Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 271
Filtrar
1.
Cell Transplant ; 33: 9636897241248942, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38712762

RESUMO

Recently, we and others generated induced tissue-specific stem/progenitor (iTS/iTP) cells. The advantages of iTS/iTP cells compared with induced pluripotent stem (iPS) cells are (1) easier generation, (2) efficient differentiation, and (3) no teratomas formation. In this study, we generated mouse induced pancreatic stem cells (iTS-P cells) by the plasmid vector expressing Yes-associated protein 1 (YAP). The iTS-P YAP9 cells expressed Foxa2 (endoderm marker) and Pdx1 (pancreatic marker) while the expressions of Oct3/4 and Nanog (marker of embryonic stem [ES] cells) in iTS-P YAP9 cells was significantly lower compared with those in ES cells. The iTS-P YAP9 cells efficiently differentiated into insulin-expressing cells compared with ES cells. The ability to generate autologous iTS cells may be applied to diverse applications of regenerative medicine.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Proteínas de Sinalização YAP , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Transativadores/metabolismo , Transativadores/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 3 de Transcrição de Octâmero/genética
2.
J Biol Chem ; 300(1): 105535, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38072043

RESUMO

Renal cell carcinoma (RCC) is a frequent malignancy of the urinary system with high mortality and morbidity. However, the molecular mechanisms underlying RCC progression are still largely unknown. In this study, we identified FOXA2, a pioneer transcription factor, as a driver oncogene for RCC. We show that FOXA2 was commonly upregulated in human RCC samples and promoted RCC proliferation, as evidenced by assays of cell viability, colony formation, migratory and invasive capabilities, and stemness properties. Mechanistically, we found that FOXA2 promoted RCC cell proliferation by transcriptionally activating HIF2α expression in vitro and in vivo. Furthermore, we found that FOXA2 could interact with VHL (von Hippel‒Lindau), which ubiquitinated FOXA2 and controlled its protein stability in RCC cells. We showed that mutation of lysine at position 264 to arginine in FOXA2 could mostly abrogate its ubiquitination, augment its activation effect on HIF2α expression, and promote RCC proliferation in vitro and RCC progression in vivo. Importantly, elevated expression of FOXA2 in patients with RCC positively correlated with the expression of HIF2α and was associated with shorter overall and disease-free survival. Together, these findings reveal a novel role of FOXA2 in RCC development and provide insights into the underlying molecular mechanisms of FOXA2-driven pathological processes in RCC.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Carcinoma de Células Renais , Fator 3-beta Nuclear de Hepatócito , Neoplasias Renais , Humanos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Fatores de Transcrição/genética , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Progressão da Doença
3.
Biol Reprod ; 110(2): 246-260, 2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-37944068

RESUMO

Uterine glands and their secretions are crucial for conceptus survival and implantation in rodents and humans. In mice, the development of uterine gland known as adenogenesis occurs after birth, whereas the adenogenesis in humans initiates from fetal life and completed at puberty. Uterine adenogenesis involves dynamic epithelial cell proliferation, differentiation, and apoptosis. However, it is largely unexplored about the mechanisms governing adenogenesis. CK1α plays important roles in regulating cell division, differentiation, and death, but it is unknown whether CK1α affects adenogenesis. In the current study, uterus-specific CK1α knockout female mice (Csnk1a1d/d) were infertile resulted from lack of uterine glands. Subsequent analysis revealed that CK1α deletion induced massive apoptosis in uterine epithelium by activating GSK3ß, which was confirmed by injections of GSK3ß inhibitor SB216763 to Csnk1a1d/d females, and the co-treatment of SB216763 and CK1 inhibitor d4476 on cultured epithelial cells. Another important finding was that our results revealed CK1α deficiency activated p53, which then blocked the expression of Foxa2, an important factor for glandular epithelium development and function. This was confirmed by that Foxa2 expression level was elevated in p53 inhibitor pifithrin-α injected Csnk1a1d/d mouse uterus and in vitro dual-luciferase reporter assay between p53 and Foxa2. Collectively, these studies reveal that CK1α is a novel factor regulating uterine adenogenesis by inhibiting epithelial cell apoptosis through GSK3ß pathway and regulating Foxa2 expression through p53 pathway. Uncovering the mechanisms of uterine adenogenesis is expected to improve pregnancy success in humans and other mammals.


