Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 237
Filter
1.
J Pathol ; 257(3): 367-378, 2022 07.
Article in English | MEDLINE | ID: mdl-35302657

ABSTRACT

Most high-grade ovarian carcinomas (HGOCs) are sensitive to carboplatin (CBP)-based chemotherapy but frequently recur within 24 months. Recurrent tumors remain CBP-sensitive and acquire resistance only after several treatment rounds. Recurrences arise from a small number of residual tumor cells not amenable to investigation in patients. We developed patient-derived xenografts (PDXs) that allow the study of these different stages of CBP-sensitive recurrence and acquisition of resistance. We generated PDX models from CBP-sensitive and intrinsically resistant HGOC. PDXs were CBP- or mock-treated and tumors were sampled, after treatment and at recurrence. We also isolated models with acquired-resistance from CBP-sensitive PDXs. Tumors were characterized at the histological and transcriptome levels. PDX models reproduced treatment response seen in the patients. CBP-sensitive residual tumors contained nonproliferating tumor cell clusters embedded in a fibrotic mesh. In nontreated PDX tumors and treated CBP-resistant tumors, fibrotic tissue was not prevalent. Residual tumors had marked differences in gene expression when compared to naïve and recurrent tumors, indicating downregulation of the cell cycle and proliferation and upregulation of interferon response and the epithelial-mesenchymal transition. This gene expression pattern resembled that described in embryonal diapause and 'drug-tolerant persister' states. Residual and acquired-resistance tumors share the overexpression of three genes: CEACAM6, CRYAB, and SOX2. Immunostaining analysis showed strong CEACAM6, CRYAB, and SOX2 protein expression in CBP-sensitive residual and acquired-resistance PDX, thus confirming the RNA profiling results. In HGOC PDX, CBP-sensitive recurrences arise from a small population of quiescent, drug-tolerant, residual cells embedded in a fibrotic mesh. These cells overexpress CEACAM6, CRYAB, and SOX2, whose overexpression is also associated with acquired resistance and poor patient prognosis. CEACAM6, CRYAB, and SOX2 may thus serve as a biomarker to predict recurrence and emergence of resistant disease in CBP-treated HGOC patients. © 2022 The Pathological Society of Great Britain and Ireland.


Subject(s)
Antigens, CD , Carcinoma, Ovarian Epithelial , Cell Adhesion Molecules , GPI-Linked Proteins , Ovarian Neoplasms , SOXB1 Transcription Factors , alpha-Crystallin B Chain , Antigens, CD/biosynthesis , Antigens, CD/genetics , Carboplatin/pharmacology , Carboplatin/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Drug Resistance, Neoplasm , Female , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Humans , Neoplasm Recurrence, Local , Neoplasm, Residual , Recurrence , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Xenograft Model Antitumor Assays , alpha-Crystallin B Chain/biosynthesis , alpha-Crystallin B Chain/genetics
2.
Dev Biol ; 466(1-2): 59-72, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32791054

ABSTRACT

Sharks and rays (elasmobranchs) have the remarkable capacity to continuously regenerate their teeth. The polyphyodont system is considered the ancestral condition of the gnathostome dentition. Despite this shared regenerative ability, sharks and rays exhibit dramatic interspecific variation in their tooth morphology. Ray (batoidea) teeth typically constitute crushing pads of flattened teeth, whereas shark teeth are pointed, multi-cuspid units. Although recent research has addressed the molecular development of the shark dentition, little is known about that of the ray. Furthermore, how dental diversity within the elasmobranch lineage is achieved remains unknown. Here, we examine dental development and regeneration in two Batoid species: the thornback skate (Raja clavata) and the little skate (Leucoraja erinacea). Using in situ hybridization and immunohistochemistry, we examine the expression of a core gnathostome dental gene set during early development of the skate dentition and compare it to development in the shark. Elasmobranch tooth development is highly conserved, with sox2 likely playing an important role in the initiation and regeneration of teeth. Alterations to conserved genes expressed in an enamel knot-like signalling centre may explain the morphological diversity of elasmobranch teeth, thereby enabling sharks and rays to occupy diverse dietary and ecological niches.


Subject(s)
Dentition , Regeneration , Skates, Fish/embryology , Animals , Fish Proteins/biosynthesis , Gene Expression Regulation, Developmental , SOXB1 Transcription Factors/biosynthesis , Species Specificity
3.
Development ; 145(1)2018 01 08.
Article in English | MEDLINE | ID: mdl-29180573

ABSTRACT

In mice, the incisors grow throughout the animal's life, and this continuous renewal is driven by dental epithelial and mesenchymal stem cells. Sox2 is a principal marker of the epithelial stem cells that reside in the mouse incisor stem cell niche, called the labial cervical loop, but relatively little is known about the role of the Sox2+ stem cell population. In this study, we show that conditional deletion of Sox2 in the embryonic incisor epithelium leads to growth defects and impairment of ameloblast lineage commitment. Deletion of Sox2 specifically in Sox2+ cells during incisor renewal revealed cellular plasticity that leads to the relatively rapid restoration of a Sox2-expressing cell population. Furthermore, we show that Lgr5-expressing cells are a subpopulation of dental Sox2+ cells that also arise from Sox2+ cells during tooth formation. Finally, we show that the embryonic and adult Sox2+ populations are regulated by distinct signalling pathways, which is reflected in their distinct transcriptomic signatures. Together, our findings demonstrate that a Sox2+ stem cell population can be regenerated from Sox2- cells, reinforcing its importance for incisor homeostasis.


