101
|
Luo X, Zhou J, Wang Z, He Y, Yu L, Ma S, Wang S, Wang X, Yuan Y, Li D, Cui T, Ding Y. An inhibitor role of Nrf2 in the regulation of myocardial senescence and dysfunction after myocardial infarction. Life Sci 2020; 259:118199. [PMID: 32781064 DOI: 10.1016/j.lfs.2020.118199] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/23/2020] [Accepted: 07/31/2020] [Indexed: 01/10/2023]
Abstract
Cellular senescence, a process whereby cells enter a state of permanent growth arrest, appears to regulate cardiac pathological remodeling and dysfunction in response to various stresses including myocardial infarction (MI). However, the precise role as well as the underlying regulatory mechanism of cardiac cellular senescence in the ischemic heart disease remain to be further determined. Herein we report an inhibitory role of Nrf2, a key transcription factor of cellular defense, in regulating cardiac senescence in infarcted hearts as well as a therapeutic potential of targeting Nrf2-mediated suppression of cardiac senescence in the treatment of MI-induced cardiac dysfunction. MI was induced by left coronary artery ligation for 28 days in mice. Heart tissues from the infarct border zone were used for the analyses. The MI-induced cardiac dysfunction was associated with increased myocardial cell senescence, oxidative stress and apoptosis in adult wild type (WT) mice. In addition, a downregulated Nrf2 activity was associated with upregulated Keap1 levels and increased phosphorylation of JAK and FYN in the infarcted border zone heart tissues. Nrf2 Knockout (Nrf2-/-) enhanced the MI-induced myocardial, cardiac dysfunction and senescence. Qiliqiangxin (QLQX), a herbal medicine which could reverse the MI-induced suppression of Nrf2 activity, significantly inhibited the MI-induced cardiac senescence, apoptosis, and cardiac dysfunction in WT mice but not in Nrf2-/- mice. These results indicate that MI downregulates Nrf2 activity thus promoting oxidative stress to accelerate cellular senescence in the infarcted heart towards cardiac dysfunction and Nrf2 may be a drug target for suppressing the cellular senescence-associated pathologies in infarcted hearts.
Collapse
Affiliation(s)
- Xinxia Luo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Junyang Zhou
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Zhixiao Wang
- Cardiovascular Department, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yun He
- Ultrasonography Department, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Li Yu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Shinan Ma
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Shan Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Xiaoli Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yahong Yuan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Dongsheng Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29208, USA.
| | - Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| |
Collapse
|
102
|
Wang T, Jian Z, Baskys A, Yang J, Li J, Guo H, Hei Y, Xian P, He Z, Li Z, Li N, Long Q. MSC-derived exosomes protect against oxidative stress-induced skin injury via adaptive regulation of the NRF2 defense system. Biomaterials 2020; 257:120264. [PMID: 32791387 DOI: 10.1016/j.biomaterials.2020.120264] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/27/2020] [Accepted: 07/26/2020] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a major cause of skin injury induced by damaging stimuli such as UV radiation. Currently, owing to their immunomodulatory properties, mesenchymal stem cell-derived exosomes (MSC-Exo), as a nanotherapeutic agent, have attracted considerable attention. Here, we investigated the therapeutic effects of MSC-Exo on oxidative injury in H2O2-stimulated epidermal keratinocytes and UV-irradiated wild type and nuclear factor-erythroid 2-related factor-2 (Nrf2) knocked down cell and animal models. Our findings showed that MSC-Exo treatment reduced reactive oxygen species generation, DNA damage, aberrant calcium signaling, and mitochondrial changes in H2O2-stimulated keratinocytes or UV-irradiated mice skin. Exosome therapy also improved antioxidant capacities shown by increased ferric ion reducing antioxidant power and glutathione peroxidase or superoxide dismutase activities in oxidative stress-induced cell and skin injury. In addition, it alleviated cellular and histological responses to inflammation and oxidation in cell or animal models. Furthermore, the NRF2 signaling pathway was involved in the antioxidation activity of MSC-Exo, while Nrf2 knockdown attenuated the antioxidant capacities of MSC-Exo in vitro and in vivo, suggesting that these effects are partially mediated by the NRF2 signaling pathway. These results indicate that MSC-Exo can repair oxidative stress-induced skin injury via adaptive regulation of the NRF2 defense system. Thus, MSC-Exo may be used as a potential dermatological nanotherapeutic agent for treating oxidative stress-induced skin diseases or disorders.
Collapse
Affiliation(s)
- Tian Wang
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Zhe Jian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China
| | - Andrius Baskys
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Junle Yang
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Jianying Li
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Hua Guo
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Yue Hei
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China
| | - Panpan Xian
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Zhongzheng He
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Zhengyu Li
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Namiao Li
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China
| | - Qianfa Long
- Mini-invasive Neurosurgery and Translational Medical Center, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an, 710003, PR China.
| |
Collapse
|
103
|
Pharmacological activation of Nrf2 promotes wound healing. Eur J Pharmacol 2020; 886:173395. [PMID: 32710954 DOI: 10.1016/j.ejphar.2020.173395] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/09/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
Wound repair and regeneration is a complex orchestrated process, comprising several phases interconnecting various cellular events and triggering multiple intracellular molecular pathways in damaged cells and tissues. In several metabolic disorders including diabetes mellitus, delay in wound healing due to elevated levels of cellular stress poses a key challenge. Several therapeutic wound dressing materials and strategies including hyperbaric oxygen therapy and negative pressure wound therapy have been developed to accelerate repair and restore cellular homeostasis at the wound site. Further, tremendous progress has been made in identification of transcriptional regulators involved in the process of wound healing. Nuclear factor erythroid 2-related factor 2 (Nrf2), a redox sensitive transcription factor, is the key regulator of intracellular redox homeostasis which induces the expression of cytoprotective genes and increases the production of antioxidants that scavenge free radicals. Activators of Nrf2 have been reported to combat oxidative stress and enhance the process of wound healing in several pathophysiological conditions, including diabetes and its complications such as diabetic foot ulcer, and chronic kidney disease, and diabetic nephropathy. Several bioactive compounds have been reported to reduce cellular stress, and thus accelerate cell proliferation, neovascularization results in repairing damaged tissues by the activation of the transcription factor, Nrf2. This review is focused on the strategies for diabetic wound healing and the highlights the role of bioactive compounds that activate the Nrf2 signaling and revitalize the cellular and molecular mechanism in the chronic wound niche, regulate and restore redox homeostasis thereby promoting wound repair and regeneration.
Collapse
|
104
|
Di Domizio J, Belkhodja C, Chenuet P, Fries A, Murray T, Mondéjar PM, Demaria O, Conrad C, Homey B, Werner S, Speiser DE, Ryffel B, Gilliet M. The commensal skin microbiota triggers type I IFN-dependent innate repair responses in injured skin. Nat Immunol 2020; 21:1034-1045. [PMID: 32661363 DOI: 10.1038/s41590-020-0721-6] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 05/27/2020] [Indexed: 01/25/2023]
Abstract
Skin wounds heal by coordinated induction of inflammation and tissue repair, but the initiating events are poorly defined. Here we uncover a fundamental role of commensal skin microbiota in this process and show that it is mediated by the recruitment and the activation of type I interferon (IFN)-producing plasmacytoid DC (pDC). Commensal bacteria colonizing skin wounds trigger activation of neutrophils to express the chemokine CXCL10, which recruits pDC and acts as an antimicrobial protein to kill exposed microbiota, leading to the formation of CXCL10-bacterial DNA complexes. These complexes and not complexes with host-derived DNA activate pDC to produce type I IFNs, which accelerate wound closure by triggering skin inflammation and early T cell-independent wound repair responses, mediated by macrophages and fibroblasts that produce major growth factors required for healing. These findings identify a key function of commensal microbiota in driving a central innate wound healing response of the skin.
Collapse
Affiliation(s)
- Jeremy Di Domizio
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Cyrine Belkhodja
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Pauline Chenuet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orleans, France
| | - Anissa Fries
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Timothy Murray
- Department of Oncology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Paula Marcos Mondéjar
- Department of Oncology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Olivier Demaria
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Curdin Conrad
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Bernhard Homey
- Department of Dermatology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Sabine Werner
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Daniel E Speiser
- Department of Oncology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orleans, France.,Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, and South Africa Medical Research Council, Cape Town, South Africa
| | - Michel Gilliet
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
105
|
Xiao C, Xu C, He N, Liu Y, Wang Y, Zhang M, Ji K, Du L, Wang J, Wang Q, Liu Q. Atractylenolide II prevents radiation damage via MAPKp38/Nrf2 signaling pathway. Biochem Pharmacol 2020; 177:114007. [DOI: 10.1016/j.bcp.2020.114007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/21/2020] [Indexed: 01/15/2023]
|
106
|
Ben-Yehuda Greenwald M, Tacconi C, Jukic M, Joshi N, Hiebert P, Brinckmann J, Tenor H, Naef R, Werner S. A Dual-Acting Nitric Oxide Donor and Phosphodiesterase 5 Inhibitor Promotes Wound Healing in Normal Mice and Mice with Diabetes. J Invest Dermatol 2020; 141:415-426. [PMID: 32598925 DOI: 10.1016/j.jid.2020.05.111] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/12/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022]
Abstract
Chronic wounds affect a large percentage of the population worldwide and cause significant morbidity. Unfortunately, efficient compounds for the treatment of chronic wounds are yet not available. Endothelial dysfunction, which is at least in part a result of compromised nitric oxide production and concomitant reduction in cGMP levels, is a major pathologic feature of chronic wounds. Therefore, we designed and synthesized a compound with a unique dual-acting activity (TOP-N53), acting as a nitric oxide donor and phosphodiesterase 5 inhibitor, and applied it locally to full-thickness skin wounds in healthy and healing-impaired mice with diabetes. TOP-N53 promoted keratinocyte proliferation, angiogenesis, and collagen maturation in healthy mice without accelerating the wound inflammatory response or scar formation. Most importantly, it partially rescued the healing impairment of mice with genetically determined type II diabetes (db/db) by stimulating re-epithelialization and granulation tissue formation, including angiogenesis. In vitro studies with human and murine primary cells showed a positive effect of TOP-N53 on keratinocyte and fibroblast migration, keratinocyte proliferation, and endothelial cell migration and tube formation. These results demonstrate a remarkable healing-promoting activity of TOP-N53 by targeting the major resident cells in the wound tissue.
Collapse
Affiliation(s)
| | - Carlotta Tacconi
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Marko Jukic
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Natasha Joshi
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Paul Hiebert
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jürgen Brinckmann
- Department of Dermatology, University of Lübeck, Lübeck, Germany; Institute of Virology and Cell Biology, University of Lubeck, Lübeck, Germany
| | | | - Reto Naef
- Topadur Pharma AG, Schlieren, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
107
|
Dengjel J, Bruckner-Tuderman L, Nyström A. Skin proteomics - analysis of the extracellular matrix in health and disease. Expert Rev Proteomics 2020; 17:377-391. [PMID: 32552150 DOI: 10.1080/14789450.2020.1773261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The skin protects the human body from external insults and regulates water and temperature homeostasis. A highly developed extracellular matrix (ECM) supports the skin and instructs its cell functions. Reduced functionality of the ECM is often associated with skin diseases that cause physical impairment and also have implications on social interactions and quality of life of affected individuals. AREAS COVERED With a focus on the skin ECM we discuss how mass spectrometry (MS)-based proteomic approaches first contributed to establishing skin protein inventories and then facilitated elucidation of molecular functions and disease mechanisms. EXPERT OPINION MS-based proteomic approaches have significantly contributed to our understanding of skin pathophysiology, but also revealed the challenges in assessing the skin ECM. The numerous posttranslational modifications of ECM proteins, like glycosylation, crosslinking, oxidation, and proteolytic maturation in disease settings can be difficult to tackle and remain understudied. Increased ease of handling of LC-MS/MS systems and automated/streamlined data analysis pipelines together with the accompanying increased usage of LC-MS/MS approaches will ensure that in the coming years MS-based proteomic approaches will continue to play a vital part in skin disease research. They will facilitate the elucidation of molecular disease mechanisms and, ultimately, identification of new druggable targets.
Collapse
Affiliation(s)
- Jörn Dengjel
- Department of Biology, University of Fribourg , Fribourg, Switzerland
| | - Leena Bruckner-Tuderman
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg , Freiburg, University of Freiburg, Freiburg, Germany Germany
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg , Freiburg, University of Freiburg, Freiburg, Germany Germany
| |
Collapse
|
108
|
Abstract
PURPOSE OF REVIEW Fibroblasts, the major cell population in all connective tissues, are best known for their role in depositing and maintaining the extracellular matrix. Recently, numerous specialised functions have been discovered revealing unpredicted fibroblast heterogeneity. We will discuss this heterogeneity, from its origins in development to alterations in fibrotic disease conditions. RECENT FINDINGS Advances in lineage tracing and single-cell transcriptional profiling techniques have revealed impressive diversity amongst fibroblasts in a range of organ systems including the skin, lung, kidney and heart. However, there are major challenges in assimilating the findings and understanding their functional significance. Certain fibroblast subsets can make specific contributions to healthy tissue functioning and to fibrotic disease processes; thus, therapeutic manipulation of particular subsets could be clinically beneficial. Here we propose that four key variables determine a fibroblast's phenotype underpinning their enormous heterogeneity: tissue status, regional features, microenvironment and cell state. We review these in different organ systems, highlighting the importance of understanding the divergent fibroblast properties and underlying mechanisms in tissue fibrosis.
Collapse
Affiliation(s)
- Tanya J Shaw
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, New Hunt’s House, Guy’s Campus, King’s College London, London, SE1 1UL UK
| | - Emanuel Rognoni
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ UK
| |
Collapse
|
109
|
Muzumdar S, Koch M, Hiebert H, Bapst A, Gravina A, Bloch W, Beer HD, Werner S, Schäfer M. Genetic activation of Nrf2 reduces cutaneous symptoms in a murine model of Netherton syndrome. Dis Model Mech 2020; 13:dmm042648. [PMID: 32457102 PMCID: PMC7286291 DOI: 10.1242/dmm.042648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 03/16/2020] [Indexed: 01/08/2023] Open
Abstract
Netherton syndrome is a monogenic autosomal recessive disorder primarily characterized by the detachment of the uppermost layer of the epidermis, the stratum corneum It results from mutations in the SPINK5 gene, which codes for a kallikrein inhibitor. Uncontrolled kallikrein activity leads to premature desquamation, resulting in a severe epidermal barrier defect and subsequent life-threatening systemic infections and chronic cutaneous inflammation. Here, we show that genetic activation of the transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nfe2l2/Nrf2) in keratinocytes of Spink5 knockout mice, a model for Netherton syndrome, significantly alleviates their cutaneous phenotype. Nrf2 activation promoted attachment of the stratum corneum and concomitant epidermal barrier function, and reduced the expression of pro-inflammatory cytokines such as tumor necrosis factor α and thymic stromal lymphopoietin. Mechanistically, we show that Nrf2 activation induces overexpression of secretory leukocyte protease inhibitor (Slpi), a known inhibitor of kallikrein 7 and elastase 2, in mouse and human keratinocytes in vivo and in vitro, respectively. In the Spink5-deficient epidermis, the upregulation of Slpi is likely to promote stabilization of corneodesmosomes, thereby preventing premature desquamation. Our results suggest pharmacological NRF2 activation as a promising treatment modality for Netherton syndrome patients.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sukalp Muzumdar
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Michael Koch
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Hayley Hiebert
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Andreas Bapst
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Alessia Gravina
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, 50933 Cologne, Germany
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 3, 8091 Zurich, Switzerland
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Matthias Schäfer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
110
|
Shrishrimal S, Chatterjee A, Kosmacek EA, Davis PJ, McDonald JT, Oberley-Deegan RE. Manganese porphyrin, MnTE-2-PyP, treatment protects the prostate from radiation-induced fibrosis (RIF) by activating the NRF2 signaling pathway and enhancing SOD2 and sirtuin activity. Free Radic Biol Med 2020; 152:255-270. [PMID: 32222469 PMCID: PMC7276298 DOI: 10.1016/j.freeradbiomed.2020.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/20/2020] [Accepted: 03/21/2020] [Indexed: 12/13/2022]
Abstract
Radiation therapy is a frequently used treatment for prostate cancer patients. Manganese (III) meso-tetrakis (N-ethylpyridinium-2-yl) porphyrin (MnTE-2-PyP or T2E or BMX-010) and other similar manganese porphyrin compounds that scavenge superoxide molecules have been demonstrated to be effective radioprotectors and prevent the development of radiation-induced fibrosis (RIF). However, understanding the molecular pathway changes associated with these compounds remains limited for radioprotection. Recent RNA-sequencing data from our laboratory revealed that MnTE-2-PyP treatment activated the nuclear factor erythroid 2-related factor 2 (NRF2) signaling pathway. Therefore, we hypothesize that MnTE-2-PyP protects the prostate from RIF by activating the NRF2 signaling pathway. We identified that MnTE-2-PyP is a post-translational activator of NRF2 signaling in prostate fibroblast cells, which plays a major role in fibroblast activation and myofibroblast differentiation. The mechanism of NRF2 activation involves an increase in hydrogen peroxide and a corresponding decrease in kelch-like ECH-associated protein 1 (KEAP1) levels. Activation of NRF2 signaling leads to an increase in expression of NAD(P)H dehydrogenase [quinone] 1 (NQO1), nicotinamide adenine dinucleotide (NAD+) levels, sirtuin activity (nuclear and mitochondrial), and superoxide dismutase 2 (SOD2) expression/activity. Increase in mitochondrial sirtuin activity correlates with a decrease in SOD2 (K122) acetylation. This decrease in SOD2 K122 acetylation correlates with an increase in SOD2 activity and mitochondrial superoxide scavenging capacity. Further, in human primary prostate fibroblast cells, the NRF2 pathway plays a major role in the fibroblast to myofibroblast transformation, which is responsible for the fibrotic phenotype. In the context of radiation protection, MnTE-2-PyP fails to prevent fibroblast to myofibroblast transformation in the absence of NRF2 signaling. Collectively, our results indicate that the activation of the NRF2 signaling pathway by MnTE-2-PyP is at least a partial mechanism of radioprotection in prostate fibroblast cells.
Collapse
Affiliation(s)
- Shashank Shrishrimal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | | | - J Tyson McDonald
- Department of Physics, Hampton University, Hampton, VA, 23668, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
111
|
Joshi N, Pohlmeier L, Ben-Yehuda Greenwald M, Haertel E, Hiebert P, Kopf M, Werner S. Comprehensive characterization of myeloid cells during wound healing in healthy and healing-impaired diabetic mice. Eur J Immunol 2020; 50:1335-1349. [PMID: 32306381 PMCID: PMC7496577 DOI: 10.1002/eji.201948438] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/18/2020] [Accepted: 04/16/2020] [Indexed: 01/13/2023]
Abstract
Wound healing involves the concerted action of various lymphoid and in particular myeloid cell populations. To characterize and quantitate different types of myeloid cells and to obtain information on their kinetics during wound healing, we performed multiparametric flow cytometry analysis. In healthy mice, neutrophil numbers increased early after injury and returned to near basal levels after completion of healing. Macrophages, monocyte‐derived dendritic cells (DCs), and eosinophils were abundant throughout the healing phase, in particular in early wounds, and Langerhans cells increased after wounding and remained elevated after epithelial closure. Major differences in healing‐impaired diabetic mice were a much higher percentage of immune cells in late wounds, mainly as a result of neutrophil, macrophage, and monocyte persistence; reduced numbers and percentages of macrophages and monocyte‐derived DCs in early wounds; and of Langerhans cells, conventional DCs, and eosinophils throughout the healing process. Finally, unbiased cluster analysis (PhenoGraph) identified a large number of different clusters of myeloid cells in skin wounds. These results provide insight into myeloid cell diversity and dynamics during wound repair and highlight the abnormal inflammatory response associated with impaired healing.
Collapse
Affiliation(s)
- Natasha Joshi
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Lea Pohlmeier
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | - Eric Haertel
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Paul Hiebert
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
112
|
Mavrogonatou E, Pratsinis H, Kletsas D. The role of senescence in cancer development. Semin Cancer Biol 2020; 62:182-191. [DOI: 10.1016/j.semcancer.2019.06.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 02/07/2023]
|
113
|
Cangkrama M, Wietecha M, Mathis N, Okumura R, Ferrarese L, Al‐Nuaimi D, Antsiferova M, Dummer R, Innocenti M, Werner S. A paracrine activin A-mDia2 axis promotes squamous carcinogenesis via fibroblast reprogramming. EMBO Mol Med 2020; 12:e11466. [PMID: 32150356 PMCID: PMC7136968 DOI: 10.15252/emmm.201911466] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 12/18/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are key regulators of tumorigenesis and promising targets for next-generation therapies. We discovered that cancer cell-derived activin A reprograms fibroblasts into pro-tumorigenic CAFs. Mechanistically, this occurs via Smad2-mediated transcriptional regulation of the formin mDia2, which directly promotes filopodia formation and cell migration. mDia2 also induces expression of CAF marker genes through prevention of p53 nuclear accumulation, resulting in the production of a pro-tumorigenic matrisome and secretome. The translational relevance of this finding is reflected by activin A overexpression in tumor cells and of mDia2 in the stroma of skin cancer and other malignancies and the correlation of high activin A/mDia2 levels with poor patient survival. Blockade of this signaling axis using inhibitors of activin, activin receptors, or mDia2 suppressed cancer cell malignancy and squamous carcinogenesis in 3D organotypic cultures, ex vivo, and in vivo, providing a rationale for pharmacological inhibition of activin A-mDia2 signaling in stratified cancer patients.
Collapse
Affiliation(s)
- Michael Cangkrama
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Mateusz Wietecha
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Nicolas Mathis
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Rin Okumura
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Luca Ferrarese
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Dunja Al‐Nuaimi
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Maria Antsiferova
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
- Present address:
Roche Glycart AGSchlierenSwitzerland
| | - Reinhard Dummer
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
| | - Metello Innocenti
- Heidelberg University Biochemistry Center (BZH)Heidelberg UniversityHeidelbergGermany
| | - Sabine Werner
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| |
Collapse
|
114
|
Lu Z, Chiu J, Lee LR, Schindeler A, Jackson M, Ramaswamy Y, Dunstan CR, Hogg PJ, Zreiqat H. Reprogramming of human fibroblasts into osteoblasts by insulin-like growth factor-binding protein 7. Stem Cells Transl Med 2020; 9:403-415. [PMID: 31904196 PMCID: PMC7031646 DOI: 10.1002/sctm.19-0281] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/16/2019] [Indexed: 12/22/2022] Open
Abstract
The induced pluripotent stem cell (iPSC) is a promising cell source for tissue regeneration. However, the therapeutic value of iPSC technology is limited due to the complexity of induction protocols and potential risks of teratoma formation. A trans-differentiation approach employing natural factors may allow better control over reprogramming and improved safety. We report here a novel approach to drive trans-differentiation of human fibroblasts into functional osteoblasts using insulin-like growth factor binding protein 7 (IGFBP7). We initially determined that media conditioned by human osteoblasts can induce reprogramming of human fibroblasts to functional osteoblasts. Proteomic analysis identified IGFBP7 as being significantly elevated in media conditioned with osteoblasts compared with those with fibroblasts. Recombinant IGFBP7 induced a phenotypic switch from fibroblasts to osteoblasts. The switch was associated with senescence and dependent on autocrine IL-6 signaling. Our study supports a novel strategy for regenerating bone by using IGFBP7 to trans-differentiate fibroblasts to osteoblasts.
Collapse
Affiliation(s)
- ZuFu Lu
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical EngineeringThe University of SydneyCamperdownNew South WalesAustralia
- ARC Training Centre for Innovative BioEngineeringThe University of SydneyCamperdownNew South WalesAustralia
| | - Joyce Chiu
- The Centenary InstituteNHMRC Clinical Trial Centre, The University of SydneyCamperdownNew South WalesAustralia
| | - Lucinda R. Lee
- Bioengineering & Molecular MedicineThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Discipline of Child and Adolescent MedicineThe University of SydneyCamperdownNew South WalesAustralia
| | - Aaron Schindeler
- Bioengineering & Molecular MedicineThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Discipline of Child and Adolescent MedicineThe University of SydneyCamperdownNew South WalesAustralia
| | - Miriam Jackson
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical EngineeringThe University of SydneyCamperdownNew South WalesAustralia
| | - Yogambha Ramaswamy
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical EngineeringThe University of SydneyCamperdownNew South WalesAustralia
- ARC Training Centre for Innovative BioEngineeringThe University of SydneyCamperdownNew South WalesAustralia
| | - Colin R. Dunstan
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical EngineeringThe University of SydneyCamperdownNew South WalesAustralia
- ARC Training Centre for Innovative BioEngineeringThe University of SydneyCamperdownNew South WalesAustralia
| | - Philip J. Hogg
- The Centenary InstituteNHMRC Clinical Trial Centre, The University of SydneyCamperdownNew South WalesAustralia
| | - Hala Zreiqat
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical EngineeringThe University of SydneyCamperdownNew South WalesAustralia
- ARC Training Centre for Innovative BioEngineeringThe University of SydneyCamperdownNew South WalesAustralia
| |
Collapse
|
115
|
Arefin S, Buchanan S, Hobson S, Steinmetz J, Alsalhi S, Shiels PG, Kublickiene K, Stenvinkel P. Nrf2 in early vascular ageing: Calcification, senescence and therapy. Clin Chim Acta 2020; 505:108-118. [PMID: 32097628 DOI: 10.1016/j.cca.2020.02.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022]
Abstract
Under normal physiological conditions, free radical generation and antioxidant defences are balanced, and reactive oxygen species (ROS) usually act as secondary messengers in a plethora of biological processes. However, when this balance is impaired, oxidative stress develops due to imbalanced redox homeostasis resulting in cellular damage. Oxidative stress is now recognized as a trigger of cellular senescence, which is associated with multiple chronic 'burden of lifestyle' diseases, including atherosclerosis, type-2 diabetes, chronic kidney disease and vascular calcification; all of which possess signs of early vascular ageing. Nuclear factor erythroid 2-related factor 2 (Nrf2), termed the master regulator of antioxidant responses, is a transcription factor found to be frequently dysregulated in conditions characterized by oxidative stress and inflammation. Recent evidence suggests that activation of Nrf2 may be beneficial in protecting against vascular senescence and calcification. Both natural and synthetic Nrf2 agonists have been introduced as promising drug classes in different phases of clinical trials. However, overexpression of the Nrf2 pathway has also been linked to tumorigenesis, which highlights the requirement for further understanding of pathways involving Nrf2 activity, especially in the context of cellular senescence and vascular calcification. Therefore, comprehensive translational pre-clinical and clinical studies addressing the targeting capabilities of Nrf2 agonists are urgently required. The present review discusses the impact of Nrf2 in senescence and calcification in early vascular ageing, with focus on the potential clinical implications of Nrf2 agonists and non-pharmacological Nrf2 therapeutics.
Collapse
Affiliation(s)
- Samsul Arefin
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Sarah Buchanan
- Institute of Cancer Sciences, Wolfson Wohl CRC, ICS, MVLS, University of Glasgow, Glasgow, UK
| | - Sam Hobson
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Julia Steinmetz
- Rheumatology Unit, Dep. of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Shno Alsalhi
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden; Research Center, Salahaddin University-Erbil, 44001 Erbil, Kurdistan-Region, Iraq
| | - Paul G Shiels
- Institute of Cancer Sciences, Wolfson Wohl CRC, ICS, MVLS, University of Glasgow, Glasgow, UK
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden.
| |
Collapse
|
116
|
Wilkinson HN, Hardman MJ. Wound senescence: A functional link between diabetes and ageing? Exp Dermatol 2020; 30:68-73. [PMID: 32009254 DOI: 10.1111/exd.14082] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 12/11/2022]
Abstract
Arguably, the two most important causes of pathological healing in the skin are diabetes and ageing. While these factors have historically been considered independent modifiers of the healing process, recent studies suggest that they may be mechanistically linked. The primary contributor to diabetic pathology is hyperglycaemia, which accelerates the production of advanced glycation end products, a characteristic of ageing tissue. Indeed, advanced age also leads to mild hyperglycaemia. Here, we discuss emerging literature that reveals a hitherto unappreciated link between cellular senescence, diabetes and wound repair. Senescent cells cause widespread destruction of normal tissue architecture in ageing and have been shown to be increased in chronic wounds. However, the role of senescence remains controversial, with several studies reporting beneficial effects for transiently induced senescence in wound healing. We recently highlighted a direct role for senescence in diabetic healing pathology, mediated by the senescence receptor, CXCR2. These findings suggest that targeting local tissue senescence may provide a therapeutic strategy applicable to a broad range of chronic wound types.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, The University of Hull, Hull, UK
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, The University of Hull, Hull, UK
| |
Collapse
|
117
|
Hinz B, Lagares D. Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases. Nat Rev Rheumatol 2020; 16:11-31. [PMID: 31792399 PMCID: PMC7913072 DOI: 10.1038/s41584-019-0324-5] [Citation(s) in RCA: 384] [Impact Index Per Article: 76.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
Abstract
Organ fibrosis is a lethal outcome of autoimmune rheumatic diseases such as systemic sclerosis. Myofibroblasts are scar-forming cells that are ultimately responsible for the excessive synthesis, deposition and remodelling of extracellular matrix proteins in fibrosis. Advances have been made in our understanding of the mechanisms that keep myofibroblasts in an activated state and control myofibroblast functions. However, the mechanisms that help myofibroblasts to persist in fibrotic tissues remain poorly understood. Myofibroblasts evade apoptosis by activating molecular mechanisms in response to pro-survival biomechanical and growth factor signals from the fibrotic microenvironment, which can ultimately lead to the acquisition of a senescent phenotype. Growing evidence suggests that myofibroblasts and senescent myofibroblasts, rather than being resistant to apoptosis, are actually primed for apoptosis owing to concomitant activation of cell death signalling pathways; these cells are poised to apoptose when survival pathways are inhibited. This knowledge of apoptotic priming has paved the way for new therapies that trigger apoptosis in myofibroblasts by blocking pro-survival mechanisms, target senescent myofibroblast for apoptosis or promote the reprogramming of myofibroblasts into scar-resolving cells. These novel strategies are not only poised to prevent progressive tissue scarring, but also have the potential to reverse established fibrosis and to regenerate chronically injured tissues.
Collapse
Affiliation(s)
- Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - David Lagares
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Fibrosis Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
118
|
Naba A, Ricard-Blum S. The Extracellular Matrix Goes -Omics: Resources and Tools. EXTRACELLULAR MATRIX OMICS 2020. [DOI: 10.1007/978-3-030-58330-9_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
119
|
Tussilagonone Ameliorates Psoriatic Features in Keratinocytes and Imiquimod-Induced Psoriasis-Like Lesions in Mice via NRF2 Activation. J Invest Dermatol 2019; 140:1223-1232.e4. [PMID: 31877316 DOI: 10.1016/j.jid.2019.12.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/31/2022]
Abstract
Psoriasis is a common inflammatory skin disorder that is characterized by keratinocyte hyperproliferation and abnormal differentiation, resulting in the thickening of the epidermis and stratum corneum. In this study, we investigated in vitro and in vivo pharmacological effects of tussilagonone (TGN), a sesquiterpenoid isolated from Tussilago farfara, on transcription factors relevant for the pathogenesis of psoriasis. TGN inhibited activation of NF-κB and STAT3, leading to the attenuated expression of psoriasis-related inflammatory genes and suppression of keratinocyte hyperproliferation. Mechanistically, we show that the inhibition of NF-κB and STAT3 by TGN is mediated through activation of the cytoprotective transcription factor NRF2. Evaluation of in vivo antipsoriatic effects of topical TGN in the imiquimod-induced psoriasis-like dermatitis mouse model demonstrated amelioration of imiquimod-induced phenotypical changes, lesion severity score, epidermal thickening, and reduction in dermal cellularity. The spleen index also diminished in TGN-treated mice, suggesting anti-inflammatory properties of TGN. Moreover, TGN significantly attenuated the imiquimod-induced mRNA levels of psoriasis-associated inflammatory cytokines and antimicrobial peptides and reduced epidermal hyperproliferation. Taken together, TGN, as a potent NRF2 activator, is a promising therapeutic candidate for the development of antipsoriatic agents derived from medicinal plants.
Collapse
|
120
|
Gruber F, Kremslehner C, Eckhart L, Tschachler E. Cell aging and cellular senescence in skin aging - Recent advances in fibroblast and keratinocyte biology. Exp Gerontol 2019; 130:110780. [PMID: 31794850 DOI: 10.1016/j.exger.2019.110780] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/07/2019] [Accepted: 11/10/2019] [Indexed: 12/17/2022]
Abstract
The aging of the skin is the most visible and obvious manifestation of organismal aging and may serve as a predictor of life expectancy and health. It is, however, also the human desire for long-lasting beauty that further raises interests in the topic, and thus considerable means and efforts are put into studying the mechanisms of skin aging in basic and applied research. Both medical und non-medical interests are of benefit for skin research in general because the results from these studies help to deepen our understanding of the complex molecular, biological, cell signaling, developmental and immunological processes in this organ. In fact, the skin is an ideal organ to observe and analyze the impact of extrinsic and intrinsic drivers of aging. Within the past five years technological advances like lineage tracing of cells in model organisms, intra-vital microscopy, nucleic acid sequencing at the single cell level, and high resolution mass spectrometry have allowed to study aging and senescence of individual skin cells within the tissue context, their signaling and communication, and to derive new hypotheses for experimental studies in vitro. In this short review we will discuss very recent developments that promise to extend the existing knowledge on cell aging and senescence of dermal fibroblasts and epidermal keratinocytes in skin aging.
