1
|
Jiang Z, Li M, Wang K, Duan H, Zhang B, Fang S. Potential Biomarkers of Post-stroke Cognitive Impairment in Chinese Population: a Systematic Review and Meta-Analysis. Mol Neurobiol 2025:10.1007/s12035-025-04792-x. [PMID: 40032750 DOI: 10.1007/s12035-025-04792-x] [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: 03/23/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
It is still controversial whether peripheral blood biomarkers have the potential to be diagnostic, prognostic, and therapeutic targets for post-stroke cognitive impairment (PSCI). All studies reporting the correlation between peripheral blood biomarkers and PSCI in Chinese acute ischemic stroke (AIS) patients were screened in eight databases and meta-analyses were performed to explore their predictive value for PSCI. The results showed that the levels of C-reactive protein (CRP), fasting blood glucose (FBG), homocysteine (Hcy), and cystatin C (CysC) were significantly higher in the PSCI group than in the post-stroke cognitive impairment no dementia (PSNCI) group. However, the differences in glycosylated hemoglobin (HbA1c), creatinine (Cr), blood urea nitrogen (BUN), and uric acid (UA) levels were not significant. The correlation between Hcy and PSCI applies to all AIS patients, whereas the correlation between CRP (p < 0.001), FBG (p = 0.005), CysC (p = 0.005), and PSCI is generalizable only to first-onset AIS. CRP may be a biomarker of cognitive impairment 3-6 months after AIS (3 months: p < 0.001; 6 months: p = 0.030) and does not appear to have a correlation in the long term. However, the correlation between FBG and PSCI may be significant 6 months to 1 year after AIS (6 months: p = 0.032; 1 year: p = 0.004), whereas the correlation between Hcy and PSCI may be significant 3 months to 1 year after AIS (3 months: p = 0.002; 6 months: p = 0.004; 1 year: p = 0.004). CRP, FBG, Hcy, and CysC may be potential biomarkers for PSCI, whereas the correlation between Cr, BUN, UA, and PSCI has not been confirmed.
Collapse
Affiliation(s)
- Zhuoya Jiang
- Department of Neurology, Neuroscience Centre, the First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, China
| | - Min Li
- Department of Neurology, Neuroscience Centre, the First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, China
| | - Kunyu Wang
- Department of Neurology, Neuroscience Centre, the First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, China
| | - Hanying Duan
- Department of Neurology, Neuroscience Centre, the First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, China
| | - Beilin Zhang
- Department of Neurology, Neuroscience Centre, the First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, China
| | - Shaokuan Fang
- Department of Neurology, Neuroscience Centre, the First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
2
|
Jiang SS, Nie HB, Hua S, Xie M, Xu RS. Preliminary Analysis of Potentially Overlapping Differentially Expressed Proteins in Both the Spinal Cord and Brain of SOD1 G93A Mice. Curr Protein Pept Sci 2025; 26:57-75. [PMID: 38984582 DOI: 10.2174/0113892037293525240621120033] [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: 01/26/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE Proteomic elucidation is an essential step in improving our understanding of the biological properties of proteins in amyotrophic lateral sclerosis (ALS). METHODS Preliminary proteomic analysis was performed on the spinal cord and brain of SOD1 G93A (TG) and wild-type (WT) mice using isobaric tags for relative and absolute quantitation. RESULTS Partial up- and downregulated proteins showing significant differences between TG and WT mice were identified, of which 105 proteins overlapped with differentially expressed proteins in both the spinal cord and brain of progression mice. Bioinformatic analyses using Gene Ontology, a cluster of orthologous groups, and Kyoto Encyclopedia of Genes and Genomes pathway revealed that the significantly up- and downregulated proteins represented multiple biological functions closely related to ALS, with 105 overlapping differentially expressed proteins in the spinal cord and brain at the progression stage of TG mice closely related to 122 pathways. Differentially expressed proteins involved in a set of molecular functions play essential roles in maintaining neural cell survival. CONCLUSION This study provides additional proteomic profiles of TG mice, including potential overlapping proteins in both the spinal cord and brain that participate in pathogenesis, as well as novel insights into the up- and downregulation of proteins involved in the pathogenesis of ALS.
Collapse
Affiliation(s)
- Shi-Shi Jiang
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Hong-Bing Nie
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Shan Hua
- Department of Ultrasonography, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Meng Xie
- Health Management Center, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Ren-Shi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
3
|
Zhou Y, Dong W, Wang L, Ren S, Wei W, Wu G. Cystatin C Attenuates Perihematomal Secondary Brain Injury by Inhibiting the Cathepsin B/NLRP3 Signaling Pathway in a Rat Model of Intracerebral Hemorrhage. Mol Neurobiol 2024; 61:9646-9662. [PMID: 38676809 DOI: 10.1007/s12035-024-04195-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
Secondary brain injury (SBI) is a noticeable contributor to the high mortality and morbidity rates associated with intracerebral hemorrhage (ICH), and effective treatment options remain limited. Cystatin C (CysC) emerges as a novel candidate for SBI intervention. The therapeutic effects and underlying mechanisms of CysC in mitigating SBI following ICH were explored in the current research. An in vivo ICH rat model was established by injecting autologous blood into the right caudate nucleus. Western blotting (WB) was utilized to assess the levels of CysC, cathepsin B (CTSB), and the NLRP3 inflammasome. Subsequently, the ICH rat model was treated with exogenous CysC supplementation or CysC knockdown plasmids. Various parameters, including Evans blue (EB) extravasation, brain water content, and neurological function in rats, were examined. RT-qPCR and WB were employed to determine the expression levels of CTSB and the NLRP3 inflammasome. The co-expression of CTSB, CysC, and NLRP3 inflammasome with GFAP, NeuN, and Iba1 was assessed through double-labeled immunofluorescence. The interaction between CysC and CTSB was investigated using double-labeled immunofluorescence and co-immunoprecipitation. The findings revealed an elevation of CysC expression level, particularly at 24 h after ICH. Exogenous CysC supplementation alleviated severe brain edema, neurological deficit scores, and EB extravasation induced by ICH. Conversely, CysC knockdown produced opposite effects. The expression levels of CTSB and the NLRP3 inflammasome were significantly risen following ICH, and exogenous CysC supplement attenuated their expression levels. Double-labeled immunofluorescence illustrated that CysC, CTSB, and the NLRP3 inflammasome were predominantly expressed in microglial cells, and the interaction between CysC and CTSB was evidenced. CysC exhibited potential in ameliorating SBI following ICH via effectively suppressing the activation of the NLRP3 inflammasome mediated by CTSB specifically in microglial cells. These findings underscore the prospective therapeutic efficacy of CysC in the treatment of ICH-induced complications.
Collapse
Affiliation(s)
- Yongfang Zhou
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Wentao Dong
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Likun Wang
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Siying Ren
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Weiqing Wei
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
4
|
Jang YO, Ahn HS, Dao TNT, Hong J, Shin W, Lim YM, Chung SJ, Lee JH, Liu H, Koo B, Kim MG, Kim K, Lee EJ, Shin Y. Magnetic transferrin nanoparticles (MTNs) assay as a novel isolation approach for exosomal biomarkers in neurological diseases. Biomater Res 2023; 27:12. [PMID: 36797805 PMCID: PMC9936675 DOI: 10.1186/s40824-023-00353-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/05/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Brain-derived exosomes released into the blood are considered a liquid biopsy to investigate the pathophysiological state, reflecting the aberrant heterogeneous pathways of pathological progression of the brain in neurological diseases. Brain-derived blood exosomes provide promising prospects for the diagnosis of neurological diseases, with exciting possibilities for the early and sensitive diagnosis of such diseases. However, the capability of traditional exosome isolation assays to specifically isolate blood exosomes and to characterize the brain-derived blood exosomal proteins by high-throughput proteomics for clinical specimens from patients with neurological diseases cannot be assured. We report a magnetic transferrin nanoparticles (MTNs) assay, which combined transferrin and magnetic nanoparticles to isolate brain-derived blood exosomes from clinical samples. METHODS The principle of the MTNs assay is a ligand-receptor interaction through transferrin on MTNs and transferrin receptor on exosomes, and electrostatic interaction via positively charged MTNs and negatively charged exosomes to isolate brain-derived blood exosomes. In addition, the MTNs assay is simple and rapid (< 35 min) and does not require any large instrument. We confirmed that the MTNs assay accurately and efficiently isolated exosomes from serum samples of humans with neurodegenerative diseases, such as dementia, Parkinson's disease (PD), and multiple sclerosis (MS). Moreover, we isolated exosomes from serum samples of 30 patients with three distinct neurodegenerative diseases and performed unbiased proteomic analysis to explore the pilot value of brain-derived blood protein profiles as biomarkers. RESULTS Using comparative statistical analysis, we found 21 candidate protein biomarkers that were significantly different among three groups of neurodegenerative diseases. CONCLUSION The MTNs assay is a convenient approach for the specific and affordable isolation of extracellular vesicles from body fluids for minimally-invasive diagnosis of neurological diseases.
Collapse
Affiliation(s)
- Yoon Ok Jang
- grid.15444.300000 0004 0470 5454Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Republic of Korea
| | - Hee-Sung Ahn
- grid.413967.e0000 0001 0842 2126Department of Convergence Medicine, Asan Medical Center, Seoul, 05505 Republic of Korea
| | - Thuy Nguyen Thi Dao
- grid.15444.300000 0004 0470 5454Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Republic of Korea
| | - JeongYeon Hong
- grid.413967.e0000 0001 0842 2126Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505 Republic of Korea ,grid.267370.70000 0004 0533 4667Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 05505 Republic of Korea
| | - Wangyong Shin
- grid.413967.e0000 0001 0842 2126Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505 Republic of Korea
| | - Young-Min Lim
- grid.413967.e0000 0001 0842 2126Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505 Republic of Korea
| | - Sun Ju Chung
- grid.413967.e0000 0001 0842 2126Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505 Republic of Korea
| | - Jae-Hong Lee
- grid.413967.e0000 0001 0842 2126Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505 Republic of Korea
| | - Huifang Liu
- grid.15444.300000 0004 0470 5454Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Republic of Korea
| | - Bonhan Koo
- grid.15444.300000 0004 0470 5454Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Republic of Korea
| | - Myoung Gyu Kim
- grid.15444.300000 0004 0470 5454Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Republic of Korea
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea. .,Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Eun-Jae Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Yong Shin
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
5
|
Bruno F, Malvaso A, Canterini S, Bruni AC. Antimicrobial Peptides (AMPs) in the Pathogenesis of Alzheimer's Disease: Implications for Diagnosis and Treatment. Antibiotics (Basel) 2022; 11:726. [PMID: 35740133 PMCID: PMC9220182 DOI: 10.3390/antibiotics11060726] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) represents the most frequent type of dementia in elderly people. There are two major forms of the disease: sporadic (SAD)-whose causes are not completely understood-and familial (FAD)-with clear autosomal dominant inheritance. The two main hallmarks of AD are extracellular deposits of amyloid-beta (Aβ) peptide and intracellular deposits of the hyperphosphorylated form of the tau protein (P-tau). An ever-growing body of research supports the infectious hypothesis of sporadic forms of AD. Indeed, it has been documented that some pathogens, such as herpesviruses and certain bacterial species, are commonly present in AD patients, prompting recent clinical research to focus on the characterization of antimicrobial peptides (AMPs) in this pathology. The literature also demonstrates that Aβ can be considered itself as an AMP; thus, representing a type of innate immune defense peptide that protects the host against a variety of pathogens. Beyond Aβ, other proteins with antimicrobial activity, such as lactoferrin, defensins, cystatins, thymosin β4, LL37, histatin 1, and statherin have been shown to be involved in AD. Here, we summarized and discussed these findings and explored the diagnostic and therapeutic potential of AMPs in AD.
Collapse
Affiliation(s)
- Francesco Bruno
- Regional Neurogenetic Centre (CRN), Department of Primary Care, ASP Catanzaro, 88046 Lamezia Terme, Italy
- Association for Neurogenetic Research (ARN), 88046 Lamezia Terme, Italy;
| | - Antonio Malvaso
- Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Sonia Canterini
- Division of Neuroscience, Department of Psychology, University La Sapienza, 00158 Rome, Italy;
| | | |
Collapse
|
6
|
Cao B, Luo M, Li J, Lu Y, Chang Y, Chen Z, Li R, Hu B, Lu Z, Qiu W, Shu Y. Cerebrospinal fluid cystatin C levels in patients with anti-NMDAR encephalitis and other neurological diseases. J Neuroimmunol 2022; 369:577900. [DOI: 10.1016/j.jneuroim.2022.577900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/29/2022] [Accepted: 05/22/2022] [Indexed: 11/29/2022]
|
7
|
Stanczykiewicz B, Gburek J, Rutkowska M, Lemieszewska M, Gołąb K, Juszczyńska K, Piotrowska A, Trziszka T, Dzięgiel P, Podhorska-Okołów M, Zabłocka A, Rymaszewska J. Ovocystatin Induced Changes in Expression of Alzheimer's Disease Relevant Proteins in APP/PS1 Transgenic Mice. J Clin Med 2022; 11:jcm11092372. [PMID: 35566501 PMCID: PMC9103311 DOI: 10.3390/jcm11092372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/08/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Background: Ovocystatin is marked by structural and biological similarities to human cystatin C, which plays an important role in the course of neurodegenerative diseases. Recently, it has been shown that ovocystatin might prevent aging-related cognitive impairment in rats and reduce memory decline in an APP/PS1 mice model. Thus, this study aimed to assess the effect of ovocystatin on histopathological changes in APP/PS1 mice. Materials and methods: Ovocystatin was administered intraperitoneally for four weeks (40 μg/mouse) to 35-weeks-old transgenic (AD, n = 14) and wild type (NCAR, n = 15) mice (stock B6C3-Tg(APPswe, PSEN1dE9)85Dbo/Mmjax). A histopathological evaluation comprised antibodies directed against β-amyloid (1:400, SIG-39320-1000, Covance) and Tau (1:4000, AHB0042, Invitrogen). Three regions of the hippocampus— the dentate gyrus (DG) and the cornu ammonis (CA1 and CA3)—were analyzed by immunohistochemistry in each animal. All differences are expressed as percentage relative to the control group. Results: The main results showed that the percentage of immunoreactive area of β-amyloid, tau protein deposits in APP/PS1+ovCYS was decreased in DG, CA1, and CA3 regions compared with the APP/PS1 control, respectively (p < 0.05). Conclusions: Ovocystatin caused significant changes in the expression pattern of all investigated proteins in hippocampal tissues both in APP/PS1 and NCAR mice.
Collapse
Affiliation(s)
- Bartlomiej Stanczykiewicz
- Department of Psychiatry, Division of Consultation Psychiatry and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.L.); (J.R.)
- Correspondence: ; Tel.: +48-71-784-1600
| | - Jakub Gburek
- Department of Pharmaceutical Biochemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.G.); (K.G.); (K.J.)
| | - Maria Rutkowska
- Department of Pharmacology, Wroclaw Medical University, 50-345 Wroclaw, Poland;
| | - Marta Lemieszewska
- Department of Psychiatry, Division of Consultation Psychiatry and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.L.); (J.R.)
| | - Krzysztof Gołąb
- Department of Pharmaceutical Biochemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.G.); (K.G.); (K.J.)
| | - Katarzyna Juszczyńska
- Department of Pharmaceutical Biochemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.G.); (K.G.); (K.J.)
| | - Aleksandra Piotrowska
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.P.); (P.D.)
| | - Tadeusz Trziszka
- Department of Animal Products Technology and Quality Management, Wroclaw University of Environmental and Life Sciences, 51-630 Wroclaw, Poland;
| | - Piotr Dzięgiel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.P.); (P.D.)
- Department of Human Biology, Faculty of Physiotherapy, University School of Physical Education, 51-612 Wroclaw, Poland
| | | | - Agnieszka Zabłocka
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Joanna Rymaszewska
- Department of Psychiatry, Division of Consultation Psychiatry and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.L.); (J.R.)
| |
Collapse
|
8
|
Katzeff JS, Bright F, Phan K, Kril JJ, Ittner LM, Kassiou M, Hodges JR, Piguet O, Kiernan MC, Halliday GM, Kim WS. Biomarker discovery and development for frontotemporal dementia and amyotrophic lateral sclerosis. Brain 2022; 145:1598-1609. [PMID: 35202463 PMCID: PMC9166557 DOI: 10.1093/brain/awac077] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 11/12/2022] Open
Abstract
Frontotemporal dementia refers to a group of neurodegenerative disorders characterized by behaviour and language alterations and focal brain atrophy. Amyotrophic lateral sclerosis is a rapidly progressing neurodegenerative disease characterized by loss of motor neurons resulting in muscle wasting and paralysis. Frontotemporal dementia and amyotrophic lateral sclerosis are considered to exist on a disease spectrum given substantial overlap of genetic and molecular signatures. The predominant genetic abnormality in both frontotemporal dementia and amyotrophic lateral sclerosis is an expanded hexanucleotide repeat sequence in the C9orf72 gene. In terms of brain pathology, abnormal aggregates of TAR-DNA-binding protein-43 are predominantly present in frontotemporal dementia and amyotrophic lateral sclerosis patients. Currently, sensitive and specific diagnostic and disease surveillance biomarkers are lacking for both diseases. This has impeded the capacity to monitor disease progression during life and the development of targeted drug therapies for the two diseases. The purpose of this review is to examine the status of current biofluid biomarker discovery and development in frontotemporal dementia and amyotrophic lateral sclerosis. The major pathogenic proteins implicated in different frontotemporal dementia and amyotrophic lateral sclerosis molecular subtypes and proteins associated with neurodegeneration and the immune system will be discussed. Furthermore, the use of mass spectrometry-based proteomics as an emerging tool to identify new biomarkers in frontotemporal dementia and amyotrophic lateral sclerosis will be summarized.
Collapse
Affiliation(s)
- Jared S Katzeff
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia
| | - Fiona Bright
- The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia.,Dementia Research Centre and Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Katherine Phan
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia
| | - Jillian J Kril
- The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia.,Dementia Research Centre and Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Lars M Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Michael Kassiou
- The University of Sydney, School of Chemistry, Sydney, NSW, Australia
| | - John R Hodges
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia
| | - Olivier Piguet
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Psychology, Sydney, NSW, Australia
| | - Matthew C Kiernan
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Glenda M Halliday
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia
| | - Woojin Scott Kim
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia.,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia
| |
Collapse
|
9
|
Cystatin C Plays a Sex-Dependent Detrimental Role in Experimental Autoimmune Encephalomyelitis. Cell Rep 2021; 33:108236. [PMID: 33027652 PMCID: PMC8603395 DOI: 10.1016/j.celrep.2020.108236] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/17/2020] [Accepted: 09/15/2020] [Indexed: 12/31/2022] Open
Abstract
The cysteine protease inhibitor Cystatin C (CST3) is highly expressed in the brains of multiple sclerosis (MS) patients and C57BL/6J mice with experimental autoimmune encephalomyelitis (EAE; a model of MS), but its roles in the diseases are unknown. Here, we show that CST3 plays a detrimental function in myelin oligodendrocyte glycoprotein 35–55 (MOG35–55)-induced EAE but only in female animals. Female Cst3 null mice display significantly lower clinical signs of disease compared to wild-type (WT) littermates. This difference is associated with reduced interleukin-6 production and lower expression of key proteins (CD80, CD86, major histocompatibility complex [MHC] II, LC3A/B) involved in antigen processing, presentation, and co-stimulation in antigen-presenting cells (APCs). In contrast, male WT and Cst3−/− mice and cells show no differences in EAE signs or APC function. Further, the sex-dependent effect of CST3 in EAE is sensitive to gonadal hormones. Altogether, we have shown that CST3 has a sex-dependent role in MOG35–55-induced EAE. Cystatin C (CST3) is increased in the brains of multiple sclerosis patients, but its role is unknown. In a mouse model of the disease, Hoghooghi et al. find that CST3 has a detrimental function but only in female animals. The effect is related to activation of antigen-presenting cells of the immune system.
Collapse
|
10
|
Zheng X, She HD, Zhang QX, Si T, Wu KS, Xiao YX. Cystatin C predicts the risk of incident cerebrovascular disease in the elderly: A meta-analysis on survival date studies. Medicine (Baltimore) 2021; 100:e26617. [PMID: 34260548 PMCID: PMC8284707 DOI: 10.1097/md.0000000000026617] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 05/26/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Stroke is the third leading cause of global year of life lost in all-age and second-ranked cause of disability adjusted life years in middle-aged and elder population. Therefore, it is critical to study the relationship between vascular-related risk factors and cerebrovascular diseases. Several cross-sectional studies have shown that Cystatin C (Cys C) is an independent risk factor for cerebrovascular diseases and levels of Cys C are significantly higher in stroke patients than in healthy individuals. In this meta-analysis, we introduce a Cox proportional hazards model to evaluate the causality between Cys C and the risk of cerebrovascular accident in the elderly. METHODS We searched PubMed, EMBASE, the Web of Science, and the Cochrane Library from 1985 to 2019 for studies on the relationship between serum Cys C and incidence stroke with Cox proportional hazards models. We conducted a subgroup analysis of the selected studies to determine a connection between atherosclerosis and stroke. Finally, 7 research studies, including 26,768 patients without a history of cerebrovascular, were studied. RESULTS After comparing the maximum and minimum Cys C levels, the hazard ratio for all types of stroke, including ischemic and hemorrhagic stroke, was 1.18 (95% confidence interval 1.04-1.31) with moderate heterogeneity (I2 = 43.0%; P = .119) in a fixed-effect model after pooled adjustment for other potential risk factors. In the subgroup analysis, the hazard ratio and 95% confidence interval for Cys C stratified by atherosclerosis was 1.85 (0.97-2.72). As shown in Egger linear regression test, there was no distinct publication bias (P = .153). CONCLUSION Increased serum Cys C is significantly associated with future stroke events in the elderly, especially in patients with carotid atherosclerosis. Thus, serum levels of Cys C could serve as a predicted biomarker for stroke attack.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Hong-da She
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Qiao-xin Zhang
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| | - Tong Si
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing
| | - Ku-sheng Wu
- Department of Public and preventive medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Ying-xiu Xiao
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong
| |
Collapse
|
11
|
PENG Q, ZHAO J, WANG P, HUANG C, CHEN B, SONG J, XU G. Expressions of plasma cystatin C, D-dimer and hypersensitive C-reactive protein in patients with intracranial progressive hemorrhagic injury after craniocerebral injury, and their clinical significance. ARQUIVOS DE NEURO-PSIQUIATRIA 2019; 77:381-386. [PMID: 31314839 DOI: 10.1590/0004-282x20190057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 01/20/2019] [Indexed: 11/22/2022]
Abstract
ABSTRACT Objective To investigate the expressions of plasma cystatin C (Cys-C), D-dimer (D-D) and hypersensitive C-reactive protein (hs-CRP) in patients with intracranial progressive hemorrhagic injury (IPHI) after craniocerebral injury, and their clinical significance. Methods Forty-two IPHI patients and 20 healthy participants (control) were enrolled. The severity and outcome of IPHI were determined according to the Glasgow Coma Scale and Glasgow Outcome Scale, and the plasma Cys-C, hs-CRP and D-D levels were measured. Results The plasma Cys-C, D-D and hs-CRP levels in the IPHI group were significantly higher than those in the control group (p < 0.01). There were significant differences of plasma Cys-C, D-D and hs-CRP levels among different IPHI patients according to the Glasgow Coma Scale and according to the Glasgow Outcome Scale (all p < 0.05). In the IPHI patients, the plasma Cys-C, D-D and hs-CRP levels were positively correlated with each other (p < 0.001). Conclusion The increase of plasma Cys-C, D-D and hs-CRP levels may be involved in IPHI after craniocerebral injury. The early detection of these indexes may help to understand the severity and outcome of IPHI.
Collapse
Affiliation(s)
- Qibin PENG
- Southern Medical University, China; Hainan Provincial People’s Hospital, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Zeng Q, Huang Z, Wei L, Fang J, Lin K. Correlations of serum cystatin C level and gene polymorphism with vascular cognitive impairment after acute cerebral infarction. Neurol Sci 2019; 40:1049-1054. [PMID: 30805744 DOI: 10.1007/s10072-019-03777-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/16/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND The aim of this study was to explore the possible correlations of serum cystatin C level and cystatin C gene (CST3) polymorphism with vascular cognitive impairment in patients who had acute cerebral infarction. METHODS A total of 152 patients with acute cerebral infarction were recruited in this case-control study. Patients were divided into vascular cognitive impairment (VCI) group (n = 71) and cognitive impairment no dementia (CIND) group (n = 81). The serum concentrations of cystatin C were measured with immunoturbidimetric assay while the gene polymorphisms of CST3 were determined by technique polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). RESULTS In the VCI group, serum cystatin C level was significantly higher than that in the control group. The frequency of the B allele was found to be higher in the VCI group as compared with that of the CIND group (18.5% vs 7.7%, p = 0.006). In logistic regression analysis, significant associations of VCI with high serum cystatin C level (OR 3.837 (1.176-12.520), p = 0.026) and CST3 B allele (OR 2.038 (1.048-3.963), p = 0.036) were also found. CONCLUSIONS A high cystatin C level and CST3 B allele confer risks for VCI after acute cerebral infarction. It is probable that measurement of the serum cystatin C level and detection of CST3 gene polymorphism would aid in the early diagnosis of VCI, but further studies are warranted.
Collapse
Affiliation(s)
- Qiong Zeng
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zhihua Huang
- Shantou University Medical College, Shantou, China
| | - Liling Wei
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jingnian Fang
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Kun Lin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
13
|
Stańczykiewicz B, Jakubik-Witkowska M, Rutkowska M, Polanowski A, Gburek J, Gołąb K, Juszczyńska K, Trziszka T, Rymaszewska J. Beneficial effect of ovocystatin on the cognitive decline in APP/PS1 transgenic mice. Adv Med Sci 2019; 64:65-71. [PMID: 30504006 DOI: 10.1016/j.advms.2018.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/16/2018] [Accepted: 08/17/2018] [Indexed: 10/27/2022]
Abstract
PURPOSE Cystatin C plays an important role in the course of neurodegenerative diseases and has a beneficial effect through inhibiting cysteine proteases and amyloid-β aggregation. It also induces proliferation and autophagy. Cystatin isolated from chicken egg white, called ovocystatin, has been widely used in the medical and pharmaceutical research due to its structural and biological similarities to human cystatin C. The aim of this study was to assess the effect of administering ovocystatin on the development of dementia-specific cognitive deficits in APP/PS1 transgenic mice. MATERIALS/METHODS The study was conducted on transgenic B6C3-Tg(APPswe,PSEN1dE9)85Dbo/Mmjax mice. Ovocystatin was administered to four-month-old transgenic (AD) and wild type (NCAR) mice in drinking water for 24 weeks (at a dose of 40 and 4 μg/ mouse). The locomotor activity and cognitive functions were determined using an actimeter and the Morris water maze test, respectively. RESULTS The results of the study indicate that ovocystatin has a beneficial effect on the cognitive functions in APP/PS1 transgenic mice. The strongest effects of ovocystatin were found in the group of AD mice, where ovocystatin was administered in drinking water at a dose of 40 μg/mouse (p < 0.05). Mice from the AD group swam statistically significantly further in the target zone during the trial in the Morris water maze compared to the AD (vehiculum) group (p < 0.05). CONCLUSIONS The obtained results encourage further research into the protective effect, which may be used as an adjuvant in the treatment of deteriorating cognitive functions.
Collapse
|
14
|
Cystatin C promotes tau protein phosphorylation and causes microtubule instability by inhibiting intracellular turnover of GSK3β in neurons. Mol Cell Neurosci 2018; 89:1-8. [PMID: 29577984 DOI: 10.1016/j.mcn.2018.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
In Alzheimer's disease (AD) tau protein hyperphosphorylation causes neurofibrillary tangle formation, microtubule instability and neurodegeneration. Determining the mechanism of tau hyperphosphorylation will provide a better understanding of AD pathology. Cystatin C (CysC) is a risk factor for late-onset AD and its level is upregulated in the brains of AD patients. The role of CysC is AD pathogenesis is not known. In this study, we found that CysC level is upregulated in 3xTg-AD mouse brain. We demonstrate that CysC does not affect cellular Aβ production. However, when overexpressed in neuron (NGF-differentiated PC12 cells), CysC inhibits turnover of GSK3β, promotes GSK3β-catalyzed tau phosphorylation at Ser396/404 and causes microtubule instability. Our data provide a novel insight into the role of CysC in AD pathogenesis.
Collapse
|
15
|
Perlenfein TJ, Murphy RM. A mechanistic model to predict effects of cathepsin B and cystatin C on β-amyloid aggregation and degradation. J Biol Chem 2017; 292:21071-21082. [PMID: 29046353 DOI: 10.1074/jbc.m117.811448] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/13/2017] [Indexed: 12/21/2022] Open
Abstract
β-Amyloid (Aβ) aggregation is thought to initiate a cascade of neurodegenerative events in Alzheimer's disease (AD). Much effort is underway to develop strategies to reduce Aβ concentration or inhibit aggregation. Cathepsin B (CatB) proteolytically degrades Aβ into non-aggregating fragments but is potently inhibited by cystatin C (CysC). It has been suggested that decreasing CysC would facilitate Aβ clearance by relieving CatB inhibition. However, CysC binds Aβ and inhibits Aβ aggregation, suggesting that an intervention that increases CysC would prevent Aβ aggregation. Both approaches have been tested in animal models, yielding contradictory results, possibly because of the opposing influences of CysC on Aβ degradation versus aggregation. Here, we sought to develop a model that quantitatively predicts the effects of CysC and CatB on Aβ aggregation. Aβ aggregation kinetics in the absence of CatB or CysC was measured. The rate constant for Aβ degradation by CatB and the equilibrium constant for binding of CysC to Aβ were determined. We derived a mathematical model that combines material balances and kinetic rate equations. The model accurately predicted Aβ aggregation kinetics at various CatB and CysC concentrations. We derived approximate expressions for the half-times of degradation and aggregation and show that their ratio can be used to estimate, at any given Aβ, CatB, or CysC concentration, whether Aβ aggregation or degradation will result. Our results may be useful for designing experiments and interpreting results from investigations of manipulation of CysC concentration as an AD therapy.
Collapse
Affiliation(s)
- Tyler J Perlenfein
- From the Department of Chemical and Biological Engineering, University of Wisconsin, Madison, Wisconsin 53706
| | - Regina M Murphy
- From the Department of Chemical and Biological Engineering, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
16
|
Deckers K, Camerino I, van Boxtel MPJ, Verhey FRJ, Irving K, Brayne C, Kivipelto M, Starr JM, Yaffe K, de Leeuw PW, Köhler S. Dementia risk in renal dysfunction: A systematic review and meta-analysis of prospective studies. Neurology 2016; 88:198-208. [PMID: 27974647 DOI: 10.1212/wnl.0000000000003482] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 09/29/2016] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Renal dysfunction has been linked with increased risk for cognitive impairment and dementia, but studies are conflicting. For that reason, the aim of the present systematic review and meta-analysis is to summarize the best available evidence on the prospective association between potential markers of renal dysfunction and development of cognitive impairment or dementia. METHODS Medline, Embase, and Cochrane Database of Systematic Reviews were searched for potential publications until August 1, 2016. Studies were eligible if they fulfilled the following criteria: population-based study, prospective design, ≥100 participants, aged ≥45 years, ≥1 year follow-up, and cognition/dementia outcomes. Where appropriate, random effects meta-analyses were conducted yielding pooled odds ratios (OR) and 95% confidence intervals (CI). RESULTS Twenty-two out of 8,494 abstracts fulfilled the eligibility criteria. Sufficient evidence was found for albuminuria, mixed results for estimated glomerular filtration rate (eGFR), insufficient support for cystatin C, and tentative evidence for serum creatinine and creatinine clearance. Meta-analyses of 5 studies representing 27,805 persons showed a 35% increased risk of cognitive impairment or dementia in those with albuminuria (OR 1.35, 95% CI 1.06-1.73, p = 0.015), whereas eGFR <60 mL/min/1.73 m2 showed no significant association (OR 1.28, 95% CI 0.99-1.65, p = 0.063). No meta-analyses could be done for serum creatinine, creatinine clearance, or cystatin C. CONCLUSIONS The overall evidence for an association between renal dysfunction and cognitive impairment or dementia is modest. Evidence suggests that albuminuria is associated with higher odds of developing cognitive impairment or dementia.
Collapse
Affiliation(s)
- Kay Deckers
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands.
| | - Ileana Camerino
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - Martin P J van Boxtel
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - Frans R J Verhey
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - Kate Irving
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - Carol Brayne
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - Miia Kivipelto
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - John M Starr
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - Kristine Yaffe
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - Peter W de Leeuw
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| | - Sebastian Köhler
- From Alzheimer Centrum Limburg (K.D., I.C., M.P.J.v.B., F.R.J.V., S.K.), School for Mental Health and Neuroscience, Maastricht University; Alzheimer Centre and Department of Neurology (I.C.), VU University Medical Centre, Amsterdam, the Netherlands; School of Nursing and Human Sciences (K.I.), Dublin City University, Ireland; Department of Public Health and Primary Care (C.B.), University of Cambridge, UK; Alzheimer Disease Research Center (M.K.), Karolinska Institute, Stockholm, Sweden; Centre for Cognitive Ageing and Cognitive Epidemiology (J.M.S.), University of Edinburgh, UK; Departments of Psychiatry, Neurology, Epidemiology and Biostatistics (K.Y.), School of Medicine, University of California, San Francisco; Department of Medicine (P.W.d.L.), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre; and Department of Medicine (P.W.d.L.), Zuyderland MC, Sittard-Geleen/Heerlen, the Netherlands
| |
Collapse
|
17
|
Chen X, Chen Y, Wei Q, Ou R, Cao B, Zhao B, Shang HF. Assessment of a multiple biomarker panel for diagnosis of amyotrophic lateral sclerosis. BMC Neurol 2016; 16:173. [PMID: 27634542 PMCID: PMC5024522 DOI: 10.1186/s12883-016-0689-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 08/29/2016] [Indexed: 02/05/2023] Open
Abstract
Background The aim of the study was to assess a panel of promising biomarkers for their ability to improve diagnosis of sporadic amyotrophic lateral sclerosis (ALS). Methods Forty patients with sporadic ALS and 40 controls with other neurological diseases were evaluated. Levels of phosphorylated neurofilament heavy chain (pNfH), S100-β, cystatin C, and chitotriosidase (CHIT) in cerebrospinal fluid were assayed using two-site solid-phase sandwich ELISA. Results Patients with sporadic ALS showed higher levels of pNfH and CHIT than controls, but lower levels of cystatin C. Multivariate logistic regression that adjusted for patient age and sex identified significant associations between sporadic ALS and levels of pNfH, CHIT and cystatin C. Levels of pNfH correlated positively with rate of progression and decline based on the Amyotrophic Lateral Sclerosis Functional Rating Scale - Revised. Based on receiver operating curve analysis, a pNfH cut-off of 437 ng/L discriminated patients from controls with a sensitivity of 97.3 % and specificity of 83.8 %. A CHIT cut-off of 1593.779 ng/L discriminated patients from controls with a sensitivity of 83.8 % and specificity of 81.1 %. Combining the two biomarkers gave a sensitivity of 83.8 % and specificity of 91.9 %. Conclusions Levels of pNfH in cerebrospinal fluid may be a reliable biomarker for diagnosing ALS, and combining this biomarker with levels of CHIT may improve diagnostic accuracy.
Collapse
Affiliation(s)
- Xueping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bi Zhao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
18
|
Chen X, Shang HF. New developments and future opportunities in biomarkers for amyotrophic lateral sclerosis. Transl Neurodegener 2015; 4:17. [PMID: 26425343 PMCID: PMC4589120 DOI: 10.1186/s40035-015-0040-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/20/2015] [Indexed: 02/05/2023] Open
Abstract
Modern technology has improved the ability to probe effectively the underlying biology of ALS by examination of genomic, proteomic and physiological changes in patients with ALS, as well as to monitor functional and structural changes during the course of disease. While effective treatments for ALS are lacking, the discovery of sensitive biomarkers to disease activity offers clinicians tools for rapid diagnosis and insights into the pathophysiology of ALS. The ultimate aim is to lessen reliance on clinical measures and survival as trial endpoints and broaden the therapeutic options for patients with this disease.
Collapse
Affiliation(s)
- Xueping Chen
- Department of Neurology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan China
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan China
| |
Collapse
|
19
|
Lysophosphatidylcholine increases the neurotoxicity of Alzheimer's amyloid β1-42 peptide: role of oligomer formation. Neuroscience 2015; 292:159-69. [PMID: 25727637 DOI: 10.1016/j.neuroscience.2015.02.034] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 11/24/2022]
Abstract
Oligomer formation is considered as a critical process for the neurotoxic effects of Alzheimer's amyloid β (Aβ) peptide. Previously we have demonstrated that lysophosphatidylcholine (LPC) increases the oligomer formation of Aβ1-42, the major Aβ peptide found Alzheimer's disease (AD) lesions. In this study, we have investigated whether LPC affects the neurotoxic effects of Aβ1-42 in a neuronal cell line (A1) culture. Dimethyl thiazolyl diphenyl tetrazolium (MTT) assay revealed that up to 10μM concentration, LPC did not affect A1 cell viability. Aβ1-42 decreased the cell viability, and such effect was dose dependently enhanced by LPC. However, neither LPC nor Aβ1-42, alone or in combination increased lactate dehydrogenase (LDH) release from A1 cells after 24-h treatment. Terminal deoxynucleotidyl transferase dUTP-biotin nick-end-labeling (TUNEL) assay showed that LPC increased Aβ1-42-induced apoptotic cell number. To determine the underlying mechanisms, the proteins implicated in apoptosis pathways including Bcl-2- and caspase-family were analyzed by Western blotting. The results demonstrated that Aβ1-42 decreased Bcl-2 in A1 cells at 24h, whereas LPC had no effect at any time point. Both LPC and Aβ1-42 increased Bax level at 24h, and their combined stimulation showed a synergistic effect. Similar synergistic effect of LPC and Aβ1-42 on caspase9 activation was observed. Dot blot immunoassay and Western blotting showed that LPC augmented Aβ1-42 oligomer formation in cell culture medium. Removing LPC-induced early-formed Aβ1-42 oligomer from the culture medium by immunoprecipitation decreased active caspase9 level and neurotoxicity, as revealed by Western blotting and MTT assay. Furthermore, dihydroethidium (DHE) assay showed that Aβ1-42 increased reactive oxygen species level in A1 cells, such effect was further enhanced by LPC. Thus, our results demonstrated that LPC increased the oligomer formation process of Aβ1-42 peptide in culture condition, and consequently increased apoptotic neuronal death. Such process might be important for the pathogenesis of AD, and inhibition of LPC generation could be a therapeutic target for the disease.
Collapse
|
20
|
Tabassum S, Sheikh AM, Yano S, Ikeue T, Handa M, Nagai A. A carboxylated Zn-phthalocyanine inhibits fibril formation of Alzheimer's amyloid β peptide. FEBS J 2014; 282:463-76. [PMID: 25404240 DOI: 10.1111/febs.13151] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 11/07/2014] [Accepted: 11/14/2014] [Indexed: 12/31/2022]
Abstract
Amyloid β (Aβ), a 39-42 amino acid peptide derived from amyloid precursor protein, is deposited as fibrils in Alzheimer's disease brains, and is considered to play a major role in the pathogenesis of the disease. We have investigated the effects of a water-soluble Zn-phthalocyanine, ZnPc(COONa)₈, a macrocyclic compound with near-infrared optical properties, on Aβ fibril formation in vitro. A thioflavin T fluorescence assay showed that ZnPc(COONa)₈ significantly inhibited Aβ fibril formation, increasing the lag time and dose-dependently decreasing the plateau level of fibril formation. Moreover, it destabilized pre-formed Aβ fibrils, resulting in an increase in low-molecular-weight species. After fibril formation in the presence of ZnPc(COONa)₈, immunoprecipitation of Aβ₁₋₄₂ using Aβ-specific antibody followed by near-infrared scanning demonstrated binding of ZnPc(COONa)₈ to Aβ₁₋₄₂. A study using the hydrophobic fluorescent probe 8-anilino-1-naphthalenesulfonic acid showed that ZnPc(COONa)8 decreased the hydrophobicity during Aβ₁₋₄₂ fibril formation. CD spectroscopy showed an increase in the α helix structure and a decrease in the β sheet structure of Aβ₁₋₄₀ in fibril-forming buffer containing ZnPc(COONa)₈. SDS/PAGE and a dot-blot immunoassay showed that ZnPc(COONa)₈ delayed the disappearance of low-molecular-weight species and the appearance of higher-molecular-weight oligomeric species of Aβ₁₋₄₂. A cell viability assay showed that ZnPc(COONa)₈ was not toxic to a neuronal cell line (A1), but instead protected A1 cells against Aβ₁₋₄₂-induced toxicity. Overall, our results indicate that ZnPc(COONa)₈ binds to Aβ and decreases the hydrophobicity, and this change is unfavorable for Aβ oligomerization and fibril formation.
Collapse
Affiliation(s)
- Shatera Tabassum
- Department of Laboratory Medicine, Shimane University School of Medicine, Izumo, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Zenón F, Segarra AC, Gonzalez M, Meléndez LM. Cocaine potentiates cathepsin B secretion and neuronal apoptosis from HIV-infected macrophages. J Neuroimmune Pharmacol 2014; 9:703-15. [PMID: 25209871 PMCID: PMC4209444 DOI: 10.1007/s11481-014-9563-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/17/2014] [Indexed: 12/16/2022]
Abstract
Substance abuse is a risk factor for HIV infection and progression to AIDS. Recent evidence establishes that cocaine use promotes brain perivascular macrophage infiltration and microglia activation. The lysosomal protease cathepsin B is increased in monocytes from patients with HIV dementia and its secretion induces 10-15% of neurotoxicity. Here we asked if cocaine potentiates cathepsin B secretion from HIV-infected monocyte-derived macrophages (MDM) and its effect in neuronal apoptosis. Samples of plasma, CSF, and post-mortem brain tissue from HIV positive patients that used cocaine were tested for cathepsin B and its inhibitors to determine the in vivo relevance of these findings. MDM were inoculated with HIV-1ADA, exposed to cocaine, and the levels of secreted and bioactive cathepsin B and its inhibitors were measured at different time-points. Cathepsin B expression (p < 0.001) and activity (p < 0.05) increased in supernatants from HIV-infected cocaine treated MDM compared with HIV-infected cocaine negative controls. Increased levels of cystatin B expression was also found in supernatants from HIV-cocaine treated MDM (p < 0.05). A significant increase in 30% of apoptotic neurons was obtained that decreased to 5% with the specific cathepsin B inhibitor (CA-074) or with cathepsin B antibody. Cathepsin B was significantly increased in the plasma and post-mortem brain tissue of HIV/cocaine users over non-drug users. Our results demonstrated that cocaine potentiates cathepsin B secretion in HIV-infected MDM and increase neuronal apoptosis. These findings provide new evidence that cocaine synergize with HIV-1 infection in increasing cathepsin B secretion and neurotoxicity.
Collapse
Affiliation(s)
- Frances Zenón
- Department of Microbiology and Medical Zoology, School of Medicine, University of Puerto Rico
- NeuroAIDS Program, Medical Sciences Campus, San Juan PR
| | | | | | - Loyda M. Meléndez
- Department of Microbiology and Medical Zoology, School of Medicine, University of Puerto Rico
- NeuroAIDS Program, Medical Sciences Campus, San Juan PR
| |
Collapse
|
22
|
Cheng XW, Sasaki T, Kuzuya M. The role of cysteinyl cathepsins in venous disorders. Thromb Haemost 2014; 112:216-8. [PMID: 24553831 DOI: 10.1160/th13-10-0889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/31/2014] [Indexed: 11/05/2022]
Affiliation(s)
- X W Cheng
- Xian Wu Cheng, MD, PhD, FAHA, Associate Professor of Geriatrics, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan, Tel.: +81 52 744 2364, Fax: +81 52 744 2371, E-mail: or
| | | | | |
Collapse
|
23
|
Sako W, Ishimoto S. Can cystatin C in cerebrospinal fluid be a biomarker for amyotrophic lateral sclerosis? A lesson from previous studies. ACTA ACUST UNITED AC 2014. [DOI: 10.1111/ncn3.82] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Wataru Sako
- Center for Neurosciences; The Feinstein Institute for Medical Research; Manhasset NY USA
| | - Shinji Ishimoto
- Department of Neurology; Wakayama Rosai Hospital; Wakayama Japan
| |
Collapse
|
24
|
Cystatin C has a dual role in post-traumatic brain injury recovery. Int J Mol Sci 2014; 15:5807-20. [PMID: 24714089 PMCID: PMC4013597 DOI: 10.3390/ijms15045807] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 03/13/2014] [Accepted: 03/25/2014] [Indexed: 11/17/2022] Open
Abstract
Cathepsin B is one of the major lysosomal cysteine proteases involved in neuronal protein catabolism. This cathepsin is released after traumatic injury and increases neuronal death; however, release of cystatin C, a cathepsin inhibitor, appears to be a self-protective brain response. Here we describe the effect of cystatin C intracerebroventricular administration in rats prior to inducing a traumatic brain injury. We observed that cystatin C injection caused a dual response in post-traumatic brain injury recovery: higher doses (350 fmoles) increased bleeding and mortality, whereas lower doses (3.5 to 35 fmoles) decreased bleeding, neuronal damage and mortality. We also analyzed the expression of cathepsin B and cystatin C in the brains of control rats and of rats after a traumatic brain injury. Cathepsin B was detected in the brain stem, cerebellum, hippocampus and cerebral cortex of control rats. Cystatin C was localized to the choroid plexus, brain stem and cerebellum of control rats. Twenty-four hours after traumatic brain injury, we observed changes in both the expression and localization of both proteins in the cerebral cortex, hippocampus and brain stem. An early increase and intralysosomal expression of cystatin C after brain injury was associated with reduced neuronal damage.
Collapse
|
25
|
Wilson ME, Boumaza I, Bowser R. Measurement of cystatin C functional activity in the cerebrospinal fluid of amyotrophic lateral sclerosis and control subjects. Fluids Barriers CNS 2013; 10:15. [PMID: 23497730 PMCID: PMC3610126 DOI: 10.1186/2045-8118-10-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/22/2013] [Indexed: 12/13/2022] Open
Abstract
Background Cystatin C is a constitutively expressed and abundant cysteine protease inhibitor within the cerebrospinal fluid (CSF). Recent studies have reported a significant reduction in cystatin C concentration in the CSF of patients with amyotrophic lateral sclerosis (ALS) and several other neurodegenerative diseases, relative to healthy controls. Cystatin C can exhibit both neuroprotective and neurotoxic properties, suggesting that altered CSF cystatin C concentrations could potentially impact the pathogenesis or progression of these disorders. However, it is unclear if alterations in cystatin C concentration result in physiologically relevant differences in its functional activity within the CSF. Measurements of the cysteine protease inhibitory activity of cystatin C within the CSF have not been reported, and the relationship between CSF cystatin C concentration and activity levels in different disease contexts has not been investigated. Methods We used a papain inhibition assay to evaluate the total cystatin C activity in CSF samples from 23 ALS patients, 23 healthy controls, and 23 neurological disease controls. Cystatin C concentrations in these samples were previously measured by ELISA. Correlations between cystatin C concentration and activity were assessed with nonparametric statistics. Activity ratios were compared among diagnostic groups using both one-way ANOVA and repeated measures statistics. Results Total cystatin C activity was found to be directly proportional to its protein concentration in all subjects, and cystatin C activity was not altered in ALS patients. In addition, our data suggest that cystatin C is the predominant cysteine protease inhibitor in human CSF. Conclusions Our data demonstrate the successful measurement of the functional activity of cystatin C in the CSF, and show that total cystatin C activity can be inferred from its total protein concentration. Our results also suggest that cystatin C is the major cysteine protease inhibitor in human CSF and altered CSF cystatin C concentration may play a role in the pathobiology of ALS and other neurological diseases.
Collapse
Affiliation(s)
- Meghan E Wilson
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
26
|
Abstract
Cystatins comprise a large superfamily of related proteins with diverse biological activities. They were initially characterised as inhibitors of lysosomal cysteine proteases, however, in recent years some alternative functions for cystatins have been proposed. Cystatins possessing inhibitory function are members of three families, family I (stefins), family II (cystatins) and family III (kininogens). Stefin A is often linked to neoplastic changes in epithelium while another family I cystatin, stefin B is supposed to have a specific role in neuredegenerative diseases. Cystatin C, a typical type II cystatin, is expressed in a variety of human tissues and cells. On the other hand, expression of other type II cystatins is more specific. Cystatin F is an endo/lysosome targeted protease inhibitor, selectively expressed in immune cells, suggesting its role in processes related to immune response. Our recent work points on its role in regulation of dendritic cell maturation and in natural killer cells functional inactivation that may enhance tumor survival. Cystatin E/M expression is mainly restricted to the epithelia of the skin which emphasizes its prominent role in cutaneous biology. Here, we review the current knowledge on type I (stefins A and B) and type II cystatins (cystatins C, F and E/M) in pathologies, with particular emphasis on their suppressive vs. promotional function in the tumorigenesis and metastasis. We proposed that an imbalance between cathepsins and cystatins may attenuate immune cell functions and facilitate tumor cell invasion.
Collapse
Affiliation(s)
- Spela Magister
- 1. Jožef Stefan Institute, Department of Biotechnology, Ljubljana, Slovenia
| | | |
Collapse
|
27
|
Rodriguez-Cerdeira C, Sanchez-Blanco E, Molares-Vila A, Alba A. Unveiling New Molecular Factors Useful for Detection of Pelvic Inflammatory Disease due to Chlamydia trachomatis Infection. ISRN OBSTETRICS AND GYNECOLOGY 2012; 2012:581725. [PMID: 23097713 PMCID: PMC3477744 DOI: 10.5402/2012/581725] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 08/31/2012] [Indexed: 11/23/2022]
Abstract
Background. Untreated Chlamydia trachomatis infections in women can result in disease sequelae such as pelvic inflammatory disease (PID), ultimately culminating in tubal occlusion and infertility. While nucleic acid amplification tests can effectively diagnose uncomplicated lower genital tract infections, they are not suitable for diagnosing upper genital tract pathological sequelae. Objective. The purpose of this paper was to provide a comprehensive review of new molecular factors associated with the diagnosis and prognosis of PID. Material and Methods. The literature was searched using the key words “Chlamydia trachomatis infections,” “pelvic inflammatory disease,” and “molecular factors” in the PubMed database. Relevant articles published between 1996 and 2012 were evaluated. Conclusions. The use of new molecular factors could potentially facilitate earlier diagnosis and prognosis in women with PID due to C. trachomatis infection.
Collapse
|
28
|
Increased intracranial pressure after diffuse traumatic brain injury exacerbates neuronal somatic membrane poration but not axonal injury: evidence for primary intracranial pressure-induced neuronal perturbation. J Cereb Blood Flow Metab 2012; 32:1919-32. [PMID: 22781336 PMCID: PMC3463883 DOI: 10.1038/jcbfm.2012.95] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Increased intracranial pressure (ICP) associated with traumatic brain injury (TBI) is linked to increased morbidity. Although our understanding of the pathobiology of TBI has expanded, questions remain regarding the specific neuronal somatic and axonal damaging consequences of elevated ICP, independent of its impact on cerebral perfusion pressure (CPP). To investigate this, Fischer rats were subjected to moderate TBI. Measurements of ICP revealed two distinct responses to injury. One population exhibited transient increases in ICP that returned to baseline levels acutely, while the other displayed persistent ICP elevation (>20 mm Hg). Utilizing these populations, the effect of elevated ICP on neuronal pathology associated with diffuse TBI was analyzed at 6 hours after TBI. No difference in axonal injury was observed, however, rats exhibiting persistently elevated ICP postinjury revealed a doubling of neurons with chronic membrane poration compared with rats exhibiting only transient increases in ICP. Elevated postinjury ICP was not associated with a concurrent increase in DNA damage; however, traditional histological assessments did reveal increased neuronal damage, potentially associated with redistribution of cathepsin-B from the lysosomal compartment into the cytosol. These findings indicate that persistently increased ICP, without deleterious alteration of CPP, exacerbates neuronal plasmalemmal perturbation that could precipitate persistent neuronal impairment and ultimate neuronal death.
Collapse
|
29
|
von Neuhoff N, Oumeraci T, Wolf T, Kollewe K, Bewerunge P, Neumann B, Brors B, Bufler J, Wurster U, Schlegelberger B, Dengler R, Zapatka M, Petri S. Monitoring CSF proteome alterations in amyotrophic lateral sclerosis: obstacles and perspectives in translating a novel marker panel to the clinic. PLoS One 2012; 7:e44401. [PMID: 22970211 PMCID: PMC3435306 DOI: 10.1371/journal.pone.0044401] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 08/02/2012] [Indexed: 11/18/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a fatal disorder of the motor neuron system with poor prognosis and marginal therapeutic options. Current clinical diagnostic criteria are based on electrophysiological examination and exclusion of other ALS-mimicking conditions. Neuroprotective treatments are, however, most promising in early disease stages. Identification of disease-specific CSF biomarkers and associated biochemical pathways is therefore most relevant to monitor disease progression, response to neuroprotective agents and to enable early inclusion of patients into clinical trials. Methods and Findings CSF from 35 patients with ALS diagnosed according to the revised El Escorial criteria and 23 age-matched controls was processed using paramagnetic bead chromatography for protein isolation and subsequently analyzed by MALDI-TOF mass spectrometry. CSF protein profiles were integrated into a Random Forest model constructed from 153 mass peaks. After reducing this peak set to the top 25%, a classifier was built which enabled prediction of ALS with high accuracy, sensitivity and specificity. Further analysis of the identified peptides resulted in a panel of five highly sensitive ALS biomarkers. Upregulation of secreted phosphoprotein 1 in ALS-CSF samples was confirmed by univariate analysis of ELISA and mass spectrometry data. Further quantitative validation of the five biomarkers was achieved in an 80-plex Multiple Reaction Monitoring mass spectrometry assay. Conclusions ALS classification based on the CSF biomarker panel proposed in this study could become a valuable predictive tool for early clinical risk stratification. Of the numerous CSF proteins identified, many have putative roles in ALS-related metabolic processes, particularly in chromogranin-mediated secretion signaling pathways. While a stand-alone clinical application of this classifier will only be possible after further validation and a multicenter trial, it could be readily used to complement current ALS diagnostics and might also provide new insights into the pathomechanisms of this disease in the future.
Collapse
Affiliation(s)
- Nils von Neuhoff
- Institute of Cell and Molecular Pathology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cheng XW, Shi GP, Kuzuya M, Sasaki T, Okumura K, Murohara T. Role for cysteine protease cathepsins in heart disease: focus on biology and mechanisms with clinical implication. Circulation 2012; 125:1551-62. [PMID: 22451605 DOI: 10.1161/circulationaha.111.066712] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Xian Wu Cheng
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Nagoya, Japan.
| | | | | | | | | | | |
Collapse
|
31
|
Dutta G, Barber DS, Zhang P, Doperalski NJ, Liu B. Involvement of dopaminergic neuronal cystatin C in neuronal injury-induced microglial activation and neurotoxicity. J Neurochem 2012; 122:752-63. [PMID: 22679891 DOI: 10.1111/j.1471-4159.2012.07826.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Factors released from injured dopaminergic (DA) neurons may trigger microglial activation and set in motion a vicious cycle of neuronal injury and inflammation that fuels progressive DA neurodegeneration in Parkinson's disease. In this study, using proteomic and immunoblotting analysis, we detected elevated levels of cystatin C in conditioned media (CM) from 1-methyl-4-phenylpyridinium and dieldrin-injured rat DA neuronal cells. Immunodepletion of cystatin C significantly reduced the ability of DA neuronal CM to induce activation of rat microglial cells as determined by up-regulation of inducible nitric oxide synthase, production of free radicals and release of proinflammatory cytokines as well as activated microglia-mediated DA neurotoxicity. Treatment of the cystatin C-containing CM with enzymes that remove O- and sialic acid-, but not N-linked carbohydrate moieties markedly reduced the ability of the DA neuronal CM to activate microglia. Taken together, these results suggest that DA neuronal cystatin C plays a role in the neuronal injury-induced microglial activation and neurotoxicity. These findings from the rat DA neuron-microglia in vitro model may help guide continued investigation to define the precise role of cystatin C in the complex interplay among neurons and glia in the pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Garima Dutta
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
32
|
Tarasiuk J, Kułakowska A, Drozdowski W, Kornhuber J, Lewczuk P. CSF markers in amyotrophic lateral sclerosis. J Neural Transm (Vienna) 2012; 119:747-57. [DOI: 10.1007/s00702-012-0806-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 04/16/2012] [Indexed: 11/29/2022]
|
33
|
Liang X, Nagai A, Terashima M, Sheikh AM, Shiota Y, Mitaki S, Kim SU, Yamaguchi S. Cystatin C induces apoptosis and tyrosine hydroxylase gene expression through JNK-dependent pathway in neuronal cells. Neurosci Lett 2011; 496:100-5. [PMID: 21504781 DOI: 10.1016/j.neulet.2011.03.091] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/28/2011] [Accepted: 03/29/2011] [Indexed: 12/01/2022]
Abstract
Cystatin C (CysC), an endogenous cysteine protease inhibitor, has been implicated in the apoptosis and differentiation processes of neuronal cells. In this study, we have investigated the pathway involved in the process. A human neuronal hybridoma cell line (A1 cell) was treated with CysC in both undifferentiated and retinoic acid (RA)-induced differentiated conditions, which decreased overall process length in both conditions. Also, CysC increased apoptotic cell number time-dependently, as revealed by TUNEL assay. Western blot analysis demonstrated that in differentiated A1 cells, CysC treatment decreased Bcl-2 and increased active caspase-9 protein level time-dependently. Immunocytochemistry results revealed that, CysC treatment significantly increased active form of Bax expressing cell number, which co-localized with mitochondria. Mitogen activated protein (MAP) kinase inhibition experiments showed that Bax mRNA induction and Bcl-2 mRNA inhibition by CysC treatment were c-Jun N-terminal kinase (JNK)-dependent. After RA-induced differentiation, choline acetyltransferase (ChAT) and neurofilament (NF) mRNA levels were increased in A1 cells. CysC treatment inhibited NF mRNA level in both undifferentiated and RA-differentiated, and increased TH mRNA in differentiated A1 neurons. Analysis of signal transduction pathway demonstrated that TH gene induction was also JNK-dependent. Thus, our results demonstrated the significance of JNK-dependent pathways on CysC-induced apoptosis and TH gene expression in neuronal cells, which might be an important target in the management of CysC dependent neurodegenerative processes.
Collapse
Affiliation(s)
- XueYun Liang
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo 693-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Wilson ME, Boumaza I, Lacomis D, Bowser R. Cystatin C: a candidate biomarker for amyotrophic lateral sclerosis. PLoS One 2010; 5:e15133. [PMID: 21151566 PMCID: PMC3000338 DOI: 10.1371/journal.pone.0015133] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 10/22/2010] [Indexed: 11/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurologic disease characterized by progressive motor neuron degeneration. Clinical disease management is hindered by both a lengthy diagnostic process and the absence of effective treatments. Reliable panels of diagnostic, surrogate, and prognostic biomarkers are needed to accelerate disease diagnosis and expedite drug development. The cysteine protease inhibitor cystatin C has recently gained interest as a candidate diagnostic biomarker for ALS, but further studies are required to fully characterize its biomarker utility. We used quantitative enzyme-linked immunosorbent assay (ELISA) to assess initial and longitudinal cerebrospinal fluid (CSF) and plasma cystatin C levels in 104 ALS patients and controls. Cystatin C levels in ALS patients were significantly elevated in plasma and reduced in CSF compared to healthy controls, but did not differ significantly from neurologic disease controls. In addition, the direction of longitudinal change in CSF cystatin C levels correlated to the rate of ALS disease progression, and initial CSF cystatin C levels were predictive of patient survival, suggesting that cystatin C may function as a surrogate marker of disease progression and survival. These data verify prior results for reduced cystatin C levels in the CSF of ALS patients, identify increased cystatin C levels in the plasma of ALS patients, and reveal correlations between CSF cystatin C levels to both ALS disease progression and patient survival.
Collapse
Affiliation(s)
- Meghan E. Wilson
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Imene Boumaza
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - David Lacomis
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Robert Bowser
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
35
|
Yamamoto-Watanabe Y, Watanabe M, Jackson M, Akimoto H, Sugimoto K, Yasujima M, Wakasaya Y, Matsubara E, Kawarabayashi T, Harigaya Y, Lyndon AR, Shoji M. Quantification of cystatin C in cerebrospinal fluid from various neurological disorders and correlation with G73A polymorphism in CST3. Brain Res 2010; 1361:140-5. [DOI: 10.1016/j.brainres.2010.09.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 09/06/2010] [Accepted: 09/08/2010] [Indexed: 12/12/2022]
|
36
|
Lee SH, Park SA, Ko SH, Yim HW, Ahn YB, Yoon KH, Cha BY, Son HY, Kwon HS. Insulin resistance and inflammation may have an additional role in the link between cystatin C and cardiovascular disease in type 2 diabetes mellitus patients. Metabolism 2010; 59:241-6. [PMID: 19765773 DOI: 10.1016/j.metabol.2009.07.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 06/16/2009] [Accepted: 07/20/2009] [Indexed: 12/26/2022]
Abstract
Recent studies suggest that serum cystatin C level is not only a sensitive marker for renal dysfunction but also a predictive marker for cardiovascular disease (CVD). However, the mechanism of this connection is not fully understood. We aimed to determine whether insulin resistance or various biomarkers of cardiovascular risk have a role in the link between cystatin C and CVD in type 2 diabetes mellitus patients. Anthropometric measurements and biochemical studies including inflammatory biomarkers were performed in 478 patients with type 2 diabetes mellitus. The degree of insulin resistance was assessed by homeostasis model assessment (HOMA-IR) and indicators of metabolic syndrome. Estimated glomerular filtration rate (eGFR) was derived from the Modification of Diet in Renal Disease study equation. After adjusting for age, sex, body mass index, and eGFR, the cystatin C level increased significantly in proportion to the number of metabolic syndrome components present (1.08 +/- 0.06, 1.19 +/- 0.04, 1.20 +/- 0.04, 1.23 +/- 0.04, and 1.37 +/- 0.06 mg/L; P < .0001); and HOMA-IR increased significantly in proportion to cystatin C quartiles (1.16 +/- 0.15, 1.40 +/- 0.13, 1.49 +/- 0.13, and 2.00 +/- 0.17; P < .0001) (means +/- SE). Albumin-creatinine ratio, fibrinogen, uric acid, homocysteine, high-sensitivity C-reactive protein, and lipoprotein(a) all showed significant correlations with cystatin C that were generally higher than those with eGFR. Cystatin C level was independently associated with HOMA-IR (beta = 0.0380, P = .0082), albumin-creatinine ratio (beta = 0.0004, P < .0001), uric acid (beta = 0.0666, P < .0001), and homocysteine (beta = 0.0087, P = .0004). In conclusion, cystatin C level was significantly associated with insulin resistance and biomarkers reflecting inflammation independent of renal function. These components may have a role in addition to that of eGFR in explaining the link between cystatin C and CVD in type 2 diabetes mellitus patients.
Collapse
Affiliation(s)
- Seung-Hwan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, St Vincent's Hospital, Suwon 442-723, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Tsai HT, Wang PH, Tee YT, Lin LY, Hsieh YS, Yang SF. Imbalanced serum concentration between cathepsin B and cystatin C in patients with pelvic inflammatory disease. Fertil Steril 2009; 91:549-55. [DOI: 10.1016/j.fertnstert.2007.12.076] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 12/27/2007] [Accepted: 12/27/2007] [Indexed: 11/27/2022]
|
39
|
Thirty years of Alzheimer's disease genetics: the implications of systematic meta-analyses. Nat Rev Neurosci 2008; 9:768-78. [PMID: 18802446 DOI: 10.1038/nrn2494] [Citation(s) in RCA: 531] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The genetic underpinnings of Alzheimer's disease (AD) remain largely elusive despite early successes in identifying three genes that cause early-onset familial AD (those that encode amyloid precursor protein (APP) and the presenilins (PSEN1 and PSEN2)), and one genetic risk factor for late-onset AD (the gene that encodes apolipoprotein E (APOE)). A large number of studies that aimed to help uncover the remaining disease-related loci have been published in recent decades, collectively proposing or refuting the involvement of over 500 different gene candidates. Systematic meta-analyses of these studies currently highlight more than 20 loci that have modest but significant effects on AD risk. This Review discusses the putative pathogenetic roles and common biochemical pathways of some of the most genetically and biologically compelling of these potential AD risk factors.
Collapse
|
40
|
Lee DC, Mason CW, Goodman CB, Holder MS, Kirksey OW, Womble TA, Severs WB, Palm DE. Hydrogen peroxide induces lysosomal protease alterations in PC12 cells. Neurochem Res 2007; 32:1499-510. [PMID: 17440810 DOI: 10.1007/s11064-007-9338-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Accepted: 03/16/2007] [Indexed: 10/23/2022]
Abstract
Alterations in lysosomal proteases have been implicated in many neurodegenerative diseases. The current study demonstrates a concentration-dependent decrease in PC12 cell viability and transient changes in cystatin C (CYSC), cathepsin B (CATB), cathepsin D (CATD) and caspase-3 following exposure to H2O2. Furthermore, activation of CATD occurred following exposure to H2O2 and cysteine protease suppression, while inhibition of CATD with pepstatin A significantly improved cell viability. Additionally, significant PARP cleavage, suggestive of caspase-3-like activity, was observed following H2O2 exposure, while inhibition of caspase-3 significantly increased cell viability compared to H2O2 administration alone. Collectively, our data suggest that H2O2 induced cell death is regulated at least in part by caspase-3 and CATD. Furthermore, cysteine protease suppression increases CATD expression and activity. These studies provide insight for alternate pathways and potential therapeutic targets of cell death associated with oxidative stress and lysosomal protease alterations.
Collapse
Affiliation(s)
- Daniel C Lee
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Wootz H, Weber E, Korhonen L, Lindholm D. Altered distribution and levels of cathepsinD and cystatins in amyotrophic lateral sclerosis transgenic mice: Possible roles in motor neuron survival. Neuroscience 2006; 143:419-30. [PMID: 16973300 DOI: 10.1016/j.neuroscience.2006.07.048] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 07/25/2006] [Accepted: 07/26/2006] [Indexed: 12/14/2022]
Abstract
In amyotrophic lateral sclerosis (ALS) there is a selective degeneration of motor neurons leading to muscle paralysis and death. The mechanism underlying cell demise in ALS is not fully understood, but involves the activation of different proteolytic enzymes, including the caspase family of cysteine proteases. We have here studied whether other proteases, such as the cathepsins, residing in lysosomes, and the cathepsin inhibitors, cystatinB and -C are changed in ALS. The expression and protein levels of the cathepsinB, -L and -D all increased in the spinal cord in ALS mice, carrying the mutant copper/zinc superoxide dismutase (SOD1) gene. At the cellular level, cathepsinB and -L were present in ventral motor neurons in controls, but in the ALS mice cathepsinB was also expressed by glial fibrillary acidic protein (GFAP) positive astrocytes. The distribution of the aspartic protease, cathepsinD also changed in ALS with a loss of the lysosomal staining in motor neurons. Inhibition of caspases by means of X-chromosome-linked inhibitor of apoptosis protein (XIAP) overexpression did not inhibit cleavage of cathepsinD in ALS mice, suggesting a caspase-independent pathway. Expression of cystatinB and -C increased slightly in the ALS spinal cords. Immunostaining showed that in ALS, cystatinC was present in motor neurons and in GFAP positive astrocytes. CystatinB that is a neuroprotective factor decreased in motor neurons in ALS but was expressed by activated microglial cells. The observed changes in the levels and distributions of cathepsinD and cystatinB and-C indicate a role of these proteins in the degeneration of motor neurons in ALS.
Collapse
Affiliation(s)
- H Wootz
- Department of Neuroscience, Unit of Neurobiology, Uppsala University, Biomedical Centre, Box 587, S-75123 Uppsala, Sweden
| | | | | | | |
Collapse
|