Subject(s)
Ameloblasts/metabolism , Antigens, Differentiation/biosynthesis , Gene Expression Regulation, Developmental , Incisor/embryology , SOXB1 Transcription Factors/biosynthesis , Stem Cells/metabolism , Ameloblasts/cytology , Animals , Antigens, Differentiation/genetics , Incisor/cytology , Mice , Mice, Transgenic , SOXB1 Transcription Factors/genetics , Stem Cells/cytology
4.
Oncology ; 99(8): 528-538, 2021.
Article in English | MEDLINE | ID: mdl-34107469

ABSTRACT

BACKGROUND: Sex-determining region Y-box 2 (SOX2) is a transcriptional factor that drives embryonic stem cells to neuroendocrine cells in lung development and is highly expressed in small-cell lung cancer (SCLC). However, the prognostic role of SOX2 and its relationship with tumor-infiltrating lymphocytes (TILs) has not been determined in SCLC. Herein, we assessed the expression of SOX2 and CD8+ TILs to obtain insights into the prognostic role of SOX2 and CD8+ TILs in limited-stage (LS)-SCLC. METHODS: A total of 75 patients with LS-SCLC was enrolled. The SOX2 expression and CD8+ TILs were evaluated by immunohistochemistry. RESULTS: High SOX2 and CD8+ TIL levels were identified in 52 (69.3%) and 40 (53.3%) patients, respectively. High SOX2 expression was correlated with increased density of CD8+ TILs (p = 0.041). Unlike SOX2, high CD8+ TIL numbers were associated with significantly longer progression-free survival (PFS; 13.9 vs. 8.0 months, p = 0.014). Patients with both high SOX2 expression and CD8+ TIL numbers (n = 29, 38.7%) had significantly longer PFS and overall survival (OS) compared to those from the other groups (median PFS 19.3 vs. 8.4 months; p = 0.002 and median OS 35.7 vs. 17.4 months; p = 0.004, respectively). Multivariate Cox regression analysis showed that the combination of high SOX2 expression and CD8+ TIL levels was an independent good prognostic factor for OS (HR = 0.471, 95% CI, 0.250-0.887, p = 0.02) and PFS (HR = 0.447, 95% CI, 0.250-0.801, p = 0.007) in SCLC. CONCLUSIONS: Evaluation of the combination of SOX2 and CD8+ TIL levels may be of a prognostic value in LS-SCLC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , SOXB1 Transcription Factors/biosynthesis , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Prognosis , Survival Rate
5.
BMC Cancer ; 21(1): 332, 2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33789601

ABSTRACT

BACKGROUND: Small cell neuroendocrine carcinoma of the uterine cervix (SCNEC) is a rare cancer involving the human papilloma virus (HPV), and has few available treatments. The present work aimed to assess the feasibility of SOX2 and HPV statuses as predictive indicators of SCNEC prognosis. METHODS: The associations of SOX2 and/or high-risk (HR)-HPV RNA in situ hybridization (RISH) levels with clinicopathological characteristics and prognostic outcomes for 88 neuroendocrine carcinoma (NEC) cases were analyzed. RESULTS: Among these patients with SCNEC, SOX2, P16INK4A and HR-HPV RISH expression and SOX2/HR-HPV RISH co-expression were detected in 68(77.3%), 76(86.4%), 73(83.0%), and 48(54.5%), respectively. SOX2-positive and HR-HPV RISH-positive SCNEC cases were associated with poorer overall survival (OS, P = 0.0170, P = 0.0451) and disease-free survival (DFS, P = 0.0334, P = 0.0309) compared with those expressing low SOX2 and negative HR-HPV RISH. Alternatively, univariate analysis revealed that SOX2 and HR-HPV RISH expression, either separately or in combination, predicted the poor prognosis of SCNEC patients. Multivariate analysis revealed that the co-expression of SOX2 with HR-HPV RISH may be an independent factor of OS [hazard ratio = 3.597; 95% confidence interval (CI): 1.085-11.928; P = 0.036] and DFS [hazard ratio = 2.880; 95% CI: 1.199-6.919; P = 0.018] prediction in SCNEC. CONCLUSIONS: Overall, the results of the present study suggest that the co-expression of SOX2 with HR-HPV RISH in SCNEC may represent a specific subgroup exhibiting remarkably poorer prognostic outcomes compared with the expression of any one marker alone.


