Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-921971

ABSTRACT

OBJECTIVE@#To explore the genetic basis of three families with recurrence of non-immune hydrops fetalis (NIHF) but negative result by copy number variation sequencing (CNV-seq).@*METHODS@#Amniotic fluid sample and/or abortive tissues of the fetuses were collected and subjected to CNV-seq analysis. Peripheral blood samples of the parents were also taken for trio whole exome sequencing (trio WES).@*RESULTS@#Fetus 1 was found to harbor heterozygous c.976G>T(p.Glu326*) variant of the SOX18 gene in addition with compound heterozygous variants c.844C>T(p.Arg282Trp) and c.9472+1G>A of the RYR1 gene. The three variants were all inherited from its parents and have been associated with the etiology of NIHF. Based on the American College of Medical Genetics and Genomics (ACMG) standards and guidelines, the c.976G>T variant of SOX18 gene and c.9472+1G>A of RYR1 gene were predicted to be pathogenic (PVS1+PM2+PP3+PP4, PVS1+PM2+PP3), and c.844C>T variant of RYR1 gene to be likely pathogenic (PM1+PM2+PP3). Fetus 2 was found to harbor compound heterozygous variants c.6682C>T(p.Gln2228*) and c.4373_4383del(p.Val1458Alafs*63) of the PIEZO1 gene. Both variants were also inherited from its parents and are associated with the etiology of NIHF. Based on ACMG standards and guidelines, both c.6682C>T and c.4373_4383del variants of PIEZO1 gene were predicted to be pathogenic (PVS1+PM2+PP4, PVS1+PM2). Fetus 3 was found to harbor compound heterozygous variants of the TTN gene c.29860G>C(p.Asp9954His) and c.21107A>T(p.Asp7036Val), which were respectively inherited from its parents. Both variants have been strongly associated with the phenotype, though the connection between the etiology of NIHF and variants of the TTN gene remains elusive. Based on ACMG standards and guidelines, the c.29860G>C and c.21107A>T variants of TTN gene were predicted to be likely pathogenic (PM1+PM2+PP3).@*CONCLUSION@#Trio WES can improve the diagnosis rate of NIHF with a negative result by CNV-seq. Considering the urgency of prenatal diagnosis, CNV-seq and trio WES should be carried out at the same time for fetuses with NIHF.


Subject(s)
Female , Humans , Pregnancy , DNA Copy Number Variations , Genomics , Heterozygote , Hydrops Fetalis/genetics , Ion Channels , SOXF Transcription Factors , United States , Exome Sequencing
15.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-272472

ABSTRACT

The sex-determining region Y-box 7 (Sox7) is a important member of SOX family containing high mobi- lity group (HMG), mapped to human chromosome 8p23.1. Wnt/β-catenin signaling pathway plays an important role in cell survival, differentiation, self-renewal, proliferation and apoptosis, and is closely related with carcinogenesis. SOX7 gene is likely to be a tumor suppressor gene in MDS and other hematological malignancies. As a negative regulator of the WNT/β-catenin signaling pathway, the function loss of this gene can lead to carcinogenesis. The methylation of SOX7 gene leads to the silence of this gene, resulting in tumorigenesis. The decision of hematopoietic stem cells to self-renew or differentiate is a stochastic process, but SOX7 can promote the differentiation into all blood cell types. This review focuses on the role of SOX7 in hematopoietic system development and hematological malignancies.


Subject(s)
Humans , DNA Methylation , Gene Silencing , Hematologic Neoplasms , Genetics , Metabolism , Hematopoietic System , SOXF Transcription Factors , Genetics , Metabolism , Wnt Signaling Pathway
16.
Chinese Journal of Pathology ; (12): 546-550, 2014.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-304455

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the expression of Sox17 and β-catenin proteins in oligodendroglioma, and its clinical significance.</p><p><b>METHODS</b>One hundred cases of oligodendroglioma of different grades and 10 cases of surrounding benign tissue from First Affiliated Hospital of Xinjiang Medical University from 2003 to 2013 were assessed by immunohistochemistry for Sox17 and β-catenin protein expression. The clinicopathologic characteristics and outcome of patients with oligodendroglioma were evaluated by Kaplan-Meien and Cox regression analyses.</p><p><b>RESULTS</b>Sox17 was expressed in 10/10, 82% (41/50) and 62% (31/50) of normal control, oligodendroglioma and anaplastic oligodendroglioma, respectively. β-catenin was expressed in 2/10, 22% (11/50), and 52% (26/50) of normal control, oligodendroglioma and anaplastic oligodendroglioma, respectively. The differences of Sox17 and β-catenin expression between normal control and different types of oligodendroglioma were statistically significant. Univariate analysis showed that the expression of Sox17 protein (P = 0.000), β-catenin protein (P = 0.033), tumor position (P = 0.001), radiotherapy (P = 0.077), and chemotherapy (P = 0.000) were significant prognostic factors.</p><p><b>CONCLUSIONS</b>Oligodendrogliomas with expression of Sox17 protein, but not β-catenin, have better prognosis. Evaluation of Sox17 and β-catenin protein expression is important for accurate pathological diagnosis, prognostication and guiding treatment.</p>


Subject(s)
Humans , Brain Neoplasms , Metabolism , Neoplasm Proteins , Metabolism , Oligodendroglioma , Metabolism , Regression Analysis , SOXF Transcription Factors , Metabolism , beta Catenin , Metabolism
17.
Article in English | WPRIM (Western Pacific) | ID: wpr-211721

ABSTRACT

Recent evidence has suggested that human skin fibroblasts may represent a novel source of therapeutic stem cells. In this study, we report a 3-stage method to induce the differentiation of skin fibroblasts into insulin-producing cells (IPCs). In stage 1, we establish the isolation, expansion and characterization of mesenchymal stem cells from human labia minora dermis-derived fibroblasts (hLMDFs) (stage 1: MSC expansion). hLMDFs express the typical mesenchymal stem cell marker proteins and can differentiate into adipocytes, osteoblasts, chondrocytes or muscle cells. In stage 2, DMEM/F12 serum-free medium with ITS mix (insulin, transferrin, and selenite) is used to induce differentiation of hLMDFs into endoderm-like cells, as determined by the expression of the endoderm markers Sox17, Foxa2, and PDX1 (stage 2: mesenchymal-endoderm transition). In stage 3, cells in the mesenchymal-endoderm transition stage are treated with nicotinamide in order to further differentiate into self-assembled, 3-dimensional islet cell-like clusters that express multiple genes related to pancreatic beta-cell development and function (stage 3: IPC). We also found that the transplantation of IPCs can normalize blood glucose levels and rescue glucose homeostasis in streptozotocin-induced diabetic mice. These results indicate that hLMDFs have the capacity to differentiate into functionally competent IPCs and represent a potential cell-based treatment for diabetes mellitus.


Subject(s)
Animals , Female , Humans , Mice , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation/drug effects , Cell Separation , Cells, Cultured , Dermis/cytology , Diabetes Mellitus, Experimental/surgery , Fibroblasts/cytology , Genitalia, Female/cytology , Glucose/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Homeodomain Proteins/metabolism , Insulin/pharmacology , Insulin-Secreting Cells/cytology , Islets of Langerhans Transplantation , Mesenchymal Stem Cells/cytology , Mice, Nude , Niacinamide/pharmacology , Recovery of Function , SOXF Transcription Factors/metabolism , Sodium Selenite/pharmacology , Trans-Activators/metabolism , Transferrin/pharmacology
18.
Chinese Medical Journal ; (24): 3526-3531, 2012.
Article in English | WPRIM (Western Pacific) | ID: wpr-256701

ABSTRACT

<p><b>BACKGROUND</b>SRY-related HMG-box 17 (SOX17) encodes a member of the SOX (SRY-related HMG-box) family of transcription factors involved in the regulation of embryonic development and in the determination of the cell fate. Recently, it was considered as a tumor suppressor gene to inhibit canonical Wnt/β-catenin signaling pathway in several malignancies. However, the function of SOX17 in thyroid cancer was unknown. Therefore, we investigated the epigenetic changes and the function of SOX17 in thyroid cancer.</p><p><b>METHODS</b>The methylation status of the promoter region of SOX17 was detected using methylation-specific PCR in 63 papillary thyroid carcinoma (PTC) tissue, 10 normal thyroid tissue, and two thyroid cancer cell lines. Semi-quantitative RT-PCR was used to assess mRNA expression of SOX17 before and after 5-aza-2'-deoxycytidine treatment in thyroid cancer cell lines. Expression of SOX17 and β-catenin were detected by immunohistochemistry in PTC and adjacent tissue. Luciferase reporter assay, colony formation, transfection, and Western blotting were employed to analyze the effect of SOX17 on thyroid cancer cell proliferation and the function of SOX17 in the Wnt signal pathway.</p><p><b>RESULTS</b>Loss of SOX17 expression was correlated to the promoter region hypermethylation in thyroid cancer cell lines. Re-expression of SOX17 was found in TPC-1 cell line after 5-aza-2'-deoxycytidine treatment. In primary thyroid cancer, 60.3% (38/63) were methylated and 39.7% (25/63) unmethylated. But no methylation was found in noncancerous thyroid tissues. Methylation of SOX17 was associated reversely with β-catenin expression in the cytoplasm or nucleus significantly in the PTC (P < 0.05). Colony formation was inhibited by re-expression of SOX17 in TPC-1 cells. SOX17 suppressed the Wnt signaling pathway and the HMG domain was essential for this effect.</p><p><b>CONCLUSIONS</b>SOX17 was frequently methylated in human PTC. Loss of SOX17 expression was induced by promoter region hypermethylation. SOX17 inhibited thyroid cancer proliferation. Methylation of SOX17 activated the Wnt signaling pathway in human thyroid cancer.</p>


Subject(s)
Humans , Blotting, Western , Carcinoma , Genetics , Metabolism , Carcinoma, Papillary , Cell Line, Tumor , DNA Methylation , Genetics , Epigenesis, Genetic , Genetics , Physiology , Immunohistochemistry , Polymerase Chain Reaction , Promoter Regions, Genetic , Genetics , SOXF Transcription Factors , Genetics , Metabolism , Thyroid Neoplasms , Genetics , Metabolism , Tumor Cells, Cultured , Wnt Signaling Pathway , Genetics , Physiology , beta Catenin , Genetics , Metabolism
19.
Article in English | WPRIM (Western Pacific) | ID: wpr-81941

ABSTRACT

Sex-determining region Y box 18 (Sox18/SOX18) gene is an important regulator of vascular development playing a role in endothelial cell specification or differentiation, angiogenesis and atherogenesis. The aim of this study was to perform comprehensive functional characterization of the human SOX18 promoter, including determination of transcription start point (tsp) and identification of control elements involved in the regulation of SOX18 gene expression, with an emphasis on angiogenesis-related transcription factors. Analyses were performed in HeLa cells, representing a tumor cell line, and in EA.hy926 cells used as an endothelial model system. We have determined unique tsp of SOX18 gene, located 172 nucleotides upstream from ATG codon. Further, we have shown that SOX18 promoter region, -726 to -89 bp relative to tsp, contains positive cis-regulatory element(s) that stimulates SOX18 promoter activity, while region -89 to + 166 represents the minimal promoter. Within this region we have recognized the presence of essential element(s), positioned from -89 to +29, which harbors cluster of three putative early growth response 1 (EGR1) binding sites. By in vitro binding assays and functional analyses we have shown that these three putative binding sites are functionally relevant and sufficient for EGR1-induced SOX18 transcription. Mutations of these binding sites significantly impaired activity of the SOX18 promoter, particularly in EA.hy926 cells, indicating the importance of these regulatory elements for SOX18 promoter activity in endothelial setting. By data presented in this study, we have established SOX18 as a novel target gene regulated by EGR1 transcription factor, thus providing the first functional link between two transcription factors previously shown to be involved in the control of angiogenesis.


Subject(s)
Humans , Early Growth Response Protein 1/genetics , Electrophoretic Mobility Shift Assay , Endothelium/metabolism , Gene Expression Regulation , HeLa Cells , Mutagenesis, Site-Directed , Neovascularization, Physiologic/genetics , Promoter Regions, Genetic , Protein Binding/genetics , SOXF Transcription Factors/genetics , Transcription Initiation Site , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL