1
|
Keizer HG, Brands R, Oosting RS, Seinen W. A comprehensive model for the biochemistry of ageing, senescence and longevity. Biogerontology 2024; 25:615-626. [PMID: 38441836 DOI: 10.1007/s10522-024-10097-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/07/2024] [Indexed: 07/02/2024]
Abstract
Various models for ageing, each focussing on different biochemical and/or cellular pathways have been proposed. This has resulted in a complex and non-coherent portrayal of ageing. Here, we describe a concise and comprehensive model for the biochemistry of ageing consisting of three interacting signalling hubs. These are the nuclear factor kappa B complex (NFκB), controlling the innate immune system, the mammalian target for rapamycin complex, controlling cell growth, and the integrated stress responses, controlling homeostasis. This model provides a framework for most other, more detailed, biochemical pathways involved in ageing, and explains why ageing involves chronic inflammation, cellular senescence, and vulnerability to environmental stress, while starting with the spontaneous formation of advanced glycation end products. The totality of data underlying this model suggest that the gradual inhibition of the AMPK-ISR probably determines the maximal lifespan. Based on this model, anti-ageing drugs in general, are expected to show hormetic dose response curves. This complicates the process of dose-optimization. Due to its specific mechanism of action, the anti-aging drug alkaline phosphatase is an exception to this rule, because it probably exhibits saturation kinetics.
Collapse
Affiliation(s)
| | - R Brands
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands
| | - Ronald Sake Oosting
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands
| | - Willem Seinen
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands
- Institute for Risk Assessment Sciences (IRAS), Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| |
Collapse
|
2
|
Liu W, Li HM, Bai G. Construction of a novel mRNA-miRNA-lncRNA/circRNA triple subnetwork associated with immunity and aging in intervertebral disc degeneration. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:1176-1195. [PMID: 38555595 DOI: 10.1080/15257770.2024.2334353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVE Intervertebral disk degeneration (IVDD) is one of the most common causes of low back pain. However, in the etiology of IVDD, the specific method by which nucleus pulposus (NP) cell senescence and the immune response induce disease is uncertain. METHODS Gene Expression Omnibus database was used to find differentially expressed genes (DEGs), differentially expressed miRNAs (DE miRNAs), differentially expressed lncRNAs (DE lncRNAs), and differentially expressed circRNAs (DE circRNAs). Functional enrichment analysis was performed through Enrichr database. Potential regulatory miRNAs, lncRNAs and circRNAs of mRNAs were predicted by ENCORI and circBank, respectively. RESULTS We identified 198 upregulated and 131 downregulated genes, 39 upregulated and 22 downregulated miRNAs, 2152 upregulated and 564 downregulated lncRNAs, and 352 upregulated and 279 downregulated circRNAs as DEGs, DE miRNAs, DE lncRNAs, DE circRNAs, respectively. Functional enrichment analysis revealed that they were significantly enriched in Toll-like receptor signaling route and the NF-kappa B signaling pathway. An mRNA-miRNA-lncRNA/circRNA network linked to the pathogenesis of NP cells in IVDD was constructed based on node degree and differential expression level. Eight immune-related DEGs (6 upregulated and 2 downregulated genes) and five aging-related DEGs (3 upregulated and 2 downregulated genes) were identified in the critical network. CONCLUSION We established a novel immune-related and aging-related triple regulatory network of mRNA-miRNA-lncRNA/circRNA ceRNA, among which all RNAs may be utilized as the pathogenesis biomarker of NP cells in IVDD.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopedics, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, P R China
| | - Hui-Min Li
- Department of Orthopedics, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, P R China
| | - Guangchao Bai
- Department of Orthopedics, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, P R China
| |
Collapse
|
3
|
Harada M, Su-Harada K, Kimura T, Ono K, Ashida N. Sustained activation of NF-κB through constitutively active IKKβ leads to senescence bypass in murine dermal fibroblasts. Cell Cycle 2024; 23:308-327. [PMID: 38461418 PMCID: PMC11057680 DOI: 10.1080/15384101.2024.2325802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Although the transcription factor nuclear factor κB (NF-κB) plays a central role in the regulation of senescence-associated secretory phenotype (SASP) acquisition, our understanding of the involvement of NF-κB in the induction of cellular senescence is limited. Here, we show that activation of the canonical NF-κB pathway suppresses senescence in murine dermal fibroblasts. IκB kinase β (IKKβ)-depleted dermal fibroblasts showed ineffective NF-κB activation and underwent senescence more rapidly than control cells when cultured under 20% oxygen conditions, as indicated by senescence-associated β-galactosidase (SA-β-gal) staining and p16INK4a mRNA levels. Conversely, the expression of constitutively active IKKβ (IKKβ-CA) was sufficient to drive senescence bypass. Notably, the expression of a degradation-resistant form of inhibitor of κB (IκB), which inhibits NF-κB nuclear translocation, abolished senescence bypass, suggesting that the inhibitory effect of IKKβ-CA on senescence is largely mediated by NF-κB. We also found that IKKβ-CA expression suppressed the derepression of INK4/Arf genes and counteracted the senescence-associated loss of Ezh2, a catalytic subunit of the Polycomb repressive complex 2 (PRC2). Moreover, pharmacological inhibition of Ezh2 abolished IKKβ-CA-induced senescence bypass. We propose that NF-κB plays a suppressive role in the induction of stress-induced senescence through sustaining Ezh2 expression.
Collapse
Affiliation(s)
- Masayuki Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kanae Su-Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Noboru Ashida
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
4
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Xu J, Shao T, Lou J, Zhang J, Xia C. Aging, cell senescence, the pathogenesis and targeted therapies of intervertebral disc degeneration. Front Pharmacol 2023; 14:1172920. [PMID: 37214476 PMCID: PMC10196014 DOI: 10.3389/fphar.2023.1172920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) refers to the aging and degenerative diseases of intervertebral disc components such as nucleus pulposus, annulus fibrosus, and cartilage endplate, and is the main cause of chronic low back pain. Over the past few years, many researchers around the world concerned that the degeneration of nucleus pulposus (NP) cells plays the main role in IVDD. The degeneration of NP cells is caused by a series of pathological processes, including oxidative stress, inflammatory response, apoptosis, abnormal proliferation, and autophagy. Interestingly, many studies have found a close relationship between the senescence of NP cells and the progression of NP degeneration. The classical aging pathways also have been confirmed to be involved in the pathological process of IVDD. Moreover, several anti-aging drugs have been used to treat IVDD by inhibiting NP cells senescence, such as proanthocyanidins, resveratrol and bone morphogenetic protein 2. Therefore, this article will systematically list and discuss aging, cell senescence, the pathogenesis and targeted therapies of IVDD, in order to provide new ideas for the treatment of IVDD in the future.
Collapse
Affiliation(s)
- Jiongnan Xu
- Center for Plastic and Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital Hangzhou Medical College, Hangzhou, China
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Shao
- Center for Plastic and Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital Hangzhou Medical College, Hangzhou, China
| | - Jianfen Lou
- Center for Plastic and Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital Hangzhou Medical College, Hangzhou, China
| | - Jun Zhang
- Department of Orthopedics, Zhejiang Provincial People’s Hospital Bijie Hospital, Bijie, Guizhou, China
| | - Chen Xia
- Center for Plastic and Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
6
|
Jin S, Wang Y, Wu X, Li Z, Zhu L, Niu Y, Zhou Y, Liu Y. Young Exosome Bio-Nanoparticles Restore Aging-Impaired Tendon Stem/Progenitor Cell Function and Reparative Capacity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211602. [PMID: 36779444 DOI: 10.1002/adma.202211602] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/19/2023] [Indexed: 05/05/2023]
Abstract
Aging impairs tendon stem/progenitor cell function and tendon homeostasis, however, effective treatments for aging-induced tendon diseases are lacking. Exosomes are naturally derived nanoparticles that contain bioactive molecules, and therefore, have attracted great interest in tissue engineering and regenerative medicine. In this study, it is shown that young exosomes secreted by stem cells from human exfoliated deciduous teeth (SHED-Exos) possess abundant anti-aging signals. These young bio-nanoparticles can alleviate the aging phenotypes of aged tendon stem/progenitor cells (AT-SCs) and maintain their tenogenic capacity. Mechanistically, SHED-Exos modulate histone methylation and inhibit nuclear factor-κB to reverse AT-SC aging. In a naturally aging mouse model, systemic administration of SHED-Exo bio-nanoparticles retards tendon degeneration. Interestingly, local delivery of SHED-Exos-loaded microspheres confers anti-aging phenotypes, including reduced senescent cells and decreased ectopic bone formation, thereby functionally and structurally rescuing endogenous tendon regeneration and repair capacity in aged rats. Overall, SHED-Exos, as natural bioactive nanoparticles, have promising translational and therapeutic potential for aging-related diseases.
Collapse
Affiliation(s)
- Shanshan Jin
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Yu Wang
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Xiaolan Wu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Zixin Li
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Lisha Zhu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Yuting Niu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| |
Collapse
|
7
|
Schlett JS, Mettang M, Skaf A, Schweizer P, Errerd A, Mulugeta EA, Hein TM, Tsesmelis K, Tsesmelis M, Büttner UFG, Wendt H, Abaei A, Rasche V, Prex V, Nespoli E, Alami NO, Tews D, Walther P, Yilmazer-Hanke D, Oswald F, Dimou L, Wirth T, Baumann B. NF-κB is a critical mediator of post-mitotic senescence in oligodendrocytes and subsequent white matter loss. Mol Neurodegener 2023; 18:24. [PMID: 37069623 PMCID: PMC10108549 DOI: 10.1186/s13024-023-00616-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/25/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Inflammaging represents an accepted concept where the immune system shifts to a low-grade chronic pro-inflammatory state without overt infection upon aging. In the CNS, inflammaging is mainly driven by glia cells and associated with neurodegenerative processes. White matter degeneration (WMD), a well-known process in the aging brain, manifests in myelin loss finally resulting in motor, sensory and cognitive impairments. Oligodendrocytes (OL) are responsible for homeostasis and maintenance of the myelin sheaths, which is a complex and highly energy demanding process sensitizing these cells to metabolic, oxidative and other forms of stress. Yet, the immediate impact of chronic inflammatory stress like inflammaging on OL homeostasis, myelin maintenance and WMD remains open. METHODS To functionally analyze the role of IKK/NF-κB signaling in the regulation of myelin homeostasis and maintenance in the adult CNS, we established a conditional mouse model allowing NF-κB activation in mature myelinating oligodendrocytes. IKK2-CAPLP-CreERT2 mice were characterized by biochemical, immunohistochemical, ultrastructural and behavioral analyses. Transcriptome data from isolated, primary OLs and microglia cells were explored by in silico pathway analysis and validated by complementary molecular approaches. RESULTS Chronic NF-κB activation in mature OLs leads to aggravated neuroinflammatory conditions phenocopying brain inflammaging. As a consequence, IKK2-CAPLP-CreERT2 mice showed specific neurological deficits and impaired motoric learning. Upon aging, persistent NF-κB signaling promotes WMD in these mice as ultrastructural analysis revealed myelination deficits in the corpus callosum accompanied by impaired myelin protein expression. RNA-Seq analysis of primary oligodendrocytes and microglia cells uncovers gene expression signatures associated with activated stress responses and increased post mitotic cellular senescence (PoMiCS) which was confirmed by elevated senescence-associated β-galactosidase activity and SASP gene expression profile. We identified an elevated integrated stress response (ISR) characterized by phosphorylation of eIF2α as a relevant molecular mechanism which is able to affect translation of myelin proteins. CONCLUSIONS Our findings demonstrate an essential role of IKK/NF-κB signaling in mature, post-mitotic OLs in regulating stress-induced senescence in these cells. Moreover, our study identifies PoMICS as an important driving force of age-dependent WMD as well as of traumatic brain injury induced myelin defects.
Collapse
Affiliation(s)
- Judith Stefanie Schlett
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Melanie Mettang
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Aladdin Skaf
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Pavel Schweizer
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Alina Errerd
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | | | - Tabea Melissa Hein
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Konstantinos Tsesmelis
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Miltiadis Tsesmelis
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Ulrike F G Büttner
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Heinrich Wendt
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Alireza Abaei
- Core Facility Small Animal Imaging (CF-SANI), Ulm University, 89081, Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal Imaging (CF-SANI), Ulm University, 89081, Ulm, Germany
| | - Vivien Prex
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Ester Nespoli
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Najwa Ouali Alami
- Institute of Clinical Neuroanatomy, Ulm University, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Daniel Tews
- Core Facility Extracellular Flux Analyzer, Ulm University Medical Center, 89081, Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Deniz Yilmazer-Hanke
- Institute of Clinical Neuroanatomy, Ulm University, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Franz Oswald
- Department of Internal Medicine I, Center for Internal Medicine, University Medical Center Ulm, 89081, Ulm, Germany
| | - Leda Dimou
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
8
|
Velotti F, Bernini R. Hydroxytyrosol Interference with Inflammaging via Modulation of Inflammation and Autophagy. Nutrients 2023; 15:nu15071774. [PMID: 37049611 PMCID: PMC10096543 DOI: 10.3390/nu15071774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/14/2023] Open
Abstract
Inflammaging refers to a chronic, systemic, low-grade inflammation, driven by immune (mainly macrophages) and non-immune cells stimulated by endogenous/self, misplaced or altered molecules, belonging to physiological aging. This age-related inflammatory status is characterized by increased inflammation and decreased macroautophagy/autophagy (a degradation process that removes unnecessary or dysfunctional cell components). Inflammaging predisposes to age-related diseases, including obesity, type-2 diabetes, cancer, cardiovascular and neurodegenerative disorders, as well as vulnerability to infectious diseases and vaccine failure, representing thus a major target for anti-aging strategies. Phenolic compounds-found in extra-virgin olive oil (EVOO)-are well known for their beneficial effect on longevity. Among them, hydroxytyrosol (HTyr) appears to greatly contribute to healthy aging by its documented potent antioxidant activity. In addition, HTyr can modulate inflammation and autophagy, thus possibly counteracting and reducing inflammaging. In this review, we reference the literature on pure HTyr as a modulatory agent of inflammation and autophagy, in order to highlight its possible interference with inflammaging. This HTyr-mediated activity might contribute to healthy aging and delay the development or progression of diseases related to aging.
Collapse
Affiliation(s)
- Francesca Velotti
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Roberta Bernini
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, Via S. Camillo de Lellis, 01100 Viterbo, Italy
| |
Collapse
|
9
|
Effects of Oleuropein and Hydroxytyrosol on Inflammatory Mediators: Consequences on Inflammaging. Int J Mol Sci 2022; 24:ijms24010380. [PMID: 36613822 PMCID: PMC9820525 DOI: 10.3390/ijms24010380] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Aging is associated with a low-grade, systemic inflammatory state defined as "inflammaging", ruled by the loss of proper regulation of the immune system leading to the accumulation of pro-inflammatory mediators. Such a condition is closely connected to an increased risk of developing chronic diseases. A number of studies demonstrate that olive oil phenolic compound oleuropein and its derivative hydroxytyrosol contribute to modulating tissue inflammation and oxidative stress, thus becoming attractive potential candidates to be used in the context of nutraceutical interventions, in order to ameliorate systemic inflammation in aging subjects. In this review, we aim to summarize the available data about the anti-inflammatory properties of oleuropein and hydroxytyrosol, discussing them in the light of molecular pathways involved in the synthesis and release of inflammatory mediators in inflammaging.
Collapse
|
10
|
New Trends in Aging Drug Discovery. Biomedicines 2022; 10:biomedicines10082006. [PMID: 36009552 PMCID: PMC9405986 DOI: 10.3390/biomedicines10082006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Aging is considered the main risk factor for many chronic diseases that frequently appear at advanced ages. However, the inevitability of this process is being questioned by recent research that suggests that senescent cells have specific features that differentiate them from younger cells and that removal of these cells ameliorates senescent phenotype and associated diseases. This opens the door to the design of tailored therapeutic interventions aimed at reducing and delaying the impact of senescence in life, that is, extending healthspan and treating aging as another chronic disease. Although these ideas are still far from reaching the bedside, it is conceivable that they will revolutionize the way we understand aging in the next decades. In this review, we analyze the main and well-validated cellular pathways and targets related to senescence as well as their implication in aging-associated diseases. In addition, the most relevant small molecules with senotherapeutic potential, with a special emphasis on their mechanism of action, ongoing clinical trials, and potential limitations, are discussed. Finally, a brief overview of alternative strategies that go beyond the small molecule field, together with our perspectives for the future of the field, is provided.
Collapse
|
11
|
Vidović T, Ewald CY. Longevity-Promoting Pathways and Transcription Factors Respond to and Control Extracellular Matrix Dynamics During Aging and Disease. FRONTIERS IN AGING 2022; 3:935220. [PMID: 35874275 PMCID: PMC9301135 DOI: 10.3389/fragi.2022.935220] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/27/2022] [Indexed: 05/28/2023]
Abstract
Aging is one of the largest risk factors for cancer, type 2 diabetes, osteoarthritis, cardiovascular diseases, and other age-related pathologies. Here, we give a detailed description of the interplay of chronic age-related pathologies with the remodeling of the extracellular matrix during disease development and progression. Longevity-promoting signaling pathways slow or prevent age-related diseases. In particular, we focus on the mTOR signaling pathway, sirtuins, and canonical longevity-promoting transcription factors, such as FOXO, NF-κB, and Nrf2. We extend our analysis using chromatin immunoprecipitation (ChIP) sequencing and transcriptomic data and report that many established and emerging longevity-promoting transcription factors, such as CREB1, FOXO1,3, GATA1,2,3,4, HIF1A, JUN, KLF4, MYC, NFE2L2/Nrf2, RELA/NF-κB, REST, STAT3,5A, and TP53/p53, directly regulate many extracellular matrix genes and remodelers. We propose that modulation of these pathways increases lifespan and protects from age-related diseases in part due to their effects on extracellular matrix remodeling. Therefore, to successfully treat age-related diseases, it is necessary to better understand the connection between extracellular matrix components and longevity pathways.
Collapse
Affiliation(s)
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
12
|
Tumor-Associated Inflammation: The Tumor-Promoting Immunity in the Early Stages of Tumorigenesis. J Immunol Res 2022; 2022:3128933. [PMID: 35733919 PMCID: PMC9208911 DOI: 10.1155/2022/3128933] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Tumorigenesis is a multistage progressive oncogenic process caused by alterations in the structure and expression level of multiple genes. Normal cells are continuously endowed with new capabilities in this evolution, leading to subsequent tumor formation. Immune cells are the most important components of inflammation, which is closely associated with tumorigenesis. There is a broad consensus in cancer research that inflammation and immune response facilitate tumor progression, infiltration, and metastasis via different mechanisms; however, their protumor effects are equally important in tumorigenesis at earlier stages. Previous studies have demonstrated that during the early stages of tumorigenesis, certain immune cells can promote the formation and proliferation of premalignant cells by inducing DNA damage and repair inhibition, releasing trophic/supporting signals, promoting immune escape, and activating inflammasomes, as well as enhance the characteristics of cancer stem cells. In this review, we focus on the potential mechanisms by which immune cells can promote tumor initiation and promotion in the early stages of tumorigenesis; furthermore, we discuss the interaction of the inflammatory environment and protumor immune cells with premalignant cells and cancer stem cells, as well as the possibility of early intervention in tumor formation by targeting these cellular mechanisms.
Collapse
|
13
|
MG132 Induces Progerin Clearance and Improves Disease Phenotypes in HGPS-like Patients’ Cells. Cells 2022; 11:cells11040610. [PMID: 35203262 PMCID: PMC8870437 DOI: 10.3390/cells11040610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Progeroid syndromes (PS), including Hutchinson-Gilford Progeria Syndrome (HGPS), are premature and accelerated aging diseases, characterized by clinical features mimicking physiological aging. Most classical HGPS patients carry a de novo point mutation within exon 11 of the LMNA gene encoding A-type lamins. This mutation activates a cryptic splice site, leading to the production of a truncated prelamin A, called prelamin A ∆50 or progerin, that accumulates in HGPS cell nuclei and is a hallmark of the disease. Some patients with PS carry other LMNA mutations and are named “HGPS-like” patients. They produce progerin and/or other truncated prelamin A isoforms (∆35 and ∆90). We previously found that MG132, a proteasome inhibitor, induced progerin clearance in classical HGPS through autophagy activation and splicing regulation. Here, we show that MG132 induces aberrant prelamin A clearance and improves cellular phenotypes in HGPS-like patients’ cells other than those previously described in classical HGPS. These results provide preclinical proof of principle for the use of a promising class of molecules toward a potential therapy for children with HGPS-like or classical HGPS.
Collapse
|
14
|
Shalaby YM, Al Aidaros A, Valappil A, Ali BR, Akawi N. Role of Ceramides in the Molecular Pathogenesis and Potential Therapeutic Strategies of Cardiometabolic Diseases: What we Know so Far. Front Cell Dev Biol 2022; 9:816301. [PMID: 35127726 PMCID: PMC8808480 DOI: 10.3389/fcell.2021.816301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
Ceramides represent a class of biologically active lipids that are involved in orchestrating vital signal transduction pathways responsible for regulating cellular differentiation and proliferation. However, accumulating clinical evidence have shown that ceramides are playing a detrimental role in the pathogenesis of several diseases including cardiovascular disease, type II diabetes and obesity, collectively referred to as cardiometabolic disease. Therefore, it has become necessary to study in depth the role of ceramides in the pathophysiology of such diseases, aiming to tailor more efficient treatment regimens. Furthermore, understanding the contribution of ceramides to the pathological molecular mechanisms of those interrelated conditions may improve not only the therapeutic but also the diagnostic and preventive approaches of the preceding hazardous events. Hence, the purpose of this article is to review currently available evidence on the role of ceramides as a common factor in the pathological mechanisms of cardiometabolic diseases as well as the mechanism of action of the latest ceramides-targeted therapies.
Collapse
Affiliation(s)
- Youssef M Shalaby
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Egypt
| | - Anas Al Aidaros
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Anjana Valappil
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Nadia Akawi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Zhang L, Zhao J, Mu X, McGowan SJ, Angelini L, O'Kelly RD, Yousefzadeh MJ, Sakamoto A, Aversa Z, LeBrasseur NK, Suh Y, Huard J, Kamenecka TM, Niedernhofer LJ, Robbins PD. Novel small molecule inhibition of IKK/NF-κB activation reduces markers of senescence and improves healthspan in mouse models of aging. Aging Cell 2021; 20:e13486. [PMID: 34734460 PMCID: PMC8672781 DOI: 10.1111/acel.13486] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
Constitutive NF-κB activation is associated with cellular senescence and stem cell dysfunction and rare variants in NF-κB family members are enriched in centenarians. We recently identified a novel small molecule (SR12343) that inhibits IKK/NF-κB activation by disrupting the association between IKKβ and NEMO. Here we investigated the therapeutic effects of SR12343 on senescence and aging in three different mouse models. SR12343 reduced senescence-associated beta-galactosidase (SA-β-gal) activity in oxidative stress-induced senescent mouse embryonic fibroblasts as well as in etoposide-induced senescent human IMR90 cells. Chronic administration of SR12343 to the Ercc1-/∆ and Zmpste24-/- mouse models of accelerated aging reduced markers of cellular senescence and SASP and improved multiple parameters of aging. SR12343 also reduced markers of senescence and increased muscle fiber size in 2-year-old WT mice. Taken together, these results demonstrate that IKK/NF-κB signaling pathway represents a promising target for reducing markers of cellular senescence, extending healthspan and treating age-related diseases.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Biochemistry, Molecular Biology and BiophysicsInstitute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Jing Zhao
- Department of Molecular MedicineScripps ResearchJupiterFloridaUSA
| | - Xiaodong Mu
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
- Shandong First Medical University (Shandong Academy of Medical Sciences)JinanChina
| | - Sara J. McGowan
- Department of Biochemistry, Molecular Biology and BiophysicsInstitute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Luise Angelini
- Department of Biochemistry, Molecular Biology and BiophysicsInstitute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Ryan D. O'Kelly
- Department of Biochemistry, Molecular Biology and BiophysicsInstitute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Matthew J. Yousefzadeh
- Department of Biochemistry, Molecular Biology and BiophysicsInstitute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Ayumi Sakamoto
- Department of Physical Medicine and Rehabilitation and Robert and Arlene Kogod Center on AgingRochesterMinnesotaUSA
| | - Zaira Aversa
- Department of Physical Medicine and Rehabilitation and Robert and Arlene Kogod Center on AgingRochesterMinnesotaUSA
| | - Nathan K. LeBrasseur
- Department of Physical Medicine and Rehabilitation and Robert and Arlene Kogod Center on AgingRochesterMinnesotaUSA
| | - Yousin Suh
- Department of Genetics and DevelopmentColumbia UniversityNew YorkNew YorkUSA
| | - Johnny Huard
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | | | - Laura J. Niedernhofer
- Department of Biochemistry, Molecular Biology and BiophysicsInstitute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Paul D. Robbins
- Department of Biochemistry, Molecular Biology and BiophysicsInstitute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
16
|
Thoms HC, Stark LA. The NF-κB Nucleolar Stress Response Pathway. Biomedicines 2021; 9:biomedicines9091082. [PMID: 34572268 PMCID: PMC8471347 DOI: 10.3390/biomedicines9091082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/09/2021] [Accepted: 08/20/2021] [Indexed: 12/20/2022] Open
Abstract
The nuclear organelle, the nucleolus, plays a critical role in stress response and the regulation of cellular homeostasis. P53 as a downstream effector of nucleolar stress is well defined. However, new data suggests that NF-κB also acts downstream of nucleolar stress to regulate cell growth and death. In this review, we will provide insight into the NF-κB nucleolar stress response pathway. We will discuss apoptosis mediated by nucleolar sequestration of RelA and new data demonstrating a role for p62 (sequestosome (SQSTM1)) in this process. We will also discuss activation of NF-κB signalling by degradation of the RNA polymerase I (PolI) complex component, transcription initiation factor-IA (TIF-IA (RRN3)), and contexts where TIF-IA-NF-κB signalling may be important. Finally, we will discuss how this pathway is targeted by aspirin to mediate apoptosis of colon cancer cells.
Collapse
|
17
|
Role of NF-κB in Ageing and Age-Related Diseases: Lessons from Genetically Modified Mouse Models. Cells 2021; 10:cells10081906. [PMID: 34440675 PMCID: PMC8394846 DOI: 10.3390/cells10081906] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/17/2021] [Accepted: 07/23/2021] [Indexed: 12/21/2022] Open
Abstract
Ageing is a complex process, induced by multifaceted interaction of genetic, epigenetic, and environmental factors. It is manifested by a decline in the physiological functions of organisms and associated to the development of age-related chronic diseases and cancer development. It is considered that ageing follows a strictly-regulated program, in which some signaling pathways critically contribute to the establishment and maintenance of the aged state. Chronic inflammation is a major mechanism that promotes the biological ageing process and comorbidity, with the transcription factor NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) as a crucial mediator of inflammatory responses. This, together with the finding that the activation or inhibition of NF-κB can induce or reverse respectively the main features of aged organisms, has brought it under consideration as a key transcription factor that acts as a driver of ageing. In this review, we focused on the data obtained entirely through the generation of knockout and transgenic mouse models of either protein involved in the NF-κB signaling pathway that have provided relevant information about the intricate processes or molecular mechanisms that control ageing. We have reviewed the relationship of NF-κB and premature ageing; the development of cancer associated with ageing and the implication of NF-κB activation in the development of age-related diseases, some of which greatly increase the risk of developing cancer.
Collapse
|
18
|
Xiao G, Zhang X, Zhang X, Chen Y, Xia Z, Cao H, Huang J, Cheng Q. Aging-related genes are potential prognostic biomarkers for patients with gliomas. Aging (Albany NY) 2021; 13:13239-13263. [PMID: 33946049 PMCID: PMC8148480 DOI: 10.18632/aging.203008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/04/2021] [Indexed: 04/25/2023]
Abstract
Aging has a significant role in the proliferation and development of cancers. This study explored the expression profiles, prognostic value, and potential roles of aging-related genes in gliomas. We designed risk score and cluster models based on aging-related genes and glioma cases using LASSO Cox regression analysis, consensus clustering analysis and univariate cox regression analyses. High risk score was related to malignant clinical features and poor prognosis based on 10 datasets, 2953 cases altogether. Genetic alterations analysis revealed that high risk scores were associated with genomic aberrations of aging-related oncogenes. GSVA analysis exhibited the potential function of the aging-related genes. More immune cell infiltration was found in high-risk group cases, and glioma patients in high-risk group may be more responsive to immunotherapy. Knock-down of CTSC, an aging-related gene, can inhibit cell cycle progression, colony formation, cell proliferation and increase cell senescence in glioma cell lines in vitro. Indeed, high expression of CTSC was associated with poor prognosis in glioma cases. In conclusion, this study revealed that aging-related genes have prognostic potential for glioma patients and further identified potential mechanisms for aging-related genes in tumorigenesis and progression in gliomas.
Collapse
Affiliation(s)
- Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
| | - Xiangyang Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Hunan, China
| | - Xun Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
| | - Yuanbing Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Changsha, Hunan, China
| | - Hui Cao
- Department of Psychiatry, The Second People’s Hospital of Hunan Province, Hunan, China
- The Hospital of Hunan University of Chinese Medicine, Hunan, China
| | - Jun Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| |
Collapse
|
19
|
FK866 Protects Human Dental Pulp Cells against Oxidative Stress-Induced Cellular Senescence. Antioxidants (Basel) 2021; 10:antiox10020271. [PMID: 33578781 PMCID: PMC7916510 DOI: 10.3390/antiox10020271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/12/2023] Open
Abstract
FK866 possesses various functional properties, such as anti-angiogenic, anti-cancer, and anti-inflammatory activities. We previously demonstrated that premature senescence of human dental pulp cells (hDPCs) was induced by hydrogen peroxide (H2O2). The present study aimed to investigate whether H2O2-induced premature senescence of hDPCs is affected by treatment with FK866. We found that FK866 markedly inhibited the senescent characteristics of hDPCs after exposure to H2O2, as revealed by an increase in the number of senescence-associated β-galactosidase (SA-β-gal)-positive hDPCs and the upregulation of the p21 and p53 proteins, which acts as molecular indicators of cellular senescence. Moreover, the stimulatory effects of H2O2 on cellular senescence are associated with oxidative stress induction, such as excessive ROS production and NADPH consumption, telomere DNA damage induction, and upregulation of senescence-associated secretory phenotype factors (IL-1β, IL-6, IL-8, COX-2, and TNF-α) as well as NF-κB activation, which were all blocked by FK866. Thus, FK866 might antagonize H2O2-induced premature senescence of hDPCs, acting as a potential therapeutic antioxidant by attenuating oxidative stress-induced pathologies in dental pulp, including inflammation and cellular senescence.
Collapse
|
20
|
Gatto N, Dos Santos Souza C, Shaw AC, Bell SM, Myszczynska MA, Powers S, Meyer K, Castelli LM, Karyka E, Mortiboys H, Azzouz M, Hautbergue GM, Márkus NM, Shaw PJ, Ferraiuolo L. Directly converted astrocytes retain the ageing features of the donor fibroblasts and elucidate the astrocytic contribution to human CNS health and disease. Aging Cell 2021; 20:e13281. [PMID: 33314575 PMCID: PMC7811849 DOI: 10.1111/acel.13281] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/20/2020] [Accepted: 10/31/2020] [Indexed: 12/17/2022] Open
Abstract
Astrocytes are highly specialised cells, responsible for CNS homeostasis and neuronal activity. Lack of human in vitro systems able to recapitulate the functional changes affecting astrocytes during ageing represents a major limitation to studying mechanisms and potential therapies aiming to preserve neuronal health. Here, we show that induced astrocytes from fibroblasts donors in their childhood or adulthood display age‐related transcriptional differences and functionally diverge in a spectrum of age‐associated features, such as altered nuclear compartmentalisation, nucleocytoplasmic shuttling properties, oxidative stress response and DNA damage response. Remarkably, we also show an age‐related differential response of induced neural progenitor cells derived astrocytes (iNPC‐As) in their ability to support neurons in co‐culture upon pro‐inflammatory stimuli. These results show that iNPC‐As are a renewable, readily available resource of human glia that retain the age‐related features of the donor fibroblasts, making them a unique and valuable model to interrogate human astrocyte function over time in human CNS health and disease.
Collapse
Affiliation(s)
- Noemi Gatto
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Cleide Dos Santos Souza
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Allan C. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Simon M. Bell
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Monika A. Myszczynska
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Samantha Powers
- The Research institute Nationwide Children’s Hospital Columbus OH USA
| | - Kathrin Meyer
- The Research institute Nationwide Children’s Hospital Columbus OH USA
| | - Lydia M. Castelli
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Evangelia Karyka
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Guillaume M. Hautbergue
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Nóra M. Márkus
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| |
Collapse
|
21
|
Checa J, Aran JM. Reactive Oxygen Species: Drivers of Physiological and Pathological Processes. J Inflamm Res 2020; 13:1057-1073. [PMID: 33293849 PMCID: PMC7719303 DOI: 10.2147/jir.s275595] [Citation(s) in RCA: 423] [Impact Index Per Article: 84.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/10/2020] [Indexed: 12/11/2022] Open
Abstract
Since the Great Oxidation Event, about 2.4 billion years ago, the Earth is immersed in an oxidizing atmosphere. Thus, it has been proposed that excess oxygen, originally a waste product of photosynthetic cyanobacteria, induced oxidative stress and the production of reactive oxygen species (ROS), which have since acted as fundamental drivers of biologic evolution and eukaryogenesis. Indeed, throughout an organism’s lifespan, ROS affect directly (as mutagens) or indirectly (as messengers and regulators) all structural and functional components of cells, and many aspects of cell biology. Whether left unchecked by protective antioxidant systems, excess ROS not only cause genomic mutations but also induce irreversible oxidative modification of proteins (protein oxidation and peroxidation), lipids and glycans (advanced lipoxidation and glycation end products), impairing their function and promoting disease or cell death. Conversely, low-level local ROS play an important role both as redox-signaling molecules in a wide spectrum of pathways involved in the maintenance of cellular homeostasis (MAPK/ERK, PTK/PTP, PI3K-AKT-mTOR), and regulating key transcription factors (NFκB/IκB, Nrf2/KEAP1, AP-1, p53, HIF-1). Consequently, ROS can shape a variety of cellular functions, including proliferation, differentiation, migration and apoptosis. In this review, we will give a brief overview of the relevance of ROS in both physiological and pathological processes, particularly inflammation and aging. In-depth knowledge of the molecular mechanisms of ROS actuation and their influence under steady-state and stressful conditions will pave the way for the development of novel therapeutic interventions. This will mitigate the harmful outcomes of ROS in the onset and progression of a variety of chronic inflammatory and age-related diseases.
Collapse
Affiliation(s)
- Javier Checa
- Immune-Inflammatory Processes and Gene Therapeutics Group, IDIBELL, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Josep M Aran
- Immune-Inflammatory Processes and Gene Therapeutics Group, IDIBELL, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona 08907, Spain
| |
Collapse
|
22
|
Giller A, Andrawus M, Gutman D, Atzmon G. Pregnancy as a model for aging. Ageing Res Rev 2020; 62:101093. [PMID: 32502628 DOI: 10.1016/j.arr.2020.101093] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 04/21/2020] [Accepted: 05/23/2020] [Indexed: 12/12/2022]
Abstract
The process of aging can be defined as the sum accumulation of damages and changes in metabolism during the life of an organism, due to both genetic predisposition and stochastic damage. During the gestational period and following parturition, similar damage can be seen due to the strenuous effect on the maternal body, exhibited on both the physiological and cellular level. In this review, we will focus on the similar physiological and cellular characteristics exhibited during pregnancy and aging, including induction of and response to oxidative stress, inflammation, and degradation of telomeres. We will evaluate any similar processes between aging and pregnancy by comparing common biomarkers, pathologies, and genetic and epigenetic effects, to establish the pregnant body as a model for aging. This review will approach the connection both in respect to current theories on aging as a byproduct of natural selection, and regarding unrelated biochemical similarities between the two, drawing on existing studies and models in humans and other species where relevant alike. Furthermore, we will show the response of the pregnant body to these changes, and through that illuminate unique areas of potential study to advance our knowledge of the maladies relating to aging and pregnancy, and an avenue for solutions.
Collapse
Affiliation(s)
- Abram Giller
- Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Ave., Mount Carmel, Haifa, 349888, Israel
| | - Mariana Andrawus
- Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Ave., Mount Carmel, Haifa, 349888, Israel
| | - Danielle Gutman
- Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Ave., Mount Carmel, Haifa, 349888, Israel
| | - Gil Atzmon
- Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Ave., Mount Carmel, Haifa, 349888, Israel; Departments of Genetics and Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, New York, 10461, USA.
| |
Collapse
|
23
|
Analysis of the intricate effects of polyunsaturated fatty acids and polyphenols on inflammatory pathways in health and disease. Food Chem Toxicol 2020; 143:111558. [PMID: 32640331 PMCID: PMC7335494 DOI: 10.1016/j.fct.2020.111558] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/16/2020] [Accepted: 06/24/2020] [Indexed: 02/08/2023]
Abstract
Prevention and treatment of non-communicable diseases (NCDs), including cardiovascular disease, diabetes, obesity, cancer, Alzheimer's and Parkinson's disease, arthritis, non-alcoholic fatty liver disease and various infectious diseases; lately most notably COVID-19 have been in the front line of research worldwide. Although targeting different organs, these pathologies have common biochemical impairments - redox disparity and, prominently, dysregulation of the inflammatory pathways. Research data have shown that diet components like polyphenols, poly-unsaturated fatty acids (PUFAs), fibres as well as lifestyle (fasting, physical exercise) are important factors influencing signalling pathways with a significant potential to improve metabolic homeostasis and immune cells' functions. In the present manuscript we have reviewed scientific data from recent publications regarding the beneficial cellular and molecular effects induced by dietary plant products, mainly polyphenolic compounds and PUFAs, and summarize the clinical outcomes expected from these types of interventions, in a search for effective long-term approaches to improve the immune system response.
Collapse
|
24
|
Buchanan S, Combet E, Stenvinkel P, Shiels PG. Klotho, Aging, and the Failing Kidney. Front Endocrinol (Lausanne) 2020; 11:560. [PMID: 32982966 PMCID: PMC7481361 DOI: 10.3389/fendo.2020.00560] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
Klotho has been recognized as a gene involved in the aging process in mammals for over 30 years, where it regulates phosphate homeostasis and the activity of members of the fibroblast growth factor (FGF) family. The α-Klotho protein is the receptor for Fibroblast Growth Factor-23 (FGF23), regulating phosphate homeostasis and vitamin D metabolism. Phosphate toxicity is a hallmark of mammalian aging and correlates with diminution of Klotho levels with increasing age. As such, modulation of Klotho activity is an attractive target for therapeutic intervention in the diseasome of aging; in particular for chronic kidney disease (CKD), where Klotho has been implicated directly in the pathophysiology. A range of senotherapeutic strategies have been developed to directly or indirectly influence Klotho expression, with varying degrees of success. These include administration of exogenous Klotho, synthetic and natural Klotho agonists and indirect approaches, via modulation of the foodome and the gut microbiota. All these approaches have significant potential to mitigate loss of physiological function and resilience accompanying old age and to improve outcomes within the diseasome of aging.
Collapse
Affiliation(s)
- Sarah Buchanan
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Emilie Combet
- School of Medicine, Dentistry & Nursing, Human Nutrition, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Peter Stenvinkel
- Division of Renal Medicine M99, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Paul G. Shiels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
25
|
Song X, Lin NH, Wang YL, Chen B, Wang HX, Hu K. Comprehensive transcriptome analysis based on RNA sequencing identifies critical genes for lipopolysaccharide-induced epididymitis in a rat model. Asian J Androl 2020; 21:605-611. [PMID: 31044753 PMCID: PMC6859662 DOI: 10.4103/aja.aja_21_19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Epididymitis is a commonly diagnosed disease associated with male infertility. However, little is known about the molecules that are involved in its development. This study was to identify critical genes associated with lipopolysaccharide-induced epididymitis and analyze the molecular mechanism of epididymitis through RNA sequencing. Experimental epididymitis models were generated by administering male Sprague–Dawley rats' lipopolysaccharide. A total of 1378 differentially expressed genes, including 531 upregulated and 847 downregulated genes, were identified in the epididymitis model rats compared with those in sham-operated rats by RNA sequencing. Functional enrichment analyses suggested that the upregulated genes were markedly enriched in inflammation-related biological processes, as well as in the tumor necrosis factor (TNF) signaling pathway, cytokine–cytokine receptor interactions, complement and coagulation cascades, and in the chemokine signaling pathway. Four downregulated genes (collagen type XXVIII alpha 1 chain [Col28α1], cyclin-dependent kinase-like 1 [Cdkl1], phosphoserine phosphatase [Psph], and fatty acid desaturase 2 [Fads2]) and ten upregulated genes (CCAAT/enhancer-binding protein beta [Cebpβ], C-X-C motif chemokine receptor 2 [Cxcr2], interleukin 11 [Il11], C-C motif chemokine ligand 20 [Ccl20], nuclear factor-kappa-B inhibitor alpha [Nfkbiα], claudin 4 [Cldn4], matrix metallopeptidase 9 [Mmp9], heat shock 70 kDa protein 8 [Hspa8], intercellular cell adhesion molecule-1 [Icam1], and Jun) were successfully confirmed by real-time polymerase chain reaction. Western blot demonstrated that CDKL1 was decreased, while MMP9 and NFKBIA were increased in the experimental model group compared with those in the sham-operated group. Our study sheds new light on the understanding of the early response of the epididymis during bacterial epididymitis.
Collapse
Affiliation(s)
- Xin Song
- Department of Urology, Shanghai Institute of Andrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Nan-He Lin
- Department of Urology, Shanghai Institute of Andrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - You-Lin Wang
- Department of Urology, Shanghai Institute of Andrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Bin Chen
- Department of Urology, Shanghai Institute of Andrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Hong-Xiang Wang
- Department of Urology, Shanghai Institute of Andrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Kai Hu
- Department of Urology, Shanghai Institute of Andrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| |
Collapse
|
26
|
Long-term every-other-day administration of DMAMCL has little effect on aging and age-associated physiological decline in mice. Aging (Albany NY) 2020; 11:2583-2609. [PMID: 31048563 PMCID: PMC6535057 DOI: 10.18632/aging.101932] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/22/2019] [Indexed: 12/19/2022]
Abstract
The activation of transcription factor NF-κB is currently identified as one of the driving forces to the aging process. Genetic impairment of NF-κB signaling pathway or pharmacological inhibition of NF-κB activity has been shown to extend healthspan and lifespan in animal models, and delay or reduce many age-related symptoms. However, the aging intervention strategies based on NF-κB inhibition by the suitable small molecular compound is currently still lacking. The water-soluble dimethylaminomicheliolide (DMAMCL), can inhibit NF-κB activity and is currently undergoing clinical trials. In this study, we showed that 15 months of DMAMCL administration started in 1-year old male mice was well-tolerated and safe, and improved or had little effect on some age-associated symptoms, such as neurobehavioral phenotypes, physical performance, cardiac function, hematological parameters, immune aging phenotypes, clinical chemistry parameters, and glucose homeostasis. At the molecular level, DMAMCL administration mitigated serum levels of several age-associated inflammatory cytokines, including IL-6, IL-1α, IL-1β, TNF-α, IFN-γ, and CXCL2, and inhibited NF-κB activity in several aged tissues. Collectively, our results indicate that current strategy of DMAMCL administration may has little effect on aging process in mice, and provide basic clues to further exploit the possibility of DMAMCL-based aging intervention to promote healthy aging.
Collapse
|
27
|
Zhang GZ, Deng YJ, Xie QQ, Ren EH, Ma ZJ, He XG, Gao YC, Kang XW. Sirtuins and intervertebral disc degeneration: Roles in inflammation, oxidative stress, and mitochondrial function. Clin Chim Acta 2020; 508:33-42. [PMID: 32348785 DOI: 10.1016/j.cca.2020.04.016] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
Intervertebral disc degeneration (IDD) is one of the main causes of low back pain, which seriously reduces the quality of life of patients and places a heavy economic burden on their families. Cellular senescence is considered to be an important factor leading to IDD, and inflammatory response, oxidative stress, and mitochondrial dysfunction are closely related to intervertebral disc (IVD) senescence. Therefore, inhibition of the inflammatory response and oxidative stress, along with maintaining mitochondrial function, may be useful in treating IDD. The sirtuins are a family of evolutionarily conserved nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, which are the major molecules mediating life extension or delay of aging-related diseases. The sirtuin protein family consist of seven members (SIRT1 - 7), which are mainly involved in various aging-related diseases by regulating inflammation, oxidative stress, and mitochondrial function. Among them, SIRT1, SIRT2, SIRT3, and SIRT6 are closely related to IDD. In addition, some activators of sirtuin proteins, such as resveratrol, melatonin, magnolol, 1,4-dihydropyridine (DHP), SRT1720, and nicotinamide mononucleotide (NMN), have been evaluated in preclinical studies for their effects in preventing IDD. This review described the biological functions of sirtuins and the important roles of SIRT1, SIRT2, SIRT3, and SIRT6 in IDD by regulating oxidative stress, inflammatory response, and mitochondrial function. In addition, we introduce the status of some sirtuin activators in IDD preclinical studies. This review will provide a background for further clarification of the molecular mechanism underlying IDD and the development of potential therapeutic drugs.
Collapse
Affiliation(s)
- Guang-Zhi Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Ya-Jun Deng
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Qi-Qi Xie
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - En-Hui Ren
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Zhan-Jun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Xue-Gang He
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Yi-Cheng Gao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Xue-Wen Kang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China; Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, PR China; The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Gansu 730000, PR China.
| |
Collapse
|
28
|
Ebert T, Pawelzik SC, Witasp A, Arefin S, Hobson S, Kublickiene K, Shiels PG, Bäck M, Stenvinkel P. Inflammation and Premature Ageing in Chronic Kidney Disease. Toxins (Basel) 2020; 12:E227. [PMID: 32260373 PMCID: PMC7232447 DOI: 10.3390/toxins12040227] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/20/2020] [Accepted: 03/29/2020] [Indexed: 02/06/2023] Open
Abstract
Persistent low-grade inflammation and premature ageing are hallmarks of the uremic phenotype and contribute to impaired health status, reduced quality of life, and premature mortality in chronic kidney disease (CKD). Because there is a huge global burden of disease due to CKD, treatment strategies targeting inflammation and premature ageing in CKD are of particular interest. Several distinct features of the uremic phenotype may represent potential treatment options to attenuate the risk of progression and poor outcome in CKD. The nuclear factor erythroid 2-related factor 2 (NRF2)-kelch-like erythroid cell-derived protein with CNC homology [ECH]-associated protein 1 (KEAP1) signaling pathway, the endocrine phosphate-fibroblast growth factor-23-klotho axis, increased cellular senescence, and impaired mitochondrial biogenesis are currently the most promising candidates, and different pharmaceutical compounds are already under evaluation. If studies in humans show beneficial effects, carefully phenotyped patients with CKD can benefit from them.
Collapse
Affiliation(s)
- Thomas Ebert
- Karolinska Institutet, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, SE-141 86 Stockholm, Sweden; (A.W.); (S.A.); (S.H.); (K.K.)
| | - Sven-Christian Pawelzik
- Karolinska Institutet, Department of Medicine Solna, Cardiovascular Medicine Unit, SE-171 76 Stockholm, Sweden; (S.-C.P.); (M.B.)
- Karolinska University Hospital, Theme Heart and Vessels, Division of Valvular and Coronary Disease, SE-171 76 Stockholm, Sweden
| | - Anna Witasp
- Karolinska Institutet, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, SE-141 86 Stockholm, Sweden; (A.W.); (S.A.); (S.H.); (K.K.)
| | - Samsul Arefin
- Karolinska Institutet, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, SE-141 86 Stockholm, Sweden; (A.W.); (S.A.); (S.H.); (K.K.)
| | - Sam Hobson
- Karolinska Institutet, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, SE-141 86 Stockholm, Sweden; (A.W.); (S.A.); (S.H.); (K.K.)
| | - Karolina Kublickiene
- Karolinska Institutet, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, SE-141 86 Stockholm, Sweden; (A.W.); (S.A.); (S.H.); (K.K.)
| | - Paul G. Shiels
- University of Glasgow, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, Institute of Cancer Sciences, Glasgow G61 1QH, UK;
| | - Magnus Bäck
- Karolinska Institutet, Department of Medicine Solna, Cardiovascular Medicine Unit, SE-171 76 Stockholm, Sweden; (S.-C.P.); (M.B.)
- Karolinska University Hospital, Theme Heart and Vessels, Division of Valvular and Coronary Disease, SE-171 76 Stockholm, Sweden
| | - Peter Stenvinkel
- Karolinska Institutet, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, SE-141 86 Stockholm, Sweden; (A.W.); (S.A.); (S.H.); (K.K.)
| |
Collapse
|
29
|
Wang D, Liu S, Xu S. Identification of hub genes, key pathways, and therapeutic agents in Hutchinson-Gilford Progeria syndrome using bioinformatics analysis. Medicine (Baltimore) 2020; 99:e19022. [PMID: 32049798 PMCID: PMC7035007 DOI: 10.1097/md.0000000000019022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Hutchinson-Gilford Progeria syndrome (HGPS) is a rare lethal premature and accelerated aging disease caused by mutations in the lamin A/C gene. Nevertheless, the mechanisms of cellular damage, senescence, and accelerated aging in HGPS are not fully understood. Therefore, we aimed to screen potential key genes, pathways, and therapeutic agents of HGPS by using bioinformatics methods in this study. METHODS The gene expression profile of GSE113648 and GSE41751 were retrieved from the gene expression omnibus database and analyzed to identify the differentially expressed genes (DEGs) between HGPS and normal controls. Then, gene ontology and the Kyoto encyclopedia of genes and genomes pathway enrichment analysis were carried out. To construct the protein-protein interaction (PPI) network, we used STRING and Cytoscape to make module analysis of these DEGs. Besides, the connectivity map (cMAP) tool was used as well to predict potential drugs. RESULTS As a result, 180 upregulated DEGs and 345 downregulated DEGs were identified, which were significantly enriched in pathways in cancer and PI3K-Akt signaling pathway. The top centrality hub genes fibroblast growth factor 2, decorin, matrix metallopeptidase2, and Fos proto-oncogene, AP-1 transcription factor subunit were screened out as the critical genes among the DEGs from the PPI network. Dexibuprofen and parthenolide were predicted to be the possible agents for the treatment of HGPS by cMAP analysis. CONCLUSION This study identified key genes, signal pathways and therapeutic agents, which might help us improve our understanding of the mechanisms of HGPS and identify some new therapeutic agents for HGPS.
Collapse
Affiliation(s)
- Dengchuan Wang
- Office of Medical Ethics, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong
| | - Shengshuo Liu
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Shi Xu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
30
|
Saban-Ruiz J, Ly-Pen D. COVID-19: A Personalized Cardiometabolic Approach for Reducing Complications and Costs. The Role of Aging beyond Topics. J Nutr Health Aging 2020; 24:550-559. [PMID: 32510105 PMCID: PMC7217344 DOI: 10.1007/s12603-020-1385-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/03/2020] [Indexed: 02/07/2023]
Abstract
COVID 19 is much more than an infectious disease by SARS-CoV-2 followed by a disproportionate immune response. An older age, diabetes and history of cardiovascular disease, especially hypertension, but also chronic heart failure and coronary artery disease among others, are between the most important risk factors. In addition, during the hospitalization both hyperglycaemia and heart failure are frequent. Less frequent are acute coronary syndrome, arrhythmias and stroke. Accordingly, not all prolonged stays or even deaths are due directly to SARS-CoV-2. To our knowledge, this is the first review, focusing both on cardiovascular and metabolic aspects of this dreadful disease, in an integrated and personalized way, following the guidelines of the Cardiometabolic Health/Medicine. Therefore, current personalized aspects such as ACEIs and ARBs, the place of statins and the most appropriate management of heart failure in diabetics are analysed. Aging, better than old age, as a dynamic process, is also considered in this review for the first time in the literature, and not only as a risk factor attributed to cardiovascular and non-cardiovascular comorbidities. Immunosenescence is also approached to build healthier elders, so they can resist present and future infectious diseases, and not only in epidemics or pandemics. In addition, to do this we must start knowing the molecular mechanisms that underlying Aging process in general, and immunosenescence in particular. Surprisingly, the endoplasmic reticulum stress and autophagy are implicated in both process. Finally, with a training in all the aspects covered in this review, not only the hospital stay, complications and costs of this frightening disease in high-risk population should be reduced. Likely, this paper will open a gate to the future for open-minded physicians.
Collapse
Affiliation(s)
- J Saban-Ruiz
- Prof. Saban-Ruiz, MD, PhD, d-médical Clinic, Madrid, Spain,
| | | |
Collapse
|
31
|
Liu C, Arnold R, Henriques G, Djabali K. Inhibition of JAK-STAT Signaling with Baricitinib Reduces Inflammation and Improves Cellular Homeostasis in Progeria Cells. Cells 2019; 8:cells8101276. [PMID: 31635416 PMCID: PMC6829898 DOI: 10.3390/cells8101276] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/05/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS), a rare premature aging disorder that leads to death at an average age of 14.7 years due to myocardial infarction or stroke, is caused by mutations in the LMNA gene. Nearly 90% of HGPS cases carry the G608G mutation within exon 11 that generates a truncated prelamin A protein “progerin”. Progerin accumulates in HGPS cells and induces premature senescence at the cellular and organismal levels. Children suffering from HGPS develop numerous clinical features that overlap with normal aging, including atherosclerosis, arthritis, hair loss and lipodystrophy. To determine whether an aberrant signaling pathway might underlie the development of these four diseases (atherosclerosis, arthritis, hair loss and lipodystrophy), we performed a text mining analysis of scientific literature and databases. We found a total of 17 genes associated with all four pathologies, 14 of which were linked to the JAK1/2-STAT1/3 signaling pathway. We report that the inhibition of the JAK-STAT pathway with baricitinib, a Food and Drug Administration-approved JAK1/2 inhibitor, restored cellular homeostasis, delayed senescence and decreased proinflammatory markers in HGPS cells. Our ex vivo data using human cell models indicate that the overactivation of JAK-STAT signaling mediates premature senescence and that the inhibition of this pathway could show promise for the treatment of HGPS and age-related pathologies.
Collapse
Affiliation(s)
- Chang Liu
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM school of Medicine, Technical University of Munich (TUM), 85748 Garching, Germany.
| | - Rouven Arnold
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM school of Medicine, Technical University of Munich (TUM), 85748 Garching, Germany.
| | - Gonçalo Henriques
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM school of Medicine, Technical University of Munich (TUM), 85748 Garching, Germany.
| | - Karima Djabali
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM school of Medicine, Technical University of Munich (TUM), 85748 Garching, Germany.
| |
Collapse
|
32
|
Neves J, Sousa-Victor P. Regulation of inflammation as an anti-aging intervention. FEBS J 2019; 287:43-52. [PMID: 31529582 DOI: 10.1111/febs.15061] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/08/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022]
Abstract
Aging is accompanied by a decline in physiological integrity and a loss of regenerative capacity in many tissues. The development of interventions that prevent or reverse age-related disease requires a better understanding of the interplay of cell intrinsic, inter-cellular communication and systemic deregulations that underlie the aging process. Immune dysfunction and changes in inflammatory pathways are transversal contributors to the aging process and are essential propagators of tissue deterioration. Here, we propose and discuss the rejuvenation potential of interventions that target chronic inflammation and how modulation of tissue repair capacity could be an important mediator of such anti-aging strategies. We highlight how current knowledge on the systemic nature of inflammatory dysregulation in old organisms, together with the development of new animal models that allow for the isolation of the inflammatory component of aging, could provide new targets for interventions in aging based on the modulation of inflammatory pathways.
Collapse
Affiliation(s)
- Joana Neves
- Faculdade de Medicina, Instituto de Medicina Molecular (iMM), Universidade de Lisboa, Portugal
| | - Pedro Sousa-Victor
- Faculdade de Medicina, Instituto de Medicina Molecular (iMM), Universidade de Lisboa, Portugal
| |
Collapse
|
33
|
Gachon F, Yeung J, Naef F. Cross-regulatory circuits linking inflammation, high-fat diet, and the circadian clock. Genes Dev 2019; 32:1359-1360. [PMID: 30385518 PMCID: PMC6217737 DOI: 10.1101/gad.320911.118] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this outlook, Gachon et al. discuss the findings of the study by Hong et al., in which the authors research the question of how acute and chronic inflammation interacts with the circadian clock. Mammalian physiology resonates with the daily changes in the external environment, allowing processes such as rest–activity cycles, metabolism, and body temperature to synchronize with daily changes in the surroundings. Studies have identified the molecular underpinnings of robust oscillations in gene expression occurring over the 24-h day, but how acute or chronic perturbations modulate gene expression rhythms, physiology, and behavior is still relatively unknown. In this issue of Genes & Development, Hong and colleagues (pp. 1367–1379) studied how acute and chronic inflammation interacts with the circadian clock. They found that NF-κB signaling can modify chromatin states and modulate expression of genes in the core clock network as well as circadian locomotor behavior. Interestingly, a high-fat diet (HFD) fed to mice also triggers this inflammation pathway, suggesting that cross-regulatory circuits link inflammation, HFD, and the circadian clock.
Collapse
Affiliation(s)
- Frédéric Gachon
- The Institute of Bioengineering (IBI), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Jake Yeung
- The Institute of Bioengineering (IBI), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Felix Naef
- The Institute of Bioengineering (IBI), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
34
|
Chen J, Stark LA. Insights into the Relationship between Nucleolar Stress and the NF-κB Pathway. Trends Genet 2019; 35:768-780. [PMID: 31434627 DOI: 10.1016/j.tig.2019.07.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
The nuclear organelle the nucleolus and the transcription factor nuclear factor of κ-light-chain-enhancer of activated B cells (NF-κB) are both central to the control of cellular homeostasis, dysregulated in common diseases and implicated in the ageing process. Until recently, it was believed that they acted independently to regulate homeostasis in health and disease. However, there is an emerging body of evidence suggesting that nucleoli and NF-κB signalling converge at multiple levels. Here we will review current understanding of this crosstalk. We will discuss activation of the NF-κB pathway by nucleolar stress and induction of apoptosis by nucleolar sequestration of NF-κB/RelA. We will also discuss the role of TIF-IA, COMMD1, and nucleophosmin, which are key players in this crosstalk, and the therapeutic relevance, particularly with respect to the antitumour effects of aspirin.
Collapse
Affiliation(s)
- Jingyu Chen
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Road, Edinburgh, Scotland EH4 2XU, UK
| | - Lesley A Stark
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Road, Edinburgh, Scotland EH4 2XU, UK.
| |
Collapse
|
35
|
Wang Y, Fan X, Xing L, Tian F. Wnt signaling: a promising target for osteoarthritis therapy. Cell Commun Signal 2019; 17:97. [PMID: 31420042 PMCID: PMC6697957 DOI: 10.1186/s12964-019-0411-x] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease worldwide and a leading cause of disability. Characterized by degradation of articular cartilage, synovial inflammation, and changes in periarticular and subchondral bone, OA can negatively impact an individual's physical and mental well-being. Recent studies have reported several critical signaling pathways as key regulators and activators of cellular and molecular processes during OA development. Wnt signaling is one such pathway whose signaling molecules and regulators were shown to be abnormally activated or suppressed. As such, agonists and antagonists of those molecules are potential candidates for OA treatment. Notably, a recent phase I clinical trial (NCT02095548) demonstrated the potential of SM04690, a small-molecule inhibitor of the Wnt signaling pathway, as a disease-modifying oseoarthritis drug (DMOAD). This review summarizes the role and mechanism of Wnt signaling and related molecules in regulating OA progression, with a view to accelerating the translation of such evidence into the development of strategies for OA treatment, particularly with respect to potential applications of molecules targeting the Wnt signaling pathway.
Collapse
Affiliation(s)
- Yudan Wang
- Medical Research Center, North China University of Science and Technology, Bohai Road 21, Caofeidian Dis, Tangshan, Hebei 063210 People’s Republic of China
| | - Xinhao Fan
- Department of Stomatology, Kailuan General Hospital, Tangshan, Hebei 063000 People’s Republic of China
| | - Lei Xing
- Department of Geriatrics, Affiliated hospital of North China University of Science and Technology, Jianshe South Road 57, Tangshan, Hebei 063000 People’s Republic of China
| | - Faming Tian
- Medical Research Center, North China University of Science and Technology, Bohai Road 21, Caofeidian Dis, Tangshan, Hebei 063210 People’s Republic of China
| |
Collapse
|
36
|
Chen J, Lobb IT, Morin P, Novo SM, Simpson J, Kennerknecht K, von Kriegsheim A, Batchelor EE, Oakley F, Stark LA. Identification of a novel TIF-IA-NF-κB nucleolar stress response pathway. Nucleic Acids Res 2019; 46:6188-6205. [PMID: 29873780 PMCID: PMC6158704 DOI: 10.1093/nar/gky455] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022] Open
Abstract
p53 as an effector of nucleolar stress is well defined, but p53 independent mechanisms are largely unknown. Like p53, the NF-κB transcription factor plays a critical role in maintaining cellular homeostasis under stress. Many stresses that stimulate NF-κB also disrupt nucleoli. However, the link between nucleolar function and activation of the NF-κB pathway is as yet unknown. Here we demonstrate that artificial disruption of the PolI complex stimulates NF-κB signalling. Unlike p53 nucleolar stress response, this effect does not appear to be linked to inhibition of rDNA transcription. We show that specific stress stimuli of NF-κB induce degradation of a critical component of the PolI complex, TIF-IA. This degradation precedes activation of NF-κB and is associated with increased nucleolar size. It is mimicked by CDK4 inhibition and is dependent upon a novel pathway involving UBF/p14ARF and S44 of the protein. We show that blocking TIF-IA degradation blocks stress effects on nucleolar size and NF-κB signalling. Finally, using ex vivo culture, we show a strong correlation between degradation of TIF-IA and activation of NF-κB in freshly resected, human colorectal tumours exposed to the chemopreventative agent, aspirin. Together, our study provides compelling evidence for a new, TIF-IA-NF-κB nucleolar stress response pathway that has in vivo relevance and therapeutic implications.
Collapse
Affiliation(s)
- Jingyu Chen
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| | - Ian T Lobb
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| | - Pierre Morin
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| | - Sonia M Novo
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| | - James Simpson
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| | - Kathrin Kennerknecht
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| | - Alex von Kriegsheim
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| | - Emily E Batchelor
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| | - Fiona Oakley
- Liver Research Group, Institute of Cellular Medicine, 4th Floor, William Leech Building, Framlington Place, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
| | - Lesley A Stark
- University of Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Rd., Edinburgh EH4 2XU, UK
| |
Collapse
|
37
|
Surai PF, Kochish II, Fisinin VI, Juniper DT. Revisiting Oxidative Stress and the Use of Organic Selenium in Dairy Cow Nutrition. Animals (Basel) 2019; 9:E462. [PMID: 31331084 PMCID: PMC6680431 DOI: 10.3390/ani9070462] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/16/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023] Open
Abstract
In commercial animals production, productive stress can negatively impact health status and subsequent productive and reproductive performance. A great body of evidence has demonstrated that as a consequence of productive stress, an overproduction of free radicals, disturbance of redox balance/signaling, and oxidative stress were observed. There is a range of antioxidants that can be supplied with animal feed to help build and maintain the antioxidant defense system of the body responsible for prevention of the damaging effects of free radicals and the toxic products of their metabolism. Among feed-derived antioxidants, selenium (Se) was shown to have a special place as an essential part of 25 selenoproteins identified in animals. There is a comprehensive body of research in monogastric species that clearly shows that Se bioavailability within the diet is very much dependent on the form of the element used. Organic Se, in the form of selenomethionine (SeMet), has been reported to be a much more effective Se source when compared with mineral forms such as sodium selenite or selenate. It has been proposed that one of the main advantages of organic Se in pig and poultry nutrition is the non-specific incorporation of SeMet into general body proteins, thus forming an endogenous Se reserve that can be utilized during periods of stress for additional synthesis of selenoproteins. Responses in ruminant species to supplementary Se tend to be much more variable than those reported in monogastric species, and much of this variability may be a consequence of the different fates of Se forms in the rumen following ingestion. It is likely that the reducing conditions found in the rumen are responsible for the markedly lower assimilation of inorganic forms of Se, thus predisposing selenite-fed animals to potential Se inadequacy that may in turn compromise animal health and production. A growing body of evidence demonstrates that organic Se has a number of benefits, particularly in dairy and beef animals; these include improved Se and antioxidant status and better Se transfer via the placenta, colostrum, and milk to the newborn. However, there is a paucity in the data concerning molecular mechanisms of SeMet assimilation, metabolism and selenoprotein synthesis regulation in ruminant animals, and as such, further investigation is required.
Collapse
Affiliation(s)
- Peter F Surai
- Department of Microbiology and Biochemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria.
- Moscow State Academy of Veterinary Medicine and Biotechnology Named after K.I. Skryabin, 109472 Moscow, Russia.
- Department of Animal Nutrition, Faculty of Agricultural and Environmental Sciences, Szent Istvan University, H-2103 Godollo, Hungary.
| | - Ivan I Kochish
- Moscow State Academy of Veterinary Medicine and Biotechnology Named after K.I. Skryabin, 109472 Moscow, Russia
| | - Vladimir I Fisinin
- All-Russian Institute of Poultry Husbandry, 141311 Sergiev Posad, Russia
| | - Darren T Juniper
- Animal, Dairy, Food Chain Sciences, School of Agriculture, Policy and Development, University of Reading, Earley Gate, Reading RG6 6AR, UK
| |
Collapse
|
38
|
Peilin W, Songsong T, Chengyu Z, Zhi C, Chunhui M, Yinxian Y, Lei Z, Min M, Zongyi W, Mengkai Y, Jing X, Tao Z, Zhuoying W, Fei Y, Chengqing Y. Directed elimination of senescent cells attenuates development of osteoarthritis by inhibition of c-IAP and XIAP. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2618-2632. [PMID: 31251987 DOI: 10.1016/j.bbadis.2019.05.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/28/2019] [Accepted: 05/27/2019] [Indexed: 01/02/2023]
Abstract
Aging drives the accumulation of senescent cells (SnCs) by secreting factors that cause the senescence-associated secretory phenotype (SASP), including stem cells in the bone marrow, which contribute to aging-related bone degradation. Osteoarthritis (OA) is a serious chronic injury disease, and increasing age is a major risk factor. The accumulation of SnCs may accelerate the development of OA, and the accumulation of SnCs may benefit from its resistance to apoptotic stimuli. Therefore, local elimination of SnCs could be a promising treatment for OA. Apoptosis inhibitor protein (IAP) is an important antiapoptotic protein in vivo. AT-406 is a small molecule inhibitor of the IAP genes and also regulates the transcription of several genes. Here, we show that SnCs upregulate the antiapoptotic proteins c-IAP1, c-IAP2 and XIAP.The combined inhibition of c-IAP1, c-IAP2 and XIAP using siRNA or AT-406 specifically induce the apoptosis of SnCs.In addition, XIAP and STX17 bind to each other to regulate the fusion of autophagosomes and lysosomes in SnCs, which in turn, affects the fate of SnCs. It is worth noting that the clearance of SnCs attenuated the secretion of SASP and created a proregenerative environment. Most importantly, local clearance of SnCs significantly attenuated the progression of osteoarthritis in rats without significant toxic effects. Thus, local elimination of SnCs may be a potential treatment for OA. This is the first report of inhibition of IAPs for clearing SnCs and suggests that eradication of SnCs may be a new strategy for the treatment of age-related diseases.
Collapse
Affiliation(s)
- Wang Peilin
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China
| | - Teng Songsong
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuang Chengyu
- Department of Orthopaedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cui Zhi
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China
| | - Ma Chunhui
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Yinxian
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Lei
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China
| | - Mao Min
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China
| | - Wang Zongyi
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China.
| | - Yang Mengkai
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China
| | - Xu Jing
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China
| | - Zhang Tao
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China.
| | - Wang Zhuoying
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China
| | - Yin Fei
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institute, Shanghai, China
| | - Yi Chengqing
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
39
|
Jęśko H, Stępień A, Lukiw WJ, Strosznajder RP. The Cross-Talk Between Sphingolipids and Insulin-Like Growth Factor Signaling: Significance for Aging and Neurodegeneration. Mol Neurobiol 2019; 56:3501-3521. [PMID: 30140974 PMCID: PMC6476865 DOI: 10.1007/s12035-018-1286-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022]
Abstract
Bioactive sphingolipids: sphingosine, sphingosine-1-phosphate (S1P), ceramide, and ceramide-1-phosphate (C1P) are increasingly implicated in cell survival, proliferation, differentiation, and in multiple aspects of stress response in the nervous system. The opposite roles of closely related sphingolipid species in cell survival/death signaling is reflected in the concept of tightly controlled sphingolipid rheostat. Aging has a complex influence on sphingolipid metabolism, disturbing signaling pathways and the properties of lipid membranes. A metabolic signature of stress resistance-associated sphingolipids correlates with longevity in humans. Moreover, accumulating evidence suggests extensive links between sphingolipid signaling and the insulin-like growth factor I (IGF-I)-Akt-mTOR pathway (IIS), which is involved in the modulation of aging process and longevity. IIS integrates a wide array of metabolic signals, cross-talks with p53, nuclear factor κB (NF-κB), or reactive oxygen species (ROS) and influences gene expression to shape the cellular metabolic profile and stress resistance. The multiple connections between sphingolipids and IIS signaling suggest possible engagement of these compounds in the aging process itself, which creates a vulnerable background for the majority of neurodegenerative disorders.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Pawińskiego, 5, 02-106, Poland
| | - Adam Stępień
- Central Clinical Hospital of the Ministry of National Defense, Department of Neurology, Military Institute of Medicine, Warsaw, Szaserów, 128, 04-141, Poland
| | - Walter J Lukiw
- LSU Neuroscience Center and Departments of Neurology and Ophthalmology, Louisiana State University School of Medicine, New Orleans, USA
| | - Robert P Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Pawińskiego, 5, 02-106, Poland.
| |
Collapse
|
40
|
Ishida T, Ishida M, Tashiro S, Takeishi Y. DNA Damage and Senescence-Associated Inflammation in Cardiovascular Disease. Biol Pharm Bull 2019; 42:531-537. [DOI: 10.1248/bpb.b18-00865] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Mari Ishida
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Satoshi Tashiro
- Department of Cellular Biology, Research Institute of Radiation Biology and Medicine, Hiroshima University
| | | |
Collapse
|
41
|
Gene variants in the NF-KB pathway (NFKB1, NFKBIA, NFKBIZ) and risk for early-onset coronary artery disease. Immunol Lett 2019; 208:39-43. [PMID: 30902734 DOI: 10.1016/j.imlet.2019.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/13/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023]
Abstract
The nuclear-factor kappa-beta (NF-KB) is a driver of inflammation, and plays an important role in the pathogenesis of atherosclerosis and coronary artery disease (CAD). Early-onset CAD is defined as a coronary ischaemic episode at an age ≤55 years, and in our population was strongly associated with male sex and smoking. Our aim was to determine whether common variants in three NF-KB genes were associated with early-onset CAD. We studied 609 patients with early-onset CAD and 423 healthy controls, all male. Allele and genotype frequencies for the NFKB1 rs28362491 (-94 delATTG) and NFKBIA rs8904 were not significantly different between the two groups. For the NFKBIZ rs3217713, the deletion allele was significantly more frequent in the patients than in controls (0.27 vs. 0.22; p = 0.004). Deletion-carriers were more frequent in the patients (p < 0.001), with an OR = 1.48 (95%CI = 1.15-1.90). We performed a multiple logistic regression (linear generalized model) with smoking, hypercholesterolemia, type 2 diabetes, hypertension, and the rs3217713 deletion carriers remained significantly associated with early-onset CAD (p = 0.01). In our population, the NFKBIZ variant was an independent risk factor for developing early-onset CAD.
Collapse
|
42
|
Gypenoside L Inhibits Proliferation of Liver and Esophageal Cancer Cells by Inducing Senescence. Molecules 2019; 24:molecules24061054. [PMID: 30889805 PMCID: PMC6471500 DOI: 10.3390/molecules24061054] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/15/2022] Open
Abstract
Senescence is an irreversible state of cell cycle arrest that can be triggered by multiple stimuli, such as oxygen reactive species and DNA damage. Growing evidence has proven that senescence is a tumor-suppressive approach in cancer treatment. Therefore, developing novel agents that modulate senescence may be an alternative strategy against cancer. In our study, we investigated the inhibitory effect of gypenoside L (Gyp-L), a saponin isolated from Gynostemma pentaphyllum, on cancer cell growth. We found that Gyp-L increased the SA-β-galactosidase activity, promoted the production of senescence-associated secretory cytokines, and inhibited cell proliferation of human liver and esophageal cancer cells. Moreover, Gyp-L caused cell cycle arrest at S phase, and activated senescence-related cell cycle inhibitor proteins (p21 and p27) and their upstream regulators. In addition, Gyp-L activated p38 and ERK MAPK pathways and NF-κB pathway to induce senescence. Consistently, adding chemical inhibitors efficiently counteracted the Gyp-L-mediated senescence, growth inhibition, and cell cycle arrest in cancer cells. Furthermore, treatment with Gyp-L, enhanced the cytotoxicity of clinic therapeutic drugs, including 5-fluorouracil and cisplatin, on cancer cells. Overall, these results indicate that Gyp-L inhibits proliferation of cancer cells by inducing senescence and renders cancer cells more sensitive to chemotherapy.
Collapse
|
43
|
Alameda JP, Ramírez Á, García-Fernández RA, Navarro M, Page A, Segovia JC, Sanchez R, Suárez-Cabrera C, Paramio JM, Bravo A, Fernández-Aceñero MJ, Casanova ML. Premature aging and cancer development in transgenic mice lacking functional CYLD. Aging (Albany NY) 2019; 11:127-159. [PMID: 30631004 PMCID: PMC6339805 DOI: 10.18632/aging.101732] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022]
Abstract
CYLD is a deubiquitinating enzyme known for its role as a tumor suppressor whose mutation leads to skin appendages tumors and other cancers. In this manuscript we report that the tumor suppressor CYLD, similarly to other renowned tumor suppressor genes, protects from premature aging and cancer. We have generated transgenic mice expressing the mutant CYLDC/S protein, lacking its deubiquitinase function, under the control of the keratin 5 promoter, the K5-CYLDC/S mice. These mice express the transgene in different organs, including those considered to be more susceptible to aging, such as skin and thymus. Our results show that K5-CYLDC/S mice exhibit epidermal, hair follicle, and sebaceous gland alterations; and, importantly, they show signs of premature aging from an early age. Typically, 3-month-old K5-CYLDC/S mice exhibit a phenotype characterized by alopecia and kyphosis, and, the histological examination reveals that transgenic mice show signs of accelerated aging in numerous organs such as skin, thymus, pancreas, liver and lung. Additionally, they spontaneously develop tumors of diverse origin. Over-activation of the NF-κB pathway, along with hyperactivation of Akt, JNK and c-Myc, and chronic inflammation, appear as the mechanisms responsible for the premature aging of the K5-CYLDC/S mice.
Collapse
Affiliation(s)
- Josefa P. Alameda
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/CIBERONC, 28040 Madrid, Spain
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
| | - Ángel Ramírez
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/CIBERONC, 28040 Madrid, Spain
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
| | | | - Manuel Navarro
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/CIBERONC, 28040 Madrid, Spain
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
| | - Angustias Page
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/CIBERONC, 28040 Madrid, Spain
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
| | - José C. Segovia
- Division of Hematopoietic Innovative Therapies, CIEMAT/CIBERER/II-FJD, 28040 Madrid, Spain
| | - Rebeca Sanchez
- Division of Hematopoietic Innovative Therapies, CIEMAT/CIBERER/II-FJD, 28040 Madrid, Spain
| | - Cristian Suárez-Cabrera
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/CIBERONC, 28040 Madrid, Spain
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
| | - Jesús M. Paramio
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/CIBERONC, 28040 Madrid, Spain
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
| | - Ana Bravo
- Department of Anatomy, Animal Production and Veterinary Clinical Sciences, Faculty of Veterinary Medicine, University of Santiago de Compostela, Lugo, Spain
| | - M. Jesús Fernández-Aceñero
- Servicio de Anatomía Patológica Hospital Clínico San Carlos, Departamento de Anatomía Patológica, Facultad de Medicina, UCM, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC),
28040 Madrid, España
| | - M. Llanos Casanova
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/CIBERONC, 28040 Madrid, Spain
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
| |
Collapse
|
44
|
Zhang L, Zhao J, Gurkar A, Niedernhofer LJ, Robbins PD. Methods to Quantify the NF-κB Pathway During Senescence. Methods Mol Biol 2019; 1896:231-250. [PMID: 30474851 DOI: 10.1007/978-1-4939-8931-7_18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Nuclear factor κB (NF-κB) is a family of transcription factors important for regulating innate and adaptive immunity, cellular proliferation, apoptosis and senescence. The NF-κB family is comprised of five subunits, RelA/p65, RelB, C-Rel, p50 (p105/NF-κB1), and p52 (p100/NF-κB2). NF-κB activity goes up with age in multiple tissues. The two subunits RelA/p65 and p50 have been implicated in senescence and aging with genetic deletion of p65 and p50 reducing or increasing senescence respectively. Pharmacologic inhibition of NF-κB also extends health span and reduces senescence in mouse models of accelerated aging. In addition, NF-κB regulates expression of many of senescence associated secretory phenotype (SASP) factors released by certain types of senescent cells that drives loss of tissue homeostasis and secondary senescence. To measure NF-κB activity with aging in vivo, multiple methods can and need to be utilized including cellular localization of p65, EMSA analysis of NF-κB DNA binding, RNA in situ hybridization, and analysis of expression of NF-κB target genes. To colocalize NF-κB activation and senescence, p65 localization or transcriptional activity can be measured by immunostaining or RNA in situ hybridization for NF-κB regulated genes along with methods such as immunostaining for γH2AX or RNA in situ for senescence markers like p16INK4a and p21. These and related methods will be described in this chapter.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Molecular Medicine and Center on Aging, The Scripps Research Institute, Jupiter, FL, USA
| | - Jing Zhao
- Department of Molecular Medicine and Center on Aging, The Scripps Research Institute, Jupiter, FL, USA
- Disease Biology and Cellular Pharmacology, Recursion Pharmaceuticals, Salt Lake, UT, USA
| | - Aditi Gurkar
- Department of Molecular Medicine and Center on Aging, The Scripps Research Institute, Jupiter, FL, USA
- Aging Institute, Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Laura J Niedernhofer
- Department of Molecular Medicine and Center on Aging, The Scripps Research Institute, Jupiter, FL, USA
| | - Paul D Robbins
- Department of Molecular Medicine and Center on Aging, The Scripps Research Institute, Jupiter, FL, USA.
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
45
|
Zhang L, Yousefzadeh MJ, Suh Y, Niedernhofer LJ, Robbins PD. Signal Transduction, Ageing and Disease. Subcell Biochem 2019; 91:227-247. [PMID: 30888655 DOI: 10.1007/978-981-13-3681-2_9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Ageing is defined by the loss of functional reserve over time, leading to a decreased tissue homeostasis and increased age-related pathology. The accumulation of damage including DNA damage contributes to driving cell signaling pathways that, in turn, can drive different cell fates, including senescence and apoptosis, as well as mitochondrial dysfunction and inflammation. In addition, the accumulation of cell autonomous damage with time also drives ageing through non-cell autonomous pathways by modulation of signaling pathways. Interestingly, genetic and pharmacologic analysis of factors able to modulate lifespan and healthspan in model organisms and even humans have identified several key signaling pathways including IGF-1, NF-κB, FOXO3, mTOR, Nrf-2 and sirtuins. This review will discuss the roles of several of these key signaling pathways, in particular NF-κB and Nrf2, in modulating ageing and age-related diseases.
Collapse
Affiliation(s)
- Lei Zhang
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Yousin Suh
- Departments of Genetics and Medicine and the Institute for Ageing Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
46
|
Sharif U, Mahmud NM, Kay P, Yang YC, Harding SP, Grierson I, Kamalden TA, Jackson MJ, Paraoan L. Advanced glycation end products-related modulation of cathepsin L and NF-κB signalling effectors in retinal pigment epithelium lead to augmented response to TNFα. J Cell Mol Med 2018; 23:405-416. [PMID: 30338926 PMCID: PMC6307775 DOI: 10.1111/jcmm.13944] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/07/2018] [Accepted: 09/10/2018] [Indexed: 01/02/2023] Open
Abstract
The retinal pigment epithelium (RPE) plays a central role in neuroretinal homoeostasis throughout life. Altered proteolysis and inflammatory processes involving RPE contribute to the pathophysiology of age‐related macular degeneration (AMD), but the link between these remains elusive. We report for the first time the effect of advanced glycation end products (AGE)—known to accumulate on the ageing RPE's underlying Bruch's membrane in situ—on both key lysosomal cathepsins and NF‐κB signalling in RPE. Cathepsin L activity and NF‐κB effector levels decreased significantly following 2‐week AGE exposure. Chemical cathepsin L inhibition also decreased total p65 protein levels, indicating that AGE‐related change of NF‐κB effectors in RPE cells may be modulated by cathepsin L. However, upon TNFα stimulation, AGE‐exposed cells had significantly higher ratio of phospho‐p65(Ser536)/total p65 compared to non‐AGEd controls, with an even higher fold increase than in the presence of cathepsin L inhibition alone. Increased proportion of active p65 indicates an AGE‐related activation of NF‐κB signalling in a higher proportion of cells and/or an enhanced response to TNFα. Thus, NF‐κB signalling modulation in the AGEd environment, partially regulated via cathepsin L, is employed by RPE cells as a protective (para‐inflammatory) mechanism but renders them more responsive to pro‐inflammatory stimuli.
Collapse
Affiliation(s)
- Umar Sharif
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Nur Musfirah Mahmud
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.,Eye Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Paul Kay
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Yit C Yang
- Ophthalmology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Simon P Harding
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Ian Grierson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | - Malcolm J Jackson
- Department of Musculoskeletal Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Luminita Paraoan
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| |
Collapse
|
47
|
Crosstalk between NF-κB and Nucleoli in the Regulation of Cellular Homeostasis. Cells 2018; 7:cells7100157. [PMID: 30301139 PMCID: PMC6210184 DOI: 10.3390/cells7100157] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 12/30/2022] Open
Abstract
Nucleoli are emerging as key sensors of cellular stress and regulators of the downstream consequences on proliferation, metabolism, senescence, and apoptosis. NF-κB signalling is activated in response to a similar plethora of stresses, which leads to modulation of cell growth and death programs. While nucleolar and NF-κB pathways are distinct, it is increasingly apparent that they converge at multiple levels. Exposure of cells to certain insults causes a specific type of nucleolar stress that is characterised by degradation of the PolI complex component, TIF-IA, and increased nucleolar size. Recent studies have shown that this atypical nucleolar stress lies upstream of cytosolic IκB degradation and NF-κB nuclear translocation. Under these stress conditions, the RelA component of NF-κB accumulates within functionally altered nucleoli to trigger a nucleophosmin dependent, apoptotic pathway. In this review, we will discuss these points of crosstalk and their relevance to anti-tumour mechanism of aspirin and small molecule CDK4 inhibitors. We will also briefly the discuss how crosstalk between nucleoli and NF-κB signalling may be more broadly relevant to the regulation of cellular homeostasis and how it may be exploited for therapeutic purpose.
Collapse
|
48
|
Hou A, Chen P, Tang H, Meng H, Cheng X, Wang Y, Zhang Y, Peng J. Cellular senescence in osteoarthritis and anti-aging strategies. Mech Ageing Dev 2018; 175:83-87. [DOI: 10.1016/j.mad.2018.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 08/03/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023]
|
49
|
Laucho‐Contreras ME, Polverino F, Rojas‐Quintero J, Wang X, Owen CA. Club cell protein 16 (Cc16) deficiency increases inflamm-aging in the lungs of mice. Physiol Rep 2018; 6:e13797. [PMID: 30084231 PMCID: PMC6079172 DOI: 10.14814/phy2.13797] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 06/26/2018] [Indexed: 12/21/2022] Open
Abstract
Low serum CC16 levels are associated with accelerated lung function decline in human population studies, but it is not known whether low serum CC16 levels contribute to lung function decline, or are an epiphenomenon. We tested the hypothesis that unchallenged Cc16-/- mice develop accelerated rates of pulmonary function test abnormalities and pulmonary pathologies over time compared with unchallenged WT mice. Respiratory mechanics, airspace enlargement, and small airway fibrosis were measured in unchallenged wild-type (WT) versus Cc16-/- mice over 6-18 months of age. Lung leukocyte counts and lung levels of metalloproteinases (Mmps), cytokines, oxidative stress, cellular senescence markers (p19 and p21), and lung cell apoptosis, and serum C-reactive protein (CRP) levels were measured in age-matched WT versus Cc16-/- mice. Unchallenged Cc16-/- mice developed greater increases in lung compliance, airspace enlargement, and small airway fibrosis than age-matched WT mice over 6-18 months of age. Cc16-/- mice had greater: (1) lung leukocyte counts; (2) lung levels of Ccl2, Ccl-5, interleukin-10, Mmp-2, and Mmp-9; (3) pulmonary oxidative stress levels, (4) alveolar septal cell apoptosis and staining for p16 and p21; and (5) serum CRP levels. Unchallenged Cc16-/- mice had greater nuclear factor-κB (NF-κB) activation in their lungs than age-matched WT mice, but similar lung levels of secretory phospholipase-A2 activity. Cc16 deficiency in mice leads spontaneously to an accelerated lung aging phenotype with exaggerated pulmonary inflammation and COPD-like lung pathologies associated with increased activation of NF- κB in the lung. CC16 augmentation strategies may reduce lung aging in CC16-deficient individuals.
Collapse
Affiliation(s)
- Maria E. Laucho‐Contreras
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Francesca Polverino
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
- The Lovelace Respiratory Research InstituteAlbuquerqueNew Mexico
| | - Joselyn Rojas‐Quintero
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Xiaoyun Wang
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusetts
- The Lovelace Respiratory Research InstituteAlbuquerqueNew Mexico
| |
Collapse
|
50
|
Gene variants in the NF-KB pathway (NFKB1, NFKBIA, NFKBIZ) and their association with type 2 diabetes and impaired renal function. Hum Immunol 2018; 79:494-498. [DOI: 10.1016/j.humimm.2018.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 03/06/2018] [Accepted: 03/26/2018] [Indexed: 12/29/2022]
|