1
|
Wang J, Huo X, Zhou H, Liu H, Li X, Lu N, Sun X. Identification of Autophagy-Related Candidate Genes in the Early Diagnosis of Alzheimer's Disease and Exploration of Potential Molecular Mechanisms. Mol Neurobiol 2024; 61:6584-6598. [PMID: 38329682 DOI: 10.1007/s12035-024-04011-z] [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: 09/21/2023] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
This study aimed to identify autophagy-related candidate genes for the early diagnosis of Alzheimer's disease (AD) and elucidate their potential molecular mechanisms. Differentially expressed genes (DEGs) and phenotype-associated significant module genes were obtained using the "limma" package and weighted gene co-expression network analysis (WGCNA) based on hippocampal tissue datasets from AD patients and control samples. The intersection between the list of autophagy-related genes (ATGs), DEGs, and module genes was further investigated to obtain AD-autophagy-related differential expression genes (ATDEGs). Subsequently, the least absolute shrinkage and selection operator (LASSO) algorithm was utilized to identify hub genes, and a second intersection was performed with important module genes from the protein-protein interaction (PPI) network to obtain co-hub genes. Finally, a diagnostic model was constructed by receiver operating characteristic (ROC) analysis to determine the candidate genes with high diagnostic efficacy in the external validation set. Moreover, immune infiltration analysis was performed on AD patient brain tissues and explore the correlation between candidate genes and immune cells. We further analyzed the expression level of candidate genes in the SH-SY5Y cells with Aβ25-35 (25 µM). Among the 17 identified AD-ATDEGs, ATP6V1E1 stood out with area under the curve (AUC) values of 0.869, 0.817, and 0.714 in the external validation set, underscoring its high diagnostic efficacy in both hippocampal and peripheral blood contexts for AD patients. Meanwhile, ATP6V1E1 expression was positively correlated with effector memory CD4 + T cells, while negatively correlated with natural killer T cells and activated CD4 + T cells. Results from quantitative PCR (qPCR) and immunofluorescence assays indicated a reduction in ATP6V1E1 expression, aligning with our database analysis findings. In summary, ATP6V1E1 as a candidate gene provides a new perspective for the early identification and pathogenesis of AD.
Collapse
Affiliation(s)
- Jian Wang
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China.
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China.
- Hunan Guangxiu Medical Imaging Diagnosis Center, Changsha, China.
| | - Xinhua Huo
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Huiqin Zhou
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Huasheng Liu
- Department of Radiology, Central South University, The Third Xiangya Hospital, Changsha, China
| | - Xiaofeng Li
- Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Na Lu
- Reproductive and Genetic Hospital of CITIC Xiangya, Changsha, China
| | - Xuan Sun
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
2
|
Buchmann Godinho D, da Silva Fiorin F, Schneider Oliveira M, Furian AF, Rechia Fighera M, Freire Royes LF. The immunological influence of physical exercise on TBI-induced pathophysiology: Crosstalk between the spleen, gut, and brain. Neurosci Biobehav Rev 2021; 130:15-30. [PMID: 34400178 DOI: 10.1016/j.neubiorev.2021.08.006] [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] [Received: 05/31/2021] [Revised: 08/04/2021] [Accepted: 08/08/2021] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a non-degenerative and non-congenital insult to the brain and is recognized as a global public health problem, with a high incidence of neurological disorders. Despite the causal relationship not being entirely known, it has been suggested that multiorgan inflammatory response involving the autonomic nervous system and the spleen-gut brain axis dysfunction exacerbate the TBI pathogenesis in the brain. Thus, applying new therapeutic tools, such as physical exercise, have been described in the literature to act on the immune modulation induced by brain injuries. However, there are caveats to consider when interpreting the effects of physical exercise on this neurological injury. Given the above, this review will highlight the main findings of the literature involving peripheral immune responses in TBI-induced neurological damage and how changes in the cellular metabolism of the spleen-gut brain axis elicited by different protocols of physical exercise alter the pathophysiology induced by this neurological injury.
Collapse
Affiliation(s)
- Douglas Buchmann Godinho
- Laboratório de Bioquímica do Exercício, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Fernando da Silva Fiorin
- Programa de Pós-Graduação em Neuroengenharia, Instituto Internacional de Neurociências Edmond e Lily Safra, Instituto Santos Dumont, Macaíba, RN, Brazil
| | - Mauro Schneider Oliveira
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Ana Flavia Furian
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Laboratório de Bioquímica do Exercício, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Departamento de Clínica Médica e Pediatria, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | |
Collapse
|
3
|
Jiang S, Gao H, Yong Y, Zhang H, Li P, Li Y, Luo Q, Yang X. Effect of Pramipexole on Inflammatory Response in Central Nervous System of Parkinson's Disease Rat Model. Arch Med Res 2021; 53:37-43. [DOI: 10.1016/j.arcmed.2021.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 05/06/2021] [Accepted: 06/10/2021] [Indexed: 11/02/2022]
|
4
|
Zaman V, Shields DC, Shams R, Drasites KP, Matzelle D, Haque A, Banik NL. Cellular and molecular pathophysiology in the progression of Parkinson's disease. Metab Brain Dis 2021; 36:815-827. [PMID: 33599945 PMCID: PMC8170715 DOI: 10.1007/s11011-021-00689-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder etiologically linked to the loss of substantia nigra (SN) dopaminergic neurons in the mid-brain. The etiopathology of sporadic PD is still unclear; however, the interaction of extrinsic and intrinsic factors may play a critical role in the onset and progression of the disease. Studies in animal models and human post-mortem tissue have identified distinct cellular and molecular changes in the diseased brain, suggesting complex interactions between different glial cell types and various molecular pathways. Small changes in the expression of specific genes in a single pathway or cell type possibly influence others at the cellular and system levels. These molecular and cellular signatures like neuroinflammation, oxidative stress, and autophagy have been observed in PD patients' brain tissue. While the etiopathology of PD is still poorly understood, the interplay between glial cells and molecular events may play a crucial role in disease onset and progression.
Collapse
Affiliation(s)
- Vandana Zaman
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
| | - Donald C Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
| | - Ramsha Shams
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Kelsey P Drasites
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Denise Matzelle
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| | - Narendra L Banik
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| |
Collapse
|
5
|
Olson KE, Namminga KL, Lu Y, Thurston MJ, Schwab AD, de Picciotto S, Tse SW, Walker W, Iacovelli J, Small C, Wipke BT, Mosley RL, Huang E, Gendelman HE. Granulocyte-macrophage colony-stimulating factor mRNA and Neuroprotective Immunity in Parkinson's disease. Biomaterials 2021; 272:120786. [PMID: 33839625 PMCID: PMC8382980 DOI: 10.1016/j.biomaterials.2021.120786] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/15/2021] [Accepted: 03/24/2021] [Indexed: 02/06/2023]
Abstract
Restoring numbers and function of regulatory T cells (Tregs) is a novel therapeutic strategy for neurodegenerative disorders. Whether Treg function is boosted by adoptive cell transfer, pharmaceuticals, or immune modulators, the final result is a robust anti-inflammatory and neuronal sparing response. Herein, a newly developed lipid nanoparticle (LNP) containing mRNA encoding granulocyte-macrophage colony-stimulating factor (Gm-csf mRNA) was developed to peripherally induce Tregs and used for treatment in preclinical Parkinson's disease (PD) models. Administration of Gm-csf mRNA to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice and rats overexpressing alpha-synuclein produced dose-dependent increases in plasma GM-CSF levels and peripheral CD4+CD25+FoxP3+ Treg populations. This upregulation paralleled nigrostriatal neuroprotection, upregulated immunosuppression-associated mRNAs that led to the detection of a treatment-induced CD4+ T cell population, and decreased reactive microgliosis. The current findings strengthen prior works utilizing immune modulation by harnessing Gm-csf mRNA to augment adaptive immune function by employing a new delivery platform to treat PD and potentially other neurodegenerative disorders.
Collapse
Affiliation(s)
- Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Krista L Namminga
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mackenzie J Thurston
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Aaron D Schwab
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | | | | | | | | | | | | | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Eric Huang
- Moderna, Inc., Cambridge, MA, 02139, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
6
|
Wang Y, Gao H, Jiang S, Luo Q, Han X, Xiong Y, Xu Z, Qiao R, Yang X. Principal component analysis of routine blood test results with Parkinson's disease: A case-control study. Exp Gerontol 2020; 144:111188. [PMID: 33279667 DOI: 10.1016/j.exger.2020.111188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/27/2020] [Accepted: 11/24/2020] [Indexed: 11/26/2022]
Abstract
This study aimed to explore the association of routine blood test values and blood cell ratios with the risk or severity of Parkinson's disease (PD). The medical records of 453 PD patients and 436 controls were retrospectively reviewed. The severity of PD was quantified by the modified Hoehn-Yahr (HY) scale. We performed principal component analysis (PCA) of significant values/ratios and used logistic regression analysis to explore the relationship between principal components (PCs) and the risk of PD. Spearman correlation and ordinal logistic regression analyses were performed to explore the relationship between indicators and the severity of PD. The PCA generated 9 PCs, which contributed to 90.86% of the total variance. Logistic regression analysis revealed positive associations of PC2 (a measure monocyte ratios) and PC6 (a measure of platelet ratios and volume) and negative associations of PC1 (a comprehensive measure of lymphocyte, eosinophil, neutrophil, and red blood cell values), PC4 (a measure of red blood cell values), and PC7 (a measure of red blood cell values and platelet volume) with the risk of PD. However, we observed no associations of variables with the severity of PD. In conclusion, PCA reduced the dimensionality of the data. Peripheral blood disorders may be associated with PD.
Collapse
Affiliation(s)
- Yuling Wang
- Medicine VIP, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushanlu Road, Urumqi 830011, China; Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, No. 38, Nanhudonglu Road, Urumqi 830054, China
| | - Hua Gao
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, No. 38, Nanhudonglu Road, Urumqi 830054, China; Department of Neurology, Fifth Affiliated Hospital of Xinjiang Medical University, No. 118, Henanxilu Road, Urumqi 830000, China
| | - Sen Jiang
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, No. 38, Nanhudonglu Road, Urumqi 830054, China
| | - Qin Luo
- Department of Medicine, Tumor Hospital Affiliated of Xinjiang Medical University, No. 789, Suzhoudongjie Road, Urumqi 830000, China
| | - Xuejie Han
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, No. 38, Nanhudonglu Road, Urumqi 830054, China
| | - Yi Xiong
- Department of Neurology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushanlu Road, Urumqi 830011, China
| | - Zeheng Xu
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, No. 38, Nanhudonglu Road, Urumqi 830054, China
| | - Rui Qiao
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, No. 38, Nanhudonglu Road, Urumqi 830054, China
| | - Xinling Yang
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, No. 38, Nanhudonglu Road, Urumqi 830054, China.
| |
Collapse
|
7
|
Stojić-Vukanić Z, Hadžibegović S, Nicole O, Nacka-Aleksić M, Leštarević S, Leposavić G. CD8+ T Cell-Mediated Mechanisms Contribute to the Progression of Neurocognitive Impairment in Both Multiple Sclerosis and Alzheimer's Disease? Front Immunol 2020; 11:566225. [PMID: 33329528 PMCID: PMC7710704 DOI: 10.3389/fimmu.2020.566225] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Neurocognitive impairment (NCI) is one of the most relevant clinical manifestations of multiple sclerosis (MS). The profile of NCI and the structural and functional changes in the brain structures relevant for cognition in MS share some similarities to those in Alzheimer's disease (AD), the most common cause of neurocognitive disorders. Additionally, despite clear etiopathological differences between MS and AD, an accumulation of effector/memory CD8+ T cells and CD8+ tissue-resident memory T (Trm) cells in cognitively relevant brain structures of MS/AD patients, and higher frequency of effector/memory CD8+ T cells re-expressing CD45RA (TEMRA) with high capacity to secrete cytotoxic molecules and proinflammatory cytokines in their blood, were found. Thus, an active pathogenetic role of CD8+ T cells in the progression of MS and AD may be assumed. In this mini-review, findings supporting the putative role of CD8+ T cells in the pathogenesis of MS and AD are displayed, and putative mechanisms underlying their pathogenetic action are discussed. A special effort was made to identify the gaps in the current knowledge about the role of CD8+ T cells in the development of NCI to "catalyze" translational research leading to new feasible therapeutic interventions.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Senka Hadžibegović
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Olivier Nicole
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Mirjana Nacka-Aleksić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Sanja Leštarević
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
8
|
Motataianu A, Barcutean L, Balasa R. Neuroimmunity in amyotrophic lateral sclerosis: focus on microglia. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:159-166. [PMID: 31903792 DOI: 10.1080/21678421.2019.1708949] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS), an incurable, devastating condition of the central nervous system, is characterized by selective destruction of motoneurons with an important contribution of innate and adaptative immunity. Microglia and immune cells are key players in neuroinflammation and active participants in ALS pathogenesis. Recent experiments in animal models have shown that microglia display both neuroprotective and neurotoxic properties, depending on the stage of disease progression and cytokine secretion. A better knowledge of the interactions between T cells and microglia in the immunopathogenesis of ALS is desirable for the development of ALS therapeutic strategies.
Collapse
Affiliation(s)
- Anca Motataianu
- Neurology Department, University of Medicine, Pharmacy, Science and Technology "George Emil Palade", Targu Mures, Romania
| | - Laura Barcutean
- Neurology Department, University of Medicine, Pharmacy, Science and Technology "George Emil Palade", Targu Mures, Romania
| | - Rodica Balasa
- Neurology Department, University of Medicine, Pharmacy, Science and Technology "George Emil Palade", Targu Mures, Romania
| |
Collapse
|
9
|
Ciccocioppo F, Bologna G, Ercolino E, Pierdomenico L, Simeone P, Lanuti P, Pieragostino D, Del Boccio P, Marchisio M, Miscia S. Neurodegenerative diseases as proteinopathies-driven immune disorders. Neural Regen Res 2020; 15:850-856. [PMID: 31719246 PMCID: PMC6990794 DOI: 10.4103/1673-5374.268971] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In the pathophysiology of neurodegenerative disorders, the role of misfolded protein deposition leading to neurodegeneration has been primarily discussed. In the last decade, however, it has been proposed a parallel involvement of innate immune activation, chronic inflammation and adaptive immunity in the neurodegeneration mechanisms triggered by proteinopathies. New insights in the neurodegenerative field strongly suggest a role for the immune system in the pathophysiology of neurodegenerative disorders. Therefore, the hypothesis underlining the modulation of the innate and the adaptive immune system in the events linked to brain deposition of misfolded proteins could open new perspectives in the setting of specific immunotherapeutic strategies for the treatment of neurodegenerative diseases. Therefore, we have reviewed the pathogenic hypothesis in neurodegenerative pathologies, underling the links between the deposition of misfolded protein mechanisms and the immune activation.
Collapse
Affiliation(s)
- Fausta Ciccocioppo
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Giuseppina Bologna
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Eva Ercolino
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Laura Pierdomenico
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Damiana Pieragostino
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT); Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Piero Del Boccio
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT); Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Marco Marchisio
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Sebastiano Miscia
- Department of Medicine and Aging Science; Centre on Aging Sciences and Translational Medicine (Ce.S.I-MeT), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
10
|
Ciccocioppo F, Lanuti P, Pierdomenico L, Simeone P, Bologna G, Ercolino E, Buttari F, Fantozzi R, Thomas A, Onofrj M, Centonze D, Miscia S, Marchisio M. The Characterization of Regulatory T-Cell Profiles in Alzheimer's Disease and Multiple Sclerosis. Sci Rep 2019; 9:8788. [PMID: 31217537 PMCID: PMC6584558 DOI: 10.1038/s41598-019-45433-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 06/04/2019] [Indexed: 01/01/2023] Open
Abstract
Regulatory T Cells (Tregs) are a T-lymphocyte subset involved in the maintenance of immune peripheral tolerance. Despite evidence of the adaptive immune system's role in Alzheimer's Disease (AD), the involvement of Tregs is still not clear. We focused on the Flow-Cytometry analysis of the Treg frequencies and phenotypes in the AD. The aim of the study is to analyse similarities and differences in Tregs profile between Alzheimer's Disease and Multiple Sclerosis. Regulatory T Cells (CD4+/CD25high/CD127low-neg) were identified using an innovative Flow Cytometry method and subtyped as Resting (analysed CD45RApos/CD25dim), Activated (CD45RAneg/CD25bright) and Secreting (CD45RAneg/CD25dim) cells. Our data demonstrate a significant decrease in the total and Resting Tregs in AD patients when compared to healthy subjects. The percentage of the results of the Resting Tregs were also reduced in MS patients together with a parallel frequency increase of Activated Tregs. Our data suggest that altered Treg phenotypes observed in both diseases could play a role in the impairment of the Treg-mediated immunological tolerance, recalling a possible link between the two pathologies. Given that this study was conducted on a restricted population, if confirmed by a further and enlarged study, the implications of the autoimmune mechanisms in AD pathophysiology could open new immunotherapeutic perspectives based on Treg modulation.
Collapse
Affiliation(s)
- Fausta Ciccocioppo
- Department of Medicine and Aging Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Laura Pierdomenico
- Department of Medicine and Aging Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Eva Ercolino
- Department of Medicine and Aging Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli, (IS), Italy
| | | | - Astrid Thomas
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Marco Onofrj
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Diego Centonze
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy.
- Unit of Neurology, IRCCS Neuromed, Pozzilli, (IS), Italy.
| | - Sebastiano Miscia
- Department of Medicine and Aging Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| | - Marco Marchisio
- Department of Medicine and Aging Sciences, University "G. D'Annunzio", Chieti-Pescara, Italy
- Center on Aging Science and Translational Medicine (Ce.S.I.-Me.T.), University "G. D'Annunzio", Chieti-Pescara, Italy
| |
Collapse
|
11
|
Kiyota T, Machhi J, Lu Y, Dyavarshetty B, Nemati M, Yokoyama I, Mosley RL, Gendelman HE. Granulocyte-macrophage colony-stimulating factor neuroprotective activities in Alzheimer's disease mice. J Neuroimmunol 2018; 319:80-92. [PMID: 29573847 PMCID: PMC5916331 DOI: 10.1016/j.jneuroim.2018.03.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/27/2018] [Accepted: 03/15/2018] [Indexed: 12/11/2022]
Abstract
We investigated the effects of granulocyte-macrophage colony stimulating factor (GM-CSF) on behavioral and pathological outcomes in Alzheimer's disease (AD) and non-transgenic mice. GM-CSF treatment in AD mice reduced brain amyloidosis, increased plasma Aβ, and rescued cognitive impairment with increased hippocampal expression of calbindin and synaptophysin and increased levels of doublecortin-positive cells in the dentate gyrus. These data extend GM-CSF pleiotropic neuroprotection mechanisms in AD and include regulatory T cell-mediated immunomodulation of microglial function, Aβ clearance, maintenance of synaptic integrity, and induction of neurogenesis. Together these data support further development of GM-CSF as a neuroprotective agent for AD.
Collapse
Affiliation(s)
- Tomomi Kiyota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bhagyalaxmi Dyavarshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maryam Nemati
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Izumi Yokoyama
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - R L Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
12
|
Olson KE, Bade AN, Schutt CR, Dong J, Shandler SJ, Boska MD, Mosley RL, Gendelman HE, Liu Y. Manganese-Enhanced Magnetic Resonance Imaging for Detection of Vasoactive Intestinal Peptide Receptor 2 Agonist Therapy in a Model of Parkinson's Disease. Neurotherapeutics 2016; 13:635-46. [PMID: 27329163 PMCID: PMC4965412 DOI: 10.1007/s13311-016-0449-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuroprotective immunity is defined by transformation of T-cell polarity for therapeutic gain. For neurodegenerative disorders and specifically for Parkinson's disease (PD), granulocyte-macrophage colony stimulating factor or vasoactive intestinal peptide receptor 2 (VIPR2) agonists elicit robust anti-inflammatory microglial responses leading to neuronal sparing in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. While neurotherapeutic potential was demonstrated for PD, there remain inherent limitations in translating these inventions from the laboratory to patients. One obstacle in translating such novel neurotherapeutics centers on the availability of suitable noninvasive methods to track disease progression and therapeutic efficacy. To this end, we developed manganese-enhanced magnetic resonance imaging (MEMRI) assays as a way to track a linkage between glial activation and VIPR2 agonist (LBT-3627)-induced neuroprotective immunity for MPTP-induced nigrostriatal degeneration. Notably, LBT-3627-treated, MPTP-intoxicated mice show reduced MEMRI brain signal intensities. These changes paralleled reduced astrogliosis and resulted in sparing of nigral tyrosine hydroxylase neurons. Most importantly, the data suggest that MEMRI can be developed as a biomarker tool to monitor neurotherapeutic responses that are relevant to common neurodegenerative disorders used to improve disease outcomes.
Collapse
Affiliation(s)
- Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aditya N Bade
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Charles R Schutt
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jingdong Dong
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Michael D Boska
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Yutong Liu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
13
|
Selective VIP Receptor Agonists Facilitate Immune Transformation for Dopaminergic Neuroprotection in MPTP-Intoxicated Mice. J Neurosci 2016; 35:16463-78. [PMID: 26674871 DOI: 10.1523/jneurosci.2131-15.2015] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
UNLABELLED Vasoactive intestinal peptide (VIP) mediates a broad range of biological responses by activating two related receptors, VIP receptor 1 and 2 (VIPR1 and VIPR2). Although the use of native VIP facilitates neuroprotection, clinical application of the hormone is limited due to VIP's rapid metabolism and inability to distinguish between VIPR1 and VIPR2 receptors. In addition, activation of both receptors by therapeutics may increase adverse secondary toxicities. Therefore, we developed metabolically stable and receptor-selective agonists for VIPR1 and VIPR2 to improve pharmacokinetic and pharmacodynamic therapeutic end points. Selective agonists were investigated for their abilities to protect mice against MPTP-induced neurodegeneration used to model Parkinson's disease (PD). Survival of tyrosine hydroxylase neurons in the substantia nigra was determined by stereological tests after MPTP intoxication in mice pretreated with either VIPR1 or VIPR2 agonist or after adoptive transfer of splenic cell populations from agonist-treated mice administered to MPTP-intoxicated animals. Treatment with VIPR2 agonist or splenocytes from agonist-treated mice resulted in increased neuronal sparing. Immunohistochemical tests showed that agonist-treated mice displayed reductions in microglial responses, with the most pronounced effects in VIPR2 agonist-treated, MPTP-intoxicated mice. In parallel studies, we observed reductions in proinflammatory cytokine release that included IL-17A, IL-6, and IFN-γ and increases in GM-CSF transcripts in CD4(+) T cells recovered from VIPR2 agonist-treated animals. Moreover, a phenotypic shift of effector to regulatory T cells was observed. These results support the use of VIPR2-selective agonists as neuroprotective agents for PD treatment. SIGNIFICANCE STATEMENT Vasoactive intestinal peptide receptor 2 can elicit immune transformation in a model of Parkinson's disease (PD). Such immunomodulatory capabilities can lead to neuroprotection by attenuating microglial activation and by slowing degradation of neuronal cell bodies and termini in MPTP-intoxicated mice. The protective mechanism arises from altering a Th1/Th2 immune cytokine response into an anti-inflammatory and neuronal sparing profile. These results are directly applicable for the development of novel PD therapies.
Collapse
|
14
|
Li M, Lin YP, Chen JL, Li H, Jiang RC, Zhang JN. Role of regulatory T cell in clinical outcome of traumatic brain injury. Chin Med J (Engl) 2015; 128:1072-8. [PMID: 25881602 PMCID: PMC4832948 DOI: 10.4103/0366-6999.155094] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a life-threatening disease worldwide. Regulatory T cells (Treg cells) were involved in the immunological system in central nervous system. It is defined as a subpopulation of CD4 + cells that express CD25 and transcription factor forkhead box P3. The level of circulating Treg cells increases in a variety of pathologic conditions. The purpose of this study was to uncover the role of circulating Treg cells in TBI. METHODS A clinical study was conducted in two neurosurgical intensive care units of Tianjin Medical University General Hospital and Second Hospital of Tianjin Medical University (Tianjin, China). Forty patients and 30 healthy controls were recruited from August 2013 to November 2013. Circulating Treg cells was detected on the follow-up period of 1, 4, 7, 14, and 21 days after TBI. Blood sample (1 ml) was withdrawn in the morning and processed within 2 h. RESULTS There was no significant difference in the level of circulating Treg cells between TBI patients and normal controls during follow-up. TBI patients exhibited higher circulating Treg level than normal controls on the 1 st day after TBI. Treg level was decreased on the 4 th day, climbed up on the 7 th day and peaked on 14 th day after TBI. Treg cells declined to the normal level on 21 th day after TBI. The level of circulating Treg cells was significantly higher in survival TBI patients when compared to nonsurvival TBI patients. TBI patients with improved conditions exhibited significantly higher circulating Treg level when compared to those with deteriorated conditions. The circulating Treg level was correlated with neurologic recovery after TBI. A better neural recovery and lower hospital mortality were found in TBI patients with circulating Treg cells more than 4.91% in total CD4 + mononuclear cells as compared to those with circulating Treg cells less than 4.91% in total CD4 + mononuclear cells in the first 14 days. CONCLUSIONS The level of circulating Treg cells is positively correlated with clinical outcome of TBI. The level of Treg cells predicts the progress for TBI patients and may be a target in TBI treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Jian-Ning Zhang
- Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
15
|
Mongillo P, Bertotto D, Pitteri E, Stefani A, Marinelli L, Gabai G. Peripheral leukocyte populations and oxidative stress biomarkers in aged dogs showing impaired cognitive abilities. AGE (DORDRECHT, NETHERLANDS) 2015; 37:9778. [PMID: 25905581 PMCID: PMC4408299 DOI: 10.1007/s11357-015-9778-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/14/2015] [Indexed: 05/02/2023]
Abstract
In the present study, the peripheral blood leukocyte phenotypes, lymphocyte subset populations, and oxidative stress parameters were studied in cognitively characterized adult and aged dogs, in order to assess possible relationships between age, cognitive decline, and the immune status. Adult (N = 16, 2-7 years old) and aged (N = 29, older than 8 years) dogs underwent two testing procedures, for the assessment of spatial reversal learning and selective social attention abilities, which were shown to be sensitive to aging in pet dogs. Based on age and performance in cognitive testing, dogs were classified as adult not cognitively impaired (ADNI, N = 12), aged not cognitively impaired (AGNI, N = 19) and aged cognitively impaired (AGCI, N = 10). Immunological and oxidative stress parameters were compared across groups with the Kruskal-Wallis test. AGCI dogs displayed lower absolute CD4 cell count (p < 0.05) than ADNI and higher monocyte absolute count and percentage (p < 0.05) than AGNI whereas these parameters were not different between AGNI and ADNI. AGNI dogs had higher CD8 cell percentage than ADNI (p < 0.05). Both AGNI and AGCI dogs showed lower CD4/CD8 and CD21 count and percentage and higher neutrophil/lymphocyte and CD3/CD21 ratios (p < 0.05). None of the oxidative parameters showed any statistically significant difference among groups. These observations suggest that alterations in peripheral leukocyte populations may reflect age-related changes occurring within the central nervous system and disclose interesting perspectives for the dog as a model for studying the functional relationship between the nervous and immune systems during aging.
Collapse
Affiliation(s)
- Paolo Mongillo
- />Department of Comparative Biomedicine and Food Science, Università degli Studi di Padova, Viale dell’Università 16, Agripolis, 35020 Legnaro, PD Italy
| | - Daniela Bertotto
- />Department of Comparative Biomedicine and Food Science, Università degli Studi di Padova, Viale dell’Università 16, Agripolis, 35020 Legnaro, PD Italy
| | - Elisa Pitteri
- />Department of Comparative Biomedicine and Food Science, Università degli Studi di Padova, Viale dell’Università 16, Agripolis, 35020 Legnaro, PD Italy
| | - Annalisa Stefani
- />Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, PD Italy
| | - Lieta Marinelli
- />Department of Comparative Biomedicine and Food Science, Università degli Studi di Padova, Viale dell’Università 16, Agripolis, 35020 Legnaro, PD Italy
| | - Gianfranco Gabai
- />Department of Comparative Biomedicine and Food Science, Università degli Studi di Padova, Viale dell’Università 16, Agripolis, 35020 Legnaro, PD Italy
| |
Collapse
|
16
|
Anderson KM, Olson KE, Estes KA, Flanagan K, Gendelman HE, Mosley RL. Dual destructive and protective roles of adaptive immunity in neurodegenerative disorders. Transl Neurodegener 2014; 3:25. [PMID: 25671101 PMCID: PMC4323229 DOI: 10.1186/2047-9158-3-25] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/28/2014] [Indexed: 12/14/2022] Open
Abstract
Inappropriate T cell responses in the central nervous system (CNS) affect the pathogenesis of a broad range of neuroinflammatory and neurodegenerative disorders that include, but are not limited to, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer’s disease and Parkinson’s disease. On the one hand immune responses can exacerbate neurotoxic responses; while on the other hand, they can lead to neuroprotective outcomes. The temporal and spatial mechanisms by which these immune responses occur and are regulated in the setting of active disease have gained significant recent attention. Spatially, immune responses that affect neurodegeneration may occur within or outside the CNS. Migration of antigen-specific CD4+ T cells from the periphery to the CNS and consequent immune cell interactions with resident glial cells affect neuroinflammation and neuronal survival. The destructive or protective mechanisms of these interactions are linked to the relative numerical and functional dominance of effector or regulatory T cells. Temporally, immune responses at disease onset or during progression may exhibit a differential balance of immune responses in the periphery and within the CNS. Immune responses with predominate T cell subtypes may differentially manifest migratory, regulatory and effector functions when triggered by endogenous misfolded and aggregated proteins and cell-specific stimuli. The final result is altered glial and neuronal behaviors that influence the disease course. Thus, discovery of neurodestructive and neuroprotective immune mechanisms will permit potential new therapeutic pathways that affect neuronal survival and slow disease progression.
Collapse
Affiliation(s)
- Kristi M Anderson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine A Estes
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Ken Flanagan
- Prothena Biosciences, South San Francisco, 650 Gateway Boulevard, CA 94080 USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
17
|
Biochemical and immunological aspects of protein aggregation in neurodegenerative diseases. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2014. [DOI: 10.1007/s13738-014-0491-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
18
|
Thymic TFH cells involved in the pathogenesis of myasthenia gravis with thymoma. Exp Neurol 2014; 254:200-5. [DOI: 10.1016/j.expneurol.2014.01.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 01/23/2014] [Accepted: 01/29/2014] [Indexed: 01/26/2023]
|
19
|
Rodrigues MCO, Sanberg PR, Cruz LE, Garbuzova-Davis S. The innate and adaptive immunological aspects in neurodegenerative diseases. J Neuroimmunol 2013; 269:1-8. [PMID: 24161471 DOI: 10.1016/j.jneuroim.2013.09.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 08/03/2013] [Accepted: 09/30/2013] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases affect a considerable percentage of the elderly population. New therapeutic approaches are warranted, aiming to at least delay and possibly reverse disease progression. Strategies to elaborate such approaches require knowledge of specific immune system involvement in disease pathogenesis. In this review, innate and adaptive immunological aspects of neurodegenerative disorders, in particular Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis (ALS), are discussed. Initiating disease factors, as well as common mechanistic pathways, are detailed and potential immunological therapeutic targets are identified.
Collapse
Affiliation(s)
- Maria C O Rodrigues
- Division of Clinical Immunology, Department of Internal Medicine, Ribeirão Preto School of Medicine, University of Sao Paulo, Brazil
| | - Paul R Sanberg
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL 33612, United States; Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL 33612, United States; Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, FL 33612, United States; Department of Psychiatry, University of South Florida, Morsani College of Medicine, Tampa, FL 33612, United States
| | - Luis Eduardo Cruz
- Cryopraxis, Cell Praxis, BioRio, Polo de Biotechnologia do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL 33612, United States; Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL 33612, United States; Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, FL 33612, United States; Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL 33612, United States.
| |
Collapse
|
20
|
Sarazin M, Dorothée G, de Souza LC, Aucouturier P. Immunotherapy in Alzheimer's disease: do we have all the pieces of the puzzle? Biol Psychiatry 2013; 74:329-32. [PMID: 23683656 DOI: 10.1016/j.biopsych.2013.04.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/26/2013] [Accepted: 04/12/2013] [Indexed: 11/28/2022]
Abstract
Results of Phase III studies involving a large number of Alzheimer's disease (AD) patients treated by passive immunotherapy with humanized anti-amyloid β monoclonal antibodies have recently been released. These approaches failed to show a significant clinical benefit in patients with mild to moderate AD. The most considered explanation is that the patients have been treated too late. Whereas targeting patients at asymptomatic stages of the disease is a critical step in the goal of improving the efficacy of such antibody-based strategies, several other important factors should be considered in the development and clinical evaluation of anti-amyloid β immunotherapies, including the as yet poorly understood relationship of AD with the immune system and the importance of cerebral amyloid angiopathy. Better understanding the role of immune responses in AD and their impact on immunotherapy appears essential in the design of alternative or combinatorial immunotherapy approaches in AD, which may imply effectors other than antibodies and even additional antigenic targets.
Collapse
Affiliation(s)
- Marie Sarazin
- Université Paris Descartes, Sorbonne Paris Cité, France.
| | | | | | | |
Collapse
|
21
|
Bisicchia E, Chiurchiù V, Viscomi MT, Latini L, Fezza F, Battistini L, Maccarrone M, Molinari M. Activation of type-2 cannabinoid receptor inhibits neuroprotective and antiinflammatory actions of glucocorticoid receptor α: when one is better than two. Cell Mol Life Sci 2013; 70:2191-204. [PMID: 23296125 PMCID: PMC11113882 DOI: 10.1007/s00018-012-1253-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/21/2012] [Accepted: 12/20/2012] [Indexed: 01/19/2023]
Abstract
Endocannabinoids (eCBs) and glucocorticoids (GCs) are two distinct classes of signaling lipids that exert both neuroprotective and immunosuppressive effects; however, the possibility of an actual interaction of their receptors [i.e., type-2 cannabinoid (CB2) and glucocorticoid receptor α (GRα), respectively] remains unexplored. Here, we demonstrate that the concomitant activation of CB2 and GRα abolishes the neuroprotective effects induced by each receptor on central neurons and on glial cells in animal models of remote cell death. We also show that the ability of eCBs and GCs, used individually, to inhibit tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) production from activated human T lymphocytes is lost when CB2 and GRα are activated simultaneously. In addition, signal transduction pathways triggered by concomitant activation of both receptors led to increased levels of GRβ, heat-shock proteins-70 and -90, and p-JNK, as well as to reduced levels of p-STAT6. These effects were reversed only by selectively antagonizing CB2, but not GRα. Overall, our study demonstrates for the first time the existence of a CB2-driven negative cross-talk between eCB and GC signaling in both rats and humans, thus paving the way to the possible therapeutic exploitation of CB2 as a new target for chronic inflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Elisa Bisicchia
- Department of Biomedical Sciences, University of Teramo, Piazza A. Moro 45, 64100 Teramo, Italy
- I.R.C.C.S. Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Valerio Chiurchiù
- Department of Biomedical Sciences, University of Teramo, Piazza A. Moro 45, 64100 Teramo, Italy
- I.R.C.C.S. Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Maria Teresa Viscomi
- I.R.C.C.S. Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Laura Latini
- I.R.C.C.S. Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Filomena Fezza
- I.R.C.C.S. Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Luca Battistini
- I.R.C.C.S. Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Mauro Maccarrone
- I.R.C.C.S. Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
- Center of Integrated Research, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Marco Molinari
- I.R.C.C.S. Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
- Fondazione S. Lucia, Via Ardeatina 306, 00179 Rome, Italy
| |
Collapse
|
22
|
Eosinophil-derived neurotoxin is elevated in patients with amyotrophic lateral sclerosis. Mediators Inflamm 2013; 2013:421389. [PMID: 23533305 PMCID: PMC3590756 DOI: 10.1155/2013/421389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 12/24/2012] [Accepted: 01/10/2013] [Indexed: 12/11/2022] Open
Abstract
Background and Objectives. Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by loss of motor neurons in the brainstem, motor cortex, and spinal cord. Oxidative stress and neuroinflammation have been implicated in the pathophysiology of ALS. Members of the family of damage-associated molecular patterns, including reactive oxygen species, high-mobility group box 1, and eosinophil-derived neurotoxin (EDN), may participate in pathological conditions. In this study, we aim to discover new biomarker for detecting ALS. Materials and Methods. We examined 44 patients with ALS, 41 patients with Alzheimer's disease, 41 patients with Parkinson's disease, and 44 healthy controls. The concentration of serum EDN was measured using an enzyme-linked immunosorbent assay. Results. EDN levels were significantly increased 2.17-fold in the serum of patients with ALS as compared with healthy controls (P < 0.05). No correlation between the levels of serum EDN and various clinical parameters of ALS was found. Moreover, the levels of serum EDN in patients with Parkinson's disease and Alzheimer's disease and healthy controls were similar. Conclusion. A higher level of serum EDN was found specifically in patients with ALS, indicating that EDN may participate in the pathophysiology of ALS.
Collapse
|
23
|
Dobbs RJ, Charlett A, Dobbs SM, Weller C, A Ibrahim MA, Iguodala O, Smee C, Plant JM, Lawson AJ, Taylor D, Bjarnason I. Leukocyte-subset counts in idiopathic parkinsonism provide clues to a pathogenic pathway involving small intestinal bacterial overgrowth. A surveillance study. Gut Pathog 2012; 4:12. [PMID: 23083400 PMCID: PMC3500215 DOI: 10.1186/1757-4749-4-12] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 09/25/2012] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED BACKGROUND Following Helicobacter pylori eradication in idiopathic parkinsonism (IP), hypokinesia improved but flexor-rigidity increased. Small intestinal bacterial-overgrowth (SIBO) is a candidate driver of the rigidity: hydrogen-breath-test-positivity is common in IP and case histories suggest that Helicobacter keeps SIBO at bay. METHODS In a surveillance study, we explore relationships of IP-facets to peripheral immune/inflammatory-activation, in light of presence/absence of Helicobacter infection (urea-breath- and/or stool-antigen-test: positivity confirmed by gastric-biopsy) and hydrogen-breath-test status for SIBO (positivity: >20 ppm increment, 2 consecutive 15-min readings, within 2h of 25G lactulose). We question whether any relationships found between facets and blood leukocyte subset counts stand in patients free from anti-parkinsonian drugs, and are robust enough to defy fluctuations in performance consequent on short t½ therapy. RESULTS Of 51 IP-probands, 36 had current or past Helicobacter infection on entry, 25 having undergone successful eradication (median 3.4 years before). Thirty-four were hydrogen-breath-test-positive initially, 42 at sometime (343 tests) during surveillance (2.8 years). Hydrogen-breath-test-positivity was associated inversely with Helicobacter-positivity (OR 0.20 (95% CI 0.04, 0.99), p<0.05).In 38 patients (untreated (17) or on stable long-t½ IP-medication), the higher the natural-killer count, the shorter stride, slower gait and greater flexor-rigidity (by mean 49 (14, 85) mm, 54 (3, 104) mm.s-1, 89 (2, 177) Nm.10-3, per 100 cells.μl-1 increment, p=0.007, 0.04 & 0.04 respectively, adjusted for patient characteristics). T-helper count was inversely associated with flexor-rigidity before (p=0.01) and after adjustment for natural-killer count (-36(-63, -10) Nm.10-3 per 100 cells.μl-1, p=0.007). Neutrophil count was inversely associated with tremor (visual analogue scale, p=0.01). Effect-sizes were independent of IP-medication, and not masked by including 13 patients receiving levodopa (except natural-killer count on flexor-rigidity). Cellular associations held after allowing for potentially confounding effect of hydrogen-breath-test or Helicobacter status. Moreover, additional reduction in stride and speed (68 (24, 112) mm & 103 (38, 168) mm.s-1, each p=0.002) was seen with Helicobacter-positivity. Hydrogen-breath-test-positivity, itself, was associated with higher natural-killer and T-helper counts, lower neutrophils (p=0.005, 0.02 & 0.008). CONCLUSION We propose a rigidity-associated subordinate pathway, flagged by a higher natural-killer count, tempered by a higher T-helper, against which Helicobacter protects by keeping SIBO at bay.
Collapse
Affiliation(s)
- R John Dobbs
- Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
- The Maudsley Hospital, Denmark Hill, London, SE5 8AZ, UK
- Gastroenterology, King’s College Hospital, Bessemer Rd, London, SE5 9PJ, UK
| | - André Charlett
- Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
- Statistics Unit, Health Protection Agency, 61 Colindale Avenue, London, NW9 5EQ, UK
| | - Sylvia M Dobbs
- Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
- The Maudsley Hospital, Denmark Hill, London, SE5 8AZ, UK
- Gastroenterology, King’s College Hospital, Bessemer Rd, London, SE5 9PJ, UK
| | - Clive Weller
- Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | | | - Owens Iguodala
- The Maudsley Hospital, Denmark Hill, London, SE5 8AZ, UK
| | - Cori Smee
- The Maudsley Hospital, Denmark Hill, London, SE5 8AZ, UK
| | | | - Andrew J Lawson
- Laboratory of Gastrointestinal Pathogens, Health Protection Agency, London, NW9 5EQ, UK
| | - David Taylor
- Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
- The Maudsley Hospital, Denmark Hill, London, SE5 8AZ, UK
| | - Ingvar Bjarnason
- Gastroenterology, King’s College Hospital, Bessemer Rd, London, SE5 9PJ, UK
| |
Collapse
|
24
|
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder. Currently, no curative treatments or treatments that interdict disease progression are available. Over the past decade, immunization strategies were developed in our laboratories to combat disease progression. These strategies were developed in laboratory and animal models of human disease. Induction of humoral immune responses can be elicited against misfolded protein aggregates. Robust cell-mediated immunity against nitrated misfolded protein(s) accelerates disease progression through effector T cell responses that facilitate neuronal death. We propose that shifting the balance between effector and regulatory T cell activity can attenuate neurotoxic inflammatory events. We now summarize our works that support immune regulation in PD with the singular goal of restoring homeostatic glial responses. New methods to optimize immunization schemes and measure their clinical efficacy are discussed.
Collapse
Affiliation(s)
- Duy Ha
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | |
Collapse
|
25
|
Hetz RA, Bedi SS, Olson S, Olsen A, Cox CS. Progenitor cells: therapeutic targets after traumatic brain injury. Transl Stroke Res 2012; 3:318-23. [PMID: 24323807 DOI: 10.1007/s12975-012-0192-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 04/24/2012] [Accepted: 05/04/2012] [Indexed: 01/15/2023]
Abstract
Traumatic brain injuries and their associated treatments carry high cost in both financial impact and morbidity to human life. Recent studies and trials present promising results in reducing secondary injury in the days and weeks following the primary insult. A number of studies, both pre-clinical and clinical, have found that different populations of stem/progenitor cells result in a reduction of inflammation, maintenance of the blood brain barrier, and an overall improved prognosis. The mechanism of action of these cellular therapies appears to rely upon the ability of the cells to influence microglia/macrophage phenotype and alter the state of the inflammatory response. The spleen has become an area of intense interest as an arena where therapeutic cells interact with reactive macrophages to cause system-level changes in immune activity. Additionally, the spleen enacts anti-inflammatory responses originating in the CNS, delivered through vagal activity with a recently described mechanism culminating in acetylcholine release. This review provides a summary of recent findings as to the mechanisms of action observed in current cellular therapies.
Collapse
Affiliation(s)
- Robert A Hetz
- Department of Pediatric Surgery, University of Texas Medical School at Houston, 6431 Fannin Street, MSB 5.234, Houston, TX, 77030, USA
| | | | | | | | | |
Collapse
|
26
|
The Neuroinflammatory Response in ALS: The Roles of Microglia and T Cells. Neurol Res Int 2012; 2012:803701. [PMID: 22666587 PMCID: PMC3362167 DOI: 10.1155/2012/803701] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/12/2012] [Indexed: 12/30/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by upper and lower motoneuron death. Mutations in the gene for superoxide dismutase 1 (SOD1) cause a familial form of ALS and have been used to develop transgenic mice which overexpress human mutant SOD1 (mSOD) and these mice exhibit a motoneuron disease which is pathologically and phenotypically similar to ALS. Neuroinflammation is a pathological hallmark of many neurodegenerative diseases including ALS and is typified by the activation and proliferation of microglia and the infiltration of T cells into the brain and spinal cord. Although the neuroinflammatory response has been considered a consequence of neuronal dysfunction and death, evidence indicates that manipulation of this response can alter disease progression. Previously viewed as deleterious to neuronal survival, recent reports suggest a trophic role for activated microglia in the mSOD mouse during the early stages of disease that is dependent on instructive signals from infiltrating T cells. However, at advanced stages of disease, activated microglia acquire increased neurotoxic potential, warranting further investigation into factors capable of skewing microglial activation towards a neurotrophic phenotype as a means of therapeutic intervention in ALS.
Collapse
|
27
|
Walsh JT, Kipnis J. Regulatory T cells in CNS injury: the simple, the complex and the confused. Trends Mol Med 2011; 17:541-7. [PMID: 21741881 PMCID: PMC3189297 DOI: 10.1016/j.molmed.2011.05.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 05/21/2011] [Accepted: 05/27/2011] [Indexed: 11/19/2022]
Abstract
Regulatory CD4(+)CD25(+)Foxp3(+) T cells (Tregs) have been the focus of significant attention for their role in controlling immune responses. Although knowledge of Treg biology has burgeoned, wide gaps remain in our understanding of Treg function under both normal and pathological conditions. Pioneering studies demonstrated roles for Tregs in cancer and autoimmune diseases, including experimental autoimmune encephalitis, and this knowledge is often applied to other pathologies including neurodegenerative conditions. However, differences between immunity in neurodegeneration and in malignancy or autoimmunity are often neglected. Thus, Treg manipulations in central nervous system (CNS) neurodegenerative conditions often yield unexpected outcomes. In this piece, we explore how the immunology of neurodegeneration differs from that of cancer and autoimmunity and how these differences create confusion about the role of Tregs in neurodegenerative conditions.
Collapse
Affiliation(s)
- James T. Walsh
- Neuroscience Graduate Program and Medical Scientist Training Program, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Neuroscience Graduate Program and Medical Scientist Training Program, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
28
|
Walker PA, Shah SK, Cox CS. Progenitor cell therapies as a novel treatment for traumatic brain injury: a pathway towards neuroprotection. ACTA ACUST UNITED AC 2011. [DOI: 10.2217/thy.11.55] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
29
|
Lee CH, Masso-Welch P, Hajishengallis G, Connell TD. TLR2-dependent modulation of dendritic cells by LT-IIa-B5, a novel mucosal adjuvant derived from a type II heat-labile enterotoxin. J Leukoc Biol 2011; 90:911-21. [PMID: 21791597 DOI: 10.1189/jlb.0511236] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A host of human pathogens invades the body at mucosal surfaces. Yet, strong, protective mucosal immune responses directed against those pathogens routinely cannot be induced without the use of adjuvants. Although the strongest mucosal adjuvants are members of the family of HLTs, the inherent toxicities of HLT holotoxins preclude their clinical use. Herein, it is shown that LT-IIa-B(5) enhances mucosal immune responses by modulating activities of DCs. i.n. immunization of mice with OVA in the presence of LT-IIa-B(5) recruited DCs to the NALT and significantly increased uptake of OVA by those DCs. Furthermore, LT-IIa-B(5) increased expression of CCR7 by DCs, which mediated enhanced migration of the cells from the NALT to the draining CLNs. LT-IIa-B(5) also enhanced maturation of DCs, as revealed by increased surface expression of CD40, CD80, and CD86. Ag-specific CD4(+) T cell proliferation was augmented in the CLNs of mice that had received i.n. LT-IIa-B(5). Finally, when used as an i.n. adjuvant, LT-IIa-B(5) dramatically increased the levels of OVA-specific salivary IgA and OVA-specific serum IgG. Strikingly, each of the activities induced by LT-IIa-B(5) was strictly TLR2-dependent. The data strongly suggest that the immunomodulatory properties of LT-IIa-B(5) depend on the productive modulation of mucosal DCs. Notably, this is the first report for any HLT to demonstrate in vivo the elicitation of strong, TLR2-dependent modulatory effects on DCs with respect to adjuvanticity.
Collapse
Affiliation(s)
- Chang Hoon Lee
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
30
|
Ferrari CC, Tarelli R. Parkinson's disease and systemic inflammation. PARKINSONS DISEASE 2011; 2011:436813. [PMID: 21403862 PMCID: PMC3049348 DOI: 10.4061/2011/436813] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 01/07/2011] [Indexed: 12/20/2022]
Abstract
Peripheral inflammation triggers exacerbation in the central brain's ongoing damage in several neurodegenerative diseases. Systemic inflammatory stimulus induce a general response known as sickness behaviour, indicating that a peripheral stimulus can induce the synthesis of cytokines in the brain. In Parkinson's disease (PD), inflammation was mainly associated with microglia activation that can underlie the neurodegeneration of neurons in the substantia nigra (SN). Peripheral inflammation can transform the “primed” microglia into an “active” state, which can trigger stronger responses dealing with neurodegenerative processes. Numerous evidences show that systemic inflammatory processes exacerbate ongoing neurodegeneration in PD patient and animal models. Anti-inflammatory treatment in PD patients exerts a neuroprotective effect. In the present paper, we analyse the effect of peripheral infections in the etiology and progression in PD patients and animal models, suggesting that these peripheral immune challenges can exacerbate the symptoms in the disease.
Collapse
Affiliation(s)
- Carina C Ferrari
- Laboratorio de Terapias Regenerativas y Protectoras del Sistema Nervioso, Fundación Instituto Leloir, Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | | |
Collapse
|
31
|
Feng J, Lu L, Hua C, Qin L, Zhao P, Wang J, Wang Y, Li W, Shi X, Jiang Y. High frequency of CD4+ CXCR5+ TFH cells in patients with immune-active chronic hepatitis B. PLoS One 2011; 6:e21698. [PMID: 21750724 PMCID: PMC3131291 DOI: 10.1371/journal.pone.0021698] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 06/05/2011] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND T follicular helper (TFH) cells are a special subpopulation of T helper cells and can regulate humoral immune responses. This study examined whether the frequency of CD4(+)CXCR5(+) TFH cells could be associated with active immunity in chronic hepatitis B (CHB) patients. METHODOLOGY AND FINDINGS The frequencies of peripheral blood CD4(+)CXCR5(+) TFH cells, inducible T cell costimulator (ICOS), and/or programmed death 1 (PD-1) positive CD4(+)CXCR5(+) TFH cells in immune-active (IA), immune-tolerant (IT) CHB, and healthy controls (HC) were characterized by flow cytometry analysis. The effect of adevofir dipivoxil treatment on the frequency of CD4(+)CXCR5(+) TFH cells, the concentrations of serum IL-2, IFN-γ, TNF-α, IL-4, IL-6, IL-10, IL-21, ALT, AST, HBsAg, HBsAb, HBeAg, HBeAb and HBV loads in IA patients were determined. The potential association of the frequency of CD4(+)CXCR5(+) TFH cells with clinical measures was analyzed. In addition, the frequency of splenic and liver CD4(+)CXCR5(+) TFH cells in HBV-transgenic mice was examined. We found that the frequency of CD4(+)CXCR5(+) TFH cells in IA patients was significantly higher than that of IT patients and HC, and the percentages of CD4(+)CXCR5(+) TFH in IA patients were positively correlated with AST. Furthermore, the percentages of ICOS(+), PD-1(+), and ICOS(+)PD-1(+) in CD4(+)CXCR5(+) TFH cells in CHB patients were significantly higher than that of HC. Treatment with adefovir dipivoxil reduced the frequency of CD4(+)CXCR5(+) TFH, PD-1(+)CD4(+)CXCR5(+) TFH cells and the concentrations of HBsAg and HBeAg, but increased the concentrations of HBsAb, HBeAb, IL-2 and IFN-γ in IA patients. Moreover, the frequency of splenic and liver CD4(+)CXCR5(+) TFH cells in HBV-transgenic mice was higher than that of wild-type controls. CONCLUSIONS These data indicate that CD4(+)CXCR5(+) TFH cells may participate in the HBV-related immune responses and that high frequency of CD4(+)CXCR5(+) TFH cells may be a biomarker for the evaluation of active immune stage of CHB patients.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Adaptive Immunity/immunology
- Adenine/analogs & derivatives
- Adenine/therapeutic use
- Adult
- Alanine Transaminase/blood
- Animals
- Antiviral Agents/therapeutic use
- Aspartate Aminotransferases/blood
- CD4 Antigens/immunology
- CD4 Antigens/metabolism
- Cytokines/blood
- DNA, Viral/blood
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Hepatitis B virus/drug effects
- Hepatitis B virus/genetics
- Hepatitis B virus/immunology
- Hepatitis B, Chronic/blood
- Hepatitis B, Chronic/drug therapy
- Hepatitis B, Chronic/immunology
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Middle Aged
- Organophosphonates/therapeutic use
- Receptors, CXCR5/immunology
- Receptors, CXCR5/metabolism
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Young Adult
Collapse
Affiliation(s)
- Junyan Feng
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Lu Lu
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Cong Hua
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Ling Qin
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Pingwei Zhao
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Juan Wang
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Ye Wang
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Wanyu Li
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Xiaodong Shi
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
| | - Yanfang Jiang
- Department of Central Laboratory, the Second Part of First Hospital, Jilin University, Changchun, China
- * E-mail:
| |
Collapse
|
32
|
Dobbs SM, Dobbs RJ, Weller C, Charlett A, Bjarnason IT, Lawson AJ, Letley D, Harbin L, Price AB, Ibrahim MAA, Oxlade NL, Bowthorpe J, Leckstroem D, Smee C, Plant JM, Peterson DW. Differential effect of Helicobacter pylori eradication on time-trends in brady/hypokinesia and rigidity in idiopathic parkinsonism. Helicobacter 2010; 15:279-94. [PMID: 20633189 PMCID: PMC2913104 DOI: 10.1111/j.1523-5378.2010.00768.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND We examine the effect of eradicating Helicobacter in idiopathic parkinsonism (IP). Marked deterioration, where eradication-therapy failed, prompted an interim report in the first 20 probands to reach de-blinding. The null-hypothesis, "eradication has no effect on principal outcome, mean stride length at free-walking speed," was rejected. We report on study completion in all 30 who had commenced post-treatment assessments. METHODS This is a randomized, placebo-controlled, parallel-group efficacy study of eradicating biopsy-proven (culture and/or organism on histopathology) Helicobacter pylori infection on the time course of facets of IP, in probands taking no, or stable long-t(1/2), anti-parkinsonian medication. Persistent infection at de-blinding (scheduled 1-year post-treatment) led to open active eradication-treatment. RESULTS Stride length improved (73 (95% CI 14-131) mm/year, p = .01) in favor of "successful" blinded active over placebo, irrespective of anti-parkinsonian medication, and despite worsening upper limb flexor rigidity (237 (57-416) Nm x 10(-3)/year, p = .01). This differential effect was echoed following open active, post-placebo. Gait did not deteriorate in year 2 and 3 post-eradication. Anti-nuclear antibody was present in all four proven (two by molecular microbiology only) eradication failures. In the remainder, it marked poorer response during the year after eradication therapy, possibly indicating residual "low-density" infection. We illustrate the importance of eradicating low-density infection, detected only by molecular microbiology, in a proband not receiving anti-parkinsonian medication. Stride length improved (424 (379-468) mm for 15 months post-eradication, p = .001), correction of deficit continuing to 3.4 years. Flexor rigidity increased before hydrogen-breath-test positivity for small intestinal bacterial overgrowth (208 (28-388) Nm x 10(-3), p = .02), increased further during (171 (67-274), p = .001) (15-31 months), and decreased (136 (6-267), p = .04) after restoration of negativity (32-41 months). CONCLUSION Helicobacter is an arbiter of progression, independent of infection-load.
Collapse
Affiliation(s)
- Sylvia M Dobbs
- Psychological Medicine and Pharmaceutical Sciences, King's College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Eder C. Ion channels in monocytes and microglia / brain macrophages: Promising therapeutic targets for neurological diseases. J Neuroimmunol 2010; 224:51-5. [DOI: 10.1016/j.jneuroim.2010.05.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 05/04/2010] [Indexed: 10/19/2022]
|
34
|
Kraft-Terry SD, Stothert AR, Buch S, Gendelman HE. HIV-1 neuroimmunity in the era of antiretroviral therapy. Neurobiol Dis 2010; 37:542-8. [PMID: 20044002 DOI: 10.1016/j.nbd.2009.12.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 12/13/2009] [Accepted: 12/17/2009] [Indexed: 01/08/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1)-associated neurocognitive disorders (HAND) can affect up to 50% of infected people during the disease course. While antiretroviral therapies have substantively increased the quality of life and reduced HIV-1-associated dementia, less severe minor cognitive and motor deficits continue. Trafficking of HIV-1 into the central nervous system (CNS), peripheral immune activation, dysregulated glial immunity, and diminished homeostatic responses are the disease-linked pathobiologic events. Monocyte-macrophage passage into the CNS remains an underlying force for disease severity. Monocyte phenotypes may change at an early stage of cell maturation and immune activation of hematopoietic stem cells. Activated monocytes are pulled into the brain in response to chemokines made as a result of glial inflammatory processes, which in turn, cause secondary functional deficits in neurons. Current therapeutic approaches are focused on adjunctive and brain-penetrating antiretroviral therapies. These may attenuate virus-associated neuroinflammatory activities thereby decreasing the severity and frequency of HAND.
Collapse
Affiliation(s)
- Stephanie D Kraft-Terry
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | |
Collapse
|
35
|
Maingat F, Viappiani S, Zhu Y, Vivithanaporn P, Ellestad KK, Holden J, Silva C, Power C. Regulation of lentivirus neurovirulence by lipopolysaccharide conditioning: suppression of CXCL10 in the brain by IL-10. THE JOURNAL OF IMMUNOLOGY 2009; 184:1566-74. [PMID: 20042580 DOI: 10.4049/jimmunol.0902575] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lentivirus infections including HIV and feline immunodeficiency virus (FIV) cause neurovirulence, which is largely mediated by innate immunity. To investigate the interactions between neurovirulence and repeated conditioning by innate immune activation, models of lentivirus infection were exposed to LPS. Gene expression in HIV-infected (HIV+) and control (HIV-) patient brains was compared by real time RT-PCR and immunocytochemistry. Supernatants from mock and HIV-infected monocyte-derived macrophages exposed to LPS were applied to human neurons. FIV-infected (FIV+) and control (FIV-) animals were exposed repeatedly to LPS postinfection together with concurrent neurobehavioral testing, viral load, and host gene analyses. Brains from HIV+ individuals exhibited induction of CD3epsilon, CXCL10, and granzyme A expression (p < 0.05). Supernatants from HIV+ monocyte-derived macrophages induced CXCL10 expression in neurons, which was diminished by IL-10 treatment (p < 0.05). LPS-exposed FIV+ animals demonstrated lower plasma and brain viral loads (p < 0.05). Neuronal CXCL10 expression was increased in FIV+ animals but was suppressed by LPS exposure, together with reduced brain CD3epsilon and granzyme A expression (p < 0.05). In conjunction with preserved NeuN-positive neuronal counts in parietal cortex (p < 0.05), FIV+ animals exposed to LPS also showed less severe neurobehavioral deficits (p < 0.05). Repeated LPS exposures suppressed CXCL10 in the brain and ensuing T cell infiltration with a concomitant reduction in neurovirulence. Thus, innate immune chronic conditioning exerted beneficial effects on neurovirulence through suppression of a specific chemotactic factor, CXCL10, mediated by IL-10, leading to reduced leukocyte infiltration and release of neurotoxic factors.
Collapse
|
36
|
Blood profile holds clues to role of infection in a premonitory state for idiopathic parkinsonism and of gastrointestinal infection in established disease. Gut Pathog 2009; 1:20. [PMID: 19941660 PMCID: PMC2795757 DOI: 10.1186/1757-4749-1-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 11/26/2009] [Indexed: 12/25/2022] Open
Abstract
The two-stage neuroinflammatory process, containment and progression, proposed to underlie neurodegeneration may predicate on systemic inflammation arising from the gastrointestinal tract. Helicobacter infection has been described as one switch in the pathogenic-circuitry of idiopathic parkinsonism (IP): eradication modifies disease progression and marked deterioration accompanies eradication-failure. Moreover, serum Helicobacter-antibody-profile predicts presence, severity and progression of IP. Slow gastrointestinal-transit precedes IP-diagnosis and becomes increasingly-apparent after, predisposing to small-intestinal bacterial-overgrowth (SIBO). Although IP is well-described as a systemic illness with a long prodrome, there has been no comprehensive overview of the blood profile. Here, it is examined in relation to Helicobacter status and lactulose-hydrogen-breath-testing for SIBO. A robust finding of reduced lymphocyte count in 126 IP-probands and 79 spouses (without clinically-definite IP), compared with that in 381 controls (p < 0.001 in each case), was not explained by Helicobacter-status or breath-hydrogen. This complements a previous report that spouses were 'down-the-pathway' to 'clinically-definite' disease. In 205 other controls without clinically-definite IP, there were strong associations between sporadic cardinal features and immunoglobulin class concentration, not explained by Helicobacter-status. Premonitory states for idiopathic parkinsonism associated with relative lymphopenia, higher serum immunoglobulin concentrations and evidence of enteric-nervous-system damage may prove viral in origin. Although only 8% of the above 79 spouses were urea-breath-test-positive for Helicobacter, all 8 spouses with clinically-definite IP were (p < 0.0001). Transmission of a 'primer' to a Helicobacter-colonised recipient might result in progression to the diagnostic threshold. Twenty-five percent of the 126 probands were seropositive for anti-nuclear autoantibody. In 20 probands, monitored before and serially after anti-Helicobacter therapy, seropositivity marked a severe hypokinetic response (p = 0.03). It may alert to continuing infection, even at low-density. Hyperhomocysteinemia is a risk factor for dementia and depression. Serum homocysteine exceeded the target in 43% of the 126 IP-probands. It was partially explained by serum B12 (12% variance, p < 0.001), but not by Helicobacter-status (gastric-atrophy uncommon in IP) or levodopa treatment. Immune-inflammatory activation increases homocysteine production. Since an estimated 60% of probands are hydrogen-breath-test positive, SIBO, with its increased bacterial utilisation of B12, is a likely cause. Thus, two prognostic indicators in established IP fit with involvement of Helicobacter and SIBO.
Collapse
|
37
|
A coat of many colors: neuroimmune crosstalk in human immunodeficiency virus infection. Neuron 2009; 64:133-45. [PMID: 19840555 DOI: 10.1016/j.neuron.2009.09.042] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2009] [Indexed: 01/20/2023]
Abstract
The use of antiretroviral therapy has reduced mortality and increased the quality of life of HIV-1-infected people, particularly in more developed countries where access to treatment is more widespread. However, morbidities continue, which include HIV-1-associated neurocognitive disorders (HAND). Subtle cognitive abnormalities and low-level viral replication underlie disease. The balance between robust antiviral adaptive immunity, neuronal homeostatic mechanisms, and neuroprotective factors on one hand and toxicities afforded by dysregulated immune activities on the other govern disease. New insights into the pathobiological processes for neuroimmune-linked disease and ways to modulate such activities for therapeutic gain are discussed. Better understanding of the complexities of immune regulation during HAND can improve diagnosis and disease outcomes but is also relevant for the pathogenesis of a broad range of neurodegenerative disorders.
Collapse
|
38
|
Nowacek A, Kosloski LM, Gendelman HE. Neurodegenerative disorders and nanoformulated drug development. Nanomedicine (Lond) 2009; 4:541-55. [PMID: 19572820 PMCID: PMC2727651 DOI: 10.2217/nnm.09.37] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Degenerative and inflammatory diseases of the CNS include, but are not limited to, Alzheimer's and Parkinson's disease, amyotrophic lateral sclerosis, stroke, multiple sclerosis and HIV-1-associated neurocognitive disorders. These are common, debilitating and, unfortunately, hold few therapeutic options. In recent years, the application of nanotechnologies as commonly used or developing medicines has served to improve pharmacokinetics and drug delivery specifically to CNS-diseased areas. In addition, nanomedical advances are leading to therapies that target CNS pathobiology and as such, can interrupt disordered protein aggregation, deliver functional neuroprotective proteins and alter the oxidant state of affected neural tissues. This article focuses on the pathobiology of common neurodegenerative disorders with a view towards how nanomedicine may be used to improve the clinical course of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ari Nowacek
- Department of Pharmacology, & Experimental Neuroscience, Center for Neurovirology &, Neurodegenerative Disorders, University of Nebraska, Medical Center, Omaha, NE 68198-5880, USA
| | - Lisa M Kosloski
- Department of Pharmacology, & Experimental Neuroscience, Center for Neurovirology &, Neurodegenerative Disorders, University of Nebraska, Medical Center, Omaha, NE 68198-5880, USA
| | - Howard E Gendelman
- Department of Pharmacology, & Experimental Neuroscience, Center for Neurovirology &, Neurodegenerative Disorders, University of Nebraska, Medical Center, Omaha, NE 68198-5880, USA
| |
Collapse
|
39
|
Abstract
A broad range of nanomedicines is being developed to improve drug delivery for CNS disorders. The structure of the blood-brain barrier (BBB), the presence of efflux pumps and the expression of metabolic enzymes pose hurdles for drug-brain entry. Nanoformulations can circumvent the BBB to improve CNS-directed drug delivery by affecting such pumps and enzymes. Alternatively, they can be optimized to affect their size, shape, and protein and lipid coatings to facilitate drug uptake, release and ingress across the barrier. This is important as the brain is a sanctuary for a broad range of pathogens including HIV-1. Improved drug delivery to the CNS would affect pharmacokinetic and drug biodistribution properties. This article focuses on how nanotechnology can serve to improve the delivery of antiretroviral medicines, termed nanoART, across the BBB and affect the biodistribution and clinical benefit for HIV-1 disease.
Collapse
Affiliation(s)
| | - Howard E Gendelman
- Author for correspondence: Department of Pharmacology & Experimental Neuroscience, Center for Neurovirology & Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA, Tel.: +1 402 559 8920, Fax: +1 402 559 3744,
| |
Collapse
|