51
|
Role of Polyamines in Asthma Pathophysiology. Med Sci (Basel) 2018; 6:medsci6010004. [PMID: 29316647 PMCID: PMC5872161 DOI: 10.3390/medsci6010004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/31/2017] [Accepted: 01/02/2018] [Indexed: 12/14/2022] Open
Abstract
Asthma is a complex disease of airways, where the interactions of immune and structural cells result in disease outcomes with airway remodeling and airway hyper-responsiveness. Polyamines, which are small-sized, natural super-cations, interact with negatively charged intracellular macromolecules, and altered levels of polyamines and their interactions have been associated with different pathological conditions including asthma. Elevated levels of polyamines have been reported in the circulation of asthmatic patients as well as in the lungs of a murine model of asthma. In various studies, polyamines were found to potentiate the pathogenic potential of inflammatory cells, such as mast cells and granulocytes (eosinophils and neutrophils), by either inducing the release of their pro-inflammatory mediators or prolonging their life span. Additionally, polyamines were crucial in the differentiation and alternative activation of macrophages, which play an important role in asthma pathology. Importantly, polyamines cause airway smooth muscle contraction and thus airway hyper-responsiveness, which is the key feature in asthma pathophysiology. High levels of polyamines in asthma and their active cellular and macromolecular interactions indicate the importance of the polyamine pathway in asthma pathogenesis; therefore, modulation of polyamine levels could be a suitable approach in acute and severe asthma management. This review summarizes the possible roles of polyamines in different pathophysiological features of asthma.
Collapse
|
52
|
Marck A, Antero J, Berthelot G, Saulière G, Jancovici JM, Masson-Delmotte V, Boeuf G, Spedding M, Le Bourg É, Toussaint JF. Are We Reaching the Limits of Homo sapiens? Front Physiol 2017; 8:812. [PMID: 29123486 PMCID: PMC5662890 DOI: 10.3389/fphys.2017.00812] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Echoing scientific and industrial progress, the Twentieth century was an unprecedented period of improvement for human capabilities and performances, with a significant increase in lifespan, adult height, and maximal physiological performance. Analyses of historical data show a major slow down occurring in the most recent years. This triggered large and passionate debates in the academic scene within multiple disciplines; as such an observation could be interpreted as our upper biological limits. Such a new phase of human history may be related to structural and functional limits determined by long term evolutionary constraints, and the interaction between complex systems and their environment. In this interdisciplinary approach, we call into question the validity of subsequent forecasts and projections through innovative and related biomarkers such as sport, lifespan, and height indicators. We set a theoretical framework based on biological and environmental relevance rather than using a typical single-variable forecasting approach. As demonstrated within the article, these new views will have major social, economical, and political implications.
Collapse
Affiliation(s)
- Adrien Marck
- Institut de Recherche bio-Médicale et d'Epidémiologie du Sport (IRMES) EA 7329, Institut National du Sport, de l'Expertise et de la Performance, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France.,Laboratoire Matière et Systèmes Complexes, UMR 7057 Université Paris Diderot, Centre National de la Recherche Scientifique, Université Sorbonne Paris Cité, Paris, France
| | - Juliana Antero
- Institut de Recherche bio-Médicale et d'Epidémiologie du Sport (IRMES) EA 7329, Institut National du Sport, de l'Expertise et de la Performance, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | - Geoffroy Berthelot
- Institut de Recherche bio-Médicale et d'Epidémiologie du Sport (IRMES) EA 7329, Institut National du Sport, de l'Expertise et de la Performance, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France.,Group Adaptation and Prospective, High Council of Public Health, Paris, France.,Research Laboratory for Interdisciplinary Studies, Paris, France
| | - Guillaume Saulière
- Institut de Recherche bio-Médicale et d'Epidémiologie du Sport (IRMES) EA 7329, Institut National du Sport, de l'Expertise et de la Performance, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | | | - Valérie Masson-Delmotte
- Laboratoire des Sciences du Climat et l'Environnement, Institut Pierre Simon Laplace, CEA-Centre National de la Recherche Scientifique, Université de Versailles Saint-Quentin en Yvelines, Gif-sur-Yvette, France
| | - Gilles Boeuf
- Muséum National d'Histoire Naturelle, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | | | - Éric Le Bourg
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Jean-François Toussaint
- Institut de Recherche bio-Médicale et d'Epidémiologie du Sport (IRMES) EA 7329, Institut National du Sport, de l'Expertise et de la Performance, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France.,Group Adaptation and Prospective, High Council of Public Health, Paris, France.,Centre d'Investigations en Médecine du Sport (CIMS), Hôtel-Dieu de Paris, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
53
|
Peng C, Hu W, Weng X, Tong R, Cheng S, Ding C, Xiao H, Lv Z, Xie H, Zhou L, Wu J, Zheng S. Over Expression of Long Non-Coding RNA PANDA Promotes Hepatocellular Carcinoma by Inhibiting Senescence Associated Inflammatory Factor IL8. Sci Rep 2017. [PMID: 28646235 PMCID: PMC5482898 DOI: 10.1038/s41598-017-04045-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
It has been reported that long non-coding RNA PANDA was disregulated in varieties types of tumor, but its expression level and biological role in hepatocellular carcinoma (HCC) remains contradictory. We detected PANDA expression in two independent cohorts (48 HCC patients following liver transplantation and 84 HCC patients following liver resection), and found that PANDA was down-regulated in HCC. Thereafter we explored its function in cancer biology by inversing its low expression. Surprisingly, overexpression of PANDA promoted HCC proliferation and carcinogenesis in vitro and in vivo. Mechanistically, PANDA repressed transcriptional activity of senescence associated inflammatory factor IL8, which leaded to inhibition of cellular senescence. Therefore, our research help to better understand the complex role of PANDA in HCC, and suggest more thoughtful strategies should be applied before it can be treated as a potential therapeutic target.
Collapse
Affiliation(s)
- Chuanhui Peng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Wendi Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Xiaoyu Weng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Rongliang Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Shaobing Cheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Chaofeng Ding
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Heng Xiao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhen Lv
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China
| | - Jian Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China. .,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China. .,Collaborative innovation center for Diagnosis treatment of infectious diseases, Hangzhou, China.
| |
Collapse
|
54
|
Melanocytic nevi and melanoma: unraveling a complex relationship. Oncogene 2017; 36:5771-5792. [PMID: 28604751 DOI: 10.1038/onc.2017.189] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022]
Abstract
Approximately 33% of melanomas are derived directly from benign, melanocytic nevi. Despite this, the vast majority of melanocytic nevi, which typically form as a result of BRAFV600E-activating mutations, will never progress to melanoma. Herein, we synthesize basic scientific insights and data from mouse models with common observations from clinical practice to comprehensively review melanocytic nevus biology. In particular, we focus on the mechanisms by which growth arrest is established after BRAFV600E mutation. Means by which growth arrest can be overcome and how melanocytic nevi relate to melanoma are also considered. Finally, we present a new conceptual paradigm for understanding the growth arrest of melanocytic nevi in vivo termed stable clonal expansion. This review builds upon the canonical hypothesis of oncogene-induced senescence in growth arrest and tumor suppression in melanocytic nevi and melanoma.
Collapse
|
55
|
|
56
|
Aravinthan AD, Alexander GJM. Senescence in chronic liver disease: Is the future in aging? J Hepatol 2016; 65:825-834. [PMID: 27245432 DOI: 10.1016/j.jhep.2016.05.030] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/04/2016] [Accepted: 05/23/2016] [Indexed: 12/25/2022]
Abstract
Cellular senescence is a fundamental, complex mechanism with an important protective role present from embryogenesis to late life across all species. It limits the proliferative potential of damaged cells thus protecting against malignant change, but at the expense of substantial alterations to the microenvironment and tissue homeostasis, driving inflammation, fibrosis and paradoxically, malignant disease if the process is sustained. Cellular senescence has attracted considerable recent interest with recognition of pathways linking aging, malignancy and insulin resistance and the current focus on therapeutic interventions to extend health-span. There are major implications for hepatology in the field of fibrosis and cancer, where cellular senescence of hepatocytes, cholangiocytes, stellate cells and immune cells has been implicated in chronic liver disease progression. This review focuses on cellular senescence in chronic liver disease and explores therapeutic opportunities.
Collapse
Affiliation(s)
- Aloysious D Aravinthan
- Department of Medicine, University of Toronto, Toronto, Canada; National Institute for Health Research (NIHR) Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Graeme J M Alexander
- UCL Institute for Liver and Digestive Health, The Royal Free Trust, London, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
57
|
George RP, Mehta AK, Perez SD, Winterberg P, Cheeseman J, Johnson B, Kwun J, Monday S, Stempora L, Warshaw B, Kirk AD. Premature T Cell Senescence in Pediatric CKD. J Am Soc Nephrol 2016; 28:359-367. [PMID: 27413076 DOI: 10.1681/asn.2016010053] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/07/2016] [Indexed: 12/29/2022] Open
Abstract
An individual's immune function, susceptibility to infection, and response to immunosuppressive therapy are influenced in part by his/her T cell maturation state. Although childhood is the most dynamic period of immune maturation, scant information regarding the variability of T cell maturation in children with renal disease is available. In this study, we compared the T cell phenotype in children with renal failure (n=80) with that in healthy children (n=20) using multiparameter flow cytometry to detect markers of T cell maturation, exhaustion, and senescence known to influence immune function. We correlated data with the degree of renal failure (dialysis or nondialysis), prior immunosuppression use, and markers of inflammation (C-reactive protein and inflammatory cytokines) to assess the influence of these factors on T cell phenotype. Children with renal disease had highly variable and often markedly skewed maturation phenotypes, including CD4/CD8 ratio reversal, increased terminal effector differentiation in CD8+ T cells, reduction in the proportion of naïve T cells, evidence of T cell exhaustion and senescence, and variable loss of T cell CD28 expression. These findings were most significant in patients who had experienced major immune insults, particularly prior immunosuppressive drug exposure. In conclusion, children with renal disease have exceptional heterogeneity in the T cell repertoire. Cognizance of this heterogeneity might inform risk stratification with regard to the balance between infectious risk and response to immunosuppressive therapy, such as that required for autoimmune disease and transplantation.
Collapse
Affiliation(s)
- Roshan P George
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Healthcare of Atlanta, Atlanta, Georgia; .,Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Aneesh K Mehta
- Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Sebastian D Perez
- Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Pamela Winterberg
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Healthcare of Atlanta, Atlanta, Georgia.,Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | | | - Brandi Johnson
- Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Jean Kwun
- Division of Surgery, Duke University, Durham, North Carolina
| | | | - Linda Stempora
- Division of Surgery, Duke University, Durham, North Carolina
| | - Barry Warshaw
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Healthcare of Atlanta, Atlanta, Georgia.,Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Allan D Kirk
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Healthcare of Atlanta, Atlanta, Georgia.,Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and.,Division of Surgery, Duke University, Durham, North Carolina
| |
Collapse
|
58
|
Bonney EA, Krebs K, Saade G, Kechichian T, Trivedi J, Huaizhi Y, Menon R. Differential senescence in feto-maternal tissues during mouse pregnancy. Placenta 2016; 43:26-34. [PMID: 27324096 PMCID: PMC5527294 DOI: 10.1016/j.placenta.2016.04.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/18/2016] [Accepted: 04/22/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Human studies show that fetal membranes have a limited lifespan and undergo telomere-dependent cellular senescence that is augmented by oxidative stress and mediated by p38 mitogen activated protein kinase (MAPK). Further, these studies suggest that fetal membranes are anatomically and physiologically positioned to transmit senescence signals that may initiate parturition at term. METHODS Longitudinal evaluation of feto-maternal tissues from mouse pregnancies was undertaken to determine the molecular progression of senescence during normal pregnancy. On days 10-18 of gestation, C57BL/6 mice were euthanized. Fetal membranes, placenta, and decidua/uterus were collected. Tissues were examined for Telomere length (TL) and the presence of Phosphorylated (P) p38MAPK and p53, p21 and senescence associated β-Galactosidase (SA- β-Gal). FINDINGS Linear regression modeling of observed telomere length as a function of gestational age revealed that beta (β), the slope of the linear regression was negative and significantly different from zero for each tissue (fetal membranes, β = -0.1901 ± 0.03125, p < 0.0001; placenta β = -0.09000 ± 0.03474, p = 0.0135; decidua/uterus β = -0.1317 ± 0.03264, p = 0.0003). Progressive activation p38MAPK was observed in all tissues from days 10 to day18, with the highest activation observed in fetal membranes. Activation of p53 was progressive in fetal membranes. In contrast, active p53 was constitutive in placenta and decidua/uterus throughout gestation. Detection of p21 indicated that pro-senescent change was higher in all compartments on day 18 as compared to other days. The number of SA-β-Gal positive cells increased in fetal membranes as gestation progressed. However, in placenta and uterus and decidua/uterus SA-β-Gal was seen only in days 15 and 18. CONCLUSIONS Telomere dependent p38 and p53 mediated senescence progressed in mouse fetal membranes as gestation advanced. Although senescence is evident, telomere dependent events were not dominant in placenta or decidua/uterus. Fetal membrane senescence may significantly contribute to mechanisms of parturition at term.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Burlington, VT 05404, USA
| | - Kendall Krebs
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Burlington, VT 05404, USA
| | - George Saade
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA
| | - Talar Kechichian
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA
| | - Jayshil Trivedi
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA
| | - Yin Huaizhi
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA.
| |
Collapse
|
59
|
Osteosarcoma: prognosis plateau warrants retinoblastoma pathway targeted therapy. Signal Transduct Target Ther 2016; 1:16001. [PMID: 29263893 PMCID: PMC5657420 DOI: 10.1038/sigtrans.2016.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 12/31/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone cancer in children and adolescents, affecting ~560 young patients in the United States annually. The term OS describes a diverse array of subtypes with varying prognoses, but the majority of tumors are high grade and aggressive. Perhaps because the true etiology of these aggressive tumors remains unknown, advances in OS treatment have reached a discouraging plateau, with only incremental improvements over the past 40 years. Thus, research surrounding the pathogenesis of OS is essential, as it promises to unveil novel therapeutic targets that can attack tumor cells with greater specificity and lower toxicity. Among the candidate molecular targets in OS, the retinoblastoma (RB) pathway demonstrates the highest frequency of inactivation and thus represents a particularly promising avenue for molecular targeted therapy. This review examines the present thinking and practices in OS treatment and specifically highlights the relevance of the RB pathway in osteosarcomagenesis. Through further investigation into RB pathway-related novel therapeutic targets, we believe that a near-term breakthrough in improved OS prognosis is possible.
Collapse
|
60
|
Smith LK, Rao AD, McArthur GA. Targeting metabolic reprogramming as a potential therapeutic strategy in melanoma. Pharmacol Res 2016; 107:42-47. [PMID: 26924126 DOI: 10.1016/j.phrs.2016.02.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/28/2022]
Abstract
Metabolic reprogramming is a recognized hallmark of cancer. In order to support continued proliferation and growth, tumor cells must metabolically adapt to balance their bioenergetic and biosynthetic needs. To achieve this, cancer cells switch from mitochondrial oxidative phosphorylation to predominantly rely on glycolysis, a process known as the "Warburg effect". The BRAF oncogene has recently emerged as a critical regulator of this process in melanoma, bringing to the fore the importance of metabolic reprogramming in the pathogenesis and treatment of metastatic melanoma. In this review, we summarize our current understanding of oncogenic reprogramming of metabolism in BRAF and NRAS mutant melanoma, and highlight emerging evidence supporting a metabolic basis for MAPK pathway inhibitor resistance and metabolic vulnerabilities that may be exploited to overcome this.
Collapse
Affiliation(s)
- Lorey K Smith
- Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Australia
| | - Aparna D Rao
- Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Australia
| | - Grant A McArthur
- Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Australia; Translational Research Laboratory, Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Australia; Department of Pathology, University of Melbourne, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Australia.
| |
Collapse
|
61
|
Idrovo JP, Jacob A, Yang WL, Wang Z, Yen HT, Nicastro J, Coppa GF, Wang P. A deficiency in cold-inducible RNA-binding protein accelerates the inflammation phase and improves wound healing. Int J Mol Med 2016; 37:423-8. [PMID: 26743936 DOI: 10.3892/ijmm.2016.2451] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/23/2015] [Indexed: 11/05/2022] Open
Abstract
Chronic or non-healing wounds are a major concern in clinical practice and these wounds are mostly associated with diabetes, and venous and pressure ulcers. Wound healing is a complex process involving overlapping phases and the primary phase in this complex cascade is the inflammatory state. While inflammation is necessary for wound healing, a prolonged inflammatory phase leads to impaired healing. Cold-inducible RNA-binding protein (CIRP) belongs to a family of cold-shock proteins that are expressed in high levels under stress conditions. Recently, we demonstrated that a deficiency in CIRP led to decreased inflammation and mortality in an experimental model of hemorrhagic shock. Thus, we hypothesized that a deficiency in CIRP would accelerate the inflammatory phase and lead to an improvement in cutaneous wound healing. In this study, to examine this hypothesis, a full-thickness wound was created on the dorsum of wild-type (WT) and CIRP-/- mice. The wound size was measured every other day for 14 days. The wound area was significantly decreased in the CIRP-/- mice by day 9 and continued to decrease until day 14 compared to the WT mice. In a separate cohort, mice were sacrificed on days 3 and 7 after wounding and the skin tissues were harvested for histological analysis and RNA measurements. On day 3, the mRNA expression of tumor necrossis factor (TNF)-α in the skin tissues was increased by 16-fold in the WT mice, whereas these levels were increased by 65-fold in the CIRP-/- mice. Of note on day 7, while the levels of TNF-α remained high in the WT mice, these levels were significantly decreased in the CIRP-/- mice. The histological analysis of the wounded skin tissue indicated an improvement as early as day 3 in the CIRP-/- mice, whereas in the WT mice, infiltrated immune cells were still present on day 7. On day 7 in the CIRP-/- mice, Gr-1 expression was low and CD31 expression was high, whereas in the WT mice, Gr-1 expression was high and CD31 expression was low, indicating that the CIRP-/- mice have already moved into the angiogenesis and tissue formation phase, whereas the WT mice were still in the inflammatory state. These data collectively suggest that a deficiency in CIRP accelerates the wound healing process.
Collapse
Affiliation(s)
- Juan Pablo Idrovo
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY, USA
| | - Asha Jacob
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY, USA
| | - Weng Lang Yang
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY, USA
| | - Zhimin Wang
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY, USA
| | - Hao Ting Yen
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY, USA
| | - Jeffrey Nicastro
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY, USA
| | - Gene F Coppa
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY, USA
| | - Ping Wang
- Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY, USA
| |
Collapse
|
62
|
Differential Proteomic Analysis of Human Placenta-Derived Mesenchymal Stem Cells Cultured on Normal Tissue Culture Surface and Hyaluronan-Coated Surface. Stem Cells Int 2015; 2016:2809192. [PMID: 27057169 PMCID: PMC4709773 DOI: 10.1155/2016/2809192] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 09/07/2015] [Accepted: 10/07/2015] [Indexed: 12/15/2022] Open
Abstract
Our previous results showed that hyaluronan (HA) preserved human placenta-derived mesenchymal stem cells (PDMSC) in a slow cell cycling mode similar to quiescence, the pristine state of stem cells in vivo, and HA was found to prevent murine adipose-derived mesenchymal stem cells from senescence. Here, stable isotope labeling by amino acid in cell culture (SILAC) proteomic profiling was used to evaluate the effects of HA on aging phenomenon in stem cells, comparing (1) old and young passage PDMSC cultured on normal tissue culture surface (TCS); (2) old passage on HA-coated surface (CHA) compared to TCS; (3) old and young passage on CHA. The results indicated that senescence-associated protein transgelin (TAGLN) was upregulated in old TCS. Protein CYR61, reportedly senescence-related, was downregulated in old CHA compared to old TCS. The SIRT1-interacting Nicotinamide phosphoribosyltransferase (NAMPT) increased by 2.23-fold in old CHA compared to old TCS, and is 0.48-fold lower in old TCS compared to young TCS. Results also indicated that components of endoplasmic reticulum associated degradation (ERAD) pathway were upregulated in old CHA compared to old TCS cells, potentially for overcoming stress to maintain cell function and suppress senescence. Our data points to pathways that may be targeted by HA to maintain stem cells youth.
Collapse
|
63
|
Abstract
Compared with traditional 2D adherent cell culture, 3D spheroidal cell aggregates, or spheroids, are regarded as more physiological, and this technique has been exploited in the field of oncology, stem cell biology, and tissue engineering. Mesenchymal stem cells (MSCs) cultured in spheroids have enhanced anti-inflammatory, angiogenic, and tissue reparative/regenerative effects with improved cell survival after transplantation. Cytoskeletal reorganization and drastic changes in cell morphology in MSC spheroids indicate a major difference in mechanophysical properties compared with 2D culture. Enhanced multidifferentiation potential, upregulated expression of pluripotency marker genes, and delayed replicative senescence indicate enhanced stemness in MSC spheroids. Furthermore, spheroid formation causes drastic changes in the gene expression profile of MSC in microarray analyses. In spite of these significant changes, underlying molecular mechanisms and signaling pathways triggering and sustaining these changes are largely unknown.
Collapse
|
64
|
Fernández Larrosa PN, Ruíz Grecco M, Mengual Gómez D, Alvarado CV, Panelo LC, Rubio MF, Alonso DF, Gómez DE, Costas MA. RAC3 more than a nuclear receptor coactivator: a key inhibitor of senescence that is downregulated in aging. Cell Death Dis 2015; 6:e1902. [PMID: 26469953 PMCID: PMC4632280 DOI: 10.1038/cddis.2015.218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 06/24/2015] [Accepted: 07/01/2015] [Indexed: 11/10/2022]
Abstract
Receptor-associated coactivator 3 (RAC3) is a nuclear receptor coactivator usually overexpressed in tumors that exerts oncogenic functions in the cytoplasm and the nucleus. Although as part of its oncogenic actions it was previously identified as an inhibitor of apoptosis and autophagy, its expression is required in order to preserve the pluripotency and embryonic stem cell self-renewal. In this work we investigated its role in cellular senescence. We found that RAC3 overexpression in the nontumoral HEK293 cells inhibits the premature senescence induced by hydrogen peroxide or rapamycin. The mechanism involves not only the inhibition of autophagy early induced by these stimuli in the pathway to senescence, but also the increase in levels and nuclear localization of both the cell cycle suppressors p53/p21 and the longevity promoters FOXO1A, FOXO3A and SIRT1. Furthermore, we found that RAC3 overexpression is required in order to maintain the telomerase activity. In tumoral HeLa cells its activity was inhibited by depletion of RAC3 inducing replicative senescence. Moreover, we demonstrated that in vivo, levels of RAC3 are downregulated in the liver from aged as compared with young rats, whereas the levels of p21 are increased, correlating with the expected senescent cell contents in aged tissues. A similar downregulation of RAC3 was observed in the premature and replicative senescence of human fetal WI-38 cells and premature senescence of hepatocyte HepG2 cell line. Taken together, all these results demonstrate that RAC3 is an inhibitor of senescence whose downregulation in aged individuals could be probably a tumor suppressor mechanism, avoiding the clonal expansion of risky old cells having damaged DNA.
Collapse
Affiliation(s)
- P N Fernández Larrosa
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - M Ruíz Grecco
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - D Mengual Gómez
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - C V Alvarado
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - L C Panelo
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - M F Rubio
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - D F Alonso
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - D E Gómez
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - M A Costas
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| |
Collapse
|
65
|
Suppression of the senescence-associated secretory phenotype (SASP) in human fibroblasts using small molecule inhibitors of p38 MAP kinase and MK2. Biogerontology 2015; 17:305-15. [PMID: 26400758 PMCID: PMC4819486 DOI: 10.1007/s10522-015-9610-z] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/18/2015] [Indexed: 01/10/2023]
Abstract
Senescent cells show an altered secretome profile termed the senescence-associated secretory phenotype (SASP). There is an increasing body of evidence that suggests that the accumulation of SASP-positive senescent cells in humans is partially causal in the observed shift to a low-level pro-inflammatory state in aged individuals. This in turn suggests the SASP as a possible therapeutic target to ameliorate inflammatory conditions in the elderly, and thus a better understanding of the signalling pathways underlying the SASP are required. Prior studies using the early generation p38 MAPK inhibitor SB203580 indicated that p38 signalling was required for the SASP. In this study, we extend these observations using two next-generation p38 inhibitors (UR-13756 and BIRB 796) that have markedly improved selectivity and specificity compared to SB203580, to strengthen the evidence that the SASP is p38-dependent in human fibroblasts. BIRB 796 has an efficacy and toxicity profile that has allowed it to reach Phase III clinical trials, suggesting its possible use to suppress the SASP in vivo. We also demonstrate for the first time a requirement for signalling through the p38 downstream MK2 kinase in the regulation of the SASP using two MK2 inhibitors. Finally, we demonstrate that a commercially-available multiplex cytokine assay technology can be used to detect SASP components in the conditioned medium of cultured fibroblasts from both young and elderly donors. This assay is a high-throughput, multiplex microtitre-based assay system that is highly sensitive, with very low sample requirements, allowing it to be used for low-volume human biological fluids. Our initial studies using existing multiplex plates form the basis for a “SASP signature” assay that could be used as a high-throughput system in a clinical study setting. Our findings therefore provide important steps towards the study of, and intervention in, the SASP in human ageing and age-related disease.
Collapse
|
66
|
Kim HS, Hong JS. Telomere length and telomerase activity are related with immortalization frequency but not with replicative senescence in mammalian embryonic fibroblasts, except human embryonic fibroblasts. Anim Cells Syst (Seoul) 2014. [DOI: 10.1080/19768354.2014.987815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
67
|
Abstract
Cellular senescence is a physiological process of irreversible cell-cycle arrest that contributes to various physiological and pathological processes of aging. Whereas replicative senescence is associated with telomere attrition after repeated cell division, stress-induced premature senescence occurs in response to aberrant oncogenic signaling, oxidative stress, and DNA damage which is independent of telomere dysfunction. Recent evidence indicates that cellular senescence provides a barrier to tumorigenesis and is a determinant of the outcome of cancer treatment. However, the senescence-associated secretory phenotype, which contributes to multiple facets of senescent cancer cells, may influence both cancer-inhibitory and cancer-promoting mechanisms of neighboring cells. Conventional treatments, such as chemo- and radiotherapies, preferentially induce premature senescence instead of apoptosis in the appropriate cellular context. In addition, treatment-induced premature senescence could compensate for resistance to apoptosis via alternative signaling pathways. Therefore, we believe that an intensive effort to understand cancer cell senescence could facilitate the development of novel therapeutic strategies for improving the efficacy of anticancer therapies. This review summarizes the current understanding of molecular mechanisms, functions, and clinical applications of cellular senescence for anticancer therapy.
Collapse
Affiliation(s)
- Minyoung Lee
- Research Center for Radio-senescence, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Korea
| | - Jae-Seon Lee
- Research Center for Radio-senescence, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Korea; Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 400-712, Korea
| |
Collapse
|
68
|
Jamal J, Mustafa MR, Wong PF. Paeonol protects against premature senescence in endothelial cells by modulating Sirtuin 1 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:428-436. [PMID: 24768807 DOI: 10.1016/j.jep.2014.04.025] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/20/2014] [Accepted: 04/12/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Paeonol is a phenolic compound isolated mainly from Moutan cortex, root bark of Chinese Peony tree. Moutan cortex holds a significant value in traditional Chinese medicine for alleviating various oxidative stress-related diseases mainly atherosclerosis and myocardial infarction. The present study seeks to identify the protective mechanisms of paeonol in oxidative stress-induced premature senescence in endothelial cells. MATERIALS AND METHODS HUVECs were pretreated with paeonol or DMSO control at different doses for 24h prior to an exposure of 200μM of reactive oxygen species (ROS) inducer, hydrogen peroxide (H2O2). The protective effects of paeonol against H2O2-induced senescence were evaluated and the activation of Sirtuin 1 pathway by paeonol pretreatment was investigated in HUVECs. RESULTS Paeonol attenuated H2O2-induced cell growth arrest at G0/G1 phase, reduced the percentage of SA-β-Gal positive cells and increased BrdU incorporation. In addition, enzymatic Sirt1 activation assay indicated that paeonol significantly increased lysyl deactylase activity of Sirt1 enzyme with a fold change of 2.4±0.195 (p<0.05). Furthermore, pretreatment with paeonol significantly decreased the levels of p53, acetyl H3K14 and H4K16 protein expression upregulated by H2O2 stimulation. The changes in the histone protein levels were accompanied with an increase in Sirt1 protein expression level. CONCLUSION These findings suggest that paeonol protects endothelial cells against oxidative stress-induced premature senescence by modulating the expressions of Sirt1 protein and its substrates.
Collapse
Affiliation(s)
- Juliana Jamal
- Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
69
|
The senescence-associated secretory phenotype promotes benign prostatic hyperplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:721-31. [PMID: 24434012 DOI: 10.1016/j.ajpath.2013.11.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/11/2013] [Accepted: 11/04/2013] [Indexed: 12/26/2022]
Abstract
Benign prostatic hyperplasia (BPH) is characterized by increased tissue mass in the transition zone of the prostate, which leads to obstruction of urine outflow and considerable morbidity in a majority of older men. Senescent cells accumulate in human tissues, including the prostate, with increasing age. Expression of proinflammatory cytokines is increased in these senescent cells, a manifestation of the senescence-associated secretory phenotype. Multiplex analysis revealed that multiple cytokines are increased in BPH, including GM-CSF, IL-1α, and IL-4, and that these are also increased in senescent prostatic epithelial cells in vitro. Tissue levels of these cytokines were correlated with a marker of senescence (cathepsin D), which was also strongly correlated with prostate weight. IHC analysis revealed the multifocal epithelial expression of cathepsin D and coexpression with IL-1α in BPH tissues. In tissue recombination studies in nude mice with immortalized prostatic epithelial cells expressing IL-1α and prostatic stromal cells, both epithelial and stromal cells exhibited increased growth. Expression of IL-1α in prostatic epithelial cells in a transgenic mouse model resulted in increased prostate size and bladder obstruction. In summary, both correlative and functional evidence support the hypothesis that the senescence-associated secretory phenotype can promote the development of BPH, which is the single most common age-related pathology in older men.
Collapse
|
70
|
|
71
|
Repeated exposure of mouse dermal fibroblasts at a sub-cytotoxic dose of UVB leads to premature senescence: a robust model of cellular photoaging. J Dermatol Sci 2013; 73:49-56. [PMID: 24054498 DOI: 10.1016/j.jdermsci.2013.08.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 08/20/2013] [Accepted: 08/26/2013] [Indexed: 01/14/2023]
Abstract
BACKGROUND Photoaging skin is due to accumulative effect of UV irradiation that mainly imposes its damage on dermal fibroblasts. To mimic the specific cellular responses invoked by long term effect of UVB, it is preferable to develop a photo-damaged model in vitro based on repeated UVB exposure instead of a single exposure. OBJECTIVE To develop a photo-damaged model of fibroblasts by repeated UVB exposure allowing for investigation of molecular mechanism underlying premature senescence and testing of potential anti-photoaging compounds. METHODS Mouse dermal fibroblasts (MDFs) at early passages (passages 1-3) were exposed to a series of 4 sub-cytotoxic dose of UVB. The senescent phenotypes were detected at 24 or 48h after the last irradiation including cell viability, ROS generation, mitochondrial membrane potential, cell cycle, production and degradation of extracellular matrix. RESULTS Repeated exposure of UVB resulted in remarkable features of senescence. It effectively avoided the disadvantages of single dose such as induction of cell death rather than senescence, inadequate stress resulting in cellular self-rehabilitation. CONCLUSION Our work confirms the possibility of detecting cellular machinery that mediates UVB damage to fibroblasts in vitro by repeated exposure, while the potential molecular mechanisms including cell surface receptors, protein kinase signal transduction pathways, and transcription factors remain to be further evaluated.
Collapse
|
72
|
Despars G, Carbonneau CL, Bardeau P, Coutu DL, Beauséjour CM. Loss of the osteogenic differentiation potential during senescence is limited to bone progenitor cells and is dependent on p53. PLoS One 2013; 8:e73206. [PMID: 24009740 PMCID: PMC3756945 DOI: 10.1371/journal.pone.0073206] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/17/2013] [Indexed: 11/18/2022] Open
Abstract
DNA damage can lead to the induction of cellular senescence. In particular, we showed that exposure to ionizing radiation (IR) leads to the senescence of bone marrow-derived multipotent stromal cells (MSC) and osteoblast-like stromal cells (OB–SC), a phenotype associated with bone loss. The mechanism by which IR leads to bone dysfunction is not fully understood. One possibility involves that DNA damage-induced senescence limits the regeneration of bone progenitor cells. Another possibility entails that bone dysfunction arises from the inability of accumulating senescent cells to fulfill their physiological function. Indeed, we show here that exposure to IR prevented the differentiation and mineralization functions of MSC, an effect we found was limited to this population as more differentiated OB–SC could still form mineralize nodules. This is in contrast to adipogenesis, which was inhibited in both IR-induced senescent MSC and 3T3-L1 pre-adipocytes. Furthermore, we demonstrate that IR-induced loss of osteogenic potential in MSC was p53-dependent, a phenotype that correlates with the inability to upregulate key osteogenic transcription factors. These results are the first to demonstrate that senescence impacts osteogenesis in a cell type dependent manner and suggest that the accumulation of senescent osteoblasts is unlikely to significantly contribute to bone dysfunction in a cell autonomous manner.
Collapse
Affiliation(s)
- Geneviève Despars
- Centre de recherche du CHU Ste-Justine, Montréal, Québec, Canada
- Département de pharmacologie Université de Montréal, Montréal, Québec, Canada
| | - Cynthia L. Carbonneau
- Centre de recherche du CHU Ste-Justine, Montréal, Québec, Canada
- Département de pharmacologie Université de Montréal, Montréal, Québec, Canada
| | - Pascal Bardeau
- Centre de recherche du CHU Ste-Justine, Montréal, Québec, Canada
- Département de pharmacologie Université de Montréal, Montréal, Québec, Canada
| | - Daniel L. Coutu
- Stem Cell Dynamics Research Unit, Helmholtz Center Munich - German Research Center for Environmental Health, Munich, Germany
| | - Christian M. Beauséjour
- Centre de recherche du CHU Ste-Justine, Montréal, Québec, Canada
- Département de pharmacologie Université de Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
73
|
|
74
|
del Nogal-Ávila M, Troyano-Suárez N, Román-García P, Cannata-Andía JB, Rodriguez-Puyol M, Rodriguez-Puyol D, Kuro-O M, Ruiz-Torres MP. Amadori products promote cellular senescence activating insulin-like growth factor-1 receptor and down-regulating the antioxidant enzyme catalase. Int J Biochem Cell Biol 2013; 45:1255-64. [DOI: 10.1016/j.biocel.2013.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 03/08/2013] [Accepted: 03/28/2013] [Indexed: 12/30/2022]
|
75
|
Hendrayani SF, Al-Khalaf HH, Aboussekhra A. Curcumin triggers p16-dependent senescence in active breast cancer-associated fibroblasts and suppresses their paracrine procarcinogenic effects. Neoplasia 2013; 15:631-40. [PMID: 23730211 PMCID: PMC3664995 DOI: 10.1593/neo.13478] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 03/18/2013] [Accepted: 03/24/2013] [Indexed: 12/12/2022]
Abstract
Activated cancer-associated fibroblasts (CAFs) or myofibroblasts not only facilitate tumor growth and spread but also affect tumor response to therapeutic agents. Therefore, it became clear that efficient therapeutic regimens should also take into account the presence of these supportive cells and inhibit their paracrine effects. To this end, we tested the effect of low concentrations of curcumin, a pharmacologically safe natural product, on patient-derived primary breast CAF cells. We have shown that curcumin treatment upregulates p16(INK4A) and other tumor suppressor proteins while inactivates the JAK2/STAT3 pathway. This reduced the level of alpha-smooth muscle actin (α-SMA) and the migration/invasion abilities of these cells. Furthermore, curcumin suppressed the expression/secretion of stromal cell-derived factor-1 (SDF-1), interleukin-6 (IL-6), matrix metalloproteinase-2 (MMP-2), MMP-9, and transforming growth factor-β, which impeded their paracrine procarcinogenic potential. Intriguingly, these effects were sustained even after curcumin withdrawal and cell splitting. Therefore, using different markers of senescence [senescence-associated β-galactosidase (SA-β-gal) activity, Ki-67 and Lamin B1 levels, and bromodeoxyuridine incorporation], we have shown that curcumin markedly suppresses Lamin B1 and triggers DNA damage-independent senescence in proliferating but not quiescent breast stromal fibroblasts. Importantly, this curcumin-related senescence was p16(INK4A)-dependent and occurred with no associated inflammatory secretory phenotype. These results indicate the possible inactivation of cancer-associated myofibroblasts and present the first indication that curcumin can trigger DNA damage-independent and safe senescence in stromal fibroblasts.
Collapse
Affiliation(s)
- Siti-Fauziah Hendrayani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Huda H Al-Khalaf
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- The Joint Center for Genomics Research, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
76
|
A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence. Nature 2013; 498:109-12. [PMID: 23685455 DOI: 10.1038/nature12154] [Citation(s) in RCA: 471] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 04/04/2013] [Indexed: 01/20/2023]
Abstract
In response to tenacious stress signals, such as the unscheduled activation of oncogenes, cells can mobilize tumour suppressor networks to avert the hazard of malignant transformation. A large body of evidence indicates that oncogene-induced senescence (OIS) acts as such a break, withdrawing cells from the proliferative pool almost irreversibly, thus crafting a vital pathophysiological mechanism that protects against cancer. Despite the widespread contribution of OIS to the cessation of tumorigenic expansion in animal models and humans, we have only just begun to define the underlying mechanism and identify key players. Although deregulation of metabolism is intimately linked to the proliferative capacity of cells, and senescent cells are thought to remain metabolically active, little has been investigated in detail about the role of cellular metabolism in OIS. Here we show, by metabolic profiling and functional perturbations, that the mitochondrial gatekeeper pyruvate dehydrogenase (PDH) is a crucial mediator of senescence induced by BRAF(V600E), an oncogene commonly mutated in melanoma and other cancers. BRAF(V600E)-induced senescence was accompanied by simultaneous suppression of the PDH-inhibitory enzyme pyruvate dehydrogenase kinase 1 (PDK1) and induction of the PDH-activating enzyme pyruvate dehydrogenase phosphatase 2 (PDP2). The resulting combined activation of PDH enhanced the use of pyruvate in the tricarboxylic acid cycle, causing increased respiration and redox stress. Abrogation of OIS, a rate-limiting step towards oncogenic transformation, coincided with reversion of these processes. Further supporting a crucial role of PDH in OIS, enforced normalization of either PDK1 or PDP2 expression levels inhibited PDH and abrogated OIS, thereby licensing BRAF(V600E)-driven melanoma development. Finally, depletion of PDK1 eradicated melanoma subpopulations resistant to targeted BRAF inhibition, and caused regression of established melanomas. These results reveal a mechanistic relationship between OIS and a key metabolic signalling axis, which may be exploited therapeutically.
Collapse
|
77
|
Sano Y, Kashiwamura Y, Abe M, Dieu LH, Huwyler J, Shimizu F, Haruki H, Maeda T, Saito K, Tasaki A, Kanda T. Stable human brain microvascular endothelial cell line retaining its barrier-specific nature independent of the passage number. ACTA ACUST UNITED AC 2012. [DOI: 10.1111/cen3.12001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yasuteru Sano
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| | - Yoko Kashiwamura
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| | - Masaaki Abe
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| | - Le-Ha Dieu
- Department of Pharmaceutical Sciences; University of Basel; Basel; Switzerland
| | - Jörg Huwyler
- Department of Pharmaceutical Sciences; University of Basel; Basel; Switzerland
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| | - Hiroyo Haruki
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| | - Toshihiko Maeda
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| | - Kazuyuki Saito
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| | - Ayako Tasaki
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi; Japan
| |
Collapse
|
78
|
Toward personalized cell therapies by using stem cells: seven relevant topics for safety and success in stem cell therapy. J Biomed Biotechnol 2012; 2012:758102. [PMID: 23226945 PMCID: PMC3514047 DOI: 10.1155/2012/758102] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 10/18/2012] [Indexed: 02/07/2023] Open
Abstract
Stem cells, both embryonic and adult, due to the potential for application in tissue regeneration have been the target of interest to the world scientific community. In fact, stem cells can be considered revolutionary in the field of medicine, especially in the treatment of a wide range of human diseases. However, caution is needed in the clinical application of such cells and this is an issue that demands more studies. This paper will discuss some controversial issues of importance for achieving cell therapy safety and success. Particularly, the following aspects of stem cell biology will be presented: methods for stem cells culture, teratogenic or tumorigenic potential, cellular dose, proliferation, senescence, karyotyping, and immunosuppressive activity.
Collapse
|
79
|
Tivey HSE, Brook AJC, Rokicki MJ, Kipling D, Davis T. p38 (MAPK) stress signalling in replicative senescence in fibroblasts from progeroid and genomic instability syndromes. Biogerontology 2012; 14:47-62. [PMID: 23112078 PMCID: PMC3627027 DOI: 10.1007/s10522-012-9407-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 10/17/2012] [Indexed: 12/26/2022]
Abstract
Werner Syndrome (WS) is a human segmental progeria resulting from mutations in a DNA helicase. WS fibroblasts have a shortened replicative capacity, an aged appearance, and activated p38 MAPK, features that can be modulated by inhibition of the p38 pathway. Loss of the WRNp RecQ helicase has been shown to result in replicative stress, suggesting that a link between faulty DNA repair and stress-induced premature cellular senescence may lead to premature ageing in WS. Other progeroid syndromes that share overlapping pathophysiological features with WS also show defects in DNA processing, raising the possibility that faulty DNA repair, leading to replicative stress and premature cellular senescence, might be a more widespread feature of premature ageing syndromes. We therefore analysed replicative capacity, cellular morphology and p38 activation, and the effects of p38 inhibition, in fibroblasts from a range of progeroid syndromes. In general, populations of young fibroblasts from non-WS progeroid syndromes do not have a high level of cells with an enlarged morphology and F-actin stress fibres, unlike young WS cells, although this varies between strains. p38 activation and phosphorylated HSP27 levels generally correlate well with cellular morphology, and treatment with the p38 inhibitor SB203580 effects cellular morphology only in strains with enlarged cells and phosphorylated HSP27. For some syndromes fibroblast replicative capacity was within the normal range, whereas for others it was significantly shorter (e.g. HGPS and DKC). However, although in most cases SB203580 extended replicative capacity, with the exception of WS and DKC the magnitude of the effect was not significantly different from normal dermal fibroblasts. This suggests that stress-induced premature cellular senescence via p38 activation is restricted to a small subset of progeroid syndromes.
Collapse
Affiliation(s)
- Hannah S E Tivey
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | | | | | | | | |
Collapse
|
80
|
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
|
81
|
David G. Regulation of oncogene-induced cell cycle exit and senescence by chromatin modifiers. Cancer Biol Ther 2012; 13:992-1000. [PMID: 22825329 DOI: 10.4161/cbt.21116] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Oncogene activation leads to dramatic changes in numerous biological pathways controlling cellular division, and results in the initiation of a transcriptional program that promotes transformation. Conversely, it also triggers an irreversible cell cycle exit called cellular senescence, which allows the organism to counteract the potentially detrimental uncontrolled proliferation of damaged cells. Therefore, a tight transcriptional control is required at the onset of oncogenic signal, coordinating both positive and negative regulation of gene expression. Not surprisingly, numerous chromatin modifiers contribute to the cellular response to oncogenic stress. While these chromatin modifiers were initially thought of as mere mediators of the cellular response to oncogenic stress, recent studies have uncovered a direct and specific regulation of chromatin modifiers by oncogenic signals. We review here the diverse functions of chromatin modifiers in the cellular response to oncogenic stress, and discuss the implications of these findings on the regulation of cell cycle progression and proliferation by activated oncogenes.
Collapse
Affiliation(s)
- Gregory David
- Department of Pharmacology and NYU Cancer Institute, NYU Langone Medical Center, New York, NY, USA.
| |
Collapse
|
82
|
Capparelli C, Whitaker-Menezes D, Guido C, Balliet R, Pestell TG, Howell A, Sneddon S, Pestell RG, Martinez-Outschoorn U, Lisanti MP, Sotgia F. CTGF drives autophagy, glycolysis and senescence in cancer-associated fibroblasts via HIF1 activation, metabolically promoting tumor growth. Cell Cycle 2012; 11:2272-84. [PMID: 22684333 PMCID: PMC3383589 DOI: 10.4161/cc.20717] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Previous studies have demonstrated that loss of caveolin-1 (Cav-1) in stromal cells drives the activation of the TGF-β signaling, with increased transcription of TGF-β target genes, such as connective tissue growth factor (CTGF). In addition, loss of stromal Cav-1 results in the metabolic reprogramming of cancer-associated fibroblasts, with the induction of autophagy and glycolysis. However, it remains unknown if activation of the TGF-β / CTGF pathway regulates the metabolism of cancer-associated fibroblasts. Therefore, we investigated whether CTGF modulates metabolism in the tumor microenvironment. For this purpose, CTGF was overexpressed in normal human fibroblasts or MDA-MB-231 breast cancer cells. Overexpression of CTGF induces HIF-1α-dependent metabolic alterations, with the induction of autophagy/mitophagy, senescence, and glycolysis. Here, we show that CTGF exerts compartment-specific effects on tumorigenesis, depending on the cell-type. In a xenograft model, CTGF overexpressing fibroblasts promote the growth of co-injected MDA-MB-231 cells, without any increases in angiogenesis. Conversely, CTGF overexpression in MDA-MB-231 cells dramatically inhibits tumor growth in mice. Intriguingly, increased extracellular matrix deposition was seen in tumors with either fibroblast or MDA-MB-231 overexpression of CTGF. Thus, the effects of CTGF expression on tumor formation are independent of its extracellular matrix function, but rather depend on its ability to activate catabolic metabolism. As such, CTGF-mediated induction of autophagy in fibroblasts supports tumor growth via the generation of recycled nutrients, whereas CTGF-mediated autophagy in breast cancer cells suppresses tumor growth, via tumor cell self-digestion. Our studies shed new light on the compartment-specific role of CTGF in mammary tumorigenesis, and provide novel insights into the mechanism(s) generating a lethal tumor microenvironment in patients lacking stromal Cav-1. As loss of Cav-1 is a stromal marker of poor clinical outcome in women with primary breast cancer, dissecting the downstream signaling effects of Cav-1 are important for understanding disease pathogenesis, and identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Claudia Capparelli
- The Jefferson Stem Cell Biology and Regenerative Medicine Center, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Stroma and pancreatic ductal adenocarcinoma: an interaction loop. Biochim Biophys Acta Rev Cancer 2012; 1826:170-8. [PMID: 22521638 DOI: 10.1016/j.bbcan.2012.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/06/2012] [Accepted: 04/08/2012] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) has two exceptional features. First, it is a highly lethal disease, with a median survival of less than 6 months and a 5-year survival rate less than 5%. Second, PDA tumor cells are surrounded by an extensive stroma, which accounts for up to 90% of the tumor volume. It is well recognized that stromal microenvironment can accelerate malignant transformation, tumor growth and progression. More importantly, the interaction loop between PDA and its stroma greatly contributes to tumor growth and progression. We propose that the extensive stroma of PDA is closely linked to its poor prognosis. An improved understanding of the mechanisms that contribute to pancreatic tumor growth and progression is therefore urgently needed. Targeting the stroma may thus provide novel prevention, earlier detection and therapeutic options to this deadly malignancy. Accordingly, in this review, we will summarize the mechanism of PDA stroma formation, the role of the stroma in tumor progression and therapy resistance and the potential of stroma-targeted therapeutics strategies.
Collapse
|
84
|
Zimmermann M, Nickl S, Lambers C, Hacker S, Mitterbauer A, Hoetzenecker K, Rozsas A, Ostoros G, Laszlo V, Hofbauer H, Renyi-Vamos F, Klepetko W, Dome B, Ankersmit HJ. Discrimination of clinical stages in non-small cell lung cancer patients by serum HSP27 and HSP70: a multi-institutional case-control study. Clin Chim Acta 2012; 413:1115-20. [PMID: 22465083 DOI: 10.1016/j.cca.2012.03.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 03/09/2012] [Accepted: 03/09/2012] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Lung cancer represents a major healthcare problem. Accordingly, there is an urgent need to identify serum biomarkers for early diagnosis of lung pathology. We have recently described that patients with manifest COPD evidence elevated levels of heat shock proteins (HSPs). Based on these data, we speculated whether HSPs are also increased in patients with diagnosed lung cancer. METHODS Serum levels of HSP27, phospho-HSP27 (pHSP27) and HSP70 in patients with non-small cell lung cancer (NSCLC) diagnosed at an early (stages I-II, n=37) or advanced (stages IIIA-IV, n=72) stage were determined by using ELISA. Healthy smokers (n=24), healthy never-smoker volunteers (n=33) and COPD patients (n=34) according to GOLD classification served as control population. RESULTS Serum levels of HSP27 were elevated in patients with NSCLC diagnosed at an early or advanced stage when compared with both healthy control groups (P<0.005 and P<0.0001 respectively). Statistically significant differences were furthermore found between the groups of patients with early vs. advanced stage NSCLC (P=0.0021). Serum levels of HSP70 were also significantly elevated in patients with NSCLC diagnosed at an early or at an advanced stage when compared with either healthy control groups (P=0.0028 and P<0.0001 respectively). In univariate logistic regression models including healthy subjects and patients with NSCLC, HSP70 had an area under the curve (AUC) of 0.779 (P<0.0001) and HSP27 showed an AUC of 0.870 (P<0.0001). CONCLUSION Our data suggest that serum HSP27 levels might serve as a possible tool to discriminate between early and advanced stages NSCLC.
Collapse
Affiliation(s)
- Matthias Zimmermann
- Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Proliferative capacity of corneal endothelial cells. Exp Eye Res 2011; 95:16-23. [PMID: 21906590 DOI: 10.1016/j.exer.2011.08.014] [Citation(s) in RCA: 222] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 08/10/2011] [Accepted: 08/23/2011] [Indexed: 12/11/2022]
Abstract
The corneal endothelial monolayer helps maintain corneal transparency through its barrier and ionic "pump" functions. This transparency function can become compromised, resulting in a critical loss in endothelial cell density (ECD), corneal edema, bullous keratopathy, and loss of visual acuity. Although penetrating keratoplasty and various forms of endothelial keratoplasty are capable of restoring corneal clarity, they can also have complications requiring re-grafting or other treatments. With the increasing worldwide shortage of donor corneas to be used for keratoplasty, there is a greater need to find new therapies to restore corneal clarity that is lost due to endothelial dysfunction. As a result, researchers have been exploring alternative approaches that could result in the in vivo induction of transient corneal endothelial cell division or the in vitro expansion of healthy endothelial cells for corneal bioengineering as treatments to increase ECD and restore visual acuity. This review presents current information regarding the ability of human corneal endothelial cells (HCEC) to divide as a basis for the development of new therapies. Information will be presented on the positive and negative regulation of the cell cycle as background for the studies to be discussed. Results of studies exploring the proliferative capacity of HCEC will be presented and specific conditions that affect the ability of HCEC to divide will be discussed. Methods that have been tested to induce transient proliferation of HCEC will also be presented. This review will discuss the effect of donor age and endothelial topography on relative proliferative capacity of HCEC, as well as explore the role of nuclear oxidative DNA damage in decreasing the relative proliferative capacity of HCEC. Finally, potential new research directions will be discussed that could take advantage of and/or improve the proliferative capacity of these physiologically important cells in order to develop new treatments to restore corneal clarity.
Collapse
|
86
|
Synthesis of the highly selective p38 MAPK inhibitor UR-13756 for possible therapeutic use in Werner syndrome. Future Med Chem 2011; 2:193-201. [PMID: 21426186 DOI: 10.4155/fmc.09.163] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND UR-13756 is a potent and selective p38 mitogen-activated protein kinase (MAPK) inhibitor, reported to have good bioavailability and pharmacokinetic properties and, thus, is of potential use in the treatment of accelerated aging in Werner syndrome. RESULTS AND DISCUSSION Irradiation of 2-chloroacrylonitrile and methylhydrazine in ethanol at 100 °C gives 1-methyl-3-aminopyrazole, which reacts with 4-fluorobenzaldehyde and a ketone, obtained by Claisen condensation of 4-picoline, in a Hantzsch-type 3-component hereocyclocondensation, to give the pyrazolopyridine UR-13756. UR-13756 shows p38 MAPK inhibitory activity in human telomerase reverse transcriptase-immortalized HCA2 dermal fibroblasts, with an IC(50) of 80 nm, as shown by ELISA, is 100% efficacious for up to 24 h at 1.0 μm and displays excellent kinase selectivity over the related stress-activated c-Jun kinases. In addition, UR-13756 is an effective p38 inhibitor at 1.0 μm in Werner syndrome cells, as shown by immunoblot. CONCLUSION The convergent synthesis of UR-13756 is realized using microwave dielectric heating and provides a highly selective inhibitor that shows excellent selectivity for p38 MAPK over c-Jun N-terminal kinase.
Collapse
|
87
|
Nguyen LTH, Liao S, Ramakrishna S, Chan CK. The role of nanofibrous structure in osteogenic differentiation of human mesenchymal stem cells with serial passage. Nanomedicine (Lond) 2011; 6:961-74. [DOI: 10.2217/nnm.11.26] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Using scaffolds with autologous stem cells is a golden strategy for the treatment of bone defects. In this strategy, human mesenchymal stem cells (hMSCs) have often been isolated and expanded in vitro on a plastic surface to obtain a sufficient cell number before seeding on a suitable scaffold. Materials & Methods: Investigating the influence of serial passages (from passage two to passage eight) on the abilities of proliferation and osteogenic differentiation of hMSCs on 24-well tissue culture polystyrene plates and poly L-lactic acid electrospun nanofibrous scaffolds was performed to determine how prolonged culture affected these cellular abilities and how the nanofibrous scaffolds supported the osteogenic differentiation potential of hMSCs. Results & Conclusion: Serial passage caused adverse changes in hMSCs characteristics, which were indicated by the decline in both proliferation and osteogenic differentiation abilities. Interestingly, the poly L-lactic acid nanofibrous scaffolds showed a significant support in recovering the osteogenic abilities of hMSCs, which had been severely affected by prolonged culture.
Collapse
Affiliation(s)
| | - Susan Liao
- Nanyang Technological University, 637551 Singapore
| | - Seeram Ramakrishna
- National University of Singapore, 117576 Singapore
- Institute of Materials Research & Engineering, 117602 Singapore
| | - Casey K Chan
- National University of Singapore, 117576 Singapore
| |
Collapse
|
88
|
Gram-scale synthesis of the p38α MAPK-inhibitor VX-745 for preclinical studies into Werner syndrome. Future Med Chem 2011; 2:1417-27. [PMID: 21426137 DOI: 10.4155/fmc.10.217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The ATP-competitive p38α MAPK inhibitor VX-745 exhibits an exquisite kinase selectivity profile, is effective in blocking p38 stress signaling in Werner syndrome dermal fibroblasts, has efficacy in clinical trials and may have therapeutic value against Werner syndrome. Previous synthetic routes, however, have only resulted in milligram quantities suitable for cell-based studies, whereas gram quantities would be required for in vivo use. RESULTS & DISCUSSION Microwave irradiation using a stop-flow monomodal microwave reactor has been found to facilitate scale-up of the synthesis of VX-745. Ullmann-type C-S bond formation using thiophenol, chloropyridazine, copper(I) catalyst and diol ligand proceeds rapidly and efficiently in this apparatus for elaboration to the pyrimido[1,6-b]pyridazinone core of VX-745 on gram scale and with good overall yield. CONCLUSION This method delivers the p38 inhibitor VX-745 in sufficient quantities for preclinical studies to rescue the aging phenotype in Werner syndrome.
Collapse
|
89
|
Kim SY, Kang HT, Choi HR, Park SC. Biliverdin reductase A in the prevention of cellular senescence against oxidative stress. Exp Mol Med 2011; 43:15-23. [PMID: 21099244 DOI: 10.3858/emm.2011.43.1.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Biliverdin reductase A (BLVRA), an enzyme that converts biliverdin to bilirubin, has recently emerged as a key regulator of the cellular redox cycle. However, the role of BLVRA in the aging process remains unclear. To study the role of BLVRA in the aging process, we compared the stress responses of young and senescent human diploid fibroblasts (HDFs) to the reactive oxygen species (ROS) inducer, hydrogen peroxide (H2O2). H2O2 markedly induced BLVRA activity in young HDFs, but not in senescent HDFs. Additionally, depletion of BLVRA reduced the H2O2-dependent induction of heme oxygenase-1 (HO-1) in young HDFs, but not in senescent cells, suggesting an aging-dependent differential modulation of responses to oxidative stress. The role of BLVRA in the regulation of cellular senescence was confirmed when lentiviral RNAi- transfected stable primary HDFs with reduced BLVRA expression showed upregulation of the CDK inhibitor family members p16, p53, and p21, followed by cell cycle arrest in G0-G1 phase with high expression of senescence-associated β-galactosidase. Taken together, these data support the notion that BLVRA contributes significantly to modulation of the aging process by adjusting the cellular oxidative status.
Collapse
Affiliation(s)
- Sung Young Kim
- Department of Biochemistry and Molecular Biology, Aging and Apoptosis Research Center, Institute on Aging, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
90
|
Ahmadbeigi N, Shafiee A, Seyedjafari E, Gheisari Y, Vassei M, Amanpour S, Amini S, Bagherizadeh I, Soleimani M. Early spontaneous immortalization and loss of plasticity of rabbit bone marrow mesenchymal stem cells. Cell Prolif 2011; 44:67-74. [PMID: 21199011 DOI: 10.1111/j.1365-2184.2010.00731.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Bone marrow-derived mesenchymal stem cells (BM-MSC) have been widely used for cell therapy and tissue engineering purposes. However, there are still controversies concerning safety of application of these cells after in vitro expansion. Therefore, we aimed to investigate the characteristics of rabbit BM-MSC during long-term culture. MATERIALS AND METHODS In this study, we have examined growth kinetics, morphological changes, differentiation potential and chromosomal abnormalities, as well as tumour formation potential of rabbit BM-MSC in long-term culture. RESULTS AND CONCLUSION We found that shortly after isolation, proliferation rate of rabbit BM-MSC decreases until they enter a dormant phase. During this period of quiescence, the cells are large and multinucleate. After some weeks of dormancy we found that several small mononuclear cells originated from each large multinucleate cell. These newly formed cells proliferated rapidly but had inferior differentiation potential. Although they were immortal, they did not have the capability for tumour formation in soft agar assay or in nude mice. This is the first report of spontaneous, non-tumorigenic immortalization of BM-MSC in rabbits. The phenomenon raises more concern for meticulous monitoring and quality control for using rabbit BM-MSC in cell-based therapies and tissue engineering experiments.
Collapse
Affiliation(s)
- N Ahmadbeigi
- Stem Cell Biology Department, Stem Cell Technology Research Center, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Anversa P, Olivetti G. Cellular Basis of Physiological and Pathological Myocardial Growth. Compr Physiol 2011. [DOI: 10.1002/cphy.cp020102] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
92
|
Ohno-Iwashita Y, Shimada Y, Hayashi M, Inomata M. Plasma membrane microdomains in aging and disease. Geriatr Gerontol Int 2010; 10 Suppl 1:S41-52. [DOI: 10.1111/j.1447-0594.2010.00600.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
93
|
Sarsour EH, Goswami M, Kalen AL, Goswami PC. MnSOD activity protects mitochondrial morphology of quiescent fibroblasts from age associated abnormalities. Mitochondrion 2010; 10:342-9. [PMID: 20206302 DOI: 10.1016/j.mito.2010.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 02/17/2010] [Accepted: 02/22/2010] [Indexed: 01/19/2023]
Abstract
Previously, we have shown manganese superoxide dismutase (MnSOD) activity protects quiescent human normal skin fibroblasts (NHFs) from age associated loss in proliferative capacity. The loss in proliferative capacity of aged vs. young quiescent cells is often characterized as the chronological life span, which is clearly distinct from replicative senescence. We investigate the hypothesis that MnSOD activity protects the mitochondrial morphology from age associated damage and preserves the chronological life span of quiescent fibroblasts. Aged quiescent NHFs exhibited abnormalities in mitochondrial morphology including abnormal cristae formation and increased number of vacuoles. These results correlate with the levels of cellular reactive oxygen species (ROS) and mitochondrial morphology in MnSOD homozygous and heterozygous knockout mouse embryonic fibroblasts. The abnormalities in mitochondrial morphology in aged quiescent NHFs cultured in presence of 21% oxygen concentration were more severe than NHFs cultured in 4% oxygen environment. The alteration in mitochondrial morphology was associated with a significant increase in cell population doubling: 54h in 21% compared to 44h in 4% oxygen environment. Overexpression of MnSOD decreased ROS levels, and preserved mitochondrial morphology in aged quiescent NHFs. These results demonstrate that MnSOD activity protects mitochondrial morphology and preserves the proliferative capacities of quiescent NHFs from age associated loss.
Collapse
Affiliation(s)
- Ehab H Sarsour
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | |
Collapse
|
94
|
Microwave-assisted synthesis of a pyrazolyl ketone library for evaluation as p38 MAPK inhibitors in Werner syndrome cells. Future Med Chem 2010; 2:203-13. [DOI: 10.4155/fmc.09.160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: The pyrazolyl ketone motif of RO3201195, which exhibits good oral bioavailability and high selectivity for p38 MAPK over other kinases, is a key pharmacophore that could find application in the treatment of Werner syndrome. Results and discussion: Microwave irradiation promotes Knoevenagel condensation of a β-ketonitrile and formamidine, to give β-aminovinyl ketones, and their subsequent cyclocondensation with a subset of hydrazines to provide rapid access to a 24-membered library of pyrazolyl ketones. The library was evaluated in human hTERT-immortalized HCA2 dermal cells and Werner syndrome cells. Conclusion: Four compounds display comparable, if not slightly improved, potency over RO3201195.
Collapse
|
95
|
Sarsour EH, Kumar MG, Chaudhuri L, Kalen AL, Goswami PC. Redox control of the cell cycle in health and disease. Antioxid Redox Signal 2009; 11:2985-3011. [PMID: 19505186 PMCID: PMC2783918 DOI: 10.1089/ars.2009.2513] [Citation(s) in RCA: 284] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 06/04/2009] [Accepted: 06/05/2009] [Indexed: 01/11/2023]
Abstract
The cellular oxidation and reduction (redox) environment is influenced by the production and removal of reactive oxygen species (ROS). In recent years, several reports support the hypothesis that cellular ROS levels could function as ''second messengers'' regulating numerous cellular processes, including proliferation. Periodic oscillations in the cellular redox environment, a redox cycle, regulate cell-cycle progression from quiescence (G(0)) to proliferation (G(1), S, G(2), and M) and back to quiescence. A loss in the redox control of the cell cycle could lead to aberrant proliferation, a hallmark of various human pathologies. This review discusses the literature that supports the concept of a redox cycle controlling the mammalian cell cycle, with an emphasis on how this control relates to proliferative disorders including cancer, wound healing, fibrosis, cardiovascular diseases, diabetes, and neurodegenerative diseases. We hypothesize that reestablishing the redox control of the cell cycle by manipulating the cellular redox environment could improve many aspects of the proliferative disorders.
Collapse
Affiliation(s)
- Ehab H Sarsour
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa , Iowa City, Iowa, USA
| | | | | | | | | |
Collapse
|
96
|
Udompataikul M, Sripiroj P, Palungwachira P. An oral nutraceutical containing antioxidants, minerals and glycosaminoglycans improves skin roughness and fine wrinkles. Int J Cosmet Sci 2009; 31:427-35. [PMID: 19570098 DOI: 10.1111/j.1468-2494.2009.00513.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Various nutraceuticals (dietary supplements) are claimed to have cutaneous antiageing properties, however, there are a limited number of research studies supporting these claims. The objective of this research was to study the effectiveness of an oral nutraceutical containing antioxidants, minerals and glycosaminoglycans on cutaneous ageing. In this double-blind, placebo-controlled trial, 60 women aged 35-60 years were randomized to receive oral dietary supplement (n = 30) or placebo (n = 30), once daily for 12 weeks. The depth of skin roughness and fine wrinkles were measured using surface evaluation of skin parameters for living skin (Visioscan) at baseline, and at the 4, 8 and 12 weeks of treatment. Surface evaluation using a replica film (Visiometer) at baseline and at the 12th week of treatment was also carried out. Statistical differences in objective skin improvement were assessed by the independent t-test. The volunteers' satisfaction was tested using the chi-squared test. The baseline depth of skin roughness and fine wrinkles in the treatment group and the placebo group were 100.5 and 100 mum, respectively. At the end of the study, the depth of skin roughness and fine wrinkles in the treatment group showed a 21.2% improvement, whereas improvement in the control group was 1.7%. This difference was statistically significant (P < 0.001). With regard to the volunteers' satisfaction, there was no statistically significant decrease in the homogenization of skin colour, however, a statistically significant reduction in pore size and depth of skin roughness and fine wrinkles were observed (P < 0.05). No side effects were noted throughout the study. The oral dietary supplement containing antioxidants, minerals and glycosaminoglycans improved skin roughness and fine wrinkles but did not affect skin colour change in female volunteers.
Collapse
Affiliation(s)
- M Udompataikul
- Skin Center, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand.
| | | | | |
Collapse
|
97
|
Liu Y, Chen Y, Richardson B. Decreased DNA methyltransferase levels contribute to abnormal gene expression in "senescent" CD4(+)CD28(-) T cells. Clin Immunol 2009; 132:257-65. [PMID: 19394279 DOI: 10.1016/j.clim.2009.03.529] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 03/20/2009] [Accepted: 03/25/2009] [Indexed: 12/29/2022]
Abstract
A senescent CD4(+)CD28(-) T cell subset develops with aging and in chronic inflammatory diseases like rheumatoid arthritis, and is implicated in plaque rupture and myocardial infarctions. This subset is pro-inflammatory, cytotoxic for endothelial cells, and aberrantly expresses genes like CD70, perforin and killer cell immunoglobulin-like receptor (KIR) genes. Why CD4(+)CD28(-) cells overexpress these genes is unclear. We found that the CD70, perforin and KIR2DL4 promoters are demethylated in CD4(+)CD28(-) T cells, and that DNA methyltransferase 1 (Dnmt1) and Dnmt3a levels are decreased in this subset. siRNA "knockdown" of Dnmt1, but not Dnmt3a, in CD4(+)CD28(+) T cells caused similar demethylation and overexpression of KIR2DL4, perforin and CD70, while simultaneous knockdown of Dnmt1 and Dnmt3a caused greater demethylation and overexpression of these genes than Dnmt1 alone. We conclude that decreased Dnmt1 and Dnmt3a cause demethylation and overexpression of these and perhaps other genes in CD4(+)CD28(-) cells, potentially contributing to pathologic functions by this subset.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medicine, University of Michigan, USA
| | | | | |
Collapse
|
98
|
Wang CY, Kim HH, Hiroi Y, Sawada N, Salomone S, Benjamin LE, Walsh K, Moskowitz MA, Liao JK. Obesity increases vascular senescence and susceptibility to ischemic injury through chronic activation of Akt and mTOR. Sci Signal 2009; 2:ra11. [PMID: 19293429 DOI: 10.1126/scisignal.2000143] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity and age are important risk factors for cardiovascular disease. However, the signaling mechanism linking obesity with age-related vascular senescence is unknown. Here we show that mice fed a high-fat diet show increased vascular senescence and vascular dysfunction compared to mice fed standard chow and are more prone to peripheral and cerebral ischemia. All of these changes involve long-term activation of the protein kinase Akt. In contrast, mice with diet-induced obesity that lack Akt1 are resistant to vascular senescence. Rapamycin treatment of diet-induced obese mice or of transgenic mice with long-term activation of endothelial Akt inhibits activation of mammalian target of rapamycin (mTOR)-rictor complex 2 and Akt, prevents vascular senescence without altering body weight, and reduces the severity of limb necrosis and ischemic stroke. These findings indicate that long-term activation of Akt-mTOR signaling links diet-induced obesity with vascular senescence and cardiovascular disease.
Collapse
Affiliation(s)
- Chao-Yung Wang
- Brigham and Women's Hospital and Harvard Medical School, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Lambers C, Hacker S, Posch M, Hoetzenecker K, Pollreisz A, Lichtenauer M, Klepetko W, Ankersmit HJ. T cell senescence and contraction of T cell repertoire diversity in patients with chronic obstructive pulmonary disease. Clin Exp Immunol 2009; 155:466-75. [PMID: 19220836 PMCID: PMC2669523 DOI: 10.1111/j.1365-2249.2008.03835.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2008] [Indexed: 01/08/2023] Open
Abstract
Pathogenetic mechanisms leading to chronic obstructive pulmonary disease (COPD) remain poorly understood. Because clonogenic T cells (CD4(+)CD28(null)) were shown to be increased in autoimmune diseases we hypothesized that CD4(+)CD28(null) T cells play a role in COPD. Here we describe that enhanced presence of CD4(+)CD28(null) cells is associated with impaired lung function. Sixty-four patients and controls were included. T cell phenotype was analysed using flow cytometry. Enzyme-linked immunosorbent assays were utilized to determine cytokines. Statistical evaluations were performed using non-parametric group comparisons and correlations. A logistic regression model was used to determine predictive values of CD4(+)CD28(null) in the diagnosis of COPD. Populations of CD4(+) T cells lacking surface co-stimulatory CD28 were enlarged significantly in evaluated patients when compared with controls. Natural killer (NK)-like T cell receptors (CD94, 158) and intracellular perforin, granzyme B were increased in CD4(+)CD28(null) cells. Cytokine production after triggering of peripheral blood mononuclear cells (PBMCs) was elevated in patients at early disease stages. Receiver operating characteristic curve plotting revealed that presence of CD4(+)CD28(null) T cells has a diagnostic value. These CD4(+)CD28(null) T cells show increased expression of NK-like T cell receptors (CD94, 158) and intracellular perforin and granzyme B. Furthermore, triggering of PBMCs obtained from patients with mild COPD led to increased interferon-gamma and tumour necrosis factor-alpha production in vitro compared with controls. Our finding of increased CD4(+)CD28(null) T cells in COPD indicates that chronic antigen exposure, e.g. through contents of smoke, leads to loss of CD28 and up-regulation of NK cell receptors expression on T cells in susceptible patients.
Collapse
Affiliation(s)
- C Lambers
- Department of Pulmonary Medicine, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Iovine B, Nino M, Irace C, Bevilacqua MA, Monfrecola G. Ultraviolet B and A irradiation induces fibromodulin expression in human fibroblasts in vitro. Biochimie 2008; 91:364-72. [PMID: 19041686 DOI: 10.1016/j.biochi.2008.10.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Accepted: 10/10/2008] [Indexed: 11/25/2022]
Abstract
Ultraviolet (UV) radiation affects the extracellular matrix (ECM) of the human skin. The small leucine-rich repeat protein fibromodulin interacts with type I and II collagen fibrils, thereby affecting ECM assembly. The aim of this study was to evaluate whether short wave UV (UVB) or long wave UV (UVA) irradiation influences fibromodulin expression. Exponentially growing human fibroblasts (IMR-90 cells) were exposed to increasing doses of UVB (2.5-60 mJ/cm(2)) or UVA (0.5-10 J/cm(2)). After UV irradiation fibromodulin, p21 and GADD45 levels were evaluated as well as cell viability, reactive oxygen species formation (ROS) and DNA damage. We found that fibromodulin expression: (i) increased after UVB and UVA irradiation; (ii) was 10-fold higher after UVA (10 J/cm(2)) versus 5-fold with UVB (10 mJ/cm(2)); (iii) correlated with reactive oxygen species formation, particularly after UVA; and (iv) was linked to the DNA damage binding protein (DDB1) translocation in the nucleus, particularly after UVB. These results further suggest that the UV-induced fibromodulin increase could counteract the UV-induced connective tissue damage, promoting the assembly of new collagen fibrils.
Collapse
Affiliation(s)
- Barbara Iovine
- Department of Biochemistry, Faculty of Biotechnology, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy
| | | | | | | | | |
Collapse
|