15 results on '"INPP5D"'
Search Results
2. Microglial INPP5D limits plaque formation and glial reactivity in the PSAPP mouse model of Alzheimers disease.
- Author
-
Castranio, Emilie, Hasel, Philip, Haure-Mirande, Jean-Vianney, Ramirez Jimenez, Angie, Hamilton, B, Kim, Rachel, Glabe, Charles, Wang, Minghui, Zhang, Bin, Gandy, Sam, Liddelow, Shane, and Ehrlich, Michelle
- Subjects
Alzheimers disease ,Inpp5d ,SHIP1 ,cystatin F ,microglia ,oligomer ,spatial transcriptomics ,Mice ,Humans ,Animals ,Infant ,Alzheimer Disease ,Microglia ,Mice ,Transgenic ,Plaque ,Amyloid ,Disease Models ,Animal ,Amyloid beta-Peptides ,Phosphatidylinositol-3 ,4 ,5-Trisphosphate 5-Phosphatases - 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 Alzheimers 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 Alzheimers disease gene networks.
- Published
- 2023
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. 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
6. SHIP1 therapeutic target enablement: Identification and evaluation of inhibitors for the treatment of late‐onset Alzheimer's disease
- Author
-
Cynthia D. Jesudason, Emily R. Mason, Shaoyou Chu, Adrian L. Oblak, June Javens‐Wolfe, Mustapha Moussaif, Greg Durst, Philip Hipskind, Daniel E. Beck, Jiajun Dong, Ovini Amarasinghe, Zhong‐Yin Zhang, Adam K. Hamdani, Kratika Singhal, Andrew D. Mesecar, Sarah Souza, Marlene Jacobson, Jerry Di Salvo, Disha M. Soni, Murugesh Kandasamy, Andrea R. Masters, Sara K Quinney, Suzanne Doolen, Hasi Huhe, Stacey J. Sukoff Rizzo, Bruce T. Lamb, Alan D. Palkowitz, and Timothy I. Richardson
- Subjects
cellular thermal shift assay (CETSA) ,INPP5D ,pharmacokinetics ,phenotypic high‐content imaging assay ,SHIP1 ,SHIP1 inhibitors ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
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.
- Published
- 2023
- Full Text
- View/download PDF
7. 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
8. SHIP1 Deficiency in Inflammatory Bowel Disease Is Associated With Severe Crohn’s Disease and Peripheral T Cell Reduction
- Author
-
Fernandes, Sandra, Srivastava, Neetu, Sudan, Raki, Middleton, Frank A, Shergill, Amandeep K, Ryan, James C, and Kerr, William G
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Genetics ,Crohn's Disease ,Digestive Diseases ,Autoimmune Disease ,Inflammatory Bowel Disease ,Clinical Research ,2.1 Biological and endogenous factors ,Aetiology ,Oral and gastrointestinal ,Alleles ,Animals ,Autophagy-Related Proteins ,Biomarkers ,Computational Biology ,Crohn Disease ,Disease Models ,Animal ,Disease Susceptibility ,Exons ,Gene Expression Profiling ,Gene Expression Regulation ,Humans ,Inflammatory Bowel Diseases ,Leukocytes ,Mononuclear ,Lymphocyte Count ,Mice ,Mice ,Transgenic ,Mutation ,Phosphatidylinositol-3 ,4 ,5-Trisphosphate 5-Phosphatases ,Proteasome Endopeptidase Complex ,Severity of Illness Index ,T-Lymphocytes ,Exome Sequencing ,SHIP1 ,Crohn's disease ,T cells ,human inflammatory bowel disease ,INPP5D ,ATG16L1 ,fusion transcript ,Crohn’s disease ,Immunology ,Medical Microbiology ,Biochemistry and cell biology - Abstract
In our previous study, we observed a severe reduction in the Src homology 2-containing-inositol-phosphatase-1 (SHIP1) protein in a subpopulation of subjects from a small adult Crohn's Disease (CD) cohort. This pilot study had been undertaken since we had previously demonstrated that engineered deficiency of SHIP1 in mice results in a spontaneous and severe CD-like ileitis. Here, we extend our analysis of SHIP1 expression in peripheral blood mononuclear cells in a second much larger adult Inflammatory Bowel Disease (IBD) cohort, comprised of both CD and Ulcerative Colitis patients, to assess contribution of SHIP1 to the pathogenesis of human IBD. SHIP1 protein and mRNA levels were evaluated from blood samples obtained from IBD subjects seen at UCSF/SFVA, and compared to healthy control samples. Western blot analyses revealed that ~15% of the IBD subjects are severely SHIP1-deficient, with less than 10% of normal SHIP1 protein present in PBMC. Further analyses by flow cytometry and sequencing were performed on secondary samples obtained from the same subjects. Pan-hematolymphoid SHIP1 deficiency was a stable phenotype and was not due to coding changes in the INPP5D gene. A very strong association between SHIP1 deficiency and the presence of a novel SHIP1:ATG16L1 fusion transcript was seen. Similar to SHIP1-deficient mice, SHIP1-deficient subjects had reduced numbers of circulating CD4+ T cell numbers. Finally, SHIP1-deficient subjects with CD had a history of severe disease requiring multiple surgeries. These studies reveal that the SHIP1 protein is crucial for normal T cell homeostasis in both humans and mice, and that it is also a potential therapeutic and/or diagnostic target in human IBD.
- Published
- 2018
9. SHIP1 Deficiency in Inflammatory Bowel Disease Is Associated With Severe Crohn’s Disease and Peripheral T Cell Reduction
- Author
-
Sandra Fernandes, Neetu Srivastava, Raki Sudan, Frank A. Middleton, Amandeep K. Shergill, James C. Ryan, and William G. Kerr
- Subjects
SHIP1 ,Crohn’s disease ,T cells ,human inflammatory bowel disease ,INPP5D ,ATG16L1 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
In our previous study, we observed a severe reduction in the Src homology 2-containing-inositol-phosphatase-1 (SHIP1) protein in a subpopulation of subjects from a small adult Crohn’s Disease (CD) cohort. This pilot study had been undertaken since we had previously demonstrated that engineered deficiency of SHIP1 in mice results in a spontaneous and severe CD-like ileitis. Here, we extend our analysis of SHIP1 expression in peripheral blood mononuclear cells in a second much larger adult Inflammatory Bowel Disease (IBD) cohort, comprised of both CD and Ulcerative Colitis patients, to assess contribution of SHIP1 to the pathogenesis of human IBD. SHIP1 protein and mRNA levels were evaluated from blood samples obtained from IBD subjects seen at UCSF/SFVA, and compared to healthy control samples. Western blot analyses revealed that ~15% of the IBD subjects are severely SHIP1-deficient, with less than 10% of normal SHIP1 protein present in PBMC. Further analyses by flow cytometry and sequencing were performed on secondary samples obtained from the same subjects. Pan-hematolymphoid SHIP1 deficiency was a stable phenotype and was not due to coding changes in the INPP5D gene. A very strong association between SHIP1 deficiency and the presence of a novel SHIP1:ATG16L1 fusion transcript was seen. Similar to SHIP1-deficient mice, SHIP1-deficient subjects had reduced numbers of circulating CD4+ T cell numbers. Finally, SHIP1-deficient subjects with CD had a history of severe disease requiring multiple surgeries. These studies reveal that the SHIP1 protein is crucial for normal T cell homeostasis in both humans and mice, and that it is also a potential therapeutic and/or diagnostic target in human IBD.
- Published
- 2018
- Full Text
- View/download PDF
10. IUSM-Purdue TREAT-AD Center Target Enabling Component; INPP5D (SHIP1) Chemical Probe
- Author
-
Timothy Richardson, Cynthia Jesudason, Shaoyou Chu, Emily Mason, Zhong-Yin Zhang, Andrew Mesecar, Adam Hamdani, Adrian Oblak, Sarah Souza, Marlene Jacobson, and Jerry Di Salvo
- Subjects
INPP5D ,TREM2 ,Alzheimer' Disease (AD) ,SHIP1 - Abstract
A SHIP1/INPP5D Target Enabling Component focused on Chemical Probes
- Published
- 2022
- Full Text
- View/download PDF
11. IUSM-Purdue TREAT-AD Center Target Enabling Package; INPP5D (SHIP1)
- Author
-
Alan Palkowitz, Bruce Lamb, Andrew Mesecar, Zhong-Yin Zhang, Timothy Richardson, Kun Huang, Karson Putt, Cynthia Jesuadason, Jie Zhang, Brent Clayton, Adam Hamdani, Jiajun Dong, Jinmin Miao, Jianping Lin, Emily Mason, Shaoyou Chu, Jun Wan, Travis Johnson, Shunian Xiang, and Members of the IUSM/Purdue TREAT-AD Center
- Subjects
INPP5D ,TREM2 ,Target Enabling Package ,Alzheimer' Disease (AD) ,SHIP1 - Abstract
A Target Enabling Package Centered around INPP5D (SHIP1) as it relates to Alzheimer's disease
- Published
- 2022
- Full Text
- View/download PDF
12. IUSM-Purdue TREAT-AD Center Target Enablement Resource; INPP5D (SHIP1) Bioinformatics
- Author
-
Huang, Kun, Zhang, Jie, Wan, Jun, Johnson, Travis, and Shunian Xiang
- Subjects
FOS: Computer and information sciences ,INPP5D ,Bioinformatics ,Alzheimer' Disease (AD) ,SHIP1 - Abstract
Target Enablement Resource for INPP5D/SHIP1 specific to bioinformatics
- Published
- 2022
- Full Text
- View/download PDF
13. IUSM-Purdue TREAT-AD Center Target Enablement Resource; INPP5D (SHIP1) Protein Structures
- Author
-
Andrew Mesecar and Adam Hamdani
- Subjects
INPP5D ,TREM2 ,Alzheimer' Disease (AD) ,SHIP1 - Abstract
Target Enablement Resource specific to INPP5D/SHIP1 Protein Structures
- Published
- 2022
- Full Text
- View/download PDF
14. IUSM-Purdue TREAT-AD Center Target Enablement Resource; INPP5D (SHIP1) Screening
- Author
-
Zhong-Yin Zhang, Karson Putt, Shaoyou Chu, Emily Mason, Jiajun Dong, Jinmin Miao, and Jianping Lin
- Subjects
INPP5D ,TREM2 ,Alzheimer' Disease (AD) ,SHIP1 - Abstract
Target Enablement Resource for INPP5D/SHIP1 specific to screening
- Published
- 2022
- Full Text
- View/download PDF
15. Define the RelationSHIP: INPP5D/SHIP1 regulates inflammasome activation in human microglia
- Author
-
Chou, Vicky
- Subjects
- alzheimers, inflammasome, Inpp5d, microglia, ship1, Neurosciences
- Abstract
Microglia and neuroinflammation are implicated in the development and progression of Alzheimer’s disease (AD). However, there are limitations to studying human microglia and utilizing mouse models to study microglia function. Various iPSC-derived microglia-like cell (iMGLs) differentiation protocols have been published within the last few years. Here, one protocol was adapted with modifications to optimize for consistency and yield across genetic backgrounds. With the iMGLs, mix culture models were generated containing iPSC-derived neurons, astrocytes, and microglia. These mixed culture models were then characterized using single cell and single nucleus sequencing to understand then transcriptional changes between a monoculture and mixed culture. To better understand microglia-mediated processes in AD, the function of INPP5D/SHIP1, a gene linked to AD through GWAS was investigated in human brain tissue and iMGL cell cultures. Immunostaining and single nucleus RNA sequencing confirmed that INPP5D expression in the adult human brain is largely restricted to microglia. Examination of prefrontal cortex across a large cohort revealed reduced full length INPP5D protein levels in AD patient brains compared to cognitively normal controls. The functional consequences of reduced INPP5D activity were evaluated in human iMGLs using both pharmacological inhibition of the phosphatase activity of INPP5D and using CRISPR-Cas9 to reduce the number of functional INPP5D copy numbers in half. Unbiased transcriptional and proteomic profiling of these iMGLs suggested an upregulation of innate immune signaling pathways, lower levels of scavenger receptors, and altered inflammasome signaling with INPP5D reduction. INPP5D inhibition induced the secretion of IL-1ß and IL-18, further implicating inflammasome activation. Inflammasome activation was confirmed through visualization of inflammasome formation through ASC immunostaining in INPP5D-inhibited iMGLs, increased cleaved caspase-1, and through rescue of elevated IL-1ß and IL-18 with caspase-1 and NLRP3 inhibitors. In the INPP5D het iMGLs, an increased secretion of both IL-1ß and IL-18 were observed, which was rescued with NLRP3 inhibitor treatment. This work implicates INPP5D as a regulator of inflammasome signaling in human microglia and provides further insight as to how INPP5D function could be linked to the development of AD.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.