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
|
Marcatti M, Tumurbaatar B, Borghi M, Guptarak J, Zhang WR, Krishnan B, Kayed R, Fracassi A, Taglialatela G. Inhibition of Calcineurin with FK506 Reduces Tau Levels and Attenuates Synaptic Impairment Driven by Tau Oligomers in the Hippocampus of Male Mouse Models. Int J Mol Sci 2024; 25:9092. [PMID: 39201779 PMCID: PMC11354963 DOI: 10.3390/ijms25169092] [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: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
Alzheimer's disease (AD) is the most common age-associated neurodegenerative disorder, characterized by progressive cognitive decline, memory impairment, and structural brain changes, primarily involving Aβ plaques and neurofibrillary tangles of hyperphosphorylated tau protein. Recent research highlights the significance of smaller Aβ and Tau oligomeric aggregates (AβO and TauO, respectively) in synaptic dysfunction and disease progression. Calcineurin (CaN), a key calcium/calmodulin-dependent player in regulating synaptic function in the central nervous system (CNS) is implicated in mediating detrimental effects of AβO on synapses and memory function in AD. This study aims to investigate the specific impact of CaN on both exogenous and endogenous TauO through the acute and chronic inhibition of CaN. We previously demonstrated the protective effect against AD of the immunosuppressant CaN inhibitor, FK506, but its influence on TauO remains unclear. In this study, we explored the short-term effects of acute CaN inhibition on TauO phosphorylation and TauO-induced memory deficits and synaptic dysfunction. Mice received FK506 post-TauO intracerebroventricular injection and TauO levels and phosphorylation were assessed, examining their impact on CaN and GSK-3β. The study investigated FK506 preventive/reversal effects on TauO-induced clustering of CaN and GSK-3β. Memory and synaptic function in TauO-injected mice were evaluated with/without FK506. Chronic FK506 treatment in 3xTgAD mice explored its influence on CaN, Aβ, and Tau levels. This study underscores the significant influence of CaN inhibition on TauO and associated AD pathology, suggesting therapeutic potential in targeting CaN for addressing various aspects of AD onset and progression. These findings provide valuable insights for potential interventions in AD, emphasizing the need for further exploration of CaN-targeted strategies.
Collapse
|
3
|
Scaduto P, Marcatti M, Bhatt N, Kayed R, Taglialatela G. Calcineurin inhibition prevents synaptic plasticity deficit induced by brain-derived tau oligomers. Brain Commun 2024; 6:fcae277. [PMID: 39239152 PMCID: PMC11375858 DOI: 10.1093/braincomms/fcae277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/08/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Compelling evidence suggests that cognitive decline in Alzheimer's disease is associated with the accumulation and aggregation of tau protein, with the most toxic aggregates being in the form of oligomers. This underscores the necessity for direct isolation and analysis of brain-derived tau oligomers from patients with Alzheimer's disease, potentially offering novel perspectives into tau toxicity. Alzheimer's brain-derived tau oligomers are potent inhibitors of synaptic plasticity; however, the involved mechanism is still not fully understood. We previously reported a significantly reduced incidence of Alzheimer's disease in ageing humans chronically treated with a Food and Drug Administration-approved calcineurin inhibitor, FK506 (tacrolimus), used as an immunosuppressant after solid organ transplant. Using a combination of electrophysiological and RNA-sequencing techniques, we provide here evidence that FK506 has the potential to block the acute toxic effect of brain-derived tau oligomers on synaptic plasticity, as well as to restore the levels of some key synaptic mRNAs. These results further support FK506 as a promising novel therapeutic strategy for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Pietro Scaduto
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| | - Michela Marcatti
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| | - Nemil Bhatt
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| | - Rakez Kayed
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| | - Giulio Taglialatela
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, TX 77555, USA
| |
Collapse
|
4
|
Del Rosario Hernandez T, Joshi NR, Gore SV, Kreiling JA, Creton R. Combining supervised and unsupervised analyses to quantify behavioral phenotypes and validate therapeutic efficacy in a triple transgenic mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597924. [PMID: 38895269 PMCID: PMC11185760 DOI: 10.1101/2024.06.07.597924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Behavioral testing is an essential tool for evaluating cognitive function and dysfunction in preclinical research models. This is of special importance in the study of neurological disorders such as Alzheimer's disease. However, the reproducibility of classic behavioral assays is frequently compromised by interstudy variation, leading to ambiguous conclusions about the behavioral markers characterizing the disease. Here, we identify age- and genotype-driven differences between 3xTg-AD and non-transgenic control mice using a low-cost, highly customizable behavioral assay that requires little human intervention. Through behavioral phenotyping combining both supervised and unsupervised behavioral classification methods, we are able to validate the preventative effects of the immunosuppressant cyclosporine A in a rodent model of Alzheimer's disease, as well as the partially ameliorating effects of candidate drugs nebivolol and cabozantinib.
Collapse
Affiliation(s)
- Thais Del Rosario Hernandez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States
| | - Narendra R Joshi
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States
| | - Sayali V Gore
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States
| | - Jill A Kreiling
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States
| | - Robbert Creton
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States
| |
Collapse
|
5
|
Niazi SK, Magoola M, Mariam Z. Innovative Therapeutic Strategies in Alzheimer's Disease: A Synergistic Approach to Neurodegenerative Disorders. Pharmaceuticals (Basel) 2024; 17:741. [PMID: 38931409 PMCID: PMC11206655 DOI: 10.3390/ph17060741] [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: 02/29/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Alzheimer's disease (AD) remains a significant challenge in the field of neurodegenerative disorders, even nearly a century after its discovery, due to the elusive nature of its causes. The development of drugs that target multiple aspects of the disease has emerged as a promising strategy to address the complexities of AD and related conditions. The immune system's role, particularly in AD, has gained considerable interest, with nanobodies representing a new frontier in biomedical research. Advances in targeting antibodies against amyloid-β (Aβ) and using messenger RNA for genetic translation have revolutionized the production of antibodies and drug development, opening new possibilities for treatment. Despite these advancements, conventional therapies for AD, such as Cognex, Exelon, Razadyne, and Aricept, often have limited long-term effectiveness, underscoring the need for innovative solutions. This necessity has led to the incorporation advanced technologies like artificial intelligence and machine learning into the drug discovery process for neurodegenerative diseases. These technologies help identify therapeutic targets and optimize lead compounds, offering a more effective approach to addressing the challenges of AD and similar conditions.
Collapse
Affiliation(s)
| | | | - Zamara Mariam
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 5FB, UK
| |
Collapse
|
6
|
Koster KP, Fyke Z, Nguyen TTA, Niqula A, Noriega-González LY, Woolfrey KM, Dell’Acqua ML, Cologna SM, Yoshii A. Akap5 links synaptic dysfunction to neuroinflammatory signaling in a mouse model of infantile neuronal ceroid lipofuscinosis. Front Synaptic Neurosci 2024; 16:1384625. [PMID: 38798824 PMCID: PMC11116793 DOI: 10.3389/fnsyn.2024.1384625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Palmitoylation and depalmitoylation represent dichotomic processes by which a labile posttranslational lipid modification regulates protein trafficking and degradation. The depalmitoylating enzyme, palmitoyl-protein thioesterase 1 (PPT1), is associated with the devastating pediatric neurodegenerative condition, infantile neuronal ceroid lipofuscinosis (CLN1). CLN1 is characterized by the accumulation of autofluorescent lysosomal storage material (AFSM) in neurons and robust neuroinflammation. Converging lines of evidence suggest that in addition to cellular waste accumulation, the symptomology of CLN1 corresponds with disruption of synaptic processes. Indeed, loss of Ppt1 function in cortical neurons dysregulates the synaptic incorporation of the GluA1 AMPA receptor (AMPAR) subunit during a type of synaptic plasticity called synaptic scaling. However, the mechanisms causing this aberration are unknown. Here, we used the Ppt1-/- mouse model (both sexes) to further investigate how Ppt1 regulates synaptic plasticity and how its disruption affects downstream signaling pathways. To this end, we performed a palmitoyl-proteomic screen, which provoked the discovery that Akap5 is excessively palmitoylated at Ppt1-/- synapses. Extending our previous data, in vivo induction of synaptic scaling, which is regulated by Akap5, caused an excessive upregulation of GluA1 in Ppt1-/- mice. This synaptic change was associated with exacerbated disease pathology. Furthermore, the Akap5- and inflammation-associated transcriptional regulator, nuclear factor of activated T cells (NFAT), was sensitized in Ppt1-/- cortical neurons. Suppressing the upstream regulator of NFAT activation, calcineurin, with the FDA-approved therapeutic FK506 (Tacrolimus) modestly improved neuroinflammation in Ppt1-/- mice. These findings indicate that the absence of depalmitoylation stifles synaptic protein trafficking and contributes to neuroinflammation via an Akap5-associated mechanism.
Collapse
Affiliation(s)
- Kevin P. Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zach Fyke
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Thu T. A. Nguyen
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Amanda Niqula
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Kevin M. Woolfrey
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Stephanie M. Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Akira Yoshii
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, United States
- Department of Neurology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
7
|
Edison P. Astroglial activation: Current concepts and future directions. Alzheimers Dement 2024; 20:3034-3053. [PMID: 38305570 PMCID: PMC11032537 DOI: 10.1002/alz.13678] [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: 08/24/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
Astrocytes are abundantly and ubiquitously expressed cell types with diverse functions throughout the central nervous system. Astrocytes show remarkable plasticity and exhibit morphological, molecular, and functional remodeling in response to injury, disease, or infection of the central nervous system, as evident in neurodegenerative diseases. Astroglial mediated inflammation plays a prominent role in the pathogenesis of neurodegenerative diseases. This review focus on the role of astrocytes as essential players in neuroinflammation and discuss their morphological and functional heterogeneity in the normal central nervous system and explore the spatial and temporal variations in astroglial phenotypes observed under different disease conditions. This review discusses the intimate relationship of astrocytes to pathological hallmarks of neurodegenerative diseases. Finally, this review considers the putative therapeutic strategies that can be deployed to modulate the astroglial functions in neurodegenerative diseases. HIGHLIGHTS: Astroglia mediated neuroinflammation plays a key role in the pathogenesis of neurodegenerative diseases. Activated astrocytes exhibit diverse phenotypes in a region-specific manner in brain and interact with β-amyloid, tau, and α-synuclein species as well as with microglia and neuronal circuits. Activated astrocytes are likely to influence the trajectory of disease progression of neurodegenerative diseases, as determined by the stage of disease, individual susceptibility, and state of astroglial priming. Modulation of astroglial activation may be a therapeutic strategy at various stages in the trajectory of neurodegenerative diseases to modify the disease course.
Collapse
Affiliation(s)
- Paul Edison
- Division of NeurologyDepartment of Brain SciencesFaculty of Medicine, Imperial College LondonLondonUK
- Division of Psychological medicine and clinical neurosciencesSchool of Medicine, Cardiff UniversityWalesUK
| |
Collapse
|
8
|
Kraner SD, Sompol P, Prateeptrang S, Promkan M, Hongthong S, Thongsopha N, Nelson PT, Norris CM. Development of a monoclonal antibody specific for a calpain-generated ∆48 kDa calcineurin fragment, a marker of distressed astrocytes. J Neurosci Methods 2024; 402:110012. [PMID: 37984591 PMCID: PMC10841921 DOI: 10.1016/j.jneumeth.2023.110012] [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: 09/14/2023] [Revised: 10/23/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Calcineurin (CN) is a Ca2+/calmodulin-dependent protein phosphatase. In healthy tissue, CN exists mainly as a full-length (∼60 kDa) highly-regulated protein phosphatase involved in essential cellular functions. However, in diseased or injured tissue, CN is proteolytically converted to a constitutively active fragment that has been causatively-linked to numerous pathophysiologic processes. These calpain-cleaved CN fragments (∆CN) appear at high levels in human brain at early stages of cognitive decline associated with Alzheimer's disease (AD). NEW METHOD We developed a monoclonal antibody to ∆CN, using an immunizing peptide corresponding to the C-terminal end of the ∆CN fragment. RESULTS We obtained a mouse monoclonal antibody, designated 26A6, that selectively detects ∆CN in Western analysis of calpain-cleaved recombinant human CN. Using this antibody, we screened both pathological and normal human brain sections provided by the University of Kentucky's Alzheimer's Disease Research Center. 26A6 showed low reactivity towards normal brain tissue, but detected astrocytes both surrounding AD amyloid plaques and throughout AD brain tissue. In brain tissue with infarcts, there was considerable concentration of 26A6-positive astrocytes within/around infarcts, suggesting a link with anoxic/ischemia pathways. COMPARISON WITH EXISTING METHOD The results obtained with the new monoclonal are similar to those obtained with a polyclonal we had previously developed. However, the monoclonal is an abundant tool available to the dementia research community. CONCLUSIONS The new monoclonal 26A6 antibody is highly selective for the ∆CN proteolytic fragment and labels a subset of astrocytes, and could be a useful tool for marking insidious brain pathology and identifying novel astrocyte phenotypes.
Collapse
Affiliation(s)
| | - Pradoldej Sompol
- Sanders Brown Center on Aging, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Siriyagon Prateeptrang
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Moltira Promkan
- Sanders Brown Center on Aging, USA; Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Suthida Hongthong
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Napasorn Thongsopha
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Peter T Nelson
- Sanders Brown Center on Aging, USA; Department of Pathology, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher M Norris
- Sanders Brown Center on Aging, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
9
|
Silva JD, Jupiter DC, Taglialatela G. Reduced Prevalence of Parkinson's Disease in Patients Prescribed Calcineurin Inhibitors. JOURNAL OF PARKINSON'S DISEASE 2024; 14:533-543. [PMID: 38427501 DOI: 10.3233/jpd-230313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Preclinical evidence suggests calcineurin inhibitors (CNIs) combat α-synuclein-induced neuronal dysfunction and motor impairments. However, whether CNIs prevent or treat Parkinson's disease (PD) in humans has never been investigated. Objective We seek to ascertain if prescription of CNIs is linked to a decreased prevalence of PD in a varied patient population and to glimpse into the mechanism(s) and target site through which CNIs might decrease PD prevalence. Methods We analyzed electronic health records (EHRs) from patients prescribed the brain penetrant CNI tacrolimus (TAC), the peripherally restricted CNI cyclosporine (CySp), or the non-CNI sirolimus (SIR). For comparison, EHRs from a diverse population from the same network served as a general population-like control. After propensity-score matching, prevalence, odds, and hazards of PD diagnoses among these cohorts were compared. Results Patients prescribed CNIs have decreased odds of PD diagnosis compared to the general population-like control, while patients prescribed SIR do not. Notably, patients prescribed TAC have a decreased prevalence of PD compared to patients prescribed SIR or CySp. Conclusions Our results suggest CNIs, especially those acting within the brain, may prevent PD. The reduced prevalence of PD in patients prescribed TAC, compared to patients prescribed SIR, suggests that mechanisms of calcineurin inhibition- other than immunosuppression, which is common to both drugs- are driving the reduction. Therefore, CNIs may provide a promising therapeutic approach for PD.
Collapse
Affiliation(s)
- Jacqueline D Silva
- Department of Neurology, Mitchell Center for Neurodegenerative Disorders, University of Texas Medical Branch, Galveston, TX, USA
- Pharmacology and Toxicology Graduate Program, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel C Jupiter
- Department of Biostatistics and Data Science, University of Texas Medical Branch, Galveston, TX, USA
- Department of Orthopaedics and Rehabilitation, University of Texas Medical Branch, Galveston, TX, USA
| | - Giulio Taglialatela
- Department of Neurology, Mitchell Center for Neurodegenerative Disorders, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
10
|
Mackiewicz J, Lisek M, Boczek T. Targeting CaN/NFAT in Alzheimer's brain degeneration. Front Immunol 2023; 14:1281882. [PMID: 38077352 PMCID: PMC10701682 DOI: 10.3389/fimmu.2023.1281882] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive functions. While the exact causes of this debilitating disorder remain elusive, numerous investigations have characterized its two core pathologies: the presence of β-amyloid plaques and tau tangles. Additionally, multiple studies of postmortem brain tissue, as well as results from AD preclinical models, have consistently demonstrated the presence of a sustained inflammatory response. As the persistent immune response is associated with neurodegeneration, it became clear that it may also exacerbate other AD pathologies, providing a link between the initial deposition of β-amyloid plaques and the later development of neurofibrillary tangles. Initially discovered in T cells, the nuclear factor of activated T-cells (NFAT) is one of the main transcription factors driving the expression of inflammatory genes and thus regulating immune responses. NFAT-dependent production of inflammatory mediators is controlled by Ca2+-dependent protein phosphatase calcineurin (CaN), which dephosphorylates NFAT and promotes its transcriptional activity. A substantial body of evidence has demonstrated that aberrant CaN/NFAT signaling is linked to several pathologies observed in AD, including neuronal apoptosis, synaptic deficits, and glia activation. In view of this, the role of NFAT isoforms in AD has been linked to disease progression at different stages, some of which are paralleled to diminished cognitive status. The use of classical inhibitors of CaN/NFAT signaling, such as tacrolimus or cyclosporine, or adeno-associated viruses to specifically inhibit astrocytic NFAT activation, has alleviated some symptoms of AD by diminishing β-amyloid neurotoxicity and neuroinflammation. In this article, we discuss the recent findings related to the contribution of CaN/NFAT signaling to the progression of AD and highlight the possible benefits of targeting this pathway in AD treatment.
Collapse
Affiliation(s)
| | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Norris CM. Is It Time to Repurpose Calcineurin Inhibitors for the Treatment of Cognitive Impairment and Dementia? J Alzheimers Dis 2023; 95:599-602. [PMID: 37661889 PMCID: PMC10947791 DOI: 10.3233/jad-230780] [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] [Indexed: 09/05/2023]
Abstract
Numerous preclinical and human tissue studies implicate the protein phosphatase calcineurin (CN) as a pathophysiologic mechanism in Alzheimer's disease (AD) and other neurodegenerative conditions. Using public electronic records of tens of thousands of individuals across the United States, Silva et al. (2023) show that use of the FDA-approved CN inhibitor, tacrolimus (for purposes of immunosuppression) is also associated with reduced prevalence of dementia-related symptoms. Notably, the study controls for age, sex, and race as well as multiple risk factors for AD. The results suggest that tacrolimus, and possibly other immunosuppressants could be repurposed for the treatment of AD-related dementia.
Collapse
Affiliation(s)
- Christopher M. Norris
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|