1
|
Kärkkäinen V, Saari T, Hannonen S, Rusanen M, Lehtola JM, Uusitalo H, Leinonen V, Thiede B, Kaarniranta K, Koivisto AM, Utheim TP. Altered tear fluid protein expression in persons with mild Alzheimer's disease in proteins involved in oxidative stress, protein synthesis, and energy metabolism. J Alzheimers Dis 2025:13872877251326868. [PMID: 40183343 DOI: 10.1177/13872877251326868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
BackgroundTear fluid (TF) is a protein-rich solution that reflects pathophysiological changes in Alzheimer's disease (AD).ObjectiveIn this study, we examined whether TF proteins were differently expressed in persons with mild AD dementia compared to cognitively healthy controls (CO).MethodsWe analyzed data from 53 study participants including 34 CO (mean age, 71 years; Mini-Mental State Examination [MMSE] score, 28.9 ± 1.4), and 19 patients with AD (Clinical Dementia Rating, 0.5-1; mean age, 72 years; MMSE score, 23.8 ± 2.8). All participants underwent cognitive testing, as well as neurological and ophthalmological examinations. TF was collected using Schirmer strips, and TF protein content was evaluated using mass spectrometry-based proteomics and label-free quantification.ResultsWe found that 16 proteins exhibited significantly upregulated expression in the AD group compared to the CO group (p ≤ 0.05). These proteins were NP1L4, BBOX1, CYTC, RNAS4, PCD, RNT2, AL1A3, SYSC, TPIS, CLH1, PGAM1, EIF3L, 5NTC, HNRNPA2B1, PYGL, and ERO1α. No proteins were significantly downregulated in the AD group compared to the CO group.ConclusionsOur results support the hypothesis that TF is a potential source of biomarkers for AD. Part of those proteins with altered expression have previously linked to increased oxidative stress, changed protein synthesis, and disturbed regulation of energy metabolism related to AD or neurodegenerative disease. The present results indicate the value of continued investigation of TF proteins in AD.
Collapse
Affiliation(s)
- Virve Kärkkäinen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- NeuroCenter, Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Toni Saari
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sanna Hannonen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Minna Rusanen
- Ceriatric Center, Wellbeing Services Country of North Karelia, Joensuu, Finland
| | - Juha-Matti Lehtola
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Hannu Uusitalo
- Eye and Vision Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ville Leinonen
- NeuroCenter, Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Anne M Koivisto
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Department of Geriatrics, Helsinki University Hospital and Department of Neurosciences, University of Helsinki, Helsinki, Finland
| | - Tor P Utheim
- Department of Ophthalmology, University of Oslo, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
2
|
Xu L, Xu W, Qin L. Association of cystatin C kidney function measures with motoric cognitive risk syndrome: evidence from two cohort studies. J Nutr Health Aging 2025; 29:100484. [PMID: 39793439 DOI: 10.1016/j.jnha.2025.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND This study aimed to examine the associations of cystatin C, cystatin C estimated glomerular filtration rate (eGFRcys), and the difference between eGFRs (eGFRdiff) using cystatin C and creatinine levels with incident motoric cognitive risk syndrome (MCR). METHODS We utilized data from two nationally representative cohort studies, the China Health and Retirement Longitudinal Study (CHARLS, 2011-2015) and the US Health and Retirement Study (HRS, 2010-2018). Baseline serum cystatin C and creatinine levels were measured, and eGFRcys and creatinine estimated GFR (eGFRcr) were calculated. MCR was defined as subjective cognitive complaints plus objectively measured slow gait speed. Multivariable logistic models were used to investigate the longitudinal associations between kidney function measurements and incident MCR. RESULTS In CHARLS (N = 2,085) and HRS (N = 1,240) cohorts, 7.4% and 7.2% developed MCR over follow-up. Each SD increment in serum cystatin C level was associated with elevated incident MCR odds, and an inverse association of eGFRcys with incident MCR was observed in both cohorts after multivariable adjustment and meta-analyses. The association between serum cystatin C and incident MCR remained significant even after adjusting for serum creatinine, suggesting that cystatin C is independently associated with MCR, regardless of kidney function levels. Additionally, each SD decrease in the absolute value of eGFRdiff was associated with lower odds of incident MCR among CHARLS participants. CONCLUSIONS Cystatin C and eGFRcys were correlated with an elevated MCR risk in two distinct populations. Specifically, eGFRdiff also related to incident MCR among Chinese older adults. Monitoring cystatin C-based kidney function could have significant clinical utility for identifying incident MCR risk, and represents a potential intervention target for healthier cognitive aging.
Collapse
Affiliation(s)
- Lijun Xu
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 463599, China
| | - Weihao Xu
- Haikou Cadre's Sanitarium of Hainan Military Region, Haikou 570203, China; Department of Geriatrics, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.
| | - Lijie Qin
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 463599, China.
| |
Collapse
|
3
|
Xu F, Liu T, Liu H, Deng J, He S, Lu Z, Zhang H, Dong H. Effect of remote ischemic preconditioning on perioperative neurocognitive disorder in elderly patients undergoing major surgery and associated genetic variant analysis: a randomized clinical trial. Perioper Med (Lond) 2025; 14:15. [PMID: 39910541 DOI: 10.1186/s13741-025-00497-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
OBJECTIVE To investigate whether remote ischemic preconditioning (RIPC) could reduce the incidence of perioperative neurocognitive disorder (PND) in elderly patients undergoing major surgery (> 2 h), to assess the potential of myeloid differentiation factor 2 (MD2) and cystatin C as biomarkers and to identify key genetic variants associated with PND. METHODS From August 2020, 250 patients scheduled for major surgeries under general anesthesia were screened and 120 patients were randomly assigned to the control group or the RIPC group. After anesthesia induction, patients in the RIPC group received a blood pressure cuff around their right upper limb, which was pressurized to 200 mmHg to induce ischemia, whereas the cuff in the control group was pressurized to only 60 mmHg. A total of five cycles were repeated with ischemia for five minutes and reperfusion for five minutes. Six neurological tests were performed before and after the surgery to assess the incidence of PND. Serum levels of myeloid differentiation factor 2 (MD2) and Cystatin C and PND-associated single nucleotide polymorphisms were analyzed by ELISA and whole genome sequencing, respectively. This study adhered to CONSORT research guidelines. RESULTS In the RIPC group, the incidence of PND (44%) was comparable to that in the control group (44%, P = 0.982). There was no significant difference in the concentrations of MD2 or cystatin C between the NPND and PND groups. A total of 3877 mutated genes were exclusively identified in PND patients. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that these mutated genes are enriched in synapse function. Notably, a Shank3 variant (SNP rs4824145) was included. CONCLUSIONS RIPC had little effect on the incidence of PND in elderly patients who underwent major surgery (> 2 h). MD2 and cystatin C were unable to predict the occurrence of PND. Patients harboring rs4824145 in the Shank3 gene may be more susceptible to PND. TRIAL REGISTRATION Chinese Clinical Trial Registry (ChiCTR2000035020(07/28/2020)).
Collapse
Affiliation(s)
- Feifei Xu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University; Key Laboratory of Anesthesiology (FMMU), Ministry of Education; Shaanxi Provincial Clinical Research Center for Anesthesiology Medicine, Xi'an, 710032, China
| | - Tingting Liu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University; Key Laboratory of Anesthesiology (FMMU), Ministry of Education; Shaanxi Provincial Clinical Research Center for Anesthesiology Medicine, Xi'an, 710032, China
| | - Huiqing Liu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University; Key Laboratory of Anesthesiology (FMMU), Ministry of Education; Shaanxi Provincial Clinical Research Center for Anesthesiology Medicine, Xi'an, 710032, China
| | - Jiao Deng
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University; Key Laboratory of Anesthesiology (FMMU), Ministry of Education; Shaanxi Provincial Clinical Research Center for Anesthesiology Medicine, Xi'an, 710032, China
| | - Shan He
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University; Key Laboratory of Anesthesiology (FMMU), Ministry of Education; Shaanxi Provincial Clinical Research Center for Anesthesiology Medicine, Xi'an, 710032, China
| | - Zhihong Lu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University; Key Laboratory of Anesthesiology (FMMU), Ministry of Education; Shaanxi Provincial Clinical Research Center for Anesthesiology Medicine, Xi'an, 710032, China.
| | - Haopeng Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University; Key Laboratory of Anesthesiology (FMMU), Ministry of Education; Shaanxi Provincial Clinical Research Center for Anesthesiology Medicine, Xi'an, 710032, China.
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University; Key Laboratory of Anesthesiology (FMMU), Ministry of Education; Shaanxi Provincial Clinical Research Center for Anesthesiology Medicine, Xi'an, 710032, China.
| |
Collapse
|
4
|
Nabizadeh F. Brain white matter damage biomarkers. Adv Clin Chem 2024; 125:55-91. [PMID: 39988408 DOI: 10.1016/bs.acc.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
White matter (WM), constituting nearly half of the human brain's mass, is pivotal for the rapid transmission of neural signals across different brain regions, significantly influencing cognitive processes like learning, memory, and problem-solving. The integrity of WM is essential for brain function, and its damage, which can occur due to conditions such as multiple sclerosis (MS), stroke, and traumatic brain injury, results in severe neurological deficits and cognitive decline. The primary objective of this book chapter is to discuss the clinical significance of fluid biomarkers in assessing WM damage within the central nervous system (CNS). It explores the biological underpinnings and pathological changes in WM due to various neurological conditions and details how alterations can be detected and quantified through fluid biomarkers. By examining biomarkers like Myelin Basic Protein (MBP), Neurofilament light chain (NFL), and others, the chapter highlights their role in enhancing diagnostic precision, monitoring disease progression, and guiding therapeutic interventions, thus providing crucial insights into maintaining WM integrity and preventing cognitive and physical disabilities.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, and Alzheimer's Disease Institute, Tehran, Iran.
| |
Collapse
|
5
|
Hamsanathan S, Anthonymuthu T, Prosser D, Lokshin A, Greenspan SL, Resnick NM, Perera S, Okawa S, Narasimhan G, Gurkar AU. A molecular index for biological age identified from the metabolome and senescence-associated secretome in humans. Aging Cell 2024; 23:e14104. [PMID: 38454639 PMCID: PMC11019119 DOI: 10.1111/acel.14104] [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: 08/13/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 03/09/2024] Open
Abstract
Unlike chronological age, biological age is a strong indicator of health of an individual. However, the molecular fingerprint associated with biological age is ill-defined. To define a high-resolution signature of biological age, we analyzed metabolome, circulating senescence-associated secretome (SASP)/inflammation markers and the interaction between them, from a cohort of healthy and rapid agers. The balance between two fatty acid oxidation mechanisms, β-oxidation and ω-oxidation, associated with the extent of functional aging. Furthermore, a panel of 25 metabolites, Healthy Aging Metabolic (HAM) index, predicted healthy agers regardless of gender and race. HAM index was also validated in an independent cohort. Causal inference with machine learning implied three metabolites, β-cryptoxanthin, prolylhydroxyproline, and eicosenoylcarnitine as putative drivers of biological aging. Multiple SASP markers were also elevated in rapid agers. Together, our findings reveal that a network of metabolic pathways underlie biological aging, and the HAM index could serve as a predictor of phenotypic aging in humans.
Collapse
Affiliation(s)
- Shruthi Hamsanathan
- Aging Institute of UPMC and the University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Tamil Anthonymuthu
- Department of Critical Care MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Denise Prosser
- Department of MedicineUniversity of Pittsburgh Medical Center and University of Pittsburgh Cancer InstitutePittsburghPennsylvaniaUSA
| | - Anna Lokshin
- Department of MedicineUniversity of Pittsburgh Medical Center and University of Pittsburgh Cancer InstitutePittsburghPennsylvaniaUSA
| | - Susan L. Greenspan
- Division of Geriatric Medicine, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Neil M. Resnick
- Aging Institute of UPMC and the University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Division of Geriatric Medicine, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Subashan Perera
- Division of Geriatric Medicine, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of BiostatisticsUniversity of Pittsburgh Graduate School of Public HealthPittsburghPennsylvaniaUSA
| | - Satoshi Okawa
- Pittsburgh Heart, Lung, and Blood Vascular Medicine InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Computational and Systems BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- McGowan Institute for Regenerative MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Giri Narasimhan
- Bioinformatics Research Group (BioRG), School of Computing and Information Sciences, Biomolecular Sciences InstituteFlorida International UniversityMiamiFloridaUSA
| | - Aditi U. Gurkar
- Aging Institute of UPMC and the University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Division of Geriatric Medicine, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
6
|
Stańczykiewicz B, Łuc M, Banach M, Zabłocka A. Cystatins: unravelling the biological implications for neuroprotection. Arch Med Sci 2023; 20:157-166. [PMID: 38414464 PMCID: PMC10895963 DOI: 10.5114/aoms/171706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/31/2023] [Indexed: 02/29/2024] Open
Abstract
Cystatins, a family of proteins known for their inhibitory role against cysteine proteases, have garnered significant attention in the field of neurodegeneration. Numerous genetic, experimental, and clinical studies concerning cystatin C suggest it plays an important role in the course of neurodegenerative diseases. Its beneficial effects are associated with cysteine protease inhibition, impact on β-amyloid aggregation, as well as regulation of cell proliferation, autophagy, and apoptosis. Cystatin isolated from chicken egg white, called ovocystatin, has been widely used in medical and pharmaceutical research due to its structural and biological similarities to human cystatin C. This article focuses on the potential use of cystatins, with special emphasis on easily obtained ovocystatin, in the treatment of neurodegenerative diseases, such as dementia. The current evidence on cystatin use has shed light on its mechanisms of action and therapeutic implications for neuroprotection and maintenance of cognitive functions.
Collapse
Affiliation(s)
- Bartłomiej Stańczykiewicz
- Division of Consultation Psychiatry and Neuroscience, Department of Psychiatry, Wroclaw Medical University, Wroclaw, Poland
| | - Mateusz Łuc
- Division of Consultation Psychiatry and Neuroscience, Department of Psychiatry, Wroclaw Medical University, Wroclaw, Poland
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Lodz, Poland
| | - Agnieszka Zabłocka
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
7
|
Nowak N, De Looze C, O’Halloran A, Kenny RA, Sexton DJ. The association between kidney function, cognitive function, and structural brain abnormalities in community-dwelling individuals aged 50+ is mediated by age and biomarkers of cardiovascular disease. Cardiovasc Res 2023; 119:2106-2116. [PMID: 37052588 PMCID: PMC10683948 DOI: 10.1093/cvr/cvad060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 04/14/2023] Open
Abstract
AIMS Cognitive impairment has been associated with kidney function and chronic kidney disease. Whether this association is due to accelerated cardiovascular disease (CVD) or an independent specific kidney function effect related to toxins is unclear. We investigated the impact of an array of clinical factors, inflammatory biomarkers, and cardiovascular biomarkers on the association between kidney function, cognitive function, and structural brain abnormalities. METHODS AND RESULTS We used data from the first and third waves of the TILDA Study, a population-representative prospective cohort of Irish adults aged 50 years and over, based on stratified random sampling (n = 3774). The MRI sub-study included participants who consented to MRI brain imaging in addition to the health assessment. Multivariable linear and mixed-effect longitudinal regression models were fitted separately for each kidney marker/estimated glomerular filtration rate (eGFR) equation after adjusting for baseline age and demographics, clinical vascular risk factors, and biomarkers. Unadjusted analyses showed an association between low eGFR, cognitive dysfunction, and cognitive decline (P < 0.001 for all kidney markers). Kidney function markers were also associated with white matter disease [OR = 3.32 (95% CI: 1.11, 9.98)], total grey matter volume (β = -0.17, 95% CI -0.27 to -0.07), and regional grey matter volumes within areas particularly susceptible to hypoxia (P < 0.001 for all). All the associations decreased after adjusting for age and were also diminished after adjusting for CVD biomarkers. Age and CVD-biomarker score were significant mediators of the adjusted associations between eGFR and cognitive status. These results remained consistent for cross-sectional and longitudinal outcomes and specific cognitive domains. CONCLUSION Decreased kidney function was associated with cerebrovascular disease. The association appeared to be mediated predominantly by age and the combination of CVD markers [namely N-terminal pro-B-type natriuretic peptide (NT-proBNP) and Growth Differentiation Factor 15 (GDF15)], supporting the idea that shared biological pathways underline both diseases. Further mechanistic studies of the specific molecular mechanisms that lead to both kidney and cognitive decline are warranted.
Collapse
Affiliation(s)
- Natalia Nowak
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, D02 R590, Dublin 2, Ireland
- School of Medicine, Trinity College Dublin, College Green, D02 PN40, Dublin 2, Ireland
| | - Celine De Looze
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, D02 R590, Dublin 2, Ireland
- School of Medicine, Trinity College Dublin, College Green, D02 PN40, Dublin 2, Ireland
| | - Aisling O’Halloran
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, D02 R590, Dublin 2, Ireland
- School of Medicine, Trinity College Dublin, College Green, D02 PN40, Dublin 2, Ireland
| | - Rose Anne Kenny
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, D02 R590, Dublin 2, Ireland
- School of Medicine, Trinity College Dublin, College Green, D02 PN40, Dublin 2, Ireland
- Saint James Hospital, Dublin, D08 NYH1, Dublin 8, Ireland
| | - Donal J Sexton
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, D02 R590, Dublin 2, Ireland
- School of Medicine, Trinity College Dublin, College Green, D02 PN40, Dublin 2, Ireland
- Saint James Hospital, Dublin, D08 NYH1, Dublin 8, Ireland
| |
Collapse
|
8
|
Choi Y, Shin S, Son HJ, Lee NH, Myeong SH, Lee C, Jang H, Choi SJ, Kim HJ, Na DL. Identification of potential biomarkers related to mesenchymal stem cell response in patients with Alzheimer's disease. Stem Cell Res Ther 2023; 14:178. [PMID: 37468918 PMCID: PMC10357744 DOI: 10.1186/s13287-023-03410-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/06/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Preclinical studies showed that mesenchymal stem cells (MSCs) ameliorate tau phosphorylation, amyloid-beta accumulation, and inflammation in Alzheimer's disease (AD) mouse models via secretion of neurotrophic factors and cytokines. We aimed to identify CSF biomarkers that can be used to predict or monitor the response to MSCs in patients with AD. METHODS AD patients were injected with human umbilical cord blood-MSCs (n = 22) or placebo (n = 12). The cerebrospinal fluid (CSF) samples were collected at baseline, one day after the first injection, and one day after the third injection. The patients injected with MSCs were classified into good responder (GR) or poor responder (PR) groups based on the rate of changes in the ratio of total-tau and phosphorylated-tau in the CSF. We selected three typical participants in each group, and their CSF protein levels were analyzed using liquid chromatography/tandem mass spectrometry (LC-MS/MS). RESULTS In the LC-MS/MS analysis, 1,667 proteins were identified. Eleven proteins showed significant differences between the typical GR and PR at baseline. Based on their significance level and known functions, two proteins, reticulocalbin-3 (RCN3) and follistatin-related protein 3 (FSTL3), were selected as potential biomarkers to predict MSC response. A total of 173 proteins showed significant change one day after the third injection compared to the baseline in typical GR. We excluded 45 proteins that showed significant change after the third injection compared to the baseline in the typical PR. Based on their significance level and known function, four proteins, scrapie-responsive protein 1 (SCRG1), neural proliferation differentiation and control protein (NPDC1), apolipoprotein E (ApoE), and cystatin C (CysC), were selected as potential biomarker to monitor MSC response. Additionally, functional analysis revealed that the increased CSF proteins after the third injection compared to the baseline in the typical GR were associated with synaptogenesis. CONCLUSIONS This study identified two proteins (RCN3 and FSTL3) that may be potential biomarkers for predicting MSC response and four proteins (SCRG1, NPDC1, ApoE, CysC) that may be potential biomarkers for monitoring MSC response in patients with AD. Further studies are needed to validate our results. Trial registration Clinical Trials.gov, NCT02054208. Registered on 4 February 2014. Samsung Medical Center IRB File No.2017-04-025. Registered on 20 June 2017.
Collapse
Affiliation(s)
- Yejoo Choi
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Sungho Shin
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyo Jin Son
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- School of Medicine, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Na-Hee Lee
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea
| | - Su Hyeon Myeong
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Cheolju Lee
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyemin Jang
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., 21, Daewangpangyo-ro 644 Beon-gil, Seongnam-si, Gyeonggi-do, 13494, Republic of Korea
| | - Hee Jin Kim
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- School of Medicine, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
| | - Duk L Na
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Higgins Tejera C, Ware EB, Kobayashi LC, Fu M, Hicken M, Zawistowski M, Mukherjee B, Bakulski KM. Decomposing interaction and mediating effects of race/ethnicity and circulating blood levels of cystatin C on cognitive status in the United States health and retirement study. Front Hum Neurosci 2023; 17:1052435. [PMID: 37323925 PMCID: PMC10267311 DOI: 10.3389/fnhum.2023.1052435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Background and objectives Elevated circulating cystatin C is associated with cognitive impairment in non-Hispanic Whites, but its role in racial disparities in dementia is understudied. In a nationally representative sample of older non-Hispanic White, non-Hispanic Black, and Hispanic adults in the United States, we use mediation-interaction analysis to understand how racial disparities in the cystatin C physiological pathway may contribute to racial disparities in prevalent dementia. Methods In a pooled cross-sectional sample of the Health and Retirement Study (n = 9,923), we employed Poisson regression to estimate prevalence ratios and to test the relationship between elevated cystatin C (>1.24 vs. ≤1.24 mg/L) and impaired cognition, adjusted for demographics, behavioral risk factors, other biomarkers, and chronic conditions. Self-reported racialized social categories were a proxy measure for exposure to racism. We calculated additive interaction measures and conducted four-way mediation-interaction decomposition analysis to test the moderating effect of race/ethnicity and mediating effect of cystatin C on the racial disparity. Results Overall, elevated cystatin C was associated with dementia (prevalence ratio [PR] = 1.2; 95% CI: 1.0, 1.5). Among non-Hispanic Black relative to non-Hispanic White participants, the relative excess risk due to interaction was 0.7 (95% CI: -0.1, 2.4), the attributable proportion was 0.1 (95% CI: -0.2, 0.4), and the synergy index was 1.1 (95% CI: 0.8, 1.8) in a fully adjusted model. Elevated cystatin C was estimated to account for 2% (95% CI: -0, 4%) for the racial disparity in prevalent dementia, and the interaction accounted for 8% (95% CI: -5, 22%). Analyses for Hispanic relative to non-white participants suggested moderation by race/ethnicity, but not mediation. Discussion Elevated cystatin C was associated with dementia prevalence. Our mediation-interaction decomposition analysis suggested that the effect of elevated cystatin C on the racial disparity might be moderated by race/ethnicity, which indicates that the racialization process affects not only the distribution of circulating cystatin C across minoritized racial groups, but also the strength of association between the biomarker and dementia prevalence. These results provide evidence that cystatin C is associated with adverse brain health and this effect is larger than expected for individuals racialized as minorities had they been racialized and treated as non-Hispanic White.
Collapse
Affiliation(s)
- César Higgins Tejera
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Erin B. Ware
- Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Lindsay C. Kobayashi
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Mingzhou Fu
- Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Margaret Hicken
- Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Matthew Zawistowski
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Bhramar Mukherjee
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Kelly M. Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Li K, Xu J, Zhao M, Wu J, Mei Y, Zhou Q, Zhao J, Li Y, Yang M, Xu Q. Serum cystatin C and mild cognitive impairment: The mediating role of glucose homeostasis. Front Aging Neurosci 2023; 15:1102762. [PMID: 37056689 PMCID: PMC10086181 DOI: 10.3389/fnagi.2023.1102762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundThis study explored the mediating role of glucose homeostasis indicators in the relationship between serum cystatin C and mild cognitive impairment (MCI).MethodsThe present study used a cross-sectional design and included 514 participants aged ≥50 years in Beijing, China. The Mini-Mental State Examination was used to assess cognitive function. Serum cystatin C and a comprehensive set of glucose homeostasis indicators were detected, including fasting blood glucose (FBG), glycosylated albumin percentage (GAP), glycated hemoglobin (HbAlc), insulin, and homeostatic model assessment of insulin resistance (HOMA-IR), and beta cell function (HOMA-β). Generalized linear models were used to investigate the associations among cystatin C, glucose homeostasis indicators, and cognitive function. Mediation analysis was conducted to explore potential mediator variables.ResultsIn this study of 514 participants, 76 (14.8%) had MCI. Those with cystatin C levels ≥1.09 mg/L had a 1.98-fold higher risk of MCI than those with levels <1.09 mg/L (95% CI, 1.05–3.69). FBG, GAP, and HbA1c increased the risk of MCI, while HOMA-β decreased the risk. Notably, the associations between MCI risk and cystatin C or glucose homeostasis were only founded in diabetes patients. Serum cystatin C was found to be positively associated with HOMA-β (beta (95% CI): 0.20 [0.06, 0.34]), HOMA-IR (0.23 [0.09, 0.36]), and insulin (0.22 [0.09, 0.34]) levels. Moreover, HOMA-β was identified as playing a negative mediating role (proportion mediated: −16%) in the relationship between cystatin C and MCI.ConclusionElevated levels of cystatin C are associated with an increased risk of MCI. The glucose homeostasis indicator, HOMA-β, plays a negative mediating role in the relationship between cystatin C and MCI risk.
Collapse
Affiliation(s)
- Kai Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Meiduo Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jingtao Wu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yayuan Mei
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Quan Zhou
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jiaxin Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yanbing Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ming Yang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qun Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- *Correspondence: Qun Xu,
| |
Collapse
|
11
|
Noori L, Filip K, Nazmara Z, Mahakizadeh S, Hassanzadeh G, Caruso Bavisotto C, Bucchieri F, Marino Gammazza A, Cappello F, Wnuk M, Scalia F. Contribution of Extracellular Vesicles and Molecular Chaperones in Age-Related Neurodegenerative Disorders of the CNS. Int J Mol Sci 2023; 24:927. [PMID: 36674442 PMCID: PMC9861359 DOI: 10.3390/ijms24020927] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/22/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Many neurodegenerative disorders are characterized by the abnormal aggregation of misfolded proteins that form amyloid deposits which possess prion-like behavior such as self-replication, intercellular transmission, and consequent induction of native forms of the same protein in surrounding cells. The distribution of the accumulated proteins and their correlated toxicity seem to be involved in the progression of nervous system degeneration. Molecular chaperones are known to maintain proteostasis, contribute to protein refolding to protect their function, and eliminate fatally misfolded proteins, prohibiting harmful effects. However, chaperone network efficiency declines during aging, prompting the onset and the development of neurological disorders. Extracellular vesicles (EVs) are tiny membranous structures produced by a wide range of cells under physiological and pathological conditions, suggesting their significant role in fundamental processes particularly in cellular communication. They modulate the behavior of nearby and distant cells through their biological cargo. In the pathological context, EVs transport disease-causing entities, including prions, α-syn, and tau, helping to spread damage to non-affected areas and accelerating the progression of neurodegeneration. However, EVs are considered effective for delivering therapeutic factors to the nervous system, since they are capable of crossing the blood-brain barrier (BBB) and are involved in the transportation of a variety of cellular entities. Here, we review the neurodegeneration process caused mainly by the inefficiency of chaperone systems as well as EV performance in neuropathies, their potential as diagnostic biomarkers and a promising EV-based therapeutic approach.
Collapse
Affiliation(s)
- Leila Noori
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran 1417653761, Iran
| | - Kamila Filip
- Department of Biology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, 35959 Rzeszow, Poland
| | - Zohreh Nazmara
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1417653761, Iran
| | - Simin Mahakizadeh
- Department of Anatomy, School of Medicine, Alborz University of Medical Sciences, Karaj 3149779453, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran 1417653761, Iran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1417653761, Iran
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Maciej Wnuk
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, 35959 Rzeszow, Poland
| | - Federica Scalia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| |
Collapse
|
12
|
Ma Y, Li C, Hua R, Yang C, Xie W, Zhang L. Association Between Serum Cystatin C and Cognitive Decline Independently from Creatinine: Evidence from Two Nationally Representative Aging Cohorts. J Alzheimers Dis 2023; 93:459-469. [PMID: 37038817 DOI: 10.3233/jad-221162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
BACKGROUND Studies on the association between cystatin C based estimated glomerular filtration rate (eGFRcys) and cognitive outcomes yielded inconsistent results. OBJECTIVE The present study aimed to examine the potential association of eGFRcys with subsequent cognitive decline rate. METHODS A total of 11,503 community-based participants were involved in our analyses, including 5,837 (aged 72.9±6.3; 58.6% women) in the Health and Retirement Study (HRS) from the US and 5,666 (aged 58.1±9.2; 49.0% women) in the China Health and Retirement Longitudinal Study (CHARLS). The association of eGFRcys with subsequent cognitive decline rate was evaluated by linear mixed models. RESULTS During 85,266 person-years of follow-up, both baseline elevated serum cystatin C (-0.048 standard deviation [SD]/year per mg/L; 95% confidence interval [CI], -0.060 to -0.036; p < 0.001) and decreased eGFRcys (0.026 SD/year per 30 mL/min/1.73m2; 95% CI, 0.020 to 0.032; p < 0.001) were associated with faster cognitive decline rate after full adjustment. Compared with those had eGFRcys ≥90 mL/min/1.73m2, participants with eGFRcys between 60 to 90 mL/min/1.73m2 (-0.012 SD/year; 95% CI, -0.020 to -0.004; p = 0.004) and those with eGFRcys <60 mL/min/1.73m2 (-0.048 SD/year; 95% CI, -0.058 to -0.039; p < 0.001) experienced statistically significantly faster cognitive decline after adjustment. The associations were independent from serum creatinine/eGFRcre (eGFR that was calculated from serum creatinine). CONCLUSION Decreased eGFRcys are significantly associated with faster cognitive decline after full adjustment, independently from serum creatinine/eGFRcre. Serum cystatin C might be a risk factor or a prodromal biomarker of cognitive decline.
Collapse
Affiliation(s)
- Yanjun Ma
- Heart and Vascular Health Research Center>, Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
- PUCRI Heart and Vascular Health Research Center at Peking University Shougang Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Chenglong Li
- Heart and Vascular Health Research Center>, Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
- PUCRI Heart and Vascular Health Research Center at Peking University Shougang Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Rong Hua
- Heart and Vascular Health Research Center>, Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
- PUCRI Heart and Vascular Health Research Center at Peking University Shougang Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Chao Yang
- National Institute of Health Data Science at Peking University, Beijing, China
- Advanced Institute of Information Technology, Peking University, Hangzhou, China
- Department of Medicine, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Wuxiang Xie
- Heart and Vascular Health Research Center>, Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
- PUCRI Heart and Vascular Health Research Center at Peking University Shougang Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Luxia Zhang
- National Institute of Health Data Science at Peking University, Beijing, China
- Advanced Institute of Information Technology, Peking University, Hangzhou, China
- Department of Medicine, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| |
Collapse
|
13
|
Wang S, Lin X, Zhou J, Li M, Song D. Association between serum cystatin C level and cognition in older adults: a cross-sectional analysis. Front Neurosci 2023; 17:1200763. [PMID: 37207178 PMCID: PMC10188921 DOI: 10.3389/fnins.2023.1200763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction Serum Cystatin C level, an indication of kidney function, has been implicated in the pathogenesis of Alzheimer's disease and cognitive impairment. In this cross-sectional study, we looked into the relation between serum Cystatin C levels and cognition in a group of U.S. older adults. Method The data of this study were from the National Health and Nutrition Examination Survey (NHANES) 1999-2002. A total of 4,832 older adults aged ≥60 who met the inclusion criteria were included. The Dade Behring N Latex Cystatin C assay, which is a particle-enhanced nephelometric assay (PENIA), was utilized to assess Cystatin C levels in participants' blood samples. Participants' cognition was examined using the digit symbol substitution test (DSST). Z-scores of the DSST were calculated based on sample means and standard deviations (SD). To investigate the relationships between the quartiles of serum Cystatin C level and DSST z scores, multiple linear regression models were developed while controlling for age, sex, race/ethnicity, and education. Results The average age of the participants was 71.1 (SD 7.8). The participants were about half female (50.5%), non-Hispanic White (61.2%), and (36.1%) who had completed at least some college. They had an average serum Cystatin C level of 1.0 mg/dl (SD 0.44). After performing multiple linear regression with a reference group consisting of participants in quartile one of plasma Cystatin C levels, we found that serum Cystatin C levels in quartiles three and four were independently associated with lower DSST z scores (β = -0.059, 95% CI -0.200 to -0.074 and β = -0.108, 95% CI -0.319 to -0.184, respectively). Conclusion Higher serum Cystatin C level is associated with worse processing speed, sustained attention, and working memory in older adults. Cystatin C level may be a biomarker for cognitive decline in older adults.
Collapse
Affiliation(s)
- Shuli Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xuechun Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zhou
- Department of Nursing, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, China
| | - Meng Li
- Kentucky STD Prevention and Control, Frankfort, KY, United States
| | - Dan Song
- Department of Nursing, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, China
- *Correspondence: Dan Song,
| |
Collapse
|
14
|
Kjaergaard AD, Ellervik C, Witte DR, Nordestgaard BG, Frikke-Schmidt R, Bojesen SE. Kidney function and risk of dementia: Observational study, meta-analysis, and two-sample mendelian randomization study. Eur J Epidemiol 2022; 37:1273-1284. [PMID: 36333541 DOI: 10.1007/s10654-022-00923-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/25/2022] [Indexed: 11/06/2022]
Abstract
Whether impaired kidney function is associated with increased risk of developing dementia is unclear. We investigated the association between estimated glomerular filtration rate (eGFR) and dementia. Using a triangulation approach, we performed (1) a prospective study in 90,369 Danes from the Copenhagen General Population Study (CGPS), (2) a meta-analysis in 468,699 Scandinavians (including CGPS) and (3) a two-sample Mendelian randomization study in 218,792-1,004,040 Europeans using summary data from largest publicly available genome wide association studies (GWASs). During up to 15 years of follow-up (CGPS), 2,468 individuals developed dementia. Age and sex standardized percentile of eGFR below versus above the median conferred a multifactorially adjusted hazard ratio of 1.09 (95% confidence interval: 1.01-1.18). In meta-analysis, random-effects risk of dementia was 1.14 (1.06-1.22) for mildly decreased eGFR (60-90 mL/min/1.73 m2), 1.31 (0.92-1.87) for moderately decreased eGFR (30-59 mL/min/1.73 m2) and 1.91 (1.21-3.01) for severely decreased eGFR (< 30 mL/min/1.73 m2), compared to reference eGFR (> 90 mL/min/1.73 m2). Using directly comparable eGFR measures (log[eGFR] scaled to one standard deviation, as well as eGFR below versus above 60 mL/min/1.73 m2), we found no association with risk of dementia in observational CGPS or in Mendelian randomization analyses. In conclusion, impaired kidney function was associated with modestly increased risk of developing dementia. This was not supported by causal, genetic analyses using a Mendelian randomization approach. However, future stronger genetic instruments for kidney function and larger GWASs with more dementia cases, particularly for the vascular dementia subtype, warrant a re-evaluation of the causal hypothesis.
Collapse
Affiliation(s)
- Alisa D Kjaergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Blvd. 11, Indgang A, Aarhus, Denmark.
| | - Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
- Department of Data and Development, Sorø, Region Zealand, Denmark
- Department of Pathology, Harvard Medical School and Department of Laboratory Medicine, Boston Children's Hospital, MA-02215, Boston, USA
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Stig E Bojesen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
| |
Collapse
|
15
|
Lin H, Himali JJ, Satizabal CL, Beiser AS, Levy D, Benjamin EJ, Gonzales MM, Ghosh S, Vasan RS, Seshadri S, McGrath ER. Identifying Blood Biomarkers for Dementia Using Machine Learning Methods in the Framingham Heart Study. Cells 2022; 11:1506. [PMID: 35563811 PMCID: PMC9100323 DOI: 10.3390/cells11091506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 11/25/2022] Open
Abstract
Blood biomarkers for dementia have the potential to identify preclinical disease and improve participant selection for clinical trials. Machine learning is an efficient analytical strategy to simultaneously identify multiple candidate biomarkers for dementia. We aimed to identify important candidate blood biomarkers for dementia using three machine learning models. We included 1642 (mean 69 ± 6 yr, 53% women) dementia-free Framingham Offspring Cohort participants attending examination, 7 who had available blood biomarker data. We developed three machine learning models, support vector machine (SVM), eXtreme gradient boosting of decision trees (XGB), and artificial neural network (ANN), to identify candidate biomarkers for incident dementia. Over a mean 12 ± 5 yr follow-up, 243 (14.8%) participants developed dementia. In multivariable models including all 38 available biomarkers, the XGB model demonstrated the strongest predictive accuracy for incident dementia (AUC 0.74 ± 0.01), followed by ANN (AUC 0.72 ± 0.01), and SVM (AUC 0.69 ± 0.01). Stepwise feature elimination by random sampling identified a subset of the nine most highly informative biomarkers. Machine learning models confined to these nine biomarkers showed improved model predictive accuracy for dementia (XGB, AUC 0.76 ± 0.01; ANN, AUC 0.75 ± 0.004; SVM, AUC 0.73 ± 0.01). A parsimonious panel of nine candidate biomarkers were identified which showed moderately good predictive accuracy for incident dementia, although our results require external validation.
Collapse
Affiliation(s)
- Honghuang Lin
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jayandra J. Himali
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- School of Public Health, Boston University, Boston, MA 02118, USA
- School of Medicine, Boston University, Boston, MA 02118, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX 77072, USA
| | - Claudia L. Satizabal
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX 77072, USA
| | - Alexa S. Beiser
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- School of Public Health, Boston University, Boston, MA 02118, USA
- School of Medicine, Boston University, Boston, MA 02118, USA
| | - Daniel Levy
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD 20824, USA
| | - Emelia J. Benjamin
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- School of Public Health, Boston University, Boston, MA 02118, USA
- School of Medicine, Boston University, Boston, MA 02118, USA
| | - Mitzi M. Gonzales
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX 77072, USA
| | - Saptaparni Ghosh
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- School of Medicine, Boston University, Boston, MA 02118, USA
| | - Ramachandran S. Vasan
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- School of Medicine, Boston University, Boston, MA 02118, USA
| | - Sudha Seshadri
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- School of Medicine, Boston University, Boston, MA 02118, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX 77072, USA
| | - Emer R. McGrath
- The Framingham Heart Study, Framingham, MA 01701, USA; (H.L.); (J.J.H.); (C.L.S.); (A.S.B.); (D.L.); (E.J.B.); (M.M.G.); (S.G.); (R.S.V.); (S.S.)
- HRB Clinical Research Facility, National University of Ireland Galway, University Road, H91TK33 Galway, Ireland
| |
Collapse
|
16
|
Saad MA, Abo-Raya SN, Waly MA, Saad NL. Association of Serum Levels of Cystatin C and Cognition in a Cohort of Egyptian Elderly Patients with Chronic Kidney Disease. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2022; 33:288-295. [PMID: 37417181 DOI: 10.4103/1319-2442.379027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023] Open
Abstract
Chronic kidney disease (CKD) and dementia are common morbidities of elders. Serum cystatin C has been suggested to be an ideal marker for kidney function. The current study aimed to detect the serum levels of cystatin C in CKD patients and to correlate these levels to cognitive performance. The study involved 90 subjects aged 65 years and more, divided into two groups: Group 1: 60 patients with CKD, and Group 2: 30 control participants. Exclusion criteria included cardiac failure, hepatic failure, thyroid diseases, dialysis for more than one month, polycystic kidney disease, organ transplantation, and immunosuppressive therapy within the past six months. All participants had routine laboratory workup, serum cystatin C using enzyme-linked immunosorbent assay kits, and cognitive assessment using mini-mental state examination (MMSE). Serum cystatin level was significantly high in CKD patients while MMSE scores were significantly lower in CKD patients. A high significant negative correlation was found between serum cystatin C levels and both degree of cognitive impairment and glomerular filtration rate (GFR). Also, a significant positive correlation was found between the degree of cognitive impairment and GFR levels. Serum cystatin levels are significantly associated with cognitive impairment in CKD patients, and this correlation becomes more evident with the worsening of CKD stages. That may help in better understanding of the pathogenesis of dementia in CKD patients with the emergence of therapeutic options depending on these data.
Collapse
Affiliation(s)
- Marwa A Saad
- Department of Internal Medicine, Geriatric Unit, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Suzan N Abo-Raya
- Department of Internal Medicine, Geriatric Unit, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Marwa A Waly
- Department of Nephrology, Shark El-Madinah Hospital, Alexandria, Egypt
| | - Neveen L Saad
- Department of Clinical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
17
|
Chen X, Huang Y, Bao T, Jia F, Ou R, Wei Q, Chen Y, Liu J, Yang J, Shang H. Changes in Serum Cystatin C Levels and the Associations With Cognitive Function in Alzheimer's Disease Patients. Front Aging Neurosci 2022; 13:790939. [PMID: 35153722 PMCID: PMC8832092 DOI: 10.3389/fnagi.2021.790939] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Objective Cystatin C is indicated to be involved in the pathogenesis of Alzheimer's disease (AD) and cognitive impairment. Our objective is to examine the serum Cystatin C levels, and to clarify the correlations between serum Cystatin C and cognitive performance in Chinese AD patients. Methods The serum Cystatin C concentrations in AD patients and age, sex, and body mass index (BMI) matched-healthy controls were measured. The cognitive functions of the AD patients were evaluated by using the Mini-mental State Examination (MMSE) and the Montreal Cognitive Assessment (MoCA). The severity of dementia was determined with clinical dementia rating (CDR). Results A total of 463 AD patients and 1,389 matched healthy subjects were included. AD patients had higher serum Cystatin C than healthy controls. Serum cystatin C levels were correlated with MoCA scores in AD patients. In an ordinal logistic regression model, AD patients with higher serum cystatin C levels had increased odds of severe cognitive dysfunction. Conclusion Our study suggested that AD patients had higher levels of serum cystatin C than age/sex/BMI-matched normal control subjects. Higher serum cystatin C may be associated with worse cognitive performance, but more studies are required to verify such association.
Collapse
|
18
|
Zheng Y, Ji B, Chen S, Zhou R, Ni R. The impact of uremic toxins on Alzheimer's disease. Curr Alzheimer Res 2022; 19:104-118. [PMID: 35048807 DOI: 10.2174/1567205019666220120113305] [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: 08/18/2021] [Revised: 11/27/2021] [Accepted: 12/14/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia, pathologically characterized by accumulation of senile plaques and neurofibrillary tangles. Chronic kidney disease (CKD) is highly prevalent in elderly population closely associated with occurrence of dementia. Recent epidemiological and experimental studies suggest a potential association of CKD with AD. Both diseases share a panel of identical risk factors, such as type 2 diabetes; and hypertension. However, the relationship between CKD and AD is unclear. Lower clearance of a panel of uremic toxin including cystatin-C, guanidine, and adiponectin due to CKD is implied to contribute to AD pathogenesis. In this review we summarize the current evidence from epidemiological, experimental and clinical studies on the potential contribution of uremic toxins to AD pathogenesis. We describe outstanding questions and propose an outlook on the link between uremic toxins and AD.
Collapse
Affiliation(s)
- Yuqi Zheng
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Sijun Chen
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Morgese MG, Bove M, Di Cesare Mannelli L, Schiavone S, Colia AL, Dimonte S, Mhillaj E, Sikora V, Tucci P, Ghelardini C, Trabace L. Precision Medicine in Alzheimer's Disease: Investigating Comorbid Common Biological Substrates in the Rat Model of Amyloid Beta-Induced Toxicity. Front Pharmacol 2022; 12:799561. [PMID: 35046821 PMCID: PMC8763383 DOI: 10.3389/fphar.2021.799561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD), one of the most widespread neurodegenerative disorder, is a fatal global burden for the elder population. Although many efforts have been made, the search of a curative therapy is still ongoing. Individuating phenotypic traits that might help in investigating treatment response is of growing interest in AD research. AD is a complex pathology characterized by many comorbidities, such as depression and increased susceptibility to pain perception, leading to postulate that these conditions may rely on common biological substrates yet to be determined. In order to investigate those biological determinants to be associable with phenotypic traits, we used the rat model of amyloid beta-induced toxicity. This established model of early phase of AD is obtained by the intracerebroventricular injection of soluble amyloid beta1-42 (Aβ) peptide 7 days before performing experiments. In this model, we have previously reported increased immobility in the forced swimming test, reduced cortical serotonin levels and subtle alterations in the cognitive domain a depressive-like phenotype associated with subtle alteration in memory processes. In light of evaluating pain perception in this animal model, we performed two different behavioral tests commonly used, such as the paw pressure test and the cold plate test, to analyze mechanical hyperalgesia and thermal allodynia, respectively. Behavioural outcomes confirmed the memory impairment in the social recognition test and, compared to sham, Aβ-injected rats showed an increased selective susceptibility to mechanical but not to thermal stimulus. Behavioural data were then corroborated by neurochemical and biochemical biomarker analyses either at central or peripheral level. Data showed that the peptide injection evoked a significant increase in hypothalamic glutamate, kynurenine and dopamine content, while serotonin levels were reduced. Plasma Cystatin-C, a cysteine protease, was increased while serotonin and melatonin levels were decreased in Aβ-injected rats. Urinary levels paralleled plasma quantifications, indicating that Aβ-induced deficits in pain perception, mood and cognitive domain may also depend on these biomarkers. In conclusion, in the present study, we demonstrated that this animal model can mimic several comorbid conditions typical of the early phase of AD. Therefore, in the perspective of generating novel therapeutic strategies relevant to precision medicine in AD, this animal model and the biomarkers evaluated herein may represent an advantageous approach.
Collapse
Affiliation(s)
- Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Bove
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Lorenzo Di Cesare Mannelli
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Anna Laura Colia
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Stefania Dimonte
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Emanuela Mhillaj
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Vladyslav Sikora
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- Department of Pathology, Sumy State University, Sumy, Ukraine
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Carla Ghelardini
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
20
|
Kaur S, Verma H, Dhiman M, Tell G, Gigli GL, Janes F, Mantha AK. Brain Exosomes: Friend or Foe in Alzheimer's Disease? Mol Neurobiol 2021; 58:6610-6624. [PMID: 34595669 DOI: 10.1007/s12035-021-02547-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/23/2021] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. It is known to be a multifactorial disease and several causes are associated with its occurrence as well as progression. However, the accumulation of amyloid beta (Aβ) is widely considered its major pathogenic hallmark. Additionally, neurofibrillary tangles (NFT), mitochondrial dysfunction, oxidative stress, and aging (cellular senescence) are considered as additional hits affecting the disease pathology. Several studies are now suggesting important role of inflammation in AD, which shifts our thought towards the brain's resident immune cells, microglia, and astrocytes; how they interact with neurons; and how these interactions are affected by intra and extracellular stressful factors. These interactions can be modulated by different mechanisms and pathways, in which exosomes could play an important role. Exosomes are multivesicular bodies secreted by nearly all types of cells. The exosomes secreted by glial cells or neurons affect the interactions and thus the physiology of these cells by transmitting miRNAs, proteins, and lipids. Exosomes can serve as a friend or foe to the neuron function, depending upon the carried signals. Exosomes, from the healthy microenvironment, may assist neuron function and health, whereas, from the stressed microenvironment, they carry oxidative and inflammatory signals to the neurons and thus prove detrimental to the neuronal function. Furthermore, exosomes can cross the blood-brain barrier (BBB), and from the blood plasma they can enter the brain cells and activate microglia and astrocytes. Exosomes can transport Aβ or Tau, cytokines, miRNAs between the cells, and alter the physiology of recipient cells. They can also assist in Aβ clearance and regulation of synaptic activity. The exosomes derived from different cells play different roles, and this field is still in its infancy stage. This review advocates exosomes' role as a friend or foe in neurodegenerative diseases, especially in the case of Alzheimer's disease.
Collapse
Affiliation(s)
- Sharanjot Kaur
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Harkomal Verma
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India
| | - Monisha Dhiman
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Gianluca Tell
- Department of Medicine, University of Udine, Udine, Italy
| | - Gian Luigi Gigli
- Department of Medicine, University of Udine, Udine, Italy
- Clinical Neurology, Udine University Hospital, Udine, Italy
| | | | - Anil K Mantha
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India.
| |
Collapse
|
21
|
Altuna-Azkargorta M, Mendioroz-Iriarte M. Blood biomarkers in Alzheimer's disease. NEUROLOGÍA (ENGLISH EDITION) 2021; 36:704-710. [PMID: 34752348 DOI: 10.1016/j.nrleng.2018.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/01/2018] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Early diagnosis of Alzheimer disease (AD) through the use of biomarkers could assist in the implementation and monitoring of early therapeutic interventions, and has the potential to significantly modify the course of the disease. DEVELOPMENT The classic cerebrospinal fluid and approved structural and functional neuroimaging biomarkers are of limited clinical application given their invasive nature and/or high cost. The identification of more accessible and less costly biomarkers, such as blood biomarkers, would increase their use in clinical practice. We review the available published evidence on the main blood biochemical biomarkers potentially useful for diagnosing AD. CONCLUSIONS Blood biomarkers are more cost- and time-effective than CSF biomarkers. However, immediate applicability in clinical practice is relatively unlikely. The main limitations come from the difficulty of measuring and standardising thresholds between different laboratories and the failure to replicate results. Of all the molecules studied, apoptosis and neurodegeneration biomarkers and the biomarker panels obtained through "omics" approaches, such as isolated or combined metabolomics, offer the most promising results.
Collapse
Affiliation(s)
- M Altuna-Azkargorta
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.
| | - M Mendioroz-Iriarte
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; Servicio de Neurología, Complejo Hospitalario de Navarra, Pamplona, Spain
| |
Collapse
|
22
|
VanDusen KW, Li YJ, Cai V, Hall A, Hiles S, Thompson JW, Moseley MA, Cooter M, Acker L, Levy JH, Ghadimi K, Quiñones QJ, Devinney MJ, Chung S, Terrando N, Moretti EW, Browndyke JN, Mathew JP, Berger M. Cerebrospinal Fluid Proteome Changes in Older Non-Cardiac Surgical Patients with Postoperative Cognitive Dysfunction. J Alzheimers Dis 2021; 80:1281-1297. [PMID: 33682719 PMCID: PMC8052629 DOI: 10.3233/jad-201544] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Postoperative cognitive dysfunction (POCD), a syndrome of cognitive deficits occurring 1–12 months after surgery primarily in older patients, is associated with poor postoperative outcomes. POCD is hypothesized to result from neuroinflammation; however, the pathways involved remain unclear. Unbiased proteomic analyses have been used to identify neuroinflammatory pathways in multiple neurologic diseases and syndromes but have not yet been applied to POCD. Objective: To utilize unbiased mass spectrometry-based proteomics to identify potential neuroinflammatory pathways underlying POCD. Methods: Unbiased LC-MS/MS proteomics was performed on immunodepleted cerebrospinal fluid (CSF) samples obtained before, 24 hours after, and 6 weeks after major non-cardiac surgery in older adults who did (n = 8) or did not develop POCD (n = 6). Linear mixed models were used to select peptides and proteins with intensity differences for pathway analysis. Results: Mass spectrometry quantified 8,258 peptides from 1,222 proteins in > 50%of patient samples at all three time points. Twelve peptides from 11 proteins showed differences in expression over time between patients with versus without POCD (q < 0.05), including proteins previously implicated in neurodegenerative disease pathophysiology. Additionally, 283 peptides from 182 proteins were identified with trend-level differences (q < 0.25) in expression over time between these groups. Among these, pathway analysis revealed that 50 were from 17 proteins mapping to complement and coagulation pathways (q = 2.44*10–13). Conclusion: These data demonstrate the feasibility of performing unbiased mass spectrometry on perioperative CSF samples to identify pathways associated with POCD. Additionally, they provide hypothesis-generating evidence for CSF complement and coagulation pathway changes in patients with POCD.
Collapse
Affiliation(s)
- Keith W VanDusen
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Yi-Ju Li
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA.,Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Victor Cai
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Ashley Hall
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Sarah Hiles
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - J Will Thompson
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - M Arthur Moseley
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Mary Cooter
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Leah Acker
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Jerrold H Levy
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Kamrouz Ghadimi
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Quintin J Quiñones
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Michael J Devinney
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Stacey Chung
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Niccolò Terrando
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Eugene W Moretti
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey N Browndyke
- Department of Psychiatry & Behavioral Sciences, Division of Geriatric Behavioral Health, Duke University Medical Center, Durham, NC, USA.,Duke Institute for Brain Sciences, Duke University, Durham, NC, USA.,Center for Cognitive Neuroscience, Duke University Medical Center, Durham, NC, USA
| | - Joseph P Mathew
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Miles Berger
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | | |
Collapse
|
23
|
Xu H, Garcia-Ptacek S, Trevisan M, Evans M, Lindholm B, Eriksdotter M, Carrero JJ. Kidney Function, Kidney Function Decline, and the Risk of Dementia in Older Adults: A Registry-Based Study. Neurology 2021; 96:e2956-e2965. [PMID: 33952656 PMCID: PMC8253567 DOI: 10.1212/wnl.0000000000012113] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/15/2021] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Community-based reports regarding the association between the estimated glomerular filtration rate (eGFR) and dementia risk show conflicting results. The aim of this study is to investigate the links among kidney function, kidney function decline, and dementia incidence. METHODS We analyzed the association of eGFR with the risk of dementia (defined as a new dementia diagnosis or initiation of dementia treatments) among 329,822 residents of Stockholm who accessed health care during 2006 to 2011, were ≥65 years of age, had no history of dementia, or underwent kidney replacement therapy. We also estimated the rate of eGFR decline among 205,622 residents with repeated eGFR measurements during the first year of observation and investigated its association with subsequent dementia risk. RESULTS We detected 18,983 cases of dementia (5.8% of participants) over a median follow-up of 5 years. Dementia incidence rates were progressively higher with lower eGFR: from 6.56/1,000 person-years in those with eGFR of 90 to 104 mL/min to 30.28/1,000 person-years in those with eGFR <30 mL/min. After multivariable adjustment, lower eGFR was associated with a higher dementia risk (hazard ratio [HR] 1.71, 95% confidence interval [CI] 1.54-1.91 in eGFR 30-59 mL/min; HR 2.62, 95% CI 1.91-3.58 in eGFR <30 mL/min) compared with eGFR of 90 to 104 mL/min. A steeper decline in eGFR (decline >2 mL/min/1.73 m2/y) within 1 year was associated with higher dementia risk. Risk magnitudes were stronger for vascular dementia than for Alzheimer dementia. As many as 10% (95% CI 6%-14%) of dementia cases could be attributed to eGFR <60 mL/min/1.73 m2, a proportion higher than that attributed to other dementia risk factors such as cardiovascular disease and diabetes. CONCLUSIONS Both lower kidney function and steeper kidney function decline are associated with the development of dementia.
Collapse
Affiliation(s)
- Hong Xu
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Department of Neurobiology, Care Sciences and Society, Department of Medical Epidemiology and Biostatistics (H.X., M.T., J.J.C.), and Division of Renal Medicine and Baxter Novum (M.E., B.L.), Department of Clinical Science, Intervention and Technology, Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset; and Theme Aging (S.G.-P., M.E.), Karolinska University Hospital, Stockholm, Sweden.
| | - Sara Garcia-Ptacek
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Department of Neurobiology, Care Sciences and Society, Department of Medical Epidemiology and Biostatistics (H.X., M.T., J.J.C.), and Division of Renal Medicine and Baxter Novum (M.E., B.L.), Department of Clinical Science, Intervention and Technology, Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset; and Theme Aging (S.G.-P., M.E.), Karolinska University Hospital, Stockholm, Sweden
| | - Marco Trevisan
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Department of Neurobiology, Care Sciences and Society, Department of Medical Epidemiology and Biostatistics (H.X., M.T., J.J.C.), and Division of Renal Medicine and Baxter Novum (M.E., B.L.), Department of Clinical Science, Intervention and Technology, Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset; and Theme Aging (S.G.-P., M.E.), Karolinska University Hospital, Stockholm, Sweden
| | - Marie Evans
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Department of Neurobiology, Care Sciences and Society, Department of Medical Epidemiology and Biostatistics (H.X., M.T., J.J.C.), and Division of Renal Medicine and Baxter Novum (M.E., B.L.), Department of Clinical Science, Intervention and Technology, Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset; and Theme Aging (S.G.-P., M.E.), Karolinska University Hospital, Stockholm, Sweden
| | - Bengt Lindholm
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Department of Neurobiology, Care Sciences and Society, Department of Medical Epidemiology and Biostatistics (H.X., M.T., J.J.C.), and Division of Renal Medicine and Baxter Novum (M.E., B.L.), Department of Clinical Science, Intervention and Technology, Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset; and Theme Aging (S.G.-P., M.E.), Karolinska University Hospital, Stockholm, Sweden
| | - Maria Eriksdotter
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Department of Neurobiology, Care Sciences and Society, Department of Medical Epidemiology and Biostatistics (H.X., M.T., J.J.C.), and Division of Renal Medicine and Baxter Novum (M.E., B.L.), Department of Clinical Science, Intervention and Technology, Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset; and Theme Aging (S.G.-P., M.E.), Karolinska University Hospital, Stockholm, Sweden
| | - Juan Jesus Carrero
- From the Division of Clinical Geriatrics (H.X., S.G.-P., M.E.), Department of Neurobiology, Care Sciences and Society, Department of Medical Epidemiology and Biostatistics (H.X., M.T., J.J.C.), and Division of Renal Medicine and Baxter Novum (M.E., B.L.), Department of Clinical Science, Intervention and Technology, Karolinska Institutet; Department of Internal Medicine (S.G.-P.), Neurology Section, Södersjukhuset; and Theme Aging (S.G.-P., M.E.), Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
24
|
Ferguson AC, Tank R, Lyall LM, Ward J, Celis-Morales C, Strawbridge R, Ho F, Whelan CD, Gill J, Welsh P, Anderson JJ, Mark PB, Mackay DF, Smith DJ, Pell JP, Cavanagh J, Sattar N, Lyall DM. Alzheimer's Disease Susceptibility Gene Apolipoprotein E (APOE) and Blood Biomarkers in UK Biobank (N = 395,769). J Alzheimers Dis 2021; 76:1541-1551. [PMID: 32651323 DOI: 10.3233/jad-200338] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative condition where the underlying etiology is still unclear. Investigating the potential influence of apolipoprotein E (APOE), a major genetic risk factor, on common blood biomarkers could provide a greater understanding of the mechanisms of AD and dementia risk. OBJECTIVE Our objective was to conduct the largest (to date) single-protocol investigation of blood biomarkers in the context of APOE genotype, in UK Biobank. METHODS After quality control and exclusions, data on 395,769 participants of White European ancestry were available for analysis. Linear regressions were used to test potential associations between APOE genotypes and biomarkers. RESULTS Several biomarkers significantly associated with APOEɛ4 'risk' and ɛ2 'protective' genotypes (versus neutral ɛ3/ɛ3). Most associations supported previous data: for example, ɛ4 genotype was associated with elevated low-density lipoprotein cholesterol (LDL) (standardized beta [b] = 0.150 standard deviations [SDs] per allele, p < 0.001) and ɛ2 with lower LDL (b = -0.456 SDs, p < 0.001). There were however instances of associations found in unexpected directions: e.g., ɛ4 and increased insulin-like growth factor (IGF-1) (b = 0.017, p < 0.001) where lower levels have been previously suggested as an AD risk factor. CONCLUSION These findings highlight biomarker differences in non-demented people at genetic risk for dementia. The evidence herein supports previous hypotheses of involvement from cardiometabolic and neuroinflammatory pathways.
Collapse
Affiliation(s)
- Amy C Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Rachana Tank
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Carlos Celis-Morales
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,GEEAFyS, Universidad Católica del Maule, Talca, Chile
| | - Rona Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK.,Health Data Research UK.,Department of Medicine Solna, Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Frederick Ho
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | | | - Jason Gill
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Jana J Anderson
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Daniel F Mackay
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Jonathan Cavanagh
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Naveed Sattar
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| |
Collapse
|
25
|
Li L, Cavuoto M, Biddiscombe K, Pike KE. Diabetes Mellitus Increases Risk of Incident Dementia in APOE ɛ4 Carriers: A Meta-Analysis. J Alzheimers Dis 2020; 74:1295-1308. [DOI: 10.3233/jad-191068] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Lily Li
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Marina Cavuoto
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Karen Biddiscombe
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Kerryn E. Pike
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Fame RM, Cortés-Campos C, Sive HL. Brain Ventricular System and Cerebrospinal Fluid Development and Function: Light at the End of the Tube: A Primer with Latest Insights. Bioessays 2020; 42:e1900186. [PMID: 32078177 DOI: 10.1002/bies.201900186] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/02/2020] [Indexed: 12/12/2022]
Abstract
The brain ventricular system is a series of connected cavities, filled with cerebrospinal fluid (CSF), that forms within the vertebrate central nervous system (CNS). The hollow neural tube is a hallmark of the chordate CNS, and a closed neural tube is essential for normal development. Development and function of the ventricular system is examined, emphasizing three interdigitating components that form a functional system: ventricle walls, CSF fluid properties, and activity of CSF constituent factors. The cellular lining of the ventricle both can produce and is responsive to CSF. Fluid properties and conserved CSF components contribute to normal CNS development. Anomalies of the CSF/ventricular system serve as diagnostics and may cause CNS disorders, further highlighting their importance. This review focuses on the evolution and development of the brain ventricular system, associated function, and connected pathologies. It is geared as an introduction for scholars with little background in the field.
Collapse
Affiliation(s)
- Ryann M Fame
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | | | - Hazel L Sive
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
27
|
Cui Z, Cao G, Wang Y, Ma Q, Wang C, Xu Y, Sun H, Ma Y. Effects of Cystatin C on Cognitive Impairment in Older Chinese Adults. Am J Alzheimers Dis Other Demen 2020; 35:1533317520965101. [PMID: 33111545 PMCID: PMC10624069 DOI: 10.1177/1533317520965101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
OBJECTIVE To find a suitable dividing value to classify cystatin C and evaluate the association between cognition and levels of cystatin C. METHODS Using data from the China Health and Retirement Longitudinal Study, We conducted a longitudinal analysis of a prospective cohort of 6,869 middle-aged and older Chinese without cognitive impairment at baseline. Levels of cystatin C were categorized into 2 groups by method of decision tree. Logistic regression models evaluated whether cystatin C was related to cognitive impairment. RESULTS Respondents were categorized as lower levels of cystatin C and higher levels of cystatin C, cut-point was 1.11 mg/L. Higher levels of cystatin C was associated with the odds of cognitive impairment (OR, 1.56; 95% CI, 1.10-2.22) after multivariable adjustment. Respondents with higher levels of cystatin C had worse cognition scores. CONCLUSIONS We found a suitable dividing value of cystatin C in middle-aged and older Chinese.
