Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Gene Ther ; 27(3-4): 256-263, 2020 04.
Article in English | MEDLINE | ID: mdl-31543512

ABSTRACT

The present study discusses the expression and effect of the SOX17 gene in endometrioid adenocarcinoma. MTT assay is performed to determine the growth inhibition ratio of the DNA methyltransferase inhibitor 5-AZA for endometrial carcinoma cells, and the real-time fluorescence quantification PCR (qRT-PCR) was used to detect the mRNA expression of SOX17, ß-catenin, and CyclinD1 in endometrial carcinoma tissues before and after using 5-AZA to treat the endometrial carcinoma cell line. There were 30 cases on endometrioid adenocarcinoma tissues and 10 cases on normal endometrial tissues. The results revealed that the expression of SOX17 in endometrioid adenocarcinoma tissues was downregulated (P < 0.05), the expression of ß-catenin and CyclinD1 was upregulated (P < 0.05), and the expression of SOX17, CyclinD1, and ß-catenin was negatively correlated (r = -0.353, P > 0.05; R = -0.463, P < 0.05). The higher the histological grade and FIGO staging were, the lower the expression level of SOX17 was (P < 0.05). After HEC1A cells were treated by 5-AZA, the cell growth inhibition was most obvious (IC50 = 12.033) at 72 h, as determined by MTT assay. After cell treatment by 5-AZA, the genetic expression of SOX17 significantly increased, when compared with that before treatment (P < 0.05), while the genetic expression of ß-catenin and CyclinD1 significantly declined (P < 0.05). These results indicate that the expression level of SOX17 in endometrioid adenocarcinoma declined, and the upregulated expression level of SOX17 in cells inhibited the growth of tumor cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Endometrioid/genetics , Cyclin D1/genetics , Endometrial Neoplasms/genetics , SOXF Transcription Factors/genetics , beta Catenin/genetics , Adult , Aged , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/pharmacology , Azacitidine/therapeutic use , Carcinoma, Endometrioid/diagnosis , Carcinoma, Endometrioid/drug therapy , Carcinoma, Endometrioid/surgery , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclin D1/analysis , Down-Regulation/drug effects , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/surgery , Endometrium/drug effects , Endometrium/pathology , Endometrium/surgery , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hysterectomy , Immunohistochemistry , Middle Aged , Neoplasm Grading , SOXF Transcription Factors/analysis , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/analysis
2.
MAbs ; 10(4): 596-606, 2018.
Article in English | MEDLINE | ID: mdl-29648920

ABSTRACT

Antibodies are routinely used to study the activity of transcription factors, using various in vitro and in vivo approaches such as electrophoretic mobility shift assay, enzyme-linked immunosorbent assay, genome-wide method analysis coupled with next generation sequencing, or mass spectrometry. More recently, a new application for antibodies has emerged as crystallisation scaffolds for difficult to crystallise proteins, such as transcription factors. Only in a few rare cases, antibodies have been used to modulate the activity of transcription factors, and there is a real gap in our knowledge on how to efficiently design antibodies to interfere with transcription. The molecular function of transcription factors is underpinned by complex networks of protein-protein interaction and in theory, setting aside intra-cellular delivery challenges, developing antibody-based approaches to modulate transcription factor activity appears a viable option. Here, we demonstrate that antibodies or an antibody single-chain variable region fragments are powerful molecular tools to unravel complex protein-DNA and protein-protein binding mechanisms. In this study, we focus on the molecular mode of action of the transcription factor SOX18, a key modulator of endothelial cell fate during development, as well as an attractive target in certain pathophysiological conditions such as solid cancer metastasis. The engineered antibody we designed inhibits SOX18 transcriptional activity, by interfering specifically with an 8-amino-acid motif in the C-terminal region directly adjacent to α-Helix 3 of SOX18 HMG domain, thereby disrupting protein-protein interaction. This new approach establishes a framework to guide the study of transcription factors interactomes using antibodies as molecular handles.


Subject(s)
SOXF Transcription Factors/analysis , SOXF Transcription Factors/chemistry , Single-Chain Antibodies , Humans
3.
J Biol Chem ; 292(46): 19122-19132, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28935668

ABSTRACT

We report here an approach to redirecting somatic cell fate under chemically defined conditions without transcription factors. We start by converting mouse embryonic fibroblasts to epithelial-like cells with chemicals and growth factors. Subsequent cell fate mapping reveals a robust induction of SOX17 in the resulting epithelial-like cells that can be further reprogrammed to endodermal progenitor cells. Interestingly, these cells can self-renew in vitro and further differentiate into albumin-producing hepatocytes that can rescue mice from acute liver injury. Our results demonstrate a rational approach to convert mouse embryonic fibroblasts to hepatocytes and suggest that this mechanism-driven approach may be generalized for other cells.


Subject(s)
Cellular Reprogramming/drug effects , Endoderm/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Stem Cells/cytology , Animals , Cell Differentiation , Cell Self Renewal , Cells, Cultured , Female , HMGB Proteins/analysis , Hepatocytes/cytology , Mice , Mice, Inbred C57BL , SOXF Transcription Factors/analysis
4.
J Hepatol ; 67(1): 72-83, 2017 07.
Article in English | MEDLINE | ID: mdl-28237397

ABSTRACT

BACKGROUND & AIMS: Cholangiocarcinoma (CCA) is a biliary malignancy linked to genetic and epigenetic abnormalities, such as hypermethylation of SOX17 promoter. Here, the role of SOX17 in cholangiocyte differentiation and cholangiocarcinogenesis was studied. METHODS: SOX17 expression/function was evaluated along the differentiation of human induced pluripotent stem cells (iPSC) into cholangiocytes, in the dedifferentiation process of normal human cholangiocytes (NHC) in culture and in cholangiocarcinogenesis. Lentiviruses for SOX17 overexpression or knockdown were used. Gene expression and DNA methylation profiling were performed. RESULTS: SOX17 expression is induced in the last stage of cholangiocyte differentiation from iPSC and regulates the acquisition of biliary markers. SOX17 becomes downregulated in NHC undergoing dedifferentiation; experimental SOX17 knockdown in differentiated NHC downregulated biliary markers and promoted baseline and Wnt-dependent proliferation. SOX17 expression is lower in human CCA than in healthy tissue, which correlates with worse survival after tumor resection. In CCA cells, SOX17 overexpression decreased their tumorigenic capacity in murine xenograft models, which was related to increased oxidative stress and apoptosis. In contrast, SOX17 overexpression in NHC did not affect their survival but inhibited their baseline proliferation. In CCA cells, SOX17 inhibited migration, anchorage-independent growth and Wnt/ß-catenin-dependent proliferation, and restored the expression of biliary markers and primary cilium length. In human CCA, SOX17 promoter was found hypermethylated and its expression inversely correlates with the methylation grade. In NHC, Wnt3a decreased SOX17 expression in a DNMT-dependent manner, whereas in CCA, DNMT1 inhibition or silencing upregulated SOX17. CONCLUSIONS: SOX17 regulates the differentiation and maintenance of the biliary phenotype and functions as a tumor suppressor for CCA, being a potential prognostic marker and a promising therapeutic target. LAY SUMMARY: Understanding the molecular mechanisms involved in the pathogenesis of CCA is key in finding new valuable diagnostic and prognostic biomarkers, as well as therapeutic targets. This study provides evidence that SOX17 regulates the differentiation and maintenance of the biliary phenotype, and its downregulation promotes their tumorigenic transformation. SOX17 acts as a tumor suppressor in CCA and its genetic, molecular and/or pharmacological restoration may represent a new promising therapeutic strategy. Moreover, SOX17 expression correlates with the outcome of patients after tumor resection, being a potential prognostic biomarker.


Subject(s)
Bile Duct Neoplasms/etiology , Bile Ducts/pathology , Cholangiocarcinoma/etiology , SOXF Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Differentiation , Gene Expression Regulation, Neoplastic , Humans , Mice , SOXF Transcription Factors/analysis , SOXF Transcription Factors/genetics
5.
Methods Mol Biol ; 1357: 375-81, 2016.
Article in English | MEDLINE | ID: mdl-25417065

ABSTRACT

Here, we describe a procedure of human iPS cells differentiation into the definitive endoderm, further into albumin-expressing and albumin-secreting hepatocyte, using M15, a mesonephros- derived cell line. Approximately 90 % of human iPS cells differentiated into SOX17-positive definitive endoderm then approximately 50 % of cells became albumin-positive cells, and secreted ALB protein. This M15 feeder system for endoderm and hepatic differentiation is a simple and efficient method, and useful for elucidating molecular mechanisms for hepatic fate decision, and could represent an attractive approach for a surrogate cell source for pharmaceutical studies.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Endoderm/cytology , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Cell Line , Cell Lineage , Cells, Cultured , Culture Media/pharmacology , Humans , Mitomycin/pharmacology , SOXF Transcription Factors/analysis , Serum Albumin/biosynthesis
6.
Biotechnol Lett ; 37(8): 1711-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25851951

ABSTRACT

OBJECTIVES: As activin/nodal signaling plays a key role in definitive endoderm (DE) differentiation, we have explored activin A-induced differentiation of DE from human parthenogenetic embryonic stem cells (hPESCs). RESULTS: Administration of 5 ng activin A/ml had no effect on the expression of markers of DE differentiation. However, higher concentrations of activin A (50 and 100 ng/ml) upregulated Sox17 and Cxcr4, as well upregulating the mesendodermal precursor marker, Brachyury. CONCLUSIONS: These findings demonstrate that low dose activin A can maintain the undifferentiated potency of hPESCs, whereas higher doses induce DE differentiation; 50 ng/ml is the optimal concentration for inducing DE from hPESCs.


Subject(s)
Activins/metabolism , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Endoderm/growth & development , Biomarkers/analysis , Endoderm/chemistry , Fetal Proteins/analysis , Humans , Receptors, CXCR4/analysis , SOXF Transcription Factors/analysis , T-Box Domain Proteins/analysis
7.
Hum Pathol ; 46(3): 376-83, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25576290

ABSTRACT

Primary mediastinal seminomas are unusual tumors that can present in a pure form or as part of a mixed germ cell tumor. Contrary to testicular seminomas, little is known about the expression of novel immunohistochemical markers in mediastinal seminomas. This study investigates the immunohistochemical features of these tumors with a focus on novel markers. Thirty-two cases of primary mediastinal seminomas were reviewed; and representative whole-tissue sections were selected for immunohistochemical studies using antibodies directed against high molecular weight cytokeratin 5/6 (CK5/6), low molecular weight cytokeratin (CAM5.2), octamer-binding transcription factor 3/4 (OCT3/4), spalt-like transcription factor 4 (SALL4), GATA binding protein 3 (GATA-3), sry-related HMG box 2 (SOX2), SOX17, human T cell leukemia/lymphoma 1 (TCL1), glypican 3, melanoma associated antigen C2 (MAGEC2), and paired box gene 8 (Pax8). The percentage of positive tumor cells as well as the intensity of staining was evaluated and scored. Thirty-one cases (97%) expressed SOX17, whereas 29 cases (91%) were positive for OCT3/4 and SALL4, respectively. Twenty-eight cases (88%) expressed MAGEC2 and CAM5.2, respectively. Two cases (6%) were positive for Pax8, and a single case (3%) was positive for TCL1. None of the cases stained with CK5/6, GATA-3, SOX2, or glypican 3. Similar to testicular seminomas, mediastinal seminomas show consistent expression of OCT3/4, SALL4, SOX17, and MAGEC2 and are negative for SOX2, glypican 3, GATA-3, and CK5/6. Pax8 positivity is only inconsistently identified in mediastinal seminomas. Contrary to their testicular counterparts, mediastinal tumors show diffuse expression of low-molecular-weight cytokeratin in up to 90% of cases and are commonly negative for TCL1. Although there is some immunohistochemical overlap between testicular and mediastinal seminomas, considerable differences also exist and should be acknowledged when dealing with these tumors.


Subject(s)
Biomarkers, Tumor/analysis , Mediastinal Neoplasms/chemistry , Seminoma/chemistry , Testicular Neoplasms/chemistry , Transcription Factors/analysis , Adult , Antigens, Neoplasm/analysis , GATA3 Transcription Factor/analysis , Glypicans/analysis , Humans , Immunohistochemistry , Keratin-5/analysis , Keratin-6/analysis , Male , Mediastinal Neoplasms/pathology , Middle Aged , Neoplasm Proteins/analysis , Octamer Transcription Factor-3/analysis , PAX8 Transcription Factor , Paired Box Transcription Factors/analysis , Proto-Oncogene Proteins/analysis , SOXB1 Transcription Factors/analysis , SOXF Transcription Factors/analysis , Seminoma/pathology , Testicular Neoplasms/pathology , Young Adult
8.
Circulation ; 131(11): 995-1005, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25596186

ABSTRACT

BACKGROUND: Intracranial aneurysm (IA) is a common vascular disorder that frequently leads to fatal vascular rupture. Although various acquired risk factors associated with IA have been identified, the hereditary basis of IA remains poorly understood. As a result, genetically modified animals accurately modeling IA and related pathogenesis have been lacking, and subsequent drug development has been delayed. METHODS AND RESULTS: The transcription factor Sox17 is robustly expressed in endothelial cells of normal intracerebral arteries. The combination of Sox17 deficiency and angiotensin II infusion in mice induces vascular abnormalities closely resembling the cardinal features of IA such as luminal dilation, wall thinning, tortuosity, and subarachnoid hemorrhages. This combination impairs junctional assembly, cell-matrix adhesion, regeneration capacity, and paracrine secretion in endothelial cells of intracerebral arteries, highlighting key endothelial dysfunctions that lead to IA pathogenesis. Moreover, human IA samples showed reduced Sox17 expression and impaired endothelial integrity, further strengthening the applicability of this animal model to clinical settings. CONCLUSIONS: Our findings demonstrate that Sox17 deficiency in mouse can induce IA under hypertensive conditions, suggesting Sox17 deficiency as a potential genetic factor for IA formation. The Sox17-deficient mouse model provides a novel platform to develop therapeutics for incurable IA.


Subject(s)
Endothelium, Vascular/pathology , HMGB Proteins/deficiency , Intracranial Aneurysm/genetics , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/physiology , Adult , Aged , Angiotensin II/toxicity , Animals , Aorta/pathology , Cells, Cultured , Cerebral Arteries/chemistry , Cerebral Arteries/pathology , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dilatation, Pathologic/genetics , Dilatation, Pathologic/pathology , Disease Models, Animal , Endothelium, Vascular/metabolism , Female , HMGB Proteins/genetics , HMGB Proteins/physiology , Humans , Hypertension/complications , Intracranial Aneurysm/etiology , Intracranial Aneurysm/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/chemistry , Paracrine Communication , RNA Interference , SOXF Transcription Factors/analysis , SOXF Transcription Factors/genetics , Specific Pathogen-Free Organisms , Subarachnoid Hemorrhage/etiology , Transcription, Genetic , Up-Regulation , Veins/chemistry
9.
Anticancer Res ; 34(8): 4029-37, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25075026

ABSTRACT

BACKGROUND: SOX18 is a transcription factor known to be involved in blood and lymphatic vessel, hair follicle development, and wound healing processes. In addition, it has been reported that SOX18 may influence cancer growth. The role of SOX18 expression in ovarian cancer (OC) has not been determined. MATERIALS AND METHODS: SOX18 expression was assessed in 85 OC cases using immunohistochemical methods and in ovarian cancer cell lines on the mRNA and protein level. RESULTS: SOX18 was expressed in cancer cell nuclei as well as the cytoplasm. Higher nuclear SOX18 expression was associated with presence of residual disease following surgical treatment (p=0.0158) and advanced disease stage (p=0.0056). Univariate survival analysis revealed that high SOX18 (p=0.0125) expression, presence of residual disease (p<0.0001) and advanced disease stage (p<0.0324) predicted poor patient outcome. CONCLUSION: SOX18 may be a new predictive marker for OC.


Subject(s)
Ovarian Neoplasms/drug therapy , SOXF Transcription Factors/physiology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Platinum/therapeutic use , SOXF Transcription Factors/analysis , SOXF Transcription Factors/genetics
10.
Int J Cancer ; 131(1): 41-8, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-21796627

ABSTRACT

SOX group F genes are important regulators of angiogenesis and lymphangiogenesis. The aim of the present study was to examine the relationships between Sox group F expression and clinicopathological factors in gastric cancer. Three hundred and fifteen gastric cancer tissues and the corresponding normal gastric tissue were obtained from the tumor bank at the National Cancer Center, Korea. SOX group F mRNA levels in these tissues were evaluated by reverse transcriptase polymerase chain reaction (RT-PCR). The serum levels of SOX 18 proteins in 219 gastric cancer patients and in 30 healthy volunteers were also measured by enzyme-linked immunosorbent assay. Furthermore, immunohistochemistry (IHC) was performed on 679 gastric cancer tissues and the clinicopathological characteristics, as well as the survival rates of SOX 18 positive and negative gastric cancers were compared. RT-PCR showed that SOX group F mRNA was increased in the gastric cancer tissues compared to the normal gastric tissues (p < 0.001, respectively). The serum levels of SOX 18 protein were also increased in gastric cancer patients compared to healthy volunteers. IHC showed that of the 679 gastric cancer cases, 177 (26.1%) were positive for SOX 18 expression in their tumor stroma, and the frequencies of both lymphovascular invasion and lymph node metastases were higher in the SOX 18 positive than in the negative group. Both the 5-year survival and the recurrence-free survival were shorter for SOX 18 positive tumors (p = 0.023 and 0.012, respectively). SOX 18 expression might be a prognostic tumor marker and a potential therapeutic target in gastric cancer.


Subject(s)
SOXF Transcription Factors/analysis , SOXF Transcription Factors/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Disease Progression , Female , Humans , Korea , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXF Transcription Factors/blood , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate
11.
Cancer Sci ; 102(7): 1313-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21457403

ABSTRACT

CD133 is a universal marker of tissue stem/progenitor cells as well as cancer stem cells, but its physiological significance remains to be elucidated. Here we examined the relationship between expression of CD133 and features of gastric epithelial cells, and found that CD133-positive (CD133[+]) tumor cell lines formed well-differentiated tumors while CD133-negative (CD133[-]) lines formed poorly differentiated ones when subcutaneously injected into nude mice. We also found that CD133(+) and CD133(-) cell populations co-existed in some cell lines. FACS analysis showed that CD133(+) cells were mother cells because CD133(+) cells formed both CD133(+) and CD133(-) cells, but CD133(-) cells did not form CD133(+) cells. In these cell lines, CD133(+) cells formed well-differentiated tumors while CD133(-) cells formed poorly differentiated ones. In human gastric cancers, CD133 was exclusively expressed on the luminal surface membrane of gland-forming cells, and it was never found on poorly differentiated diffuse-type cells. Considering that poorly differentiated tumors often develop from well-differentiated tumors during tumor progression, these results suggest that loss of expression of CD133 might be related to gastric tumor progression. Microarray analysis showed that CD133(+) cells specifically expressed Sox17, a tumor suppressor in gastric carcinogenesis. Forced expression of SOX17 induced expression of CD133 in CD133(-) cells, and reduction of SOX17 caused by siRNA in CD133(+) cells induced a reduction in the level of CD133. These results indicate that Sox17 might be a key transcription factor controlling CD133 expression, and that it might also play a role in the control of gastric tumor progression.


Subject(s)
Antigens, CD/physiology , Glycoproteins/physiology , HMGB Proteins/physiology , Neoplastic Stem Cells/chemistry , Peptides/physiology , SOXF Transcription Factors/physiology , Stomach Neoplasms/pathology , AC133 Antigen , Animals , Antigens, CD/analysis , Female , Gastric Mucosa/pathology , Gene Expression Profiling , Glycoproteins/analysis , HMGB Proteins/analysis , Humans , Mice , Peptides/analysis , SOXF Transcription Factors/analysis , Stomach Neoplasms/chemistry
12.
Assay Drug Dev Technol ; 9(4): 403-19, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21395400

ABSTRACT

Embryonic stem (ES) cell technology may serve as a platform for the discovery of drugs to treat diseases such as diabetes. However, because of difficulties in establishing reliable ES cell differentiation methods and in creating cost-effective plating conditions for the high-throughput format, screening for molecules that regulate pancreatic beta cells and their immediate progenitors has been limited. A relatively simple and inexpensive differentiation protocol that allows efficient generation of insulin-expressing cells from murine ES cells was previously established in our laboratories. In this report, this system is characterized in greater detail to map developmental cell stages for future screening experiments. Our results show that sequential activation of multiple gene markers for undifferentiated ES cells, epiblast, definitive endoderm, foregut, and pancreatic lineages was found to follow the sequence of events that mimics pancreatic ontogeny. Cells that expressed enhanced green fluorescent protein, driven by pancreatic and duodenal homeobox 1 or insulin 1 promoter, correctly expressed known beta cell lineage markers. Overexpression of Sox17, an endoderm fate-determining transcription factor, at a very early stage of differentiation (days 2-3) enhanced pancreatic gene expression. Overexpression of neurogenin3, an endocrine progenitor cell marker, induced glucagon expression at stages when pancreatic and duodenal homeobox 1 message was present (days 10-16). Forced expression (between days 16 and 25) of MafA, a pancreatic maturation factor, resulted in enhanced expression of insulin genes, glucose transporter 2 and glucokinase, and glucose-responsive insulin secretion. Day 20 cells implanted in vivo resulted in pancreatic-like cells. Together, our differentiation assay recapitulates the proceedings and behaviors of pancreatic development and will be valuable for future screening of beta cell effectors.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Insulin-Secreting Cells/metabolism , Pancreas/growth & development , Animals , Cell Differentiation/drug effects , Cell Lineage/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Endoderm/cytology , Endoderm/metabolism , Fibroblast Growth Factor 10/biosynthesis , Fibroblast Growth Factor 10/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/analysis , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin/biosynthesis , Insulin/genetics , Insulin/metabolism , Male , Mice , Mice, SCID , Pancreas/cytology , Pancreas/metabolism , SOXF Transcription Factors/analysis , SOXF Transcription Factors/biosynthesis , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Trans-Activators/analysis , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/analysis , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Dev Biol ; 350(2): 393-404, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21146513

ABSTRACT

Cells of the primitive endoderm (PrE) and the pluripotent epiblast (EPI), the two lineages specified within the inner cell mass (ICM) of the mouse blastocyst stage embryo, are segregated into adjacent tissue layers by the end of the preimplantation period. The PrE layer which emerges as a polarized epithelium adjacent to the blastocoel, with a basement membrane separating it from the EPI, has two derivatives, the visceral and parietal endoderm. In this study we have investigated the localization of two transcriptional regulators of the SOX family, SOX17 and SOX7, within the PrE and its derivatives. We noted that SOX17 was first detected in a salt-and-pepper distribution within the ICM, subsequently becoming restricted to the nascent PrE epithelium. This dynamic distribution of SOX17 resembled the localization of GATA6 and GATA4, two other PrE lineage-specific transcription factors. By contrast, SOX7 was only detected in PrE cells positioned in contact with the blastocoel, raising the possibility that these cells are molecularly distinct. Our observations support a model of sequential GATA6 > SOX17 > GATA4 > SOX7 transcription factor activation within the PrE lineage, perhaps correlating with the consecutive periods of cell lineage 'naïvete', commitment and sorting. Furthermore our data suggest that co-expression of SOX17 and SOX7 within sorted PrE cells could account for the absence of a detectable phenotype of Sox17 mutant blastocysts. However, analysis of implantation-delayed blastocysts, revealed a role for SOX17 in the maintenance of PrE epithelial integrity, with the absence of SOX17 leading to premature delamination and migration of parietal endoderm.


Subject(s)
Blastocyst/physiology , Cell Differentiation , Cell Lineage , Endoderm/physiology , HMGB Proteins/physiology , SOXF Transcription Factors/physiology , Transcription Factors/physiology , Animals , Blastocyst/cytology , Cell Movement , Cell Polarity , Endoderm/cytology , Female , HMGB Proteins/analysis , Male , Mice , Mice, Inbred ICR , SOXF Transcription Factors/analysis , SOXF Transcription Factors/genetics
14.
J Pathol ; 218(2): 146-62, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19253916

ABSTRACT

Human germ cell tumours (GCTs) comprise several types of neoplasias with different pathogeneses and clinical behaviours. A classification into five subtypes has been proposed. Here, the so-called type II testicular GCTs (TGCTs), ie the seminomas and non-seminomas, will be reviewed with emphasis on pathogenesis and clinical implications. Various risk factors have been identified that define subpopulations of men who are amenable to early diagnosis. TGCTs are omnipotent, able to generate all differentiation lineages, both embryonic and extra-embryonic, as well as the germ cell lineage itself. The precursor lesion, composed of primordial germ cells/gonocytes, is referred to as carcinoma in situ of the testis (CIS) and gonadoblastoma of the dysgenetic gonad. These pre-malignant cells retain embryonic characteristics, which probably explains the unique responsiveness of the derived tumours to DNA-damaging agents. Development of CIS and gonadoblastoma is crucially dependent on the micro-environment created by Sertoli cells in the testis, and granulosa cells in the dysgenetic gonad. OCT3/4 has high sensitivity and specificity for CIS/gonadoblastoma, seminoma, and embryonal carcinoma, and is useful for the detection of CIS cells in semen, thus a promising tool for non-invasive screening. Overdiagnosis of CIS due to germ cell maturation delay can be avoided using immunohistochemical detection of stem cell factor (SCF). Immunohistochemistry is helpful in making the distinction between seminoma and embryonal carcinoma, especially SOX17 and SOX2. The different non-seminomatous histological elements can be recognized using various markers, such as AFP and hCG, while others need confirmation. The value of micro-satellite instability as well as BRAF mutations in predicting treatment resistance needs validation in prospective trials. The availability of representative cell lines, both for seminoma and for embryonal carcinoma, allows mechanistic studies into the initiation and progression of this disease.


Subject(s)
Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/pathology , Adult , Biomarkers, Tumor/analysis , Carcinoma in Situ/chemistry , Carcinoma in Situ/pathology , Gonadoblastoma/chemistry , Gonadoblastoma/pathology , Humans , Immunohistochemistry , Male , SOXB1 Transcription Factors/analysis , SOXF Transcription Factors/analysis , Stem Cell Factor/analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...