Fact-checked by Grok 2 weeks ago

SOX9

SOX9, also known as SRY-box transcription factor 9, is a gene located on the human chromosome 17q24.3 that encodes a transcription factor belonging to the SOX family of proteins, characterized by a high-mobility group (HMG) DNA-binding domain similar to that of the sex-determining region Y (SRY) gene. This protein plays a pivotal role in embryonic development, particularly in sex determination, chondrogenesis, and the formation of multiple organs such as the testis, skeleton, pancreas, lung, heart, and neural crest-derived structures. Recent research in 2025 identified a Y-chromosome-specific duplication near SOX9 that enhances its expression in males, potentially influencing male development and fertility. SOX9 functions primarily as a regulator of gene expression, activating or repressing target genes to direct cell differentiation, proliferation, and extracellular matrix production across mesodermal, ectodermal, and endodermal lineages. In sex determination, SOX9 is indispensable for testicular development; it is expressed in the bipotential of XY embryos around embryonic day 11.5 in mice, where it cooperates with SRY to specify Sertoli cells and upregulate genes like AMH, FGF9, and PTGDS, leading to male differentiation. Mutations or in SOX9 cause , a severe skeletal disorder characterized by bowing of long bones, hypoplastic scapulae, and often 46, sex reversal in approximately 75% of affected males due to . Beyond reproduction, SOX9 acts as a master regulator of chondrogenesis by activating cartilage-specific genes such as Col2a1 while repressing hypertrophic markers like Col10a1, ensuring proper skeletogenesis and . SOX9 also maintains stem and progenitor cell pools in various adult tissues, including intestinal crypts, hair follicles, neural progenitors, and retinal cells, where it balances self-renewal and . In the , it sustains endocrine and exocrine progenitor populations during , and in the , it coordinates branching by modulating , , and extracellular matrix remodeling. Dysregulation of SOX9 is implicated in several pathologies; for instance, it contributes to in organs like the liver and through excessive deposition, and exhibits context-dependent roles in cancer, acting as an in , pancreatic, and ovarian tumors by promoting , epithelial-mesenchymal , and (as of 2025), or as a tumor suppressor in certain cancers where it influences immune evasion and . Duplications of SOX9, conversely, can lead to 46,XX , highlighting its dosage-sensitive nature in gonadal development. Overall, SOX9's multifaceted transcriptional activities underscore its importance as a therapeutic target for developmental disorders, , and malignancies.

Gene and Protein Overview

Gene Structure and Expression

The SOX9 is located on the long arm of human chromosome 17 at the 17q24.3 cytogenetic band, with genomic coordinates spanning approximately 5.4 kb from 72,121,020 to 72,126,416 (GRCh38/hg38 assembly). It consists of three exons separated by two small introns, producing a mature mRNA transcript of 3.9 kb (NM_000346.4; ENST00000245479 in Ensembl) that encodes a 509-amino-acid protein. According to Ensembl, there are two transcripts, with the canonical ENST00000245479 producing the full-length protein. The was identified in 1994 through positional cloning efforts targeting translocation breakpoints in patients with , a skeletal disorder often accompanied by . Its official OMIM entry is 608160. Alternative splicing of SOX9 pre-mRNA yields at least two isoforms in humans, though the predominant full-length transcript (NM_000346.4) includes all three exons and translates into the canonical 509-residue protein essential for its transcriptional activity. Minor isoforms may arise from or , but these are less abundant and their functional roles remain under investigation. SOX9 expression is dynamically regulated during embryonic and persists in select adult tissues, driven by multiple cis-regulatory enhancers located upstream of the within a large gene desert spanning over 1 Mb. High levels are observed in chondroprogenitors and differentiating chondrocytes of the developing , Sertoli cells of the embryonic testis and adult gonads, neural crest-derived structures such as branchial arches and otic vesicles, and mesodermal derivatives including the heart and gut . In adults, SOX9 maintains elevated expression in testis, (particularly glial cells), cardiac chondrocytes, and intestinal crypts. A key 32.5-kb regulatory region approximately 600 kb upstream, known as the XY sex reversal region (XYSR), contains enhancers critical for SOX9 activation in limb bud and prominences during craniofacial . The HMG box domain of SOX9, a 79-amino-acid DNA-binding central to its function as a , exhibits strong evolutionary conservation across , with sequence identity exceeding 90% between and other mammals, birds, and fish, underscoring its ancient role in developmental processes. This conservation extends to non-vertebrate chordates, reflecting the SOX family's origins over 500 million years ago.

Protein Structure and Domains

SOX9 is a 56 kDa transcription factor belonging to the SOX (SRY-related HMG-box) family of proteins, characterized by a conserved high-mobility group (HMG) DNA-binding domain. The human SOX9 protein consists of 509 amino acids and is encoded by the SOX9 gene on chromosome 17. Its structure includes an N-terminal transactivation domain (TAD), a central HMG domain responsible for DNA binding, and a C-terminal proline- and glutamine-rich region that serves as an additional transactivation domain. These domains enable SOX9 to function as a potent transcriptional activator in various developmental processes. The central HMG domain, spanning approximately 79 , binds with high affinity to a DNA , such as AACAAT flanked by AG and GG nucleotides, with a dissociation constant (Kd) of approximately 12.4 nM. Crystallographic studies of the SOX9 HMG domain in complex with DNA reveal that it induces a significant bend in the DNA helix, typically by 80-90 degrees, facilitating interactions with other regulatory proteins and enhancing transcriptional specificity. This bending is achieved through the L-shaped of the HMG box, which inserts into the minor groove of DNA. Post-translational modifications play crucial roles in modulating SOX9 activity. Phosphorylation occurs at specific serine residues, such as Ser181, often mediated by the MAPK/ERK signaling pathway, which can enhance nuclear localization and transcriptional potency. Additionally, sumoylation at conserved lysine residues, such as Lys61 and Lys398, influences SOX9's stability and interaction with co-factors, generally repressing its potential. These modifications allow fine-tuned regulation of SOX9 function in response to cellular signals. Alternative isoforms of SOX9, including truncated variants that lack the N-terminal or C-terminal domains due to or mutations, exhibit significantly reduced transcriptional activity compared to the full-length protein. For instance, forms missing the /glutamine-rich C-terminal region fail to effectively recruit the transcriptional machinery, underscoring the importance of these domains for SOX9's regulatory roles.

Regulation and Localization

Transcriptional and Post-Translational Regulation

The expression of SOX9 is primarily initiated during male gonadal development by the SRY protein, which binds to enhancers such as the testis-specific enhancer core (TESCO) within the SOX9 regulatory region to directly activate transcription and drive differentiation. This activation is amplified through a loop involving 9 (FGF9) and (PGD2), where SOX9 upregulates FGF9 expression, and FGF9 in turn promotes SOX9 transcription independently of PGD2 in some contexts, ensuring robust amplification in the developing testis. Key cis-regulatory enhancers, including a 1.45 Mb upstream region (EC1.45), respond to extrinsic signals such as , which collaborates with Wnt/β-catenin signaling to modulate SOX9 in chondrogenic differentiation of pluripotent stem cells. Wnt signaling also influences SOX9 enhancers in , though it often acts repressively in gonadal contexts to prevent ectopic activation. In female gonads, SOX9 expression is actively repressed to favor ovarian development. The transcription factor WT1 suppresses SOX9 by directing lineage specification and inhibiting SOX9 promoter activity through interactions with receptor 2 (KDR) signaling. Similarly, (SF1, also known as NR5A1) contributes to repression in XX gonads, as Wnt/β-catenin signaling inhibits SF1-mediated activation of the SOX9 promoter and (AMH) expression, thereby blocking male pathway progression. Beyond transcription factors, epigenetic mechanisms enforce SOX9 silencing in non-expressing tissues; lysine 27 trimethylation () deposits repressive marks near the SOX9 locus, including within transposable elements in the TESCO enhancer, maintaining in a closed state during ovarian . Post-translational modifications further fine-tune SOX9 protein levels and function. of SOX9 by the p300 promotes its transcriptional activity by facilitating recruitment to and at target loci, though this primarily enhances activation rather than directly stabilizing the protein itself.48191-X/fulltext) Conversely, ubiquitination targets SOX9 for proteasomal via the ubiquitin-proteasome system, with in ubiquitin-target sites stabilizing the protein and increasing its activity; while their direct role in SOX9 turnover remains under . of SOX9 by (PKC) modulates its activity in chondrogenesis, with PKC downregulation altering SOX9 states to influence transcriptional output, and related kinases like enhancing nuclear import through sites such as Ser181 to promote localization and function. SOX9 exhibits dose-dependent effects critical for developmental processes, particularly chondrogenesis, where levels below 50% of wild-type activity are insufficient to fully activate target genes like (COL2A1) and support formation. This threshold sensitivity underscores SOX9's role as a rheostat in lineage commitment, with leading to skeletal defects as seen in . Recent genetic studies have revealed evolutionary influences on SOX9 , including Neanderthal-derived in the 1.45 Mb upstream enhancer that increase SOX9 activity in craniofacial progenitors, potentially contributing to differences in between Neanderthals and modern humans.

Subcellular Localization and Dynamics

SOX9 is primarily a protein, directed to the through a bipartite localization signal (NLS) embedded within its high-mobility group (HMG) domain. This NLS consists of an N-terminal sequence ( 106–122) and a C-terminal sequence ( 175–180), which facilitate recognition by proteins for efficient import. Within the nucleus, SOX9 exhibits high mobility, characterized by a diffusion coefficient of approximately 10–20 μm²/s in the nucleoplasm. Fluorescence recovery after photobleaching (FRAP) analyses reveal that roughly 50% of SOX9 is chromatin-bound, with the remainder freely diffusing. The residence half-time of SOX9 on DNA is about 14 seconds, indicating transient interactions that allow rapid scanning and binding to target sites. External signals modulate SOX9's nuclear dynamics; for instance, bone morphogenetic protein (BMP) signaling enhances chromatin retention by increasing the immobile fraction from ~54% to ~66% and extending the recovery half-time from ~14 seconds to ~19 seconds, thereby promoting transcriptional activity. Conversely, SOX9 undergoes CRM1-dependent nuclear export in response to cellular stress, mediated by a nuclear export signal (NES) within the HMG domain (amino acids 134–147), which facilitates translocation to the cytoplasm under conditions such as calcification or signaling perturbations. Subcellular fractionation studies in chondrocytes demonstrate that 80–90% of SOX9 localizes to the under steady-state conditions, underscoring its role as a dedicated regulator. shuttling of SOX9 is further influenced by ; for example, ()-mediated at serine residues S64 and S181 enhances nuclear import and retention by altering conformational exposure of the NLS.

Biological Functions

Role in Skeletal and Cartilage Development

SOX9 plays a pivotal role in chondrogenesis by driving the of mesenchymal cells into within precartilaginous condensations. It directly transactivates key matrix genes, including Col2a1 (encoding ) and Acan (encoding aggrecan), which are essential for production and formation. Inactivation of SOX9 prior to mesenchymal condensation abolishes expression of these genes, preventing overt and leading to complete loss of skeletal elements. SOX9 significantly impairs this process, resulting in defective primordia and reduced expression. In limb development, SOX9 coordinates with SOX5 and SOX6 as transcriptional co-factors to regulate proliferation and matrix production, ensuring proper skeletal patterning. These factors collectively bind to enhancers of cartilage-specific genes, promoting robust chondrogenesis in developing limbs. Absence of SOX9 leads to delayed , increased in condensations, and severe chondrodysplasia characterized by underdeveloped long bones. Post-condensation SOX9 loss further disrupts proliferation and formation, highlighting its stage-specific necessity. SOX9 is crucial for craniofacial skeletal development, where it influences -derived progenitors essential for formation. It supports the migration of cranial cells to pharyngeal arches and is required for their subsequent differentiation into chondrocytes, as evidenced by species-specific loss-of-function studies showing depleted skeletal elements like Meckel's upon SOX9 knockdown. In palatal development, SOX9 expression in subepithelial drives shelf growth and ; its conditional inactivation results in cleft due to arrested shelf . A 2025 study identified Neanderthal-derived variants in the EC1.45 enhancer region upstream of SOX9 that enhance its activity in craniofacial progenitors, leading to increased precartilaginous volume and potentially contributing to Neanderthal-specific , such as enlarged retromolar . Through negative feedback with , SOX9 inhibits hypertrophic chondrocyte differentiation to preserve cartilage integrity and prevent premature . SOX9 directly upregulates Bapx1, which in turn represses expression and activity, blocking the transition to and maintaining identity. This regulatory loop ensures balanced skeletogenesis, with SOX9 dominance over during early stages.

Role in Sexual Differentiation

SOX9 plays a central role in mammalian sex determination by acting as the primary effector downstream of the Y-linked SRY gene, driving the of s in the bipotential of embryos. In gonads, SOX9 expression is rapidly upregulated following transient SRY activation, establishing a mutual loop that amplifies the male developmental signal. This loop involves SOX9 autoregulation and synergistic interactions with fibroblast growth factor 9 (FGF9) and (PGD2), which further enhance SOX9 transcription and promote specification while suppressing ovarian fates. A key aspect of SOX9's function is the repression of the ovarian differentiation pathway, achieved by inhibiting key pro-ovarian factors such as FOXL2 and WNT4, thereby preventing granulosa cell development and ensuring commitment to testis formation. The dosage of SOX9 is critical for this balance; in XY mouse models, complete knockout of Sox9 results in full sex reversal, with gonads developing ovarian structures including follicles, due to unrestrained activation of the WNT4/β-catenin pathway. Conversely, in XX individuals lacking SRY, upstream duplications of SOX9 regulatory elements, such as those in the RevSex region, cause SOX9 overexpression and trigger testicular or ovotesticular development, as documented in recent genetic analyses of 46,XX disorders of sex development (DSD).01433-0) The role of SOX9 in sex determination is evolutionarily conserved across mammals, where it serves as an ancestral trigger for testis formation, independent of SRY in some non-mammalian vertebrates. In humans, heterozygous in SOX9 underlie and cause 46,XY complete with in approximately 70-80% of affected XY individuals, highlighting its dosage sensitivity. Temporally, SOX9 expression peaks around embryonic day 11.5 (E11.5) in mouse XY gonads, coinciding with the initiation of differentiation and the upregulation of (AMH), which SOX9 directly activates to induce regression of the Müllerian ducts and prevent female reproductive tract development.

Roles in Neural and Stem Cell Development

SOX9 plays a pivotal role in neural development by promoting the of and while inhibiting premature differentiation, particularly in the ventricular zone of the developing . In the , SOX9 expression in basal enhances their proliferative capacity, contributing to neocortical expansion during embryogenesis. High levels of SOX9 in radial glial cells extend the duration, thereby delaying the onset of and allowing for a larger pool. This function is essential for transforming neuroepithelial into multipotent , as evidenced by studies showing SOX9's necessity in cerebellar development where its absence disrupts maintenance. In gliogenesis, SOX9 is critical for directing neural progenitors toward glial fates, including the specification of . SOX9 regulates the expression of Olig2, a key for (OPC) generation and differentiation, ensuring timely production of these cells in regions like the and . Loss of SOX9 leads to sparse Olig2 expression in migrating OPCs and impairs their survival and migration, highlighting its role in the neurogenic-to-gliogenic switch during midgestation when ventricular zone stem cells transition from neuronal to glial output. Beyond core neural lineages, SOX9 maintains pluripotency and multipotency in stem cells, enabling their broad differentiation potential. SOX9 induces neural crest-like properties in progenitors, promoting ectomesenchymal fates at the expense of neuronal differentiation. In human pluripotent -derived models, SOX9-positive ectomesenchymal cells derived from neural crest progenitors exhibit long-term expandability while retaining chondrogenic potential, underscoring SOX9's role in sustaining stemness. Additionally, SOX9 interacts with the to drive gliogenesis, where maintains SOX9 expression to promote astrogliogenesis and proliferation in the developing . Knockdown of Notch-induced SOX9 reverses astroglial differentiation and stem cell expansion, confirming SOX9 as a downstream of Notch in glial fate commitment. In the adult brain, SOX9 is expressed in the (SVZ), where it acts as a nuclear marker in functioning as neural stem cells, influencing the balance between and gliogenesis. SOX9 overexpression in SVZ cells suppresses neuronal production while supporting the gliogenic switch, and its knockdown increases formation, indicating a regulatory role in maintenance. SOX9 loss disrupts this balance, impairing gliogenesis and reducing glial output, with studies showing up to a 50% decrease in glial differentiation efficiency in SOX9-deficient models. Recent investigations have linked SOX9 modulation to enhanced neurological recovery post-stroke, with conditional SOX9 mice exhibiting reduced levels, increased tissue sparing, and improved functional outcomes through promoted axonal . These findings suggest SOX9 inhibition as a therapeutic target for limiting inhibitory deposition after ischemic injury. SOX9 also contributes to development, particularly in specification within the otic placode. Early SOX9 expression in the otic is required for placode and maintenance, with depletion leading to and failure in neurosensory domain formation. SOX9 coordinates with factors like and to pattern the cochlear duct, ensuring proper specification of s and supporting fluid in the mature .

Molecular Interactions

Protein-Protein Interactions

SOX9 engages in direct protein-protein interactions with several key partners that modulate its transcriptional activity, particularly in developmental contexts. In mammalian sex determination, SOX9 cooperates with the sex-determining region Y (SRY) protein through shared binding to target promoters, facilitating mutual reinforcement of their expression in precursors to drive testis differentiation. SOX9 also forms a direct complex with (SF1, also known as NR5A1), where the high-mobility-group (HMG) domain of SOX9 binds the C-terminal ligand-binding domain of SF1; this interaction cooperatively activates the (AMH) promoter, with the SF1-SOX9 complex increasing AMH transcription by approximately 10- to 20-fold in cotransfection assays. Additionally, SOX9 interacts with MED12, a subunit of complex, via its transcriptional activation domain, enabling recruitment of to enhance target gene transcription during chondrogenesis and other processes. Beyond these core partners, SOX9 associates with factors that fine-tune dynamics and epigenetic modifications. In chondrocytes, SOX9 synergizes with the MAF (specifically the long isoform Lc-MAF) to co-activate enhancers of cartilage-specific genes like Col2a1, promoting chondrogenic differentiation through direct binding and cooperative transcriptional enhancement. SOX9 further recruits components of the complex, including ARID1A/B and SMARCD2, to increase accessibility at closed enhancers, thereby facilitating SOX9-dependent gene in stem cell fate transitions. For epigenetic , SOX9 directly interacts with the histone methyltransferases MLL3 and MLL4, recruiting them to enhancers where they deposit H3K4me1 marks to prime transcriptional prior to opening. SOX9 activity is also counteracted by inhibitory interactions. In ovarian development, FOXL2 competes with SOX9 for binding sites, repressing SOX9 target genes to maintain female identity and prevent toward a male fate. SOX9 stability is negatively regulated by the ubiquitin-proteasome system, with in its target site leading to increased protein and enhanced transcriptional output. Recent studies have extended SOX9's interactome to immune regulation, where it modulates , though direct protein-level engagements with T-cell receptor components remain under investigation.

Target Genes and Regulatory Pathways

SOX9 directly regulates several key transcriptional targets essential for development and . In chondrogenesis, SOX9 activates expression of Col2a1 and Acan by binding to their enhancers, often in cooperation with SOX5 and SOX6, to promote matrix production. In , SOX9 induces (AMH) in Sertoli cells of the developing testis, driving Müllerian duct regression and male gonad formation. Additionally, SOX9 forms a loop with FGF9 in gonads, where SOX9 upregulates Fgf9, and FGF9 signaling reinforces SOX9 expression to stabilize male fate. In neural development, SOX9 promotes gliogenesis by inducing Olig2 expression in basal progenitors, shifting cell fate from toward lineage commitment. Genome-wide ChIP-seq studies reveal that SOX9 binds to over 1,000 sites across the genome, with peaks predominantly enriched in enhancers and intronic regions rather than promoters. The consensus binding motif for SOX9 is the palindromic sequence MAWWMAWR, facilitating both monomeric and dimeric binding to DNA for transcriptional activation or repression. SOX9 modulates multiple signaling pathways through these targets. In cartilage development, it activates the BMP/SMAD pathway by regulating noggin expression, providing feedback to fine-tune chondrocyte differentiation. In gonadal development, SOX9 inhibits the Wnt/β-catenin pathway by promoting β-catenin ubiquitination and degradation, suppressing female differentiation cues. For limb patterning, SOX9 integrates with Hedgehog signaling, where Shh induces SOX9 in mesenchymal condensations to coordinate proximal-distal and anteroposterior skeletal organization. SOX9 dosage exerts context-specific effects on target gene expression, with low levels activating proliferation-associated genes such as Ccnd1 to support expansion, while high levels drive programs including Sox5 and Sox6 for lineage commitment. In cancer contexts, recent analyses highlight SOX9's role in regulating stemness genes like Nanog, promoting a stem-like transcriptional state that confers resistance to platinum-based in high-grade serous .

Clinical Significance

Genetic Disorders and Mutations

Mutations in the SOX9 gene are primarily associated with (CD), a rare autosomal dominant skeletal dysplasia syndrome caused by heterozygous loss-of-function variants, including frameshift, nonsense, and missense mutations that disrupt the protein's DNA-binding or transactivation domains. These mutations typically occur and lead to of SOX9, impairing chondrogenesis and gonadal development. Clinical features include bowing and angulation of long bones (campomelia), hypoplastic scapulae, 11 pairs of ribs, and craniofacial abnormalities such as a flat face, high forehead, and cleft palate; approximately 75% of affected 46,XY individuals exhibit manifesting as complete and female external genitalia. The condition is lethal in about 80% of cases due to respiratory insufficiency from or thoracic , though milder variants can allow survival into adulthood with medical interventions. SOX9 mutations account for nearly all cases of CD, with over 100 distinct pathogenic variants reported, predominantly clustered in the high-mobility group (HMG) . A related but milder condition, acampomelic campomelic dysplasia (ACD), arises from specific SOX9 mutations that preserve partial protein function, resulting in skeletal manifestations without the characteristic long bone bowing or . Affected individuals typically present with , axial skeletal defects, and craniofacial dysmorphism but lack campomelia and gonadal involvement, highlighting the genotype-phenotype correlation where less disruptive mutations correlate with attenuated severity. ACD represents a clinical spectrum overlapping with CD, emphasizing SOX9's dosage-sensitive role in skeletal patterning. Isolated Pierre Robin sequence (PRS), characterized by micrognathia, glossoptosis, and cleft palate, can result from disruptions in SOX9 upstream regulatory enhancers rather than coding mutations, leading to reduced SOX9 expression in neural crest-derived tissues. For instance, a 117 kb deletion approximately 1.5 Mb upstream of SOX9 abolishes enhancer activity critical for craniofacial development, causing PRS without broader skeletal or gonadal defects. These non-coding variants underscore the importance of long-range cis-regulatory elements in SOX9 dosage control during embryogenesis. SOX9 mutations contribute to 10-20% of cases of 46,XY gonadal dysgenesis, particularly those accompanied by skeletal anomalies as seen in CD. In these instances, loss-of-function variants prevent SOX9 upregulation in the bipotential gonad, blocking Sertoli cell differentiation and testis formation, resulting in streak gonads and female phenotype. Recent studies have expanded the spectrum of SOX9-related disorders. A 2025 report identified missense variants in the transactivation middle (TAM) domain of SOX9 that reduce protein stability without abolishing DNA binding, causing a milder axial skeletal dysplasia phenotype including scoliosis and vertebral anomalies but sparing limb and gonadal development. These variants highlight the TAM domain's role in modulating SOX9 activity for axial patterning. Additionally, a 2024 analysis of upstream duplications encompassing SOX9 enhancers revealed their association with 46,XX ovotesticular disorders of sex development (DSD), where ectopic SOX9 overexpression drives testis differentiation and ovotesticular histology in the absence of SRY. Such duplications, often inherited, represent a key genetic mechanism in non-SRY 46,XX testicular/ovotesticular DSD, second only to SRY translocations in prevalence among identified causes.

Role in Oncogenesis and Cancer Progression

SOX9 is frequently overexpressed in various solid tumors, contributing to oncogenic transformation and progression. In , SOX9 expression is upregulated by (AR) signaling, where it cooperates with AR to promote tumor initiation and invasion; for instance, ERG fusion proteins induce SOX9 via AR binding sites, enhancing neoplastic growth in murine models. Similarly, in , SOX9 maintains properties by activating stemness programs that inhibit intestinal differentiation, thereby sustaining tumor propagation and resistance to therapy. In , elevated SOX9 drives endocrine resistance by promoting lineage plasticity and , as highlighted in a 2025 review emphasizing its role in therapy-refractory . Overexpression of SOX9 also confers resistance in high-grade serous by inducing a stem-like transcriptional state that enhances survival under chemotherapeutic stress, according to a 2025 study. Mechanistically, SOX9 promotes oncogenesis through multiple pathways, including enhancement of epithelial-mesenchymal transition (EMT), which facilitates invasion and metastasis across cancer types; for example, SOX9 upregulates EMT markers like vimentin and snail in non-small cell lung cancer cells, driving distant spread. Its oncogenic effects are dose-dependent, with high SOX9 levels stimulating proliferation via direct regulation of Ccnd1 (cyclin D1), a key cell cycle promoter, while lower levels may support stemness without aggressive growth. Notably, SOX9 loss in colon cancer models paradoxically accelerates tumor progression by disrupting differentiation barriers, leading to increased invasion and poor prognosis, as demonstrated in 2025 research. In lung adenocarcinoma, SOX9 remodels the immune microenvironment by suppressing anti-tumor immunity, fostering an immunosuppressive niche that supports KRAS-driven progression, per a 2024 analysis. High SOX9 expression serves as a prognostic indicator, correlating with poor overall survival in advanced and cases, positioning it as a potential for aggressive disease. Recent studies underscore SOX9's targetability in specific malignancies; in , elevated SOX9 in dormant cells facilitates MYC-driven recurrence, suggesting therapeutic windows for SOX9 inhibition in 2025 preclinical models. Likewise, in , SOX9 regulates cancer maintenance and self-renewal, promoting tumor heterogeneity and , as reviewed in 2024.

Emerging Roles in Non-Neoplastic Diseases

SOX9 has been implicated in the of organ , where its upregulation in fibrotic tissues contributes to (ECM) deposition and scarring. In liver fibrosis, SOX9 expression is elevated in activated hepatic stellate cells, promoting the transcription of ECM components such as type I and inhibiting antifibrotic pathways, thereby exacerbating tissue stiffness and dysfunction. Similarly, in kidney , SOX9 drives the of epithelial cells into myofibroblasts, enhancing I synthesis and renal scarring, as observed in models of . A 2025 review highlights SOX9 as a central regulator across multiple fibrotic organs, suggesting its inhibition as a potential therapeutic target to mitigate progressive scarring without affecting developmental roles. In metabolic disorders, SOX9 exhibits protective effects against hepatic lipid accumulation in metabolic dysfunction-associated (MASH). Overexpression of SOX9 in hepatocytes activates the (AMPK) pathway, which suppresses and promotes oxidation, thereby reducing and in high-fat diet models. Hepatocyte-specific SOX9 deletion, conversely, worsens MASH progression by impairing AMPK signaling and increasing formation, underscoring its role in maintaining metabolic in the liver. These findings from a 2024 study position SOX9 as a promising modulator for MASH therapy, potentially through targeted gene delivery to enhance AMPK-mediated lipid clearance. SOX9 functions as a Janus-faced in immune responses, influencing T-cell development while also contributing to autoimmune . In thymic T-cell maturation, SOX9 maintains stability but its dysregulation promotes autoreactive T-cell escape, fostering in conditions like . A 2025 study reveals that SOX9 modulates T-cell infiltration and production in inflammatory milieus, with elevated levels correlating to exacerbated immune-mediated tissue damage, yet controlled expression could suppress overactive responses. This dual role highlights SOX9's therapeutic potential in immune disorders, where inhibitors might restore T-cell tolerance without compromising adaptive immunity. In neurological contexts, SOX9 hinders post-stroke recovery by upregulating proteoglycans (CSPGs), which form inhibitory glial scars that impede axonal regeneration. Conditional SOX9 in murine models reduces CSPG deposition in the peri-infarct zone, leading to enhanced tissue sparing, , and improved motor function compared to wild-type controls. This suggests that SOX9-driven proteoglycan barriers limit reparative processes after ischemic injury, with conferring neuroprotective benefits by facilitating synaptic reconnection. Recent investigations have expanded SOX9's relevance to acquired fibro-inflammatory diseases, emphasizing targeted modulation for disease amelioration.

Experimental Models and Therapeutics

Knockout and Transgenic Animal Models

Homozygous global of Sox9 in mice results in perinatal lethality characterized by the complete absence of formation throughout the skeleton, as Sox9 is essential for initiating chondrogenesis from mesenchymal precursors. Sox9 null mice exhibit full gonadal , developing ovaries instead of testes due to failure in differentiation and maintenance of male gonadal identity. These mutants also display defects, including open s and truncated forebrains, along with cardiac looping abnormalities, underscoring Sox9's broad roles in . Conditional knockout models have revealed tissue-specific functions of Sox9. In limb bud , targeted inactivation using Prx1-Cre leads to severe appendicular skeletal defects, including profoundly shortened limbs and from disrupted mesenchymal condensations and impaired differentiation. In , conditional ablation with Col2a1-Cre prevents proper maturation and hypertrophy, blocking and resulting in with hypoplastic long bones. -specific knockout in the testes causes rapid testicular regression and partial toward ovarian structures, as Sox9 sustains function and represses ovarian pathways postnatally. Transgenic overexpression of Sox9 demonstrates its dose-dependent effects on development. Constitutive or conditional Sox9 transgenics in mesenchymal tissues enhance chondrogenesis by upregulating cartilage matrix genes like Col2a1, promoting ectopic cartilage formation in non-skeletal sites. However, in cranial neural crest-derived , elevated Sox9 levels accelerate suture fusion, inducing through premature osteogenic differentiation. In gonadal models, Sox9 overexpression in XX mice, as seen in the Odsex insertional mutant, drives testis differentiation and produces phenotypic XX males with sterile testes. Recent studies using models have provided insights into Sox9's roles in contexts. In a 2025 mouse model of colon cancer combining Apc inactivation with Sox9 loss, tumor progression accelerates with increased invasion and due to enhanced epithelial-mesenchymal transition and properties. For ischemic stroke, conditional Sox9 ablation in glial cells reduces chondroitin sulfate proteoglycan (CSPG) deposition in the , leading to improved neurological recovery in behavioral assays compared to controls, via enhanced axonal sprouting and tissue sparing. Humanized mouse models have recapitulated patient-specific variants. CRISPR/Cas9 knock-in of transactivation middle (TAM) domain variants in Sox9, such as Asp272del, generates mice with axial skeletal dysplasia including and anomalies, mirroring mild human phenotypes and highlighting dosage in vertebral development.

Therapeutic Targeting and Recent Advances

Therapeutic strategies targeting SOX9 have emerged as promising approaches for modulating its activity in various diseases, particularly cancers where it drives stemness and resistance, as well as in regenerative contexts like skeletal disorders. In , inhibiting SOX9 has shown potential to overcome resistance; for instance, CRISPR-mediated knockdown of SOX9 in high-grade serous cells reverts a stem-like transcriptional state and enhances to platinum-based drugs like , reducing tumor cell survival in preclinical models. Similarly, targeting the SOX9-USP28 axis with the specific USP28 inhibitor AZ1 destabilizes SOX9 protein levels, thereby impairing DNA damage repair and increasing cell susceptibility to such as . These findings highlight SOX9's role in therapy resistance and position upstream modulators like USP28 as viable intervention points. In pediatric tumors, a novel utilizing a "" viral vector has been developed to selectively target SOX9-high cells, which are often therapy-resistant. This approach employs an engineered (AAV) vector that recognizes SOX9 expression via a specific , delivering a cytotoxic payload that induces tumor while sparing normal ; preclinical testing in models demonstrated significant reduction in tumor burden without systemic toxicity. For , where SOX9 typically acts as a tumor suppressor by inhibiting epithelial-mesenchymal and stemness, peptide mimics of SOX9's functional domains have been shown to restore its activity, thereby suppressing tumor growth and in cell lines and xenografts. Conversely, in contexts like , super-enhancer inhibitors targeting SOX9-driven transcriptional programs have reprogrammed oncogenic pathways, suggesting broader applicability of epigenetic modulators to disrupt SOX9-dependent tumor progression. Activation of SOX9 holds therapeutic promise beyond cancer, particularly in metabolic and skeletal disorders. In metabolic dysfunction-associated (MASH), SOX9 overexpression activates the AMPK pathway, reducing hepatic lipid accumulation, inflammation, and fibrosis in mouse models; while direct agonists are under exploration, alleviating disease severity. For skeletal dysplasias caused by SOX9 , such as , engineered SOX9 variants with enhanced potential have been developed to promote differentiation, offering a foundation for future approaches to restore skeletal development. In immune-related conditions, SOX9's dual role as a regulator of immune cell infiltration and positions it as a target for autoimmune diseases. A 2025 review outlines how SOX9 modulates T-cell responses and production in disorders like and systemic sclerosis, proposing small-molecule modulators to dampen its pro-fibrotic effects while preserving anti-inflammatory functions. Despite these advances, challenges persist, including SOX9's context-dependent functions—tumor-suppressive in some cancers but oncogenic in others—and dose-dependent toxicity in activation strategies, necessitating precise delivery systems. As of 2025, no SOX9-specific inhibitors have entered clinical trials for , though preclinical data support their evaluation in phase I studies targeting endocrine-resistant subtypes.

References

  1. [1]
    SOX9 - an overview | ScienceDirect Topics
    SOX9 (locus: 17q24.3) encodes a transcription factor essential for normal testicular differentiation as well as the development of the skeleton and other ...
  2. [2]
    The versatile functions of Sox9 in development, stem cells, and ...
    This review provides a summary of the various roles of Sox9 in cell fate specification, stem cell biology, and related human diseases.
  3. [3]
  4. [4]
    6662 - Gene ResultSOX9 SRY-box transcription factor 9 [ (human)]
    Sep 27, 2025 · It acts during chondrocyte differentiation and, with steroidogenic factor 1, regulates transcription of the anti-Muellerian hormone (AMH) gene.Missing: chromosome size
  5. [5]
    Entry - *608160 - SRY-BOX 9; SOX9 - OMIM
    The genomic arrangement of SOX9 is such that the 5-prime end is oriented toward the centromere of chromosome 17 and closest to the breakpoint. It is possible ...Missing: size | Show results with:size
  6. [6]
    SOX9 has distinct regulatory roles in alternative splicing and ...
    Jun 13, 2018 · Since its discovery 30 years ago, SOX9 has been described as a key player during embryogenesis, especially in the maintenance of the progenitor ...
  7. [7]
    Comparative analysis demonstrates cell type-specific conservation ...
    Aug 29, 2019 · Among vertebrates, SOX9's functions in chondrogenesis are well conserved, while they vary in sex determination. To investigate the conservation ...
  8. [8]
    Diverse Regulation but Conserved Function: SOX9 in Vertebrate ...
    This article explores the cellular, morphological, and genetic mechanisms initiated by SOX9 for male gonad differentiation.
  9. [9]
    SOX9 - Transcription factor SOX-9 - Homo sapiens (Human) - UniProt
    SOX9 is a transcription factor key for chondrocyte differentiation and skeletal development, binding to DNA and promoting cartilage gene expression.Missing: span | Show results with:span<|separator|>
  10. [10]
    SOX9 in organogenesis: shared and unique transcriptional functions
    The SOX9 upstream region prone to chromosomal aberrations causing campomelic dysplasia contains multiple cartilage enhancers. Nucleic Acids Res. 2015;43(11): ...
  11. [11]
    Dimerization and Transactivation Domains as Candidates for ...
    Sox9 plays an important role in a large variety of developmental pathways in vertebrates. It is composed of three domains: high-mobility group box (HMG box) ...
  12. [12]
  13. [13]
    Crystal structure of a POU/HMG/DNA ternary complex suggests ...
    The HMG domain severely bends the DNA toward the major groove with an approximate bend angle of 90°. Side chains from residues of helix 1 and helix 2 of the ...
  14. [14]
    TGF-β regulates phosphorylation and stabilization of Sox9 protein in ...
    Dec 8, 2016 · Sox9, a key transcriptional regulator of chondrogenesis, is required for TGF-β-mediated regulation of specific cartilage genes. TGF-β can signal ...
  15. [15]
    Influence of Sox protein SUMOylation on neural development and ...
    Here, we review the mechanisms of three Sox proteins in neuronal development and disease, along with their transcriptional changes under SUMOylation.
  16. [16]
    Recurrent, truncating SOX9 mutations are associated with SOX9 ...
    As the majority of SOX9 mutations in CRC are truncating and result in deletion of the C-terminal protein including the transactivation domain, the protein ...
  17. [17]
    Two Sox9 messenger RNA isoforms: isolation of cDNAs and their ...
    Jan 28, 2000 · In this study, we report the isolation of two isoforms of Sox9 cDNAs and their expressions during gonadal development in the frog Rana rugosa.
  18. [18]
    The molecular and cellular basis of gonadal sex reversal in mice ...
    Feb 28, 2012 · It seems likely that SRY acts to regulate one transcriptional target: Sox9. This positive regulation is a requirement for Sertoli cell ...
  19. [19]
    The PGD2 pathway, independently of FGF9, amplifies SOX9 activity ...
    To define the in vivo genetic relationships between Sry, Sox9 and L-Pgds, we used real-time RT-PCR and in situ hybridization to analyze the expression of L-Pgds ...Missing: retinoic Wnt
  20. [20]
    Interactions between Sox9 and β-catenin control chondrocyte ...
    Our results strongly suggest that chondrogenesis is controlled by interactions between Sox9 and the Wnt/β-catenin signaling pathway. Keywords: Chondrocyte ...
  21. [21]
    Wilms tumor protein-dependent transcription of VEGF receptor 2 and ...
    Repression of Sox9 by WT1 in XX gonads depends on KDR signaling. A, gonadal explant cultures (XX and XY gonads, 12.5 dpc) were treated with Wt1 vivo-morpholino, ...
  22. [22]
    Wnt Signaling in Ovarian Development Inhibits Sf1 Activation of ...
    The decisive effect of the Wnt/β-catenin action that occurs in the ovary is therefore to repress Sox9 expression, allowing female development to proceed.
  23. [23]
    The transcription factor Sox9 is degraded by the ubiquitin ... - PubMed
    Our in vitro studies indicate that the ubiquitin-proteasome proteolytic system degrades Sox9 and regulates its transcriptional activity.Missing: TRIM26 | Show results with:TRIM26
  24. [24]
    Regulation of chondrogenesis by protein kinase C: Emerging new ...
    The authors suggested that by downregulating PKC, phorbol esters could have altered the phosphorylation state and activity of the Sox9 transcription factor, ...
  25. [25]
    Up-regulation of the chondrogenic Sox9 gene by fibroblast ... - PNAS
    Signals from FGF receptors lead to the activation of MEK, which in turn phosphorylates and activates MAPK/ERK. MAPK then phosphorylates and modulates the ...
  26. [26]
    Precise modulation of transcription factor levels identifies features ...
    Apr 6, 2023 · Of all SOX9-dependent genes, 76% have ED50 < 30 (buffered), 12% have ED50 between 30 and 40 (moderately sensitive), and 12% have ED50 > 40 ( ...
  27. [27]
  28. [28]
    SRY-Box transcription factor 9 triggers YAP nuclear entry via direct ...
    Apr 24, 2024 · In this study, we have uncovered a crucial role of SOX9 in the activation of YAP. SOX9 promotes the nuclear translocation of YAP by direct interaction.Missing: PKC | Show results with:PKC
  29. [29]
    Two independent nuclear localization signals are present ... - PubMed
    The HMG domain is a DNA-binding and DNA-bending motif comprising about 80 amino acid residues. It has been shown that SRY and SOX9 are nuclear proteins. Using ...
  30. [30]
  31. [31]
  32. [32]
    A nuclear export signal within the high mobility group domain ...
    These results indicate that SOX9 nuclear export signal is essential for SOX9 sex-specific subcellular localization and could be part of a regulatory switch.Missing: stress | Show results with:stress
  33. [33]
    Prostaglandin D2 induces nuclear import of the sex‐determining ...
    We find that activation of cAMP‐dependent protein kinase A (PKA) induces phosphorylation of SOX9 on its two S64 and S181 PKA sites, and its nuclear localization ...
  34. [34]
  35. [35]
  36. [36]
    Sex determination and the control of Sox9 expression in mammals
    Jan 31, 2011 · Sox9 is the master regulator of Sertoli cell differentiation, crucial for testis development. Its expression is upregulated in XY gonads and ...
  37. [37]
    Fgf9 and Wnt4 Act as Antagonistic Signals to Regulate Mammalian ...
    In the mouse XY gonad, Sry normally initiates a feed-forward loop between Sox9 and Fgf9, which up-regulates Fgf9 and represses Wnt4 to establish the testis ...Missing: PGD2 | Show results with:PGD2
  38. [38]
    The PGD2 pathway, independently of FGF9, amplifies SOX9 activity ...
    In this study, we demonstrated genetically that the L-Pgds/PGD2 pathway acts as a second amplification loop of Sox9 expression.Missing: mutual | Show results with:mutual
  39. [39]
    Gonadal Identity in the Absence of Pro-Testis Factor SOX9 and Pro ...
    These studies implicate an antagonism between the pro-testis SOX9 and pro-ovary RSPO1/WNT4/β-catenin pathways in shaping the fate of the bipotential gonad. The ...
  40. [40]
    Testicular differentiation in 46,XX DSD: an overview of genetic causes
    Overexpression of SOX9, often caused by gene duplications or copy number variations in the upstream promoter region, has been linked to testis determination in ...
  41. [41]
    Sox9 expression during gonadal development implies a conserved ...
    Our results imply that Sox9 plays an essential role in sex determination, possibly immediately downstream of Sry in mammals.
  42. [42]
    A novel SOX9 mutation, 972delC, causes 46,XY sex-reversed ...
    In 65% to 75% of the XY patients, male-to-female sex-reversal occurs, typically with streak gonads. Campomelic dysplasia is generally lethal in the neonatal ...
  43. [43]
    Testis cord differentiation after the sex determination stage is ...
    Mar 15, 2009 · AMH causes degeneration of the Müllerian ducts in males, and the Amh gene has been shown to be a downstream target of SOX9 and SOX8 (de Santa ...
  44. [44]
    The Sex-Determining Factors SRY and SOX9 Regulate Similar ...
    Aug 7, 2014 · SRY and SOX9 bind to the promoters of many common targets involved in testis differentiation and regulate their expression in Sertoli cells.Missing: PGD2 | Show results with:PGD2
  45. [45]
    Direct interaction of SRY-related protein SOX9 and steroidogenic ...
    In this study, we demonstrate that the canonical SOX-binding site within the human AMH proximal promoter can bind the transcription factor SOX9.Missing: coactivation | Show results with:coactivation
  46. [46]
    Zebrafish Trap230/Med12 is required as a coactivator for Sox9 ...
    Mediator is a coactivator complex transducing the interaction of DNA-binding transcription factors with RNA polymerase II, and our results reveal a critical ...
  47. [47]
    The pioneer factor SOX9 competes for epigenetic factors to switch ...
    Jul 24, 2023 · Here we tackle this question with SOX9, a master regulator that diverts embryonic epidermal stem cells (EpdSCs) into becoming hair follicle stem cells.
  48. [48]
    Genome-wide identification of FOXL2 binding and characterization ...
    Comparison of FOXL2 genome occupancy in the fetal ovary with SOX9 and DMRT1 ... FOXL2 could compete with SOX9 for chromatin accessibility and therefore repress ...<|separator|>
  49. [49]
    SOX9: a novel janus-faced regulator in immunity and its promise as ...
    Its ability to suppress anti-tumor immunity through multiple, synergistic pathways, makes it a central node in the network of cancer immune resistance (Table 1) ...
  50. [50]
    In mammalian foetal testes, SOX9 regulates expression of its target ...
    May 2, 2017 · In mammalian foetal testes, SOX9 regulates expression of its target genes by binding to genomic regions with conserved signatures.
  51. [51]
    Extracellular matrix-inducing Sox9 promotes both basal progenitor ...
    Mar 19, 2020 · Transcription factor Sox9 has an important role in neocortex expansion, where its expression in basal progenitors increases proliferation, ...
  52. [52]
    SOX9 Regulates Multiple Genes in Chondrocytes, Including Genes ...
    Possible direct targets of SOX9 suggested by RNA-Seq and Chip-Seq results. We next examined which genes identified as differentially regulated by SOX9 in RNA ...
  53. [53]
    Context-specific role of SOX9 in NF-Y mediated gene regulation in ...
    Jun 3, 2015 · Pathway analysis of ChIP-Seq-identified SOX9 target genes. SOX9 target genes identified based on ChIP-seq were classified into two groups ...
  54. [54]
    SOX9 haploinsufficiency reveals SOX9-Noggin interaction in BMP ...
    Mar 2, 2025 · In summary, SOX9 acts as the downstream of various signaling pathways and promotes chondrogenic gene transcriptions, orchestrating the skeletal ...Missing: threshold | Show results with:threshold
  55. [55]
    Sox9 Inhibits Wnt Signaling by Promoting β-Catenin ... - NIH
    Chondrocyte fate determination and maintenance are regulated by both intrinsic and extrinsic factors such as Sox9 and Wnt/β-catenin signaling, respectively.Missing: retinoic | Show results with:retinoic
  56. [56]
    The transcription factor Sox9 has essential roles in successive steps ...
    No evidence for patterning defects in limb buds of Sox9flox/flox; Prx1–Cre mutant embryos. To determine whether patterning defects occurred in Sox9-deficient ...
  57. [57]
    SOX9 drives a stem-like transcriptional state and platinum ...
    Oct 1, 2025 · The transcription factor SOX9 is sufficient to induce a stem-like transcriptional state and significant resistance to platinum treatment in high ...Missing: Nanog | Show results with:Nanog
  58. [58]
    Campomelic Dysplasia - GeneReviews® - NCBI Bookshelf
    Jul 31, 2008 · Campomelic dysplasia (CD) is an autosomal dominant disorder typically caused by a de novo SOX9 pathogenic variant. Rarely, CD is the result of a ...Summary · Diagnosis · Clinical Characteristics · Genetic Counseling
  59. [59]
    Mutations in SOX9, the Gene Responsible for Campomelic ... - NIH
    Five different mutations have been identified in six CD patients: two missense mutations in the SOX9 putative DNA binding domain (high mobility group, or HMG, ...
  60. [60]
    Dominant‐negative SOX9 mutations in campomelic dysplasia - NIH
    The data identify a novel molecular mechanism of disease in CD in which the truncated protein leads to a distinct and more significant effect on SOX9 function.
  61. [61]
    Acampomelic campomelic dysplasia with SOX9 mutation - PubMed
    Acampomelic campomelic dysplasia is a rare clinical variant of the more commonly encountered campomelic dysplasia (CMD1), characterized by absence of long ...
  62. [62]
    Acampomelic campomelic dysplasia with SOX9 mutation - Nature
    Jan 1, 2000 · Mutations in the SOX9 gene are reported in affected children with CMPD. Acampomelic CMPD is a clinical variant of the more commonly encountered CMPD.
  63. [63]
    Acampomelic Form of Campomelic Dysplasia with SOX9 Missense ...
    Apr 7, 2013 · Campomelic dysplasia is a skeletal dysplasia which results from a SOX9 gene mutation. The authors report a newborn with the acampomelic variant ...
  64. [64]
    Loss of Extreme Long-Range Enhancers in Human Neural Crest ...
    Sep 28, 2020 · Non-coding mutations at the far end of a large gene desert surrounding the SOX9 gene result in a human craniofacial disorder called Pierre Robin ...
  65. [65]
    Integration of 3D genome topology and local chromatin features ...
    Nov 23, 2022 · Disrupting a noncoding region distal from the human SOX9 gene causes the Pierre Robin sequence (PRS) characterized by the undersized lower jaw.
  66. [66]
    2 Genetics of 46,XY gonadal dysgenesis - ScienceDirect.com
    Approximately 15% of all cases of 46,XY complete gonadal dysgenesis result from mutations involving the SRY gene. ... Mutations in other genes including SOX9, ...
  67. [67]
    Failure of SOX9 Regulation in 46XY Disorders of Sex Development ...
    Mar 11, 2011 · Significantly, almost all 46,XY female patients with SRY mutations show complete gonadal dysgenesis [2], [3], consistent with the function of ...
  68. [68]
    Variants in the SOX9 transactivation middle domain induce axial ...
    Jan 24, 2025 · SOX9 protein contains an SRY-related high-mobility-group-box (HMG-box) DNA-binding domain, which is conserved in all SOX family proteins (5, 6).
  69. [69]
    Phenotypes Linked to Duplication Upstream of SOX9: New Insights ...
    Duplications occurring upstream of the SOX9 gene have been identified in a limited subset of patients with 46,XX testicular/ovotesticular differences/disorders ...
  70. [70]
    SOX9: a key transcriptional regulator in organ fibrosis - Frontiers
    SOX9 may be the key regulators in fibrosis through different signaling pathways which has shown in Figure 1. This study offers a summary of the latest ...
  71. [71]
    SOX9 Overexpression Ameliorates Metabolic Dysfunction ...
    Dec 20, 2024 · Our findings demonstrate that SOX9 overexpression alleviates hepatic lipid accumulation in MASH by activating the AMPK pathway.
  72. [72]
    Sox9 knockout mice have improved recovery following stroke
    Feb 6, 2018 · We demonstrate that conditional Sox9 ablation leads to reduced CSPG levels, increased tissue sparing and improved post-stroke neurological recovery.Missing: 2024 | Show results with:2024
  73. [73]
    USP28 promotes PARP inhibitor resistance by enhancing SOX9 ...
    Apr 16, 2025 · Our study demonstrated that elevated expression of SRY-box 9 (SOX9) contributes to olaparib resistance in ovarian cancer.
  74. [74]
    Pancreatic endocrine and exocrine signaling and crosstalk ... - Nature
    Feb 14, 2025 · SOX9 plays a crucial role in pancreatic duct development and is currently recognized as one of the markers for the ductal lineage. It is ...
  75. [75]
    SOX Genes and Their Role in Disorders of Sex Development
    Jun 27, 2022 · Copy number variation of two separate regulatory regions upstream of SOX9 causes isolated 46,XY or 46,XX disorder of sex development . J Med ...
  76. [76]
    Sox9 and Sox8 protect the adult testis from male-to-female genetic ...
    Here we show that after ablation of Sox9 in Sertoli cells of adult, fertile Sox8 -/- mice, testis-to-ovary genetic reprogramming occurs.
  77. [77]
    Generation of transgenic mice for conditional overexpression of Sox9
    This Sox9 conditional transgenic mouse line will be a valuable tool to uncover tissue-specific and developmental stage-specific functions of Sox9.Missing: knockout brachydactyly
  78. [78]
    Analysis of the Fgfr2C342Y mouse model shows condensation ...
    Summary: Mutation of FGFR2 causes a misregulation of Sox9, leading to disrupted mesenchymal condensation, and thus skeletal and craniofacial birth defects in ...
  79. [79]
    A transgenic insertion upstream of sox9 is associated with dominant ...
    We report here a new dominant insertional mutation, Odsex (Ods), in which XX mice carrying a 150-kb deletion (approximately 1 Mb upstream of Sox9) develop as ...Missing: chondrogenesis craniosynostosis
  80. [80]
    SOX9 suppresses colon cancer via inhibiting epithelial ... - JCI
    Apr 3, 2025 · Our results indicate SOX9 has tumor suppressor function in CRC; its loss may promote progression, invasion, and poor prognosis by enhancing EMT and stem cell ...Missing: accelerated | Show results with:accelerated
  81. [81]
    Sox9 knockout mice have improved recovery following stroke
    We demonstrate that conditional Sox9 ablation leads to reduced CSPG levels, increased tissue sparing and improved post-stroke neurological recovery.Missing: benefits | Show results with:benefits
  82. [82]
  83. [83]
    A Short SOX9 Peptide Mimics SOX9 Tumor Suppressor Activity and ...
    A Short SOX9 Peptide Mimics SOX9 Tumor Suppressor Activity and Is Sufficient to Inhibit Colon Cancer Cell Growth Available. Philippe Blache Corresponding ...
  84. [84]
    Super‐Enhancer Reprograming Driven by SOX9 and TCF7L2 ...
    Nov 4, 2024 · Super-enhancer reprograming driven by SOX9 and TCF7L2 represents transcription-targeted therapeutic vulnerability for treating gallbladder cancer.
  85. [85]
    Engineering Critical Residues of SOX9 Discovers a Variant With ...
    Dec 14, 2023 · Our results show that the novel SOX9 variant may be useful for efficient induction of chondrocytes and illuminate the strategic feasibility of ...
  86. [86]
    SOX9: an important factor in regulating breast cancer
    Aug 18, 2025 · SOX9 controls tumor growth, regulates tumor initiation, proliferation, immune evasion, and treatment resistance in breast cancer.