51
|
Zhu H, Wang Z, Yu J, Yang X, He F, Liu Z, Che F, Chen X, Ren H, Hong M, Wang J. Role and mechanisms of cytokines in the secondary brain injury after intracerebral hemorrhage. Prog Neurobiol 2019; 178:101610. [PMID: 30923023 DOI: 10.1016/j.pneurobio.2019.03.003] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 12/18/2022]
Abstract
Intracerebral hemorrhage (ICH) is a common and severe cerebrovascular disease that has high mortality. Few survivors achieve self-care. Currently, patients receive only symptomatic treatment for ICH and benefit poorly from this regimen. Inflammatory cytokines are important participants in secondary injury after ICH. Increases in proinflammatory cytokines may aggravate the tissue injury, whereas increases in anti-inflammatory cytokines might be protective in the ICH brain. Inflammatory cytokines have been studied as therapeutic targets in a variety of acute and chronic brain diseases; however, studies on ICH are limited. This review summarizes the roles and functions of various pro- and anti-inflammatory cytokines in secondary brain injury after ICH and discusses pathogenic mechanisms and emerging therapeutic strategies and directions for treatment of ICH.
Collapse
Affiliation(s)
- Huimin Zhu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Zhiqiang Wang
- Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Jixu Yu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China; Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China; Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xiuli Yang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Feng He
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Zhenchuan Liu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China.
| | - Fengyuan Che
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China; Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China.
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Honglei Ren
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Hong
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
52
|
Rahman MR, Islam T, Turanli B, Zaman T, Faruquee HM, Rahman MM, Mollah MNH, Nanda RK, Arga KY, Gov E, Moni MA. Network-based approach to identify molecular signatures and therapeutic agents in Alzheimer's disease. Comput Biol Chem 2018; 78:431-439. [PMID: 30606694 DOI: 10.1016/j.compbiolchem.2018.12.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/25/2018] [Indexed: 01/15/2023]
Abstract
Alzheimer's disease (AD) is a dynamic degeneration of the brain with progressive dementia. Considering the uncertainties in its molecular mechanism, in the present study, we employed network-based integrative analyses, and aimed to explore the key molecules and their associations with small drugs to identify potential biomarkers and therapeutic agents for the AD. First of all, we studied a transcriptome dataset and identified 1521 differentially expressed genes (DEGs). Integration of transcriptome data with protein-protein and transcriptional regulatory interactions resulted with central (hub) proteins (UBA52, RAC1, CREBBP, AR, RPS11, SMAD3, RPS6, RPL12, RPL15, and UBC), regulatory transcription factors (FOXC1, GATA2, YY1, FOXL1, NFIC, E2F1, USF2, SRF, PPARG, and JUN) and microRNAs (mir-335-5p, mir-26b-5p, mir-93-5p, mir-124-3p, mir-17-5p, mir-16-5p, mir-20a-5p, mir-92a-3p, mir-106b-5p, and mir-192-5p) as key signaling and regulatory molecules associated with transcriptional changes for the AD. Considering these key molecules as potential therapeutic targets and Connectivity Map (CMap) architecture, candidate small molecular agents (such as STOCK1N-35696) were identified as novel potential therapeutics for the AD. This study presents molecular signatures at RNA and protein levels which might be useful in increasing discernment of the molecular mechanisms, and potential drug targets and therapeutics to design effective treatment strategies for the AD.
Collapse
Affiliation(s)
- Md Rezanur Rahman
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh; Department of Biochemistry and Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajgonj, Bangladesh
| | - Tania Islam
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
| | - Beste Turanli
- Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey; Department of Bioengineering, Marmara University, Istanbul, Turkey
| | - Toyfiquz Zaman
- Department of Biochemistry and Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajgonj, Bangladesh
| | - Hossain Md Faruquee
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh; Translational Health, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Md Mafizur Rahman
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
| | - Md Nurul Haque Mollah
- Laboratory of Bioinformatics, Department of Statistics, University of Rajshahi, Rajshahi, Bangladesh
| | - Ranjan Kumar Nanda
- Translational Health, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | | | - Esra Gov
- Department of Bioengineering, Adana Science and Technology University, Adana, Turkey.
| | - Mohammad Ali Moni
- The University of Sydney, Sydney Medical School, School of Medical Sciences, Discipline of Biomedical Science, Sydney, New South Wales, Australia.
| |
Collapse
|
53
|
Ding J, Kong W, Mou X, Wang S. Construction of Transcriptional Regulatory Network of Alzheimer’s Disease Based on PANDA Algorithm. Interdiscip Sci 2018; 11:226-236. [DOI: 10.1007/s12539-018-0297-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/09/2018] [Accepted: 04/03/2018] [Indexed: 11/27/2022]
|
54
|
Chong JR, Chai YL, Lee JH, Howlett D, Attems J, Ballard CG, Aarsland D, Francis PT, Chen CP, Lai MKP. Increased Transforming Growth Factor β2 in the Neocortex of Alzheimer's Disease and Dementia with Lewy Bodies is Correlated with Disease Severity and Soluble Aβ42 Load. J Alzheimers Dis 2018; 56:157-166. [PMID: 27911312 DOI: 10.3233/jad-160781] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Of the three transforming growth factor (TGF)-β isoforms known, TGFβ1 deficits have been widely reported in Alzheimer's disease (AD) and studied as a potential therapeutic target. In contrast, the status of TGFβ2, which has been shown to mediate amyloid-β (Aβ)-mediated neuronal death, are unclear both in AD and in Lewy body dementias (LBD) with differential neuritic plaque and neurofibrillary tangle burden. OBJECTIVE To measure neocortical TGFβ2 levels and their correlations with neuropathological and clinical markers of disease severity in a well-characterized cohort of AD as well as two clinical subtypes of LBD, dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD), known to manifest relatively high and low Aβ plaque burden, respectively. METHODS Postmortem samples from temporal cortex (BA21) were measured for TGFβ2 using a Luminex-based platform, and correlated with scores for neuritic plaques, neurofibrillary tangles, α-synuclein pathology, dementia severity (as measured by annual decline of Mini-Mental State Examination scores) as well as soluble and total fractions of brain Aβ42. RESULTS TGFβ2 was significantly increased in AD and DLB, but not in PDD. TGFβ2 also correlated with scores for neurofibrillary tangles, Lewy bodies (within the LBD group), dementia severity, and soluble Aβ42 concentration, but not with neuritic plaque scores, total Aβ42, or monomeric α-synuclein immunoreactivity. CONCLUSIONS TGFβ2 is increased in the temporal cortex of AD and DLB, and its correlations with neuropathological and clinical markers of disease severity as well as with soluble Aβ42 load suggest a potential pathogenic role in mediating the neurotoxicity of non-fibrillar Aβ. Our study also indicates the potential utility of targeting TGFβ2 in pharmacotherapeutic approaches to AD and DLB.
Collapse
Affiliation(s)
- Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jasinda H Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Howlett
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Johannes Attems
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Clive G Ballard
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Dag Aarsland
- Department of Neurobiology, Ward Sciences and Society, Karolinska Institute, Stockholm, Sweden.,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Paul T Francis
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| |
Collapse
|
55
|
Monacelli F, Cea M, Borghi R, Odetti P, Nencioni A. Do Cancer Drugs Counteract Neurodegeneration? Repurposing for Alzheimer's Disease. J Alzheimers Dis 2018; 55:1295-1306. [PMID: 27834781 DOI: 10.3233/jad-160840] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In spite of in depth investigations in the field of the amyloid cascade hypothesis, so far, no disease modifying therapy has been developed for Alzheimer's disease (AD). The pathophysiology provides some evidence of the inverse correlation between cancer and AD. Both AD and cancer are characterized by abnormal cellular behaviors; trigger factors along with a meta synchronously action is expected to drive cancer or neurodegeneration, supporting, respectively, progressive neuronal loss or uncontrolled cell proliferation in cancer cells. So far, cancer and AD are seemingly two opposite ends of the same biological spectrum. Basic science increasingly indicates shared molecular mechanisms between cancer and AD and gives weight to key relevant biological theories; according to them, the inverse tuning of clustered gene expression, the sharing of mutual independent pathway or the deregulated unfolded proteins system (UPR) may count for this inverse association. Additionally, the common biological background gave credibility to the recent discovery of a repurposing role for cancer drugs in AD. It refers to the development of new uses for existing pharmaceuticals having the same role as the original mechanism or to the discovery of a new drug action with disease modifying effects. The present review summarizes the most important biological theories that link neurodegeneration and cancer and provides an up-to-date revision of the repurposing cancer agents for AD. The review also addresses the gap of knowledge, since drug cancer repositioning holds an important promise but further investigations are warranted to ascertain the clinical relevance of such attractive clinical candidate compounds for AD.
Collapse
Affiliation(s)
- Fiammetta Monacelli
- Section of Geriatrics, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Michele Cea
- Section of Haematology, Department of Internal Medicine and Medical Specialties, (DIMI), University of Genoa, Genoa, Italy
| | - Roberta Borghi
- Section of Geriatrics, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Patrizio Odetti
- Section of Geriatrics, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Alessio Nencioni
- Section of Geriatrics, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| |
Collapse
|
56
|
Tapella L, Cerruti M, Biocotino I, Stevano A, Rocchio F, Canonico PL, Grilli M, Genazzani AA, Lim D. TGF-β2 and TGF-β3 from cultured β-amyloid-treated or 3xTg-AD-derived astrocytes may mediate astrocyte-neuron communication. Eur J Neurosci 2018; 47:211-221. [DOI: 10.1111/ejn.13819] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Matteo Cerruti
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Isabella Biocotino
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Alessio Stevano
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Francesca Rocchio
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| |
Collapse
|
57
|
TGF-β1 protection against Aβ1–42-induced hippocampal neuronal inflammation and apoptosis by TβR-I. Neuroreport 2018; 29:141-146. [DOI: 10.1097/wnr.0000000000000940] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
58
|
Kashima R, Hata A. The role of TGF-β superfamily signaling in neurological disorders. Acta Biochim Biophys Sin (Shanghai) 2018; 50:106-120. [PMID: 29190314 PMCID: PMC5846707 DOI: 10.1093/abbs/gmx124] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
The TGF-β superfamily signaling is involved in a variety of biological processes during embryogenesis and in adult tissue homeostasis. Faulty regulation of the signaling pathway that transduces the TGF-β superfamily signals accordingly leads to a number of ailments, such as cancer and cardiovascular, metabolic, urinary, intestinal, skeletal, and immune diseases. In recent years, a number of studies have elucidated the essential roles of TGF-βs and BMPs during neuronal development in the maintenance of appropriate innervation and neuronal activity. The new advancement implicates significant roles of the aberrant TGF-β superfamily signaling in the pathogenesis of neurological disorders. In this review, we compile a number of reports implicating the deregulation of TGF-β/BMP signaling pathways in the pathogenesis of cognitive and neurodegenerative disorders in animal models and patients. We apologize in advance that the review falls short of providing details of the role of TGF-β/BMP signaling or mechanisms underlying the pathogenesis of neurological disorders. The goal of this article is to reveal a gap in our knowledge regarding the association between TGF-β/BMP signaling pathways and neuronal tissue homeostasis and development and facilitate the research with a potential to develop new therapies for neurological ailments by modulating the pathways.
Collapse
Affiliation(s)
- Risa Kashima
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
59
|
Fraga VG, Guimarães HC, Teixeira AL, Barbosa MT, Carvalho MG, Caramelli P, Gomes KB. Polymorphisms in cytokine genes influence cognitive and functional performance in a population aged 75 years and above. Int J Geriatr Psychiatry 2017; 32:1401-1410. [PMID: 27891653 DOI: 10.1002/gps.4627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 10/20/2016] [Accepted: 10/21/2016] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To investigate the frequency of the cytokine single nucleotide polymorphisms (SNPs) tumor necrosis factor (TNF)-α -308G > A, tumor growth factor (TGF)-β1 codon +10C > T, TGF-β1 codon +25G > C, interleukin (IL)-10 -1082A > G, IL-10 -819C > T, IL-10 -592C > A, IL-6 -174G > C, and IFN-γ +874T > A in a sample of healthy and cognitively impaired elderlies and to verify the probable association between these SNPs and cognitive and functional performance of subjects aged 75 years and above. METHODS 259 Brazilian subjects were included, comprising 81 with cognitive impairment no dementia (CIND) and 54 demented seniors (together made up the cognitively impaired group, CI) and 124 age-matched and gender-matched cognitively healthy controls (CHS). The genotyping was performed by multiplex polymerase chain reaction. The cognitive performance was evaluated by Mini-Mental State Examination Brief Cognitive Screening Battery. The functional performance was accessed by Functional Activities Questionnaire. RESULTS The CClower genotype of TGF-β1 codon +25G > C was frequent in both patient groups. The TThigher genotype of INF-γ +874T > A was less frequent in the dementia group. IL-10 haplotypes of lower expression were more frequent among CIND and demented patients. In CI, individuals with genetic variants that produce higher expression of TGF-β1, INF-γ, and IL-10 showed better normalized cognitive performance. Additionally, the Alower allele of INF-γ +874T > A was related to worse functional performance in CI, while the Alower allele of TNF-α -308G > A was associated with better cognitive and functional scores in the CIND group. CONCLUSIONS Our findings suggest a potential role for certain cytokine SNPs in the development of CIND and dementia, which may influence the functional and cognitive performance of these patients. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Vanessa G Fraga
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Henrique C Guimarães
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Antônio L Teixeira
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maira T Barbosa
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria G Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paulo Caramelli
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina B Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
60
|
Ham S, Kim TK, Lee S, Tang YP, Im HI. MicroRNA Profiling in Aging Brain of PSEN1/PSEN2 Double Knockout Mice. Mol Neurobiol 2017; 55:5232-5242. [PMID: 28879407 DOI: 10.1007/s12035-017-0753-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 08/23/2017] [Indexed: 01/09/2023]
Abstract
MicroRNAs are small non-coding RNAs that function as regulators of gene expression. The altered expression of microRNAs influences the pathogenesis of Alzheimer's disease. Many researchers have focused on studies based on the relatively distinctive etiology of familial Alzheimer's disease due to the absence of risk factors in the pathogenesis of sporadic Alzheimer's disease. Although there is a limitation in Alzheimer's disease studies, both Alzheimer's disease types have a common risk factor-aging. No study to date has examined the aging factor in Alzheimer's disease animal models with microRNAs. To investigate the effect of aging on the changes in microRNA expressions in the Alzheimer's disease animal model, we selected 37 hippocampal microRNAs whose expression in 12- and 18-month aged mice changed significantly using microRNA microarray. On the basis of bioinformatics databases, 30 hippocampal microRNAs and their putative targets of PSEN1/PSEN2 double knockout mice were included in 28 pathways such as the wnt signaling pathway and ubiquitin-mediated proteolysis pathway. Cortical microRNAs and its putative targets involved in pathological aging were included in only four pathways such as the heparin sulfate biosynthesis. The altered expressions of these hippocampal microRNAs were associated to the imbalance between neurotoxic and neuroprotective functions and seemed to affect neurodegeneration in PSEN1/PSEN2 double knockout mice more severely than in wild-type mice. This microRNA profiling suggests that microRNAs play potential roles in the normal aging process, as well as in the Alzheimer's disease process.
Collapse
Affiliation(s)
- Suji Ham
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.,Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Tae Kyoo Kim
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.,Department of Biology, Boston University, Boston, 02215, USA
| | - Sangjoon Lee
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Ya-Ping Tang
- Neuroscience Center of Excellence, Louisiana State University Health New Orleans Sciences Center, New Orleans, LA, 70112, USA
| | - Heh-In Im
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea. .,Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea. .,Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
61
|
GULP1/CED-6 ameliorates amyloid-β toxicity in a Drosophila model of Alzheimer's disease. Oncotarget 2017; 8:99274-99283. [PMID: 29245900 PMCID: PMC5725091 DOI: 10.18632/oncotarget.20062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/30/2017] [Indexed: 01/24/2023] Open
Abstract
Amyloidogenic processing of APP by β- and γ-secretases leads to the generation of amyloid-β peptide (Aβ), and the accumulation of Aβ in senile plaques is a hallmark of Alzheimer’s disease (AD). Understanding the mechanisms of APP processing is therefore paramount. Increasing evidence suggests that APP intracellular domain (AICD) interacting proteins influence APP processing. In this study, we characterized the overexpression of AICD interactor GULP1 in a Drosophila AD model expressing human BACE and APP695. Transgenic GULP1 significantly lowered the levels of both Aβ1-40 and Aβ1-42 without decreasing the BACE and APP695 levels. Overexpression of GULP1 also reduced APP/BACE-mediated retinal degeneration, rescued motor dysfunction and extended longevity of the flies. Our results indicate that GULP1 regulate APP processing and reduce neurotoxicity in a Drosophila AD model.
Collapse
|
62
|
Jansson AM, Csiszar A, Maier J, Nyström AC, Ax E, Johansson P, Schiavone LH. The interleukin-like epithelial-mesenchymal transition inducer ILEI exhibits a non-interleukin-like fold and is active as a domain-swapped dimer. J Biol Chem 2017; 292:15501-15511. [PMID: 28751379 DOI: 10.1074/jbc.m117.782904] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/12/2017] [Indexed: 01/07/2023] Open
Abstract
Production and secretion of pro-metastatic proteins is a feature of many tumor cells. The FAM3C interleukin-like epithelial-to-mesenchymal-transition (EMT) inducer (ILEI) has been shown to be strongly up-regulated in several cancers and to be essential for tumor formation and metastasis in epithelial cells, correlating with a significant decrease in overall survival in colon and breast cancer patients. ILEI has been seen to interact with the γ-secretase presenilin 1 subunit (PS1). However, not much is known about the mechanism-of-action or the detailed ILEI structure. We present here the crystal structures of FAM3C ILEI and show that it exists as monomers but also as covalent dimers. The observed ILEI β-β-α fold confirmed previous indications that the FAM3C proteins do not form classical four-helix-bundle structures as was initially predicted. This provides the first experimental evidence that the interleukin-like EMT inducers are not evolutionarily related to the interleukins. However, more surprisingly, the ILEI dimer structure was found to feature a trans-linked domain swap, converting an intramolecular disulfide to intermolecular. Interestingly, dimeric but not monomeric ILEI was subsequently found to cause a dose-dependent increase in EpRas cell invasiveness comparable with TGF-β, indicating that the dimer might be the active ILEI species. This is in line with a parallel study showing that covalent oligomerization of ILEI is essential for EMT and tumor progression in vivo The structures and the activity data give some first insight into the relationship between dimerization and ILEI function as well as indicate an intriguing link between ILEI, the PS1-protease, TGF-β, and the TGF-β receptor 1.
Collapse
Affiliation(s)
- Anna M Jansson
- From the Reagents and Assay Development Division, Discovery Sciences Department
| | - Agnes Csiszar
- the Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Joachim Maier
- From the Reagents and Assay Development Division, Discovery Sciences Department
| | - Ann-Christin Nyström
- Translational Sciences Division, Cardiovascular and Metabolic Diseases Department, and
| | - Elisabeth Ax
- From the Reagents and Assay Development Division, Discovery Sciences Department
| | - Patrik Johansson
- Structure and Biophysics Division, Discovery Sciences Department, AstraZeneca, Pepparedsleden 1, 431 83 Mölndal, Sweden and
| | | |
Collapse
|
63
|
Romano GL, Platania CBM, Drago F, Salomone S, Ragusa M, Barbagallo C, Di Pietro C, Purrello M, Reibaldi M, Avitabile T, Longo A, Bucolo C. Retinal and Circulating miRNAs in Age-Related Macular Degeneration: An In vivo Animal and Human Study. Front Pharmacol 2017; 8:168. [PMID: 28424619 PMCID: PMC5371655 DOI: 10.3389/fphar.2017.00168] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 03/14/2017] [Indexed: 01/05/2023] Open
Abstract
Age related macular degeneration (AMD) is the leading cause of blindness among people aged 50 and over. Retinal deposition of amyloid-β (Aβ) aggregates in AMD patients has suggested a potential link between AMD and Alzheimer's disease (AD). We have evaluated the differential retinal expression profile of miRNAs in a rat model of AMD elicited by Aβ. A serum profile of miRNAs in AMD patients has been also assessed using single TaqMan assay. Analysis of retina from rats intravitreally injected with Aβ revealed that miR-27a, miR-146a, and miR-155 were up-regulated in comparison to control rats. Seven miRNA (miR-9, miR-23a, miR-27a, miR-34a, miR-126, miR-146a, and miR-155) have been found to be dysregulated in serum of AMD patients in comparison to control group. Analysis of pathways has revealed that dysregulated miRNAs, both in the AMD animal model and in AMD patients, can target genes regulating pathways linked to neurodegeneration and inflammation, reinforcing the hypothesis that AMD is a protein misfolding disease similar to AD. In fact, miR-9, miR-23a, miR-27a, miR-34a, miR-146a, miR-155 have been found to be dysregulated both in AMD and AD. In conclusion, we suggest that miR-9, miR-23a, miR-27a, miR-34a, miR-146a, miR-155 represent potential biomarkers and new pharmacological targets for AMD.
Collapse
Affiliation(s)
- Giovanni L Romano
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of CataniaCatania, Italy
| | - Chiara B M Platania
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of CataniaCatania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of CataniaCatania, Italy
| | - Salvatore Salomone
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of CataniaCatania, Italy
| | - Marco Ragusa
- BioMolecular, Genome and Complex Systems BioMedicine Unit, Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of CataniaCatania, Italy
| | - Cristina Barbagallo
- BioMolecular, Genome and Complex Systems BioMedicine Unit, Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of CataniaCatania, Italy
| | - Cinzia Di Pietro
- BioMolecular, Genome and Complex Systems BioMedicine Unit, Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of CataniaCatania, Italy
| | - Michele Purrello
- BioMolecular, Genome and Complex Systems BioMedicine Unit, Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of CataniaCatania, Italy
| | - Michele Reibaldi
- Department of Ophthalmology, School of Medicine, University of CataniaCatania, Italy
| | - Teresio Avitabile
- Department of Ophthalmology, School of Medicine, University of CataniaCatania, Italy
| | - Antonio Longo
- Department of Ophthalmology, School of Medicine, University of CataniaCatania, Italy
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of CataniaCatania, Italy
| |
Collapse
|
64
|
Caraci F, Tascedda F, Merlo S, Benatti C, Spampinato SF, Munafò A, Leggio GM, Nicoletti F, Brunello N, Drago F, Sortino MA, Copani A. Fluoxetine Prevents Aβ 1-42-Induced Toxicity via a Paracrine Signaling Mediated by Transforming-Growth-Factor-β1. Front Pharmacol 2016; 7:389. [PMID: 27826242 PMCID: PMC5078904 DOI: 10.3389/fphar.2016.00389] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/05/2016] [Indexed: 01/01/2023] Open
Abstract
Selective reuptake inhibitors (SSRIs), such as fluoxetine and sertraline, increase circulating Transforming-Growth-Factor-β1 (TGF-β1) levels in depressed patients, and are currently studied for their neuroprotective properties in Alzheimer’s disease. TGF-β1 is an anti-inflammatory cytokine that exerts neuroprotective effects against β-amyloid (Aβ)-induced neurodegeneration. In the present work, the SSRI, fluoxetine, was tested for the ability to protect cortical neurons against 1 μM oligomeric Aβ1-42-induced toxicity. At therapeutic concentrations (100 nM–1 μM), fluoxetine significantly prevented Aβ-induced toxicity in mixed glia-neuronal cultures, but not in pure neuronal cultures. Though to a lesser extent, also sertraline was neuroprotective in mixed cultures, whereas serotonin (10 nM–10 μM) did not mimick fluoxetine effects. Glia-conditioned medium collected from astrocytes challenged with fluoxetine protected pure cortical neurons against Aβ toxicity. The effect was lost in the presence of a neutralizing antibody against TGF-β1 in the conditioned medium, or when the specific inhibitor of type-1 TGF-β1 receptor, SB431542, was added to pure neuronal cultures. Accordingly, a 24 h treatment of cortical astrocytes with fluoxetine promoted the release of active TGF-β1 in the culture media through the conversion of latent TGF-β1 to mature TGF-β1. Unlike fluoxetine, both serotonin and sertraline did not stimulate the astrocyte release of active TGF-β1. We conclude that fluoxetine is neuroprotective against Aβ toxicity via a paracrine signaling mediated by TGF-β1, which does not result from a simplistic SERT blockade.
Collapse
Affiliation(s)
- Filippo Caraci
- Department of Drug Sciences, University of CataniaCatania, Italy; Istituto di Ricovero e Cura a Carattere Scientifico Oasi Maria SantissimaTroina, Italy
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia Modena, Italy
| | - Sara Merlo
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Cristina Benatti
- Department of Life Sciences, University of Modena and Reggio Emilia Modena, Italy
| | - Simona F Spampinato
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Antonio Munafò
- Department of Drug Sciences, University of Catania Catania, Italy
| | - Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Ferdinando Nicoletti
- Istituto di Ricovero e Cura a Carattere Scientifico NeuromedPozzilli, Italy; Department of Physiology and Pharmacology, University of Rome SapienzaRome, Italy
| | - Nicoletta Brunello
- Istituto di Ricovero e Cura a Carattere Scientifico Oasi Maria Santissima Troina, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Agata Copani
- Department of Drug Sciences, University of CataniaCatania, Italy; Institute of Biostructure and Bioimaging, National Research CouncilCatania, Italy
| |
Collapse
|
65
|
Inflammatory Cytokines and Alzheimer's Disease: A Review from the Perspective of Genetic Polymorphisms. Neurosci Bull 2016; 32:469-80. [PMID: 27568024 DOI: 10.1007/s12264-016-0055-4] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/03/2016] [Indexed: 12/30/2022] Open
Abstract
Neuroinflammatory processes are a central feature of Alzheimer's disease (AD) in which microglia are over-activated, resulting in the increased production of pro-inflammatory cytokines. Moreover, deficiencies in the anti-inflammatory system may also contribute to neuroinflammation. Recently, advanced methods for the analysis of genetic polymorphisms have further supported the relationship between neuroinflammatory factors and AD risk because a series of polymorphisms in inflammation-related genes have been shown to be associated with AD. In this review, we summarize the polymorphisms of both pro- and anti-inflammatory cytokines related to AD, primarily interleukin-1 (IL-1), IL-6, tumor necrosis factor alpha, IL-4, IL-10, and transforming growth factor beta, as well as their functional activity in AD pathology. Exploration of the relationship between inflammatory cytokine polymorphisms and AD risk may facilitate our understanding of AD pathogenesis and contribute to improved treatment strategies.
Collapse
|
66
|
TGF-β1 prevents rat retinal insult induced by amyloid-β (1-42) oligomers. Eur J Pharmacol 2016; 787:72-7. [PMID: 26845696 DOI: 10.1016/j.ejphar.2016.02.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/25/2016] [Accepted: 02/01/2016] [Indexed: 11/21/2022]
Abstract
To set up a retinal degenerative model in rat that mimics pathologic conditions such as age-related macular degeneration (AMD) using amyloid-β (Aβ) oligomers, and assess the effect of TGF-β1. Sprague-Dawley male rats were used. Human Aβ1-42 oligomers were intravitreally (ITV) injected (10µM) in the presence or in the absence of recombinant human TGF-β1 (1ng/μl ITV injected). After 48h, the animals were sacrificed and the eyes removed and dissected. The apoptotic markers Bax and Bcl-2 were assessed by western blot analysis in retina lysates. Gene-pathway network analysis was carried out in order to identify pathways involved in AMD. Treatment with Aβ oligomers induced a strong increase in Bax protein level (about 4-fold; p<0.01) and a significant reduction in Bcl-2 protein level (about 2-fold; p<0.05). Co-injection of TGF-β1 triggered a significant reduction of Bax protein induced by Aβ oligomers. Bioinformatic analysis revealed that Bcl-2 and PI3K-Akt are the most connected nodes, for genes and pathways respectively, in the enriched gene-pathway network common to AMD and Alzheimer disease (AD). Overall, these data indicate that ITV injection of Aβ1-42 oligomers in rat induces molecular changes associated with apoptosis in rat retina, highlighting a potential pathogenetic role of Aβ oligomers in AMD. Bioinformatics analysis confirms that apoptosis pathways can take part in AMD. Furthermore, these findings suggest that human recombinant TGF-β1 can prevent retinal damage elicited by Aβ oligomers.
Collapse
|
67
|
Anti-Inflammatory Effects of Ginsenoside-Rh2 Inhibits LPS-Induced Activation of Microglia and Overproduction of Inflammatory Mediators Via Modulation of TGF-β1/Smad Pathway. Neurochem Res 2016; 41:951-7. [PMID: 26738987 DOI: 10.1007/s11064-015-1804-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 12/13/2022]
Abstract
Microglia activation plays an important role in neuroinflammation and contributes to several neurological disorders. Hence, inhibition of both microglia activation and pro-inflammatory cytokines may lead to the effective treatment of neurodegenerative diseases. In this study, we found that GRh2 inhibited the inflammatory response to lipopolysaccharide (LPS) and prevented the LPS-induced neurotoxicity in microglia cells. GRh2 significantly decreased the generation of nitric oxide production, and tumor necrosis factor-α, interleukin (IL)-6, IL-1β, cyclooxygenase-2 and inducible nitric oxide synthase in LPS-induced activated microglia cells. Furthermore, GRh2 (20 and 50 μM) significantly increased TGF-β1 expression and reduced the expression of Smad. These results suggest that GRh2 effectively inhibits microglia activation and production of pro-inflammatory cytokines via modulating the TGF-β1/Smad pathway.
Collapse
|
68
|
Li Q, Cheng Q, Chen Z, Peng R, Chen R, Ma Z, Wan X, Liu J, Meng M, Peng Z, Jiang B. MicroRNA-663 inhibits the proliferation, migration and invasion of glioblastoma cells via targeting TGF-β1. Oncol Rep 2015; 35:1125-34. [PMID: 26717894 DOI: 10.3892/or.2015.4432] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/23/2015] [Indexed: 11/06/2022] Open
Abstract
Cell migration and invasion are key processes involved during tumor metastasis. Recently, microRNAs (miRs) have been demonstrated to play important roles in the regulation of cancer metastasis. However, the underlying mechanisms remain unknown. Here, we aimed to investigate the exact role of miR-663 in the metastasis of glioblastoma as well as the underlying mechanisms. By performing quantitative reverse transcription-polymerase chain reaction (RT-PCR) analysis, we demonstrated that miR-663 was significantly downregulated in glioblastoma tissues (n=25), when compared to that in normal brain tissues (n=15). In addition, its expression levels were also reduced in human glioblastoma cell lines, A172 and U87. Furthermore, restoration of miR-663 expression led to a significant decrease in the cell proliferation, migration and invasion of human glioblastoma A172 and U87 cells. We further identified TGF-β1 as a direct target of miR-663, and found that the expression of TGF-β1 was negatively mediated by miR-663 at the post-transcriptional level in glioblastoma cells. Moreover, overexpression of TGF-β1 significantly reversed the inhibitory effects of miR-663 upregulation on the proliferation, migration and invasion in A172 and U87 cells. In addition, our data suggest that MMP2 and E-cadherin, a key factor in epithelial-mesenchymal transition (EMT), are involved in the miR-633/TGF-β1-mediated metastasis of glioblastoma. In summary, miR-663 plays an inhibitory role in the regulation of proliferation, migration and invasion of glioblastoma cells, partly at least, via direct mediation of TGF-β1 as well as downstream MMP2 and E-cadherin. Therefore, we suggest that miR-663 is a potential candidate for the prevention of glioblastoma metastasis.
Collapse
Affiliation(s)
- Qizhuang Li
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Zigui Chen
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Renjun Peng
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Rui Chen
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhiming Ma
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Xin Wan
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Jincan Liu
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Ming Meng
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhigang Peng
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| | - Bing Jiang
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
69
|
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: 114] [Impact Index Per Article: 11.4] [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
|
70
|
A key role for TGF-β1 in hippocampal synaptic plasticity and memory. Sci Rep 2015; 5:11252. [PMID: 26059637 PMCID: PMC4462026 DOI: 10.1038/srep11252] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 05/08/2015] [Indexed: 01/06/2023] Open
Abstract
Transforming Growth Factor β1 (TGF-β1) is a well-known neuroprotective and neurotrophic factor demonstrated to play a role in synaptic transmission. However, its involvement in physiological mechanisms underlying synaptic plasticity and memory at hippocampal level has not been thoroughly investigated. Here, we examine the role of TGF-β1 in hippocampal long-term potentiation (LTP) and memory in adult wild type mice. Our data provide evidence that administration of exogenous TGF-β1 is able to convert early-phase-LTP into late-phase-LTP. Furthermore, we show that the block of the endogenous TGF-β1 signaling pathway by the specific TGF-β1 inhibitor SB431542, impairs LTP and object recognition memory. The latter impairment was rescued by administration of exogenous TGF-β1, suggesting that endogenously produced TGF-β1 plays a role in physiological mechanisms underlying LTP and memory. Finally, TGF-β1 functional effect correlates with an increased expression of the phosphorylated transcription factor cAMP-Responsive Element Binding protein.
Collapse
|
71
|
Chen JH, Ke KF, Lu JH, Qiu YH, Peng YP. Protection of TGF-β1 against neuroinflammation and neurodegeneration in Aβ1-42-induced Alzheimer's disease model rats. PLoS One 2015; 10:e0116549. [PMID: 25658940 PMCID: PMC4319949 DOI: 10.1371/journal.pone.0116549] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/08/2014] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation has been reported to be associated with Alzheimer’s disease (AD) pathogenesis. Neuroinflammation is generally considered as an outcome of glial activation; however, we recently demonstrated that T helper (Th)17 cells, a subpopulation of proinflammatory CD4+ T cells, are also involved in AD pathogenesis. Transforming growth factor (TGF)-β1, a cytokine that can be expressed in the brain, can be immunosuppressive, but its effects on lymphocyte-mediated neuroinflammation in AD pathogenesis have not been well addressed. In the current study we administered TGF-β1 via intracerebroventricle (ICV) and intranasal (IN) routes in AD model rats to investigate its antiinflammatory and neuroprotective effects. The AD rat model was prepared by bilateral hippocampal injection of amyloid-β (Aβ)1–42. TGF-β1 was administered via ICV one hour prior to Aβ1–42 injection or via both nares seven days after Aβ1–42 injection. ICV administration of TGF-β1 before Aβ1–42 injection remarkably ameliorated Aβ1–42-induced neurodegeneration and prevented Aβ1–42-induced increases in glia-derived proinflammatory mediators (TNF-α, IL-1β and iNOS), as well as T cell-derived proinflammatory cytokines (IFN-γ, IL-2, IL-17 and IL-22), in the hypothalamus, serum or cerebrospinal fluid (CSF) in a concentration-dependent manner. TGF-β1 pretreatment also prevented Aβ1–42-induced decreases in the neurotrophic factors, IGF-1, GDNF and BDNF, and in the antiinflammatory cytokine, IL-10. Similarly, IN administration of TGF-β1 after Aβ1–42 injection reduced neurodegeneration, elevation of proinflammatory mediators and cytokines, and reduction of neurotrophic and antiinflammatory factors, in the hypothalamus, serum or CSF. These findings suggest that TGF-β1 suppresses glial and T cell-mediated neuroinflammation and thereby alleviates AD-related neurodegeneration. The effectiveness of IN administered TGF-β1 in reducing Aβ1–42 neurotoxicity suggests a possible therapeutic approach in patients with AD.
Collapse
Affiliation(s)
- Jia-Hui Chen
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Neurology, Affiliated Hospital, Nantong University, Nantong, China
| | - Kai-Fu Ke
- Department of Neurology, Affiliated Hospital, Nantong University, Nantong, China
| | - Jian-Hua Lu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- * E-mail: (YHQ); (YPP)
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- * E-mail: (YHQ); (YPP)
| |
Collapse
|
72
|
Battaglia G, Riozzi B, Bucci D, Di Menna L, Molinaro G, Pallottino S, Nicoletti F, Bruno V. Activation of mGlu3 metabotropic glutamate receptors enhances GDNF and GLT-1 formation in the spinal cord and rescues motor neurons in the SOD-1 mouse model of amyotrophic lateral sclerosis. Neurobiol Dis 2015; 74:126-36. [DOI: 10.1016/j.nbd.2014.11.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/29/2014] [Accepted: 11/19/2014] [Indexed: 11/28/2022] Open
|
73
|
Prakash A, Kalra J, Mani V, Ramasamy K, Majeed ABA. Pharmacological approaches for Alzheimer’s disease: neurotransmitter as drug targets. Expert Rev Neurother 2014; 15:53-71. [DOI: 10.1586/14737175.2015.988709] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
74
|
Celik M, Kose A, Kose D, Karakus E, Akpinar E, Calik M, Dostbil A, Calikoglu C, Aksoy M, Ozel L. The double-edged sword: effects of pregabalin on experimentally induced sciatic nerve transection and crush injury in rats. Int J Neurosci 2014; 125:845-54. [PMID: 25340254 DOI: 10.3109/00207454.2014.978976] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM The aim of this study was to research the effects of pregabalin on experimentally induced peripheral nerve crush injuries in rats. MATERIAL AND METHOD Forty-two adult female Wistar albino rats were divided into seven groups: 1st group: healthy; 2nd group: axonotmesis control; 3rd group: anastomosis control; 4th group: axonotmesis+30 mg/kg of pregabalin; 5th group: axonotmesis+60 mg/kg of pregabalin; 6th group: anastomosis+30 mg/kg of pregabalin; 7th group: anastomosis+60 mg/kg of pregabalin. Evaluation of the sciatic functional index (SFI) was performed one day before and on days 7, 14, 21, and 28 following surgery. The right sciatic nerves of all animals were examined histopathologically and molecularly. RESULTS After 28 days post-injury, the histopathological regeneration in peripheral nerve injuries for pregabalin 30 mg/kg treated groups was significantly better than that of the control groups. Also the SFI increases and TGF-β gene expression up-regulation were significantly better in pregabalin 30 mg/kg treated groups. CONCLUSION The histopathological, functional and molecular data suggest that pregabalin 30 mg/kg treatment in axonotmesis and anostomosis groups improves nerve regeneration and increases SFI in peripheral nerve injuries by activating antiinflammatory cytokine TGF-β1.
Collapse
Affiliation(s)
- Mine Celik
- a Ataturk University, Faculty of Medicine, Department of Anesthesiology and Reanimation , Erzurum , Turkey
| | - Ahmet Kose
- b Department of Orthophedics and Traumatology, Horasan Public Hospital , Erzurum , Turkey
| | - Duygu Kose
- c Ataturk University, Faculty of Medicine, Department of Pharmacology , Erzurum , Turkey
| | - Emre Karakus
- d Ataturk University, Faculty of Veterinary, Department of Pharmacology and Toxicology , Erzurum , Turkey
| | - Erol Akpinar
- c Ataturk University, Faculty of Medicine, Department of Pharmacology , Erzurum , Turkey
| | - Muhammed Calik
- e Ataturk University, Faculty of Medicine, Department of Pathology , Erzurum , Turkey
| | - Aysenur Dostbil
- a Ataturk University, Faculty of Medicine, Department of Anesthesiology and Reanimation , Erzurum , Turkey
| | - Cagatay Calikoglu
- f Ataturk University, Faculty of Medicine, Department of Brain Surgery , Erzurum , Turkey
| | - Mehmet Aksoy
- a Ataturk University, Faculty of Medicine, Department of Anesthesiology and Reanimation , Erzurum , Turkey
| | - Lutfu Ozel
- g Ataturk University, Faculty of Medicine, Department of Neurology , Erzurum , Turkey
| |
Collapse
|
75
|
Song L, Gu Y, Jie J, Bai X, Yang Y, Liu C, Liu Q. Dab2 attenuates brain injury in APP/PS1 mice via targeting transforming growth factor-beta/SMAD signaling. Neural Regen Res 2014; 9:41-50. [PMID: 25206742 PMCID: PMC4146324 DOI: 10.4103/1673-5374.125328] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2013] [Indexed: 02/04/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) type II receptor (TβRII) levels are extremely low in the brain tissue of patients with Alzheimer's disease. This receptor inhibits TGF-β1/SMAD signaling and thereby aggravates amyolid-beta deposition and neuronal injury. Dab2, a specific adapter protein, protects TβRII from degradation and ensures the effective conduction of TGF-β1/SMAD signaling. In this study, we used an adenoviral vector to overexpress the Dab2 gene in the mouse hippocampus and investigated the regulatory effect of Dab2 protein on TGF-β1/SMAD signaling in a mouse model of Alzheimer's disease, and the potential neuroprotective effect. The results showed that the TβRII level was lower in APP/PS1 mouse hippocampus than in normal mouse hippocampus. After Dab2 expression, hippocampal TβRII and p-SMAD2/3 levels were significantly increased, while amyloid-beta deposition, microglia activation, tumor necrosis factor-α and interleulin-6 levels and neuronal loss were significantly attenuated in APP/PS1 mouse brain tissue. These results suggest that Dab2 can exhibit neuroprotective effects in Alzheimer's disease by regulating TGF-β1/SMAD signaling.
Collapse
Affiliation(s)
- Lei Song
- Department of Neurology, Norman Bethune First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yue Gu
- Department of Respiratory Medicine, Norman Bethune First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jing Jie
- Department of Respiratory Medicine, Norman Bethune First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiaoxue Bai
- Department of Respiratory Medicine, Norman Bethune First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ying Yang
- Department of Respiratory Medicine, Norman Bethune First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Chaoying Liu
- Department of Respiratory Medicine, Norman Bethune First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qun Liu
- Department of Neurology, Norman Bethune First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
76
|
Cong L, Jia J, Qin W, Ren Y, Sun Y. Genome-wide analysis of DNA methylation in an APP/PS1 mouse model of Alzheimer's disease. Acta Neurol Belg 2014; 114:195-206. [PMID: 24347181 DOI: 10.1007/s13760-013-0267-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/29/2013] [Indexed: 12/16/2022]
Abstract
To investigate aberrant genome-wide CpG methylation patterns in cortex brain tissue of APP/PS1 mice and as compared to controls, which allows for identification of novel disease-associated genes. This study investigates the genome-wide DNA methylation profiles of the cortex from APP/PS1 transgenic mice and control mice using the Roche NimbleGen chip platform. Functional analysis was then conducted by Ingenuity Pathways Analysis system. The methylated DNA fragments in the genome of each sample were enriched by MeDIP and the whole-genome interrogations were hybridized to the Roche NimbleGen Human DNA Methylation 3x720 K CpG Island Plus RefSeq Promoter Array that cover 15,980 CpG islands and 20,404 reference gene promoter regions of the entire human genome. Analysis reveals 2346 CpG sites representing 485 unique genes as potentially associated with AD disease status pending confirmation in additional study. At the same time, these hyper-methylated genes display familial aggregation. An impairment of the transforming growth factor-β1 (TGF-β1) signaling pathway has been demonstrated to be specific to the AD brain and, particularly, to the early phase of the disease, supporting a role for epigenetic change of TGF-β1 in AD pathology. In future research, we will focus on TGF-β1, as it appeared to be the most promising candidate for AD.
Collapse
|
77
|
Chen NF, Huang SY, Chen WF, Chen CH, Lu CH, Chen CL, Yang SN, Wang HM, Wen ZH. TGF-β1 attenuates spinal neuroinflammation and the excitatory amino acid system in rats with neuropathic pain. THE JOURNAL OF PAIN 2014; 14:1671-85. [PMID: 24290447 DOI: 10.1016/j.jpain.2013.08.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/16/2013] [Accepted: 08/28/2013] [Indexed: 01/28/2023]
Abstract
UNLABELLED Previous studies have reported that the intrathecal (i.t.) administration of transforming growth factor β1 (TGF-β1) prevents and reverses neuropathic pain. However, only limited information is available regarding the possible role and effects of spinal TGF-β1 in neuropathic pain. We aimed to investigate the antinociceptive effects of exogenous TGF-β1 on chronic constriction injury (CCI)-induced neuropathic pain in rats. We demonstrated that sciatic nerve injury caused a downregulation of endogenous TGF-β1 levels on the ipsilateral side of the lumbar spinal dorsal gray matter, and that the i.t. administration of TGF-β1 (.01-10 ng) significantly attenuated CCI-induced thermal hyperalgesia in neuropathic rats. TGF-β1 significantly inhibited CCI-induced spinal neuroinflammation, microglial and astrocytic activation, and upregulation of tumor necrosis factor-α. Moreover, i.t. TGF-β1 significantly attenuated the CCI-induced downregulation of glutamate transporter 1, the glutamate aspartate transporter, and the excitatory amino acid carrier 1 on the ipsilateral side. Furthermore, i.t. TGF-β1 significantly decreased the concentrations of 2 excitatory amino acids, aspartate and glutamate, in the spinal dialysates in CCI rats. In summary, we conclude that the mechanisms of the antinociceptive effects of i.t. TGF-β1 in neuropathy may include attenuation of spinal neuroinflammation, attenuation, or upregulation of glutamate transporter downregulation, and a decrease of spinal extracellular excitatory amino acids. PERSPECTIVE Clinically, medical treatment is usually initiated after the onset of intractable pain. Therefore, in the present study, i.t. TGF-β1 was designed to be administered 2 weeks after the establishment of CCI pain. Compared to the continuous TGF-β1 infusion mode, single-dose administration seems more convenient and practical to use.
Collapse
Affiliation(s)
- Nan-Fu Chen
- Department of Marine Biotechnology and Resources, Asia-Pacific Ocean Research Center, National Sun Yat-sen University, Kaohsiung, Taiwan; Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Kandasamy M, Lehner B, Kraus S, Sander PR, Marschallinger J, Rivera FJ, Trümbach D, Ueberham U, Reitsamer HA, Strauss O, Bogdahn U, Couillard-Despres S, Aigner L. TGF-beta signalling in the adult neurogenic niche promotes stem cell quiescence as well as generation of new neurons. J Cell Mol Med 2014; 18:1444-1459. [PMID: 24779367 PMCID: PMC4124027 DOI: 10.1111/jcmm.12298] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 03/10/2014] [Indexed: 12/16/2022] Open
Abstract
Members of the transforming growth factor (TGF)-β family govern a wide range of mechanisms in brain development and in the adult, in particular neuronal/glial differentiation and survival, but also cell cycle regulation and neural stem cell maintenance. This clearly created some discrepancies in the field with some studies favouring neuronal differentiation/survival of progenitors and others favouring cell cycle exit and neural stem cell quiescence/maintenance. Here, we provide a unifying hypothesis claiming that through its regulation of neural progenitor cell (NPC) proliferation, TGF-β signalling might be responsible for (i) maintaining stem cells in a quiescent stage, and (ii) promoting survival of newly generated neurons and their functional differentiation. Therefore, we performed a detailed histological analysis of TGF-β1 signalling in the hippocampal neural stem cell niche of a transgenic mouse that was previously generated to express TGF-β1 under a tetracycline regulatable Ca-Calmodulin kinase promoter. We also analysed NPC proliferation, quiescence, neuronal survival and differentiation in relation to elevated levels of TGF-β1 in vitro and in vivo conditions. Finally, we performed a gene expression profiling to identify the targets of TGF-β1 signalling in adult NPCs. The results demonstrate that TGF-β1 promotes stem cell quiescence on one side, but also neuronal survival on the other side. Thus, considering the elevated levels of TGF-β1 in ageing and neurodegenerative diseases, TGF-β1 signalling presents a molecular target for future interventions in such conditions.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
| | - Bernadette Lehner
- Department of Neurology, University Hospital RegensburgRegensburg, Germany
| | - Sabrina Kraus
- Department of Experimental Ophthalmology, University of RegensburgRegensburg, Germany
| | - Paul Ramm Sander
- Department of Neurology, University Hospital RegensburgRegensburg, Germany
- Institute of Biophysics and Physical Biochemistry, University of RegensburgRegensburg, Germany
| | - Julia Marschallinger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
| | - Francisco J Rivera
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health (GmbH), Technical University MunichNeuherberg, Germany
| | - Uwe Ueberham
- Paul Flechsig Institute for Brain Research, Department of Neuroanatomy, University of LeipzigLeipzig, Germany
| | - Herbert A Reitsamer
- Department of Ophthalmology, SALK, Paracelsus Medical UniversitySalzburg, Austria
| | - Olaf Strauss
- Department of Experimental Ophthalmology, University of RegensburgRegensburg, Germany
| | - Ulrich Bogdahn
- Department of Neurology, University Hospital RegensburgRegensburg, Germany
| | - Sebastien Couillard-Despres
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical UniversitySalzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
| |
Collapse
|
79
|
Flexibilide obtained from cultured soft coral has anti-neuroinflammatory and analgesic effects through the upregulation of spinal transforming growth factor-β1 in neuropathic rats. Mar Drugs 2014; 12:3792-817. [PMID: 24979268 PMCID: PMC4113799 DOI: 10.3390/md12073792] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 05/28/2014] [Accepted: 05/29/2014] [Indexed: 12/27/2022] Open
Abstract
Chronic neuroinflammation plays an important role in the development and maintenance of neuropathic pain. The compound flexibilide, which can be obtained from cultured soft coral, possesses anti-inflammatory and analgesic effects in the rat carrageenan peripheral inflammation model. In the present study, we investigated the antinociceptive properties of flexibilide in the rat chronic constriction injury (CCI) model of neuropathic pain. First, we found that a single intrathecal (i.t.) administration of flexibilide significantly attenuated CCI-induced thermal hyperalgesia at 14 days after surgery. Second, i.t. administration of 10-μg flexibilide twice daily was able to prevent the development of thermal hyperalgesia and weight-bearing deficits in CCI rats. Third, i.t. flexibilide significantly inhibited CCI-induced activation of microglia and astrocytes, as well as the upregulated proinflammatory enzyme, inducible nitric oxide synthase, in the ipsilateral spinal dorsal horn. Furthermore, flexibilide attenuated the CCI-induced downregulation of spinal transforming growth factor-β1 (TGF-β1) at 14 days after surgery. Finally, i.t. SB431542, a selective inhibitor of TGF-β type I receptor, blocked the analgesic effects of flexibilide in CCI rats. Our results suggest that flexibilide may serve as a therapeutic agent for neuropathic pain. In addition, spinal TGF-β1 may be involved in the anti-neuroinflammatory and analgesic effects of flexibilide.
Collapse
|
80
|
The FAM3 superfamily member ILEI ameliorates Alzheimer's disease-like pathology by destabilizing the penultimate amyloid-β precursor. Nat Commun 2014; 5:3917. [PMID: 24894631 DOI: 10.1038/ncomms4917] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/16/2014] [Indexed: 01/12/2023] Open
Abstract
Accumulation of amyloid-β peptide (Aβ) in the brain underlies the pathogenesis of Alzheimer's disease (AD). Aβ is produced by β- and γ-secretase-mediated sequential proteolysis of amyloid-β precursor protein (APP). Here we identify a secretory protein named interleukin-like epithelial-mesenchymal transition inducer (ILEI, also known as FAM3 superfamily member C) as a negative regulator of Aβ production. ILEI destabilizes the β-secretase-cleaved APP carboxy-terminal fragment, the penultimate precursor of Aβ, by binding to the γ-secretase complex and interfering with its chaperone properties. Notch signalling and γ-secretase activity are not affected by ILEI. We also show neuronal expression of ILEI and its induction by transforming growth factor-β signalling. The level of secreted ILEI is markedly decreased in the brains of AD patients. Transgenic (Tg) overexpression of ILEI significantly reduces the brain Aβ burden and ameliorates the memory deficit in AD model mice. ILEI may be a plausible target for the development of disease-modifying therapies.
Collapse
|
81
|
Grosso G, Pajak A, Marventano S, Castellano S, Galvano F, Bucolo C, Drago F, Caraci F. Role of omega-3 fatty acids in the treatment of depressive disorders: a comprehensive meta-analysis of randomized clinical trials. PLoS One 2014; 9:e96905. [PMID: 24805797 PMCID: PMC4013121 DOI: 10.1371/journal.pone.0096905] [Citation(s) in RCA: 306] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/11/2014] [Indexed: 12/14/2022] Open
Abstract
Background Despite omega-3 polyunsaturated fatty acids (PUFA) supplementation in depressed patients have been suggested to improve depressive symptomatology, previous findings are not univocal. Objectives To conduct an updated meta-analysis of randomized controlled trials (RCTs) of omega-3 PUFA treatment of depressive disorders, taking into account the clinical differences among patients included in the studies. Methods A search on MEDLINE, EMBASE, PsycInfo, and the Cochrane Database of RCTs using omega-3 PUFA on patients with depressive symptoms published up to August 2013 was performed. Standardized mean difference in clinical measure of depression severity was primary outcome. Type of omega-3 used (particularly eicosapentaenoic acid [EPA] and docosahexaenoic acid [DHA]) and omega-3 as mono- or adjuvant therapy was also examined. Meta-regression analyses assessed the effects of study size, baseline depression severity, trial duration, dose of omega-3, and age of patients. Results Meta-analysis of 11 and 8 trials conducted respectively on patients with a DSM-defined diagnosis of major depressive disorder (MDD) and patients with depressive symptomatology but no diagnosis of MDD demonstrated significant clinical benefit of omega-3 PUFA treatment compared to placebo (standardized difference in random-effects model 0.56 SD [95% CI: 0.20, 0.92] and 0.22 SD [95% CI: 0.01, 0.43], respectively; pooled analysis was 0.38 SD [95% CI: 0.18, 0.59]). Use of mainly EPA within the preparation, rather than DHA, influenced final clinical efficacy. Significant clinical efficacy had the use of omega-3 PUFA as adjuvant rather than mono-therapy. No relation between efficacy and study size, baseline depression severity, trial duration, age of patients, and study quality was found. Omega-3 PUFA resulted effective in RCTs on patients with bipolar disorder, whereas no evidence was found for those exploring their efficacy on depressive symptoms in young populations, perinatal depression, primary disease other than depression and healthy subjects. Conclusions The use of omega-3 PUFA is effective in patients with diagnosis of MDD and on depressive patients without diagnosis of MDD.
Collapse
Affiliation(s)
- Giuseppe Grosso
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry, University of Catania, Catania, Italy
- * E-mail:
| | - Andrzej Pajak
- Department of Epidemiology and Population Studies, Jagiellonian University Medical College, Krakow, Poland
| | - Stefano Marventano
- Department “G.F. Ingrassia”, Section of Hygiene and Public Health, University of Catania, Catania, Italy
| | - Sabrina Castellano
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry, University of Catania, Catania, Italy
| | - Fabio Galvano
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry, University of Catania, Catania, Italy
| | - Claudio Bucolo
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry, University of Catania, Catania, Italy
| | - Filippo Drago
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry, University of Catania, Catania, Italy
| | - Filippo Caraci
- Department of Educational Sciences, University of Catania, Catania, Italy
- IRCCS Associazione Oasi Maria S.S. – Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy
| |
Collapse
|
82
|
Durand D, Carniglia L, Beauquis J, Caruso C, Saravia F, Lasaga M. Astroglial mGlu3 receptors promote alpha-secretase-mediated amyloid precursor protein cleavage. Neuropharmacology 2014; 79:180-9. [DOI: 10.1016/j.neuropharm.2013.11.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/26/2013] [Accepted: 11/20/2013] [Indexed: 12/21/2022]
|
83
|
Richens JL, Morgan K, O'Shea P. Reverse engineering of Alzheimer's disease based on biomarker pathways analysis. Neurobiol Aging 2014; 35:2029-38. [PMID: 24684789 DOI: 10.1016/j.neurobiolaging.2014.02.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 02/18/2014] [Accepted: 02/26/2014] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) poses an increasingly profound problem to society, yet progress toward a genuine understanding of the disease remains worryingly slow. Perhaps, the most outstanding problem with the biology of AD is the question of its mechanistic origins, that is, it remains unclear wherein the molecular failures occur that underlie the disease. We demonstrate how molecular biomarkers could help define the nature of AD in terms of the early biochemical events that correlate with disease progression. We use a novel panel of biomolecules that appears in cerebrospinal fluid of AD patients. As changes in the relative abundance of these molecular markers are associated with progression to AD from mild cognitive impairment, we make the assumption that by tracking their origins we can identify the biochemical conditions that predispose their presence and consequently cause the onset of AD. We couple these protein markers with an analysis of a series of genetic factors and together this hypothesis essentially allows us to redefine AD in terms of the molecular pathways that underlie the disease.
Collapse
Affiliation(s)
- Joanna L Richens
- Cell Biophysics Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University Park, University of Nottingham, Nottingham, UK
| | - Kevin Morgan
- Humans Genetics Research Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Paul O'Shea
- Cell Biophysics Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University Park, University of Nottingham, Nottingham, UK.
| |
Collapse
|
84
|
Dambach H, Hinkerohe D, Prochnow N, Stienen MN, Moinfar Z, Haase CG, Hufnagel A, Faustmann PM. Glia and epilepsy: Experimental investigation of antiepileptic drugs in an astroglia/microglia co-culture model of inflammation. Epilepsia 2013; 55:184-92. [DOI: 10.1111/epi.12473] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2013] [Indexed: 01/22/2023]
Affiliation(s)
- Hannes Dambach
- Department of Neuroanatomy and Molecular Brain Research; Ruhr University Bochum; Bochum Germany
| | - Daniel Hinkerohe
- Department of Neurology; Ruhr-University Bochum; Knappschafts Hospital; Bochum Germany
| | - Nora Prochnow
- Department of Neuroanatomy and Molecular Brain Research; Ruhr University Bochum; Bochum Germany
- International Graduate School of Neuroscience (IGSN); Ruhr University Bochum; Bochum Germany
| | - Martin N. Stienen
- Department of Neuroanatomy and Molecular Brain Research; Ruhr University Bochum; Bochum Germany
| | - Zahra Moinfar
- Department of Neuroanatomy and Molecular Brain Research; Ruhr University Bochum; Bochum Germany
- International Graduate School of Neuroscience (IGSN); Ruhr University Bochum; Bochum Germany
| | - Claus G. Haase
- Department of Neurology; Holy Spirit Hospital; Köln Germany
| | | | - Pedro M. Faustmann
- Department of Neuroanatomy and Molecular Brain Research; Ruhr University Bochum; Bochum Germany
- International Graduate School of Neuroscience (IGSN); Ruhr University Bochum; Bochum Germany
| |
Collapse
|
85
|
Villapol S, Wang Y, Adams M, Symes AJ. Smad3 deficiency increases cortical and hippocampal neuronal loss following traumatic brain injury. Exp Neurol 2013; 250:353-65. [PMID: 24120438 DOI: 10.1016/j.expneurol.2013.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 09/11/2013] [Accepted: 10/03/2013] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGF-β) signaling is involved in pathological processes following brain injury. TGF-β signaling through Smad3 contributes significantly to the immune response and glial scar formation after brain injury. However, TGF-β is also neuroprotective, suggesting that Smad3 signaling may also be involved in neuroprotection after injury. We found expression of the TGF-β type II receptor (TβRII) and Smad3 protein to be strongly and rapidly induced in neurons in the ipsilateral cortex and CA1 region of the hippocampus after stab wound injury. In contrast, astrocytic expression of TβRII and Smad3 was induced more slowly. Comparison of the response of wild-type and Smad3 null mice to cortical stab wound injury showed a more pronounced loss of neuronal viability in Smad3 null mice. Neuronal density was more strongly reduced in Smad3 null mice than in wild-type mice at 1 and 3days post lesion in both the ipsilateral cortex and hippocampal CA1 region. Fluoro-Jade B, TUNEL staining, and cleaved caspase-3 staining also demonstrated increased neuronal degeneration at early time points after injury in the ipsilateral hemisphere in Smad3 null mice. Taken together, our results suggest that TGF-β cytokine family signaling through Smad3 protects neurons in the damaged cortex and hippocampus at early time points after injury.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | | |
Collapse
|
86
|
Aprahamian I, Stella F, Forlenza OV. New treatment strategies for Alzheimer's disease: is there a hope? Indian J Med Res 2013; 138:449-60. [PMID: 24434253 PMCID: PMC3868059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Indexed: 10/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disease, and corresponds to the most common cause of dementia worldwide. Although not fully understood, the pathophysiology of AD is largely represented by the neurotoxic events triggered by the beta-amyloid cascade and by cytoskeletal abnormalities subsequent to the hyperphosphorylation of microtubule-associated Tau protein in neurons. These processes lead respectively to the formation of neuritic plaques and neurofibrillary tangles, which are the pathological hallmarks of the disease. Clinical benefits of the available pharmacological treatment for AD with antidementia drugs (namely cholinesterase inhibitors and memantine) are unquestionable, although limited to a temporary, symptomatic support to cognitive and related functions. Over the past decade, substantial funding and research have been dedicated to the search and development of new pharmaceutical compounds with disease-modifying properties. The rationale of such approach is that by tackling key pathological processes in AD it may be possible to attenuate or even change its natural history. In the present review, we summarize the available evidence on the new therapeutic approaches that target amyloid and Tau pathology in AD, focusing on pharmaceutical compounds undergoing phase 2 and phase 3 randomized controlled trials.
Collapse
Affiliation(s)
- Ivan Aprahamian
- Laboratory of Neuroscience (LIM 27) Department & Institute of Psychiatry, Faculty of Medicine, University of São Paulo, Brazil
| | - Florindo Stella
- Laboratory of Neuroscience (LIM 27) Department & Institute of Psychiatry, Faculty of Medicine, University of São Paulo, Brazil
- UNESP, Biosciences Institute, Campus of Rio Claro-SP, Brazil
| | - Orestes V. Forlenza
- Laboratory of Neuroscience (LIM 27) Department & Institute of Psychiatry, Faculty of Medicine, University of São Paulo, Brazil
| |
Collapse
|
87
|
Bosco P, Ferri R, Salluzzo MG, Castellano S, Signorelli M, Nicoletti F, Nuovo SD, Drago F, Caraci F. Role of the Transforming-Growth-Factor-β1 Gene in Late-Onset Alzheimer's Disease: Implications for the Treatment. Curr Genomics 2013; 14:147-56. [PMID: 24082824 PMCID: PMC3637679 DOI: 10.2174/1389202911314020007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 01/31/2013] [Accepted: 02/01/2013] [Indexed: 11/22/2022] Open
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common form of dementia in the elderly. LOAD has a complex and largely unknown etiology with strong genetic determinants. Genetics of LOAD is known to involve several genetic risk factors among which the Apolipoprotein E (APOE) gene seems to be the major recognized genetic determinant. Recent efforts have been made to identify other genetic factors involved in the pathophysiology of LOAD such as genes associated with a deficit of neurotrophic factors in the AD brain. Genetic variations of neurotrophic factors, such as brain-derived neurotrophic factor (BDNF), and transforming-growth-factor-β1 (TGF-β1) are known to increase the risk to develop LOAD and have also been related to depression susceptibility in LOAD. Transforming-Growth-Factor-β1 (TGF-β1) is a neurotrophic factor that exerts neuroprotective effects against ß-amyloid-induced neurodegeneration. Recent evidence suggests that a specific impairment in the signaling of TGF-β is an early event in the pathogenesis of AD. TGF-β1 protein levels are predominantly under genetic control, and the TGF-β1 gene, located on chromosome 19q13.1–3, con-tains several single nucleotide polymorphisms (SNPs) upstream and in the transcript region, such as the SNP at codon +10 (T/C) and +25 (G/C), which is known to influence the level of expression of TGF-β1. In the present review, we summarize the current literature on genetic risk factors for LOAD, focusing on the role of the TGF-β1 gene, finally discussing the possible implications of these genetic studies for the selection of patients eligible for neuroprotective strategies in AD.
Collapse
Affiliation(s)
- Paolo Bosco
- IRCCS Associazione Oasi Maria S.S. - Institute for Research on Mental Retardation and Brain Aging, 94018 Troina, Enna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Kraus S, Lehner B, Reichhart N, Couillard-Despres S, Wagner K, Bogdahn U, Aigner L, Strauß O. Transforming growth factor-β1 primes proliferating adult neural progenitor cells to electrophysiological functionality. Glia 2013; 61:1767-83. [DOI: 10.1002/glia.22551] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 06/19/2013] [Accepted: 06/27/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Sabrina Kraus
- Department of Experimental Ophthalmology, Eye Clinic; University Medical Center Regensburg; Regensburg Germany
| | - Bernadette Lehner
- Department of Neurology; University Medical Center Regensburg; Regensburg Germany
| | - Nadine Reichhart
- Department of Experimental Ophthalmology, Eye Clinic; University Medical Center Regensburg; Regensburg Germany
- Department of Experimental Ophthalmology, Ophthalmology; Charite Universitaetsmedizin Berlin; Berlin Germany
| | - Sebastien Couillard-Despres
- Institute of Molecular Regenerative Medicine; Paracelsus Medical University; Salzburg Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg; Paracelsus Medical University; Salzburg Austria
| | - Katrin Wagner
- Institute of Molecular Regenerative Medicine; Paracelsus Medical University; Salzburg Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg; Paracelsus Medical University; Salzburg Austria
| | - Ulrich Bogdahn
- Department of Neurology; University Medical Center Regensburg; Regensburg Germany
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine; Paracelsus Medical University; Salzburg Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg; Paracelsus Medical University; Salzburg Austria
| | - Olaf Strauß
- Department of Experimental Ophthalmology, Eye Clinic; University Medical Center Regensburg; Regensburg Germany
- Department of Experimental Ophthalmology, Ophthalmology; Charite Universitaetsmedizin Berlin; Berlin Germany
| |
Collapse
|
89
|
Huang L, Jia J, Liu R. Decreased serum levels of the angiogenic factors VEGF and TGF-β1 in Alzheimer's disease and amnestic mild cognitive impairment. Neurosci Lett 2013; 550:60-3. [PMID: 23827227 DOI: 10.1016/j.neulet.2013.06.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 06/03/2013] [Accepted: 06/20/2013] [Indexed: 12/30/2022]
Abstract
Growing evidence suggests that angiogenesis might represent a new pathogenic mechanism involved in the progression of Alzheimer's disease (AD). Vascular endothelial growth factor (VEGF) and transforming growth factor-β1 (TGF-β1) are two cytokines having a pivotal role in angiogenesis. In the present study, serum VEGF and TGF-β1 levels were measured with ELISA in 31 AD patients, 28 amnestic mild cognitive impairment (aMCI) patients and 29 controls. VEGF concentration in serum of AD patients was significantly lower than that in aMCI patients and controls (p<0.05). Serum VEGF levels in aMCI patients were also significantly decreased compared to controls (p<0.05). Serum TGF-β1 levels in AD patients were significantly lower than those in controls (p<0.05). There was a negative correlation between serum VEGF/TGF-β1 levels and the Clinical Dementia Rating (CDR) scores (p<0.05) and a positive correlation between serum VEGF levels and TGF-β1 levels (p<0.05). These observations suggest that angiogenesis might be involved in the onset process of AD and the decrease of angiogenic factors might be related to the severity of cognitive impairment.
Collapse
Affiliation(s)
- Li Huang
- Department of Neurology, Xuan Wu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, PR China.
| | | | | |
Collapse
|
90
|
Araki W. Potential repurposing of oncology drugs for the treatment of Alzheimer's disease. BMC Med 2013; 11:82. [PMID: 23531187 PMCID: PMC3655040 DOI: 10.1186/1741-7015-11-82] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 03/26/2013] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative dementia, affecting about 30 million people worldwide. Despite recent advances in understanding its molecular pathology, no mechanism-based drugs are currently available that can halt the progression of AD. Because amyloid-β-peptide (Aβ), a primary component of senile plaques, is thought to be a central pathogenic culprit, several disease-modifying therapies are being developed, including inhibitors of Aβ-producing proteases and immunotherapies with anti-Aβ antibodies. Drug repositioning or repurposing is regarded as a complementary and reasonable approach to identify new drug candidates for AD. This commentary will discuss the clinical relevance of an attractive candidate compound reported in a recent paper by Hayes et al. (BMC Medicine 2013) as well as perspectives regarding the possible repositioning of oncology drugs for the treatment of AD. See related research article here http://www.biomedcentral.com/1741-7015/11/81.
Collapse
Affiliation(s)
- Wataru Araki
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan.
| |
Collapse
|
91
|
Cheng L, Xiang GY, Chen D. Role of TGF-β1 and its receptors in malignant progression of hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2012; 20:3231-3236. [DOI: 10.11569/wcjd.v20.i33.3231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) is a cytokine which exerts a wide range of biological activities by initiating downstream signaling. As transmembrane receptors, TGFBRⅠ, TGFBRⅡ and TGFBRⅢ have been shown to play an important role in mediating TGF-β1 signal transduction. Owing to roles in promoting cell growth and development, alterations in TGF-β1 and its receptors may result in many diseases, especially tumors. Hepatocellular carcinoma (HCC) is a common malignant solid tumor with high risk of metastasis and recurrence and is associated with a high fatality. Recent studies point to a close relationship between hepatocellular carcinoma and the dysregulation of TGF-β1 signaling pathway. The changes in the levels of TGF-β1 and its receptors in tumor microenvironment may facilitate the invasive potential of HCC. In this article we mainly summarize the structure and function of TGF-β1 and its receptors and assess their role in metastasis and recurrence of hepatocellular carcinoma.
Collapse
|
92
|
Centelles JJ. General aspects of colorectal cancer. ISRN ONCOLOGY 2012; 2012:139268. [PMID: 23209942 PMCID: PMC3504424 DOI: 10.5402/2012/139268] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Accepted: 10/11/2012] [Indexed: 12/14/2022]
Abstract
Colorectal cancer (CRC) is one of the main causes of death. Cancer is initiated by several DNA damages, affecting proto-oncogenes, tumour suppressor genes, and DNA repairing genes. The molecular origins of CRC are chromosome instability (CIN), microsatellite instability (MSI), and CpG island methylator phenotype (CIMP). A brief description of types of CRC cancer is presented, including sporadic CRC, hereditary nonpolyposis colorectal cancer (HNPCC) or Lynch syndromes, familiar adenomatous polyposis (FAP), MYH-associated polyposis (MAP), Peutz-Jeghers syndrome (PJS), and juvenile polyposis syndrome (JPS). Some signalling systems for CRC are also described, including Wnt-β-catenin pathway, tyrosine kinase receptors pathway, TGF-β pathway, and Hedgehog pathway. Finally, this paper describes also some CRC treatments.
Collapse
Affiliation(s)
- Josep J. Centelles
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Avenida Diagonal 643, Catalunya, 08028 Barcelona, Spain
| |
Collapse
|
93
|
Pál G, Vincze C, Renner É, Wappler EA, Nagy Z, Lovas G, Dobolyi A. Time course, distribution and cell types of induction of transforming growth factor betas following middle cerebral artery occlusion in the rat brain. PLoS One 2012; 7:e46731. [PMID: 23056426 PMCID: PMC3466286 DOI: 10.1371/journal.pone.0046731] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 09/03/2012] [Indexed: 01/04/2023] Open
Abstract
Transforming growth factor-βs (TGF-β1–3) are cytokines that regulate the proliferation, differentiation, and survival of various cell types. The present study describes the induction of TGF-β1–3 in the rat after focal ischemia at 3 h, 24 h, 72 h and 1 month after transient (1 h) or permanent (24 h) middle cerebral artery occlusion (MCAO) using in situ hybridization histochemistry and quantitative analysis. Double labeling with different markers was used to identify the localization of TGF-β mRNA relative to the penumbra and glial scar, and the types of cells expressing TGF-βs. TGF-β1 expression increased 3 h after MCAO in the penumbra and was further elevated 24 h after MCAO. TGF-β1 was present mostly in microglial cells but also in some astrocytes. By 72 h and 1 month after the occlusion, TGF-β1 mRNA-expressing cells also appeared in microglia within the ischemic core and in the glial scar. In contrast, TGF-β2 mRNA level was increased in neurons but not in astrocytes or microglial cells in layers II, III, and V of the ipsilateral cerebral cortex 24 h after MCAO. TGF-β3 was not induced in cells around the penumbra. Its expression increased in only a few cells in layer II of the cerebral cortex 24 h after MCAO. The levels of TGF-β2 and -β3 decreased at subsequent time points. Permanent MCAO further elevated the levels of all 3 subtypes of TGF-βs suggesting that reperfusion is not a major factor in their induction. TGF-β1 did not co-localize with either Fos or ATF-3, while the co-localization of TGF-β2 with Fos but not with ATF-3 suggests that cortical spreading depolarization, but not damage to neural processes, might be the mechanism of induction for TGF-β2. The results imply that endogenous TGF-βs are induced by different mechanisms following an ischemic attack in the brain suggesting that they are involved in distinct spatially and temporally regulated inflammatory and neuroprotective processes.
Collapse
Affiliation(s)
- Gabriella Pál
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
| | - Csilla Vincze
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
- Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Éva Renner
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
| | - Edina A. Wappler
- Cardiovascular Center, Department Section of Vascular Neurology, Semmelweis University, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Nagy
- Cardiovascular Center, Department Section of Vascular Neurology, Semmelweis University, Budapest, Hungary
| | - Gábor Lovas
- Department of Neurology, Semmelweis University, Budapest, Hungary
- Department of Neurology, Jahn Ferenc Teaching Hospital, Budapest, Hungary
| | - Arpád Dobolyi
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
- * E-mail:
| |
Collapse
|
94
|
Dong S, Duan Y, Hu Y, Zhao Z. Advances in the pathogenesis of Alzheimer's disease: a re-evaluation of amyloid cascade hypothesis. Transl Neurodegener 2012; 1:18. [PMID: 23210692 PMCID: PMC3526416 DOI: 10.1186/2047-9158-1-18] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 09/13/2012] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is a common neurodegenerative disease characterized clinically by progressive deterioration of memory, and pathologically by histopathological changes including extracellular deposits of amyloid-beta (A-beta) peptides forming senile plaques (SP) and the intracellular neurofibrillary tangles (NFT) of hyperphosphorylated tau in the brain. This review focused on the new developments of amyloid cascade hypothesis with details on the production, metabolism and clearance of A-beta, and the key roles of some important A-beta-related genes in the pathological processes of AD. The most recent research advances in genetics, neuropathology and pathogenesis of the disease were also discussed.
Collapse
Affiliation(s)
- Suzhen Dong
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, 3663 Zhongshan Road (N), Shanghai, 200062, China.
| | | | | | | |
Collapse
|
95
|
Li LY, Li JL, Zhang HM, Yang WM, Wang K, Fang Y, Wang Y. TGFβ1 treatment reduces hippocampal damage, spontaneous recurrent seizures, and learning memory deficits in pilocarpine-treated rats. J Mol Neurosci 2012; 50:109-23. [PMID: 22936246 DOI: 10.1007/s12031-012-9879-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 08/15/2012] [Indexed: 11/28/2022]
Abstract
Studies have demonstrated the neuroprotective activity of transforming growth factor beta-1 (TGFβ1), protecting neurons against different kinds of insults. However, the role of exogenous TGFβ1 in the neuronal damage following status epilepticus (SE) and the related spontaneous recurrent seizures (SRS) is unknown. The present study aimed to determine the effect of intranasal TGFβ1 administration on SRS and cognitive function following lithium-pilocarpine-induced SE and associated hippocampal damage. We found that intranasal TGFβ1 significantly attenuated the hippocampal insults marked by hematoxylin and eosin, terminal deoxynucleotidyl transferase dUTP nick end labeling, and Fluoro-Jade B staining by 24, 48, and 72 h after SE was induced. The expression of the apoptosis-suppressing protein, Bcl-2, was elevated, whereas the expression of the apoptosis-promoting proteins, Bax and Caspase-3, was suppressed in TGFβ1-treated rats compared to rats without TGFβ1 treatment by 24, 48, and 72 h following induction of SE. The seizure number, severity, and duration of SRS over a 1-month period of monitoring starting 15 days after SE induction as well as the cognitive deficits detected 45 days after SE induction were significantly reduced in TGFβ1-treated rats compared to those without TGFβ1 treatment. Our results indicate that intranasal delivery of TGFβ1 immediately after SE induction not only protected against SRS but also improved cognitive function. The anti-epileptogenic properties of TGFβ1 may be related to its effect of neuroprotection or to its effect of apoptosis pathway changes.
Collapse
Affiliation(s)
- Liang-Yong Li
- Epilepsy and Headache Group, Department of Neurology, the First Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | | | | | | | | | | | | |
Collapse
|
96
|
Chacón PJ, Rodríguez-Tébar A. Increased expression of the homologue of enhancer-of-split 1 protects neurons from beta amyloid neurotoxicity and hints at an alternative role for transforming growth factor beta1 as a neuroprotector. ALZHEIMERS RESEARCH & THERAPY 2012; 4:31. [PMID: 22849569 PMCID: PMC3506945 DOI: 10.1186/alzrt134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 06/11/2012] [Accepted: 07/31/2012] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the deposition of β-amyloid (Aβ) in the brain, which produces progressive neuronal loss and dementia. We recently demonstrated that the noxious effects of Aβ on cultured hippocampal neurons are in part provoked by the antagonism of nerve growth factor (NGF) signalling, which impairs the activation of nuclear factor κB (NF-κB) by impeding the tyrosine phosphorylation of I-κBα. As a result, the expression of the homologue of Enhancer-of split 1 (Hes1) gene is downregulated and ultimately, gamma-aminobutyric acid (GABA)-ergic connectivity is lost. METHODS Hes1 activity was promoted in cultured hippocampal neurons by overexpressing a Hes1-encoding plasmid or by upregulating this gene by activating NF-κB through different approaches (overexpressing either the I-κB kinaseβ, or p65/RelA/NF-κB). Alternatively neurons were exposed to TGFβ1. Dendrite patterning, GABAergic connectivity and cell survival were analyzed by immunofluorescence microscopy. Hes1 expression was determined by real-time PCR. NF-κB activation was measured using the dual-luciferase reporter assay. RESULTS The expression of Hes1 abolished the effects of Aβ on dendritic patterning and GABAergic input, and it prevented the death of the cultured neurons. TGFβ1, a known neuroprotector, could counteract the deleterious effects of Aβ by inducing NF-κB activation following the serine phosphorylation of I-κBα. Indeed, the number of GABAergic terminals generated by inducing Hes1 expression was doubled. CONCLUSION Our data define some of the mechanisms involved in Aβ-mediated cell death and they point to potential means to counteract this noxious activity.
Collapse
Affiliation(s)
- Pedro J Chacón
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Americo Vespucio s/n, Isla de la Cartuja, 41092 Seville, Spain.
| | | |
Collapse
|
97
|
Meta-analysis of the transforming growth factor-β1 polymorphisms and susceptibility to Alzheimer’s disease. J Neural Transm (Vienna) 2012; 120:353-60. [DOI: 10.1007/s00702-012-0850-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Accepted: 06/21/2012] [Indexed: 01/13/2023]
|
98
|
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: 185] [Impact Index Per Article: 14.2] [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
|
99
|
Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F. New pharmacological strategies for treatment of Alzheimer's disease: focus on disease modifying drugs. Br J Clin Pharmacol 2012; 73:504-17. [PMID: 22035455 DOI: 10.1111/j.1365-2125.2011.04134.x] [Citation(s) in RCA: 206] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Current approved drug treatments for Alzheimer disease (AD) include cholinesterase inhibitors (donepezil, rivastigmine, galantamine) and the NMDA receptor antagonist memantine. These drugs provide symptomatic relief but poorly affect the progression of the disease. Drug discovery has been directed, in the last 10 years, to develop 'disease modifying drugs' hopefully able to counteract the progression of AD. Because in a chronic, slow progressing pathological process, such as AD, an early start of treatment enhances the chance of success, it is crucial to have biomarkers for early detection of AD-related brain dysfunction, usable before clinical onset. Reliable early biomarkers need therefore to be prospectively tested for predictive accuracy, with specific cut off values validated in clinical practice. Disease modifying drugs developed so far include drugs to reduce β amyloid (Aβ) production, drugs to prevent Aβ aggregation, drugs to promote Aβ clearance, drugs targeting tau phosphorylation and assembly and other approaches. Unfortunately none of these drugs has demonstrated efficacy in phase 3 studies. The failure of clinical trials with disease modifying drugs raises a number of questions, spanning from methodological flaws to fundamental understanding of AD pathophysiology and biology. Recently, new diagnostic criteria applicable to presymptomatic stages of AD have been published. These new criteria may impact on drug development, such that future trials on disease modifying drugs will include populations susceptible to AD, before clinical onset. Specific problems with completed trials and hopes with ongoing trials are discussed in this review.
Collapse
Affiliation(s)
- Salvatore Salomone
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry Department of Formative Processes, University of Catania, Viale Andrea Doria 6, Catania, Italy
| | | | | | | | | |
Collapse
|
100
|
Liu WY, Wang ZB, Zhang LC, Wei X, Li L. Tight junction in blood-brain barrier: an overview of structure, regulation, and regulator substances. CNS Neurosci Ther 2012; 18:609-15. [PMID: 22686334 DOI: 10.1111/j.1755-5949.2012.00340.x] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Blood-brain barrier (BBB) is a dynamic interference that regulates the nutrition and toxic substance in and out of the central nervous system (CNS), and plays a crucial role in maintaining a stable circumstance of the CNS. Tight junctions among adjacent cells form the basic structure of BBB to limiting paracellular permeability. In the present review, the constituents of tight junction proteins are depicted in detail, together with the regulation of tight junction under stimulation and in pathological conditions. Tight junction modulators are also discussed.
Collapse
Affiliation(s)
- Wei-Ye Liu
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | | | | | | | | |
Collapse
|