Assuntos
Indóis , Maleimidas , Proteína Supressora de Tumor p53 , Útero , Gravidez , Animais , Feminino , Camundongos , Humanos , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Útero/metabolismo , Apoptose , Células Epiteliais/metabolismo , Camundongos Knockout , Mamíferos/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo
4.
Environ Toxicol ; 39(2): 708-722, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37665156

RESUMO

BACKGROUND: Gallbladder cancer (GBC), a highly malignant gastrointestinal tumor, lacks effective therapies. Foxhead box A2 (FOXA2) is a tumor suppressor that is poorly expressed in various human malignancies. This study aimed to ascertain FOXA2 expression in GBC and its relevance to tumor metastasis, and to elucidate its regulatory mechanism with epithelial-mesenchymal transition (EMT) as an entry point, in the hope of providing a potential therapeutic target for GBC. METHODS: FOXA2 expression in GBC tissues was first detected using immunohistochemistry (IHC), followed by correlation analysis with clinicopathological characteristics and survival prognosis. Subsequently, the effects of FOXA2 on GBC cell migration and invasion, as well as EMT induction, were evaluated by scratch, Transwell, RT-PCR, and Western blot assays, together with animal experimentation. Ultimately, mRNA sequencing was carried out to identify the key downstream target genes of FOXA2 in controlling the EMT process in GBC cells, and dual-luciferase reporter and chromatin immunoprecipitation assays were used to determine its regulatory mechanism. RESULTS: FOXA2 was underexpressed in GBC tissues and inversely correlated with tumor node metastasis stage, lymph node metastasis, and poor patient prognosis. FOXA2 exerts suppressive effects on EMT and metastasis of GBC in vivo and in vitro. FOXA2 can impede GBC cell migratory and invasive functions and EMT by positively mediating serine protein kinase inhibitor B5 (SERPINB5) expression. CONCLUSION: FOXA2 directly binds to the SERPINB5 promoter region to stimulate its transcription, thereby modulating the migration and invasion behaviors of GBC cells as well as the EMT process, which might be an effective therapeutic target against GBC.


Assuntos
Neoplasias da Vesícula Biliar , Animais , Humanos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Transição Epitelial-Mesenquimal/genética , Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/patologia , Regulação Neoplásica da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo
5.
Virchows Arch ; 484(4): 709-713, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38141134

RESUMO

In the post-chemotherapy setting, germ cell tumors of the testis (GCTT) that resemble non-specific sarcomas and co-express cytokeratins and glypican-3 (GPC3) are diagnosed as "sarcomatoid yolk sac tumor postpubertal-type (YSTpt)". The diagnosis of sarcomatoid YSTpt is clinically relevant but challenging due to its rarity, non-specific histology, and negative α-fetoprotein (AFP) staining. Recently, FOXA2 has emerged as a key-gene in the reprogramming of GCTT (activating the transcription of several genes, among which GATA3), and immunohistochemical studies showed that GATA3 and FOXA2 have a higher sensitivity for non-sarcomatoid YSTpt than GPC3 and AFP. We found that sarcomatoid YSTpt did not express FOXA2 [0: 14/14 (100%)] and showed focal expression of GATA3 [0: 12/14 (85.7%), 1 + : 2/14 (14.3%)], thus suggesting that these markers are not useful in diagnosing this tumor. Furthermore, we proposed a potential mechanism of sarcomatoid transformation in the post-chemotherapy setting of GCTT, mediated by the downregulation of FOXA2 and GATA3.


Assuntos
Biomarcadores Tumorais , Regulação para Baixo , Tumor do Seio Endodérmico , Fator de Transcrição GATA3 , Fator 3-beta Nuclear de Hepatócito , Fenótipo , Neoplasias Testiculares , Fator de Transcrição GATA3/metabolismo , Fator de Transcrição GATA3/genética , Humanos , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Masculino , Neoplasias Testiculares/patologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Tumor do Seio Endodérmico/patologia , Tumor do Seio Endodérmico/genética , Tumor do Seio Endodérmico/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Imuno-Histoquímica , Glipicanas/genética , Glipicanas/metabolismo , Adulto , Sarcoma/genética , Sarcoma/patologia , Sarcoma/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Embrionárias de Células Germinativas/patologia , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/metabolismo , Adulto Jovem , Adolescente
6.
Biochim Biophys Acta Gen Subj ; 1868(1): 130500, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37914145

