1
|
Hagen KM, Ousman SS. The Neuroimmunology of Guillain-Barré Syndrome and the Potential Role of an Aging Immune System. Front Aging Neurosci 2021; 12:613628. [PMID: 33584245 PMCID: PMC7873882 DOI: 10.3389/fnagi.2020.613628] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Guillain-Barré syndrome (GBS) is a paralyzing autoimmune condition affecting the peripheral nervous system (PNS). Within GBS there are several variants affecting different aspects of the peripheral nerve. In general, there appears to be a role for T cells, macrophages, B cells, and complement in initiating and perpetuating attacks on gangliosides of Schwann cells and axons. Of note, GBS has an increased prevalence and severity with increasing age. In addition, there are alterations in immune cell functioning that may play a role in differences in GBS with age alongside general age-related declines in reparative processes (e.g., delayed de-differentiation of Schwann cells and decline in phagocytic ability of macrophages). The present review will explore the immune response in GBS as well as in animal models of several variants of the disorder. In addition, the potential involvement of an aging immune system in contributing to the increased prevalence and severity of GBS with age will be theorized.
Collapse
Affiliation(s)
- Kathleen M. Hagen
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Shalina S. Ousman
- Departments of Clinical Neurosciences and Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
2
|
Li L, Liu MS, Li GQ, Tang J, Liao Y, Zheng Y, Guo TL, Kang X, Yuan MT. Relationship between Apolipoprotein Superfamily and Parkinson's Disease. Chin Med J (Engl) 2018; 130:2616-2623. [PMID: 29067960 PMCID: PMC5678263 DOI: 10.4103/0366-6999.217092] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective: Parkinson's disease (PD) is featured with motor disorder and nonmotor manifestations including psychological symptoms, autonomic nervous system dysfunction, and paresthesia, which results in great inconvenience to the patients’ life. The apolipoprotein (Apo) superfamily, as a group of potentially modifiable biomarkers in clinical practice, is of increasing significance in the diagnosis, evaluation, and prognosis of PD. The present review summarized the current understanding and emerging findings of the relationship between Apo superfamily and PD. Data Sources: All literatures were identified by systematically searching PubMed, Embase, and Cochrane electronic databases with terms “Parkinson disease,” “apolipoprotein,” and their synonyms until May 2017. Study Selection: We have thoroughly examined titles and abstracts of all the literatures that met our search strategy and the full text if the research is identified or not so definite. Reference lists of retrieved articles were also scrutinized for additional relevant studies. Results: The levels of plasma ApoA1 are inversely correlated with the risk of PD and the lower levels of ApoA1 trend toward association with poorer motor performance. Higher ApoD expression in neurons represents more puissant protection against PD, which is critical in delaying the neurodegeneration process of PD. It is suggested that APOE alleles are related to development and progression of cognitive decline and age of PD onset, but conclusions are not completely identical, which may be attributed to different ApoE isoforms. APOJ gene expressions are upregulated in PD patients and it is possible that high ApoJ level is an indicator of PD dementia and correlates with specific phenotypic variations in PD. Conclusions: The Apo superfamily has been proved to be closely involved in the initiation, progression, and prognosis of PD. Apos and their genes are of great value in predicting the susceptibility of PD and hopeful to become the target of medical intervention to prevent the onset of PD or slow down the progress. Therefore, further large-scale studies are warranted to elucidate the precise mechanisms of Apos in PD.
Collapse
Affiliation(s)
- Lin Li
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Ming-Su Liu
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Guang-Qin Li
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Jie Tang
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yan Liao
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yang Zheng
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Tong-Li Guo
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Xin Kang
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Mao-Ting Yuan
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
3
|
Brick RM, Sun AX, Tuan RS. Neurotrophically Induced Mesenchymal Progenitor Cells Derived from Induced Pluripotent Stem Cells Enhance Neuritogenesis via Neurotrophin and Cytokine Production. Stem Cells Transl Med 2017; 7:45-58. [PMID: 29215199 PMCID: PMC5746147 DOI: 10.1002/sctm.17-0108] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/06/2017] [Indexed: 12/11/2022] Open
Abstract
Adult tissue‐derived mesenchymal stem cells (MSCs) are known to produce a number of bioactive factors, including neurotrophic growth factors, capable of supporting and improving nerve regeneration. However, with a finite culture expansion capacity, MSCs are inherently limited in their lifespan and use. We examined here the potential utility of an alternative, mesenchymal‐like cell source, derived from induced pluripotent stem cells, termed induced mesenchymal progenitor cells (MiMPCs). We found that several genes were upregulated and proteins were produced in MiMPCs that matched those previously reported for MSCs. Like MSCs, the MiMPCs secreted various neurotrophic and neuroprotective factors, including brain‐derived neurotrophic factor (BDNF), interleukin‐6 (IL‐6), leukemia inhibitory factor (LIF), osteopontin, and osteonectin, and promoted neurite outgrowth in chick embryonic dorsal root ganglia (DRG) cultures compared with control cultures. Cotreatment with a pharmacological Trk‐receptor inhibitor did not result in significant decrease in MiMPC‐induced neurite outgrowth, which was however inhibited upon Jak/STAT3 blockade. These findings suggest that the MiMPC induction of DRG neurite outgrowth is unlikely to be solely dependent on BDNF, but instead Jak/STAT3 activation by IL‐6 and/or LIF is likely to be critical neurotrophic signaling pathways of the MiMPC secretome. Taken together, these findings suggest MiMPCs as a renewable, candidate source of therapeutic cells and a potential alternative to MSCs for peripheral nerve repair, in view of their ability to promote nerve growth by producing many of the same growth factors and cytokines as Schwann cells and signaling through critical neurotrophic pathways. stemcellstranslational Medicine2018;7:45–58
Collapse
Affiliation(s)
- Rachel M Brick
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Aaron X Sun
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Martin-Vaquero P, da Costa RC, Allen MJ, Moore SA, Keirsey JK, Green KB. Proteomic analysis of cerebrospinal fluid in canine cervical spondylomyelopathy. Spine (Phila Pa 1976) 2015; 40:601-12. [PMID: 26030213 PMCID: PMC4451599 DOI: 10.1097/brs.0000000000000831] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Prospective study. OBJECTIVE To identify proteins with differential expression in the cerebrospinal fluid (CSF) from 15 clinically normal (control) dogs and 15 dogs with cervical spondylomyelopathy (CSM). SUMMARY OF BACKGROUND DATA Canine CSM is a spontaneous, chronic, compressive cervical myelopathy similar to human cervical spondylotic myelopathy. There is a limited knowledge of the molecular mechanisms underlying these conditions. Differentially expressed CSF proteins may contribute with novel information about the disease pathogenesis in both dogs and humans. METHODS Protein separation was performed with 2-dimensional electrophoresis. A Student t test was used to detect significant differences between groups (P < 0.05). Three comparisons were made: (1) control versus CSM-affected dogs, (2) control versus non-corticosteroid-treated CSM-affected dogs, and (3) non-corticosteroid-treated CSM-affected versus corticosteroid-treated CSM-affected dogs. Protein spots exhibiting at least a statistically significant 1.25-fold change between groups were selected for subsequent identification with capillary-liquid chromatography tandem mass spectrometry. RESULTS A total of 96 spots had a significant average change of at least 1.25-fold in 1 of the 3 comparisons. Compared with the CSF of control dogs, CSM-affected dogs demonstrated increased CSF expression of 8 proteins including vitamin D-binding protein, gelsolin, creatine kinase B-type, angiotensinogen, α-2-HS-glycoprotein, SPARC (secreted protein, acidic, rich in cysteine), calsyntenin-1, and complement C3, and decreased expression of pigment epithelium-derived factor, prostaglandin-H2 D-isomerase, apolipoprotein E, and clusterin. In the CSF of CSM-affected dogs, corticosteroid treatment increased the expression of haptoglobin, transthyretin isoform 2, cystatin C-like, apolipoprotein E, and clusterin, and decreased the expression of angiotensinogen, α-2-HS-glycoprotein, and gelsolin. CONCLUSION Many of the differentially expressed proteins are associated with damaged neural tissue, bone turnover, and/or compromised blood-spinal cord barrier. The knowledge of the protein changes that occur in CSM and upon corticosteroid treatment of CSM-affected patients will aid in further understanding the pathomechanisms underlying this disease. LEVEL OF EVIDENCE N/A.
Collapse
Affiliation(s)
- Paula Martin-Vaquero
- Department of Veterinary Clinical Sciences, The Ohio State University, College of Veterinary Medicine, Columbus, OH
- Dr. Martin-Vaquero’s current address is Centro Médico Veterinario Delicias, Calle Delicias 35, C.P. 28045, Madrid, Spain
| | - Ronaldo C. da Costa
- Department of Veterinary Clinical Sciences, The Ohio State University, College of Veterinary Medicine, Columbus, OH
| | - Matthew J. Allen
- Department of Veterinary Clinical Sciences, The Ohio State University, College of Veterinary Medicine, Columbus, OH
| | - Sarah A. Moore
- Department of Veterinary Clinical Sciences, The Ohio State University, College of Veterinary Medicine, Columbus, OH
| | - Jeremy K. Keirsey
- Mass Spectrometry and Proteomics Facility, The Ohio State University, College of Medicine, Department of Molecular and Cellular Biochemistry, Columbus, OH
| | | |
Collapse
|
5
|
Malekzadeh A, Teunissen C. Recent progress in omics-driven analysis of MS to unravel pathological mechanisms. Expert Rev Neurother 2014; 13:1001-16. [PMID: 24053344 DOI: 10.1586/14737175.2013.835602] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
At present, the pathophysiology and specific biological markers reflecting pathology of multiple sclerosis (MS) remain undetermined. The risk of developing MS is considered to depend on genetic susceptibility and environmental factors. The interaction of environmental factors with epigenetic mechanisms could affect the transcriptional level and therefore also the translational level. In the last decade, growing amount of hypothesis-free 'omics' studies have shed light on the potential MS mechanisms and raised potential biomarker targets. To understand MS pathophysiology and discover a subset of biomarkers, it is becoming essential to take a step forward and integrate the findings of the different fields of 'omics' into a systems biology network. In this review, we will discuss the recent findings of the genomic, transcriptomic and proteomic fields for MS and aim to make a unifying model.
Collapse
Affiliation(s)
- Arjan Malekzadeh
- Department of Clinical Chemistry, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | |
Collapse
|
6
|
Novel roles for osteopontin and clusterin in peripheral motor and sensory axon regeneration. J Neurosci 2014; 34:1689-700. [PMID: 24478351 DOI: 10.1523/jneurosci.3822-13.2014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Previous studies demonstrated that Schwann cells (SCs) express distinct motor and sensory phenotypes, which impact the ability of these pathways to selectively support regenerating neurons. In the present study, unbiased microarray analysis was used to examine differential gene expression in denervated motor and sensory pathways in rats. Several genes that were significantly upregulated in either denervated sensory or motor pathways were identified and two secreted factors were selected for further analysis: osteopontin (OPN) and clusterin (CLU) which were upregulated in denervated motor and sensory pathways, respectively. Sciatic nerve transection induced upregulation of OPN and CLU and expression of both returned to baseline levels with ensuing regeneration. In vitro analysis using exogenously applied OPN induced outgrowth of motor but not sensory neurons. CLU, however, induced outgrowth of sensory neurons, but not motor neurons. To assess the functional importance of OPN and CLU, peripheral nerve regeneration was examined in OPN and CLU(-/-) mice. When compared with OPN(+/+) mice, motor neuron regeneration was reduced in OPN(-/-) mice. Impaired regeneration through OPN(-/-) peripheral nerves grafted into OPN(+/+) mice indicated that loss of OPN in SCs was responsible for reduced motor regeneration. Sensory neuron regeneration was impaired in CLU(-/-) mice following sciatic nerve crush and impaired regeneration nerve fibers through CLU(-/-) nerve grafts transplanted into CLU(+/+) mice indicated that reduced sensory regeneration is likely due to SC-derived CLU. Together, these studies suggest unique roles for SC-derived OPN and CLU in regeneration of peripheral motor and sensory axons.
Collapse
|
7
|
Wu Z, Yang L, Wang Y. Applying Imaging Genetics to ADHD: the Promises and the Challenges. Mol Neurobiol 2014; 50:449-62. [DOI: 10.1007/s12035-014-8683-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 03/13/2014] [Indexed: 12/31/2022]
|
8
|
Development- and activity-dependent expression of clusterin in the mouse olfactory bulb. Int J Dev Neurosci 2013; 31:550-9. [PMID: 23831077 DOI: 10.1016/j.ijdevneu.2013.06.009] [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: 04/30/2013] [Revised: 06/15/2013] [Accepted: 06/25/2013] [Indexed: 11/23/2022] Open
Abstract
Clusterin, a protein involved in many biological processes, is expressed broadly in the central nervous system, but its functions remain largely unknown. As preparations for elucidating some possible functions, we examined the spatiotemporal expression patterns of clusterin in the mouse olfactory bulb at different developmental stages and under different neuronal activity levels. Our results revealed a dynamic expression of the protein during development. Clusterin signal was seemingly diffuse during the early stages of development, shifted to the cell somas later and then predominantly to the axons of projection neurons in the adult stage, with a transition point at approximately postnatal day 18. The effects of olfactory deficits on the clusterin expression level in an anosmic mouse model were neuron-specific: the signals increased remarkably from faint to strong in olfactory sensory neurons, reduced considerably from moderate/strong to faint in the centrifugal projection neurons, decreased moderately from moderate to faint in the local bulbar projection neurons, and remained intense in long-distance bulbar projection neurons. These results showed that clusterin expression is modulated dynamically during development and by sensory activity. These findings deepen our understanding of this broadly expressed protein.
Collapse
|
9
|
Charnay Y, Imhof A, Vallet PG, Kovari E, Bouras C, Giannakopoulos P. Clusterin in neurological disorders: Molecular perspectives and clinical relevance. Brain Res Bull 2012; 88:434-43. [DOI: 10.1016/j.brainresbull.2012.05.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 05/07/2012] [Indexed: 10/28/2022]
|
10
|
The role of clusterin in Alzheimer's disease: pathways, pathogenesis, and therapy. Mol Neurobiol 2012; 45:314-26. [PMID: 22274961 DOI: 10.1007/s12035-012-8237-1] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/12/2012] [Indexed: 10/14/2022]
Abstract
Genetic variation in clusterin gene, also known as apolipoprotein J, has been associated with Alzheimer's disease (AD) through replicated genome-wide studies, and plasma clusterin levels are associated with brain atrophy, baseline prevalence and severity, and rapid clinical progression in patients with AD, highlighting the importance of clusterin in AD pathogenesis. Emerging data suggest that clusterin contributes to AD through various pathways, including amyloid-β aggregation and clearance, lipid metabolism, neuroinflammation, and neuronal cell cycle control and apoptosis. Moreover, epigenetic regulation of the clusterin expression also seems to play an important role in the pathogenesis of AD. Emerging knowledge of the contribution of clusterin to the pathogenesis of AD presents new opportunities for AD therapy.
Collapse
|
11
|
Ding J, List EO, Bower BD, Kopchick JJ. Differential effects of growth hormone versus insulin-like growth factor-I on the mouse plasma proteome. Endocrinology 2011; 152:3791-802. [PMID: 21791560 PMCID: PMC3176651 DOI: 10.1210/en.2011-1217] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 07/07/2011] [Indexed: 12/23/2022]
Abstract
The GH/IGF-I axis has both pre- and postpubertal metabolic effects. However, the differential effects of GH and/or IGF-I on animal physiology or the plasma proteome are still being unraveled. In this report, we analyzed several physiological effects along with the plasma proteome after treatment of mice with recombinant bovine GH or recombinant human IGF-I. GH and IGF-I showed similar effects in increasing body length, body weight, lean and fluid masses, and organ weights including muscle, kidney, and spleen. However, GH significantly increased serum total cholesterol, whereas IGF-I had no effect on it. Both acute and longer-term effects on the plasma proteome were determined. Proteins found to be significantly changed by recombinant bovine GH and/or recombinant human IGF-I injections were identified by mass spectrometry (MS) and MS/MS. The identities of these proteins were further confirmed by Western blotting analysis. Isoforms of apolipoprotein A4, apolipoprotein E, serum amyloid protein A-1, clusterin, transthyretin, and several albumin fragments were found to be differentially regulated by GH vs. IGF-I in mouse plasma. Thus, we have identified several plasma protein biomarkers that respond specifically and differentially to GH or IGF-I and may represent new physiological targets of these hormones. These findings may lead to better understanding of the independent biological effects of GH vs. IGF-I. In addition, these novel biomarkers may be useful for the development of tests to detect illicit use of GH or IGF-I.
Collapse
Affiliation(s)
- Juan Ding
- Edison Biotechnology Institute, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| | | | | | | |
Collapse
|
12
|
Common Alzheimer's disease risk variant within the CLU gene affects white matter microstructure in young adults. J Neurosci 2011; 31:6764-70. [PMID: 21543606 DOI: 10.1523/jneurosci.5794-10.2011] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
There is a strong genetic risk for late-onset Alzheimer's disease (AD), but so far few gene variants have been identified that reliably contribute to that risk. A newly confirmed genetic risk allele C of the clusterin (CLU) gene variant rs11136000 is carried by ∼88% of Caucasians. The C allele confers a 1.16 greater odds of developing late-onset AD than the T allele. AD patients have reductions in regional white matter integrity. We evaluated whether the CLU risk variant was similarly associated with lower white matter integrity in healthy young humans. Evidence of early brain differences would offer a target for intervention decades before symptom onset. We scanned 398 healthy young adults (mean age, 23.6 ± 2.2 years) with diffusion tensor imaging, a variation of magnetic resonance imaging sensitive to white matter integrity in the living brain. We assessed genetic associations using mixed-model regression at each point in the brain to map the profile of these associations with white matter integrity. Each C allele copy of the CLU variant was associated with lower fractional anisotropy--a widely accepted measure of white matter integrity--in multiple brain regions, including several known to degenerate in AD. These regions included the splenium of the corpus callosum, the fornix, cingulum, and superior and inferior longitudinal fasciculi in both brain hemispheres. Young healthy carriers of the CLU gene risk variant showed a distinct profile of lower white matter integrity that may increase vulnerability to developing AD later in life.
Collapse
|
13
|
Braskie MN, Ringman JM, Thompson PM. Neuroimaging measures as endophenotypes in Alzheimer's disease. Int J Alzheimers Dis 2011; 2011:490140. [PMID: 21547229 PMCID: PMC3087508 DOI: 10.4061/2011/490140] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 01/08/2011] [Accepted: 02/07/2011] [Indexed: 01/06/2023] Open
Abstract
Late onset Alzheimer's disease (AD) is moderately to highly heritable. Apolipoprotein E allele ε4 (APOE4) has been replicated consistently as an AD risk factor over many studies, and recently confirmed variants in other genes such as CLU, CR1, and PICALM each increase the lifetime risk of AD. However, much of the heritability of AD remains unexplained. AD is a complex disease that is diagnosed largely through neuropsychological testing, though neuroimaging measures may be more sensitive for detecting the incipient disease stages. Difficulties in early diagnosis and variable environmental contributions to the disease can obscure genetic relationships in traditional case-control genetic studies. Neuroimaging measures may be used as endophenotypes for AD, offering a reliable, objective tool to search for possible genetic risk factors. Imaging measures might also clarify the specific mechanisms by which proposed risk factors influence the brain.
Collapse
Affiliation(s)
- Meredith N Braskie
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, 635 Charles Young Drive South, Suite 225, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
14
|
Aronovich R, Katzav A, Chapman J. The Strategies Used for Treatment of Experimental Autoimmune Neuritis (EAN): A Beneficial Effect of Glatiramer Acetate Administered Intraperitoneally. Clin Rev Allergy Immunol 2011; 42:181-8. [DOI: 10.1007/s12016-010-8246-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
15
|
Sleegers K, Lambert JC, Bertram L, Cruts M, Amouyel P, Van Broeckhoven C. The pursuit of susceptibility genes for Alzheimer's disease: progress and prospects. Trends Genet 2010; 26:84-93. [PMID: 20080314 DOI: 10.1016/j.tig.2009.12.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 12/10/2009] [Accepted: 12/11/2009] [Indexed: 11/19/2022]
Abstract
The recent discoveries in genome-wide association studies (GWAS) of novel susceptibility loci (CLU, CR1 and PICALM) for Alzheimer's disease (AD) have elicited considerable interest in the AD community. But what are the implications of these purely epidemiological findings for our understanding of disease etiology and patient care? In this review, we attempt to place these findings in the context of current and future AD genetics research. CLU, CR1 and PICALM support existing hypotheses about the amyloid, lipid, chaperone and chronic inflammatory pathways in AD pathogenesis. We discuss how these and future findings can be translated into efforts to ameliorate patient care by genetic profiling for risk prediction and pharmacogenetics and by guiding drug development.
Collapse
Affiliation(s)
- Kristel Sleegers
- Neurodegenerative Brain Diseases Group, VIB-Department of Molecular Genetics; Universiteitsplein 1, B-2610 Antwerp, Belgium
| | | | | | | | | | | |
Collapse
|
16
|
Nuutinen T, Suuronen T, Kauppinen A, Salminen A. Clusterin: a forgotten player in Alzheimer's disease. ACTA ACUST UNITED AC 2009; 61:89-104. [PMID: 19651157 DOI: 10.1016/j.brainresrev.2009.05.007] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 05/13/2009] [Accepted: 05/14/2009] [Indexed: 11/16/2022]
Abstract
Clusterin, also known as apolipoprotein J, is a versatile chaperone molecule which contains several amphipathic and coiled-coil alpha-helices, typical characteristics of small heat shock proteins. In addition, clusterin has three large intrinsic disordered regions, so-called molten globule domains, which can stabilize stressed protein structures. Twenty years ago, it was demonstrated that the expression of clusterin was clearly increased in Alzheimer's disease (AD). Later it was observed that clusterin can bind amyloid-beta peptides and prevent their fibrillization. Clusterin is also involved in the clearance of amyloid-beta peptides and fibrils by binding to megalin receptors and enhancing their endocytosis within glial cells. Clusterin is a complement inhibitor and can suppress complement activation observed in AD. Clusterin is also present in lipoprotein particles and regulates cholesterol and lipid metabolism of brain which is disturbed in AD. Clusterin is a stress-induced chaperone which is normally secreted but in conditions of cellular stress, it can be transported to cytoplasm where it can bind to Bax protein and inhibit neuronal apoptosis. Clusterin can also bind to Smad2/3 proteins and potentiate the neuroprotective TGFbeta signaling. An alternative splicing can produce a variant isoform of clusterin which can be translocated to nuclei where it induces apoptosis. The role of nuclear clusterin in AD needs to be elucidated. We will review here the extensive literature linking clusterin to AD and examine the recent progress in clusterin research with the respect to AD pathology. Though clusterin can be viewed as a multipotent guardian of brain, it is unable to prevent the progressive neuropathology in chronic AD.
Collapse
Affiliation(s)
- Tapio Nuutinen
- Department of Neuroscience and Neurology, University of Kuopio, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | | | | | | |
Collapse
|
17
|
Hakkoum D, Imhof A, Vallet PG, Boze H, Moulin G, Charnay Y, Stoppini L, Aronow B, Bouras C, Giannakopoulos P. Clusterin increases post-ischemic damages in organotypic hippocampal slice cultures. J Neurochem 2008; 106:1791-803. [PMID: 18554319 DOI: 10.1111/j.1471-4159.2008.05519.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Clusterin or apolipoprotein J is a heterodimeric glycoprotein which is known to be increased during tissue involution in response to hormonal changes or injury and under circumstances leading to apoptosis. Previous studies in wild-type (WT) and clusterin-null (Clu-/-) mice indicated a protective role of clusterin over-expression in astrocytes lasting up to 90 days post-ischemia. However, in in vitro and in vivo models of neonatal hypoxia-ischemia, clusterin exacerbates necrotic cell death. We developed recombinant forms of clusterin and examined their effect on propidium iodide uptake, neuronal and synaptic markers as well as electrophysiological recordings in hippocampal slice cultures from Clu-/- and WT mice subjected to oxygen-glucose deprivation (OGD). WT mice displayed a marked up-regulation of clusterin associated with electrophysiological deficits and dramatic increase of propidium iodide uptake 5 days post-OGD. Immunocytochemical and western blot analyses revealed a substantial decrease of neuronal nuclei and synaptophysin immunoreactivity that predominated in WT mice. These findings contrasted with the relative post-OGD resistance of Clu-/- mice. The addition of biologically active recombinant forms of human clusterin for 24 h post-OGD led to the abolishment of the ischemic tolerance in Clu-/- slices. This deleterious effect of clusterin was reverted by the concomitant administration of the NMDA receptor antagonist, d-2-amino-5-phosphonopentanoate. The present data indicate that in an in vitro model of ischemia characterized by the predominance of NMDA-mediated cell death, clusterin exerts a negative effect on the structural integrity and functionality of hippocampal neurons.
Collapse
Affiliation(s)
- David Hakkoum
- Department of Psychiatry, University Hospital and Faculty of Medicine of Geneva, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Charnay Y, Imhof A, Vallet PG, Hakkoum D, Lathuiliere A, Poku N, Aronow B, Kovari E, Bouras C, Giannakopoulos P. Clusterin expression during fetal and postnatal CNS development in mouse. Neuroscience 2008; 155:714-24. [PMID: 18620027 DOI: 10.1016/j.neuroscience.2008.06.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 05/21/2008] [Accepted: 06/06/2008] [Indexed: 01/10/2023]
Abstract
Clusterin (or apolipoprotein J) is a widely distributed multifunctional glycoprotein involved in CNS plasticity and post-traumatic remodeling. Using biochemical and morphological approaches, we investigated the clusterin ontogeny in the CNS of wild-type (WT) mice and explored developmental consequences of clusterin gene knock-out in clusterin null (Clu-/-) mice. A punctiform expression of clusterin mRNA was detected through the hypothalamic region, neocortex and hippocampus at embryonic stages E14/E15. From embryonic stage E16 to the first week of the postnatal life, the vast majority of CNS neurons expressed low levels of clusterin mRNA. In contrast, a very strong hybridizing signal mainly localized in pontobulbar and spinal cord motor nuclei was observed from the end of the first postnatal week to adulthood. Astrocytes expressing clusterin mRNA were often detected through the hippocampus and neocortex in neonatal mice. Real-time polymerase chain amplification and clusterin-immunoreactivity dot-blot analyses indicated that clusterin levels paralleled mRNA expression. Comparative analyses between WT and Clu-/- mice during postnatal development showed no significant differences in brain weight, neuronal, synaptic and astrocyte markers as well myelin basic protein expression. However, quantitative estimation of large motor neuron populations in the facial nucleus revealed a significant deficit in motor cells (-16%) in Clu-/- compared with WT mice. Our data suggest that clusterin expression is already present in fetal life mainly in subcortical structures. Although the lack of this protein does not significantly alter basic aspects of the CNS development, it may have a negative impact on neuronal development in certain motor nuclei.
Collapse
Affiliation(s)
- Y Charnay
- Division of Neuropsychiatry, Department of Psychiatry, University Hospitals of Geneva, 2, Ch du petit-Bel-Air, CH-1225 Chene-Bourg, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Neuroprotection: VEGF, IL-6, and clusterin: the dark side of the moon. PROGRESS IN BRAIN RESEARCH 2008; 173:555-73. [PMID: 18929134 DOI: 10.1016/s0079-6123(08)01138-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Growth factors and their respective receptors are key regulators in development and homeostasis of the nervous system, and changes in the function, expression, or downstream signaling of growth factors are involved in many neuropathological disorders. Recently, research has yielded a rich harvest of information about molecules and gene, and currently the assumption "a gene-a protein", where each gene encodes the structure of a single protein, is becoming a paradox. In the past years, the discovery of synergic or antagonistic proteins deriving from the same gene is a novelty upsetting. In some way, the conventional function of proteins involved in DNA repair, cell death/growth induction, vascularization, and metabolism is inhibited or shifted toward other pathways by soluble mediators that orchestrate such change depending on the microenvironment conditions. In this chapter, we focus on the antithetic properties that proteins could exert, depending on the microenvironment that orchestrates the complex networks among proteins and their respective partners.
Collapse
|
20
|
Créange A. Should targeting immunosuppression, immunoregulation or remyelination be used to treat inflammatory neuropathies? J Neuroimmunol 2007; 190:3-4. [PMID: 17910984 DOI: 10.1016/j.jneuroim.2007.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Accepted: 08/07/2007] [Indexed: 11/16/2022]
|