1
|
Parekh P, Serra M, Allaw M, Perra M, Pinna A, Manconi M, Morelli M. Extract from Nasco pomace loaded in nutriosomes exerts anti-inflammatory effects in the MPTP mouse model of Parkinson's disease. Exp Neurol 2024; 382:114958. [PMID: 39303846 DOI: 10.1016/j.expneurol.2024.114958] [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: 05/08/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Neuroinflammation has recently emerged as a key event in Parkinson's disease (PD) pathophysiology and as a potential target for disease-modifying therapies. Plant-derived extracts, rich in bioactive phytochemicals with antioxidant properties, have shown potential in this regard. Yet their clinical utility is hampered by poor systemic availability and rapid metabolism. Recently, our group demonstrated that intragastric delivery of Nasco pomace extract via nutriosomes (NN), a novel nanoliposome formulation, contrasts the degeneration of nigrostriatal dopaminergic neurons in a subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. In the present study, we investigated the impact of intragastric NN treatment on the reactivity of glial cells in the substantia nigra pars compacta (SNc) and caudate-putamen (CPu) of MPTP-treated mice. To this scope, in mice exposed to MPTP (20 mg/kg/day, × 4 days), we conducted immunohistochemistry analyses of glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (IBA1) to assess the responsiveness of astrocytes and microglial cells, respectively. Additionally, we studied the co-localization of the pro-inflammatory interleukin (IL)-1β and tumor necrosis factor (TNF)-α with IBA1 to obtain insights into microglial phenotype. Immunohistochemical results showed that NN administration significantly mitigated astrogliosis and microgliosis in the CPu and SNc of mice receiving subacute MPTP treatment, with region-specific variations in anti-inflammatory efficacy. Remarkably, the CPu showed a heightened response to NN treatment, including a pronounced decrease in microglial IL-1β and TNF-α production. Altogether, these findings underscore the anti-inflammatory effects of NN treatment and provide a potential mechanism underlying the neuroprotective effects previously observed in a subacute MPTP mouse model of PD.
Collapse
Affiliation(s)
- Pathik Parekh
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.
| | - Mohamad Allaw
- Department of Life and Environmental Sciences, University of Cagliari, Italy
| | - Matteo Perra
- Department of Life and Environmental Sciences, University of Cagliari, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Maria Manconi
- Department of Life and Environmental Sciences, University of Cagliari, Italy.
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| |
Collapse
|
2
|
Sun X, Gu R, Bai J. Differentiation and regulation of CD4 + T cell subsets in Parkinson's disease. Cell Mol Life Sci 2024; 81:352. [PMID: 39153043 PMCID: PMC11335276 DOI: 10.1007/s00018-024-05402-0] [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: 03/09/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and its hallmark pathological features are the loss of dopaminergic (DA) neurons in the midbrain substantia nigra pars compacta (SNpc) and the accumulation of alpha-synuclein (α-syn). It has been shown that the integrity of the blood-brain barrier (BBB) is damaged in PD patients, and a large number of infiltrating T cells and inflammatory cytokines have been detected in the cerebrospinal fluid (CSF) and brain parenchyma of PD patients and PD animal models, including significant change in the number and proportion of different CD4+ T cell subsets. This suggests that the neuroinflammatory response caused by CD4+ T cells is an important risk factor for the development of PD. Here, we systematically review the differentiation of CD4+ T cell subsets, and focus on describing the functions and mechanisms of different CD4+ T cell subsets and their secreted cytokines in PD. We also summarize the current immunotherapy targeting CD4+ T cells with a view to providing assistance in the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Xiaowei Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
- Southwest United Graduate School, Kunming, 650500, China
| | - Rou Gu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
- Southwest United Graduate School, Kunming, 650500, China.