Collapse
Affiliation(s)
- Zhizhen Cui
- Department of Child Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, People’s Republic of China
| | - Guizhen Cao
- Department of Child Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, People’s Republic of China
| | - Youyi Wang
- Department of Child Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, People’s Republic of China
| | - Qinghua Ma
- The 3rd People’s Hospital of Xiangcheng District, Suzhou, People’s Republic of China
| | - Congju Wang
- Centers for Disease Control and Prevention of Suzhou High-tech Zone, Suzhou, People’s Republic of China
| | - Yong Xu
- Department of Child Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, People’s Republic of China
| | - Hongpeng Sun
- Department of Child Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, People’s Republic of China
| | - Yana Ma
- Department of Child Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
28
|
Ivanova MV, Chekanova EO, Belugin BV, Tutykhina IL, Dolzhikova IV, Zakroishchikova IV, Vasil’ev AV, Zakharova MN. Exosomal Transport and Progression of Neurodegeneration in Amyotrophic Lateral Sclerosis. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419030085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Moderately increased albuminuria, chronic kidney disease and incident dementia: the HUNT study. BMC Nephrol 2019; 20:261. [PMID: 31299931 PMCID: PMC6626412 DOI: 10.1186/s12882-019-1425-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 06/20/2019] [Indexed: 12/29/2022] Open
Abstract
Background Epidemiologic studies has shown an association of albuminuria and low estimated glomerular filtration rate (eGFR) with dementia, but the findings are inconsistent. This study examines the association between eGFR, MA with dementia and its subtypes: AD, VaD, a mixture of AD/VaD, and other dementias. Methods Data from the second wave of the HUNT 2 Study (1995–1997) were linked with a dementia register known as the Health and Memory Study (HMS) collected during 1995–2011 in Nord-Trøndelag County, Norway. Dementia was ascertained using World Health Organization’s ICD-10 criteria into subtypes: AD,VaD, mixed AD/VaD, and other dementia. eGFR and its association with dementia was examined in 48,508 participants of the HUNT Study, of which 668 were diagnosed with all-cause dementia. Association between MA and dementia were studied in a subset of 7024 participants, and 214 were diagnosed with all-cause dementia. Cox regression models were conducted analyzing the association between dementia and MA using albumin creatine ratio (ACR). Cox regression models and Fine-Gray models were used to examine the association between dementia and eGFR. Results A positive association was found between increasing ACR and dementia. ACR in the fourth quartile (> 1.78 mg/mmol) with increased hazard ratio of VaD, 3.97 (1.12 to 14.07), compared with ACR in the first quartile (<.53 mg/mmol). There was no association between eGFR and dementia or its subgroups. Conclusions Our results strengthens the hypothesis that vascular mechanisms may affect both kidney and brain as an association between MA and dementia was found. However, eGFR was not significantly associated with dementia independent of diabetes mellitus or hypertension. Electronic supplementary material The online version of this article (10.1186/s12882-019-1425-8) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Korolenko TA, Shintyapina AB, Pupyshev AB, Akopyan AA, Russkikh GS, Dikovskaya MA, Vavilin VA, Zavjalov EL, Tikhonova MA, Amstislavskaya TG. The regulatory role of cystatin C in autophagy and neurodegeneration. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Autophagy is a dynamic cellular process involved in the turnover of proteins, protein complexes, and organelles through lysosomal degradation. It is particularly important in neurons, which do not have a proliferative option for cellular repair. Autophagy has been shown to be suppressed in the striatum of a transgenic mouse model of Parkinson’s disease. Cystatin C is one of the potent regulators of autophagy. Changes in the expression and secretion of cystatin C in the brain have been shown in amyotrophic lateral sclerosis, Alzheimer’s and Parkinson’s diseases, and in some animal models of neurodegeneration, thus proving a protective function of cystatin C. It has been suggested that cystatin C plays the primary role in amyloidogenesis and shows promise as a therapeutic agent for neurodegenerative diseases (Alzheimer’s and Parkinson’s diseases). Cystatin C colocalizes with the amyloid β-protein in the brain during Alzheimer’s disease. Controlled expression of a cystatin C peptide has been proposed as a new approach to therapy for Alzheimer’s disease. In Parkinson’s disease, serum cystatin C levels can predict disease severity and cognitive dysfunction, although the exact involvement of cystatin C remains unclear. The aim: to study the role of cystatin C in neurodegeneration and evaluate the results in relation to the mechanism of autophagy. In our study on humans, a higher concentration of cystatin C was noted in cerebrospinal fluid than in serum; much lower concentrations were observed in other biological fluids (intraocular fluid, bile, and sweat). In elderly persons (61–80 years old compared to practically healthy people at 40–60 years of age), we revealed increased cystatin C levels both in serum and intraocular fluid. In an experiment on C57Bl/6J mice, cystatin C concentration was significantly higher in brain tissue than in the liver and spleen: an indication of an important function of this cysteine protease inhibitor in the brain. Using a transgenic mouse model of Parkinson’s disease (5 months old), we demonstrated a significant increase in osmotic susceptibility of brain lysosomes, depending on autophagy, while in a murine model of Alzheimer’s disease, this parameter did not differ from that in the appropriate control.
Collapse
Affiliation(s)
- T. A. Korolenko
- Scientific Research Institute of Physiology and Basic Medicine
| | - A. B. Shintyapina
- Scientific Research Institute of Molecular Biology and Biophysics, Federal Research Center for Basic and Translational Medicine
| | - A. B. Pupyshev
- Scientific Research Institute of Physiology and Basic Medicine
| | - A. A. Akopyan
- Scientific Research Institute of Physiology and Basic Medicine
| | - G. S. Russkikh
- Scientific Research Institute of Biochemistry, Federal Research Center for Basic and Translational Medicine
| | - M. A. Dikovskaya
- Scientific Research Institute of Physiology and Basic Medicine; S.N. Fedorov NMRC “MNTK “Eye Microsurgery”, Novosibirsk Branch
| | - V. A. Vavilin
- Scientific Research Institute of Molecular Biology and Biophysics, Federal Research Center for Basic and Translational Medicine; Institute of Cytology and Genetics, SB RAS
| | | | - M. A. Tikhonova
- Scientific Research Institute of Physiology and Basic Medicine; Novosibirsk State University
| | - T. G. Amstislavskaya
- Scientific Research Institute of Physiology and Basic Medicine; Novosibirsk State University
| |
Collapse
|
31
|
Jiang L, Dong H, Cao H, Ji X, Luan S, Liu J. Exosomes in Pathogenesis, Diagnosis, and Treatment of Alzheimer's Disease. Med Sci Monit 2019; 25:3329-3335. [PMID: 31056537 PMCID: PMC6515980 DOI: 10.12659/msm.914027] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by the accumulation of β-amyloid peptide 1-42 and phosphorylation of tau protein in the brain. Thus far, the transfer mechanism of these cytotoxic proteins between nerve cells remains unclear. Recent studies have shown that nanoscale extracellular vesicles (exosomes) originating from cells may play important roles in this transfer process. In addition, several genetic materials and proteins are also involved in intercellular communication by the secretion of the exosomes. That proposes novel avenues for early diagnosis and biological treatment in AD, based on exosome detection and intervention. In this review, exosome-related pathways of cytotoxic protein intercellular transfer in AD, and the effect of membrane proteins on exosomes targeting cells are first introduced. The advances in exosome-related biomarker detection in AD are summarized. Finally, the advantages and challenges of reducing cytotoxic protein accumulation via exosomal intervention for AD treatment are discussed. It is envisaged that future research in exosomes may well provide new insights into the pathogenesis, diagnosis, and treatment of AD.
Collapse
Affiliation(s)
- Liqun Jiang
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Huijie Dong
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Hua Cao
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Xiaofei Ji
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Siyu Luan
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Sargent L, Nalls M, Starkweather A, Hobgood S, Thompson H, Amella EJ, Singleton A. Shared biological pathways for frailty and cognitive impairment: A systematic review. Ageing Res Rev 2018; 47:149-158. [PMID: 30102995 PMCID: PMC6376483 DOI: 10.1016/j.arr.2018.08.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/30/2018] [Accepted: 08/01/2018] [Indexed: 11/20/2022]
Affiliation(s)
- Lana Sargent
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Virginia Commonwealth University School of Nursing, Richmond, VA, USA; Medical University of South Carolina School of Nursing, Charleston, NC, USA.
| | - Mike Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International, Glen Echo, MD, USA
| | | | - Sarah Hobgood
- Virginia Commonwealth School of Medicine, Richmond, VA, USA
| | - Holly Thompson
- National Institutes of Health Library, Division of Library Services, Office of Research Services, National Institutes of Health, Bethesda, MD, USA
| | - Elaine J Amella
- Medical University of South Carolina School of Nursing, Charleston, NC, USA
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Altuna-Azkargorta M, Mendioroz-Iriarte M. Blood biomarkers in Alzheimer's disease. Neurologia 2018; 36:S0213-4853(18)30091-4. [PMID: 29752036 DOI: 10.1016/j.nrl.2018.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/20/2018] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION The early diagnosis of Alzheimer's disease (AD) via the use of biomarkers could facilitate the implementation and monitoring of early therapeutic interventions with the potential capacity to significantly modify the course of the disease. DEVELOPMENT Classic cerebrospinal fluid biomarkers and approved structural and functional neuroimaging have a limited clinical application given their invasive nature and/or high cost. The identification of more accessible and less costly biomarkers, such as blood biomarkers, would facilitate application in clinical practice. We present a literature review of the main blood biochemical biomarkers with potential use for diagnosing Alzheimer's disease. CONCLUSIONS Blood biomarkers are cost and time effective with regard to cerebrospinal fluid biomarkers. However, the immediate applicability of blood biochemical biomarkers in clinical practice is not very likely. The main limitations come from the difficulties in measuring and standardising thresholds between different laboratories and in failures to replicate results. Among all the molecules studied, apoptosis and neurodegeneration biomarkers and the biomarker panels obtained through omics approaches, such as isolated or combined metabolomics, offer the most promising results.
Collapse
Affiliation(s)
- M Altuna-Azkargorta
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, España.
| | - M Mendioroz-Iriarte
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, España; Servicio de Neurología, Complejo Hospitalario de Navarra, Pamplona, España
| |
Collapse
|
36
|
Benussi L, Binetti G, Ghidoni R. Loss of Neuroprotective Factors in Neurodegenerative Dementias: The End or the Starting Point? Front Neurosci 2017; 11:672. [PMID: 29249935 PMCID: PMC5717017 DOI: 10.3389/fnins.2017.00672] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/20/2017] [Indexed: 01/05/2023] Open
Abstract
Recent clinical, genetic and biochemical experimental evidences highlight the existence of common molecular pathways underlying neurodegenerative diseases. In this review, we will explore a key common pathological mechanism, i.e., the loss of neuroprotective factors, across the three major neurodegenerative diseases leading to dementia: Alzheimer's disease (AD), Frontotemporal dementia (FTD) and Lewy body dementia (LBD). We will report evidences that the Brain Derived Neurotrophic Factor (BDNF), the most investigated and characterized brain neurotrophin, progranulin, a multi-functional adipokine with trophic and growth factor properties, and cystatin C, a neuroprotective growth factor, are reduced in AD, FTD, and LBD. Moreover, we will review the molecular mechanism underlying the loss of neuroprotective factors in neurodegenerative diseases leading to dementia, with a special focus on endo-lysosomal pathway and intercellular communication mediated by extracellular vesicles. Exploring the shared commonality of disease mechanisms is of pivotal importance to identify novel potential therapeutic targets and to develop treatments to delay, slow or block disease progression.
Collapse
Affiliation(s)
- Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,MAC Memory Center, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
37
|
Brinkmalm G, Sjödin S, Simonsen AH, Hasselbalch SG, Zetterberg H, Brinkmalm A, Blennow K. A Parallel Reaction Monitoring Mass Spectrometric Method for Analysis of Potential CSF Biomarkers for Alzheimer's Disease. Proteomics Clin Appl 2017; 12. [PMID: 29028155 DOI: 10.1002/prca.201700131] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Indexed: 01/04/2023]
Abstract
SCOPE The aim of this study was to develop and evaluate a parallel reaction monitoring mass spectrometry (PRM-MS) assay consisting of a panel of potential protein biomarkers in cerebrospinal fluid (CSF). EXPERIMENTAL DESIGN Thirteen proteins were selected based on their association with neurodegenerative diseases and involvement in synaptic function, secretory vesicle function, or innate immune system. CSF samples were digested and two to three peptides per protein were quantified using stable isotope-labeled peptide standards. RESULTS Coefficients of variation were generally below 15%. Clinical evaluation was performed on a cohort of 10 patients with Alzheimer's disease (AD) and 15 healthy subjects. Investigated proteins of the granin family exhibited the largest difference between the patient groups. Secretogranin-2 (p<0.005) and neurosecretory protein VGF (p<0.001) concentrations were lowered in AD. For chromogranin A, two of three peptides had significantly lowered AD concentrations (p<0.01). The concentrations of the synaptic proteins neurexin-1 and neuronal pentraxin-1, as well as neurofascin were also significantly lowered in AD (p<0.05). The other investigated proteins, β2-microglobulin, cystatin C, amyloid precursor protein, lysozyme C, neurexin-2, neurexin-3, and neurocan core protein, were not significantly altered. CONCLUSION AND CLINICAL RELEVANCE PRM-MS of protein panels is a valuable tool to evaluate biomarker candidates for neurodegenerative disorders.
Collapse
Affiliation(s)
- Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Simon Sjödin
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Anja Hviid Simonsen
- Danish Dementia Research Centre, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, University College London Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute, London, UK
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
38
|
Ciregia F, Urbani A, Palmisano G. Extracellular Vesicles in Brain Tumors and Neurodegenerative Diseases. Front Mol Neurosci 2017; 10:276. [PMID: 28912682 PMCID: PMC5583211 DOI: 10.3389/fnmol.2017.00276] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/15/2017] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) can be classified into apoptotic bodies, microvesicles (MVs), and exosomes, based on their origin or size. Exosomes are the smallest and best characterized vesicles which derived from the endosomal system. These vesicles are released from many different cell types including neuronal cells and their functions in the nervous system are investigated. They have been proposed as novel means for intercellular communication, which takes part not only to the normal neuronal physiology but also to the transmission of pathogenic proteins. Indeed, exosomes are fundamental to assemble and transport proteins during development, but they can also transfer neurotoxic misfolded proteins in pathogenesis. The present review will focus on their roles in neurological diseases, specifically brain tumors, such as glioblastoma (GBM), neuroblastoma (NB), medulloblastoma (MB), and metastatic brain tumors and chronic neurodegenerative diseases, such as Alzheimer, Parkinson, multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Huntington, and Prion diseseases highlighting their involvement in spreading neurotoxicity, in therapeutics, and in pathogenesis.
Collapse
Affiliation(s)
- Federica Ciregia
- Department of Pharmacy, University of PisaPisa, Italy.,Department of Clinical and Experimental Medicine, SOD Endocrinology and Metabolism of Organ and Cell Transplants, University of PisaPisa, Italy
| | - Andrea Urbani
- Istituto di Biochimica e Biochimica Clinica, Università CattolicaRome, Italy.,Proteomics and Metabonomics Unit, IRCCS-Fondazione Santa LuciaRome, Italy
| | - Giuseppe Palmisano
- Proteomics and Metabonomics Unit, IRCCS-Fondazione Santa LuciaRome, Italy.,GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of Sao PauloSao Paulo, Brazil
| |
Collapse
|
39
|
Stillman CM, Lopez OL, Becker JT, Kuller LH, Mehta PD, Tracy RP, Erickson KI. Physical activity predicts reduced plasma β amyloid in the Cardiovascular Health Study. Ann Clin Transl Neurol 2017; 4:284-291. [PMID: 28491896 PMCID: PMC5420805 DOI: 10.1002/acn3.397] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 01/27/2023] Open
Abstract
Objective Higher levels of physical activity (PA) reduce the risk of cognitive impairment, but the underlying mechanisms are unclear. Using longitudinal data from the Cardiovascular Health Study, we examined whether PA predicted plasma Aβ levels and risk for cognitive decline 9–13 years later. Methods Linear and logistic regressions (controlling for APOE status, age, gender, body mass index, cardiovascular disease, brain white matter lesions, and cystatin C levels) tested associations between PA, Aβ, and cognitive impairment in a sample of 149 cognitively normal older adults (mean age 83 years). Results More PA at baseline predicted lower levels of Aβ 9–13 years later. Higher Aβ levels at year 9 predicted greater risk for cognitive impairment at year 13. Levels of Aβ at year 9 mediated the relationship between PA and cognitive impairment. Interpretation Greater PA may reduce plasma levels of a neurotoxic peptide at an age when the risk for cognitive impairment is especially high.
Collapse
Affiliation(s)
- Chelsea M Stillman
- Department of Psychiatry University of Pittsburgh School of Medicine Pittsburgh Pennsylvania
| | - Oscar L Lopez
- Department of Psychiatry University of Pittsburgh School of Medicine Pittsburgh Pennsylvania.,Department of Neurology University of Pittsburgh School of Medicine Pittsburgh Pennsylvania
| | - James T Becker
- Department of Psychiatry University of Pittsburgh School of Medicine Pittsburgh Pennsylvania.,Department of Neurology University of Pittsburgh School of Medicine Pittsburgh Pennsylvania.,Department of Psychology University of Pittsburgh Pittsburgh Pennsylvania
| | - Lewis H Kuller
- Graduate School of Public Health University of Pittsburgh Pittsburgh Pennsylvania
| | - Pankaj D Mehta
- New York State Institute for Basic Research in Developmental Disabilities Staten Island New York
| | - Russell P Tracy
- Department of Pathology & Laboratory Medicine University of Vermont College of Medicine Burlington Vermont
| | - Kirk I Erickson
- Department of Psychology University of Pittsburgh Pittsburgh Pennsylvania
| |
Collapse
|
40
|
Wang R, Chen Z, Fu Y, Wei X, Liao J, Liu X, He B, Xu Y, Zou J, Yang X, Weng R, Tan S, McElroy C, Jin K, Wang Q. Plasma Cystatin C and High-Density Lipoprotein Are Important Biomarkers of Alzheimer's Disease and Vascular Dementia: A Cross-Sectional Study. Front Aging Neurosci 2017; 9:26. [PMID: 28223934 PMCID: PMC5294921 DOI: 10.3389/fnagi.2017.00026] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/27/2017] [Indexed: 01/02/2023] Open
Abstract
Objectives: Cystatin C (Cys C) and high-density lipoprotein (HDL) play critical roles in neurodegenerative diseases, such as dementia, Alzheimer’s disease (AD) and vascular dementia (VaD). However, whether they can be used as reliable biomarkers to distinguish patients with dementia from healthy subjects and to determine disease severity remain largely unknown. Methods: We conducted a cross-sectional study to determine plasma Cys C and HDL levels of 88 patients with dementia (43 AD patients, 45 VaD patients) and 45 healthy age-matched controls. The severity of dementia was determined based on the Schwab and England Activities of Daily Living (ADL) Scale, the Mini-mental State Examination (MMSE), the Global Deterioration Scale (GDS), the Lawton Instrumental ADL (IADL) Scale, and the Hachinski Ischemia Scale (Hachinski). Receiver operating characteristic (ROC) curves were calculated to determine the diagnostic accuracy of Cys C and HDL levels in distinguishing patients with dementia from healthy subjects. Results: We found that plasma Cys C levels were higher, but HDL levels were lower in AD and VaD patients respectively, compared to healthy control subjects. Yet, Cys C levels were highest among patients with VaD. Interestingly, plasma Cys C levels were significantly correlated with IADL Scale scores. In addition, the ROC curves for Cys C (area under the curve, AUC 0.816 for AD, AUC 0.841 for VaD) and HDL (AUC 0.800 for AD, AUC 0.731 for VaD) exhibited potential diagnostic value in distinguishing AD/VaD patients from healthy subjects. While the ROC curve for the combination of Cys C and HDL (AUC 0.873 for AD, AUC 0.897 for VaD) showed higher diagnostic accuracy in distinguishing AD/VaD patients from healthy subjects than the separate curves for each parameter. Conclusions: Our findings suggest that the inflammatory mediators Cys C and HDL may play important roles in the pathogenesis of dementia, and plasma Cys C and HDL levels may be useful screening tools for differentiating AD/VaD patients from healthy subjects.
Collapse
Affiliation(s)
- Rui Wang
- Departments of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Zhaoyu Chen
- Departments of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Yongmei Fu
- Departments of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Xiaobo Wei
- Departments of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Jinchi Liao
- Departments of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Xu Liu
- Departments of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Bingjun He
- Departments of Radiology, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Yunqi Xu
- Department of Neurology, Nanfang Hospital, Southern Medical University Guangzhou, China
| | - Jing Zou
- Departments of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Xiaoyan Yang
- Departments of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Ruihui Weng
- Departments of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, China
| | - Sheng Tan
- Departments of Neurology, Zhujiang Hospital, Southern Medical University Guangzhou, China
| | - Christopher McElroy
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Qing Wang
- Departments of Neurology, The Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, China; Guangdong Province Key Laboratory of Brain Function and DiseaseGuangzhou, China
| |
Collapse
|
41
|
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: 99] [Impact Index Per Article: 11.0] [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
|
42
|
Mathews PM, Levy E. Cystatin C in aging and in Alzheimer's disease. Ageing Res Rev 2016; 32:38-50. [PMID: 27333827 DOI: 10.1016/j.arr.2016.06.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/08/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
Under normal conditions, the function of catalytically active proteases is regulated, in part, by their endogenous inhibitors, and any change in the synthesis and/or function of a protease or its endogenous inhibitors may result in inappropriate protease activity. Altered proteolysis as a result of an imbalance between active proteases and their endogenous inhibitors can occur during normal aging, and such changes have also been associated with multiple neuronal diseases, including Amyotrophic Lateral Sclerosis (ALS), rare heritable neurodegenerative disorders, ischemia, some forms of epilepsy, and Alzheimer's disease (AD). One of the most extensively studied endogenous inhibitor is the cysteine-protease inhibitor cystatin C (CysC). Changes in the expression and secretion of CysC in the brain have been described in various neurological disorders and in animal models of neurodegeneration, underscoring a role for CysC in these conditions. In the brain, multiple in vitro and in vivo findings have demonstrated that CysC plays protective roles via pathways that depend upon the inhibition of endosomal-lysosomal pathway cysteine proteases, such as cathepsin B (Cat B), via the induction of cellular autophagy, via the induction of cell proliferation, or via the inhibition of amyloid-β (Aβ) aggregation. We review the data demonstrating the protective roles of CysC under conditions of neuronal challenge and the protective pathways induced by CysC under various conditions. Beyond highlighting the essential role that balanced proteolytic activity plays in supporting normal brain aging, these findings suggest that CysC is a therapeutic candidate that can potentially prevent brain damage and neurodegeneration.
Collapse
Affiliation(s)
- Paul M Mathews
- Departments of Psychiatry, New York University School of Medicine, USA; Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Efrat Levy
- Departments of Psychiatry, New York University School of Medicine, USA; Biochemistry and Molecular Pharmacology, New York University School of Medicine, USA; Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA.
| |
Collapse
|
43
|
Grewal R, Haghighi M, Huang S, Smith AG, Cao C, Lin X, Lee DC, Teten N, Hill AM, Selenica MLB. Identifying biomarkers of dementia prevalent among amnestic mild cognitively impaired ethnic female patients. ALZHEIMERS RESEARCH & THERAPY 2016; 8:43. [PMID: 27756387 PMCID: PMC5067885 DOI: 10.1186/s13195-016-0211-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022]
Abstract
Background There is a need to investigate biomarkers that are indicative of the progression of dementia in ethnic patient populations. The disparity of information in these populations has been the focus of many clinical and academic centers, including ours, to contribute to a higher success rate in clinical trials. In this study, we have investigated plasma biomarkers in amnestic mild cognitively impaired (aMCI) female patient cohorts in the context of ethnicity and cognitive status. Method A panel of 12 biomarkers involved in the progression of brain pathology, inflammation, and cardiovascular disorders were investigated in female cohorts of African American, Hispanic, and White aMCI patients. Both biochemical and algorithmic analyses were applied to correlate biomarker levels measured during the early stages of the disease for each ethnicity. Results We report elevated plasma Aβ40, Aβ42, YKL-40, and cystatin C levels in the Hispanic cohort at early aMCI status. In addition, elevated plasma Aβ40 levels were associated with the aMCI status in both White and African American patient cohorts by the decision tree algorithm. Eotaxin-1 levels, as determined by the decision tree algorithm and biochemically measured total tau levels, were associated with the aMCI status in the African American cohort. Conclusions Overall, our data displayed novel differences in the plasma biomarkers of the aMCI female cohorts where the plasma levels of several biomarkers distinguished between each ethnicity at an early aMCI stage. Identification of these plasma biomarkers encourages new areas of investigation among aMCI ethnic populations, including larger patient cohorts and longitudinal study designs. Electronic supplementary material The online version of this article (doi:10.1186/s13195-016-0211-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rinko Grewal
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Mona Haghighi
- Department of Industrial and Systems Engineering, University of Washington, 3900 Northeast Stevens Way, Seattle, WA, 98195, USA
| | - Shuai Huang
- Department of Industrial and Systems Engineering, University of Washington, 3900 Northeast Stevens Way, Seattle, WA, 98195, USA.,School of Aging Studies, University of South Florida, 4202 E Fowler Ave, Tampa, FL, 33620, USA
| | - Amanda G Smith
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Psychiatry and Behavioral Medicine, College of Medicine, University of South Florida, 3515 E Fletcher Ave, Tampa, FL, 33613, USA
| | - Chuanhai Cao
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Xiaoyang Lin
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Daniel C Lee
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Nancy Teten
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Angela M Hill
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA.,Department of Pharmacotherapeutics and Clinical Research, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Maj-Linda B Selenica
- Byrd Alzheimer's Institute, University of South Florida, 4001 E. Fletcher Ave, Tampa, FL, 33613, USA. .,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
44
|
Babić M, Svob Štrac D, Mück-Šeler D, Pivac N, Stanić G, Hof PR, Simić G. Update on the core and developing cerebrospinal fluid biomarkers for Alzheimer disease. Croat Med J 2015; 55:347-65. [PMID: 25165049 PMCID: PMC4157375 DOI: 10.3325/cmj.2014.55.347] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Alzheimer disease (AD) is a complex neurodegenerative disorder, whose prevalence will dramatically rise by 2050. Despite numerous clinical trials investigating this disease, there is still no effective treatment. Many trials showed negative or inconclusive results, possibly because they recruited only patients with severe disease, who had not undergone disease-modifying therapies in preclinical stages of AD before severe degeneration occurred. Detection of AD in asymptomatic at risk individuals (and a few presymptomatic individuals who carry an autosomal dominant monogenic AD mutation) remains impractical in many of clinical situations and is possible only with reliable biomarkers. In addition to early diagnosis of AD, biomarkers should serve for monitoring disease progression and response to therapy. To date, the most promising biomarkers are cerebrospinal fluid (CSF) and neuroimaging biomarkers. Core CSF biomarkers (amyloid β1-42, total tau, and phosphorylated tau) showed a high diagnostic accuracy but were still unreliable for preclinical detection of AD. Hence, there is an urgent need for detection and validation of novel CSF biomarkers that would enable early diagnosis of AD in asymptomatic individuals. This article reviews recent research advances on biomarkers for AD, focusing mainly on the CSF biomarkers. In addition to core CSF biomarkers, the potential usefulness of novel CSF biomarkers is discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Goran Simić
- Goran Šimić, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000 Zagreb, Croatia,
| |
Collapse
|
45
|
Abstract
Alzheimer disease (AD) is a dementing, neurodegenerative disorder that affects approximately 500,000 Canadians and its prevalence is expected to double over the next 30 years. Although several medications may temporarily augment cognitive abilities in AD, there presently exists no proven method to avoid the inevitable clinical deterioration in this devastating condition. The delineation of risk factors for the development of AD offers hope for the advent of effective prevention or interventions that might retard the onset of symptoms. In this article, we provide a comprehensive review of midlife risk factors implicated in the etiopathogenesis of sporadic AD. Although some risk factors are heritable and largely beyond our control, others are determined by lifestyle or environment and are potentially modifiable. In a companion paper, we introduce the concept of an Alzheimer Risk Assessment Clinic for ascertainment and mitigation of these and other putative dementia risk factors in middle-aged adults.
Collapse
|
46
|
Slinin Y, Peters KW, Ishani A, Yaffe K, Fink HA, Stone KL, Steffes M, Ensrud KE. Cystatin C and cognitive impairment 10 years later in older women. J Gerontol A Biol Sci Med Sci 2014; 70:771-8. [PMID: 25362662 DOI: 10.1093/gerona/glu189] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/08/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Results of prospective studies examining the association between cystatin C and incident cognitive impairment have been inconsistent. We tested the hypothesis that there is a U-shaped association in older women between cystatin C and risk of incident cognitive impairment 10 years later. METHODS We conducted a longitudinal analysis of a prospective cohort of 1,332 community-dwelling elderly women without dementia at baseline who had baseline cystatin C and serum creatinine measurements and completed an extended cognitive battery of neuropsychological tests with determination of cognitive status 10 years later. Incident cognitive impairment was defined as either new onset of adjudicated diagnosis of mild cognitive impairment or dementia. RESULTS Incident mild cognitive impairment or dementia was identified among 140 (26.0%) women in quartile 1 (Q1), 122 (22.6%) in Q2, 121 (22.5%) in Q3, and 156 (28.9%) in Q4 of cystatin C. In the fully adjusted model, compared to women in Q2-Q3 of cystatin C, adjusted odds ratios (95% CI) for incident cognitive impairment were 1.31 (0.98-1.75) for Q1, and 1.25 (0.94-1.66) for Q4 Compared to women in Q2-Q3 of estimated glomerular filtration rate (eGFRCysC), adjusted odds ratios (95% CI) for incident cognitive impairment after 10 years of follow-up were 1.18 (0.88-1.58) for Q4 (eGFRCysC 76.1-109.4mL/min/1.73 m(2)) and 1.26 (0.94-1.67) for Q1 (eGFRCysC 21.8-55.5mL/min/1.73 m(2)). CONCLUSIONS These results support a U-shaped association between cystatin C concentration and risk of cognitive impairment or dementia 10 years later, but the association is not independent of potential confounding factors.
Collapse
Affiliation(s)
- Yelena Slinin
- Department of Medicine, University of Minnesota, Minneapolis. Department of Medicine, Minneapolis VA Health Care System, Minnesota.
| | | | - Areef Ishani
- Department of Medicine, University of Minnesota, Minneapolis. Department of Medicine, Minneapolis VA Health Care System, Minnesota
| | - Kristine Yaffe
- Department of Psychiatry, Neurology, and Epidemiology & Biostatistics, University of California, San Francisco
| | - Howard A Fink
- Department of Medicine, University of Minnesota, Minneapolis. Department of Medicine, Minneapolis VA Health Care System, Minnesota. Division of Epidemiology & Community Health and
| | - Katie L Stone
- California Pacific Medical Center Research Institute, San Francisco
| | - Michael Steffes
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Kristine E Ensrud
- Department of Medicine, University of Minnesota, Minneapolis. Department of Medicine, Minneapolis VA Health Care System, Minnesota. Division of Epidemiology & Community Health and
| | | |
Collapse
|
47
|
Yaffe K, Kurella-Tamura M, Ackerson L, Hoang TD, Anderson AH, Duckworth M, Go AS, Krousel-Wood M, Kusek JW, Lash JP, Ojo A, Robinson N, Sehgal AR, Sondheimer JH, Steigerwalt S, Townsend RR. Higher levels of cystatin C are associated with worse cognitive function in older adults with chronic kidney disease: the chronic renal insufficiency cohort cognitive study. J Am Geriatr Soc 2014; 62:1623-9. [PMID: 25125225 DOI: 10.1111/jgs.12986] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES To determine the association between cognition and levels of cystatin C in persons with chronic kidney disease (CKD). DESIGN Prospective observational study. SETTING Chronic Renal Insufficiency Cohort Cognitive Study. PARTICIPANTS Individuals with a baseline cognitive assessment completed at the same visit as serum cystatin C measurement (N = 821; mean age 64.9, 50.6% male, 48.6% white). MEASUREMENTS Levels of serum cystatin C were categorized into tertiles; cognitive function was assessed using six neuropsychological tests. Scores on these tests were compared across tertiles of cystatin C using linear regression and logistic regression to examine the association between cystatin C level and cognitive performance (1 standard deviation difference from the mean). RESULTS After multivariable adjustment for age, race, education, and medical comorbidities in linear models, higher levels of cystatin C were associated with worse cognition on the modified Mini-Mental State Examination, Buschke Delayed Recall, Trail-Making Test Part (Trails) A and Part B, and Boston Naming (P < .05 for all). This association remained statistically significant for Buschke Delayed Recall (P = .01) and Trails A (P = .03) after additional adjustment for estimated glomerular filtration rate (eGFR). The highest tertile of cystatin C was associated with greater likelihood of poor performance on Trails A (odds ratio (OR) = 2.17, 95% confidence interval (CI) = 1.16-4.06), Trails B (OR = 1.89, 95% CI = 1.09-3.27), and Boston Naming (OR = 1.85, 95% CI = 1.07-3.19) than the lowest tertile after multivariate adjustment in logistic models. CONCLUSION In individuals with CKD, higher serum cystatin C levels were associated with worse cognition and greater likelihood of poor cognitive performance on attention, executive function, and naming. Cystatin C is a marker of cognitive impairment and may be associated with cognition independent of eGFR.
Collapse
Affiliation(s)
- Kristine Yaffe
- Department of Psychiatry, School of Medicine, University of California at San Francisco, San Francisco, California; Department of Neurology, School of Medicine, University of California at San Francisco, San Francisco, California; Department of Epidemiology and Biostatistics, School of Medicine, University of California at San Francisco, San Francisco, California; San Francisco Veterans Affairs Medical Center, San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Greco V, Pieragostino D, Piras C, Aebersold R, Wiltfang J, Caltagirone C, Bernardini S, Urbani A. Direct analytical sample quality assessment for biomarker investigation: qualifying cerebrospinal fluid samples. Proteomics 2014; 14:1954-62. [PMID: 25044759 DOI: 10.1002/pmic.201300565] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/09/2014] [Accepted: 07/10/2014] [Indexed: 01/05/2023]
Abstract
Measurement of biochemical markers represents an important aid to clinicians in the early diagnosis and prognosis of neurological diseases. Many factors can contribute to increase the chances that a biomarker study becomes successful. In a cerebrospinal fluid analysis (CSF), more than 84% of laboratory errors can be attributed to several preanalytical variables that include CSF collection, storage, and freeze thawing cycles. In this concept paper, we focus on some critical issues arising from basic proteomics investigation in order to highlight some key elements of CSF quality control. Furthermore, we propose a direct assessment of sample quality (DASQ) by applying a fast MALDI-TOF-MS methodology to evaluate molecular features of sample degradation and oxidation. We propose DASQ as a fast and simple initial step to be included in large-scale projects for neurological biomarker studies. In fact, such a procedure will improved the development of standardized protocols in order to have well-characterized CSF biobanks.
Collapse
Affiliation(s)
- Viviana Greco
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy; IRCCS-Santa Lucia Foundation, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Gabelle A, Schraen S, Gutierrez LA, Pays C, Rouaud O, Buée L, Touchon J, Helmer C, Lambert JC, Berr C. Plasma β-amyloid 40 levels are positively associated with mortality risks in the elderly. Alzheimers Dement 2014; 11:672-80. [PMID: 25022539 DOI: 10.1016/j.jalz.2014.04.515] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/13/2014] [Accepted: 04/16/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND We evaluated if plasma β-amyloid (Aβ) levels were associated with mortality risks in a subsample of the French Three-City (3C) prospective cohort study. METHODS Analyses were based on 1254 participants randomly selected from the initial 3C cohort stratified by center, sex, and age in the context of a nested case-cohort study to investigate biological variables. Associations between plasma Aβ and mortality were assessed with the Cox regression model with delayed entry including various potential confounding factors and testing possible mediators. RESULTS A relationship between high plasma Aβ1-40 concentrations and risk of mortality (hazards ratio, 1.15; 95% confidence interval, 1.01-1.31, P = .03) was unveiled independently of age, educational level, vascular risk factors, diet, physical activity, cognitive impairment, or frailty status. It was only modified when we included cystatin C levels. CONCLUSIONS Further investigations are needed to determine precisely the pathophysiological roles of plasma Aβ1-40 and cystatin C and before envisioning any future clinical applications.
Collapse
Affiliation(s)
- Audrey Gabelle
- Department of Neurology, Centre Mémoire Ressources Recherche Languedoc-Roussillon, CHRU Gui de Chauliac Hospital, Montpellier, France; University Montpellier 1, Montpellier, France
| | - Susanna Schraen
- Université Droit et Santé de Lille, Lille, France; CHRU de Lille, Lille, France; INSERM UMR837, Lille, France
| | - Laure-Anne Gutierrez
- Department of Neurology, Centre Mémoire Ressources Recherche Languedoc-Roussillon, CHRU Gui de Chauliac Hospital, Montpellier, France; INSERM U1061, Hôpital La Colombière, Montpellier, France
| | - Cecile Pays
- Department of Neurology, Centre Mémoire Ressources Recherche Languedoc-Roussillon, CHRU Gui de Chauliac Hospital, Montpellier, France; INSERM U1061, Hôpital La Colombière, Montpellier, France
| | - Olivier Rouaud
- Department of Neurology, Centre Mémoire Ressources Recherche, CHRU Dijon, Dijon, France
| | - Luc Buée
- Université Droit et Santé de Lille, Lille, France; CHRU de Lille, Lille, France; INSERM UMR837, Lille, France
| | - Jacques Touchon
- Department of Neurology, Centre Mémoire Ressources Recherche Languedoc-Roussillon, CHRU Gui de Chauliac Hospital, Montpellier, France; INSERM U1061, Hôpital La Colombière, Montpellier, France
| | | | - Jean-Charles Lambert
- CHRU de Lille, Lille, France; INSERM U744, Lille, France; Institut Pasteur de Lille, Lille, France; Université de Lille Nord de France, Lille, France
| | - Claudine Berr
- Department of Neurology, Centre Mémoire Ressources Recherche Languedoc-Roussillon, CHRU Gui de Chauliac Hospital, Montpellier, France; INSERM U1061, Hôpital La Colombière, Montpellier, France; Université Montpellier 1, Hôpital La Colombière, Montpellier, France.
| |
Collapse
|
50
|
Watanabe K, Watanabe T, Nakayama M. Cerebro-renal interactions: impact of uremic toxins on cognitive function. Neurotoxicology 2014; 44:184-93. [PMID: 25003961 DOI: 10.1016/j.neuro.2014.06.014] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 06/13/2014] [Accepted: 06/27/2014] [Indexed: 01/21/2023]
Abstract
Cognitive impairment (CI) associated with chronic kidney disease (CKD) has received attention as an important problem in recent years. Causes of CI with CKD are multifactorial, and include cerebrovascular disease, renal anemia, secondary hyperparathyroidism, dialysis disequilibrium, and uremic toxins (UTs). Among these causes, little is known about the role of UTs. We therefore selected 21 uremic compounds, and summarized reports of cerebro-renal interactions associated with UTs. Among the compounds, uric acid, indoxyl sulfate, p-cresyl sulfate, interleukin 1-β, interleukin 6, TNF-α, and PTH were most likely to affect the cerebro-renal interaction dysfunction; however, sufficient data have not been obtained for other UTs. Notably, most of the data were not obtained under uremic conditions; therefore, the impact and mechanism of each UT on cognition and central nervous system in uremic state remains unknown. At present, impacts and mechanisms of UT effects on cognition are poorly understood. Clarifying the mechanisms and establishing novel therapeutic strategies for cerebro-renal interaction dysfunction is expected to be subject of future research.
Collapse
Affiliation(s)
- Kimio Watanabe
- Department of Nephrology, Hypertension, Diabetology, Endocrinology and Metabolism, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Tsuyoshi Watanabe
- Department of Nephrology, Hypertension, Diabetology, Endocrinology and Metabolism, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Masaaki Nakayama
- Department of Nephrology, Hypertension, Diabetology, Endocrinology and Metabolism, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan.
| |
Collapse
|