1
|
Sullivan MA, Lane SD, McKenzie ADJ, Ball SR, Sunde M, Neely GG, Moreno CL, Maximova A, Werry EL, Kassiou M. iPSC-derived PSEN2 (N141I) astrocytes and microglia exhibit a primed inflammatory phenotype. J Neuroinflammation 2024; 21:7. [PMID: 38178159 PMCID: PMC10765839 DOI: 10.1186/s12974-023-02951-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/07/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Widescale evidence points to the involvement of glia and immune pathways in the progression of Alzheimer's disease (AD). AD-associated iPSC-derived glial cells show a diverse range of AD-related phenotypic states encompassing cytokine/chemokine release, phagocytosis and morphological profiles, but to date studies are limited to cells derived from PSEN1, APOE and APP mutations or sporadic patients. The aim of the current study was to successfully differentiate iPSC-derived microglia and astrocytes from patients harbouring an AD-causative PSEN2 (N141I) mutation and characterise the inflammatory and morphological profile of these cells. METHODS iPSCs from three healthy control individuals and three familial AD patients harbouring a heterozygous PSEN2 (N141I) mutation were used to derive astrocytes and microglia-like cells and cell identity and morphology were characterised through immunofluorescent microscopy. Cellular characterisation involved the stimulation of these cells by LPS and Aβ42 and analysis of cytokine/chemokine release was conducted through ELISAs and multi-cytokine arrays. The phagocytic capacity of these cells was then indexed by the uptake of fluorescently-labelled fibrillar Aβ42. RESULTS AD-derived astrocytes and microglia-like cells exhibited an atrophied and less complex morphological appearance than healthy controls. AD-derived astrocytes showed increased basal expression of GFAP, S100β and increased secretion and phagocytosis of Aβ42 while AD-derived microglia-like cells showed decreased IL-8 secretion compared to healthy controls. Upon immunological challenge AD-derived astrocytes and microglia-like cells showed exaggerated secretion of the pro-inflammatory IL-6, CXCL1, ICAM-1 and IL-8 from astrocytes and IL-18 and MIF from microglia. CONCLUSION Our study showed, for the first time, the differentiation and characterisation of iPSC-derived astrocytes and microglia-like cells harbouring a PSEN2 (N141I) mutation. PSEN2 (N141I)-mutant astrocytes and microglia-like cells presented with a 'primed' phenotype characterised by reduced morphological complexity, exaggerated pro-inflammatory cytokine secretion and altered Aβ42 production and phagocytosis.
Collapse
Affiliation(s)
- Michael A Sullivan
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Samuel D Lane
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - André D J McKenzie
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Sarah R Ball
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Margaret Sunde
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - G Gregory Neely
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camperdown, Australia
| | - Cesar L Moreno
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camperdown, Australia
| | - Alexandra Maximova
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Eryn L Werry
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
- School of Chemistry, The Faculty of Science, The University of Sydney, Camperdown, Australia.
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
| | - Michael Kassiou
- School of Chemistry, The Faculty of Science, The University of Sydney, Camperdown, Australia.
| |
Collapse
|
2
|
Nystuen KL, McNamee SM, Akula M, Holton KM, DeAngelis MM, Haider NB. Alzheimer's Disease: Models and Molecular Mechanisms Informing Disease and Treatments. Bioengineering (Basel) 2024; 11:45. [PMID: 38247923 PMCID: PMC10813760 DOI: 10.3390/bioengineering11010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex neurodegenerative disease resulting in progressive loss of memory, language and motor abilities caused by cortical and hippocampal degeneration. This review captures the landscape of understanding of AD pathology, diagnostics, and current therapies. Two major mechanisms direct AD pathology: (1) accumulation of amyloid β (Aβ) plaque and (2) tau-derived neurofibrillary tangles (NFT). The most common variants in the Aβ pathway in APP, PSEN1, and PSEN2 are largely responsible for early-onset AD (EOAD), while MAPT, APOE, TREM2 and ABCA7 have a modifying effect on late-onset AD (LOAD). More recent studies implicate chaperone proteins and Aβ degrading proteins in AD. Several tests, such as cognitive function, brain imaging, and cerebral spinal fluid (CSF) and blood tests, are used for AD diagnosis. Additionally, several biomarkers seem to have a unique AD specific combination of expression and could potentially be used in improved, less invasive diagnostics. In addition to genetic perturbations, environmental influences, such as altered gut microbiome signatures, affect AD. Effective AD treatments have been challenging to develop. Currently, there are several FDA approved drugs (cholinesterase inhibitors, Aß-targeting antibodies and an NMDA antagonist) that could mitigate AD rate of decline and symptoms of distress.
Collapse
Affiliation(s)
- Kaden L. Nystuen
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Shannon M. McNamee
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Monica Akula
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Kristina M. Holton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Margaret M. DeAngelis
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Neena B. Haider
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Sun W, Feng Y, Li H, He X, Lu Y, Shan Z, Teng W, Li J. The effects of maternal anti-alpha-enolase antibody expression on the brain development in offspring. Clin Exp Immunol 2022; 210:187-198. [PMID: 36149061 PMCID: PMC9750830 DOI: 10.1093/cei/uxac086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/25/2022] [Accepted: 09/21/2022] [Indexed: 01/25/2023] Open
Abstract
Anti-alpha-enolase autoantibodies have not only been found to play an important role in autoimmune diseases but also cause neurological damage in adults. In this study, a pregnant mouse model with high serum alpha-enolase (ENO1)-specific antibody (ENO1Ab) was established by immunization with ENO1 protein to explore the effects of maternal circulatory ENO1Ab on the brain development in offspring. The pups showed impaired learning and memory abilities with obviously thinner tight junctions in the brain tissue. IgG deposits colocalized with both ENO1 protein and complement 3 (C3), and the membrane attack complex was obviously detectable in the brain tissues of pups from dams with high serum ENO1Ab expression. Our findings suggest that highly expressed ENO1Ab in the maternal circulation can pass through the blood-placenta-barrier and the compromised blood-brain barrier into the brain tissues of offspring and may cause neurological development impairment mainly through complement-dependent cytotoxicity.
Collapse
Affiliation(s)
- Wei Sun
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Yan Feng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Hui Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Xiaoqing He
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Yihan Lu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| | - Jing Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang110001, PR China
| |
Collapse
|
4
|
Fan M, Li Z, Hu M, Zhao H, Wang T, Jia Y, Yang R, Wang S, Song J, Liu Y, Jin W. Formononetin attenuates Aβ 25-35-induced adhesion molecules in HBMECs via Nrf2 activation. Brain Res Bull 2022; 183:162-171. [PMID: 35304289 DOI: 10.1016/j.brainresbull.2022.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/21/2022] [Accepted: 03/12/2022] [Indexed: 01/14/2023]
Abstract
Brain vascular inflammation plays a crucial role in the pathogenesis of Alzheimer's disease (AD). As a central pathogenic factor in AD, the extracellular buildup of amyloid-β (Aβ) induces brain microvascular endothelial cells activation, impairs endothelial structure and function. Formononetin (FMN) has been reported to protect against Alzheimer's disease (AD) and attenuates vascular inflammation in atherosclerosis. However, its involvement in regulating vascular inflammation of AD has not been investigated. In the study, we found that FMN significantly attenuates Aβ25-35-induced expression of adhesion molecules, including intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in the human brain microvascular endothelial cells (HBMECs), suggesting that FMN inhibits Aβ25-35-induced brain endothelial cells inflammatory response. Moreover, we observed that FMN attenuates Aβ25-35-induced translocation of NFκB (p65) into the nucleus of HBMECs, and found that FMN treatment induces Nrf2 expression and attenuates Nrf2-Keap1 association in a dose-dependent manner in HBMECs. Furthermore, we demonstrated that Nrf2 silencing significantly attenuates FMN-reduced NFκB (p65) activation and nuclear translocation. Lastly, our results showed that FMN treatment attenuates Aβ25-35-induced adhesion of THP-1 cell to endothelial cell monolayer. Collectively, these findings suggest that FMN attenuates Aβ25-35-induced activation in human brain microvascular endothelial cells, which at least in part was mediated through Nrf2 pathways.
Collapse
Affiliation(s)
- Mingyue Fan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Zhe Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Ming Hu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Haifeng Zhao
- Department of Anesthesiology, Shijiazhuang Obstetrics and Gynecology Hospital, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, P.R. China
| | - Tianjun Wang
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Yanqiu Jia
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Rui Yang
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Shuo Wang
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Jiaxi Song
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Yang Liu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Wei Jin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China.
| |
Collapse
|
5
|
Aso Y, Kimura N, Matsubara E. Novel Serum Biomarkers of Neurovascular Unit Associated with Cortical Amyloid Deposition. J Alzheimers Dis 2021; 84:905-914. [PMID: 34602488 DOI: 10.3233/jad-215135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Whether blood biomarkers of neurovascular unit are associated with cortical amyloid deposition on positron emission tomography (PET) imaging remains unclear. OBJECTIVE To investigate the association between novel serum biomarkers of neurovascular unit, such as protein tyrosine phosphatase receptor type B (PTPRB), gap junction protein alpha-5 (GJA5), adenosine triphosphate-sensitive inward rectifier potassium channel-8 (KCNJ8), and von Willebrand factor (vWF), and cortical amyloid deposition. METHODS Between 2012 and 2018, 68 elderly individuals with amnestic mild cognitive impairment (32 men and 36 women; mean age 75.2 years) were enrolled. All participants underwent 11C-Pittsburgh compound-B (PiB)-PET, 18F-fluorodeoxyglucose-PET, and measurement of serum PTPRB, GJA5, KCNJ8, and vWF levels using commercially available human enzyme-linked immunosorbent assay kits. Based on the mean cortical standardized uptake value ratio, the participants were divided into two groups: PiB-negative group and PiB-positive group. Serum levels of PTPRB, GJA5, KCNJ8, and vWF were compared between the two groups. Multiple linear regression analysis was performed to investigate the relationship between serum PTPRB, GJA5, KCNJ8, and vWF levels and cortical amyloid deposition. RESULTS PTPRB and GJA5 levels were significantly lower and KCNJ8 and vWF levels were significantly higher in the PiB-positive group than in the PiB-negative group. PTPRB and GJA5 levels inversely correlated with mean PiB uptake, whereas KCNJ8 and vWF levels positively correlated with mean PiB uptake. CONCLUSION Serum levels of PTPRB, GJA5, KCNJ8, and vWF correlate with cortical amyloid deposition. These novel blood biomarkers of neurovascular unit are useful for identifying elderly individuals at risk of developing Alzheimer's disease.
Collapse
Affiliation(s)
- Yasuhiro Aso
- Department of Neurology, Oita University, Faculty of Medicine, Yufu, Oita, Japan
| | - Noriyuki Kimura
- Department of Neurology, Oita University, Faculty of Medicine, Yufu, Oita, Japan
| | - Etsuro Matsubara
- Department of Neurology, Oita University, Faculty of Medicine, Yufu, Oita, Japan
| |
Collapse
|
6
|
Ruffini N, Klingenberg S, Schweiger S, Gerber S. Common Factors in Neurodegeneration: A Meta-Study Revealing Shared Patterns on a Multi-Omics Scale. Cells 2020; 9:E2642. [PMID: 33302607 PMCID: PMC7764447 DOI: 10.3390/cells9122642] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are heterogeneous, progressive diseases with frequently overlapping symptoms characterized by a loss of neurons. Studies have suggested relations between neurodegenerative diseases for many years (e.g., regarding the aggregation of toxic proteins or triggering endogenous cell death pathways). We gathered publicly available genomic, transcriptomic, and proteomic data from 177 studies and more than one million patients to detect shared genetic patterns between the neurodegenerative diseases on three analyzed omics-layers. The results show a remarkably high number of shared differentially expressed genes between the transcriptomic and proteomic levels for all conditions, while showing a significant relation between genomic and proteomic data between AD and PD and AD and ALS. We identified a set of 139 genes being differentially expressed in several transcriptomic experiments of all four diseases. These 139 genes showed overrepresented gene ontology (GO) Terms involved in the development of neurodegeneration, such as response to heat and hypoxia, positive regulation of cytokines and angiogenesis, and RNA catabolic process. Furthermore, the four analyzed neurodegenerative diseases (NDDs) were clustered by their mean direction of regulation throughout all transcriptomic studies for this set of 139 genes, with the closest relation regarding this common gene set seen between AD and HD. GO-Term and pathway analysis of the proteomic overlap led to biological processes (BPs), related to protein folding and humoral immune response. Taken together, we could confirm the existence of many relations between Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis on transcriptomic and proteomic levels by analyzing the pathways and GO-Terms arising in these intersections. The significance of the connection and the striking relation of the results to processes leading to neurodegeneration between the transcriptomic and proteomic data for all four analyzed neurodegenerative diseases showed that exploring many studies simultaneously, including multiple omics-layers of different neurodegenerative diseases simultaneously, holds new relevant insights that do not emerge from analyzing these data separately. Furthermore, the results shed light on processes like the humoral immune response that have previously been described only for certain diseases. Our data therefore suggest human patients with neurodegenerative diseases should be addressed as complex biological systems by integrating multiple underlying data sources.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
- Leibniz Institute for Resilience Research, Leibniz Association, Wallstraße 7, 55122 Mainz, Germany
| | - Susanne Klingenberg
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susann Schweiger
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susanne Gerber
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| |
Collapse
|
7
|
Chiarini A, Armato U, Hu P, Dal Prà I. CaSR Antagonist (Calcilytic) NPS 2143 Hinders the Release of Neuroinflammatory IL-6, Soluble ICAM-1, RANTES, and MCP-2 from Aβ-Exposed Human Cortical Astrocytes. Cells 2020; 9:cells9061386. [PMID: 32498476 PMCID: PMC7349863 DOI: 10.3390/cells9061386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023] Open
Abstract
Available evidence shows that human cortical neurons’ and astrocytes’ calcium-sensing receptors (CaSRs) bind Amyloid-beta (Aβ) oligomers triggering the overproduction/oversecretion of several Alzheimer’s disease (AD) neurotoxins—effects calcilytics suppress. We asked whether Aβ•CaSR signaling might also play a direct pro-neuroinflammatory role in AD. Cortical nontumorigenic adult human astrocytes (NAHAs) in vitro were untreated (controls) or treated with Aβ25–35 ± NPS 2143 (a calcilytic) and any proinflammatory agent in their protein lysates and growth media assayed via antibody arrays, enzyme-linked immunosorbent assays (ELISAs), and immunoblots. Results show Aβ•CaSR signaling upregulated the synthesis and release/shedding of proinflammatory interleukin (IL)-6, intercellular adhesion molecule-1 (ICAM-1) (holoprotein and soluble [s] fragment), Regulated upon Activation, normal T cell Expressed and presumably Secreted (RANTES), and monocyte chemotactic protein (MCP)-2. Adding NPS 2143 (i) totally suppressed IL-6′s oversecretion while remarkably reducing the other agents’ over-release; and (ii) more effectively than Aβ alone increased over controls the four agents’ distinctive intracellular accumulation. Conversely, NPS 2143 did not alter Aβ-induced surges in IL-1β, IL-3, IL-8, and IL-16 secretion, consequently revealing their Aβ•CaSR signaling-independence. Finally, Aβ25–35 ± NPS 2143 treatments left unchanged MCP-1′s and TIMP-2′s basal expression. Thus, NAHAs Aβ•CaSR signaling drove four proinflammatory agents’ over-release that NPS 2143 curtailed. Therefore, calcilytics would also abate NAHAs’ Aβ•CaSR signaling direct impact on AD’s neuroinflammation.
Collapse
Affiliation(s)
- Anna Chiarini
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics and Gynecology, Medical School, University of Verona, Veneto, 37134 Verona, Italy; (U.A.); (P.H.)
- Correspondence: (A.C.); (I.D.P.); Tel.: +39-045-802-7646 (A.C.); +39-045-802-7161 (I.D.P)
| | - Ubaldo Armato
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics and Gynecology, Medical School, University of Verona, Veneto, 37134 Verona, Italy; (U.A.); (P.H.)
- Burns Department, Shenzhen Second People’s Hospital, University of Shenzhen, Shenzhen 518000, China
| | - Peng Hu
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics and Gynecology, Medical School, University of Verona, Veneto, 37134 Verona, Italy; (U.A.); (P.H.)
| | - Ilaria Dal Prà
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics and Gynecology, Medical School, University of Verona, Veneto, 37134 Verona, Italy; (U.A.); (P.H.)
- Burns Department, Shenzhen Second People’s Hospital, University of Shenzhen, Shenzhen 518000, China
- Correspondence: (A.C.); (I.D.P.); Tel.: +39-045-802-7646 (A.C.); +39-045-802-7161 (I.D.P)
| |
Collapse
|
8
|
Plasma levels of soluble NCAM in multiple sclerosis. J Neurol Sci 2019; 396:36-41. [DOI: 10.1016/j.jns.2018.10.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/24/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022]
|
9
|
Janelidze S, Mattsson N, Stomrud E, Lindberg O, Palmqvist S, Zetterberg H, Blennow K, Hansson O. CSF biomarkers of neuroinflammation and cerebrovascular dysfunction in early Alzheimer disease. Neurology 2018; 91:e867-e877. [PMID: 30054439 PMCID: PMC6133624 DOI: 10.1212/wnl.0000000000006082] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/29/2018] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To measure CSF levels of biomarkers reflecting microglia and astrocytes activation, neuroinflammation, and cerebrovascular changes and study their associations with the core biomarkers of Alzheimer disease (AD) pathology (β-amyloid [Aβ] and tau), structural imaging correlates, and clinical disease progression over time. METHODS The study included cognitively unimpaired elderly (n = 508), patients with mild cognitive impairment (MCI, n = 256), and patients with AD dementia (n = 57) from the longitudinal Swedish BioFINDER cohort. CSF samples were analyzed for YKL-40, interleukin (IL)-6, IL-7, IL-8, IL-15, IP-10, monocyte chemoattractant protein 1, intercellular adhesion molecule 1 (ICAM-1), vascular adhesion molecule 1 (VCAM-1), placental growth factor, and fms-related tyrosine kinase 1 (Flt-1). MRI data were available from 677 study participants. Longitudinal clinical assessments were conducted in control individuals and patients with MCI (mean follow-up 3 years, range 1-6 years). RESULTS CSF levels of YKL-40, ICAM-1, VCAM-1, IL-15, and Flt-1 were increased during the preclinical, prodromal, and dementia stages of AD. High levels of these biomarkers were associated with increased CSF levels of total tau, with the associations, especially for YKL-40, being stronger in Aβ-positive individuals. The results were similar for associations between phosphorylated tau and YKL-40, ICAM-1, and VCAM-1. High levels of the biomarkers were also associated with cortical thinning (primarily in the precuneus and superior parietal regions) and with subsequent cognitive deterioration in patients without dementia as measured with Mini-Mental State Examination (YKL-40) and Clinical Dementia Rating Sum of Boxes (YKL-40, ICAM-1, VCAM-1 and IL-15). Finally, higher levels of CSF YKL-40, ICAM-1, and Flt-1 increased risk of development of AD dementia in patients without dementia. CONCLUSIONS Neuroinflammation and cerebrovascular dysfunction are early events occurring already at presymptomatic stages of AD and contribute to disease progression.
Collapse
Affiliation(s)
- Shorena Janelidze
- From the Clinical Memory Research Unit (S.J., N.M., E.S., O.L., S.P., O.H.), Department of Clinical Sciences, Lund University; Department of Neurology (N.M., S.P.) and Memory Clinic (E.S., O.H.), Skåne University Hospital; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London
| | - Niklas Mattsson
- From the Clinical Memory Research Unit (S.J., N.M., E.S., O.L., S.P., O.H.), Department of Clinical Sciences, Lund University; Department of Neurology (N.M., S.P.) and Memory Clinic (E.S., O.H.), Skåne University Hospital; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London
| | - Erik Stomrud
- From the Clinical Memory Research Unit (S.J., N.M., E.S., O.L., S.P., O.H.), Department of Clinical Sciences, Lund University; Department of Neurology (N.M., S.P.) and Memory Clinic (E.S., O.H.), Skåne University Hospital; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London
| | - Olof Lindberg
- From the Clinical Memory Research Unit (S.J., N.M., E.S., O.L., S.P., O.H.), Department of Clinical Sciences, Lund University; Department of Neurology (N.M., S.P.) and Memory Clinic (E.S., O.H.), Skåne University Hospital; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (S.J., N.M., E.S., O.L., S.P., O.H.), Department of Clinical Sciences, Lund University; Department of Neurology (N.M., S.P.) and Memory Clinic (E.S., O.H.), Skåne University Hospital; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London
| | - Henrik Zetterberg
- From the Clinical Memory Research Unit (S.J., N.M., E.S., O.L., S.P., O.H.), Department of Clinical Sciences, Lund University; Department of Neurology (N.M., S.P.) and Memory Clinic (E.S., O.H.), Skåne University Hospital; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London
| | - Kaj Blennow
- From the Clinical Memory Research Unit (S.J., N.M., E.S., O.L., S.P., O.H.), Department of Clinical Sciences, Lund University; Department of Neurology (N.M., S.P.) and Memory Clinic (E.S., O.H.), Skåne University Hospital; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London
| | - Oskar Hansson
- From the Clinical Memory Research Unit (S.J., N.M., E.S., O.L., S.P., O.H.), Department of Clinical Sciences, Lund University; Department of Neurology (N.M., S.P.) and Memory Clinic (E.S., O.H.), Skåne University Hospital; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London.
| |
Collapse
|
10
|
Scaini G, Tonon T, Moura de Souza CF, Schuck PF, Ferreira GC, Quevedo J, Neto JS, Amorim T, Camelo JS, Margutti AVB, Hencke Tresbach R, Sperb-Ludwig F, Boy R, de Medeiros PFV, Schwartz IVD, Streck EL. Evaluation of plasma biomarkers of inflammation in patients with maple syrup urine disease. J Inherit Metab Dis 2018; 41:10.1007/s10545-018-0188-x. [PMID: 29740775 DOI: 10.1007/s10545-018-0188-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 03/27/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Maple syrup urine disease (MSUD) is an autosomal recessive inherited disorder that affects branched-chain amino acid (BCAA) catabolism and is associated with acute and chronic brain dysfunction. Recent studies have shown that inflammation may be involved in the neuropathology of MSUD. However, these studies have mainly focused on single or small subsets of proteins or molecules. Here we performed a case-control study, including 12 treated-MSUD patients, in order to investigate the plasmatic biomarkers of inflammation, to help to establish a possible relationship between these biomarkers and the disease. Our results showed that MSUD patients in treatment with restricted protein diets have high levels of pro-inflammatory cytokines [IFN-γ, TNF-α, IL-1β and IL-6] and cell adhesion molecules [sICAM-1 and sVCAM-1] compared to the control group. However, no significant alterations were found in the levels of IL-2, IL-4, IL-5, IL-7, IL-8, and IL-10 between healthy controls and MSUD patients. Moreover, we found a positive correlation between number of metabolic crisis and IL-1β levels and sICAM-1 in MSUD patients. In conclusion, our findings in plasma of patients with MSUD suggest that inflammation may play an important role in the pathogenesis of MSUD, although this process is not directly associated with BCAA blood levels. Overall, data reported here are consistent with the working hypothesis that inflammation may be involved in the pathophysiological mechanism underlying the brain damage observed in MSUD patients.
Collapse
Affiliation(s)
- Giselli Scaini
- Laboratório de Bioenergética e Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Tássia Tonon
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Post Graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Patricia F Schuck
- Laboratório de Erros Inatos do Metabolismo, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gustavo C Ferreira
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Quevedo
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | | | - Tatiana Amorim
- Associação de Pais e Amigos dos Excepcionais (APAE), Salvador, Brazil
| | - Jose S Camelo
- Pediatrics Department, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Rafael Hencke Tresbach
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Fernanda Sperb-Ludwig
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Raquel Boy
- Pediatrics Department, Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula F V de Medeiros
- Unidade Acadêmica de Medicina, Hospital Universitário Alcides Carneiro, Universidade Federal de Campina Grande, Campina Grande, Brazil
| | - Ida Vanessa D Schwartz
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Emilio Luiz Streck
- Laboratório de Bioenergética e Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
11
|
A Novel Association of Polymorphism in the ITGA4 Gene Encoding the VLA-4 α4 Subunit with Increased Risk of Alzheimer's Disease. Mediators Inflamm 2018; 2018:7623823. [PMID: 29769839 PMCID: PMC5892238 DOI: 10.1155/2018/7623823] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/11/2018] [Accepted: 02/28/2018] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia in elderly people worldwide. Many studies support the hypothesis that the inflammation of the CNS contributes to the neurodegeneration and disease progression. The integrin molecule α4β1, also known as very late antigen 4 (VLA-4), belongs to adhesion molecules that activate the inflammatory process through the migration of immune cells into the CNS. Therefore, the objective of our study was to analyze the association between two polymorphisms located in the ITGA4 gene encoding the α4 subunit of VLA-4 and the risk of AD. 104 late-onset AD patients and 206 control subjects from Slovakia were genotyped for ITGA4 gene SNP polymorphism rs113276800 (-269C/A) and rs1143676 (+3061A/G). The same study cohorts were also genotyped for the APOE-ε4, which is a known genetic factor associated with increased risk of AD developing. ITGA4 polymorphism analysis revealed significantly higher frequency of the +3061AG carriers in AD group compared to the controls (P ≤ 0.05). Following the APOE-ε4 stratification of study groups, the association remained significant only in APOE-ε4 noncarriers. Our study suggests a novel association of ITGA4 +3061A/G polymorphism with AD and its possible contribution to the disease pathology.
Collapse
|
12
|
Zenaro E, Piacentino G, Constantin G. The blood-brain barrier in Alzheimer's disease. Neurobiol Dis 2017; 107:41-56. [PMID: 27425887 PMCID: PMC5600438 DOI: 10.1016/j.nbd.2016.07.007] [Citation(s) in RCA: 446] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/06/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder characterized by the pathological accumulation of amyloid beta (Aβ) peptides and neurofibrillary tangles containing hyperphosphorylated neuronal tau protein. AD pathology is also characterized by chronic brain inflammation, which promotes disease pathogenesis. In this context, the blood-brain barrier (BBB), a highly specialized endothelial cell membrane that lines cerebral microvessels, represents the interface between neural cells and circulating cells of the immune system. The BBB thus plays a key role in the generation and maintenance of chronic inflammation during AD. The BBB operates within the neurovascular unit (NVU), which includes clusters of glial cells, neurons and pericytes. The NVU becomes dysfunctional during AD, and each of its components may undergo functional changes that contribute to neuronal injury and cognitive deficit. In transgenic animals with AD-like pathology, recent studies have shown that circulating leukocytes migrate through the activated brain endothelium when certain adhesion molecules are expressed, penetrating into the brain parenchyma, interacting with the NVU components and potentially affecting their structural integrity and functionality. Therefore, migrating immune system cells in cerebral vessels act in concert with the modified BBB and may be integrated into the dysfunctional NVU. Notably, blocking the adhesion mechanisms controlling leukocyte-endothelial interactions inhibits both Aβ deposition and tau hyperphosphorylation, and reduces memory loss in AD models. The characterization of molecular mechanisms controlling vascular inflammation and leukocyte trafficking could therefore help to determine the basis of BBB dysfunction during AD and may lead to the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Elena Zenaro
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy
| | - Gennj Piacentino
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy.
| |
Collapse
|
13
|
Hu Y, Cao C, Qin XY, Yu Y, Yuan J, Zhao Y, Cheng Y. Increased peripheral blood inflammatory cytokine levels in amyotrophic lateral sclerosis: a meta-analysis study. Sci Rep 2017; 7:9094. [PMID: 28831083 PMCID: PMC5567306 DOI: 10.1038/s41598-017-09097-1] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/20/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with poorly understood etiology. Increasing evidence suggest that inflammation may play a critical role in the pathogenesis of ALS. Several studies have demonstrated altered levels of blood cytokines in ALS, but results were inconsistent. Therefore, we did a systematic review of studies comparing blood inflammatory cytokines between ALS patients and control subjects, and quantitatively combined the clinical data with a meta-analysis. The systematic review of Pubmed and Web of Science identified 25 studies encompassing 812 ALS patients and 639 control subjects. Random-effects meta-analysis demonstrated that blood tumor necrosis factor-α (TNF; Hedges' g = 0.655; p = 0.001), TNF receptor 1 (Hedges' g = 0.741; p < 0.001), interleukin 6 (IL-6; Hedges' g = 0.25; p = 0.005), IL-1β (Hedges' g = 0.296; p = 0.038), IL-8 (Hedges' g = 0.449; p < 0.001) and vascular endothelial growth factor (Hedges' g = 0.891; p = 0.003) levels were significantly elevated in patients with ALS compared with control subjects. These results substantially enhance our knowledge of the inflammatory response in ALS, and peripheral blood inflammatory cytokines may be used as diagnostic biomarkers for ALS in the future.
Collapse
Affiliation(s)
- Yang Hu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Chang Cao
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Xiao-Yan Qin
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Yun Yu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Jing Yuan
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Yu Zhao
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China.
| |
Collapse
|
14
|
Bagyinszky E, Giau VV, Shim K, Suk K, An SSA, Kim S. Role of inflammatory molecules in the Alzheimer's disease progression and diagnosis. J Neurol Sci 2017; 376:242-254. [PMID: 28431620 DOI: 10.1016/j.jns.2017.03.031] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a complex disorder and the most common form of neurodegenerative dementia. Several genetic, environmental, and physiological factors, including inflammations and metabolic influences, are involved in the progression of AD. Inflammations are composed of complicated networks of many chemokines and cytokines with diverse cells. Inflammatory molecules are needed for the protection against pathogens, and maintaining their balances is important for normal physiological function. Recent studies demonstrated that inflammation may be involved in neurodegenerative dementia. Cellular immune components, such as microglia or astrocytes, mediate the release of inflammatory molecules, including tumor necrosis factor, growth factors, adhesion molecules, or chemokines. Over- and underexpression of pro- and anti-inflammatory molecules, respectively, may result in neuroinflammation and thus disease initiation and progression. In addition, levels of several inflammatory factors were reported to be altered in the brain or bodily fluids of patients with AD, reflecting their neuropathological changes. Therefore, simultaneous detection of several inflammatory molecules in the early or pre-symptomatic stage may improve the early diagnosis of AD. Further studies are needed to determine, how induction or inhibition of inflammatory factors could be used for AD therapies. This review summarizes the role or possible role of immune cells and inflammatory molecules in disease progression or prevention.
Collapse
Affiliation(s)
- Eva Bagyinszky
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - Vo Van Giau
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - Kyuhwan Shim
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea.
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| |
Collapse
|
15
|
Hammouda NE, Salah El-Din MA, El-Shishtawy MM, El-Gayar AM. Serum Cystatin C as a Biomarker in Diffuse Large B-Cell Lymphoma. Sci Pharm 2017; 85:scipharm85010009. [PMID: 28282874 PMCID: PMC5388146 DOI: 10.3390/scipharm85010009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 11/16/2022] Open
Abstract
Elevated serum levels of cystatin C are found to be related to poor outcome and metastatic potential of some malignant disorders. To evaluate the clinical prominence of serum cystatin C in diffuse large B-cell lymphoma (DLBCL), blood samples were obtained from 58 patients at the time of diagnosis and paired blood samples were obtained from 22 patients at the time of remission. Also, serum cystatin C level was measured in matched healthy controls. Serum cystatin C levels were significantly more elevated in DLBCL patients than in controls (p < 0.0001). Furthermore, paired-sample analysis revealed that pretreatment cystatin C levels were reduced significantly in patients who achieved remission after therapy (p = 0.016). High serum cystatin C levels were correlated with age over 60 years (p = 0.049), extra-nodal involvement (p = 0.005) and with high serum lactate dehydrogenase (LDH) (p < 0.013). Elevated serum cystatin C levels were associated with extra-nodal involvement and they were significantly reduced to normal range after the remission. However, Kaplan-Meier curves revealed no survival difference in the pretreatment serum cystatin C levels. Therefore, serum cystatin C may be a novel biomarker that reflects tumor burden in DLBCL but bears no prognostic significance regarding survival.
Collapse
Affiliation(s)
- Nada E Hammouda
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Manal A Salah El-Din
- Oncology Center, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Mamdouh M El-Shishtawy
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Amal M El-Gayar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
16
|
Pate KM, Murphy RM. Cerebrospinal Fluid Proteins as Regulators of Beta-amyloid Aggregation and Toxicity. Isr J Chem 2017; 57:602-612. [PMID: 29129937 DOI: 10.1002/ijch.201600078] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Amyloid disorders, such as Alzheimer's, are almost invariably late-onset diseases. One defining diagnostic feature of Alzheimer's disease is the deposition of beta-amyloid as extracellular plaques, primarily in the hippocampus. This raises the question: are there natural protective agents that prevent beta-amyloid from depositing, and is it loss of this protection that leads to onset of disease? Proteins in cerebrospinal fluid (CSF) have been suggested to act as just such natural protective agents. Here, we describe some of the early evidence that led to this suggestion, and we discuss, in greater detail, two CSF proteins that have garnered the bulk of the attention.
Collapse
Affiliation(s)
- Kayla M Pate
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison WI 53706 (USA)
| | - Regina M Murphy
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison WI 53706 (USA)
| |
Collapse
|
17
|
Henderson-Smith A, Corneveaux JJ, De Both M, Cuyugan L, Liang WS, Huentelman M, Adler C, Driver-Dunckley E, Beach TG, Dunckley TL. Next-generation profiling to identify the molecular etiology of Parkinson dementia. NEUROLOGY-GENETICS 2016; 2:e75. [PMID: 27275011 PMCID: PMC4881621 DOI: 10.1212/nxg.0000000000000075] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/21/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVE We sought to determine the underlying cortical gene expression changes associated with Parkinson dementia using a next-generation RNA sequencing approach. METHODS In this study, we used RNA sequencing to evaluate differential gene expression and alternative splicing in the posterior cingulate cortex from neurologically normal control patients, patients with Parkinson disease, and patients with Parkinson disease with dementia. RESULTS Genes overexpressed in both disease states were involved with an immune response, whereas shared underexpressed genes functioned in signal transduction or as components of the cytoskeleton. Alternative splicing analysis produced a pattern of immune and RNA-processing disturbances. CONCLUSIONS Genes with the greatest degree of differential expression did not overlap with genes exhibiting significant alternative splicing activity. Such variation indicates the importance of broadening expression studies to include exon-level changes because there can be significant differential splicing activity with potential structural consequences, a subtlety that is not detected when examining differential gene expression alone, or is underrepresented with probe-limited array technology.
Collapse
Affiliation(s)
- Adrienne Henderson-Smith
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Jason J Corneveaux
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Matthew De Both
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Lori Cuyugan
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Winnie S Liang
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Matthew Huentelman
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Charles Adler
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Erika Driver-Dunckley
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Thomas G Beach
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Travis L Dunckley
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| |
Collapse
|
18
|
Role of sICAM-1 and sVCAM-1 as biomarkers in early and late stages of schizophrenia. J Psychiatr Res 2016; 73:45-52. [PMID: 26679764 DOI: 10.1016/j.jpsychires.2015.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/31/2015] [Accepted: 11/02/2015] [Indexed: 12/14/2022]
Abstract
Schizophrenia (SZ) is a neuroprogressive disorder presenting with biochemical, functional, and structural changes, which differ from early to late stages of the illness. We explored the differences in serum levels of soluble intercellular cell adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1) between early and late stages of SZ, in regard to clinical characteristics and treatment application. Serum levels of sICAM-1 and sVCAM-1 were measured in 80 patients with SZ (40 early stage; 40 late stage), and compared with 80 healthy controls, matched by age, gender, body mass index, and smoking habits with each SZ group. Serum levels of sICAM-1 and sVCAM-1 were measured using ELISA. The severity of psychopathology was assessed using the Clinical Global Impression Scale and five-factor Positive and Negative Symptoms in Schizophrenia Scale. After adjustment for confounders, we noticed normal levels of sICAM-1 in the early stage, and elevated levels of sICAM-1 in the late stage of SZ. sVCAM-1 levels were decreased in both stages of SZ. Higher sICAM-1 levels have been related to more pronounced cognitive deficit and excitement symptoms in the early stage of SZ and to favorable characteristics of treatment application in both stages. SZ is associated with changes in the levels of adhesion molecules that vary from early to late stages of the illness. This implies that the concept of biochemical staging is applicable in SZ, at least for markers of cellular adhesion.
Collapse
|
19
|
Yang X, Hou D, Jiang W, Zhang C. Intercellular protein-protein interactions at synapses. Protein Cell 2014; 5:420-44. [PMID: 24756565 PMCID: PMC4026422 DOI: 10.1007/s13238-014-0054-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 03/23/2014] [Indexed: 12/11/2022] Open
Abstract
Chemical synapses are asymmetric intercellular junctions through which neurons send nerve impulses to communicate with other neurons or excitable cells. The appropriate formation of synapses, both spatially and temporally, is essential for brain function and depends on the intercellular protein-protein interactions of cell adhesion molecules (CAMs) at synaptic clefts. The CAM proteins link pre- and post-synaptic sites, and play essential roles in promoting synapse formation and maturation, maintaining synapse number and type, accumulating neurotransmitter receptors and ion channels, controlling neuronal differentiation, and even regulating synaptic plasticity directly. Alteration of the interactions of CAMs leads to structural and functional impairments, which results in many neurological disorders, such as autism, Alzheimer's disease and schizophrenia. Therefore, it is crucial to understand the functions of CAMs during development and in the mature neural system, as well as in the pathogenesis of some neurological disorders. Here, we review the function of the major classes of CAMs, and how dysfunction of CAMs relates to several neurological disorders.
Collapse
Affiliation(s)
- Xiaofei Yang
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, 430074 China
| | - Dongmei Hou
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, 430074 China
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871 China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871 China
| | - Wei Jiang
- Key Laboratory of Cognitive Science, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, Wuhan, 430074 China
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871 China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871 China
| | - Chen Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871 China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871 China
| |
Collapse
|
20
|
Chang KC, Chang WC, Chang Y, Hung LY, Lai CH, Yeh YM, Chou YW, Chen CH. Ran GTPase-activating protein 1 is a therapeutic target in diffuse large B-cell lymphoma. PLoS One 2013; 8:e79863. [PMID: 24223200 PMCID: PMC3819250 DOI: 10.1371/journal.pone.0079863] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 09/25/2013] [Indexed: 01/07/2023] Open
Abstract
Lymphoma-specific biomarkers contribute to therapeutic strategies and the study of tumorigenesis. Diffuse large B-cell lymphoma (DLBCL) is the most common type of malignant lymphoma. However, only 50% of patients experience long-term survival after current treatment; therefore, developing novel therapeutic strategies is warranted. Comparative proteomic analysis of two DLBCL lines with a B-lymphoblastoid cell line (LCL) showed differential expression of Ran GTPase-activating protein 1 (RanGAP1) between them, which was confirmed using immunoblotting. Immunostaining showed that the majority of DLBCLs (92%, 46/50) were RanGAP1(+), while reactive lymphoid hyperplasia (n = 12) was RanGAP1(+) predominantly in germinal centers. RanGAP1 was also highly expressed in other B-cell lymphomas (BCL, n = 180) with brisk mitotic activity (B-lymphoblastic lymphoma/leukemia: 93%, and Burkitt lymphoma: 95%) or cell-cycle dysregulation (mantle cell lymphoma: 83%, and Hodgkin's lymphoma 91%). Interestingly, serum RanGAP1 level was higher in patients with high-grade BCL (1.71 ± 2.28 ng/mL, n = 62) than in low-grade BCL (0.75 ± 2.12 ng/mL, n = 52) and healthy controls (0.55 ± 1.58 ng/mL, n = 75) (high-grade BCL vs. low-grade BCL, p = 0.002; high-grade BCL vs. control, p < 0.001, Mann-Whitney U test). In vitro, RNA interference of RanGAP1 showed no effect on LCL but enhanced DLBCL cell death (41% vs. 60%; p = 0.035) and cell-cycle arrest (G0/G1: 39% vs. 49%, G2/M: 19.0% vs. 7.5%; p = 0.030) along with decreased expression of TPX2 and Aurora kinases, the central regulators of mitotic cell division. Furthermore, ON 01910.Na (Estybon), a multikinase inhibitor induced cell death, mitotic cell arrest, and hyperphosphorylation of RanGAP1 in DLBCL cell lines but no effects in normal B and T cells. Therefore, RanGAP1 is a promising marker and therapeutic target for aggressive B-cell lymphoma, especially DLBCL.
Collapse
Affiliation(s)
- Kung-Chao Chang
- Department of Pathology, College of Medicine, National Cheng Kung University and Hospital, Tainan, Taiwan
- * E-mail: (KCC); (CHC)
| | - Wei-Chao Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yao Chang
- Division of Infectious Diseases, National Health Research Institute, Tainan, Taiwan
| | - Liang-Yi Hung
- Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Hsien Lai
- Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Min Yeh
- Department of Internal Medicine, College of Medicine, National Cheng Kung University and Hospital, Tainan, Taiwan,
| | - Yu-Wei Chou
- Department of Pathology, College of Medicine, National Cheng Kung University and Hospital, Tainan, Taiwan
| | - Chung-Hsuan Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- * E-mail: (KCC); (CHC)
| |
Collapse
|
21
|
Hall JR, Wiechmann AR, Johnson LA, Edwards M, Barber RC, Winter AS, Singh M, O'Bryant SE. Biomarkers of vascular risk, systemic inflammation, and microvascular pathology and neuropsychiatric symptoms in Alzheimer's disease. J Alzheimers Dis 2013; 35:363-71. [PMID: 23403534 DOI: 10.3233/jad-122359] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Numerous serum and plasma based biomarkers of systemic inflammation have been linked to both neuropsychiatric disorders and Alzheimer's disease (AD). The present study investigated the relationship of clinical biomarkers of cardiovascular risk (cholesterol, triglycerides, and homocysteine) and a panel of markers of systemic inflammation (CRP, TNF-α, IL1-ra, IL-7, IL-10, IL-15, IL-18) and microvascular pathology (ICAM-1, VCAM-1) to neuropsychiatric symptoms in a sample with mild AD. Biomarker data was analyzed on a sample of 194 diagnosed with mild to moderate probable AD. The sample was composed of 127 females and 67 males. The presence of neuropsychiatric symptoms was gathered from interview with caretakers/family members using the Neuropsychiatric Inventory. For the total sample, IL-15, VCAM (vascular adhesion molecule), and triglycerides were significantly and negatively related to number of neuropsychiatric symptoms, and total cholesterol and homocysteine were positively related and as a group accounted for 16.1% of the variance. When stratified by gender, different patterns of significant biomarkers were found with relationships more robust for males for both total symptoms and symptom clusters. A combination of biomarkers of systemic inflammation, microvascular pathology, and clinical biomarkers of cardiovascular risk can account for a significant portion of the variance in the occurrence of neuropsychiatric symptoms in AD supporting a vascular and inflammatory component of psychiatric disorders found in AD. Gender differences suggest distinct impact of specific risks with total cholesterol, a measure of cardiovascular risk, being the strongest marker for males and IL-15, a marker of inflammation, being the strongest for females.
Collapse
Affiliation(s)
- James R Hall
- Institute of Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Mruk DD, Xiao X, Lydka M, Li MWM, Bilinska B, Cheng CY. Intercellular adhesion molecule 1: recent findings and new concepts involved in mammalian spermatogenesis. Semin Cell Dev Biol 2013; 29:43-54. [PMID: 23942142 DOI: 10.1016/j.semcdb.2013.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 07/26/2013] [Accepted: 07/29/2013] [Indexed: 01/05/2023]
Abstract
Spermatogenesis, the process of spermatozoa production, is regulated by several endocrine factors, including testosterone, follicle stimulating hormone, luteinizing hormone and estradiol 17β. For spermatogenesis to reach completion, developing germ cells must traverse the seminiferous epithelium while remaining transiently attached to Sertoli cells. If germ cell adhesion were to be compromised for a period of time longer than usual, germ cells would slough from the seminiferous epithelium and infertility would result. Presently, Sertoli-germ cell adhesion is known to be mediated largely by classical and desmosomal cadherins. More recent studies, however, have begun to expand long-standing concepts and to examine the roles of other proteins such as intercellular adhesion molecules. In this review, we focus on the biology of intercellular adhesion molecules in the mammalian testis, hoping that this information is useful in the design of future studies.
Collapse
Affiliation(s)
- Dolores D Mruk
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States.
| | - Xiang Xiao
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Marta Lydka
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Michelle W M Li
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Barbara Bilinska
- Institute of Zoology, Department of Endocrinology, The Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - C Yan Cheng
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| |
Collapse
|
23
|
Evans MC, Couch Y, Sibson N, Turner MR. Inflammation and neurovascular changes in amyotrophic lateral sclerosis. Mol Cell Neurosci 2013; 53:34-41. [PMID: 23110760 DOI: 10.1016/j.mcn.2012.10.008] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 10/17/2012] [Accepted: 10/19/2012] [Indexed: 11/16/2022] Open
Abstract
Neuroinflammation in now established as an important factor in the pathogenesis of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). At various time points, astrocytes and microglia are markedly activated, either producing neuroprotective or pro-inflammatory molecules, which can decrease or increase the rate of primary motor neuron degeneration respectively. Recent research has shown that this neuroinflammatory component is affected by the peripheral immune system; T lymphocytes in particular are able to cross into the brain and spinal cord parenchyma, where they interact with resident microglia, either inducing them to adopt an M1 (cytotoxic) or M2 (protective) phenotype, depending on the stage of disease. Clearly understanding the changes that occur to allow the interaction between peripheral and central immune responses will be essential in any attempt to manipulate the disease process via neuroinflammatory mechanisms. However, our understanding of the endothelial changes, which facilitate the infiltration of peripheral immune cells into the brain and spinal cord, is still in its infancy. There are suggestions, though, of up-regulation of cellular adhesion molecules, which are able to arrest circulating leukocytes and facilitate diapedesis into the brain parenchyma. In addition, tight junction proteins appear to be down-regulated, leading to an increase in vascular permeability, an effect that is amplified by vascular damage late in the disease process. This review summarises our current knowledge regarding neuroinflammation, peripheral immune involvement, and endothelial changes in ALS. This article is part of a Special Issue entitled 'Neuroinflammation in neurodegeneration and neurodysfunction'.
Collapse
Affiliation(s)
- M C Evans
- Nuffield Department of Clinical Neurosciences, Oxford University, UK
| | | | | | | |
Collapse
|
24
|
Wennström M, Nielsen HM. Cell adhesion molecules in Alzheimer's disease. Degener Neurol Neuromuscul Dis 2012; 2:65-77. [PMID: 30890880 DOI: 10.2147/dnnd.s19829] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell adhesion molecules (CAMs) mediate interactions between cells and their surroundings that are vital to processes controlling for cell survival, activation, migration, and plasticity. However, increasing evidence suggests that CAMs also mediate mechanisms involved in several neurological diseases. This article reviews the current knowledge on the role of CAMs in amyloid-β (Aβ) metabolism, cell plasticity, neuroinflammation, and vascular changes, all of which are considered central to the pathogenesis and progression of Alzheimer's disease (AD). This paper also outlines the possible roles of CAMs in current and novel AD treatment strategies.
Collapse
Affiliation(s)
- Malin Wennström
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden,
| | | |
Collapse
|
25
|
Abstract
The central nervous system (CNS) is a complex and precise mechanism that controls the most highest functions of the body. All of them depend on the cellular and molecular interactions called by neurobiologists "cellular plasticity". The CNS is a flexible structure but its regeneration after damage is strongly limited. Better understanding of cellular and molecular basis of brain repair can open new way in the development of therapeutic tools for neurodegeneration. Among many molecules that participate in the formation of neuronal networks, neural cell adhesion molecule (NCAM) and its sialylated derivative seem to play crucial role in the life of brain. In particular, polysialylated cell adhesion molecule (PSA-NCAM) is proposed to participate in the neuroprotective response in neurodegeneration by reducing of AMPA/NMDA receptors sensitivity to glutamate and facilitating disconnection of cell-cell interactions. These mechanisms protect from excitotoxic damage and promote dendritic/spine re-growth. This review briefly focuses on the expression and role of PSA-NCAM in neurodegenerative diseases and its potential application in therapy.
Collapse
|
26
|
Reale M, Kamal MA, Velluto L, Gambi D, Di Nicola M, Greig NH. Relationship between inflammatory mediators, Aβ levels and ApoE genotype in Alzheimer disease. Curr Alzheimer Res 2012; 9:447-57. [PMID: 22272623 PMCID: PMC5215089 DOI: 10.2174/156720512800492549] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 11/05/2011] [Accepted: 11/09/2011] [Indexed: 11/22/2022]
Abstract
Activation of inflammatory processes is observed within the brain as well as periphery of subjects with Alzheimer's disease (AD). Whether or not inflammation represents a possible cause of AD or occurs as a consequence of the disease process, or, alternatively, whether the inflammatory response might be beneficial to slow the disease progression remains to be elucidated. The cytokine IL-18 shares with IL-1 the same pro-inflammatory features. Consequent to these similarities, IL-18 and its endogenous inhibitor, IL-18BP, were investigated in the plasma of AD patients versus healthy controls (HC). An imbalance of IL-18 and IL-18BP was observed in AD, with an elevated IL-18/IL-18BP ratio that might be involved in disease pathogenesis. As part of the inflammatory response, altered levels of RANTES, MCP-1 and ICAM- 1, molecules involved in cell recruitment to inflammatory sites, were observed in AD. Hence, correlations between IL-18 and other inflammatory plasma markers were analyzed. A negative correlation was observed between IL-18 and IL-18BP in both AD and HC groups. A positive correlation was observed between IL-18 and ICAM-1 in AD patients, whereas a negative correlation was evident in the HC group. IL-18 positively correlated with Aβ in both groups, and no significant correlations were observed between IL-18, RANTES and MCP-1. An important piece of evidence supporting a pathophysiologic role for inflammation in AD is the number of inflammatory mediators that have been found to be differentially regulated in AD patients, and specific ones may provide utility as part of a biomarker panel to not only aid early AD diagnosis, but follow its progression.
Collapse
Affiliation(s)
- M Reale
- Dept. of Oncology and Experimental Medicine, Unit of Immunodiagnostic, University "G. D'Annunzio", Chieti, Italy.
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
IL-15 is a proinflammatory cytokine. It is produced by activated blood monocytes, macrophages, dendritic cells, and activated glial cells. It promotes T-cell proliferation, induction of cytolytic effector cells including natural killer and cytotoxic cells and stimulates B-cell to proliferate and secrete immunoglobulins. Little information is available on the exact role of IL-15 in the neurological diseases. Microglial cells are the main regulators of both innate and adaptive immune responses in the central nervous system (CNS). IL-15 may be involved in the inflammatory reactions and microglial activation of some common CNS disorders such as multiple sclerosis, Alzheimer's and Parkinson's disease, but its exact role in their pathogenesis is not clear.
Collapse
Affiliation(s)
- M Rentzos
- Department of Neurology, Aeginition Hospital, Athens National University, School of Medicine, Greece.
| | | |
Collapse
|
28
|
Abstract
Experimental and clinical evidence have demonstrated the increased synthesis of specific inflammatory mediators, and the upregulation of their cognate receptors in the chronic epileptic brain, indicating that some proinflammatory pathways are activated in seizure foci. Inhibition of experimental seizures by pharmacological interference with specific proinflammatory signaling, together with evidence of changes in intrinsic susceptibility to seizures in transgenic mice with perturbed inflammatory pathways, was instrumental to establish the concept that brain inflammation has a role in the etiopathogenesis of seizures. Increasing evidence also highlights the possible involvement of inflammatory processes arising in the injured brain in the development of epilepsy (i.e., in epileptogenesis). Since brain inflammation in epilepsy is not a mere epiphenomenon of the pathology but is likely involved in the mechanisms underlying neuronal hyperexcitability, the onset of seizures and their recurrence, it might be considered as a biomarker of disease development and severity, and, as such, could be used for diagnostic, prognostic or therapeutic purposes, provided that adequate noninvasive methodologies are developed to detect and quantify brain inflammation in humans.
Collapse
Affiliation(s)
- Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Via G. La Masa 19, 20156 Milano, Italy .
| | | |
Collapse
|
29
|
Reduced plasma levels of P-selectin and L-selectin in a pilot study from Alzheimer disease: relationship with neuro-degeneration. Biogerontology 2011; 12:451-4. [PMID: 21484243 DOI: 10.1007/s10522-011-9335-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 04/04/2011] [Indexed: 10/16/2022]
Abstract
Neurodegenerative processes associated with Alzheimer's disease (AD) are accompanied by reactive astrogliosis and microglia activation and a role for chronic inflammation in the brain degeneration of these patients has been suggested. Moreover impaired immune functions in AD brains might also influence the disease's progression. Therefore, it is of interest to further characterized inflammatory molecules in the peripheral blood of patients with AD and its relationship with cognitive decline. A complex picture emerged in this pilot study and IL-8, IFN-gamma, MCP-1 and VEGF levels were increased in AD. Levels of P-selectin and L-selectin were decreased in AD and lowest in AD patients with highest cognitive decline. Our findings suggest that these molecules may induce alterations of endothelial regulation and influence neurodegenerative processes of AD.
Collapse
|
30
|
Sabayan B, Zamiri N, Farshchizarabi S, Sabayan B. Phosphodiesterase-5 inhibitors: novel weapons against Alzheimer's disease? Int J Neurosci 2010; 120:746-51. [PMID: 20942592 DOI: 10.3109/00207454.2010.520381] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Although Alzheimer's disease (AD) poses a major health problem in both developing and developed countries, no definite treatment is available for its cure; hence efforts are being focused on introducing disease-modifying agents for slowing down its course. Recent studies on the effects of sildenafil on different organs have shown that PDE-5 inhibitors may offer new horizons in therapeutic treatment of pulmonary hypertension, multiple sclerosis, neuropathic pain, and age-related memory impairment. In this paper we introduce PDE-5 inhibitors as novel disease-modifying agents against AD and review the different impacts of PDE-5 inhibition on various pathogenic mechanisms leading to AD, including neuronal apoptosis, neurovascular dysfunction, neurotransmitter modulation, and impairment of neurogenesis.
Collapse
Affiliation(s)
- Behnam Sabayan
- Department of Geriatrics and Gerontology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | |
Collapse
|
31
|
Soluble cell adhesion molecules in monocytes of Alzheimer's disease and mild cognitive impairment. Exp Gerontol 2009; 45:70-4. [PMID: 19836440 DOI: 10.1016/j.exger.2009.10.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2009] [Revised: 09/24/2009] [Accepted: 10/12/2009] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with characteristic neuropathological features that are accompanied by inflammatory processes and release of pro-inflammatory cytokines. There is evidence that microglial cells are a key mediator of damage in AD. The microglial compartment may arise to a greater part from activation and transmigration of circulating monocytes. The aim of the present pilot study was to explore, if different cell adhesion molecules (ICAM-1 and -3, PECAM-1, VCAM-1, P-, L- and E-selectins, E-cadherin), RAGE and CD14 are affected in monocytes of healthy subjects compared to patients suffering from AD or mild cognitive impairment (MCI). Monocytes were isolated by negative magnetic selection (MACS) from EDTA blood samples, and extracts were analyzed by Searchlight Multiplex ELISAs. When compared to healthy subjects, the ratio of monocytic ICAM-3/CD14 was significantly decreased in MCI and AD patients and the ratio of the monocytic P-selectin/CD14 was specifically decreased in AD patients. In conclusion, our data show that monocytic cell adhesion molecules are decreased in AD and MCI patients. Further larger longitudinal studies should then clarify whether any of these parameters may be useful as a diagnostic biomarker.
Collapse
|
32
|
Janciauskiene S, Erikson C, Warkentin S. A link between sICAM-1, ACE and parietal blood flow in the aging brain. Neurobiol Aging 2009; 30:1504-11. [DOI: 10.1016/j.neurobiolaging.2007.08.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2006] [Revised: 05/05/2007] [Accepted: 08/16/2007] [Indexed: 02/07/2023]
|
33
|
Zuliani G, Cavalieri M, Galvani M, Passaro A, Munari MR, Bosi C, Zurlo A, Fellin R. Markers of endothelial dysfunction in older subjects with late onset Alzheimer's disease or vascular dementia. J Neurol Sci 2008; 272:164-70. [PMID: 18597785 DOI: 10.1016/j.jns.2008.05.020] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 04/30/2008] [Accepted: 05/23/2008] [Indexed: 12/13/2022]
Abstract
A consistent amount of evidence suggests that vascular factors might be involved in the pathogenesis of late onset Alzheimer's disease (LOAD). We evaluated the presence of endothelial dysfunction by measuring the plasma levels of soluble E-selectin and vascular cell adhesion molecule 1 (VCAM-1) in a sample of patients affected by LOAD (n. 60) or vascular dementia (VD: n. 80). They were compared with a sample of older patients with cerebrovascular disease but not-dementia (CDND: n. 40), and with a sample of healthy older controls (n. 30). sVCAM-1 plasma levels were higher in LOAD and VD compared with controls. Among patients (LOAD, VD, and CDND), sE-selectin levels were higher in individuals with most severe cerebrovascular disease on CT scan. At multivariate regression analysis, fasting glucose (p<0.05) and TNF-alpha levels (p<0.02) were positively correlated with sE-selectin levels (adjusted r(2): 20%), while sVCAM-1 was positively correlated with age (p<0.01), and alcohol consumption (p: 0.03), and negatively associated with HDL-C levels (p: 0.005), (p<0.01; adjusted r(2): 44%), independent of possible confounders. Increased sVCAM-1 plasma levels in LOAD and VD suggest the existence of endothelial dysfunction in both types of dementia. The possible role of E-selectin in the pathogenesis of cerebrovascular disease is also supported by our data.
Collapse
Affiliation(s)
- G Zuliani
- Department of Clinical and Experimental Medicine, Section of Internal Medicine, Gerontology and Geriatrics, University of Ferrara, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Freudenberg JA, Bembas K, Greene MI, Zhang H. Non-invasive, ultra-sensitive, high-throughput assays to quantify rare biomarkers in the blood. Methods 2008; 46:33-8. [PMID: 18573345 DOI: 10.1016/j.ymeth.2008.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 05/22/2008] [Indexed: 11/27/2022] Open
Abstract
Many diseases are easier to treat and control when detected at an early stage of disease progression. Often, disease-related antigens or biomarkers are shed from the primary site and present in the blood. Unfortunately, there are very few tests capable of detecting these rare biomarkers in the blood. A blood test would be very useful to diagnose the disease earlier, monitor effectiveness of treatments, predict recurrence, and monitor recurrence. There is certainly a need to develop assays that are ultra-sensitive, non-invasive, and high-throughput. Here we describe several highly sensitive immunological assays we have developed to detect rare serum antigens. Initially we created an assay named immuno-detection amplified by T7 RNA polymerase (IDAT). To enhance the effectiveness and streamline the procedure, this assay was amended to the facile amplification system termed fluorescent amplification catalyzed by T7 polymerase technique (FACTT). These assays have been used to analyze the tumor antigen HER2 and the prion protein PrPSc. They can also be applied to other tumor markers or antigens from a variety of diseases such as cardiovascular disease, rheumatoid arthritis, Alzheimer's disease, Parkinson's disease, and hepatitis. These tests are not limited to testing only serum, but may also be applicable to detecting biomarkers in tissue, saliva, urine, cerebrospinal fluid, etc. Clearly, the FACTT-based technology represents an important step in the detection of rare molecules in fluids or tissues for a variety of diseases.
Collapse
Affiliation(s)
- Jaclyn A Freudenberg
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, 252 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104-6082, USA
| | | | | | | |
Collapse
|
35
|
Nielsen HM, Londos E, Minthon L, Janciauskiene SM. Soluble adhesion molecules and angiotensin-converting enzyme in dementia. Neurobiol Dis 2007; 26:27-35. [PMID: 17270454 DOI: 10.1016/j.nbd.2006.11.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Revised: 10/22/2006] [Accepted: 11/22/2006] [Indexed: 12/13/2022] Open
Abstract
We aimed to determine plasma and cerebrospinal fluid (CSF) levels of angiotensin-converting enzyme (ACE) and the soluble forms of intercellular adhesion molecule-1 (sICAM-1), vascular cell adhesion molecule-1 (sVCAM-1) and platelet endothelial cell adhesion molecule-1 (sPECAM-1) as surrogate markers for endothelial cell activation in clinically diagnosed patients with Alzheimer's disease (AD, n=260), dementia with Lewy bodies (DLB, n=39) and non-demented controls (n=34). Plasma sICAM-1 and sPECAM-1 were higher and CSF sVCAM-1 were lower in AD and DLB patients than in controls (p<0.001). DLB patients had higher CSF sICAM-1, but lower CSF sVCAM-1 (p<0.001). No difference in ACE levels was found between the dementia groups and controls. In controls and AD patients CSF sICAM and sVCAM-1 strongly correlated with each other and with blood barrier permeability whereas in DLB group these correlations were weaker. The observed patterns in adhesion molecules may reflect distinctions in the pathophysiological basis of their generation in dementia patients.
Collapse
Affiliation(s)
- Henrietta M Nielsen
- Chronic Inflammatory and Degenerative Disease Research Unit, Department of Clinical Sciences, Lund University, 205 02 Malmö, Sweden.
| | | | | | | |
Collapse
|
36
|
Rentzos M, Paraskevas GP, Kapaki E, Nikolaou C, Zoga M, Rombos A, Tsoutsou A, Vassilopoulos D D. Interleukin-12 is reduced in cerebrospinal fluid of patients with Alzheimer's disease and frontotemporal dementia. J Neurol Sci 2006; 249:110-4. [PMID: 16843497 DOI: 10.1016/j.jns.2006.05.063] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 04/14/2006] [Accepted: 05/26/2006] [Indexed: 11/19/2022]
Abstract
UNLABELLED Interleukin-12 is a heterodimeric cytokine produced by activated blood monocytes, macrophages and glial cells. It enhances differentiation and proliferation of T cells and increases production of proinflammatory cytokines, such as Interferon-gamma and Tumor Necrosis Factor-alpha. There is little information about the involvement of IL-12 in the pathophysiology of Alzheimer's disease (AD) and other tauopathies. OBJECTIVES The objective of our study was to assess the role of IL-12 as a potential marker of immune reactions in patients with AD and frontotemporal dementia (FTD). PATIENTS AND METHODS We measured by immunoassay cerebrospinal fluid (CSF) IL-12 levels in 19 patients with AD and 7 patients with FTD in comparison with CSF IL-12 levels in 30 patients with non-inflammatory neurological diseases served as neurological control patients (NCTRL). IL-12 levels were correlated with age, age of disease onset, disease duration, MMSE score, and rate of dementia progression. Abeta42 and Total tau (tau(T)) levels in CSF were also measured. RESULTS Patients with AD had significantly lower CSF IL-12 levels compared with NCTRL patients (p<0.001). Patients with FTD had also lower CSF IL-12 levels compared with NCTRL patients (p<0.05). Age, sex, disease duration and MMSE score did not affect IL-12 levels in any of the groups. In AD a significant positive correlation was noted between IL-12 levels and tau(T) levels (Rs=0.46, p=0.048). CONCLUSIONS Our findings may suggest a reduced inflammatory reaction during the course of AD and FTD. A neurotrophic role of IL-12 and other proinflammatory cytokines cannot be excluded.
Collapse
Affiliation(s)
- Michael Rentzos
- Department of Neurology, Aeginition Hospital, Athens National University, School of Medicine, 72-74 Vas. Sophias Ave, Athens 11528, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Rentzos M, Zoga M, Paraskevas GP, Kapaki E, Rombos A, Nikolaou C, Tsoutsou A, Vassilopoulos D. IL-15 is elevated in cerebrospinal fluid of patients with Alzheimer's disease and frontotemporal dementia. J Geriatr Psychiatry Neurol 2006; 19:114-7. [PMID: 16690997 DOI: 10.1177/0891988706286226] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Interleukin-15 is a novel proinflammatory cytokine. It is produced by activated blood monocytes, macrophages, and glial cells. The objective of our study was to assess the role of interleukin-15 as a marker of increased proinflammatory activity in patients with Alzheimer's disease and frontotemporal dementia. We measured cerebrospinal fluid interleukin-15 levels in 17 patients with Alzheimer's disease and 7 patients with frontotemporal dementia in comparison with 17 patients with amyotrophic lateral sclerosis and 15 patients with Parkinson's disease. Patients with Alzheimer's disease and frontotemporal dementia had significantly higher cerebrospinal fluid interleukin-15 levels compared with patients with noninflammatory neurological diseases (P < .05 and P < .01, respectively). In Alzheimer's disease, a significant positive correlation was noted between interleukin-15 levels and age of onset (R = .48, P = .05). Our findings suggest that interleukin-15 may be implicated in the pathophysiology of Alzheimer's disease and frontotemporal dementia.
Collapse
Affiliation(s)
- M Rentzos
- Department of Neurology, Aeginition Hospital-Athens Medical School.
| | | | | | | | | | | | | | | |
Collapse
|