101
|
Estaras M, Marchena AM, Fernandez-Bermejo M, Mateos JM, Vara D, Roncero V, Salido GM, Gonzalez A. The melatonin receptor antagonist luzindole induces the activation of cellular stress responses and decreases viability of rat pancreatic stellate cells. J Appl Toxicol 2020; 40:1554-1565. [PMID: 32567733 DOI: 10.1002/jat.4018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022]
Abstract
In this study, we have examined the effects of luzindole, a melatonin receptor-antagonist, on cultured pancreatic stellate cells. Intracellular free-Ca2+ concentration, production of reactive oxygen species (ROS), activation of mitogen-activated protein kinases (MAPK), endoplasmic reticulum stress and cell viability were analyzed. Stimulation of cells with the luzindole (1, 5, 10 and 50 μm) evoked a slow and progressive increase in intracellular free Ca2+ ([Ca2+ ]i ) towards a plateau. The effect of the compound on Ca2+ mobilization depended on the concentration used. Incubation of cells with the sarcoendoplasmic reticulum Ca2+ -ATPase inhibitor thapsigargin (1 μm), in the absence of Ca2+ in the extracellular medium, induced a transient increase in [Ca2+ ]i . In the presence of thapsigargin, the addition of luzindole to the cells failed to induce further mobilization of Ca2+ . Luzindole induced a concentration-dependent increase in ROS generation, both in the cytosol and in the mitochondria. This effect was smaller in the absence of extracellular Ca2+ . In the presence of luzindole the phosphorylation of p44/42 and p38 MAPKs was increased, whereas no changes in the phosphorylation of JNK could be noted. Moreover, the detection of the endoplasmic reticulum stress-sensor BiP was increased in the presence of luzindole. Finally, viability was decreased in cells treated with luzindole. Because cellular membrane receptors for melatonin have not been detected in pancreatic stellate cells, we conclude that luzindole could exert direct effects that are not mediated through its action on melatonin membrane receptors.
Collapse
Affiliation(s)
- Matias Estaras
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | - Ana M Marchena
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | | | - Jose M Mateos
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Caceres, Spain
| | - Daniel Vara
- Department of Gastroenterology, San Pedro de Alcantara Hospital, Caceres, Spain
| | - Vicente Roncero
- Unit of Histology and Pathological Anatomy, Veterinary Faculty, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| | - Antonio Gonzalez
- Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres, Spain
| |
Collapse
|
102
|
Polinski JM, Kron N, Smith DR, Bodnar AG. Unique age-related transcriptional signature in the nervous system of the long-lived red sea urchin Mesocentrotus franciscanus. Sci Rep 2020; 10:9182. [PMID: 32514014 PMCID: PMC7280269 DOI: 10.1038/s41598-020-66052-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/12/2020] [Indexed: 11/23/2022] Open
Abstract
The red sea urchin, Mesocentrotus franciscanus, is one the earth’s longest-lived animals, reported to live more than 100 years with indeterminate growth, life-long reproduction and no increase in mortality rate with age. To gain insight into mechanisms associated with longevity and negligible senescence, age-related transcriptional profiles were examined in tissues of the red sea urchin. Genome-wide transcriptional profiling using RNA-Seq revealed few age-related changes in gene expression in muscle and esophagus tissue. In contrast, radial nerve showed an unexpected level of complexity with the expression of 3,370 genes significantly altered more than two-fold with age, including genes involved in nerve function, signaling, metabolism, transcriptional regulation and chromatin modification. There was an age-related upregulation in expression of genes involved in synaptogenesis, axonogenesis and neuroprotection suggesting preservation of neuronal processes with age. There was also an upregulation in expression of positive regulators and key components of the AMPK pathway, autophagy, proteasome function, and the unfolded protein response. This unique age-related gene expression profile in the red sea urchin nervous system may play a role in mitigating the detrimental effects of aging in this long-lived animal.
Collapse
Affiliation(s)
- Jennifer M Polinski
- Gloucester Marine Genomics Institute, 417 Main Street, Gloucester, MA, 01930, USA
| | - Nicholas Kron
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, 4600 Rickenbacker Causeway, Miami, FL, 33149, United States
| | - Douglas R Smith
- Gloucester Marine Genomics Institute, 417 Main Street, Gloucester, MA, 01930, USA
| | - Andrea G Bodnar
- Gloucester Marine Genomics Institute, 417 Main Street, Gloucester, MA, 01930, USA.
| |
Collapse
|
103
|
Teissier T, Boulanger E, Deramecourt V. Normal ageing of the brain: Histological and biological aspects. Rev Neurol (Paris) 2020; 176:649-660. [PMID: 32418702 DOI: 10.1016/j.neurol.2020.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 02/02/2023]
Abstract
All the hallmarks of ageing are observed in the brain, and its cells, especially neurons, are characterized by their remarkably long lifetime. Like any organ or system, the brain is exposed to ageing processes which affect molecules, cells, blood vessels, gross morphology and, uniquely for this organ, cognition. The preponderant cerebral structures are characterized by the cellular processes of neurons and glial cells and while the quantity of cerebral interstitial fluid is limited, it is now recognized as playing a crucial role in maintaining cerebral homeostasis. Most of our current knowledge of the ageing brain derives from studies of neurodegenerative disorders. It is interesting to note that common features of these disorders, like Tau, phosphoTau and amyloid peptide accumulation, can begin relatively early in life as a result of physiological ageing and are present in subclinical cases while also being used as early-stage markers of neurodegenerative diseases in progression. In this article, we review tissue and cellular modifications in the ageing brain. Commonly described macroscopic, microscopic and vascular changes that in the ageing brain are contrasted with those seen in neurodegenerative contexts. We also review the molecular changes that occur with age in the brain, such as modifications in gene expression, insulin/insulin-like growth factor 1 signalling dysfunction, post-translational protein modifications, mitochondrial dysfunction, autophagy and calcium conductance changes.
Collapse
Affiliation(s)
- T Teissier
- Inserm, université de Lille, CHU de Lille, Institut Pasteur de Lille, U1167 - RID-AGE - facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, équipe « de l'inflammation au vieillissement, 59000 Lille, France.
| | - E Boulanger
- Inserm, université de Lille, CHU de Lille, Institut Pasteur de Lille, U1167 - RID-AGE - facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, équipe « de l'inflammation au vieillissement, 59000 Lille, France; Pôle de gérontologie, CHU de Lille, 59000 Lille, France
| | - V Deramecourt
- Inserm, UMR-S 1172 « Alzheimer et Tauopathies », centre mémoire de ressources et de recherche, Labex DISTALZ, université de Lille, CHU de Lille, 59000 Lille, France; Pôle de neurologie, CHU de Lille, 59000 Lille, France
| |
Collapse
|
104
|
Malerba A, Roth F, Harish P, Dhiab J, Lu-Nguyen N, Cappellari O, Jarmin S, Mahoudeau A, Ythier V, Lainé J, Negroni E, Abgueguen E, Simonelig M, Guedat P, Mouly V, Butler-Browne G, Voisset C, Dickson G, Trollet C. Pharmacological modulation of the ER stress response ameliorates oculopharyngeal muscular dystrophy. Hum Mol Genet 2020; 28:1694-1708. [PMID: 30649389 DOI: 10.1093/hmg/ddz007] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a rare late onset genetic disease leading to ptosis, dysphagia and proximal limb muscles at later stages. A short abnormal (GCN) triplet expansion in the polyA-binding protein nuclear 1 (PABPN1) gene leads to PABPN1-containing aggregates in the muscles of OPMD patients. Here we demonstrate that treating mice with guanabenz acetate (GA), an FDA-approved antihypertensive drug, reduces the size and number of nuclear aggregates, improves muscle force, protects myofibers from the pathology-derived turnover and decreases fibrosis. GA targets various cell processes, including the unfolded protein response (UPR), which acts to attenuate endoplasmic reticulum (ER) stress. We demonstrate that GA increases both the phosphorylation of the eukaryotic translation initiation factor 2α subunit and the splicing of Xbp1, key components of the UPR. Altogether these data show that modulation of protein folding regulation is beneficial for OPMD and promote the further development of GA or its derivatives for treatment of OPMD in humans. Furthermore, they support the recent evidences that treating ER stress could be therapeutically relevant in other more common proteinopathies.
Collapse
Affiliation(s)
- Alberto Malerba
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Fanny Roth
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Pradeep Harish
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Jamila Dhiab
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Ngoc Lu-Nguyen
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Ornella Cappellari
- Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Susan Jarmin
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Alexandrine Mahoudeau
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Victor Ythier
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Jeanne Lainé
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Elisa Negroni
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | | | - Martine Simonelig
- Institute of Human Genetics, CNRS UMR9002-University of Montpellier, mRNA Regulation and Development, Montpellier, France
| | | | - Vincent Mouly
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| | - Cécile Voisset
- UMR1078 'Genetic, Functional Genomic and Biotechnologies', INSERM, EFS, Brest University, IBSAM, Brest, France
| | - George Dickson
- School of Biological Sciences, Centers of Gene and Cell Therapy and Biomedical Sciences, Royal Holloway, University of London, TW20 OEX Surrey, UK
| | - Capucine Trollet
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, 47 bd de l'Hôpital, Paris, France
| |
Collapse
|
105
|
Midorikawa R, Takakura D, Morise J, Wakazono Y, Kawasaki N, Oka S, Takamiya K. Monitoring the glycosylation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionate-type glutamate receptors using specific antibodies reveals a novel regulatory mechanism of N-glycosylation occupancy by molecular chaperones in mice. J Neurochem 2020; 153:567-585. [PMID: 31958346 DOI: 10.1111/jnc.14964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 01/11/2023]
Abstract
In the mammalian nervous system, protein N-glycosylation plays an important role in neuronal physiology. In this study, we performed a comprehensive N-glycosylation analysis of mouse GluA1, one of the major subunits of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionate type glutamate receptor, which possesses six potential N-glycosylation sites in the N-terminal domain. By mass spectrometry-based analysis, we identified the N-glycoforms and semiquantitatively determined the site-specific N-glycosylation occupancy of GluA1. In addition, only the N401-glycosylation site demonstrated incomplete N-glycosylation occupancy. Therefore, we generated a peptide antibody that specifically detects the N401-glycan-free form to precisely quantify N401-glycosylation occupancy. Using this antibody, we clarified that N401 occupancy varies between cell types and increases in an age-dependent manner in mouse forebrains. To address the regulatory mechanism of N401-glycosylation, binding proteins of GluA1 around the N401 site were screened. HSP70 family proteins, including Bip, were identified as candidates. Bip has been known as a molecular chaperone that plays a key role in protein folding in the ER (endoplasmic reticulum). To examine the involvement of Bip in N401-glycosylation, the effect of Bip over-expression on N401 occupancy was evaluated in HEK293T cells, and the results demonstrated Bip increases the N401 glycan-free form by mediating selective prolongation of its protein half-life. Taken together, we propose that the N401-glycosite of GluA1 receives a unique control of modification, and we also propose a novel N-glycosylation occupancy regulatory mechanism by Bip that might be associated with α-amino-3-hydroxy-5-methyl-4-isoxazole-propionate receptors function in the brain.
Collapse
Affiliation(s)
- Ryosuke Midorikawa
- Department of Neuroscience, University of Miyazaki Faculty of Medicine, Miyazaki, Japan
| | - Daisuke Takakura
- Project for Utilizing Glycans in the Development of Innovative Drug Discovery Technologies, Shinanomachi Research Park, Keio University School of Medicine, Tokyo, Japan
| | - Jyoji Morise
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihiko Wakazono
- Department of Neuroscience, University of Miyazaki Faculty of Medicine, Miyazaki, Japan
| | - Nana Kawasaki
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Shogo Oka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kogo Takamiya
- Department of Neuroscience, University of Miyazaki Faculty of Medicine, Miyazaki, Japan
| |
Collapse
|
106
|
Daniele JR, Higuchi-Sanabria R, Durieux J, Monshietehadi S, Ramachandran V, Tronnes SU, Kelet N, Sanchez M, Metcalf MG, Garcia G, Frankino PA, Benitez C, Zeng M, Esping DJ, Joe L, Dillin A. UPR ER promotes lipophagy independent of chaperones to extend life span. SCIENCE ADVANCES 2020; 6:eaaz1441. [PMID: 31911951 PMCID: PMC6938708 DOI: 10.1126/sciadv.aaz1441] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/05/2019] [Indexed: 05/13/2023]
Abstract
Longevity is dictated by a combination of environmental and genetic factors. One of the key mechanisms to regulate life-span extension is the induction of protein chaperones for protein homeostasis. Ectopic activation of the unfolded protein response of the endoplasmic reticulum (UPRER) specifically in neurons is sufficient to enhance organismal stress resistance and extend life span. Here, we find that this activation not only promotes chaperones but also facilitates ER restructuring and ER function. This restructuring is concomitant with lipid depletion through lipophagy. Activation of lipophagy is distinct from chaperone induction and is required for the life-span extension found in this paradigm. Last, we find that overexpression of the lipophagy component, ehbp-1, is sufficient to deplete lipids, remodel ER, and promote life span. Therefore, UPR induction in neurons triggers two distinct programs in the periphery: the proteostasis arm through protein chaperones and metabolic changes through lipid depletion mediated by EH domain binding protein 1 (EHBP-1).
Collapse
Affiliation(s)
- Joseph R. Daniele
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Ryo Higuchi-Sanabria
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Jenni Durieux
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Samira Monshietehadi
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Vidhya Ramachandran
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
- Thermo Fisher Scientific, Genetic Sciences Division, Santa Clara, CA 95051, USA
| | - Sarah U. Tronnes
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Naame Kelet
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Melissa Sanchez
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Melissa G. Metcalf
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Gilberto Garcia
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Phillip A. Frankino
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Camila Benitez
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Mandy Zeng
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Daniel J. Esping
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Larry Joe
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Andrew Dillin
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
- Corresponding author.
| |
Collapse
|
107
|
Abstract
Sleep is a universal phenomenon occurring in all species studied thus far. Sleep loss results in adverse physiological effects at both the organismal and cellular levels suggesting an adaptive role for sleep in the maintenance of overall health. This review examines the bidirectional relationship between sleep and cellular stress. Cellular stress in this review refers to a shift in cellular homeostasis in response to an external stressor. Studies that illustrate the fact that sleep loss induces cellular stress and those that provide evidence that cellular stress in turn promotes sleep will be discussed.
Collapse
Affiliation(s)
- Julie A Williams
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nirinjini Naidoo
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Division of Sleep Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
108
|
Nowakowska M, Gualtieri F, von Rüden EL, Hansmann F, Baumgärtner W, Tipold A, Potschka H. Profiling the Expression of Endoplasmic Reticulum Stress Associated Heat Shock Proteins in Animal Epilepsy Models. Neuroscience 2019; 429:156-172. [PMID: 31887356 DOI: 10.1016/j.neuroscience.2019.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
Unfolded protein response is a signaling cascade triggered by misfolded proteins in the endoplasmic reticulum. Heat shock protein H4 (HSPH4) and A5 (HSPA5) are two chaperoning proteins present within the organelle, which target misfolded peptides during prolonged stress conditions. Epileptogenic insults and epileptic seizures are a notable source of stress on cells. To investigate whether they influence expression of these chaperones, we performed immunohistochemical stainings in brains from rats that experienced a status epilepticus (SE) as a trigger of epileptogenesis and from canine epilepsy patients. Quantification of HSPA5 and HSPH4 revealed alterations in hippocampus and parahippocampal cortex. In rats, SE induced up-regulation of HSPA5 in the piriform cortex and down-regulation of HSPA5 and HSPH4 in the hippocampus. Regionally restricted increases in expression of the two proteins has been observed in the chronic phase with spontaneous recurrent seizures. Confocal microscopy revealed a predominant expression of both proteins in neurons, no expression in microglia and circumscribed expression in astroglia. In canine patients, only up-regulation of HSPH4 expression was observed in Cornu Ammonis 1 region in animals diagnosed with structural epilepsy. This characterization of HSPA5 and HSPH4 expression provided extensive information regarding spatial and temporal alterations of the two proteins during SE-induced epileptogenesis and following epilepsy manifestations. Up-regulation of both proteins implies stress exerted on ER during these disease phases. Taken together suggest a differential impact of epileptogenesis on HSPA5 and HSPH4 expression and indicate them as a possible target for pharmacological modulation of unfolded protein response.
Collapse
Affiliation(s)
- Marta Nowakowska
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University Munich, Germany
| | - Fabio Gualtieri
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University Munich, Germany
| | - Eva-Lotta von Rüden
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University Munich, Germany
| | - Florian Hansmann
- Department of Pathology, University of Veterinary Medicine Hannover, Germany
| | | | - Andrea Tipold
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University Munich, Germany.
| |
Collapse
|
109
|
Capurso C, Bellanti F, Lo Buglio A, Vendemiale G. The Mediterranean Diet Slows Down the Progression of Aging and Helps to Prevent the Onset of Frailty: A Narrative Review. Nutrients 2019; 12:nu12010035. [PMID: 31877702 PMCID: PMC7019245 DOI: 10.3390/nu12010035] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 02/05/2023] Open
Abstract
The aging population is rapidly increasing all over the world. This results in significant implications for the planning and provision of health and social care. Aging is physiologically characterized by a decrease in lean mass, bone mineral density and, to a lesser extent, fat mass. The onset of sarcopenia leads to weakness and a further decrease in physical activity. An insufficient protein intake, which we often observe in patients of advanced age, certainly accelerates the progression of sarcopenia. In addition, many other factors (e.g., insulin resistance, impaired protein digestion and absorption of amino acids) reduce the stimulation of muscle protein synthesis in the elderly, even if the protein intake is adequate. Inadequate intake of foods can also cause micronutrient deficiencies that contribute to the development of frailty. We know that a healthy eating style in middle age predisposes to so-called "healthy and successful" aging, which is the condition of the absence of serious chronic diseases or of an important decline in cognitive or physical functions, or mental health. The Mediterranean diet is recognized to be a "healthy food" dietary pattern; high adherence to this dietary pattern is associated with a lower incidence of chronic diseases and lower physical impairment in old age. The aim of our review was to analyze observational studies (cohort and case-control studies) that investigated the effects of following a healthy diet, and especially the effect of adherence to a Mediterranean diet (MD), on the progression of aging and on onset of frailty.
Collapse
|
110
|
Schinzel RT, Higuchi-Sanabria R, Shalem O, Moehle EA, Webster BM, Joe L, Bar-Ziv R, Frankino PA, Durieux J, Pender C, Kelet N, Kumar SS, Savalia N, Chi H, Simic M, Nguyen NT, Dillin A. The Hyaluronidase, TMEM2, Promotes ER Homeostasis and Longevity Independent of the UPR ER. Cell 2019; 179:1306-1318.e18. [PMID: 31761535 PMCID: PMC6913896 DOI: 10.1016/j.cell.2019.10.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/31/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022]
Abstract
Cells have evolved complex mechanisms to maintain protein homeostasis, such as the UPRER, which are strongly associated with several diseases and the aging process. We performed a whole-genome CRISPR-based knockout (KO) screen to identify genes important for cells to survive ER-based protein misfolding stress. We identified the cell-surface hyaluronidase (HAase), Transmembrane Protein 2 (TMEM2), as a potent modulator of ER stress resistance. The breakdown of the glycosaminoglycan, hyaluronan (HA), by TMEM2 within the extracellular matrix (ECM) altered ER stress resistance independent of canonical UPRER pathways but dependent upon the cell-surface receptor, CD44, a putative HA receptor, and the MAPK cell-signaling components, ERK and p38. Last, and most surprisingly, ectopic expression of human TMEM2 in C. elegans protected animals from ER stress and increased both longevity and pathogen resistance independent of canonical UPRER activation but dependent on the ERK ortholog mpk-1 and the p38 ortholog pmk-1.
Collapse
Affiliation(s)
- Robert Thomas Schinzel
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ryo Higuchi-Sanabria
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ophir Shalem
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Erica Ann Moehle
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Brant Michael Webster
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Larry Joe
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Raz Bar-Ziv
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Phillip Andrew Frankino
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jenni Durieux
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Corinne Pender
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Naame Kelet
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Saranya Santhosh Kumar
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nupur Savalia
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hannah Chi
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Milos Simic
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ngoc-Tram Nguyen
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA
| | - Andrew Dillin
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Regenerative Medicine, Berkeley, CA 94720, USA; University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
111
|
Kokubun H, Jin H, Aoe T. Pathogenic Effects of Impaired Retrieval between the Endoplasmic Reticulum and Golgi Complex. Int J Mol Sci 2019; 20:ijms20225614. [PMID: 31717602 PMCID: PMC6888596 DOI: 10.3390/ijms20225614] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular activities, such as growth and secretion, are dependent on correct protein folding and intracellular protein transport. Injury, like ischemia, malnutrition, and invasion of toxic substances, affect the folding environment in the endoplasmic reticulum (ER). The ER senses this information, following which cells adapt their response to varied situations through the unfolded protein response. Activation of the KDEL receptor, resulting from the secretion from the ER of chaperones containing the KDEL sequence, plays an important role in this adaptation. The KDEL receptor was initially shown to be necessary for the retention of KDEL sequence-containing proteins in the ER. However, it has become clear that the activated KDEL receptor also regulates bidirectional transport between the ER and the Golgi complex, as well as from the Golgi to the secretory pathway. In addition, it has been suggested that the signal for KDEL receptor activation may also affect several other cellular activities. In this review, we discuss KDEL receptor-mediated bidirectional transport and signaling and describe disease models and human diseases related to KDEL receptor dysfunction.
Collapse
Affiliation(s)
- Hiroshi Kokubun
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomohiko Aoe
- Department of Medicine, Pain Center, Chiba Medical Center, Teikyo University, Ichihara 299-0111, Japan
- Correspondence: ; Tel.: +81-436-62-1211
| |
Collapse
|
112
|
Zhao H, Ma N, Chen Q, You X, Liu C, Wang T, Yuan D, Zhang C. Decline in testicular function in ageing rats: Changes in the unfolded protein response and mitochondrial apoptotic pathway. Exp Gerontol 2019; 127:110721. [DOI: 10.1016/j.exger.2019.110721] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/03/2019] [Accepted: 09/02/2019] [Indexed: 10/26/2022]
|
113
|
4-Phenylbutyric Acid Reduces Endoplasmic Reticulum Stress in Chondrocytes That Is Caused by Loss of the Protein Disulfide Isomerase ERp57. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6404035. [PMID: 31781343 PMCID: PMC6875354 DOI: 10.1155/2019/6404035] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/01/2019] [Indexed: 01/30/2023]
Abstract
Objective The integrity of cartilage depends on the correct synthesis of extracellular matrix (ECM) components. In case of insufficient folding of proteins in the endoplasmic reticulum (ER) of chondrocytes, ECM proteins aggregate, ER stress evolves, and the unfolded protein response (UPR) is initiated. By this mechanism, chondrocytes relieve the stress condition or initiate cell death by apoptosis. Especially persistent ER stress has emerged as a pathogenic mechanism in cartilage diseases, such as chondrodysplasias and osteoarthritis. As pharmacological intervention is not available yet, it is of great interest to understand cartilage ER stress in detail and to develop therapeutics to intervene. Methods ERp57-deficient chondrocytes were generated by CRISPR/Cas9-induced KO. ER stress and autophagy were studied on mRNA and protein level as well as by transmission electron microscopy (TEM) in chondrocyte micromass or cartilage explant cultures of ERp57 KO mice. Thapsigargin (Tg), an inhibitor of the ER-residing Ca2+-ATPase, and 4-Phenylbutyric acid (4-PBA), a small molecular chemical chaperone, were applied to induce or inhibit ER stress. Results Our data reveal that the loss of the protein disulfide isomerase ERp57 is sufficient to induce ER stress in chondrocytes. 4-PBA efficiently diffuses into cartilage explant cultures and diminishes excessive ER stress in chondrocytes dose dependently, no matter if it is induced by ERp57 KO or stimulation with Tg. Conclusion ER-stress-related diseases have different sources; therefore, various targets for therapeutic treatment exist. In the future, 4-PBA may be used alone or in combination with other drugs for the treatment of ER-stress-related skeletal disorders in patients.
Collapse
|
114
|
Ahn C, Jung EM, An BS, Hong EJ, Yoo YM, Jeung EB. The Protective Role of Calbindin-D 9k on Endoplasmic Reticulum Stress-Induced Beta Cell Death. Int J Mol Sci 2019; 20:ijms20215317. [PMID: 31731478 PMCID: PMC6862009 DOI: 10.3390/ijms20215317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 11/16/2022] Open
Abstract
Intracellular calcium ion content is tightly regulated for the maintenance of cellular functions and cell survival. Calbindin-D9k (CaBP-9k) is responsible for regulating the distribution of cytosolic free-calcium ions. In this study, we aimed to investigate the effect of CaBP-9k on cell survival in pancreatic beta cells. Six-month-old wildtype CaBP-9k, CaBP-28k, and CaBP-9k/28k knockout (KO) mice were used to compare the pathological phenotypes of calcium-binding protein-deleted mice. Subsequently, the endoplasmic reticulum (ER) stress reducer tauroursodeoxycholic acid (TUDCA) was administered to wildtype and CaBP-9k KO mice. In vitro assessment of the role of CaBP-9k was performed following CaBP-9k overexpression and treatment with the ER stress inducer thapsigargin. Six-month-old CaBP-9k KO mice showed reduced islet volume and up-regulation of cell death markers resulting from ER stress, which led to pancreatic beta cell death. TUDCA treatment recovered islet volume, serum insulin level, and abdominal fat storage by CaBP-9k ablation. CaBP-9k overexpression elevated insulin secretion and recovered thapsigargin-induced ER stress in the INS-1E cell line. The results of this study show that CaBP-9k can protect pancreatic beta cell survival from ER stress and contribute to glucose homeostasis, which can reduce the risk of type 1 diabetes and provide the molecular basis for calcium supplementation to diabetic patients.
Collapse
Affiliation(s)
- Changhwan Ahn
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Korea; (C.A.); (E.-M.J.); (Y.-M.Y.)
| | - Eui-Man Jung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Korea; (C.A.); (E.-M.J.); (Y.-M.Y.)
| | - Beum-Soo An
- Department of Biomaterials Science, College of Natural Resources & Life Science, Pusan National University, Miryang 50463, Korea;
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary Medicine Bldg., Yuseong, Daejeon 34134, Korea;
| | - Yeong-Min Yoo
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Korea; (C.A.); (E.-M.J.); (Y.-M.Y.)
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Korea; (C.A.); (E.-M.J.); (Y.-M.Y.)
- Correspondence:
| |
Collapse
|
115
|
Zhang Z, Liu S, Huang S. Effects of thymosin β4 on neuronal apoptosis in a rat model of cerebral ischemia‑reperfusion injury. Mol Med Rep 2019; 20:4186-4192. [PMID: 31545437 PMCID: PMC6797993 DOI: 10.3892/mmr.2019.10683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/30/2019] [Indexed: 01/15/2023] Open
Abstract
The aim of the present study was to investigate the protective effects of thymosin β4 (Tβ4) on neuronal apoptosis in rat middle cerebral artery occlusion ischemia/reperfusion (MCAO I/R) injury, and determine the mechanisms involved in this process. Forty-eight adult male Sprague-Dawley rats were randomly divided into three groups (n=16 per group): A sham control group, an ischemia/reperfusion group (I/R group), and a Tβ4 group. The focal cerebral I/R model was established by blocking the right MCA for 2 h, followed by reperfusion for 24 h. The Zea-Longa method was used to assess neurological deficits. Cerebral infarct volume was assessed using 2,3,5-triphenyltetrazolium chloride staining, and pathological changes were observed via hematoxylin and eosin staining. The terminal dexynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay was used to detect apoptosis. The expression of glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), and caspase-12 (CASP12) protein was assessed using immunohistochemistry and western blotting 24 h after reperfusion. Infarct volume and neuronal damage in the I/R and Tβ4 groups were significantly greater than those observed in the sham group. The Zea-Longa score, neuronal apoptosis, and expression of GRP78, CHOP, and CASP12 in the I/R and Tβ4 groups were significantly higher than those reported in the sham group. However, the Longa score and neuronal apoptosis were lower in the Tβ4 group compared to the I/R group. The expression of GRP78 was significantly increased, whereas that of CHOP and CASP12 was significantly decreased in the Tβ4 group compared to the I/R group. The present data revealed that Tβ4 can inhibit neuronal apoptosis by upregulating GRP78 and downregulating CHOP and CASP12, thereby reducing cerebral I/R injury.
Collapse
Affiliation(s)
- Zhongsheng Zhang
- Department of Neurology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| | - Shuangfeng Liu
- Department of Neurology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| | - Sichun Huang
- Department of Neurology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| |
Collapse
|
116
|
Lebeau J, Saunders JM, Moraes VWR, Madhavan A, Madrazo N, Anthony MC, Wiseman RL. The PERK Arm of the Unfolded Protein Response Regulates Mitochondrial Morphology during Acute Endoplasmic Reticulum Stress. Cell Rep 2019. [PMID: 29539413 PMCID: PMC5870888 DOI: 10.1016/j.celrep.2018.02.055] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is transmitted to mitochondria and is associated with pathologic mitochondrial dysfunction in diverse diseases. The PERK arm of the unfolded protein response (UPR) protects mitochondria during ER stress through the transcriptional and translational remodeling of mitochondrial molecular quality control pathways. Here, we show that ER stress also induces dynamic remodeling of mitochondrial morphology by promoting protective stress-induced mitochondrial hyperfusion (SIMH). ER-stress-associated SIMH is regulated by the PERK arm of the UPR and activated by eIF2α phosphorylation-dependent translation attenuation. We show that PERK-regulated SIMH is a protective mechanism to prevent pathologic mitochondrial fragmentation and promote mitochondrial metabolism in response to ER stress. These results identify PERK-dependent SIMH as a protective stress-responsive mechanism that regulates mitochondrial morphology during ER stress. Furthermore, our results show that PERK integrates transcriptional and translational signaling to coordinate mitochondrial molecular and organellar quality control in response to pathologic ER insults.
Collapse
Affiliation(s)
- Justine Lebeau
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jaclyn M Saunders
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vivian W R Moraes
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Aparajita Madhavan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nicole Madrazo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mary C Anthony
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
117
|
Dietary restriction improves proteostasis and increases life span through endoplasmic reticulum hormesis. Proc Natl Acad Sci U S A 2019; 116:17383-17392. [PMID: 31413197 PMCID: PMC6717303 DOI: 10.1073/pnas.1900055116] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The endoplasmic reticulum (ER) deteriorates with age and fails to mount an effective stress response against misfolded proteins (UPRER), leading to protein folding disorders. However, preconditioning the ER using a mild ER stress (ER hormesis) can protect against future insults. We show that dietary restriction, an intervention that protects against protein misfolding disorders and increases life span across species, uses ER hormesis as a mechanism of action. Simply mimicking the ER hormesis in Caenorhabditis elegans by transient treatment with pharmacological reagents leads to delayed age-onset failure of UPRER, better capacity to process misfolded proteins, and increased life span. We also show that this process may be conserved in a mammalian cellular model of neurodegenerative disease. Unfolded protein response (UPR) of the endoplasmic reticulum (UPRER) helps maintain proteostasis in the cell. The ability to mount an effective UPRER to external stress (iUPRER) decreases with age and is linked to the pathophysiology of multiple age-related disorders. Here, we show that a transient pharmacological ER stress, imposed early in development on Caenorhabditis elegans, enhances proteostasis, prevents iUPRER decline with age, and increases adult life span. Importantly, dietary restriction (DR), that has a conserved positive effect on life span, employs this mechanism of ER hormesis for longevity assurance. We found that only the IRE-1–XBP-1 branch of UPRER is required for the longevity effects, resulting in increased ER-associated degradation (ERAD) gene expression and degradation of ER resident proteins during DR. Further, both ER hormesis and DR protect against polyglutamine aggregation in an IRE-1–dependent manner. We show that the DR-specific FOXA transcription factor PHA-4 transcriptionally regulates the genes required for ER homeostasis and is required for ER preconditioning-induced life span extension. Finally, we show that ER hormesis improves proteostasis and viability in a mammalian cellular model of neurodegenerative disease. Together, our study identifies a mechanism by which DR offers its benefits and opens the possibility of using ER-targeted pharmacological interventions to mimic the prolongevity effects of DR.
Collapse
|
118
|
Lee EJ, Cárdenes N, Álvarez D, Sellarés J, Sembrat J, Aranda P, Peng Y, Bullock J, Nouraie SM, Mora AL, Rojas M. Mesenchymal stem cells reduce ER stress via PERK-Nrf2 pathway in an aged mouse model. Respirology 2019; 25:417-426. [PMID: 31364255 DOI: 10.1111/resp.13646] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 04/01/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE Mesenchymal stem cells (MSC) have been shown to ameliorate the deleterious effects of bleomycin in murine models. However, the mechanism responsible for protection from pulmonary fibrosis by stem cell therapy is still poorly understood, especially in terms of endoplasmic reticulum (ER) stress. We hypothesized that during bleomycin-induced lung injury, markers of ER stress, specifically the activation of the unfolded protein response (UPR), increase during injury, resembling the kinetics of collagen deposition in the lung described for the bleomycin model. We aimed to elucidate the possible role of MSC in ER stress modulation. METHODS To determine the kinetics of ER stress in aged mice, the expression of ER stress markers after bleomycin lung injury was measured in old mice at different time points (days 0, 3, 7, 14 and 21). To evaluate the consequences of systemic delivery of MSC on lung ER stress in the bleomycin model, we evaluated changes in body weight, lung histology and protein expression of ER stress markers. RESULTS The level of expression of UPR transcription factor XBP-1 and its regulator BiP was elevated at day 7 and progressively increased up to day 21. MSC inhibited BiP expression in bleomycin-induced ER stress, attenuating ER stress via the protein kinase RNA-like ER kinase (PERK)-Nrf2 pathway. The expression levels of other ER stress markers were not perturbed by MSC. CONCLUSION Our data suggest that MSC operate on ER stress via several pathways, but the PERK-Nrf2 pathway revealed to be the main functioning pathway in our bleomycin model.
Collapse
Affiliation(s)
- Eun Joo Lee
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nayra Cárdenes
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Diana Álvarez
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jacobo Sellarés
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Interstitial Lung Disease Program, Servei de Pneumologia, Hospital Clínic, Barcelona, Spain
| | - John Sembrat
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paola Aranda
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yating Peng
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jordan Bullock
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seyed M Nouraie
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana L Mora
- Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Vascular Medicine Institute University of Pittsburgh, Pittsburgh, PA, USA
| | - Mauricio Rojas
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Respiratory, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Vascular Medicine Institute University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
119
|
Chadwick SR, Fazio EN, Etedali-Zadeh P, Genereaux J, Duennwald ML, Lajoie P. A functional unfolded protein response is required for chronological aging in Saccharomyces cerevisiae. Curr Genet 2019; 66:263-277. [PMID: 31346745 DOI: 10.1007/s00294-019-01019-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/08/2019] [Accepted: 07/16/2019] [Indexed: 12/29/2022]
Abstract
Progressive impairment of proteostasis and accumulation of toxic misfolded proteins are associated with the cellular aging process. Here, we employed chronologically aged yeast cells to investigate how activation of the unfolded protein response (UPR) upon accumulation of misfolded proteins in the endoplasmic reticulum (ER) affects lifespan. We found that cells lacking a functional UPR display a significantly reduced chronological lifespan, which contrasts previous findings in models of replicative aging. We find exacerbated UPR activation in aged cells, indicating an increase in misfolded protein burden in the ER during the course of aging. We also observed that caloric restriction, which promotes longevity in various model organisms, extends lifespan of UPR-deficient strains. Similarly, aging in pH-buffered media extends lifespan, albeit independently of the UPR. Thus, our data support a role for caloric restriction and reduced acid stress in improving ER homeostasis during aging. Finally, we show that UPR-mediated upregulation of the ER chaperone Kar2 and functional ER-associated degradation (ERAD) are essential for proper aging. Our work documents the central role of secretory protein homeostasis in chronological aging in yeast and highlights that the requirement for a functional UPR can differ between post-mitotic and actively dividing eukaryotic cells.
Collapse
Affiliation(s)
- Sarah R Chadwick
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, N6A 5C1, Canada
| | - Elena N Fazio
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, N6A 5C1, Canada
| | - Parnian Etedali-Zadeh
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, N6A 5C1, Canada
| | - Julie Genereaux
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, N6A 5C1, Canada.,Department of Biochemistry, The University of Western Ontario, London, N6A 5C1, Canada
| | - Martin L Duennwald
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, N6A 5C1, Canada.,Department of Pathology and Laboratory Medicine, The University of Western Ontario, London, N6A 5C1, Canada
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, N6A 5C1, Canada.
| |
Collapse
|
120
|
Hafycz JM, Naidoo NN. Sleep, Aging, and Cellular Health: Aged-Related Changes in Sleep and Protein Homeostasis Converge in Neurodegenerative Diseases. Front Aging Neurosci 2019; 11:140. [PMID: 31244649 PMCID: PMC6579877 DOI: 10.3389/fnagi.2019.00140] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/23/2019] [Indexed: 01/17/2023] Open
Abstract
Many neurodegenerative diseases manifest in an overall aged population, the pathology of which is hallmarked by abnormal protein aggregation. It is known that across aging, sleep quality becomes less efficient and protein homeostatic regulatory mechanisms deteriorate. There is a known relationship between extended wakefulness and poorly consolidated sleep and an increase in cellular stress. In an aged population, when sleep is chronically poor, and proteostatic regulatory mechanisms are less efficient, the cell is inundated with misfolded proteins and suffers a collapse in homeostasis. In this review article, we explore the interplay between aging, sleep quality, and proteostasis and how these processes are implicated in the development and progression of neurodegenerative diseases like Alzheimer's disease (AD). We also present data suggesting that reducing cellular stress and improving proteostasis and sleep quality could serve as potential therapeutic solutions for the prevention or delay in the progression of these diseases.
Collapse
Affiliation(s)
- Jennifer M Hafycz
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, United States
| | - Nirinjini N Naidoo
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, United States
| |
Collapse
|
121
|
Phang CW, Gandah NA, Abd Malek SN, Karsani SA. Proteomic analysis of flavokawain C-induced cell death in HCT 116 colon carcinoma cell line. Eur J Pharmacol 2019; 853:388-399. [DOI: 10.1016/j.ejphar.2019.04.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 12/15/2022]
|
122
|
Castelli V, Benedetti E, Antonosante A, Catanesi M, Pitari G, Ippoliti R, Cimini A, d'Angelo M. Neuronal Cells Rearrangement During Aging and Neurodegenerative Disease: Metabolism, Oxidative Stress and Organelles Dynamic. Front Mol Neurosci 2019; 12:132. [PMID: 31191244 PMCID: PMC6546816 DOI: 10.3389/fnmol.2019.00132] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/07/2019] [Indexed: 12/22/2022] Open
Abstract
Brain cells normally respond adaptively to oxidative stress or bioenergetic challenges, resulting from ongoing activity in neuronal circuits. During aging and in neurodegenerative disorders, these mechanisms are compromised. In fact, neurons show unique age-related changes in functions and metabolism, resulting in greater susceptibility to insults and disease. Aging affects the nervous system as well as other organs. More precisely, as the nervous system ages, neuron metabolism may change, inducing glucose hypometabolism, impaired transport of critical substrates underlying metabolism, alterations in calcium signaling, and mitochondrial dysfunction. Moreover, in neuronal aging, an accumulation of impaired and aggregated proteins in the cytoplasm and in mitochondria is observed, as the result of oxidative stress: reduced antioxidant defenses and/or increase of reactive oxygen species (ROS). These changes lead to greater vulnerability of neurons in various regions of the brain and increased susceptibility to several diseases. Specifically, the first part of the review article will focus on the major neuronal cells’ rearrangements during aging in response to changes in metabolism and oxidative stress, while the second part will cover the neurodegenerative disease areas in detail.
Collapse
Affiliation(s)
- Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Abruzzo, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Abruzzo, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Abruzzo, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Abruzzo, Italy
| | - Giuseppina Pitari
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Abruzzo, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Abruzzo, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Abruzzo, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Temple University, Philadelphia, PA, United States
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Abruzzo, Italy
| |
Collapse
|
123
|
Chadwick SR, Lajoie P. Endoplasmic Reticulum Stress Coping Mechanisms and Lifespan Regulation in Health and Diseases. Front Cell Dev Biol 2019; 7:84. [PMID: 31231647 PMCID: PMC6558375 DOI: 10.3389/fcell.2019.00084] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/03/2019] [Indexed: 12/30/2022] Open
Abstract
Multiple factors lead to proteostatic perturbations, often resulting in the aberrant accumulation of toxic misfolded proteins. Cells, from yeast to humans, can respond to sudden accumulation of secretory proteins within the endoplasmic reticulum (ER) through pathways such as the Unfolded Protein Response (UPR). The ability of cells to adapt the ER folding environment to the misfolded protein burden ultimately dictates cell fate. The aging process is a particularly important modifier of the proteostasis network; as cells age, both their ability to maintain this balance in protein folding/degradation and their ability to respond to insults in these pathways can break down, a common element of age-related diseases (including neurodegenerative diseases). ER stress coping mechanisms are central to lifespan regulation under both normal and disease states. In this review, we give a brief overview of the role of ER stress response pathways in age-dependent neurodegeneration.
Collapse
Affiliation(s)
- Sarah R Chadwick
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON, Canada
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
124
|
Emodin inhibits zinc-induced neurotoxicity in neuroblastoma SH-SY5Y cells. Biosci Rep 2019; 39:BSR20182378. [PMID: 31023967 PMCID: PMC6522748 DOI: 10.1042/bsr20182378] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
Emodin is a natural anthraquinone derivative with numerous beneficial effects, including antioxidant properties, anti-tumor activities, and protecting the nerves. Zinc-induced neurotoxicity plays a crucial role in the pathogenesis of vascular dementia (VD) and Parkinson’s disease (PD). Here, the protective activity of emodin inhibiting zinc-induced neurotoxicity and its molecular mechanisms such as cellular Zn2+ influx and zinc-induced gene expression were examined using human neuroblastoma cells (SH-SY5Y cells). Our findings showed that emodin obviously enhanced cell viability and reduced cell apoptosis and lactate dehydrogenase release. Bedsides, we detected a decrease of intracellular Zn2+ concentration after SH-SY5Y cells were pretreated with emodin. Simultaneously, the expression of zinc transporter-1, metallothionein-1, and metallothionein-2 were weakened in emodin-pretreated SH-SY5Y cells. In addition, emodin prevented the depletion of NAD+ and ATP induced by zinc. Emodin also reduced intracellular reactive oxygen species and endoplasmic reticulum-stress levels. Strikingly, emodin elevated SH-SY5Y cell viability and inhibited cell apoptosis caused by AMP-activated protein kinase signaling pathway activation. Thus, emodin could protect against neurotoxicity induced by Zn2+ in neuroblastoma SH-SY5Y cells. It is expected to have future therapeutic potential for VD or PD and other neurodegenerative diseases.
Collapse
|
125
|
Amatore D, Celestino I, Brundu S, Galluzzi L, Coluccio P, Checconi P, Magnani M, Palamara AT, Fraternale A, Nencioni L. Glutathione increase by the n-butanoyl glutathione derivative (GSH-C4) inhibits viral replication and induces a predominant Th1 immune profile in old mice infected with influenza virus. FASEB Bioadv 2019; 1:296-305. [PMID: 32123833 PMCID: PMC6996388 DOI: 10.1096/fba.2018-00066] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022] Open
Abstract
During aging, glutathione (GSH) content declines and the immune system undergoes a deficiency in the induction of Th1 response. Reduced secretion of Th1 cytokines, which is associated with GSH depletion, could weaken the host defenses against viral infections. We first evaluated the concentration of GSH and cysteine in organs of old mice; then, the effect of the administration of the N-butanoyl GSH derivative (GSH-C4) on the response of aged mice infected with influenza A PR8/H1N1 virus was studied through the determination of GSH concentration in organs, lung viral titer, IgA and IgG1/IgG2a production, and Th1/Th2 cytokine profile. Old mice had lower GSH than young mice in organs. Also the gene expression of endoplasmic reticulum (ER) stress markers involved in GSH metabolism and folding of proteins, that is, Nrf2 and PDI, was reduced. Following infection, GSH content remained low and neither infection nor GSH-C4 treatment affected Nrf2 expression. In contrast, PDI expression was upregulated during infection and appeared counterbalanced by GSH-C4. Moreover, the treatment with GSH-C4 increased GSH content in organs, reduced viral replication and induced a predominant Th1 response. In conclusion, GSH-C4 treatment could be used in the elderly to contrast influenza virus infection by inducing immune response, in particular the Th1 profile.
Collapse
Affiliation(s)
- Donatella Amatore
- Deparment of Public Health and Infectious DiseasesIstituto Pasteur Italia‐Fondazione Cenci‐Bolognetti, Sapienza University of RomeRomeItaly
| | - Ignacio Celestino
- Deparment of Public Health and Infectious DiseasesIstituto Pasteur Italia‐Fondazione Cenci‐Bolognetti, Sapienza University of RomeRomeItaly
| | - Serena Brundu
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbino (PU)Italy
| | - Luca Galluzzi
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbino (PU)Italy
| | - Paolo Coluccio
- Deparment of Public Health and Infectious DiseasesIstituto Pasteur Italia‐Fondazione Cenci‐Bolognetti, Sapienza University of RomeRomeItaly
| | - Paola Checconi
- Department of Human Sciences and Promotion of the Quality of LifeIRCCS San Raffaele Pisana, San Raffaele Roma Open UniversityRomeItaly
| | - Mauro Magnani
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbino (PU)Italy
| | - Anna Teresa Palamara
- Deparment of Public Health and Infectious DiseasesIstituto Pasteur Italia‐Fondazione Cenci‐Bolognetti, Sapienza University of RomeRomeItaly
- Department of Human Sciences and Promotion of the Quality of LifeIRCCS San Raffaele Pisana, San Raffaele Roma Open UniversityRomeItaly
| | | | - Lucia Nencioni
- Deparment of Public Health and Infectious DiseasesIstituto Pasteur Italia‐Fondazione Cenci‐Bolognetti, Sapienza University of RomeRomeItaly
| |
Collapse
|
126
|
Volpi VG, Ferri C, Fregno I, Del Carro U, Bianchi F, Scapin C, Pettinato E, Solda T, Feltri ML, Molinari M, Wrabetz L, D’Antonio M. Schwann cells ER-associated degradation contributes to myelin maintenance in adult nerves and limits demyelination in CMT1B mice. PLoS Genet 2019; 15:e1008069. [PMID: 30995221 PMCID: PMC6488099 DOI: 10.1371/journal.pgen.1008069] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 04/29/2019] [Accepted: 03/06/2019] [Indexed: 12/19/2022] Open
Abstract
In the peripheral nervous system (PNS) myelinating Schwann cells synthesize large amounts of myelin protein zero (P0) glycoprotein, an abundant component of peripheral nerve myelin. In humans, mutations in P0 cause the demyelinating Charcot-Marie-Tooth 1B (CMT1B) neuropathy, one of the most diffused genetic disorders of the PNS. We previously showed that several mutations, such as the deletion of serine 63 (P0-S63del), result in misfolding and accumulation of P0 in the endoplasmic reticulum (ER), with activation of the unfolded protein response (UPR). In addition, we observed that S63del mouse nerves display the upregulation of many ER-associated degradation (ERAD) genes, suggesting a possible involvement of this pathway in the clearance of the mutant P0. In ERAD in fact, misfolded proteins are dislocated from the ER and targeted for proteasomal degradation. Taking advantage of inducible cells that express the ER retained P0, here we show that the P0-S63del glycoprotein is degraded via ERAD. Moreover, we provide strong evidence that the Schwann cell-specific ablation of the ERAD factor Derlin-2 in S63del nerves exacerbates both the myelin defects and the UPR in vivo, unveiling a protective role for ERAD in CMT1B neuropathy. We also found that lack of Derlin-2 affects adult myelin maintenance in normal nerves, without compromising their development, pinpointing ERAD as a previously unrecognized player in preserving Schwann cells homeostasis in adulthood. Finally, we provide evidence that treatment of S63del peripheral nerve cultures with N-Acetyl-D-Glucosamine (GlcNAc), known to enhance protein quality control pathways in C.elegans, ameliorates S63del nerve myelination ex vivo. Overall, our study suggests that potentiating adaptive ER quality control pathways might represent an appealing strategy to treat both conformational and age-related PNS disorders. Charcot-Marie-Tooth neuropathies are a large family of peripheral nerve disorders, showing extensive clinical and genetic heterogeneity. Although strong advances have been made in the identification of genes and mutations involved, effective therapies are still lacking. Intracellular retention of abnormal proteins has been recently suggested as one of the pathogenetic events that might underlie several conformational neuropathies. To limit the toxic effects of accumulated mutant proteins, cells have developed efficient protein quality control systems aimed at optimizing both protein folding and degradation. Here we show that ER-associated degradation limits Schwann cells stress and myelin defects caused by the accumulation of a mutant myelin protein into the ER. In addition, we also describe for the first time the importance of Schwann cells ERAD in preserving myelin integrity in adult nerves, showing that genetic ERAD impairment leads to a late onset, motor-predominant, peripheral neuropathy in vivo. Effort in the design of strategies that potentiate ERAD and ER quality controls is therefore highly desirable.
Collapse
Affiliation(s)
- Vera G. Volpi
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Ferri
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Fregno
- Instuitute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Bellinzona, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Ubaldo Del Carro
- Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Bianchi
- Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Scapin
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuela Pettinato
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tatiana Solda
- Instuitute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - M. Laura Feltri
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York, United States of America
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Maurizio Molinari
- Instuitute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Bellinzona, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York, United States of America
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Maurizio D’Antonio
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- * E-mail:
| |
Collapse
|
127
|
Izquierdo V, Palomera-Ávalos V, López-Ruiz S, Canudas AM, Pallàs M, Griñán-Ferré C. Maternal Resveratrol Supplementation Prevents Cognitive Decline in Senescent Mice Offspring. Int J Mol Sci 2019; 20:ijms20051134. [PMID: 30845644 PMCID: PMC6429303 DOI: 10.3390/ijms20051134] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 02/24/2019] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
A variety of environmental factors contribute significantly to age-related cognitive decline and memory impairment in Alzheimer’s Disease (AD) and other neurodegenerative diseases. Nutrition can alter epigenetics, improving health outcomes, which can be transmitted across generations; this process is called epigenetic inheritance. We investigate the beneficial effects of maternal resveratrol supplementation in the direct exposed F1 generation and the transgenerational F2 generation. The offspring was generated from females Senescence Accelerated Mouse-Prone (SAMP8) fed a resveratrol-enriched diet for two months prior to mating. Object novel recognition and Morris Water Maze (MWM) demonstrated improvements in cognition in the 6-month-old F1 and F2 generations from resveratrol fed mothers. A significant increase in global DNA methylation with a decrease in hydroxymethylation in F1 and F2 were found. Accordingly, Dnmt3a/b and Tet2 gene expression changed. Methylation levels of Nrf2 and NF-kβ genes promoters raised in offspring, inducing changes in target genes expression, as well as hydrogen peroxide levels. Offspring that resulted from a resveratrol fed mother showed increase AMPKα activation, mTOR inhibition, and an increase in Pgc-1α gene expression and Beclin-1 protein levels. Endoplasmic reticulum stress sensors were found changed both in F1 and F2 generations. Overall, our results demonstrated that maternal resveratrol supplementation could prevent cognitive impairment in the SAMP8 mice offspring through epigenetic changes and cell signaling pathways.
Collapse
Affiliation(s)
- Vanesa Izquierdo
- Department of Pharmacology and Therapeutic Chemistry. Institut de Neurociències-University of Barcelona, Avda. Joan XXIII, 27. 08028 Barcelona, Spain.
| | - Verónica Palomera-Ávalos
- Department of Pharmacology and Therapeutic Chemistry. Institut de Neurociències-University of Barcelona, Avda. Joan XXIII, 27. 08028 Barcelona, Spain.
- Department of Cellular and Molecular Biology, University Center of Biological and Agricultural Sciences, University of Guadalajara, km 15.5 Guadalajara-Nogales highway, C.P. 45110 Zapopan, Jalisco, Mexico.
| | - Sergio López-Ruiz
- Department of Pharmacology and Therapeutic Chemistry. Institut de Neurociències-University of Barcelona, Avda. Joan XXIII, 27. 08028 Barcelona, Spain.
| | - Anna-Maria Canudas
- Department of Pharmacology and Therapeutic Chemistry. Institut de Neurociències-University of Barcelona, Avda. Joan XXIII, 27. 08028 Barcelona, Spain.
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry. Institut de Neurociències-University of Barcelona, Avda. Joan XXIII, 27. 08028 Barcelona, Spain.
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry. Institut de Neurociències-University of Barcelona, Avda. Joan XXIII, 27. 08028 Barcelona, Spain.
| |
Collapse
|
128
|
Dickens JA, Malzer E, Chambers JE, Marciniak SJ. Pulmonary endoplasmic reticulum stress-scars, smoke, and suffocation. FEBS J 2019; 286:322-341. [PMID: 29323786 DOI: 10.1111/febs.14381] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/11/2017] [Accepted: 01/08/2018] [Indexed: 12/14/2022]
Abstract
Protein misfolding within the endoplasmic reticulum (ER stress) can be a cause or consequence of pulmonary disease. Mutation of proteins restricted to the alveolar type II pneumocyte can lead to inherited forms of pulmonary fibrosis, but even sporadic cases of pulmonary fibrosis appear to be strongly associated with activation of the unfolded protein response and/or the integrated stress response. Inhalation of smoke can impair protein folding and may be an important cause of pulmonary ER stress. Similarly, tissue hypoxia can lead to impaired protein homeostasis (proteostasis). But the mechanisms linking smoke and hypoxia to ER stress are only partially understood. In this review, we will examine the role of ER stress in the pathogenesis of lung disease by focusing on fibrosis, smoke, and hypoxia.
Collapse
Affiliation(s)
- Jennifer A Dickens
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| | - Elke Malzer
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| | - Joseph E Chambers
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| | - Stefan J Marciniak
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| |
Collapse
|
129
|
Different Forms of ER Stress in Chondrocytes Result in Short Stature Disorders and Degenerative Cartilage Diseases: New Insights by Cartilage-Specific ERp57 Knockout Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8421394. [PMID: 30647818 PMCID: PMC6311764 DOI: 10.1155/2018/8421394] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
Abstract
Cartilage is essential for skeletal development by endochondral ossification. The only cell type within the tissue, the chondrocyte, is responsible for the production of macromolecules for the extracellular matrix (ECM). Before proteins and proteoglycans are secreted, they undergo posttranslational modification and folding in the endoplasmic reticulum (ER). However, the ER folding capacity in the chondrocytes has to be balanced with physiological parameters like energy and oxygen levels. Specific cellular conditions, e.g., a high protein demand, or pathologic situations disrupt ER homeostasis and lead to the accumulation of poorly folded or misfolded proteins. This state is called ER stress and induces a cellular quality control system, the unfolded protein response (UPR), to restore homeostasis. Different mouse models with ER stress in chondrocytes display comparable skeletal phenotypes representing chondrodysplasias. Therefore, ER stress itself seems to be involved in the pathogenesis of these diseases. It is remarkable that chondrodysplasias with a comparable phenotype arise independent from the sources of ER stress, which are as follows: (1) mutations in ECM proteins leading to aggregation, (2) deficiencies in ER chaperones, (3) mutations in UPR signaling factors, or (4) deficiencies in the degradation of aggregated proteins. In any case, the resulting UPR substantially impairs ECM protein synthesis, chondrocyte proliferation, and/or differentiation or regulation of autophagy and apoptosis. Notably, chondrodysplasias arise no matter if single or multiple events are affected. We analyzed cartilage-specific ERp57 knockout mice and demonstrated that the deficiency of this single protein disulfide isomerase, which is responsible for formation of disulfide bridges in ECM glycoproteins, is sufficient to induce ER stress and to cause an ER stress-related bone phenotype. These mice therefore qualify as a novel model for the analysis of ER stress in chondrocytes. They give new insights in ER stress-related short stature disorders and enable the analysis of ER stress in other cartilage diseases, such as osteoarthritis.
Collapse
|
130
|
Potential Application of Ixeris dentata in the Prevention and Treatment of Aging-Induced Dry Mouth. Nutrients 2018; 10:nu10121989. [PMID: 30558302 PMCID: PMC6316753 DOI: 10.3390/nu10121989] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022] Open
Abstract
Dry mouth is a common complaint among the elderly population. The aim of this study was to investigate the effect of Ixeris dentata (IXD) extract on aging-induced dry mouth. We used young (two months) and aged (20 months) SD rats in our study. Using water as the vehicle, IXD extract (25, 50, and 100 mg/kg) was given via oral gavage to the young and aged rats for eight weeks. We found that the salivary flow rate relative to the submandibular gland weight was differently influenced by IXD extract treatment. IXD extract augmented the submandibular gland acinar cells, which are depleted during aging. In addition, the decreased salivary alpha-amylase, inositol triphosphate receptor, and aquaporin-5 in the aging rats were upregulated by IXD treatment. Free radical-induced oxidative stress in the aging rats was also alleviated in the IXD-treated group. The formation of high molecular weight complexes of protein disulfide isomerase, decreased expression of an ER chaperone (GRP78), and increased ER stress response (ATF-4, CHOP and p-JNK) in aging rats was regulated with IXD treatment, and eventually increased salivary secretions from the aging submandibular glands. These are the first data to suggest that IXD extract might ameliorate aging-associated oral dryness by regulating the ER environment.
Collapse
|
131
|
Utility of curcumin for the treatment of diabetes mellitus: Evidence from preclinical and clinical studies. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
132
|
Kim S, Woo CH. Laminar Flow Inhibits ER Stress-Induced Endothelial Apoptosis through PI3K/Akt-Dependent Signaling Pathway. Mol Cells 2018; 41:964-970. [PMID: 30396238 PMCID: PMC6277562 DOI: 10.14348/molcells.2018.0111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 10/15/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis preferentially involves in prone area of low and disturbed blood flow while steady and high levels of laminar blood flow are relatively protected from atherosclerosis. Disturbed flow induces endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). ER stress is caused under stress that disturbs the processing and folding of proteins resulting in the accumulation of misfolded proteins in the ER and activation of the UPR. Prolonged or severe UPR leads to activate apoptotic signaling. Recent studies have indicated that disturbed flow significantly up-regulated p-ATF6α, p-IRE1α, and its target spliced XBP-1. However, the role of laminar flow in ER stress-mediated endothelial apoptosis has not been reported yet. The present study thus investigated the role of laminar flow in ER stress-dependent endothelial cell death. The results demonstrated that laminar flow protects ER stress-induced cleavage forms of PARP-1 and caspase-3. Also, laminar flow inhibits ER stress-induced p-eIF2α, ATF4, CHOP, spliced XBP-1, ATF6 and JNK pathway; these effects are abrogated by pharmacological inhibition of PI3K with wortmannin. Finally, nitric oxide affects thapsigargin-induced cell death in response to laminar flow but not UPR. Taken together, these findings indicate that laminar flow inhibits UPR and ER stress-induced endothelial cell death via PI3K/Akt pathway.
Collapse
Affiliation(s)
- Suji Kim
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415,
Korea
| | - Chang-Hoon Woo
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415,
Korea
| |
Collapse
|
133
|
Madreiter-Sokolowski CT, Waldeck-Weiermair M, Bourguignon MP, Villeneuve N, Gottschalk B, Klec C, Stryeck S, Radulovic S, Parichatikanond W, Frank S, Madl T, Malli R, Graier WF. Enhanced inter-compartmental Ca 2+ flux modulates mitochondrial metabolism and apoptotic threshold during aging. Redox Biol 2018; 20:458-466. [PMID: 30458321 PMCID: PMC6243020 DOI: 10.1016/j.redox.2018.11.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 01/02/2023] Open
Abstract
Background Senescence is characterized by a gradual decline in cellular functions, including changes in energy homeostasis and decreased proliferation activity. As cellular power plants, contributors to signal transduction, sources of reactive oxygen species (ROS) and executors of programmed cell death, mitochondria are in a unique position to affect aging-associated processes of cellular decline. Notably, metabolic activation of mitochondria is tightly linked to Ca2+ due to the Ca2+ -dependency of several enzymes in the Krebs cycle, however, overload of mitochondria with Ca2+ triggers cell death pathways. Consequently, a machinery of proteins tightly controls mitochondrial Ca2+ homeostasis as well as the exchange of Ca2+ between the different cellular compartments, including Ca2+ flux between mitochondria and the endoplasmic reticulum (ER). Methods In this study, we investigated age-related changes in mitochondrial Ca2+ homeostasis, mitochondrial-ER linkage and the activity of the main ROS production site, the mitochondrial respiration chain, in an in vitro aging model based on porcine aortic endothelial cells (PAECs), using high-resolution live cell imaging, proteomics and various molecular biological methods. Results We describe that in aged endothelial cells, increased ER-mitochondrial Ca2+ crosstalk occurs due to enhanced ER-mitochondrial tethering. The close functional inter-organelle linkage increases mitochondrial Ca2+ uptake and thereby the activity of the mitochondrial respiration, but also makes senescent cells more vulnerable to mitochondrial Ca2+-overload-induced cell death. Moreover, we identified the senolytic properties of the polyphenol resveratrol, triggering cell death via mitochondrial Ca2+ overload exclusively in senescent cells. Conclusion By unveiling aging-related changes in the inter-organelle tethering and Ca2+ communications we have advanced the understanding of endothelial aging and highlighted a potential basis to develop drugs specifically targeting senescent cells.
Collapse
Affiliation(s)
- Corina T Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; Department of Health Sciences and Technology, ETH Zurich, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland.
| | - Markus Waldeck-Weiermair
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | | | - Nicole Villeneuve
- Servier Research Institute, Cardiovascular Unit, 11 rue des Moulineaux, 92150 Suresnes, France
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Christiane Klec
- Division of Oncology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Sarah Stryeck
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Snjezana Radulovic
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | | | - Saša Frank
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Tobias Madl
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; BioTechMed, Graz, Austria
| | - Wolfgang F Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; BioTechMed, Graz, Austria.
| |
Collapse
|
134
|
Jin H, Komita M, Aoe T. Decreased Protein Quality Control Promotes the Cognitive Dysfunction Associated With Aging and Environmental Insults. Front Neurosci 2018; 12:753. [PMID: 30443201 PMCID: PMC6221900 DOI: 10.3389/fnins.2018.00753] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 10/01/2018] [Indexed: 11/29/2022] Open
Abstract
Background: Most neurodegenerative diseases are sporadic and develop with age. Degenerative neural tissues often contain intra- and extracellular protein aggregates, suggesting that the proteostasis network that combats protein misfolding could be dysfunctional in the setting of neurodegenerative disease. Binding immunoglobulin protein (BiP) is an endoplasmic reticulum (ER) chaperone that is crucial for protein folding and modulating the adaptive response in early secretory pathways. The interaction between BiP and unfolded proteins is mediated by the substrate-binding domain and nucleotide-binding domain with ATPase activity. The interaction facilitates protein folding and maturation. BiP has a recovery motif at the carboxyl terminus. The aim of this study is to examine cognitive function in model mice with an impaired proteostasis network by expressing a mutant form of BiP lacking the recovery motif. We also investigated if impairments of cognitive function were exacerbated by exposure to environmental insults, such as inhaled anesthetics. Methods: We examined cognitive function by performing radial maze testing with mutant BiP mice and assessed the additional impact of general anesthesia in the context of proteostasis dysfunction. Testing over 8 days was performed 10 weeks, 6 months, and 1 year after birth. Results: Age-related cognitive decline occurred in both forms of mice. The mutant BiP and anesthetic exposure promoted cognitive dysfunction prior to the senile period. After senescence, when mice were tested at 6 months of age and at 1 year old, there were no significant differences between the two genotypes in terms of the radial maze testing; furthermore, there was no significant difference when tested with and without anesthetic exposure. Conclusion: Our data suggest that aging was the predominant factor underlying the impairment of cognitive function in this study. Impairment of the proteostasis network may promote age-related neurodegeneration, and this is exacerbated by external insults.
Collapse
Affiliation(s)
- Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Mari Komita
- Department of Anesthesiology, Chiba Rosai Hospital, Ichihara, Japan
| | - Tomohiko Aoe
- Department of Medicine, Pain Center, Chiba Medical Center, Teikyo University, Ichihara, Japan
| |
Collapse
|
135
|
CDNF induces the adaptive unfolded protein response and attenuates endoplasmic reticulum stress-induced cell death. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1579-1589. [DOI: 10.1016/j.bbamcr.2018.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/28/2018] [Accepted: 08/15/2018] [Indexed: 02/07/2023]
|
136
|
Mukai S, Ogawa Y, Urano F, Kawakami Y, Tsubota K. Novel elucidation and treatment of pancreatic chronic graft-versus-host disease in mice. ROYAL SOCIETY OPEN SCIENCE 2018; 5:181067. [PMID: 30473850 PMCID: PMC6227968 DOI: 10.1098/rsos.181067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/20/2018] [Indexed: 06/09/2023]
Abstract
Chronic graft-versus-host disease (cGVHD) is a severe complication of allogeneic haematopoietic stem cell transplantation. There is a growing understanding of cGVHD, and several effective therapies for cGVHD have been reported. However, pancreatic cGVHD is a potentially untapped study field. Our thought-provoking study using a mouse model of cGVHD suggested that the pancreas could be impaired by cGVHD-induced inflammation and fibrosis and that endoplasmic reticulum (ER) stress was augmented in the pancreas affected by cGVHD. These findings urged us to treat pancreatic cGVHD through reduction of ER stress, and we used 4-phenylbutyric acid (PBA) as an ER stress reducer. A series of experiments has indicated that PBA can suppress cGVHD-elicited ER stress in the pancreas and accordingly alleviate pancreatic cGVHD. Furthermore, we focused on a correlation between epithelial to mesenchymal transition (EMT) and fibrosis in the cGVHD-affected pancreas, because EMT was conceivably implicated in various fibrosis-associated diseases. Our investigation has suggested that the expression of EMT markers was increased in the cGVHD-disordered pancreas and that it could be reduced by PBA. Taken together, we have provided a clue to elucidate the pathogenic process of pancreatic cGVHD and created a potentially effective treatment of this disease using the ER stress alleviator PBA.
Collapse
Affiliation(s)
- Shin Mukai
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Division of Cellular Signalling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Yutaka Kawakami
- Division of Cellular Signalling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
137
|
Hui CW, St-Pierre MK, Detuncq J, Aumailley L, Dubois MJ, Couture V, Skuk D, Marette A, Tremblay JP, Lebel M, Tremblay MÈ. Nonfunctional mutant Wrn protein leads to neurological deficits, neuronal stress, microglial alteration, and immune imbalance in a mouse model of Werner syndrome. Brain Behav Immun 2018; 73:450-469. [PMID: 29908963 DOI: 10.1016/j.bbi.2018.06.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/25/2018] [Accepted: 06/06/2018] [Indexed: 12/30/2022] Open
Abstract
Werner syndrome (WS) is a premature aging disorder caused by mutations in a RecQ-family DNA helicase, WRN. Mice lacking part of the helicase domain of the WRN orthologue exhibit many phenotypic features of WS, including metabolic abnormalities and a shorter lifespan. Yet, little is known about the impact of WRN mutations on the central nervous system in both humans and mouse models of WS. In the current study, we have performed a longitudinal behavioral assessment on mice bearing a Wrn helicase deletion. Behavioral tests demonstrated a loss of motor activity and coordination, reduction in perception, increase in repetitive behavior, and deficits in both spatial and social novelty memories in Wrn mutant mice compared to age-matched wild type mice. These neurological deficits were associated with biochemical and histological changes in the brain of aged Wrn mutant mice. Microglia, resident immune cells that regulate neuronal plasticity and function in the brain, were hyper-ramified in multiple regions involved with the behavioral deficits of Wrn mutant mice. Furthermore, western analyses indicated that Wrn mutant mice exhibited an increase of oxidative stress markers in the prefrontal cortex. Supporting these findings, electron microscopy studies revealed increased cellular aging and oxidative stress features, among microglia and neurons respectively, in the prefrontal cortex of aged Wrn mutant mice. In addition, multiplex immunoassay of serum identified significant changes in the expression levels of several pro- and anti-inflammatory cytokines. Taken together, these findings indicate that microglial dysfunction and neuronal oxidative stress, associated with peripheral immune system alterations, might be important driving forces leading to abnormal neurological symptoms in WS thus suggesting potential therapeutic targets for interventions.
Collapse
Affiliation(s)
- Chin Wai Hui
- Axe neurosciences, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada
| | - Marie-Kim St-Pierre
- Axe neurosciences, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada
| | - Jérôme Detuncq
- Axe neurosciences, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada
| | - Lucie Aumailley
- Axe endocrinologie/néphrologie, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada
| | - Marie-Julie Dubois
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Sainte-Foy, Québec City, Québec G1V 4G5, Canada
| | - Vanessa Couture
- Axe neurosciences, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada
| | - Daniel Skuk
- Axe neurosciences, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada
| | - André Marette
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Sainte-Foy, Québec City, Québec G1V 4G5, Canada
| | - Jacques P Tremblay
- Axe neurosciences, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada
| | - Michel Lebel
- Axe endocrinologie/néphrologie, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada.
| | - Marie-Ève Tremblay
- Axe neurosciences, Centre de recherche du CHU de Québec, Centre Hospitalier de l'Université Laval (CHUL), 2705 Laurier Blvd., Québec City, Québec G1V 4G2, Canada.
| |
Collapse
|
138
|
Zhou X, Sen I, Lin XX, Riedel CG. Regulation of Age-related Decline by Transcription Factors and Their Crosstalk with the Epigenome. Curr Genomics 2018; 19:464-482. [PMID: 30258277 PMCID: PMC6128382 DOI: 10.2174/1389202919666180503125850] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 04/20/2018] [Accepted: 04/30/2018] [Indexed: 12/16/2022] Open
Abstract
Aging is a complex phenomenon, where damage accumulation, increasing deregulation of biological pathways, and loss of cellular homeostasis lead to the decline of organismal functions over time. Interestingly, aging is not entirely a stochastic process and progressing at a constant rate, but it is subject to extensive regulation, in the hands of an elaborate and highly interconnected signaling network. This network can integrate a variety of aging-regulatory stimuli, i.e. fertility, nutrient availability, or diverse stresses, and relay them via signaling cascades into gene regulatory events - mostly of genes that confer stress resistance and thus help protect from damage accumulation and homeostasis loss. Transcription factors have long been perceived as the pivotal nodes in this network. Yet, it is well known that the epigenome substantially influences eukaryotic gene regulation, too. A growing body of work has recently underscored the importance of the epigenome also during aging, where it not only undergoes drastic age-dependent changes but also actively influences the aging process. In this review, we introduce the major signaling pathways that regulate age-related decline and discuss the synergy between transcriptional regulation and the epigenetic landscape.
Collapse
Affiliation(s)
| | | | | | - Christian G. Riedel
- Address correspondence to this author at the Integrated Cardio Metabolic Centre (ICMC), Department of Biosciences and Nutrition, Karolinska Institutet, Blickagången 6, Novum, 7 floor Huddinge, Stockholm 14157, Sweden; Tel: +46-736707008; E-mail:
| |
Collapse
|
139
|
Glucose-regulated protein 78 in lipid rafts elevates vascular smooth muscle cell proliferation of spontaneously hypertensive rats by controlling platelet-derived growth factor receptor signaling. Pflugers Arch 2018; 470:1831-1843. [PMID: 30155775 DOI: 10.1007/s00424-018-2199-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/09/2018] [Accepted: 08/16/2018] [Indexed: 10/28/2022]
Abstract
The multifunctional glucose-regulated protein 78 (GRP78) is known to be differentially expressed in the lipid rafts of vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats (SHRs) and normal Wistar-Kyoto (WKY) rats. However, its role in VSMCs from SHRs remains to be elucidated. This work was conducted to investigate the contribution made by GRP78 in VSMCs. GRP78 expression in VSMC lipid rafts decreased in WKY rats with age, but not in SHRs. Transfection with GRP78-siRNA attenuated not only platelet-derived growth factor (PDGF)-BB-induced VSMC proliferation and aortic sprout outgrowth but also the phosphorylation of PDGF receptor (PDGFR)-β, Akt, and extracellular signal-regulated kinase (Erk) 1/2 in VSMCs in response to PDGF-BB. Moreover, GRP78 knockdown also reduced the PDGF-BB-induced dimerization of PDGFR-β and GRP78 in SHR VSMCs. The phosphorylation of GRP78 and PDGFR-β was elevated in VSMCs treated with PDGF-BB and was completely abolished by AG1296 (a PDGFR inhibitor). Moreover, the binding of PDGFR-β to GRP78 and the co-localization of GRP78 to PDGFR-β in VSMCs were stronger in SHRs than in WKY rat controls. This study demonstrates that the PDGF-BB-induced proliferation of SHR VSMCs is mediated by the expressional upregulation of GRP78 on VSMC lipid rafts in SHRs, probably via the regulation of PDGFR-β-GRP78 binding and their cross-activation. These observations indicate that GRP78 may play important roles in the pathological progression of SHR VSMCs.
Collapse
|
140
|
Fuhrmann-Stroissnigg H, Niedernhofer LJ, Robbins PD. Hsp90 inhibitors as senolytic drugs to extend healthy aging. Cell Cycle 2018; 17:1048-1055. [PMID: 29886783 DOI: 10.1080/15384101.2018.1475828] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aging is characterized by progressive decay of biological systems and although it is not considered a disease, it is one of the main risk factors for chronic diseases and many types of cancers. The accumulation of senescent cells in various tissues is thought to be a major factor contributing to aging and age-related diseases. Removal of senescent cells during aging by either genetic or therapeutic methods have led to an improvement of several age related disease in mice. In this preview, we highlight the significance of developing senotherapeutic approaches to specifically kill senescent cells (senolytics) or suppress the senescence-associated secretory phenotype (SASP) that drives sterile inflammation (senomorphics) associated with aging to extend healthspan and potentially lifespan. Also, we provide an overview of the senotherapeutic drugs identified to date. In particular, we discuss and expand upon the recent identification of inhibitors of the HSP90 co-chaperone as a new class of senolytics.
Collapse
Affiliation(s)
- Heike Fuhrmann-Stroissnigg
- a Department of Molecular Medicine and The Center on Aging , The Scripps Research Institute , Jupiter , FL , USA
| | - Laura J Niedernhofer
- a Department of Molecular Medicine and The Center on Aging , The Scripps Research Institute , Jupiter , FL , USA
| | - Paul D Robbins
- a Department of Molecular Medicine and The Center on Aging , The Scripps Research Institute , Jupiter , FL , USA
| |
Collapse
|
141
|
Tesei A, Cortesi M, Zamagni A, Arienti C, Pignatta S, Zanoni M, Paolillo M, Curti D, Rui M, Rossi D, Collina S. Sigma Receptors as Endoplasmic Reticulum Stress "Gatekeepers" and their Modulators as Emerging New Weapons in the Fight Against Cancer. Front Pharmacol 2018; 9:711. [PMID: 30042674 PMCID: PMC6048940 DOI: 10.3389/fphar.2018.00711] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
Despite the interest aroused by sigma receptors (SRs) in the area of oncology, their role in tumor biology remains enigmatic. The predominant subcellular localization and main site of activity of SRs are the endoplasmic reticulum (ER). Current literature data, including recent findings on the sigma 2 receptor subtype (S2R) identity, suggest that SRs may play a role as ER stress gatekeepers. Although SR endogenous ligands are still unknown, a wide series of structurally unrelated compounds able to bind SRs have been identified. Currently, the identification of novel antiproliferative molecules acting via SR interaction is a challenging task for both academia and industry, as shown by the fact that novel anticancer drugs targeting SRs are in the preclinical-stage pipeline of pharmaceutical companies (i.e., Anavex Corp. and Accuronix). So far, no clinically available anticancer drugs targeting SRs are still available. The present review focuses literature advancements and provides a state-of-the-art overview of SRs, with emphasis on their involvement in cancer biology and on the role of SR modulators as anticancer agents. Findings from preclinical studies on novel anticancer drugs targeting SRs are presented in brief.
Collapse
Affiliation(s)
- Anna Tesei
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Michela Cortesi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Alice Zamagni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Chiara Arienti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Sara Pignatta
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Mayra Paolillo
- Pharmacology Section, Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Daniela Curti
- Laboratory of Cellular and Molecular Neuropharmacology, Department of Biology and Biotechnology 'L. Spallanzani', University of Pavia, Pavia, Italy
| | - Marta Rui
- Medicinal Chemistry and Pharmaceutical Technology Section, Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Daniela Rossi
- Medicinal Chemistry and Pharmaceutical Technology Section, Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Simona Collina
- Medicinal Chemistry and Pharmaceutical Technology Section, Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
142
|
Zhu B, Dacso CC, O’Malley BW. Unveiling "Musica Universalis" of the Cell: A Brief History of Biological 12-Hour Rhythms. J Endocr Soc 2018; 2:727-752. [PMID: 29978151 PMCID: PMC6025213 DOI: 10.1210/js.2018-00113] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022] Open
Abstract
"Musica universalis" is an ancient philosophical concept claiming the movements of celestial bodies follow mathematical equations and resonate to produce an inaudible harmony of music, and the harmonious sounds that humans make were an approximation of this larger harmony of the universe. Besides music, electromagnetic waves such as light and electric signals also are presented as harmonic resonances. Despite the seemingly universal theme of harmonic resonance in various disciplines, it was not until recently that the same harmonic resonance was discovered also to exist in biological systems. Contrary to traditional belief that a biological system is either at stead-state or cycles with a single frequency, it is now appreciated that most biological systems have no homeostatic "set point," but rather oscillate as composite rhythms consisting of superimposed oscillations. These oscillations often cycle at different harmonics of the circadian rhythm, and among these, the ~12-hour oscillation is most prevalent. In this review, we focus on these 12-hour oscillations, with special attention to their evolutionary origin, regulation, and functions in mammals, as well as their relationship to the circadian rhythm. We further discuss the potential roles of the 12-hour clock in regulating hepatic steatosis, aging, and the possibility of 12-hour clock-based chronotherapy. Finally, we posit that biological rhythms are also musica universalis: whereas the circadian rhythm is synchronized to the 24-hour light/dark cycle coinciding with the Earth's rotation, the mammalian 12-hour clock may have evolved from the circatidal clock, which is entrained by the 12-hour tidal cues orchestrated by the moon.
Collapse
Affiliation(s)
- Bokai Zhu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Clifford C Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Bert W O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
143
|
Inflammation and Monocyte Recruitment due to Aging and Mechanical Stretch in Alveolar Epithelium are Inhibited by the Molecular Chaperone 4-phenylbutyrate. Cell Mol Bioeng 2018; 11:495-508. [PMID: 30581495 DOI: 10.1007/s12195-018-0537-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Introduction Ventilator-Induced lung injury (VILI) is a form of acute lung injury that is initiated or exacerbated by mechanical ventilation. The aging lung is also more susceptible to injury. Harmful mechanical stretch of the alveolar epithelium is a recognized mechanism of VILI, yet little is known about how mechanical stretch affects aged epithelial cells. Disruption to Endoplasmic Reticulum (ER) homeostasis results in a condition known as ER stress that leads to disruption of cellular homeostasis, apoptosis, and inflammation. ER stress is increased with aging and other pathological stimuli. We hypothesized that age and mechanical stretch increase alveolar epithelial cells' proinflammatory responses that are mediated by ER stress. Furthermore, we believed that inhibition of this upstream mechanism with 4PBA, an ER stress reducer, alleviates subsequent inflammation and monocyte recruitment. Methods Type II alveolar epithelial cells (ATII) were harvested from C57Bl6/J mice 2 months (young) and 20 months (old) of age. The cells were cyclically stretched at 15% change in surface area for up to 24 hours. Prior to stretch, groups were administered 4PBA or vehicle as a control. Results Mechanical stretch and age upregulated ER stress and proinflammatory MCP-1/CCL2 and MIP-1β/CCL4 chemokine expression in ATIIs. Age-matched and mismatched monocyte recruitment by ATII conditioned media was also quantified. Conclusions Age increases susceptibility to stretch-induced ER stress and downstream inflammatory gene expression in a primary ATII epithelial cell model. Administration of 4PBA attenuated the increased ER stress and proinflammatory responses from stretch and/or age and significantly reduced monocyte migration to ATII conditioned media.
Collapse
|
144
|
Ma N, Liu HM, Xia T, Liu JD, Wang XZ. Chronic aerobic exercise training alleviates myocardial fibrosis in aged rats through restoring bioavailability of hydrogen sulfide. Can J Physiol Pharmacol 2018; 96:902-908. [PMID: 29862831 DOI: 10.1139/cjpp-2018-0153] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Age-related fibrosis is attenuated by aerobic exercise; however, little is known concerning the underlying molecular mechanism. To address this question, aged rats were given moderate-intensity exercise for 12 weeks. After exercise in aged rats, hydrogen sulfide levels in plasma and heart increased 39.8% and 90.9%, respectively. Exercise upregulated expression of cystathionine γ-lyase and 3-mercaptopyruvate sulfurtransferase in heart of aged rats. Furthermore, aged rats were given moderate-intensity exercise for 12 weeks or treated with NaHS (intraperitoneal injection of 0.1 mL/kg per day of 0.28 mol/L NaHS). After exercise in aged rats, Masson-trichrome staining area decreased 34.8% and myocardial hydroxyproline levels decreased 29.6%. Exercise downregulated expression of collagen-I and α- smooth muscle actin in heart of aged rats. Exercise in aged rats reduced malondialdehyde levels in plasma and heart and 3-nitrotyrosine in heart. Exercise in aged rats reduced mRNA and protein expression of C/EBP homologous protein, glucose regulated protein 78, and X-box protein 1. Exercise also reduced mRNA and protein expression of interleukin 6 and monocyte chemotactic protein 1and suppressed activation of c-Jun N-terminal kinase in aging heart. Similar effects were demonstrated in aged rats treated with NaHS. Collectively, exercise restored bioavailability of hydrogen sulfide in the heart of aged rats, which partly explained the benefits of exercise against myocardial fibrosis of aged population.
Collapse
Affiliation(s)
- Ning Ma
- a School of Physical Education, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Hong-Mei Liu
- b Yunnan College of Business Management, Kunming, Yunnan, China
| | - Ting Xia
- c Department of Critical Care Medicine, Weinan First Hospital, Weinan, Shaanxi, China
| | - Jian-Dong Liu
- d Department of Gastroenterology, The Third People's Hospital of Datong, Datong, Shanxi, China
| | - Xiao-Ze Wang
- e Department of General Surgery, Yuquan Hospital of Tsinghua University, Beijing, China
| |
Collapse
|
145
|
Mishra N, Lata S, Deshmukh P, Kamat K, Surolia A, Banerjee T. Insulin signaling pathway protects neuronal cell lines by Sirt3 mediated IRS2 activation. Biofactors 2018; 44:224-236. [PMID: 29411439 DOI: 10.1002/biof.1413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/28/2017] [Accepted: 01/05/2018] [Indexed: 01/15/2023]
Abstract
Cellular stress like ER and oxidative stress are the principle causative agents of various proteinopathies. Multifunctional protein PARK7/DJ-1 provides protection against cellular stress. Recently, insulin/IGF also has emerged as a neuro-protective molecule. However, it is not known whether DJ-1 and insulin/IGF complement each other for cellular protection in response to stress. In this study, we show for the first time, that in human and mouse neuronal cell lines, down regulation of DJ-1 for 48 h leads to compensatory upregulation of insulin/IGF signaling (IIS) pathway genes, namely, insulin receptor, insulin receptor substrate, and Akt under normal physiological conditions as well as in cellular stress conditions. Moreover, upon exogenous supply of insulin there is a marked increase in the IIS components both at gene and protein levels leading to down regulation and inactivation of GSK3β. By immunoprecipitation, it was observed that Sirt3 mediated deacetylation and activation of FoxO3a could not occur under DJ-1 downregulation. Transient DJ-1 downregulation also led to Akt mediated increased phosphorylation and nuclear exclusion of FoxO3a. When DJ-1 was downregulated increased interaction of Sirt3 with IRS2 was observed leading to its activation resulting in IIS upregulation. Thus, transient downregulation of DJ-1 leads to stimulation of IIS pathway by Sirt3 mediated IRS2 activation. Consequently, antiapoptotic program is triggered in neuronal cells via Akt-GSK3β-FoxO3a axis. © 2018 BioFactors, 44(3):224-236, 2018.
Collapse
Affiliation(s)
- Neha Mishra
- Department of Biotechnology, Savitribai Phule Pune University (Former Pune University), Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Sonam Lata
- Department of Biotechnology, Savitribai Phule Pune University (Former Pune University), Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Priyanka Deshmukh
- Department of Biotechnology, Savitribai Phule Pune University (Former Pune University), Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Kajal Kamat
- Department of Biotechnology, Savitribai Phule Pune University (Former Pune University), Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Avadhesha Surolia
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Tanushree Banerjee
- Department of Biotechnology, Savitribai Phule Pune University (Former Pune University), Ganeshkhind Road, Pune, Maharashtra 411007, India
| |
Collapse
|
146
|
Naidoo N, Zhu J, Galante RJ, Lian J, Strus E, Lee A, Keenan BT, Pack AI. Reduction of the molecular chaperone binding immunoglobulin protein (BiP) accentuates the effect of aging on sleep-wake behavior. Neurobiol Aging 2018; 69:10-25. [PMID: 29843048 DOI: 10.1016/j.neurobiolaging.2018.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 04/05/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022]
Abstract
Sleep and wake quality, quantity, and architecture become modified with aging. Sleep and wake quality decline coinciding with increased fragmentation of both states across aging. We have previously shown that this age-related decline in sleep-wake quality is associated with increased endoplasmic reticular (ER) stress and decreased expression of the major ER chaperone binding immunoglobulin protein (BiP). BiP, also known as glucose-regulated protein 78, plays a key role in controlling the cellular response to ER stress, acting as a regulator of a protein homeostatic signaling pathway known as the unfolded protein response. Induction of BiP during cellular stress is part of an adaptive prosurvival mechanism. Here, using mice heterozygous for BiP, we investigated the effect of reduced BiP expression on sleep-wake behavior across aging; complete knockdown of BiP is embryonic lethal. We report that BiP heterozygosity accentuates the aging sleep-wake phenotype. Sleep and wake fragmentation was more pronounced in the BiP heterozygotes across the 3 ages examined. In mice lacking 1 functional copy of BiP, we observed an age-related significant reduction in wake bout duration and increase in wake bout numbers during the active period, as well as an increase in non rapid eye movement and rapid eye movement bout numbers accompanied by reduced bout durations of both non rapid eye movement and rapid eye movement during the sleep period. In addition, we observed increased ER stress in orexin neurons and occurrence of aggregates immunopositive for orexin at the terminals and projections of orexin neurons in the middle-aged BiP heterozygotes. Taken together, our data indicate that a reduction in the molecular chaperone BiP impacts sleep architecture across aging and that orexin processing is likely to be affected.
Collapse
Affiliation(s)
- Nirinjini Naidoo
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Sleep Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Jingxu Zhu
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond J Galante
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jie Lian
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ewa Strus
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Lee
- Department of Biochemistry and Molecular Biology, University of Southern California, Keck, School of Medicine, USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Brendan T Keenan
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Allan I Pack
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Sleep Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
147
|
Zhang SJ, Xu TT, Li L, Xu YM, Qu ZL, Wang XC, Huang SQ, Luo Y, Luo NC, Lu P, Shi YF, Yang X, Wang Q. Bushen-Yizhi formula ameliorates cognitive dysfunction through SIRT1/ER stress pathway in SAMP8 mice. Oncotarget 2018; 8:49338-49350. [PMID: 28521305 PMCID: PMC5564772 DOI: 10.18632/oncotarget.17638] [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] [Received: 04/03/2017] [Accepted: 04/25/2017] [Indexed: 12/22/2022] Open
Abstract
The Chinese formula Bushen-Yizhi (BSYZ) has been reported to ameliorate cognitive dysfunction. However the mechanism is still unclear. In this study, we employ an aging model, SAMP8 mice, to explore whether BSYZ could protect dementia through SIRT1/endoplasmic reticulum (ER) stress pathway. Morris water maze and the fearing condition test results show that oral administration of BSYZ (1.46 g/kg/d, 2.92 g/kg/d and 5.84 g/kg/d) and donepezil (3 mg/kg/d) shorten the escape latency, increase the crossing times of the original position of the platform and the time spent in the target quadrant, and increase the freezing time. BSYZ decreases the activity of acetylcholinesterase (AChE), and increases the activity of choline acetyltransferase (ChAT) and the concentration of acetylcholine (Ach) in both hippocampus and cortex. In addition, western blot results (Bcl-2, Bax and Caspase-3) and TUNEL staining show that BSYZ prevents neuron from apoptosis, and elevates the expression of neurotrophic factors, including nerve growth factor (NGF), postsynapticdensity 95 (PSD95) and synaptophysin (SYN), in both hippocampus and cortex. BSYZ also increases the protein expression of SIRT1 and alleviates ER stress-associated proteins (PERK, IRE-1α, eIF-2α, BIP, PDI and CHOP). These results indicate that the neuroprotective mechanism of BSYZ might be related with SIRT1/ER stress pathway.
Collapse
Affiliation(s)
- Shi-Jie Zhang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ting-Ting Xu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Lin Li
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yu-Min Xu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zi-Ling Qu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xin-Chen Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shui-Qing Huang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yi Luo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Na-Chuan Luo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ping Lu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ya-Fei Shi
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xin Yang
- Department of Pharmacy, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
148
|
Long-Term Exercise Protects against Cellular Stresses in Aged Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2894247. [PMID: 29765493 PMCID: PMC5889853 DOI: 10.1155/2018/2894247] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/07/2018] [Accepted: 01/22/2018] [Indexed: 01/09/2023]
Abstract
The current study examined the effect of aging and long-term wheel-running on the expression of heat shock protein (HSP), redox regulation, and endoplasmic reticulum (ER) stress markers in tibialis anterior (T.A.) and soleus muscle of mice. Male mice were divided into young (Y, 3-month-old), old-sedentary (OS, 24-month-old), and old-exercise (OE, 24-month-old) groups. The OE group started voluntary wheel-running at 3 months and continued until 24 months of age. Aging was associated with a higher thioredoxin-interacting protein (TxNiP) level, lower thioredoxin-1 (TRX-1) to TxNiP ratio—a determinant of redox regulation and increased CHOP, an indicator of ER stress-related apoptosis signaling in both muscles. Notably, GRP78, a key indicator of ER stress, was selectively elevated in T.A. Long-term exercise decreased TxNiP in T.A. and soleus muscles and increased the TRX-1/TxNiP ratio in soleus muscle of aged mice. Inducible HSP70 and constituent HSC70 were upregulated, whereas CHOP was reduced after exercise in soleus muscle. Thus, our data demonstrated that aging induced oxidative stress and activated ER stress-related apoptosis signaling in skeletal muscle, whereas long-term wheel-running improved redox regulation, ER stress adaptation and attenuated ER stress-related apoptosis signaling. These findings suggest that life-long exercise can protect against age-related cellular stress.
Collapse
|
149
|
Aumailley L, Roux-Dalvai F, Kelly I, Droit A, Lebel M. Vitamin C alters the amount of specific endoplasmic reticulum associated proteins involved in lipid metabolism in the liver of mice synthesizing a nonfunctional Werner syndrome (Wrn) mutant protein. PLoS One 2018; 13:e0193170. [PMID: 29494634 PMCID: PMC5832228 DOI: 10.1371/journal.pone.0193170] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/06/2018] [Indexed: 11/19/2022] Open
Abstract
Werner syndrome (WS) is a premature aging disorder caused by mutations in a protein containing both a DNA exonuclease and DNA helicase domain. Mice lacking the helicase domain of the Wrn protein orthologue exhibit transcriptomic and metabolic alterations, some of which are reversed by vitamin C. Recent studies on these animals indicated that the mutant protein is associated with enriched endoplasmic reticulum (ER) fractions of tissues resulting in an ER stress response. In this study, we identified proteins that exhibit actual level differences in the ER enriched fraction between the liver of wild type and Wrn mutant mice using quantitative proteomic profiling with label-free Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS). Multiple Reaction Monitoring (MRM) and immunoblotting were performed to validate findings in a secondary independent cohort of wild type and Wrn mutant mice. DAVID 6.7 (NIH) was used for functional annotation analysis and indicated that the identified proteins exhibiting level changes between untreated wild type, Wrn mutant, and vitamin C treated Wrn mutant mice (ANOVA P–value < 0.05) were involved in fatty acid and steroid metabolism pathways (Bonferroni P-value = 0.0137). Finally, when we compared the transcriptomic and the proteomic data of our mouse cohorts only ~7% of the altered mRNA profiles encoding for ER gene products were consistent with their corresponding protein profiles measured by the label-free quantification methods. These results suggest that a great number of ER gene products are regulated at the post-transcriptional level in the liver of Wrn mutant mice exhibiting an ER stress response.
Collapse
Affiliation(s)
- Lucie Aumailley
- Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
| | - Florence Roux-Dalvai
- Proteomics Platform Center, Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
| | - Isabelle Kelly
- Proteomics Platform Center, Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
| | - Arnaud Droit
- Proteomics Platform Center, Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
| | - Michel Lebel
- Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Quebec City Québec, Canada
- * E-mail:
| |
Collapse
|
150
|
Aumailley L, Dubois MJ, Brennan TA, Garand C, Paquet ER, Pignolo RJ, Marette A, Lebel M. Serum vitamin C levels modulate the lifespan and endoplasmic reticulum stress response pathways in mice synthesizing a nonfunctional mutant WRN protein. FASEB J 2018; 32:3623-3640. [PMID: 29452565 DOI: 10.1096/fj.201701176r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Werner syndrome (WS) is a premature aging disorder caused by mutations in a RecQ-family DNA helicase (WRN). Mice lacking part of the helicase domain of the WRN ortholog exhibit several phenotypic features of WS. In this study, we generated a Wrn mutant line that, like humans, relies entirely on dietary sources of vitamin C (ascorbate) to survive, by crossing them to mice that lack the gulonolactone oxidase enzyme required for ascorbate synthesis. In the presence of 0.01% ascorbate (w/v) in drinking water, double-mutant mice exhibited a severe reduction in lifespan, small size, sterility, osteopenia, and metabolic profiles different from wild-type (WT) mice. Although increasing the dose of ascorbate to 0.4% improved dramatically the phenotypes of double-mutant mice, the metabolic and cytokine profiles were different from age-matched WT mice. Finally, double-mutant mice treated with 0.01% ascorbate revealed a permanent activation of all the 3 branches of the ER stress response pathways due to a severe chronic oxidative stress in the ER compartment. In addition, markers associated with the ubiquitin-proteasome-dependent ER-associated degradation pathway were increased. Augmenting the dose of ascorbate reversed the activation of this pathway to WT levels rendering this pathway a potential therapeutic target in WS.-Aumailley, L., Dubois, M. J., Brennan, T. A., Garand, C., Paquet, E. R., Pignolo, R. J., Marette, A., Lebel, M. Serum vitamin C levels modulate the lifespan and endoplasmic reticulum stress response pathways in mice synthesizing a nonfunctional mutant WRN protein.
Collapse
Affiliation(s)
- Lucie Aumailley
- Centre de Recherche du Centre Hospitalier de l'Université (CHU) de Québec, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Marie Julie Dubois
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Tracy A Brennan
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chantal Garand
- Centre de Recherche du Centre Hospitalier de l'Université (CHU) de Québec, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Eric R Paquet
- Centre de Recherche sur le Cancer de l'Université Laval, Hôpital Hôtel-Dieu de Québec, Quebec City, Quebec, Canada
| | - Robert J Pignolo
- Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - André Marette
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Michel Lebel
- Centre de Recherche du Centre Hospitalier de l'Université (CHU) de Québec, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|