1
|
Javali PS, Sekar M, Kumar A, Thirumurugan K. Dynamics of redox signaling in aging via autophagy, inflammation, and senescence. Biogerontology 2023; 24:663-678. [PMID: 37195483 DOI: 10.1007/s10522-023-10040-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
Review paper attempts to explain the dynamic aspects of redox signaling in aging through autophagy, inflammation, and senescence. It begins with ROS source in the cell, then states redox signaling in autophagy, and regulation of autophagy in aging. Next, we discuss inflammation and redox signaling with various pathways involved: NOX pathway, ROS production via TNF-α, IL-1β, xanthine oxidase pathway, COX pathway, and myeloperoxidase pathway. Also, we emphasize oxidative damage as an aging marker and the contribution of pathophysiological factors to aging. In senescence-associated secretory phenotypes, we link ROS with senescence, aging disorders. Relevant crosstalk between autophagy, inflammation, and senescence using a balanced ROS level might reduce age-related disorders. Transducing the context-dependent signal communication among these three processes at high spatiotemporal resolution demands other tools like multi-omics aging biomarkers, artificial intelligence, machine learning, and deep learning. The bewildering advancement of technology in the above areas might progress age-related disorders diagnostics with precision and accuracy.
Collapse
Affiliation(s)
- Prashanth S Javali
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Mouliganesh Sekar
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Ashish Kumar
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Kavitha Thirumurugan
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
2
|
Morrow CS, Arndt ZP, Klosa PC, Peng B, Zewdie EY, Benayoun BA, Moore DL. Adult fibroblasts use aggresomes only in distinct cell-states. Sci Rep 2022; 12:15001. [PMID: 36056070 PMCID: PMC9440096 DOI: 10.1038/s41598-022-19055-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
The aggresome is a protein turnover system in which proteins are trafficked along microtubules to the centrosome for degradation. Despite extensive focus on aggresomes in immortalized cell lines, it remains unclear if the aggresome is conserved in all primary cells and all cell-states. Here we examined the aggresome in primary adult mouse dermal fibroblasts shifted into four distinct cell-states. We found that in response to proteasome inhibition, quiescent and immortalized fibroblasts formed aggresomes, whereas proliferating and senescent fibroblasts did not. Using this model, we generated a resource to provide a characterization of the proteostasis networks in which the aggresome is used and transcriptomic features associated with the presence or absence of aggresome formation. Using this resource, we validate a previously reported role for p38 MAPK signaling in aggresome formation and identify TAK1 as a novel driver of aggresome formation upstream of p38 MAPKs. Together, our data demonstrate that the aggresome is a non-universal protein degradation system which can be used cell-state specifically and provide a resource for studying aggresome formation and function.
Collapse
Affiliation(s)
| | - Zachary P Arndt
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Payton C Klosa
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Bo Peng
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Eden Y Zewdie
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Darcie L Moore
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
3
|
Woo J, Shin S, Ji H, Ryu D, Cho E, Kim Y, Kim J, Park D, Jung E. Isatis tinctoria L. Leaf Extract Inhibits Replicative Senescence in Dermal Fibroblasts by Regulating mTOR-NF-κB-SASP Signaling. Nutrients 2022; 14:nu14091979. [PMID: 35565945 PMCID: PMC9102489 DOI: 10.3390/nu14091979] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023] Open
Abstract
Senescent fibroblasts progressively deteriorate the functional properties of skin tissue. Senescent cells secrete senescence-associated secretory phenotype (SASP) factor, which causes the aging of surrounding non-senescent cells and accelerates aging in the individuals. Recent findings suggested the senomorphic targeting of the SASP regulation as a new generation of effective therapeutics. We investigated whether Isatis tinctoria L. leaf extract (ITE) inhibited senescence biomarkers p53, p21CDKN1A, and p16INK4A gene expression, and SASP secretions by inhibiting cellular senescence in the replicative senescent human dermal fibroblast (RS-HDF). ITE has been demonstrated to inhibit the secretion of SASP factors in several senomorphic types by regulating the MAPK/NF-κB pathway via its inhibitory effect on mTOR. ITE suppressed the inflammatory response by inhibiting mTOR, MAPK, and IκBα phosphorylation, and blocking the nuclear translocation of NF-κB. In addition, we observed that autophagy pathway was related to inhibitory effect of ITE on cellular senescence. From these results, we concluded that ITE can prevent and restore senescence by blocking the activation and secretion of senescence-related factors generated from RS-HDFs through mTOR-NF-κB regulation.
Collapse
Affiliation(s)
- Jieun Woo
- BioSpectrum Life Science Institute, 767, Sinsu-ro, Yongin-si 16827, Korea; (J.W.); (S.S.); (H.J.); (D.R.); (E.C.); (D.P.)
| | - Seoungwoo Shin
- BioSpectrum Life Science Institute, 767, Sinsu-ro, Yongin-si 16827, Korea; (J.W.); (S.S.); (H.J.); (D.R.); (E.C.); (D.P.)
| | - Hyanggi Ji
- BioSpectrum Life Science Institute, 767, Sinsu-ro, Yongin-si 16827, Korea; (J.W.); (S.S.); (H.J.); (D.R.); (E.C.); (D.P.)
| | - Dehun Ryu
- BioSpectrum Life Science Institute, 767, Sinsu-ro, Yongin-si 16827, Korea; (J.W.); (S.S.); (H.J.); (D.R.); (E.C.); (D.P.)
| | - Eunae Cho
- BioSpectrum Life Science Institute, 767, Sinsu-ro, Yongin-si 16827, Korea; (J.W.); (S.S.); (H.J.); (D.R.); (E.C.); (D.P.)
| | - Youngseok Kim
- Shinsegae International Technology Innovation Center, 449, Dosan-daero, Seoul 06015, Korea; (Y.K.); (J.K.)
| | - Junoh Kim
- Shinsegae International Technology Innovation Center, 449, Dosan-daero, Seoul 06015, Korea; (Y.K.); (J.K.)
| | - Deokhoon Park
- BioSpectrum Life Science Institute, 767, Sinsu-ro, Yongin-si 16827, Korea; (J.W.); (S.S.); (H.J.); (D.R.); (E.C.); (D.P.)
| | - Eunsun Jung
- BioSpectrum Life Science Institute, 767, Sinsu-ro, Yongin-si 16827, Korea; (J.W.); (S.S.); (H.J.); (D.R.); (E.C.); (D.P.)
- Correspondence:
| |
Collapse
|
4
|
Maity P, Singh K, Krug L, Koroma A, Hainzl A, Bloch W, Kochanek S, Wlaschek M, Schorpp-Kistner M, Angel P, Ignatius A, Geiger H, Scharffetter-Kochanek K. Persistent JunB activation in fibroblasts disrupts stem cell niche interactions enforcing skin aging. Cell Rep 2021; 36:109634. [PMID: 34469740 DOI: 10.1016/j.celrep.2021.109634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/15/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023] Open
Abstract
Fibroblasts residing in the connective tissues constitute the stem cell niche, particularly in organs such as skin. Although the effect of fibroblasts on stem cell niches and organ aging is an emerging concept, the underlying mechanisms are largely unresolved. We report a mechanism of redox-dependent activation of transcription factor JunB, which, through concomitant upregulation of p16INK4A and repression of insulin growth factor-1 (IGF-1), initiates the installment of fibroblast senescence. Fibroblast senescence profoundly disrupts the metabolic and structural niche, and its essential interactions with different stem cells thus enforces depletion of stem cells pools and skin tissue decline. In fact, silencing of JunB in a fibroblast-niche-specific manner-by reinstatement of IGF-1 and p16 levels-restores skin stem cell pools and overall skin tissue integrity. Here, we report a role of JunB in the control of connective tissue niche and identified targets to combat skin aging and associated pathologies.
Collapse
Affiliation(s)
- Pallab Maity
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany; Aging Research Center (ARC), 89081 Ulm, Germany.
| | - Karmveer Singh
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany; Aging Research Center (ARC), 89081 Ulm, Germany
| | - Linda Krug
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Albert Koroma
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany; Aging Research Center (ARC), 89081 Ulm, Germany
| | - Adelheid Hainzl
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Wilhelm Bloch
- Institute of Cardiology and Sports Medicine, Molecular and cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany
| | - Stefan Kochanek
- Department of Gene Therapy, University of Ulm, 89081 Ulm, Germany
| | - Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Marina Schorpp-Kistner
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Ulm University, 89081 Ulm, Germany
| | - Hartmut Geiger
- Aging Research Center (ARC), 89081 Ulm, Germany; Institute of Molecular Medicine and Stem Cell Aging, Ulm University, 89081 Ulm, Germany; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Karin Scharffetter-Kochanek
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany; Aging Research Center (ARC), 89081 Ulm, Germany.
| |
Collapse
|
5
|
Ngoi NY, Liew AQ, Chong SJF, Davids MS, Clement MV, Pervaiz S. The redox-senescence axis and its therapeutic targeting. Redox Biol 2021; 45:102032. [PMID: 34147844 PMCID: PMC8220395 DOI: 10.1016/j.redox.2021.102032] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/14/2022] Open
Abstract
Significance Cellular growth arrest, associated with ‘senescence’, helps to safeguard against the accumulation of DNA damage which is often recognized as the underlying mechanism of a wide variety of age-related pathologies including cancer. Cellular senescence has also been described as a ‘double-edged sword’. In cancer, for example, the creation of an immune-suppressive milieu by senescent tumor cells through the senescence-associated secretory phenotype contributes toward carcinogenesis and cancer progression. Recent advances The potential for cellular senescence to confer multi-faceted effects on tissue fate has led to a rejuvenated interest in its landscape and targeting. Interestingly, redox pathways have been described as both triggers and propagators of cellular senescence, leading to intricate cross-links between both pathways. Critical issues In this review, we describe the mechanisms driving cellular senescence, the interface with cellular redox metabolism as well as the role that chemotherapy-induced senescence plays in secondary carcinogenesis. Notably, the role that anti-apoptotic proteins of the Bcl-2 family play in inducing drug resistance via mechanisms that involve senescence induction. Future directions Though the therapeutic targeting of senescent cells as cancer therapy remains in its infancy, we summarize the current development of senotherapeutics, including recognized senotherapies, as well as the repurposing of drugs as senomorphic/senolytic candidates.
Collapse
Affiliation(s)
- Natalie Yl Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Angeline Qx Liew
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Stephen J F Chong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew S Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marie-Veronique Clement
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Medicine Healthy Longevity Program, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Medicine Healthy Longevity Program, National University of Singapore, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; Faculté de Medicine, University of Paris, Paris, France.
| |
Collapse
|
6
|
Effect of Antioxidants on the Fibroblast Replicative Lifespan In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6423783. [PMID: 33029282 PMCID: PMC7530501 DOI: 10.1155/2020/6423783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/04/2020] [Accepted: 09/12/2020] [Indexed: 12/20/2022]
Abstract
Replicative senescence is an unalterable growth arrest of primary cells in the culture system. It has been reported that aging in vivo is related to the limited replicative capacity that normal somatic cells show in vitro. If oxidative damage contributes to the lifespan limitation, antioxidants are expected to extend the replicative lifespan of fibroblasts. This article critically reviews the results of experiments devoted to this problem performed within the last decades under conditions of in vitro culture. The results of studied are heterogeneous, some papers showing no effects of antioxidants; most finding limited enhancement of reproductive capacity of fibroblasts, some reporting a significant extension of replicative lifespan (RLS). Both natural and synthetic antioxidants were found to extend the RLS of fibroblasts, either by a direct antioxidant effect or, indirectly, by activation of signaling pathways and activation of proteasomes or hormetic effects. Most significant prolongation of RLS was reported so far for nicotinamide, N-hydroxylamines, carnosine and Methylene Blue. These results may be of importance for the design of skin-protecting cosmetics.
Collapse
|
7
|
Clement M, Luo L. Organismal Aging and Oxidants beyond Macromolecules Damage. Proteomics 2020; 20:e1800400. [DOI: 10.1002/pmic.201800400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/20/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Marie‐Veronique Clement
- Department of BiochemistryYong Loo Lin School of MedicineNational University of Singapore Singapore 117596 Singapore
- National University of Singapore Graduate School for Integrative Sciences and Engineering Singapore 117456 Singapore
| | - Le Luo
- Department of BiochemistryYong Loo Lin School of MedicineNational University of Singapore Singapore 117596 Singapore
| |
Collapse
|
8
|
Komaravolu RK, Waltmann MD, Konaniah E, Jaeschke A, Hui DY. ApoER2 (Apolipoprotein E Receptor-2) Deficiency Accelerates Smooth Muscle Cell Senescence via Cytokinesis Impairment and Promotes Fibrotic Neointima After Vascular Injury. Arterioscler Thromb Vasc Biol 2019; 39:2132-2144. [PMID: 31412739 PMCID: PMC6761011 DOI: 10.1161/atvbaha.119.313194] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Genome-wide studies showed that mutation in apoER2 (apolipoprotein E receptor-2) is additive with ε4 polymorphism in the APOE gene on cardiovascular disease risk in humans. ApoE or apoER2 deficiency also accelerates atherosclerosis lesion necrosis in hypercholesterolemic mice and promotes neointima formation after vascular injury. This study tests the hypothesis that apoE and apoER2 modulate vascular occlusive diseases through distinct mechanisms. Approach and Results: Carotid endothelial denudation induced robust neointima formation in both apoE-/- and apoER2-deficient Lrp8-/- mice. The intima in apoE-/- mice was rich in smooth muscle cells, but the intima in Lrp8-/- mice was cell-poor and rich in extracellular matrix. Vascular smooth muscle cells isolated from apoE-/- mice were hyperplastic whereas Lrp8-/- smooth muscle cells showed reduced proliferation but responded robustly to TGF (transforming growth factor)-β-induced fibronectin synthesis indicative of a senescence-associated secretory phenotype, which was confirmed by increased β-galactosidase activity, p16INK4a immunofluorescence, and number of multinucleated cells. Western blot analysis of cell cycle-associated proteins showed that apoER2 deficiency promotes cell cycle arrest at the metaphase/anaphase. Coimmunoprecipitation experiments revealed that apoER2 interacts with the catalytic subunit of protein phosphatase 2A. In the absence of apoER2, PP2A-C (protein phosphatase 2A catalytic subunit) failed to interact with CDC20 (cell-division cycle protein 20) thus resulting in inactive anaphase-promoting complex and impaired cell cycle exit. CONCLUSIONS This study showed that apoER2 participates in APC (anaphase-promoting complex)/CDC20 complex formation during mitosis, and its absence impedes cytokinesis abscission thereby accelerating premature cell senescence and vascular disease. This mechanism is distinct from apoE deficiency, which causes smooth muscle cell hyperplasia to accelerate vascular disease.
Collapse
Affiliation(s)
- Ravi K. Komaravolu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Meaghan D. Waltmann
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Eddy Konaniah
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - David Y. Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| |
Collapse
|
9
|
Rezatabar S, Karimian A, Rameshknia V, Parsian H, Majidinia M, Kopi TA, Bishayee A, Sadeghinia A, Yousefi M, Monirialamdari M, Yousefi B. RAS/MAPK signaling functions in oxidative stress, DNA damage response and cancer progression. J Cell Physiol 2019; 234:14951-14965. [PMID: 30811039 DOI: 10.1002/jcp.28334] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/12/2019] [Accepted: 01/15/2019] [Indexed: 01/24/2023]
Abstract
Mitogen-activated protein kinase (MAPK) signaling pathways organize a great constitution network that regulates several physiological processes, like cell growth, differentiation, and apoptotic cell death. Due to the crucial importance of this signaling pathway, dysregulation of the MAPK signaling cascades is involved in the pathogenesis of various human cancer types. Oxidative stress and DNA damage are two important factors which in common lead to carcinogenesis through dysregulation of this signaling pathway. Reactive oxygen species (ROS) are a common subproduct of oxidative energy metabolism and are considered to be a significant physiological modulator of several intracellular signaling pathways including the MAPK pathway. Studies demonstrated that the MAP kinases extracellular signal-regulated kinase (ERK) 1/2 and p38 were activated in response to oxidative stress. In addition, DNA damage is a partly common circumstance in cell life and may result in mutation, cancer, and even cell death. Recently, accumulating evidence illustrated that the MEK/ERK pathway is associated with the suitable performance of cellular DNA damage response (DDR), the main pathway of tumor suppression. During DDR, the MEK/ERK pathway is regularly activated, which contributes to the appropriate activation of DDR checkpoints to inhibit cell division. Therefore, the aim of this review is to comprehensively discuss the critical function of MAPK signaling in oxidative stress, DNA damage, and cancer progression.
Collapse
Affiliation(s)
- Setareh Rezatabar
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ansar Karimian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Cancer & Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Vahid Rameshknia
- Faculty of Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Tayebeh Azramezani Kopi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, Florida
| | - Ali Sadeghinia
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Fujiwara T, Dohi T, Maan ZN, Rustad KC, Kwon SH, Padmanabhan J, Whittam AJ, Suga H, Duscher D, Rodrigues M, Gurtner GC. Age-associated intracellular superoxide dismutase deficiency potentiates dermal fibroblast dysfunction during wound healing. Exp Dermatol 2017; 28:485-492. [PMID: 28677217 DOI: 10.1111/exd.13404] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2017] [Indexed: 01/05/2023]
Abstract
Reactive oxygen species (ROS) impair wound healing through destructive oxidation of intracellular proteins, lipids and nucleic acids. Intracellular superoxide dismutase (SOD1) regulates ROS levels and plays a critical role in tissue homoeostasis. Recent evidence suggests that age-associated wound healing impairments may partially result from decreased SOD1 expression. We investigated the mechanistic basis by which increased oxidative stress links to age-associated impaired wound healing. Fibroblasts were isolated from unwounded skin of young and aged mice, and myofibroblast differentiation was assessed by measuring α-smooth muscle actin and collagen gel contraction. Excisional wounds were created on young and aged mice to study the healing rate, ROS levels and SOD1 expression. A mechanistic link between oxidative stress and fibroblast function was explored by assessing the TGF-β1 signalling pathway components in young and aged mice. Age-related wounds displayed reduced myofibroblast differentiation and delayed wound healing, consistent with a decrease in the in vitro capacity for fibroblast-myofibroblast transition following oxidative stress. Young fibroblasts with normal SOD1 expression exhibited increased phosphorylation of ERK in response to elevated ROS. In contrast, aged fibroblasts with reduced SOD1 expression displayed a reduced capacity to modulate intracellular ROS. Collectively, age-associated wound healing impairments are associated with fibroblast dysfunction that is likely the result of decreased SOD1 expression and subsequent dysregulation of intracellular ROS. Strategies targeting these mechanisms may suggest a new therapeutic approach in the treatment of chronic non-healing wounds in the aged population.
Collapse
Affiliation(s)
- Toshihiro Fujiwara
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Teruyuki Dohi
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zeshaan N Maan
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristine C Rustad
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sun Hyung Kwon
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Alexander J Whittam
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hirotaka Suga
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Dominik Duscher
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Melanie Rodrigues
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Noh EM, Kim JM, Hong OY, Song HK, Kim JS, Kwon KB, Lee YR. PTEN inhibits replicative senescence-induced MMP-1 expression by regulating NOX4-mediated ROS in human dermal fibroblasts. J Cell Mol Med 2017; 21:3113-3116. [PMID: 28557373 PMCID: PMC5661253 DOI: 10.1111/jcmm.13220] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/03/2017] [Indexed: 12/01/2022] Open
Abstract
The biological function of NADPH oxidase (NOX) is the generation of reactive oxygen species (ROS). ROS, primarily arising from oxidative cell metabolism, play a major role in both chronological ageing and photoageing. ROS in extrinsic and intrinsic skin ageing may be assumed to induce the expression of matrix metalloproteinases. NADPH oxidase is closely linked with phosphatidylinositol 3‐OH kinase (PI3K) signalling. Protein kinase C (PKC), a downstream molecule of PI3K, is essential for superoxide generation by NADPH oxidase. However, the effect of PTEN and NOX4 in replicative‐aged MMPs expression has not been determined. In this study, we confirmed that inhibition of the PI3K signalling pathway by PTEN gene transfer abolished the NOX‐4 and MMP‐1 expression. Also, NOX‐4 down‐expression of replicative‐aged skin cells abolished the MMP‐1 expression and ROS generation. These results suggest that increase of MMP‐1 expression by replicative‐induced ROS is related to the change in the PTEN and NOX expression.
