1
|
Faller KME, Chaytow H, Gillingwater TH. Targeting common disease pathomechanisms to treat amyotrophic lateral sclerosis. Nat Rev Neurol 2025; 21:86-102. [PMID: 39743546 DOI: 10.1038/s41582-024-01049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
The motor neuron disease amyotrophic lateral sclerosis (ALS) is a devastating condition with limited treatment options. The past few years have witnessed a ramping up of translational ALS research, offering the prospect of disease-modifying therapies. Although breakthroughs using gene-targeted approaches have shown potential to treat patients with specific disease-causing mutations, the applicability of such therapies remains restricted to a minority of individuals. Therapies targeting more general mechanisms that underlie motor neuron pathology in ALS are therefore of considerable interest. ALS pathology is associated with disruption to a complex array of key cellular pathways, including RNA processing, proteostasis, metabolism and inflammation. This Review details attempts to restore cellular homeostasis by targeting these pathways in order to develop effective, broadly-applicable ALS therapeutics.
Collapse
Affiliation(s)
- Kiterie M E Faller
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Helena Chaytow
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
2
|
Wu CC, Meyer DN, Haimbaugh A, Baker TR. Implications of Lead (Pb)-Induced Transcriptomic and Phenotypic Alterations in the Aged Zebrafish ( Danio rerio). TOXICS 2024; 12:745. [PMID: 39453165 PMCID: PMC11511149 DOI: 10.3390/toxics12100745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024]
Abstract
Lead (Pb) is a well-known neurotoxin with established adverse effects on the neurological functions of children and younger adults, including motor, learning, and memory abilities. However, its potential impact on older adults has received less attention. Using the zebrafish model, our study aims to characterize the dose-response relationship between environmentally relevant Pb exposure levels and their effects on changes in behavior and transcriptomics during the geriatric periods. We exposed two-year-old zebrafish to waterborne lead acetate (1, 10, 100, 1000, or 10,000 µg/L) or a vehicle (DMSO) for 5 days. While lower concentrations (1-100 µg/L) reflect environmentally relevant Pb levels, higher concentrations (1000-10,000 µg/L) were included to assess acute toxicity under extreme exposure scenarios. We conducted adult behavior assessment to evaluate the locomotor activity following exposure. The same individual fish were subsequently sacrificed for brain dissection after a day of recovery in the aquatic system. RNA extraction and sequencing were then performed to evaluate the Pb-induced transcriptomic changes. Higher (1000-10,000 ug/L) Pb levels induced hyperactive locomotor patterns in aged zebrafish, while lower (10-100 ug/L) Pb levels resulted in the lowest locomotor activity compared to the control group. Exposure to 100 µg/L led to the highest number of differentially expressed genes (DEGs), while 10,000 µg/L induced larger fold changes in both directions. The neurological pathways impacted by Pb exposure include functions related to neurotransmission, such as cytoskeletal regulation and synaptogenesis, and oxidative stress response, such as mitochondrial dysfunction and downregulation of heat shock protein genes. These findings emphasize a U-shape dose-response relationship with Pb concentrations in locomotor activity and transcriptomic changes in the aging brain.
Collapse
Affiliation(s)
- Chia-Chen Wu
- Institute of Environmental Engineering, National Yang Ming Chiao Tung University, 1001, Daxue Rd, East District, Hsinchu City 300093, Taiwan;
- Department of Environmental and Global Health, University of Florida, 1225 Center Drive, Gainesville, FL 32610, USA; (D.N.M.)
| | - Danielle N. Meyer
- Department of Environmental and Global Health, University of Florida, 1225 Center Drive, Gainesville, FL 32610, USA; (D.N.M.)
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Alex Haimbaugh
- Department of Environmental and Global Health, University of Florida, 1225 Center Drive, Gainesville, FL 32610, USA; (D.N.M.)
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
| | - Tracie R. Baker
- Department of Environmental and Global Health, University of Florida, 1225 Center Drive, Gainesville, FL 32610, USA; (D.N.M.)
- Department of Pharmacology, School of Medicine, Wayne State University, 540 E. Canfield, Detroit, MI 48201, USA
- UF Genetics Institute, University of Florida, 2033 Mowry Road, Gainesville, FL 32610, USA
| |
Collapse
|
3
|
Koloko Ngassie ML, Drake LY, Roos BB, Koenig-Kappes A, Pabelick CM, Gosens R, Brandsma CA, Burgess JK, Prakash YS. Endoplasmic reticulum stress-induced senescence in human lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2024; 327:L126-L139. [PMID: 38771153 PMCID: PMC11380945 DOI: 10.1152/ajplung.00264.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/08/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
Loss of proteostasis and cellular senescence have been previously established as characteristics of aging; however, their interaction in the context of lung aging and potential contributions to aging-associated lung remodeling remains understudied. In this study, we aimed to characterize endoplasmic reticulum (ER) stress response, cellular senescence, and their interaction in relation to extracellular matrix (ECM) production in lung fibroblasts from young (25-45 yr) and old (>60 yr) humans. Fibroblasts from young and old patients without significant preexisting lung disease were exposed to vehicle, MG132, etoposide, or salubrinal. Afterward, cells and cell lysates or supernatants were analyzed for ER stress, cellular senescence, and ECM changes using protein analysis, proliferation assay, and senescence-associated beta-galactosidase (SA-β-Gal) staining. At baseline, fibroblasts from aging individuals showed increased levels of ER stress (ATF6 and PERK), senescence (p21 and McL-1), and ECM marker (COL1A1) compared to those from young individuals. Upon ER stress induction and etoposide exposure, fibroblasts showed an increase in senescence (SA-β-Gal, p21, and Cav-1), ER stress (PERK), and ECM markers (COL1A1 and LUM) compared to vehicle. Additionally, IL-6 and IL-8 levels were increased in the supernatants of MG132- and etoposide-treated fibroblasts, respectively. Finally, the ER stress inhibitor salubrinal decreased the expression of p21 compared to vehicle and MG132 treatments; however, salubrinal inhibited COL1A1 but not p21 expression in MG132-treated fibroblasts. Our study suggests that ER stress response plays an important role in establishment and maintenance of a senescence phenotype in lung fibroblasts and therefore contributes to altered remodeling in the aging lung.NEW & NOTEWORTHY The current study establishes functional links between endoplasmic reticulum (ER) stress and cellular senescence per se in the specific context of aging human lung fibroblasts. Recognizing that the process of aging per se is complex, modulated by the myriad of lifelong and environmental exposures, it is striking to note that chronic ER stress may play a crucial role in the establishment and maintenance of cellular senescence in lung fibroblasts.
Collapse
Affiliation(s)
- Maunick Lefin Koloko Ngassie
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Li Y Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Benjamin B Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Amanda Koenig-Kappes
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janette K Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
4
|
Françon A, Behar-Cohen F, Torriglia A. The blue light hazard and its use on the evaluation of photochemical risk for domestic lighting. An in vivo study. ENVIRONMENT INTERNATIONAL 2024; 184:108471. [PMID: 38335626 DOI: 10.1016/j.envint.2024.108471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Nowadays artificial light highly increases human exposure to light leading to circadian rhythm and sleep perturbations. Moreover, excessive exposure of ocular structures to photons can induce irreversible retinal damage. Meta-analyses showed that sunlight exposure influences the age of onset and the progression of Age-related macular degeneration (AMD), the leading cause of blindness in people over fifty-year old. Currently, the blue-light hazard (BLH) curve is used in the evaluation of the phototoxicity of a light source for domestic lighting regulations. OBJECTIVES Here, we analyze the phototoxicity threshold in rats and investigate the role played by the light spectrum, assessing the relevance of the use of the BLH-weighting to define phototoxicity. METHODS We exposed albino rats to increasing doses of blue and white light, or to lights of different colors to evaluate the impact of each component of the white light spectrum on phototoxicity. Cellular mechanisms of cell death and cellular stress induced by light were analyzed. RESULTS Our results show that the phototoxicity threshold currently accepted for rats is overestimated by a factor of 50 when considering blue light and by a factor of 550 concerning white light. This is the result of the toxicity induced by green light that increases white light toxicity by promoting an inflammatory response. The content of green in white light induces 8 fold more invasion of macrophages in the retina than the content of blue light. Moreover, the use of BLH-weighting does not evaluate the amount of red radiations contained in white light that mitigates damage by inhibiting the nuclear translocation of L-DNase II and reducing by 33% the number of TUNEL-positive cells. DISCUSSION These findings question the current methods to determine the phototoxicity of a light source and show the necessity to take into account the entire emission spectrum. As current human phototoxicity thresholds were estimated with the same methods used for rats, our results suggest that they might need to be reconsidered.
Collapse
Affiliation(s)
- Anaïs Françon
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université. Team: Physiopathology of Ocular Diseases: Therapeutic Innovations. 15, rue de l'école de Médecine, 75006 Paris, France
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université. Team: Physiopathology of Ocular Diseases: Therapeutic Innovations. 15, rue de l'école de Médecine, 75006 Paris, France; Assistance Publique, Hôpitaux de Paris, Hôpital Cochin, Ophtalmopole, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Alicia Torriglia
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université. Team: Physiopathology of Ocular Diseases: Therapeutic Innovations. 15, rue de l'école de Médecine, 75006 Paris, France.
| |
Collapse
|
5
|
Hebishy M, Shintouo CM, Dufait I, Debacq-Chainiaux F, Bautmans I, Njemini R. Heat shock proteins and cellular senescence in humans: A systematic review. Arch Gerontol Geriatr 2023; 113:105057. [PMID: 37207540 DOI: 10.1016/j.archger.2023.105057] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/07/2023] [Indexed: 05/21/2023]
Abstract
Cellular senescence (CS) is a permanent arrest of cell growth and exit of the cell cycle. It is an important tumor suppression mechanism and has a key role in wound healing, tissue regeneration, and prevention of tissue fibrosis. Despite the short-term benefits of CS, accumulation of senescent cells has deleterious effects and is associated with several pathological age-related phenotypes. As Heat Shock Proteins (HSP) are associated with cyto-protection, their role in longevity and CS became a research interest. However, an overview of the relationship between HSP and CS in humans still lacks in the literature. To provide an overview of the current state of the literature, this systematic review focused on the role of HSP in the development of CS in humans. PubMed, Web of Science and Embase were systematically screened for studies on the relationship between HSP and CS in humans. A total of 14 articles were eligible for inclusion. The heterogeneity and lack of numerical reporting of outcomes obstructed the conduction of a meta-analysis. The results consistently show that HSP depletion results in increased CS, while overexpression of HSP decreases CS, whether in cancer, fibroblasts, or stem cell lines. This systematic review summarized the literature on the prospective role of HSP in the development of CS in humans.
Collapse
Affiliation(s)
- Mariam Hebishy
- Department of Gerontology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium
| | - Cabirou Mounchili Shintouo
- Department of Gerontology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium; Frailty in Ageing Research Group, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium; Department of Biochemistry and Molecular Biology, Faculty of Science, University of Buea, P.O. Box 63 Buea, Cameroon
| | - Ines Dufait
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, Brussels 1090, Belgium
| | - Florence Debacq-Chainiaux
- Research Unit on Cellular Biology (URBC), Department of Biology, University of Namur, Rue de Bruxelles, 61, Namur B-5000, Belgium
| | - Ivan Bautmans
- Department of Gerontology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium; Frailty in Ageing Research Group, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium
| | - Rose Njemini
- Department of Gerontology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium; Frailty in Ageing Research Group, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels 1090, Belgium.
| |
Collapse
|
6
|
Liu AY, Minetti CA, Remeta DP, Breslauer KJ, Chen KY. HSF1, Aging, and Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:23-49. [PMID: 35995906 DOI: 10.1007/5584_2022_733] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heat shock factor 1 (HSF1) is a master transcription regulator that mediates the induction of heat shock protein chaperones for quality control (QC) of the proteome and maintenance of proteostasis as a protective mechanism in response to stress. Research in this particular area has accelerated dramatically over the past three decades following successful isolation, cloning, and characterization of HSF1. The intricate multi-protein complexes and transcriptional activation orchestrated by HSF1 are fundamental processes within the cellular QC machinery. Our primary focus is on the regulation and function of HSF1 in aging and neurodegenerative diseases (ND) which represent physiological and pathological states of dysfunction in protein QC. This chapter presents an overview of HSF1 structural, functional, and energetic properties in healthy cells while addressing the deterioration of HSF1 function viz-à-viz age-dependent and neuron-specific vulnerability to ND. We discuss the structural domains of HSF1 with emphasis on the intrinsically disordered regions and note that disease proteins associated with ND are often structurally disordered and exquisitely sensitive to changes in cellular environment as may occur during aging. We propose a hypothesis that age-dependent changes of the intrinsically disordered proteome likely hold answers to understand many of the functional, structural, and organizational changes of proteins and signaling pathways in aging - dysfunction of HSF1 and accumulation of disease protein aggregates in ND included.Structured AbstractsIntroduction: Heat shock factor 1 (HSF1) is a master transcription regulator that mediates the induction of heat shock protein chaperones for quality control (QC) of the proteome as a cyto-protective mechanism in response to stress. There is cumulative evidence of age-related deterioration of this QC mechanism that contributes to disease vulnerability. OBJECTIVES Herein we discuss the regulation and function of HSF1 as they relate to the pathophysiological changes of protein quality control in aging and neurodegenerative diseases (ND). METHODS We present an overview of HSF1 structural, functional, and energetic properties in healthy cells while addressing the deterioration of HSF1 function vis-à-vis age-dependent and neuron-specific vulnerability to neurodegenerative diseases. RESULTS We examine the impact of intrinsically disordered regions on the function of HSF1 and note that proteins associated with neurodegeneration are natively unstructured and exquisitely sensitive to changes in cellular environment as may occur during aging. CONCLUSIONS We put forth a hypothesis that age-dependent changes of the intrinsically disordered proteome hold answers to understanding many of the functional, structural, and organizational changes of proteins - dysfunction of HSF1 in aging and appearance of disease protein aggregates in neurodegenerative diseases included.
Collapse
Affiliation(s)
- Alice Y Liu
- Department of Cell Biology and Neuroscience, Rutgers The State University of New Jersey, Piscataway, NJ, USA.
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA.
| | - Conceição A Minetti
- Department of Chemistry and Chemical Biology, Rutgers The State University of New Jersey, Piscataway, NJ, USA
| | - David P Remeta
- Department of Chemistry and Chemical Biology, Rutgers The State University of New Jersey, Piscataway, NJ, USA
| | - Kenneth J Breslauer
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Department of Chemistry and Chemical Biology, Rutgers The State University of New Jersey, Piscataway, NJ, USA
| | - Kuang Yu Chen
- Department of Chemistry and Chemical Biology, Rutgers The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
7
|
Kim H, Gomez-Pastor R. HSF1 and Its Role in Huntington's Disease Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1410:35-95. [PMID: 36396925 DOI: 10.1007/5584_2022_742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
PURPOSE OF REVIEW Heat shock factor 1 (HSF1) is the master transcriptional regulator of the heat shock response (HSR) in mammalian cells and is a critical element in maintaining protein homeostasis. HSF1 functions at the center of many physiological processes like embryogenesis, metabolism, immune response, aging, cancer, and neurodegeneration. However, the mechanisms that allow HSF1 to control these different biological and pathophysiological processes are not fully understood. This review focuses on Huntington's disease (HD), a neurodegenerative disease characterized by severe protein aggregation of the huntingtin (HTT) protein. The aggregation of HTT, in turn, leads to a halt in the function of HSF1. Understanding the pathways that regulate HSF1 in different contexts like HD may hold the key to understanding the pathomechanisms underlying other proteinopathies. We provide the most current information on HSF1 structure, function, and regulation, emphasizing HD, and discussing its potential as a biological target for therapy. DATA SOURCES We performed PubMed search to find established and recent reports in HSF1, heat shock proteins (Hsp), HD, Hsp inhibitors, HSF1 activators, and HSF1 in aging, inflammation, cancer, brain development, mitochondria, synaptic plasticity, polyglutamine (polyQ) diseases, and HD. STUDY SELECTIONS Research and review articles that described the mechanisms of action of HSF1 were selected based on terms used in PubMed search. RESULTS HSF1 plays a crucial role in the progression of HD and other protein-misfolding related neurodegenerative diseases. Different animal models of HD, as well as postmortem brains of patients with HD, reveal a connection between the levels of HSF1 and HSF1 dysfunction to mutant HTT (mHTT)-induced toxicity and protein aggregation, dysregulation of the ubiquitin-proteasome system (UPS), oxidative stress, mitochondrial dysfunction, and disruption of the structural and functional integrity of synaptic connections, which eventually leads to neuronal loss. These features are shared with other neurodegenerative diseases (NDs). Currently, several inhibitors against negative regulators of HSF1, as well as HSF1 activators, are developed and hold promise to prevent neurodegeneration in HD and other NDs. CONCLUSION Understanding the role of HSF1 during protein aggregation and neurodegeneration in HD may help to develop therapeutic strategies that could be effective across different NDs.
Collapse
Affiliation(s)
- Hyuck Kim
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
8
|
Perez FP, Bandeira JP, Perez Chumbiauca CN, Lahiri DK, Morisaki J, Rizkalla M. Multidimensional insights into the repeated electromagnetic field stimulation and biosystems interaction in aging and age-related diseases. J Biomed Sci 2022; 29:39. [PMID: 35698225 PMCID: PMC9190166 DOI: 10.1186/s12929-022-00825-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/07/2022] [Indexed: 11/25/2022] Open
Abstract
We provide a multidimensional sequence of events that describe the electromagnetic field (EMF) stimulation and biological system interaction. We describe this process from the quantum to the molecular, cellular, and organismal levels. We hypothesized that the sequence of events of these interactions starts with the oscillatory effect of the repeated electromagnetic stimulation (REMFS). These oscillations affect the interfacial water of an RNA causing changes at the quantum and molecular levels that release protons by quantum tunneling. Then protonation of RNA produces conformational changes that allow it to bind and activate Heat Shock Transcription Factor 1 (HSF1). Activated HSF1 binds to the DNA expressing chaperones that help regulate autophagy and degradation of abnormal proteins. This action helps to prevent and treat diseases such as Alzheimer's and Parkinson's disease (PD) by increasing clearance of pathologic proteins. This framework is based on multiple mathematical models, computer simulations, biophysical experiments, and cellular and animal studies. Results of the literature review and our research point towards the capacity of REMFS to manipulate various networks altered in aging (Reale et al. PloS one 9, e104973, 2014), including delay of cellular senescence (Perez et al. 2008, Exp Gerontol 43, 307-316) and reduction in levels of amyloid-β peptides (Aβ) (Perez et al. 2021, Sci Rep 11, 621). Results of these experiments using REMFS at low frequencies can be applied to the treatment of patients with age-related diseases. The use of EMF as a non-invasive therapeutic modality for Alzheimer's disease, specifically, holds promise. It is also necessary to consider the complicated and interconnected genetic and epigenetic effects of the REMFS-biological system's interaction while avoiding any possible adverse effects.
Collapse
Affiliation(s)
- Felipe P Perez
- Indiana University School of Medicine, Indianapolis, IN, USA.
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Joseph P Bandeira
- Indiana University School of Medicine, Indianapolis, IN, USA
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cristina N Perez Chumbiauca
- Indiana University School of Medicine, Indianapolis, IN, USA
- Division of Rheumatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Debomoy K Lahiri
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jorge Morisaki
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Maher Rizkalla
- Department of Electrical and Computer Engineering, Indiana University-Purdue University, Indianapolis, IN, USA
| |
Collapse
|
9
|
Madelaire CB, Klink AC, Israelsen WJ, Hindle AG. Fibroblasts as an experimental model system for the study of comparative physiology. Comp Biochem Physiol B Biochem Mol Biol 2022; 260:110735. [PMID: 35321853 DOI: 10.1016/j.cbpb.2022.110735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 10/18/2022]
Abstract
Mechanistic evaluations of processes that underlie organism-level physiology often require reductionist approaches. Dermal fibroblasts offer one such approach. These cells are easily obtained from minimally invasive skin biopsy, making them appropriate for the study of protected and/or logistically challenging species. Cell culture approaches permit extensive and fine-scale sampling regimes as well as gene manipulation techniques that are not feasible in vivo. Fibroblast isolation and culture protocols are outlined here for primary cells, and the benefits and drawbacks of immortalization are discussed. We show examples of physiological metrics that can be used to characterize primary cells (oxygen consumption, translation, proliferation) and readouts that can be informative in understanding cell-level responses to environmental stress (lactate production, heat shock protein induction). Importantly, fibroblasts may display fidelity to whole animal physiological phenotypes, facilitating their study. Fibroblasts from Antarctic Weddell seals show greater resilience to low temperatures and hypoxia exposure than fibroblasts from humans or rats. Fibroblast oxygen consumption rates are not affected by temperature stress in the heat-tolerant camel, whereas similar temperature exposures depress mitochondrial metabolism in fibroblasts from rhinoceros. Finally, dermal fibroblasts from a hibernator, the meadow jumping mouse, better resist experimental cooling than a fibroblast line from the laboratory mouse, with the hibernator demonstrating a greater maintenance of homeostatic processes such as protein translation. These results exemplify the parallels that can be drawn between fibroblast physiology and expectations in vivo, and provide evidence for the power of fibroblasts as a model system to understand comparative physiology and biomedicine.
Collapse
Affiliation(s)
- Carla B Madelaire
- School of Life Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Amy C Klink
- School of Life Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - William J Israelsen
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Skroot Laboratory, Inc., Ames, IA, USA
| | - Allyson G Hindle
- School of Life Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA.
| |
Collapse
|
10
|
Koloko Ngassie ML, Brandsma CA, Gosens R, Prakash YS, Burgess JK. The Stress of Lung Aging: Endoplasmic Reticulum and Senescence Tête-à-Tête. Physiology (Bethesda) 2021; 36:150-159. [PMID: 33904785 DOI: 10.1152/physiol.00039.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Beyond the structural changes, features including the dysregulation of endoplasmic reticulum (ER) stress response and increased senescence characterize the lung aging. ER stress response and senescence have been reported to be induced by factors like cigarette smoke. Therefore, deciphering the mechanisms underlying ER and senescent pathways interaction has become a challenge. In this review we highlight the known and unknown regarding ER stress response and senescence and their cross talk in aged lung.
Collapse
Affiliation(s)
- M L Koloko Ngassie
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - C A Brandsma
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - R Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD; University of Groningen, Department of Molecular Pharmacology, Groningen, The Netherlands
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - J K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| |
Collapse
|
11
|
Jurivich DA, Manocha GD, Trivedi R, Lizakowski M, Rakoczy S, Brown-Borg H. Multifactorial Attenuation of the Murine Heat Shock Response With Age. J Gerontol A Biol Sci Med Sci 2021; 75:1846-1852. [PMID: 31612204 DOI: 10.1093/gerona/glz204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Indexed: 01/08/2023] Open
Abstract
Age-dependent perturbation of the cellular stress response affects proteostasis and other key functions relevant to cellular action and survival. Central to age-related changes in the stress response is loss of heat shock factor 1 (HSF1)-DNA binding and transactivation properties. This report elucidates how age alters different checkpoints of HSF1 activation related to posttranslational modification and protein interactions. When comparing liver extracts from middle aged (12 M) and old (24 M) mice, significant differences are found in HSF1 phosphorylation and acetylation. HSF1 protein levels and messenger RNA decline with age, but its protein levels are stress-inducible and exempt from age-dependent changes. This surprising adaptive change in the stress response has additional implications for aging and chronic physiological stress that might explain an age-dependent dichotomy of HSF1 protein levels that are low in neurodegeneration and elevated in cancer.
Collapse
Affiliation(s)
- Donald A Jurivich
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Gunjan D Manocha
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Rachana Trivedi
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Mary Lizakowski
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Sharlene Rakoczy
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Holly Brown-Borg
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| |
Collapse
|
12
|
Korfei M, MacKenzie B, Meiners S. The ageing lung under stress. Eur Respir Rev 2020; 29:29/156/200126. [DOI: 10.1183/16000617.0126-2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 01/10/2023] Open
Abstract
Healthy ageing of the lung involves structural changes but also numerous cell-intrinsic and cell-extrinsic alterations. Among them are the age-related decline in central cellular quality control mechanisms such as redox and protein homeostasis. In this review, we would like to provide a conceptual framework of how impaired stress responses in the ageing lung, as exemplified by dysfunctional redox and protein homeostasis, may contribute to onset and progression of COPD and idiopathic pulmonary fibrosis (IPF). We propose that age-related imbalanced redox and protein homeostasis acts, amongst others (e.g.cellular senescence), as a “first hit” that challenges the adaptive stress-response pathways of the cell, increases the level of oxidative stress and renders the lung susceptible to subsequent injury and disease. In both COPD and IPF, additional environmental insults such as smoking, air pollution and/or infections then serve as “second hits” which contribute to persistently elevated oxidative stress that overwhelms the already weakened adaptive defence and repair pathways in the elderly towards non-adaptive, irremediable stress thereby promoting development and progression of respiratory diseases. COPD and IPF are thus distinct horns of the same devil, “lung ageing”.
Collapse
|
13
|
Molecular Chaperones and Proteolytic Machineries Regulate Protein Homeostasis In Aging Cells. Cells 2020; 9:cells9051308. [PMID: 32456366 PMCID: PMC7291254 DOI: 10.3390/cells9051308] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022] Open
Abstract
Throughout their life cycles, cells are subject to a variety of stresses that lead to a compromise between cell death and survival. Survival is partially provided by the cell proteostasis network, which consists of molecular chaperones, a ubiquitin-proteasome system of degradation and autophagy. The cooperation of these systems impacts the correct function of protein synthesis/modification/transport machinery starting from the adaption of nascent polypeptides to cellular overcrowding until the utilization of damaged or needless proteins. Eventually, aging cells, in parallel to the accumulation of flawed proteins, gradually lose their proteostasis mechanisms, and this loss leads to the degeneration of large cellular masses and to number of age-associated pathologies and ultimately death. In this review, we describe the function of proteostasis mechanisms with an emphasis on the possible associations between them.
Collapse
|
14
|
Hwang HV, Lin Y, Rebuffatti MN, Tran DT, Lee L, Gomes AV, Li CS, Knowlton AA. Impaired proteostasis in senescent vascular endothelial cells: a perspective on estrogen and oxidative stress in the aging vasculature. Am J Physiol Heart Circ Physiol 2018; 316:H421-H429. [PMID: 30499713 DOI: 10.1152/ajpheart.00318.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The heat shock response is an important cytoprotective mechanism for protein homeostasis and is an essential protective response to cellular stress and injury. Studies on changes in the heat shock response with aging have been mixed with regard to whether it is inhibited, and this, at least in part, reflects different tissues and different models. Cellular senescence is a key feature in aging, but work on the heat shock response in cultured senescent (SEN) cells has largely been limited to fibroblasts. Given the prevalence of oxidative injury in the aging cardiovascular system, we investigated whether SEN primary human coronary artery endothelial cells have a diminished heat shock response and impaired proteostasis. In addition, we tested whether this downregulation of heat shock response can be mitigated by 17β-estradiol (E2), which has a critical cardioprotective role in women, as we have previously reported that E2 improves the heat shock response in endothelial cells (Hamilton KL, Mbai FN, Gupta S, Knowlton AA. Arterioscler Thromb Vasc Biol 24: 1628-1633, 2004). We found that SEN endothelial cells, despite their unexpectedly increased proteasome activity, had a diminished heat shock response and had more protein aggregation than early passage cells. SEN cells had increased oxidative stress, which promoted protein aggregation. E2 treatment did not decrease protein aggregation or improve the heat shock response in either early passage or SEN cells. In summary, cellular senescence in adult human endothelial cells is accompanied by increased oxidative stress and a blunting of proteostasis, and E2 did not mitigate these changes. NEW & NOTEWORTHY Senescent human endothelial cells have a diminished heat shock response and increased protein aggregates. Senescent human endothelial cells have increased basal oxidative stress, which increases protein aggregates. Physiological level of 17β-estradiol did not improve proteostasis in endothelial cells.
Collapse
Affiliation(s)
- HyunTae V Hwang
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Yun Lin
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Michelle N Rebuffatti
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Darlene T Tran
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Lily Lee
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California.,Department of Physiology and Membrane Biology, University of California, Davis, California
| | - Chin-Shang Li
- School of Nursing, The State University of New York, University at Buffalo, Buffalo, New York
| | - Anne A Knowlton
- Veterans Affairs Medical Center, Sacramento, California.,Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California.,Department of Pharmacology, University of California, Davis, California
| |
Collapse
|
15
|
Proteomic profiling of senescent human diploid fibroblasts treated with gamma-tocotrienol. Altern Ther Health Med 2018; 18:314. [PMID: 30497457 PMCID: PMC6267793 DOI: 10.1186/s12906-018-2383-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 11/22/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Replicative senescence of human diploid fibroblasts (HDFs) has been used as a model to study mechanisms of cellular aging. Gamma-tocotrienol (γT3) is one of the members of vitamin E family which has been shown to increase proliferation of senescent HDFs. However, the modulation of protein expressions by γT3 in senescent HDFs remains to be elucidated. Therefore, this study aimed to determine the differentially expressed proteins (DEPs) in young and senescent HDFs; and in vehicle- and γT3-treated senescent HDFs using label-free quantitative proteomics. METHODS Whole proteins were extracted and digested in-gel with trypsin. Peptides were detected by Orbitrap liquid chromatography mass spectrometry. Mass spectra were identified and quantitated by MaxQuant software. The data were further filtered and analyzed statistically using Perseus software to identify DEPs. Functional annotations of DEPs were performed using Panther Classification System. RESULTS A total of 1217 proteins were identified in young and senescent cells, while 1218 proteins in vehicle- and γT3-treated senescent cells. 11 DEPs were found in young and senescent cells which included downregulation of platelet-derived growth factor (PDGF) receptor beta and upregulation of tubulin beta-2A chain protein expressions in senescent cells. 51 DEPs were identified in vehicle- and γT3-treated senescent cells which included upregulation of 70 kDa heat shock protein, triosephosphate isomerase and malate dehydrogenase protein expressions in γT3-treated senescent cells. CONCLUSIONS PDGF signaling and cytoskeletal structure may be dysregulated in senescent HDFs. The pro-proliferative effect of γT3 on senescent HDFs may be mediated through the stimulation of cellular response to stress and carbohydrate metabolism. The expressions and roles of these proteins in relation to cellular senescence are worth further investigations. Data are available via ProteomeXchange with identifier PXD009933.
Collapse
|
16
|
Abstract
Amyloid fibrils are protein homopolymers that adopt diverse cross-β conformations. Some amyloid fibrils are associated with the pathogenesis of devastating neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. Conversely, functional amyloids play beneficial roles in melanosome biogenesis, long-term memory formation and release of peptide hormones. Here, we showcase advances in our understanding of amyloid assembly and structure, and how distinct amyloid strains formed by the same protein can cause distinct neurodegenerative diseases. We discuss how mutant steric zippers promote deleterious amyloidogenesis and aberrant liquid-to-gel phase transitions. We also highlight effective strategies to combat amyloidogenesis and related toxicity, including: (1) small-molecule drugs (e.g. tafamidis) to inhibit amyloid formation or (2) stimulate amyloid degradation by the proteasome and autophagy, and (3) protein disaggregases that disassemble toxic amyloid and soluble oligomers. We anticipate that these advances will inspire therapeutics for several fatal neurodegenerative diseases. Summary: This Review showcases important advances in our understanding of amyloid structure, assembly and disassembly, which are inspiring novel therapeutic strategies for amyloid disorders.
Collapse
Affiliation(s)
- Edward Chuang
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Acacia M Hori
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christina D Hesketh
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA .,Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
Xing W, Gao W, Su H, Wang S, Zhang J, Mao G, Yan J. Salidroside influences the cellular cross-talk of human fetal lung diploid fibroblasts: A proteomic approach. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 58:114-121. [PMID: 29329019 DOI: 10.1016/j.etap.2018.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/04/2018] [Indexed: 06/07/2023]
Abstract
Senescence is a complex multiple factor proces, which is still poorly understood. The purpose of this study was to find the proteome of cultured human fetal lung diploid fibroblasts (2BS) of different population doubling (PD), as well as the altered proteome induced by salidroside (SAL) in 2BS cells. Proteins were identified by two-dimensional electrophoresis (2-DE) combining matrix-assisted laser desorption/ionization-time and flight mass spectrometry (MAL DI-TOF/MS). As a result, we found 16 proteins with two-fold variations in senescent cells or after SAL treatment, some being reduced such as reticulocalbin-1, heat shock protein beta-6, elongation factor 1-delta, F-actin-capping protein subunit alpha-1, and chloride intracellular channel 1. In contrast, 40S ribosomal protein SA, proteasome subunit alpha type-5, and zinc finger BED domain-containing protein 5 increased with cell age. Furthermore, heat shock protein beta-6, Zinc finger BED domain-containing protein 5 was increased in PD30 cells after 10 μM SAL treatment, whereas, elongation factor 1-delta, 6-phosphogluconolactonase, Nucleoside diphosphate kinase A, F-actin-capping protein subunit alpha-1, Probable ATP-dependent RNA helicase DDX41, Chloride intracellular channel 1, and Peroxiredoxin-6 were increased in PD50 cells after 10 μM SAL treatment. Some of these proteins were involved in the protein synthetic and degradative pathways, which emphasizes the metabolic disorder or functional impairment of cell senescence. Moreover, these proteins could be candidate biomarkers for evaluating the SAL anti-senescence effect.
Collapse
Affiliation(s)
- Wenmin Xing
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Wenyan Gao
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, China
| | - Huili Su
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Sanying Wang
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Jing Zhang
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China.
| | - Jing Yan
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China.
| |
Collapse
|
18
|
Hwang HV, Tran DT, Rebuffatti MN, Li CS, Knowlton AA. Investigation of quercetin and hyperoside as senolytics in adult human endothelial cells. PLoS One 2018; 13:e0190374. [PMID: 29315311 PMCID: PMC5760026 DOI: 10.1371/journal.pone.0190374] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/13/2017] [Indexed: 01/09/2023] Open
Abstract
Quercetin has been reported to act as a senolytic by selectively removing senescent endothelial cells, and thus it would seem quercetin could revolutionize the field of gerontology. However, given quercetin's narrow therapeutic index reported in work done with human umbilical vein endothelial cells (HUVECs), we hypothesized that quercetin is not innocuous for non-senescent adult human vascular endothelial cells at concentrations that have been reported to be safe for proliferating HUVECs. Furthermore, we investigated quercetin 3-D-galactoside (Q3G; hyperoside), an inactive quercetin derivative that needs to be cleaved by beta-galactosidase overexpressed in senescent cells to release quercetin, as a potential safer senolytic. We compared the effectiveness of quercetin and Q3G in primary human coronary artery endothelial cells (HCAEC), which are adult microvascular cells. We found that quercetin caused cell death in non-senescent endothelial cells at a concentration that has been reported to selectively remove senescent cells, and that Q3G was not cytotoxic to either young or senescent cells. Thus, in primary adult human endothelial cells, quercetin and Q3G are not senolytics. Earlier work reporting positive results was done with HUVECs, and given their origin and the disparate findings from the current study, these may not be the best cells for evaluating potential senolytics in clinically relevant endothelial cells.
Collapse
Affiliation(s)
- HyunTae V. Hwang
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Darlene Thuy Tran
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Michelle Nicole Rebuffatti
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Chin-Shang Li
- Division of Biostatistics, Department of Public Health Sciences, University of California-Davis, Davis, CA, United States of America
| | - Anne A. Knowlton
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
- VA Medical Center, Sacramento, CA, United States of America
- Pharmacology Department, University of California-Davis, Davis, CA, United States of America
- * E-mail:
| |
Collapse
|
19
|
Brain Aging: Hsp90 and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1086:93-103. [PMID: 30232754 DOI: 10.1007/978-981-13-1117-8_6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The brain is the most complex organ in the human body and the main component of the central nervous system. Because it lacks the ability of regeneration, age is a major risk factor for most common neurodegenerative diseases, which caused an irreversible cognitive impairment. It has been shown that the function of molecular chaperones, majorly heat shock proteins, was compromised and then causes the imbalance of protein homeostasis inside the cell, which is the most influential reason of brain aging. Here, in this review, we discuss the mechanisms underneath the impairment of heat shock protein function during brain aging, including transcriptional regulation, posttranslational modification, and communication across cells and organs.
Collapse
|
20
|
Triplett JC, Tramutola A, Swomley A, Kirk J, Grimes K, Lewis K, Orr M, Rodriguez K, Cai J, Klein JB, Perluigi M, Buffenstein R, Butterfield DA. Age-related changes in the proteostasis network in the brain of the naked mole-rat: Implications promoting healthy longevity. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1852:2213-24. [PMID: 26248058 PMCID: PMC4845741 DOI: 10.1016/j.bbadis.2015.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/13/2015] [Accepted: 08/01/2015] [Indexed: 12/18/2022]
Abstract
The naked mole-rat (NMR) is the longest-lived rodent and possesses several exceptional traits: marked cancer resistance, negligible senescence, prolonged genomic integrity, pronounced proteostasis, and a sustained health span. The underlying molecular mechanisms that contribute to these extraordinary attributes are currently under investigation to gain insights that may conceivably promote and extend human health span and lifespan. The ubiquitin-proteasome and autophagy-lysosomal systems play a vital role in eliminating cellular detritus to maintain proteostasis and have been previously shown to be more robust in NMRs when compared with shorter-lived rodents. Using a 2-D PAGE proteomics approach, differential expression and phosphorylation levels of proteins involved in proteostasis networks were evaluated in the brains of NMRs in an age-dependent manner. We identified 9 proteins with significantly altered levels and/or phosphorylation states that have key roles involved in proteostasis networks. To further investigate the possible role that autophagy may play in maintaining cellular proteostasis, we examined aspects of the PI3K/Akt/mammalian target of rapamycin (mTOR) axis as well as levels of Beclin-1, LC3-I, and LC3-II in the brain of the NMR as a function of age. Together, these results show that NMRs maintain high levels of autophagy throughout the majority of their lifespan and may contribute to the extraordinary health span of these rodents. The potential of augmenting human health span via activating the proteostasis network will require further studies.
Collapse
Affiliation(s)
- Judy C Triplett
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, United States
| | - Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Aaron Swomley
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, United States
| | - Jessime Kirk
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, United States
| | - Kelly Grimes
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX 78245, United States; Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78245, United States
| | - Kaitilyn Lewis
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX 78245, United States; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78245, United States
| | - Miranda Orr
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX 78245, United States; Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78245, United States
| | - Karl Rodriguez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX 78245, United States; Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78245, United States
| | - Jian Cai
- Department of Nephrology and Proteomics Center, University of Louisville, Louisville, KY 40202, United States
| | - Jon B Klein
- Department of Nephrology and Proteomics Center, University of Louisville, Louisville, KY 40202, United States
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Rochelle Buffenstein
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX 78245, United States; Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78245, United States.
| | - D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, United States; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
21
|
Babizhayev MA, Yegorov YE. Tissue formation and tissue engineering through host cell recruitment or a potential injectable cell-based biocomposite with replicative potential: Molecular mechanisms controlling cellular senescence and the involvement of controlled transient telomerase activation therapies. J Biomed Mater Res A 2015; 103:3993-4023. [PMID: 26034007 DOI: 10.1002/jbm.a.35515] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 05/18/2015] [Indexed: 01/04/2023]
Abstract
Accumulated data indicate that wound-care products should have a composition equivalent to that of the skin: a combination of particular growth factors and extracellular matrix (ECM) proteins endogenous to the skin, together with viable epithelial cells, fibroblasts, and mesenchymal stem cells (MSCs). Strategies consisting of bioengineered dressings and cell-based products have emerged for widespread clinical use; however, their performance is not optimal because chronic wounds persist as a serious unmet medical need. Telomerase, the ribonucleoprotein complex that adds telomeric repeats to the ends of chromosomes, is responsible for telomere maintenance, and its expression is associated with cell immortalization and, in certain cases, cancerogenesis. Telomerase contains a catalytic subunit, the telomerase reverse transcriptase (hTERT). Introduction of TERT into human cells extends both their lifespan and their telomeres to lengths typical of young cells. The regulation of TERT involves transcriptional and posttranscriptional molecular biology mechanisms. The manipulation, regulation of telomerase is multifactorial in mammalian cells, involving overall telomerase gene expression, post-translational protein-protein interactions, and protein phosphorylation. Reactive oxygen species (ROS) have been implicated in aging, apoptosis, and necrosis of cells in numerous diseases. Upon production of high levels of ROS from exogenous or endogenous generators, the redox balance is perturbed and cells are shifted into a state of oxidative stress, which subsequently leads to modifications of intracellular proteins and membrane lipid peroxidation and to direct DNA damage. When the oxidative stress is severe, survival of the cell is dependent on the repair or replacement of damaged molecules, which can result in induction of apoptosis in the injured with ROS cells. ROS-mediated oxidative stress induces the depletion of hTERT from the nucleus via export through the nuclear pores. Nuclear export is initiated by ROS-induced phosphorylation of tyrosine 707 within hTERT by the Src kinase family. It might be presumed that protection of mitochondria against oxidative stress is an important telomere length-independent function for telomerase in cell survival. Biotechnology companies are focused on development of therapeutic telomerase vaccines, telomerase inhibitors, and telomerase promoter-driven cell killing in oncology, have a telomerase antagonist in late preclinical studies. Anti-aging medicine-oriented groups have intervened on the market with products working on telomerase activation for a broad range of degenerative diseases in which replicative senescence or telomere dysfunction may play an important role. Since oxidative damage has been shown to shorten telomeres in tissue culture models, the adequate topical, transdermal, or systemic administration of antioxidants (such as, patented ocular administration of 1% N-acetylcarnosine lubricant eye drops in the treatment of cataracts) may be beneficial at preserving telomere lengths and delaying the onset or in treatment of disease in susceptible individuals. Therapeutic strategies toward controlled transient activation of telomerase are targeted to cells and replicative potential in cell-based therapies, tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mark A Babizhayev
- Innovative Vision Products, Inc., 3511 Silverside Road, Suite 105, County of New Castle, Delaware, 19810
| | - Yegor E Yegorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov Street, Moscow, 119991, Russian Federation
| |
Collapse
|
22
|
Stimulation of the Fibrillar Collagen and Heat Shock Proteins by Nicotinamide or Its Derivatives in Non-Irradiated or UVA Radiated Fibroblasts, and Direct Anti-Oxidant Activity of Nicotinamide Derivatives. COSMETICS 2015. [DOI: 10.3390/cosmetics2020146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
23
|
Differential translocation of heat shock factor-1 after mild and severe stress to human skin fibroblasts undergoing aging in vitro. J Cell Commun Signal 2014; 8:333-9. [PMID: 25193128 DOI: 10.1007/s12079-014-0244-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 09/01/2014] [Indexed: 01/09/2023] Open
Abstract
Repeated exposure to mild heat shock (HS) has been shown to induce a wide range of health promoting hormetic effects in various biological systems, including human cells undergoing aging in vitro. In order to understand how cells distinguish between mild and severe stress, we have investigated the extent of early and immediate HS response by analyzing the nuclear translocation of the transcription factor heat shock factor-1 (HSF1), in serially passaged normal adult human facial skin fibroblasts exposed to mild (41 °C) or severe (43 °C) HS. Cells respond differently when exposed to mild and severe HS at different passage levels in terms of the extent of HSF1 translocation. In early passage young cells there was a 5-fold difference between mild and severe HS in the extent of HSF1 translocation. However, in near senescent late passage cells, the difference between mild and severe stress in terms of the extent of HSF1 translocation was reduced to less than 2-fold. One of the reasons for this age-related attenuation of heat shock response is due to the fact there was a higher basal level of HSF1 in the nuclei of late passage cells, which is indicative of increased intrinsic stress during cellular aging. These observations are consistent with previously reported data that whereas repeated mild stress given at younger ages can slow down aging and increase the lifespan, the same level of stress given at older ages may not provide the same benefits. Therefore, elucidating the early and immediate steps in the induction of stress response can be useful in deciding whether a particular level of stress is potentially hormetically beneficial or not.
Collapse
|
24
|
Beneficial Regulation of Elastase Activity and Expression of Tissue Inhibitors of Matrixmetalloproteinases, Fibrillin, Transforming Growth Factor-β, and Heat Shock Proteins by P. leucotomos in Nonirradiated or Ultraviolet-Radiated Epidermal Keratinocytes. ACTA ACUST UNITED AC 2013. [DOI: 10.1155/2013/257463] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is loss of the structural integrity of the extracellular matrix (ECM) with intrinsic aging as well as photoaging, largely due to reactive oxygen species (ROS). The structural ECM proteins include the microfibrils that are composed of fibrillin. The structural ECM proteins are primarily degraded by the matrixmetalloproteinases (MMPs) and elastase enzymes. The MMPs are inhibited by the tissue inhibitors of MMPs (TIMPs). A primary regulator of the ECM proteins is transforming growth factor-β (TGF-β), and the chaperone proteins important for its formation are the heat shock proteins (HSP). P. leucotomos extract beneficially regulates of MMPs, TIMPs, and TGF-β in nonirradiated or ultraviolet (UV) radiated fibroblasts and melanoma cells. The hypothesis of this research was that the antioxidant activity or chemistry of P. leucotomos extract would also directly inhibit elastase activity, stimulate the cellular expression of TIMPs, fibrillins, and TGF-β, and regulate HSPs in nonirradiated and UVA or UVB radiated epidermal keratinocytes. P. leucotomos directly inhibited elastase activity, stimulated the cellular expression of TIMPs, fibrillins, and TGF-β, and differentially regulated HSPs in nonirradiated and UVA or UVB radiated epidermal keratinocytes. We infer that the P. leucotomos extract strengthens the ECM and is effective in the prevention or treatment of intrinsic and photoaging of skin.
Collapse
|
25
|
Cloutier P, Coulombe B. Regulation of molecular chaperones through post-translational modifications: decrypting the chaperone code. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:443-54. [PMID: 23459247 DOI: 10.1016/j.bbagrm.2013.02.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 02/15/2013] [Accepted: 02/19/2013] [Indexed: 12/30/2022]
Abstract
Molecular chaperones and their associated cofactors form a group of highly specialized proteins that orchestrate the folding and unfolding of other proteins and the assembly and disassembly of protein complexes. Chaperones are found in all cell types and organisms, and their activity must be tightly regulated to maintain normal cell function. Indeed, deregulation of protein folding and protein complex assembly is the cause of various human diseases. Here, we present the results of an extensive review of the literature revealing that the post-translational modification (PTM) of chaperones has been selected during evolution as an efficient mean to regulate the activity and specificity of these key proteins. Because the addition and reciprocal removal of chemical groups can be triggered very rapidly, this mechanism provides an efficient switch to precisely regulate the activity of chaperones on specific substrates. The large number of PTMs detected in chaperones suggests that a combinatory code is at play to regulate function, activity, localization, and substrate specificity for this group of biologically important proteins. This review surveys the core information currently available as a starting point toward the more ambitious endeavor of deciphering the "chaperone code".
Collapse
|
26
|
Yoshihisa Y, Hassan MA, Furusawa Y, Tabuchi Y, Kondo T, Shimizu T. Alkannin, HSP70 inducer, protects against UVB-induced apoptosis in human keratinocytes. PLoS One 2012; 7:e47903. [PMID: 23110127 PMCID: PMC3478268 DOI: 10.1371/journal.pone.0047903] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 09/17/2012] [Indexed: 12/05/2022] Open
Abstract
Alkannin is an active constituent from the root extract of Alkanna tinctoria of the Boraginaceae family and it may have utility as a heat shock protein 70 (HSP70) inducer in living organisms. Here, the effects of alkannin-induced HSP70 on ultraviolet (UV) B (40 mJ/cm2)-induced apoptosis were investigated in human keratinocyte HaCaT cells. Pretreatment of cells with alkannin (1 µM) caused significant inhibition of UVB-induced apoptosis and caspase-3 cleavage. On the other hand, the addition of KNK437 (HSP70 inhibitor) reversed the action of alkannin increasing UVB-induced apoptosis in a dose-dependent manner. In addition, differences in gene expression associated with the suppression of UVB-induced apoptosis in the presence of alkannin were investigated using Gene Chip assay. Our results indicate that alkannin suppresses UVB-induced apoptosis through the induction of HSP70 in human keratinocytes, and therefore, we suggest the usefulness of using alkannin as an antiaging agent.
Collapse
Affiliation(s)
- Yoko Yoshihisa
- Department of Dermatology, Life Science Research Center, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Mariame Ali Hassan
- Department of Radiological Sciences, Life Science Research Center, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Yukihiro Furusawa
- Department of Radiological Sciences, Life Science Research Center, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Life Science Research Center, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Tadamichi Shimizu
- Department of Dermatology, Life Science Research Center, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- * E-mail:
| |
Collapse
|
27
|
Alekseenko LL, Zemelko VI, Zenin VV, Pugovkina NA, Kozhukharova IV, Kovaleva ZV, Grinchuk TM, Fridlyanskaya II, Nikolsky NN. Heat shock induces apoptosis in human embryonic stem cells but a premature senescence phenotype in their differentiated progeny. Cell Cycle 2012; 11:3260-9. [PMID: 22895173 DOI: 10.4161/cc.21595] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Embryonic stem cells (ESC) are able to self-renew and to differentiate into any cell type. To escape error transmission to future cell progeny, ESC require robust mechanisms to ensure genomic stability. It was stated that stress defense of mouse and human ESC against oxidative stress and irradiation is superior compared with differentiated cells. Here, we investigated heat shock response of human ESC (hESC) and their differentiated progeny. Fibroblast-like cells were generated by spontaneous hESC differentiation via embryoid bodies. Like normal human diploid fibroblasts, these cells have a finite lifespan in culture, undergo replicative senescence and die. We found that sublethal heat shock affected survival of both cell types, but in hESC it induced apoptosis, whereas in differentiated cells it produced cell cycle arrest and premature senescence phenotype. Heat shock survived hESC and differentiated cells restored the properties of initial cells. Heated hESC progeny exhibited pluripotent markers and the capacity to differentiate into the cells of three germ layers. Fibroblast-like cells resisted heat shock, proliferated for a limited number of passages and entered replicative senescence as unheated parental cells. Taken together, these results show for the first time that both hESC and their differentiated derivatives are sensitive to heat shock, but the mechanisms of their stress response are different: hESC undergo apoptosis, whereas differentiated cells under the same conditions exhibit stress-induced premature senescence (SIPS) phenotype. Both cell types that survived sublethal heat shock sustain parental cell properties.
Collapse
|
28
|
Misa Agustiño MJ, Leiro JM, Jorge Mora MT, Rodríguez-González JA, Jorge Barreiro FJ, Ares-Pena FJ, López-Martín E. Electromagnetic fields at 2.45 GHz trigger changes in heat shock proteins 90 and 70 without altering apoptotic activity in rat thyroid gland. Biol Open 2012; 1:831-8. [PMID: 23213477 PMCID: PMC3507243 DOI: 10.1242/bio.20121297] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/07/2012] [Indexed: 12/17/2022] Open
Abstract
Non-ionizing radiation at 2.45 GHz may modify the expression of genes that codify heat shock proteins (HSP) in the thyroid gland. Using the enzyme-linked immunosorbent assay (ELISA) technique, we studied levels of HSP-90 and HSP-70. We also used hematoxilin eosin to look for evidence of lesions in the gland and applied the DAPI technique of fluorescence to search for evidence of chromatin condensation and nuclear fragmentation in the thyroid cells of adult female Sprague-Dawley rats. Fifty-four rats were individually exposed for 30 min to 2.45 GHz radiation in a Gigahertz transverse electromagnetic (GTEM) cell at different levels of non-thermal specific absorption rate (SAR), which was calculated using the finite difference time domain (FDTD) technique. Ninety minutes after radiation, HSP-90 and HSP-70 had decreased significantly (P<0.01) after applying a SAR of 0.046±1.10 W/Kg or 0.104±5.10−3 W/Kg. Twenty-four hours after radiation, HSP-90 had partially recovered and HSP-70 had recovered completely. There were few indications of lesions in the glandular structure and signs of apoptosis were negative in all radiated animals. The results suggest that acute sub-thermal radiation at 2.45 GHz may alter levels of cellular stress in rat thyroid gland without initially altering their anti-apoptotic capacity.
Collapse
|
29
|
Abstract
Estrogen is a potent steroid with pleiotropic effects, which have yet to be fully elucidated. Estrogen has both nuclear and non-nuclear effects. The rapid response to estrogen, which involves a membrane associated estrogen receptor(ER) and is protective, involves signaling through PI3K, Akt, and ERK 1/2. The nuclear response is much slower, as the ER-estrogen complex moves to the nucleus, where it functions as a transcription factor, both activating and repressing gene expression. Several different ERs regulate the specificity of response to estrogen, and appear to have specific effects in cardiac remodeling and the response to injury. However, much remains to be understood about the selectivity of these receptors and their specific effects on gene expression. Basic studies have demonstrated that estrogen treatment prevents apoptosis and necrosis of cardiac and endothelial cells. Estrogen also attenuates pathologic cardiac hypertrophy. Estrogen may have great benefit in aging as an anti-inflammatory agent. However, clinical investigations of estrogen have had mixed results, and not shown the clear-cut benefit of more basic investigations. This can be explained in part by differences in study design: in basic studies estrogen treatment was used immediately or shortly after ovariectomy, while in some key clinical trials, estrogen was given years after menopause. Further basic research into the underlying molecular mechanisms of estrogen's actions is essential to provide a better comprehension of the many properties of this powerful hormone.
Collapse
Affiliation(s)
- A A Knowlton
- Molecular and Cellular Cardiology, Department of Medicine, University of California, Davis, CA 95616, USA.
| | | |
Collapse
|
30
|
Kästle M, Grune T. Interactions of the Proteasomal System with Chaperones. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 109:113-60. [DOI: 10.1016/b978-0-12-397863-9.00004-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Heat shock transcription factor 1 as a therapeutic target in neurodegenerative diseases. Nat Rev Drug Discov 2011; 10:930-44. [PMID: 22129991 DOI: 10.1038/nrd3453] [Citation(s) in RCA: 214] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and prion-based neurodegeneration are associated with the accumulation of misfolded proteins, resulting in neuronal dysfunction and cell death. However, current treatments for these diseases predominantly address disease symptoms, rather than the underlying protein misfolding and cell death, and are not able to halt or reverse the degenerative process. Studies in cell culture, fruitfly, worm and mouse models of protein misfolding-based neurodegenerative diseases indicate that enhancing the protein-folding capacity of cells, via elevated expression of chaperone proteins, has therapeutic potential. Here, we review advances in strategies to harness the power of the natural cellular protein-folding machinery through pharmacological activation of heat shock transcription factor 1--the master activator of chaperone protein gene expression--to treat neurodegenerative diseases.
Collapse
|
32
|
Anckar J, Sistonen L. Regulation of HSF1 function in the heat stress response: implications in aging and disease. Annu Rev Biochem 2011; 80:1089-115. [PMID: 21417720 DOI: 10.1146/annurev-biochem-060809-095203] [Citation(s) in RCA: 550] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
To dampen proteotoxic stresses and maintain protein homeostasis, organisms possess a stress-responsive molecular machinery that detects and neutralizes protein damage. A prominent feature of stressed cells is the increased synthesis of heat shock proteins (Hsps) that aid in the refolding of misfolded peptides and restrain protein aggregation. Transcriptional activation of the heat shock response is orchestrated by heat shock factor 1 (HSF1), which rapidly translocates to hsp genes and induces their expression. Although the role of HSF1 in protecting cells and organisms against severe stress insults is well established, many aspects of how HSF1 senses qualitatively and quantitatively different forms of stresses have remained poorly understood. Moreover, recent discoveries that HSF1 controls life span have prompted new ways of thinking about an old transcription factor. Here, we review the established role of HSF1 in counteracting cell stress and prospect the role of HSF1 as a regulator of disease states and aging.
Collapse
Affiliation(s)
- Julius Anckar
- Department of Biosciences, Åbo Akademi University, BioCity, 20520 Turku, Finland.
| | | |
Collapse
|
33
|
Kalia SK, Kalia LV, McLean PJ. Molecular chaperones as rational drug targets for Parkinson's disease therapeutics. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2011; 9:741-53. [PMID: 20942788 DOI: 10.2174/187152710793237386] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 03/30/2010] [Indexed: 12/19/2022]
Abstract
Parkinson's disease is a neurodegenerative movement disorder that is caused, in part, by the loss of dopaminergic neurons within the substantia nigra pars compacta of the basal ganglia. The presence of intracellular protein aggregates, known as Lewy bodies and Lewy neurites, within the surviving nigral neurons is the defining neuropathological feature of the disease. Accordingly, the identification of specific genes mutated in families with Parkinson's disease and of genetic susceptibility variants for idiopathic Parkinson's disease has implicated abnormalities in proteostasis, or the handling and elimination of misfolded proteins, in the pathogenesis of this neurodegenerative disorder. Protein folding and the refolding of misfolded proteins are regulated by a network of interactive molecules, known as the chaperone system, which is composed of molecular chaperones and co-chaperones. The chaperone system is intimately associated with the ubiquitin-proteasome system and the autophagy-lysosomal pathway which are responsible for elimination of misfolded proteins and protein quality control. In addition to their role in proteostasis, some chaperone molecules are involved in the regulation of cell death pathways. Here we review the role of the molecular chaperones Hsp70 and Hsp90, and the cochaperones Hsp40, BAG family members such as BAG5, CHIP and Hip in modulating neuronal death with a focus on dopaminergic neurodegeneration in Parkinson's disease. We also review current progress in preclinical studies aimed at targetting the chaperone system to prevent neurodegeneration. Finally, we discuss potential future chaperone-based therapeutics for the symptomatic treatment and possible disease modification of Parkinson's disease.
Collapse
Affiliation(s)
- S K Kalia
- Department of Neurology, Massachusetts General Hospital, Mass General Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA 02129, USA
| | | | | |
Collapse
|
34
|
Dehbi M, Baturcam E, Eldali A, Ahmed M, Kwaasi A, Chishti MA, Bouchama A. Hsp-72, a candidate prognostic indicator of heatstroke. Cell Stress Chaperones 2010; 15:593-603. [PMID: 20174993 PMCID: PMC3006628 DOI: 10.1007/s12192-010-0172-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Revised: 01/17/2010] [Accepted: 01/21/2010] [Indexed: 12/28/2022] Open
Abstract
Exposure of rats to environmental heat enhances the expression of heat shock protein-72 (Hsp-72) in most of their organs proportionally to heat stress severity. Pre-induction or over-expression of Hsp-72 prevents organ damage and lethality, suggesting that heat shock proteins (Hsps) may have a pathogenic role in this condition. We investigated the expression profile of Hsps in baboons subjected to environmental heat stress until the core temperature attained 42.5 degrees C (moderate heatstroke) or occurrence of hypotension associated with core temperature > or = 43.5 degrees C (severe heatstroke). Western blot analysis demonstrated a differential induction of Hsp-72 among organs of heat-stressed animals with the highest induction in the liver and the lowest in lung. Hsp-60 and Hsc-70 expression was similar between control and heat-stressed animals. ELISA studies indicated a marked release of Hsp-72 into the circulation of baboons with severe heatstroke with a peak at 24 h post-heatstroke onset and remained sustained up to 72 h. Hsp-72 release was not associated with core temperature or systolic blood pressure, but correlated with markers of liver, myocardium, and skeletal muscle tissue necrosis. Non-survivors displayed significantly higher Hsp-72 levels than survivors. No Hsp-60 was detected in the circulation. These findings add further evidence that increased expression of Hsp-72 may be an important component of the host response to severe heatstroke. They also suggest that extracellular Hsp-72 is a marker of multiple organs tissue damage. Whether extracellular Hsp-72 plays a role in the host immune response to heat stress merits further studies.
Collapse
Affiliation(s)
- Mohammed Dehbi
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia.
| | | | | | | | | | | | | |
Collapse
|
35
|
Dmitrieva NI, Chen HT, Nussenzweig A, Burg MB. Knockout of Ku86 accelerates cellular senescence induced by high NaCl. Aging (Albany NY) 2010; 1:245-53. [PMID: 19946467 PMCID: PMC2783634 DOI: 10.18632/aging.100022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
NaCl induces DNA breaks, thus leading to cellular senescence. Here we showed that Ku86 deficiency accelerated the high NaCl-induced cellular senescence. We find that 1) high NaCl induces rapid cellular senescence in Ku86 deficient(xrs5) cells, 2) Ku86 deficiency shortens lifespan of C. elegans in high NaCl, and 3) cellular senescence is greatly accelerated in renal inner medullas of Ku86 (-/-) mice. Further, although water balance is known to be compromised in old mice, this occurs at much earlier age in Ku86(-/-) mice. When subjected to mild water restriction, 3 month old Ku86(-/-), but not Ku86(+/+),mice rapidly become dehydrated as evidenced by decrease in body weight, increased production of antidiuretic hormone,increased urine osmolality and decreased urine volume. The deficiency in water balance does not occur in Ku86(+/+)mice until they are much older (14 months). We conclude that Ku86 deficiency accelerates high NaCl(-) induced cellular senescence,particularly in the renal medulla where NaCl normally is high.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- Laboratory of Kidney and Electrolyte Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
36
|
Hoshino T, Matsuda M, Yamashita Y, Takehara M, Fukuya M, Mineda K, Maji D, Ihn H, Adachi H, Sobue G, Funasaka Y, Mizushima T. Suppression of melanin production by expression of HSP70. J Biol Chem 2010; 285:13254-63. [PMID: 20177067 DOI: 10.1074/jbc.m110.103051] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Skin hyperpigmentation disorders due to abnormal melanin production induced by ultraviolet (UV) irradiation are both a clinical and cosmetic problem. UV irradiation stimulates melanin production in melanocytes by increasing intracellular cAMP. Expression of heat shock proteins (HSPs), especially HSP70, is induced by various stressors, including UV irradiation, to provide cellular resistance to such stressors. In this study we examined the effect of expression of HSP70 on melanin production both in vitro and in vivo. 3-Isobutyl-1-methylxanthine (IBMX), a cAMP-elevating agent, stimulated melanin production in cultured mouse melanoma cells, and this stimulation was suppressed in cells overexpressing HSP70. IBMX-dependent transcriptional activation of the tyrosinase gene was also suppressed in HSP70-overexpressing cells. Expression of microphthalmia-associated transcription factor (MITF), which positively regulates transcription of the tyrosinase gene, was up-regulated by IBMX; however, this up-regulation was not suppressed in HSP70-overexpressing cells. On the other hand, immunoprecipitation and immunostaining analyses revealed a physical interaction between and co-localization of MITF and HSP70, respectively. Furthermore, the transcription of tyrosinase gene in nuclear extract was inhibited by HSP70. In vivo, UV irradiation of wild-type mice increased the amount of melanin in the basal layer of the epidermis, and this increase was suppressed in transgenic mice expressing HSP70. This study provides the first evidence of an inhibitory effect of HSP70 on melanin production both in vitro and in vivo. This effect seems to be mediated by modulation of MITF activity through a direct interaction between HSP70 and MITF.
Collapse
Affiliation(s)
- Tatsuya Hoshino
- Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gruber HE, Hoelscher GL, Ingram JA, Zinchenko N, Hanley EN. Senescent vs. non-senescent cells in the human annulus in vivo: cell harvest with laser capture microdissection and gene expression studies with microarray analysis. BMC Biotechnol 2010; 10:5. [PMID: 20109216 PMCID: PMC2828399 DOI: 10.1186/1472-6750-10-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 01/28/2010] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Senescent cells are well-recognized in the aging/degenerating human disc. Senescent cells are viable, cannot divide, remain metabolically active and accumulate within the disc over time. Molecular analysis of senescent cells in tissue offers a special challenge since there are no cell surface markers for senescence which would let one use fluorescence-activated cell sorting as a method for separating out senescent cells. METHODS We employed a novel laser capture microdissection (LCM) design to selectively harvest senescent and non-senescent annulus cells in paraffin-embedded tissue, and compared their gene expression with microarray analysis. LCM was used to separately harvest senescent and non-senescent cells from 11 human annulus specimens. RESULTS Microarray analysis revealed significant differences in expression levels in senescent cells vs non-senescent cells: 292 genes were upregulated, and 321 downregulated. Genes with established relationships to senescence were found to be significantly upregulated in senescent cells vs. non-senescent cells: p38 (MPAK14), RB-Associated KRAB zinc finger, Discoidin, CUB and LCCL domain, growth arrest and DNA-damage inducible beta, p28ING5, sphingosine-1-phosphate receptor 2 and somatostatin receptor 3; cyclin-dependent kinase 8 showed significant downregulation in senescent cells. Nitric oxidase synthase 1, and heat shock 70 kDa protein 6, both of which were significantly down-regulated in senescent cells, also showed significant changes. Additional genes related to cytokines, cell proliferation, and other processes were also identified. CONCLUSIONS Our LCM-microarray analyses identified a set of genes associated with senescence which were significantly upregulated in senescent vs non-senescent cells in the human annulus. These genes include p38 MAP kinase, discoidin, inhibitor of growth family member 5, and growth arrest and DNA-damage-inducible beta. Other genes, including genes associated with cell proliferation, extracellular matrix formation, cell signaling and other cell functions also showed significant modulation in senescent vs non-senescent cells. The aging/degenerating disc undergoes a well-recognized loss of cells; understanding senescent cells is important since their presence further reduces the disc's ability to generate new cells to replace those lost to necrosis or apoptosis.
Collapse
Affiliation(s)
- Helen E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, NC, USA.
| | | | | | | | | |
Collapse
|
38
|
|
39
|
Lindner AB, Demarez A. Protein aggregation as a paradigm of aging. Biochim Biophys Acta Gen Subj 2009; 1790:980-96. [PMID: 19527771 DOI: 10.1016/j.bbagen.2009.06.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 06/08/2009] [Accepted: 06/09/2009] [Indexed: 12/23/2022]
Abstract
The process of physiological decline leading to death of the individual is driven by the deteriorating capacity to withstand extrinsic and intrinsic hazards, resulting in damage accumulation with age. The dynamic changes with time of the network governing the outcome of misfolded proteins, exemplifying as intrinsic hazards, is considered here as a paradigm of aging. The main features of the network, namely, the non-linear increase of damage and the presence of amplifying feedback loops within the system are presented through a survey of the different components of the network and related cellular processes in aging and disease.
Collapse
Affiliation(s)
- Ariel B Lindner
- INSERM U571, Paris Descartes University, Paris, F-75015, France.
| | | |
Collapse
|
40
|
Oh S, Lee E, Lee J, Lim Y, Kim J, Woo S. Comparison of the effects of 40% oxygen and two atmospheric absolute air pressure conditions on stress-induced premature senescence of normal human diploid fibroblasts. Cell Stress Chaperones 2008; 13:447-58. [PMID: 18465208 PMCID: PMC2673923 DOI: 10.1007/s12192-008-0041-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2008] [Revised: 03/24/2008] [Accepted: 03/24/2008] [Indexed: 01/28/2023] Open
Abstract
The pressure during hyperbaric oxygen treatment may increase oxygen toxicity via an augmented oxygen pressure in the gas. Nevertheless, only a few reports have been published on the effect of cells grown under 2 atmospheric absolute (ATA) pressure. To evaluate the effect of pressure on oxygen toxicity and to study effects in addition to oxygen toxicity, we designed an experiment to compare the effects of normobaric mild hyperoxia (NMH, 40% oxygen) and hyperbaric air condition (HA, air with 2 ATA) on human diploid fibroblasts (HDF) in a hyperbaric incubator. HDFs in both the NMH and the HA condition had a similar oxidative stress response and exhibited premature senescence. To investigate differences in gene profiling in cells grown in the NMH and HA conditions, samples from cells exposed to each condition were applied to microarrays. We found no expression difference in genes related to aging and deoxyribonucleic acid damage, but the expression of genes including cell adhesion, stress response, and transcription were significantly increased in fibroblasts that were responsive to pressure. Among 26 statistically reliable genes, the expression of apoptosis related genes such as ADAM22, Bax, BCL2L14, and UBD, as well as tumor suppressor-related genes like Axin2 and ATF, and also mitogen-activated protein kinase-related genes like mitogen-activated protein kinase kinase kinase 1, histamine receptor, and RAB24, were significantly changed in cells responsive to pressure-induced oxidative stress.
Collapse
Affiliation(s)
- Sangnam Oh
- Department of Preventive Medicine and Medical Research Center for Environmental Toxico-Genomics and Proteomics, College of Medicine, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
- Cellular and Developmental Biology, Division of Brain Korea 21 Program for Biomedical Science, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
| | - Eunil Lee
- Department of Preventive Medicine and Medical Research Center for Environmental Toxico-Genomics and Proteomics, College of Medicine, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
- Cellular and Developmental Biology, Division of Brain Korea 21 Program for Biomedical Science, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
- Postgraduate Studies of Public Health, Graduate School, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
| | - Joohyun Lee
- Department of Preventive Medicine and Medical Research Center for Environmental Toxico-Genomics and Proteomics, College of Medicine, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
- Postgraduate Studies of Public Health, Graduate School, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
| | - Yongchul Lim
- Department of Preventive Medicine and Medical Research Center for Environmental Toxico-Genomics and Proteomics, College of Medicine, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
- Cellular and Developmental Biology, Division of Brain Korea 21 Program for Biomedical Science, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
| | - Joonhee Kim
- Department of Preventive Medicine and Medical Research Center for Environmental Toxico-Genomics and Proteomics, College of Medicine, Korea University, Anam-dong 5ga 126-1, Seongbuk-gu, Seoul, 136-705 South Korea
| | - Samyong Woo
- Korea Research Institute of Standards and Science, Yuseng, Deajeon, 305-340 Korea
| |
Collapse
|
41
|
Calabrese V, Calafato S, Puleo E, Cornelius C, Sapienza M, Morganti P, Mancuso C. Redox regulation of cellular stress response by ferulic acid ethyl ester in human dermal fibroblasts: role of vitagenes. Clin Dermatol 2008; 26:358-63. [PMID: 18691515 DOI: 10.1016/j.clindermatol.2008.01.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Skin is one of the main targets for reactive oxygen species; thus, reactive oxygen species-induced damage and protein and lipid modifications occur, and skin can undergo a wide array of diseases, from photosensitivity to cancer. In this study, human dermal fibroblasts exposed to hydrogen peroxide (0-1000 micromol/L) exhibited a marked increase in both protein carbonyls and 4-hydroxy-2-nonenal, which are indices of protein and lipid oxidation, respectively. An amount of 25 micromol/L ferulic acid ethyl ester, a well-known nutritional antioxidant, significantly counteracted both protein and lipid oxidation and reduced the loss in cell viability elicited by 500 micromol/L of hydrogen peroxide. A common way for cells to react to oxidative stress is up-regulation of vitagenes. To the vitagene family belong the heat shock proteins heme oxygenase-1 and heat shock protein-70, which are involved in the cellular defense against oxidative stress by different mechanisms. The administration of 25 micromol/L ferulic acid ethyl ester significantly decreased hydrogen peroxide-induced protein and lipid oxidation. Dermal fibroblasts exposed to 25 micromol/L ferulic acid ethyl ester in the presence of 500 micromol/L hydrogen peroxide showed an increased level of both heme oxygenase-1 and heat shock protein-70 compared with dermal fibroblasts treated with hydrogen peroxide alone. These findings provide evidence for the protective role of vitagenes in free radical-induced skin damage and highlight the potential protective use of nutritional antioxidants, such as ferulic acid and its derivatives.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Chemistry, Biochemistry and Molecular Biology Section, Faculty of Medicine, University of Catania, 95100 Catania, Italy.
| | | | | | | | | | | | | |
Collapse
|
42
|
Lee YH, Lee JC, Moon HJ, Jung JE, Sharma M, Park BH, Yi HK, Jhee EC. Differential effect of oxidative stress on the apoptosis of early and late passage human diploid fibroblasts: implication of heat shock protein 60. Cell Biochem Funct 2008; 26:502-8. [DOI: 10.1002/cbf.1473] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
43
|
Stoka V, Turk V, Bredesen DE. Differential regulation of Smac/DIABLO and Hsp-70 during brain maturation. Neuromolecular Med 2007; 9:255-63. [PMID: 17914183 PMCID: PMC2755584 DOI: 10.1007/s12017-007-8007-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 11/30/1999] [Accepted: 03/14/2007] [Indexed: 12/23/2022]
Abstract
The heat shock protein (Hsp) system is a cell defense mechanism constitutively expressed at the basal state and essential for cell survival in response to damaging stimuli. Apoptosis is a physiological cell death program that preserves tissue homeostasis. We investigated the intrinsic pathway of apoptosis at various stages of brain maturation in CD-1 mice, triggered by two mitochondrial proapoptotic proteins, cytochrome c and Smac/DIABLO, and the pathway's regulation by Hsp-70. Smac/DIABLO and Hsp-70 proteins were upregulated 2-fold and 1.5-3-fold, respectively, after birth. In contrast, in the presence of cytochrome c/2'-deoxyadenosine 5'-triphosphate (dATP), caspase activity in mouse brain cell-free extracts increased 90-fold and 61-fold, at fetal and neonatal stages, whereas no activation was detected 15 days postnatally or at any subsequent times. These results indicate that the activation pattern of the intrinsic pathway of apoptosis undergoes a marked shift during postnatal maturation.
Collapse
Affiliation(s)
- Veronika Stoka
- Buck Institute for Age Research, 8001 Redwood Blvd., Novato, CA 94945, USA
- Department of Biochemistry, Molecular and Structural Biology, J. Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Vito Turk
- Department of Biochemistry, Molecular and Structural Biology, J. Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Dale E. Bredesen
- Buck Institute for Age Research, 8001 Redwood Blvd., Novato, CA 94945, USA
- Department of Neurology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
44
|
Bagchi M, Besser D, Reddy TR, Skoff R, Maisel H. Effect of thermal stress on early and late passaged mouse lens epithelial cells. J Cell Biochem 2007; 102:1036-42. [PMID: 17427954 DOI: 10.1002/jcb.21339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cataract is an age related disease of protein aggregation. It has been suggested that aging affects the cells ability to protect protein integrity. The protein integrity, which is essential for cellular homeostasis, is maintained by a complex system of refolding or degradation of damaged proteins. The heat shock proteins (hsps) are the major contributors in the maintenance of protein integrity. The heat shock transcription factor (HSF-1) is the master regulator of all hsp synthesis in response to stress. This investigation examined the role of HSF-1 in the regulation of hsp synthesis in early and late passaged alphaTN-4 cells. Data collected in this study revealed that the nucleotide sequence of HSF-1 mRNA obtained from early and late passaged alphaTN-4 cells were identical. When early and late passaged cell were exposed to thermal stress, their hsp expression were also similar. HSP-40 expression was detected after 2 h of heat stress, whereas HSP-70 and low molecular weight heat shock protein alphabeta crystallin showed significantly increased synthesis 18 h post heat stress. The late passaged alphaTN-4 cells ability to upregulate hsps in response to heat stress could be due to its high replicative activities. The data presented here suggests a relationship between the presence of functional HSF-1 and sustained proliferative activities of the late passaged alphaTN-4 cell.
Collapse
Affiliation(s)
- M Bagchi
- Department of Anatomy and Cell Biology, School of Medicine, Wayne State University, Detroit, Michigan 48201, USA.
| | | | | | | | | |
Collapse
|
45
|
Chinnathambi S, Tomanek-Chalkley A, Bickenbach JR. HSP70 and EndoG modulate cell death by heat in human skin keratinocytes in vitro. Cells Tissues Organs 2007; 187:131-40. [PMID: 17938562 DOI: 10.1159/000109941] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2007] [Indexed: 01/20/2023] Open
Abstract
We examined how young and old keratinocytes died from heat stress in vitro. We found that keratinocyte cell death was not due to oxidative stress as neither Mn-SOD nor Cu-Zn-SOD was produced in either young or old heated keratinocytes. Instead, analysis of the anti-apoptotic factors, Bcl2 and HSP70, and the pro-apoptotic factors, caspase 3, caspase 8, Apaf-1, cytochrome c, AIF, and EndoG, indicated that keratinocyte cell death occurred via the caspase-independent EndoG apoptotic pathway. We found that both young and old keratinocytes died via the same pathway, and that we could specifically reduce both young and old keratinocyte death by addition of the EndoG inhibitor NEM. Further analysis suggested that the difference between young and old keratinocyte death was due to the synthesis of HSP70 protein, with the increase in response to heat more pronounced in young keratinocytes than in old keratinocytes. When we inhibited HSP70 by adding quercetin, death was increased in both young and old keratinocytes, but more so in old keratinocytes. These data suggest that old keratinocytes may die more readily than young keratinocytes when heated because they synthesize HSP70 at a lower efficiency. Such findings suggest that HSP70 production may be age-dependent.
Collapse
Affiliation(s)
- Sathivel Chinnathambi
- Department of Anatomy and Cell Biology, The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|
46
|
Dai C, Whitesell L, Rogers AB, Lindquist S. Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis. Cell 2007; 130:1005-18. [PMID: 17889646 PMCID: PMC2586609 DOI: 10.1016/j.cell.2007.07.020] [Citation(s) in RCA: 654] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Revised: 06/11/2007] [Accepted: 07/13/2007] [Indexed: 11/15/2022]
Abstract
Heat shock factor 1 (HSF1) is the master regulator of the heat shock response in eukaryotes, a very highly conserved protective mechanism. HSF1 function increases survival under a great many pathophysiological conditions. How it might be involved in malignancy remains largely unexplored. We report that eliminating HSF1 protects mice from tumors induced by mutations of the RAS oncogene or a hot spot mutation in the tumor suppressor p53. In cell culture, HSF1 supports malignant transformation by orchestrating a network of core cellular functions including proliferation, survival, protein synthesis, and glucose metabolism. The striking effects of HSF1 on oncogenic transformation are not limited to mouse systems or tumor initiation; human cancer lines of diverse origins show much greater dependence on HSF1 function to maintain proliferation and survival than their nontransformed counterparts. While it enhances organismal survival and longevity under most circumstances, HSF1 has the opposite effect in supporting the lethal phenomenon of cancer.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene
- Animals
- Carcinogens
- Cell Line, Tumor
- Cell Proliferation
- Cell Survival
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Gene Expression Regulation, Neoplastic
- Genotype
- Glucose/metabolism
- Heat Shock Transcription Factors
- Humans
- Methylnitronitrosoguanidine
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mutation
- Phenotype
- Protein Biosynthesis
- Proto-Oncogene Proteins c-sis/genetics
- Proto-Oncogene Proteins c-sis/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Signal Transduction/genetics
- Skin/metabolism
- Skin/pathology
- Skin Neoplasms/chemically induced
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Tetradecanoylphorbol Acetate
- Time Factors
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transduction, Genetic
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- ras Proteins/genetics
- ras Proteins/metabolism
Collapse
Affiliation(s)
- Chengkai Dai
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Luke Whitesell
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Arlin B. Rogers
- Divison of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
47
|
Wieten L, Broere F, van der Zee R, Koerkamp EK, Wagenaar J, van Eden W. Cell stress induced HSP are targets of regulatory T cells: A role for HSP inducing compounds as anti-inflammatory immuno-modulators? FEBS Lett 2007; 581:3716-22. [PMID: 17507013 DOI: 10.1016/j.febslet.2007.04.082] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Revised: 04/25/2007] [Accepted: 04/27/2007] [Indexed: 01/06/2023]
Abstract
T cell responses to heat shock proteins (HSP) have disease suppressive activities through production of anti-inflammatory cytokines in patients and in models of inflammatory diseases. There is evidence that the anti-inflammatory activity of HSP-specific T cells depends on their recognition of endogenous HSP epitopes as expressed by stressed cells at sites of inflammation. Previously, we have demonstrated that such T cells can be induced by conserved sequences of microbial HSP. Now we propose that drug induced up-regulation of endogenous HSP can contribute to anti-inflammatory T cell regulation.
Collapse
Affiliation(s)
- Lotte Wieten
- Division of Immunology, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, University of Utrecht, Yalelaan 1, 3584CL Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
48
|
Kletsas D, Pratsinis H, Gioni V, Pilichos K, Yiacoumettis AM, Tsagarakis S. Prior Chronic in Vivo Glucocorticoid Excess Leads to an Anabolic Phenotype and an Extension of Cellular Life Span of Skin Fibroblasts in Vitro. Ann N Y Acad Sci 2007; 1100:449-54. [PMID: 17460210 DOI: 10.1196/annals.1395.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Intense stress can be detrimental for tissue homeostasis and accelerates aging. On the other hand, repeated mild stresses can have beneficial and even life-prolonging effects. Hypersecretion of glucocorticoids (GCs) represents the major hormonal response to stress. However, besides its life-sustaining role, GC excess can promote a "catabolic" phenotype. Accordingly, we have studied the effect of long-lasting exposure to high GC levels in vivo on several parameters of tissue homeostasis, as well as cellular senescence, in cells removed from the high-GC milieu in vivo and then cultured in vitro. To this end, we have used human skin fibroblasts from (a) Cushing's syndrome patients that are characterized by chronic endogenous GC excess and (b) patients treated with exogenous GC administration. Interestingly, when Cushing's syndrome fibroblasts were cultured in vitro under standard conditions they express an "anabolic" phenotype, i.e., they restore their ability for collagen synthesis, secrete reduced levels of metalloproteases, and have an increased proliferative capacity and contractility. Furthermore, these cells exhibit a significant extension of their proliferative life span, while they respond better to exogenous stress by producing significantly higher levels of heat-shock protein-70 (HSP70). In addition, preliminary results with fibroblasts from patients subjected to chronic exogenous GC administration indicate that they express a similar behavior in vitro, at least with regard to the restoration of collagen expression. These data suggest that prior exposure to elevated GC concentrations is not associated with persisting adverse effects on fibroblasts and may also have a beneficial outcome in some aspects of cell physiology, including longevity in vitro.
Collapse
Affiliation(s)
- Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biology, National Centre for Scientific Research Demokritos, 15310 Athens, Greece.
| | | | | | | | | | | |
Collapse
|
49
|
Valen G, Hinokiyama K, Vedin J, Vaage J. Preoperative unstable angina causes venous adaptation to surgical graft injury. Basic Res Cardiol 2007; 102:265-73. [PMID: 17268886 DOI: 10.1007/s00395-007-0642-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 11/29/2006] [Accepted: 12/19/2006] [Indexed: 10/23/2022]
Abstract
Ischemic preconditioning may provide a systemic organ protection, evident as the phenomenon known as remote preconditioning. Unstable angina may be a clinical analogue to ischemic preconditioning. Vein graft harvesting induces inflammation of the graft wall. We hypothesized that preoperative unstable angina preconditions vein grafts and reduces the inflammatory response to graft harvesting. Consecutive patients with stable or unstable angina undergoing open heart surgery (n = 12 in each group) were studied. Saphenous vein biopsies were collected at the start of graft harvesting, and when the last proximal anastomosis to the aorta was finished (average 112 minutes later). Gene expression of inflammatory mediators (tumor necrosis factor alpha, interleukin-1beta (IL-1beta), E-selectin (CD62E), intercellular leukocyte adhesion molecule 1, inducible nitric oxide synthase, endothelin-1) increased after surgical handling (semiquantitative RT-PCR). In vein grafts from unstable patients the increase was attenuated for Il-1beta (p < 0.004) and CD62E (p < 0.001). In stable patients the protein expression of IkappaBalpha and heat shock protein72 was reduced by surgical handling (p < 0.04), but was not influenced in unstable patients (immunoblotting). In vitro relaxation to acetylcholine was enhanced, and contractions to phenylephrine and endothelin-1 were attenuated in veins rings from unstable patients (p < 0.003). In conclusion, surgical handling of vein grafts induces inflammation of the vessel wall. This response was reduced in grafts from patients with unstable angina, indicating a possible systemic preconditioning-like effect of acute coronary syndromes.
Collapse
Affiliation(s)
- Guro Valen
- Institute of Basic Medical Science, Dept. of Physiology University of Oslo, 1103 Blindern, 0317, Oslo, Norway.
| | | | | | | |
Collapse
|
50
|
Pratsinis H, Tsagarakis S, Zervolea I, Stathakos D, Thalassinos N, Kletsas D. The unexpected anabolic phenotype and extended longevity of skin fibroblasts after chronic glucocorticoid excess. Dose Response 2006; 4:133-44. [PMID: 18648637 DOI: 10.2203/dose-response.05-007.pratsinis] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Intense stress can challenge tissue homeostasis and accelerate the ageing process. However, several lines of evidence indicate that repeated mild stresses can have beneficial and even life-prolonging effects. Hypersecretion of glucocorticoids (GC) represents the major hormonal response to stress. Besides its life-sustaining role, GC excess, usually due to several side-effects that promote a "catabolic" phenotype, can be detrimental for several tissues. Cushing's syndrome patients are characterized by chronic endogenous GC excess and consequently at the time of diagnosis they have an atrophic elderly-like skin. Interestingly, when Cushing's syndrome fibroblasts were removed from the high-GC milieu in vivo and cultured in vitro under standard conditions they express an "anabolic" phenotype, i.e. they restore their ability for collagen synthesis, they secrete reduced levels of metalloproteases (MMP-1 and MMP-2) and have an increased proliferative capacity and contractility. Furthermore, these cells exhibit a significant extension of their proliferative lifespan, while they respond better to exogenous stress by producing significantly higher levels of heat-shock protein-70 (HSP70). These results imply that long-term hypercortisolism in vivo can have beneficial consequences on fibroblast physiology in vitro.
Collapse
Affiliation(s)
- Harris Pratsinis
- Laboratory of Cell Proliferation and Ageing, Institute of Biology, National Centre for Scientific Research Demokritos, Athens, Greece
| | | | | | | | | | | |
Collapse
|