16 results on '"INPP5D"'
Search Results
2. Inpp5d haplodeficiency alleviates tau pathology in the PS19 mouse model of Tauopathy.
- Author
-
Soni, Disha M., Lin, Peter Bor‐Chian, Lee‐Gosselin, Audrey, Lloyd, Christopher D., Mason, Emily, Ingraham, Cynthia M., Perkins, Abigail, Moutinho, Miguel, Lamb, Bruce T., Chu, Shaoyou, and Oblak, Adrian L.
- Abstract
INTRODUCTION: A noncoding variant (rs35349669) within INPP5D, a lipid and protein phosphatase restricted to microglia in the brain, is linked to increased susceptibility to Alzheimer's disease (AD). While Inpp5d is well‐studied in amyloid pathology, its role in tau pathology remains unclear. METHODS: PS19 Tauopathy mice were crossed with Inpp5d‐haplodeficient (Inpp5d+/−) mice to examine the impact of Inpp5d in tau pathology. RESULTS: Increased INPP5D expression correlated positively with phospho‐Tau AT8 in PS19 mice. Inpp5d haplodeficiency mitigated hyperphosphorylated tau levels (AT8, AT180, AT100, and PHF1) and motor deficits in PS19 mice. Transcriptomic analysis revealed an up‐regulation of genes associated with immune response and cell migration. DISCUSSION: Our findings define an association between INPP5D expression and tau pathology in PS19 mice. Alleviation in hyperphosphorylated tau, motor deficits, and transcriptomics changes in haplodeficient‐Inpp5d PS19 mice indicate that modulation in INPP5D expression may provide therapeutic potential for mitigating tau pathology and improving motor deficits. Highlights: The impact of Inpp5d in the context of tau pathology was studied in the PS19 mouse model.INPP5D expression is associated with tau pathology.Reduced Inpp5d expression in PS19 mice improved motor functions and decreased total and phospho‐Tau levels.Inpp5d haplodeficiency in PS19 mice modulates gene expression patterns linked to immune response and cell migration.These data suggest that inhibition of Inpp5d may be a therapeutic approach in tauopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Microglial function, INPP5D/SHIP1 signaling, and NLRP3 inflammasome activation: implications for Alzheimer’s disease
- Author
-
Gizem Terzioglu and Tracy L. Young-Pearse
- Subjects
NLRP3 inflammasome ,SHIP1 ,INPP5D ,Alzheimer’s Disease ,Inflammation ,Phosphoinositide signaling ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Recent genetic studies on Alzheimer’s disease (AD) have brought microglia under the spotlight, as loci associated with AD risk are enriched in genes expressed in microglia. Several of these genes have been recognized for their central roles in microglial functions. Increasing evidence suggests that SHIP1, the protein encoded by the AD-associated gene INPP5D, is an important regulator of microglial phagocytosis and immune response. A recent study from our group identified SHIP1 as a negative regulator of the NLRP3 inflammasome in human iPSC-derived microglial cells (iMGs). In addition, we found evidence for a connection between SHIP1 activity and inflammasome activation in the AD brain. The NLRP3 inflammasome is a multiprotein complex that induces the secretion of pro-inflammatory cytokines as part of innate immune responses against pathogens and endogenous damage signals. Previously published studies have suggested that the NLRP3 inflammasome is activated in AD and contributes to AD-related pathology. Here, we provide an overview of the current understanding of the microglial NLRP3 inflammasome in the context of AD-related inflammation. We then review the known intracellular functions of SHIP1, including its role in phosphoinositide signaling, interactions with microglial phagocytic receptors such as TREM2 and evidence for its intersection with NLRP3 inflammasome signaling. Through rigorous examination of the intricate connections between microglial signaling pathways across several experimental systems and postmortem analyses, the field will be better equipped to tailor newly emerging therapeutic strategies targeting microglia in neurodegenerative diseases.
- Published
- 2023
- Full Text
- View/download PDF
4. Microglial function, INPP5D/SHIP1 signaling, and NLRP3 inflammasome activation: implications for Alzheimer's disease.
- Author
-
Terzioglu, Gizem and Young-Pearse, Tracy L.
- Subjects
ALZHEIMER'S disease ,NLRP3 protein ,INFLAMMASOMES ,MICROGLIA ,POSTMORTEM changes ,IMMUNE response - Abstract
Recent genetic studies on Alzheimer's disease (AD) have brought microglia under the spotlight, as loci associated with AD risk are enriched in genes expressed in microglia. Several of these genes have been recognized for their central roles in microglial functions. Increasing evidence suggests that SHIP1, the protein encoded by the AD-associated gene INPP5D, is an important regulator of microglial phagocytosis and immune response. A recent study from our group identified SHIP1 as a negative regulator of the NLRP3 inflammasome in human iPSC-derived microglial cells (iMGs). In addition, we found evidence for a connection between SHIP1 activity and inflammasome activation in the AD brain. The NLRP3 inflammasome is a multiprotein complex that induces the secretion of pro-inflammatory cytokines as part of innate immune responses against pathogens and endogenous damage signals. Previously published studies have suggested that the NLRP3 inflammasome is activated in AD and contributes to AD-related pathology. Here, we provide an overview of the current understanding of the microglial NLRP3 inflammasome in the context of AD-related inflammation. We then review the known intracellular functions of SHIP1, including its role in phosphoinositide signaling, interactions with microglial phagocytic receptors such as TREM2 and evidence for its intersection with NLRP3 inflammasome signaling. Through rigorous examination of the intricate connections between microglial signaling pathways across several experimental systems and postmortem analyses, the field will be better equipped to tailor newly emerging therapeutic strategies targeting microglia in neurodegenerative diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. The Alzheimer's disease risk factor INPP5D restricts neuroprotective microglial responses in amyloid beta‐mediated pathology.
- Author
-
Samuels, Joshua D., Moore, Katelyn A., Ennerfelt, Hannah E., Johnson, Alexis M., Walsh, Adeline E., Price, Richard J., and Lukens, John R.
- Abstract
Introduction: Mutations in INPP5D, which encodes for the SH2‐domain‐containing inositol phosphatase SHIP‐1, have recently been linked to an increased risk of developing late‐onset Alzheimer's disease. While INPP5D expression is almost exclusively restricted to microglia in the brain, little is known regarding how SHIP‐1 affects neurobiology or neurodegenerative disease pathogenesis. Methods: We generated and investigated 5xFAD Inpp5dfl/flCx3cr1Ert2Cre mice to ascertain the function of microglial SHIP‐1 signaling in response to amyloid beta (Aβ)‐mediated pathology. Results: SHIP‐1 deletion in microglia led to substantially enhanced recruitment of microglia to Aβ plaques, altered microglial gene expression, and marked improvements in neuronal health. Further, SHIP‐1 loss enhanced microglial plaque containment and Aβ engulfment when compared to microglia from Cre‐negative 5xFAD Inpp5dfl/fl littermate controls. Discussion: These results define SHIP‐1 as a pivotal regulator of microglial responses during Aβ‐driven neurological disease and suggest that targeting SHIP‐1 may offer a promising strategy to treat Alzheimer's disease. Highlights: Inpp5d deficiency in microglia increases plaque‐associated microglia numbers.Loss of Inpp5d induces activation and phagocytosis transcriptional pathways.Plaque encapsulation and engulfment by microglia are enhanced with Inpp5d deletion.Genetic ablation of Inpp5d protects against plaque‐induced neuronal dystrophy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. SHIP1 therapeutic target enablement: Identification and evaluation of inhibitors for the treatment of late‐onset Alzheimer's disease.
- Author
-
Jesudason, Cynthia D., Mason, Emily R., Chu, Shaoyou, Oblak, Adrian L., Javens‐Wolfe, June, Moussaif, Mustapha, Durst, Greg, Hipskind, Philip, Beck, Daniel E., Dong, Jiajun, Amarasinghe, Ovini, Zhang, Zhong‐Yin, Hamdani, Adam K., Singhal, Kratika, Mesecar, Andrew D., Souza, Sarah, Jacobson, Marlene, Salvo, Jerry Di, Soni, Disha M., and Kandasamy, Murugesh
- Subjects
ALZHEIMER'S disease ,DISEASE risk factors ,ORAL drug administration ,POLYPHOSPHATES - Abstract
INTRODUCTION: The risk of developing Alzheimer's disease is associated with genes involved in microglial function. Inositol polyphosphate‐5‐phosphatase (INPP5D), which encodes Src homology 2 (SH2) domain–containing inositol polyphosphate 5‐phosphatase 1 (SHIP1), is a risk gene expressed in microglia. Because SHIP1 binds receptor immunoreceptor tyrosine‐based inhibitory motifs (ITIMs), competes with kinases, and converts PI(3,4,5)P3 to PI(3,4)P2, it is a negative regulator of microglia function. Validated inhibitors are needed to evaluate SHIP1 as a potential therapeutic target. METHODS: We identified inhibitors and screened the enzymatic domain of SHIP1. A protein construct containing two domains was used to evaluate enzyme inhibitor potency and selectivity versus SHIP2. Inhibitors were tested against a construct containing all ordered domains of the human and mouse proteins. A cellular thermal shift assay (CETSA) provided evidence of target engagement in cells. Phospho‐AKT levels provided further evidence of on‐target pharmacology. A high‐content imaging assay was used to study the pharmacology of SHIP1 inhibition while monitoring cell health. Physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties were evaluated to select a compound suitable for in vivo studies. RESULTS: SHIP1 inhibitors displayed a remarkable array of activities and cellular pharmacology. Inhibitory potency was dependent on the protein construct used to assess enzymatic activity. Some inhibitors failed to engage the target in cells. Inhibitors that were active in the CETSA consistently destabilized the protein and reduced pAKT levels. Many SHIP1 inhibitors were cytotoxic either at high concentration due to cell stress or they potently induced cell death depending on the compound and cell type. One compound activated microglia, inducing phagocytosis at concentrations that did not result in significant cell death. A pharmacokinetic study demonstrated brain exposures in mice upon oral administration. DISCUSSION: 3‐((2,4‐Dichlorobenzyl)oxy)‐5‐(1‐(piperidin‐4‐yl)‐1H‐pyrazol‐4‐yl) pyridine activated primary mouse microglia and demonstrated exposures in mouse brain upon oral dosing. Although this compound is our recommended chemical probe for investigating the pharmacology of SHIP1 inhibition at this time, further optimization is required for clinical studies. Highlights: Cellular thermal shift assay (CETSA) and signaling (pAKT) assays were developed to provide evidence of src homology 2 (SH2) domain‐contaning inositol phosphatase 1 (SHIP1) target engagement and on‐target activity in cellular assays.A phenotypic high‐content imaging assay with simultaneous measures of phagocytosis, cell number, and nuclear intensity was developed to explore cellular pharmacology and monitor cell health.SHIP1 inhibitors demonstrate a wide range of activity and cellular pharmacology, and many reported inhibitors are cytotoxic.The chemical probe 3‐((2,4‐dichlorobenzyl)oxy)−5‐(1‐(piperidin‐4‐yl)−1H‐pyrazol‐4‐yl) pyridine is recommended to explore SHIP1 pharmacology. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Microglial INPP5D limits plaque formation and glial reactivity in the PSAPP mouse model of Alzheimer's disease.
- Author
-
Castranio, Emilie L., Hasel, Philip, Haure‐Mirande, Jean‐Vianney, Ramirez Jimenez, Angie V., Hamilton, B. Wade, Kim, Rachel D., Glabe, Charles G., Wang, Minghui, Zhang, Bin, Gandy, Sam, Liddelow, Shane A., and Ehrlich, Michelle E.
- Abstract
Introduction: The inositol polyphosphate‐5‐phosphatase D (INPP5D) gene encodes a dual‐specificity phosphatase that can dephosphorylate both phospholipids and phosphoproteins. Single nucleotide polymorphisms in INPP5D impact risk for developing late onset sporadic Alzheimer's disease (LOAD). Methods: To assess the consequences of inducible Inpp5d knockdown in microglia of APPKM670/671NL/PSEN1Δexon9 (PSAPP) mice, we injected 3‐month‐old Inpp5dfl/fl/Cx3cr1CreER/+ and PSAPP/Inpp5dfl/fl/Cx3cr1CreER/+ mice with either tamoxifen (TAM) or corn oil (CO) to induce recombination. Results: At age 6 months, we found that the percent area of 6E10+ deposits and plaque‐associated microglia in Inpp5d knockdown mice were increased compared to controls. Spatial transcriptomics identified a plaque‐specific expression profile that was extensively altered by Inpp5d knockdown. Discussion: These results demonstrate that conditional Inpp5d downregulation in the PSAPP mouse increases plaque burden and recruitment of microglia to plaques. Spatial transcriptomics highlighted an extended gene expression signature associated with plaques and identified CST7 (cystatin F) as a novel marker of plaques. Highlights: Inpp5d knockdown increases plaque burden and plaque‐associated microglia number.Spatial transcriptomics identifies an expanded plaque‐specific gene expression profile.Plaque‐induced gene expression is altered by Inpp5d knockdown in microglia.Our plaque‐associated gene signature overlaps with human Alzheimer's disease gene networks. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Expression of INPP5D Isoforms in Human Brain: Impact of Alzheimer's Disease Neuropathology and Genetics.
- Author
-
Zajac, Diana J., Simpson, James, Zhang, Eric, Parikh, Ishita, and Estus, Steven
- Subjects
- *
ALZHEIMER'S disease , *DISEASE risk factors , *SINGLE nucleotide polymorphisms , *GENETICS , *NEUROLOGICAL disorders - Abstract
The single nucleotide polymorphisms rs35349669 and rs10933431 within Inositol Polyphosphate-5-Phosphatase D (INPP5D) are strongly associated with Alzheimer's Disease risk. To better understand INPP5D expression in the brain, we investigated INPP5D isoform expression as a function of rs35349669 and rs10933431, as well as Alzheimer's disease neuropathology, by qPCR and isoform-specific primers. In addition, INPP5D allelic expression imbalance was evaluated relative to rs1141328 within exon 1. Expression of INPP5D isoforms associated with transcription start sites in exon 1 and intron 14 was increased in individuals with high Alzheimer's disease neuropathology. In addition, a novel variant with 47bp lacking from exon 12 increased expression in Alzheimer's Disease brains, accounting for 13% of total INPP5D expression, and was found to undergo nonsense-mediated decay. Although inter-individual variation obscured a possible polymorphism effect on INPP5D isoform expression as measured by qPCR, rs35349669 was associated with rs1141328 allelic expression imbalance, suggesting that rs35349669 is significantly associated with full-length INPP5D isoform expression. In summary, expression of INPP5D isoforms with start sites in exon 1 and intron 14 are increased in brains with high Alzheimer's Disease neuropathology, a novel isoform lacking the phosphatase domain was significantly increased with the disease, and the polymorphism rs35349669 correlates with allele-specific full-length INPP5D expression. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. Targeted genetic analysis of cerebral blood flow imaging phenotypes implicates the INPP5D gene.
- Author
-
Yao, Xiaohui, Risacher, Shannon L., Nho, Kwangsik, Saykin, Andrew J., Wang, Ze, and Shen, Li
- Subjects
- *
CEREBRAL circulation , *MAGNETIC resonance angiography , *AMYLOID plaque , *BLOOD testing , *ALZHEIMER'S disease , *SPIN labels - Abstract
The vascular hypothesis of Alzheimer's disease (AD) has proposed the involvement of brain hypoperfusion in AD pathogenesis, where cognitive decline and dysfunction result from dwindling cerebral blood flow (CBF). Based on the vascular hypothesis of Alzheimer's disease, we focused on exploring how genetic factors influence AD pathogenesis via the cerebrovascular system. To investigate the role of CBF endophenotypes in AD pathogenesis, we performed a targeted genetic analysis of 258 subjects from the Alzheimer's Disease Neuroimaging Initiative cohort to examine associations between 4033 single-nucleotide polymorphisms of 24 AD genes and CBF measures in 4 brain regions. A novel association with CBF measure in the left angular gyrus was identified in an INPP5D single-nucleotide polymorphism (i.e., rs61068452; p = 1.48E-7; corrected p = 2.39E-3). The gene-based analysis discovered both INPP5D and CD2AP associated with the left angular gyrus CBF. Further analyses on nonoverlapping samples revealed that rs61068452-G was associated with lower CSF t-tau/Aβ 1–42 ratio. Our findings suggest a protective role of rs61068452-G in an AD-relevant cerebrovascular endophenotype, which has the potential to provide novel insights for better mechanistic understanding of AD. • We performed a targeted genetic study of CBF phenotypes captured by arterial spin labeling (ASL) perfusion magnetic resonance imaging (pMRI). • We discovered a novel locus in INPP5D (rs61068452) significantly associated with the CBF measure in the L-AG. • The identified genetic variation was also associated with several AD biomarkers. • This work is among the first genetic association studies of CBF in AD, and our finding has the potential to help understand AD molecular mechanism. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
10. Associations of Alzheimer’s disease risk variants with gene expression, amyloidosis, tauopathy, and neurodegeneration
- Author
-
John Suckling, Meng-Shan Tan, Yu-Xiang Yang, Wei Xu, Lan Tan, Jin-Tai Yu, Chuantao Zuo, Lin Tan, Hui-Fu Wang, Qiang Dong, Yu, Jin-Tai [0000-0002-7686-0547], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Amyloid ,Cognitive Neuroscience ,Biology ,lcsh:RC346-429 ,lcsh:RC321-571 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,INPP5D ,Neurodegeneration ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,lcsh:Neurology. Diseases of the nervous system ,Genetic association ,Genetics ,Risk variants ,Amyloidosis ,Research ,Human brain ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Tauopathies ,Neurology (clinical) ,Tauopathy ,Gene expression ,Alzheimer's disease ,Alzheimer disease ,Tau ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Funder: ZHANGJIANG LAB, Tianqiao and Chrissy Chen Institute, and the State Key Laboratory of Neurobiology and Frontiers Center for Brain Science of Ministry of Education, Fudan University, Background: Genome-wide association studies have identified more than 30 Alzheimer’s disease (AD) risk genes, although the detailed mechanism through which all these genes are associated with AD pathogenesis remains unknown. We comprehensively evaluate the roles of the variants in top 30 non-APOE AD risk genes, based on whether these variants were associated with altered mRNA transcript levels, as well as brain amyloidosis, tauopathy, and neurodegeneration. Methods: Human brain gene expression data were obtained from the UK Brain Expression Consortium (UKBEC), while other data used in our study were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort. We examined the association of AD risk allele carrier status with the levels of gene expression in blood and brain regions and tested the association with brain amyloidosis, tauopathy, and neurodegeneration at baseline, using a multivariable linear regression model. Next, we analyzed the longitudinal effects of these variants on the change rates of pathology using a mixed effect model. Results: Altogether, 27 variants were detected to be associated with the altered expression of 21 nearby genes in blood and brain regions. Eleven variants (especially novel variants in ADAM10, IGHV1-68, and SLC24A4/RIN3) were associated with brain amyloidosis, 7 variants (especially in INPP5D, PTK2B) with brain tauopathy, and 8 variants (especially in ECHDC3, HS3ST1) with brain neurodegeneration. Variants in ADAMTS1, BZRAP1-AS1, CELF1, CD2AP, and SLC24A4/RIN3 participated in more than one cerebral pathological process. Conclusions: Genetic variants might play functional roles and suggest potential mechanisms in AD pathogenesis, which opens doors to uncover novel targets for AD treatment.
- Published
- 2021
11. INPP5D Modulates TREM2 Loss-of-Function Phenotypes in a Mouse Model of Alzheimer Disease
- Author
-
Taisuke Tomita, Shingo Kimura, Tsuneya Ikezu, Akihiro Iguchi, Yukiko Hori, Takehiko Sasaki, Takaomi C. Saido, Toshiyuki Takai, Junko Sasaki, Sho Takatori, and Takashi Saito
- Subjects
Pathogenesis ,medicine.anatomical_structure ,Microglia ,TREM2 ,Phosphatase ,medicine ,INPP5D ,Alzheimer's disease ,Biology ,medicine.disease ,Phenotype ,Astrogliosis ,Cell biology - Abstract
The genetic associations of TREM2 loss-of-function variants with Alzheimer disease (AD) indicate the protective roles of microglia in AD pathogenesis. Functional deficiencies of TREM2 disrupt microglial clustering around amyloid β (Aβ) plaques, impair their transcriptional response to Aβ, and worsen neuritic dystrophy. However, the molecular mechanism underlying these phenotypes remains unclear. In this study, we investigated the pathological role of another AD risk gene, INPP5D, encoding a phosphoinositide PI(3,4,5)P3 phosphatase expressed in microglia. In a Tyrobp-deficient TREM2 loss-of-function mouse model, Inpp5d haplodeficiency restored the association of microglia with Aβ plaques, partially restored plaque compaction and astrogliosis, and reduced phosphorylated tau+ dystrophic neurites. RNA-sequencing of isolated microglia demonstrated that the expression of “disease-associated microglia” genes is not responsible for these beneficial effects. Our results suggest that INPP5D acts downstream of TREM2/TYROBP to regulate the microglial barrier against Aβ toxicity, thereby modulates the pathological progression from Aβ deposition to tau pathology.
- Published
- 2021
- Full Text
- View/download PDF
12. INPP5D rs35349669 polymorphism with late-onset Alzheimer's disease: A replication study and meta-analysis
- Author
-
Ling-Li Kong, Hua Jing, Chen-Chen Tan, Zi-Xuan Wang, Lin Tan, Wei Zhang, Zhan-Jie Zheng, Hui-Fu Wang, Jun-Xia Zhu, and Lan Tan
- Subjects
Male ,0301 basic medicine ,China ,medicine.medical_specialty ,Genotype ,INPP5D ,Single-nucleotide polymorphism ,Disease ,association study ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Research Paper: Gerotarget (Focus on Aging) ,0302 clinical medicine ,Asian People ,Gene Frequency ,Alzheimer Disease ,Internal medicine ,medicine ,Genetic predisposition ,Humans ,SNP ,Genetic Predisposition to Disease ,Allele ,Allele frequency ,Alleles ,rs35349669 ,Aged ,Aged, 80 and over ,Traditional medicine ,Gerotarget ,business.industry ,Alzheimer's disease ,meta-analysis ,030104 developmental biology ,Oncology ,Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases ,Meta-analysis ,Female ,business ,030217 neurology & neurosurgery - Abstract
// Hua Jing 1,* , Jun-Xia Zhu 1,* , Hui-Fu Wang 1 , Wei Zhang 2 , Zhan-Jie Zheng 3 , Ling-Li Kong 3 , Chen-Chen Tan 1 , Zi-Xuan Wang 1 , Lin Tan 4 and Lan Tan 1,4 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China 2 Department of Emergency, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China 3 Department of Geriatric, Qingdao Mental Health Center, Qingdao, PR China 4 College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, PR China * The first two authors should be regarded as co-first authors Correspondence to: Lan Tan, email: // Keywords : Alzheimer’s disease; INPP5D; rs35349669; association study; meta-analysis; Gerotarget Received : May 05, 2016 Accepted : October 02, 2016 Published : October 13, 2016 Abstract Inositol polyphosphate-5-phosphatase ( INPP5D ) was reported to be associated with Alzheimer’s disease (AD) through modulating the inflammatory process and immune response. A recent genome-wide association study discovered a new locus single nucleotide polymorphism (SNP, rs35349669) of INPP5D which was significantly associated with susceptibility to late-onset Alzheimer’s disease (LOAD) in Caucasians. In this study, we investigated the relations between the INPP5D polymorphism rs35349669 and LOAD in Han Chinese population comprising 984 LOAD cases and 1352 healthy controls being matched for age and gender. Our results showed no obvious differences in the genotypic or allelic distributions of rs35349669 polymorphism between LOAD cases and healthy controls (genotype: p = 0.167; allele: p = 0.094). Additionally, when these data were stratified by APOE e4 status, there are still no evident differences in the genotypic or allelic distributions in APOEe4 carriers ( p > 0.05). Furthermore, meta-analysis of 81964 individuals confirmed that rs35349669 was significantly associated with the risk for LOAD (OR=1.08, 95%CI=1.06-1.11), but the results remained negative in Chinese subgroup (OR=0.77, 95%CI=0.53-1.13). Overall, the current evidence did not indicate that INPP5D rs35349669 polymorphism play a role in the genetic predisposition to LOAD in Chinese population.
- Published
- 2016
- Full Text
- View/download PDF
13. INPP5D expression is associated with risk for Alzheimer's disease and induced by plaque-associated microglia.
- Author
-
Tsai, Andy P., Lin, Peter Bor-Chian, Dong, Chuanpeng, Moutinho, Miguel, Casali, Brad T., Liu, Yunlong, Lamb, Bruce T., Landreth, Gary E., Oblak, Adrian L., and Nho, Kwangsik
- Subjects
- *
ALZHEIMER'S disease , *MICROGLIA , *AMYLOID plaque - Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, robust microgliosis, neuroinflammation, and neuronal loss. Genome-wide association studies recently highlighted a prominent role for microglia in late-onset AD (LOAD). Specifically, inositol polyphosphate-5-phosphatase (INPP5D), also known as SHIP1, is selectively expressed in brain microglia and has been reported to be associated with LOAD. Although INPP5D is likely a crucial player in AD pathophysiology, its role in disease onset and progression remains unclear. We performed differential gene expression analysis to investigate INPP5D expression in AD and its association with plaque density and microglial markers using transcriptomic (RNA-Seq) data from the Accelerating Medicines Partnership for Alzheimer's Disease (AMP-AD) cohort. We also performed quantitative real-time PCR, immunoblotting, and immunofluorescence assays to assess INPP5D expression in the 5xFAD amyloid mouse model. Differential gene expression analysis found that INPP5D expression was upregulated in LOAD and positively correlated with amyloid plaque density. In addition, in 5xFAD mice, Inpp5d expression increased as the disease progressed, and selectively in plaque-associated microglia. Increased Inpp5d expression levels in 5xFAD mice were abolished entirely by depleting microglia with the colony-stimulating factor receptor-1 antagonist PLX5622. Our findings show that INPP5D expression increases as AD progresses, predominantly in plaque-associated microglia. Importantly, we provide the first evidence that increased INPP5D expression might be a risk factor in AD, highlighting INPP5D as a potential therapeutic target. Moreover, we have shown that the 5xFAD mouse model is appropriate for studying INPP5D in AD. • INPP5D was positively associated with amyloid plaque density in the human brain. • In the 5xFAD mouse model, Inpp5d increased in a disease-progression-dependent manner • Inpp5d was selectively expressed in plaque-associated microglia. • The increased Inpp5d expression levels were attenuated following microglia depletion. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
14. Alzheimer’s Disease Risk Genes and Mechanisms of Disease Pathogenesis
- Author
-
Alison Goate and Celeste M. Karch
- Subjects
TREM2 ,SORL1 ,Genome-wide association study ,Disease ,Biology ,medicine.disease ,Bioinformatics ,Endocytosis ,Article ,Presenilin ,PICALM ,Amyloid beta-Protein Precursor ,Cholesterol ,Phenotype ,Alzheimer Disease ,Risk Factors ,Presenilin-2 ,Presenilin-1 ,medicine ,Humans ,Genetic Predisposition to Disease ,INPP5D ,Alzheimer's disease ,Biological Psychiatry ,Genome-Wide Association Study - Abstract
Here, we review the genetic risk factors for late onset Alzheimer's disease (AD) and their role in AD pathogenesis. Recent advances in our understanding of the human genome, namely technological advances in methods to analyze millions of polymorphisms in thousands of subjects, have revealed new genes associated with AD risk: ABCA7, BIN1, CASS4, CD33, CD2AP, CELF1, CLU, CR1, DSG2, EPHA1, FERMT2, HLA-DRB5-DBR1, INPP5D, MS4A, MEF2C, NME8, PICALM, PTK2B, SLC24H4 RIN3, SORL1, ZCWPW1. Emerging technologies to analyze the entire genome in large datasets have also revealed coding variants that increase AD risk: PLD3 and TREM2. We review the relationship between these AD risk genes and the cellular and neuropathological features of AD. Together, understanding the mechanisms underlying the association of these genes with risk for disease will provide the most meaningful targets for therapeutic development to date.
- Published
- 2015
- Full Text
- View/download PDF
15. Late onset Alzheimer’s disease genetics implicates microglial pathways in disease risk
- Author
-
Alison Goate and Anastasia G. Efthymiou
- Subjects
Myeloid ,0301 basic medicine ,Gene regulatory network ,Context (language use) ,Review ,Disease ,lcsh:Geriatrics ,Biology ,lcsh:RC346-429 ,ABCA7 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Alzheimer Disease ,Risk Factors ,Genetics ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,INPP5D ,Age of Onset ,Molecular Biology ,lcsh:Neurology. Diseases of the nervous system ,Microglia ,TREM2 ,medicine.disease ,3. Good health ,lcsh:RC952-954.6 ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,biology.protein ,Neurology (clinical) ,Alzheimer's disease ,Alzheimer’s disease ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Alzheimer’s disease (AD) is a highly heritable complex disease with no current effective prevention or treatment. The majority of drugs developed for AD focus on the amyloid cascade hypothesis, which implicates Aß plaques as a causal factor in the disease. However, it is possible that other underexplored disease-associated pathways may be more fruitful targets for drug development. Findings from gene network analyses implicate immune networks as being enriched in AD; many of the genes in these networks fall within genomic regions that contain common and rare variants that are associated with increased risk of developing AD. Of these genes, several (including CR1, SPI1, the MS4As, TREM2, ABCA7, CD33, and INPP5D) are expressed by microglia, the resident immune cells of the brain. We summarize the gene network and genetics findings that implicate that these microglial genes are involved in AD, as well as several studies that have looked at the expression and function of these genes in microglia and in the context of AD. We propose that these genes are contributing to AD in a non-Aß-dependent fashion.
- Published
- 2017
- Full Text
- View/download PDF
16. Alzheimer's disease genes and autophagy
- Author
-
Dong-Hou Kim and Seung-Yong Yoon
- Subjects
0301 basic medicine ,Autophagosome ,Cellular homeostasis ,Biology ,Presenilin ,03 medical and health sciences ,Alzheimer Disease ,Lysosome ,medicine ,Autophagy ,Animals ,Humans ,INPP5D ,Molecular Biology ,General Neuroscience ,Brain ,Membrane Proteins ,BECN1 ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,Neurology (clinical) ,Alzheimer's disease ,Neuroscience ,Developmental Biology - Abstract
Autophagy is a process to degrade and recycle cellular constituents via the lysosome for regulating cellular homeostasis. Its dysfunction is now considered to be involved in many diseases, including neurodegenerative diseases. Many features reflecting autophagy impairment, such as autophagosome accumulation and lysosomal dysfunction, have been also revealed to be involved in Alzheimer's disease (AD). Recent genetic studies such as genome-wide association studies in AD have identified a number of novel genes associated with AD. Some of the identified genes have demonstrated dysfunction in autophagic processes in AD, while others remain under investigation. Since autophagy is strongly regarded to be one of the major pathogenic mechanisms of AD, it is necessary to review how the AD-associated genes are related to autophagy. We anticipate our current review to be a starting point for future studies regarding AD-associated genes and autophagy. This article is part of a Special Issue entitled SI:Autophagy.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.