1
|
Attia FM, Kassab RB, Ahmed-Farid OA, Abdel Moneim AE, El-Yamany NA. Zinc Oxide Nanoparticles Attenuated Neurochemical and Histopathological Alterations Associated with Aluminium Chloride Intoxication in Rats. Biol Trace Elem Res 2025; 203:2058-2071. [PMID: 38963645 DOI: 10.1007/s12011-024-04292-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
The present investigation explored the potential neuroprotective role of zinc oxide nanoparticles (ZnONPs) on aluminum chloride (AlCl3)-mediated Alzheimer's disease (AD)-like symptoms. Rats were distributed into four treatment groups equally: control, ZnONPs (4 mg/kg b.wt.), AlCl3 (100 mg/kg b.wt.), and ZnONPs + AlCl3 groups. Rats were treated for 42 consecutive days. ZnONPs injection into AlCl3-treated rats suppressed the development of oxidative challenge in the cortical and hippocampal tissues, as demonstrated by the decreased neuronal pro-oxidants (malondialdehyde and nitric oxide), and the increased glutathione and catalase levels. Additionally, ZnONPs injection showed anti-inflammatory potency in response to AlCl3 by decreasing levels of tumor necrosis factor-α and interleukin-1β. Moreover, pretreatment with ZnONPs prevented neuronal cell loss by decreasing the level of pro-apoptotic caspase-3 and enhancing the anti-apoptotic B cell lymphoma 2. Furthermore, ZnONPs ameliorated the disturbed acetylcholinesterase activity, monoamines (norepinephrine, dopamine, and serotonin), excitatory (glutamic and aspartic acids), and inhibitory amino acids (GABA and glycine) in response to AlCl3 exposure. These findings indicate that ZnONPs may have the potential as an alternative therapy to minimize or prevent the neurological deficits in AD model by exhibiting antioxidative, anti-inflammation, anti-apoptosis, and neuromodulatory effects.
Collapse
Affiliation(s)
- Fatma M Attia
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Rami B Kassab
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt.
- Biology Department, Faculty of Science and Arts, Almakhwah, Al Baha University, Al Baha, Saudi Arabia.
| | | | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Nabil A El-Yamany
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
2
|
Melchiorri D, Merlo S, Micallef B, Borg JJ, Dráfi F. Alzheimer's disease and neuroinflammation: will new drugs in clinical trials pave the way to a multi-target therapy? Front Pharmacol 2023; 14:1196413. [PMID: 37332353 PMCID: PMC10272781 DOI: 10.3389/fphar.2023.1196413] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/02/2023] [Indexed: 06/20/2023] Open
Abstract
Despite extensive research, no disease-modifying therapeutic option, able to prevent, cure or halt the progression of Alzheimer's disease [AD], is currently available. AD, a devastating neurodegenerative pathology leading to dementia and death, is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of neurofibrillary tangles (NFTs) consisting of altered hyperphosphorylated tau protein. Both have been widely studied and pharmacologically targeted for many years, without significant therapeutic results. In 2022, positive data on two monoclonal antibodies targeting Aβ, donanemab and lecanemab, followed by the 2023 FDA accelerated approval of lecanemab and the publication of the final results of the phase III Clarity AD study, have strengthened the hypothesis of a causal role of Aβ in the pathogenesis of AD. However, the magnitude of the clinical effect elicited by the two drugs is limited, suggesting that additional pathological mechanisms may contribute to the disease. Cumulative studies have shown inflammation as one of the main contributors to the pathogenesis of AD, leading to the recognition of a specific role of neuroinflammation synergic with the Aβ and NFTs cascades. The present review provides an overview of the investigational drugs targeting neuroinflammation that are currently in clinical trials. Moreover, their mechanisms of action, their positioning in the pathological cascade of events that occur in the brain throughout AD disease and their potential benefit/limitation in the therapeutic strategy in AD are discussed and highlighted as well. In addition, the latest patent requests for inflammation-targeting therapeutics to be developed in AD will also be discussed.
Collapse
Affiliation(s)
- Daniela Melchiorri
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Sara Merlo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | | | - John-Joseph Borg
- Malta Medicines Authority, San Ġwann, Malta
- School of Pharmacy, Department of Biology, University of Tor Vergata, Rome, Italy
| | - František Dráfi
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine SAS Bratislava, Bratislava, Slovakia
- State Institute for Drug Control, Bratislava, Slovakia
| |
Collapse
|
3
|
Azargoonjahromi A. Dual role of nitric oxide in Alzheimer's Disease. Nitric Oxide 2023; 134-135:23-37. [PMID: 37019299 DOI: 10.1016/j.niox.2023.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Nitric oxide (NO), an enzymatic product of nitric oxide synthase (NOS), has been associated with a variety of neurological diseases such as Alzheimer's disease (AD). NO has long been thought to contribute to neurotoxic insults caused by neuroinflammation in AD. This perception shifts as more attention is paid to the early stages before cognitive problems manifest. However, it has revealed a compensatory neuroprotective role for NO that protects synapses by increasing neuronal excitability. NO can positively affect neurons by inducing neuroplasticity, neuroprotection, and myelination, as well as having cytolytic activity to reduce inflammation. NO can also induce long-term potentiation (LTP), a process by which synaptic connections among neurons become more potent. Not to mention that such functions give rise to AD protection. Notably, it is unquestionably necessary to conduct more research to clarify NO pathways in neurodegenerative dementias because doing so could help us better understand their pathophysiology and develop more effective treatment options. All these findings bring us to the prevailing notion that NO can be used either as a therapeutic agent in patients afflicted with AD and other memory impairment disorders or as a contributor to the neurotoxic and aggressive factor in AD. In this review, after presenting a general background on AD and NO, various factors that have a pivotal role in both protecting and exacerbating AD and their correlation with NO will be elucidated. Following this, both the neuroprotective and neurotoxic effects of NO on neurons and glial cells among AD cases will be discussed in detail.
Collapse
|
4
|
Lamie PF, Abdel-Fattah MM, Philoppes JN. Design and synthesis of new indole drug candidates to treat Alzheimer's disease and targeting neuro-inflammation using a multi-target-directed ligand (MTDL) strategy. J Enzyme Inhib Med Chem 2022; 37:2660-2678. [PMID: 36146947 PMCID: PMC9518246 DOI: 10.1080/14756366.2022.2126464] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A novel series of indole-based compounds was designed, synthesised, and evaluated as anti-Alzheimer’s and anti-neuroinflammatory agents. The designed compounds were in vitro evaluated for their AChE and BuChE inhibitory activities. The obtained results revealed that compound 3c had higher selectivity for AChE than BuChE, while, 4a, 4b, and 4d showed selectivity for BuChE over AChE. Compounds 5b, 6b, 7c, and 10b exerted dual AChE/BuChE inhibitory activities at nanomolar range. Compounds 5b and 6b had the ability to inhibit the self-induced Aβ amyloid aggregation. Different anti-inflammatory mediators (NO, COX-2, IL-1β, and TNF-α) were assessed for compounds 5b and 6b. Cytotoxic effect of 5b and 6b against human neuroblastoma (SH-SY5Y) and normal hepatic (THLE2) cell lines was screened in vitro. Molecular docking study inside rhAChE and hBuChE active sites, drug-likeness, and ADMET prediction were performed.
Collapse
Affiliation(s)
- Phoebe F Lamie
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Maha M Abdel-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - John N Philoppes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
5
|
Kumar A, Sinha N, Kodidela S, Zhou L, Singh UP, Kumar S. Effect of benzo(a)pyrene on oxidative stress and inflammatory mediators in astrocytes and HIV-infected macrophages. PLoS One 2022; 17:e0275874. [PMID: 36240258 PMCID: PMC9565757 DOI: 10.1371/journal.pone.0275874] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Benzo(a)pyrene (BaP), an important polycyclic aromatic hydrocarbons (PAH) component of cigarette/tobacco smoking, is known to cause adverse health effects and is responsible for various life-threatening conditions including cancer. However, it is not yet clear whether BaP contributes to the macrophage- and astrocyte-mediated inflammatory response. METHODS We examined the acute (up to 72 h) effects of BaP on the expression of antioxidant enzymes (AOEs), cytokines/chemokines, and cytochromes P450 (CYP) enzymes in astrocytic cell lines, SVGA, and chronically HIV-infected U1 macrophage. The treated cells were examined for mRNA, protein levels of CYPs, AOEs superoxide dismutase-1 (SOD1) and catalase (CAT), cytokines/chemokines, using Western blot, multiplex ELISA, and reactive oxygen species (ROS) by flow cytometry analysis. RESULTS Upon acute exposure, BaP (1 μM) showed a significant increase in the mRNA levels of CYPs (CYP1A1 and CYP1B1), and pro-inflammatory cytokine IL-1β in SVGA cells following BaP for 24, 48, and 72h. In addition, we observed a significant increase in the mRNA levels of SOD1 and CAT at 24h of BaP treatment. In contrast, BaP did not exert any change in the protein expression of AOEs and CYP enzymes. In U1 cells, however, we noticed an interesting increase in the levels of MCP-1 as well as a modest increase in TNFα, IL-8 and IL-1β levels observed at 72 h of BaP treatment but could not reach to statistically significant level. CONCLUSIONS Overall, these results suggest that BaP contributes in part to macrophage and astrocyte-mediated neuroinflammation by mainly inducing IL-1β and MCP-1 production, which is likely to occur with the involvement of CYP and/or oxidative stress pathways.
Collapse
Affiliation(s)
- Asit Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Namita Sinha
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Sunitha Kodidela
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Lina Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
- * E-mail:
| |
Collapse
|
6
|
The Therapeutic Prospects of Targeting IL-1R1 for the Modulation of Neuroinflammation in Central Nervous System Disorders. Int J Mol Sci 2022; 23:ijms23031731. [PMID: 35163653 PMCID: PMC8915186 DOI: 10.3390/ijms23031731] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 11/16/2022] Open
Abstract
The interleukin-1 receptor type 1 (IL-1R1) holds pivotal roles in the immune system, as it is positioned at the “epicenter” of the inflammatory signaling networks. Increased levels of the cytokine IL-1 are a recognized feature of the immune response in the central nervous system (CNS) during injury and disease, i.e., neuroinflammation. Despite IL-1/IL-1R1 signaling within the CNS having been the subject of several studies, the roles of IL-1R1 in the CNS cellular milieu still cause controversy. Without much doubt, however, the persistent activation of the IL-1/IL-1R1 signaling pathway is intimately linked with the pathogenesis of a plethora of CNS disease states, ranging from Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS), all the way to schizophrenia and prion diseases. Importantly, a growing body of evidence is showing that blocking IL-1R1 signaling via pharmacological or genetic means in different experimental models of said CNS diseases leads to reduced neuroinflammation and delayed disease progression. The aim of this paper is to review the recent progress in the study of the biological roles of IL-1R1, as well as to highlight key aspects that render IL-1R1 a promising target for the development of novel disease-modifying treatments for multiple CNS indications.
Collapse
|
7
|
de Oliveira J, Kucharska E, Garcez ML, Rodrigues MS, Quevedo J, Moreno-Gonzalez I, Budni J. Inflammatory Cascade in Alzheimer's Disease Pathogenesis: A Review of Experimental Findings. Cells 2021; 10:cells10102581. [PMID: 34685563 PMCID: PMC8533897 DOI: 10.3390/cells10102581] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia worldwide. Most AD patients develop the disease in late life, named late onset AD (LOAD). Currently, the most recognized explanation for AD pathology is the amyloid cascade hypothesis. It is assumed that amyloid beta (Aβ) aggregation and deposition are critical pathogenic processes in AD, leading to the formation of amyloid plaques, as well as neurofibrillary tangles, neuronal cell death, synaptic degeneration, and dementia. In LOAD, the causes of Aβ accumulation and neuronal loss are not completely clear. Importantly, the blood–brain barrier (BBB) disruption seems to present an essential role in the induction of neuroinflammation and consequent AD development. In addition, we propose that the systemic inflammation triggered by conditions like metabolic diseases or infections are causative factors of BBB disruption, coexistent inflammatory cascade and, ultimately, the neurodegeneration observed in AD. In this regard, the use of anti-inflammatory molecules could be an interesting strategy to treat, delay or even halt AD onset and progression. Herein, we review the inflammatory cascade and underlying mechanisms involved in AD pathogenesis and revise the anti-inflammatory effects of compounds as emerging therapeutic drugs against AD.
Collapse
Affiliation(s)
- Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90050-000, Brazil; (J.d.O.); (M.S.R.)
| | - Ewa Kucharska
- Faculty of Education, Institute of Educational Sciences, Jesuit University Ignatianum in Krakow, 31-501 Krakow, Poland;
| | - Michelle Lima Garcez
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis 88040-900, Santa Catarina, Brazil;
| | - Matheus Scarpatto Rodrigues
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90050-000, Brazil; (J.d.O.); (M.S.R.)
| | - João Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA;
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, MD Anderson Cancer Center, UTHealth, The University of Texas Houston, Houston, TX 77030, USA
- Graduate Program in Health Sciences, Translational Psychiatry Laboratory, University of Southern Santa Catarina (UNESC), Criciuma 88806-000, Brazil
| | - Ines Moreno-Gonzalez
- Department of Cell Biology, Faculty of Sciences, University of Malaga, IBIMA, 29010 Malaga, Spain;
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 29010 Malaga, Spain
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Josiane Budni
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Neurologia Experimental, Universidade do Extremo Sul Catarinense, Criciuma 88806-000, Brazil
- Correspondence: ; Tel.: +55-48431-2539
| |
Collapse
|
8
|
Mohamed RA, Abdallah DM, El-brairy AI, Ahmed KA, El-Abhar HS. Palonosetron/Methyllycaconitine Deactivate Hippocampal Microglia 1, Inflammasome Assembly and Pyroptosis to Enhance Cognition in a Novel Model of Neuroinflammation. Molecules 2021; 26:5068. [PMID: 34443654 PMCID: PMC8401912 DOI: 10.3390/molecules26165068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/30/2022] Open
Abstract
Since westernized diet-induced insulin resistance is a risk factor in Alzheimer's disease (AD) development, and lipopolysaccharide (LPS) coexists with amyloid β (Aβ)1-42 in these patients, our AD novel model was developed to resemble sporadic AD by injecting LPS into high fat/fructose diet (HFFD)-fed rats. The neuroprotective potential of palonosetron and/or methyllycaconitine, 5-HT3 receptor and α7 nAChR blockers, respectively, was evaluated after 8 days of daily administration in HFFD/LPS rats. All regimens improved histopathological findings and enhanced spatial memory (Morris Water Maze); however, palonosetron alone or with methyllycaconitine promoted animal performance during novel object recognition tests. In the hippocampus, all regimens reduced the expression of glial fibrillary acidic protein and skewed microglia M1 to M2 phenotype, indicated by the decreased M1 markers and the enhanced M2 related parameters. Additionally, palonosetron and its combination regimen downregulated the expression of ASC/TMS1, as well as levels of inflammasome downstream molecules and abated cleaved caspase-1, interleukin (IL)-1β, IL-18 and caspase-11. Furthermore, ACh and 5-HT were augmented after being hampered by the insult. Our study speculates that blocking 5-HT3 receptor using palonosetron overrides methyllycaconitine to combat AD-induced neuroinflammation and inflammasome cascade, as well as to restore microglial function in a HFFD/LPS novel model for sporadic AD.
Collapse
Affiliation(s)
- Reem A. Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 26 July Mehwar Road Intersection with Wahat Road, 6th of October City, Giza 12451, Egypt; (R.A.M.); (A.I.E.-b.)
| | - Dalaal M. Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., Cairo 11562, Egypt;
| | - Amany I. El-brairy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 26 July Mehwar Road Intersection with Wahat Road, 6th of October City, Giza 12451, Egypt; (R.A.M.); (A.I.E.-b.)
| | - Kawkab A. Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Hanan S. El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., Cairo 11562, Egypt;
| |
Collapse
|
9
|
Babić Leko M, Nikolac Perković M, Klepac N, Štrac DŠ, Borovečki F, Pivac N, Hof PR, Šimić G. IL-1β, IL-6, IL-10, and TNFα Single Nucleotide Polymorphisms in Human Influence the Susceptibility to Alzheimer's Disease Pathology. J Alzheimers Dis 2021; 75:1029-1047. [PMID: 32390629 DOI: 10.3233/jad-200056] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neuroinflammation plays an important role in Alzheimer's disease (AD). During this process, activated microglia release pro-inflammatory cytokines such as interleukin (IL)-1α, IL-1β, IL-6, and tumor necrosis factor α (TNFα) that participate in neuron damage, but also anti-inflammatory cytokines (such as IL-10), which maintain homeostasis of immune response. Previous studies showed the association of IL-1α -889C/T (rs1800587), IL-1β-1473G/C (rs1143623), IL-6 -174C/G (rs1800795), IL-10 -1082G/A (rs1800896), and TNFα -308A/G (rs1800629) polymorphisms with AD. OBJECTIVE We aimed to investigate whether people with certain IL-1α, IL-1β, IL-6, IL-10, and TNFα genotypes in these polymorphisms are more prone to develop AD-related pathology, reflected by pathological levels of cerebrospinal fluid (CSF) AD biomarkers including amyloid-β1-42, total tau (t-tau), tau phosphorylated at Thr 181 (p-tau181), Ser 199 (p-tau199), and Thr 231 (p-tau231), and visinin-like protein 1 (VILIP-1). METHODS The study included 115 AD patients, 53 patients with mild cognitive impairment, and 11 healthy controls. The polymorphisms were determined using real-time polymerase chain reaction. Levels of CSF biomarkers were determined by enzyme-linked immunosorbent assay. RESULTS A significant increase in p-tau CSF levels was found in patients with the AA IL-10 -1082G/A and GG TNFα -308A/G genotypes, and in carriers of a G allele in IL-1β -1473C/G and IL-6 -174C/G polymorphisms. t-tau levels were increased in carriers of a G allele in IL-1β -1473C/G polymorphism. An increase in VILIP-1 levels was observed in patients with CG and GG IL-1β -1473C/G, GC IL-6 -174C/G, and GG TNFα -308A/G genotype. CONCLUSION These results suggest that persons carrying certain genotypes in IL10 (-1082G/A), IL1β (1473C/G), IL6 (-174C/G), and TNFIα (-308A/G) could be more vulnerable to development of neuroinflammation, and consequently of AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | | | - Nataša Klepac
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nela Pivac
- Department of Molecular Medicine, Institute Ruđer Bošković, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
10
|
Lonnemann N, Hosseini S, Marchetti C, Skouras DB, Stefanoni D, D'Alessandro A, Dinarello CA, Korte M. The NLRP3 inflammasome inhibitor OLT1177 rescues cognitive impairment in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2020; 117:32145-32154. [PMID: 33257576 PMCID: PMC7749353 DOI: 10.1073/pnas.2009680117] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Numerous studies demonstrate that neuroinflammation is a key player in the progression of Alzheimer's disease (AD). Interleukin (IL)-1β is a main inducer of inflammation and therefore a prime target for therapeutic options. The inactive IL-1β precursor requires processing by the the nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome into a mature and active form. Studies have shown that IL-1β is up-regulated in brains of patients with AD, and that genetic inactivation of the NLRP3 inflammasome improves behavioral tests and synaptic plasticity phenotypes in a murine model of the disease. In the present study, we analyzed the effect of pharmacological inhibition of the NLRP3 inflammasome using dapansutrile (OLT1177), an oral NLRP3-specific inhibitor that is safe in humans. Six-month-old WT and APP/PS1 mice were fed with standard mouse chow or OLT1177-enriched chow for 3 mo. The Morris water maze test revealed an impaired learning and memory ability of 9-mo-old APP/PS1 mice (P = 0.001), which was completely rescued by OLT1177 fed to mice (P = 0.008 to untreated APP/PS1). Furthermore, our findings revealed that 3 mo of OLT1177 diet can rescue synaptic plasticity in this mouse model of AD (P = 0.007 to untreated APP/PS1). In addition, microglia were less activated (P = 0.07) and the number of plaques was reduced in the cortex (P = 0.03) following NLRP3 inhibition with OLT1177 administration. We also observed an OLT1177 dose-dependent normalization of plasma metabolic markers of AD to those of WT mice. This study suggests the therapeutic potential of treating neuroinflammation with an oral inhibitor of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Niklas Lonnemann
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
- Neuroinflammation and Neurodegeneration Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Carlo Marchetti
- Department of Medicine, University of Colorado, Denver, Aurora, CO 80045
| | | | - Davide Stefanoni
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO 80045
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO 80045
| | - Charles A Dinarello
- Department of Medicine, University of Colorado, Denver, Aurora, CO 80045;
- Department of Medicine, Radboud University, Medical Center, 6525 Nijmegen, The Netherlands
| | - Martin Korte
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany;
- Neuroinflammation and Neurodegeneration Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| |
Collapse
|
11
|
Antonyová V, Kejík Z, Brogyányi T, Kaplánek R, Pajková M, Talianová V, Hromádka R, Masařík M, Sýkora D, Mikšátková L, Martásek P, Jakubek M. Role of mtDNA disturbances in the pathogenesis of Alzheimer's and Parkinson's disease. DNA Repair (Amst) 2020; 91-92:102871. [PMID: 32502755 DOI: 10.1016/j.dnarep.2020.102871] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (e.g. Alzheimer's and Parkinson's disease) are becoming increasingly problematic to healthcare systems. Therefore, their underlying mechanisms are trending topics of study in medicinal research. Numerous studies have evidenced a strong association between mitochondrial DNA disturbances (e.g. oxidative damage, mutations, and methylation shifts) and the initiation and progression of neurodegenerative diseases. Therefore, this review discusses the risk and development of neurodegenerative diseases in terms of disturbances in mitochondrial DNA and as a part of a complex ecosystem that includes other important mechanisms (e.g. neuroinflammation and the misfolding and aggregation of amyloid-β peptides, α-synuclein, and tau proteins). In addition, the influence of individual mitochondrial DNA haplogroups on the risk and development of neurodegenerative diseases is also described and discussed.
Collapse
Affiliation(s)
- Veronika Antonyová
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - Zdeněk Kejík
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic
| | - Tereza Brogyányi
- Depertment of Pathological Physiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 121 00 Prague 2, Czech Republic
| | - Robert Kaplánek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic
| | - Martina Pajková
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - Veronika Talianová
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - Róbert Hromádka
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - Michal Masařík
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - David Sýkora
- BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic
| | - Lucie Mikšátková
- BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic
| | - Pavel Martásek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic.
| | - Milan Jakubek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic.
| |
Collapse
|
12
|
Jiang H, Jayadev S, Lardelli M, Newman M. A Review of the Familial Alzheimer's Disease Locus PRESENILIN 2 and Its Relationship to PRESENILIN 1. J Alzheimers Dis 2019; 66:1323-1339. [PMID: 30412492 DOI: 10.3233/jad-180656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PRESENILIN 1 (PSEN1) and PRESENILIN 2 (PSEN2) genes are loci for mutations causing familial Alzheimer's disease (fAD). However, the function of these genes and how they contribute to fAD pathogenesis has not been fully determined. This review provides a summary of the overlapping and independent functions of the PRESENILINS with a focus on the lesser studied PSEN2. As a core component of the γ-secretase complex, the PSEN2 protein is involved in many γ-secretase-related physiological activities, including innate immunity, Notch signaling, autophagy, and mitochondrial function. These physiological activities have all been associated with AD progression, indicating that PSEN2 plays a particular role in AD pathogenesis.
Collapse
Affiliation(s)
- Haowei Jiang
- Alzheimer's Disease Genetics Laboratory, Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Michael Lardelli
- Alzheimer's Disease Genetics Laboratory, Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Morgan Newman
- Alzheimer's Disease Genetics Laboratory, Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
13
|
Martini AC, Gomez-Arboledas A, Forner S, Rodriguez-Ortiz CJ, McQuade A, Danhash E, Phan J, Javonillo D, Ha JV, Tram M, Trujillo-Estrada L, da Cunha C, Ager RR, Davila JC, Kitazawa M, Blurton-Jones M, Gutierrez A, Baglietto-Vargas D, Medeiros R, LaFerla FM. Amyloid-beta impairs TOM1-mediated IL-1R1 signaling. Proc Natl Acad Sci U S A 2019; 116:21198-21206. [PMID: 31570577 PMCID: PMC6800331 DOI: 10.1073/pnas.1914088116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Defects in interleukin-1β (IL-1β)-mediated cellular responses contribute to Alzheimer's disease (AD). To decipher the mechanism associated with its pathogenesis, we investigated the molecular events associated with the termination of IL-1β inflammatory responses by focusing on the role played by the target of Myb1 (TOM1), a negative regulator of the interleukin-1β receptor-1 (IL-1R1). We first show that TOM1 steady-state levels are reduced in human AD hippocampi and in the brain of an AD mouse model versus respective controls. Experimentally reducing TOM1 affected microglia activity, substantially increased amyloid-beta levels, and impaired cognition, whereas enhancing its levels was therapeutic. These data show that reparation of the TOM1-signaling pathway represents a therapeutic target for brain inflammatory disorders such as AD. A better understanding of the age-related changes in the immune system will allow us to craft therapies to limit detrimental aspects of inflammation, with the broader purpose of sharply reducing the number of people afflicted by AD.
Collapse
Affiliation(s)
- Alessandra Cadete Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Angela Gomez-Arboledas
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Carlos J Rodriguez-Ortiz
- Center for Occupational and Environmental Health, School of Medicine, University of California, Irvine, CA 92697
| | - Amanda McQuade
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Emma Danhash
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Dominic Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Jordan-Vu Ha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Melanie Tram
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Rahasson R Ager
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Jose C Davila
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health, School of Medicine, University of California, Irvine, CA 92697
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Antonia Gutierrez
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| | - Rodrigo Medeiros
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697;
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697;
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| |
Collapse
|
14
|
Novak P, Cente M, Kosikova N, Augustin T, Kvetnansky R, Novak M, Filipcik P. Stress-Induced Alterations of Immune Profile in Animals Suffering by Tau Protein-Driven Neurodegeneration. Cell Mol Neurobiol 2018; 38:243-259. [PMID: 28405903 PMCID: PMC11481851 DOI: 10.1007/s10571-017-0491-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/29/2017] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial disorder; neurofibrillary pathology composed of tau protein is found side by side with amyloid-β deposits and extensive neuroinflammation. The immune system of the brain is considered as one of the factors that could influence the speed of the progression of AD neuropathology as a potential mediator of the damage induced by AD protein deposits. Alzheimer's disease pathology can be impacted by psychological stress; however, signalling pathways in background are not well known. We have explored possible avenues of how stress could influence the brain's immune system in a rat model of AD. Animals were subjected either to a single or multiple instances of immobilization stress. The analysis of a panel of immunity-related genes was used to evaluate the impact of stress on the immune response in the brain. We have identified 19 stress-responsive genes that are involved in neuroinflammation accompanying tau pathology: Nos2, Ptgs2, IL-8rb, C5, Mmp9, Cx3cr1, CD40lg, Adrb2, IL-6, IL-6r, IL-1r2, Ccl2, Ccl3, Ccl4, Ccl12, TNF-α, IL-1α, IL-1β, IL-10. Most of them are deregulated under the stress conditions also in control animals; however, the magnitude of the response to either acute or chronic stress differs. This can lead to serious influence, most probably to acceleration of neurodegenerative phenotype in diseased animals. Several of the genes (IL-1β, Casp1, Cx3cr1 and C5) are deregulated solely in tauopathic animals. The stress-induced changes in the inflammatory picture of the brain highlight the fact that the brain's immune response is highly responsive to environmental stimuli. The pattern of changes is indicative of an attempt to protect the brain in the short term, while being potentially detrimental to the response against a long-term pathological process such as neurofibrillary degeneration.
Collapse
Affiliation(s)
- Petr Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Martin Cente
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Nina Kosikova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Augustin
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Richard Kvetnansky
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Peter Filipcik
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia.
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia.
| |
Collapse
|
15
|
Khazim K, Azulay EE, Kristal B, Cohen I. Interleukin 1 gene polymorphism and susceptibility to disease. Immunol Rev 2017; 281:40-56. [DOI: 10.1111/imr.12620] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Khaled Khazim
- Faculty of Medicine in the Galilee; Bar Ilan University; Safed Israel
- Department of Nephrology and Hypertension; Galilee Medical Center; Nahariya Israel
| | - Etti Ester Azulay
- Faculty of Medicine in the Galilee; Bar Ilan University; Safed Israel
- Research Institute; Galilee Medical Center; Nahariya Israel
| | - Batya Kristal
- Faculty of Medicine in the Galilee; Bar Ilan University; Safed Israel
- Department of Nephrology and Hypertension; Galilee Medical Center; Nahariya Israel
| | - Idan Cohen
- Faculty of Medicine in the Galilee; Bar Ilan University; Safed Israel
- Research Institute; Galilee Medical Center; Nahariya Israel
| |
Collapse
|
16
|
Ebrahimie E, Moussavi Nik SH, Newman M, Van Der Hoek M, Lardelli M. The Zebrafish Equivalent of Alzheimer's Disease-Associated PRESENILIN Isoform PS2V Regulates Inflammatory and Other Responses to Hypoxic Stress. J Alzheimers Dis 2017; 52:581-608. [PMID: 27031468 DOI: 10.3233/jad-150678] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Dominant mutations in the PRESENILIN genes PSEN1 and PSEN2 cause familial Alzheimer's disease (fAD) that usually shows onset before 65 years of age. In contrast, genetic variation at the PSEN1 and PSEN2 loci does not appear to contribute to risk for the sporadic, late onset form of the disease (sAD), leading to doubts that these genes play a role in the majority of AD cases. However, a truncated isoform of PSEN2, PS2V, is upregulated in sAD brains and is induced by hypoxia and high cholesterol intake. PS2V can increase γ-secretase activity and suppress the unfolded protein response (UPR), but detailed analysis of its function has been hindered by lack of a suitable, genetically manipulable animal model since mice and rats lack this PRESENILIN isoform. We recently showed that zebrafish possess an isoform, PS1IV, that is cognate to human PS2V. Using an antisense morpholino oligonucleotide, we can block specifically the induction of PS1IV that normally occurs under hypoxia. Here, we exploit this ability to identify gene regulatory networks that are modulated by PS1IV. When PS1IV is absent under hypoxia-like conditions, we observe changes in expression of genes controlling inflammation (particularly sAD-associated IL1B and CCR5), vascular development, the UPR, protein synthesis, calcium homeostasis, catecholamine biosynthesis, TOR signaling, and cell proliferation. Our results imply an important role for PS2V in sAD as a component of a pathological mechanism that includes hypoxia/oxidative stress and support investigation of the role of PS2V in other diseases, including schizophrenia, when these are implicated in the pathology.
Collapse
Affiliation(s)
- Esmaeil Ebrahimie
- Department of Genetics and Evolution, School of Biological Sciences, University of Adelaide, Adelaide, Australia.,School of Information Technology and Mathematical Sciences, Division of Information Technology, Engineering and the Environment, University of South Australia, Adelaide, Australia.,School of Biological Sciences, Faculty of Science and Engineering, Flinders University, Adelaide, Australia
| | - Seyyed Hani Moussavi Nik
- Department of Genetics and Evolution, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Morgan Newman
- Department of Genetics and Evolution, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Mark Van Der Hoek
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, Australia
| | - Michael Lardelli
- Department of Genetics and Evolution, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
17
|
Ardura-Fabregat A, Boddeke EWGM, Boza-Serrano A, Brioschi S, Castro-Gomez S, Ceyzériat K, Dansokho C, Dierkes T, Gelders G, Heneka MT, Hoeijmakers L, Hoffmann A, Iaccarino L, Jahnert S, Kuhbandner K, Landreth G, Lonnemann N, Löschmann PA, McManus RM, Paulus A, Reemst K, Sanchez-Caro JM, Tiberi A, Van der Perren A, Vautheny A, Venegas C, Webers A, Weydt P, Wijasa TS, Xiang X, Yang Y. Targeting Neuroinflammation to Treat Alzheimer's Disease. CNS Drugs 2017; 31:1057-1082. [PMID: 29260466 PMCID: PMC5747579 DOI: 10.1007/s40263-017-0483-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Over the past few decades, research on Alzheimer's disease (AD) has focused on pathomechanisms linked to two of the major pathological hallmarks of extracellular deposition of beta-amyloid peptides and intra-neuronal formation of neurofibrils. Recently, a third disease component, the neuroinflammatory reaction mediated by cerebral innate immune cells, has entered the spotlight, prompted by findings from genetic, pre-clinical, and clinical studies. Various proteins that arise during neurodegeneration, including beta-amyloid, tau, heat shock proteins, and chromogranin, among others, act as danger-associated molecular patterns, that-upon engagement of pattern recognition receptors-induce inflammatory signaling pathways and ultimately lead to the production and release of immune mediators. These may have beneficial effects but ultimately compromise neuronal function and cause cell death. The current review, assembled by participants of the Chiclana Summer School on Neuroinflammation 2016, provides an overview of our current understanding of AD-related immune processes. We describe the principal cellular and molecular players in inflammation as they pertain to AD, examine modifying factors, and discuss potential future therapeutic targets.
Collapse
Affiliation(s)
- A. Ardura-Fabregat
- grid.5963.9Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - E. W. G. M. Boddeke
- 0000 0004 0407 1981grid.4830.fDepartment of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A. Boza-Serrano
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| | - S. Brioschi
- grid.5963.9Department of Psychiatry and Psychotherapy, Medical Center University of Freiburg, Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - S. Castro-Gomez
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - K. Ceyzériat
- grid.457334.2Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, MIRCen, 92260 Fontenay-aux-Roses, France ,0000 0001 2171 2558grid.5842.bNeurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, UMR 9199, F-92260 Fontenay-aux-Roses, France
| | - C. Dansokho
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - T. Dierkes
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany ,0000 0000 8786 803Xgrid.15090.3dBiomedical Centre, Institute of Innate Immunity, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - G. Gelders
- 0000 0001 0668 7884grid.5596.fDepartment of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Michael T. Heneka
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany ,0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - L. Hoeijmakers
- 0000000084992262grid.7177.6Center for Neuroscience (SILS-CNS), Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - A. Hoffmann
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - L. Iaccarino
- grid.15496.3fVita-Salute San Raffaele University, Milan, Italy ,0000000417581884grid.18887.3eIn Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S. Jahnert
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - K. Kuhbandner
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - G. Landreth
- 0000 0001 2287 3919grid.257413.6Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - N. Lonnemann
- 0000 0001 1090 0254grid.6738.aDepartment of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - R. M. McManus
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - A. Paulus
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| | - K. Reemst
- 0000000084992262grid.7177.6Center for Neuroscience (SILS-CNS), Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - J. M. Sanchez-Caro
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - A. Tiberi
- grid.6093.cBio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - A. Van der Perren
- 0000 0001 0668 7884grid.5596.fDepartment of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - A. Vautheny
- grid.457334.2Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, MIRCen, 92260 Fontenay-aux-Roses, France ,0000 0001 2171 2558grid.5842.bNeurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, UMR 9199, F-92260 Fontenay-aux-Roses, France
| | - C. Venegas
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - A. Webers
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - P. Weydt
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - T. S. Wijasa
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - X. Xiang
- 0000 0004 1936 973Xgrid.5252.0Biomedical Center (BMC), Biochemistry, Ludwig-Maximilians-University Munich, 81377 Munich, Germany ,0000 0004 1936 973Xgrid.5252.0Graduate School of Systemic Neuroscience, Ludwig-Maximilians-University, Munich, 82152 Munich, Germany
| | - Y. Yang
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| |
Collapse
|
18
|
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: 164] [Impact Index Per Article: 18.2] [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
|
19
|
Santos LE, Beckman D, Ferreira ST. Microglial dysfunction connects depression and Alzheimer's disease. Brain Behav Immun 2016; 55:151-165. [PMID: 26612494 DOI: 10.1016/j.bbi.2015.11.011] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 11/19/2015] [Accepted: 11/19/2015] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) and major depressive disorder (MDD) are highly prevalent neuropsychiatric conditions with intriguing epidemiological overlaps. Depressed patients are at increased risk of developing late-onset AD, and around one in four AD patients are co-diagnosed with MDD. Microglia are the main cellular effectors of innate immunity in the brain, and their activation is central to neuroinflammation - a ubiquitous process in brain pathology, thought to be a causal factor of both AD and MDD. Microglia serve several physiological functions, including roles in synaptic plasticity and neurogenesis, which may be disrupted in neuroinflammation. Following early work on the 'sickness behavior' of humans and other animals, microglia-derived inflammatory cytokines have been shown to produce depressive-like symptoms when administered exogenously or released in response to infection. MDD patients consistently show increased circulating levels of pro-inflammatory cytokines, and anti-inflammatory drugs show promise for treating depression. Activated microglia are abundant in the AD brain, and concentrate around senile plaques, hallmark lesions composed of aggregated amyloid-β peptide (Aβ). The Aβ burden in affected brains is regulated largely by microglial clearance, and the complex activation state of microglia may be crucial for AD progression. Intriguingly, recent reports have linked soluble Aβ oligomers, toxins that accumulate in AD brains and are thought to cause memory impairment, to increased brain cytokine production and depressive-like behavior in mice. Here, we review recent findings supporting the inflammatory hypotheses of AD and MDD, focusing on microglia as a common player and therapeutic target linking these devastating disorders.
Collapse
Affiliation(s)
- Luís Eduardo Santos
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Danielle Beckman
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Sergio T Ferreira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| |
Collapse
|
20
|
Dong X, Zhang L, Meng Q, Gao Q. Association Between Interleukin-1A, Interleukin-1B, and Bridging integrator 1 Polymorphisms and Alzheimer's Disease: a standard and Cumulative Meta-analysis. Mol Neurobiol 2016; 54:736-747. [PMID: 26768592 DOI: 10.1007/s12035-015-9683-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 12/28/2015] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) has been one of the most prevalent health problems among senior population. Interleukin-1A (IL-1A) and IL-1B are two isoforms of IL-1. Recent studies suggested that certain polymorphisms on these two genes are associated with AD. Bridging integrator 1 (BIN1) is considered as common genetic risk factors for AD, whereas different studies have provided various conclusions regarding its role in AD. This study was designed to justify the association between multiple gene polymorphisms and AD through an evidence synthesis approach. We conducted a literature search to identify relevant articles published from 2000 to 2015 from PubMed, Embase, and Cochrane Library, in accordance with inclusion criteria. Pooled odds ratios (ORs) were calculated for the allele model. The effect estimates were summarized by both standard and cumulative meta-analysis. Finally, 54 articles with 88 independent studies were enrolled in this meta-analysis. Mutants in rs1800587 of IL-1A, rs1143634 of IL-1B, rs12989701, and rs744373 of BIN1 were significantly associated with AD onset. The difference effect of same single nucleotide polymorphisms (SNPs) on various ethnicities was also observed in our results. The present meta-analysis suggested that IL-1A, IL-1B, and BIN1 were candidate genes for AD pathogenesis. Polymorphisms of IL-1A, IL-1B, and BIN1 are associated with AD onset.
Collapse
Affiliation(s)
- Xiaoliu Dong
- Department of Neurology, Tangshan People's Hospital, Tangshan, 063000, China.
| | - Li Zhang
- Department of Neurosurgery, Tangshan People's Hospital, Tangshan, 063000, China
| | - Qingling Meng
- Department of Internal Medicine, The People's Hospital of Qianxi County, Qianxi, 064300, China
| | - Qiuyan Gao
- Department of Ultrasound, Tangshan People's Hospital, Tangshan, 063000, China
| |
Collapse
|
21
|
Mun MJ, Kim JH, Choi JY, Jang WC. Genetic polymorphisms of interleukin genes and the risk of Alzheimer's disease: An update meta-analysis. Meta Gene 2016; 8:1-10. [PMID: 27014584 PMCID: PMC4792847 DOI: 10.1016/j.mgene.2016.01.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/30/2015] [Accepted: 01/07/2016] [Indexed: 01/08/2023] Open
Abstract
Objectives Recently, several meta-analyses have reported an association between interleukin (IL) gene polymorphisms and the risk of Alzheimer's disease (AD). Several further papers discussing the relationship with the risk of AD have recently been published. The aim of this meta-analysis was to re-evaluate and update the associations between IL gene polymorphisms and the risk of AD. Methods The search sources were PubMed, Science Direct, Scopus, and Google Scholar up to July 2015, and the following search terms were used: “interleukin 1 or interleukin 6 or interleukin 10” and “variant or polymorphism or SNP” in combination with “Alzheimer's disease”. A meta-analysis using the pooled odds ratios and 95% confidence intervals was carried out to assess the associations between four polymorphisms of IL genes (− 889C > T in IL-1α, − 511C > T in IL-1β, − 174G > C in IL-6 and − 1082G > A in IL-10) and the risk of AD under the heterozygous, homozygous, dominant, and recessive models with fixed- or random-effects models. Results A total of 21,864 cases and 40,321 controls from 93 individual studies were included in this meta-analysis. Our results indicated that the − 889C > T polymorphism was strongly associated with the increased risk of AD. However, three polymorphisms were not associated with the risk of AD. Conclusions Similar to previous meta-analyses, our updated meta-analysis suggested that the − 889C > T polymorphism may be a factor in AD. However, the results of our meta-analysis of the − 174G > C polymorphism differed from those of previous meta-analyses. Consequently, we suggest that the − 174G > C polymorphism may not be a risk factor for AD. 889 C > T polymorphism of IL-1α was significantly associated with increased risk of Alzheimer's disease Three polymorphisms (− 511C > T in IL − 1β, − 174G > C in IL-6 and − 1082G > A in IL-10) were no associated with risk of Alzheimer's disease The results of our meta-analyses for three polymorphisms (− 889C > T, − 511C > T and − 1082G > A) were similar to those previous meta-analyses. However, the results of the − 174G > C polymorphism were different.
Collapse
Affiliation(s)
- Myung-Jin Mun
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University Graduate School, South Korea; Department of Chemistry, School of Natural Science, Dankook University, Cheonan 330-714, South Korea; Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, South Korea
| | - Jin-Ho Kim
- Department of Chemistry, School of Natural Science, Dankook University, Cheonan 330-714, South Korea
| | - Ji-Young Choi
- Department of Chemistry, School of Natural Science, Dankook University, Cheonan 330-714, South Korea
| | - Won-Cheoul Jang
- Department of Chemistry, School of Natural Science, Dankook University, Cheonan 330-714, South Korea
| |
Collapse
|
22
|
Giil LM, Kristoffersen EK, Vedeler CA, Aarsland D, Nordrehaug JE, Winblad B, Cedazo-Minguez A, Lund A, Reksten TR. Autoantibodies Toward the Angiotensin 2 Type 1 Receptor: A Novel Autoantibody in Alzheimer’s Disease. J Alzheimers Dis 2015; 47:523-9. [DOI: 10.3233/jad-150053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Lasse M. Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Einar K. Kristoffersen
- Institute of Clinical Science, University of Bergen, Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | | | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
- Centre for Age-Related Diseases (SESAM), Stavanger University Hospital, Stavanger, Norway
| | - Jan Erik Nordrehaug
- Institute of Clinical Science, University of Bergen, Bergen, Norway
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Anders Lund
- Institute of Clinical Science, University of Bergen, Bergen, Norway
| | - Tove Ragna Reksten
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
- Centre for Age-Related Diseases (SESAM), Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
23
|
Chiappelli M, Tumini E, Porcellini E, Licastro F. Impaired regulation of immune responses in cognitive decline and Alzheimer’s disease: lessons from genetic association studies. Expert Rev Neurother 2014; 6:1327-36. [PMID: 17009920 DOI: 10.1586/14737175.6.9.1327] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Altered levels of cytokines and acute-phase proteins have been described in the blood and brain of patients with Alzheimer's disease. Microglia are resident cells of the brain and metabolic upregulation of these cells may play a crucial role in the development of the neurodegeneration associated with Alzheimer's disease. Studies focusing on gene polymorphisms of molecules with immune regulatory function have demonstrated an association with increased risk of the disease and confirmed the pivotal role of immune responses in Alzheimer's disease pathogenesis. Several gene variants may also influence the rate of the cognitive decline associated with the disease. A definite immune-related gene polymorphism profile may be a feature of a limited group of patients with early onset of the disease and fast clinical deterioration. Only this group of patients may benefit from anti-inflammatory treatment.
Collapse
Affiliation(s)
- Martina Chiappelli
- University of Bologna, Department of Experimental Pathology, School of Medicine, Via S. Giacomo 14, 40126 Bologna, Italy.
| | | | | | | |
Collapse
|
24
|
Yuan H, Xia Q, Ge P, Wu S. Genetic polymorphism of interleukin 1β -511C/T and susceptibility to sporadic Alzheimer's disease: a meta-analysis. Mol Biol Rep 2012; 40:1827-34. [PMID: 23079713 DOI: 10.1007/s11033-012-2237-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 10/10/2012] [Indexed: 10/27/2022]
Abstract
A large number of epidemiological studies have been performed to investigate the association between Alzheimer's disease (AD) risk and interleukin-1β -511C/T genetic polymorphism, however, inconsistent results have been reported. The effect of the IL-1β -511C/T polymorphism on AD susceptibility was evaluated by a meta-analysis. Series of databases were researched. 14 studies involving 2640 AD case and 3493 control subjects were identified. The pooled results showed there were no statistical associations of interleukin-1β -511C/T genetic polymorphism with susceptibility to AD for five analysis models in all subjects. However, obvious heterogeneity among studies was detected. When stratifying for age at onset, ethnicity and geographic distribution of population to explore the original source of heterogeneity, the meta-analysis results based on geographic distribution of population showed the significant difference (CC vs CT, OR 1.26, 95 % CI: 1.03, 1.54, z = 2.25, P = 0.025; CC vs CT+TT, OR 1.24, 95 % CI: 1.03, 1.50, z = 2.24, P = 0.025) only in non-Europe. These findings indicate that the IL-1β -511C/T polymorphism might be associated with AD risk, and individuals with IL-1β -511C/C genotype might be at higher risk of AD in non-Europe. Further larger sample research would be warranted to confirm these conclusions.
Collapse
Affiliation(s)
- Hai Yuan
- Department of Rehabilitation Medicine, The Second People's Hospital of Hefei City, 246th Heping Road, Hefei, 230011, Anhui Province, China.
| | | | | | | |
Collapse
|
25
|
Matsumoto T, Samuel MD, Bollinger T, Pybus M, Coltman DW. Association mapping of genetic risk factors for chronic wasting disease in wild deer. Evol Appl 2012; 6:340-52. [PMID: 23467626 PMCID: PMC3586622 DOI: 10.1111/eva.12003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 07/11/2012] [Indexed: 01/06/2023] Open
Abstract
Chronic wasting disease (CWD) is a fatal transmissible spongiform encephalopathy affecting North American cervids. We assessed the feasibility of association mapping CWD genetic risk factors in wild white-tailed deer (Odocoileus virginianus) and mule deer (Odocoileus hemionus) using a panel of bovine microsatellite markers from three homologous deer linkage groups predicted to contain candidate genes. These markers had a low cross-species amplification rate (27.9%) and showed weak linkage disequilibrium (<1 cM). Markers near the prion protein and the neurofibromin 1 (NF1) genes were suggestively associated with CWD status in white-tailed deer (P = 0.006) and mule deer (P = 0.02), respectively. This is the first time an association between the NF1 region and CWD has been reported.
Collapse
Affiliation(s)
- Tomomi Matsumoto
- Department of Biological Sciences, University of Alberta Edmonton, AB, Canada
| | | | | | | | | |
Collapse
|
26
|
Li BH, Zhang LL, Yin YW, Pi Y, Guo L, Yang QW, Gao CY, Fang CQ, Wang JZ, Xiang J, Li JC. Association between interleukin-1α C(-889)T polymorphism and Alzheimer's disease: a meta-analysis including 12,817 subjects. J Neural Transm (Vienna) 2012; 120:497-506. [PMID: 23322030 DOI: 10.1007/s00702-012-0867-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 07/09/2012] [Indexed: 02/06/2023]
Abstract
Epidemiological studies have evaluated the association between interleukin-1 (IL-1)α C(-889)T polymorphism and Alzheimer's disease (AD), but the results remain inconclusive. This meta-analysis was, therefore, designed to clarify these controversies. Systematic searches of electronic databases Embase, PubMed, and Web of Science as well as hand searching of the references of identified articles and the meeting abstracts were performed. Statistical analyses were performed using software Review Manager (Version 5.1.2) and Stata (Version 11.0). The pooled odds ratios (ORs) with 95 % confidence intervals (95 % CIs) were calculated. A total of 28 publications including 29 studies were involved. There was a significant association between IL-1α C(-889)T polymorphism and AD (for T allele vs. C allele: OR = 1.14, 95 % CI = 1.07-1.21; for T/T vs. C/C: OR = 1.39, 95 % CI = 1.18-1.63; for dominant model: OR = 1.13, 95 % CI = 1.04-1.22; and for recessive model: OR = 1.39, 95 % CI = 1.20-1.60). Significant association was found for Asians, Caucasians, and early-onset Alzheimer's disease (EOAD) but for late-onset Alzheimer's disease (LOAD). This meta-analysis indicates that there is a significant association between IL-1α C(-889)T polymorphism and AD as well as EOAD.
Collapse
Affiliation(s)
- Bing-Hu Li
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Association of interleukin 1β polymorphisms and haplotypes with Alzheimer's disease. J Neuroimmunol 2012; 247:59-62. [PMID: 22498095 DOI: 10.1016/j.jneuroim.2012.03.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 02/07/2012] [Accepted: 03/16/2012] [Indexed: 11/21/2022]
Abstract
Our study aimed to associate IL-1β and IL-1RN polymorphisms with AD disease in comparison with elderly control group from São Paulo - Brazil. We genotyped 199 Alzheimer's disease (AD) patients, 165 elderly control and 122 young control samples, concerning VNTR (IL-1RN) and -511C>T and -31T>C (IL-1β) polymorphisms. Our findings revealed that -511C/-31T/2-repetitions VNTR haplotype had a protective effect for AD when compared to EC (p=0.005), whereas -511C/-31C/1-repetition VNTR haplotype was associated as a risk factor for AD (p=0.021). Taken together, we may suggest that there is a relevant role of IL-1 genes cluster in AD pathogenesis in this Brazilian population.
Collapse
|
28
|
Albani D, Tettamanti M, Batelli S, Polito L, Dusi S, Ateri E, Forloni G, Lucca U. Interleukin-1α, interleukin-1β and tumor necrosis factor-α genetic variants and risk of dementia in the very old: evidence from the "Monzino 80-plus" prospective study. AGE (DORDRECHT, NETHERLANDS) 2012; 34:519-526. [PMID: 21509504 PMCID: PMC3312635 DOI: 10.1007/s11357-011-9249-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 04/04/2011] [Indexed: 05/30/2023]
Abstract
The association among single nucleotide polymorphisms in inflammatory genes as interleukin-1 alpha (IL-1α), interleukin-1 beta (IL-1β) or tumor necrosis factor alpha (TNF-α) and dementia has been explored mostly in Alzheimer's disease, while few data addressing their association with dementia in very old people are available. We performed a prospective, door-to-door population-based study of 80 years or older residents in eight municipalities of Varese province, Italy (the Monzino 80-plus study). No difference was found by a cross-sectional approach comparing IL-1α rs1800587, IL-1β rs3087258 and TNF-α rs1799724 genotypic and allelic frequencies between those affected and not affected by dementia. After a 5-year follow-up, the elderly carriers of T-allele of TNF-α rs1799724 were at an increased risk of dementia (p = 0.03). This association was no more significant adjusting for the apolipoprotein E epsilon-4 allele (APOE-ε4, p = 0.26), which was an independent predictor of dementia onset (p = 0.0002). In short, in this Italian population of oldest olds, dementia was associated to the APOE-ε4 allele only.
Collapse
Affiliation(s)
- Diego Albani
- Laboratory of Biology of Neurodegenerative Disorders, Mario Negri Institute for Pharmacological Research, Milan, Italy
- Mario Negri Institute for Pharmacological Research, Via Giuseppe La Masa 19, 20156 Milan, Italy
| | - Mauro Tettamanti
- Laboratory of Geriatric Neuropsychiatry, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Sara Batelli
- Laboratory of Biology of Neurodegenerative Disorders, Mario Negri Institute for Pharmacological Research, Milan, Italy
- Gene Center, University of Munich, Munich, Germany
| | - Letizia Polito
- Laboratory of Biology of Neurodegenerative Disorders, Mario Negri Institute for Pharmacological Research, Milan, Italy
- Golgi Cenci Foundation, Abbiategrasso, Italy
| | - Sabrina Dusi
- Laboratory of Biology of Neurodegenerative Disorders, Mario Negri Institute for Pharmacological Research, Milan, Italy
- Unit of Molecular Neurogenetics, The Foundation ‘Carlo Besta’ Institute of Neurology, Milan, Italy
| | - Eleonora Ateri
- Laboratory of Biology of Neurodegenerative Disorders, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Gianluigi Forloni
- Laboratory of Biology of Neurodegenerative Disorders, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Ugo Lucca
- Laboratory of Geriatric Neuropsychiatry, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| |
Collapse
|
29
|
Ma SL, Tang NLS, Tam CWC, Lui VWC, Lam LCW, Chiu HFK, Driver JA, Pastorino L, Lu KP. A PIN1 polymorphism that prevents its suppression by AP4 associates with delayed onset of Alzheimer's disease. Neurobiol Aging 2012; 33:804-13. [PMID: 20580132 PMCID: PMC2988914 DOI: 10.1016/j.neurobiolaging.2010.05.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 05/12/2010] [Accepted: 05/16/2010] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by the presence of neurofibrillary tangles composed of tau and senile plaques of amyloid-beta peptides (Aβ) derived from amyloid precursor protein (APP). Pin1 is a unique prolyl isomerase that has been shown to protect against age-dependent neurodegeneration by acting on phosphorylated tau and APP to suppress tangle formation and amyloidogenic APP processing. Here we report a functional polymorphism, rs2287839, in the Pin1 promoter that is significantly associated with a 3-year delay in the average age at onset (AAO) of late-onset AD in a Chinese population. More significantly, the Pin1 polymorphism rs2287839 is located within the consensus binding motif for the brain-selective transcription factor, AP4 (CAGCTG) and almost completely abolishes the ability of AP4 to bind and suppress the Pin1 promoter, as shown by chromatin immunoprecipitation, electrophoretic mobility shift assay, and promoter luciferase assay. Moreover, overexpression or knockdown of AP4 resulted in an 80% reduction or 2-fold increase in endogenous Pin1 levels, respectively. Thus, AP4 is a novel transcriptional repressor of Pin1 expression and the Pin1 promoter single nucleotide polymorphism (SNP) identified in this study that prevents such suppression is associated with delayed onset of AD. These results indicate that regulation of Pin1 by AP4 plays a critical role in determining age at onset of AD and might be a novel therapeutic target to delay the onset of AD.
Collapse
Affiliation(s)
- Suk Ling Ma
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Center for Life Science, Room 0408, 3 Blackfan Circle, Boston, Massachusetts 02215, USA
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Nelson Leung Sang Tang
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Cindy Woon Chi Tam
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Victor Wing Cheong Lui
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Linda Chiu Wa Lam
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Helen Fung Kum Chiu
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jane Ann Driver
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Center for Life Science, Room 0408, 3 Blackfan Circle, Boston, Massachusetts 02215, USA
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston VA Health Care System, Boston, Massachusetts 02215, USA
| | - Lucia Pastorino
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Center for Life Science, Room 0408, 3 Blackfan Circle, Boston, Massachusetts 02215, USA
| | - Kun Ping Lu
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Center for Life Science, Room 0408, 3 Blackfan Circle, Boston, Massachusetts 02215, USA
| |
Collapse
|
30
|
Posthuma D, Meulenbelt I, de Craen AJM, de Geus EJC, Slagboom PE, Boomsma DI, Westendorp RGJ. Human Cytokine Response to ex vivo Amyloid-β Stimulation is Mediated by Genetic Factors. Twin Res Hum Genet 2012. [DOI: 10.1375/twin.8.2.132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractThrough its ability to induce the enhanced release and production of cytokines, amyloid-β is responsible for the chronic inflammatory response that contributes to Alzheimer's disease (AD). Determining whether the response of monocytes to amyloid-β stimulation is under genetic control may help understand the basis of why some people are more prone to develop neuronal degeneration than others. In the current study we investigated the heritability of the cytokine (IL-10, IL-6, IL-1β, IL-1ra, TNF-[.alpha]) production capacity upon ex vivo stimulation with amyloid-β in whole blood samples of 222 twins and 85 singleton siblings from 139 extended twin families. It was found that individual differences in amyloid-β-induced cytokine production capacity are to a large extent of genetic origin, with heritability estimates ranging from 55% (IL-1β) to 68% (IL-6). We conclude that genes influencing amyloid-β-induced cytokine response may provide clues to the progression of AD pathology.
Collapse
|
31
|
Um JY, Rim HK, Kim SJ, Kim HL, Hong SH. Functional polymorphism of IL-1 alpha and its potential role in obesity in humans and mice. PLoS One 2011; 6:e29524. [PMID: 22216303 PMCID: PMC3246492 DOI: 10.1371/journal.pone.0029524] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 11/30/2011] [Indexed: 11/19/2022] Open
Abstract
Proinflammatory cytokines secreted from adipose tissue contribute to the morbidity associated with obesity. IL-1α is one of the proinflammatory cytokines; however, it has not been clarified whether IL-1α may also cause obesity. In this study, we investigated whether polymorphisms in IL-1α contribute to human obesity. A total of 260 obese subjects were genotyped for IL-1α C-889T (rs1800587) and IL-1α G+4845T (rs17561). Analyses of genotype distributions revealed that both IL-1α polymorphisms C-889T (rs1800587) and G+4845T (rs17561) were associated with an increase in body mass index in obese healthy women. In addition, the effect of rs1800587 on the transcriptional activity of IL-1α was explored in pre-adipocyte 3T3-L1 cells. Significant difference was found between the rs1800587 polymorphism in the regulatory region of the IL-1α gene and transcriptional activity. We extended these observations in vivo to a high-fat diet-induced obese mouse model and in vitro to pre-adipocyte 3T3-L1 cells. IL-1α levels were dramatically augmented in obese mice, and triglyceride was increased 12 hours after IL-1α injection. Taken together, IL-1α treatment regulated the differentiation of preadipocytes. IL-1α C-889T (rs1800587) is a functional polymorphism of IL-1α associated with obesity. IL-1α may have a critical function in the development of obesity.
Collapse
Affiliation(s)
- Jae-Young Um
- Department of Pharmacology, College of Oriental Medicine, Institute of Oriental Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea.
| | | | | | | | | |
Collapse
|
32
|
Sardi F, Fassina L, Venturini L, Inguscio M, Guerriero F, Rolfo E, Ricevuti G. Alzheimer's disease, autoimmunity and inflammation. The good, the bad and the ugly. Autoimmun Rev 2011; 11:149-53. [DOI: 10.1016/j.autrev.2011.09.005] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 09/19/2011] [Indexed: 11/26/2022]
|
33
|
Immunological responses of astroglia in the rat brain under acute stress: interleukin 1 beta co-localized in astroglia. Neuroscience 2011; 192:429-37. [DOI: 10.1016/j.neuroscience.2011.06.051] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 06/16/2011] [Accepted: 06/17/2011] [Indexed: 11/19/2022]
|
34
|
Ma SL, Lam LCW. Panel of Genetic Variations as a Potential Non-invasive Biomarker for Early Diagnosis of Alzheimer's Disease. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2011; 9:54-66. [PMID: 23429712 PMCID: PMC3569084 DOI: 10.9758/cpn.2011.9.2.54] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/06/2011] [Accepted: 05/23/2011] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Biomarkers such as levels of amyloid beta (Aβ) in cerebrospinal fluid and ApoE genotyping were suggested for the diagnosis of AD, however, the result is either non-conclusive or with invasive procedure. Genome-wide association studies (GWASs) for AD suggested single nucleotide polymorphisms (SNPs) in many genes are associated with the risk of AD, but each only contributed with small effect to the disease. By incorporating a panel of established genetic susceptibility factors, the risk of an individual in getting AD could be better estimated. Further research will be required to reveal if adding to the current well-developed clinical diagnosis protocol, the accuracy and specificity of diagnosis of AD would be greatly improved and if this might also be beneficial in identifying pre-symptomatic AD patients for early diagnosis and intervention of the disease.
Collapse
Affiliation(s)
- Suk Ling Ma
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA. ; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | | |
Collapse
|
35
|
Déniz-Naranjo MC, Muñoz-Fernandez C, Alemany-Rodríguez MJ, Pérez-Vieitez MC, Aladro-Benito Y, Irurita-Latasa J, Sánchez-García F. Cytokine IL-1 beta but not IL-1 alpha promoter polymorphism is associated with Alzheimer disease in a population from the Canary Islands, Spain. Eur J Neurol 2008; 15:1080-4. [DOI: 10.1111/j.1468-1331.2008.02252.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
Di Bona D, Plaia A, Vasto S, Cavallone L, Lescai F, Franceschi C, Licastro F, Colonna-Romano G, Lio D, Candore G, Caruso C. Association between the interleukin-1beta polymorphisms and Alzheimer's disease: a systematic review and meta-analysis. ACTA ACUST UNITED AC 2008; 59:155-63. [PMID: 18675847 DOI: 10.1016/j.brainresrev.2008.07.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Revised: 07/04/2008] [Accepted: 07/08/2008] [Indexed: 11/26/2022]
Abstract
The pro-inflammatory cytokine interleukin(IL)-1beta is a main component in inflammatory pathways and is overexpressed in the brain of Alzheimer's disease (AD) patients. Several studies report associations between IL-1beta polymorphisms and AD, but findings from different studies are controversial. Our aim was to verify the correlation between the single nucleotide polymorphisms (SNPs) of the IL-1beta, at sites -511 and +3953, and AD by meta-analysis. Computerized bibliographic searches of PUBMED and AlzGene database (http://www.alzgene.org) were supplemented with manual searches of reference lists. There is evidence for association between IL-1beta +3953 SNP and AD, with an OR=1.60 (95% C.I.: 1.16-2.22; Z=2.83 p=0.005) for TT genotype. No significant difference in genotype distribution of the IL-1beta -511 SNP in AD was obtained, but high between-study heterogeneity was found. To reduce heterogeneity, subgroup analyses were performed using, as stratifying variables, characteristics of the population under study (age, gender, type of AD diagnosis, Mini Mental State Examination of the controls) and characteristics related to the study design (statistical power of individual studies). The frequency of the IL-1beta -511 TT genotype resulted significantly higher than other genotypes only when the Caucasian studies with the highest statistical power were included in the subgroup analysis (OR=1.32; 95% C.I.: 1.03-1.69; p=0.03), with no evidence of between-study heterogeneity. Our data support an association between the TT genotype of IL-1beta +3953 SNP and AD, and suggest a possible association of the -511 TT genotype. Unreplicability of the results seems to be due mainly to the lack of statistical power of the individual studies.
Collapse
Affiliation(s)
- Danilo Di Bona
- Istituto di Biomedicina ed Immunologia Molecolare (IBIM), CNR, Palermo Università di Palermo, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Samuelsson M, Ramberg V, Iverfeldt K. Alzheimer amyloid-β peptides block the activation of C/EBPβ and C/EBPδ in glial cells. Biochem Biophys Res Commun 2008; 370:619-22. [DOI: 10.1016/j.bbrc.2008.03.150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 03/31/2008] [Indexed: 12/31/2022]
|
38
|
Stress and glucocorticoid footprints in the brain-the path from depression to Alzheimer's disease. Neurosci Biobehav Rev 2008; 32:1161-73. [PMID: 18573532 DOI: 10.1016/j.neubiorev.2008.05.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 04/23/2008] [Accepted: 05/05/2008] [Indexed: 01/02/2023]
Abstract
Increasingly, stress is recognized as a trigger of depressive episodes and recent evidence suggests a causal role of stress in the onset and progression of Alzheimer's disease (AD) pathology. Besides aging, sex is an important determinant of prevalence rates for both AD and mood disorders. In light of a recent meta-analysis indicating that depressed subjects have a higher likelihood of developing AD, a key message in this article will be that both depression and AD are stress-related disorders and may represent a continuum that should receive more attention in future neurobiological studies. Accordingly, this review considers some of the cellular mechanisms that may be involved in regulating this transition threshold. In addition, it highlights the importance of addressing the question of how aging and sex interplay with stress to influence mood and cognition, with a bias towards consideration of neuroplastic events in particular brain regions, as the basis of AD and depressive disorders.
Collapse
|
39
|
Baune BT, Ponath G, Rothermundt M, Riess O, Funke H, Berger K. Association between genetic variants of IL-1beta, IL-6 and TNF-alpha cytokines and cognitive performance in the elderly general population of the MEMO-study. Psychoneuroendocrinology 2008; 33:68-76. [PMID: 17988804 DOI: 10.1016/j.psyneuen.2007.10.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 10/02/2007] [Accepted: 10/04/2007] [Indexed: 11/27/2022]
Abstract
This study is to investigate the associations between specific polymorphisms in three cytokine genes and domains of cognitive functioning in a population based study in the elderly. In a cross-sectional study of 369 community dwelling elderly subjects we examined the relationships between the polymorphisms IL-1beta-1418C-->T, IL-6-572G-->C and TNF-alpha-308G-->A and the cognitive function domains memory, processing speed and motor function using an extensive neuropsychological test battery. Linear regression models were used in the analysis and results adjusted for multiple comparisons. A significant association between the IL-1beta-1418C-->T polymorphism and memory performance was found with carriers of the T allele (dominant model) having worse memory performance than those with the C allele. In addition, a significant association between the TNF-alpha-308G-->A polymorphism and processing speed was observed, indicating better performance for heterozygous or homozygous carriers of the A allele. These results remained significant after adjustment for known confounders of cognitive function and additional Bonferroni correction for multiple comparisons. Our study provides first results on detrimental effects of the IL-1beta-1418C-->T polymorphism on memory performance and neuroprotective effects of the TNF-alpha-308G-->A polymorphism on processing speed in elderly individuals. Further research is needed to prospectively examine changes in cognitive performance in relation to cytokine genotypes.
Collapse
Affiliation(s)
- Bernhard T Baune
- Department of Psychiatry, School of Medicine, James Cook University, QLD 4814, Australia.
| | | | | | | | | | | |
Collapse
|
40
|
Rojo LE, Fernández JA, Maccioni AA, Jimenez JM, Maccioni RB. Neuroinflammation: implications for the pathogenesis and molecular diagnosis of Alzheimer's disease. Arch Med Res 2007; 39:1-16. [PMID: 18067990 DOI: 10.1016/j.arcmed.2007.10.001] [Citation(s) in RCA: 252] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2007] [Accepted: 05/31/2007] [Indexed: 12/27/2022]
Abstract
During the past few years, an increasing set of evidence has supported the major role of deregulation of the interaction patterns between glial cells and neurons in the pathway toward neuronal degeneration. Neurons and glial cells, together with brain vessels, constitute an integrated system for brain function. Inflammation is a process related with the onset of several neurodegenerative disorders, including Alzheimer's disease (AD). Several hypotheses have been postulated to explain the pathogenesis of AD, but none provides insight into the early events that trigger metabolic and cellular alterations in neuronal degeneration. The amyloid hypothesis was sustained on the basis that Abeta-peptide deposition into senile plaques is responsible for neurodegeneration. However, recent findings point to Abeta oligomers as responsible for synaptic impairment in neuronal degeneration. Amyloid is only one among many other major factors affecting the quality of neuronal cells. Another explanation derives from the tau hypothesis, supported by the observations that tau hyperphosphorylations constitute a common feature of most of the altered signaling pathways in degenerating neurons. Altered tau patterns have been detected in the cerebrospinal fluids of AD patients, and a close correlation was observed between the levels of hyperphosphorylated tau isoforms and the degree of cognitive impairment. On the other hand, the anomalous effects of cytokines and trophic factors share in common the activation of tau hyperphosphorylation patterns. In this context, a neuroimmunological approach to AD becomes relevant. When glial cells that normally provide neurotrophic factors essential for neurogenesis are activated by a set of stressing events, they overproduce cytokines and NGF, thus triggering altered signaling patterns in the etiopathogenesis of AD. A solid set of discoveries has strengthened the idea that altered patterns in the glia-neuron interactions constitute early molecular events within the cascade of cellular signals that lead to neurodegeneration in AD. A direct correlation has been established between the Abeta-induced neurodegeneration and cytokine production and its subsequent release. In effect, neuroinflammation is responsible for an abnormal secretion of proinflammatory cytokines that trigger signaling pathways that activate brain tau hyperphosphorylation in residues that are not modified under normal physiological conditions. Other cytokines such as IL-3 and TNF-alpha seem to display neuroprotective activities. Elucidation of the events that control the transitions from neuroprotection to neurodegeneration should be a critical point toward elucidation of AD pathogenesis.
Collapse
Affiliation(s)
- Leonel E Rojo
- Laboratory of Cellular and Molecular Neurosciences, Faculty of Sciences, University of Chile, Santiago, Chile
| | | | | | | | | |
Collapse
|
41
|
Leonard BE. Inflammation, Depression and Dementia: Are they Connected? Neurochem Res 2007; 32:1749-56. [PMID: 17705097 DOI: 10.1007/s11064-007-9385-y] [Citation(s) in RCA: 256] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 05/10/2007] [Indexed: 01/18/2023]
Abstract
Chronic inflammation is now considered to be central to the pathogenesis not only of such medical disorders as cardiovascular disease, multiple sclerosis, diabetes and cancer but also of major depression. If chronic inflammatory changes are a common feature of depression, this could predispose depressed patients to neurodegenerative changes in later life. Indeed there is now clinical evidence that depression is a common antecedent of Alzheimer's disease and may be an early manifestation of dementia before the cognitive declines becomes apparent. This review summarises the evidence that links chronic low grade inflammation with changes in brain structure that could precipitate neurodegenerative changes associated with Alzheimer's disease and other dementias. For example, neuronal loss is a common feature of major depression and dementia. It is hypothesised that the progress from depression to dementia could result from the activation of macrophages in the blood, and microglia in the brain, that release pro-inflammatory cytokines. Such cytokines stimulate a cascade of inflammatory changes (such as an increase in prostaglandin E2, nitric oxide in addition to more pro-inflammatory cytokines) and a hypersecretion of cortisol. The latter steroid inhibits protein synthesis thereby reducing the synthesis of neurotrophic factors and preventing reairto damages neuronal networks. In addition, neurotoxic end products of the tryptophan-kynurenine pathway, such as quinolinic acid, accumulate in astrocytes and neurons in both depression and dementia. Thus increased neurodegeneration, reduced neuroprotection and neuronal repair are common pathological features of major depression and dementia. Such changes may help to explain why major depression is a frequent prelude to dementia in later life.
Collapse
Affiliation(s)
- Brian E Leonard
- Department of Psychiatry and Neuropsychology, Brain and Behaviour Research Institute, University of Maastricht, Maastricht, The Netherlands.
| |
Collapse
|
42
|
Harris SE, Fox H, Wright AF, Hayward C, Starr JM, Whalley LJ, Deary IJ. A genetic association analysis of cognitive ability and cognitive ageing using 325 markers for 109 genes associated with oxidative stress or cognition. BMC Genet 2007; 8:43. [PMID: 17601350 PMCID: PMC1933580 DOI: 10.1186/1471-2156-8-43] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 07/02/2007] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Non-pathological cognitive ageing is a distressing condition affecting an increasing number of people in our 'ageing society'. Oxidative stress is hypothesised to have a major role in cellular ageing, including brain ageing. RESULTS Associations between cognitive ageing and 325 single nucleotide polymorphisms (SNPs), located in 109 genes implicated in oxidative stress and/or cognition, were examined in a unique cohort of relatively healthy older people, on whom we have cognitive ability scores at ages 11 and 79 years (LBC1921). SNPs showing a significant positive association were then genotyped in a second cohort for whom we have cognitive ability scores at the ages of 11 and 64 years (ABC1936). An intronic SNP in the APP gene (rs2830102) was significantly associated with cognitive ageing in both LBC1921 and a combined LBC1921/ABC1936 analysis (p < 0.01), but not in ABC1936 alone. CONCLUSION This study suggests a possible role for APP in normal cognitive ageing, in addition to its role in Alzheimer's disease.
Collapse
Affiliation(s)
- Sarah E Harris
- Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, UK
| | - Helen Fox
- Department of Mental Health, University of Aberdeen, Clinical Research Centre, Royal Cornhill Hospital, Cornhill Road, Aberdeen, AB25 2ZH, UK
| | - Alan F Wright
- Medical Research Council Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - John M Starr
- Department of Geriatric Medicine, University of Edinburgh, Royal Victoria Hospital, Craigleith Road, Edinburgh EH4 2DN, UK
| | - Lawrence J Whalley
- Department of Mental Health, University of Aberdeen, Clinical Research Centre, Royal Cornhill Hospital, Cornhill Road, Aberdeen, AB25 2ZH, UK
| | - Ian J Deary
- Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, UK
| |
Collapse
|
43
|
Arra M, Emanuele E, Martinelli V, Minoretti P, Bertona M, Geroldi D. The M694V variant of the familial Mediterranean fever gene is associated with sporadic early-onset Alzheimer's disease in an Italian population sample. Dement Geriatr Cogn Disord 2007; 23:55-9. [PMID: 17090974 DOI: 10.1159/000096743] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2006] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Inflammation is deemed to play a crucial role in the pathogenesis of Alzheimer's disease (AD). We sought to determine whether the proinflammatory M694V mutation of pyrin, the gene responsible for familial Mediterranean fever, could lead to an increased risk for AD. METHODS We compared the M694V variant genotypes in 378 sporadic AD patients and 384 healthy control subjects of Italian descent. RESULTS After adjustment for potential confounders, the M694V mutation was found to be associated with an increased risk for AD in subjects with an age at onset of 65 years or younger (multivariate-adjusted odds ratio, OR: 3.01, 95% confidence interval, CI: 1.24-6.72, p = 0.021), but not in patients with an age at onset older than 65 years (multivariate-adjusted OR: 0.81, 95% CI: 0.34-1.99, p = 0.847). Kaplan-Meier analysis indicated that AD patients bearing the M694V mutation presented with disease onset 7 years earlier than carriers of the wild-type genotype (log rank = 41.61, p < 0.001). CONCLUSION Our data indicate that the M694V sequence variant in the pyrin gene might influence the age at onset of AD in the Italian population.
Collapse
Affiliation(s)
- Mariarosa Arra
- Interdepartmental Center for Research in Molecular Medicine, University of Pavia, Pavia, Italy
| | | | | | | | | | | |
Collapse
|
44
|
Marcos-Carcavilla A, Calvo JH, González C, Moazami-Goudarzi K, Laurent P, Bertaud M, Hayes H, Beattie AE, Serrano C, Lyahyai J, Martín-Burriel I, Alves E, Zaragoza P, Badiola JJ, Serrano M. IL-1 family members as candidate genes modulating scrapie susceptibility in sheep: localization, partial characterization, and expression. Mamm Genome 2007; 18:53-63. [PMID: 17242860 DOI: 10.1007/s00335-006-0095-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Accepted: 10/30/2006] [Indexed: 01/08/2023]
Abstract
Scrapie (SC) is a transmissible spongiform encephalopathy (TSE) in sheep and goats. Susceptibility to this neurodegenerative disease is controlled mainly by point mutations at the PRNP locus. Other genes, apart from PRNP, have been reported to modulate resistance/susceptibility to SC. On the basis of several studies on Alzheimer's disease and different TSE models, and of requirement for correct homeostasis of cytokines in brain, IL1B and IL1RN were chosen as putative positional and functional candidate genes that might be involved in the polygenic variance mentioned above. In the present work, ovine IL1B and IL1RN genes were partially isolated and characterized, including promoter and other regulatory regions. In addition, several sequence polymorphisms were identified. Furthermore, their cytogenetic positions on sheep chromosomes were determined by FISH and confirmed by linkage analysis, localizing both genes in OAR3p22, a region previously described as carrying a QTL for SC incubation period in sheep. Finally, expression analyses were carried out in eight naturally SC-infected and five uninfected sheep with the same genotype for PRNP (ARQ/ARQ). This comparison was performed using real-time RT-PCR in samples of spleen and cerebellum. Results showed differences in the expression of both cytokines in cerebellum (p < 0.05) but not in spleen (p > 0.05).
Collapse
Affiliation(s)
- Ane Marcos-Carcavilla
- Departamento de Mejora Genética Animal, INIA, Ctra La Coruña Km 7.5, 28040, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Konsman JP, Drukarch B, Van Dam AM. (Peri)vascular production and action of pro-inflammatory cytokines in brain pathology. Clin Sci (Lond) 2006; 112:1-25. [PMID: 17132137 DOI: 10.1042/cs20060043] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In response to tissue injury or infection, the peripheral tissue macrophage induces an inflammatory response through the release of IL-1β (interleukin-1β) and TNFα (tumour necrosis factor α). These cytokines stimulate macrophages and endothelial cells to express chemokines and adhesion molecules that attract leucocytes into the peripheral site of injury or infection. The aims of the present review are to (i) discuss the relevance of brain (peri)vascular cells and compartments to bacterial meningitis, HIV-1-associated dementia, multiple sclerosis, ischaemic and traumatic brain injury, and Alzheimer's disease, and (ii) to provide an overview of the production and action of pro-inflammatory cytokines by (peri)vascular cells in these pathologies of the CNS (central nervous system). The brain (peri)vascular compartments are highly relevant to pathologies affecting the CNS, as infections are almost exclusively blood-borne. Insults disrupt blood and energy flow to neurons, and active brain-to-blood transport mechanisms, which are the bottleneck in the clearance of unwanted molecules from the brain. Perivascular macrophages are the most reactive cell type and produce IL-1β and TNFα after infection or injury to the CNS. The main cellular target for IL-1β and TNFα produced in the brain (peri)vascular compartment is the endothelium, where these cytokines induce the expression of adhesion molecules and promote leucocyte infiltration. Whether this and other effects of IL-1 and TNF in the brain (peri)vascular compartments are detrimental or beneficial in neuropathology remains to be shown and requires a clear understanding of the role of these cytokines in both damaging and repair processes in the CNS.
Collapse
Affiliation(s)
- Jan P Konsman
- Laboratory of Integrative Neurobiology, CNRS FRE 2723/INRA UR 1244/University Bordeaux2, Institut François Magendie, Bordeaux, France
| | | | | |
Collapse
|
46
|
Abstract
The neuropathology of Alzheimer's disease (AD) is characterized by deposits of amyloid beta (Abeta) peptides and neurofibrillary tangles, but also, among other aspects, by signs of a chronic inflammatory process. Epidemiological studies have shown that long-term use of nonsteroidal antiinflammatory drugs (NSAIDs) reduces the risk of developing AD and delays its onset. The classic target of NSAIDs is the prevention of cyclooxygenase (COX) activation. The main mechanism of action of COXs is the synthesis of prostaglandins, some of which have potent inflammatory activity. The discovery of two isoforms of this enzyme, COX-1 and COX-2, and that the latter is inducible by inflammatory cytokines supported the hypothesis that its inhibition would result in a potent antiinflammatory effect and led to the rapid development of selective COX-2 inhibitors, collectively called coxibs. Based on this rationale, some coxibs have been used in clinical trials for AD patients, but all the results obtained so far have been negative. Here, we review our knowledge in terms of COX-2 in the central nervous system, COX-2 and Abeta formation, and finally COX-2 and AD pathogenesis to understand the reasons why these drugs have failed and whether there is any scientific support to keep them as therapeutic tools for this chronic disease.
Collapse
Affiliation(s)
- Omidreza Firuzi
- Department of Pharmacology, University of Pennsylvania, School of Medicine. Philadelphia, PA 19014, USA
| | | |
Collapse
|
47
|
Weisman D, Hakimian E, Ho GJ. Interleukins, inflammation, and mechanisms of Alzheimer's disease. VITAMINS AND HORMONES 2006; 74:505-30. [PMID: 17027528 DOI: 10.1016/s0083-6729(06)74020-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Alzheimer's disease (AD) is the most common progressive neurodegenerative form of dementia in the elderly and is characterized neuropathologically by neurofibrillary tangles (NFT), amyloid neuritic plaques (NP), and prominent synaptic and eventually neuronal loss. Although the molecular basis of AD is not clearly understood, a neuroinflammatory process, triggered by Abeta42, plays a central role in the neurodegenerative process. This inflammatory process is driven by activated microglia, astrocytes and the induction of proinflammatory molecules and related signaling pathways, leading to both synaptic and neuronal damage as well as further inflammatory cell activation. Epidemiologic data as well as clinical trial evidence suggest that nonsteroidal anti-inflammatory drug (NSAID) use may decrease the incidence of AD, further supporting a role for inflammation in AD pathogenesis. Although the precise molecular and cellular relationship between AD and inflammation remains unclear, interleukins and cytokines might induce activation of signaling pathways leading to futher inflammation and neuronal injury. This chapter will discuss the association between interleukins and neurodegeneration in AD and highlight the significance of genetic and clinical aspects of interleukins in disease expression and progression. As part of an emerging inflammatory signaling network underlying AD pathogenesis, beta-amyloid (Abeta) stimulates the glial and microglial production of interleukins and other cytokines, leading to an ongoing inflammatory cascade and contributing to synaptic dysfunction and loss, and later, neuronal death. Inflammatory pathways involving interleukin and cytokine signaling might suggest potential targets for intervention and influence the development of novel therapies to circumvent synaptic and neuronal dysfunction ultimately leading to AD neurodegeneration.
Collapse
Affiliation(s)
- David Weisman
- Department of Neurosciences and the Alzheimer's Disease Research Center, University of California, San Diego, California 92093, USA
| | | | | |
Collapse
|
48
|
Walker DG, Lue LF. Investigations with cultured human microglia on pathogenic mechanisms of Alzheimer's disease and other neurodegenerative diseases. J Neurosci Res 2005; 81:412-25. [PMID: 15957156 DOI: 10.1002/jnr.20484] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Inflammation-mediated mechanisms for human neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) have evolved from being on the fringe of medical hypotheses to mainstream thinking. Pioneering immunopathology studies with human brain tissues identified microglia associated with neuropathologic hallmarks of these diseases. As activated macrophages were known to produce many potential toxic products, this gave rise to the hypothesis that activated microglia (brain resident macrophages) could be contributing to the degeneration of key target neurons in these diseases, as well as potential vascular dysfunction. Studies with microglia derived from different sources, including human brains, have confirmed that activated microglia can mediate neuronal cell death. Based on these theories, a number of human clinical trials with antiinflammatory agents have been carried out on AD patients. Results to date have indicated a lack of effectiveness at slowing disease progression and have begun to cast doubt on the significance of inflammation in AD. It has been shown recently that activating microglia through immunization of amyloid plaque-developing mice with amyloid beta peptide (Abeta) has promise as a therapeutic strategy and despite some setbacks, has potential as a treatment for AD patients. This article will consider experimental data with microglia to determine whether the additional targets need to be investigated. The use of human microglia cultures, in particular those derived from elderly diseased human brains, offers an experimental system that can closely model the cell type activated in human neurodegenerative diseases. Experimental data produced by our laboratory and others is reviewed to determine the contribution of this unique experimental model to understanding disease mechanisms and possibly discovering new therapeutic targets.
Collapse
Affiliation(s)
- D G Walker
- Laboratory of Neuroinflammation, Sun Health Research Institute, Sun City, Arizona 85351, USA.
| | | |
Collapse
|
49
|
Tuppo EE, Arias HR. The role of inflammation in Alzheimer's disease. Int J Biochem Cell Biol 2005; 37:289-305. [PMID: 15474976 DOI: 10.1016/j.biocel.2004.07.009] [Citation(s) in RCA: 494] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2004] [Indexed: 11/17/2022]
Abstract
Considerable evidence gained over the past decade has supported the conclusion that neuroinflammation is associated with Alzheimer's disease (AD) pathology. Inflammatory components related to AD neuroinflammation include brain cells such as microglia and astrocytes, the classic and alternate pathways of the complement system, the pentraxin acute-phase proteins, neuronal-type nicotinic acetylcholine receptors (AChRs), peroxisomal proliferators-activated receptors (PPARs), as well as cytokines and chemokines. Both the microglia and astrocytes have been shown to generate beta-amyloid protein (Abeta), one of the main pathologic features of AD. Abeta itself has been shown to act as a pro-inflammatory agent causing the activation of many of the inflammatory components. Further substantiation for the role of neuroinflammation in AD has come from studies that demonstrate patients who took non-steroidal anti-inflammatory drugs had a lower risk of AD than those who did not. These same results have led to increased interest in pursuing anti-inflammatory therapy for AD but with poor results. On the other hand, increasing amount of data suggest that AChRs and PPARs are involved in AD-induced neuroinflammation and in this regard, future therapy may focus on their specific targeting in the AD brain.
Collapse
Affiliation(s)
- Ehab E Tuppo
- Center for Aging, University of Medicine and Dentistry of New Jersey-School of Osteopathic Medicine, Stratford, NJ 08084, USA.
| | | |
Collapse
|
50
|
Patel NS, Paris D, Mathura V, Quadros AN, Crawford FC, Mullan MJ. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer's disease. J Neuroinflammation 2005; 2:9. [PMID: 15762998 PMCID: PMC555557 DOI: 10.1186/1742-2094-2-9] [Citation(s) in RCA: 223] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2005] [Accepted: 03/11/2005] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND: Inflammation is believed to play an important role in the pathology of Alzheimer's disease (AD) and cytokine production is a key pathologic event in the progression of inflammatory cascades. The current study characterizes the cytokine expression profile in the brain of two transgenic mouse models of AD (TgAPPsw and PS1/APPsw) and explores the correlations between cytokine production and the level of soluble and insoluble forms of Abeta. METHODS: Organotypic brain slice cultures from 15-month-old mice (TgAPPsw, PS1/APPsw and control littermates) were established and multiple cytokine levels were analyzed using the Bio-plex multiple cytokine assay system. Soluble and insoluble forms of Abeta were quantified and Abeta-cytokine relationships were analyzed. RESULTS: Compared to control littermates, transgenic mice showed a significant increase in the following pro-inflammatory cytokines: TNF-alpha, IL-6, IL-12p40, IL-1beta, IL-1alpha and GM-CSF. TNF-alpha, IL-6, IL-1alpha and GM-CSF showed a sequential increase from control to TgAPPsw to PS1/APPsw suggesting that the amplitude of this cytokine response is dependent on brain Abeta levels, since PS1/APPsw mouse brains accumulate more Abeta than TgAPPsw mouse brains. Quantification of Abeta levels in the same slices showed a wide range of Abeta soluble:insoluble ratio values across TgAPPsw and PS1/APPsw brain slices. Abeta-cytokine correlations revealed significant relationships between Abeta1-40, 1-42 (both soluble and insoluble) and all the above cytokines that changed in the brain slices. CONCLUSION: Our data confirm that the brains of transgenic APPsw and PS1/APPsw mice are under an active inflammatory stress, and that the levels of particular cytokines may be directly related to the amount of soluble and insoluble Abeta present in the brain suggesting that pathological accumulation of Abeta is a key driver of the neuroinflammatory response.
Collapse
Affiliation(s)
- Nikunj S Patel
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL34243, USA
| | - Daniel Paris
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL34243, USA
| | | | - Amita N Quadros
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL34243, USA
| | - Fiona C Crawford
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL34243, USA
| | - Michael J Mullan
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL34243, USA
| |
Collapse
|