1
|
Carregosa D, Loncarevic-Vasiljkovic N, Feliciano R, Moura-Louro D, Mendes CS, Dos Santos CN. Locomotor and gait changes in the LPS model of neuroinflammation are correlated with inflammatory cytokines in blood and brain. J Inflamm (Lond) 2024; 21:39. [PMID: 39379968 PMCID: PMC11463041 DOI: 10.1186/s12950-024-00412-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
Lipopolysaccharide (LPS) challenge in mice has been used to identify the mechanisms and therapeutics for neuroinflammation. In this study, we aimed to comprehensively evaluate the behavioral changes including locomotion, exploration, and memory, correlating them with a panel of thirteen inflammatory cytokines in both blood and brain.We found that acute LPS administration (0.83 mg/Kg i.p.) reduced body weight, food intake, and glucose levels compared to the saline-injected mice, concomitant with decreased activity in home cage monitoring. Locomotion was significantly reduced in Open Field, Introduced Object, and Y-Maze tests. Decreased exploratory behavior in the Y-Maze and Introduced Object tests was noticed, by measuring the number of arms explored and object interaction time, respectively. Additionally, in rotarod, LPS administration led to a significant decrease in the distance achieved, while in the MouseWalker, LPS led to a reduction in average velocity.LPS induced a decrease in microglia ramification index in the motor cortex and the striatum, while surprisingly a reduction in microglia number was observed in the motor cortex.The concentrations of thirteen cytokines in the blood were significantly altered, while only CXCL1, CCL22, CCL17, G-CSF, and IL-12p40 were changed in the brain. Correlations between cytokine levels in blood and brain were found, most notably for CCL17 and CCL22. TGFβ was the only one with negative correlations to other cytokines. Correlations between cytokines and behavior changes were also disclosed, especially for CCL17, CCL22, G-CSF, and IL-6 and negatively for TGFβ and IL-10.In summary, our study employing acute LPS challenge in mice has revealed a comprehensive profile of behavioral alterations alongside significant changes in inflammatory cytokine levels, both in peripheral blood and brain tissue. These findings contribute to a deeper understanding of the interplay between inflammation and behavior, with possible implications for identifying prognostics and therapeutic targets for neuroinflammatory conditions.
Collapse
Affiliation(s)
- Diogo Carregosa
- iNOVA4Health, NOVA Medical School | Faculdade Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Natasa Loncarevic-Vasiljkovic
- iNOVA4Health, NOVA Medical School | Faculdade Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Raquel Feliciano
- iNOVA4Health, NOVA Medical School | Faculdade Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Diogo Moura-Louro
- iNOVA4Health, NOVA Medical School | Faculdade Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - César S Mendes
- iNOVA4Health, NOVA Medical School | Faculdade Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Cláudia Nunes Dos Santos
- iNOVA4Health, NOVA Medical School | Faculdade Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal.
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal.
| |
Collapse
|
2
|
Jagadeesan N, Roules GC, Chandrashekar DV, Yang J, Kolluru S, Sumbria RK. Modulation of hippocampal protein expression by a brain penetrant biologic TNF-α inhibitor in the 3xTg Alzheimer's disease mice. J Transl Med 2024; 22:291. [PMID: 38500108 PMCID: PMC10946165 DOI: 10.1186/s12967-024-05008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Biologic TNF-α inhibitors (bTNFIs) can block cerebral TNF-α in Alzheimer's disease (AD) if these macromolecules can cross the blood-brain barrier (BBB). Thus, a model bTNFI, the extracellular domain of type II TNF-α receptor (TNFR), which can bind to and sequester TNF-α, was fused with a mouse transferrin receptor antibody (TfRMAb) to enable brain delivery via BBB TfR-mediated transcytosis. Previously, we found TfRMAb-TNFR to be protective in a mouse model of amyloidosis (APP/PS1) and tauopathy (PS19), and herein we investigated its effects in mice that combine both amyloidosis and tauopathy (3xTg-AD). METHODS Eight-month-old female 3xTg-AD mice were injected intraperitoneally with saline (n = 11) or TfRMAb-TNFR (3 mg/kg; n = 11) three days per week for 12 weeks. Age-matched wild-type (WT) mice (n = 9) were treated similarly with saline. Brains were processed for immunostaining and high-resolution multiplex NanoString GeoMx spatial proteomics. RESULTS We observed regional differences in proteins relevant to Aβ, tau, and neuroinflammation in the hippocampus of 3xTg-AD mice compared with WT mice. From 64 target proteins studied using spatial proteomics, a comparison of the Aβ-plaque bearing vs. plaque-free regions in the 3xTg-AD mice yielded 39 differentially expressed proteins (DEP) largely related to neuroinflammation (39% of DEP) and Aβ and tau pathology combined (31% of DEP). Hippocampal spatial proteomics revealed that the majority of the proteins modulated by TfRMAb-TNFR in the 3xTg-AD mice were relevant to microglial function (⁓ 33%). TfRMAb-TNFR significantly reduced mature Aβ plaques and increased Aβ-associated microglia around larger Aβ deposits in the 3xTg-AD mice. Further, TfRMAb-TNFR increased mature Aβ plaque-associated microglial TREM2 in 3xTg-AD mice. CONCLUSION Overall, despite the low visual Aβ load in the 11-month-old female 3xTg-AD mice, our results highlight region-specific AD-relevant DEP in the hippocampus of these mice. Chronic TfRMAb-TNFR dosing modulated several DEP involved in AD pathology and showed a largely microglia-centric mechanism of action in the 3xTg-AD mice.
Collapse
Affiliation(s)
- Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - G Chuli Roules
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Devaraj V Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Joshua Yang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Sanjana Kolluru
- Rancho Cucamonga High School, 11801 Lark Dr, Rancho Cucamonga, CA, 91701, USA
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA.
- Department of Neurology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
3
|
Kloske CM, Barnum CJ, Batista AF, Bradshaw EM, Brickman AM, Bu G, Dennison J, Gearon MD, Goate AM, Haass C, Heneka MT, Hu WT, Huggins LKL, Jones NS, Koldamova R, Lemere CA, Liddelow SA, Marcora E, Marsh SE, Nielsen HM, Petersen KK, Petersen M, Piña-Escudero SD, Qiu WQ, Quiroz YT, Reiman E, Sexton C, Tansey MG, Tcw J, Teunissen CE, Tijms BM, van der Kant R, Wallings R, Weninger SC, Wharton W, Wilcock DM, Wishard TJ, Worley SL, Zetterberg H, Carrillo MC. APOE and immunity: Research highlights. Alzheimers Dement 2023; 19:2677-2696. [PMID: 36975090 DOI: 10.1002/alz.13020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 03/29/2023]
Abstract
INTRODUCTION At the Alzheimer's Association's APOE and Immunity virtual conference, held in October 2021, leading neuroscience experts shared recent research advances on and inspiring insights into the various roles that both the apolipoprotein E gene (APOE) and facets of immunity play in neurodegenerative diseases, including Alzheimer's disease and other dementias. METHODS The meeting brought together more than 1200 registered attendees from 62 different countries, representing the realms of academia and industry. RESULTS During the 4-day meeting, presenters illuminated aspects of the cross-talk between APOE and immunity, with a focus on the roles of microglia, triggering receptor expressed on myeloid cells 2 (TREM2), and components of inflammation (e.g., tumor necrosis factor α [TNFα]). DISCUSSION This manuscript emphasizes the importance of diversity in current and future research and presents an integrated view of innate immune functions in Alzheimer's disease as well as related promising directions in drug development.
