Abstract Background Acute myeloid leukemia (AML) is a highly heterogeneous disease characterized by complex genetic and molecular features that contribute to poor prognosis and low cure rates. Therefore, identifying novel therapeutic targets is crucial for improving treatment efficacy and patient survival. This study investigated the potential role of cyclin-dependent kinase 9 (CDK9), a known regulator of gene expression, in AML pathogenesis and prognosis. Methods This study employed multiple bioinformatics approaches, including analysis of CDK9 expression across various cancers using the Tumor Immune Estimation Resource (TIMER2.0) database and further investigation of its expression and prognostic significance in AML using data from the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. Survival analysis and Cox regression analysis were used to assess the association between CDK9 expression and patient prognosis. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were performed to elucidate the pathways and biological processes influenced by CDK9. Furthermore, the relationship between CDK9 expression and tumor immune infiltration was evaluated, and a protein–protein interaction (PPI) network was constructed. In vitro experiments, including Western blotting, CCK-8 assays, and flow cytometry, were conducted to validate the bioinformatics findings. Results Bioinformatics analysis revealed significantly elevated CDK9 expression in AML samples, which correlated with poor patient prognosis. Functional enrichment analysis indicated that CDK9 is involved in key pathways related to cell proliferation, differentiation, and the tumor microenvironment. Moreover, the study observed a strong correlation between CDK9 expression and altered immune cell infiltration, suggesting a potential role in immune evasion. In vitro experiments confirmed that CDK9 overexpression promoted AML cell proliferation and inhibited apoptosis. Additionally, CDK9 showed a strong correlation with epithelial-mesenchymal transition (EMT)-related proteins, suggesting a potential role in AML progression and the EMT process. Conclusions This study demonstrates that CDK9 is a potential prognostic biomarker and therapeutic target in AML. Its involvement in multiple key pathways during AML development and its influence on the tumor immune microenvironment support further exploration of CDK9-targeted therapies to improve AML treatment outcomes. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-02841-4. Keywords: CDK9, Acute myeloid leukemia, Biomarkers, Immune infiltration, Prognosis Introduction Acute myeloid leukemia (AML) is a clonal hematopoietic malignancy originating from multipotent stem cells or early progenitor cells and is the most common acute leukemia in adults. It is characterized by the accumulation of leukemic blasts in the bone marrow and blood, with infiltration into organs and tissues [[40]1]. Prognosis, beyond age and comorbidities, is largely determined by AML's intrinsic biological characteristics [[41]2]. Recent advances in understanding AML biology and treatment have led to several therapeutic strategies that significantly benefit patients [[42]3]. However, AML prognosis remains poor, with a 5-year survival rate below 30% and a high relapse risk, presenting significant challenges [[43]4]. Therefore, discovering novel therapeutic targets and developing more effective treatments are crucial for improving patient outcomes. Cyclin-dependent kinases (CDKs) are serine/threonine (Ser/Thr) protein kinases essential for cell growth, proliferation, and transcriptional regulation [[44]5–[45]7]. Cyclin-dependent kinase 9 (CDK9), a member of the CDK family, regulates cell cycle progression by forming complexes with cyclin T1. These complexes, known as positive transcriptional elongation factor b (P-TEFb), control transcriptional elongation and mRNA maturation [[46]8]. CDK9 is frequently overexpressed in various cancers and is closely associated with tumorigenesis, invasion, metastasis, and prognosis, making it a promising cancer therapeutic target [[47]9–[48]12]. The importance of CDK9 has driven the development of CDK9 inhibitors, which have shown promising efficacy in preclinical studies across multiple cancers, including some demonstrating potential therapeutic value in AML [[49]13–[50]15]. However, previous studies on the role of CDK9 in AML have primarily focused on its involvement in pathways affecting anti-apoptotic factors such as myeloid cell leukemia 1 (MCL-1), B-cell lymphoma-2 (Bcl-2), and X-linked inhibitor of apoptosis protein (XIAP), which are crucial for cancer cell survival [[51]16, [52]17]. The prognostic potential of CDK9 in AML and its potential immunomodulatory functions, among other molecular mechanisms, warrant further investigation. This study integrates bioinformatics analysis to assess the diagnostic and prognostic significance of CDK9, coupled with Western blotting to detect CDK9 expression in AML cell lines and its correlation with epithelial-mesenchymal transition (EMT)-related proteins. In vitro CCK-8 proliferation assays and flow cytometry were used to analyze the impact of CDK9 on cell proliferation and apoptosis. Furthermore, we constructed a CDK9 co-expression network and performed functional enrichment analysis to identify associated signaling pathways and explore the regulatory role of CDK9 in the AML immune microenvironment. Through this integrated analytical approach, we aim to provide valuable insights into the significant role and underlying mechanisms of CDK9 in AML. Materials and methods Data download AML datasets were retrieved from The Cancer Genome Atlas (TCGA)-LAML via the Cancer Genomics Hub (CGHub) and TCGA data portal ([53]https://portal.gdc.cancer.gov/), comprising 150 AML samples [[54]18]. Clinical data corresponding to TCGA-LAML samples were obtained from the UCSC Xena database ([55]http://genome.ucsc.edu) [[56]19]. Additionally, the TCGA_GTEx-LAML dataset, which included 70 GTEx normal samples and 173 TCGA tumor samples, was sourced from the Xena database. RNA expression data were normalized using log2(value + 1). CDK9 expression analysis in AML Pan-cancer analysis was first performed using the"Gene_DE module"of the Tumor Immune Estimation Resource (TIMER2.0, [57]https://cistrome.shinyapps.io/timer/) to assess CDK9 expression in various tumor and normal tissues [[58]20]. To further validate its expression in AML, we evaluated the differential expression between 173 AML samples and 70 normal samples from the TCGA_GTEx-LAML dataset. Data visualization was performed using the R package “ggplot2” (v3.4.4). Clinical correlation analysis of CDK9 expression in AML The TCGA-LAML dataset was used to analyze the differential expression of CDK9 across various clinical subgroups in AML to assess the correlation between CDK9 expression levels and clinicopathological characteristics. The Wilcoxon rank-sum test was used for clinical variables with only two groups (e.g., age, FL3 mutation), while for variables with more than two groups, a one-way ANOVA was performed, followed by Tukey's post-hoc test for multiple comparisons. Data visualization was performed using the R packages ggplot2, stats (v4.2.1), and car (3.1-0). Independent prognostic and diagnostic value in AML patients Survival analyses involved 139 AML samples with complete survival data from the TCGA-LAML dataset. Kaplan–Meier (K-M) survival curves were plotted using the survminer R package (v0.4.9) to examine the association between CDK9 expression and survival. Diagnostic performance in distinguishing AML patients from healthy individuals was evaluated by plotting receiver operating characteristic (ROC) curves using “ggplot2.” Univariate and multivariate Cox regression analyses were performed with the “survival” package (v3.3.1), and forest plots were visualized using “ggplot2.” The proportional hazard assumption was tested using the “survival” package, and Cox regression analyses predicted overall survival. A nomogram was constructed and visualized using the “rms” package (v6.3-0). Risk scores were calculated based on the median expression of CDK9 and visualized with “ggplot2.” Predictive performance was further validated with calibration curves and time-dependent ROC (tROC) curves using the “rms,” “survival,” “ggplot2,” and “timeROC (v0.4)” packages. Co-expression analysis of CDK9 in AML The expression and transcriptional landscape of CDK9 in AML was analyzed using the LinkedOmics online platform ([59]http://www.linkedomics.org/login.php) [[60]21]. We identified 9120 and 10,328 genes down-regulated and up-regulated in co-expression with CDK9, respectively. A heatmap visualized the top 50 most significantly downregulated and upregulated genes. Subsequently, Pearson correlation analysis assessed the strength of correlation among the top three up-regulated and down-regulated genes, with visualization using"ggplot2.". Gene ontology (GO) and Kyoto rncyclopedia of genes and genomes (KEGG) enrichment analyses AML patients in the TCGA-LAML dataset were divided into low and high CDK9 expression groups based on the median expression level. The DESeq2 R package (v1.36.0) was then used to identify 126 differentially expressed genes (DEGs) with thresholds of |log2-fold-change (FC)| > 2 and adjusted P-value < 0.05. Metascape ([61]http://www.metascape.org/) was used for functional annotation of CDK9 and its associated genes from the TCGA-LAML dataset [[62]22]. Gene Ontology (GO) enrichment analysis categorized biological processes (BP), cellular components (CC), and molecular functions (MF), identifying CDK9's roles in key metabolic pathways. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis identified significant pathways associated with CDK9. Gene set enrichment analysis (GSEA) Gene set enrichment analysis (GSEA) was performed using the “clusterProfiler” (v4.4.4) R package, with significance determined by an adjusted P-value < 0.05 and FDR q-value < 0.25, to identify functional and pathway differences between the high and low CDK9 expression groups. CDK9 expression, immune infiltration, and protein interaction network analysis To assess the potential relationship between CDK9 expression and immune cell infiltration, single-sample gene set enrichment analysis (ssGSEA) was performed using the"GSVA"package (v1.46.0) for 24 immune cell types in the context of CDK9. The Wilcoxon rank-sum test compared enrichment scores for the 24 immune cell types between high and low CDK9 expression groups. Spearman correlation analysis evaluated the correlation between CDK9 expression and immune cell infiltration. A protein–protein interaction network was constructed using the STRING database ([63]http://string-db.org/) by setting the minimum confidence to “medium confidence (0.400)” and using other default parameters [[64]23]. Cell culture Human AML cell lines (HL-60, THP-1, Kasumi-1) and normal HMSC-BM bone marrow cells were obtained from Anhui Baioujing Technology Co., LTD., China. Cells were cultured in 1640 medium supplemented with 10% fetal bovine serum and 1% penicillin–streptomycin at 37 °C in a 5% CO2 incubator. Cells were passaged when the growth density reached 80–90%. Cell transfection THP-1 cells at the logarithmic growth stage were seeded into 6-well plates at a density of 1 × 10^5 cells per well. Transfections were performed for 24–48 h using the NanoTrans 60™ transfection kit (China Biomedical). Cells were grouped as NC, OE-CDK9, siCtrl, siCDK9-1, siCDK9-2, and siCDK9-3 based on transfection conditions, following the manufacturer’s instructions. After transfection, cells were incubated at 37 °C for 48 h. CDK9 protein expression levels were assessed using Western blot analysis. Compared to the siCtrl group, the siCDK9-1, siCDK9-2, and siCDK9-3 groups exhibited reduced CDK9 protein levels, while the OE-CDK9 group showed increased CDK9 expression relative to the NC group, confirming successful CDK9 plasmid transfection. Quantitative RT-qPCR analysis cDNA synthesis was performed using AG RNAex Pro RNA extraction reagent (Ecorui Bio, China), followed by PCR amplification using SYBR Green Pro Taq HS premix (Ecorui Bio, China). Real-Time PCR conditions and reaction details are listed in Supplementary Table [65]S1. Each sample was analyzed in triplicate, with GAPDH serving as the internal reference gene for both experimental and control groups. Relative gene expression levels were calculated using the 2^−ΔΔCt method. Primer sequences are provided in Supplementary Table [66]S2. Western blot assay Total protein was extracted from cells or tissues using RIPA lysis buffer (R0010, Solarbio). Protein concentration was determined with the BCA assay (P0010, Beyotime). A total of 30 μg of protein was separated by SDS-PAGE and transferred to PVDF membranes. Membranes were blocked with 5% skim milk in PBS and incubated overnight at 4 °C with primary antibodies. They were then incubated with Goat anti-Rabbit IgG-HRP (abs20040, Ambition) or Goat anti-Mouse IgG-HRP (abs20039, Ambition) at 37 °C for 2 h. Immunoreactive bands were visualized using an ECL kit (PK10002, Proteintech). Cell proliferation test Cell Counting Kit-8 (CCK-8) assay was performed. Cells at the logarithmic growth phase were seeded into 96-well plates at a density of 1 × 10^4 cells per well. After incubation, MTT solution (5 g/L) was added and incubated for 4 h. Following centrifugation and removal of the supernatant, 150 μL of DMSO was added to each well and incubated in the dark for 15 min. Absorbance at 490 nm was measured with a microplate reader to evaluate cell proliferation activity. Apoptosis test Flow cytometry was used to analyze apoptosis. Cells at the logarithmic growth phase were seeded into 24-well plates at a density of 1 × 10^5 cells per well. After transfection and 48 h of incubation, cells were collected, washed with cold PBS, and resuspended in 100 μL of Binding Buffer. Cells were stained with 5 μL of Annexin V-FITC and 5 μL of PI and incubated in the dark at room temperature for 10 min. Apoptosis rates were measured using flow cytometry within 1 h of staining. Statistical analysis Statistical analyses were conducted using GraphPad Prism (v9.5.1), R software (v4.2.1), and online tools. Kaplan–Meier survival curves were analyzed with the log-rank test. Correlation coefficients were calculated using Pearson or Spearman tests, as appropriate. Data are presented as mean ± standard deviation (SD) from at least three independent experiments. Statistical significance was defined as P < 0.05 (*P < 0.05; **P < 0.01; ***P < 0.001; N.S., not significant). Results CDK9 expression analysis in pancarcinoma and AML Using the TIMER2.0 database, we observed differential CDK9 expression between tumor and adjacent normal tissues across 33 different cancers (Fig. [67]1A). Significantly different CDK9 expression (P < 0.05) was observed in ten cancer types: breast invasive carcinoma (BRCA), cholangiocarcinoma (CHOL), esophageal carcinoma (ESCA), head and neck squamous cell carcinoma (HNSC), clear cell renal cell carcinoma (KIRC), lung adenocarcinoma (LUAD), rectum adenocarcinoma (READ), stomach adenocarcinoma (STAD), thyroid carcinoma (THCA), and uterine corpus endometrial carcinoma (UCEC). However, due to the lack of normal AML samples in this database, we used the TCGA_GTEx-LAML dataset to compare CDK9 expression in 173 AML samples and 70 normal samples. Results (Fig. [68]1B) showed significantly higher CDK9 expression in AML samples compared to normal samples (P < 0.001). We further investigated the correlation between CDK9 expression and clinicopathological features (Fig. [69]1C–H). CDK9 expression was significantly lower in AML samples with t(8;21) chromosomal abnormalities compared to those with normal cytogenetics. Higher CDK9 expression was observed in the intermediate/normal and poor cytogenetic risk groups compared to the favorable group. The M1 FAB subtype showed higher CDK9 expression than the M4 subtype. No significant differences in CDK9 expression were observed among other clinicopathological subgroups. Fig. 1. [70]Fig. 1 [71]Open in a new tab Differential expression analysis of CDK9. A Differential expression of CDK9 in tumor and adjacent normal tissues across 33 different cancers in the TIMER2 database; B Difference in CDK9 expression levels between AML and normal tissues based on the TCGA_GTEx-LAML dataset; C-H correlation between CDK9 expression levels and clinicopathological features of AML, including age, FLT3 mutation, NPM1 mutation, prognostic stratification, and FAB classification. (*p < 0.05; **p < 0.01; ***p < 0.001) Prognostic analysis of CDK9 in AML AML patients from the TCGA database were categorized into high- and low-CDK9 expression groups based on average CDK9 expression levels. Kaplan–Meier (K-M) survival analysis revealed that patients in the low-CDK9 expression group had better survival rates (P < 0.05) (Fig. [72]2A), indicating that low CDK9 expression is associated with a favorable prognosis. ROC curve analysis demonstrated high accuracy of CDK9 expression in assessing tumorigenesis (Fig. [73]2B). Univariate Cox regression analysis showed significant correlations between AML prognosis and CDK9 expression, age, and prognostic stratification (Fig. [74]2C, Supplementary Table [75]S3). Fig. 2. [76]Fig. 2 [77]Open in a new tab The prognostic and diagnostic value of CDK9 in AML. A CDK9 expression and survival analysis for patient prognosis; B ROC curves illustrating the diagnostic value of CDK9; C Screening of clinically relevant prognostic features associated with AML prognosis; D Nomograph prediction model based on CDK9 expression levels and associated clinical features; E Calibration curves with one, two, and three years for the Cox regression prediction model; F Risk scores and maps of survival status for prognostic models that are clinically relevant; G Time-dependent ROC curve results of risk scores versus patient prognostic OS for clinically relevant prognostic models To develop a prognostic model, CDK9 expression was combined with clinical characteristics. The model demonstrated higher predictive value for age (Fig. [78]2D). Multivariate analysis confirmed the model's predictive performance. Prognostic calibration curves indicated high predictive accuracy (Fig. [79]2E), and risk factor plots visually displayed prediction results (Fig. [80]2F). Time-dependent ROC analysis further validated the model's predictive accuracy for 1-, 2-, and 3-year survival rates (Fig. [81]2G). CDK9 coexpression network To investigate the potential mechanisms of CDK9 in AML, a co-expression network analysis was performed using the LinkedOmics database. This identified 10,328 genes positively correlated and 9,120 genes negatively correlated with CDK9 expression (Fig. [82]3A). A heatmap highlighted the top 50 positively and negatively correlated genes (Fig. [83]3B, C), emphasizing CDK9's impact on the transcriptome. The top three positively correlated genes were WBP1 (R = 0.660, P < 0.001), FPGS (R = 0.819, P < 0.001), and RASGRP2 (R = 0.737, P < 0.001), while the top three negatively correlated genes were CPSF2 (R = − 0.201, P = 0.013), ZMPSTE24 (R = − 0.225, P = 0.006), and PGM2 (R = − 0.103, P = 0.211) (F[84]ig. [85]3D–I). Fig. 3. [86]Fig. 3 [87]Open in a new tab Coexpression network of CDK9 in LAML. A Volcano plot showing CDK9 coexpressed genes; B, C Heat maps illustrating the top 50 genes positively and negatively correlated with CDK9 expression in AML; D–I Pearson correlation scatter plots showing the relationship between CDK9 expression and the expression levels of WBP1, FPGS, RASGRP2, CPSF2, ZMPSTE24, and PGM2 CDK9 differential gene screening, functional enrichment, and pathway analysis The TCGA-LAML dataset was stratified into high and low CDK9 expression groups. R software was used to identify 126 differentially expressed genes (DEGs) using a cutoff of |log2 FoldChange|> 2 and P < 0.05. GO functional annotation and KEGG pathway enrichment analyses of these DEGs revealed enrichment in biological processes such as cell differentiation and signal receptor binding (Fig. [88]4A–C). KEGG analysis highlighted the involvement of cancer pathways, TGF-β signaling, and cytokine-cytokine receptor interaction (Fig. [89]4D). Gene Set Enrichment Analysis (GSEA) comparing the high and low CDK9 expression groups identified key AML-related pathways, including mitochondrial complex III assembly (Fig. [90]4E), downstream CD8 signaling (Fig. [91]4F), BCR signaling (Fig. [92]4G), and corticotropin-releasing hormone signaling (Fig. [93]4H). These findings suggest a role for CDK9 in immune infiltration and highlight its importance in AML immunooncology. Fig. 4. [94]Fig. 4 [95]Open in a new tab Identification of CDK9 DEGs and enrichment analysis of biological functions. A-C GO bubble plots; D KEGG enrichment analysis bubble plot; E Mitochondrial Complex III assembly pathway; F CD8 downstream signaling pathway; G BCR signaling pathway. H corticotropin-releasing hormone signaling pathway Relationship between CDK9, immune cell infiltration, and the protein interaction network To further explore the role of CDK9 expression in AML immune infiltration, the ssGSEA algorithm was used to estimate the infiltration proportions of 24 immune cells in each sample. Seven immune cells showed differential infiltration between high and low CDK9 expression groups (Fig. [96]5A). High CDK9 expression was associated with increased infiltration of NK CD56bright cells, TFH cells, Treg cells, CD8 T cells, pDCs, and Th17 cells and decreased infiltration of Th2 cells compared to the low expression group. Spearman correlation analysis was then performed to investigate the correlation between CDK9 and immune cells. Setting a threshold of |R|≥ 0.3 and p < 0.05 (as |R|< 0.3 indicates weak correlation), NK CD56bright cells (R = 0.439, P < 0.001) and follicular helper T cells (TFH) (R = 0.300, P < 0.001) were found to be significantly correlated with CDK9 (Fig. [97]5B, C). A CDK9 protein–protein interaction (PPI) network was constructed using the STRING database to assess its functional impact on AML progression. The top 10 proteins identified in the PPI analysis were MLLT1, CCNT1, BRD4, CCNT2, AFF4, HEXIM1, CCNK, AFF1, MEPCE, and MED12 (details in Supplementary Table [98]S4, Fig. [99]5D). Fig. 5. [100]Fig. 5 [101]Open in a new tab Relationship between CDK9 and immune cell infiltration and protein interaction network. A Assessment of immune infiltration levels in high and low CDK9 expression groups; B correlation of CDK9 gene expression with immune cell infiltration; C Scatter plots showing the correlation between CDK9 and NK CD56bright cells and TFH cells; D Protein–protein interaction (PPI) network of CDK9 Effect of CDK9 on proliferation and apoptosis in AML cells Western blot analysis showed elevated CDK9 protein levels in HL-60, THP-1, and Kasumi-1 cells compared to HMSC-BM cells (Fig. [102]6A). CDK9 knockdown (siCDK9-1, siCDK9-2, siCDK9-3) and overexpression (OE-CDK9) plasmids were introduced in THP-1 cells. CDK9 protein expression decreased in the siCDK9 groups compared to siCtrl, while it increased in the OE-CDK9 group compared to NC (Fig. [103]6B). Fig. 6. [104]Fig. 6 [105]Open in a new tab AML cell proliferation is stimulated, apoptosis is inhibited, and the expression of proteins related to EMT is regulated by CDK9. A The abundance of CDK9 protein cells was detected by WB; B WB identified CDK9's interference and overexpression efficiency; C CCK8 cell viability was detected (Each experimental group's OD450 multiplicity from day 1 to day 5 in relation to day 1 is expressed as OD450/fold, which shows the multiplicity of proliferation on each day.); D Apoptosis was detected by flow cytometry. E qPCR and WB were used to detect the expression of EMT-related proteins. (*p < 0.05; **p < 0.01; ***p < 0.001) CCK-8 assays demonstrated that OE-CDK9 significantly enhanced THP-1 proliferation (**P < 0.01), while siCDK9 significantly reduced it (**P < 0.01) (Fig. [106]6C). Flow cytometry analysis indicated that CDK9 overexpression inhibited apoptosis, whereas CDK9 knockdown promoted apoptosis (Fig. [107]6D). Subsequently, epithelial-mesenchymal transition (EMT)-related protein expression was assessed. In the OE-CDK9 group, mRNA and protein levels of E-cadherin decreased significantly, while N-cadherin, Vimentin, and Snail1 levels increased compared to NC. Conversely, the siCDK9 group showed the opposite trends, with increased E-cadherin and decreased N-cadherin, Vimentin, and Snail1 levels (Fig. [108]6E). Discussion Recent years have witnessed extensive research dedicated to elucidating the pathogenesis and treatment of AML, aiming to identify novel clinical targets and therapeutic agents with improved specificity and reduced side effects [[109]24, [110]25]. CDK9, the catalytic subunit of P-TEFb, has been shown to be widely expressed in various cancers and plays a crucial role in their development and progression, including some hematological malignancies and solid tumors [[111]13]. Given CDK9's established role in various malignancies, we first investigated its differential expression between normal and tumor samples across different cancer types using public databases. We observed differential CDK9 expression in 10 cancer types with comparable sample availability. However, due to the lack of normal AML samples, we further evaluated CDK9 expression using the TCGA_GTEx_LAML database, revealing increased CDK9 expression in AML patients. Interestingly, a recent study reported no significant difference in CDK9 expression between AML and normal samples [[112]26]. This discrepancy may be attributed to the different data filtering methods used; they employed the GEPIA database. Subsequently, Western blot analysis confirmed our findings, demonstrating increased CDK9 expression in AML, thus highlighting its potential significance. We further investigated the association between CDK9 expression and AML clinicopathological features and prognosis. Prognostic stratification and French-American-British (FAB) subtypes were significantly associated with CDK9 expression in AML patients. Survival analysis and both univariate and multivariate Cox regression analyses revealed that high CDK9 expression was correlated with poor prognosis. A nomogram prediction model further underscored the importance of CDK9 expression in predicting AML prognosis. In summary, these results highlight the significance of CDK9 as a prognostic biomarker and potential therapeutic target in AML. To explore the potential functional mechanisms of CDK9 in AML, we analyzed CDK9 co-expressed genes in the TCGA-LAML dataset using the LinkedOmics database. The results showed that a large number of genes were significantly positively or negatively correlated with CDK9 expression, suggesting a broad impact of CDK9 on the AML transcriptome. Specifically, FPGs showed a significant positive correlation with CDK9, consistent with previous reports associating FPGs with poor prognosis in acute leukemia [[113]27]. Subsequently, we stratified the TCGA-LAML dataset into high and low CDK9 expression groups and identified 126 differentially expressed genes (DEGs). GO functional enrichment analysis revealed that these DEGs were significantly enriched in biological processes such as cell differentiation and signal receptor binding. KEGG pathway enrichment analysis showed that CDK9 is involved in the regulation of cancer-related pathways, the TGF-β signaling pathway, and cytokine-cytokine receptor interactions. Furthermore, GSEA revealed significant associations between CDK9 and key pathways, including mitochondrial complex III assembly, downstream CD8 signaling, BCR signaling, and corticotropin-releasing hormone signaling. These results suggest that CDK9 may play a crucial role in AML development and progression by regulating multiple key signaling pathways, influencing AML cell proliferation, differentiation, apoptosis, and the immune microenvironment. The tumor microenvironment (TME) in AML is characterized by immunosuppression, promoting immune tolerance and immune evasion of malignant cells [[114]28, [115]29]. Therefore, studying immune cell infiltration within the tumor may provide valuable insights into the efficacy and mechanisms of immunotherapy in AML. Given the potential impact of CDK9 on the immune microenvironment, we further assessed immune cell changes at different CDK9 expression levels using the ssGSEA algorithm. NK CD56bright cells and TFH cells showed a significant positive correlation with high CDK9 expression. Notably, CD56bright CD16- NK cells are a major component of NK cells in various immune-tolerant organs, and their function is known to be suppressed in the tumor immune environment [[116]30]. Therefore, we hypothesize that high CDK9 expression may lead to increased infiltration of NK CD56bright cells in the TME of AML patients, resulting in tumor immune evasion and ultimately poor prognosis. Furthermore, protein–protein interaction (PPI) analysis identified interactions between CDK9 and proteins such as MLLT1, CCNT1, BRD4, and CCNT2, which also play important roles in AML development and progression, providing new avenues for further investigation into CDK9's mechanism of action. In vitro experiments confirmed CDK9's role in promoting proliferation and inhibiting apoptosis in AML cells. qPCR and western blot analyses of epithelial-mesenchymal transition (EMT)-related proteins in CDK9 knockdown and overexpression groups revealed reduced E-cadherin mRNA and protein levels in the OE-CDK9 group, while N-cadherin, Vimentin, and Snail1 levels were significantly elevated. These findings suggest that CDK9 is closely associated with EMT-related proteins, potentially contributing to AML progression. This study has several limitations. First, our findings are based on analyses of public databases with limited sample sizes. Current datasets lack information on specific epigenetic mutations for all patients, including those in key genes highlighted by the recently updated WHO classification [[117]31], which is crucial for a more detailed understanding of CDK9's mechanism in AML. Future studies should include larger and more comprehensive cohorts for validation. Second, we only confirmed the effects of CDK9 on AML cell proliferation, apoptosis, and EMT marker expression through in vitro experiments. Further validation of CDK9 co-expressed genes, related signaling pathways, and interacting proteins associated with CDK9 expression is lacking. Future studies should include more functional experiments and in vivo studies. Finally, this study did not evaluate the efficacy and sensitivity of immune-targeted drugs and CDK9 inhibitors. Given that studies have shown that selective CDK9 inhibitors and degraders such as Dinaciclib, NVP-2, LDC000067, FIT-039, Atuveciclib, PROTAC 2, and Compound 45 [[118]26, [119]32–[120]36] can inhibit tumor growth in hematological and solid tumors in vitro and in vivo, and that combination therapy may be a promising strategy to enhance the efficacy of selective CDK9 inhibitors and degraders in tumors, future research should explore the efficacy of these drugs in AML and combination therapy strategies to promote the clinical translation of CDK9 inhibitors. Conclusions In conclusion, we have identified a correlation between high CDK9 expression in AML and poor prognosis, suggesting that CDK9 may promote AML development and progression by influencing immune cell infiltration and the EMT process. This study enhances our understanding of CDK9's role in AML. Supplementary Information [121]Additional file 1.^ (26.3KB, docx) Acknowledgements