RESUMO

BACKGROUND: Excessive inflammation is the main cause of treatment failure in neonatal pneumonia (NP). CCCTC-binding factor (CTCF) represents an important node in various inflammatory diseases. In the present study, we tried to clarify the function and underlying molecular mechanism of CTCF on an in vitro cellular model of NP, which was generated by simulating the human lung fibroblast cell line WI-38 with lipopolysaccharide (LPS). METHODS: The SUMOylation level and protein interaction were verified by Co-immunoprecipitation assay. Cell viability was measured by Cell Counting Kit-8 assay. Inflammatory factors were examined by Enzyme-linked immunosorbent assay. Cell apoptosis was evaluated by TUNEL assay. The binding activity of CTCF to target promoter was tested by chromatin immunoprecipitation and luciferase reporter assay. RESULTS: LPS treatment restrained cell viability, promoted the production of inflammatory factors, and enhanced cell apoptosis. CTCF overexpression played anti-inflammatory and anti-apoptotic roles. Furthermore, CTCF was modified by SUMOylation with small ubiquitin-like modifier protein 1 (SUMO1). Interfering with sumo-specific protease 1 (SENP1) facilitated CTCF SUMOylation and protein stability, thus suppressing LPS-evoked inflammatory and apoptotic injuries. Moreover, CTCF could bind to the forkhead box protein A2 (FOXA2) promoter region to promote FOXA2 expression. The anti-inflammatory and anti-apoptotic roles of CTCF are associated with FOXA2 activation. In addition, SENP1 knockdown increased FOXA2 expression by enhancing the abundance and binding ability of CTCF. CONCLUSIONS: SUMOylation of CTCF by SENP1 knockdown enhanced its protein stability and binding ability and it further alleviated LPS-evoked inflammatory injury in human lung fibroblasts by positively regulating FOXA2 transcription.


Assuntos
Lipopolissacarídeos , Peptídeo Hidrolases , Recém-Nascido , Humanos , Peptídeo Hidrolases/metabolismo , Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Sumoilação , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Fibroblastos/metabolismo , Pulmão/metabolismo , Anti-Inflamatórios , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo
7.
Biochim Biophys Acta Mol Cell Res ; 1871(2): 119655, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38135007

RESUMO

Farnesoid X receptor (FXR) is a nuclear ligand-activated receptor of bile acids that plays a role in the modulation of insulin content. However, the underlying molecular mechanisms remain unclear. Forkhead box a2 (Foxa2) is an important nuclear transcription factor in pancreatic ß-cells and is involved in ß-cell function. We aimed to explore the signaling mechanism downstream of FXR to regulate insulin content and underscore its association with Foxa2 and insulin gene (Ins) transcription. All experiments were conducted on FXR transgenic mice, INS-1 823/13 cells, and diabetic Goto-Kakizaki (GK) rats undergoing sham or Roux-en-Y gastric bypass (RYGB) surgery. Islets from FXR knockout mice and INS-1823/13 cells with FXR knockdown exhibited substantially lower insulin levels than that of controls. This was accompanied by decreased Foxa2 expression and Ins transcription. Conversely, FXR overexpression increased insulin content, concomitant with enhanced Foxa2 expression and Ins transcription in INS-1 823/13 cells. Moreover, FXR knockdown reduced FXR recruitment and H3K27 trimethylation in the Foxa2 promoter. Importantly, Foxa2 overexpression abrogated the adverse effects of FXR knockdown on Ins transcription and insulin content in INS-1 823/13 cells. Notably, RYGB surgery led to improved insulin content in diabetic GK rats, which was accompanied by upregulated FXR and Foxa2 expression and Ins transcription. Collectively, these data suggest that Foxa2 serves as the target gene of FXR in ß-cells and mediates FXR-enhanced Ins transcription. Additionally, the upregulated FXR/Foxa2 signaling cascade could contribute to the enhanced insulin content in diabetic GK rats after RYGB.


Assuntos
Diabetes Mellitus , Insulina , Camundongos , Ratos , Animais , Insulina/metabolismo , Fatores de Transcrição/metabolismo , Regulação da Expressão Gênica , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo
8.
Elife ; 122023 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-37861292

RESUMO

Millions suffer from incurable lung diseases, and the donor lung shortage hampers organ transplants. Generating the whole organ in conjunction with the thymus is a significant milestone for organ transplantation because the thymus is the central organ to educate immune cells. Using lineage-tracing mice and human pluripotent stem cell (PSC)-derived lung-directed differentiation, we revealed that gastrulating Foxa2 lineage contributed to both lung mesenchyme and epithelium formation. Interestingly, Foxa2 lineage-derived cells in the lung mesenchyme progressively increased and occupied more than half of the mesenchyme niche, including endothelial cells, during lung development. Foxa2 promoter-driven, conditional Fgfr2 gene depletion caused the lung and thymus agenesis phenotype in mice. Wild-type donor mouse PSCs injected into their blastocysts rescued this phenotype by complementing the Fgfr2-defective niche in the lung epithelium and mesenchyme and thymic epithelium. Donor cell is shown to replace the entire lung epithelial and robust mesenchymal niche during lung development, efficiently complementing the nearly entire lung niche. Importantly, those mice survived until adulthood with normal lung function. These results suggest that our Foxa2 lineage-based model is unique for the progressive mobilization of donor cells into both epithelial and mesenchymal lung niches and thymus generation, which can provide critical insights into studying lung transplantation post-transplantation shortly.


Assuntos
Células Endoteliais , Células-Tronco Pluripotentes , Camundongos , Humanos , Animais , Adulto , Células-Tronco Pluripotentes/metabolismo , Diferenciação Celular , Pulmão , Blastocisto/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo
9.
Exp Cell Res ; 426(1): 113539, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36889571

RESUMO

BACKGROUND: Endometriosis is a severe disease which is associated with excessive activation of pyroptosis. Our present research aimed to investigate the function of Forkhead Box A2 (FoxA2) in regulating pyroptosis in endometriosis. METHODS: IL-1ß and IL-18 concentrations were assessed using ELISA. Cell pyroptosis was analyzed using flow cytometry. TUNEL staining was performed to determine human endometrial stromal cells (HESC) death. Moreover, ERß mRNA stability was assessed using RNA degradation assay. Finally, the binding relationships between FoxA2, IGF2BP1 and ERß were verified by dual-luciferase reporter system, ChIP, RIP and RNA pull-down assays. RESULTS: Our results revealed that IGF2BP1 and ERß were significantly upregulated in ectopic endometrium (EC) tissues of endometriosis patients compared to that in eutopic endometrium (EU) tissues as well as IL-18 and IL-1ß levels. Loss-of-function experiments subsequently demonstrated that either IGF2BP1 knockdown or ERß knockdown could repress HESC pyroptosis. In addition, IGF2BP1 upregulation promoted the pyroptosis in endometriosis by binding to ERß and promoting ERß mRNA stability. Our further research displayed that FoxA2 upregulation suppressed HESC pyroptosis by interacting with IGF2BP1 promoter. CONCLUSION: Our research proved that FoxA2 upregulation downregulated ERß by transcriptionally inhibiting IGF2BP1, thereby repressing pyroptosis in endometriosis.


Assuntos
Endometriose , Feminino , Humanos , Endometriose/genética , Endometriose/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Piroptose/genética , Interleucina-18/metabolismo , Endométrio , Células Estromais/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo
10.
Clin Exp Pharmacol Physiol ; 50(7): 561-572, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36946190

RESUMO

Aerobic glycolysis is critical for the energy metabolism of cancer cells. This study focuses on the regulation of forkhead box A2 (FOXA2) on pyruvate kinase M2 (PKM2) and their effects on the glycolytic activity and malignant phenotype of thyroid carcinoma (THCA) cells. By analysing four Gene Expression Omnibus datasets and querying bioinformatics systems, we obtained FOXA2 as a poorly expressed transcription factor in THCA. Later, we validated decreased mRNA and protein levels of FOXA2 in THCA cells by quantitative polymerase chain reaction and western blot assays. FOXA2 upregulation in THCA cells suppressed the glucose uptake and lactate production, and it reduced the extracellular acidification rate, but increased the oxygen consumption rate of cells. Meanwhile, the FOXA2 overexpression blocked the proliferation and mobility, and the tumourigenic activity of cancer cells. The chromatin immunoprecipitation and luciferase assays showed that FOXA2 bound to PKM2 promoter and suppressed the transcription of PKM2, which was highly expressed in THCA cells. Further upregulation of PKM2 elevated the ß-catenin, c-Myc and cyclin D1 levels and restored the glycolytic activity as well as the malignant properties of cancer cells. Collectively, this work reveals that FOXA2 suppresses aerobic glycolysis and progression of THCA by blocking PKM2 transcription and inactivating the Wnt/ß-catenin pathway.


Assuntos
Neoplasias da Glândula Tireoide , beta Catenina , Humanos , beta Catenina/genética , beta Catenina/metabolismo , Via de Sinalização Wnt/genética , Regulação para Cima , Neoplasias da Glândula Tireoide/genética , Glicólise/genética , Proliferação de Células/genética , Linhagem Celular Tumoral , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo
11.
Biol Trace Elem Res ; 201(11): 5201-5212, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36689145

RESUMO

Arsenic (AS) is a metalloid element that widely exists and can cause different degrees of liver damage. The molecular mechanism of arsenic-induced liver injury has yet to be fully elucidated. Clinically, glutathione (GSH) is often used as an antidote for heavy metal poisoning and hepatoprotective drugs. However, the hepatoprotective effect of glutathione remains unknown in arsenic-induced liver injury. The regulatory relationship between Foxa2 and XIAP may play an important role in mitochondrial survival and death. Therefore, we took Foxa2-XIAP as the axis to explore the protective mechanism of GSH. In this study, we first established a mouse model of chronic arsenic exposure and examined liver function as reflected by quantitative parameters such as aspartate aminotransferase and alanine aminotransferase. Also, redox parameters in the liver were measured, including malondialdehyde, superoxide dismutase, 8-hydroxy-2'-deoxyguanosin, and glutathione peroxidase. RT-qPCR and western-blotting were used to detect the levels of related genes and proteins, such as Foxa2, XIAP, Smac, Bax, Bcl2, Caspase9, and Caspase3. Subsequently, GSH was administered at the same time as high arsenic exposure, and changes in the above parameters were observed. After a comprehensive analysis of the above results, we demonstrate that GSH treatment alleviates arsenic-induced oxidative stress and inhibits the mitochondrial pathway of apoptosis, which can be regulated through the Foxa2 and XIAP axis. The present study would be helpful in elucidating the molecular mechanism of arsenic-induced liver injury and identifying a new potential therapeutic target. And we also provided new theoretical support for glutathione in the treatment of liver damage caused by arsenic.


Assuntos
Arsênio , Doença Hepática Crônica Induzida por Substâncias e Drogas , Hepatopatias , Camundongos , Animais , Arsênio/metabolismo , Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Estresse Oxidativo , Fígado/metabolismo , Glutationa/metabolismo , Apoptose , Hepatopatias/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/farmacologia
12.
Biol Reprod ; 108(3): 359-362, 2023 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-36611017

RESUMO

Forkhead box protein A2 (FOXA2) is a pioneer transcription factor important for epithelial budding and morphogenesis in different organs. It has been used as a specific marker for uterine glandular epithelial cells (GE). FOXA2 has close interactions with estrogen receptor α (ERα). ERα binding to Foxa2 gene in the uterus indicates its regulation of Foxa2. The intimate interactions between ERα and FOXA2 and their essential roles in early pregnancy led us to investigate the expression of FOXA2 in the female reproductive tract of pre-implantation epiERα-/- (Esr1fl/flWnt7aCre/+) mice, in which ERα is conditionally deleted in the epithelium of reproductive tract. In the oviduct, FOXA2 is detected in the ciliated epithelial cells of ampulla but absent in the isthmus of day 3.5 post-coitum (D3.5) Esr1fl/fl control and epiERα-/- mice. In the uterus, FOXA2 expression in the GE appears to be comparable between Esr1fl/fl and epiERα-/- mice. However, FOXA2 is upregulated in the D0.5 and D3.5 but not PND25-28 epiERα-/- uterine luminal epithelial cells (LE). In the vagina, FOXA2 expression is low in the basal layer and increases toward the superficial layer of the D3.5 Esr1fl/fl vaginal epithelium, but FOXA2 is detected in the basal, intermediate, and superficial layers, with the strongest FOXA2 expression in the intermediate layers of the D3.5 epiERα-/- vaginal epithelium. This study demonstrates that loss of ERα in LE and vaginal basal layer upregulates FOXA2 expression in these epithelial cells during early pregnancy. The mechanisms for epithelial cell-type specific regulation of FOXA2 by ERα remain to be elucidated.


Assuntos
Receptor alfa de Estrogênio , Útero , Animais , Feminino , Camundongos , Gravidez , Epitélio/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Regulação para Cima , Útero/metabolismo , Vagina/metabolismo
14.
Gut ; 72(3): 549-559, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35444014

RESUMO

OBJECTIVE: Multidrug resistance protein 2 (MRP2) is a bottleneck in bilirubin excretion. Its loss is sufficient to induce hyperbilirubinaemia, a prevailing characteristic of acute liver failure (ALF) that is closely associated with clinical outcome. This study scrutinises the transcriptional regulation of MRP2 under different pathophysiological conditions. DESIGN: Hepatic MRP2, farnesoid X receptor (FXR) and Forkhead box A2 (FOXA2) expression and clinicopathologic associations were examined by immunohistochemistry in 14 patients with cirrhosis and 22 patients with ALF. MRP2 regulatory mechanisms were investigated in primary hepatocytes, Fxr -/- mice and lipopolysaccharide (LPS)-treated mice. RESULTS: Physiologically, homeostatic MRP2 transcription is mediated by the nuclear receptor FXR/retinoid X receptor complex. Fxr-/- mice lack apical MRP2 expression and rapidly progress into hyperbilirubinaemia. In patients with ALF, hepatic FXR expression is undetectable, however, patients without infection maintain apical MRP2 expression and do not suffer from hyperbilirubinaemia. These patients express FOXA2 in hepatocytes. FOXA2 upregulates MRP2 transcription through binding to its promoter. Physiologically, nuclear FOXA2 translocation is inhibited by insulin. In ALF, high levels of glucagon and tumour necrosis factor α induce FOXA2 expression and nuclear translocation in hepatocytes. Impressively, ALF patients with sepsis express low levels of FOXA2, lose MRP2 expression and develop severe hyperbilirubinaemia. In this case, LPS inhibits FXR expression, induces FOXA2 nuclear exclusion and thus abrogates the compensatory MRP2 upregulation. In both Fxr -/- and LPS-treated mice, ectopic FOXA2 expression restored apical MRP2 expression and normalised serum bilirubin levels. CONCLUSION: FOXA2 replaces FXR to maintain MRP2 expression in ALF without sepsis. Ectopic FOXA2 expression to maintain MRP2 represents a potential strategy to prevent hyperbilirubinaemia in septic ALF.


Assuntos
Fator 3-beta Nuclear de Hepatócito , Falência Hepática Aguda , Proteína 2 Associada à Farmacorresistência Múltipla , Animais , Camundongos , Bilirrubina , Fator 3-beta Nuclear de Hepatócito/metabolismo , Hepatócitos/metabolismo , Hiperbilirrubinemia/metabolismo , Hiperbilirrubinemia/patologia , Lipopolissacarídeos/metabolismo , Fígado/metabolismo , Falência Hepática Aguda/metabolismo , Proteína 2 Associada à Farmacorresistência Múltipla/metabolismo , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
15.
Cancer Cell ; 40(11): 1306-1323.e8, 2022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-36332622

RESUMO

Prostate cancer adeno-to-neuroendocrine lineage transition has emerged as a mechanism of targeted therapeutic resistance. Identifying the direct molecular drivers and developing pharmacological strategies using clinical-grade inhibitors to overcome lineage transition-induced therapeutic resistance are imperative. Here, using single-cell multiomics analyses, we investigate the dynamics of cellular heterogeneity, transcriptome regulation, and microenvironmental factors in 107,201 cells from genetically engineered mouse prostate cancer samples with complete time series of tumor evolution seen in patients. We identify that FOXA2 orchestrates prostate cancer adeno-to-neuroendocrine lineage transition and that Foxa2 expression is significantly induced by androgen deprivation. Moreover, Foxa2 knockdown induces the reversal of adeno-to-neuroendocrine transition. The KIT pathway is directly regulated by FOXA2 and specifically activated in neuroendocrine prostate cancer (NEPC). Pharmacologic inhibition of KIT pathway significantly suppresses mouse and human NEPC tumor growth. These findings reveal that FOXA2 drives adeno-to-neuroendocrine lineage plasticity in prostate cancer and provides a potential pharmacological strategy for castration-resistant NEPC.


Assuntos
Carcinoma Neuroendócrino , Neoplasias da Próstata , Masculino , Humanos , Camundongos , Animais , Neoplasias da Próstata/patologia , Antagonistas de Androgênios/uso terapêutico , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Carcinoma Neuroendócrino/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo
16.
Metabolism ; 136: 155292, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35995281

RESUMO

Mitochondrial FAM3A has been revealed to be a viable target for treating diabetes and nonalcoholic fatty liver disease (NAFLD). However, its distinct mechanism in ameliorating hepatic steatosis remained unrevealed. High-throughput RNA sequencing revealed that carnitine palmityl transferase 2 (CPT2), one of the key enzymes for lipid oxidation, is the downstream molecule of FAM3A signaling pathway in hepatocytes. Intensive study demonstrated that FAM3A-induced ATP release activated P2 receptor to promote the translocation of calmodulin (CaM) from cytoplasm into nucleus, where it functioned as a co-activator of forkhead box protein A2 (FOXA2) to promote the transcription of CPT2, increasing free fatty acid oxidation and reducing lipid deposition in hepatocytes. Furthermore, antidepressant imipramine activated FAM3A-ATP-P2 receptor-CaM-FOXA2-CPT2 pathway to reduce lipid deposition in hepatocytes. In FAM3A-deficient hepatocytes, imipramine failed to activate CaM-FOXA2-CPT2 axis to increase lipid oxidation. Imipramine administration significantly ameliorated hepatic steatosis, hyperglycemia and obesity of obese mice mainly by activating FAM3A-ATP-CaM-FOXA2-CPT2 pathway in liver and thermogenesis in brown adipose tissue (BAT). In FAM3A-deficient mice fed on high-fat-diet, imipramine treatment failed to correct the dysregulated lipid and glucose metabolism, and activate thermogenesis in BAT. In conclusion, imipramine activates FAM3A-ATP-CaM-FOXA2-CPT2 pathway to ameliorate steatosis. For depressive patients complicated with metabolic disorders, imipramine may be recommended in priority as antidepressive drug.


Assuntos
Imipramina , Hepatopatia Gordurosa não Alcoólica , Trifosfato de Adenosina/metabolismo , Animais , Calmodulina/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Citocinas/metabolismo , Dieta Hiperlipídica , Ácidos Graxos não Esterificados/metabolismo , Glucose/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Imipramina/farmacologia , Imipramina/uso terapêutico , Metabolismo dos Lipídeos , Fígado/metabolismo , Camundongos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo
17.
Biochem Biophys Res Commun ; 625: 16-22, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35944359

RESUMO

Esophageal cancer is one of the most common malignant tumors worldwide and half of the patients present tumor metastasis at initial diagnosis. CXCR4 has been reported to be upregulated in esophageal cancer tissues and associated with tumor metastasis. However, the upstream transcriptional regulator of CXCR4 in esophageal cancer is still unclear. In this study, we found that transcription factor FOXA2 directly bound on the CXCR4 promoter region and activated its expression in esophageal cancer cells. The expression of FOXA2 was upregulated in esophageal cancer tissues and positively correlated with CXCR4. Moreover, knockout of FOXA2 significantly inhibited esophageal cancer migration and metastasis, which can be rescued by ectopically expressed CXCR4. Taken together, we reveal a novel transcriptional activator of CXCR4 in esophageal cancer, which might provide new strategies for esophageal cancer treatment.


Assuntos
Neoplasias Esofágicas , Receptores CXCR4 , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Esofágicas/patologia , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo
18.
Nat Commun ; 13(1): 3907, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35798741

RESUMO

Existing knowledge of the role of epigenetic modifiers in pancreas development has exponentially increased. However, the function of TET dioxygenases in pancreatic endocrine specification remains obscure. We set out to tackle this issue using a human embryonic stem cell (hESC) differentiation system, in which TET1/TET2/TET3 triple knockout cells display severe defects in pancreatic ß-cell specification. The integrative whole-genome analysis identifies unique cell-type-specific hypermethylated regions (hyper-DMRs) displaying reduced chromatin activity and remarkable enrichment of FOXA2, a pioneer transcription factor essential for pancreatic endoderm specification. Intriguingly, TET depletion leads to significant changes in FOXA2 binding at the pancreatic progenitor stage, in which gene loci with decreased FOXA2 binding feature low levels of active chromatin modifications and enriches for bHLH motifs. Transduction of full-length TET1 but not the TET1-catalytic-domain in TET-deficient cells effectively rescues ß-cell differentiation accompanied by restoring PAX4 hypomethylation. Taking these findings together with the defective generation of functional ß-cells upon TET1-inactivation, our study unveils an essential role of TET1-dependent demethylation in establishing ß-cell identity. Moreover, we discover a physical interaction between TET1 and FOXA2 in endodermal lineage intermediates, which provides a mechanistic clue regarding the complex crosstalk between TET dioxygenases and pioneer transcription factors in epigenetic regulation during pancreas specification.


Assuntos
Dioxigenases , Fator 3-beta Nuclear de Hepatócito , Oxigenases de Função Mista , Proteínas Proto-Oncogênicas , 5-Metilcitosina/metabolismo , Diferenciação Celular/genética , Cromatina/genética , Montagem e Desmontagem da Cromatina , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Epigênese Genética , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo
19.
Elife ; 112022 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-35861728

RESUMO

Embryonic diapause is a reproductive strategy in which embryo development and growth is temporarily arrested within the uterus to ensure the survival of neonates and mothers during unfavorable conditions. Pregnancy is reinitiated when conditions become favorable for neonatal survival. The mechanism of how the uterus enters diapause in various species remains unclear. Mice with uterine depletion of Foxa2, a transcription factor, are infertile. In this study, we show that dormant blastocysts are recovered from these mice on day 8 of pregnancy with persistent expression of uterine Msx1, a gene critical to maintaining the uterine quiescent state, suggesting that these mice enter embryonic diapause. Leukemia inhibitory factor (LIF) can resume implantation in these mice. Although estrogen is critical for implantation in progesterone-primed uterus, our current model reveals that FOXA2-independent estrogenic effects are detrimental to sustaining uterine quiescence. Interestingly, progesterone and anti-estrogen can prolong uterine quiescence in the absence of FOXA2. Although we find that Msx1 expression persists in the uterus deficient in Foxa2, the complex relationship of FOXA2 with Msx genes and estrogen receptors remains to be explored.


Assuntos
Diapausa , Progesterona , Animais , Blastocisto/metabolismo , Implantação do Embrião , Desenvolvimento Embrionário , Estrogênios/metabolismo , Feminino , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Camundongos , Gravidez , Progesterona/metabolismo , Útero/metabolismo
20.
J Clin Invest ; 132(12)2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35703180

RESUMO

FOXA2 encodes a transcription factor mutated in 10% of endometrial cancers (ECs), with a higher mutation rate in aggressive variants. FOXA2 has essential roles in embryonic and uterine development. However, FOXA2's role in EC is incompletely understood. Functional investigations using human and mouse EC cell lines revealed that FOXA2 controls endometrial epithelial gene expression programs regulating cell proliferation, adhesion, and endometrial-epithelial transition. In live animals, conditional inactivation of Foxa2 or Pten alone in endometrial epithelium did not result in ECs, but simultaneous inactivation of both genes resulted in lethal ECs with complete penetrance, establishing potent synergism between Foxa2 and PI3K signaling. Studies in tumor-derived cell lines and organoids highlighted additional invasion and cell growth phenotypes associated with malignant transformation and identified key mediators, including Myc and Cdh1. Transcriptome and cistrome analyses revealed that FOXA2 broadly controls gene expression programs through modification of enhancer activity in addition to regulating specific target genes, rationalizing its tumor suppressor functions. By integrating results from our cell lines, organoids, animal models, and patient data, our findings demonstrated that FOXA2 is an endometrial tumor suppressor associated with aggressive disease and with shared commonalities among its roles in endometrial function and carcinogenesis.


Assuntos
Neoplasias do Endométrio , Transição Epitelial-Mesenquimal , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Neoplasias do Endométrio/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Camundongos , Fosfatidilinositol 3-Quinases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA