Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 686
Filter
1.
BMC Biol ; 22(1): 222, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354506

ABSTRACT

BACKGROUND: Abnormalities of in utero testis development are strongly associated with reproductive health conditions, including male infertility and testis cancer. In mouse testes, SOX9 and FGF9 support Sertoli cell development, while VEGF signalling is essential for the establishment of vasculature. The mitogen-activated protein kinase (MAPK) pathway is a major signalling cascade, essential for cell proliferation, differentiation and activation of Sry during primary sex-determination, but little is known about its function during fetal testis morphogenesis. We explored potential functions of MAPK signalling immediately after the establishment of testis cords in embryonic day (E)12.5 Oct4-eGFP transgenic mouse testes cultured using a MEK1/2 inhibitor. RESULTS: RNA sequencing in isolated gonadal somatic cells identified 116 and 114 differentially expressed genes after 24 and 72 h of MEK1/2 inhibition, respectively. Ingenuity Pathway Analysis revealed an association of MEK1/2 signalling with biological functions such as angiogenesis, vasculogenesis and cell migration. This included a failure to upregulate the master transcriptional regulators of vascular development, Sox7 and Sox17, VEGF receptor genes, the cell adhesion factor gene Cd31 and a range of other endothelial cell markers such as Cdh5 (encoding VE-cadherin) and gap junction genes Gja4 and Gja5. In contrast, only a small number of Sertoli cell enriched genes were affected. Immunofluorescent analyses of control testes revealed that the MEK1/2 downstream target, ERK1/2 was phosphorylated in endothelial cells and Sertoli cells. Inhibition of MEK1/2 eliminated pERK1/2 in fetal testes, and CD31, VE-cadherin, SOX7 and SOX17 and endothelial cells were lost. Consistent with a role for VEGF in driving endothelial cell development in the testis, inhibition of VEGFR also abrogated pERK1/2 and SOX7 and SOX17 expressing endothelial cells. Moreover, while Sertoli cell proliferation and localisation to the testis cord basement membrane was disrupted by inhibition of MEK1/2, it was unaffected by VEGFR inhibition. Instead, inhibition of FGF signalling compromised Sertoli cell proliferation and localisation to the testis cord basement membrane. CONCLUSIONS: Together, our data highlight an essential role for VEGF-dependent MEK1/2 signalling in promoting vasculature and indicate that FGF signalling through MEK1/2 regulates Sertoli cell organisation in the developing mouse testis.


Subject(s)
Mice, Transgenic , SOXF Transcription Factors , Testis , Animals , Male , SOXF Transcription Factors/metabolism , SOXF Transcription Factors/genetics , Mice , Testis/metabolism , Testis/embryology , Testis/blood supply , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/metabolism , MAP Kinase Kinase 2/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Signal Transduction , MAP Kinase Signaling System , Neovascularization, Physiologic , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 1/genetics , Angiogenesis , HMGB Proteins
3.
Mol Biomed ; 5(1): 36, 2024 09 04.
Article in English | MEDLINE | ID: mdl-39227479

ABSTRACT

Bladder cancer (BCa) stands out as a highly prevalent malignant tumor affecting the urinary system. The Sex determining region Y-box protein family is recognized for its crucial role in BCa progression. However, the effect of Sex determining region Y-box 7 (SOX7) on BCa progression has not been fully elucidated. Herein, RNA-sequencing, western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF) and tissue microarray were utilized to assess SOX7 expression in vitro and in vivo. Additionally, SOX7 expression, prognosis, and SOX7 + cytoglobin (CYGB) score were analyzed using R software. In vitro and vivo experiments were performed with BCa cell lines to validate the effect of SOX7 knockdown and overexpression on the malignant progression of BCa. The results showed that SOX7 exhibits low expression in BCa. It functions in diverse capacities, inhibiting the proliferative, migratory, and invasive capabilities of BCa. In addition, the experimental database demonstrated that SOX7 binds to the promoter of DNA methyltransferase 3 beta (DNMT3B), leading to the transcriptional inhibition of DNMT3B. This subsequently results in a reduced methylation of CYGB promoter, ultimately inhibiting the tumor progression of BCa. SOX7 + CYGB scores were significantly linked to patient prognosis. In conclusion, SOX7 inhibits the malignant progression of BCa via the DNMT3B/CYGB axis. Additionally, the SOX7 + CYGB score is capable of predicting the prognostic outcomes of BCa patients. Therefore, SOX7 and CYGB may play an important role in the progression of bladder cancer, and they can be used as prognostic markers of bladder cancer patients.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA Methyltransferase 3B , Disease Progression , Gene Expression Regulation, Neoplastic , Urinary Bladder Neoplasms , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Mice, Nude , Prognosis , Promoter Regions, Genetic/genetics , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Cytoglobin/genetics , Cytoglobin/metabolism
4.
Histopathology ; 85(5): 820-825, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39245863

ABSTRACT

AIMS: Recently, SOX17 has emerged as a promising biomarker for non-mucinous Müllerian (ovarian and endometrial) carcinomas, demonstrating increased specificity in comparison to PAX8 while maintaining similar sensitivity. However, expression of SOX17 in mesonephric-like adenocarcinoma (MLA), a carcinoma of the female genital tract with uncertain, but probably Müllerian histogenesis, remains unexplored. This study aims to address this gap. METHODS AND RESULTS: SOX17 immunohistochemistry was performed on whole tissue sections from 68 MLAs originating from the endometrium or ovary and seven cervical mesonephric carcinomas, as well as six mesonephric remnants/hyperplasias. Using a four-tiered scoring system based on distribution and intensity of staining, 68% of MLA displayed a negative/low (< 10%) SOX17 expression pattern, which contrasts with the high expression observed in most Müllerian carcinomas. However, 22% of MLA demonstrated high SOX17 expression, similar to other endometrial and ovarian carcinomas. Similarly, five of seven (72%) mesonephric carcinomas of the cervix were SOX17-negative, but two cases (28%) were positive. All mesonephric remnants/hyperplasias were SOX17 negative. CONCLUSIONS: The majority of MLA are negative or exhibit low SOX17 expression, in contrast to the diffuse and strong expression commonly seen in other types of Müllerian carcinoma. However, a subset of MLAs demonstrate high SOX17 expression. Therefore, absence of SOX17 staining is supportive for MLA when the differential includes another non-mucinous Müllerian carcinoma. SOX17 may also be useful for differentiating mesonephric remnants/hyperplasias from Müllerian malignancies and benign Müllerian glandular lesions.


Subject(s)
Adenocarcinoma , Biomarkers, Tumor , SOXF Transcription Factors , Humans , Female , SOXF Transcription Factors/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Middle Aged , Aged , Adult , Immunohistochemistry , Mesonephroma/pathology , Mesonephroma/diagnosis , Mesonephroma/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/diagnosis , Endometrial Neoplasms/pathology , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/metabolism , Aged, 80 and over
5.
FASEB J ; 38(15): e23495, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39126242

ABSTRACT

Hepatic stellate cell (HSC) activation is the essential pathological process of liver fibrosis (LF). The molecular mechanisms regulating HSC activation and LF are incompletely understood. Here, we explored the effect of transcription factor SRY-related high mobility group box 7 (SOX7) on HSC activation and LF, and the underlying molecular mechanism. We found the expression levels of SOX7 were decreased in human and mouse fibrotic livers, particularly at the fibrotic foci. SOX7 was also downregulated in primary activated HSCs and TGF-ß1 stimulated LX-2 cells. SOX7 knockdown promoted activation and proliferation of LX-2 cells while inhibiting their apoptosis. On the other hand, overexpression of SOX7 suppressed the activation and proliferation of HSCs. Mechanistically, SOX7 attenuates HSC activation and LF by decreasing the expression of ß-catenin and phosphorylation of Smad2 and Smad3 induced by TGF-ß1. Furthermore, overexpression of SOX7 using AAV8-SOX7 mouse models ameliorated the extent of LF in response to CCl4 treatment in vivo. Collectively, SOX7 suppressed HSC activation and LF. Targeting SOX7, therefore, could be a potential novel strategy to protect against LF.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , SOXF Transcription Factors , Hepatic Stellate Cells/metabolism , Animals , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Humans , Male , SOXF Transcription Factors/metabolism , SOXF Transcription Factors/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cell Proliferation , Mice, Inbred C57BL , beta Catenin/metabolism , beta Catenin/genetics , Apoptosis , Smad2 Protein/metabolism , Smad2 Protein/genetics , Cell Line , Smad3 Protein/metabolism , Smad3 Protein/genetics
7.
J Cancer Res Clin Oncol ; 150(7): 345, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38981872

ABSTRACT

BACKGROUND: Endometrial cancer (EC) is the sixth most frequent cancer in women worldwide and has higher fatality rates. The pathophysiology of EC is complex, and there are currently no reliable methods for diagnosing and treating the condition. Long non-coding RNA (lncRNA), according to mounting evidence, is vital to the pathophysiology of EC. HOTAIR is regarded as a significant prognostic indicator of EC. ZBTB7A decreased EC proliferation and migration, according to recent studies, however the underlying mechanism still needs to be clarified. METHODS: The research utilized RT-qPCR to measure HOTAIR expression in clinical EC tissues and various EC cell lines. Kaplan-Meier survival analysis was employed to correlate HOTAIR levels with patient prognosis. Additionally, the study examined the interaction between ZBTB7A and HOTAIR using bioinformatics tools and ChIP assays. The experimental approach also involved manipulating the expression levels of HOTAIR and ZBTB7A in EC cell lines and assessing the impact on various cellular processes and gene expression. RESULTS: The study found significantly higher levels of HOTAIR in EC tissues compared to adjacent normal tissues, with high HOTAIR expression correlating with poorer survival rates and advanced cancer characteristics. EC cell lines like HEC-1 A and KLE showed higher HOTAIR levels compared to normal cells. Knockdown of HOTAIR in these cell lines reduced proliferation, angiogenesis, and migration. ZBTB7A was found to be inversely correlated with HOTAIR, and its overexpression led to a decrease in HOTAIR levels and a reduction in malignant cell behaviors. The study also uncovered that HOTAIR interacts with ELAVL1 to regulate SOX17, which in turn activates the Wnt/ß-catenin pathway, promoting malignant behaviors in EC cells. CONCLUSION: HOTAIR is a critical regulator in EC, contributing to tumor growth and poor prognosis. Its interaction with ZBTB7A and regulation of SOX17 via the Wnt/ß-catenin pathway underlines its potential as a therapeutic target.


Subject(s)
Cell Proliferation , ELAV-Like Protein 1 , Endometrial Neoplasms , RNA, Long Noncoding , SOXF Transcription Factors , Humans , RNA, Long Noncoding/genetics , Female , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , ELAV-Like Protein 1/metabolism , ELAV-Like Protein 1/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Prognosis , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Animals , Mice , Middle Aged , Wnt Signaling Pathway/genetics , Angiogenesis
8.
Nat Commun ; 15(1): 4170, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755186

ABSTRACT

Endothelial cells are a heterogeneous population with various organ-specific and conserved functions that are critical to organ development, function, and regeneration. Here we report a Sox17-Erg direct reprogramming approach that uses cardiac fibroblasts to create differentiated endothelial cells that demonstrate endothelial-like molecular and physiological functions in vitro and in vivo. Injection of these induced endothelial cells into myocardial infarct sites after injury results in improved vascular perfusion of the scar region. Furthermore, we use genomic analyses to illustrate that Sox17-Erg reprogramming instructs cardiac fibroblasts toward an arterial-like identity. This results in a more efficient direct conversion of fibroblasts into endothelial-like cells when compared to traditional Etv2-based reprogramming. Overall, this Sox17-Erg direct reprogramming strategy offers a robust tool to generate endothelial cells both in vitro and in vivo, and has the potential to be used in repairing injured tissue.


Subject(s)
Cellular Reprogramming , Endothelial Cells , Fibroblasts , SOXF Transcription Factors , Transcriptional Regulator ERG , Animals , Mice , Cell Differentiation , Cellular Reprogramming/genetics , Endothelial Cells/metabolism , Endothelial Cells/cytology , Fibroblasts/metabolism , Fibroblasts/cytology , HMGB Proteins/metabolism , HMGB Proteins/genetics , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardium/cytology , Myocardium/metabolism , SOXF Transcription Factors/metabolism , SOXF Transcription Factors/genetics , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
9.
Trends Mol Med ; 30(7): 609-611, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38594095

ABSTRACT

In a recent report in Nature, Goto et al. reveal a novel immune-evasion mechanism adopted by early colorectal cancer (CRC) cells that is based on the transcription factor sex determining region Y (SRY)-box transcription factor 17 (SOX17). Leveraging colorectal adenoma and cancer models to perform comprehensive transcriptomic/chromatin analyses, this work shows that SOX17 generates immune-silent leucine-rich repeat-containing G protein-coupled receptor 5- (LGR5-) tumor cells, which suppress interferon gamma (IFNγ) signaling and promote immune escape.


Subject(s)
Colorectal Neoplasms , SOXF Transcription Factors , Humans , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , SOXF Transcription Factors/metabolism , SOXF Transcription Factors/genetics , Animals , Tumor Escape , Signal Transduction , Gene Expression Regulation, Neoplastic
10.
Shanghai Kou Qiang Yi Xue ; 33(1): 30-35, 2024 Feb.
Article in Chinese | MEDLINE | ID: mdl-38583021

ABSTRACT

PURPOSE: To explore the mechanism of SETDB1 inhibiting epithelial mesenchymal transition (EMT),migration and invasion in oral cancer via SOX 7 methylation. METHODS: SETDB1 and SOX7 mRNA and protein expression levels in KB cells of oral cancer and oral mucosal epithelial ATCC cells were determined by qRT-PCR and Western blot (WB). SETDB1 si-RNA was structured, then transfect into KB cells of oral cancer by liposome-mediated method. siRNA-SETDB1 was the experimental group (si-S), siRNA empty vector was the negative control group (si-N), and untransfected KB cells were the blank control group(NC). SETDB1 mRNA and protein expression levels were detected by qRT-PCR and Western blot(WB), to verify the transfection effect. The methylation levels of SOX7 were determined by pyrosequencing. The expression of N-cadherin, Vimentin, ß-catenin, and Slug proteins was detected by WB. Cell viability was measured by MTT assay, migration ability was tested by scratch healing assay, and invasion ability was tested by Transwell chamber assay. Statistical analysis was performed with SPSS 21.0 software package. RESULTS: The results of Rt-qPCR and WB showed that the SETDB1 mRNA and protein expression decreased significantly in si-S group(P<0.05). Pyrosequencing test results showed that the regulation of SETDB1 could significantly reduce the SOX7 methylation rate and increased the SOX7 protein expression. WB results showed that knockdown of SETDB1 significantly inhibited the expression of EMT-related proteins N-cadherin, Vimentin, ß-catenin and Slug in oral cancer KB cells (P<0.05). The results of cell functology experiments showed that knockdown of SETDB1 could significantly inhibit survival, migration and invasion of KB cells. CONCLUSIONS: Downregulation of SETDB1 could suppress EMT, migration and invasion of oral cancer cells by regulating SOX7 methylation level, providing new ideas and targets for the diagnosis and treatment of oral cancer.


Subject(s)
Mouth Neoplasms , SOXF Transcription Factors , beta Catenin , Humans , beta Catenin/genetics , beta Catenin/metabolism , Down-Regulation , Cell Line, Tumor , Vimentin/genetics , Vimentin/metabolism , Cadherins/genetics , Cadherins/metabolism , RNA, Small Interfering/metabolism , Mouth Neoplasms/genetics , Epithelial-Mesenchymal Transition , RNA, Messenger/metabolism , Methylation , Cell Movement/genetics , Cell Proliferation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism
11.
Cell Mol Life Sci ; 81(1): 137, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38478109

ABSTRACT

Improving the function of the blood-spinal cord barrier (BSCB) benefits the functional recovery of mice following spinal cord injury (SCI). The death of endothelial cells and disruption of the BSCB at the injury site contribute to secondary damage, and the ubiquitin-proteasome system is involved in regulating protein function. However, little is known about the regulation of deubiquitinated enzymes in endothelial cells and their effect on BSCB function after SCI. We observed that Sox17 is predominantly localized in endothelial cells and is significantly upregulated after SCI and in LPS-treated brain microvascular endothelial cells. In vitro Sox17 knockdown attenuated endothelial cell proliferation, migration, and tube formation, while in vivo Sox17 knockdown inhibited endothelial regeneration and barrier recovery, leading to poor functional recovery after SCI. Conversely, in vivo overexpression of Sox17 promoted angiogenesis and functional recovery after injury. Additionally, immunoprecipitation-mass spectrometry revealed the interaction between the deubiquitinase UCHL1 and Sox17, which stabilized Sox17 and influenced angiogenesis and BSCB repair following injury. By generating UCHL1 conditional knockout mice and conducting rescue experiments, we further validated that the deubiquitinase UCHL1 promotes angiogenesis and restoration of BSCB function after injury by stabilizing Sox17. Collectively, our findings present a novel therapeutic target for treating SCI by revealing a potential mechanism for endothelial cell regeneration and BSCB repair after SCI.


Subject(s)
Endothelial Cells , Spinal Cord Injuries , Animals , Mice , Rats , Angiogenesis , Blood-Brain Barrier/metabolism , Deubiquitinating Enzymes/metabolism , Endothelial Cells/metabolism , HMGB Proteins/metabolism , HMGB Proteins/pharmacology , Rats, Sprague-Dawley , Recovery of Function/physiology , SOXF Transcription Factors/genetics , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
12.
Cell Rep Methods ; 4(3): 100732, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38503291

ABSTRACT

An autologous source of vascular endothelial cells (ECs) is valuable for vascular regeneration and tissue engineering without the concern of immune rejection. The transcription factor ETS variant 2 (ETV2) has been shown to directly convert patient fibroblasts into vascular EC-like cells. However, reprogramming efficiency is low and there are limitations in EC functions, such as eNOS expression. In this study, we directly reprogram adult human dermal fibroblasts into reprogrammed ECs (rECs) by overexpressing SOX17 in conjunction with ETV2. We find several advantages to rEC generation using this approach, including improved reprogramming efficiency, increased enrichment of EC genes, formation of large blood vessels carrying blood from the host, and, most importantly, expression of eNOS in vivo. From these results, we present an improved method to reprogram adult fibroblasts into functional ECs and posit ideas for the future that could potentially further improve the reprogramming process.


Subject(s)
Endothelial Cells , Transcription Factors , Adult , Humans , Endothelial Cells/metabolism , Cells, Cultured , Tissue Engineering , Fibroblasts/metabolism , SOXF Transcription Factors/metabolism
13.
Stem Cell Reports ; 19(4): 579-595, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38518781

ABSTRACT

Transcription factors (TFs) are pivotal in guiding stem cell behavior, including their maintenance and differentiation. Using single-cell RNA sequencing, we investigated TFs expressed in endothelial progenitors (EPs) derived from human pluripotent stem cells (hPSCs) and identified upregulated expression of SOXF factors SOX7, SOX17, and SOX18 in the EP population. To test whether overexpression of these factors increases differentiation efficiency, we established inducible hPSC lines for each SOXF factor and found only SOX17 overexpression robustly increased the percentage of cells expressing CD34 and vascular endothelial cadherin (VEC). Conversely, SOX17 knockdown via CRISPR-Cas13d significantly compromised EP differentiation. Intriguingly, we discovered SOX17 overexpression alone was sufficient to generate CD34+VEC+CD31- cells, and, when combined with FGF2 treatment, more than 90% of CD34+VEC+CD31+ EP was produced. These cells are capable of further differentiating into endothelial cells. These findings underscore an undiscovered role of SOX17 in programming hPSCs toward an EP lineage, illuminating pivotal mechanisms in EP differentiation.


Subject(s)
Endothelial Cells , Fibroblast Growth Factor 2 , Pluripotent Stem Cells , SOXF Transcription Factors , Humans , Antigens, CD34/metabolism , Cell Differentiation/genetics , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Pluripotent Stem Cells/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
14.
Gastroenterology ; 167(2): 264-280, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38417530

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is characterized by an immune-suppressive microenvironment, which contributes to tumor progression, metastasis, and immunotherapy resistance. Identification of HCC-intrinsic factors regulating the immunosuppressive microenvironment is urgently needed. Here, we aimed to elucidate the role of SYR-Related High-Mobility Group Box 18 (SOX18) in inducing immunosuppression and to validate novel combination strategies for SOX18-mediated HCC progression and metastasis. METHODS: The role of SOX18 in HCC was investigated in orthotopic allografts and diethylinitrosamine/carbon tetrachloride-induced spontaneous models by using murine cell lines, adeno-associated virus 8, and hepatocyte-specific knockin and knockout mice. The immune cellular composition in the HCC microenvironment was evaluated by flow cytometry and immunofluorescence. RESULTS: SOX18 overexpression promoted the infiltration of tumor-associated macrophages (TAMs) and regulatory T cells (Tregs) while diminishing cytotoxic T cells to facilitate HCC progression and metastasis in cell-derived allografts and chemically induced HCC models. Mechanistically, transforming growth factor-beta 1 (TGF-ß1) upregulated SOX18 expression by activating the Smad2/3 complex. SOX18 transactivated chemokine (C-X-C motif) ligand 12 (CXCL12) and programmed death ligand 1 (PD-L1) to induce the immunosuppressive microenvironment. CXCL12 knockdown significantly attenuated SOX18-induced TAMs and Tregs accumulation and HCC dissemination. Antagonism of chemokine receptor 4 (CXCR4), the cognate receptor of CXCL12, or selective knockout of CXCR4 in TAMs or Tregs likewise abolished SOX18-mediated effects. TGFßR1 inhibitor Vactosertib or CXCR4 inhibitor AMD3100 in combination with anti-PD-L1 dramatically inhibited SOX18-mediated HCC progression and metastasis. CONCLUSIONS: SOX18 promoted the accumulation of immunosuppressive TAMs and Tregs in the microenvironment by transactivating CXCL12 and PD-L1. CXCR4 inhibitor or TGFßR1 inhibitor in synergy with anti-PD-L1 represented a promising combination strategy to suppress HCC progression and metastasis.


Subject(s)
B7-H1 Antigen , Benzylamines , Carcinoma, Hepatocellular , Chemokine CXCL12 , Cyclams , Disease Progression , Liver Neoplasms , Receptors, CXCR4 , SOXF Transcription Factors , T-Lymphocytes, Regulatory , Transforming Growth Factor beta1 , Tumor Microenvironment , Tumor-Associated Macrophages , Up-Regulation , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , SOXF Transcription Factors/metabolism , SOXF Transcription Factors/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Tumor Microenvironment/immunology , Humans , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Transforming Growth Factor beta1/metabolism , Mice , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Cyclams/pharmacology , Benzylamines/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Cell Line, Tumor , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Mice, Knockout , Gene Expression Regulation, Neoplastic , Signal Transduction , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice, Inbred C57BL , Diethylnitrosamine/toxicity , Male
15.
Nature ; 627(8004): 636-645, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38418875

ABSTRACT

A hallmark of cancer is the avoidance of immune destruction. This process has been primarily investigated in locally advanced or metastatic cancer1-3; however, much less is known about how pre-malignant or early invasive tumours evade immune detection. Here, to understand this process in early colorectal cancers (CRCs), we investigated how naive colon cancer organoids that were engineered in vitro to harbour Apc-null, KrasG12D and Trp53-null (AKP) mutations adapted to the in vivo native colonic environment. Comprehensive transcriptomic and chromatin analyses revealed that the endoderm-specifying transcription factor SOX17 became strongly upregulated in vivo. Notably, whereas SOX17 loss did not affect AKP organoid propagation in vitro, its loss markedly reduced the ability of AKP tumours to persist in vivo. The small fraction of SOX17-null tumours that grew displayed notable interferon-γ (IFNγ)-producing effector-like CD8+ T cell infiltrates in contrast to the immune-suppressive microenvironment in wild-type counterparts. Mechanistically, in both endogenous Apc-null pre-malignant adenomas and transplanted organoid-derived AKP CRCs, SOX17 suppresses the ability of tumour cells to sense and respond to IFNγ, preventing anti-tumour T cell responses. Finally, SOX17 engages a fetal intestinal programme that drives differentiation away from LGR5+ tumour cells to produce immune-evasive LGR5- tumour cells with lower expression of major histocompatibility complex class I (MHC-I). We propose that SOX17 is a transcription factor that is engaged during the early steps of colon cancer to orchestrate an immune-evasive programme that permits CRC initiation and progression.


Subject(s)
Adenoma , Colorectal Neoplasms , Immune Evasion , SOXF Transcription Factors , Animals , Humans , Mice , Adenoma/immunology , Adenoma/pathology , CD8-Positive T-Lymphocytes/immunology , Chromatin/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Gene Expression Profiling , Interferon-gamma/immunology , Organoids/immunology , Organoids/pathology , SOXF Transcription Factors/metabolism , Tumor Microenvironment/immunology , Mutation , Endoderm/metabolism , Disease Progression
16.
Crit Rev Immunol ; 44(3): 1-12, 2024.
Article in English | MEDLINE | ID: mdl-38421701

ABSTRACT

Endothelial dysfunction is associated with the progression of sepsis. This study sought to probe the molecular route of sex-determining region on the Y chromosome-box transcription factor 18 (SOX18) in sepsis-associated endothelial injury. Human umbilical vein endothelial cells (HUVECs) were treated with lipopolysaccharide (LPS) to establish the sepsis cell model. Cell viability, lactate dehydrogenase (LDH) release, oxidative stress (reactive oxygen species/malondialdehyde/superoxide dismutase), and inflammation (interleukin-1ß/tumor necrosis factor-α/interleukin-6) were evaluated by cell counting kit-8 assay and relevant assay kits. The expression levels of SOX18, microRNA (miR)-204-5p, and cadherin-2 (CDH2) in cells were determined by real-time quantitative polymerase chain reaction and Western blot assay. The interaction of SOX18, miR-204-5p, and CDH2 was analyzed by chromatin immunoprecipitation and dual-luciferase assay. LPS induced HUVECs injury and downregulation of SOX18. SOX18 overexpression increased cell viability, while decreased LDH activity, oxidative stress, and inflammation. SOX18 bound to the miR-204-5p promoter to promote miR-204-5p expression, and further repressed CDH2 expression. miR-204-5p knockdown and CDH2 overexpression abrogated the protective role of SOX18 in HUVECs injury. Overall, SOX18 alleviated LPS-induced injury of HUVECs by promoting miR-204-5p and repressing CDH2, suggesting it as a potential target for sepsis treatment.


Subject(s)
MicroRNAs , Sepsis , Humans , Human Umbilical Vein Endothelial Cells , Lipopolysaccharides , Inflammation , MicroRNAs/genetics , SOXF Transcription Factors/genetics
17.
Methods Mol Biol ; 2770: 87-97, 2024.
Article in English | MEDLINE | ID: mdl-38351448

ABSTRACT

Human primordial germ cell (PGC) development initiates about 2 weeks after fertilization during embryogenesis. Unique molecular events follow, including epigenetic resetting, to establish functional gametes (egg and sperm). Due to the inaccessibility of human embryos, it is essential to have an amenable experimental platform to investigate the mechanisms and potential dysfunctions of the events. We previously established a PGC-like cell (PGCLC) differentiation method using human pluripotent stem cells (PSCs) via induction of precursor cells followed by stimulation with a cytokine cocktail including BMP. We also revealed that the expression of PGC specifiers, SOX17 and PRDM1, can robustly induce PGCLCs from PSCs without the cytokines. The balance of SOX17 and PRDM1 is critical for germ cell fate since the two factors also regulate endoderm differentiation. Here we describe a detailed procedure for PGCLC differentiation with the balanced induction of SOX17 and PRDM1. The protocol can be used for PGC induction in other mammalian species exhibiting PGCs with SOX17 expression. Together, these studies will advance the understanding of germ cell biology and its applications in reproductive technology and medicine.


Subject(s)
Pluripotent Stem Cells , Semen , Animals , Humans , Male , Cell Differentiation/physiology , Germ Cells/metabolism , Embryo, Mammalian , Mammals , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism
18.
Gen Comp Endocrinol ; 350: 114472, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38373462

ABSTRACT

Heart development is a delicate and complex process regulated by coordination of various signaling pathways. In this study, we investigated the role of sox18 in heart development by modulating Wnt/ß-Catenin signaling pathways. Our spatiotemporal expression analysis revealed that sox18 is mainly expressed in the heart, branchial arch, pharyngeal arch, spinal cord, and intersegmental vessels at the tailbud stage of Xenopus tropicalis embryo. Overexpression of sox18 in the X. tropicalis embryos causes heart edema, while loss-of-function of sox18 can change the signal of developmental heart marker gata4 at different stages, suggesting that sox18 plays an essential role in the development of the heart. Knockdown of SOX18 in human umbilical vein endothelial cells suggests a link between Sox18 and ß-CATENIN, a key regulator of the Wnt signaling pathway. Sox18 negatively regulates islet1 and tbx3, the downstream factors of Wnt/ß-Catenin signaling, during the linear heart tube formation and the heart looping stage. Taken together, our findings highlight the crucial role of Sox18 in the development of the heart via inhibiting Wnt/ß-Catenin signaling.


Subject(s)
SOXF Transcription Factors , Xenopus Proteins , beta Catenin , Animals , Humans , beta Catenin/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Wnt Signaling Pathway , Xenopus/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
19.
Int J Mol Sci ; 24(23)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38068994

ABSTRACT

SRY-box transcription factor 18 (SOX18) is known to play a crucial role in the growth and development of hair follicles (HF) in both humans and mice. However, the specific effect of SOX18 on sheep hair follicles remains largely unknown. In our previous study, we observed that SOX18 was specifically expressed within dermal papilla cells (DPCs) in ovine hair follicles, leading us to investigate its potential role in the growth of hair follicles in sheep. In the present study, we aimed to examine the effect of SOX18 in DPCs and preliminarily study its regulatory mechanism through RNA-seq. We initially found that the overexpression of SOX18 promoted the proliferation of DPCs compared to the negative control group, while the interference of SOX18 had the opposite effect. To gain further insight into the regulatory mechanism of SOX18, we conducted RNA-seq analysis after knocking down SOX18 in Hu sheep DPCs. The result showed that the Wnt/ß-Catenin signaling pathway was involved in the growth process of DPC after SOX18 knockdown. Subsequently, we investigated the effect of SOX18 on the Wnt/ß-Catenin signaling pathway in DPCs using TOP/FOP-flash, qRT-PCR, and Western blot (WB) analysis. Our data demonstrated that SOX18 could activate the Wnt/ß-Catenin signaling pathway in DPCs. Additionally, we observed that SOX18 could rescue the proliferation of DPCs after inhibiting the Wnt/ß-Catenin signaling pathway. These findings underscore the essential role of SOX18 as a functional molecule governing the proliferation of DPCs. Additionally, these findings also greatly enhance our understanding of the role of SOX18 in the proliferation of DPCs and the growth of wool in Hu sheep.


Subject(s)
Hair Follicle , Sheep , Wnt Signaling Pathway , Animals , Cell Proliferation , Cells, Cultured , Hair Follicle/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL