions with and without serum and at 24 or 72 h exposure. Deduced EC50 values were compared between the cell lines and to the reported in vivo LC50s. Tissues sensitive to the effects of TFM in vivo correlated with cell lines from the same tissues being most sensitive to TFM in vitro. EC50 values for the LSliver-e cells was significantly lower than the EC50 for the rainbow trout (RBT) liver cells RTL-W1, reaffirming the in vivo observation that LS was generally more TFM-sensitive than rainbow trout. Our data suggests that whole-fish sensitivity of LS to TFM is likely attributable to sensitivity at the cellular level. Thus, LS cell lines, as well as those of RBT, can be used to screen and evaluate the toxicity of the next generation of lampricides on non-target fish such as lake sturgeon.
Collapse
Affiliation(s)
- Nguyen T K Vo
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Levi C Moore
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Katelin W Spiteri
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Robert Hanner
- Department of Integrative Biology, University of Guelph, Guelph, ON, Canada
| | - Michael P Wilkie
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Stephanie J DeWitte-Orr
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada; Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada.
| |
Collapse
|
17
|
Tsuboi T, Maeda M, Hayashi T. Administration of L-arginine plus L-citrulline or L-citrulline alone successfully retarded endothelial senescence. PLoS One 2018; 13:e0192252. [PMID: 29415069 PMCID: PMC5802914 DOI: 10.1371/journal.pone.0192252] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/18/2018] [Indexed: 11/19/2022] Open
Abstract
L-citrulline and L-arginine supplementation has been shown to have several beneficial effects on the cardiovascular system. Nitric oxide (NO) protects against the progression of atherosclerosis and is synthesized by nitric oxide synthase (NOS), which converts L-arginine (L-Arg) into L-citrulline (L-Cit). Our previous study revealed that chronic administration of a combination of L-Cit and L- Arg has a better therapeutic effect on high cholesterol-induced atherosclerosis in rabbits. We investigated how L-Arg and L-Cit affect endothelial function, aging and atherosclerosis. Following a 3-day stimulation of human umbilical venous endothelial cells (HUVECs) with high glucose (HG: 22 mM) and L-Arg (300 μM), L-Cit (300 μM) or L-Arg plus L-Cit (LALC: each 150 μM) supplementation, endothelial senescence and function were evaluated. These amino acids were also administered to dyslipidemic type 2 diabetic (ZDFM) rats fed a high cholesterol diet. They were fed L-Arg or L-Cit or LALC for four weeks. Aortic senescence was investigated by measuring senescence-associated ß-galactosidase (SA-ß-gal), telomerase activity, DNA damage and p16INK4a protein expression. Only L-Cit and LALC supplementation retarded the HG-induced endothelial senescence, as evaluated by SA-ß-gal activity, a widely used marker of cellular senescence, p16INK4a expression, a senescence-related protein, and DNA damage. Under HG conditions, L-Cit and LCLA restored telomerase activity to levels observed under normal glucose (NG) conditions. Under HG conditions, L-Cit decreased ROS production, as measured by CM-H2DCFDA and the expression of p67phox, a major component of NADPH oxidase. Under HG conditions, L-Cit and LALC increased NO production, as measured by DAF-2AM. Endothelial NO synthase (eNOS) and phosphorylated eNOS were decreased under HG conditions and L-Cit and LALC significantly increased these levels. Arginase 2 protein expression increased under the HG conditions, and L-Cit and LALC significantly attenuated this effect. In ZDFM rats, SA-ß-gal activity was detected on the aortic endothelial surface; however, L-Cit and LALC reduced these levels. L-Cit and LALC both decreased the proportion of senescent cells. Furthermore, treatment with LALC for 4 weeks increased plasma NO production. Therefore conclusively, L-citrulline supplementation rescued NO levels better than L-arginine supplementation by inhibiting ROS production and arginase 2 protein expression. Consequently, L-Cit and LCLA supplementation retaeded HG-induced endothelial senescence.
Collapse
Affiliation(s)
- Tomoe Tsuboi
- Department of Geriatrics, Nagoya University Graduate School of Medicine School of Health Sciences, Daiko-Minami, Higashi-ku, Nagoya, Aichi, Japan
- Chubu University Graduate School of Bioscience and Biotechnology, Matsumoto-cho, Kasugai, Aichi, Japan
| | - Morihiko Maeda
- Department of Geriatrics, Nagoya University Graduate School of Medicine School of Health Sciences, Daiko-Minami, Higashi-ku, Nagoya, Aichi, Japan
| | - Toshio Hayashi
- Department of Geriatrics, Nagoya University Graduate School of Medicine School of Health Sciences, Daiko-Minami, Higashi-ku, Nagoya, Aichi, Japan
- * E-mail:
| |
Collapse
|
18
|
Zhu R, Wang S, Xue Z, Han J, Han S. Senescence-associated sialidase revealed by an activatable fluorescence-on labeling probe. Chem Commun (Camb) 2018; 54:11566-11569. [DOI: 10.1039/c8cc07024e] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A fluorescence-quenched substrate of sialidase enables fluorescence-on live cell imaging of sialidases, revealing up-regulation of lysosome-associated sialidase in cell senescence.
Collapse
Affiliation(s)
- Rui Zhu
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- The Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| | - Siyu Wang
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- The Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| | - Zhongwei Xue
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- The Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology
- Innovation Center for Cell Signalling Network
- School of Life Sciences
- Xiamen University
- Xiamen
| | - Shoufa Han
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- The Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| |
Collapse
|
19
|
Boraldi F, Burns JS, Bartolomeo A, Dominici M, Quaglino D. Mineralization by mesenchymal stromal cells is variously modulated depending on commercial platelet lysate preparations. Cytotherapy 2017; 20:335-342. [PMID: 29289444 DOI: 10.1016/j.jcyt.2017.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/13/2017] [Accepted: 11/22/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AIMS Numerous cellular models have been developed to investigate calcification for regenerative medicine applications and for the identification of therapeutic targets in various complications associated with age-related diseases. However, results have often been contradictory due to specific culture conditions, cell type ontogeny and aging status. Human platelet lysate (hPL) has been recently investigated as valuable alternative to fetal bovine serum (FBS) in cell culture and bone regeneration. A parallel comparison of how all these multiple factors may converge to influence mineralization has yet to be reported. METHODS To compare mineralization of human mesenchymal cell types known to differ in extracellular matrix calcification potency, bone marrow-derived mesenchymal stromal cells and dermal fibroblasts from neonatal and adult donors, at both low and high passages, were investigated in an ex vivo experimental model by supplementing the osteogenic induction medium with FBS or with hPL. Four commercial hPL preparations were profiled by liquid chromatography/electrospray ionization quadrupole time-of-flight spectrometry, and mineralization was visualized by von Kossa staining and quantified by morphometric evaluations after 9, 14 and 21 days of culture. RESULTS Data demonstrate that (i) commercial hPL preparations differ according to mass spectra profiles, (ii) hPL variously influences mineral deposition depending on cell line and possibly on platelet product preparation methods, (iii) donor age modifies mineral deposition in the presence of the same hPL and (iv) reduced in vitro proliferative capacity affects osteogenic induction and response to hPL. CONCLUSION Despite the standardized procedures applied to obtain commercial hPL, this study highlights the divergent effects of different preparations and emphasizes the importance of cellular ontology, donor age and cell proliferative capacity to optimize the osteogenic induction capabilities of mesenchymal stromal cells and design more effective cell-based therapeutic protocols.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jorge S Burns
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy; Fondazione Democenter-Sipe, Tecnopolo Mirandola-TPM, Science and Technology Park for Medicine, Modena, Italy
| | - Angelica Bartolomeo
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy; Fondazione Democenter-Sipe, Tecnopolo Mirandola-TPM, Science and Technology Park for Medicine, Modena, Italy
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
20
|
Salt-induced Na+/K+-ATPase-α/β expression involves soluble adenylyl cyclase in endothelial cells. Pflugers Arch 2017; 469:1401-1412. [DOI: 10.1007/s00424-017-1999-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 04/03/2017] [Accepted: 05/15/2017] [Indexed: 12/28/2022]
|
21
|
Altieri P, Murialdo R, Barisione C, Lazzarini E, Garibaldi S, Fabbi P, Ruggeri C, Borile S, Carbone F, Armirotti A, Canepa M, Ballestrero A, Brunelli C, Montecucco F, Ameri P, Spallarossa P. 5-fluorouracil causes endothelial cell senescence: potential protective role of glucagon-like peptide 1. Br J Pharmacol 2017; 174:3713-3726. [PMID: 28127745 DOI: 10.1111/bph.13725] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 01/09/2017] [Accepted: 01/19/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE 5-fluorouracil (5FU) and its prodrug, capecitabine, can damage endothelial cells, whilst endothelial integrity is preserved by glucagon-like peptide 1 (GLP-1). Here, we studied the effect of 5FU on endothelial senescence and whether GLP-1 antagonizes it. EXPERIMENTAL APPROACH EA.hy926 cells were exposed to 5FU or sera from patients taking capecitabine, with or without pre-incubation with GLP-1. Senescence was identified by expression of senescence-associated β-galactosidase and p16INK4a and reduced cell proliferation. Soluble vascular cell adhesion molecule-1 (sVCAM-1), soluble intercellular adhesion molecule-1 (sICAM-1) and CD146 (marker of endothelial injury) were measured by ELISA before and at completion of capecitabine chemotherapy. RT-PCR, western blotting, functional experiments with signalling inhibitors and ERK1/2 silencing were performed to characterize 5FU-induced phenotype and elucidate the pathways underlying 5FU and GLP-1 activity. KEY RESULTS Both 5FU and sera from capecitabine-treated patients stimulated endothelial cell senescence. 5FU-elicited senescence occurred via activation of p38 and JNK, and was associated with decreased eNOS and SIRT-1 levels. Furthermore, 5FU up-regulated VCAM1 and TYMP (encodes enzyme activating capecitabine and 5FU), and sVCAM-1 and CD146 concentrations were higher after than before capecitabine chemotherapy. A non-significant trend for higher ICAM1 levels was also observed. GLP-1 counteracted 5FU-initiated senescence and reduced eNOS and SIRT-1 expression, this protection being mediated by GLP-1 receptor, ERK1/2 and, possibly, PKA and PI3K. CONCLUSIONS AND IMPLICATIONS 5FU causes endothelial cell senescence and dysfunction, which may contribute to its cardiovascular side effects. 5FU-triggered senescence was prevented by GLP-1, raising the possibility of using GLP-1 analogues and degradation inhibitors to treat 5FU and capecitabine vascular toxicity. LINKED ARTICLES This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.
Collapse
Affiliation(s)
- Paola Altieri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | | | - Chiara Barisione
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Edoardo Lazzarini
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Silvano Garibaldi
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Patrizia Fabbi
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Clarissa Ruggeri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Silvia Borile
- Cardiovascular Disease Unit, IRCCS AOU San Martino-IST, Genova, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, IRCCS AOU San Martino - IST, Genova, Italy.,Department of Internal Medicine, University of Genova, Genova, Italy
| | - Andrea Armirotti
- Drug Discovery and Development Department, Italian Institute of Technology (IIT), Genova, Italy
| | - Marco Canepa
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy.,Cardiovascular Disease Unit, IRCCS AOU San Martino-IST, Genova, Italy
| | | | - Claudio Brunelli
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy.,Cardiovascular Disease Unit, IRCCS AOU San Martino-IST, Genova, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, IRCCS AOU San Martino - IST, Genova, Italy.,Department of Internal Medicine, University of Genova, Genova, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Pietro Ameri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy.,Cardiovascular Disease Unit, IRCCS AOU San Martino-IST, Genova, Italy
| | - Paolo Spallarossa
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy.,Cardiovascular Disease Unit, IRCCS AOU San Martino-IST, Genova, Italy
| |
Collapse
|
22
|
Miller JW, Bagheri S, Vavvas DG. Advances in Age-related Macular Degeneration Understanding and Therapy. ACTA ACUST UNITED AC 2017; 10:119-130. [PMID: 29142592 PMCID: PMC5683729 DOI: 10.17925/usor.2017.10.02.119] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
While the development of anti-vascular endothelial growth factor (anti-VEGF) as a therapy for neovascular age-related macular degeneration (AMD) was a great success, the pathologic processes underlying dry AMD that eventually leads to photoreceptor dysfunction, death, and vision loss remain elusive to date, with a lack of effective therapies and increasing prevalence of the disease. There is an overwhelming need to improve the classification system of AMD, to increase our understanding of cell death mechanisms involved in both neovascular and non-neovascular AMD, and to develop better biomarkers and clinical endpoints to eventually be able to identify better therapeutic targets—especially early in the disease process. There is no doubt that it is a matter of time before progress will be made and better therapies will be developed for non-neovascular AMD.
Collapse
Affiliation(s)
- Joan W Miller
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, US
| | - Saghar Bagheri
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, US
| | - Demetrios G Vavvas
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, US
| |
Collapse
|
23
|
Kang MK, Park NH. Conversion of Normal To Malignant Phenotype: Telomere Shortening, Telomerase Activation, and Genomic Instability During Immortalization of Human Oral Keratinocytes. ACTA ACUST UNITED AC 2016; 12:38-54. [PMID: 11349961 DOI: 10.1177/10454411010120010301] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Normal somatic cells terminate their replicative life span through a pathway leading to cellular senescence, which is triggered by activation of p53 and/or pRb in response to critically shortened telomere DNA. Potentially neoplastic cells must first overcome the senescence checkpoint mechanisms and subsequently activate telomerase to propagate indefinitely. Although telomerase activation is closely associated with cellular immortality, telomerase alone is not sufficient to warrant tumorigenicity. Environmental factors, including chemical carcinogens and viral infection, often contribute to aberrant changes leading to tumorigenic conversion of normal cells. Of particular importance in oral cancer development are tobacco-related chemical carcinogens and human papillomavirus (HPV) infection. To describe the molecular mechanisms by which these environmental factors facilitate the genesis of oral cancer, we first established an in vitro multistep oral carcinogenesis model by sequential exposure of normal human oral keratinocytes (NHOK) to "high risk" HPV and chemical carcinogens. Upon introduction of the HPV genome, the cells bypassed the senescence checkpoint and entered into an extended, but not immortal, life span during which telomere DNA continued to shorten. In a few immortal clones surviving beyond the crisis, we found a marked elevation of telomerase activity and stabilization of telomere length. Furthermore, the E6 and E7 oncoproteins of "high risk" HPV disrupted the cell cycle control and DNA repair in immortalized HOK, and enhanced mutation frequency resulting from genomic instability. However, HPV infection alone failed to give rise to a tumorigenic cell population, which required further exposure to chemical carcinogens in addition to HPV infection. Analysis of the data presented suggests that oral carcinogenesis is a series of discrete genetic alterations that result from a continued genotoxic challenge by environmental risk factors. Our in vitro model may be useful for investigators with interest in furthering our understanding of oral carcinogenesis.
Collapse
Affiliation(s)
- M K Kang
- School of Dentistry and Dental Research Institute, University of California, Los Angeles 90095, USA
| | | |
Collapse
|
24
|
Huang KC, Yang J, Ng MC, Ng SK, Welch WR, Muto MG, Berkowitz RS, Ng SW. Cyclin A1 expression and paclitaxel resistance in human ovarian cancer cells. Eur J Cancer 2016; 67:152-163. [PMID: 27669502 PMCID: PMC5080661 DOI: 10.1016/j.ejca.2016.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 05/30/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND The development of intrinsic and acquired resistance to antineoplastic agents is a major obstacle to successful chemotherapy in ovarian cancers. Identification and characterisation of chemoresponse-associated biomarkers are of paramount importance for novel therapeutic development. METHODS Global RNA expression profiles were obtained by high-throughput microarray analysis. Cell cycle, proliferation rate, and paclitaxel sensitivity of ovarian cancer cells harbouring cyclin A1-inducible expression construct were compared with and without tetracycline induction, as well as when the cyclin A1 expression was suppressed by short inhibiting RNA (siRNA). Cellular senescence was evaluated by β-galactosidase activity staining. RESULTS Global RNA expression profiling and subsequent correlation studies of gene expression level and drug response has identified that elevated expression of cyclin A1 (CCNA1) was significantly associated with cellular resistance to paclitaxel, doxorubicin and 5-fluorouracil. The role of cyclin A1 in paclitaxel resistance was confirmed in ovarian cancer cells that harbour an inducible cyclin A1 expression construct, which showed reduced paclitaxel-mediated growth inhibition and apoptosis when cyclin A1 expression was induced, whereas downregulation of cyclin A1 expression in the same cell lines using cyclin A1-specific siRNAs sensitised the cells to paclitaxel toxicity. However, ovarian cancer cells with ectopic expression of cyclin A1 demonstrated slowdown of proliferation and senescence-associated β-galactosidase activity. CONCLUSIONS Our profiling and correlation studies have identified cyclin A1 as one chemoresistance-associated biomarker in ovarian cancer. The results of the characterisation studies suggest that cyclin A1 functions as an oncogene that controls proliferative and survival activities in tumourigenesis and chemoresistance of ovarian cancer.
Collapse
Affiliation(s)
- Kuan-Chun Huang
- Laboratory of Gynecologic Oncology, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Boston, MA 02115, USA
| | - Junzheng Yang
- Laboratory of Gynecologic Oncology, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Boston, MA 02115, USA
| | - Michelle C Ng
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shu-Kay Ng
- School of Medicine and Menzies Health Institute Queensland, Griffith University, Meadowbrook, Australia
| | - William R Welch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael G Muto
- Laboratory of Gynecologic Oncology, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Boston, MA 02115, USA
| | - Ross S Berkowitz
- Laboratory of Gynecologic Oncology, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Boston, MA 02115, USA
| | - Shu-Wing Ng
- Laboratory of Gynecologic Oncology, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Katsurabayashi S, Kawano H, Ii M, Nakano S, Tatsumi C, Kubota K, Takasaki K, Mishima K, Fujiwara M, Iwasaki K. Overexpression of Swedish mutant APP in aged astrocytes attenuates excitatory synaptic transmission. Physiol Rep 2016; 4:4/1/e12665. [PMID: 26733247 PMCID: PMC4760399 DOI: 10.14814/phy2.12665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Amyloid precursor protein (APP), a type I transmembrane protein, has different aspects, namely, performs essential physiological functions and produces β‐amyloid peptide (Aβ). Overexpression of neuronal APP is responsible for synaptic dysfunction. In the central nervous system, astrocytes – a major glial cell type – have an important role in the regulation of synaptic transmission. Although APP is expressed in astrocytes, it remains unclear whether astrocytic overexpression of mutant APP affects synaptic transmission. In this study, the effect of astrocytic overexpression of a mutant APP on the excitatory synaptic transmission was investigated using coculture system of the transgenic (Tg) cortical astrocytes that express the human APP695 polypeptide with the double mutation K670N + M671L found in a large Swedish family with early onset Alzheimer's disease, and wild‐type hippocampal neuron. Significant secretion of Aβ 1–40 and 1–42 was observed in cultured cortical astrocytes from the Tg2576 transgenic mouse that genetically overexpresses Swedish mutant APP. Under the condition, Tg astrocytes did not affect excitatory synaptic transmission of cocultured wild‐type neurons. However, aged Tg astrocytes cultured for 9 weeks elicited a significant decrease in excitatory synaptic transmission in cocultured neurons. Moreover, a reduction in the number of readily releasable synaptic vesicles accompanied a decrease in the number of excitatory synapses in neurons cocultured with aged Tg astrocytes. These observations indicate that astrocytic expression of the mutant APP is involved in the downregulation of synaptic transmission with age.
Collapse
Affiliation(s)
- Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Hiroyuki Kawano
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Miyuki Ii
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Sachiko Nakano
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Chihiro Tatsumi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| | - Kotaro Takasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kenichi Mishima
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| | - Michihiro Fujiwara
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
26
|
Characterization and Expression of Senescence Marker in Prolonged Passages of Rat Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:8487264. [PMID: 27579045 PMCID: PMC4989133 DOI: 10.1155/2016/8487264] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
The present study is aimed at optimizing the in vitro culture protocol for generation of rat bone marrow- (BM-) derived mesenchymal stem cells (MSCs) and characterizing the culture-mediated cellular senescence. The initial phase of generation and characterization was conducted using the adherent cells from Sprague Dawley (SD) rat's BM via morphological analysis, growth kinetics, colony forming unit capacity, immunophenotyping, and mesodermal lineage differentiation. Mesenchymal stem cells were successfully generated and characterized as delineated by the expressions of CD90.1, CD44H, CD29, and CD71 and lack of CD11b/c and CD45 markers. Upon induction, rBM-MSCs differentiated into osteocytes and adipocytes and expressed osteocytes and adipocytes genes. However, a decline in cell growth was observed at passage 4 onwards and it was further deciphered through apoptosis, cell cycle, and senescence assays. Despite the enhanced cell viability at later passages (P4-5), the expression of senescence marker, β-galactosidase, was significantly increased at passage 5. Furthermore, the cell cycle analysis has confirmed the in vitro culture-mediated cellular senescence where cells were arrested at the G0/G1 phase of cell cycle. Although the currently optimized protocols had successfully yielded rBM-MSCs, the culture-mediated cellular senescence limits the growth of rBM-MSCs and its potential use in rat-based MSC research.
Collapse
|
27
|
|
28
|
Vitamin E Supplementation Delays Cellular Senescence In Vitro. BIOMED RESEARCH INTERNATIONAL 2015; 2015:563247. [PMID: 26613084 PMCID: PMC4647025 DOI: 10.1155/2015/563247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/08/2015] [Indexed: 01/25/2023]
Abstract
Vitamin E is an important antioxidant that protects cells from oxidative stress-induced damage, which is an important contributor to the progression of ageing. Ageing can be studied in vitro using primary cells reaching a state of irreversible growth arrest called senescence after a limited number of cellular divisions. Generally, the most utilized biomarker of senescence is represented by the expression of the senescence associated β-galactosidase (SA-β-gal). We aimed here to study the possible effects of vitamin E supplementation in two different human primary cell types (HUVECs and fibroblasts) during the progression of cellular senescence. Utilizing an unbiased automated system, based on the detection of the SA-β-gal, we quantified cellular senescence in vitro and showed that vitamin E supplementation reduced the numbers of senescent cells during progression of ageing. Acute vitamin E supplementation did not affect cellular proliferation, whereas it was decreased after chronic treatment. Mechanistically, we show that vitamin E supplementation acts through downregulation of the expression of the cycline dependent kinase inhibitor P21. The data obtained from this study support the antiageing properties of vitamin E and identify possible mechanisms of action that warrant further investigation.
Collapse
|
29
|
Boraldi F, Bartolomeo A, Di Bari C, Cocconi A, Quaglino D. Donor's age and replicative senescence favour the in-vitro mineralization potential of human fibroblasts. Exp Gerontol 2015; 72:218-26. [PMID: 26494600 DOI: 10.1016/j.exger.2015.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/16/2015] [Accepted: 10/19/2015] [Indexed: 11/30/2022]
Abstract
Aberrant mineralization of soft connective tissues (ectopic calcification) may occur as a frequent age-related complication. Still, it remains unclear the role of mesenchymal cell donor's age and of replicative senescence on ectopic calcification. Therefore, the ability of cells to deposit in-vitro hydroxyapatite crystals and the expression of progressive ankylosis protein homolog (ANKH), ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), tissue non specific alkaline phosphatase (TNAP) and osteopontin (OPN) have been evaluated in human dermal fibroblasts derived from neonatal (nHDF) and adult (aHDF) donors (ex-vivo ageing model) or at low and high cumulative population doublings (CPD) up to replicative senescence (in-vitro ageing model). This study demonstrates that: 1) replicative senescence favours hydroxyapatite formation in cultured fibroblasts; 2) donor's age acts as a major modulator of the mineralizing potential of HDF, since nHDF are less prone than aHDF to induce calcification; 3) donor's age and replicative senescence play in concert synergistically increasing the calcification process; 4) the ANKH+ENPP1/TNAP ratio, being crucial for pyrophosphate/inorganic phosphate balance, is greatly influenced by donor's age, as well as by replicative senescence, and regulates mineral deposition; 5) OPN is only modulated by replicative senescence.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Angelica Bartolomeo
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Caterina Di Bari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Cocconi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
30
|
Quantitative and integrated proteome and microRNA analysis of endothelial replicative senescence. J Proteomics 2015; 126:12-23. [DOI: 10.1016/j.jprot.2015.05.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/05/2015] [Accepted: 05/20/2015] [Indexed: 12/31/2022]
|
31
|
Lunt SY, Muralidhar V, Hosios AM, Israelsen WJ, Gui DY, Newhouse L, Ogrodzinski M, Hecht V, Xu K, Acevedo PNM, Hollern DP, Bellinger G, Dayton TL, Christen S, Elia I, Dinh AT, Stephanopoulos G, Manalis SR, Yaffe MB, Andrechek ER, Fendt SM, Vander Heiden MG. Pyruvate kinase isoform expression alters nucleotide synthesis to impact cell proliferation. Mol Cell 2015; 57:95-107. [PMID: 25482511 PMCID: PMC4289430 DOI: 10.1016/j.molcel.2014.10.027] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 08/19/2014] [Accepted: 10/28/2014] [Indexed: 01/15/2023]
Abstract
Metabolic regulation influences cell proliferation. The influence of pyruvate kinase isoforms on tumor cells has been extensively studied, but whether PKM2 is required for normal cell proliferation is unknown. We examine how PKM2 deletion affects proliferation and metabolism in nontransformed, nonimmortalized PKM2-expressing primary cells. We find that deletion of PKM2 in primary cells results in PKM1 expression and proliferation arrest. PKM1 expression, rather than PKM2 loss, is responsible for this effect, and proliferation arrest cannot be explained by cell differentiation, senescence, death, changes in gene expression, or prevention of cell growth. Instead, PKM1 expression impairs nucleotide production and the ability to synthesize DNA and progress through the cell cycle. Nucleotide biosynthesis is limiting, as proliferation arrest is characterized by severe thymidine depletion, and supplying exogenous thymine rescues both nucleotide levels and cell proliferation. Thus, PKM1 expression promotes a metabolic state that is unable to support DNA synthesis.
Collapse
Affiliation(s)
- Sophia Y Lunt
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Vinayak Muralidhar
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard-MIT Health Sciences and Technology Division, Harvard Medical School, Boston, MA 02115, USA
| | - Aaron M Hosios
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - William J Israelsen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dan Y Gui
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lauren Newhouse
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Martin Ogrodzinski
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Vivian Hecht
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kali Xu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Paula N Marín Acevedo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel P Hollern
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Gary Bellinger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Talya L Dayton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stefan Christen
- Vesalius Research Center, VIB, 3000 Leuven, Belgium; Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Ilaria Elia
- Vesalius Research Center, VIB, 3000 Leuven, Belgium; Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Anh T Dinh
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Scott R Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael B Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eran R Andrechek
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Sarah-Maria Fendt
- Vesalius Research Center, VIB, 3000 Leuven, Belgium; Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Cheung TM, Yan JB, Fu JJ, Huang J, Yuan F, Truskey GA. Endothelial Cell Senescence Increases Traction Forces due to Age-Associated Changes in the Glycocalyx and SIRT1. Cell Mol Bioeng 2014; 8:63-75. [PMID: 25914755 DOI: 10.1007/s12195-014-0371-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Endothelial cell (EC) aging and senescence are key events in atherogenesis and cardiovascular disease development. Age-associated changes in the local mechanical environment of blood vessels have also been linked to atherosclerosis. However, the extent to which cell senescence affects mechanical forces generated by the cell is unclear. In this study, we sought to determine whether EC senescence increases traction forces through age-associated changes in the glycocalyx and antioxidant regulator deacetylase Sirtuin1 (SIRT1), which is downregulated during aging. Traction forces were higher in cells that had undergone more population doublings and changes in traction force were associated with altered actin localization. Older cells also had increased actin filament thickness. Depletion of heparan sulfate in young ECs elevated traction forces and actin filament thickness, while addition of heparan sulfate to the surface of aged ECs by treatment with angiopoietin-1 had the opposite effect. While inhibition of SIRT1 had no significant effect on traction forces or actin organization for young cells, activation of SIRT1 did reduce traction forces and increase peripheral actin in aged ECs. These results show that EC senescence increases traction forces and alters actin localization through changes to SIRT1 and the glycocalyx.
Collapse
Affiliation(s)
- Tracy M Cheung
- Department of Biomedical Engineering Duke University Durham, NC 27708
| | - Jessica B Yan
- Department of Biomedical Engineering Duke University Durham, NC 27708
| | - Justin J Fu
- Department of Biomedical Engineering Duke University Durham, NC 27708
| | - Jianyong Huang
- Department of Biomedical Engineering Duke University Durham, NC 27708
| | - Fan Yuan
- Department of Biomedical Engineering Duke University Durham, NC 27708
| | - George A Truskey
- Department of Biomedical Engineering Duke University Durham, NC 27708
| |
Collapse
|
33
|
Senescence-associated β-galactosidase staining in fish cell lines and primary cultures from several tissues and species, including rainbow trout coelomic fluid and milt. In Vitro Cell Dev Biol Anim 2014; 51:361-71. [DOI: 10.1007/s11626-014-9837-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 10/16/2014] [Indexed: 12/11/2022]
|
34
|
Ribeiro-Varandas E, Pereira HS, Monteiro S, Neves E, Brito L, Boavida Ferreira R, Viegas W, Delgado M. Bisphenol A disrupts transcription and decreases viability in aging vascular endothelial cells. Int J Mol Sci 2014; 15:15791-805. [PMID: 25207595 PMCID: PMC4200871 DOI: 10.3390/ijms150915791] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 05/04/2014] [Accepted: 06/13/2014] [Indexed: 12/16/2022] Open
Abstract
Bisphenol A (BPA) is a widely utilized endocrine disruptor capable of mimicking endogenous hormones, employed in the manufacture of numerous consumer products, thereby interfering with physiological cellular functions. Recent research has shown that BPA alters epigenetic cellular mechanisms in mammals and may be correlated to enhanced cellular senescence. Here, the effects of BPA at 10 ng/mL and 1 µg/mL, concentrations found in human samples, were analyzed on HT29 human colon adenocarcinona cell line and Human Umbilical Vein Endothelial Cells (HUVEC). Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) transcriptional analysis of the Long Interspersed Element-1 (LINE-1) retroelement showed that BPA induces global transcription deregulation in both cell lines, although with more pronounced effects in HUVEC cells. Whereas there was an increase in global transcription in HT29 exclusively after 24 h of exposure, this chemical had prolonged effects on HUVEC. Immunoblotting revealed that this was not accompanied by alterations in the overall content of H3K9me2 and H3K4me3 epigenetic marks. Importantly, cell viability assays and transcriptional analysis indicated that prolonged BPA exposure affects aging processes in senescent HUVEC. To our knowledge this is the first report that BPA interferes with senescence in primary vascular endothelial cells, therefore, suggesting its association to the etiology of age-related human pathologies, such as atherosclerosis.
Collapse
Affiliation(s)
- Edna Ribeiro-Varandas
- Centro de Botânica Aplicada à Agricultura (CBAA), Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; E-Mails: (E.R.-V.); (H.S.P.); (S.M.); (E.N.); (L.B.); (R.B.F.); (W.V.)
| | - H. Sofia Pereira
- Centro de Botânica Aplicada à Agricultura (CBAA), Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; E-Mails: (E.R.-V.); (H.S.P.); (S.M.); (E.N.); (L.B.); (R.B.F.); (W.V.)
| | - Sara Monteiro
- Centro de Botânica Aplicada à Agricultura (CBAA), Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; E-Mails: (E.R.-V.); (H.S.P.); (S.M.); (E.N.); (L.B.); (R.B.F.); (W.V.)
| | - Elsa Neves
- Centro de Botânica Aplicada à Agricultura (CBAA), Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; E-Mails: (E.R.-V.); (H.S.P.); (S.M.); (E.N.); (L.B.); (R.B.F.); (W.V.)
- Escola Superior de Tecnologia e Gestão Jean Piaget do Litoral Alentejano (ESTGJPLA), Instituto Piaget, Campus Académico de Santo André, Ap. 38, 7500-999 Vila Nova de Santo André, Portugal
| | - Luísa Brito
- Centro de Botânica Aplicada à Agricultura (CBAA), Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; E-Mails: (E.R.-V.); (H.S.P.); (S.M.); (E.N.); (L.B.); (R.B.F.); (W.V.)
| | - Ricardo Boavida Ferreira
- Centro de Botânica Aplicada à Agricultura (CBAA), Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; E-Mails: (E.R.-V.); (H.S.P.); (S.M.); (E.N.); (L.B.); (R.B.F.); (W.V.)
- Instituto de Tecnologia Química e Biológica (ITQB), Universidade Nova de Lisboa (UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - Wanda Viegas
- Centro de Botânica Aplicada à Agricultura (CBAA), Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; E-Mails: (E.R.-V.); (H.S.P.); (S.M.); (E.N.); (L.B.); (R.B.F.); (W.V.)
| | - Margarida Delgado
- Centro de Botânica Aplicada à Agricultura (CBAA), Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017 Lisboa, Portugal; E-Mails: (E.R.-V.); (H.S.P.); (S.M.); (E.N.); (L.B.); (R.B.F.); (W.V.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +351-213-653-100 (ext. 3281); Fax: +351-213-653-195
| |
Collapse
|
35
|
Blue EK, DiGiuseppe R, Derr-Yellin E, Acosta JC, Pay SL, Hanenberg H, Schellinger MM, Quinney SK, Mund JA, Case J, Haneline LS. Gestational diabetes induces alterations in the function of neonatal endothelial colony-forming cells. Pediatr Res 2014; 75:266-72. [PMID: 24232636 PMCID: PMC3944713 DOI: 10.1038/pr.2013.224] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/30/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Children born to mothers with gestational diabetes mellitus (GDM) experience increased risk of developing hypertension, type 2 diabetes mellitus, and obesity. Disrupted function of endothelial colony-forming cells (ECFCs) may contribute to this enhanced risk. The goal of this study was to determine whether cord blood ECFCs from GDM pregnancies exhibit altered functionality. METHODS ECFCs isolated from the cord blood of control and GDM pregnancies were assessed for proliferation, senescence, and Matrigel network formation. The requirement for p38MAPK in hyperglycemia-induced senescence was determined using inhibition and overexpression studies. RESULTS GDM-exposed ECFCs were more proliferative than control ECFCs. However, GDM-exposed ECFCs exhibited decreased network-forming ability in Matrigel. Aging of ECFCs by serial passaging led to increased senescence and reduced proliferation of GDM-exposed ECFCs. ECFCs from GDM pregnancies were resistant to hyperglycemia-induced senescence compared with those from controls. In response to hyperglycemia, control ECFCs activated p38MAPK, which was required for hyperglycemia-induced senescence. In contrast, GDM-exposed ECFCs showed no change in p38MAPK activation under equivalent conditions. CONCLUSION Intrauterine exposure of ECFCs to GDM induces unique phenotypic alterations. The resistance of GDM-exposed ECFCs to hyperglycemia-induced senescence and decreased p38MAPK activation suggest that these progenitor cells have undergone changes that induce tolerance to a hyperglycemic environment.
Collapse
Affiliation(s)
- Emily K. Blue
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN,Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| | - Robert DiGiuseppe
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Ethel Derr-Yellin
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN,Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| | - Juan Carlos Acosta
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - S. Louise Pay
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| | - Helmut Hanenberg
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN,Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN,Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Megan M. Schellinger
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN
| | - Sara K. Quinney
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN
| | - Julie A. Mund
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN,Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN,Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Jamie Case
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN,Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN,Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Laura S. Haneline
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN,Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN,Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN,Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, IN,Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
36
|
Endothelial cellular senescence is inhibited by liver X receptor activation with an additional mechanism for its atheroprotection in diabetes. Proc Natl Acad Sci U S A 2014; 111:1168-73. [PMID: 24398515 DOI: 10.1073/pnas.1322153111] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Senescence of vascular endothelial cells leads to endothelial dysfunction and contributes to the progression of atherosclerosis. Liver X receptors (LXRs) are nuclear receptors whose activation protects against atherosclerosis by transcriptional regulation of genes important in promoting cholesterol efflux and inhibiting inflammation. Here we found that LXR activation with specific ligands reduced the increase in senescence-associated (SA) β-gal activity, a senescence marker, and reversed the decrease in telomerase activity, a replicative senescence marker, in human endothelial cells under high glucose. This effect of LXR activation was associated with reduced reactive oxygen species and increased endothelial NO synthase activity. A series of experiments that used siRNAs indicated that LXRβ mediates the prevention of endothelial cellular senescence, and that sterol regulatory element binding protein-1, which was up-regulated as a direct LXRβ target gene, may act as a brake of endothelial cellular senescence. Although oral administration of the LXR ligand led to severe fatty liver in diabetic rats, concomitant therapy with metformin avoided the development of hepatic steatosis. However, the preventive effect of the LXR ligand on SA β-gal-stained cells in diabetic aortic endothelium was preserved even if metformin was coadministered. Taken together, our studies demonstrate that an additional mechanism, such as the regulation of endothelial cellular senescence, is related to the antiatherogenic properties of LXRs, and concomitant treatment with metformin may provide a clinically useful therapeutic strategy to alleviate an LXR activation-mediated adverse effects on liver triglyceride metabolism.
Collapse
|
37
|
AbuBakar S, Shu MH, Johari J, Wong PF. Senescence affects endothelial cells susceptibility to dengue virus infection. Int J Med Sci 2014; 11:538-44. [PMID: 24782642 PMCID: PMC4003538 DOI: 10.7150/ijms.7896] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 01/06/2014] [Indexed: 01/17/2023] Open
Abstract
Alteration in the endothelium leading to increased vascular permeability contributes to plasma leakage seen in dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). An earlier study showed that senescent endothelial cells (ECs) altered the ECs permeability. Here we investigated the susceptibility of senescing human umbilical vein endothelial cells (HUVECs) to dengue virus infection and determined if dengue virus infection induces HUVECs senescence. Our results suggest that DENV type-2 (DENV-2) foci forming unit (FFU) and extracellular virus RNA copy number were reduced by at least 35% and 85% in infection of the intermediate young and early senescent HUVECs, respectively, in comparison to infection of young HUVECs. No to low infectivity was recovered from infection of late senescent HUVECs. DENV infection also increases the percentage of HUVECs expressing senescence-associated (SA)-β-gal, cells arrested at the G2/M phase or 4N DNA content stage and cells with enlarged morphology, indicative of senescing cells. Alteration of HUVECs morphology was recorded using impedance-based real-time cell analysis system following DENV-2 infection. These results suggest that senescing HUVECs do not support DENV infection and DENV infection induces HUVECs senescence. The finding highlights the possible role of induction of senescence in DENV infection of the endothelial cells.
Collapse
Affiliation(s)
- Sazaly AbuBakar
- 1. Tropical Infectious Diseases Research and Education Centre (TIDREC), Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Meng-Hooi Shu
- 1. Tropical Infectious Diseases Research and Education Centre (TIDREC), Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Jefree Johari
- 1. Tropical Infectious Diseases Research and Education Centre (TIDREC), Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- 2. Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Gao SG, Zeng C, Li LJ, Luo W, Zhang FJ, Tian J, Cheng C, Tu M, Xiong YL, Jiang W, Xu M, Lei GH. Correlation between senescence-associated beta-galactosidase expression in articular cartilage and disease severity of patients with knee osteoarthritis. Int J Rheum Dis 2013; 19:226-32. [PMID: 26112901 DOI: 10.1111/1756-185x.12096] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM The purposes of this study were to investigate senescence-associated beta-galactosidase (SA-beta-Gal) levels in articular cartilage of knee osteoarthritis (OA) and the relationship with severity of the disease. METHODS All the 50 cartilage tissues, including normal (controls) and OA cartilage were ascribed to four groups of normal, mild lesions, moderate lesions and severe lesions on the basis of the modified Mankin score. Immunohistochemistry was used to assess the SA-beta-Gal expression in articular cartilage. RESULTS No SA-beta-Gal staining was observed in the normal articular cartilage samples. SA-beta-Gal staining was found in a subset of the chondrocytes close to the lesion site of mild, moderate and severe damaged knee OA cartilage. The percentage of SA-beta-Gal-positive chondrocytes in articular cartilage was 0% in controls, 13.00 ± 5.77% in mild lesions, 31.65 ± 6.91% in moderate lesions and 51.95 ± 6.21% in severe lesions. SA-beta-Gal expression in mild lesions, moderate lesions and severe lesions was higher compared with that of controls (P < 0.0001). SA-beta-Gal expression in moderate lesions and severe lesions were higher with respect to mild lesion samples (P < 0.0001). SA-beta-Gal expression in severe lesions was elevated compared with those of moderate lesions (P < 0.0001). Subsequent analysis showed that articular cartilage SA-beta-Gal levels correlated with severity of disease (Spearman's ρ = 0.94, P < 0.0001). CONCLUSION SA-beta-Gal expression in articular cartilage is associated with progressive knee OA joint damage and is a potential indictor of disease severity.
Collapse
Affiliation(s)
- Shu-Guang Gao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Orthopaedics Institute of Central South University, Changsha, Hunan, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liang-Jun Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Orthopaedics, Changsha Central Hospital, Changsha, Hunan, China
| | - Wei Luo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang-Jie Zhang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Tian
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chao Cheng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Tu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi-Lin Xiong
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Jiang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mai Xu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guang-Hua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Orthopaedics Institute of Central South University, Changsha, Hunan, China
| |
Collapse
|
39
|
Smith MC, Goddard ET, Perusina Lanfranca M, Davido DJ. hTERT extends the life of human fibroblasts without compromising type I interferon signaling. PLoS One 2013; 8:e58233. [PMID: 23472163 PMCID: PMC3589264 DOI: 10.1371/journal.pone.0058233] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 02/05/2013] [Indexed: 12/24/2022] Open
Abstract
Primary cells are often used to study viral replication and host-virus interactions as their antiviral pathways have not been altered or inactivated; however, their use is restricted by their short lifespan. Conventional methods to extend the life of primary cultures typically utilize viral oncogenes. Many of these oncogenes, however, perturb or inactivate cellular antiviral pathways, including the interferon (IFN) response. It has been previously shown that expression of the telomerase reverse transcriptase (TERT) gene extends the life of certain cell types. The effect that TERT expression has on the innate antiviral response to RNA- and DNA-containing viruses has not been examined. In the current study, we introduced the human TERT (hTERT) gene into a primary human embryonic lung (HEL-299) cell strain, which is known to respond to the type I IFN, IFN-β. We show that the resulting HEL-TERT cell line is capable of replicating beyond 100 population doublings without exhibiting signs of senescence. Treatment with IFN-β resulted in the upregulation of four model IFN stimulated genes (ISGs) in HEL-299 and HEL-TERT cells. Both cell lines supported the replication of herpes simplex virus type 1 (HSV-1) and vesicular stomatitis virus (VSV) and impaired the replication of both viruses upon IFN-β pretreatment. Introduction of the viral oncoprotein, simian virus 40 (SV40) large T-antigen, which is frequently used to immortalize cells, largely negated this effect. Taken together, our data indicate that expression of hTERT does not alter type 1 IFN signaling and/or the growth of two viruses, making this cell line a useful reagent for studying viral replication and virus-cell interactions.
Collapse
Affiliation(s)
- Miles C. Smith
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Erica T. Goddard
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Mirna Perusina Lanfranca
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - David J. Davido
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
40
|
Lee YS, Park CO, Noh JY, Jin S, Lee NR, Noh S, Lee JH, Lee KH. Knockdown of paraoxonase 1 expression influences the ageing of human dermal microvascular endothelial cells. Exp Dermatol 2013; 21:682-7. [PMID: 22897574 DOI: 10.1111/j.1600-0625.2012.01555.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Skin is one of the most commonly studied tissues for microcirculation research owing to its close correlation of cutaneous vascular function, ageing and age-related cardiovascular events. To elucidate proteins that determine this correlation between endothelial cell function and ageing in the vascular environment of the skin, we performed a proteomic analysis of plasma samples from six donors in their 20s (young) and six donors in their 60s (old). Among identified proteins, paraoxonase 1 (PON1) was selected in this study. To elucidate the role of PON1 on skin ageing and determine how it controls cellular senescence, the characteristics of PON1 in human dermal microvascular endothelial cells (HDMECs) were determined. When the expression of endogenous PON1 was knocked-down by small interfering RNA (siRNA) targeting PON1, HDMECs showed characteristic features of cellular senescence such as increases in senescence-associated β-galactosidase stained cells and enlarged and flattened cell morphology. At 48 h post-transfection, the protein expression of p16 in PON1 siRNA-treated HDMECs was higher than that in scrambled siRNA-treated HDMECs. In addition, the expressions of moesin and rho GTP dissociation inhibitor, additional age-related candidate biomarkers, were decreased by PON1 knock-down in HDMECs. In conclusion, these results suggest that PON1 functions as an ageing-related protein and plays an important role in the cellular senescence of HDMECs.
Collapse
Affiliation(s)
- Yun Sun Lee
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
PURPOSE We sought to determine the effect of stress-induced senescence on the permeability to albumin of aging endothelial progenitor cells. METHODS Human umbilical cord blood derived endothelial cells (hCB-ECs) and human aortic endothelial cells (HAECs) were treated with 200 μM H2O2 and permeability to FITC-bovine serum albumin was measured. Some samples were subsequently treated with 100μM 8-pCPT-2'-O-Me-cAMP, a cAMP analog that activates the Epac1-Rap1 pathway. Cell proliferation was measured with the EdU assay. Phase contrast, and immunofluorescence images were taken to observe morphological changes in cells after exposure to H2O2. RESULTS hCB-ECs exposed to H2O2 exhibited a significant increase in permeability, but their response differed from the HAECs. Low passage hCB-ECs had a permeability increase of about 82% (p<0.01) compared to aged cells which had a permeability increase of about 37% (p<0.05). This increase in permeability was reduced by treating the cells with 100 μM 8-pCPT-2'-O-Me-cAMP. The younger cells exhibited a significant decrease in proliferation after being subjected to various concentrations of H2O2 whereas the aged cells exhibited a more gradual decrease in the percent of cells in S-phase. These changes also correlated with changes in cell morphology and junction staining. When placed back in the original media, the morphology and permeability of the hCB-ECs returned to the control condition, while the HAECs did not. CONCLUSIONS The permeability of low and high passage hCB-ECs and HAECs initially increases in response to oxidative stress. hCB-ECs, but not HAECs, were able to recover from the stress 24 hours later. Early passage hCB-ECs were more susceptible to exogenous H2O2 than late passage hCB-ECs. The increase in permeability of hCB-ECs to H2O2 also correlated with decreased cell proliferation and changes in cell junctions.
Collapse
|
42
|
Low-dose etoposide-treatment induces endoreplication and cell death accompanied by cytoskeletal alterations in A549 cells: Does the response involve senescence? The possible role of vimentin. Cancer Cell Int 2013; 13:9. [PMID: 23383739 PMCID: PMC3599314 DOI: 10.1186/1475-2867-13-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/30/2013] [Indexed: 02/07/2023] Open
Abstract
Background Senescence in the population of cells is often described as a program of restricted proliferative capacity, which is manifested by broad morphological and biochemical changes including a metabolic shift towards an autophagic-like response and a genotoxic-stress related induction of polyploidy. Concomitantly, the cell cycle progression of a senescent cell is believed to be irreversibly arrested. Recent reports suggest that this phenomenon may have an influence on the therapeutic outcome of anticancer treatment. The aim of this study was to verify the possible involvement of this program in the response to the treatment of the A549 cell population with low doses of etoposide, as well as to describe accompanying cytoskeletal alterations. Methods After treatment with etoposide, selected biochemical and morphological parameters were examined, including: the activity of senescence-associated ß-galactosidase, SAHF formation, cell cycle progression, the induction of p21Cip1/Waf1/Sdi1 and cyclin D1, DNA strand breaks, the disruption of cell membrane asymmetry/integrity and ultrastructural alterations. Vimentin and G-actin cytoskeleton was evaluated both cytometrically and microscopically. Results and conclusions Etoposide induced a senescence-like phenotype in the population of A549 cells. Morphological alterations were nevertheless not directly coupled with other senescence markers including a stable cell cycle arrest, SAHF formation or p21Cip1/Waf1/Sdi1 induction. Instead, a polyploid, TUNEL-positive fraction of cells visibly grew in number. Also upregulation of cyclin D1 was observed. Here we present preliminary evidence, based on microscopic analyses, that suggest a possible role of vimentin in nuclear alterations accompanying polyploidization-depolyploidization events following genotoxic insults.
Collapse
|
43
|
Hsu CY, Chan YP. The use of honeybees reared in a thermostatic chamber for aging studies. AGE (DORDRECHT, NETHERLANDS) 2013; 35:149-158. [PMID: 22124884 PMCID: PMC3543731 DOI: 10.1007/s11357-011-9344-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 11/07/2011] [Indexed: 05/31/2023]
Abstract
Honeybees (Apis mellifera) are an attractive model system for studying aging. However, the aging level of worker honeybees from the field hive is in dispute. To eliminate the influence of task performance and confirm the relationship between chronological age and aging, we reared newly emerged workers in a thermostat at 34°C throughout their lives. A survivorship curve was obtained, indicating that workers can be reared away from the field hive, and the only difference between these workers is age. To confirm that these workers can be used for aging studies, we assayed age-related molecules in the trophocytes and fat cells of young and old workers. Old workers expressed more senescence-associated β-galactosidase, lipofuscin granules, lipid peroxidation, and protein oxidation than young workers. Furthermore, cellular energy metabolism molecules were also assayed. Old workers exhibited less ATP concentration, β-oxidation, and microtubule-associated protein light chain 3 (LC3) than young workers. These results demonstrate that honeybees reared in a thermostatic chamber can be used for aging studies and cellular energy metabolism in the trophocytes and fat cells of workers changes with advancing age.
Collapse
Affiliation(s)
- Chin-Yuan Hsu
- Department of Biomedical Sciences, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, Taiwan.
| | | |
Collapse
|
44
|
Bachschmid MM, Schildknecht S, Matsui R, Zee R, Haeussler D, Cohen RA, Pimental D, Loo BVD. Vascular aging: chronic oxidative stress and impairment of redox signaling-consequences for vascular homeostasis and disease. Ann Med 2013; 45:17-36. [PMID: 22380696 PMCID: PMC3717565 DOI: 10.3109/07853890.2011.645498] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Characteristic morphological and molecular alterations such as vessel wall thickening and reduction of nitric oxide occur in the aging vasculature leading to the gradual loss of vascular homeostasis. Consequently, the risk of developing acute and chronic cardiovascular diseases increases with age. Current research of the underlying molecular mechanisms of endothelial function demonstrates a duality of reactive oxygen and nitrogen species in contributing to vascular homeostasis or leading to detrimental effects when formed in excess. Furthermore, changes in function and redox status of vascular smooth muscle cells contribute to age-related vascular remodeling. The age-dependent increase in free radical formation causes deterioration of the nitric oxide signaling cascade, alters and activates prostaglandin metabolism, and promotes novel oxidative posttranslational protein modifications that interfere with vascular and cell signaling pathways. As a result, vascular dysfunction manifests. Compensatory mechanisms are initially activated to cope with age-induced oxidative stress, but become futile, which results in irreversible oxidative modifications of biological macromolecules. These findings support the 'free radical theory of aging' but also show that reactive oxygen and nitrogen species are essential signaling molecules, regulating vascular homeostasis.
Collapse
Affiliation(s)
- Markus M Bachschmid
- Vascular Biology Unit, Whitaker Cardiovascular Institute, Boston University Medical Center, Boston, MA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhang G, Morin C, Zhu X, Bao Huynh M, Ouidir Ouidja M, Sepulveda-Diaz JE, Raisman-Vozari R, Li P, Papy-Garcia D. Self-evolving oxidative stress with identifiable pre- and postmitochondrial phases in PC12 cells. J Neurosci Res 2012; 91:273-84. [DOI: 10.1002/jnr.23146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/25/2012] [Accepted: 08/25/2012] [Indexed: 11/10/2022]
|
46
|
Kawano H, Katsurabayashi S, Kakazu Y, Yamashita Y, Kubo N, Kubo M, Okuda H, Takasaki K, Kubota K, Mishima K, Fujiwara M, Harata NC, Iwasaki K. Long-term culture of astrocytes attenuates the readily releasable pool of synaptic vesicles. PLoS One 2012; 7:e48034. [PMID: 23110166 PMCID: PMC3482238 DOI: 10.1371/journal.pone.0048034] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 09/19/2012] [Indexed: 12/24/2022] Open
Abstract
The astrocyte is a major glial cell type of the brain, and plays key roles in the formation, maturation, stabilization and elimination of synapses. Thus, changes in astrocyte condition and age can influence information processing at synapses. However, whether and how aging astrocytes affect synaptic function and maturation have not yet been thoroughly investigated. Here, we show the effects of prolonged culture on the ability of astrocytes to induce synapse formation and to modify synaptic transmission, using cultured autaptic neurons. By 9 weeks in culture, astrocytes derived from the mouse cerebral cortex demonstrated increases in β-galactosidase activity and glial fibrillary acidic protein (GFAP) expression, both of which are characteristic of aging and glial activation in vitro. Autaptic hippocampal neurons plated on these aging astrocytes showed a smaller amount of evoked release of the excitatory neurotransmitter glutamate, and a lower frequency of miniature release of glutamate, both of which were attributable to a reduction in the pool of readily releasable synaptic vesicles. Other features of synaptogenesis and synaptic transmission were retained, for example the ability to induce structural synapses, the presynaptic release probability, the fraction of functional presynaptic nerve terminals, and the ability to recruit functional AMPA and NMDA glutamate receptors to synapses. Thus the presence of aging astrocytes affects the efficiency of synaptic transmission. Given that the pool of readily releasable vesicles is also small at immature synapses, our results are consistent with astrocytic aging leading to retarded synapse maturation.
Collapse
Affiliation(s)
- Hiroyuki Kawano
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
- * E-mail: (SK); (KI)
| | - Yasuhiro Kakazu
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Yuta Yamashita
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - Natsuko Kubo
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - Masafumi Kubo
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - Hideto Okuda
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - Kotaro Takasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - Kenichi Mishima
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
- A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - Michihiro Fujiwara
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
| | - N. Charles Harata
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
- A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Fukuoka, Japan
- * E-mail: (SK); (KI)
| |
Collapse
|
47
|
Cheung TM, Ganatra MP, Peters EB, Truskey GA. Effect of cellular senescence on the albumin permeability of blood-derived endothelial cells. Am J Physiol Heart Circ Physiol 2012; 303:H1374-83. [PMID: 23023872 DOI: 10.1152/ajpheart.00182.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this study, we tested the hypotheses that endothelial cells (ECs) derived from human umbilical cord blood (hCB-ECs) exhibit low permeability, which increases as hCB-ECs age and undergo senescence, and that the change in the permeability of hCB-ECs is due to changes in tight junction protein localization and the activity of exchange protein activated by cAMP (Epac)1. Albumin permeability across low-passage hCB-EC monolayers on Transwell membranes was 10 times lower than for human aortic ECs (HAECs) (P < 0.01) but similar to in vivo values in arteries. Expression of the tight junction protein occludin and tyrosine phosphorylation of occludin were less in hCB-ECs than in HAECs (P < 0.05). More hCB-ECs than HAECs underwent mitosis (P < 0.01). hCB-ECs that underwent >44 population doublings since isolation had a significantly higher permeability than hCB-ECs that underwent <31 population doublings (P < 0.05). This age-related increase in hCB-EC permeability was associated with an increase in tyrosine phosphorylation of occludin (P < 0.01); permeability and occludin phosphorylation were reduced by treatment with 2 μM resveratrol. Tyrosine phosphorylation of occludin and cell age influence the permeability of hCB-ECs, whereas levels of EC proliferation and expression of tight junction proteins did not explain the differences between hCB-EC and HAEC permeability. The elevated permeability in late passage hCB-ECs was reduced by 25-40% by elevation of membrane-associated cAMP and activation of the Epac1 pathway. Given the similarity to in vivo permeability to albumin and the high proliferation potential, hCB-ECs may be a suitable in vitro model to study transport-related pathologies and cell aging.
Collapse
Affiliation(s)
- Tracy M Cheung
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
48
|
Effects of resveratrol on longevity, cognitive ability and aging-related histological markers in the annual fish Nothobranchius guentheri. Exp Gerontol 2012; 47:940-9. [PMID: 22960591 DOI: 10.1016/j.exger.2012.08.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 08/24/2012] [Accepted: 08/27/2012] [Indexed: 12/29/2022]
Abstract
Aging research was hindered because of the long lifespan of available vertebrates. Annual fishes of Nothobranchius have become a new model organism for aging studies. Resveratrol, a natural plant-derived chemical, prolongs lifespan in many animals. Here we used the wild strain of N. guentheri, which has the mean lifespan of 12months, to detect the effects of resveratrol on the longevity, cognitive ability and aging-related histological markers. Our results showed that the pharmaceutical treatment of resveratrol prolonged the lifespan of N. guentheri but did not affect their body size. Three behavioral assays for cognitive ability and locomotor activity demonstrated that the resveratrol-treated fish exhibited the higher rate of performances than the fish in the control group. Further data indicated that resveratrol not only had the property of protecting N. guentheri from neurodegeneration, but retarded the aging-related histological markers in lipofuscin formation and in the expression of senescence-associated beta-galactosidase activity.
Collapse
|
49
|
Taranum S, Vaylann E, Meinke P, Abraham S, Yang L, Neumann S, Karakesisoglou I, Wehnert M, Noegel AA. LINC complex alterations in DMD and EDMD/CMT fibroblasts. Eur J Cell Biol 2012; 91:614-28. [PMID: 22555292 PMCID: PMC3778752 DOI: 10.1016/j.ejcb.2012.03.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 03/08/2012] [Accepted: 03/09/2012] [Indexed: 11/29/2022] Open
Abstract
Emery-Dreifuss muscular dystrophy (EDMD) is a late onset-disease characterized by skeletal muscle wasting and heart defects with associated risk of sudden death. The autosomal dominant form of the disease is caused by mutations in the LMNA gene encoding LaminA and C, the X-linked form results from mutations in the gene encoding the inner nuclear membrane protein Emerin (STA). Both Emerin and LaminA/C interact with the nuclear envelope proteins Nesprin-1 and -2 and mutations in genes encoding C-terminal isoforms of Nesprin-1 and -2 have also been implicated in EDMD. Here we analyse primary fibroblasts from patients affected by either Duchenne muscular dystrophy (DMD) or Emery-Dreifuss muscular dystrophy/Charcot-Marie-Tooth syndrome (EDMD/CMT) that in addition to the disease causing mutations harbour mutations in the Nesprin-1 gene and in the SUN1 and SUN2 gene, respectively. SUN proteins together with the Nesprins form the core of the LINC complex which connects the nucleus with the cytoskeleton. The mutations are accompanied by changes in cell adhesion, cell migration, senescence, and stress response, as well as in nuclear shape and nuclear envelope composition which are changes characteristic for laminopathies. Our results point to a potential influence of mutations in components of the LINC complex on the clinical outcome and the molecular pathology in the patients.
Collapse
Affiliation(s)
- Surayya Taranum
- Institute for Biochemistry I, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 52, 50931 Cologne, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Wolf J, Weinberger B, Grubeck-Loebenstein B. The immunoregulatory effects of CMV-infection in human fibroblasts and the impact on cellular senescence. IMMUNITY & AGEING 2012; 9:1. [PMID: 22455503 PMCID: PMC3364840 DOI: 10.1186/1742-4933-9-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/29/2012] [Indexed: 11/10/2022]
Abstract
BACKGROUND As a chronic antigenic stressor human Cytomegalovirus (CMV) contributes substantially to age-related alterations of the immune system. Even though monocytes have the greatest propensity for CMV-infection and seem to be an important host for the virus during latency, fibroblasts are also discussed to be target cells of CMV in vivo. However, little is known so far about general immunoregulatory properties of CMV in fibroblasts. We therefore investigated the immunoregulatory effects of CMV-infection in human lung fibroblasts and the impact on replicative senescence. FINDINGS We observed that CMV-infection led to the induction of several immunoregulatory host cell genes associated with the innate and adaptive immune system. These were genes of different function such as genes regulating apoptosis, cytokines/chemokines and genes that are responsible for the detection of pathogens. Some of the genes upregulated following CMV-infection are also upregulated during cellular senescence, indicating that CMV causes an immunological phenotype in fibroblasts, which is partially reminiscent of replicative senescent cells. CONCLUSION In summary our results demonstrate that CMV not only affects the T cell pool but also induces inflammatory processes in human fibroblasts.
Collapse
Affiliation(s)
- Juliane Wolf
- Immunology Division, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, 6020 Innsbruck, Austria.
| | | | | |
Collapse
|