Subject(s)
Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/virology , Carcinoma, Small Cell/genetics , Papillomaviridae/isolation & purification , Papillomavirus Infections/metabolism , SOXB1 Transcription Factors/biosynthesis , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/virology , Adult , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Carcinoma, Small Cell/pathology , Female , Humans , In Situ Hybridization , Middle Aged , Neoplasm Staging , Nomograms , Papillomaviridae/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Prognosis , RNA, Viral/genetics , Retrospective Studies , SOXB1 Transcription Factors/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
6.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946876

ABSTRACT

The hair follicle dermal papilla is critical for hair generation and de novo regeneration. When cultured in vitro, dermal papilla cells from different species demonstrate two distinguishable growth patterns under the conventional culture condition: a self-aggregative three dimensional spheroidal (3D) cell pattern and a two dimensional (2D) monolayer cell pattern, correlating with different hair inducing properties. Whether the loss of self-aggregative behavior relates to species-specific differences or the improper culture condition remains unclear. Can the fixed 2D patterned dermal papilla cells recover the self-aggregative behavior and 3D pattern also remains undetected. Here, we successfully constructed the two growth patterns using sika deer (Cervus nippon) dermal papilla cells and proved it was the culture condition that determined the dermal papilla growth pattern. The two growth patterns could transit mutually as the culture condition was exchanged. The fixed 2D patterned sika deer dermal papilla cells could recover the self-aggregative behavior and transit back to 3D pattern, accompanied by the restoration of hair inducing capability when the culture condition was changed. In addition, the global gene expressions during the transition from 2D pattern to 3D pattern were compared to detect the potential regulating genes and pathways involved in the recovery of 3D pattern and hair inducing capability.


Subject(s)
Deer/anatomy & histology , Hair Follicle/cytology , AC133 Antigen/biosynthesis , AC133 Antigen/genetics , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/genetics , Animals , Biomarkers , Cell Aggregation , Cell Culture Techniques , Cell Division , Cells, Cultured , Deer/genetics , Gene Expression Regulation , Gene Ontology , Hair , Hair Follicle/growth & development , Hair Follicle/metabolism , Mesoderm/cytology , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Species Specificity , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Transcriptome , Versicans/biosynthesis , Versicans/genetics
7.
J Neurosci ; 39(7): 1320-1331, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30587544

ABSTRACT

Individuals with sleep apnea often exhibit changes in cognitive behaviors consistent with alterations in the hippocampus. It is hypothesized that adult neurogenesis in the dentate gyrus is an ongoing process that maintains normal hippocampal function in many mammalian species, including humans. However, the impact of chronic intermittent hypoxia (IH), a principal consequence of sleep apnea, on hippocampal adult neurogenesis remains unclear. Using a murine model, we examined the impact of 30 d of IH (IH30) on adult neurogenesis and synaptic plasticity in the dentate gyrus. Although IH30 did not affect paired-pulse facilitation, IH30 suppressed long-term potentiation (LTP). Immunohistochemical experiments also indicate that IH perturbs multiple aspects of adult neurogenesis. IH30 increased the number of proliferating Sox2+ neural progenitor cells in the subgranular zone yet reduced the number of doublecortin-positive neurons. Consistent with these findings, cell lineage tracing revealed that IH30 increased the proportion of radial glial cells in the subgranular zone, yet decreased the proportion of adult-born neurons in the dentate gyrus. While administration of a superoxide anion scavenger during IH did not prevent neural progenitor cell proliferation, it mitigated the IH-dependent suppression of LTP and prevented adult-born neuron loss. These data demonstrate that IH causes both reactive oxygen species-dependent and reactive oxygen species-independent effects on adult neurogenesis and synaptic plasticity in the dentate gyrus. Our findings identify cellular and neurophysiological changes in the hippocampus that may contribute to cognitive and behavioral deficits occurring in sleep apnea.SIGNIFICANCE STATEMENT Individuals with sleep apnea experience periods of intermittent hypoxia (IH) that can negatively impact many aspects of brain function. Neurons are continually generated throughout adulthood to support hippocampal physiology and behavior. This study demonstrates that IH exposure attenuates hippocampal long-term potentiation and reduces adult neurogenesis. Antioxidant treatment mitigates these effects indicating that oxidative signaling caused by IH is a significant factor that impairs synaptic plasticity and reduces adult neurogenesis in the hippocampus.


Subject(s)
Dentate Gyrus/pathology , Hypoxia, Brain/pathology , Neurogenesis , Neuronal Plasticity , Animals , Cell Lineage , Cell Proliferation , Doublecortin Domain Proteins , Excitatory Postsynaptic Potentials , Female , Free Radical Scavengers/pharmacology , Hypoxia, Brain/etiology , Long-Term Potentiation , Male , Mice , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/pathology , Neuroglia/pathology , Neuropeptides/metabolism , Reactive Nitrogen Species/metabolism , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Sleep Apnea Syndromes/complications , Sleep Apnea Syndromes/physiopathology
8.
Prostate ; 80(13): 1134-1144, 2020 09.
Article in English | MEDLINE | ID: mdl-32628304

ABSTRACT

BACKGROUND: Although androgen deprivation therapy (ADT) is the initial treatment strategy for prostate cancer (PCa), recurrent castration-resistant prostate cancer (CRPC) eventually ensues. In this study, cancer-derived immunoglobulin G (CIgG) is found to be induced after ADT, identifying CIgG as a potential CRPC driver gene. METHODS: The expression of CIgG and its clinical significance in PCa tissue was analyzed by The Cancer Genome Atlas database and immunohistochemistry. Subsequently, the sequence features of prostate cell line VHDJH rearrangements were analyzed. We also assessed the effect of CIgG on the migratory, invasive and proliferative abilities of PCa cells in vitro and vivo. Suspended microsphere, colony formation and drug-resistant assays were performed using PC3 cells with high CIgG expression (CIgGhigh ) and low CIgG expression (CIgG-/low ), and A nonobese diabetic/severe combined immunodeficiency mouse tumor xenograft model was developed for the study of the tumorigenic effects of the different cell populations. The SOX2-CIgG signaling pathway was validated by immunohistochemistry, immunofluorescence, quantitative reverse transcription-polymerase chain reaction, Western blot, luciferase, and chromatin immunoprecipitation assays and bioinformatics analyses. Finally, we investigated the effect of RP215 inhibition on the progression of PCa in vivo using a Babl/c nude mouse xenograft model. RESULTS: CIgG is frequently expressed in PCa and associated with clinicopathological characteristics, moreover, CIgG transcripts with unique patterns of VHDJH rearrangements are found in PCa cells. Functional analyses identified that CIgG was induced by ADT and upregulated by SOX2 (SRY (sex determining region Y)-box 2) in PCa, promoting the development of PCa. In addition, our findings underscore a novel role of CIgG signaling in the maintenance of stemness and the progression of cancer through mitogen activated protein kinase/extracellular-signal-regulated kinase and AKT in PCa. In vivo experiments further demonstrated that depleting CIgG significantly suppressed the growth of PCa cell xenografts. Furthermore, a CIgG monoclonal antibody named RP215 exhibits tumor inhibitory effect as well. CONCLUSION: Our data suggests that CIgG could be a driver of PCa development, and that targeting the SOX2-CIgG axis may therefore inhibit PCa development after ADT.


Subject(s)
Immunoglobulin G/immunology , Prostatic Neoplasms, Castration-Resistant/immunology , SOXB1 Transcription Factors/immunology , Animals , HEK293 Cells , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Signal Transduction/immunology , Tissue Array Analysis
9.
Development ; 144(24): 4522-4529, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29084802

ABSTRACT

Elongation of the body axis is a key aspect of body plan development. Bipotential neuromesoderm progenitors (NMPs) ensure axial growth of embryos by contributing both to the spinal cord and mesoderm. The current model for the mechanism controlling NMP deployment invokes Tbx6, a T-box factor, to drive mesoderm differentiation of NMPs. Here, we identify a new population of Tbx6+ cells in a subdomain of the NMP niche in mouse embryos. Based on co-expression of a progenitor marker, Sox2, we identify this population as representing a transient cell state in the mesoderm-fated NMP lineage. Genetic lineage tracing confirms the presence of the Tbx6+ NMP cell state. Furthermore, we report a novel aspect of the documented Tbx6 mutant phenotype, namely an increase from two to four ectopic neural tubes, corresponding to the switch in NMP niche, thus highlighting the importance of Tbx6 function in NMP fate decision. This study emphasizes the function of Tbx6 as a bistable switch that turns mesoderm fate 'on' and progenitor state 'off', and thus has implications for the molecular mechanism driving NMP fate choice.


Subject(s)
Embryonic Stem Cells/cytology , Mesoderm/cytology , Neural Tube/embryology , SOXB1 Transcription Factors/biosynthesis , Spinal Cord/embryology , Transcription Factors/biosynthesis , Animals , Body Patterning/genetics , Body Patterning/physiology , Cell Differentiation , Cell Lineage , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Neural Tube/cytology , SOXB1 Transcription Factors/genetics , T-Box Domain Proteins , Transcription Factors/genetics
10.
Respir Res ; 21(1): 302, 2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33208157

ABSTRACT

BACKGROUND: Characterized by abnormal lung growth or maturation, congenital diaphragmatic hernia (CDH) affects 1:3000 live births. Cellular studies report proximal (SOX2+) and distal (SOX9+) progenitor cells as key modulators of branching morphogenesis and epithelial differentiation, whereas transcriptome studies demonstrate ROBO/SLIT as potential therapeutic targets for diaphragm defect repair in CDH. In this study, we tested the hypothesis that (a) experimental-CDH could changes the expression profile of ROBO1, ROBO2, SOX2 and SOX9; and (b) ROBO1 or ROBO2 receptors are regulators of branching morphogenesis and SOX2/SOX9 balance. METHODS: The expression profile for receptors and epithelial progenitor markers were assessed by Western blot and immunohistochemistry in a nitrofen-induced CDH rat model. Immunohistochemistry signals by pulmonary structure were also quantified from embryonic-to-saccular stages in normal and hypoplastic lungs. Ex vivo lung explant cultures were harvested at E13.5, cultures during 4 days and treated with increasing doses of recombinant rat ROBO1 or human ROBO2 Fc Chimera proteins for ROBO1 and ROBO2 inhibition, respectively. The lung explants were analyzed morphometrically and ROBO1, ROBO2, SOX2, SOX9, BMP4, and ß-Catenin were quantified by Western blot. RESULTS: Experimental-CDH induces distinct expression profiles by pulmonary structure and developmental stage for both receptors (ROBO1 and ROBO2) and epithelial progenitor markers (SOX2 and SOX9) that provide evidence of the impairment of proximodistal patterning in experimental-CDH. Ex vivo functional studies showed unchanged branching morphogenesis after ROBO1 inhibition; increased fetal lung growth after ROBO2 inhibition in a mechanism-dependent on SOX2 depletion and overexpression of SOX9, non-phospho ß-Catenin, and BMP4. CONCLUSIONS: These studies provided evidence of receptors and epithelial progenitor cells which are severely affected by CDH-induction from embryonic-to-saccular stages and established the ROBO2 inhibition as promoter of branching morphogenesis through SOX2/SOX9 balance.


Subject(s)
Hernias, Diaphragmatic, Congenital/metabolism , Lung/embryology , Phenyl Ethers/toxicity , Receptors, Immunologic/biosynthesis , SOX9 Transcription Factor/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Animals , Female , Herbicides/toxicity , Hernias, Diaphragmatic, Congenital/chemically induced , Hernias, Diaphragmatic, Congenital/genetics , Lung/drug effects , Morphogenesis/drug effects , Morphogenesis/physiology , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/genetics , SOX9 Transcription Factor/genetics , SOXB1 Transcription Factors/genetics , Signal Transduction/drug effects , Signal Transduction/physiology
11.
J Microencapsul ; 37(4): 332-340, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32223347

ABSTRACT

Aim: In this study, we aimed to develop a polycationic non-viral carrier for the delivery of the reprogramming factors to the L929 fibroblast cell.Methods: We have prepared (3-hydroxybutyrate-co-3-hydroxyhexanoate) PHBHHx-based nanoparticles with the solvent diffusion method. Cytotoxicity of PXNs was determined via MTT assay. Transfection efficiency was evaluated via screening GFP expression by fluorescence microscopy. The expression of reprogramming factors (Oct4, Klf4, and Sox2) was determined by RT-qPCR.Results: PXNs with 32.9 ± 0.41 mV zeta potential and 177.6 ± 0.80 nm size were used for transfection of L929 Fbroblast cells. The percentage of cell viability of PXN were between 91.8%(±2.9) and 42.1%(±1.3). The transfection efficiency was found as 71.6%(±3,5). According to RT-qPCR data, the rate of transfection factors was significantly increased after the 11th cycle compared to non-transfected cells. Based on these results, it can be concluded that newly developed PXN is thought to be an effective tool for reprogramming cells.


Subject(s)
Caproates/chemistry , Nanoparticles/chemistry , Cellular Reprogramming , Gene Expression , Green Fluorescent Proteins , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Particle Size , Paxillin/genetics , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Transfection/methods
12.
Int J Mol Sci ; 21(14)2020 Jul 11.
Article in English | MEDLINE | ID: mdl-32664542

ABSTRACT

Stemness and reprogramming involve transcriptional master regulators that suppress cell differentiation while promoting self-renewal. A distinguished example thereof is SOX2, a high mobility group (HMG)-box transcription factor (TF), whose subcellular localization and turnover regulation in embryonic, induced-pluripotent, and cancer stem cells (ESCs, iPSCs, and CSCs, respectively) is mediated by the PI3K/AKT/SOX2 axis, a stem cell-specific branch of the PI3K/AKT signaling pathway. Further effector functions associated with PI3K/AKT induction include cell cycle progression, cellular (mass) growth, and the suppression of apoptosis. Apoptosis, however, is a central element of DNA damage response (DDR), where it provides a default mechanism for cell clearance when DNA integrity cannot be maintained. A key player in DDR is tumor suppressor p53, which accumulates upon DNA-damage and is counter-balanced by PI3K/AKT enforced turnover. Accordingly, stemness sustaining SOX2 expression and p53-dependent DDR mechanisms show molecular-functional overlap in PI3K/AKT signaling. This constellation proves challenging for stem cells whose genomic integrity is a functional imperative for normative ontogenesis. Unresolved mutations in stem and early progenitor cells may in fact provoke transformation and cancer development. Such mechanisms are also particularly relevant for iPSCs, where genetic changes imposed through somatic cell reprogramming may promote DNA damage. The current review aims to summarize the latest advances in the understanding of PI3K/AKT/SOX2-driven stemness and its intertwined relations to p53-signaling in DDR under conditions of pluripotency, reprogramming, and transformation.


Subject(s)
Cell Self Renewal/physiology , Cell Transformation, Neoplastic/genetics , Cellular Reprogramming/physiology , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , SOXB1 Transcription Factors/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Apoptosis , Cell Cycle/physiology , DNA Damage , DNA Repair , Humans , Induced Pluripotent Stem Cells/metabolism , Neoplasms/enzymology , Neoplastic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , SOXB1 Transcription Factors/biosynthesis , Tumor Suppressor Protein p53/biosynthesis
13.
Glia ; 67(6): 1062-1075, 2019 06.
Article in English | MEDLINE | ID: mdl-30648289

ABSTRACT

Chronic pain is one of the most prevalent chronic diseases in the world. The plastic changes of sensory neurons in dorsal root ganglia (DRG) have been extensively studied as the underlying periphery mechanism. Recent studies revealed that satellite cells, the major glial cells in DRG, also played important roles in the development/modulation of chronic pain. Whether DRG satellite glial cells generate new neurons as their counterparts in enteric nerve ganglia and carotid body do under pathological conditions remains poorly investigated. Here, we report that chronic pain induces proliferation and upregulation of progenitor markers in the sex-determining region Y-box 2 (Sox2)- and platelet-derived growth factor receptor alpha (PDGFRα)-positive satellite glial cells. BrdU incorporation assay revealed the generation of IB4- and CGRP-positive neurons, but not NF200-positive neurons in DRG ipsilateral to injury. Genetic fate tracings showed that PDGFRα-positive cells did not generate neurons, whereas Sox2-positive cells produced both IB4- and CGRP-positive neurons. Interestingly, glial fibrillary acidic protein-positive cells, a subpopulation of Sox2-positive satellites, only gave birth to IB4-positive neurons. Local persistent delivery of tetrodotoxin to the sciatic nerve trunk significantly reduced the pain-induced neurogenesis. Furthermore, patch-clamp studies demonstrated that these glia-derived new neurons could fire action potentials and respond to capsaicin. Taken together, our data demonstrated a chronic pain-induced nociceptive neurogenesis in DRG from Sox2-positive satellite cells, indicating a possible contribution of DRG neurogenesis to the pathology of chronic pain.


Subject(s)
Chronic Pain/metabolism , Ganglia, Spinal/metabolism , Neurogenesis/physiology , SOXB1 Transcription Factors/biosynthesis , Satellite Cells, Perineuronal/metabolism , Animals , Chronic Pain/pathology , Ganglia, Spinal/chemistry , Ganglia, Spinal/pathology , Male , Mice , Mice, Inbred C57BL , SOXB1 Transcription Factors/analysis , Satellite Cells, Perineuronal/chemistry , Satellite Cells, Perineuronal/pathology
14.
EMBO J ; 34(7): 896-910, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25527292

ABSTRACT

Major efforts are invested to characterize the factors controlling the proliferation of neural stem cells. During mammalian corticogenesis, our group has identified a small pool of genes that are transiently downregulated in the switch of neural stem cells to neurogenic division and reinduced in newborn neurons. Among these switch genes, we found Tox, a transcription factor with hitherto uncharacterized roles in the nervous system. Here, we investigated the role of Tox in corticogenesis by characterizing its expression at the tissue, cellular and temporal level. We found that Tox is regulated by calcineurin/Nfat signalling. Moreover, we combined DNA adenine methyltransferase identification (DamID) with deep sequencing to characterize the chromatin binding properties of Tox including its motif and downstream transcriptional targets including Sox2, Tbr2, Prox1 and other key factors. Finally, we manipulated Tox in the developing brain and validated its multiple roles in promoting neural stem cell proliferation and neurite outgrowth of newborn neurons. Our data provide a valuable resource to study the role of Tox in other tissues and highlight a novel key player in brain development.


Subject(s)
Calcineurin/metabolism , Cerebral Cortex/embryology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , NFATC Transcription Factors/metabolism , Signal Transduction/physiology , Animals , Calcineurin/genetics , Cell Proliferation/physiology , Cerebral Cortex/cytology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , NFATC Transcription Factors/genetics , Neural Stem Cells/metabolism , Neurons/metabolism , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Site-Specific DNA-Methyltransferase (Adenine-Specific)/genetics , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
15.
Stem Cells ; 36(12): 1851-1862, 2018 12.
Article in English | MEDLINE | ID: mdl-30270540

ABSTRACT

There are no effective treatments for corneal endothelial diseases, except for corneal transplantation, as human corneal endothelial cells (hCECs) do not regenerate. The regeneration of hCECs could be induced through regulation of the expression of specific genes. In this study, we investigated whether the overexpression of sex-determining region Y-box 2 (SOX2) can regenerate hCECs in vivo and in vitro. SOX2 was activated using the clustered regularly interspaced short palindromic repeats (CRISPR)/deactivated CRISPR-associated protein 9 (dCas9) activation system. Genes were transfected into the corneal endothelium of Sprague-Dawley rats. Central corneal thickness and opacity were measured, and alizarin red S staining was performed. Corneal opacity and central corneal thickness were reduced in the SOX2 group compared with the control group. The density of CECs was higher in the SOX2 group compared with the control group. Additionally, hCECs were cultured and analyzed after overexpressing SOX2. Cell viability, proliferation rate, and the number of cells in S-phase were increased after SOX2 overexpression (p < .05). Cyclin-dependent kinase 1 and cyclin D1 were found to be overexpressed (p < .05). WNT signaling was repressed, and the AKT pathway was activated by SOX2 overexpression. Mitochondrial oxidative stress and energy production were increased by SOX2 overexpression (p < .05). In conclusion, SOX2 activation promotes wound healing and regeneration in CECs. SOX2 activation using the CRISPR/dCas9 system may thus be useful for the treatment of hCEC diseases. Stem Cells 2018;36:1851-12.


Subject(s)
Corneal Diseases/pathology , Endothelial Cells/pathology , Endothelium, Corneal/pathology , SOXB1 Transcription Factors/biosynthesis , Wound Healing/physiology , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Corneal Diseases/genetics , Corneal Diseases/metabolism , Endothelial Cells/metabolism , Endothelium, Corneal/metabolism , Humans , Rats , Rats, Sprague-Dawley , Regeneration/physiology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
16.
Stem Cells ; 36(10): 1514-1524, 2018 10.
Article in English | MEDLINE | ID: mdl-29893054

ABSTRACT

Stem cell surface markers may facilitate a better understanding of stem cell biology through molecular function studies or serve as tools to monitor the differentiation status and behavior of stem cells in culture or tissue. Thus, it is important to identify additional novel stem cell markers. We used glycoproteomics to discover surface glycoproteins on human embryonic stem cells (hESCs) that may be useful stem cell markers. We found that a surface glycoprotein, leucine-rich repeat neuronal protein 1 (LRRN1), is expressed abundantly on the surface of hESCs before differentiation into embryoid bodies (EBs). Silencing of LRRN1 with short hairpin RNA (shLRRN1) in hESCs resulted in decreased capacity of self-renewal, and skewed differentiation toward endoderm/mesoderm lineages in vitro and in vivo. Meanwhile, the protein expression levels of the pluripotency factors OCT4, NANOG, and SOX2 were reduced. Interestingly, the mRNA levels of these pluripotency factors were not affected in LRRN1 silenced cells, but protein half-lives were substantially shortened. Furthermore, we found LRRN1 silencing led to nuclear export and proteasomal degradation of all three pluripotency factors. In addition, the effects on nuclear export were mediated by AKT phosphorylation. These results suggest that LRRN1 plays an important role in maintaining the protein stability of pluripotency factors through AKT phosphorylation, thus maintaining hESC self-renewal capacity and pluripotency. Overall, we found that LRRN1 contributes to pluripotency of hESC by preventing translocation of OCT4, NANOG, and SOX2 from nucleus to cytoplasm, thereby lessening their post-translational modification and degradation. Stem Cells 2018;36:1514-1524.


Subject(s)
Embryonic Stem Cells/metabolism , Membrane Proteins/metabolism , Nanog Homeobox Protein/metabolism , Neoplasm Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Humans , Nanog Homeobox Protein/biosynthesis , Nanog Homeobox Protein/genetics , Nerve Tissue Proteins , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/cytology , Protein Processing, Post-Translational , Protein Stability , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics
17.
Dev Biol ; 422(2): 155-170, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28017643

ABSTRACT

Classical grafting experiments in the Mexican axolotl had shown that the posterior neural plate of the neurula is no specified neuroectoderm but gives rise to somites of the tail and posterior trunk. The bipotentiality of this region with neuromesodermal progenitor cell populations was revealed more recently also in zebrafish, chick, and mouse. We reinvestigated the potency of the posterior plate in axolotl using grafts from transgenic embryos, immunohistochemistry, and in situ hybridization. The posterior plate is brachyury-positive except for its more anterior parts which express sox2. Between anterior and posterior regions of the posterior plate a small domain with sox2+ and bra+ cells exists. Lineage analysis of grafted GFP-labeled posterior plate tissue revealed that posterior GFP+ cells move from dorsal to ventral, form the posterior wall, turn anterior bilaterally, and join the gastrulated paraxial presomitic mesoderm. More anterior sox2+/GFP+ cells, however, are integrated into the developing spinal cord. Tail notochord is formed from axial mesoderm involuted already during gastrulation. Thus the posterior neural plate is a postgastrula source of paraxial mesoderm, which performs an anterior turn, a novel morphogenetic movement. More anterior plate cells, in contrast, do not turn anteriorly but become specified to form tail spinal cord.


Subject(s)
Ambystoma mexicanum/embryology , Mesoderm/embryology , Neural Plate/embryology , Neural Tube/embryology , Spinal Cord/embryology , Tail/embryology , Animals , Animals, Genetically Modified , Cells, Cultured , Fetal Proteins/metabolism , Gastrulation/physiology , Green Fluorescent Proteins/genetics , Notochord/embryology , SOXB1 Transcription Factors/biosynthesis , Somites/embryology , Stem Cells/cytology , T-Box Domain Proteins/metabolism
18.
J Biol Chem ; 292(23): 9840-9854, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28298438

ABSTRACT

The mammalian post-implantation embryo has been extensively investigated at the tissue level. However, to unravel the molecular basis for the cell-fate plasticity and determination, it is essential to study the characteristics of individual cells. In particular, the individual definitive endoderm (DE) cells have not been characterized in vivo Here, we report gene expression patterns in single cells freshly isolated from mouse embryos on days 5.5 and 6.5. Initial transcriptome data from 124 single cells yielded signature genes for the epiblast, visceral endoderm, and extra-embryonic ectoderm and revealed a unique distribution pattern of fibroblast growth factor (FGF) ligands and receptors. Further analysis indicated that early-stage epiblast cells do not segregate into lineages of the major germ layers. Instead, some cells began to diverge from epiblast cells, displaying molecular features of the premesendoderm by expressing higher levels of mesendoderm markers and lower levels of Sox3 transcripts. Analysis of single-cell high-throughput quantitative RT-PCR data from 441 cells identified a late stage of the day 6.5 embryo in which mesoderm and DE cells emerge, with many of them coexpressing Oct4 and Gata6 Analysis of single-cell RNA-sequence data from 112 cells of the late-stage day 6.5 embryos revealed differentially expressed signaling genes and networks of transcription factors that might underlie the segregation of the mesoderm and DE lineages. Moreover, we discovered a subpopulation of mesoderm cells that possess molecular features of the extraembryonic mesoderm. This study provides fundamental insight into the molecular basis for lineage segregation in post-implantation mouse embryos.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Lineage/physiology , Embryo, Mammalian , Embryonic Development/physiology , Gene Expression Regulation, Developmental/physiology , Transcriptome/physiology , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , GATA6 Transcription Factor/biosynthesis , Mice , Octamer Transcription Factor-3/biosynthesis , SOXB1 Transcription Factors/biosynthesis
19.
Biol Reprod ; 99(5): 1057-1069, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29931041

ABSTRACT

Severe uterine damage and infection lead to intrauterine adhesions, which result in hypomenorrhea, amenorrhea and infertility. Cell sheet engineering has shown great promise in clinical applications. Adipose-derived stem cells (ADSCs) are emerging as an alternative source of stem cells for cell-based therapies. In the present study, we investigated the feasibility of applying ADSCs as seed cells to form scaffold-free cell sheet. Data showed that ADSC sheets expressed higher levels of FGF, Col I, TGFß, and VEGF than ADSCs in suspension, while increased expression of this gene set was associated with stemness, including Nanog, Oct4, and Sox2. We then investigated the therapeutic effects of 3D ADSCs sheet on regeneration in a rat model. We found that ADSCs were mainly detected in the basal layer of the regenerating endometrium in the cell sheet group at 21 days after transplantation. Additionally, some ADSCs differentiated into stromal-like cells. Moreover, ADSC sheets transplanted into partially excised uteri promoted regeneration of the endometrium cells, muscle cells and stimulated angiogenesis, and also resulted in better pregnancy outcomes. Therefore, ADSC sheet therapy shows considerable promise as a new treatment for severe uterine damage.


Subject(s)
Adipose Tissue/cytology , Adnexa Uteri/growth & development , Stem Cell Transplantation , Animals , Cell Differentiation , Endometrium/cytology , Endometrium/growth & development , Feasibility Studies , Female , Nanog Homeobox Protein/biosynthesis , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Regeneration , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics
20.
Mol Reprod Dev ; 85(7): 590-598, 2018 07.
Article in English | MEDLINE | ID: mdl-29719081

ABSTRACT

Upregulation of Cdx2 expression in outer cells is a key event responsible for cell lineage segregation between the inner cell mass and the trophoderm (TE) in mouse morula-stage embryos. In TE cells, polarization can regulate Hippo and Rho-associated kinase (Rho-ROCK) signaling to induce the nuclear location of YAP, which has been demonstrated to further induce the expression of Cdx2. However, we found that CDX2 expression could not be detected in the outer cells of porcine morula-stage embryos but only in some TE cells at the early blastocyst stage. The biological significance and the regulation mechanism of this species-specific CDX2 expression pattern have still not been determined. We show here that an asynchronous CDX2 expression pattern exists in porcine TE cells during the development of the blastocyst. We demonstrate that CDX2 expression in porcine TE cells depends on the nuclear localization of YAP and polarization of the embryo through Y27632 treatment. We found that the polarization process in the morula to the late blastocyst stage porcine embryos was asynchronous, which was revealed by the apical localization of phosphorylated EZRIN staining. Artificially enhancing the number of polarized blastomeres by culturing the separated blastomeres of four-cell stage porcine embryos resulted in increased CDX2-positive cell numbers. These results indicate that the mechanism of CDX2 expression regulation is conserved, but the polarization progress is not conserved between the pig and the mouse, and results in a species-specific trophoblast determination progress model.


Subject(s)
Blastocyst/metabolism , Blastomeres/metabolism , CDX2 Transcription Factor/biosynthesis , Morula/metabolism , SOXB1 Transcription Factors/biosynthesis , Trophoblasts/metabolism , Amides/pharmacology , Animals , Cell Lineage , Cells, Cultured , Cytoskeletal Proteins/metabolism , Embryo Culture Techniques , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice , Phosphorylation , Proto-Oncogene Proteins c-yes/metabolism , Pyridines/pharmacology , Swine , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL