Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 923
Filter
Add more filters

Publication year range
1.
Cell ; 184(25): 6081-6100.e26, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34861191

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable success in hematological malignancies but remains ineffective in solid tumors, due in part to CAR T cell exhaustion in the solid tumor microenvironment. To study dysfunction of mesothelin-redirected CAR T cells in pancreatic cancer, we establish a robust model of continuous antigen exposure that recapitulates hallmark features of T cell exhaustion and discover, both in vitro and in CAR T cell patients, that CAR dysregulation is associated with a CD8+ T-to-NK-like T cell transition. Furthermore, we identify a gene signature defining CAR and TCR dysregulation and transcription factors, including SOX4 and ID3 as key regulators of CAR T cell exhaustion. Our findings shed light on the plasticity of human CAR T cells and demonstrate that genetic downmodulation of ID3 and SOX4 expression can improve the efficacy of CAR T cell therapy in solid tumors by preventing or delaying CAR T cell dysfunction.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Pancreatic Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , HEK293 Cells , Humans , Inhibitor of Differentiation Proteins/immunology , Male , Mice , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplasm Proteins/immunology , SOXC Transcription Factors/immunology
2.
Cell ; 164(5): 1015-30, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26898331

ABSTRACT

TGF-ß signaling can be pro-tumorigenic or tumor suppressive. We investigated this duality in pancreatic ductal adenocarcinoma (PDA), which, with other gastrointestinal cancers, exhibits frequent inactivation of the TGF-ß mediator Smad4. We show that TGF-ß induces an epithelial-mesenchymal transition (EMT), generally considered a pro-tumorigenic event. However, in TGF-ß-sensitive PDA cells, EMT becomes lethal by converting TGF-ß-induced Sox4 from an enforcer of tumorigenesis into a promoter of apoptosis. This is the result of an EMT-linked remodeling of the cellular transcription factor landscape, including the repression of the gastrointestinal lineage-master regulator Klf5. Klf5 cooperates with Sox4 in oncogenesis and prevents Sox4-induced apoptosis. Smad4 is required for EMT but dispensable for Sox4 induction by TGF-ß. TGF-ß-induced Sox4 is thus geared to bolster progenitor identity, whereas simultaneous Smad4-dependent EMT strips Sox4 of an essential partner in oncogenesis. Our work demonstrates that TGF-ß tumor suppression functions through an EMT-mediated disruption of a lineage-specific transcriptional network.


Subject(s)
Carcinoma, Ductal/genetics , Epithelial-Mesenchymal Transition , Gene Regulatory Networks , Pancreatic Neoplasms/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis , Carcinoma, Ductal/pathology , Kruppel-Like Transcription Factors/metabolism , Mice , Organoids/metabolism , Organoids/pathology , Pancreatic Neoplasms/pathology , SOXC Transcription Factors/metabolism , Smad4 Protein/metabolism
3.
Nature ; 599(7885): 453-457, 2021 11.
Article in English | MEDLINE | ID: mdl-34754107

ABSTRACT

Interconnectivity between neocortical areas is critical for sensory integration and sensorimotor transformations1-6. These functions are mediated by heterogeneous inter-areal cortical projection neurons (ICPN), which send axon branches across cortical areas as well as to subcortical targets7-9. Although ICPN are anatomically diverse10-14, they are molecularly homogeneous15, and how the diversity of their anatomical and functional features emerge during development remains largely unknown. Here we address this question by linking the connectome and transcriptome in developing single ICPN of the mouse neocortex using a combination of multiplexed analysis of projections by sequencing16,17 (MAPseq, to identify single-neuron axonal projections) and single-cell RNA sequencing (to identify corresponding gene expression). Focusing on neurons of the primary somatosensory cortex (S1), we reveal a protracted unfolding of the molecular and functional differentiation of motor cortex-projecting ([Formula: see text]) ICPN compared with secondary somatosensory cortex-projecting ([Formula: see text]) ICPN. We identify SOX11 as a temporally differentially expressed transcription factor in [Formula: see text] versus [Formula: see text] ICPN. Postnatal manipulation of SOX11 expression in S1 impaired sensorimotor connectivity and disrupted selective exploratory behaviours in mice. Together, our results reveal that within a single cortical area, different subtypes of ICPN have distinct postnatal paces of molecular differentiation, which are subsequently reflected in distinct circuit connectivities and functions. Dynamic differences in the expression levels of a largely generic set of genes, rather than fundamental differences in the identity of developmental genetic programs, may thus account for the emergence of intra-type diversity in cortical neurons.


Subject(s)
Cell Differentiation , Neural Pathways , Neurons/cytology , Neurons/physiology , Somatosensory Cortex/cytology , Somatosensory Cortex/physiology , Animals , Axons/physiology , Connectome , Female , Male , Mice , Mice, Inbred C57BL , Motor Cortex/cytology , Motor Cortex/physiology , Neocortex/cytology , Neocortex/physiology , SOXC Transcription Factors/genetics , Time Factors , Transcriptome
4.
Blood ; 143(19): 1953-1964, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38237141

ABSTRACT

ABSTRACT: Sterile alpha motif and histidine-aspartate (HD) domain-containing protein 1 (SAMHD1) is a deoxynucleoside triphosphate triphosphohydrolase with ara-CTPase activity that confers cytarabine (ara-C) resistance in several hematological malignancies. Targeting SAMHD1's ara-CTPase activity has recently been demonstrated to enhance ara-C efficacy in acute myeloid leukemia. Here, we identify the transcription factor SRY-related HMG-box containing protein 11 (SOX11) as a novel direct binding partner and first known endogenous inhibitor of SAMHD1. SOX11 is aberrantly expressed not only in mantle cell lymphoma (MCL), but also in some Burkitt lymphomas. Coimmunoprecipitation of SOX11 followed by mass spectrometry in MCL cell lines identified SAMHD1 as the top SOX11 interaction partner, which was validated by proximity ligation assay. In vitro, SAMHD1 bound to the HMG box of SOX11 with low-micromolar affinity. In situ crosslinking studies further indicated that SOX11-SAMHD1 binding resulted in a reduced tetramerization of SAMHD1. Functionally, expression of SOX11 inhibited SAMHD1 ara-CTPase activity in a dose-dependent manner resulting in ara-C sensitization in cell lines and in a SOX11-inducible mouse model of MCL. In SOX11-negative MCL, SOX11-mediated ara-CTPase inhibition could be mimicked by adding the recently identified SAMHD1 inhibitor hydroxyurea. Taken together, our results identify SOX11 as a novel SAMHD1 interaction partner and its first known endogenous inhibitor with potentially important implications for clinical therapy stratification.


Subject(s)
Lymphoma, Mantle-Cell , SAM Domain and HD Domain-Containing Protein 1 , SOXC Transcription Factors , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Humans , SAM Domain and HD Domain-Containing Protein 1/metabolism , SAM Domain and HD Domain-Containing Protein 1/genetics , Animals , Mice , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Protein Binding , Cell Line, Tumor , Cytarabine/pharmacology
5.
Blood ; 143(19): 1953-1964, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38774451

ABSTRACT

The sterile alpha motif and histidine-aspartate (HD) domain containing protein 1 (SAMHD1) is a deoxynucleoside triphosphate triphosphohydrolase with ara-CTPase activity that confers cytarabine (ara-C) resistance in several haematological malignancies. Targeting SAMHD1's ara-CTPase activity has recently been demonstrated to enhance ara-C efficacy in acute myeloid leukemia. Here, we identify the transcription factor SRY-related HMG-box containing protein 11 (SOX11) as a novel direct binding partner and first known endogenous inhibitor of SAMHD1. SOX11 is aberrantly expressed not only in mantle cell lymphoma (MCL), but also in some Burkitt lymphomas. Co-immunoprecipitation of SOX11 followed by mass spectrometry in MCL cell lines identified SAMHD1 as the top SOX11 interaction partner which was validated by proximity ligation assay. In vitro, SAMHD1 bound to the HMG box of SOX11 with low-micromolar affinity. In situ crosslinking studies further indicated that SOX11-SAMHD1 binding resulted in a reduced tetramerization of SAMHD1. Functionally, expression of SOX11 inhibited SAMHD1 ara-CTPase activity in a dose-dependent manner resulting in ara-C sensitization in cell lines and in a SOX11-inducible mouse model of MCL. In SOX11-negative MCL, SOX11-mediated ara-CTPase inhibition could be mimicked by adding the recently identified SAMHD1 inhibitor hydroxyurea. Taken together, our results identify SOX11 as a novel SAMHD1 interaction partner and its first known endogenous inhibitor with potentially important implications for clinical therapy stratification.


Subject(s)
Lymphoma, Mantle-Cell , SAM Domain and HD Domain-Containing Protein 1 , SOXC Transcription Factors , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Humans , SAM Domain and HD Domain-Containing Protein 1/metabolism , SAM Domain and HD Domain-Containing Protein 1/genetics , Animals , Mice , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Protein Binding , Cell Line, Tumor , Cytarabine/pharmacology
6.
Nature ; 580(7805): 669-672, 2020 04.
Article in English | MEDLINE | ID: mdl-32350470

ABSTRACT

'Pioneer' transcription factors are required for stem-cell pluripotency, cell differentiation and cell reprogramming1,2. Pioneer factors can bind nucleosomal DNA to enable gene expression from regions of the genome with closed chromatin. SOX2 is a prominent pioneer factor that is essential for pluripotency and self-renewal of embryonic stem cells3. Here we report cryo-electron microscopy structures of the DNA-binding domains of SOX2 and its close homologue SOX11 bound to nucleosomes. The structures show that SOX factors can bind and locally distort DNA at superhelical location 2. The factors also facilitate detachment of terminal nucleosomal DNA from the histone octamer, which increases DNA accessibility. SOX-factor binding to the nucleosome can also lead to a repositioning of the N-terminal tail of histone H4 that includes residue lysine 16. We speculate that this repositioning is incompatible with higher-order nucleosome stacking, which involves contacts of the H4 tail with a neighbouring nucleosome. Our results indicate that pioneer transcription factors can use binding energy to initiate chromatin opening, and thereby facilitate nucleosome remodelling and subsequent transcription.


Subject(s)
Chromatin Assembly and Disassembly , Cryoelectron Microscopy , Nucleosomes/metabolism , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/metabolism , SOXC Transcription Factors/chemistry , SOXC Transcription Factors/metabolism , Base Sequence , DNA, Superhelical/chemistry , DNA, Superhelical/genetics , DNA, Superhelical/metabolism , Histones/chemistry , Histones/metabolism , Humans , Lysine/metabolism , Models, Biological , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Nucleosomes/chemistry , Nucleosomes/ultrastructure , SOXB1 Transcription Factors/ultrastructure , SOXC Transcription Factors/ultrastructure
7.
Nature ; 585(7824): 283-287, 2020 09.
Article in English | MEDLINE | ID: mdl-32814897

ABSTRACT

The risk of cancer and associated mortality increases substantially in humans from the age of 65 years onwards1-6. Nonetheless, our understanding of the complex relationship between age and cancer is still in its infancy2,3,7,8. For decades, this link has largely been attributed to increased exposure time to mutagens in older individuals. However, this view does not account for the established role of diet, exercise and small molecules that target the pace of metabolic ageing9-12. Here we show that metabolic alterations that occur with age can produce a systemic environment that favours the progression and aggressiveness of tumours. Specifically, we show that methylmalonic acid (MMA), a by-product of propionate metabolism, is upregulated in the serum of older people and functions as a mediator of tumour progression. We traced this to the ability of MMA to induce SOX4 expression and consequently to elicit transcriptional reprogramming that can endow cancer cells with aggressive properties. Thus, the accumulation of MMA represents a link between ageing and cancer progression, suggesting that MMA is a promising therapeutic target for advanced carcinomas.


Subject(s)
Aging/metabolism , Disease Progression , Methylmalonic Acid/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology , Adult , Aged , Aging/blood , Aging/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Methylmalonic Acid/blood , Mice , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasms/blood , Neoplasms/genetics , SOXC Transcription Factors/metabolism , Signal Transduction , Transcriptome/genetics , Transforming Growth Factor beta/metabolism
8.
Mol Cell ; 72(3): 553-567.e5, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30401432

ABSTRACT

In mammals, neurons in the peripheral nervous system (PNS) have regenerative capacity following injury, but it is generally absent in the CNS. This difference is attributed, at least in part, to the intrinsic ability of PNS neurons to activate a unique regenerative transcriptional program following injury. Here, we profiled gene expression following sciatic nerve crush in mice and identified long noncoding RNAs (lncRNAs) that act in the regenerating neurons and which are typically not expressed in other contexts. We show that two of these lncRNAs regulate the extent of neuronal outgrowth. We then focus on one of these, Silc1, and show that it regulates neuroregeneration in cultured cells and in vivo, through cis-acting activation of the transcription factor Sox11.


Subject(s)
Nerve Regeneration/genetics , RNA, Long Noncoding/physiology , Animals , Cell Line, Tumor , Ganglia, Spinal , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/metabolism , Neurites/physiology , Neurons/physiology , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/physiopathology , RNA, Long Noncoding/genetics , RNA, Messenger , SOXC Transcription Factors , Sciatic Nerve/metabolism
9.
Proc Natl Acad Sci U S A ; 120(34): e2301301120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37585469

ABSTRACT

The auditory organ of Corti is comprised of only two major cell types-the mechanosensory hair cells and their associated supporting cells-both specified from a single pool of prosensory progenitors in the cochlear duct. Here, we show that competence to respond to Atoh1, a transcriptional master regulator necessary and sufficient for induction of mechanosensory hair cells, is established in the prosensory progenitors between E12.0 and 13.5. The transition to the competent state is rapid and is associated with extensive remodeling of the epigenetic landscape controlled by the SoxC group of transcription factors. Conditional loss of Sox4 and Sox11-the two homologous family members transiently expressed in the inner ear at the time of competence establishment-blocks the ability of prosensory progenitors to differentiate as hair cells. Mechanistically, we show that Sox4 binds to and establishes accessibility of early sensory lineage-specific regulatory elements, including ones associated with Atoh1 and its direct downstream targets. Consistent with these observations, overexpression of Sox4 or Sox11 prior to developmental establishment of competence precociously induces hair cell differentiation in the cochlear progenitors. Further, reintroducing Sox4 or Sox11 expression restores the ability of postnatal supporting cells to differentiate as hair cells in vitro and in vivo. Our findings demonstrate the pivotal role of SoxC family members as agents of epigenetic and transcriptional changes necessary for establishing competence for sensory receptor differentiation in the inner ear.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , SOXC Transcription Factors , Animals , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cochlea/metabolism , Hair Cells, Auditory/metabolism , Cell Differentiation , Transcription Factors/metabolism , Epigenesis, Genetic , Organ of Corti , Gene Expression Regulation, Developmental , Mammals/metabolism
10.
EMBO J ; 40(19): e108041, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34431536

ABSTRACT

The role of WNT/ß-catenin signalling in mouse neocortex development remains ambiguous. Most studies demonstrate that WNT/ß-catenin regulates progenitor self-renewal but others suggest it can also promote differentiation. Here we explore the role of WNT/STOP signalling, which stabilizes proteins during G2/M by inhibiting glycogen synthase kinase (GSK3)-mediated protein degradation. We show that mice mutant for cyclin Y and cyclin Y-like 1 (Ccny/l1), key regulators of WNT/STOP signalling, display reduced neurogenesis in the developing neocortex. Specifically, basal progenitors, which exhibit delayed cell cycle progression, were drastically decreased. Ccny/l1-deficient apical progenitors show reduced asymmetric division due to an increase in apical-basal astral microtubules. We identify the neurogenic transcription factors Sox4 and Sox11 as direct GSK3 targets that are stabilized by WNT/STOP signalling in basal progenitors during mitosis and that promote neuron generation. Our work reveals that WNT/STOP signalling drives cortical neurogenesis and identifies mitosis as a critical phase for neural progenitor fate.


Subject(s)
Mitosis , Neocortex/embryology , Neocortex/metabolism , Neurogenesis , Wnt Signaling Pathway , Amino Acid Sequence , Animals , Biomarkers , Cell Cycle , Cell Differentiation/genetics , Cyclins/genetics , Cyclins/metabolism , Embryo, Mammalian , Fluorescent Antibody Technique , Gene Expression , Glycogen Synthase Kinase 3/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Mitosis/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Phosphorylation , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
11.
EMBO J ; 40(21): e107277, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34558085

ABSTRACT

The dorsal and ventral human telencephalons contain different neuronal subtypes, including glutamatergic, GABAergic, and cholinergic neurons, and how these neurons are generated during early development is not well understood. Using scRNA-seq and stringent validations, we reveal here a developmental roadmap for human telencephalic neurons. Both dorsal and ventral telencephalic radial glial cells (RGs) differentiate into neurons via dividing intermediate progenitor cells (IPCs_div) and early postmitotic neuroblasts (eNBs). The transcription factor ASCL1 plays a key role in promoting fate transition from RGs to IPCs_div in both regions. RGs from the regionalized neuroectoderm show heterogeneity, with restricted glutamatergic, GABAergic, and cholinergic differentiation potencies. During neurogenesis, IPCs_div gradually exit the cell cycle and branch into sister eNBs to generate distinct neuronal subtypes. Our findings highlight a general RGs-IPCs_div-eNBs developmental scheme for human telencephalic progenitors and support that the major neuronal fates of human telencephalon are predetermined during dorsoventral regionalization with neuronal diversity being further shaped during neurogenesis and neural circuit integration.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Neurons/metabolism , Telencephalon/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/genetics , Cell Differentiation , Choline/metabolism , Doublecortin Protein/genetics , Doublecortin Protein/metabolism , Fetus , Gene Ontology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Molecular Sequence Annotation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/classification , Neurons/cytology , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Signal Transduction , Stathmin/genetics , Stathmin/metabolism , Telencephalon/cytology , Telencephalon/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism
12.
Exp Cell Res ; 439(1): 114059, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38705228

ABSTRACT

Filopodia are thin, actin-rich projection from the plasma membrane that promote cancer cell invasion and migration. Sex-determining region Y-related high-mobility group-box 4 (SOX4) is a crucial transcription factor that plays a role in the development and metastasis of colorectal cancer (CRC). However, the involvement of SOX4 in cytoskeleton remodeling in CRC remains unknown. For the first time, we demonstrate that SOX4 is a potent regulator of filopodia formation in CRC cells. Overexpression of SOX4 protein enhances both migration and invasion ability of HCT116, and CACO2 cells, which is relevant to the metastasis. Furthermore, through phalloidin staining, cytoskeleton re-assembly was observed in SOX4-modified cell lines. Enhanced expression of SOX4 increased the number and length of filopodia on cell surface. In contrast, silencing SOX4 in SW620 cells with higher endogenous expression of SOX4, impeded the filopodia formation. Moreover, SOX4 was found to be positively regulating the expression of central regulators of actin cytoskeleton - N-Wiskott-Aldrich syndrome protein (N-WASP); WAVE2; Actin related proteins, ARP2 and ARP3. Inhibiting the N-WASP/ARP2/3 pathway diminishes the filopodia formation and the migration of CRC cells. These results indicate the crucial role of SOX4 in the regulation of filopodia formation mediated by N-WASP/ARP2/3 pathway in CRC cells.


Subject(s)
Actin-Related Protein 2-3 Complex , Cell Movement , Colorectal Neoplasms , Cytoskeleton , Pseudopodia , SOXC Transcription Factors , Wiskott-Aldrich Syndrome Protein, Neuronal , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Cell Movement/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2-3 Complex/genetics , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , Cytoskeleton/metabolism , Pseudopodia/metabolism , Caco-2 Cells , Signal Transduction , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , HCT116 Cells , Actin Cytoskeleton/metabolism
13.
Lab Invest ; 104(5): 102042, 2024 May.
Article in English | MEDLINE | ID: mdl-38431117

ABSTRACT

Esophageal squamous cell carcinoma stands as a notably aggressive malignancy within the digestive system. In cases of early esophageal cancer without lymph node metastasis, endoscopic surgical resection offers a viable alternative, often resulting in improved patient quality of life. However, the paucity of methods to preoperatively ascertain lymph node involvement complicates surgical planning. SOX4 gene was previously found to be highly associated with invasive metastasis in our work through single-cell RNA sequencing on 5 paired tumor/peritumor tissues. This research included the collection of 124 tissue samples from 106 patients (106 tumor and 18 lymph node specimens). Samples were methodically arranged into a tissue microarray and treated with immunohistochemical staining. Statistical analysis was conducted to assess the relationship between them. In the univariate analysis, 3 factors were identified as statistically significant in relation to lymph node metastasis: T category (P = .014), vascular invasion (P < .001), and SOX4 intensity (P = .001). Additionally, when evaluating SOX4 intensity alongside other clinical indicators, SOX4 was shown to independently influence lymph node metastasis. Further, the multivariate analysis revealed that vascular invasion (P < .001) and SOX4 intensity (P = .003) were significantly associated with lymph node metastasis, exhibiting hazard ratios of 10.174 and 7.142, respectively. The results of our study indicate that both SOX4 expression and vascular invasion serve as predictors of lymph node metastasis in patients diagnosed with category T1 esophageal squamous cell carcinoma, underscoring the potential utility of SOX4 in prognostic evaluations.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lymphatic Metastasis , SOXC Transcription Factors , Humans , Male , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Female , Middle Aged , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/secondary , Esophageal Squamous Cell Carcinoma/surgery , Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Lymph Nodes/pathology , Lymph Nodes/metabolism , Adult , Prognosis
14.
Biochem Biophys Res Commun ; 705: 149738, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38447391

ABSTRACT

The proliferation and apoptosis of ovarian granulosa cells are important for folliculogenesis. As a transcription factor, SRY-box transcription factor 4 (SOX4) has important roles in regulating cellular proliferation and apoptosis. Nonetheless, the regulatory mechanisms of SOX4 on proliferation and apoptosis of granulosa cells remain elusive. Therefore, a stably overexpressed SOX4 ovarian granulosa cell line KGN was generated by lentivirus encapsulation. We observed that overexpression of SOX4 inhibits apoptosis, promotes proliferation and migration of KGN cells. Comparative analysis of the transcriptome revealed 868 upregulated and 696 downregulated DEGs in LV-SOX4 in comparison with LV-CON KGN cell lines. Afterward, further assessments were performed to explore the possible functions about these DEGs. The data showed their involvement in many biological processes, particularly the Hippo signaling pathway. Moreover, the expression levels of YAP1, WWTR1, WTIP, DLG3, CCN2, and AMOT, which were associated with the Hippo signaling pathway, were further validated by qRT-PCR. In addition, the protein expression levels of YAP1 were markedly elevated, while p-YAP1 were notably reduced after overexpression of SOX4 in KGN cells. Thus, these results suggested that SOX4 regulates apoptosis, proliferation and migration of KGN cells, at least partly, through activation of the Hippo signaling pathway, which might be implicated in mammalian follicle development.


Subject(s)
Granulosa Cells , Hippo Signaling Pathway , Female , Animals , Humans , Cell Line, Tumor , Granulosa Cells/metabolism , Cell Proliferation , Apoptosis , Mammals/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Cytoskeletal Proteins/metabolism , Co-Repressor Proteins/metabolism
15.
Biochem Biophys Res Commun ; 708: 149815, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38531220

ABSTRACT

Sesamin, a special compound present in sesame and sesame oil, has been reported a role in regulating lipid metabolism, while the underlying mechanisms remain unclear. Autophagy has been reported associated with lipid metabolism and regarded as a key modulator in liver steatosis. The present work aimed to investigate whether sesamin could exert its protective effects against lipid accumulation via modulating autophagy in HepG2 cells stimulated with oleic acid (OA). Cell viability was evaluated using the CCK-8 method, and triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein, cholesterol (LDL-C), alanine aminotransferase (ALT), along with aspartate aminotransferase (AST) were assessed by oil red O staining, transmission electron microscopy (TEM), and biochemical kits to investigate the lipid-lowering effects of sesamin. Differentially expressed genes were screened by RNA sequencing and validated using real-time quantitative PCR and Western blot. Autophagy and mitophagy related molecules were analyzed employing TEM, Western blot, and immunofluorescence. The data shows that in HepG2 cells stimulated by OA, sesamin reduces levels of TG, TC, LDL-C, ALT, and AST while elevating HDL-C, alleviates the lipid accumulation and improves fatty acid metabolism through modulating the levels of fat metabolism related genes including PCSK9, FABP1, CD36, and SOX4. Sesamin restores the suppressed autophagy in HepG2 cells caused by OA, which could be blocked by autophagy inhibitors. This indicates that sesamin improves fatty acid metabolism by enhancing autophagy levels, thereby mitigating the intracellular lipid accumulation. Furthermore, sesamin significantly enhances the mitophagy and improves mitochondrial homeostasis via activating the PINK/Parkin pathway. These data suggest that sesamin alleviates the excessive lipid accumulation in HepG2 caused by OA by restoring the impaired mitophagy via the PINK1/Parkin pathway, probably playing a preventive or therapeutic role in hepatic steatosis.


Subject(s)
Dioxoles , Fatty Liver , Lignans , Proprotein Convertase 9 , SOXC Transcription Factors , Humans , Hep G2 Cells , Proprotein Convertase 9/metabolism , Mitophagy , Oleic Acid/metabolism , Cholesterol, LDL/metabolism , Cholesterol, LDL/pharmacology , Fatty Liver/metabolism , Lipid Metabolism , Cholesterol/metabolism , Triglycerides/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Liver/metabolism
16.
Development ; 148(14)2021 07 15.
Article in English | MEDLINE | ID: mdl-34184026

ABSTRACT

Transcription factor 4 (TCF4) is a crucial regulator of neurodevelopment and has been linked to the pathogenesis of autism, intellectual disability and schizophrenia. As a class I bHLH transcription factor (TF), it is assumed that TCF4 exerts its neurodevelopmental functions through dimerization with proneural class II bHLH TFs. Here, we aim to identify TF partners of TCF4 in the control of interhemispheric connectivity formation. Using a new bioinformatic strategy integrating TF expression levels and regulon activities from single cell RNA-sequencing data, we find evidence that TCF4 interacts with non-bHLH TFs and modulates their transcriptional activity in Satb2+ intercortical projection neurons. Notably, this network comprises regulators linked to the pathogenesis of neurodevelopmental disorders, e.g. FOXG1, SOX11 and BRG1. In support of the functional interaction of TCF4 with non-bHLH TFs, we find that TCF4 and SOX11 biochemically interact and cooperatively control commissure formation in vivo, and regulate the transcription of genes implicated in this process. In addition to identifying new candidate interactors of TCF4 in neurodevelopment, this study illustrates how scRNA-Seq data can be leveraged to predict TF networks in neurodevelopmental processes.


Subject(s)
RNA, Small Cytoplasmic/metabolism , Single-Cell Analysis , Transcription Factor 4/genetics , Transcription Factor 4/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , DNA Helicases , Embryo, Mammalian , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Intellectual Disability , Matrix Attachment Region Binding Proteins , Mice , Mice, Knockout , Nerve Tissue Proteins , Neurons/physiology , Nuclear Proteins , Protein Interaction Domains and Motifs , RNA, Small Cytoplasmic/genetics , SOXC Transcription Factors , Schizophrenia/genetics , Schizophrenia/metabolism
17.
Mod Pathol ; 37(2): 100405, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38104893

ABSTRACT

Large or blastoid B-cell neoplasms that are SOX11+ are a diagnostic dilemma and raise a differential diagnosis of cyclin D1-negative blastoid/pleomorphic mantle cell lymphoma (MCL) versus diffuse large B-cell lymphoma (DLBCL) or blastoid high-grade B-cell lymphoma (HGBL) with aberrant SOX11 expression. Here we report a study cohort of 13 SOX11+ large/blastoid B-cell neoplasms. Fluorescence in situ hybridization analysis was negative for CCND1 rearrangement in all 13 cases; 1 of 8 (12.5%) cases tested showed CCND2 rearrangement and 2 (25%) cases had extracopies of CCND2. Gene expression profiling showed that the study group had a gene expression signature similar to cyclin D1+ blastoid/pleomorphic MCL but different from DLBCL. Principal component analysis revealed that the cohort cases overlapped with cyclin D1+ blastoid/pleomorphic MCL but had minimal overlap with DLBCL. All patients in the cohort had clinicopathologic features similar to those reported for patients with cyclin D1+ MCL. We also performed a survey of SOX11 expression in a group of 85 cases of DLBCL and 24 cases of blastoid HGBL. SOX11 expression showed a 100% specificity and positive predictive value for the diagnosis of MCL. Overall, the results support the conclusion that large or blastoid B-cell neoplasms that are positive for SOX11 are best classified as cyclin D1-negative blastoid/pleomorphic MCL, and not as DLBCL or blastoid HGBL. We also conclude that SOX11 is a specific marker for the diagnosis of MCL, including cyclin D1-negative blastoid/pleomorphic MCL cases and should be performed routinely on blastoid/large B-cell neoplasms to help identify potential cases of cyclin D1-negative blastoid/pleomorphic MCL.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Lymphoma, Mantle-Cell , Adult , Humans , Lymphoma, Mantle-Cell/metabolism , Cyclin D1/genetics , In Situ Hybridization, Fluorescence , Immunohistochemistry , Lymphoma, Large B-Cell, Diffuse/pathology , SOXC Transcription Factors/genetics
18.
J Transl Med ; 22(1): 602, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943117

ABSTRACT

OBJECTIVE: This study aims to elucidate the functional role of IQGAP1 phosphorylation modification mediated by the SOX4/MAPK1 regulatory axis in developing pancreatic cancer through phosphoproteomics analysis. METHODS: Proteomics and phosphoproteomics data of pancreatic cancer were obtained from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database. Differential analysis, kinase-substrate enrichment analysis (KSEA), and independent prognosis analysis were performed on these datasets. Subtype analysis of pancreatic cancer patients was conducted based on the expression of prognostic-related proteins, and the prognosis of different subtypes was evaluated through prognosis analysis. Differential analysis of proteins in different subtypes was performed to identify differential proteins in the high-risk subtype. Clinical correlation analysis was conducted based on the expression of prognostic-related proteins, pancreatic cancer typing results, and clinical characteristics in the pancreatic cancer proteomics dataset. Functional pathway enrichment analysis was performed using GSEA/GO/KEGG, and most module proteins correlated with pancreatic cancer were selected using WGCNA analysis. In cell experiments, pancreatic cancer cells were grouped, and the expression levels of SOX4, MAPK1, and the phosphorylation level of IQGAP1 were detected by RT-qPCR and Western blot experiments. The effect of SOX4 on MAPK1 promoter transcriptional activity was assessed using a dual-luciferase assay, and the enrichment of SOX4 on the MAPK1 promoter was examined using a ChIP assay. The proliferation, migration, and invasion functions of grouped pancreatic cancer cells were assessed using CCK-8, colony formation, and Transwell assays. In animal experiments, the impact of SOX4 on tumor growth and metastasis through the regulation of MAPK1-IQGAP1 phosphorylation modification was studied by constructing subcutaneous and orthotopic pancreatic cancer xenograft models, as well as a liver metastasis model in nude mice. RESULTS: Phosphoproteomics and proteomics data analysis revealed that the kinase MAPK1 may play an important role in pancreatic cancer progression by promoting IQGAP1 phosphorylation modification. Proteomics analysis classified pancreatic cancer patients into two subtypes, C1 and C2, where the high-risk C2 subtype was associated with poor prognosis, malignant tumor typing, and enriched tumor-related pathways. SOX4 may promote the occurrence of the high-risk C2 subtype of pancreatic cancer by regulating MAPK1-IQGAP1 phosphorylation modification. In vitro cell experiments confirmed that SOX4 promoted IQGAP1 phosphorylation modification by activating MAPK1 transcription while silencing SOX4 inhibited the proliferation, migration, and invasion of pancreatic cancer cells by reducing the phosphorylation level of MAPK1-IQGAP1. In vivo, animal experiments further confirmed that silencing SOX4 suppressed the growth and metastasis of pancreatic cancer by reducing the phosphorylation level of MAPK1-IQGAP1. CONCLUSION: The findings of this study suggest that SOX4 promotes the phosphorylation modification of IQGAP1 by activating MAPK1 transcription, thereby facilitating the growth and metastasis of pancreatic cancer.


Subject(s)
Disease Progression , Pancreatic Neoplasms , Proteomics , SOXC Transcription Factors , ras GTPase-Activating Proteins , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Humans , Phosphorylation , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Cell Line, Tumor , Animals , Mitogen-Activated Protein Kinase 1/metabolism , Mice, Nude , Gene Expression Regulation, Neoplastic , Cell Proliferation , Prognosis , Mice , Male , Female , Phosphoproteins/metabolism , Signal Transduction , Cell Movement
19.
J Transl Med ; 22(1): 9, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38169402

ABSTRACT

Epigenetic regulation is reported to play a significant role in the pathogenesis of various kidney diseases, including renal cell carcinoma, acute kidney injury, renal fibrosis, diabetic nephropathy, and lupus nephritis. However, the role of epigenetic regulation in calcium oxalate (CaOx) crystal deposition-induced kidney injury remains unclear. Our study demonstrated that the upregulation of enhancer of zeste homolog 2 (EZH2)-mediated ferroptosis facilitates CaOx-induced kidney injury. CaOx crystal deposition promoted ferroptosis in vivo and in vitro. Usage of liproxstatin-1 (Lip-1), a ferroptosis inhibitor, mitigated CaOx-induced kidney damage. Single-nucleus RNA-sequencing, RNA-sequencing, immunohistochemical and western blotting analyses revealed that EZH2 was upregulated in kidney stone patients, kidney stone mice, and oxalate-stimulated HK-2 cells. Experiments involving in vivo EZH2 knockout, in vitro EZH2 knockdown, and in vivo GSK-126 (an EZH2 inhibitor) treatment confirmed the protective effects of EZH2 inhibition on kidney injury and ferroptosis. Mechanistically, the results of RNA-sequencing and chromatin immunoprecipitation assays demonstrated that EZH2 regulates ferroptosis by suppressing solute carrier family 7, member 11 (SLC7A11) expression through trimethylation of histone H3 lysine 27 (H3K27me3) modification. Additionally, SOX4 regulated ferroptosis by directly modulating EZH2 expression. Thus, this study demonstrated that SOX4 facilitates ferroptosis in CaOx-induced kidney injury through EZH2/H3K27me3-mediated suppression of SLC7A11.


Subject(s)
Diabetic Nephropathies , Ferroptosis , Kidney Calculi , Humans , Mice , Animals , Enhancer of Zeste Homolog 2 Protein/metabolism , Calcium Oxalate , Histones/metabolism , Epigenesis, Genetic , Kidney/pathology , Diabetic Nephropathies/metabolism , Kidney Calculi/pathology , RNA/metabolism , SOXC Transcription Factors/metabolism , Amino Acid Transport System y+
20.
Nat Immunol ; 13(8): 778-86, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22751141

ABSTRACT

Sox4 is a transcription factor that regulates various developmental processes. Here we show that Sox4 was induced by TGF-ß and negatively regulated the transcription factor GATA-3, the master regulator of function of T helper type 2 (T(H)2) cells, by two distinct mechanisms. First, Sox4 bound directly to GATA-3, preventing its binding to GATA-3 consensus DNA sequences. Second, Sox4 bound to the promoter region of the gene encoding interleukin 5 (IL-5), a T(H)2 cytokine, and prevented binding of GATA-3 to this promoter. T(H)2 cell-driven airway inflammation was modulated by alterations in Sox4 expression. Thus, Sox4 acted as a downstream target of TGF-ß to inhibit GATA-3 function, T(H)2 differentiation and T(H)2 cell-mediated inflammation.


Subject(s)
GATA3 Transcription Factor/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Th2 Cells/cytology , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , GATA3 Transcription Factor/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-5/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/immunology , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering , Signal Transduction , Th2 Cells/immunology , Th2 Cells/metabolism , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL