1
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-w] [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: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
2
|
Kim D, Kiprov DD, Luellen C, Lieb M, Liu C, Watanabe E, Mei X, Cassaleto K, Kramer J, Conboy MJ, Conboy IM. Old plasma dilution reduces human biological age: a clinical study. GeroScience 2022; 44:2701-2720. [PMID: 35999337 PMCID: PMC9398900 DOI: 10.1007/s11357-022-00645-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/10/2022] [Indexed: 01/07/2023] Open
Abstract
This work extrapolates to humans the previous animal studies on blood heterochronicity and establishes a novel direct measurement of biological age. Our results support the hypothesis that, similar to mice, human aging is driven by age-imposed systemic molecular excess, the attenuation of which reverses biological age, defined in our work as a deregulation (noise) of 10 novel protein biomarkers. The results on biological age are strongly supported by the data, which demonstrates that rounds of therapeutic plasma exchange (TPE) promote a global shift to a younger systemic proteome, including youthfully restored pro-regenerative, anticancer, and apoptotic regulators and a youthful profile of myeloid/lymphoid markers in circulating cells, which have reduced cellular senescence and lower DNA damage. Mechanistically, the circulatory regulators of the JAK-STAT, MAPK, TGF-beta, NF-κB, and Toll-like receptor signaling pathways become more youthfully balanced through normalization of TLR4, which we define as a nodal point of this molecular rejuvenation. The significance of our findings is confirmed through big-data gene expression studies.
Collapse
Affiliation(s)
- Daehwan Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | | | - Connor Luellen
- Biophysics, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Michael Lieb
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Chao Liu
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Etsuko Watanabe
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Xiaoyue Mei
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | | | - Joel Kramer
- Brain Aging Center, UCSF, San Francisco, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
3
|
Fessel J. Ineffective levels of transforming growth factors and their receptor account for old age being a risk factor for Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:899-905. [PMID: 31890854 PMCID: PMC6926356 DOI: 10.1016/j.trci.2019.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
After the midninth decade of age, the incidence rates of Alzheimer's disease (AD) and the presence of active TGF-β1 show comparable increases. The hypothesis is proposed that the reason why advanced age is a major risk factor for AD is a progressive decrease with advancing age in the numbers of TGFR2 receptors in the brain, with the consequence of a decline in the neurotrophic efficacy of TGF-β1 and 2 despite their already increased levels in older persons. Alternative, possible reasons are discussed but rejected because either those reasons may also affect young persons or because they cannot be validated in a clinical trial. The proposed hypothesis may be validated in persons with aMCI after raising their brain levels of TGF-β1 and 2 by using a combination of three drugs, lithium, memantine, plus either glatiramer or venlafaxine, and then assessing their progression to AD.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Emeritus, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
4
|
TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. Int J Mol Sci 2019; 20:ijms20205002. [PMID: 31658594 PMCID: PMC6834140 DOI: 10.3390/ijms20205002] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022] Open
Abstract
Aging is broadly defined as the functional decline that occurs in all body systems. The accumulation of senescent cells is considered a hallmark of aging and thought to contribute to the aging pathologies. Transforming growth factor-β (TGF-β) is a pleiotropic cytokine that regulates a myriad of cellular processes and has important roles in embryonic development, physiological tissue homeostasis, and various pathological conditions. TGF-β exerts potent growth inhibitory activities in various cell types, and multiple growth regulatory mechanisms have reportedly been linked to the phenotypes of cellular senescence and stem cell aging in previous studies. In addition, accumulated evidence has indicated a multifaceted association between TGF-β signaling and aging-associated disorders, including Alzheimer’s disease, muscle atrophy, and obesity. The findings regarding these diseases suggest that the impairment of TGF-β signaling in certain cell types and the upregulation of TGF-β ligands contribute to cell degeneration, tissue fibrosis, inflammation, decreased regeneration capacity, and metabolic malfunction. While the biological roles of TGF-β depend highly on cell types and cellular contexts, aging-associated changes are an important additional context which warrants further investigation to better understand the involvement in various diseases and develop therapeutic options. The present review summarizes the relationships between TGF-β signaling and cellular senescence, stem cell aging, and aging-related diseases.
Collapse
|
5
|
Ongali B, Nicolakakis N, Tong XK, Lecrux C, Imboden H, Hamel E. Transforming growth factor-β1 induces cerebrovascular dysfunction and astrogliosis through angiotensin II type 1 receptor-mediated signaling pathways. Can J Physiol Pharmacol 2018; 96:527-534. [PMID: 29505736 DOI: 10.1139/cjpp-2017-0640] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transgenic mice constitutively overexpressing the cytokine transforming growth factor-β1 (TGF-β1) (TGF mice) display cerebrovascular alterations as seen in Alzheimer's disease (AD) and vascular cognitive impairment and dementia (VCID), but no or only subtle cognitive deficits. TGF-β1 may exert part of its deleterious effects through interactions with angiotensin II (AngII) type 1 receptor (AT1R) signaling pathways. We test such interactions in the brain and cerebral vessels of TGF mice by measuring cerebrovascular reactivity, levels of protein markers of vascular fibrosis, nitric oxide synthase activity, astrogliosis, and mnemonic performance in mice treated (6 months) with the AT1R blocker losartan (10 mg/kg per day) or the angiotensin converting enzyme inhibitor enalapril (3 mg/kg per day). Both treatments restored the severely impaired cerebrovascular reactivity to acetylcholine, calcitonin gene-related peptide, endothelin-1, and the baseline availability of nitric oxide in aged TGF mice. Losartan, but not enalapril, significantly reduced astrogliosis and cerebrovascular levels of profibrotic protein connective tissue growth factor while raising levels of antifibrotic enzyme matrix metallopeptidase-9. Memory was unaffected by aging and treatments. The results suggest a pivotal role for AngII in TGF-β1-induced cerebrovascular dysfunction and neuroinflammation through AT1R-mediated mechanisms. Further, they suggest that AngII blockers could be appropriate against vasculopathies and astrogliosis associated with AD and VCID.
Collapse
Affiliation(s)
- Brice Ongali
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Nektaria Nicolakakis
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Xin-Kang Tong
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Clotilde Lecrux
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Hans Imboden
- b Institute of Cell Biology, University of Bern Baltzerstrasse 43012 Bern, Switzerland
| | - Edith Hamel
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| |
Collapse
|
6
|
Chio CC, Lin MT, Chang CP, Lin HJ. A positive correlation exists between neurotrauma and TGF-β1-containing microglia in rats. Eur J Clin Invest 2016; 46:1063-1069. [PMID: 27759956 DOI: 10.1111/eci.12693] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 10/17/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Transforming growth factor-beta 1 (TGF-β1) regulates many processes after traumatic brain injury (TBI). Both Neuro AiD™ (MLC601) and astragaloside (AST) attenuate microglia activation in rats with TBI. The purpose of this study was to investigate whether MLC601 or AST improves output of TBI by affecting microglial expression of TGF-β1. MATERIALS AND METHODS Adult male Sprague-Dawley rats (120 in number) were used to investigate the contribution of TGF-β1-containing microglia in the MLC601-mediated or the AST-mediated neuroprotection in the brain trauma condition using lateral fluid percussion injury. RESULTS Pearson correlation analysis revealed that there was a positive correlation between brain injury (evidenced by both brain contused volume and neurological severity score) and the cortical numbers of TGF-β1-containing microglia for the rats (n = 12) 4 days post-TBI. MLC601 or AST significantly (P < 0·05) attenuated TBI-induced brain contused volume (119 ± 14 mm3 or 108 ± 11 mm3 vs. 160 ± 21 mm3 ), neurological severity score (7·8 ± 0·3 or 8·1 ± 0·4 vs. 10·2 ± 0·5) and numbers of TGF-β1-containing microglia (6% ± 2% or 11% ± 3% vs. 79% ± 7%) for the rats 4 days post-TBI. CONCLUSIONS There was a positive correlation between TBI and cortical numbers of TGF-β1-containing microglia which could be significantly attenuated by astragaloside or NeuroAiD™ (MLC601) in rats.
Collapse
Affiliation(s)
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Hung-Jung Lin
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| |
Collapse
|
7
|
Corbi G, Conti V, Davinelli S, Scapagnini G, Filippelli A, Ferrara N. Dietary Phytochemicals in Neuroimmunoaging: A New Therapeutic Possibility for Humans? Front Pharmacol 2016; 7:364. [PMID: 27790141 PMCID: PMC5062465 DOI: 10.3389/fphar.2016.00364] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/21/2016] [Indexed: 12/12/2022] Open
Abstract
Although several efforts have been made in the search for genetic and epigenetic patterns linked to diseases, a comprehensive explanation of the mechanisms underlying pathological phenotypic plasticity is still far from being clarified. Oxidative stress and inflammation are two of the major triggers of the epigenetic alterations occurring in chronic pathologies, such as neurodegenerative diseases. In fact, over the last decade, remarkable progress has been made to realize that chronic, low-grade inflammation is one of the major risk factor underlying brain aging. Accumulated data strongly suggest that phytochemicals from fruits, vegetables, herbs, and spices may exert relevant immunomodulatory and/or anti-inflammatory activities in the context of brain aging. Starting by the evidence that a common denominator of aging and chronic degenerative diseases is represented by inflammation, and that several dietary phytochemicals are able to potentially interfere with and regulate the normal function of cells, in particular neuronal components, aim of this review is to summarize recent studies on neuroinflammaging processes and proofs indicating that specific phytochemicals may act as positive modulators of neuroinflammatory events. In addition, critical pathways involved in mediating phytochemicals effects on neuroinflammaging were discussed, exploring the real impact of these compounds in preserving brain health before the onset of symptoms leading to inflammatory neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Graziamaria Corbi
- Department of Medicine and Health Sciences, University of Molise Campobasso, Italy
| | - Valeria Conti
- Department of Medicine, Surgery and Dentistry, University of Salerno Salerno, Italy
| | - Sergio Davinelli
- Department of Medicine and Health Sciences, University of Molise Campobasso, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences, University of Molise Campobasso, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry, University of Salerno Salerno, Italy
| | - Nicola Ferrara
- Department of Translational Medical Sciences, Federico II University of NaplesNaples, Italy; Salvatore Maugeri Foundation, IRCCS, Scientific Institute of TeleseTelese Terme, Italy
| |
Collapse
|
8
|
Cornejo F, von Bernhardi R. Age-Dependent Changes in the Activation and Regulation of Microglia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:205-226. [DOI: 10.1007/978-3-319-40764-7_10] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
9
|
von Bernhardi R, Cornejo F, Parada GE, Eugenín J. Role of TGFβ signaling in the pathogenesis of Alzheimer's disease. Front Cell Neurosci 2015; 9:426. [PMID: 26578886 PMCID: PMC4623426 DOI: 10.3389/fncel.2015.00426] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 10/09/2015] [Indexed: 12/19/2022] Open
Abstract
Aging is the main risk factor for Alzheimer’s disease (AD); being associated with conspicuous changes on microglia activation. Aged microglia exhibit an increased expression of cytokines, exacerbated reactivity to various stimuli, oxidative stress, and reduced phagocytosis of β-amyloid (Aβ). Whereas normal inflammation is protective, it becomes dysregulated in the presence of a persistent stimulus, or in the context of an inflammatory environment, as observed in aging. Thus, neuroinflammation can be a self-perpetuating deleterious response, becoming a source of additional injury to host cells in neurodegenerative diseases. In aged individuals, although transforming growth factor β (TGFβ) is upregulated, its canonical Smad3 signaling is greatly reduced and neuroinflammation persists. This age-related Smad3 impairment reduces protective activation while facilitating cytotoxic activation of microglia through several cellular mechanisms, potentiating microglia-mediated neurodegeneration. Here, we critically discuss the role of TGFβ-Smad signaling on the cytotoxic activation of microglia and its relevance in the pathogenesis of AD. Other protective functions, such as phagocytosis, although observed in aged animals, are not further induced by inflammatory stimuli and TGFβ1. Analysis in silico revealed that increased expression of receptor scavenger receptor (SR)-A, involved in Aβ uptake and cell activation, by microglia exposed to TGFβ, through a Smad3-dependent mechanism could be mediated by transcriptional co-factors Smad2/3 over the MSR1 gene. We discuss that changes of TGFβ-mediated regulation could at least partially mediate age-associated microglia changes, and, together with other changes on inflammatory response, could result in the reduction of protective activation and the potentiation of cytotoxicity of microglia, resulting in the promotion of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Francisca Cornejo
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Guillermo E Parada
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Jaime Eugenín
- Laboratory of Neural Systems, Faculty of Chemistry and Biology, Department of Biology, Universidad de Santiago de Chile Santiago, Chile
| |
Collapse
|
10
|
von Bernhardi R, Eugenín-von Bernhardi L, Eugenín J. Microglial cell dysregulation in brain aging and neurodegeneration. Front Aging Neurosci 2015; 7:124. [PMID: 26257642 PMCID: PMC4507468 DOI: 10.3389/fnagi.2015.00124] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 06/22/2015] [Indexed: 12/29/2022] Open
Abstract
Aging is the main risk factor for neurodegenerative diseases. In aging, microglia undergoes phenotypic changes compatible with their activation. Glial activation can lead to neuroinflammation, which is increasingly accepted as part of the pathogenesis of neurodegenerative diseases, including Alzheimer’s disease (AD). We hypothesize that in aging, aberrant microglia activation leads to a deleterious environment and neurodegeneration. In aged mice, microglia exhibit an increased expression of cytokines and an exacerbated inflammatory response to pathological changes. Whereas LPS increases nitric oxide (NO) secretion in microglia from young mice, induction of reactive oxygen species (ROS) predominates in older mice. Furthermore, there is accumulation of DNA oxidative damage in mitochondria of microglia during aging, and also an increased intracellular ROS production. Increased ROS activates the redox-sensitive nuclear factor kappa B, which promotes more neuroinflammation, and can be translated in functional deficits, such as cognitive impairment. Mitochondria-derived ROS and cathepsin B, are also necessary for the microglial cell production of interleukin-1β, a key inflammatory cytokine. Interestingly, whereas the regulatory cytokine TGFβ1 is also increased in the aged brain, neuroinflammation persists. Assessing this apparent contradiction, we have reported that TGFβ1 induction and activation of Smad3 signaling after inflammatory stimulation are reduced in adult mice. Other protective functions, such as phagocytosis, although observed in aged animals, become not inducible by inflammatory stimuli and TGFβ1. Here, we discuss data suggesting that mitochondrial and endolysosomal dysfunction could at least partially mediate age-associated microglial cell changes, and, together with the impairment of the TGFβ1-Smad3 pathway, could result in the reduction of protective activation and the facilitation of cytotoxic activation of microglia, resulting in the promotion of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | | | - Jaime Eugenín
- Laboratory of Neural Systems, Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile (USACH) Santiago, Chile
| |
Collapse
|
11
|
Lu F, Xu Y, Wang T. Advance of
transforming growth factor beta
in traumatic brain injury. IBRAIN 2015. [DOI: 10.1002/j.2769-2795.2015.tb00003.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Fan Lu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yang Xu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ting‐Hua Wang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
12
|
Context-Dependent Regulation of Autophagy by IKK-NF-κB Signaling: Impact on the Aging Process. Int J Cell Biol 2012; 2012:849541. [PMID: 22899934 PMCID: PMC3412117 DOI: 10.1155/2012/849541] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 06/21/2012] [Indexed: 12/19/2022] Open
Abstract
The NF-κB signaling system and the autophagic degradation pathway are crucial cellular survival mechanisms, both being well conserved during evolution. Emerging studies have indicated that the IKK/NF-κB signaling axis regulates autophagy in a context-dependent manner. IKK complex and NF-κB can enhance the expression of Beclin 1 and other autophagy-related proteins and stimulate autophagy whereas as a feedback response, autophagy can degrade IKK components. Moreover, NF-κB signaling activates the expression of autophagy inhibitors (e.g., A20 and Bcl-2/xL) and represses the activators of autophagy (BNIP3, JNK1, and ROS). Several studies have indicated that NF-κB signaling is enhanced both during aging and cellular senescence, inducing a proinflammatory phenotype. The aging process is also associated with a decline in autophagic degradation. It seems that the activity of Beclin 1 initiation complex could be impaired with aging, since the expression of Beclin 1 decreases as does the activity of type III PI3K. On the other hand, the expression of inhibitory Bcl-2/xL proteins increases with aging. We will review the recent literature on the control mechanisms of autophagy through IKK/NF-κB signaling and emphasize that NF-κB signaling could be a potent repressor of autophagy with ageing.
Collapse
|
13
|
The neuroprotective functions of transforming growth factor beta proteins. Int J Mol Sci 2012; 13:8219-8258. [PMID: 22942700 PMCID: PMC3430231 DOI: 10.3390/ijms13078219] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 05/24/2012] [Accepted: 06/19/2012] [Indexed: 12/26/2022] Open
Abstract
Transforming growth factor beta (TGF-β) proteins are multifunctional cytokines whose neural functions are increasingly recognized. The machinery of TGF-β signaling, including the serine kinase type transmembrane receptors, is present in the central nervous system. However, the 3 mammalian TGF-β subtypes have distinct distributions in the brain suggesting different neural functions. Evidence of their involvement in the development and plasticity of the nervous system as well as their functions in peripheral organs suggested that they also exhibit neuroprotective functions. Indeed, TGF-β expression is induced following a variety of types of brain tissue injury. The neuroprotective function of TGF-βs is most established following brain ischemia. Damage in experimental animal models of global and focal ischemia was shown to be attenuated by TGF-βs. In addition, support for their neuroprotective actions following trauma, sclerosis multiplex, neurodegenerative diseases, infections, and brain tumors is also accumulating. The review will also describe the potential mechanisms of neuroprotection exerted by TGF-βs including anti-inflammatory, -apoptotic, -excitotoxic actions as well as the promotion of scar formation, angiogenesis, and neuroregeneration. The participation of these mechanisms in the neuroprotective effects of TGF-βs during different brain lesions will also be discussed.
Collapse
|
14
|
Stress by noise produces differential effects on the proliferation rate of radial astrocytes and survival of neuroblasts in the adult subgranular zone. Neurosci Res 2011; 70:243-50. [DOI: 10.1016/j.neures.2011.03.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/28/2011] [Accepted: 03/31/2011] [Indexed: 01/22/2023]
|
15
|
Doyle KP, Cekanaviciute E, Mamer LE, Buckwalter MS. TGFβ signaling in the brain increases with aging and signals to astrocytes and innate immune cells in the weeks after stroke. J Neuroinflammation 2010; 7:62. [PMID: 20937129 PMCID: PMC2958905 DOI: 10.1186/1742-2094-7-62] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 10/11/2010] [Indexed: 01/18/2023] Open
Abstract
Background TGFβ is both neuroprotective and a key immune system modulator and is likely to be an important target for future stroke therapy. The precise function of increased TGF-β1 after stroke is unknown and its pleiotropic nature means that it may convey a neuroprotective signal, orchestrate glial scarring or function as an important immune system regulator. We therefore investigated the time course and cell-specificity of TGFβ signaling after stroke, and whether its signaling pattern is altered by gender and aging. Methods We performed distal middle cerebral artery occlusion strokes on 5 and 18 month old TGFβ reporter mice to get a readout of TGFβ responses after stroke in real time. To determine which cell type is the source of increased TGFβ production after stroke, brain sections were stained with an anti-TGFβ antibody, colocalized with markers for reactive astrocytes, neurons, and activated microglia. To determine which cells are responding to TGFβ after stroke, brain sections were double-labelled with anti-pSmad2, a marker of TGFβ signaling, and markers of neurons, oligodendrocytes, endothelial cells, astrocytes and microglia. Results TGFβ signaling increased 2 fold after stroke, beginning on day 1 and peaking on day 7. This pattern of increase was preserved in old animals and absolute TGFβ signaling in the brain increased with age. Activated microglia and macrophages were the predominant source of increased TGFβ after stroke and astrocytes and activated microglia and macrophages demonstrated dramatic upregulation of TGFβ signaling after stroke. TGFβ signaling in neurons and oligodendrocytes did not undergo marked changes. Conclusions We found that TGFβ signaling increases with age and that astrocytes and activated microglia and macrophages are the main cell types that undergo increased TGFβ signaling in response to post-stroke increases in TGFβ. Therefore increased TGFβ after stroke likely regulates glial scar formation and the immune response to stroke.
Collapse
Affiliation(s)
- Kristian P Doyle
- Department of Neurology and Neurological Sciences, Stanford University Medical School, Stanford, CA 94305-5489, USA
| | | | | | | |
Collapse
|
16
|
Mathieu P, Battista D, Depino A, Roca V, Graciarena M, Pitossi F. The more you have, the less you get: the functional role of inflammation on neuronal differentiation of endogenous and transplanted neural stem cells in the adult brain. J Neurochem 2009; 112:1368-85. [PMID: 20028453 DOI: 10.1111/j.1471-4159.2009.06548.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The differentiation of neural stem cells toward a neuronal phenotype is determined by the extracellular and intracellular factors that form the neurogenic niche. In this review, we discuss the available data on the functional role of inflammation and in particular, pro- and anti-inflammatory cytokines, on neuronal differentiation from endogenous and transplanted neural stem/progenitor cells. In addition, we discuss the role of microglial cell activation on these processes and the fact that microglial cell activation is not univocally associated with a pro-inflammatory milieu. We conclude that brain cytokines could be regarded as part of the endogenous neurogenic niche. In addition, we propose that accumulating evidence suggests that pro-inflammatory cytokines have a negative effect on neuronal differentiation, while anti-inflammatory cytokines exert an opposite effect. The clarification of the functional role of cytokines on neuronal differentiation will be relevant not only to better understand adult neurogenesis, but also to envisage complementary treatments to modulate cytokine action that could increase the therapeutic benefit of future progenitor/stem cell-based therapies.
Collapse
Affiliation(s)
- Patricia Mathieu
- Institute Leloir Foundation-IIBBA-CONICET, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
17
|
von Bernhardi R, Tichauer JE, Eugenín J. Aging-dependent changes of microglial cells and their relevance for neurodegenerative disorders. J Neurochem 2009; 112:1099-114. [PMID: 20002526 DOI: 10.1111/j.1471-4159.2009.06537.x] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Among multiple structural and functional brain changes, aging is accompanied by an increase of inflammatory signaling in the nervous system as well as a dysfunction of the immune system elsewhere. Although the long-held view that aging involves neurocognitive impairment is now dismissed, aging is a major risk factor for neurodegenerative diseases such as Alzheimer;s disease, Parkinson;s disease and Huntington's disease, among others. There are many age-related changes affecting the brain, contributing both to certain declining in function and increased frailty, which could singly and collectively affect neuronal viability and vulnerability. Among those changes, both inflammatory responses in aged brains and the altered regulation of toll like receptors, which appears to be relevant for understanding susceptibility to neurodegenerative processes, are linked to pathogenic mechanisms of several diseases. Here, we review how aging and pro-inflammatory environment could modulate microglial phenotype and its reactivity and contribute to the genesis of neurodegenerative processes. Data support our idea that age-related microglial cell changes, by inducing cytotoxicity in contrast to neuroprotection, could contribute to the onset of neurodegenerative changes. This view can have important implications for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta, Santiago, Chile.
| | | | | |
Collapse
|
18
|
Couillard-Despres S, Wuertinger C, Kandasamy M, Caioni M, Stadler K, Aigner R, Bogdahn U, Aigner L. Ageing abolishes the effects of fluoxetine on neurogenesis. Mol Psychiatry 2009; 14:856-864. [PMID: 19139747 DOI: 10.1038/mp.2008.147] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 11/26/2008] [Accepted: 12/15/2008] [Indexed: 11/09/2022]
|
19
|
Deng B, Glanzman D, Tidball JG. Nitric oxide generated by muscle corrects defects in hippocampal neurogenesis and neural differentiation caused by muscular dystrophy. J Physiol 2009; 587:1769-78. [PMID: 19237426 DOI: 10.1113/jphysiol.2008.166256] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) results from null mutation of dystrophin, a membrane-associated structural protein that is expressed in skeletal muscle. Dystrophin deficiency causes muscle membrane lesions, muscle degeneration and eventually death in afflicted individuals. However, dystrophin deficiency also causes cognitive defects that are difficult to relate to the loss of dystrophin. We assayed neurogenesis in the dentate gyrus (DG) in the mdx mouse model of DMD, using bromodeoxyuridine incorporation as a marker of proliferation and NeuN expression as a marker of differentiation. Our findings show that dystrophin mutation disrupts adult neurogenesis by promoting cell proliferation in the DG and suppressing neuronal differentiation. Because loss of dystrophin from muscle results in the secondary loss of neuronal nitric oxide synthase (nNOS), and NO is able to modulate neurogenesis, we assayed whether the genetic restoration of nNOS to mdx muscles corrected defects in adult, hippocampal neurogenesis. Assays of NO in the sera of active mice showed significant reductions in NO caused by the dystrophin mutation. However, over-expression of nNOS in the muscles of mdx mice increased serum NO and normalized cell proliferation and neuronal differentiation in the DG. These findings indicate that muscle-derived NO regulates adult neurogenesis in the brain and loss of muscle nNOS may underlie defects in the central nervous system in DMD.
Collapse
Affiliation(s)
- Bo Deng
- Molecular, Cellular and Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA
| | | | | |
Collapse
|
20
|
Choy KHC, de Visser Y, Nichols NR, van den Buuse M. Combined neonatal stress and young-adult glucocorticoid stimulation in rats reduce BDNF expression in hippocampus: effects on learning and memory. Hippocampus 2008; 18:655-67. [PMID: 18398848 DOI: 10.1002/hipo.20425] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Epidemiological studies suggest that multiple developmental disruptions are involved in the etiology of psychiatric illnesses including schizophrenia. In addition, altered expression of brain-derived neurotrophic factor (BDNF) has been implicated in these illnesses. In the present study, we examined the combined long-term effect of an early stress, in the form of maternal deprivation, and a later stress, simulated by chronic young-adult treatment with the stress hormone, corticosterone, on BDNF expression in the hippocampus of rats. To assess whether there were behavioral effects, which may correlate with the BDNF changes, learning and memory was tested in the Y-maze test for short term spatial memory, the Morris water maze for long-term spatial memory, and the T-maze test for working memory. Four groups of rats received either no stress, maternal deprivation, corticosterone treatment, or both. Dorsal hippocampus sections obtained from parallel groups were used for BDNF mRNA in situ hybridization. Rats which had undergone both maternal deprivation and corticosterone treatment displayed a unique and significant 25-35% reduction of BDNF expression in the dentate gyrus (DG), and similar trends in the CA1 and CA3 regions of the hippocampus. These "two-hit" animals exhibited a learning delay in the Morris water maze test, a marked deficit in the Y-maze, but little change in the T-maze test. However, some aspects of cognition were also altered in rats with either maternal deprivation or corticosterone treatment. This study demonstrates a persistent effect of two developmental disruptions on BDNF expression in the hippocampus, with parallel, but not completely correlative changes in learning and memory.
Collapse
Affiliation(s)
- Kwok Ho Christopher Choy
- Behavioural Neuroscience Laboratory, Mental Health Research Institute of Victoria, Melbourne, Australia
| | | | | | | |
Collapse
|
21
|
Rodricks CL, Gibbs ME, Jenkin G, Miller SL. The effect of hypoxia at different embryonic ages on impairment of memory ability in chicks. Int J Dev Neurosci 2007; 26:113-8. [PMID: 17904781 DOI: 10.1016/j.ijdevneu.2007.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 08/12/2007] [Accepted: 08/16/2007] [Indexed: 11/25/2022] Open
Abstract
Hypoxia during the prenatal period is a principal antecedent to cognitive impairment after birth. In this study we have investigated the duration, severity and timing of acute hypoxia during chick embryonic development to elucidate the relative importance of these factors. Our results show that 24h of hypoxia (exposure to 14% oxygen) at embryonic day 10 (E10) results in significant impairment of intermediate and long-term memory in the post-hatch chick, which is the same as we observed with 4 days of hypoxia. At E14, 24h of hypoxia, 5min of anoxia, but not 1h of hypoxia, resulted only in impaired long-term memory; the same as 4 days of hypoxia from E14. Corticosterone levels, measured post-hatch as an indicator of a stress response, were significantly elevated in response to E10 hypoxia, and E14 hypoxia (both 1 and 24h) and anoxia. In a separate experiment we exposed embryos to 24h of hypoxia from E6 to E16, and found that memory deficits resulted from hypoxia at E9 and E10, and E13-E15, while corticosterone concentrations at hatch were significantly raised following E10-E16 hypoxia. These results demonstrate that the developmental age when the insult occurs determines the nature of the cognitive deficit and, if the severity of the insult is sufficient, then the outcome, or deficits in memory ability, are consistent whether the insult is acute or chronic. Importantly, there are two critical stages in development, which in the chick are around E10 and E14, when acute hypoxia results in significant adverse cognitive effects after hatch. These time-points correspond to two different stages in growth and development.
Collapse
Affiliation(s)
- Candice L Rodricks
- Department of Physiology, Monash University, Clayton, 3800 Victoria, Australia
| | | | | | | |
Collapse
|
22
|
Aigner L, Bogdahn U. TGF-beta in neural stem cells and in tumors of the central nervous system. Cell Tissue Res 2007; 331:225-41. [PMID: 17710437 DOI: 10.1007/s00441-007-0466-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 07/04/2007] [Indexed: 10/22/2022]
Abstract
Mechanisms that regulate neural stem cell activity in the adult brain are tightly coordinated. They provide new neurons and glia in regions associated with high cellular and functional plasticity, after injury, or during neurodegeneration. Because of the proliferative and plastic potential of neural stem cells, they are currently thought to escape their physiological control mechanisms and transform to cancer stem cells. Signals provided by proteins of the transforming growth factor (TGF)-beta family might represent a system by which neural stem cells are controlled under physiological conditions but released from this control after transformation to cancer stem cells. TGF-beta is a multifunctional cytokine involved in various physiological and patho-physiological processes of the brain. It is induced in the adult brain after injury or hypoxia and during neurodegeneration when it modulates and dampens inflammatory responses. After injury, although TGF-beta is neuroprotective, it may limit the self-repair of the brain by inhibiting neural stem cell proliferation. Similar to its effect on neural stem cells, TGF-beta reveals anti-proliferative control on most cell types; however, paradoxically, many brain tumors escape from TGF-beta control. Moreover, brain tumors develop mechanisms that change the anti-proliferative influence of TGF-beta into oncogenic cues, mainly by orchestrating a multitude of TGF-beta-mediated effects upon matrix, migration and invasion, angiogenesis, and, most importantly, immune escape mechanisms. Thus, TGF-beta is involved in tumor progression. This review focuses on TGF-beta and its role in the regulation and control of neural and of brain-cancer stem cells.
Collapse
Affiliation(s)
- Ludwig Aigner
- Department of Neurology, University of Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany.
| | | |
Collapse
|
23
|
Rodricks CL, Miller SL, Jenkin G, Gibbs ME. The role of corticosterone in prehatch-induced memory deficits in chicks. Brain Res 2006; 1123:34-41. [PMID: 17045973 DOI: 10.1016/j.brainres.2006.09.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Revised: 09/06/2006] [Accepted: 09/11/2006] [Indexed: 11/27/2022]
Abstract
We have previously shown that prehatch hypoxia (14% oxygen for 24 h), at E10 or E14 of chick embryonic development, produces significant memory deficits, with E10 hypoxia significantly affecting short-term memory and the subsequent formation of long-term memory, whereas E14 hypoxia only affects long-term memory. One of the consequences of hypoxia is the release of stress hormones and we found in this study that hypoxia at E10 or E14 induced a significant increase in circulating corticosterone immediately after the cessation of hypoxia (E11 and E15, respectively). Corticosterone levels remained significantly elevated at hatch in the E14 hypoxia group. This study describes the effect of a single, in ovo, injection of corticosterone on subsequent memory ability in hatched chicks. It was found that corticosterone (0.2 nmol/egg) at E10 or E14 mimicked the memory deficits produced by hypoxia at the same prehatch ages. Embryos treated with corticosterone at E10 had poor short-term memory at hatch, whereas corticosterone administration at E14 resulted in poor long-term memory. Embryos treated with corticosterone at E16 had raised circulating corticosterone levels at hatch, but did not have impaired memory. Treatment with corticosterone at E10, E12, E14 and E16 produced the same cognitive outcomes as hypoxia at the same prehatch ages. However, elevated plasma corticosterone levels at hatch did not necessarily cause the impaired memory processing. Raised levels were observed after treatment at E14 when memory processing was impaired, at E16 when memory was not impaired and not at E10 when memory was impaired. This suggests that an acute rather than sustained increase in plasma corticosterone at particular developmental ages is the cause of impaired memory processing seen at hatch.
Collapse
Affiliation(s)
- Candice L Rodricks
- Department of Pharmacology, Monash University, Clayton 3800, Victoria, Australia
| | | | | | | |
Collapse
|
24
|
Buckwalter MS, Yamane M, Coleman BS, Ormerod BK, Chin JT, Palmer T, Wyss-Coray T. Chronically increased transforming growth factor-beta1 strongly inhibits hippocampal neurogenesis in aged mice. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:154-64. [PMID: 16816369 PMCID: PMC1698757 DOI: 10.2353/ajpath.2006.051272] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
There is increasing evidence that hippocampal learning correlates strongly with neurogenesis in the adult brain. Increases in neurogenesis after brain injury also correlate with improved outcomes. With aging the capacity to generate new neurons decreases dramatically, both under normal conditions and after injury. How this decrease occurs is not fully understood, but we hypothesized that transforming growth factor (TGF)-beta1, a cell cycle regulator that rapidly increases after injury and with age, might play a role. We found that chronic overproduction of TGF-beta1 from astrocytes almost completely blocked the generation of new neurons in aged transgenic mice. Even young adult TGF-beta1 mice had 60% fewer immature, doublecortin-positive, hippocampal neurons than wild-type littermate controls. Bromodeoxyuridine labeling of dividing cells in 2-month-old TGF-beta1 mice confirmed this decrease in neuro-genesis and revealed a similar decrease in astrogenesis. Treatment of early neural progenitor cells with TGF-beta1 inhibited their proliferation. This strongly suggests that TGF-beta1 directly affects these cells before their differentiation into neurons and astrocytes. Together, these data show that TGF-beta1 is a potent inhibitor of hippocampal neural progenitor cell proliferation in adult mice and suggest that it plays a key role in limiting injury and age-related neurogenesis.
Collapse
Affiliation(s)
- Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, SUMC Rm. 343A, Stanford, CA 94305-5235, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Paratore S, Parenti R, Torrisi A, Copani A, Cicirata F, Cavallaro S. Genomic profiling of cortical neurons following exposure to beta-amyloid. Genomics 2006; 88:468-79. [PMID: 16904863 DOI: 10.1016/j.ygeno.2006.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 06/12/2006] [Accepted: 06/16/2006] [Indexed: 10/24/2022]
Abstract
In vitro and in vivo studies have shown that beta-amyloid peptide induces neuronal cell death. To explore the molecular basis underlying beta-amyloid-induced toxicity, we analyzed gene expression profiles of cultured rat cortical neurons treated for 24 and 48 h with synthetic beta-amyloid peptide. From the 8740 genes interrogated by oligonucleotide microarray analysis, 241 genes were found to be differentially expressed and segregated into distinct clusters. Functional clustering based on gene ontologies showed coordinated expression of genes with common biological functions and metabolic pathways. The comparison with genes differentially expressed in cerebellar granule neurons following serum and potassium deprivation indicates the existence of common regulatory mechanisms underlying neuronal cell death. Our results offer a genomic view of the changes that accompany beta-amyloid-induced neurodegeneration.
Collapse
Affiliation(s)
- Sabrina Paratore
- Functional Genomics Center, Institute of Neurological Sciences, Italian National Research Council, Italy
| | | | | | | | | | | |
Collapse
|
26
|
Bravo JA, Parra CS, Arancibia S, Andrés S, Morales P, Herrera-Marschitz M, Herrera L, Lara HE, Fiedler JL. Adrenalectomy promotes a permanent decrease of plasma corticoid levels and a transient increase of apoptosis and the expression of Transforming Growth Factor beta1 (TGF-beta1) in hippocampus: effect of a TGF-beta1 oligo-antisense. BMC Neurosci 2006; 7:40. [PMID: 16712723 PMCID: PMC1481618 DOI: 10.1186/1471-2202-7-40] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Accepted: 05/19/2006] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Corticosterone reduction produced by adrenalectomy (ADX) induces apoptosis in dentate gyrus (DG) of the hippocampus, an effect related to an increase in the expression of the pro-apoptotic gene bax. However it has been reported that there is also an increase of the anti-apoptotic gene bcl-2, suggesting the promotion of a neuroprotective phenomenon, perhaps related to the expression of transforming growth factor beta1 (TGF-beta1). Thus, we have investigated whether TGF-beta1 levels are induced by ADX, and whether apoptosis is increased by blocking the expression of TGF-beta1 with an antisense oligonucleotide (ASO) administered intracerebrally in corticosterone depleted rats. RESULTS It was observed an increase of apoptosis in DG, 2 and 5 days after ADX, in agreement with a reduction of corticosterone levels. However, the effect of ADX on the number of apoptotic positive cells in DG was decreased 5 days after the lesion. In CA1-CA3 regions, the effect was only observed 2 days after ADX. TGF-beta1 mRNA levels were increased 2 days after ADX. The sustained intracerebro-ventricular administration of a TGF-beta1 ASO via an osmotic mini pump increased apoptosis levels in CA and DG regions 5 days after ADX as well as sham-operated control animals. No significant effect was observed following a scrambled-oligodeoxynucleotide treatment. CONCLUSION The changes in both the pattern and the magnitude of apoptotic-cell morphology observed 2 and 5 days after ADX suggest that, as a consequence of the reduction of corticosteroids, some trophic mechanisms restricting cell death to a particular time window are elicited. Sustained intracerebral administration of TGF-beta1 ASO increased the apoptosis promoted by ADX, suggesting that TGF-beta1 plays an anti-apoptotic role in vivo in hippocampus.
Collapse
Affiliation(s)
- Javier A Bravo
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| | - Claudio S Parra
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| | - Sandor Arancibia
- Laboratory of Molecular Mechanisms of Neurodegenerative Diseases, Université de Montpellier, Montpellier, France
| | - Sergio Andrés
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| | - Paola Morales
- Programmes of Molecular & Clinical Pharmacology ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Mario Herrera-Marschitz
- Programmes of Molecular & Clinical Pharmacology ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Luisa Herrera
- Human Genetics, ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Hernán E Lara
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| | - Jenny L Fiedler
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| |
Collapse
|
27
|
Magri F, Cravello L, Barili L, Sarra S, Cinchetti W, Salmoiraghi F, Micale G, Ferrari E. Stress and dementia: the role of the hypothalamicpituitary-adrenal axis. Aging Clin Exp Res 2006; 18:167-70. [PMID: 16702789 DOI: 10.1007/bf03327435] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hippocampus plays a crucial role in learning and memory and, in spite of its remarkable plasticity, it is also particularly sensitive to stress hormones due to its high concentration of corticosteroid receptors. Indeed, adrenal steroids modulate hippocampal plasticity, acting on excitability and long term potentiation or depression. By a chronobiological approach, we studied the cortisol and DHEAS secretion in clinically healthy old subjects and in age-matched demented patients, including both the degenerative and the vascular type. When compared to young controls, both clinically healthy elderly subjects and demented patients, particularly those with AD, had significantly higher cortisol levels at night time, i.e. at the moment of the maximal sensitivity of HPA axis to stimulatory or inhibitory inputs. At the same time, a clear age- and disease-dependent reduction of DHEAS secretion was found. Thus the cortisol to DHEAS molar ratio was significantly higher in healthy old subjects, and even more in demented patients, when compared to young controls, and significantly linked to both age and cognitive impairment. Finally, the quantitative and qualitative changes of the adrenal secretory pattern were significantly correlated with the decline of hippocampal volumes, measured by MRI. In conclusion, several lines of evidence deal with a pathogenetic role of stress hormones in the occurrence and progression of cognitive disorders in elderly subjects. The consequent hippocampal neuronal impairment may in turn be responsible for the continuous activation of HPA axis and the increased hypothalamic expression of vasopressin and corticotropin releasing hormone.
Collapse
Affiliation(s)
- Flavia Magri
- UO of Internal Medicine and Endocrinology, IRCCS S. Maugeri Foundation, Pavia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Herrera-Molina R, von Bernhardi R. Transforming growth factor-β1 produced by hippocampal cells modulates microglial reactivity in culture. Neurobiol Dis 2005; 19:229-36. [PMID: 15837578 DOI: 10.1016/j.nbd.2005.01.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Revised: 11/23/2004] [Accepted: 01/04/2005] [Indexed: 11/30/2022] Open
Abstract
Activated microglia produce superoxide anion (O2-) and nitric oxide (NO), both of which can be neurotoxic. To identify regulatory mechanisms that might modulate over-activation of microglia, we evaluated the inhibition of microglial activation by factors secreted by hippocampal cells. Supernatants from hippocampal cell cultures (Hippocampal-Cm) prevented microglial O2- and NO production. LAP-TGF beta1 was present in the Hippocampal-Cm as shown by immunoblot and a TGF beta1-dependent proliferation-inhibition bioassay. LAP-TGF beta1 and TGFbeta activity increased in hippocampal cultures exposed to proinflammatory conditions (LPS and Interferon-gamma). The inhibition of O2- and NO production by Hippocampal-Cm was mimicked by the addition of recombinant TGF beta1. Treating Hippocampal-Cm with an antibody against TGF beta1 to neutralize its activity eliminated its ability to inhibit O2- and NO production. Our findings suggest that the TGF beta1 secreted by hippocampal cells modulated microglial activity. We propose that in pathological conditions, impairment of this modulatory mechanism could enhance microglia-mediated neurotoxicity.
Collapse
Affiliation(s)
- Rodrigo Herrera-Molina
- Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | | |
Collapse
|
29
|
Nichols NR, Agolley D, Zieba M, Bye N. Glucocorticoid regulation of glial responses during hippocampal neurodegeneration and regeneration. ACTA ACUST UNITED AC 2005; 48:287-301. [PMID: 15850668 DOI: 10.1016/j.brainresrev.2004.12.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2004] [Accepted: 12/09/2004] [Indexed: 10/25/2022]
Abstract
Glucocorticoids can prevent or accelerate neurodegeneration in the adult rat hippocampus. To investigate these actions of glucocorticoids, we previously cloned genes from the hippocampus. Adrenalectomy specifically increased glial fibrillary acidic protein and transforming growth factor (TGF)-beta1 mRNAs in the dentate gyrus and these effects were dependent on induced apoptosis. Corticosterone treatment prevented apoptosis, and decreased glial activation and the influx of activated microglia. Since these effects are opposite to injury and neurodegeneration, we propose that they represent adaptive actions of glucocorticoids, preventing cellular defense mechanisms from overshooting. We used adrenalectomy as a model to investigate how adult granule neurons die in vivo and the effects of neurotrophic factors in protecting against apoptosis. Neurotrophin-4/5 and TGF-beta1 protected granule neurons against adrenalectomy-induced apoptosis. Since neurogenesis is also greatly increased in the dentate gyrus following adrenalectomy, we compared the time course of birth and death with glial responses. TGF-beta1 mRNA increased before the detection of dying cells in the dentate gyrus, which was coincident with increased proliferation in the neurogenic zone. Glucocorticoids also increased Ndrg2 mRNA in glia in the neurogenic zone; Ndrg2 is a member of a novel gene family involved in neural differentiation and synapse formation. Therefore, studying the effects of glucocorticoid manipulation on the dentate gyrus is increasing our understanding of how mature neurons die by apoptosis and the role of glia in induced apoptosis and neurogenesis. Discovering how endocrine and inflammatory responses regulate neuron birth and survival is important for developing successful neuron replacement strategies to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Nancy R Nichols
- Department of Physiology, Building 13F, Monash University, Clayton, Australia.
| | | | | | | |
Collapse
|
30
|
Rodricks CL, Rose IA, Camm EJ, Jenkin G, Miller SL, Gibbs ME. The effect of prenatal hypoxia and malnutrition on memory consolidation in the chick. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 148:113-9. [PMID: 14757525 DOI: 10.1016/j.devbrainres.2003.10.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The contribution of hypoxia and malnutrition to cognitive impairments was investigated in chicks incubated in conditions of reduced gas exchange. Previous research has shown that reducing gas exchange during incubation by wrapping half the eggshell with an impermeable membrane results in impaired cognitive ability in young chicks. The results were interpreted within a three stage sequential model of memory using discriminated bead avoidance learning. Reducing gas exchange for 4 days from day 10 or 14, of the 21-day incubation, inhibits memory formation and consolidation into permanent storage. The nature of the cognitive deficit depended on the timing of the insult. Environmental hypoxia (14% oxygen), induced from days 10 to 14 and from days 14 to 18, replicated the memory deficits found previously when eggs were partially wrapped with a membrane. Oxygen is necessary to break down food and to provide energy to build tissue proteins, and therefore hypoxia (partial wrapping or environmental incubation) may indirectly cause malnutrition. Malnutrition, induced by removing 5%, 7.5% or 10% albumin from the egg prior to incubation, had no significant effect on memory consolidation. Raised corticosterone levels occurred in chicks malnourished by 5% and 7.5%, but brain sparing was only evident in chicks with 7.5% albumin removal. Hatch rates were very low in 10% malnourished chicks. Using the chick as a model of prenatal stress, we have been able to isolate the effects of hypoxia from contributing maternal factors.
Collapse
Affiliation(s)
- Candice L Rodricks
- Department of Physiology, Monash University, Melbourne, Victoria 3800, Australia.
| | | | | | | | | | | |
Collapse
|
31
|
Morale MC, Serra PA, Delogu MR, Migheli R, Rocchitta G, Tirolo C, Caniglia S, Testa N, L'Episcopo F, Gennuso F, Scoto GM, Barden N, Miele E, Desole MS, Marchetti B. Glucocorticoid receptor deficiency increases vulnerability of the nigrostriatal dopaminergic system: critical role of glial nitric oxide. FASEB J 2003; 18:164-6. [PMID: 14630699 DOI: 10.1096/fj.03-0501fje] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Glucocorticoids (GCs) exert via glucocorticoid receptors (GRs) potent anti-inflammatory and immunosuppressive effects. Emerging evidence indicates that an inflammatory process is involved in dopaminergic nigro-striatal neuronal loss in Parkinson's disease. We here report that the GR deficiency of transgenic (Tg) mice expressing GR antisense RNA from early embryonic life has a dramatic impact in "programming" the vulnerability of dopaminergic neurons to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The GR deficiency of Tg mice exacerbates MPTP-induced toxicity to dopaminergic neurons, as revealed by both severe loss of tyrosine hydroxylase positive nigral neurons and sharp decreases in striatal levels of dopamine and its metabolites. In addition, the late increase in dopamine oxidative metabolism and ascorbic acid oxidative status in GR-deficient mice was far greater than in wild-type (Wt) mice. Inducible nitric oxide synthase (iNOS) was sharply increased in activated astrocytes, macrophages/microglia of GR-deficient as compared with Wt mice. Moreover, GR-deficient microglia produced three- to fourfold higher nitrite levels than Wt mice; these increases preceded the loss of dopaminergic function and were resistant to GR the inhibitory effect of GC, pointing to peroxynitrites as candidate neurotoxic effectors. The iNOS inhibitor N6-(1-iminoethyl)-L-lysine normalized vulnerability of Tg mice, thus establishing a novel link between genetic impairment of GR function and vulnerability to MPTP.
Collapse
Affiliation(s)
- Maria Concetta Morale
- OASI Institute for Research and Care on Mental Retardation and Brain Aging (IRCCS), Neuropharmacology Section, 94018 Troina, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Nichols NR, Zieba M, Bye N. Do glucocorticoids contribute to brain aging? BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2001; 37:273-86. [PMID: 11744092 DOI: 10.1016/s0165-0173(01)00131-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The hippocampus, an area with abundant glucocorticoid receptors, continues to be the focus of research on effects of glucocorticoids on the aging brain. Based on recent studies, the primary structural change found during aging is synaptic loss, rather than neuronal loss. High levels of glucocorticoids are associated with synaptic loss in the hippocampus, hippocampal atrophy, and cognitive decline during aging in some individuals. However, increasing levels of glucocorticoid are not always found since early experiences can alter sensitivity to negative feedback and the level of activation of the hypothalamic-pituitary-adrenal axis in aged individuals. New ways in which glucocorticoids may contribute to brain aging are discussed, including decreased responses to glucocorticoids possibly as a result of decreased glucocorticoid receptors and also altered regulation of neuronal turnover in the dentate gyrus. Decreased responsiveness of glial fibrillary acidic protein to glucocorticoids during aging could facilitate reactive gliosis and loss of synapses by altering neuron-astrocyte interactions. Neuronal turnover is regulated by glucocorticoids in the dentate gyrus where ongoing neurogenesis may be important for hippocampal-based memory formation in adulthood. Although the age-related decline in neurogenesis can be reversed by removal of adrenal steroids, the death of dentate granule neurons is also greatly increased by this treatment. Recent studies show age-related resistance to induced apoptosis and neurogenesis in the dentate gyrus following adrenalectomy, which is associated with increased expression of transforming growth factor-beta1. Therefore, the contribution of glucocorticoids to brain aging depends on the physiological and cellular context and some of these effects are reversible.
Collapse
Affiliation(s)
- N R Nichols
- Department of Physiology, Monash University, PO Box 13F, 3800, Victoria, Australia.
| | | | | |
Collapse
|