Multiomic characterization, immunological and prognostic potential of smad3 in pan-cancer and validation in lihc

Multiomic characterization, immunological and prognostic potential of smad3 in pan-cancer and validation in lihc

Play all audios:

Loading...

ABSTRACT SMAD3, a protein-coding gene, assumes a pivotal role within the transforming growth factor-beta (TGF-β) signaling pathway. Notably, aberrant SMAD3 expression has been linked to


various malignancies. Nevertheless, an extensive examination of the comprehensive pan-cancer impact on SMAD3’s diagnostic, prognostic, and immunological predictive utility has yet to be


undertaken. Bioinformatics methods were employed to systematically investigate the potential carcinogenic impact of SMAD3. We extensively harnessed data from authoritative sources, including


The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), cBioPortal, Human Protein Atlas (HPA), UALCAN, and various other databases. Our study encompassed a comprehensive analysis


of the following aspects: differential SMAD3 expression and its association with prognosis across diverse cancer types, gene mutations, immune cell infiltration, single-cell sequencing


analysis, DNA methylation patterns, and drug sensitivity profiles. In vitro experiments were conducted with the primary objective of appraising both the expression profile and the precise


functional attributes of SMAD3 within the milieu of Liver Hepatocellular Carcinoma (LIHC). Our findings revealed significant variations in SMAD3 expression between cancerous and adjacent


normal tissues. High levels of SMAD3 expression were consistently associated with unfavorable prognoses across multiple cancer types,. Additionally, our analysis of SMAD3 methylation


patterns in human cancers unveiled a favorable prognosis linked to elevated DNA methylation levels in pan-cancer. Furthermore, we identified positive associations between SMAD3 expression


and RNAm6A methylation-related genes in the majority of cancers. Moreover, SMAD3 expression displayed substantial correlations with immune cell infiltration. Notably, immune checkpoint genes


exhibited significant associations with SMAD3 expression across diverse cancers. Single-cell sequencing results elucidated the pan-cancer single-cell expression landscape of SMAD3. Within


specific cancer subtypes, SMAD3 expression exhibited a noteworthy positive association with distinctive facets of malignancy. Finally, in our comprehensive analysis of drug sensitivity, we


discerned a catalog of prospective therapeutic agents. In our comprehensive analysis across multiple cancer types, we observed a significant disparity in SMAD3 expression compared to normal


tissues, and this findings suggest that SMAD3 holds promise as both a prognostic biomarker and a therapeutic target against various cancers. Difference displayed a noteworthy association


with patient prognosis. SIMILAR CONTENT BEING VIEWED BY OTHERS LZTS2 METHYLATION AS A POTENTIAL DIAGNOSTIC AND PROGNOSTIC MARKER IN LIHC AND STAD: EVIDENCE FROM BIOINFORMATICS AND IN VITRO


ANALYSES Article Open access 22 May 2025 _ZC3H12D_ GENE EXPRESSION EXHIBITS DUAL EFFECTS ON THE DEVELOPMENT AND PROGRESSION OF LUNG ADENOCARCINOMA Article Open access 18 May 2025 TMEM101


EXPRESSION AND ITS IMPACT ON IMMUNE CELL INFILTRATION AND PROGNOSIS IN HEPATOCELLULAR CARCINOMA Article Open access 30 December 2024 INTRODUCTION Cancer ranks as the second leading global


cause of mortality, responsible for approximately 14.1 million new cases and 8.2 million annual fatalities1. While recent years have witnessed the emergence of immunotherapy and targeted


therapy as novel cancer treatment strategies, their applicability remains limited to a subset of patients2. Moreover, the five-year survival rates continue to be relatively modest3.


Consequently, enhancing the effectiveness of cancer treatment necessitates the identification of more potent biomarkers4. SMAD3 is a pivotal transcription factor that modulates the


transforming growth factor-beta (TGF-β) signal and assumes a dual role in both restraining and promoting cancer. It features highly conserved regions at the N- and C-termini, known as the


mad-homology domain 1 (MH1) and MH2 regions, respectively. The MH1 region primarily governs DNA binding, while the phosphorylated MH2 region plays a critical role in the canonical TGF-β


signaling pathway, As the C-terminal phosphorylated form, known as R-SMAD, SMAD3 functions as an intracellular signaling transducer and transcriptional regulator. Activation is initiated by


TGF-β and activin type 1 receptor kinases. R-SMAD binds to TGF-β responsive elements (TREs) in the promoter regions of numerous TGF-β-regulated genes. Following the formation of the


SMAD3/SMAD4 complex, it translocates to the nucleus to activate transcription, modulate gene activity, and stimulate cell proliferation. Numerous studies have illuminated SMAD3’s involvement


in various disease states, with a growing recognition of its role in cancer5. Research has revealed that SMAD3 promotes the formation of cancer-associated fibroblasts through


macrophage-myofibroblast transition. Chromatin immunoprecipitation sequencing (ChIP-seq) has demonstrated substantial enrichment of Smad3 binding to genes related to fibroblast


differentiation in LLC tumor macrophage lineage cells. Notably, targeted deletion in macrophages and pharmacological inhibition of Smad3 effectively thwart macrophage-myofibroblast


transition, thereby impeding cancer-associated fibroblast formation and cancer progression in vivo6. Other studies have shown that macrophage-specific gene deletion and systemic


pharmacological inhibition of TGF-β1/Smad3/Runx1 signal transduction effectively prevent MMT driven CAF and tumor formation in vitro and in vivo, reflecting the important role of SMAD3 in


MTT driven CAF7. Aberrant SMAD3 activation is a pivotal driver of breast cancer metastasis. This occurs via the acetylation of SMAD3 by KAT6A at K20 and K117, promoting interactions with


TRIM24, and disrupting the interaction with the tumor suppressor TRIM33. Consequently, this leads to heightened SMAD3 activation and the expression of immune response-related cytokines,


culminating in increased breast cancer stemness, myeloid-derived suppressor cells (MDSC), and metastasis in triple-negative breast cancer (TNBC)8. Furthermore, SMAD3 plays a role in


promoting cancer progression by impeding E4BP4-mediated natural killer (NK) cell development9. Subsequent studies unveiled that NK-3-S1KD, a genetically engineered stable Smad3-silenced


human NK cell line, effectively curbs cancer progression in xenografted mouse models with human liver cancer and melanoma10. Elevated SMAD3 expression levels also correlate with the


progression of various cancers. Studies indicate that SEPHS1 promotes SMAD3 expression and enhances invasion of hepatocellular carcinoma cells11. SMAD3 also plays a crucial role in bladder


cancer; for instance, it participates in miR-5581-3p-mediated inhibition of bladder cancer cell migration and proliferation12. Additionally, cirRIP2 accelerates bladder cancer progression


through the miR-1305/TGF-β2/SMAD3 pathway13.Moreover, miR-195 inhibits cervical cancer migration and invasion by targeting SMAD314, while Circ_0001686 promotes prostate cancer progression by


upregulating SMAD3/TGFBR2 via miR-411-5p15. Additionally, numerous drugs have been found to regulate tumor proliferation and metastasis through SMAD3, including CDK4 inhibitors and


doxorubicin, which induce breast cancer cell apoptosis via SMAD3. Docetaxel inhibits SMAD3 binding to the HIF-1α gene promoter, thereby curtailing the transcriptional activation of HIF-1 and


inhibiting cell proliferation16,17. In summation, the significance of SMAD3 in the development of multiple cancers is gaining increasing recognition, rendering it a promising candidate as a


novel diagnostic and prognostic biomarker and therapeutic target for these malignancies. In this study, we conducted a comprehensive bioinformatics analysis of SMAD3-related data. Utilizing


a range of databases including HPA, cBioPortal, UALCAN, GSCA, GEPIA, CancerSEA, TISMO, TISCH, and others, we assessed the expression levels of SMAD3 across various tissues and its


associations with different cancer types. Furthermore, we delved into the prognostic significance, mutation patterns, and single-cell expression profiles of SMAD3. Our analysis extended to


exploring the connections between SMAD3 and immune cell infiltration, immune-related genes, immune checkpoints, immunomodulatory genes, methylation patterns, tumor mutation burden (TMB),


microsatellite instability (MSI), tumor stemness, and drug sensitivity. Additionally, we conducted functional enrichment analysis of genes related to SMAD3 to unveil the underlying molecular


mechanisms contributing to diverse cancers. This study collectively sheds light on the potential of SMAD3 as a prognostic marker in various cancer contexts. Our in-depth in vitro


experiments have provided substantial evidence regarding the atypical expression and biological roles of SMAD3 in Liver Hepatocellular Carcinoma (LIHC). The findings firmly establish SMAD3


as an independent and promising prognostic marker, underscoring its potential as a fresh therapeutic target for LIHC. MATERIALS AND METHODS GENE EXPRESSION ANALYSIS OF SMAD3 IN PAN-CANCER


The TIMER2.0 database18 stands as an intricate network for the assessment of immune cell infiltration within tumor tissues. It not only facilitates the scrutiny of immune cell infiltration


levels in tumors but also enables the comparative examination of SMAD3 expression profiles across various cancer types and their adjacent normal tissues. The unified and standardized


pan-cancer dataset was acquired through retrieval from the UCSC database19.Subsequently, the SMAD3 gene expression data were meticulously extracted from each sample, resulting in


comprehensive profiles across 27 distinct cancer types. Comparative analyses involved assessing expression levels in cancer samples versus paired standard samples across the same 27 cancers.


Subsequently, log2 transformation and t-tests were conducted on the expression data within each tumor type. Significance of expression discrepancies between tumor and normal tissue samples


was determined using the standard criterion of p-value < 0.05. Data analysis utilized R software (Version 4.0.2, https://www.Rproject.org), with visualization accomplished using the


“ggplot2” R package. The Human Protein Atlas (HPA) constitutes a repository of human proteins within the framework of diverse anatomical compartments, encompassing organs, tissues, and


cellular constituents, leveraging a multitude of omics methodologies. The expression of SMAD3 mRNA in normal tissues was detected using the HPA database. In addition, histological and


pathological immunohistochemical images of the SMAD3 protein were obtained from the database, and the subcellular localization of SMAD3 was determined through the utilization of indirect


immunofluorescence microscopy20,21. PROGNOSTIC ANALYSIS OF SMAD3 Our data acquisition process entailed the retrieval of a meticulously standardized pan-cancer dataset originating from the


UCSC database, specifically the TCGA TARGET GTEx dataset, which comprised a substantial cohort (PAN-CANCER _N_ = 19131 G = 60499). Within this dataset, we meticulously extracted the gene


expression data for SMAD3 from each sample. In addition, we secured a high-quality TCGA prognostic dataset, sourced from prior TCGA prognostic studies as published in Cell22. To complement


this dataset, we incorporated the follow-up data obtained from TARGET through UCSC.our study encompassed the examination of various endpoints, including overall survival (OS),


disease-specific survival (DSS), disease-free interval (DFI), and progression-free interval (PFI), to scrutinize the association between SMAD3 expression and cancer prognosis. For this


purpose, we harnessed the robust analytical tools of Kaplan-Meier survival analysis and Cox regression analysis to delve into the intricate relationship between SMAD3 and survival


prognosis23 . DNA METHYLATION AND RNA MODIFICATION ANALYSIS The occurrence and proliferation of cancer are regulated by epigenetic inheritance and genetic events, in which DNA methylation


plays an important role24. We used data from the UALCAN database to determine SMAD3 promoter DNA methylation levels to determine the difference between the cancers and normal tissues. We


also obtained DNA methylation prognostic data from the GSCA database. More and more evidence show that RNA methylation is closely related to cancer cell proliferation, cell stress,


metastasis, and the immune response. RNA methylation-related proteins have become promising targets for cancer therapy25. Pan-cancer data were retrieved from the UCSC database, and from this


dataset, we extracted the expression profiles of the SMAD3 gene along with 44 marker genes related to Class III RNA modification (comprising m1A10, m5C13, and m6A21 genes) within each


sample. Subsequently, Pearson correlation coefficients were computed to elucidate the associations between SMAD3 and these marker genes within the context of five distinct immune pathways.


GENE MUTATION ANALYSIS OF SMAD3 The cBioPortal database26 serves as a repository for the comprehensive compilation of data pertaining to mutation frequency, mutation types, specific mutation


locations, and the three-dimensional (3D) structural attributes of prospective proteins across the entirety of TCGA cancer datasets. We downloaded the uniformly standardized pan-cancer


dataset from the UCSC database. In addition, we also downloaded the copy number variation dataset at the level-4 gene level for all TCGA samples by GISTIC27 software from the GDC. We


integrated the copy number data and gene expression data of the samples and analyzed the CNV mutation differences between pan-cancers by using R software. IMMUNE-RELATED CHARACTERISTICS OF


SMAD3 We conducted an extensive analysis of immune cell infiltration by leveraging the resources available in the TIMER 2.0 database. Leveraging a range of algorithms, including TIMER,


XCELL, EPIC, CIBERSORT, TIDE, QUANTISEQ, and others, we meticulously examined the associations between SMAD3 expression and diverse immune cell subsets. These subsets encompassed CD8 + T


cells, CD4 + T cells, B cells, macrophages, and T cell regulators (Tregs) within distinct TCGA cancer types. We extracted the expression data of SMAD3 in each sample from the TCGA TARGET


GTEx database, and calculated stromal, immune, and estimate scores according to gene expression by using the R software package ESTIMATE28. In addition, expression data of SMAD3 gene and 150


marker genes of five types of immune pathways (chemokine41, receptor18, MHC21, immunoinhibitor24 and immunostimulator45) were extracted from the database, Correlative studies on SMAD3


expression with immunoregulatory genes were also discussed by Spearman analysis. In our study, we harnessed Pearson correlation analysis to assess the associations between SMAD3 gene


expression and key parameters, including immune checkpoints (ICP), tumor mutation burden (TMB), microsatellite instability (MSI), and tumor stemness. Additionally, we conducted a comparative


analysis of SMAD3 expression levels in different tumor cell lines before and after immune checkpoint blockade (ICB) treatment, as well as between responders and non-responders. These


analyses were carried out using data from the TIDE database.Furthermore, we examined the correlation between SMAD3 expression and overall survival (OS) within the human immunotherapy cohort


and evaluated response outcomes utilizing the TISMO database29. SINGLE-CELL FUNCTIONAL ANALYSIS OF SMAD3 TISCH, an extensive and meticulously curated database, amalgamates the single-cell


transcriptome profiles of approximately 2 million cells sourced from high-caliber tumor datasets spanning 27 distinct cancer types30. We acquired the single-cell expression data of SMAD3


encompassing various pan-cancer datasets from the database. Our analysis delved into the single-cell composition of SMAD3 expression, and we meticulously examined this within the context of


three distinct datasets. CancerSEA is a database that studies tumor cell function at the single-cell level31. In our research, we mapped the correlations between SMAD3 expression and diverse


tumor functions by leveraging single-cell sequencing data. Furthermore, we provided a demonstration of the spatial transcriptomic characteristics specific to Breast Cancer (BRCA) and


Prostate Adenocarcinoma (PRAD) based on data sourced from the SpatialDB database30. CO-EXPRESSED GENES AND ENRICHMENT ANALYSIS OF SMAD3 Our research methodology involved several key steps.


Initially, we procured a set of 50 Smad3-interacting genes through the STRING website. Subsequently, to acquire a practical list of Smad3-binding proteins, we utilized the “similar gene


detection” feature of GEPIA2. This process relied on the TCGA tumor and normal tissue dataset, from which we extracted the first 100 SMAD3-related genes. Concurrently, we conducted a Pearson


correlation analysis to further refine the selection of genes that exhibited a significant correlation with SMAD3. Moving forward, we conducted comprehensive Gene Ontology (GO) and Kyoto


Encyclopedia of Genes and Genomes (KEGG) enrichment analyses32,33,34. Additionally, transcription factor enrichment among associated genes was explored using the Metascape database. SMAD3


promoter sequence was obtained from NCBI, and candidate transcription factor protein sequences were acquired from UniProt. Interactions with SMAD3 promoters were predicted using AlphaFold


3.0. Transcription factor binding sites were predicted using the JASPAR website (https://jaspar.elixir.no/).focusing on the top ten genes associated with SMAD3 across pan-cancer datasets35,


p-value < 0.05 was considered to be statistically significant. GSEA ENRICHMENT ANALYSIS AND DRUG SENSITIVITY PREDICTION OF SMAD3 Within the PAAD and LIHC groups, we harnessed Gene Set


Enrichment Analysis (GSEA) to probe the biological signaling pathways. In this analysis, we focused on the top 5 terms derived from both the KEGG and HALLMARK databases. Notably, we


considered KEGG pathways with notable enrichment results, as indicated by the enrichment score (ES) and gene ratio. Specifically, gene sets with |NES| ≥ 1 and NOM _p_ ≤ 0.05 were deemed to


be significantly enriched36. In our quest to explore the nexus between drug sensitivity and the expression of SMAD3, we engaged in a rigorous drug sensitivity analysis employing the GSCALite


platform. For this endeavor, Spearman correlation analysis served as our indispensable tool. Within our analytical framework, a positive correlation unveiled that neoplastic cell


characterized by heightened SMAD3 expression displayed an elevated proclivity towards drug resistance, whereas a negative correlation signified that those same cells manifested an increased


receptivity to pharmacological agents. CELL CULTURE AND TRANSFECTION The human Liver Hepatocellular Carcinoma (LIHC) cell lines JHH-5, HepG2, SK-HEP-1, Hep3B, Hu-7, LI-7, and LM3 were


procured from the Shanghai Institute of Cell Biology, China. The immortalized liver cell line HL-7702 was obtained from Shanghai Fuxiang Biotechnology Co., Ltd. These cell lines were


cultivated in DMEM (Gibco) supplemented with 10% FBS (Hyclone) and exposed to 5% carbon dioxide environment at 37 °C. We acquired SMAD3 small interfering RNA (siRNA) from GenePharma in


Shanghai, China. In LM3 cells, the multiplicity of infection (MOI) was set at 10. To enhance transfection efficiency, Polybrene was employed, and positive cells were subsequently selected


using puromycin screening. WESTERN BLOT ANALYSIS The Ethics Committee of the Second Affiliated Hospital of Nanchang University granted authorization for the utilization of human tissue in


this research. To extract cellular and human tissue lysates, a radioimmunoprecipitation assay buffer (Solarbio, China), fortified with a cocktail of protease inhibitors, was employed.


Subsequently, a 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis technique was implemented to facilitate the separation of the resulting pyrolysis products. These products were


subsequently transferred onto polyvinylidene fluoride (PVDF) membranes, followed by incubation with primary antibodies. Notably, the primary antibodies employed included SMAD3 (1:1000,


66516-1-Ig, Proteintech, China) and GAPDH (1:2000, 60004-1-Ig, Proteintech, China). In the subsequent steps, PVDF membranes were subjected to further incubation with relevant secondary


antibodies. Ultimately, the membranes were exposed to an enhanced chemiluminescence (ECL) substrate (Proteintech, China), and the discernible protein bands were documented via the SH-Cute


523 imaging system for analysis. WOUND HEALING AND TRANSWELL EXPERIMENTS Wound Healing Assay: Hu7, LM3, and Hep3B cells, at a cell density of 2.5 × 105 per well, were seeded into 6-well


plates. Subsequently, transfection of siSMAD3#1 and siSMAD3#2 was conducted in these plates. After a 24-hour incubation period, a 200 mL pipetting head was employed to create cell scratches


within the plates. The cells were then provided with serum-free medium. Subsequent images were captured at intervals of 0 h, 12 h, and 24 h using an inverted microscope (IX81, Olympus


Company, Japan). Transwell Assay: Cells were suspended and enumerated using serum-free medium. These cells were then introduced into the upper chamber at a density of 5 × 104, while the


lower chamber received 500 mL of full medium. Following a 24-hour incubation period, the cells were fixed with 4% paraformaldehyde for 10 min. Subsequently, 1% crystal violet was utilized to


stain the cells for a period of 10 s. Any remaining cells in the upper chamber were gently removed, and images were captured using a brightfield microscope (BX53, Olympus Company, Japan).


Furthermore, the number of cells that had traversed the chamber was counted across four randomly selected fields under the microscope. EDU ASSAY The transfected Hu-7, LM3, and Hep3B cells,


with a seeding density of 2 × 104, were plated in 24-well plates and subsequently incubated for a duration of 72 h. Following this incubation period, the cells were subjected to culture with


EdU reagent for a duration of 2 h. Following these steps, cellular specimens were meticulously subjected to fixation employing a solution of 4% paraformaldehyde and 0.5% Triton X-100.


Subsequent to the fixation process, the specimens underwent staining procedures utilizing Hoechst stain. Quantitative assessment of the EdU inclusion rate was accomplished through the


utilization of ImageJ software. STATISTICAL ANALYSIS Through the online databases mentioned above, the statistical analysis was automatically computed in this study. These results were


considered as statistically significant at *_P_ < 0.0001. RESULTS ANALYSIS OF GENE EXPRESSION Figure 1 presents a flowchart illustrating the complete research process. In the HPA


database, we conducted an examination of SMAD3 expression in various normal tissues. The results of our analysis revealed heightened levels of SMAD3 mRNA expression in several tissues,


including the esophagus, skeletal muscle, bladder, tongue, choroid plexus, ovary, and skin (Fig. 2A). Furthermore, we performed an analysis of SMAD3’s differential expression in both tumor


and normal tissues by leveraging the TIMER 2.0 database. Our investigation revealed a substantial upregulation of SMAD3 expression in six cancer types, including ESCA, STAD, LUSC, LIHC,


CHOL, and HNSC. Conversely, we observed significant downregulation in seven other cancers, specifically COAD, BRCA, PRAD, UCEC, KIRC, THCA, and SKCM (Fig. 2B). Simultaneously, we extracted


SMAD3 expression data from normal tissues in the GTEx database and integrated it with the data from TCGA. Our analysis unveiled a remarkable up-regulation of SMAD3 in 15 cancer types, which


include ACC, CHOL, COAD, ESCA, GBM, HNSC, KICH, LAML, LGG, LIHC, LUAD, LUSC, PAAD, STAD, and TGCT. In contrast, we observed significant down-regulation in 7 other cancer types, specifically


BRCA, KIRC, OV, PRAD, SKCM, UCEC, and UCS (Fig. 2C). Additionally, we conducted a paired difference analysis for SMAD3 expression across pan-cancer datasets. Our analysis revealed a


significant increase in SMAD3 expression in LIHC, LUSC, OSCC, CHOL, STAD, and HNSC. Conversely, a decrease in SMAD3 expression was observed in BRCA, UCEC, PRAD, and COAD (Fig. 2D). We


employed R software to compute the variations in gene expression across tumor samples at different clinical stages. Utilizing an unpaired Student’s t-Test, we performed a significance


analysis of the differences between pairs of samples. In addition, we conducted variance analysis to test the differences among multiple groups of samples. Our observations indicated


significant differences in eight types of tumors, including LUAD, BRCA, KIPAN, KIRC-PAAD, OV, TGCT, and BLCA (Fig. 2E-F). Furthermore, we acquired the IHC results from the HPA database to


corroborate the protein expression of SMAD3. As depicted in Fig. 2G-H, tumor tissues displayed substantial SMAD3 IHC staining, whereas normal tissues from the liver, lung, pancreas, and


brain exhibited low to moderate staining. Notably, these findings align with the respective levels of gene expression. To gain insight into SMAD3’s subcellular localization, we conducted


immunofluorescence localization in A-549 and U251-MG cells. Our observations indicated that SMAD3 predominantly localized within the nucleus and cytosols. PROGNOSTIC VALUE OF SMAD3 We


employed Overall Survival (OS), Disease-Specific Survival (DSS), and Progression-Free Interval (PFI) to investigate the prognostic implications of SMAD3 across various cancer types. Notably,


high expression of SMAD3 was associated with unfavorable outcomes in LAML, PAAD, ACC, UVM, and LUAD (Fig. 3A-B). Conversely, SMAD3 exhibited high expression in GBMLGG, KIRC, and KIPAN,


which correlated with favorable OS. In terms of DSS, SMAD3 was highly expressed in ACC, PCPG, LUAD, and PAAD, and this high expression was associated with shorter DSS (Supplementary Fig. 1S


A-B). Additionally, our results revealed that SMAD3 was overexpressed in ACC, PAAD, STAD, UVM, LIHC, and STES, and this overexpression was linked to a shorter PFI (Supplementary Fig. 2S


A-B). Consequently, SMAD3 overexpression was particularly prominent in PAAD and ACC, where OS, DSS, and PFI were notably abbreviated. CORRELATION OF SMAD3 EXPRESSION WITH DNA METHYLATION AND


RNA MODIFICATION The UALCAN online tool and GSCA database furnished a valuable platform for our exploration of SMAD3 promoter methylation levels across different patient groups and normal


populations in various cancer types. Notably, our findings indicated a considerable increase in the promoter methylation levels of SMAD3 in eight tumor groups (BRCA, READ, HNSC, KIRP, UCEC,


PRAD, COAD, KIRC) compared to their respective normal counterparts (Fig. 4B). Conversely, in seven tumor groups (LUAD, LUSC, TGCT, CHOL, THCA, BLCA, LIHC), the SMAD3 methylation levels were


significantly lower than those observed in normal groups. Moreover, we evaluated the impact of SMAD3 methylation expression on the survival prognosis using the GSCA database. The results


were rather promising, demonstrating that high SMAD3 methylation expression tended to predict a more favorable prognosis in nearly all statistically significant prognostic analyses (Fig. 4C,


Supplementary Fig. 3SD-G). This suggests that SMAD3 may influence the prognosis of cancer patients through its methylation status. Furthermore, we observed a positive correlation between


SMAD3 expression and m6A, m5C, and m1A regulatory genes in most cancers, with the exception of NB and CHOL (Fig. 4A). This intriguing finding implies that SMAD3 may play a role in


carcinogenesis by affecting the expression of RNA methylation regulatory genes. SMAD3 MUTATION IN PAN-CANCER Our analysis of genetic alterations across various cancers, conducted through the


cBioPortal platform, allowed us to delve into the landscape of SMAD3 gene variations in pan-cancer. The results unveiled distinct patterns in different malignancies, with COAD exhibiting


the highest frequency of SMAD3 variations at 6.40%, primarily in the form of mutations. Following COAD, the top five cancers with the highest variation frequencies were UCEC (5.48%), SKCM


(3.83%), MESO (3.45%), and KICH (3.08%). Notably, MESO was unique in its pattern of variation, characterized by amplifications (Fig. 5A). In further exploration of these variations, we


identified missense and truncating mutations as the primary mutation types affecting SMAD3, as depicted in Fig. 5B. These insights into the genetic alterations in SMAD3 shed light on its


role in the context of different cancers. Five independent studies conducted on endometrial, lung, gastric, and colorectal adenocarcinomas revealed that R268C/H missense mutations underwent


changes within the MH2 domain. Additionally, SMAD3 mutations were correlated with various post-translational modifications, including phosphorylation, acetylation, ubiquitination, and


malonylation. The alterations in the conformation of the SMAD3 protein resulting from the R268C/H mutations are visually depicted in Fig. 5C. Changes in copy number in parts of the genome


are known to be a feature of many cancers37. We utilized the R software to compute disparities in tumor gene expression across clinical stage-specific samples. Significance analysis for


pairwise disparities was conducted using unpaired Wilcoxon rank sum and signed rank tests, while the Kruskal test was employed to assess differences among multiple sample groups. Figure 5D


illustrates notable disparities in copy number variations (CNVs) across 14 distinct cancer types (GBMLGG, LGG, CESC, COAD, COADREAD, BRCA, STES, SARC, STAD, HNSC, LIHC, READ, SKCM, UCS).


ANALYSIS OF IMMUNE CELL INFILTRATION AND IMMUNOINFILTRATION OF SMAD3 The UCSC database was used to download a unified and standardized pan-cancer dataset, and the R software package ESTIMATE


was used to calculate stromal, immune, and ESTIMATE scores for each patient in each tumor based on gene expression. To ascertain the statistically significant associations pertaining to


immune infiltration scores, we computed the Spearman’s correlation coefficient between gene expression and immunoinfiltration scores for each tumor sample. This analysis was conducted by


means of the corr.test function within the R package. The results showed that LGG, COAD, COADREAD, PRAD, NB, READ, and UVM showed a significant positive correlation among the three immune


infiltration scores. At the same time, STES, KIRP, KIPAN, HNSC, KIRC, LUSC, LIHC, WT, BLCA, THCA, and ACC were all negatively correlated (Supplementary Fig. 4S、Figure 5S、Figure 6S). In


recent investigations, a compelling body of evidence has emerged, affirming the intricate relationship between immune cell infiltration and the genesis, progression, and metastatic cascade


of an array of malignancies38,39. In the quest to discern the interplay of SMAD3 expression with the presence of tumor-infiltrating immune cell populations, we conducted a comprehensive


analysis utilizing the TIMER 2.0 database. Employing a repertoire of algorithms, such as TIMER, EPIC, QUANTISEQ, XCELL, CIBERSORT, TIDE, and others, we unveiled noteworthy findings. Our


analysis revealed a significant positive correlation between SMAD3 expression and the infiltration levels of various immune cell types, including B cells, cancer-associated fibroblasts


(CAFs), NK cells, neutrophils, monocytes, and endothelial cells (Endo). Interestingly, there was no significant immune-related association between SMAD3 expression and macrophages,


progenitor cells, eosinophils, hematopoietic stem cells (HSCs), myeloid-derived suppressor cells (MDSCs), follicular helper T cells (FhTs), dermal epithelial cells, and mast cells. Moreover,


our investigation identified a strong positive correlation between SMAD3 expression and CD4 + T cell infiltration in prostate adenocarcinoma (PRAD), liver hepatocellular carcinoma (LIHC),


cervical squamous cell carcinoma (CESC), and diffuse large B-cell lymphoma (DLBC). Conversely, we observed a significant negative correlation between SMAD3 expression and CD8 + T cell


infiltration in glioblastoma (GBM), head and neck squamous cell carcinoma (HNSC), kidney renal papillary cell carcinoma (KIRP), low-grade glioma (LGG), and lung squamous cell carcinoma


(LUSC). as illustrated in Fig. 6. RELATIONSHIP BETWEEN SMAD3 AND IMMUNE CHECKPOINTS, IMMUNE MODULATORS In recent years, immune checkpoint inhibitors (ICIs) have achieved significant


milestones in the realm of cancer immunotherapy. However, a substantial number of patients continue to exhibit either a lack of response or develop resistance to immune checkpoint


inhibitors40. We observed that SMAD3 displayed associations with multiple immune checkpoint inhibitors and immune checkpoint stimulators. Subsequently, we conducted an investigation into the


relationships between SMAD3 and immune checkpoints as well as immune modulators. Across the majority of cancer types, SMAD3 expression displayed a substantial association with the


expression profiles of established immune checkpoint molecules, a trend that was consistently observed. Notable exceptions to this pattern included kidney chromophobe (KICH), esophageal


carcinoma (ESCA), acute lymphoblastic leukemia (ALL), cholangiocarcinoma (CHOL), and mesothelioma (MESO), where the correlation between SMAD3 expression and immune checkpoint markers was not


statistically significant. The encompassed immune checkpoints encompassed lymphocyte-activation gene 3 (LAG3), CD40, cytotoxic T-lymphocyte-associated protein 4 (CTLA4), CD80, programmed


cell death 1 (PDCD1), T cell immunoreceptor with Ig and ITIM domains (TIGIT), CD86, and tumor necrosis factor receptor superfamily, member 9 (TNFRSF9) (Fig. 7A). These findings portend a


prospect of synergy between SMAD3 and established immune checkpoint molecules. To appraise the influence of SMAD3 expression on immunotherapeutic outcomes, we conducted a comparative


analysis of SMAD3 expression levels in distinct tumor cell lines, both antecedent to and post-administration of immune checkpoint blockade (ICB) interventions, and across respondent and


non-respondent subpopulations. Eminent among our observations is the salient decrease in SMAD3 expression within CT26 and T11 cells subsequent to anti-CTLA4 and anti-PD-1 treatments.


Furthermore, a conspicuous reduction in SMAD3 expression was discerned in CT26 cells displaying favorable responses to anti-PDL-1 treatment, alongside the corresponding downregulation


observed in YTN16 cells that manifested a positive response to anti-CTLA4 intervention (Fig. 7C). Subsequently, we proceeded to assess the relationship between SMAD3 expression and the


resultant response patterns, as well as overall survival (OS), within the human immunotherapy cohort (Fig. 7B). The findings unveiled that, within the ambit of 24 immunotherapy cohorts,


SMAD3 emerged with an Area Under the Curve (AUC) value exceeding 0.5 in nine instances, denoting its predictive utility. Remarkably, SMAD3 exhibited superior predictive efficacy when


contrasted with Tumor Mutation Burden (TMB), T cell clonality (T clonality), and B cell clonality (B clonality). Nevertheless, SMAD3’s predictive performance, while notable, was surpassed by


the predictive value of the CD274 score (AUC > 0.5 in 21 immunotherapy cohorts), the CD8 score (AUC > 0.5 in 18 immunotherapy cohorts), the Interferon Gamma (IFNG) score (AUC > 0.5


in 17 immunotherapy cohorts), and the Merck 18 score (AUC > 0.5 in 16 immunotherapy cohorts). In the aggregate, SMAD3 expression emerges as a potent biomarker, signifying its efficacy in


the anticipation of immunotherapeutic responses. Finally, an examination of SMAD3 expression levels across various datasets revealed its pronounced upregulation in METABRIC,


ICB-Lauss2017-ACT, Patel 2017, and MDSC, thereby emphasizing its salient role in these contexts (Fig. 7D). We additionally conducted an analysis of the correlation between SMAD3 and immune


regulatory genes across 40 tumor types. This analysis encompassed Major Histocompatibility Complex (MHC), immunostimulator genes, immunoinhibitor genes, chemokines, and chemokine receptors,


aimed at gaining deeper insights into the interplay between SMAD3 and immune regulatory genes within the context of cancer. The results revealed a robust association between SMAD3 expression


and immune regulatory genes in malignant tumors (Supplementary Fig. 7S). RELATIONSHIP BETWEEN SMAD3 EXPRESSION AND TUMOR STEMNESS, AND GENOMIC HETEROGENEITY In addition to metastasis and


heterogeneity, it is well-established that immune checkpoint gene expression and immune system cell infiltration are intricately linked to tumor stemness38. Our analysis of SMAD3 expression


alongside tumor stemness markers, DNAss and RNAss, revealed noteworthy findings. We observed significant negative correlations between SMAD3 expression and DNAss in four tumor types,


specifically COAD, COADREAD, BRCA, and UCEC. Conversely, significant positive correlations between SMAD3 expression and DNAss were identified in several tumor types, including CESC, STES,


THYM, THCA, UVM, and HNSC, as depicted in Fig. 7G. Furthermore, our analysis unveiled significant negative correlations between SMAD3 expression and RNAss in 18 tumor types, encompassing


LGG, COADREAD, LAML, BRCA, KIRP, STAD, PRAD, UCEC, THYM, LIHC, THCA, READ, OV, TGCT, UCS, BLCA, and CHOL, as illustrated in Fig. 7H. Cancer stem cells (CSCs) are a population of


self-renewing cells that play a pivotal role in tumor development, facilitate metastasis, and contribute to increased resistance to cancer treatments41. It is important to note that the


impact of immunotherapy can be influenced by tumor heterogeneity. Therefore, we conducted an examination of the Pearson’s correlation coefficients between SMAD3 and tumor heterogeneity


indicators, including Tumor Mutational Burden (TMB) and Microsatellite Instability (MSI). The analysis revealed that SMAD3 expression was significantly negatively correlated with TMB in


tumor types such as GBMLGG, BRCA, LUAD, KIPAN, PRAD, KIRC, THCA, SKCM, BLCA, and DLBC. Conversely, SMAD3 expression exhibited a significant positive association with TMB in SARC, HNSC, ACC,


THYM, and PAAD (Fig. 7E). Furthermore, the Microsatellite Instability (MSI) displayed a positive correlation with SMAD3 expression in tumor types including GBMLGG, COAD, SARC, KIPAN, LUSC,


and UVM. Conversely, a negative correlation was observed between MSI and SMAD3 expression in BRCA, PRAD, HNSC, THCA, and DLBC (Fig. 7F). SINGLE-CELL ANALYSIS OF SMAD3 Utilizing the TISCH


database, we conducted an in-depth exploration of SMAD3’s single-cell expression patterns across diverse cancer types. Our analysis unveiled that SMAD3 exhibited notably high expression


levels in various cell types, including CD4Tconv, Treg, Tprolif, CD8T + cells, CD8Tex, B cells, dendritic cells (DC), endothelial cells, and fibroblasts. Particularly striking was the high


expression of SMAD3 in monocytes (Mono) and macrophages (Macaro) (Fig. 8A). To delve deeper, we acquired precise SMAD3 expression data within specific cell groups from liver hepatocellular


carcinoma (LIHC), prostate adenocarcinoma (PRAD), and pancreatic adenocarcinoma (PAAD). This information was sourced from datasets GSE125449, GSE143791, and GSE111672. Our analysis revealed


the expression of SMAD3 within key cell groups, including CD8 + T cells, monocytes/macrophages, and endothelial cells (Fig. 8B). Our exploration extended to the investigation of SMAD3’s


functions at the single-cell level using the CancerSEA database. The heatmap results unveiled significant insights into the correlations between SMAD3 and various biological processes. SMAD3


exhibited positive correlations with critical processes such as angiogenesis, hypoxia, inflammation, metastasis, and stemness across diverse cancer types. In contrast, SMAD3 displayed


negative correlations with processes related to DNA damage, DNA repair, invasion, and quiescence (Fig. 8C). In addition, our investigation extended to the SpatialDB database, revealing that


SMAD3 is consistently expressed in a majority of monocyte- and macrophage-rich tumors. Our analysis determined that SMAD3, in conjunction with monocyte/macrophage markers such as CD68 and


CD14, exhibited varying levels of spatial transcriptomic expression patterns in breast cancer (BRCA) and prostate adenocarcinoma (PRAD) (Fig. 8D). ENRICHMENT OF SMAD3-RELATED PARTNERS A


total of 50 SMAD3-binding proteins were identified, each supported by experimental data acquired through the STRING tool, which illustrates the interaction network of these proteins (Fig. 


9A). Utilizing the GEPIA2 tool to integrate TCGA pan-cancer expression data, we identified the top 100 genes that exhibit a correlation with SMAD3 expression. Among these correlated genes,


SMAD3 expression displayed positive associations with the top 10 genes, namely, BICD2 (correlation coefficient, _R_ = 0.5), FAM160A1 (_R_ = 0.52), KLF3 (_R_ = 0.49), MAPK6 (_R_ = 0.48),


SEMA4B (_R_ = 0.52), SLK (_R_ = 0.48), TMOD3 (_R_ = 0.52), TP63 (_R_ = 0.53), ZDHHC5 (_R_ = 0.49), and PCYT1A (_R_ = 0.48), with all correlations being statistically significant (_P_ < 


0.001) (Fig. 9J). In an intersection analysis, two of the aforementioned genes, SMURF1 and SP1, were identified as belonging to both the SMAD3-binding proteins and the top correlated genes


with SMAD3 expression (Fig. 9B). To perform KEGG and GO enrichment analyses, the two datasets were combined. KEGG data suggest that the effect of SMAD3 on tumor pathogenesis, which was


depicted in Fig. 7C, may involve the “TGF-beta signaling pathway,” “Transcriptional misregulation in cancer,” and “Human T-cell leukemia virus 1 infection.” (Fig. 9C). Additionally, we


performed GO enrichment analysis and discovered that SMAD3-interacting genes are related to the biological processes “reaction to endogenous stimuli,” “positive regulation of nucleic


acid-templated transcription,” and “positive regulation “of biological process (BP) (Fig. 9D). Second, we found that Smad3-related genes were primarily enriched in “nuclear part,” “nuclear


lumen,” and “nucleoplasm” in the cellular component (CC) (Fig. 9E), and that “transcription regulatory region DNA binding”, “regulatory region nucleic acid binding” and “double-stranded DNA


binding” were enriched in cellular function (MF) (Fig. 9F). Additionally, we assessed Smad3-related genes using the Metascape database and conducted transcription factor enrichment analysis.


The top three enriched transcription factors identified were “GGGYGTGNY UNKNOWN,” “PAX Q6,” and “CEBPB 01” (Fig. 9G). Using AlphaFold 3.0, we predicted the molecular docking of CEBPB with


the SMAD3 promoter, and using PyMOL software to visualize protein-DNA interactions, the docking result is: Docking Score: -194.16, Confidence Score: 0.7612 we identified specific binding


sites of CEBPB on the SMAD3 promoter using the JASPAR website. The highest sequence scores were observed at positions − 1794 to -1785 and − 1523 to -1514 (Fig. 9H-I). GSEA ENRICHMENT OF


SMAD3 AND RELATIONSHIP BETWEEN SMAD3 EXPRESSION AND DRUG SENSITIVITY We performed functional enrichment analysis of SMAD3 expression in pancreatic adenocarcinoma (PAAD) and liver


hepatocellular carcinoma (LIHC) using Gene Set Enrichment Analysis (GSEA). In PAAD, SMAD3 expression was predominantly associated with KEGG enrichment terms related to Glycine, Serine, and


Threonine Metabolism, the Renin-Angiotensin System, Apoptosis, and Neuroactive Ligand Receptor Interaction (Fig. 10A). Additionally, Apoptosis, Notch Signaling Pathway, TNFA Signaling VIA


NFKB, P53 Pathway, and Interferon Gamma Response in PAAD were all linked to SMAD3 in HALLMARK terms (Fig. 10C). According to KEGG enrichment terms, Complement and Coagulation, Prostate


Cancer, Ubiquitin Mediated Proteolysis, ERBB Signaling Pathway and Oocyte Meiosis are primarily associated with SMAD3 expression in LIHC (Fig. 10B). In LIHC, the analysis of SMAD3 expression


revealed significant associations with HALLMARK enrichment terms. Specifically, SMAD3 was linked to processes such as PI3k AKT MTOR Signaling, Wnt-β Catenin Signaling, Mitotic Spindle, Heme


Metabolism, and Unfolded protein response (Fig. 10D). We assessed the drug sensitivity associated with SMAD3 expression in various cancers using GSCALite. The analysis revealed that the 50%


inhibitory concentration (IC50) values of nine drugs, namely, 17-AAG, Dasatinib, Docetaxel, Lapatinib, PD-0325901, RDEA119, Trametinib, Selumetinib, and Bleomycin, exhibited an inverse


correlation with KIF18A expression. On the other hand, the IC50 values of 15 different medications, including AR-42, BX-912, CAY10603, FX866, GSK1070916, I-BET-762, KIN001-102,


NPK76-II-72-1, Navitoclax, QL-XL138, THZ-2-102-1, UNC0638, Vorinostat, WZ3105, and YM201636, were positively correlated with SMAD3 expression (Fig. 10E). SMAD3 PROMOTES THE PROLIFERATION AND


MIGRATION OF LIVER CANCER CELLS The protein expression levels of SMAD3 in Liver Hepatocellular Carcinoma (LIHC) tissues exhibited a notable elevation, with the outcomes meticulously


analyzed utilizing ImageJ software (Fig. 11A). We extended our scrutiny to assess the protein expressions of SMAD3 in a spectrum of LIHC cell lines, encompassing JHH-5, HepG2, SK-HEP-1,


Hep3B, Hu7, Li-7, and LM3. Strikingly, we observed that the expression of SMAD3 in the Hep3B, Hu7, and LM3 cell lines surpassed that in other LIHC cell lines (Fig. 11B). Subsequently, we


employed specific siRNAs targeting SMAD3 to achieve knockdown of SMAD3 expression in the Hep3B, Hu7, and LM3 cells (Fig. 11C, D, E). Following this, In our pursuit of unraveling the


intricate interplay between LIHC cells and SMAD3 expression across discrete cohorts, we embarked on a series of functional experiments. Notably, the outcomes derived from EDU assays, wound


healing assays, and Transwell assays synergistically unveiled a compelling narrative. The act of SMAD3 knock-down ushered forth a marked diminution in the proliferation and migratory


potential of Hep3B, Hu7, and LM3 cells (Fig. 11F, G, H). These findings compellingly underscore the promoting role of SMAD3 in the proliferation and migration of LIHC cells. DISCUSSION The


similarities and differences between tumors can be shown by pan-cancer analysis, which can support to the theoretical framework guiding efforts in cancer prevention, the design of


therapeutic targets, and the prospective screening of therapeutic drugs42. SMAD3 is central to the TGF-β pathway and plays an important role in immune-inflammatory responses. Recent research


suggests that SMAD3 is connected to the survival and prognosis of malignant tumors and plays a role in the etiology and advancement of colorectal cancer, lung cancer, breast cancer, bladder


cancer, prostate cancer, cervical cancer, gastric cancer, and osteosarcoma12,14,15,43,44,45,46. We embarked on an exhaustive and all-encompassing analysis of 33 different global cancer


types to discern the molecular characteristics of SMAD3. This analysis involved the utilization of diverse databases, including TCGA, GTEx, UALCAN, TIMER2.0, GSCA, and cBioportal. Our


investigation aimed to elucidate SMAD3’s role in tumor development and potential regulatory mechanisms, encompassing gene expression, prognosis, gene mutations, immunological infiltration,


single-cell analysis, immune checkpoints, DNA methylation, RNA methylation, tumor mutation burden (TMB), microsatellite instability (MSI), tumor characteristics, and drug sensitivity.


Previous research has demonstrated that SMAD3 has a role in the progression and development of tumors and is expressed differently in cancer47. we utilized data from the GTEx and HPA


databases to validate the tissue-specific expression of SMAD3. Our results indicate a significant increase in SMAD3 mRNA expression across most cancer types (including ACC, CHOL, COAD, ESCA,


GBM, HNSC, KICH, LAML, LGG, LIHC, LUAD, LUSC, PAAD, STAD, and TGCT) compared to normal samples. Combined pairwise difference analysis results, SMAD3 showed significantly increased


expression in LIHC, STAD, LUSC, CHOL, and HNSC, whereas it was decreased in BRCA, PRAD, and UCEC. These findings suggest that SMAD3 expression changes in different cancer types may be


influenced by various tumor biological characteristics, gene regulatory mechanisms and the tumor microenvironment. DNA methylation in tumors is widely acknowledged as a crucial epigenetic


regulatory mechanism in cancer development24. Specifically, hypermethylation of certain genes often leads to their downregulation or silencing. In this study, we also analyzed the DNA


methylation status of SMAD3 across multiple cancer types. We found that lower levels of DNA methylation in LIHC and CHOL were associated with increased SMAD3 expression, whereas higher


methylation levels in BRCA, PRAD, and UCEC correlated with decreased SMAD3 expression. This suggests that DNA methylation modifications of SMAD3 may play a significant role in these cancer


types. Further researches are needed to elucidate the potential function of SMAD3 methylation in different cancers. Furthermore, we explored the potential of SMAD3 as a biomarker for cancer


diagnosis and prognosis. By analyzing SMAD3 expression levels in cancer patients, we observed that high SMAD3 expression in tumor tissues correlated with poor prognosis. This association was


particularly pronounced in PAAD and ACC, where high SMAD3 expression was significantly linked to shortened overall survival (OS), disease-specific survival (DSS), and progression-free


interval (PFI). Therefore, SMAD3 may serve as a critical diagnostic and prognostic biomarker in personalized and precision cancer therapies, especially notable in PAAD and ACC. An integral


component of the tumor microenvironment (TME), tumor immunity is closely related to tumor immunity and drives tumor growth48,49,50. Data from the TIMER2.0 database showed a significant link


between SMAD3 expression and various immune cell infiltrations in different cancers. Our study also evaluated immune and stromal elements in the tumor microenvironment using immunological


and stromal scores, and indirectly assessed tumor purity through the ESTIMATE score51. The results showed a positive correlation with three immune infiltration scores in seven cancer types.


In contrast, negative associations were shown in 11 other cancer types. These findings suggest SMAD3 might play important roles in the importance of tumor immune microenvironment. Tumor


mutation burden (TMB), microsatellite instability (MSI), and programmed death-ligand 1 (PD-L1) are frequently employed as markers to anticipate the efficacy of immune checkpoint inhibitors


(ICIs). TMB and MSI strongly correlated with PD-1/PD-L1 inhibitor efficacy, and most clinical studies using TMB and MSI as predictive markers reached endpoints with significant success


rates52. Notably, high SMAD3 expression was positively correlated with increased TMB in GBMLGG, BRCA, LUAD, KIPAN, PRAD, KIRC, THCA, SKCM, BLCA, and DLBC, and decreased TMB in SARC, HNSC,


ACC, THYM, and PAAD. In addition, SMAD3 was positively correlated with microsatellite instability (MSI) expression in GBMLGG, COAD, SARC, SARC KIPAN, LUSC and UVM. Additionally, we


investigated the relationship between SMAD3 and tumor stemness. Cancer stem cells possess the unique ability to regenerate and generate a variety of tumor cells, making them indispensable in


the contexts of tumor survival, growth, metastasis, and recurrence53,54. Our analysis of SMAD3 expression and tumor stem cell markers DNAss and RNAss showed that SMAD3 was negatively


correlated with COADREAD, BRCA and UCEC. In terms of immune checkpoints, we found more than 30 immune checkpoints that were positively associated with most cancers, except KICH, ESCA, ALL,


CHOL, and MESO. Furthermore, our analysis shows that low expression of SMAD3 generally predicts a favorable response to immunotherapy. Taken together, these results demonstrate a close


relationship between SMAD3, TMB, MSI, MMR, DNAss, RNAss, and immune checkpoints, highlighting the potential of SMAD3 as a novel biomarker for predicting immune checkpoint inhibitor (ICI)


efficacy. However, the predictive abilities of TMB and MSI vary significantly across different types of cancer, warranting further research to validate these relationships. Using STRING and


GEPIA2 databases, we integrated and analyzed information on SMAD3-related genes and proteins.Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis can


analyze pivotal genes55. Our findings indicate a significant association of SMAD3 with key biological pathways including the “TGF-beta signaling pathway,” “Transcriptional misregulation in


cancer,” and “Human T-cell leukemia virus 1 infection.” Particularly noteworthy is SMAD3’s pivotal role within the TGF-beta signaling pathway, which is intricately linked to various aspects


of tumorigenesis56. This pathway notably influences tumor progression by modulating epithelial-mesenchymal transition, thereby impacting the migration and invasion capabilities of tumor


cells57.Furthermore, Bioinformatics analysis showed that SMAD3 has important diagnostic and prognostic significance in LIHC, and its expression may be related to the effectiveness of


immunotherapy for LIHC. Here, we use molecular biology techniques to verify the role of SMAD3 in LIHC. We demonstrate that SMAD3 expression is elevated in LIHC tissues and cell lines. SMAD3


knockdown inhibits proliferation and migration of hepatoma cells. These findings suggest that SMAD3 is involved in carcinogenesis of liver cancer and has therapeutic promise. Further


molecular studies are needed to elucidate the specific mechanism of SMAD3 in LIHC. In summary, SMAD3, as a signaling molecule, exhibits significant expression variations across different


types of tumors, profoundly influencing tumor development and treatment responses. According to data from the TIMER2.0 database, SMAD3 expression correlates significantly with immune cell


infiltration in various cancers, suggesting its potential role in the tumor immune microenvironment (TME). Additionally, SMAD3 expression correlates closely with immune checkpoint inhibitor


(ICI) treatment response predictors such as tumor mutational burden (TMB) and microsatellite instability (MSI). High SMAD3 expression positively correlates with increased TMB and MSI in


several cancers, while it correlates negatively in others, indicating complex relationships that warrant further investigation between SMAD3 expression patterns and TMB/MSI. Furthermore,


SMAD3’s DNA methylation status correlates with its expression changes, reflecting its involvement in intricate regulatory mechanisms during tumor development.In cancers like pancreatic


adenocarcinoma (PAAD) and adrenocortical carcinoma (ACC), elevated SMAD3 expression significantly correlates with poor prognosis, highlighting its potential as a biomarker for cancer


diagnosis and prognosis. However, SMAD3’s varied roles across different cancers underscore the need for further research to validate its potential as a universal cancer biomarker. Moreover,


its critical role in the TGF-β signaling pathway and its potential applications in regulating tumor cell migration and invasion are suggested. While this study briefly validates SMAD3’s


impact on proliferation and migration of liver hepatocellular carcinoma (LIHC) cells, further molecular biology experiments are necessary to elucidate specific mechanisms, including


immunohistochemistry and immunocytochemistry approaches.Lastly, considering data extraction from multiple databases, potential systematic biases should not be overlooked. These limitations


emphasize the necessity for broader research efforts to fully clarify SMAD3’s roles in cancer and its potential as a therapeutic target for cancer treatment. CONCLUSION Our study


systematically examined the significant correlation between SMAD3 expression and clinical features, prognosis, mutation status, DNA methylation, RNA methylation, immune checkpoints,


immunomodulatory genes, TMB, MSI, DNAss, RNAss, and drug susceptibility in several cancers, which helps us better understand the potential role of SMAD3 in pan cancer. DATA AVAILABILITY The


datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found in the article/Supplementary Material.


REFERENCES * Ferlay, J. et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. _Int. J. Cancer_. 136, E359–386.


https://doi.org/10.1002/ijc.29210 (2015). Article  CAS  PubMed  MATH  Google Scholar  * Zhang, Y. & Zhang, Z. The history and advances in cancer immunotherapy: understanding the


characteristics of tumor-infiltrating immune cells and their therapeutic implications. _Cell. Mol. Immunol._ 17, 807–821. https://doi.org/10.1038/s41423-020-0488-6 (2020). Article  CAS 


PubMed  PubMed Central  MATH  Google Scholar  * Ferlay, J. et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018. _Eur. J.


Cancer_. 103, 356–387. https://doi.org/10.1016/j.ejca.2018.07.005 (2018). Article  CAS  PubMed  Google Scholar  * Zou, J. & Wang, E. Cancer Biomarker Discovery for Precision Medicine:


New Progress. _Curr. Med. Chem._ 26, 7655–7671. https://doi.org/10.2174/0929867325666180718164712 (2019). Article  CAS  PubMed  MATH  Google Scholar  * Brown, K. A., Pietenpol, J. A. &


Moses, H. L. A tale of two proteins: differential roles and regulation of Smad2 and Smad3 in TGF-beta signaling. _J. Cell. Biochem._ 101, 9–33. https://doi.org/10.1002/jcb.21255 (2007).


Article  CAS  PubMed  Google Scholar  * Tang, P. C. T. et al. Smad3 promotes Cancer-Associated fibroblasts Generation via Macrophage-Myofibroblast transition. _Adv. Sci. (Weinh)_. 9,


e2101235. https://doi.org/10.1002/advs.202101235 (2022). Article  CAS  PubMed  Google Scholar  * Tang, P. C. T. et al. Hematopoietic transcription factor RUNX1 is essential for promoting


macrophage-myofibroblast transition in Non-small-cell Lung Carcinoma. _Adv. Sci. (Weinh)_. 11, e2302203. https://doi.org/10.1002/advs.202302203 (2024). Article  CAS  PubMed  Google Scholar 


* Yu, B. et al. KAT6A acetylation of SMAD3 regulates myeloid-derived suppressor cell recruitment, metastasis, and Immunotherapy in Triple-negative breast Cancer. _Adv. Sci. (Weinh)_. 8,


e2100014. https://doi.org/10.1002/advs.202100014 (2021). Article  CAS  PubMed  Google Scholar  * Tang, P. M. K. et al. Smad3 promotes cancer progression by inhibiting E4BP4-mediated NK cell


development. _Nat. Commun._ 8, 14677. https://doi.org/10.1038/ncomms14677 (2017). Article  ADS  PubMed  PubMed Central  Google Scholar  * Wang, Q-M. et al. Enhanced Cancer Immunotherapy with


Smad3-Silenced NK-92 cells. _Cancer Immunol. Res._ 6, 965–977. https://doi.org/10.1158/2326-6066.CIR-17-0491 (2018). Article  CAS  PubMed  MATH  Google Scholar  * Yang, S. et al. SEPHS1


promotes SMAD2/3/4 expression and hepatocellular carcinoma cells invasion. _Exp. Hematol. Oncol._ 10, 17. https://doi.org/10.1186/s40164-021-00212-7 (2021). Article  CAS  PubMed  PubMed


Central  Google Scholar  * Sun, J. et al. SMAD3 and FTO are involved in mir-5581-3p-mediated inhibition of cell migration and proliferation in bladder cancer. _Cell. Death Discov_. 8, 199.


https://doi.org/10.1038/s41420-022-01010-8 (2022). Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Su, Y. et al. circRIP2 accelerates bladder cancer progression via


miR-1305/Tgf-β2/smad3 pathway. _Mol. Cancer_. 19, 23. https://doi.org/10.1186/s12943-019-1129-5 (2020). Article  CAS  PubMed  PubMed Central  Google Scholar  * Zhou, Q., Han, L. R., Zhou, Y.


X. & Li, Y. MiR-195 suppresses cervical Cancer Migration and Invasion through Targeting Smad3. _Int. J. Gynecol. Cancer_. 26, 817–824. https://doi.org/10.1097/IGC.0000000000000686


(2016). Article  PubMed  MATH  Google Scholar  * Pan, J. et al. Circ_0001686 promotes prostate Cancer progression by Up-Regulating SMAD3/TGFBR2 via miR-411-5p. _World J. Mens Health_. 40,


149–161. https://doi.org/10.5534/wjmh.200204 (2022). Article  PubMed  Google Scholar  * Tarasewicz, E. et al. CDK4 inhibition and doxorubicin mediate breast cancer cell apoptosis through


Smad3 and survivin. _Cancer Biol. Ther._ 15, 1301–1311. https://doi.org/10.4161/cbt.29693 (2014). Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Peng, J. et al. Docetaxel


suppressed cell proliferation through Smad3/HIF-1α-mediated glycolysis in prostate cancer cells. _Cell. Commun. Signal._ 20, 194. https://doi.org/10.1186/s12964-022-00950-z (2022). Article 


CAS  PubMed  PubMed Central  Google Scholar  * Li, T. et al. TIMER: a web server for Comprehensive Analysis of Tumor-infiltrating Immune cells. _Cancer Res._ 77, e108–e110.


https://doi.org/10.1158/0008-5472.CAN-17-0307 (2017). Article  ADS  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Goldman, M. J. et al. Visualizing and interpreting cancer genomics


data via the Xena platform. _Nat. Biotechnol._ 38, 675–678. https://doi.org/10.1038/s41587-020-0546-8 (2020). Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Uhlen, M. et al. A


pathology atlas of the human cancer transcriptome. _Science_ 357, eaan2507. https://doi.org/10.1126/science.aan2507 (2017). Article  CAS  PubMed  MATH  Google Scholar  * Uhlén, M. et al.


Proteomics. Tissue-based map of the human proteome. _Science_ 347, 1260419. https://doi.org/10.1126/science.1260419 (2015). Article  CAS  PubMed  MATH  Google Scholar  * Liu, J. et al. An


Integrated TCGA Pan-cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. _Cell_ 173, 400–416e11. https://doi.org/10.1016/j.cell.2018.02.052 (2018). Article  CAS 


PubMed  PubMed Central  MATH  Google Scholar  * Liu, X., Wu, H. & Liu, Z. An Integrative Human Pan-cancer Analysis of Cyclin-Dependent Kinase 1 (CDK1). _Cancers (Basel)_. 14 (2658).


https://doi.org/10.3390/cancers14112658 (2022). * Pan, Y., Liu, G., Zhou, F., Su, B. & Li, Y. DNA methylation profiles in cancer diagnosis and therapeutics. _Clin. Exp. Med._ 18, 1–14.


https://doi.org/10.1007/s10238-017-0467-0 (2018). Article  CAS  PubMed  MATH  Google Scholar  * Yang, B. et al. RNA methylation and cancer treatment. _Pharmacol. Res._ 174, 105937.


https://doi.org/10.1016/j.phrs.2021.105937 (2021). Article  CAS  PubMed  MATH  Google Scholar  * Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using


the cBioPortal. _Sci. Signal._ 6, pl1. https://doi.org/10.1126/scisignal.2004088 (2013). Article  CAS  PubMed  PubMed Central  Google Scholar  * Mermel, C. H. et al. GISTIC2.0 facilitates


sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. _Genome Biol._ 12, R41. https://doi.org/10.1186/gb-2011-12-4-r41 (2011). Article


  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Yoshihara, K. et al. Inferring tumour purity and stromal and immune cell admixture from expression data. _Nat. Commun._ 4, 2612.


https://doi.org/10.1038/ncomms3612 (2013). Article  ADS  CAS  PubMed  MATH  Google Scholar  * Zeng, Z. et al. TISMO: syngeneic mouse tumor database to model tumor immunity and immunotherapy


response. _Nucleic Acids Res._ 50, D1391–D1397. https://doi.org/10.1093/nar/gkab804 (2022). Article  CAS  PubMed  Google Scholar  * Sun, D. et al. TISCH: a comprehensive web resource


enabling interactive single-cell transcriptome visualization of tumor microenvironment. _Nucleic Acids Res._ 49, D1420–D1430. https://doi.org/10.1093/nar/gkaa1020 (2021). Article  CAS 


PubMed  Google Scholar  * Yuan, H. et al. CancerSEA: a cancer single-cell state atlas. _Nucleic Acids Res._ 47, D900–D908. https://doi.org/10.1093/nar/gky939 (2019). Article  CAS  PubMed 


Google Scholar  * Kanehisa, M., Furumichi, M., Sato, Y., Kawashima, M. & Ishiguro-Watanabe, M. KEGG for taxonomy-based analysis of pathways and genomes. _Nucleic Acids Res._ 51,


D587–D592. https://doi.org/10.1093/nar/gkac963 (2023). Article  CAS  PubMed  Google Scholar  * Kanehisa, M. & Goto, S. KEGG: kyoto encyclopedia of genes and genomes. _Nucleic Acids Res._


28, 27–30. https://doi.org/10.1093/nar/28.1.27 (2000). Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Kanehisa, M. Toward understanding the origin and evolution of cellular


organisms. _Protein Sci._ 28, 1947–1951. https://doi.org/10.1002/pro.3715 (2019). Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Zhou, Y. et al. Metascape provides a


biologist-oriented resource for the analysis of systems-level datasets. _Nat. Commun._ 10, 1523. https://doi.org/10.1038/s41467-019-09234-6 (2019). Article  ADS  CAS  PubMed  PubMed Central


  MATH  Google Scholar  * Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. _Proc. Natl. Acad. Sci. U S A_.


102, 15545–15550. https://doi.org/10.1073/pnas.0506580102 (2005). Article  ADS  CAS  PubMed  PubMed Central  Google Scholar  * Dear, P. H. Copy-number variation: the end of the human genome?


_Trends Biotechnol._ 27, 448–454. https://doi.org/10.1016/j.tibtech.2009.05.003 (2009). Article  CAS  PubMed  MATH  Google Scholar  * Sui, S. et al. An immune cell infiltration-based immune


score model predicts prognosis and chemotherapy effects in breast cancer. _Theranostics_ 10, 11938–11949. https://doi.org/10.7150/thno.49451 (2020). Article  CAS  PubMed  PubMed Central 


MATH  Google Scholar  * Chen, F. et al. Signatures of immune cell infiltration for predicting immune escape and immunotherapy in cervical cancer. _Aging (Albany NY)_. 15, 1685–1698.


https://doi.org/10.18632/aging.204583 (2023). Article  ADS  PubMed  MATH  Google Scholar  * Bagchi, S., Yuan, R. & Engleman, E. G. Immune Checkpoint inhibitors for the treatment of


Cancer: clinical impact and mechanisms of response and resistance. _Annu. Rev. Pathol._ 16, 223–249. https://doi.org/10.1146/annurev-pathol-042020-042741 (2021). Article  CAS  PubMed  Google


Scholar  * Chen, P., Hsu, W-H., Han, J., Xia, Y. & DePinho, R. A. Cancer Stemness meets immunity: from mechanism to Therapy. _Cell. Rep._ 34, 108597.


https://doi.org/10.1016/j.celrep.2020.108597 (2021). Article  CAS  PubMed  PubMed Central  Google Scholar  * Schaub, F. X. et al. Pan-cancer alterations of the MYC Oncogene and its Proximal


Network across the Cancer Genome Atlas. _Cell. Syst._ 6, 282–300e2. https://doi.org/10.1016/j.cels.2018.03.003 (2018). Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Fleming,


N. I. et al. SMAD2, SMAD3 and SMAD4 mutations in colorectal cancer. _Cancer Res._ 73, 725–735. https://doi.org/10.1158/0008-5472.CAN-12-2706 (2013). Article  CAS  PubMed  MATH  Google


Scholar  * Huang, Y-T. et al. Chen W-Y. USP7 facilitates SMAD3 autoregulation to repress cancer progression in p53-deficient lung cancer. _Cell. Death Dis._ 12, 880.


https://doi.org/10.1038/s41419-021-04176-8 (2021). Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Zhao, Y. et al. TGF-β transactivates EGFR and facilitates breast cancer


migration and invasion through canonical Smad3 and ERK/Sp1 signaling pathways. _Mol. Oncol._ 12, 305–321. https://doi.org/10.1002/1878-0261.12162 (2018). Article  CAS  PubMed  PubMed Central


  MATH  Google Scholar  * Li, Y. et al. IGHG1 induces EMT in gastric cancer cells by regulating TGF-β/SMAD3 signaling pathway. _J. Cancer_. 12, 3458–3467. https://doi.org/10.7150/jca.56056


(2021). Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Huang, C. et al. EZH2-triggered methylation of SMAD3 promotes its activation and tumor metastasis. _J. Clin. Invest._


132, e152394. https://doi.org/10.1172/JCI152394 (2022). Article  CAS  PubMed  PubMed Central  Google Scholar  * Ali, S. R., Jordan, M., Nagarajan, P. & Amit, M. Nerve density and


neuronal biomarkers in Cancer. _Cancers (Basel)_. 14, 4817. https://doi.org/10.3390/cancers14194817 (2022). Article  CAS  PubMed  MATH  Google Scholar  * Lei, X. et al. Immune cells within


the tumor microenvironment: Biological functions and roles in cancer immunotherapy. _Cancer Lett._ 470, 126–133. https://doi.org/10.1016/j.canlet.2019.11.009 (2020). Article  CAS  PubMed 


MATH  Google Scholar  * Yang, K. et al. Glioma targeted therapy: insight into future of molecular approaches. _Mol. Cancer_. 21, 39. https://doi.org/10.1186/s12943-022-01513-z (2022).


Article  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Ju, Q. et al. NFE2L2 is a potential prognostic biomarker and is correlated with Immune Infiltration in Brain Lower Grade Glioma:


a Pan-cancer Analysis. _Oxid. Med. Cell. Longev._ 2020, 3580719. https://doi.org/10.1155/2020/3580719 (2020). Article  CAS  PubMed  PubMed Central  Google Scholar  * Lin, A., Zhang, J.


& Luo, P. Crosstalk between the MSI Status and Tumor Microenvironment in Colorectal Cancer. _Front. Immunol._ 11, 2039. https://doi.org/10.3389/fimmu.2020.02039 (2020). Article  CAS 


PubMed  PubMed Central  MATH  Google Scholar  * Wei, F. et al. PD-L1 promotes colorectal cancer stem cell expansion by activating HMGA1-dependent signaling pathways. _Cancer Lett._ 450,


1–13. https://doi.org/10.1016/j.canlet.2019.02.022 (2019). Article  ADS  CAS  PubMed  Google Scholar  * Nallasamy, P. et al. Pancreatic tumor microenvironment factor promotes Cancer Stemness


via SPP1-CD44 Axis. _Gastroenterology_ 161, 1998–2013e7. https://doi.org/10.1053/j.gastro.2021.08.023 (2021). Article  CAS  PubMed  Google Scholar  * Zhang, X. et al. Application of


weighted gene co-expression network analysis to identify key modules and hub genes in oral squamous cell carcinoma tumorigenesis. _Onco Targets Ther._ 11, 6001–6021.


https://doi.org/10.2147/OTT.S171791 (2018). Article  ADS  CAS  PubMed  PubMed Central  MATH  Google Scholar  * Kuburich, N. A. et al. Proactive and reactive roles of TGF-β in cancer. _Semin


Cancer Biol._ 95, 120–139. https://doi.org/10.1016/j.semcancer.2023.08.002 (2023). Article  CAS  PubMed  PubMed Central  Google Scholar  * Iser, I. C. et al. The crossroads of adenosinergic


pathway and epithelial-mesenchymal plasticity in cancer. _Semin Cancer Biol._ 86, 202–213. https://doi.org/10.1016/j.semcancer.2022.06.012 (2022). Article  CAS  PubMed  MATH  Google Scholar


  Download references ACKNOWLEDGEMENTS We would like to express our sincere gratitude for the contributions provided by the TCGA Pan-cancer Project and Sangerbox websites. FUNDING This study


was supported by the National Natural Science Foundation of China [82060049, 82170289 and 82060048] and Jiangxi Natural Science Foundation [20202BAB216009]. AUTHOR INFORMATION AUTHORS AND


AFFILIATIONS * Jiangxi Key Laboratory of Molecular Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, China Tao


Zhou, Jiejing jin, Jinyang Xie & Rong Wan * Department of General Surgery, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang,


330006, China Dan Dan Zhang * Department of Cardiovascular Medicine, Jiangxi Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006,


China Jianhua Yu * Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China Chao Zhu & Rong Wan Authors * Tao Zhou View author


publications You can also search for this author inPubMed Google Scholar * Dan Dan Zhang View author publications You can also search for this author inPubMed Google Scholar * Jiejing jin


View author publications You can also search for this author inPubMed Google Scholar * Jinyang Xie View author publications You can also search for this author inPubMed Google Scholar *


Jianhua Yu View author publications You can also search for this author inPubMed Google Scholar * Chao Zhu View author publications You can also search for this author inPubMed Google


Scholar * Rong Wan View author publications You can also search for this author inPubMed Google Scholar CONTRIBUTIONS ZC and WR contributed to the conception and design of the study. ZT,


ZDD, JJJ, YJH and XJY drafted the manuscript. ZT and ZDD collected and analyzed the data. ZC and WR revised the manuscript. All authors contributed to the manuscript revision and read and


approved the submitted version. CORRESPONDING AUTHORS Correspondence to Chao Zhu or Rong Wan. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing interests. ETHICS


STATEMENT This study was conducted in accordance with all appropriate guidelines and regulations stipulated by the Declaration of Helsinki. The experimental received approval from the


institutional review board at The Second Affiliated Hospital of Nanchang University. Written informed consent was secured from all participants who agreed to partake in this research.


ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. ELECTRONIC SUPPLEMENTARY


MATERIAL Below is the link to the electronic supplementary material. SUPPLEMENTARY MATERIAL 1 SUPPLEMENTARY MATERIAL 2 SUPPLEMENTARY MATERIAL 3 SUPPLEMENTARY MATERIAL 4 SUPPLEMENTARY


MATERIAL 5 SUPPLEMENTARY MATERIAL 6 SUPPLEMENTARY MATERIAL 7 SUPPLEMENTARY MATERIAL 8 SUPPLEMENTARY MATERIAL 9 RIGHTS AND PERMISSIONS OPEN ACCESS This article is licensed under a Creative


Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as


you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have


permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s


Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not


permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit


http://creativecommons.org/licenses/by-nc-nd/4.0/. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Zhou, T., Zhang, D.D., jin, J. _et al._ Multiomic characterization,


immunological and prognostic potential of SMAD3 in pan-cancer and validation in LIHC. _Sci Rep_ 15, 657 (2025). https://doi.org/10.1038/s41598-024-84553-3 Download citation * Received: 19


March 2024 * Accepted: 24 December 2024 * Published: 03 January 2025 * DOI: https://doi.org/10.1038/s41598-024-84553-3 SHARE THIS ARTICLE Anyone you share the following link with will be


able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing


initiative KEYWORDS * smad3 * Pan-cancer * TGF-β * LIHC * Diagnosis * Prognosis * Immune infiltration