Collapse
Affiliation(s)
- Eun-Mi Noh
- Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan City, Jeonbuk, South Korea
| | - Jeong-Mi Kim
- Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan City, Jeonbuk, South Korea
| | - On-Yu Hong
- Department of Biochemistry, Institute of Medical Science, Chonbuk National University Medical School, Jeonju, South Korea
| | - Hyun-Kyung Song
- Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan City, Jeonbuk, South Korea
| | - Jong-Suk Kim
- Department of Biochemistry, Institute of Medical Science, Chonbuk National University Medical School, Jeonju, South Korea
| | - Kang-Beom Kwon
- Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan City, Jeonbuk, South Korea.,Department of Korean Physiology, Wonkwang University School of Korean Medicine, Iksan City, Jeonbuk, South Korea
| | - Young-Rae Lee
- Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan City, Jeonbuk, South Korea.,Department of Oral Biochemistry, Institute of Biomaterials, Implant, School of Dentistry, Wonkwang University, Iksan City, Jeonbuk, South Korea
| |
Collapse
|
12
|
Eberhardt K, Beleites C, Marthandan S, Matthäus C, Diekmann S, Popp J. Raman and Infrared Spectroscopy Distinguishing Replicative Senescent from Proliferating Primary Human Fibroblast Cells by Detecting Spectral Differences Mainly Due to Biomolecular Alterations. Anal Chem 2017; 89:2937-2947. [DOI: 10.1021/acs.analchem.6b04264] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Katharina Eberhardt
- Leibniz Institute of Photonic Technology e. V., Albert-Einstein-Str. 9, 07745 Jena, Germany
- Institute
for Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Claudia Beleites
- Leibniz Institute of Photonic Technology e. V., Albert-Einstein-Str. 9, 07745 Jena, Germany
- Chemometric Consulting and Chemometrix GmbH, Södeler Weg 19, 61200 Wölfersheim, Germany
| | - Shiva Marthandan
- Department
of Molecular Biology, Leibniz Institute on Aging − Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Christian Matthäus
- Leibniz Institute of Photonic Technology e. V., Albert-Einstein-Str. 9, 07745 Jena, Germany
- Institute
for Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Stephan Diekmann
- Department
of Molecular Biology, Leibniz Institute on Aging − Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology e. V., Albert-Einstein-Str. 9, 07745 Jena, Germany
- Institute
for Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| |
Collapse
|
13
|
Skin Aging-Dependent Activation of the PI3K Signaling Pathway via Downregulation of PTEN Increases Intracellular ROS in Human Dermal Fibroblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6354261. [PMID: 28003865 PMCID: PMC5149682 DOI: 10.1155/2016/6354261] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 07/04/2016] [Accepted: 07/26/2016] [Indexed: 01/04/2023]
Abstract
Reactive oxygen species (ROS) play a major role in both chronological aging and photoaging. ROS induce skin aging through their damaging effect on cellular constituents. However, the origins of ROS have not been fully elucidated. We investigated that ROS generation of replicative senescent fibroblasts is generated by the modulation of phosphatidylinositol 3,4,5-triphosphate (PIP3) metabolism. Reduction of the PTEN protein, which dephosphorylates PIP3, was responsible for maintaining a high level of PIP3 in replicative cells and consequently mediated the activation of the phosphatidylinositol-3-OH kinase (PI3K)/Akt pathway. Increased ROS production was blocked by inhibition of PI3K or protein kinase C (PKC) or by NADPH oxidase activating in replicative senescent cells. These data indicate that the signal pathway to ROS generation in replicative aged skin cells can be stimulated by reduced PTEN level. Our results provide new insights into skin aging-associated modification of the PI3K/NADPH oxidase signaling pathway and its relationship with a skin aging-dependent increase of ROS in human dermal fibroblasts.
Collapse
|
14
|
Panebianco C, Oben JA, Vinciguerra M, Pazienza V. Senescence in hepatic stellate cells as a mechanism of liver fibrosis reversal: a putative synergy between retinoic acid and PPAR-gamma signalings. Clin Exp Med 2016; 17:269-280. [PMID: 27655446 DOI: 10.1007/s10238-016-0438-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 09/08/2016] [Indexed: 12/16/2022]
Abstract
Hepatic stellate cells (HSCs), also known as perisinusoidal cells, are pericytes found in the perisinusoidal space of the liver. HSCs are the major cell type involved in liver fibrosis, which is the formation of scar tissue in response to liver damage. When the liver is damaged, stellate cells can shift into an activated state, characterized by proliferation, contractility and chemotaxis. The activated HSCs secrete collagen scar tissue, which can lead to cirrhosis. Recent studies have shown that in vivo activation of HSCs by fibrogenic agents can eventually lead to senescence of these cells, which would contribute to reversal of fibrosis although it may also favor the insurgence of liver cancer. HSCs in their non-active form store huge amounts of retinoic acid derivatives in lipid droplets, which are progressively depleted upon cell activation in injured liver. Retinoic acid is a metabolite of vitamin A (retinol) that mediates the functions of vitamin A, generally required for growth and development. The precise function of retinoic acid and its alterations in HSCs has yet to be elucidated, and nonetheless in various cell types retinoic acid and its receptors (RAR and RXR) are known to act synergistically with peroxisome proliferator-activated receptor gamma (PPAR-gamma) signaling through the activity of transcriptional heterodimers. Here, we review the recent advancements in the understanding of how retinoic acid signaling modulates the fibrogenic potential of HSCs and proposes a synergistic combined action with PPAR-gamma in the reversal of liver fibrosis.
Collapse
Affiliation(s)
- Concetta Panebianco
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, Viale dei Cappuccini, 1, San Giovanni Rotondo, FG, Italy
| | - Jude A Oben
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London (UCL), London, UK
| | - Manlio Vinciguerra
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London (UCL), London, UK.,Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czech Republic.,Centro Studi Fegato (CSF)-Liver Research Center, Fondazione Italiana Fegato, Trieste, Italy
| | - Valerio Pazienza
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, Viale dei Cappuccini, 1, San Giovanni Rotondo, FG, Italy.
| |
Collapse
|
15
|
Mitochondria: Are they causal players in cellular senescence? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1373-9. [DOI: 10.1016/j.bbabio.2015.05.017] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 12/25/2022]
|
16
|
Abstract
SIGNIFICANCE The molecular mechanism of aging is still vigorously debated, although a general consensus exists that mitochondria are significantly involved in this process. However, the previously postulated role of mitochondrial-derived reactive oxygen species (ROS) as the damaging agents inducing functional loss in aging has fallen out of favor in the recent past. In this review, we critically examine the role of ROS in aging in the light of recent advances on the relationship between mitochondrial structure and function. RECENT ADVANCES The functional mitochondrial respiratory chain is now recognized as a reflection of the dynamic association of respiratory complexes in the form of supercomplexes (SCs). Besides providing kinetic advantage (channeling), SCs control ROS generation by the respiratory chain, thus providing a means to regulate ROS levels in the cell. Depending on their concentration, these ROS are either physiological signals essential for the life of the cell or toxic species that damage cell structure and functions. CRITICAL ISSUES We propose that under physiological conditions the dynamic nature of SCs reversibly controls the generation of ROS as signals involved in mitochondrial-nuclear communication. During aging, there is a progressive loss of control of ROS generation so that their production is irreversibly enhanced, inducing a vicious circle in which signaling is altered and structural damage takes place. FUTURE DIRECTIONS A better understanding on the forces affecting SC association would allow the manipulation of ROS generation, directing these species to their physiological signaling role.
Collapse
Affiliation(s)
- Maria Luisa Genova
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| | - Giorgio Lenaz
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| |
Collapse
|
17
|
Fu WL, Li J, Chen G, Li Q, Tang X, Zhang CH. Mesenchymal Stem Cells Derived from Peripheral Blood Retain Their Pluripotency, but Undergo Senescence During Long-Term Culture. Tissue Eng Part C Methods 2015; 21:1088-97. [PMID: 25996678 DOI: 10.1089/ten.tec.2014.0595] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Peripheral blood-derived mesenchymal stem cells (PB-MSCs) show promise as a source of cells for autologous transplantation because they can be harvested through minimally invasive procedures. To ensure adequate numbers of cells for transplantation and tissue regeneration, PB-MSCs must first be cultured and expanded in vitro, but whether long-term passage modifies their properties has been poorly understood. In this study we triggered production of PB-MSCs in rabbits using granulocyte colony-stimulating factor (G-CSF) and AMD3100, and then isolated and expanded the cells in culture until they reached a state of senescence, usually after about 20 passages. Cultures of low-, middle-, and high-passage numbers were compared in terms of morphology, proliferative capacity, phenotype, differentiation potential, apoptosis, metabolic indicators, and senescence. As passage number increased, MSCs retained their elongated spindle shape, but became larger and flatter, slowed in growth gradually, and increased proportion of cells showed G1 arrest. The proportions of apoptotic cells, production of reactive oxygen species (ROS), and ADP/ATP ratio increased with passage number. Expression of senescence-associated β-galactosidase increased, while telomerase activity decreased. On the other hand, cultures did not show significant changes in phenotype or lose their ability to differentiate into three lineages as passage number increased. These results suggest that PB-MSCs maintain their stem cell properties during prolonged culturing, but they undergo senescence that may be due to apoptosis and production of ROS. These findings may help to standardize in vitro production of PB-MSCs for tissue engineering.
Collapse
Affiliation(s)
- Wei-Li Fu
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Jian Li
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Gang Chen
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Qi Li
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Xin Tang
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Cheng-Hao Zhang
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| |
Collapse
|
18
|
Hwang ES. Senescence suppressors: their practical importance in replicative lifespan extension in stem cells. Cell Mol Life Sci 2014; 71:4207-19. [PMID: 25052377 PMCID: PMC11113678 DOI: 10.1007/s00018-014-1685-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 06/23/2014] [Accepted: 07/14/2014] [Indexed: 02/06/2023]
Abstract
Recent animal and clinical studies report promising results for the therapeutic utilization of stem cells in regenerative medicine. Mesenchymal stem cells (MSCs), with their pluripotent nature, have advantages over embryonic stem cells in terms of their availability and feasibility. However, their proliferative activity is destined to slow by replicative senescence, and the limited proliferative potential of MSCs not only hinders the preparation of sufficient cells for in vivo application, but also draws a limitation on their potential for differentiation. This calls for the development of safe and efficient means to increase the proliferative as well as differentiation potential of MSCs. Recent advances have led to a better understanding of the underlying mechanisms and significance of cellular senescence, facilitating ways to manipulate the replicative lifespan of a variety of primary cells, including MSCs. This paper introduces a class of proteins that function as senescence suppressors. Like tumor suppressors, these proteins are lost in senescence, while their forced expression delays the onset of senescence. Moreover, treatments that increase the expression or the activity of senescence suppressors, therefore, cause expansion of the replicative and differentiation potential of MSCs. The nature of the activities and putative underlying mechanisms of the senescence suppressors will be discussed to facilitate their evaluation.
Collapse
Affiliation(s)
- Eun Seong Hwang
- Department of Life Science, University of Seoul, Dongdaemungu, Jeonnongdong 90, Seoul, 130-743, Republic of Korea,
| |
Collapse
|
19
|
Alili L, Diekmann J, Giesen M, Holtkötter O, Brenneisen P. A drug-induced accelerated senescence (DIAS) is a possibility to study aging in time lapse. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9658. [PMID: 24833306 PMCID: PMC4082584 DOI: 10.1007/s11357-014-9658-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 04/14/2014] [Indexed: 05/28/2023]
Abstract
Currently, the oxidative stress (or free radical) theory of aging is the most popular explanation of how aging occurs at the molecular level. Accordingly, a stress-induced senescence-like phenotype of human dermal fibroblasts can be induced in vitro by the exposure of human diploid fibroblasts to subcytotoxic concentrations of hydrogen peroxide. However, several biomarkers of replicative senescence e.g. cell cycle arrest and enlarged morphology are abrogated 14 days after treatment, indicating that reactive oxygen species (ROS) rather acts as a trigger for short-term senescence (1-3 days) than being responsible for the maintenance of the senescence-like phenotype. Further, DNA-damaging factors are discussed resulting in a permanent senescent cell type. To induce long-term premature senescence and to understand the molecular alterations occurring during the aging process, we analyzed mitomycin C (MMC) as an alkylating DNA-damaging agent and ROS producer. Human dermal fibroblasts (HDF), used as model for skin aging, were exposed to non-cytotoxic concentrations of MMC and analyzed for potential markers of cellular aging, for example enlarged morphology, activity of senescence-associated-ß-galactosidase, cell cycle arrest, increased ROS production and MMP1-activity, which are well-documented for HDF in replicative senescence. Our data show that mitomycin C treatment results in a drug-induced accelerated senescence (DIAS) with long-term expression of senescence markers, demonstrating that a combination of different susceptibility factors, here ROS and DNA alkylation, are necessary to induce a permanent senescent cell type.
Collapse
Affiliation(s)
- Lirija Alili
- Institute of Biochemistry & Molecular Biology I, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany,
| | | | | | | | | |
Collapse
|
20
|
Correia-Melo C, Hewitt G, Passos JF. Telomeres, oxidative stress and inflammatory factors: partners in cellular senescence? LONGEVITY & HEALTHSPAN 2014; 3:1. [PMID: 24472138 PMCID: PMC3922784 DOI: 10.1186/2046-2395-3-1] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 12/02/2013] [Indexed: 12/31/2022]
Abstract
Senescence, the state of irreversible cell-cycle arrest, plays paradoxical albeit important roles in vivo: it protects organisms against cancer but also contributes to age-related loss of tissue function. The DNA damage response (DDR) has a central role in cellular senescence. Not only does it contribute to the irreversible loss of replicative capacity but also to the production and secretion of reactive oxygen species (ROS), and bioactive peptides collectively known as the senescence-associated secretory phenotype (SASP). Both ROS and the SASP have been shown to impact on senescence in an autocrine as well as paracrine fashion; however, the underlying mechanisms are not well understood. In this review we describe our current understanding of cellular senescence, examine in detail the intricate pathways linking the DDR, ROS and SASP, and evaluate their impact on the stability of the senescent phenotype.
Collapse
Affiliation(s)
| | | | - João F Passos
- Ageing Research Laboratories, Centre for Integrated Systems Biology of Ageing and Nutrition, Institute for Ageing and Health, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| |
Collapse
|
21
|
Yang TK, Lee YH, Paudel U, Bhattarai G, Yun BS, Hwang PH, Yi HK. Davallialactone from mushroom reduced premature senescence and inflammation on glucose oxidative stress in human diploid fibroblast cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:7089-7095. [PMID: 23802590 DOI: 10.1021/jf401691y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Mushrooms are both food and a source of natural compounds of biopharmaceutical interest. The purpose of this study was to clarify whether davallialactone from mushroom extract affected the pathogenesis of hyperglycemia oxidative stress and the aging process in human diploid fibroblast (HDF) cells. The high-glucose state with glucose oxidase resulted in glucose oxidative stress, induction of inflammatory molecules, dysfunction of antioxidant molecules, and activation of mitogen-activated protein kinase (MAPKs) and its downstream signaling in old HDF cells. The exposure of glucose oxidative stress in middle-stage cells led to stress-induced premature senescence (SIPS) via senescence-associated β-galactosidase (SA β-gal) activity and displayed replicative senescence phenomena. However, davallialactone reduces the pathogenesis of glucose oxidative stress and the aging process through down-regulation of SA β-gal activity. These results strongly suggest that natural compounds, especially mushroom extract davallialactone, improve the pathogenesis of glucose oxidative stress and the aging process. Hence, davallialactone has potential in the treatment of diabetes mellitus or age-related disease complications.
Collapse
Affiliation(s)
- Tae-Ki Yang
- Department of Pediatrics, School of Medicine, Chonbuk National University , Jeonju 561-712, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
22
|
Treiber N, Maity P, Singh K, Ferchiu F, Wlaschek M, Scharffetter-Kochanek K. The role of manganese superoxide dismutase in skin aging. DERMATO-ENDOCRINOLOGY 2013; 4:232-5. [PMID: 23467724 PMCID: PMC3583882 DOI: 10.4161/derm.21819] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The free radical theory of aging postulates that the production of mitochondrial reactive oxygen species is the major determinant of aging and lifespan. The skin represents an excellent and accessible model organ to study aging that is characterized by atrophy, wrinkle formation, reduced tensile strength and impaired wound healing. Oxidative stress as a consequence of an imbalance in prooxidants and antioxidants with increased ROS concentrations has been demonstrated in the aged skin in vitro and in vivo, suggesting the important role of the antioxidant balance. Here we will summarize recent data on the role of the mitochondrial superoxide dismutase 2 in skin aging.
Collapse
Affiliation(s)
- Nicolai Treiber
- Department of Dermatology and Allergic Diseases; University of Ulm; Ulm, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Treiber N, Maity P, Singh K, Kohn M, Keist AF, Ferchiu F, Sante L, Frese S, Bloch W, Kreppel F, Kochanek S, Sindrilaru A, Iben S, Högel J, Ohnmacht M, Claes LE, Ignatius A, Chung JH, Lee MJ, Kamenisch Y, Berneburg M, Nikolaus T, Braunstein K, Sperfeld AD, Ludolph AC, Briviba K, Wlaschek M, Florin L, Angel P, Scharffetter-Kochanek K. Accelerated aging phenotype in mice with conditional deficiency for mitochondrial superoxide dismutase in the connective tissue. Aging Cell 2011; 10:239-54. [PMID: 21108731 DOI: 10.1111/j.1474-9726.2010.00658.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The free radical theory of aging postulates that the production of mitochondrial reactive oxygen species is the major determinant of aging and lifespan. Its role in aging of the connective tissue has not yet been established, even though the incidence of aging-related disorders in connective tissue-rich organs is high, causing major disability in the elderly. We have now addressed this question experimentally by creating mice with conditional deficiency of the mitochondrial manganese superoxide dismutase in fibroblasts and other mesenchyme-derived cells of connective tissues in all organs. Here, we have shown for the first time that the connective tissue-specific lack of superoxide anion detoxification in the mitochondria results in reduced lifespan and premature onset of aging-related phenotypes such as weight loss, skin atrophy, kyphosis (curvature of the spine), osteoporosis and muscle degeneration in mutant mice. Increase in p16(INK4a) , a robust in vivo marker for fibroblast aging, may contribute to the observed phenotype. This novel model is particularly suited to decipher the underlying mechanisms and to develop hopefully novel connective tissue-specific anti-aging strategies.
Collapse
Affiliation(s)
- Nicolai Treiber
- Department of Dermatology and Allergic Diseases, University of Ulm, Maienweg 12, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Cho S, Hwang ES. Fluorescence-based detection and quantification of features of cellular senescence. Methods Cell Biol 2011; 103:149-88. [PMID: 21722803 DOI: 10.1016/b978-0-12-385493-3.00007-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cellular senescence is a spontaneous organismal defense mechanism against tumor progression which is raised upon the activation of oncoproteins or other cellular environmental stresses that must be circumvented for tumorigenesis to occur. It involves growth-arrest state of normal cells after a number of active divisions. There are multiple experimental routes that can drive cells into a state of senescence. Normal somatic cells and cancer cells enter a state of senescence upon overexpression of oncogenic Ras or Raf protein or by imposing certain kinds of stress such as cellular tumor suppressor function. Both flow cytometry and confocal imaging analysis techniques are very useful in quantitative analysis of cellular senescence phenomenon. They allow quantitative estimates of multiple different phenotypes expressed in multiple cell populations simultaneously. Here we review the various types of fluorescence methodologies including confocal imaging and flow cytometry that are frequently utilized to study a variety of senescence. First, we discuss key cell biological changes occurring during senescence and review the current understanding on the mechanisms of these changes with the goal of improving existing protocols and further developing new ones. Next, we list specific senescence phenotypes associated with each cellular trait along with the principles of their assay methods and the significance of the assay outcomes. We conclude by selecting appropriate references that demonstrate a typical example of each method.
Collapse
Affiliation(s)
- Sohee Cho
- Department of Life Science, University of Seoul, Seoul, Republic of Korea
| | | |
Collapse
|
25
|
Sun C, Liu X, Qi L, Xu J, Zhao J, Zhang Y, Zhang S, Miao J. Modulation of vascular endothelial cell senescence by integrin β4. J Cell Physiol 2010; 225:673-81. [PMID: 20509141 DOI: 10.1002/jcp.22262] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Increasing evidence has demonstrated that the senescence of vascular endothelial cells (VECs) has critical roles in the pathogenesis of vascular dysfunction. Finding important factors that regulate VEC senescence will help provide novel therapeutic strategies for vascular disorders. Previously, we found that integrin β4 was involved in VEC senescence. However, the mechanism underlying VEC senescence mediated by integrin β4 remains poorly understand. In this study, we used a mouse in vivo model and showed that the level of integrin β4 in the endothelium of mouse thoracic aorta was increased during natural aging and atherosclerosis. Furthermore, we found that H-ras, caveolin-1, and AP-1 were implicated in the senescent signal pathway mediated by integrin β4 in human umbilical vein ECs (HUVECs). Knockdown of integrin β4 could attenuate HUVEC senescent features, including increased interleukin-8 (IL-8) release and decreased endothelial nitric oxide synthase (eNOS) and NO levels and mitochondrial membrane potential in vitro. Our findings provide new clues illustrating the mechanism of VEC senescence. Integrin β4 might be a potential target for therapy in cardiovascular diseases.
Collapse
Affiliation(s)
- ChunHui Sun
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Mallette FA, Moiseeva O, Calabrese V, Mao B, Gaumont-Leclerc MF, Ferbeyre G. Transcriptome analysis and tumor suppressor requirements of STAT5-induced senescence. Ann N Y Acad Sci 2010; 1197:142-51. [DOI: 10.1111/j.1749-6632.2010.05192.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
27
|
Hackl M, Brunner S, Fortschegger K, Schreiner C, Micutkova L, Mück C, Laschober GT, Lepperdinger G, Sampson N, Berger P, Herndler-Brandstetter D, Wieser M, Kühnel H, Strasser A, Rinnerthaler M, Breitenbach M, Mildner M, Eckhart L, Tschachler E, Trost A, Bauer JW, Papak C, Trajanoski Z, Scheideler M, Grillari-Voglauer R, Grubeck-Loebenstein B, Jansen-Dürr P, Grillari J. miR-17, miR-19b, miR-20a, and miR-106a are down-regulated in human aging. Aging Cell 2010; 9:291-6. [PMID: 20089119 PMCID: PMC2848978 DOI: 10.1111/j.1474-9726.2010.00549.x] [Citation(s) in RCA: 285] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aging is a multifactorial process where deterioration of body functions is driven by stochastic damage while counteracted by distinct genetically encoded repair systems. To better understand the genetic component of aging, many studies have addressed the gene and protein expression profiles of various aging model systems engaging different organisms from yeast to human. The recently identified small non-coding miRNAs are potent post-transcriptional regulators that can modify the expression of up to several hundred target genes per single miRNA, similar to transcription factors. Increasing evidence shows that miRNAs contribute to the regulation of most if not all important physiological processes, including aging. However, so far the contribution of miRNAs to age-related and senescence-related changes in gene expression remains elusive. To address this question, we have selected four replicative cell aging models including endothelial cells, replicated CD8+ T cells, renal proximal tubular epithelial cells, and skin fibroblasts. Further included were three organismal aging models including foreskin, mesenchymal stem cells, and CD8+ T cell populations from old and young donors. Using locked nucleic acid-based miRNA microarrays, we identified four commonly regulated miRNAs, miR-17 down-regulated in all seven; miR-19b and miR-20a, down-regulated in six models; and miR-106a down-regulated in five models. Decrease in these miRNAs correlated with increased transcript levels of some established target genes, especially the cdk inhibitor p21/CDKN1A. These results establish miRNAs as novel markers of cell aging in humans.
Collapse
Affiliation(s)
- Matthias Hackl
- Aging and Immortalization Research, Department of Biotechnology, University of Natural Resources and Applied Life SciencesVienna, Austria, Muthgasse 18, A-1190 Vienna
| | - Stefan Brunner
- Departments of Immunology, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Klaus Fortschegger
- Aging and Immortalization Research, Department of Biotechnology, University of Natural Resources and Applied Life SciencesVienna, Austria, Muthgasse 18, A-1190 Vienna
| | - Carina Schreiner
- Aging and Immortalization Research, Department of Biotechnology, University of Natural Resources and Applied Life SciencesVienna, Austria, Muthgasse 18, A-1190 Vienna
| | - Lucia Micutkova
- Molecular and Cell Biology, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Christoph Mück
- Molecular and Cell Biology, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Gerhard T Laschober
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Günter Lepperdinger
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Natalie Sampson
- Endocrinology, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Peter Berger
- Endocrinology, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Dietmar Herndler-Brandstetter
- Departments of Immunology, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Matthias Wieser
- Aging and Immortalization Research, Department of Biotechnology, University of Natural Resources and Applied Life SciencesVienna, Austria, Muthgasse 18, A-1190 Vienna
| | - Harald Kühnel
- Department of Natural Sciences, Institute of Physiology, University of Veterinary Medicine ViennaVeterinärplatz 1, A-1210 Wien, Austria
| | - Alois Strasser
- Department of Natural Sciences, Institute of Physiology, University of Veterinary Medicine ViennaVeterinärplatz 1, A-1210 Wien, Austria
| | - Mark Rinnerthaler
- Department of Genetics, University of SalzburgHeilbrunnerstraße 34, 5020 Salzburg, Austria
| | - Michael Breitenbach
- Department of Genetics, University of SalzburgHeilbrunnerstraße 34, 5020 Salzburg, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of ViennaA-1090 Vienna, Austria
| | - Leopold Eckhart
- Department of Dermatology, Medical University of ViennaA-1090 Vienna, Austria
| | - Erwin Tschachler
- Department of Dermatology, Medical University of ViennaA-1090 Vienna, Austria
| | - Andrea Trost
- Department of Dermatology, SALK and Paracelsus Medical UniversitySalzburg, Austria
| | - Johann W Bauer
- Department of Dermatology, SALK and Paracelsus Medical UniversitySalzburg, Austria
| | - Christine Papak
- Institute for Genomics and Bioinformatics and Christian Doppler Laboratory for Genomics and Bioinformatics, Graz University of TechnologyPetersgasse 14, 8010 Graz, Austria
| | - Zlatko Trajanoski
- Institute for Genomics and Bioinformatics and Christian Doppler Laboratory for Genomics and Bioinformatics, Graz University of TechnologyPetersgasse 14, 8010 Graz, Austria
| | - Marcel Scheideler
- Institute for Genomics and Bioinformatics and Christian Doppler Laboratory for Genomics and Bioinformatics, Graz University of TechnologyPetersgasse 14, 8010 Graz, Austria
| | - Regina Grillari-Voglauer
- Aging and Immortalization Research, Department of Biotechnology, University of Natural Resources and Applied Life SciencesVienna, Austria, Muthgasse 18, A-1190 Vienna
| | - Beatrix Grubeck-Loebenstein
- Departments of Immunology, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Pidder Jansen-Dürr
- Molecular and Cell Biology, Institute for Biomedical Aging Research, Austrian Academy of SciencesRennweg 10, 6020 Innsbruck, Austria (IBA)
| | - Johannes Grillari
- Aging and Immortalization Research, Department of Biotechnology, University of Natural Resources and Applied Life SciencesVienna, Austria, Muthgasse 18, A-1190 Vienna
| |
Collapse
|
28
|
Ralph SJ, Rodríguez-Enríquez S, Neuzil J, Saavedra E, Moreno-Sánchez R. The causes of cancer revisited: "mitochondrial malignancy" and ROS-induced oncogenic transformation - why mitochondria are targets for cancer therapy. Mol Aspects Med 2010; 31:145-70. [PMID: 20206201 DOI: 10.1016/j.mam.2010.02.008] [Citation(s) in RCA: 216] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Accepted: 02/19/2010] [Indexed: 12/17/2022]
Abstract
The role of oncoproteins and tumor suppressor proteins in promoting the malignant transformation of mammalian cells by affecting properties such as proliferative signalling, cell cycle regulation and altered adhesion is well established. Chemicals, viruses and radiation are also generally accepted as agents that commonly induce mutations in the genes encoding these cancer-causing proteins, thereby giving rise to cancer. However, more recent evidence indicates the importance of two additional key factors imposed on proliferating cells that are involved in transformation to malignancy and these are hypoxia and/or stressful conditions of nutrient deprivation (e.g. lack of glucose). These two additional triggers can initiate and promote the process of malignant transformation when a low percentage of cells overcome and escape cellular senescence. It is becoming apparent that hypoxia causes the progressive elevation in mitochondrial ROS production (chronic ROS) which over time leads to stabilization of cells via increased HIF-2alpha expression, enabling cells to survive with sustained levels of elevated ROS. In cells under hypoxia and/or low glucose, DNA mismatch repair processes are repressed by HIF-2alpha and they continually accumulate mitochondrial ROS-induced oxidative DNA damage and increasing numbers of mutations driving the malignant transformation process. Recent evidence also indicates that the resulting mutated cancer-causing proteins feedback to amplify the process by directly affecting mitochondrial function in combinatorial ways that intersect to play a major role in promoting a vicious spiral of malignant cell transformation. Consequently, many malignant processes involve periods of increased mitochondrial ROS production when a few cells survive the more common process of oxidative damage induced cell senescence and death. The few cells escaping elimination emerge with oncogenic mutations and survive to become immortalized tumors. This review focuses on evidence highlighting the role of mitochondria as drivers of elevated ROS production during malignant transformation and hence, their potential as targets for cancer therapy. The review is organized into five main sections concerning different aspects of "mitochondrial malignancy". The first concerns the functions of mitochondrial ROS and its importance as a pacesetter for cell growth versus senescence and death. The second considers the available evidence that cellular stress in the form of hypoxic and/or hypoglycaemic conditions represent two of the major triggering events for cancer and how oncoproteins reinforce this process by altering gene expression to bring about a common set of changes in mitochondrial function and activity in cancer cells. The third section presents evidence that oncoproteins and tumor suppressor proteins physically localize to the mitochondria in cancer cells where they directly regulate malignant mitochondrial programs, including apoptosis. The fourth section covers common mutational changes in the mitochondrial genome as they relate to malignancy and the relationship to the other three areas. The last section concerns the relevance of these findings, their importance and significance for novel targeted approaches to anti-cancer therapy and selective triggering in cancer cells of the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Stephen J Ralph
- Genomic Research Centre, Griffith Institute of Health and Medical Research, School of Medical Science, Griffith University, Parklands Avenue, Southport, 4222 Qld, Australia.
| | | | | | | | | |
Collapse
|
29
|
Perumalsamy A, Fernandes R, Lai I, Detmar J, Varmuza S, Casper RF, Jurisicova A. Developmental consequences of alternative Bcl-x splicing during preimplantation embryo development. FEBS J 2010; 277:1219-33. [PMID: 20136652 DOI: 10.1111/j.1742-4658.2010.07554.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Elevated cell death in human preimplantation embryos is one of the cellular events compromising pregnancy rates after assisted reproductive technology treatments. We therefore explored the molecular pathways regulating cell death at the blastocyst stage in human embryos cultured in vitro. Owing to limited availability of human embryos, these pathways were further characterized in mouse blastocysts. Gene expression studies revealed a positive correlation between the cell death index and the expression of Bcl-x transcript. Cell death activation in human blastocysts was accompanied by changes in Bcl-x splicing, favoring production of Bcl-xS, an activator of cell death. Expression of Bcl-xS was detected in a subset of human blastocysts that show particular clustering in dying and/or dead cells. Altering the Bcl-xL/Bcl-xS ratio in mouse embryos, in antisense experiments, confirmed that upregulation of Bcl-xS, with concomitant downregulation of Bcl-xL, compromised developmental potential and committed a subset of cells to undergoing cell death. This was accompanied by increased accumulation of reactive oxygen species levels without any impact on mtDNA content. In addition, altered Bcl-x splicing in favor of Bcl-xS was stimulated by culture in HTF medium or by addition of excessive glucose, leading to compromised embryo development. Thus, we conclude that inappropriate culture conditions affect Bcl-x isoform expression, contributing to compromised preimplantation embryo development.
Collapse
|
30
|
Passos JF, Simillion C, Hallinan J, Wipat A, von Zglinicki T. Cellular senescence: unravelling complexity. AGE (DORDRECHT, NETHERLANDS) 2009; 31:353-363. [PMID: 19618294 PMCID: PMC2813046 DOI: 10.1007/s11357-009-9108-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 06/19/2009] [Indexed: 05/28/2023]
Abstract
Cellular senescence might be a tumour suppressing mechanism as well as a contributor to age-related loss of tissue function. It has been characterised classically as the result of the loss of DNA sequences called telomeres at the end of chromosomes. However, recent studies have revealed that senescence is in fact an intricate process, involving the sequential activation of multiple cellular processes, which have proven necessary for the establishment and maintenance of the phenotype. Here, we review some of these processes, namely, the role of mitochondrial function and reactive oxygen species, senescence-associated secreted proteins and chromatin remodelling. Finally, we illustrate the use of systems biology to address the mechanistic, functional and biochemical complexity of senescence.
Collapse
Affiliation(s)
- João F Passos
- Ageing Biology Laboratories and Centre for Integrated Systems Biology of Ageing and Nutrition (CISBAN), Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK.
| | | | | | | | | |
Collapse
|
31
|
Lee SLO, Hong SW, Shin JS, Kim JS, Ko SG, Hong NJ, Kim DJ, Lee WJ, Jin DH, Lee MS. p34SEI-1 inhibits doxorubicin-induced senescence through a pathway mediated by protein kinase C-delta and c-Jun-NH2-kinase 1 activation in human breast cancer MCF7 cells. Mol Cancer Res 2009; 7:1845-53. [PMID: 19903772 DOI: 10.1158/1541-7786.mcr-09-0086] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, we describe a novel function of the p34(SEI-1) protein, which is both an oncogenic protein and a positive regulator of the cell cycle. The p34(SEI-1) protein was found to inhibit doxorubicin-induced senescence. We investigated the molecular mechanisms of the inhibitory effect of p34(SEI-1) on senescence. First, we found that the activation of protein kinase C-delta (PKC-delta), which is cleaved into a 38 kDa active form from a 78 kDa pro-form, induced after doxorubicin treatment, was inhibited by p34(SEI-1). Furthermore, p34(SEI-1) induced the ubiquitination of PKC-delta. Yet, there is no interaction between p34(SEI-1) and PKC-delta. We also found that the phosphorylation of c-Jun-NH(2)-kinase 1 (JNK1) induced after doxorubicin treatment was suppressed by p34(SEI-1), but not in JNK2. Consistently, pharmacologic or genetic inactivation of either PKC-delta or JNK1 was found to inhibit doxorubicin-induced senescence. In addition, the genetic inactivation of PKC-delta by PKC-delta small interfering RNA resulted in an inhibition of JNK1 activation, but PKC-delta expression was not inactivated by JNK1 small interfering RNA, implying that the activation of JNK1 could be dependently induced by PKC-delta. Therefore, p34(SEI-1) inhibits senescence by inducing PKC-delta ubiquitination and preventing PKC-delta-dependent phosphorylation of JNK1.
Collapse
Affiliation(s)
- Sae Lo Oom Lee
- Research Center for Women's Diseases, Division of Biological Sciences, Sookmyung Women's University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Muller M. Cellular senescence: molecular mechanisms, in vivo significance, and redox considerations. Antioxid Redox Signal 2009; 11:59-98. [PMID: 18976161 DOI: 10.1089/ars.2008.2104] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cellular senescence is recognized as a critical cellular response to prolonged rounds of replication and environmental stresses. Its defining characteristics are arrested cell-cycle progression and the development of aberrant gene expression with proinflammatory behavior. Whereas the mechanistic events associated with senescence are generally well understood at the molecular level, the impact of senescence in vivo remains to be fully determined. In addition to the role of senescence as an antitumor mechanism, this review examines cellular senescence as a factor in organismal aging and age-related diseases, with particular emphasis on aberrant gene expression and abnormal paracrine signaling. Senescence as an emerging factor in tissue remodeling, wound repair, and infection is considered. In addition, the role of oxidative stress as a major mediator of senescence and the role of NAD(P)H oxidases and changes to intracellular GSH/GSSG status are reviewed. Recent findings indicate that senescence and the behavior of senescent cells are amenable to therapeutic intervention. As the in vivo significance of senescence becomes clearer, the challenge will be to modulate the adverse effects of senescence without increasing the risks of other diseases, such as cancer. The uncoupled relation between cell-cycle arrest and the senescent phenotype suggests that this is an achievable outcome.
Collapse
Affiliation(s)
- Michael Muller
- Centre for Education and Research on Ageing, ANZAC Research Institute, University of Sydney, Concord RG Hospital, Concord, Sydney, Australia.
| |
Collapse
|
33
|
Kusumi A, Kusumi T, Miura J, Tateishi T. Passage-affected competitive regulation of osteoprotegerin synthesis and the receptor activator of nuclear factor-kappaB ligand mRNA expression in normal human osteoblasts stimulated by the application of cyclic tensile strain. J Bone Miner Metab 2009; 27:653-62. [PMID: 19449178 DOI: 10.1007/s00774-009-0085-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2008] [Accepted: 03/25/2009] [Indexed: 11/24/2022]
Abstract
Mechanical stress application is a unique method for bone studies. We have reported regulation via the p38 mitogen-activated protein kinase (MAPK) pathway in osteoblasts under application of cyclic tensile strain (CTS), among many reports on the extracellular signal-regulated kinase (ERK) 1/2 pathway during mechanical stress, and questions remain as to the differences between our findings and those of others regarding types of MAPK activation. In the present study, osteoblasts were used after the third passage and stimulated by the application of 7%, 0.25 Hz CTS for 3 days, 4 h/day. CTS-induced osteoprotegerin (OPG) synthesis in osteoblasts increased at the third passage and decreased at the fifth passage, whereas CTS-induced receptor activator of nuclear factor-kappaB ligand (RANKL) mRNA expression decreased in osteoblasts at the third passage and increased at the fifth passage. Increases in CTS-induced osteopontin (OPN) synthesis, cyclooxygenase-2 (Cox-2) mRNA expression, and nitric oxide (NO) production by osteoblasts did not change at the third and fifth passages. Furthermore, p38 MAPK at the third passage and ERK1/2 at the fifth passage were found to be competitively activated in osteoblasts by the application of CTS. Based on these results, osteoblasts were shown to be affected by the number of passages. It was suggested that the examination of passage-affected characteristics of osteoblasts might not only be pertinent to the analysis of cellular senescence and in vivo models of bone remodelling with aging but could also be useful in the development of bone tissue engineering.
Collapse
Affiliation(s)
- Akinori Kusumi
- Department of Clinical Pharmacology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan.
| | | | | | | |
Collapse
|
34
|
Pekovic V, Hutchison CJ. Adult stem cell maintenance and tissue regeneration in the ageing context: the role for A-type lamins as intrinsic modulators of ageing in adult stem cells and their niches. J Anat 2008; 213:5-25. [PMID: 18638067 DOI: 10.1111/j.1469-7580.2008.00928.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adult stem cells have been identified in most mammalian tissues of the adult body and are known to support the continuous repair and regeneration of tissues. A generalized decline in tissue regenerative responses associated with age is believed to result from a depletion and/or a loss of function of adult stem cells, which itself may be a driving cause of many age-related disease pathologies. Here we review the striking similarities between tissue phenotypes seen in many degenerative conditions associated with old age and those reported in age-related nuclear envelope disorders caused by mutations in the LMNA gene. The concept is beginning to emerge that nuclear filament proteins, A-type lamins, may act as signalling receptors in the nucleus required for receiving and/or transducing upstream cytosolic signals in a number of pathways central to adult stem cell maintenance as well as adaptive responses to stress. We propose that during ageing and in diseases caused by lamin A mutations, dysfunction of the A-type lamin stress-resistant signalling network in adult stem cells, their progenitors and/or stem cell niches leads to a loss of protection against growth-related stress. This in turn triggers an inappropriate activation or a complete failure of self-renewal pathways with the consequent initiation of stress-induced senescence. As such, A-type lamins should be regarded as intrinsic modulators of ageing within adult stem cells and their niches that are essential for survival to old age.
Collapse
Affiliation(s)
- Vanja Pekovic
- School of Biological and Biomedical Science, Integrated Cell Biology Laboratories, Durham University, South Road, Durham DH1 3LE, UK.
| | | |
Collapse
|
35
|
SIRT1 overexpression antagonizes cellular senescence with activated ERK/S6k1 signaling in human diploid fibroblasts. PLoS One 2008; 3:e1710. [PMID: 18320031 PMCID: PMC2249701 DOI: 10.1371/journal.pone.0001710] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Accepted: 01/23/2008] [Indexed: 12/28/2022] Open
Abstract
Sir2, a NAD-dependent deacetylase, modulates lifespan in yeasts, worms and flies. The SIRT1, mammalian homologue of Sir2, regulates signaling for favoring survival in stress. But whether SIRT1 has the function to influence cell viability and senescence under non-stressed conditions in human diploid fibroblasts is far from unknown. Our data showed that enforced SIRT1 expression promoted cell proliferation and antagonized cellular senescence with the characteristic features of delayed Senescence-Associated beta-galactosidase (SA-beta-gal) staining, reduced Senescence-Associated Heterochromatic Foci (SAHF) formation and G1 phase arrest, increased cell growth rate and extended cellular lifespan in human fibroblasts, while dominant-negative SIRT1 allele (H363Y) did not significantly affect cell growth and senescence but displayed a bit decreased lifespan. Western blot results showed that SIRT1 reduced the expression of p16(INK4A) and promoted phosphorylation of Rb. Our data also exposed that overexpression of SIRT1 was accompanied by enhanced activation of ERK and S6K1 signaling. These effects were mimicked in both WI38 cells and 2BS cells by concentration-dependent resveratrol, a SIRT1 activator. It was noted that treatment of SIRT1-.transfected cells with Rapamycin, a mTOR inhibitor, reduced the phosphorylation of S6K1 and the expression of Id1, implying that SIRT1-induced phosphorylation of S6K1 may be partly for the decreased expression of p16(INK4A) and promoted phosphorylation of Rb in 2BS. It was also observed that the expression of SIRT1 and phosphorylation of ERK and S6K1 was declined in senescent 2BS. These findings suggested that SIRT1-promoted cell proliferation and antagonized cellular senescence in human diploid fibroblasts may be, in part, via the activation of ERK/ S6K1 signaling.
Collapse
|
36
|
Passos JF, Saretzki G, von Zglinicki T. DNA damage in telomeres and mitochondria during cellular senescence: is there a connection? Nucleic Acids Res 2007; 35:7505-13. [PMID: 17986462 PMCID: PMC2190715 DOI: 10.1093/nar/gkm893] [Citation(s) in RCA: 233] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cellular senescence is the ultimate and irreversible loss of replicative capacity occurring in primary somatic cell culture. It is triggered as a stereotypic response to unrepaired nuclear DNA damage or to uncapped telomeres. In addition to a direct role of nuclear DNA double-strand breaks as inducer of a DNA damage response, two more subtle types of DNA damage induced by physiological levels of reactive oxygen species (ROS) can have a significant impact on cellular senescence: Firstly, it has been established that telomere shortening, which is the major contributor to telomere uncapping, is stress dependent and largely caused by a telomere-specific DNA single-strand break repair inefficiency. Secondly, mitochondrial DNA (mtDNA) damage is closely interrelated with mitochondrial ROS production, and this might also play a causal role for cellular senescence. Improvement of mitochondrial function results in less telomeric damage and slower telomere shortening, while telomere-dependent growth arrest is associated with increased mitochondrial dysfunction. Moreover, telomerase, the enzyme complex that is known to re-elongate shortened telomeres, also appears to have functions independent of telomeres that protect against oxidative stress. Together, these data suggest a self-amplifying cycle between mitochondrial and telomeric DNA damage during cellular senescence.
Collapse
Affiliation(s)
- João F Passos
- Henry Wellcome Laboratory for Biogerontology Research, Institute for Ageing and Health, University of Newcastle, Newcastle upon Tyne NE4 6BE, UK
| | | | | |
Collapse
|
37
|
Davis T, Wyllie FS, Rokicki MJ, Bagley MC, Kipling D. The role of cellular senescence in Werner syndrome: toward therapeutic intervention in human premature aging. Ann N Y Acad Sci 2007; 1100:455-69. [PMID: 17460211 DOI: 10.1196/annals.1395.051] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Werner syndrome (WS) is a premature aging disorder used as a model of normal human aging. WS individuals have several characteristics of normal aging, such as cataracts, hair graying, and skin aging, but manifest these at an early age. Additionally, WS individuals have high levels of inflammatory diseases, such as atherosclerosis and type 2 diabetes. The in vivo aging in WS is associated with accelerated aging of fibroblasts in culture. The cause of the accelerated senescence is not understood, but may be due to the genomic instability that is a hallmark of WS. Genome instability results in activation of stress kinases, such as p38, and the p38-specific inhibitor SB203580, prevents the accelerated senescence seen in WS fibroblasts. However, oxidative damage plays a role, as low oxygen conditions and antioxidant treatment revert some of the accelerated senescence phenotype. The effects of oxidative stress appear to be suppressible by SB203580; however, it does not appear to be transduced by p38. As SB203580 is known to inhibit other kinases in addition to p38, this suggests that more than one kinase pathway is involved. The recent development of p38 inhibitors with different binding properties, specificities, and oral bioavailability, and of new potent and selective inhibitors of JNK and MK2, will make it possible to dissect the roles of various kinase pathways in the accelerated senescence of WS cells. If this accelerated senescence is reflective of WS aging in vivo, these kinase inhibitors may well form the basis of antiaging therapies for individuals with WS.
Collapse
Affiliation(s)
- Terence Davis
- Department of Pathology, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | | | | | | | | |
Collapse
|
38
|
Favetta LA, St John EJ, King WA, Betts DH. High levels of p66shc and intracellular ROS in permanently arrested early embryos. Free Radic Biol Med 2007; 42:1201-10. [PMID: 17382201 DOI: 10.1016/j.freeradbiomed.2007.01.018] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 01/09/2007] [Accepted: 01/09/2007] [Indexed: 11/17/2022]
Abstract
A high incidence of permanent embryo arrest occurs during the first week of in vitro development. We hypothesize that this developmental arrest event is regulated by the stress adaptor protein p66shc, a genetic determinant of life span in mammals, which regulates ROS metabolism, apoptosis, and cellular senescence. The aim of this study was to assess the relationship between intracellular oxidative stress levels with the incidence of embryo arrest and the expression of senescent-associated genes in embryos produced under different oxygen tensions. Embryos cultured under 20% oxygen conditions showed approximately 10-fold increase in oxidative stress, 2-fold increase in the percentage of 2- to 4-cell arrest, and significantly lower developmental capabilities compared to embryos cultured under a 5% oxygen environment. Quantification by real-time PCR and by semiquantitative immunofluorescence showed significantly higher p66shc mRNA and protein levels, respectively, in embryos cultured in 20% versus those cultured in 5% oxygen atmosphere. No significant changes in p53 mRNA and protein levels were detected among embryos derived from both oxygen tensions. Taken together, these results demonstrate that p66shc, but not p53, is significantly more abundant in an embryo population that exhibits higher frequencies of embryo arrest and quantities of intracellular ROS. These results further substantiate that p66shc and oxidative stress are associated with a p53-independent embryonic arrest event for in vitro-produced embryos.
Collapse
Affiliation(s)
- Laura A Favetta
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Canada
| | | | | | | |
Collapse
|
39
|
Han YH, Kwon JH, Yu DY, Moon EY. Inhibitory effect of peroxiredoxin II (Prx II) on Ras-ERK-NFkappaB pathway in mouse embryonic fibroblast (MEF) senescence. Free Radic Res 2007; 40:1182-9. [PMID: 17050172 DOI: 10.1080/10715760600868552] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Intracellular reactive oxygen species (ROS) were attenuated by the expression of peroxiredoxin II (Prx II). Cellular senescence as judged by senescence-associated (SA)-beta-galactosidase (Gal) positive cell formation was increased in Prx II-deficient mouse embryonic fibroblast (MEF). Ras expression was increased following passages. The level of Ras expression was higher in Prx II-/- MEF than wild type MEF. ERK activity was also augmented by the deletion of Prx II. SA-beta-Gal-positive cell formation was reduced by PD98059, ERK inhibitor. Activated nuclear transcription factor, nuclear factor-kappaB (NFkappaB) by the deletion of Prx II was inhibited by the treatment with PD98059. In contrast, no changes in SA-beta-Gal-positive cell formation were detected by NFkappaB inhibitor, N-alpha-tosyl-L-phenylalanyl chloromethyl ketone (TPCK). Collectively, results suggest that Prx II deletion activate Ras-ERK-NFkappaB pathways and cellular senescence in Prx II-/- MEF cells was mediated by ERK activation but not by NFkappaB activation.
Collapse
Affiliation(s)
- Ying-Hao Han
- Department of Human Genomics, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Taejeon, 305-806, South Korea
| | | | | | | |
Collapse
|
40
|
Passos JF, Von Zglinicki T. Oxygen free radicals in cell senescence: are they signal transducers? Free Radic Res 2007; 40:1277-83. [PMID: 17090417 DOI: 10.1080/10715760600917151] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxygen free radicals have a major impact on senescence of primary human cells. In replicative senescence, which is induced by uncapping of telomeres, the rate of telomere shortening is largely determined by telomere-specific accumulation of DNA damage induced by reactive oxygen species (ROS). More intense ROS-generating stressors can induce premature senescence via generation of telomere-independent DNA damage. Interestingly, ROS levels were also elevated when premature senescence was triggered by pathways downstream or independent of DNA damage. This has led to the suggestion that ROS generation could be a specific component of the signalling pathways inducing senescence. However, the available data are compatible with the concept that senescence is triggered as a DNA damage response. ROS appear to be involved as inducers of DNA damage rather than as specific signalling molecules. The upregulation of ROS production often seen in premature senescence might be related to retrograde response initiated by mitochondria.
Collapse
Affiliation(s)
- João F Passos
- Henry Wellcome Laboratory for Biogerontology Research, Institute for Ageing and Health, Newcastle upon Tyne, UK
| | | |
Collapse
|
41
|
Abstract
Cellular senescence, the ultimate and irreversible loss of replicative capacity of cells in primary culture, has been a popular model for studying the aging process. However, the replicative life span of human fibroblasts is heterogeneous even in clonal populations, with the fraction of senescent cells increasing at each population doubling, rather than all cells entering senescence simultaneously. Thus, the study of individual cells in a mass culture is of extreme importance to the understanding of replicative senescence. Cell sorting is a method that allows physical separation of cells with different characteristics when measured by flow cytometry. Here, we describe various methods by which cells that reach senescence early can be physically sorted out of a bulk of growing cells, and discuss how different methods can affect the posterior analysis of the sorted populations.
Collapse
Affiliation(s)
- João F Passos
- Henry Wellcome Laboratory for Biogerontology Research Newcastle University, Newcastle, UK
| | | |
Collapse
|
42
|
Passos JF, von Zglinicki T, Kirkwood TBL. Mitochondria and ageing: winning and losing in the numbers game. Bioessays 2007; 29:908-17. [PMID: 17688237 DOI: 10.1002/bies.20634] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mitochondrial dysfunction has long been considered a key mechanism in the ageing process but surprisingly little attention has been paid to the impact of mitochondrial number or density within cells. Recent reports suggest a positive association between mitochondrial density, energy homeostasis and longevity. However, mitochondrial number also determines the number of sites generating reactive oxygen species (ROS) and we suggest that the links between mitochondrial density and ageing are more complex, potentially acting in both directions. The idea that increased density, especially when combined with mitochondrial dysfunction, might accelerate ageing is supported by a negative correlation between mitochondrial density and maximum longevity in an interspecies comparison in mammals, and by evidence for an intimate interconnection between cellular ROS levels, mitochondrial density and cellular ageing. Recent data suggest that retrograde response, which activates mitochondrial biogenesis, accompanies cellular ageing processes. We hypothesise that increased mitochondrial biogenesis, and possibly also impaired degradation and segregation of mitochondria, if occurring as adaptation to pre-existing mitochondrial dysfunction, might aggravate ROS production and thus actively contribute to ageing.
Collapse
Affiliation(s)
- João F Passos
- Centre for Integrated Systems Biology of Ageing and Nutrition, Henry Wellcome Laboratory for Biogerontology Research, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | | | | |
Collapse
|
43
|
Abstract
We found that an ongoing application of nicotinamide to normal human fibroblasts not only attenuated expression of the aging phenotype but also increased their replicative lifespan, causing a greater than 1.6-fold increase in the number of population doublings. Although nicotinamide by itself does not act as an antioxidant, the cells cultured in the presence of nicotinamide exhibited reduced levels of reactive oxygen species (ROS) and oxidative damage products associated with cellular senescence, and a decelerated telomere shortening rate without a detectable increase in telomerase activity. Furthermore, in the treated cells growing beyond the original Hayflick limit, the levels of p53, p21WAF1, and phospho-Rb proteins were similar to those in actively proliferating cells. The nicotinamide treatment caused a decrease in ATP levels, which was stably maintained until the delayed senescence point. Nicotinamide-treated cells also maintained high mitochondrial membrane potential but a lower respiration rate and superoxide anion level. Taken together, in contrast to its demonstrated pro-aging effect in yeast, nicotinamide extends the lifespan of human fibroblasts, possibly through reduction in mitochondrial activity and ROS production.
Collapse
Affiliation(s)
- Hyun Tae Kang
- Department of Life Science, University of Seoul, Dongdaemungu, Jeonnongdong, Seoul, Korea
| | | | | |
Collapse
|
44
|
Passos JF, von Zglinicki T. Mitochondria, telomeres and cell senescence. Exp Gerontol 2006; 40:466-72. [PMID: 15963673 DOI: 10.1016/j.exger.2005.04.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2005] [Revised: 04/14/2005] [Accepted: 04/14/2005] [Indexed: 01/21/2023]
Abstract
The accumulation of oxidative damage is one of the most widely accepted causes of ageing. Mitochondrial dysfunction, in particular damage to the mitochondrial DNA has been hypothesised, more than thirty years ago, as responsible for increased production of reactive oxygen species (ROS) and, thus, as one possible causal factor for ageing. There is now a wealth of data that supports this hypothesis, which is mostly derived from models considering the ageing of post-mitotic or slowly dividing cells in vivo. One major cellular model of ageing, however, is replicative senescence, the irreversible loss of division potential of somatic cells after a more or less constant number of cell divisions. Not much data exists concerning the role of mitochondria in this model. Here, we review evidence supporting an involvement of mitochondria in replicative senescence and a possible link to telomere shortening.
Collapse
Affiliation(s)
- João F Passos
- Henry Wellcome Laboratory for Biogerontology Research, Newcastle University, Newcastle NE4 6BE, UK
| | | |
Collapse
|
45
|
Kregel KC, Zhang HJ. An integrated view of oxidative stress in aging: basic mechanisms, functional effects, and pathological considerations. Am J Physiol Regul Integr Comp Physiol 2006; 292:R18-36. [PMID: 16917020 DOI: 10.1152/ajpregu.00327.2006] [Citation(s) in RCA: 534] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Aging is an inherently complex process that is manifested within an organism at genetic, molecular, cellular, organ, and system levels. Although the fundamental mechanisms are still poorly understood, a growing body of evidence points toward reactive oxygen species (ROS) as one of the primary determinants of aging. The "oxidative stress theory" holds that a progressive and irreversible accumulation of oxidative damage caused by ROS impacts on critical aspects of the aging process and contributes to impaired physiological function, increased incidence of disease, and a reduction in life span. While compelling correlative data have been generated to support the oxidative stress theory, a direct cause-and-effect relationship between the accumulation of oxidatively mediated damage and aging has not been strongly established. The goal of this minireview is to broadly describe mechanisms of in vivo ROS generation, examine the potential impact of ROS and oxidative damage on cellular function, and evaluate how these responses change with aging in physiologically relevant situations. In addition, the mounting genetic evidence that links oxidative stress to aging is discussed, as well as the potential challenges and benefits associated with the development of anti-aging interventions and therapies.
Collapse
Affiliation(s)
- Kevin C Kregel
- Department of Integrative Physiology and Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA.
| | | |
Collapse
|
46
|
Lenaz G, Baracca A, Fato R, Genova ML, Solaini G. New insights into structure and function of mitochondria and their role in aging and disease. Antioxid Redox Signal 2006; 8:417-37. [PMID: 16677088 DOI: 10.1089/ars.2006.8.417] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This review covers some novel findings on mitochondrial biochemistry and discusses diseases due to mitochondrial DNA mutations as a model of the changes occurring during physiological aging. The random collision model of organization of the mitochondrial respiratory chain has been recently challenged on the basis of findings of supramolecular organization of respiratory chain complexes. The source of superoxide in Complex I is discussed on the basis of laboratory experiments using a series of specific inhibitors and is presumably iron sulfur center N2. Maternally inherited diseases due to mutations of structural genes in mitochondrial DNA are surveyed as a model of alterations mimicking those occurring during normal aging. The molecular defects in senescence are surveyed on the basis of the "Mitochondrial Theory of Aging", establishing mitochondrial DNA somatic mutations, caused by accumulation of oxygen radical damage, to be at the basis of cellular senescence. Mitochondrial production of reactive oxygen species increases with aging and mitochondrial DNA mutations and deletions accumulate and may be responsible for oxidative phosphorylation defects. Evidence is presented favoring the mitochondrial theory, with primary mitochondrial alterations, although the problem is made more complex by changes in the cross-talk between nuclear and mitochondrial DNA.
Collapse
Affiliation(s)
- Giorgio Lenaz
- Dipartimento di Biochimica, Università di Bologna, Bologna, Italy.
| | | | | | | | | |
Collapse
|
47
|
Brégégère F, Milner Y, Friguet B. The ubiquitin-proteasome system at the crossroads of stress-response and ageing pathways: a handle for skin care? Ageing Res Rev 2006; 5:60-90. [PMID: 16330259 DOI: 10.1016/j.arr.2005.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2005] [Revised: 09/22/2005] [Accepted: 09/26/2005] [Indexed: 02/07/2023]
Abstract
The regulation of gene expression at the transcriptional level has been considered for long as the main mechanism of cellular adaptive responses. Since the turn of the century, however, it is becoming clear that higher organisms developed a complex, sensitive and maybe equally important network of regulatory pathways, relying largely on protein interactions, post-translational modifications and proteolysis. Here we review the involvement of the ubiquitin-proteasome pathway of protein degradation at different levels of cellular life in relation with ageing, and with a special focus on skin. It comes out that the ubiquitin system plays a major role in signal transduction associated with stress and ageing, in skin in particular through the control of retinoid and NF-kappaB pathways. The understanding of specific proteolytic targeting by E3 ubiquitin-ligases paves the way for a new generation of active molecules that may control particular steps of normal and pathological ageing.
Collapse
Affiliation(s)
- François Brégégère
- Laboratoire de Biologie et Biochimie Cellulaire du Vieillissement, Université Denis Diderot-Paris 7, C.C.7128, 2 Place Jussieu, 75251 Paris Cédex 05, France.
| | | | | |
Collapse
|
48
|
Herbig U, Sedivy JM. Regulation of growth arrest in senescence: Telomere damage is not the end of the story. Mech Ageing Dev 2006; 127:16-24. [PMID: 16229875 DOI: 10.1016/j.mad.2005.09.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Revised: 08/30/2005] [Accepted: 09/02/2005] [Indexed: 12/15/2022]
Abstract
After a limited number of divisions, most eukaryotic cells grown in culture will undergo a terminal growth arrest called cellular senescence. This growth arrest is thought to be a consequence of progressive telomere shortening that occurs due to incomplete DNA replication of the chromosome ends. In addition, cellular senescence can also be induced by a number of environmental stresses and signaling imbalances which are independent of telomere shortening. The cyclin dependent kinase inhibitors p21 and p16(INK4a) have been shown to execute and maintain the cell cycle arrest in senescence but the nature of the signals that cause upregulation of these inhibitors in senescent cells are only now starting to be discovered. Here we will review the current literature that leads us to propose a model how independent signals activate distinct signaling pathways to regulate p21 and p16(INK4a) levels in senescent cells.
Collapse
Affiliation(s)
- Utz Herbig
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 70 Ship Street, Box G-E438, Providence, RI 02903, USA
| | | |
Collapse
|
49
|
Noble M, Mayer-Pröschel M, Pröschel C. Redox regulation of precursor cell function: insights and paradoxes. Antioxid Redox Signal 2005; 7:1456-67. [PMID: 16356108 DOI: 10.1089/ars.2005.7.1456] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Studies on oligodendrocytes, the myelin-forming cells of the central nervous system, and on the progenitor cells from which they are derived, have provided several novel insights into the role of intracellular redox state in cell function. This review discusses our findings indicating that intracellular redox state is utilized by the organism as a means of regulating the balance between progenitor cell division and differentiation. This regulation is achieved in part through cell-intrinsic differences that modify the response of cells to extracellular signaling molecules, such that cells that are slightly more reduced are more responsive to inducers of cell survival and division and less responsive to inducers of differentiation or cell death. Cells that are slightly more oxidized, in contrast, show a greater response to inducers of differentiation or cell death, but less response to inducers of proliferation or survival. Regulation is also achieved by the ability of exogenous signaling molecules to modify intracellular redox state in a highly predictable manner, such that signaling molecules that promote self-renewal make progenitor cells more reduced and those that promote differentiation make cells more oxidized. In both cases, the redox changes induced by exposure to exogenous signaling molecules are a necessary component of their mode of action. Paradoxically, the results obtained through studies on the oligodendrocyte lineage are precisely the opposite of what might be predicted from a large number of studies demonstrating the ability of reactive oxidative species to enhance the effects of signaling through receptor tyrosine kinase receptors and to promote cell proliferation. Taken in sum, available data demonstrate clearly the existence of two distinct programs of cellular responses to changes in oxidative status. In one of these, becoming even slightly more oxidized is sufficient to inhibit proliferation and induce differentiation. In the second program, similar changes enhance proliferation. It is not yet clear how cells can interpret putatively identical signals in such opposite manners, but it does already seem clear that resolving this paradox will provide insights of considerable relevance to the understanding of normal development, tissue repair, and tumorigenesis.
Collapse
Affiliation(s)
- Mark Noble
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
50
|
Abstract
Aging has often been viewed as a random process arising from the accumulation of both genetic and epigenetic changes. Increasingly, the notion that aging is a stochastic process is being supplanted by the concept that maximum lifespan of an organism is tightly regulated. This knowledge has led to a growing overlap between classical signal transduction paradigms and the biology of aging. We review certain specific examples where these seemingly disparate disciplines intersect. In particular, we review the concept that intracellular reactive oxygen species function as signalling molecules and that oxidants play a central role as mediators of cellular senescence.
Collapse
Affiliation(s)
- Renata Colavitti
- Cardiovascular Branch, NHLBI, NIH, Bethesda, Maryland 20892-1454, USA
| | | |
Collapse
|