| |
Collapse
|
3
|
Stoll AC, Kemp CJ, Patterson JR, Kubik M, Kuhn N, Benskey M, Duffy MF, Luk KC, Sortwell CE. Alpha-synuclein inclusion responsive microglia are resistant to CSF1R inhibition. J Neuroinflammation 2024; 21:108. [PMID: 38664840 PMCID: PMC11045433 DOI: 10.1186/s12974-024-03108-5] [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: 05/03/2023] [Accepted: 04/22/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by the presence of proteinaceous alpha-synuclein (α-syn) inclusions (Lewy bodies), markers of neuroinflammation and the progressive loss of nigrostriatal dopamine (DA) neurons. These pathological features can be recapitulated in vivo using the α-syn preformed fibril (PFF) model of synucleinopathy. We have previously determined that microglia proximal to PFF-induced nigral α-syn inclusions increase in soma size, upregulate major-histocompatibility complex-II (MHC-II) expression, and increase expression of a suite of inflammation-associated transcripts. This microglial response is observed months prior to degeneration, suggesting that microglia reacting to α-syn inclusion may contribute to neurodegeneration and could represent a potential target for novel therapeutics. The goal of this study was to determine whether colony stimulating factor-1 receptor (CSF1R)-mediated microglial depletion impacts the magnitude of α-syn aggregation, nigrostriatal degeneration, or the response of microglial in the context of the α-syn PFF model. METHODS Male Fischer 344 rats were injected intrastriatally with either α-syn PFFs or saline. Rats were continuously administered Pexidartinib (PLX3397B, 600 mg/kg), a CSF1R inhibitor, to deplete microglia for a period of either 2 or 6 months. RESULTS CSF1R inhibition resulted in significant depletion (~ 43%) of ionized calcium-binding adapter molecule 1 immunoreactive (Iba-1ir) microglia within the SNpc. However, CSF1R inhibition did not impact the increase in microglial number, soma size, number of MHC-II immunoreactive microglia or microglial expression of Cd74, Cxcl10, Rt-1a2, Grn, Csf1r, Tyrobp, and Fcer1g associated with phosphorylated α-syn (pSyn) nigral inclusions. Further, accumulation of pSyn and degeneration of nigral neurons was not impacted by CSF1R inhibition. Paradoxically, long term CSF1R inhibition resulted in increased soma size of remaining Iba-1ir microglia in both control and PFF rats, as well as expression of MHC-II in extranigral regions. CONCLUSIONS Collectively, our results suggest that CSF1R inhibition does not impact the microglial response to nigral pSyn inclusions and that CSF1R inhibition is not a viable disease-modifying strategy for PD.
Collapse
Affiliation(s)
- Anna C Stoll
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Christopher J Kemp
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - Joseph R Patterson
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - Michael Kubik
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - Nathan Kuhn
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - Matthew Benskey
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - Megan F Duffy
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
4
|
Kandpal M, Varshney N, Rawal KS, Jha HC. Gut dysbiosis and neurological modalities: An engineering approach via proteomic analysis of gut-brain axis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:199-248. [PMID: 38762270 DOI: 10.1016/bs.apcsb.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
The human gut microbiota is a complex and dynamic community of microorganisms, that influence metabolic, neurodevelopmental, and immune pathways. Microbial dysbiosis, characterized by changes in microbial diversity and relative abundances, is implicated in the development of various chronic neurological and neurodegenerative disorders. These disorders are marked by the accumulation of pathological protein aggregates, leading to the progressive loss of neurons and behavioural functions. Dysregulations in protein-protein interaction networks and signalling complexes, critical for normal brain function, are common in neurological disorders but challenging to unravel, particularly at the neuron and synapse-specific levels. To advance therapeutic strategies, a deeper understanding of neuropathogenesis, especially during the progressive disease phase, is needed. Biomarkers play a crucial role in identifying disease pathophysiology and monitoring disease progression. Proteomics, a powerful technology, shows promise in accelerating biomarker discovery and aiding in the development of novel treatments. In this chapter, we provide an in-depth overview of how proteomic techniques, utilizing various biofluid samples from patients with neurological conditions and diverse animal models, have contributed valuable insights into the pathogenesis of numerous neurological disorders. We also discuss the current state of research, potential challenges, and future directions in proteomic approaches to unravel neuro-pathological conditions.
Collapse
Affiliation(s)
- Meenakshi Kandpal
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Nidhi Varshney
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Kunal Sameer Rawal
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India; Centre for Rural Development & Technology, IIT Indore, Indore, India.
| |
Collapse
|
5
|
de Fàbregues O, Sellés M, Ramos-Vicente D, Roch G, Vila M, Bové J. Relevance of tissue-resident memory CD8 T cells in the onset of Parkinson's disease and examination of its possible etiologies: infectious or autoimmune? Neurobiol Dis 2023; 187:106308. [PMID: 37741513 DOI: 10.1016/j.nbd.2023.106308] [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: 12/16/2022] [Revised: 05/05/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023] Open
Abstract
Tissue-resident memory CD8 T cells are responsible for local immune surveillance in different tissues, including the brain. They constitute the first line of defense against pathogens and cancer cells and play a role in autoimmunity. A recently published study demonstrated that CD8 T cells with markers of residency containing distinct granzymes and interferon-γ infiltrate the parenchyma of the substantia nigra and contact dopaminergic neurons in an early premotor stage of Parkinson's disease. This infiltration precedes α-synuclein aggregation and neuronal loss in the substantia nigra, suggesting a relevant role for CD8 T cells in the onset of the disease. To date, the nature of the antigen that initiates the adaptive immune response remains unknown. This review will discuss the role of tissue-resident memory CD8 T cells in brain immune homeostasis and in the onset of Parkinson's disease and other neurological diseases. We also discuss how aging and genetic factors can affect the CD8 T cell immune response and how animal models can be misleading when studying human-related immune response. Finally, we speculate about a possible infectious or autoimmune origin of Parkinson's disease.
Collapse
Affiliation(s)
- Oriol de Fàbregues
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain; Movement Disorders Unit, Neurology Department, Vall d'Hebron University Hospital
| | - Maria Sellés
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - David Ramos-Vicente
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Gerard Roch
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain; Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Catalonia, Spain; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| | - Jordi Bové
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain.
| |
Collapse
|
6
|
Dillon ST, Vasunilashorn SM, Otu HH, Ngo L, Fong T, Gu X, Cavallari M, Touroutoglou A, Shafi M, Inouye SK, Xie Z, Marcantonio ER, Libermann TA. Aptamer-Based Proteomics Measuring Preoperative Cerebrospinal Fluid Protein Alterations Associated with Postoperative Delirium. Biomolecules 2023; 13:1395. [PMID: 37759795 PMCID: PMC10526755 DOI: 10.3390/biom13091395] [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: 08/10/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Delirium is a common postoperative complication among older patients with many adverse outcomes. Due to a lack of validated biomarkers, prediction and monitoring of delirium by biological testing is not currently feasible. Circulating proteins in cerebrospinal fluid (CSF) may reflect biological processes causing delirium. Our goal was to discover and investigate candidate protein biomarkers in preoperative CSF that were associated with the development of postoperative delirium in older surgical patients. We employed a nested case-control study design coupled with high multiplex affinity proteomics analysis to measure 1305 proteins in preoperative CSF. Twenty-four matched delirium cases and non-delirium controls were selected from the Healthier Postoperative Recovery (HiPOR) cohort, and the associations between preoperative protein levels and postoperative delirium were assessed using t-test statistics with further analysis by systems biology to elucidate delirium pathophysiology. Proteomics analysis identified 32 proteins in preoperative CSF that significantly associate with delirium (t-test p < 0.05). Due to the limited sample size, these proteins did not remain significant by multiple hypothesis testing using the Benjamini-Hochberg correction and q-value method. Three algorithms were applied to separate delirium cases from non-delirium controls. Hierarchical clustering classified 40/48 case-control samples correctly, and principal components analysis separated 43/48. The receiver operating characteristic curve yielded an area under the curve [95% confidence interval] of 0.91 [0.80-0.97]. Systems biology analysis identified several key pathways associated with risk of delirium: inflammation, immune cell migration, apoptosis, angiogenesis, synaptic depression and neuronal cell death. Proteomics analysis of preoperative CSF identified 32 proteins that might discriminate individuals who subsequently develop postoperative delirium from matched control samples. These proteins are potential candidate biomarkers for delirium and may play a role in its pathophysiology.
Collapse
Affiliation(s)
- Simon T. Dillon
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.); (X.G.)
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
| | - Sarinnapha M. Vasunilashorn
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Divisions of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Hasan H. Otu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA;
| | - Long Ngo
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Divisions of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Tamara Fong
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew Senior Life, Boston, MA 02131, USA;
| | - Xuesong Gu
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.); (X.G.)
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
| | - Michele Cavallari
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Center for Neurological Imaging, Department of Radiology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Alexandra Touroutoglou
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mouhsin Shafi
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Sharon K. Inouye
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew Senior Life, Boston, MA 02131, USA;
- Divisions of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Zhongcong Xie
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Edward R. Marcantonio
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
- Divisions of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Divisions of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Towia A. Libermann
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.); (X.G.)
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA; (S.M.V.); (L.N.); (T.F.); (M.C.); (A.T.); (M.S.); (Z.X.); (E.R.M.)
| |
Collapse
|
7
|
Yin Z, Chen J, Xia M, Zhang X, Li Y, Chen Z, Bao Q, Zhong W, Yao J, Wu K, Zhao L, Liang F. Assessing causal relationship between circulating cytokines and age-related neurodegenerative diseases: a bidirectional two-sample Mendelian randomization analysis. Sci Rep 2023; 13:12325. [PMID: 37516812 PMCID: PMC10387057 DOI: 10.1038/s41598-023-39520-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023] Open
Abstract
Numerous studies have reported that circulating cytokines (CCs) are linked to age-related neurodegenerative diseases (ANDDs); however, there is a lack of systematic investigation for the causal association. A two-sample bidirectional Mendelian Randomisation (MR) method was utilized to evaluate the causal effect. We applied genetic variants correlated with concentrations of CCs from a genome-wide association study meta-analysis (n = 8293) as instrumental variables. Summary data of three major ANDDs [Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic lateral sclerosis (ALS)] were identified from the IEU OpenGWAS platform (n = 627, 266). Inverse-variance weighted method is the main approach to analyse causal effect, and MR results are verified by several sensitivity and pleiotropy analyses. In directional MR, it suggested that several CCs were nominally correlated with the risk of ANDDs, with a causal odds ratio (OR) of Interleukin (IL)-5 of 0.909 for AD; OR of IL-2 of 1.169 for PD; and OR of Beta nerve growth factor of 1.142 for ALS). In reverse MR, there were some suggestively causal effects of ANDDs on CCs (AD on increased Basic fibroblast growth factor and IL-12 and decreased Stem cell growth factor beta; PD on decreased Monokine induced by interferon-gamma; ALS on decreased Basic fibroblast growth factor and IL-17). The findings were stable across sensitivity and pleiotropy analyses. However, after Bonferroni correction, there is no statistically significant association between CCs and ANDDs. Through the genetic epidemiological approach, our study assessed the role and presented possible causal associations between CCs and ANDDs. Further studies are warranted to verify the causal associations.
Collapse
Affiliation(s)
- Zihan Yin
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Jiao Chen
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Manze Xia
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Xinyue Zhang
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Yaqin Li
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
| | - Zhenghong Chen
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Qiongnan Bao
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Wanqi Zhong
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Jin Yao
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Kexin Wu
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China
| | - Ling Zhao
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China.
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China.
| | - Fanrong Liang
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, 37 Shierqiao Road, Chengdu, 610075, Sichuan, China.
- Acupuncture Clinical Research Center of Sichuan Province, Chengdu, China.
| |
Collapse
|
8
|
Gerasimova T, Stepanenko E, Novosadova L, Arsenyeva E, Shimchenko D, Tarantul V, Grivennikov I, Nenasheva V, Novosadova E. Glial Cultures Differentiated from iPSCs of Patients with PARK2-Associated Parkinson's Disease Demonstrate a Pro-Inflammatory Shift and Reduced Response to TNFα Stimulation. Int J Mol Sci 2023; 24:ijms24032000. [PMID: 36768317 PMCID: PMC9916517 DOI: 10.3390/ijms24032000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/20/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative diseases characterized by progressive loss of midbrain dopaminergic neurons in the substantia nigra. Mutations in the PARK2 gene are a frequent cause of familial forms of PD. Sustained chronic neuroinflammation in the central nervous system makes a significant contribution to neurodegeneration events. In response to inflammatory factors produced by activated microglia, astrocytes change their transcriptional programs and secretion profiles, thus acting as immunocompetent cells. Here, we investigated iPSC-derived glial cell cultures obtained from healthy donors (HD) and from PD patients with PARK2 mutations in resting state and upon stimulation by TNFα. The non-stimulated glia of PD patients demonstrated higher IL1B and IL6 expression levels and increased IL6 protein synthesis, while BDNF and GDNF expression was down-regulated when compared to that of the glial cells of HDs. In the presence of TNFα, all of the glial cultures displayed a multiplied expression of genes encoding inflammatory cytokines: TNFA, IL1B, and IL6, as well as IL6 protein synthesis, although PD glia responded to TNFα stimulation less strongly than HD glia. Our results demonstrated a pro-inflammatory shift, a suppression of the neuroprotective gene program, and some depletion of reactivity to TNFα in PARK2-deficient glia compared to glial cells of HDs.
Collapse
Affiliation(s)
- Tatiana Gerasimova
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Correspondence:
| | - Ekaterina Stepanenko
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Lyudmila Novosadova
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Elena Arsenyeva
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Darya Shimchenko
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Vyacheslav Tarantul
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Igor Grivennikov
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Valentina Nenasheva
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Ekaterina Novosadova
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| |
Collapse
|
9
|
Deus CM, Tavares H, Beatriz M, Mota S, Lopes C. Mitochondrial Damage-Associated Molecular Patterns Content in Extracellular Vesicles Promotes Early Inflammation in Neurodegenerative Disorders. Cells 2022; 11:2364. [PMID: 35954208 PMCID: PMC9367540 DOI: 10.3390/cells11152364] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is a common hallmark in different neurodegenerative conditions that share neuronal dysfunction and a progressive loss of a selectively vulnerable brain cell population. Alongside ageing and genetics, inflammation, oxidative stress and mitochondrial dysfunction are considered key risk factors. Microglia are considered immune sentinels of the central nervous system capable of initiating an innate and adaptive immune response. Nevertheless, the pathological mechanisms underlying the initiation and spread of inflammation in the brain are still poorly described. Recently, a new mechanism of intercellular signalling mediated by small extracellular vesicles (EVs) has been identified. EVs are nanosized particles (30-150 nm) with a bilipid membrane that carries cell-specific bioactive cargos that participate in physiological or pathological processes. Damage-associated molecular patterns (DAMPs) are cellular components recognised by the immune receptors of microglia, inducing or aggravating neuroinflammation in neurodegenerative disorders. Diverse evidence links mitochondrial dysfunction and inflammation mediated by mitochondrial-DAMPs (mtDAMPs) such as mitochondrial DNA, mitochondrial transcription factor A (TFAM) and cardiolipin, among others. Mitochondrial-derived vesicles (MDVs) are a subtype of EVs produced after mild damage to mitochondria and, upon fusion with multivesicular bodies are released as EVs to the extracellular space. MDVs are particularly enriched in mtDAMPs which can induce an immune response and the release of pro-inflammatory cytokines. Importantly, growing evidence supports the association between mitochondrial dysfunction, EV release and inflammation. Here, we describe the role of extracellular vesicles-associated mtDAMPS in physiological conditions and as neuroinflammation activators contributing to neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Sandra Mota
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, III-Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; (C.M.D.); (H.T.); (M.B.)
| | - Carla Lopes
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, III-Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; (C.M.D.); (H.T.); (M.B.)
| |
Collapse
|
10
|
Alhowail A, Alsikhan R, Alsaud M, Aldubayan M, Rabbani SI. Protective Effects of Pioglitazone on Cognitive Impairment and the Underlying Mechanisms: A Review of Literature. Drug Des Devel Ther 2022; 16:2919-2931. [PMID: 36068789 PMCID: PMC9441149 DOI: 10.2147/dddt.s367229] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Ahmad Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
- Correspondence: Ahmad Alhowail, Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia, Tel +9665672025858, Email
| | - Rawan Alsikhan
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Unaizah, Al Qassim, 51911, Kingdom of Saudi Arabia
| | - May Alsaud
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
| | - Maha Aldubayan
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
| | - Syed Imam Rabbani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
| |
Collapse
|
11
|
Abdi IY, Ghanem SS, El-Agnaf OM. Immune-related biomarkers for Parkinson's disease. Neurobiol Dis 2022; 170:105771. [DOI: 10.1016/j.nbd.2022.105771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 12/13/2022] Open
|
12
|
Xiao Z, Lei T, Liu Y, Yang Y, Bi W, Du H. The potential therapy with dental tissue-derived mesenchymal stem cells in Parkinson's disease. Stem Cell Res Ther 2021; 12:5. [PMID: 33407864 PMCID: PMC7789713 DOI: 10.1186/s13287-020-01957-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/27/2020] [Indexed: 12/19/2022] Open
Abstract
Parkinson’s disease (PD), the second most common neurodegenerative disease worldwide, is caused by the loss of dopaminergic (DAergic) neurons in the substantia nigra resulting in a series of motor or non-motor disorders. Current treatment methods are unable to stop the progression of PD and may bring certain side effects. Cell replacement therapy has brought new hope for the treatment of PD. Recently, human dental tissue-derived mesenchymal stem cells have received extensive attention. Currently, dental pulp stem cells (DPSCs) and stem cells from human exfoliated deciduous teeth (SHED) are considered to have strong potential for the treatment of these neurodegenerative diseases. These cells are considered to be ideal cell sources for the treatment of PD on account of their unique characteristics, such as neural crest origin, immune rejection, and lack of ethical issues. In this review, we briefly describe the research investigating cell therapy for PD and discuss the application and progress of DPSCs and SHED in the treatment of PD. This review offers significant and comprehensive guidance for further clinical research on PD.
Collapse
Affiliation(s)
- Zhuangzhuang Xiao
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 XueYuan Road, Haidian District, Beijing, 100083, China
| | - Tong Lei
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 XueYuan Road, Haidian District, Beijing, 100083, China
| | - Yanyan Liu
- Kangyanbao (Beijing) Stem Cell Technology Co., Ltd, Beijing, 102600, China
| | - Yanjie Yang
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 XueYuan Road, Haidian District, Beijing, 100083, China
| | - Wangyu Bi
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 XueYuan Road, Haidian District, Beijing, 100083, China
| | - Hongwu Du
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 XueYuan Road, Haidian District, Beijing, 100083, China.
| |
Collapse
|
13
|
Fonteles AA, Neves JCS, Menezes APF, Pereira JF, Silva ATA, Cunha RA, Andrade GM. ATP Signaling Controlling Dyskinesia Through P2X7 Receptors. Front Mol Neurosci 2020; 13:111. [PMID: 32848592 PMCID: PMC7427508 DOI: 10.3389/fnmol.2020.00111] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
Dopamine replacement therapy with L-3,4-dihydroxyphenylalanine (L-DOPA) is the only temporary therapy for Parkinson's disease (PD), but it triggers dyskinesia over time. Since dyskinesia is associated with increased neuronal firing that bolsters purinergic signaling, we now tested whether the selective and blood-brain barrier-permeable P2X7 receptor antagonist Brilliant Blue-G (BBG, 22.5-45 mg/kg ip) attenuated behavioral, neurochemical and biochemical alterations in rats turned hemiparkinsonian upon unilateral striatal injection of 6-hydroxydopamine (6-OHDA) and treated daily with L-DOPA (30 mg/kg by gavage) for 22 days. The blockade of P2X7 receptors decreased L-DOPA-induced dyskinesia and motor incoordination in hemiparkinsonian rats. In parallel, BBG treatment rebalanced the altered dopamine D1 and D2 receptor density and signaling as well as some neuroinflammation-associated parameters in the striatum and substantia nigra. These findings herald a hitherto unrecognized role for purinergic signaling in the etiopathology of dyskinesia and prompt P2X7 receptor antagonists as novel candidate anti-dyskinesia drugs.
Collapse
Affiliation(s)
- Analu A Fonteles
- Post-Graduate Program in Pharmacology, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Julliana C S Neves
- Post-Graduate Program in Pharmacology, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Ana Paula F Menezes
- Post-Graduate Program in Pharmacology, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Juliana F Pereira
- Post-Graduate Program in Medical Sciences, Department of Medicine, Faculty of Medicine, Center for Research and Drug Development (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Ana Thais A Silva
- Post-Graduate Program in Pharmacology, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Geanne M Andrade
- Post-Graduate Program in Pharmacology, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil.,Post-Graduate Program in Medical Sciences, Department of Medicine, Faculty of Medicine, Center for Research and Drug Development (NPDM), Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
14
|
Santaella A, Kuiperij HB, van Rumund A, Esselink RAJ, van Gool AJ, Bloem BR, Verbeek MM. Inflammation biomarker discovery in Parkinson's disease and atypical parkinsonisms. BMC Neurol 2020; 20:26. [PMID: 31952511 PMCID: PMC6967088 DOI: 10.1186/s12883-020-1608-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/09/2020] [Indexed: 01/09/2023] Open
Abstract
Background Parkinson’s disease (PD) and atypical parkinsonisms (APD) have overlapping symptoms challenging an early diagnosis. Diagnostic accuracy is important because PD and APD have different prognosis and response to treatment. We aimed to identify diagnostic inflammatory biomarkers of PD and APD in cerebrospinal fluid (CSF) using the multiplex proximity extension assay (PEA) technology and to study possible correlations of biomarkers with disease progression. Methods CSF from a longitudinal cohort study consisting of PD and APD patients (PD, n = 44; multiple system atrophy (MSA), n = 14; vascular parkinsonism (VaP), n = 9; and PD with VaP, n = 7) and controls (n = 25) were analyzed. Results Concentrations of CCL28 were elevated in PD compared to controls (p = 0.0001). Five other biomarkers differentiated both MSA and PD from controls (p < 0.05) and 10 biomarkers differentiated MSA from controls, of which two proteins, i.e. beta nerve growth factor (β-NGF) and Delta and Notch like epidermal growth factor-related receptor (DNER), were also present at lower levels in MSA compared to PD (both p = 0.032). Two biomarkers (MCP-1 and MMP-10) positively correlated with PD progression (rho > 0.650; p < 0.01). Conclusions PEA technique identified potential new CSF biomarkers to help to predict the prognosis of PD. Also, we identified new candidate biomarkers to distinguish MSA from PD.
Collapse
Affiliation(s)
- Anna Santaella
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Laboratory Medicine, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Parkinson Center Nijmegen, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Laboratory Medicine, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Anouke van Rumund
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Rianne A J Esselink
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Parkinson Center Nijmegen, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Alain J van Gool
- Laboratory Medicine, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Parkinson Center Nijmegen, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Laboratory Medicine, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Parkinson Center Nijmegen, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
15
|
Microglial Phenotyping in Neurodegenerative Disease Brains: Identification of Reactive Microglia with an Antibody to Variant of CD105/Endoglin. Cells 2019; 8:cells8070766. [PMID: 31340569 PMCID: PMC6678308 DOI: 10.3390/cells8070766] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammation is considered a key pathological process in neurodegenerative diseases, including Alzheimer’s disease (AD) and Parkinson’s disease (PD), but there are still mechanisms not understood. In the brain, most microglia are performing essential homeostatic functions, but can also respond to pathogenic stimuli by producing harmful pro-inflammatory cytokines or free radicals. Distinguishing between damaging and homeostatic microglia in human diseased brain tissues is a challenge. This report describes findings using a monoclonal antibody to CD105/Endoglin (R&D Systems MAB1097) that identifies subtypes of activated microglia. CD105/Endoglin is a co-receptor for transforming growth factor beta (TGFβ) receptor that antagonizes TGFβ signaling. CD105/Endoglin is a marker for vascular endothelial cells, but was originally identified as a marker for activated macrophages. This antibody did not identify endothelial cells in brain sections, only microglia-like cells. In this study, we examined with this antibody tissue section from middle temporal gyrus derived from human brains from normal control subjects with low-plaque pathology, high-plaque pathology, and AD cases, and also substantia nigra samples from control and PD cases, in conjunction with antibodies to markers of pathology and microglia. In low-plaque pathology cases, CD105-positive microglia were mostly absent, but noticeably increased with increasing pathology. CD105-positive cells strongly colocalized with amyloid-beta plaques, but not phosphorylated tau positive tangles. In substantia nigra, strong microglial CD105 staining was observed in microglia associated with degenerating dopaminergic neurons and neuromelanin. In PD cases with few surviving dopaminergic neurons, this staining had decreased. By Western blot, this antibody identified polypeptide bands of 70 kDa in brain samples, and samples from microglia, macrophages, and brain endothelial cells. In comparison with other tested CD105 antibodies, this antibody did not recognize the glycosylated forms of CD105 on Western blots. Overall, the data indicate that this antibody and this marker could have utility for subtyping of microglia in pathologically-involved tissue.
Collapse
|
16
|
Soares NM, Pereira GM, Altmann V, de Almeida RMM, Rieder CRM. Cortisol levels, motor, cognitive and behavioral symptoms in Parkinson's disease: a systematic review. J Neural Transm (Vienna) 2018; 126:219-232. [PMID: 30374595 DOI: 10.1007/s00702-018-1947-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/22/2018] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is a progressive and multifactorial neurodegenerative disease. It has been suggested that a dysregulation of the hypothalamic-pituitary-adrenal axis (HPA) occurs in PD. Furthermore, this dysregulation may be involved in triggering, exacerbation or progression of disease. The objective of this study was to systematically review the literature regarding cortisol levels and their relation with motor, cognitive and behavioral symptoms in patients with PD. A systematic search was performed in PubMed and Embase databases, according to PRISMA norms. Twenty-one studies were included, which evaluated baseline levels of cortisol and motor, cognitive, behavioral symptoms, drugs administration or deep brain stimulation to PD treatment. Sample size ranged from 7 to 249 individuals. In 14 studies that assessed cortisol levels in PD patients, seven showed elevation of cortisol levels. In relation to symptomatology, high levels of cortisol were associated with worst functional scores evaluated by UPDRS, depression and behavior in risk preference. Medication interactions showed an influence on the regulation of cortisol release, mainly, conventional drugs used in the PD's treatment, such as levodopa. The results found in this review point to a possible relationship between cortisol levels and symptoms in PD, indicating that an HPA axis dysfunction related to cortisol level occurs in PD.
Collapse
Affiliation(s)
- Nayron Medeiros Soares
- Medical Science Post Graduation Program, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2400, Porto Alegre, RS, Brazil.
- Institute of Psychology, Laboratory of Psychology, Neuroscience and Behavior (LPNeC), Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, Brazil.
- Hospital de Clinicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, Brazil.
- Federal University of Health Science of Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, RS, Brazil.
| | - Gabriela Magalhães Pereira
- Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2400, Porto Alegre, RS, Brazil
- Institute of Psychology, Laboratory of Psychology, Neuroscience and Behavior (LPNeC), Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, Brazil
| | - Vivian Altmann
- Institute of Biosciences, Federal University of Rio Grande do Sul, Av. Bento Gonçalves, 9500, Porto Alegre, RS, Brazil
- Hospital de Clinicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, Brazil
| | - Rosa Maria Martins de Almeida
- Institute of Psychology, Laboratory of Psychology, Neuroscience and Behavior (LPNeC), Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, Brazil
| | - Carlos R M Rieder
- Medical Science Post Graduation Program, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2400, Porto Alegre, RS, Brazil
- Hospital de Clinicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, Brazil
- Federal University of Health Science of Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, RS, Brazil
| |
Collapse
|
17
|
Santpere G, Garcia-Esparcia P, Andres-Benito P, Lorente-Galdos B, Navarro A, Ferrer I. Transcriptional network analysis in frontal cortex in Lewy body diseases with focus on dementia with Lewy bodies. Brain Pathol 2017; 28:315-333. [PMID: 28321951 DOI: 10.1111/bpa.12511] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/15/2017] [Indexed: 12/13/2022] Open
Abstract
The present study investigates global transcriptional changes in frontal cortex area 8 in incidental Lewy Body disease (iLBD), Parkinson disease (PD) and Dementia with Lewy bodies (DLB). We identified different coexpressed gene sets associated with disease stages, and gene ontology categories enriched in gene modules and differentially expressed genes including modules or gene clusters correlated to iLBD comprising upregulated dynein genes and taste receptors, and downregulated innate inflammation. Focusing on DLB, we found modules with genes significantly enriched in functions related to RNA and DNA production, mitochondria and energy metabolism, purine metabolism, chaperone and protein folding system and synapses and neurotransmission (particularly the GABAergic system). The expression of more than fifty selected genes was assessed with real time quantitative polymerase chain reaction. Our findings provide, for the first time, evidence of molecular cortical alterations in iLBD and involvement of several key metabolic pathways and gene hubs in DLB which may underlie cognitive impairment and dementia.
Collapse
Affiliation(s)
- Gabriel Santpere
- Department of Neurobiology, Yale School of Medicine, New Haven, CT.,Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain
| | - Paula Garcia-Esparcia
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Pol Andres-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Belen Lorente-Galdos
- Department of Neurobiology, Yale School of Medicine, New Haven, CT.,Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain
| | - Arcadi Navarro
- Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain.,Institute of Science and Technology, Centre for Genomic Regulation (CRG), Barcelona, Spain.,National Institute for Bioinformatics (INB), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain.,Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain.,Institute of Neurosciences, University of Barcelona, Hospitalet de Llobregat, Spain.,CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain
| |
Collapse
|
18
|
Walker DG, Lue LF, Tang TM, Adler CH, Caviness JN, Sabbagh MN, Serrano GE, Sue LI, Beach TG. Changes in CD200 and intercellular adhesion molecule-1 (ICAM-1) levels in brains of Lewy body disorder cases are associated with amounts of Alzheimer's pathology not α-synuclein pathology. Neurobiol Aging 2017; 54:175-186. [PMID: 28390825 DOI: 10.1016/j.neurobiolaging.2017.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/30/2017] [Accepted: 03/07/2017] [Indexed: 12/21/2022]
Abstract
Enhanced inflammation has been associated with Alzheimer's disease (AD) and diseases with Lewy body (LB) pathology, such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB). One issue is whether amyloid and tangle pathology, features of AD, or α-synuclein LB pathology have similar or different effects on brain inflammation. An aim of this study was to examine if certain features of inflammation changed in brains with increasing LB pathology. To assess this, we measured levels of the anti-inflammatory protein CD200 and the pro-inflammatory protein intercellular adhesion molecule-1 (ICAM-1) in cingulate and temporal cortex from a total of 143 cases classified according to the Unified Staging System for LB disorders. Changes in CD200 and ICAM-1 levels did not correlate with LB pathology, but with AD pathology. CD200 negatively correlated with density of neurofibrillary tangles, phosphorylated tau, and amyloid plaque density. ICAM-1 positively correlated with these AD pathology measures. Double immunohistochemistry for phosphorylated α-synuclein and markers for microglia showed limited association of microglia with LB pathology, but microglia strongly associated with amyloid plaques or phosphorylated tau. These results suggest that there are different features of inflammatory pathology in diseases associated with abnormal α-synuclein compared with AD.
Collapse
Affiliation(s)
- Douglas G Walker
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, USA; Banner Sun Health Research Institute, Sun City, AZ, USA.
| | - Lih-Fen Lue
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, USA; Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Tiffany M Tang
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Scottsdale, AZ, USA
| | - John N Caviness
- Department of Neurology, Mayo Clinic College of Medicine, Scottsdale, AZ, USA
| | | | | | - Lucia I Sue
- Banner Sun Health Research Institute, Sun City, AZ, USA
| | | |
Collapse
|
19
|
Del-Bel E, Bortolanza M, Dos-Santos-Pereira M, Bariotto K, Raisman-Vozari R. l-DOPA-induced dyskinesia in Parkinson's disease: Are neuroinflammation and astrocytes key elements? Synapse 2016; 70:479-500. [DOI: 10.1002/syn.21941] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Elaine Del-Bel
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Physiology; FMRP; São Paulo Brazil
- Department of Neurology and Behavioral Neuroscience; FMRP, Campus USP, University of São Paulo; Av. Bandeirantes 13400 Ribeirão Preto SP 14049-900 Brazil
| | - Mariza Bortolanza
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
| | - Maurício Dos-Santos-Pereira
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Physiology; FMRP; São Paulo Brazil
| | - Keila Bariotto
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Neurology and Behavioral Neuroscience; FMRP, Campus USP, University of São Paulo; Av. Bandeirantes 13400 Ribeirão Preto SP 14049-900 Brazil
| | - Rita Raisman-Vozari
- INSERM UMR 1127, CNRS UMR 7225, UPMC; Thérapeutique Expérimentale de la Neurodégénérescence, Hôpital de la Salpetrière-ICM (Institut du cerveau et de la moelle épinière); Paris France
| |
Collapse
|
20
|
Kondo T, Funayama M, Miyake M, Tsukita K, Era T, Osaka H, Ayaki T, Takahashi R, Inoue H. Modeling Alexander disease with patient iPSCs reveals cellular and molecular pathology of astrocytes. Acta Neuropathol Commun 2016; 4:69. [PMID: 27402089 PMCID: PMC4940830 DOI: 10.1186/s40478-016-0337-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/15/2016] [Indexed: 01/28/2023] Open
Abstract
Alexander disease is a fatal neurological illness characterized by white-matter degeneration and formation of Rosenthal fibers, which contain glial fibrillary acidic protein as astrocytic inclusion. Alexander disease is mainly caused by a gene mutation encoding glial fibrillary acidic protein, although the underlying pathomechanism remains unclear. We established induced pluripotent stem cells from Alexander disease patients, and differentiated induced pluripotent stem cells into astrocytes. Alexander disease patient astrocytes exhibited Rosenthal fiber-like structures, a key Alexander disease pathology, and increased inflammatory cytokine release compared to healthy control. These results suggested that Alexander disease astrocytes contribute to leukodystrophy and a variety of symptoms as an inflammatory source in the Alexander disease patient brain. Astrocytes, differentiated from induced pluripotent stem cells of Alexander disease, could be a cellular model for future translational medicine.
Collapse
|