Collapse
Affiliation(s)
| | | | - Andre F Batista
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Departments of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth M Bradshaw
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, G.H. Sergievsky Center, and Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jessica Dennison
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mary D Gearon
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alison M Goate
- Department of Genetics & Genomic Sciences, Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christian Haass
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany 3 Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB) University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - William T Hu
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School and Center for Healthy Aging, Rutgers Institute for Health, Health Care Policy, and Aging Research, New Brunswick, New Jersey, USA
| | - Lenique K L Huggins
- Department of Biology, Duke University, Durham, North Carolina, USA
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Nahdia S Jones
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, USA
| | - Radosveta Koldamova
- EOH, School of Public Health University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cynthia A Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Departments of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Shane A Liddelow
- Neuroscience Institute and Departments of Neuroscience & Physiology and of Ophthalmology, NYU Grossman School of Medicine, New York, New York, USA
| | - Edoardo Marcora
- Ronald M. Loeb Center for Alzheimer's disease, Dept. of Genetics & Genomic Sciences, Dept. of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Samuel E Marsh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Kellen K Petersen
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Melissa Petersen
- Department of Family Medicine, Institute of Translational Research, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Stefanie D Piña-Escudero
- Global Brain Health Institute, Department of Neurology, University of California, San Francisco, California, USA
| | - Wei Qiao Qiu
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Yakeel T Quiroz
- Departments of Psychiatry and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Reiman
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
- Banner Research, Phoenix, Arizona, USA
| | | | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Julia Tcw
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Clinical Chemistry department, Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Rik van der Kant
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Rebecca Wallings
- CTRND, Department of Neuroscience, University of Florida, Florida, USA
| | | | | | - Donna M Wilcock
- Sanders-Brown Center on Aging and Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Tyler James Wishard
- Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Susan L Worley
- Independent science writer, Bryn Mawr, Pennsylvania, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | | |
Collapse
|
4
|
Wei ZH, Koya J, Acharekar N, Trejos J, Dong XD, Schanne FA, Ashby CR, Reznik SE. N,N-dimethylacetamide targets neuroinflammation in Alzheimer's disease in in-vitro and ex-vivo models. Sci Rep 2023; 13:7077. [PMID: 37127686 PMCID: PMC10151369 DOI: 10.1038/s41598-023-34355-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/28/2023] [Indexed: 05/03/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic degenerative brain disorder with no clear pathogenesis or effective cure, accounting for 60-80% of cases of dementia. In recent years, the importance of neuroinflammation in the pathogenesis of AD and other neurodegenerative disorders has come into focus. Previously, we made the serendipitous discovery that the widely used drug excipient N,N-dimethylacetamide (DMA) attenuates endotoxin-induced inflammatory responses in vivo. In the current work, we investigate the effect of DMA on neuroinflammation and its mechanism of action in in-vitro and ex-vivo models of AD. We show that DMA significantly suppresses the production of inflammatory mediators, such as reactive oxygen species (ROS), nitric oxide (NO) and various cytokines and chemokines, as well as amyloid-β (Aβ), in cultured microglia and organotypic hippocampal slices induced by lipopolysaccharide (LPS). We also demonstrate that DMA inhibits Aβ-induced inflammation. Finally, we show that the mechanism of DMA's effect on neuroinflammation is inhibition of the nuclear factor kappa-B (NF-κB) signaling pathway and we show how DMA dismantles the positive feedback loop between NF-κB and Aβ synthesis. Taken together, our findings suggest that DMA, a generally regarded as safe compound that crosses the blood brain barrier, should be further investigated as a potential therapy for Alzheimer's disease and neuroinflammatory disorders.
Collapse
Affiliation(s)
- Zeng-Hui Wei
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Nikita Acharekar
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jesus Trejos
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Xing-Duo Dong
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Francis A Schanne
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, 11439, USA.
- Departments of Pathology and Obstetrics and Gynecology and Women's Health, The University Hospital for Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, 10461, USA.
| |
Collapse
|
5
|
Qi C, Liu F, Zhang W, Han Y, Zhang N, Liu Q, Li H. Alzheimer's disease alters the transcriptomic profile of natural killer cells at single-cell resolution. Front Immunol 2022; 13:1004885. [PMID: 36405736 PMCID: PMC9666759 DOI: 10.3389/fimmu.2022.1004885] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/12/2022] [Indexed: 01/25/2023] Open
Abstract
Alzheimer's disease (AD) is the most common dementia without an effective cure at least partially due to incomplete understanding of the disease. Inflammation has emerged as a central player in the onset and progression of AD. As innate lymphoid cells, natural killer (NK) cells orchestrate the initiation and evolution of inflammatory responses. Yet, the transcriptomic features of NK cells in AD remain poorly understood. We assessed the diversity of NK cells using web-based single-cell RNA sequencing data of blood NK cells from patients with AD and control subjects and flow cytometry. We identified a contraction of NK cell compartment in AD, accompanied by a reduction of cytotoxicity. Unbiased clustering revealed four subsets of NK cells in AD, i.e., CD56bright NK cells, CD56dim effector NK cells, adaptive NK cells, and a unique NK cell subset that is expanded and characterized by upregulation of CX3CR1, TBX21, MYOM2, DUSP1, and ZFP36L2, and negatively correlated with cognitive function in AD patients. Pseudo-temporal analysis revealed that this unique NK cell subset was at a late stage of NK cell development and enriched with transcription factors TBX21, NFATC2, and SMAD3. Together, our study identified a distinct NK cell subset and its potential involvement in AD.
Collapse
Affiliation(s)
| | | | | | | | - Nan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Handong Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
6
|
Di Benedetto G, Burgaletto C, Serapide MF, Caltabiano R, Munafò A, Bellanca CM, Di Mauro R, Bernardini R, Cantarella G. TRAIL-R Deficient Mice Are Protected from Neurotoxic Effects of Amyloid-β. Int J Mol Sci 2022; 23:ijms231911625. [PMID: 36232931 PMCID: PMC9569968 DOI: 10.3390/ijms231911625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/19/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
TRAIL, a member of TNF superfamily, is a potent inducer of neuronal death. Neurotoxic effects of TRAIL appear mediated by its death receptor TRAIL-R2/DR5. To assess the role of TRAIL/TRAIL-R2 pathway in AD-related neurodegeneration, we studied the impact of the treatment with amyloid-β (Aβ) upon cell viability and inflammation in TRAIL-R-deficient mice (TRAIL-R−/−). Here, we demonstrate that the lack of TRAIL-R2 protects from death cultured TRAIL-R−/− mouse embryonic hippocampal cells after treatment with either Aβ1-42 or TRAIL. Consistently, stereotaxic injection of Aβ1-42 resulted in blunted caspase activation, as well as in reduction of JNK phosphorylation and increased AKT phosphorylation in TRAIL-R−/− mice. Moreover, the lack of TRAIL-R2 was associated with blunted constitutive p53 expression in mice that have undergone Aβ1-42 treatment, as well as in decrease of phosphorylated forms of tau and GSK3β proteins. Likewise, TRAIL-R2 appears essential to both TRAIL and Aβ-mediated neurotoxicity and inflammation. Indeed, hippocampi of TRAIL-R−/− mice challenged with Aβ1-42, showed a slight expression of microglial (Iba-1) and astrocytic (GFAP) markers along with attenuated levels of IL-1β, TNF-α, NOS2 and COX2. In conclusion, the bulk of these results demonstrate that the constitutive lack of TRAIL-R2 is associated with a substantial reduction of noxious effects of Aβ1-42, providing further evidence on the prominent role played by TRAIL in course of Aβ-related neurodegeneration and confirming that the TRAIL system represents a potential target for innovative AD therapy.
Collapse
Affiliation(s)
- Giulia Di Benedetto
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Chiara Burgaletto
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Maria Francesca Serapide
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rosario Caltabiano
- Section of Anatomic Pathology, Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy
| | - Antonio Munafò
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Carlo Maria Bellanca
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rosaria Di Mauro
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Clinical Toxicology Unit, University Hospital of Catania, 95123 Catania, Italy
| | - Renato Bernardini
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Clinical Toxicology Unit, University Hospital of Catania, 95123 Catania, Italy
- Correspondence: ; Tel.: +39-0954781190
| | - Giuseppina Cantarella
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| |
Collapse
|
7
|
Tamura Y, Yamato M, Kataoka Y. Animal Models for Neuroinflammation and Potential Treatment Methods. Front Neurol 2022; 13:890217. [PMID: 35832182 PMCID: PMC9271866 DOI: 10.3389/fneur.2022.890217] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/03/2022] [Indexed: 11/25/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic disease of unknown etiology and without effective treatment options. The onset of ME/CFS is often associated with neuroinflammation following bacterial or viral infection. A positron emission tomography imaging study revealed that the degree of neuroinflammation was correlated with the severity of several symptoms in patients with ME/CFS. In animal studies, lipopolysaccharide- and polyinosinic-polycytidylic acid-induced models are thought to mimic the pathological features of ME/CFS and provoke neuroinflammation, characterized by increased levels of proinflammatory cytokines and activation of microglia. In this review, we described the anti-inflammatory effects of three compounds on neuroinflammatory responses utilizing animal models. The findings of the included studies suggest that anti-inflammatory substances may be used as effective therapies to ameliorate disease symptoms in patients with ME/CFS.
Collapse
Affiliation(s)
- Yasuhisa Tamura
- Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, RIKEN, Kobe, Japan
| | - Masanori Yamato
- Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, RIKEN, Kobe, Japan
| | - Yosky Kataoka
- Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, RIKEN, Kobe, Japan
- *Correspondence: Yosky Kataoka
| |
Collapse
|
8
|
Hudák A, Letoha A, Vizler C, Letoha T. Syndecan-3 as a Novel Biomarker in Alzheimer's Disease. Int J Mol Sci 2022; 23:3407. [PMID: 35328830 PMCID: PMC8955174 DOI: 10.3390/ijms23063407] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/17/2022] Open
Abstract
Early diagnosis of Alzheimer's disease (AD) is of paramount importance in preserving the patient's mental and physical health in a fairly manageable condition for a longer period. Reliable AD detection requires novel biomarkers indicating central nervous system (CNS) degeneration in the periphery. Members of the syndecan family of transmembrane proteoglycans are emerging new targets in inflammatory and neurodegenerative disorders. Reviewing the growing scientific evidence on the involvement of syndecans in the pathomechanism of AD, we analyzed the expression of the neuronal syndecan, syndecan-3 (SDC3), in experimental models of neurodegeneration. Initial in vitro studies showed that prolonged treatment of tumor necrosis factor-alpha (TNF-α) increases SDC3 expression in model neuronal and brain microvascular endothelial cell lines. In vivo studies revealed elevated concentrations of TNF-α in the blood and brain of APPSWE-Tau transgenic mice, along with increased SDC3 concentration in the brain and the liver. Primary brain endothelial cells and peripheral blood monocytes isolated from APPSWE-Tau mice exhibited increased SDC3 expression than wild-type controls. SDC3 expression of blood-derived monocytes showed a positive correlation with amyloid plaque load in the brain, demonstrating that SDC3 on monocytes is a good indicator of amyloid pathology in the brain. Given the well-established role of blood tests, the SDC3 expression of monocytes could serve as a novel biomarker for early AD detection.
Collapse
Affiliation(s)
| | - Annamária Letoha
- Albert Szent-Györgyi Clinical Center, Department of Medicine, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary;
| | - Csaba Vizler
- Biological Research Centre, Institute of Biochemistry, H-6726 Szeged, Hungary;
| | | |
Collapse
|
9
|
Sunkaria A, Bhardwaj S. Sleep Disturbance and Alzheimer's Disease: The Glial Connection. Neurochem Res 2022; 47:1799-1815. [PMID: 35303225 DOI: 10.1007/s11064-022-03578-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
Poor quality and quantity of sleep are very common in elderly people throughout the world. Growing evidence has suggested that sleep disturbances could accelerate the process of neurodegeneration. Recent reports have shown a positive correlation between sleep deprivation and amyloid-β (Aβ)/tau aggregation in the brain of Alzheimer's patients. Glial cells have long been implicated in the progression of Alzheimer's disease (AD) and recent findings have also suggested their role in regulating sleep homeostasis. However, how glial cells control the sleep-wake balance and exactly how disturbed sleep may act as a trigger for Alzheimer's or other neurological disorders have recently gotten attention. In an attempt to connect the dots, the present review has highlighted the role of glia-derived sleep regulatory molecules in AD pathogenesis. Role of glia in sleep disturbance and Alzheimer's progression.
Collapse
Affiliation(s)
- Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| | - Supriya Bhardwaj
- Department of Dermatology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| |
Collapse
|
10
|
Wei Z, Koya J, Reznik SE. Insulin Resistance Exacerbates Alzheimer Disease via Multiple Mechanisms. Front Neurosci 2021; 15:687157. [PMID: 34349617 PMCID: PMC8326507 DOI: 10.3389/fnins.2021.687157] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is a chronic neurodegenerative disease that accounts for 60–70% of dementia and is the sixth leading cause of death in the United States. The pathogenesis of this debilitating disorder is still not completely understood. New insights into the pathogenesis of AD are needed in order to develop novel pharmacologic approaches. In recent years, numerous studies have shown that insulin resistance plays a significant role in the development of AD. Over 80% of patients with AD have type II diabetes (T2DM) or abnormal serum glucose, suggesting that the pathogenic mechanisms of insulin resistance and AD likely overlap. Insulin resistance increases neuroinflammation, which promotes both amyloid β-protein deposition and aberrant tau phosphorylation. By increasing production of reactive oxygen species, insulin resistance triggers amyloid β-protein accumulation. Oxidative stress associated with insulin resistance also dysregulates glycogen synthase kinase 3-β (GSK-3β), which leads to increased tau phosphorylation. Both insulin and amyloid β-protein are metabolized by insulin degrading enzyme (IDE). Defects in this enzyme are the basis for a strong association between T2DM and AD. This review highlights multiple pathogenic mechanisms induced by insulin resistance that are implicated in AD. Several pharmacologic approaches to AD associated with insulin resistance are presented.
Collapse
Affiliation(s)
- Zenghui Wei
- Department of Pharmaceutical Sciences, St. John's University, New York, NY, United States
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, St. John's University, New York, NY, United States
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, New York, NY, United States.,Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States.,Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
11
|
Imran M, Shah FA, Nadeem H, Zeb A, Faheem M, Naz S, Bukhari A, Ali T, Li S. Synthesis and Biological Evaluation of Benzimidazole Derivatives as Potential Neuroprotective Agents in an Ethanol-Induced Rodent Model. ACS Chem Neurosci 2021; 12:489-505. [PMID: 33430586 DOI: 10.1021/acschemneuro.0c00659] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is the most devastating and progressive neurodegenerative disease in middle to elder aged people, which can be exacerbated by lifestyle factors. Recent longitudinal studies demonstrated that alcohol consumption exacerbates memory impairments in adults. However, the underlying mechanism of alcohol-induced memory impairment is still elusive. The increased cellular manifestation of reactive oxygen species (ROS) and the production of numerous proinflammatory markers play a critical role in the neurodegeneration and pathogenesis of AD. Therefore, reducing neurodegeneration by decreasing oxidative stress and neuroinflammation may provide a potential therapeutic roadmap for the treatment of AD. In this study, eight new benzimidazole acetamide derivatives (FP1, FP2, FP5-FP10) were synthesized and characterized to investigate its neuroprotective effects in ethanol-induced neurodegeneration in a rat model. Further, three derivatives (FP1, FP7, and FP8) were selected for in vivo molecular analysis based on preliminary in vitro antioxidant screening assay. Molecular docking analysis was performed to assess the affinity of synthesized benzimidazole acetamide derivatives against selected proinflammatory targets (TNF-α, IL-6). Biochemical analysis revealed elevated expression of neuroinflammatory markers (TNF-α, NF-κB, IL-6, NLRP3), increased cellular oxidative stress, and reduced antioxidant enzymes in ethanol-exposed rats brain. Notably, pretreatment with new benzimidazole acetamide derivatives (FP1, FP7, and FP8) significantly modulated the ethanol-induced memory deficits, oxidative stress, and proinflammatory markers (TNF-α, NF-κB, IL-6, NLRP3) in the cortex. The multipurpose nature of acetamide containing benzimidazole nucleus and its versatile affinity toward numerous receptors highlight its multistep targeting potential. These results indicated the neuroprotective potential of benzimidazole acetamide derivatives (FP1, FP7, and FP8) as novel therapeutic candidates in ethanol-induced neurodegeneration which may partially be due to inhibition of the neuroinflammatory-oxidative stress vicious cycle.
Collapse
Affiliation(s)
- Muhammad Imran
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Fawad Ali Shah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Alam Zeb
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Muhammad Faheem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Shagufta Naz
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Asma Bukhari
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Tahir Ali
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen 518055, China
| |
Collapse
|
12
|
Abdul Manap AS, Madhavan P, Vijayabalan S, Chia A, Fukui K. Explicating anti-amyloidogenic role of curcumin and piperine via amyloid beta (A β) explicit pathway: recovery and reversal paradigm effects. PeerJ 2020; 8:e10003. [PMID: 33062432 PMCID: PMC7532763 DOI: 10.7717/peerj.10003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/30/2020] [Indexed: 12/28/2022] Open
Abstract
Previously, we reported the synergistic effects of curcumin and piperine in cell cultures as potential anti-cholinesterase and anti-amyloidogenic agents. Due to limited findings on the enrolment of these compounds on epigenetic events in AD, we aimed at elucidating the expression profiles of Aβ42-induced SH-SY5Y cells using microarray profiling. In this study, an optimized concentration of 35 µM of curcumin and piperine in combination was used to treat Aβ42 fibril and high-throughput microarray profiling was performed on the extracted RNA. This was then compared to curcumin and piperine used singularly at 49.11 µM and 25 µM, respectively. Our results demonstrated that in the curcumin treated group, from the top 10 upregulated and top 10 downregulated significantly differentially expressed genes (p < 0.05; fold change ≥ 2 or ≤ -2), there were five upregulated and three downregulated genes involved in the amyloidogenic pathway. While from top 10 upregulated and top 10 downregulated significantly differentially expressed genes (p < 0.05; fold change ≥ 2 or ≤ - 2) in the piperine treated group, there were four upregulated and three downregulated genes involved in the same pathway, whereas there were five upregulated and two downregulated genes involved (p < 0.05; fold change ≥ 2 or ≤ - 2) in the curcumin-piperine combined group. Four genes namely GABARAPL1, CTSB, RAB5 and AK5 were expressed significantly in all groups. Other genes such as ITPR1, GSK3B, PPP3CC, ERN1, APH1A, CYCS and CALM2 were novel putative genes that are involved in the pathogenesis of AD. We revealed that curcumin and piperine have displayed their actions against Aβ via the modulation of various mechanistic pathways. Alterations in expression profiles of genes in the neuronal cell model may explain Aβ pathology post-treatment and provide new insights for remedial approaches of a combined treatment using curcumin and piperine.
Collapse
Affiliation(s)
- Aimi Syamima Abdul Manap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Shantini Vijayabalan
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Adeline Chia
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Koji Fukui
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| |
Collapse
|
13
|
Prenatal exposure to bisphenol A alters the transcriptome-interactome profiles of genes associated with Alzheimer's disease in the offspring hippocampus. Sci Rep 2020; 10:9487. [PMID: 32528016 PMCID: PMC7289845 DOI: 10.1038/s41598-020-65229-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/27/2020] [Indexed: 01/01/2023] Open
Abstract
Our recent study revealed that prenatal exposure to bisphenol A (BPA) disrupted the transcriptome profiles of genes in the offspring hippocampus. In addition to genes linked to autism, several genes associated with Alzheimer’s disease (AD) were found to be differentially expressed, although the association between BPA-responsive genes and AD-related genes has not been thoroughly investigated. Here, we demonstrated that in utero BPA exposure also disrupted the transcriptome profiles of genes associated with neuroinflammation and AD in the hippocampus. The level of NF-κB protein and its AD-related target gene Bace1 were significantly increased in the offspring hippocampus in a sex-dependent manner. Quantitative RT-PCR analysis also showed an increase in the expression of Tnf gene. Moreover, the reanalysis of transcriptome profiling data from several previously published BPA studies consistently showed that BPA-responsive genes were significantly associated with top AD candidate genes. The findings from this study suggest that maternal BPA exposure may increase AD risk in offspring by dysregulating genes associated with AD neuropathology and inflammation and reveal a possible relationship between AD and autism, which are linked to the same environmental factor. Sex-specific effects of prenatal BPA exposure on the susceptibility of AD deserve further investigation.
Collapse
|
14
|
Kim S, Nam Y, Kim C, Lee H, Hong S, Kim HS, Shin SJ, Park YH, Mai HN, Oh SM, Kim KS, Yoo DH, Chung WK, Chung H, Moon M. Neuroprotective and Anti-Inflammatory Effects of Low-Moderate Dose Ionizing Radiation in Models of Alzheimer's Disease. Int J Mol Sci 2020; 21:E3678. [PMID: 32456197 PMCID: PMC7279400 DOI: 10.3390/ijms21103678] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. The neuropathological features of AD include amyloid-β (Aβ) deposition and hyperphosphorylated tau accumulation. Although several clinical trials have been conducted to identify a cure for AD, no effective drug or treatment has been identified thus far. Recently, the potential use of non-pharmacological interventions to prevent or treat AD has gained attention. Low-dose ionizing radiation (LDIR) is a non-pharmacological intervention which is currently being evaluated in clinical trials for AD patients. However, the mechanisms underlying the therapeutic effects of LDIR therapy have not yet been established. In this study, we examined the effect of LDIR on Aβ accumulation and Aβ-mediated pathology. To investigate the short-term effects of low-moderate dose ionizing radiation (LMDIR), a total of 9 Gy (1.8 Gy per fraction for five times) were radiated to 4-month-old 5XFAD mice, an Aβ-overexpressing transgenic mouse model of AD, and then sacrificed at 4 days after last exposure to LMDIR. Comparing sham-exposed and LMDIR-exposed 5XFAD mice indicated that short-term exposure to LMDIR did not affect Aβ accumulation in the brain, but significantly ameliorated synaptic degeneration, neuronal loss, and neuroinflammation in the hippocampal formation and cerebral cortex. In addition, a direct neuroprotective effect was confirmed in SH-SY5Y neuronal cells treated with Aβ1-42 (2 μM) after single irradiation (1 Gy). In BV-2 microglial cells exposed to Aβ and/or LMDIR, LMDIR therapy significantly inhibited the production of pro-inflammatory molecules and activation of the nuclear factor-kappa B (NF-κB) pathway. These results indicate that LMDIR directly ameliorated neurodegeneration and neuroinflammation in vivo and in vitro. Collectively, our findings suggest that the therapeutic benefits of LMDIR in AD may be mediated by its neuroprotective and anti-inflammatory effects.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Chanyang Kim
- Department of Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea;
| | - Hyewon Lee
- Department of Occupational Therapy, Konyang University, Daejeon 35365, Korea; (H.L.); (D.-H.Y.)
| | - Seojin Hong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Han Ngoc Mai
- Department of Radiation Oncology, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea;
| | - Sang-Muk Oh
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| | - Kyoung Soo Kim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University School of Medicine, Seoul 02447, Korea;
| | - Doo-Han Yoo
- Department of Occupational Therapy, Konyang University, Daejeon 35365, Korea; (H.L.); (D.-H.Y.)
| | - Weon Kuu Chung
- Department of Radiation Oncology, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea;
| | - Hyunju Chung
- Department of Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea;
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea; (S.K.); (Y.N.); (S.H.); (H.S.K.); (S.J.S.); (Y.H.P.); (S.-M.O.)
| |
Collapse
|
15
|
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. The AD pathophysiology entails chronic inflammation involving innate immune cells including microglia, astrocytes, and other peripheral blood cells. Inflammatory mediators such as cytokines and complements are also linked to AD pathogenesis. Despite increasing evidence supporting the association between abnormal inflammation and AD, no well-established inflammatory biomarkers are currently available for AD. Since many reports have shown that abnormal inflammation precedes the outbreak of the disease, non-invasive and readily available peripheral inflammatory biomarkers should be considered as possible biomarkers for early diagnosis of AD. In this mini-review, we introduce the peripheral biomarker candidates related to abnormal inflammation in AD and discuss their possible molecular mechanisms. Furthermore, we also summarize the current state of inflammatory biomarker research in clinical practice and molecular diagnostics. We believe this review will provide new insights into biomarker candidates for the early diagnosis of AD with systemic relevance to inflammation during AD pathogenesis.
Collapse
Affiliation(s)
- Jong-Chan Park
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
| | - Sun-Ho Han
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
| | - Inhee Mook-Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
16
|
Hampel H, Caraci F, Cuello AC, Caruso G, Nisticò R, Corbo M, Baldacci F, Toschi N, Garaci F, Chiesa PA, Verdooner SR, Akman-Anderson L, Hernández F, Ávila J, Emanuele E, Valenzuela PL, Lucía A, Watling M, Imbimbo BP, Vergallo A, Lista S. A Path Toward Precision Medicine for Neuroinflammatory Mechanisms in Alzheimer's Disease. Front Immunol 2020; 11:456. [PMID: 32296418 PMCID: PMC7137904 DOI: 10.3389/fimmu.2020.00456] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 02/27/2020] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation commences decades before Alzheimer's disease (AD) clinical onset and represents one of the earliest pathomechanistic alterations throughout the AD continuum. Large-scale genome-wide association studies point out several genetic variants—TREM2, CD33, PILRA, CR1, MS4A, CLU, ABCA7, EPHA1, and HLA-DRB5-HLA-DRB1—potentially linked to neuroinflammation. Most of these genes are involved in proinflammatory intracellular signaling, cytokines/interleukins/cell turnover, synaptic activity, lipid metabolism, and vesicle trafficking. Proteomic studies indicate that a plethora of interconnected aberrant molecular pathways, set off and perpetuated by TNF-α, TGF-β, IL-1β, and the receptor protein TREM2, are involved in neuroinflammation. Microglia and astrocytes are key cellular drivers and regulators of neuroinflammation. Under physiological conditions, they are important for neurotransmission and synaptic homeostasis. In AD, there is a turning point throughout its pathophysiological evolution where glial cells sustain an overexpressed inflammatory response that synergizes with amyloid-β and tau accumulation, and drives synaptotoxicity and neurodegeneration in a self-reinforcing manner. Despite a strong therapeutic rationale, previous clinical trials investigating compounds with anti-inflammatory properties, including non-steroidal anti-inflammatory drugs (NSAIDs), did not achieve primary efficacy endpoints. It is conceivable that study design issues, including the lack of diagnostic accuracy and biomarkers for target population identification and proof of mechanism, may partially explain the negative outcomes. However, a recent meta-analysis indicates a potential biological effect of NSAIDs. In this regard, candidate fluid biomarkers of neuroinflammation are under analytical/clinical validation, i.e., TREM2, IL-1β, MCP-1, IL-6, TNF-α receptor complexes, TGF-β, and YKL-40. PET radio-ligands are investigated to accomplish in vivo and longitudinal regional exploration of neuroinflammation. Biomarkers tracking different molecular pathways (body fluid matrixes) along with brain neuroinflammatory endophenotypes (neuroimaging markers), can untangle temporal–spatial dynamics between neuroinflammation and other AD pathophysiological mechanisms. Robust biomarker–drug codevelopment pipelines are expected to enrich large-scale clinical trials testing new-generation compounds active, directly or indirectly, on neuroinflammatory targets and displaying putative disease-modifying effects: novel NSAIDs, AL002 (anti-TREM2 antibody), anti-Aβ protofibrils (BAN2401), and AL003 (anti-CD33 antibody). As a next step, taking advantage of breakthrough and multimodal techniques coupled with a systems biology approach is the path to pursue for developing individualized therapeutic strategies targeting neuroinflammation under the framework of precision medicine.
Collapse
Affiliation(s)
- Harald Hampel
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy.,Oasi Research Institute-IRCCS, Troina, Italy
| | - A Claudio Cuello
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | | - Robert Nisticò
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, Italy.,School of Pharmacy, Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Filippo Baldacci
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,Department of Radiology, "Athinoula A. Martinos" Center for Biomedical Imaging, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,Casa di Cura "San Raffaele Cassino", Cassino, Italy
| | - Patrizia A Chiesa
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | | | | | - Félix Hernández
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jesús Ávila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | | | | | - Alejandro Lucía
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain.,Research Institute of the Hospital 12 de Octubre ("imas"), Madrid, Spain.,Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | | | - Bruno P Imbimbo
- Research & Development Department, Chiesi Farmaceutici, Parma, Italy
| | - Andrea Vergallo
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| |
Collapse
|
17
|
Zhou M, Xu R, Kaelber DC, Gurney ME. Tumor Necrosis Factor (TNF) blocking agents are associated with lower risk for Alzheimer's disease in patients with rheumatoid arthritis and psoriasis. PLoS One 2020; 15:e0229819. [PMID: 32203525 PMCID: PMC7089534 DOI: 10.1371/journal.pone.0229819] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/16/2020] [Indexed: 01/03/2023] Open
Abstract
This large, retrospective case-control study of electronic health records from 56 million unique adult patients examined whether or not treatment with a Tumor Necrosis Factor (TNF) blocking agent is associated with lower risk for Alzheimer’s disease (AD) in patients with rheumatoid arthritis (RA), psoriasis, and other inflammatory diseases which are mediated in part by TNF and for which a TNF blocker is an approved treatment. The analysis compared the diagnosis of AD as an outcome measure in patients receiving at least one prescription for a TNF blocking agent (etanercept, adalimumab, and infliximab) or for methotrexate. Adjusted odds ratios (AORs) were estimated using the Cochran-Mantel-Haenszel (CMH) method and presented with 95% confidence intervals (CIs) and p-values. RA was associated with a higher risk for AD (Adjusted Odds Ratio (AOR) = 2.06, 95% Confidence Interval: (2.02–2.10), P-value <0.0001) as did psoriasis (AOR = 1.37 (1.31–1.42), P <0.0001), ankylosing spondylitis (AOR = 1.57 (1.39–1.77), P <0.0001), inflammatory bowel disease (AOR = 2.46 (2.33–2.59), P < 0.0001), ulcerative colitis (AOR = 1.82 (1.74–1.91), P <0.0001), and Crohn’s disease (AOR = 2.33 (2.22–2.43), P <0.0001). The risk for AD in patients with RA was lower among patients treated with etanercept (AOR = 0.34 (0.25–0.47), P <0.0001), adalimumab (AOR = 0.28 (0.19–0.39), P < 0.0001), or infliximab (AOR = 0.52 (0.39–0.69), P <0.0001). Methotrexate was also associated with a lower risk for AD (AOR = 0.64 (0.61–0.68), P <0.0001), while lower risk was found in patients with a prescription history for both a TNF blocker and methotrexate. Etanercept and adalimumab also were associated with lower risk for AD in patients with psoriasis: AOR = 0.47 (0.30–0.73 and 0.41 (0.20–0.76), respectively. There was no effect of gender or race, while younger patients showed greater benefit from a TNF blocker than did older patients. This study identifies a subset of patients in whom systemic inflammation contributes to risk for AD through a pathological mechanism involving TNF and who therefore may benefit from treatment with a TNF blocking agent.
Collapse
Affiliation(s)
- Mengshi Zhou
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
- * E-mail: (R.X.); (M.E.G.)
| | - David C. Kaelber
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
- Departments of Internal Medicine and Pediatrics and the Center for Clinical Informatics Research and Education, The MetroHealth System, Cleveland, OH, United States of America
| | - Mark E. Gurney
- Tetra Therapeutics, Grand Rapids, MI, United States of America
- * E-mail: (R.X.); (M.E.G.)
| |
Collapse
|
18
|
Pardridge WM. Blood-Brain Barrier and Delivery of Protein and Gene Therapeutics to Brain. Front Aging Neurosci 2020; 11:373. [PMID: 31998120 PMCID: PMC6966240 DOI: 10.3389/fnagi.2019.00373] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/19/2019] [Indexed: 01/02/2023] Open
Abstract
Alzheimer’s disease (AD) and treatment of the brain in aging require the development of new biologic drugs, such as recombinant proteins or gene therapies. Biologics are large molecule therapeutics that do not cross the blood-brain barrier (BBB). BBB drug delivery is the limiting factor in the future development of new therapeutics for the brain. The delivery of recombinant protein or gene medicines to the brain is a binary process: either the brain drug developer re-engineers the biologic with BBB drug delivery technology, or goes forward with brain drug development in the absence of a BBB delivery platform. The presence of BBB delivery technology allows for engineering the therapeutic to enable entry into the brain across the BBB from blood. Brain drug development may still take place in the absence of BBB delivery technology, but with a reliance on approaches that have rarely led to FDA approval, e.g., CSF injection, stem cells, small molecules, and others. CSF injection of drug is the most widely practiced approach to brain delivery that bypasses the BBB. However, drug injection into the CSF results in limited drug penetration to the brain parenchyma, owing to the rapid export of CSF from the brain to blood. A CSF injection of a drug is equivalent to a slow intravenous (IV) infusion of the pharmaceutical. Given the profound effect the existence of the BBB has on brain drug development, future drug or gene development for the brain will be accelerated by future advances in BBB delivery technology in parallel with new drug discovery.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
19
|
Phosphorylation Signaling in APP Processing in Alzheimer's Disease. Int J Mol Sci 2019; 21:ijms21010209. [PMID: 31892243 PMCID: PMC6981488 DOI: 10.3390/ijms21010209] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/13/2022] Open
Abstract
The abnormal accumulation of amyloid-β (Aβ) in the central nervous system is a hallmark of Alzheimer’s disease (AD). The regulation of the processing of the single- transmembrane amyloid precursor protein (APP) plays an important role in the generation of Aβ in the brain. The phosphorylation of APP and key enzymes involved in the proteolytic processing of APP has been demonstrated to be critical for modulating the generation of Aβ by either altering the subcellular localization of APP or changing the enzymatic activities of the secretases responsible for APP processing. In addition, the phosphorylation may also have an impact on the physiological function of these proteins. In this review, we summarize the kinases and signaling pathways that may participate in regulating the phosphorylation of APP and secretases and how this further affects the function and processing of APP and Aβ pathology. We also discuss the potential of approaches that modulate these phosphorylation-signaling pathways or kinases as interventions for AD pathology.
Collapse
|
20
|
Pillai JA, Maxwell S, Bena J, Bekris LM, Rao SM, Chance M, Lamb BT, Leverenz JB. Key inflammatory pathway activations in the MCI stage of Alzheimer's disease. Ann Clin Transl Neurol 2019; 6:1248-1262. [PMID: 31353852 PMCID: PMC6649519 DOI: 10.1002/acn3.50827] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/30/2019] [Accepted: 05/31/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To determine the key inflammatory pathways that are activated in the peripheral and CNS compartments at the mild cognitive impairment (MCI) stage of Alzheimer's disease (AD). METHODS A cross-sectional study of patients with clinical and biomarker characteristics consistent with MCI-AD in a discovery cohort, with replication in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Inflammatory analytes were measured in the CSF and plasma with the same validated multiplex analyte platform in both cohorts and correlated with AD biomarkers (CSF Aβ42, total tau (t-tau), phosphorylated tau (p-tau) to identify key inflammatory pathway activations. The pathways were additionally validated by evaluating genes related to all analytes in coexpression networks of brain tissue transcriptome from an autopsy confirmed AD cohort to interrogate if the same pathway activations were conserved in the brain tissue gene modules. RESULTS Analytes of the tumor necrosis factor (TNF) signaling pathway (KEGG ID:4668) in the CSF and plasma best correlated with CSF t-tau and p-tau levels, and analytes of the complement and coagulation pathway (KEGG ID:4610) best correlated with CSF Aβ42 levels. The top inflammatory signaling pathways of significance were conserved in the peripheral and the CNS compartments. They were also confirmed to be enriched in AD brain transcriptome gene clusters. INTERPRETATION A cell-protective rather than a proinflammatory analyte profile predominates in the CSF in relation to neurodegeneration markers among MCI-AD patients. Analytes from the TNF signaling and the complement and coagulation pathways are relevant in evaluating disease severity at the MCI stage of AD.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhio44195
- Neurological InstituteCleveland ClinicClevelandOhio44195
- Department of NeurologyCleveland ClinicClevelandOhio44195
| | - Sean Maxwell
- Center for Proteomics and BioinformaticCase Western Reserve UniversityClevelandOhio44106
- Department of NutritionCase Western Reserve UniversityClevelandOhio44106
| | - James Bena
- Quantitative Health SciencesCleveland ClinicClevelandOhio44195
| | - Lynn M. Bekris
- Genomic Medicine InstituteCleveland ClinicClevelandOhio44195
| | - Stephen M. Rao
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhio44195
- Neurological InstituteCleveland ClinicClevelandOhio44195
| | - Mark Chance
- Center for Proteomics and BioinformaticCase Western Reserve UniversityClevelandOhio44106
- Department of NutritionCase Western Reserve UniversityClevelandOhio44106
| | - Bruce T. Lamb
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndiana46202
| | - James B. Leverenz
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhio44195
- Neurological InstituteCleveland ClinicClevelandOhio44195
- Department of NeurologyCleveland ClinicClevelandOhio44195
| | | |
Collapse
|
21
|
Ortí-Casañ N, Wu Y, Naudé PJW, De Deyn PP, Zuhorn IS, Eisel ULM. Targeting TNFR2 as a Novel Therapeutic Strategy for Alzheimer's Disease. Front Neurosci 2019; 13:49. [PMID: 30778285 PMCID: PMC6369349 DOI: 10.3389/fnins.2019.00049] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/18/2019] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia. Accumulating experimental evidence shows the important linkage between tumor necrosis factor-α (TNF) and AD, but the exact role of TNF in AD is still not completely understood. Although TNF-inhibitors are successfully used for treating several diseases, total inhibition of TNF can cause side effects, particularly in neurological diseases. This is attributed to the opposing roles of the two TNF receptors. TNF receptor 1 (TNFR1) predominantly mediates inflammatory and pro-apoptotic signaling pathways, whereas TNF receptor 2 (TNFR2) is neuroprotective and promotes tissue regeneration. Therefore, the specific activation of TNFR2 signaling, either by directly targeting TNFR2 via TNFR2 agonists or by blocking TNFR1 signaling with TNFR1-selective antagonists, seems a promising strategy for AD therapy. This mini-review discusses the involvement of TNFR2 and its signaling pathway in AD and outlines its potential application as therapeutic target. A better understanding of the function of TNFR2 may lead to the development of a treatment for AD.
Collapse
Affiliation(s)
- Natalia Ortí-Casañ
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Yingying Wu
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Petrus J W Naudé
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands.,Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Peter P De Deyn
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Inge S Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| |
Collapse
|
22
|
Ravichandran S, Michelucci A, Del Sol A. Integrative Computational Network Analysis Reveals Site-Specific Mediators of Inflammation in Alzheimer's Disease. Front Physiol 2018; 9:154. [PMID: 29551980 PMCID: PMC5840953 DOI: 10.3389/fphys.2018.00154] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/14/2018] [Indexed: 12/02/2022] Open
Abstract
Alzheimer's disease (AD) is a major neurodegenerative disease and is one of the most common cause of dementia in older adults. Among several factors, neuroinflammation is known to play a critical role in the pathogenesis of chronic neurodegenerative diseases. In particular, studies of brains affected by AD show a clear involvement of several inflammatory pathways. Furthermore, depending on the brain regions affected by the disease, the nature and the effect of inflammation can vary. Here, in order to shed more light on distinct and common features of inflammation in different brain regions affected by AD, we employed a computational approach to analyze gene expression data of six site-specific neuronal populations from AD patients. Our network based computational approach is driven by the concept that a sustained inflammatory environment could result in neurotoxicity leading to the disease. Thus, our method aims to infer intracellular signaling pathways/networks that are likely to be constantly activated or inhibited due to persistent inflammatory conditions. The computational analysis identified several inflammatory mediators, such as tumor necrosis factor alpha (TNF-a)-associated pathway, as key upstream receptors/ligands that are likely to transmit sustained inflammatory signals. Further, the analysis revealed that several inflammatory mediators were mainly region specific with few commonalities across different brain regions. Taken together, our results show that our integrative approach aids identification of inflammation-related signaling pathways that could be responsible for the onset or the progression of AD and can be applied to study other neurodegenerative diseases. Furthermore, such computational approaches can enable the translation of clinical omics data toward the development of novel therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Srikanth Ravichandran
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Alessandro Michelucci
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg.,Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
23
|
Decourt B, Lahiri DK, Sabbagh MN. Targeting Tumor Necrosis Factor Alpha for Alzheimer's Disease. Curr Alzheimer Res 2017; 14:412-425. [PMID: 27697064 DOI: 10.2174/1567205013666160930110551] [Citation(s) in RCA: 254] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/10/2016] [Accepted: 09/22/2016] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD) affects an estimated 44 million individuals worldwide, yet no therapeutic intervention is available to stop the progression of the dementia. Neuropathological hallmarks of AD are extracellular deposits of amyloid beta (Aβ) peptides assembled in plaques, intraneuronal accumulation of hyperphosphorylated tau protein forming tangles, and chronic inflammation. A pivotal molecule in inflammation is the pro-inflammatory cytokine TNF-α. Several lines of evidence using genetic and pharmacological manipulations indicate that TNF-α signaling exacerbates both Aβ and tau pathologies in vivo. Interestingly, preventive and intervention anti-inflammatory strategies demonstrated a reduction in brain pathology and an amelioration of cognitive function in rodent models of AD. Phase I and IIa clinical trials suggest that TNF-α inhibitors might slow down cognitive decline and improve daily activities in AD patients. In the present review, we summarize the evidence pointing towards a beneficial role of anti-TNF-α therapies to prevent or slow the progression of AD. We also present possible physical and pharmacological interventions to modulate TNF-α signaling in AD subjects along with their limitations.
Collapse
Affiliation(s)
- Boris Decourt
- Banner Sun Health Research Institute, 10515 W. Santa Fe Dr., Sun City AZ 85351, United States
| | - Debomoy K Lahiri
- Institute of Psychiatry Research, Department of Psychiatry, School of Medicine, Indiana University-Purdue University, Indianapolis, IN, United States
| | - Marwan N Sabbagh
- Alzheimer's and Memory Disorders Division, Barrow Neurological Institute, 240 West Thomas, Ste 301, Phoenix, AZ 85013, United States
| |
Collapse
|
24
|
The adaptive immune system restrains Alzheimer's disease pathogenesis by modulating microglial function. Proc Natl Acad Sci U S A 2016; 113:E1316-25. [PMID: 26884167 DOI: 10.1073/pnas.1525466113] [Citation(s) in RCA: 306] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The innate immune system is strongly implicated in the pathogenesis of Alzheimer's disease (AD). In contrast, the role of adaptive immunity in AD remains largely unknown. However, numerous clinical trials are testing vaccination strategies for AD, suggesting that T and B cells play a pivotal role in this disease. To test the hypothesis that adaptive immunity influences AD pathogenesis, we generated an immune-deficient AD mouse model that lacks T, B, and natural killer (NK) cells. The resulting "Rag-5xfAD" mice exhibit a greater than twofold increase in β-amyloid (Aβ) pathology. Gene expression analysis of the brain implicates altered innate and adaptive immune pathways, including changes in cytokine/chemokine signaling and decreased Ig-mediated processes. Neuroinflammation is also greatly exacerbated in Rag-5xfAD mice as indicated by a shift in microglial phenotype, increased cytokine production, and reduced phagocytic capacity. In contrast, immune-intact 5xfAD mice exhibit elevated levels of nonamyloid reactive IgGs in association with microglia, and treatment of Rag-5xfAD mice or microglial cells with preimmune IgG enhances Aβ clearance. Last, we performed bone marrow transplantation studies in Rag-5xfAD mice, revealing that replacement of these missing adaptive immune populations can dramatically reduce AD pathology. Taken together, these data strongly suggest that adaptive immune cell populations play an important role in restraining AD pathology. In contrast, depletion of B cells and their appropriate activation by T cells leads to a loss of adaptive-innate immunity cross talk and accelerated disease progression.
Collapse
|
25
|
Gabbita SP, Johnson MF, Kobritz N, Eslami P, Poteshkina A, Varadarajan S, Turman J, Zemlan F, Harris-White ME. Oral TNFα Modulation Alters Neutrophil Infiltration, Improves Cognition and Diminishes Tau and Amyloid Pathology in the 3xTgAD Mouse Model. PLoS One 2015; 10:e0137305. [PMID: 26436670 PMCID: PMC4593589 DOI: 10.1371/journal.pone.0137305] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 08/15/2015] [Indexed: 01/22/2023] Open
Abstract
Cytokines such as TNFα can polarize microglia/macrophages into different neuroinflammatory types. Skewing of the phenotype towards a cytotoxic state is thought to impair phagocytosis and has been described in Alzheimer’s Disease (AD). Neuroinflammation can be perpetuated by a cycle of increasing cytokine production and maintenance of a polarized activation state that contributes to AD progression. In this study, 3xTgAD mice, age 6 months, were treated orally with 3 doses of the TNFα modulating compound isoindolin-1,3 dithione (IDT) for 10 months. We demonstrate that IDT is a TNFα modulating compound both in vitro and in vivo. Following long-term IDT administration, mice were assessed for learning & memory and tissue and serum were collected for analysis. Results demonstrate that IDT is safe for long-term treatment and significantly improves learning and memory in the 3xTgAD mouse model. IDT significantly reduced paired helical filament tau and fibrillar amyloid accumulation. Flow cytometry of brain cell populations revealed that IDT increased the infiltrating neutrophil population while reducing TNFα expression in this population. IDT is a safe and effective TNFα and innate immune system modulator. Thus small molecule, orally bioavailable modulators are promising therapeutics for Alzheimer’s disease.
Collapse
Affiliation(s)
| | - Ming F. Johnson
- Veterans Administration-Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
| | - Naomi Kobritz
- Veterans Administration-Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
| | - Pirooz Eslami
- Veterans Administration-Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
| | - Aleksandra Poteshkina
- Veterans Administration-Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
| | - Sridhar Varadarajan
- University of North Carolina Wilmington, Department of Chemistry and Biochemistry, Wilmington, North Carolina, United States of America
| | - John Turman
- University of North Carolina Wilmington, Department of Chemistry and Biochemistry, Wilmington, North Carolina, United States of America
| | - Frank Zemlan
- P2D Bioscience, Inc., Cincinnati, Ohio, United States of America
| | - Marni E. Harris-White
- Veterans Administration-Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
- University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
26
|
Reale M, Di Nicola M, Velluto L, D'Angelo C, Costantini E, Lahiri DK, Kamal MA, Yu QS, Greig NH. Selective acetyl- and butyrylcholinesterase inhibitors reduce amyloid-β ex vivo activation of peripheral chemo-cytokines from Alzheimer's disease subjects: exploring the cholinergic anti-inflammatory pathway. Curr Alzheimer Res 2015; 11:608-22. [PMID: 24359497 DOI: 10.2174/1567205010666131212113218] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/28/2013] [Accepted: 11/02/2013] [Indexed: 12/22/2022]
Abstract
Increasing evidence suggests that elevated production and/or reduced clearance of amyloid-β peptide (Aβ) drives the early pathogenesis of Alzheimer's disease (AD). Aβ soluble oligomers trigger a neurotoxic cascade that leads to neuronal dysfunction, neurodegeneration and, ultimately, clinical dementia. Inflammation, both within brain and systemically, together with a deficiency in the neurotransmitter acetylcholine (ACh) that underpinned the development of anticholinesterases for AD symptomatic treatment, are invariable hallmarks of the disease. The inter-relation between Aβ, inflammation and cholinergic signaling is complex, with each feeding back onto the others to drive disease progression. To elucidate these interactions plasma samples and peripheral blood mononuclear cells (PBMCs) were evaluated from healthy controls (HC) and AD patients. Plasma levels of acetylcholinesterase (AChE), butyrylcholinesterase (BuChE) and Aβ were significantly elevated in AD vs. HC subjects, and ACh showed a trend towards reduced levels. Aβ challenge of PBMCs induced a greater release of inflammatory cytokines interleukin-1β (IL-1β), monocyte chemotactic protein-1 (MCP-1) and tumor necrosis factor-alpha (TNF-α) from AD vs. HC subjects, with IL-10 being similarly affected. THP-1 monocytic cells, a cell culture counterpart of PBMCs and brain microglial cells, responded similarly to Aβ as well as to phytohaemagglutinin (PHA) challenge, to allow preliminary analysis of the cellular and molecular pathways underpinning Aβ-induced changes in cytokine expression. As amyloid-β precursor protein expression, and hence Aβ, has been reported regulated by particular cytokines and anticholinesterases, the latter were evaluated on Aβ- and PHA-induced chemocytokine expression. Co-incubation with selective AChE/BuChE inhibitors, (-)-phenserine (AChE) and (-)-cymserine analogues (BuChE), mitigated the rise in cytokine levels and suggest that augmentation of the cholinergic anti-inflammatory pathway may prove valuable in AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Nigel H Greig
- Dept. of Experimental and Clinical Sciences, Unit ofImmunodiagnostic and Molecular Pathology, University "G. D'Annunzio", N.P.D., Ed. C, III lev., Via dei Vestini, 31, 66123 Chieti, Italy.
| |
Collapse
|
27
|
Modeling Alzheimer's disease in mouse without mutant protein overexpression: cooperative and independent effects of Aβ and tau. PLoS One 2013; 8:e80706. [PMID: 24278307 PMCID: PMC3835479 DOI: 10.1371/journal.pone.0080706] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 10/15/2013] [Indexed: 01/23/2023] Open
Abstract
Background Alzheimer’s disease (AD), the most common cause of dementia in the elderly, has two pathological hallmarks: Aβ plaques and aggregation of hyperphosphorylated tau (p-tau). Aβ is a cleavage product of Amyloid Precursor Protein (APP). Presenilin 1 (PS1) and presenilin 2 (PS2) are the catalytic subunit of γ-secretase, which cleaves APP and mediates Aβ production. Genetic mutations in APP, PSEN1 or PSEN2 can lead to early onset of familial AD (FAD). Although mutations in the tau encoding gene MAPT leads to a subtype of frontotemporal dementia and these mutations have been used to model AD tauopathy, no MAPT mutations have been found to be associated with AD. Results To model AD pathophysiology in mice without the gross overexpression of mutant transgenes, we created a humanized AD mouse model by crossing the APP and PSEN1 FAD knock-in mice with the htau mice which express wildtype human MAPT genomic DNA on mouse MAPT null background (APP/PS1/htau). The APP/PS1/htau mice displayed mild, age-dependent, Aβ plaques and tau hyperphosphorylation, thus successfully recapitulating the late-onset AD pathological hallmarks. Selected biochemical analyses, including p-tau western blot, γ-secretase activity assay, and Aβ ELISA, were performed to study the interaction between Aβ and p-tau. Subsequent behavioral studies revealed that the APP/PS1/htau mice showed reduced mobility in old ages and exaggerated fear response. Genetic analysis suggested that the fear phenotype is due to a synergic interaction between Aβ and p-tau, and it can be completely abolished by tau deletion. Conclusion The APP/PS1/htau model represents a valuable and disease-relevant late-onset pre-clinical AD animal model because it incorporates human AD genetics without mutant protein overexpression. Analysis of the mice revealed both cooperative and independent effects of Aβ and p-tau.
Collapse
|
28
|
Sama DM, Mohmmad Abdul H, Furman JL, Artiushin IA, Szymkowski DE, Scheff SW, Norris CM. Inhibition of soluble tumor necrosis factor ameliorates synaptic alterations and Ca2+ dysregulation in aged rats. PLoS One 2012; 7:e38170. [PMID: 22666474 PMCID: PMC3362564 DOI: 10.1371/journal.pone.0038170] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 05/01/2012] [Indexed: 12/14/2022] Open
Abstract
The role of tumor necrosis factor α (TNF) in neural function has been investigated extensively in several neurodegenerative conditions, but rarely in brain aging, where cognitive and physiologic changes are milder and more variable. Here, we show that protein levels for TNF receptor 1 (TNFR1) are significantly elevated in the hippocampus relative to TNF receptor 2 (TNFR2) in aged (22 months) but not young adult (6 months) Fischer 344 rats. To determine if altered TNF/TNFR1 interactions contribute to key brain aging biomarkers, aged rats received chronic (4–6 week) intracranial infusions of XPro1595: a soluble dominant negative TNF that preferentially inhibits TNFR1 signaling. Aged rats treated with XPro1595 showed improved Morris Water Maze performance, reduced microglial activation, reduced susceptibility to hippocampal long-term depression, increased protein levels for the GluR1 type glutamate receptor, and lower L-type voltage sensitive Ca2+ channel (VSCC) activity in hippocampal CA1 neurons. The results suggest that diverse functional changes associated with brain aging may arise, in part, from selective alterations in TNF signaling.
Collapse
Affiliation(s)
- Diana M. Sama
- Graduate Center for Gerontology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Hafiz Mohmmad Abdul
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jennifer L. Furman
- Molecular & Biomedical Pharmacology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Irina A. Artiushin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | | | - Stephen W. Scheff
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Christopher M. Norris
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Molecular & Biomedical Pharmacology, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|