Abstract This study investigates NADPH oxidase 4 (NOX4) involvement in iron-mediated astrocyte cell death in Alzheimer’s Disease (AD) using single-cell sequencing data and transcriptomes. We analyzed AD single-cell RNA sequencing data, identified astrocyte marker genes, and explored biological processes in astrocytes. We integrated AD-related chip data with ferroptosis-related genes, highlighting NOX4. We validated NOX4’s role in ferroptosis and AD in vitro and in vivo. Astrocyte marker genes were enriched in AD, emphasizing their role. NOX4 emerged as a crucial player in astrocytic ferroptosis in AD. Silencing NOX4 mitigated ferroptosis, improved cognition, reduced Aβ and p-Tau levels, and alleviated mitochondrial abnormalities. NOX4 promotes astrocytic ferroptosis, underscoring its significance in AD progression. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-024-01266-w. Keywords: Single-cell sequencing, Ferroptosis, NADPH oxidase 4, Astrocytes, Alzheimer’s disease, Differential gene analysis, Immunofluorescence staining, Mouse model validation Introduction Alzheimer’s disease (AD) is a prevalent neurodegenerative condition characterized by memory loss, cognitive decline, and impaired behavioral abilities [[35]1–[36]3]. Despite some advances in understanding AD’s pathological mechanisms, the specific molecular underpinnings remain elusive, and effective treatments targeting its root causes are lacking [[37]1, [38]4–[39]6]. A comprehensive investigation into AD’s pathogenesis holds paramount importance for its prevention and treatment. In recent years, ferroptosis, an emerging form of cell death reliant on iron, has garnered significant attention in the realm of neurodegenerative diseases [[40]7–[41]10]. Astrocytes are one of the most abundant subtypes of glial cells in the central nervous system [[42]11, [43]12]. They are associated with brain development and function, such as regulating synaptic formation and function, controlling neurotransmitter release and uptake, producing trophic factors, and maintaining neuronal survival [[44]13–[45]16]. As major glial cells in the central nervous system, astrocytes also play a role in various physiological and pathological processes in the brain and may have significant implications for the pathogenesis of AD [[46]17–[47]19]. Previous studies have indicated higher levels of astrocyte damage in Alzheimer’s disease patients [[48]20]. Ferroptosis, characterized by heightened lipid peroxidation and iron-dependent reactive oxygen species generation, constitutes a novel iron-dependent cell demise pathway [[49]21–[50]23]. There is a close relationship between oxidative stress and iron death. Both iron excess and deficiency can induce oxidative stress, leading to cell death and other related diseases [[51]24]. Studies suggest that iron death may play a significant role in neurodegenerative diseases, including Alzheimer’s disease [[52]25]. Furthermore, literature indicates the presence of high concentrations of iron in the brains of AD patients and transgenic mouse models, where excess iron can exacerbate oxidative damage and cause cognitive impairment. Disruption of iron homeostasis is considered to be associated with Alzheimer’s disease [[53]26, [54]27]. Increasing evidence indicates that iron death can lead to AD-mediated neuronal cell death [[55]28]. Nevertheless, iron’s precise roles and regulatory mechanisms in Alzheimer’s, particularly its interplay with astrocytes, remain enigmatic [[56]29]. Star-shaped glial cells play pivotal roles in the central nervous system’s physiological and pathological processes, with certain key regulatory genes or pathways potentially offering valuable insights into Alzheimer’s pathogenesis [[57]30–[58]32]. Single-cell sequencing technology, noted for its remarkable sensitivity and high resolution, has gained widespread adoption in biomedical research [[59]33–[60]35]. This innovative approach facilitates the analysis of gene expression and regulatory networks at the single-cell level, revealing cellular heterogeneity and dynamic changes under physiological and pathological conditions [[61]36]. Single-cell sequencing affords the capacity to dissect distinct neural cell types, including neurons and glial cells, and their roles in disease progression, offering invaluable insights into neurodegenerative conditions such as Alzheimer’s [[62]37]. NADPH oxidase 4 (NOX4), an enzyme with protein catalytic activity that generates reactive oxygen species (ROS), exerts pivotal roles in various physiological and pathological processes encompassing cell proliferation, migration, and cell death [[63]38–[64]40]. Current research highlights NOX4’s potential significance in neurodegenerative diseases, including Parkinson’s and Alzheimer’s diseases. Nonetheless, the precise mechanisms by which NOX4 influences Alzheimer’s, particularly its association with astrocytic iron-mediated cell death, remain to be fully elucidated [[65]41, [66]42]. This study’s primary objective is to elucidate the critical role of NADPH oxidase 4 (NOX4) in iron-triggered astrocytic cell death and its implications for Alzheimer’s disease (AD) pathogenesis. Employing single-cell sequencing technology, the GEO database, and transcriptome sequencing data, this study delves deeply into the cellular populations and associated genes of AD patients. Researchers meticulously label and analyze various cell types, identifying distinctive marker genes for astrocytes. Furthermore, this study uncovers NOX4’s pivotal role in iron-induced astrocyte demise. These findings substantially augment our comprehension of Alzheimer’s disease etiology, particularly elucidating the nexus between NOX4, astrocytic iron-mediated death, and AD. Importantly, these insights hold clinical relevance for the diagnosis and treatment of Alzheimer’s disease. Materials and methods Transcriptome sequencing data acquisition The single-cell transcriptome sequencing data of AD-related samples in the [67]GSE164089 dataset was analyzed using the Seurat package in R software. To ensure data quality, quality control criteria were applied, including nFeature_RNA > 500, 1000 < nCount_RNA < 20,000, and percent.mt < 10%. Additionally, the top 1000 highly variable genes were selected based on their variance. Furthermore, AD-related microarray dataset [68]GSE48350 was obtained from the GEO database ([69]https://www.ncbi.nlm.nih.gov/geo/). This dataset consists of 173 normal brain tissue samples and 80 AD brain tissue samples [[70]43]. TSNE clustering analysis To reduce the dimensionality of scRNA-Seq datasets, we employ principal component analysis (PCA) based on the top 1000 genes with the highest variance in expression. We used the Elbowplot function of the Seurat package and selected the top 15 principal components for downstream analysis. Using the FindClusters function provided by Seurat, we identified different subpopulations of cells with the default resolution (res = 0.5). Next, we use the t-SNE algorithm to reduce nonlinear dimensionality on scRNA-seq sequencing data. We also used the Seurat package to identify marker genes for individual cell subpopulations and combined the single package with the online website CellMarker ([71]http://xteam.xbio.top/CellMarker) for cell type annotation analysis [[72]44, [73]45]. GO and KEGG enrichment analysis The differential expression genes (DEGs) were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis using the “clusterProfiler”, “org.Hs.eg.db”, “enrichplot”, and “ggplot2” packages in the R language. Bubble plots and circular plots were generated to visualize the enrichment results of the three categories, namely Biological Processes (BP), Cellular Components (CC), and Molecular Functions (MF), in the Gene Ontology (GO). Additionally, a bubble plot was generated to display the enrichment results of the KEGG pathway analysis [[74]46]. Differential gene expression screening The “limma” package in R software was utilized for the selection of differentially expressed genes. Differentially expressed genes between normal samples and AD samples were filtered based on the criteria |logFC| > 0 and P.adjust < 0.05 [[75]47]. Lentivirus infection To construct a lentivirus-mediated NOX4 silencing vector, the pSIH1-H1-copGFP (sh-) interference vector (catalog number SI501A-1, System Biosciences, USA) was purchased. The silencing sequence can be found in Table [76]S1. The lentiviral particles carrying the vector were packaged into HEK-293T cells (CRL-3216, ATCC, USA) using the lentivirus packaging reagent kit (catalog number A35684CN, Invitrogen, USA). After 48 h, the supernatant was collected to obtain lentivirus with a titer of 1 × 10^8 TU/ml. Researchers interested in rapidly and efficiently constructing a lentiviral vector that mediates NOX4 silencing may consider adopting the methodology utilized in our laboratory [[77]48, [78]49]. Cell culture and screening Human normal astrocytes were purchased from ATCC (ATCC, USA) and cultured in human astrocyte medium (catalog number 1801, ScienCell, USA). The medium consisted of a basal medium (catalog number 1801), 2% (v/v) fetal bovine serum (FBS, catalog number 0010), 1% (v/v) astrocyte growth supplement (AGS, catalog number 1852), and 1% (v/v) penicillin/streptomycin solution (P/S, catalog number 0503). The cells were cultured at 37 °C in a 5% CO2 incubator. To simulate Aβ-induced neuronal injury, the cells were treated with Aβ25–35 peptide (catalog number A107853-25 mg, Aladdin, Shanghai, China) at a concentration of 20 μM for 24 h. The cells were divided into the following groups: control, AD, AD + sh-NC (infected with negative control lentivirus expressing sh-NC), and AD + sh-NOX4 (infected with lentivirus expressing sh-NOX4). After adding 1 × 10^5 TU lentivirus to the astrocytes, the cells were incubated for 48 h, except for the control group, which was incubated for an additional 24 h in the medium containing 20 μM Aβ25–35 peptide [[79]50, [80]51]. Alzheimer’s disease APP/PS1 mouse model The male transgenic mice with overexpression of human amyloid precursor protein (APP) and mutant forms of presenilin 1 (PS1) were purchased from Jackson Laboratory (Bar Harbor, ME, USA, Stock #034829). The wild-type C57 male mice were purchased from Weitonlihua Experimental Animal Technology Co., Ltd. in Beijing, China for the Alzheimer’s disease model experiments. They were maintained under non-pathogenic conditions at a temperature of 26–28 ℃ and humidity of 50–65%, with free access to food and water. All mice were acclimated for one week prior to the experiments. The experimental procedures were conducted in accordance with ethical standards and were approved by our institution’s Animal Ethics Committee. For the experiments, the mice were randomly divided into five groups: WT, APP/PS1, APP/PS1 + sh-NC, APP/PS1 + sh-NOX4, and APP/PS1 + sh-NOX4 + erastin, with six mice in each group. To silence NOX4 in the neurons in vivo, sh-NOX4 (4 × 10^5 TU) or sh-NC (4 × 10^5 TU) was slowly injected into the bilateral hippocampi of APP/PS1 mice. In the APP/PS1 + sh-NOX4 + erastin group, erastin (10 μM; HY-15,763, MedChem Express, New Jersey, USA) was dissolved in a water bath at 37℃ with gentle shaking, and then 5% dimethyl sulfoxide with corn oil (C8267, Sigma-Aldrich, USA) was added. The mice were treated for 20 days. At the end of the experiment, all mice were euthanized with an overdose of anesthetic (pentobarbital sodium). The tissues were processed by perfusing the ascending aorta with a 0.9% sodium chloride solution, followed by fixation of the brain tissues in 4% paraformaldehyde solution and embedding in paraffin [[81]38, [82]52–[83]54]. The behavior test experiment was finished, and the above processing methods were executed. Immunofluorescent staining The brain tissue embedded in paraffin was sectioned into 4 μm thick slices and permeabilized using 0.5% Triton-X (T8787, Sigma-Aldrich, USA). The slices were then blocked in CAS-Block™ tissue blocking reagent (008120, Thermo Fisher Scientific, Waltham, MA, USA). Immunostaining was performed using the following antibodies: rabbit anti-GFAP antibody (ab207165, Abcam, Cambridge, UK), rabbit anti-NOX4 antibody (MA5-32090, 1:50, ThermoFisher, USA), rabbit anti-4-HNE antibody (MA5-45789, 1:50, ThermoFisher, USA), and rabbit anti-malondialdehyde (MDA) antibody (MA5-45803, 1:50, ThermoFisher, USA), as well as rabbit monoclonal anti-GFAP antibody (ab207165, Abcam, Cambridge, UK). The slices were then incubated with secondary antibodies goat anti-rabbit IgG (H + L) Alexa Fluor 488 (A11008, 1:100, Thermo Fisher Scientific, USA) and goat anti-mouse IgG (H&L) Texas Red (ab6787, 1:100, Abcam, Cambridge, UK) at 25 °C for 2 h. Nuclear staining was performed using Fluoroshield™ with DAPI (F6057, Sigma-Aldrich, USA). The stained brain tissue sections were analyzed using THUNDER Imager Tissue (Leica Microsystems Ltd., Wetzlar, Germany) and quantified using LAS X imaging software (Leica Microsystems Ltd, Wetzlar, Germany) and ImageJ software v1.52a (Bethesda, MD, USA) [[84]38]. Flow cytometric cell sorting The mouse brain tissue samples were cut into small pieces and digested at 37 °C in PBS solution containing 0.8 mg/mL Collagenase IV (Merck, C4-BIOC, USA). After a wash with PBS buffer, the cell suspension was filtered through a 50 μm sieve to remove residual solid components such as organic matter and neurons. The cell suspension was then centrifuged at 1000 rpm for 5 min. The supernatant was discarded, and the cell pellet was retained. Washing with cell culture medium was performed, followed by grinding the remaining tissue cell clusters using a homogenizer. After repeated washes, the cell suspension was added to approximately 1 mL of washing buffer for selection. Subsequently, the cell suspension was placed in a column containing magnetic beads labeled with S100β antibody. In the spatial sliding column, cells were bound to the S100β antibody (# 9550 S, Cell Signaling Technology, Danvers, MA, USA). Through negative selection, non-stellar glial cells were removed while stellar glial cells were retained. Subsequent washing steps were performed to eliminate cells and impurities not bound to the magnetic beads. The cell suspension was then transferred to a sterile culture dish for inspecting the integrity and activity of stellar glial cells under a microscope. After identifying and collecting the target cell samples, the cells were fixed using PBS solution containing 3.7% formaldehyde. Permeabilization of the fixed cells was done using 0.1% Triton X-100, followed by labeling of the stellar glial cells using GFAP antibody (ab207165, Abcam, Cambridge, UK). The labeled cell samples were injected into a flow cytometer, and cell fluorescence intensity was measured by laser excitation to obtain a purity of 90% for stellar glial cells. Finally, the collected stellar glial cells were subjected to Western blot, lipid peroxidation detection, and measurement of iron ion content using the same culture conditions [[85]55]. Western blot Tissue total protein was extracted using RIPA lysis buffer (P0013C, Beyotime, Shanghai, China) containing PMSF. The extraction process involved incubation on ice for 30 min, followed by centrifugation at 4 °C and 8000 g for 10 min to collect the supernatant. The total protein concentration was measured using a BCA assay kit (Catalog number: 23,227, ThermoFisher, USA). 50 μg of protein was dissolved in 2x SDS loading buffer and boiled for 5 min at 100 °C prior to SDS-PAGE gel electrophoresis. The proteins were then transferred to a PVDF membrane. The PVDF membrane was blocked with 5% non-fat milk at room temperature for 1 h, followed by incubation overnight at 4 °C with the diluted primary antibodies: rabbit anti-GPX4 (ab125066, 1:1000, Abcam, Cambridge, UK), rabbit anti-NOX4 (MA5-32090, 1:1000, ThermoFisher, USA), rabbit anti-4-HNE antibody (MA5-45789, 1:1000, ThermoFisher, USA), rabbit anti-malondialdehyde (MDA) antibody (MA5-45803, 1:1000, ThermoFisher, USA), rabbit anti-SAT1 antibody (ab105220, 1:500, Abcam, Cambridge, UK), rabbit anti-FTH1 antibody (PA5-27500, 1:500, ThermoFisher, USA), rabbit anti-SLC7A11 (711,589, 1:1000, ThermoFisher, USA), rabbit anti-ACSL4 (PA5-27137, 1:1000, ThermoFisher, USA), and rabbit anti-GAPDH (ab181602, 1:10000, Abcam, Cambridge, UK) as internal references. Subsequently, the membrane was washed three times with TBST