Collapse
Affiliation(s)
- Florian Gruber
- Division for Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for the Biotechnology of Skin Aging, Vienna, Austria.
| | - Christopher Kremslehner
- Division for Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for the Biotechnology of Skin Aging, Vienna, Austria
| | - Leopold Eckhart
- Division for Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Erwin Tschachler
- Division for Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
121
|
Weavers H, Wood W, Martin P. Injury Activates a Dynamic Cytoprotective Network to Confer Stress Resilience and Drive Repair. Curr Biol 2019; 29:3851-3862.e4. [PMID: 31668626 PMCID: PMC6868510 DOI: 10.1016/j.cub.2019.09.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 02/07/2023]
Abstract
In healthy individuals, injured tissues rapidly repair themselves following damage. Within a healing skin wound, recruited inflammatory cells release a multitude of bacteriocidal factors, including reactive oxygen species (ROS), to eliminate invading pathogens. Paradoxically, while these highly reactive ROS confer resistance to infection, they are also toxic to host tissues and may ultimately delay repair. Repairing tissues have therefore evolved powerful cytoprotective "resilience" machinery to protect against and tolerate this collateral damage. Here, we use in vivo time-lapse imaging and genetic manipulation in Drosophila to dissect the molecular and cellular mechanisms that drive tissue resilience to wound-induced stress. We identify a dynamic, cross-regulatory network of stress-activated cytoprotective pathways, linking calcium, JNK, Nrf2, and Gadd45, that act to both "shield" tissues from oxidative damage and promote efficient damage repair. Ectopic activation of these pathways confers stress protection to naive tissue, while their inhibition leads to marked delays in wound closure. Strikingly, the induction of cytoprotection is tightly linked to the pathways that initiate the inflammatory response, suggesting evolution of a fail-safe mechanism for tissue protection each time inflammation is triggered. A better understanding of these resilience mechanisms-their identities and precise spatiotemporal regulation-is of major clinical importance for development of therapeutic interventions for all pathologies linked to oxidative stress, including debilitating chronic non-healing wounds.
Collapse
Affiliation(s)
- Helen Weavers
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK.
| | - Will Wood
- School of Cellular and Molecular Medicine, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK; Centre for Inflammation Research, University of Edinburgh, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK; School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK; School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
122
|
Fang J, Yan Y, Teng X, Wen X, Li N, Peng S, Liu W, Donadeu FX, Zhao S, Hua J. Melatonin prevents senescence of canine adipose-derived mesenchymal stem cells through activating NRF2 and inhibiting ER stress. Aging (Albany NY) 2019; 10:2954-2972. [PMID: 30362962 PMCID: PMC6224246 DOI: 10.18632/aging.101602] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022]
Abstract
Transplantation of adipose-derived mesenchymal stem cells (ADMSCs) can aid in the treatment of numerous diseases in animals. However, natural aging during in vitro expansion of ADMSCs prior to their use in transplantation restricts their beneficial effects. Melatonin is reported to exert biorhythm regulation, anti-oxidation, and anti-senescence effects in various animal and cell models. Herein, by using a senescent canine ADMSCs (cADMSCs) cell model subjected to multiple passages in vitro, we investigated the effects of melatonin on ADMSCs senescence. We found that melatonin alleviates endoplasmic reticulum stress (ERS) and cell senescence. MT1/MT2 melatonin receptor inhibitor, luzindole, diminished the mRNA expression levels and rhythm expression amplitude of Bmal1 and Nrf2 genes. Nrf2 knockdown blocked the stimulatory effects of melatonin on endoplasmic reticulum-associated degradation (ERAD)-related gene expression and its inhibitory effects on ERS-related gene expression. At the same time, the inhibitory effects of melatonin on the NF-κB signaling pathway and senescence-associated secretory phenotype (SASP) were blocked by Nrf2 knockdown in cADMSCs. Melatonin pretreatment improved the survival of cADMSCs and enhanced the beneficial effects of cADMSCs transplantation in canine acute liver injury. These results indicate that melatonin activates Nrf2 through the MT1/MT2 receptor pathway, stimulates ERAD, inhibits NF-κB and ERS, alleviates cADMSCs senescence, and improves the efficacy of transplanted cADMSCs.
Collapse
Affiliation(s)
- Jia Fang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A and F University, Yangling, Shaanxi Province, China
| | - Yuan Yan
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A and F University, Yangling, Shaanxi Province, China
| | - Xin Teng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A and F University, Yangling, Shaanxi Province, China
| | - Xinyu Wen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A and F University, Yangling, Shaanxi Province, China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A and F University, Yangling, Shaanxi Province, China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A and F University, Yangling, Shaanxi Province, China
| | - Wenshuai Liu
- Department of Pathology, Yangling Demonstration Zone Hospital, Yangling, Shaanxi Province, China
| | - F Xavier Donadeu
- Division of Developmental Biology, The Roslin Institute Reader, Royal (Dick) School of Veterinary Studies University of Edinburgh, Easter Bush, Midlothian, EH25 9RG Scotland, UK
| | - Shanting Zhao
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A and F University, Yangling, Shaanxi Province, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A and F University, Yangling, Shaanxi Province, China
| |
Collapse
|
123
|
Abstract
Following tissue injury, cells produce reactive molecules that fight off invading pathogens, but these factors might also damage the host tissue. A new study has characterized a network of defense pathways that synergize to protect cells from collateral damage and drive repair.
Collapse
Affiliation(s)
- Paul Hiebert
- Institute of Molecular Health Sciences, ETH Zurich, Switzerland.
| | - Sabine Werner
- Institute of Molecular Health Sciences, ETH Zurich, Switzerland.
| |
Collapse
|
124
|
Moos WH, Faller DV, Glavas IP, Harpp DN, Kanara I, Pinkert CA, Powers WR, Sampani K, Steliou K, Vavvas DG, Kodukula K, Zamboni RJ. Epigenetic treatment of dermatologic disorders. Drug Dev Res 2019. [DOI: 10.1002/ddr.21562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Walter H. Moos
- Department of Pharmaceutical Chemistry, School of PharmacyUniversity of California, San Francisco San Francisco California
- ShangPharma Innovation Inc. South San Francisco California
| | - Douglas V. Faller
- Department of MedicineBoston University School of Medicine Boston Massachusetts
- Cancer Research CenterBoston University School of Medicine Boston Massachusetts
| | - Ioannis P. Glavas
- Department of OphthalmologyNew York University School of Medicine New York City New York
| | - David N. Harpp
- Department of ChemistryMcGill University Montreal Quebec Canada
| | | | - Carl A. Pinkert
- Department of Pathobiology, College of Veterinary MedicineAuburn University Auburn Alabama
| | - Whitney R. Powers
- Department of Health SciencesBoston University Boston Massachusetts
- Department of AnatomyBoston University School of Medicine Boston Massachusetts
| | - Konstantina Sampani
- Beetham Eye InstituteJoslin Diabetes Center Boston Massachusetts
- Department of MedicineHarvard Medical School Boston Massachusetts
| | - Kosta Steliou
- Cancer Research CenterBoston University School of Medicine Boston Massachusetts
- PhenoMatriX, Inc. Natick Massachusetts
| | - Demetrios G. Vavvas
- Retina Service, Angiogenesis LaboratoryMassachusetts Eye and Ear Infirmary Boston Massachusetts
- Department of OphthalmologyHarvard Medical School Boston Massachusetts
| | - Krishna Kodukula
- ShangPharma Innovation Inc. South San Francisco California
- PhenoMatriX, Inc. Natick Massachusetts
| | | |
Collapse
|
125
|
Sun T, Ghosh AK, Eren M, Miyata T, Vaughan DE. PAI-1 contributes to homocysteine-induced cellular senescence. Cell Signal 2019; 64:109394. [PMID: 31472244 DOI: 10.1016/j.cellsig.2019.109394] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/12/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022]
Abstract
Cellular Senescence is associated with organismal aging and related pathologies. Previously, we reported that plasminogen activator inhibitor-1 (PAI-1) is an essential mediator of senescence and a potential therapeutic target for preventing aging-related pathologies. In this study, we investigate the efficacies of PAI-1 inhibitors in both in vitro and in vivo models of homocysteine (Hcy)-induced cardiovascular aging. Elevated Hcy, a known risk factor of cardiovascular diseases, induces endothelial senescence as evidenced by increased senescence-associated β-Gal positivity (SA-β-Gal), flattened cellular morphology, and cylindrical appearance of cellular nuclei. Importantly, inhibition of PAI-1 by small molecule inhibitors reduces the number of SA-β-Gal positive cells, normalizes cellular morphology and nuclear shape. Furthermore, while Hcy induces the levels of senescence regulators PAI-1, p16, p53 and integrin β3, and suppresses catalase expression, treatment with PAI-1 inhibitors blocks the Hcy-induced stimulation of senescence cadres, and reverses the Hcy-induced suppression of catalase, indicating that PAI-1 specific small molecule inhibitors are efficient to prevent Hcy-induced cellular senescence. Our in vivo study shows that the levels of integrin β3, a recently identified potential regulator of cellular senescence, and its interaction with PAI-1 are significantly elevated in Hcy-treated heart tissues. In contrast, Hcy suppresses antioxidant gene regulator Nrf2 expression in hearts. However, co-treatment with PAI-1 inhibitor completely blocks the stimulation of Hcy-induced induction of integrin β3 and reverses Nrf2 expression. Collectively these in vitro and in vivo studies indicate that pharmacological inhibition of PAI-1 improves endothelial and cardiac health by suppressing the pro-senescence effects of hyperhomocysteinemia through suppression of Hcy-induced master regulators of cellular senescence PAI-1 and integrin β3. Therefore, PAI-1 inhibitors are promising drugs for amelioration of hyperhomocysteinemia-induced vascular aging and aging-related disease.
Collapse
Affiliation(s)
- Tianjiao Sun
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Asish K Ghosh
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Mesut Eren
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Toshio Miyata
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA; United Centers for Advanced Research and Translational Medicine, Tohoku University, Miyagi, Japan
| | - Douglas E Vaughan
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
126
|
Hiebert P, Werner S. Regulation of Wound Healing by the NRF2 Transcription Factor-More Than Cytoprotection. Int J Mol Sci 2019; 20:ijms20163856. [PMID: 31398789 PMCID: PMC6720615 DOI: 10.3390/ijms20163856] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/31/2022] Open
Abstract
The nuclear factor-erythroid 2-related factor 2 (NRF2) transcription factor plays a central role in mediating the cellular stress response. Due to their antioxidant properties, compounds activating NRF2 have received much attention as potential medications for disease prevention, or even for therapy. Accumulating evidence suggests that activation of the NRF2 pathway also has a major impact on wound healing and may be beneficial in the treatment of chronic wounds, which remain a considerable health and economic burden. While NRF2 activation indeed shows promise, important considerations need to be made in light of corresponding evidence that also points towards pro-tumorigenic effects of NRF2. In this review, we discuss the evidence to date, highlighting recent advances using gain- and loss-of-function animal models and how these data fit with observations in humans.
Collapse
Affiliation(s)
- Paul Hiebert
- Institute for Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zürich, 8093 Zurich, Switzerland.
| | - Sabine Werner
- Institute for Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zürich, 8093 Zurich, Switzerland.
| |
Collapse
|
127
|
The TGF-β1/p53/PAI-1 Signaling Axis in Vascular Senescence: Role of Caveolin-1. Biomolecules 2019; 9:biom9080341. [PMID: 31382626 PMCID: PMC6723262 DOI: 10.3390/biom9080341] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022] Open
Abstract
Stress-induced premature cellular senescence is a significant factor in the onset of age-dependent disease in the cardiovascular system. Plasminogen activator inhibitor-1 (PAI-1), a major TGF-β1/p53 target gene and negative regulator of the plasmin-based pericellular proteolytic cascade, is elevated in arterial plaques, vessel fibrosis, arteriosclerosis, and thrombosis, correlating with increased tissue TGF-β1 levels. Additionally, PAI-1 is necessary and sufficient for the induction of p53-dependent replicative senescence. The mechanism of PAI-1 transcription in senescent cells appears to be dependent on caveolin-1 signaling. Src kinases are upstream effectors of both FAK and caveolin-1 activation as FAKY577,Y861 and caveolin-1Y14 phosphorylation are not detected in TGF-β1-stimulated src family kinase (pp60c-src, Yes, Fyn) triple-deficient (SYF−/−/−) cells. However, restoration of pp60c-src expression in SYF-null cells rescued both caveolin-1Y14 phosphorylation and PAI-1 induction in response to TGF-β1. Furthermore, TGF-β1-initiated Src phosphorylation of caveolin-1Y14 is critical in Rho-ROCK-mediated suppression of the SMAD phosphatase PPM1A maintaining and, accordingly, SMAD2/3-dependent transcription of the PAI-1 gene. Importantly, TGF-β1 failed to induce PAI-1 expression in caveolin-1-null cells, correlating with reductions in both Rho-GTP loading and SMAD2/3 phosphorylation. These findings implicate caveolin-1 in expression controls on specific TGF-β1/p53 responsive growth arrest genes. Indeed, up-regulation of caveolin-1 appears to stall cells in G0/G1 via activation of the p53/p21 cell cycle arrest pathway and restoration of caveolin-1 in caveolin-1-deficient cells rescues TGF-β1 inducibility of the PAI-1 gene. Although the mechanism is unclear, caveolin-1 inhibits p53/MDM2 complex formation resulting in p53 stabilization, induction of p53-target cell cycle arrest genes (including PAI-1), and entrance into premature senescence while stimulating the ATM→p53→p21 pathway. Identification of molecular events underlying senescence-associated PAI-1 expression in response to TGF-β1/src kinase/p53 signaling may provide novel targets for the therapy of cardiovascular disease.
Collapse
|
128
|
Wang Z, Chen Z, Jiang Z, Luo P, Liu L, Huang Y, Wang H, Wang Y, Long L, Tan X, Liu D, Jin T, Wang Y, Wang Y, Liao F, Zhang C, Chen L, Gan Y, Liu Y, Yang F, Huang C, Miao H, Chen J, Cheng T, Fu X, Shi C. Cordycepin prevents radiation ulcer by inhibiting cell senescence via NRF2 and AMPK in rodents. Nat Commun 2019; 10:2538. [PMID: 31182708 PMCID: PMC6557849 DOI: 10.1038/s41467-019-10386-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 05/08/2019] [Indexed: 12/13/2022] Open
Abstract
The pathological mechanisms of radiation ulcer remain unsolved and there is currently no effective medicine. Here, we demonstrate that persistent DNA damage foci and cell senescence are involved in radiation ulcer development. Further more, we identify cordycepin, a natural nucleoside analogue, as a potent drug to block radiation ulcer (skin, intestine, tongue) in rats/mice by preventing cell senescence through the increase of NRF2 nuclear expression (the assay used is mainly on skin). Finally, cordycepin is also revealed to activate AMPK by binding with the α1 and γ1 subunit near the autoinhibitory domain of AMPK, then promotes p62-dependent autophagic degradation of Keap1, to induce NRF2 dissociate from Keap1 and translocate to the nucleus. Taken together, our findings identify cordycepin prevents radiation ulcer by inhibiting cell senescence via NRF2 and AMPK in rodents, and activation of AMPK or NRF2 may thus represent therapeutic targets for preventing cell senescence and radiation ulcer.
Collapse
Affiliation(s)
- Ziwen Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Zelin Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Zhongyong Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Peng Luo
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Lang Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
- Department of Toxicology, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Yu Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
- Department of Toxicology, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Huilan Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
- Institute of Clinical Medicine, Southwest Medical University, 646000, Luzhou, China
| | - Yu Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Lei Long
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Xu Tan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Dengqun Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Taotao Jin
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yawei Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Fengying Liao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chi Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Long Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yibo Gan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yunsheng Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Fan Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chunji Huang
- College of Basic Medical Sciences, Third Military Medical University, 400038, Chongqing, China
| | - Hongming Miao
- College of Basic Medical Sciences, Third Military Medical University, 400038, Chongqing, China
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University, 40038, Chongqing, China
| | - Tianmin Cheng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, the First Affiliated Hospital, Chinese PLA General Hospital, Trauma Center of Postgraduate Medical College, 100000, Beijing, China.
| | - Chunmeng Shi
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force Medicine, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| |
Collapse
|
129
|
Brown Y, Hua S, Tanwar PS. Extracellular matrix-mediated regulation of cancer stem cells and chemoresistance. Int J Biochem Cell Biol 2019; 109:90-104. [DOI: 10.1016/j.biocel.2019.02.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/03/2019] [Accepted: 02/05/2019] [Indexed: 12/12/2022]
|
130
|
Muzumdar S, Hiebert H, Haertel E, Ben-Yehuda Greenwald M, Bloch W, Werner S, Schäfer M. Nrf2-Mediated Expansion of Pilosebaceous Cells Accelerates Cutaneous Wound Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:568-579. [PMID: 30593821 DOI: 10.1016/j.ajpath.2018.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/14/2018] [Accepted: 11/19/2018] [Indexed: 12/15/2022]
Abstract
The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcription factor is a key regulator of the cellular stress response. Therefore, pharmacologic Nrf2 activation is a promising strategy for skin protection and cancer prevention. This study found that genetic Nrf2 activation in keratinocytes accelerates wound repair. Enhanced proliferation of cells of the pilosebaceous unit peripheral to the wound and a concomitant acceleration of re-epithelialization were identified as the underlying mechanism. Nrf2 specifically promoted the expansion of pilosebaceous cells expressing markers of junctional zone and upper isthmus follicular stem cells. This may result, at least in part, from the up-regulation of the direct Nrf2 target epigen and a concomitant increase in epidermal growth factor receptor signaling. The increase in pilosebaceous cells provided a larger pool of keratinocytes that migrate into the wound, resulting in faster wound closure. These results unravel a novel function of Nrf2 in wound repair and suggest the use of NRF2-activating compounds in patients with impaired healing.
Collapse
Affiliation(s)
- Sukalp Muzumdar
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Hayley Hiebert
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Eric Haertel
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Matthias Schäfer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
131
|
Song S, Wu S, Wang Y, Wang Z, Ye C, Song R, Song D, Ruan Y. 17β-estradiol inhibits human umbilical vascular endothelial cell senescence by regulating autophagy via p53. Exp Gerontol 2018; 114:57-66. [PMID: 30399406 DOI: 10.1016/j.exger.2018.10.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/30/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022]
Abstract
Vascular endothelial cell (VEC) senescence is an initiating factor in numerous cardiovascular diseases. Recent studies showed that 17β-estradiol (17β-E2), an estrogen with numerous biological activities such as inhibition of atherosclerosis, protects VECs from senescence. However, the effects of 17β-E2 on human umbilical VECs (HUVECs) remain unknown. This study investigated the anti-senescent effect of 17β-E2 on HUVECs and explored the underlying mechanism with respect to autophagy and p53 activity. First, rapamycin and 3-methyladenine were used to clarify the relationship between autophagy and senescence in HUVECs, and an inverse relationship was demonstrated. Next, the effect of 17β-E2 on H2O2-induced senescence of HUVECs was examined. Increased autophagy induced by 17β-E2 inhibited H2O2-induced senescence of HUVECs, increased cell viability, and maintained HUVEC morphology. 17β-E2 pre-treatment also decreased cell cycle arrest, decreased the dephosphorylation of Rb, decreased the production of ET-1, and increased the production of NO. Most importantly, 17β-E2 pre-treatment increased autophagy by activating p53 and its downstream effector p53-upregulated modulator of apoptosis (PUMA). Overall, our data indicate the critical role of autophagy in the anti-senescent effect of 17β-E2 on HUVECs.
Collapse
Affiliation(s)
- Shicong Song
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Saizhu Wu
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuyan Wang
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Wang
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changxiong Ye
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rui Song
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongqing Song
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yunjun Ruan
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
132
|
Schmidt A, Bekeschus S. Redox for Repair: Cold Physical Plasmas and Nrf2 Signaling Promoting Wound Healing. Antioxidants (Basel) 2018; 7:E146. [PMID: 30347767 PMCID: PMC6210784 DOI: 10.3390/antiox7100146] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/12/2018] [Accepted: 10/18/2018] [Indexed: 12/15/2022] Open
Abstract
Chronic wounds and ulcers are major public health threats. Being a substantial burden for patients and health care systems alike, better understanding of wound pathophysiology and new avenues in the therapy of chronic wounds are urgently needed. Cold physical plasmas are particularly effective in promoting wound closure, irrespective of its etiology. These partially ionized gases deliver a therapeutic cocktail of reactive oxygen and nitrogen species safely at body temperature and without genotoxic side effects. This field of plasma medicine reanimates the idea of redox repair in physiological healing. This review compiles previous findings of plasma effects in wound healing. It discusses new links between plasma treatment of cells and tissues, and the perception and intracellular translation of plasma-derived reactive species via redox signaling pathways. Specifically, (i) molecular switches governing redox-mediated tissue response; (ii) the activation of the nuclear E2-related factor (Nrf2) signaling, together with antioxidative and immunomodulatory responses; and (iii) the stabilization of the scaffolding function and actin network in dermal fibroblasts are emphasized in the light of wound healing.
Collapse
Affiliation(s)
- Anke Schmidt
- Plasma Life Science, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany.
| | - Sander Bekeschus
- ZIK-PRE, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany.
| |
Collapse
|