Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
PLoS Biol ; 22(5): e3002621, 2024 May.
Article in English | MEDLINE | ID: mdl-38805565

ABSTRACT

Cholesterol metabolism is vital for multiple cancer progression, while how cholesterol affects lung, a low-cholesterol tissue, for cancer metastasis and the underlying mechanism remain unclear. In this study, we found that metastatic lung adenocarcinoma cells acquire cellular dehydrocholesterol and cholesterol by endogenous cholesterol biosynthesis, instead of uptake upon cholesterol treatment. Besides, we demonstrated that exogenous cholesterol functions as signaling molecule to induce FOXA3, a key transcription factor for lipid metabolism via GLI2. Subsequently, ChIP-seq analysis and molecular studies revealed that FOXA3 transcriptionally activated Hmgcs1, an essential enzyme of cholesterol biosynthesis, to induce endogenous dehydrocholesterol and cholesterol level for membrane composition change and cell migration. Conversely, FOXA3 knockdown or knockout blocked cholesterol biosynthesis and lung adenocarcinoma metastasis in mice. In addition, the potent FOXA3 inhibitor magnolol suppressed metastatic gene programs in lung adenocarcinoma patient-derived organoids (PDOs). Altogether, our findings shed light onto unique cholesterol metabolism and FOXA3 contribution to lung adenocarcinoma metastasis.


Subject(s)
Adenocarcinoma of Lung , Cholesterol , Disease Progression , Hepatocyte Nuclear Factor 3-gamma , Lung Neoplasms , Cholesterol/metabolism , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Animals , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Mice , Hepatocyte Nuclear Factor 3-gamma/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement
2.
Chin Med J (Engl) ; 137(11): 1351-1359, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38721807

ABSTRACT

BACKGROUND: The ability to generate functional hepatocytes without relying on donor liver organs holds significant therapeutic promise in the fields of regenerative medicine and potential liver disease treatments. Clustered regularly interspaced short palindromic repeats (CRISPR) activator (CRISPRa) is a powerful tool that can conveniently and efficiently activate the expression of multiple endogenous genes simultaneously, providing a new strategy for cell fate determination. The main purpose of this study is to explore the feasibility of applying CRISPRa for hepatocyte reprogramming and its application in the treatment of mouse liver fibrosis. METHOD: The differentiation of mouse embryonic fibroblasts (MEFs) into functional induced hepatocyte-like cells (iHeps) was achieved by utilizing the CRISPRa synergistic activation mediator (SAM) system, which drove the combined expression of three endogenous transcription factors- Gata4, Foxa3 , and Hnf1a -or alternatively, the expression of two transcription factors, Gata4 and Foxa3 . In vivo , we injected adeno-associated virus serotype 6 (AAV6) carrying the CRISPRa SAM system into liver fibrotic Col1a1-CreER ; Cas9fl/fl mice, effectively activating the expression of endogenous Gata4 and Foxa3 in fibroblasts. The endogenous transcriptional activation of genes was confirmed using real-time quantitative polymerase chain reaction (RT-qPCR) and RNA-seq, and the morphology and characteristics of the induced hepatocytes were observed through microscopy. The level of hepatocyte reprogramming in vivo is detected by immunofluorescence staining, while the improvement of liver fibrosis is evaluated through Sirius red staining, alpha-smooth muscle actin (α-SMA) immunofluorescence staining, and blood alanine aminotransferase (ALT) examination. RESULTS: Activation of only two factors, Gata4 and Foxa3 , via CRISPRa was sufficient to successfully induce the transformation of MEFs into iHeps. These iHeps could be expanded in vitro and displayed functional characteristics similar to those of mature hepatocytes, such as drug metabolism and glycogen storage. Additionally, AAV6-based delivery of the CRISPRa SAM system effectively induced the hepatic reprogramming from fibroblasts in mice with live fibrosis. After 8 weeks of induction, the reprogrammed hepatocytes comprised 0.87% of the total hepatocyte population in the mice, significantly reducing liver fibrosis. CONCLUSION: CRISPRa-induced hepatocyte reprogramming may be a promising strategy for generating functional hepatocytes and treating liver fibrosis caused by hepatic diseases.


Subject(s)
Fibroblasts , GATA4 Transcription Factor , Hepatocyte Nuclear Factor 3-gamma , Hepatocytes , Animals , Mice , Hepatocyte Nuclear Factor 3-gamma/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , GATA4 Transcription Factor/metabolism , GATA4 Transcription Factor/genetics , Fibroblasts/metabolism , Hepatocytes/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cellular Reprogramming/physiology , Cellular Reprogramming/genetics , Cell Differentiation/physiology , Cell Differentiation/genetics , Cells, Cultured
3.
J Lipid Res ; 65(4): 100527, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38447926

ABSTRACT

Forkhead transcription factor 3 (FOXA3) has been shown to regulate metabolism and development. Hepatic FOXA3 is reduced in obesity and fatty liver disease. However, the role of hepatic FOXA3 in regulating obesity or steatohepatitis remains to be investigated. In this work, C57BL/6 mice were i.v. injected with AAV8-ALB-FOXA3 or the control virus. The mice were then fed a chow or Western diet for 16 weeks. The role of hepatic FOXA3 in energy metabolism and steatohepatitis was investigated. Plasma bile acid composition and the role of Takeda G protein-coupled receptor 5 (TGR5) in mediating the metabolic effects of FOXA3 were determined. Overexpression of hepatic FOXA3 reduced hepatic steatosis in chow-fed mice and attenuated Western diet-induced obesity and steatohepatitis. FOXA3 induced lipolysis and inhibited hepatic genes involved in bile acid uptake, resulting in elevated plasma bile acids. The beneficial effects of hepatic FOXA3 overexpression on Western diet-induced obesity and steatohepatitis were abolished in Tgr5-/- mice. Our data demonstrate that overexpression of hepatic FOXA3 prevents Western diet-induced obesity and steatohepatitis via activation of TGR5.


Subject(s)
Diet, Western , Hepatocyte Nuclear Factor 3-gamma , Liver , Mice, Inbred C57BL , Obesity , Receptors, G-Protein-Coupled , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Obesity/metabolism , Obesity/genetics , Obesity/etiology , Mice , Hepatocyte Nuclear Factor 3-gamma/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Liver/metabolism , Diet, Western/adverse effects , Male , Fatty Liver/metabolism , Fatty Liver/genetics , Fatty Liver/etiology , Bile Acids and Salts/metabolism
4.
Food Funct ; 14(23): 10591-10604, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37955610

ABSTRACT

Ergothioneine (EGT) is a bioactive compound derived from certain edible mushrooms. The activation of hepatic stellate cells (HSCs) is critically involved in the etiology of liver fibrosis (LF). Here, we report that in LX-2 HSCs, EGT upregulates the expression of Hint1 and Smad7 and suppresses their activation provoked by TGFß1. The EGT-triggered inhibition of HSC activation is abolished by knocking down the expression of Hint1. Overexpression of Hint1 increases Smad7 and represses TGFß1-provoked activation of LX-2 HSCs. In silico predictions unveiled that in the promoter region of the human Hint1 gene, there are two conserved cis-acting elements that have the potential to interact with the transcription factor Foxa3 termed hFBS1 and hFBS2, respectively. The knockdown of Foxa3 obviously declined Hint1 expression at both mRNA and protein levels. Transfection of Foxa3 or EGT treatment increased the activity of the luciferase reporter driven by the Hint1 promoter in an hFBS2-dependent manner. The knockdown of Foxa3 eliminated EGT-mediated upregulation of Hint1 promoter activity. Additionally, EGT triggered the nuclear translocation of Foxa3 without obviously affecting its expression level. Molecular docking analysis showed that EGT has the potential to directly interact with the Foxa3 protein. Moreover, Foxa3 played a critical role in EGT-mediated hepatoprotection. EGT modulated the Foxa3/Hint1/Smad7 signaling in mouse primary HSCs and inhibited their activation. The gavage of EGT considerably relieved CCl4-induced LF in mice. Our data provide new insights into the anti-LF activity of EGT. Mechanistically, EGT triggers the nuclear translocation of Foxa3 in HSCs, which promotes Hint1 transcription and subsequently elevates Smad7.


Subject(s)
Ergothioneine , Mice , Humans , Animals , Ergothioneine/pharmacology , Hepatic Stellate Cells/metabolism , Molecular Docking Simulation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/genetics , Gene Expression Regulation , Nerve Tissue Proteins/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/metabolism
5.
Exp Mol Med ; 54(6): 848-860, 2022 06.
Article in English | MEDLINE | ID: mdl-35764883

ABSTRACT

Growing evidence has revealed that hypoxia is involved in multiple stages of cancer development. However, there are limited reports on the effects of long noncoding RNAs (lncRNAs) on hepatocellular carcinoma (HCC) progression under hypoxia. The main purposes of this study were to analyze the effect of the novel lncRNA DACT3-AS1 on metastasis in HCC and to elucidate the related molecular mechanism. Bioinformatics tools were employed. RT-qPCR or western blot assays were conducted to detect RNA or protein expression. Clinical samples and in vivo assays were utilized to reveal the role of DACT3-AS1 in HCC. Other mechanism and functional analyses were specifically designed and performed as well. Based on the collected data, this study revealed that HIF-1α transcriptionally activates DACT3-AS1 expression under hypoxia. DACT3-AS1 was verified to promote metastasis in HCC. Mechanistically, DACT3-AS1 promotes the interaction between HDAC2 and FOXA3 to stimulate FOXA3 deacetylation, which consequently downregulates the FOXA3 protein. Furthermore, FOXA3 serves as a transcription factor that can bind to the PKM2 promoter region, thus hindering PKM2 expression. To summarize, this study uncovered that HIF-1α-induced DACT3-AS1 promotes metastasis in HCC and can upregulate PKM2 via the HDAC2/FOXA3 pathway in HCC cells.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/metabolism , Histone Deacetylase 2/genetics , Humans , Hypoxia , Liver Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
6.
Nephrol Dial Transplant ; 37(10): 1833-1843, 2022 09 22.
Article in English | MEDLINE | ID: mdl-34473308

ABSTRACT

BACKGROUND: Congenital anomalies of the kidneys and urinary tract (CAKUT) constitute the most common cause of chronic kidney disease in the first three decades of life. Variants in four Forkhead box (FOX) transcription factors have been associated with CAKUT. We hypothesized that other FOX genes, if highly expressed in developing kidneys, may also represent monogenic causes of CAKUT. METHODS: We here performed whole-exome sequencing (WES) in 541 families with CAKUT and generated four lists of CAKUT candidate genes: (A) 36 FOX genes showing high expression during renal development, (B) 4 FOX genes known to cause CAKUT to validate list A, (C) 80 genes that we identified as unique potential novel CAKUT candidate genes when performing WES in 541 CAKUT families and (D) 175 genes identified from WES as multiple potential novel CAKUT candidate genes. RESULTS: To prioritize potential novel CAKUT candidates in the FOX gene family, we overlapped 36 FOX genes (list A) with lists C and D of WES-derived CAKUT candidates. Intersection with list C identified a de novo FOXL2 in-frame deletion in a patient with eyelid abnormalities and ureteropelvic junction obstruction, and a homozygous FOXA2 missense variant in a patient with horseshoe kidney. Intersection with list D identified a heterozygous FOXA3 missense variant in a CAKUT family with multiple affected individuals. CONCLUSIONS: We hereby identified FOXL2, FOXA2 and FOXA3 as novel monogenic candidate genes of CAKUT, supporting the utility of a paralog-based approach to discover mutated genes associated with human disease.


Subject(s)
Urinary Tract , Urogenital Abnormalities , Forkhead Box Protein L2/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Humans , Kidney/abnormalities , Urinary Tract/abnormalities , Urogenital Abnormalities/genetics , Vesico-Ureteral Reflux , Exome Sequencing
7.
J Clin Lab Anal ; 35(3): e23686, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33368532

ABSTRACT

OBJECTIVE: In this research paper, we aimed to study the role of FOXA3 in hepatoblastoma (HB) and the molecular mechanism. METHODS: Immunohistochemistry was applied to determine the expression situation of FOXA3 and AFP in HB tissues and the adjacent normal tissues. FOXA3, HNF1A, and ZFHX3 expressions in HB tissues and the normal tissues were measured by Western blot. HB cell lines were randomly divided into 4 groups: Model, si-NC, si-FOXA3-1, and si-FOXA3-2 group. The HB cell viability and colony formation characteristics in the 4 groups were explored by CCK-8 and cell cloning formation assay, respectively. The expression of FOXA3, AFP, HNF1A, ZFHX3, and MYC in HB cells after knockdown of FOXA3 was measured. RESULTS: FOXA3, AFP, and HNF1A expressions were significantly up-regulated in HB tissues, while ZFHX3 expression was down-regulated. Knockdown of FOXA3 markedly inhibited HB cell viability and cloning formation ability. Knockdown of FOXA3 decreased FOXA3, AFP, and HNF1A/MYC expression, while increased ZFHX3 expression. CONCLUSION: FOXA3 promotes the occurrence and development of HB by up-regulating AFP and HNF1A/MYC expression, and down-regulating ZFHX3 expression.


Subject(s)
Hepatoblastoma/pathology , Hepatocyte Nuclear Factor 3-gamma/metabolism , Liver Neoplasms/pathology , alpha-Fetoproteins/metabolism , Cell Survival , Child, Preschool , Female , Hepatoblastoma/genetics , Hepatoblastoma/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Homeodomain Proteins/metabolism , Humans , Infant , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male
8.
Nat Commun ; 11(1): 5292, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087715

ABSTRACT

Recent advances have enabled the direct induction of human tissue-specific stem and progenitor cells from differentiated somatic cells. However, it is not known whether human hepatic progenitor cells (hHepPCs) can be generated from other cell types by direct lineage reprogramming with defined transcription factors. Here, we show that a set of three transcription factors, FOXA3, HNF1A, and HNF6, can induce human umbilical vein endothelial cells to directly acquire the properties of hHepPCs. These induced hHepPCs (hiHepPCs) propagate in long-term monolayer culture and differentiate into functional hepatocytes and cholangiocytes by forming cell aggregates and cystic epithelial spheroids, respectively, under three-dimensional culture conditions. After transplantation, hiHepPC-derived hepatocytes and cholangiocytes reconstitute damaged liver tissues and support hepatic function. The defined transcription factors also induce hiHepPCs from endothelial cells circulating in adult human peripheral blood. These expandable and bipotential hiHepPCs may be useful in the study and treatment of human liver diseases.


Subject(s)
Cellular Reprogramming Techniques/methods , Endothelial Cells/cytology , Hepatocytes/cytology , Stem Cells/cytology , Animals , Bile Ducts/cytology , Bile Ducts/physiology , Cell Aggregation , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Endothelial Cells/physiology , Female , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/physiology , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/physiology , Hepatocyte Nuclear Factor 6/genetics , Hepatocyte Nuclear Factor 6/physiology , Hepatocytes/physiology , Hepatocytes/transplantation , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Spheroids, Cellular/cytology , Spheroids, Cellular/physiology , Stem Cells/physiology
9.
Mol Cell ; 79(4): 660-676.e8, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32755593

ABSTRACT

Specific combinations of two transcription factors (Hnf4α plus Foxa1, Foxa2, or Foxa3) can induce direct conversion of mouse fibroblasts into hepatocyte-like cells. However, the molecular mechanisms underlying hepatic reprogramming are largely unknown. Here, we show that the Foxa protein family members and Hnf4α sequentially and cooperatively bind to chromatin to activate liver-specific gene expression. Although all Foxa proteins bind to and open regions of closed chromatin as pioneer factors, Foxa3 has the unique potential of transferring from the distal to proximal regions of the transcription start site of target genes, binding RNA polymerase II, and co-traversing target genes. These distinctive characteristics of Foxa3 are essential for inducing the hepatic fate in fibroblasts. Similar functional coupling of transcription factors to RNA polymerase II may occur in other contexts whereby transcriptional activation can induce cell differentiation.


Subject(s)
Hepatocyte Nuclear Factor 3-gamma/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Liver/cytology , Liver/physiology , Transcriptional Activation , Animals , Binding Sites , Cells, Cultured , Cellular Reprogramming/physiology , Chromatin/metabolism , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 4/genetics , Mice, Inbred C57BL , Protein Domains , Transcription Initiation Site
10.
Genes Dev ; 34(15-16): 1039-1050, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32561546

ABSTRACT

The FoxA transcription factors are critical for liver development through their pioneering activity, which initiates a highly complex regulatory network thought to become progressively resistant to the loss of any individual hepatic transcription factor via mutual redundancy. To investigate the dispensability of FoxA factors for maintaining this regulatory network, we ablated all FoxA genes in the adult mouse liver. Remarkably, loss of FoxA caused rapid and massive reduction in the expression of critical liver genes. Activity of these genes was reduced back to the low levels of the fetal prehepatic endoderm stage, leading to necrosis and lethality within days. Mechanistically, we found FoxA proteins to be required for maintaining enhancer activity, chromatin accessibility, nucleosome positioning, and binding of HNF4α. Thus, the FoxA factors act continuously, guarding hepatic enhancer activity throughout adult life.


Subject(s)
Forkhead Transcription Factors/physiology , Gene Regulatory Networks , Liver/metabolism , Animals , Binding Sites , Chromatin/metabolism , Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 4/metabolism , Liver/pathology , Liver Failure/etiology , Liver Failure/pathology , Male , Mice , Nucleosomes
11.
Toxins (Basel) ; 12(3)2020 03 22.
Article in English | MEDLINE | ID: mdl-32235689

ABSTRACT

Bee venom phospholipase A2 is a lipolytic enzyme in bee venom that catalyzes hydrolysis of the sn-2 ester bond of membrane phospholipids to produce free fatty acid and lysophospholipids. Current evidence suggests that bee venom phospholipase A2 (bvPLA2) induces regulatory T cell expansion and attenuates several immune system-related diseases, including Alzheimer's disease. The induction of Treg cells is directly mediated by binding to mannose receptors on dendritic cells. This interaction induces the PGE2-EP2 signaling pathway, which promotes Treg induction in CD4+ T cells. In this study, we investigated the effects of bvPLA2 treatment on the apoptotic signaling pathway in Treg populations. Flow cytometry was performed to identify early apoptotic cells. As a result, early apoptotic cells were dramatically decreased in bvPLA2-treated splenocytes, whereas rapamycin-treated cells showed levels of apoptotic cells similar to those of PBS-treated cells. Furthermore, bvPLA2 treatment increased expression of anti-apoptotic molecules including CTLA-4 and PD-1. The survival rate increased in bvPLA2-treated Tregs. Our findings indicate that bvPLA2-mediated modulation of apoptotic signaling is strongly associated with the Treg induction, which exhibits protective effects against various immune-related diseases. To our knowledge, this study is the first to demonstrate that bvPLA2 is the major bee venom (BV) compound capable of inducing Treg expansion through altering apoptotic signal.


Subject(s)
Apoptosis/drug effects , Bee Venoms/enzymology , Phospholipases A2/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis/immunology , Bee Venoms/pharmacology , CD4 Antigens/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Mice , Mice, Transgenic , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/pathology
12.
Int J Mol Sci ; 21(5)2020 Mar 05.
Article in English | MEDLINE | ID: mdl-32151057

ABSTRACT

Cholangiocarcinoma (CCA), a malignancy of biliary epithelium, is related to liver stem cell deregulation. FoxAs are a group of transcription factors that play critical roles in liver stem cell differentiation. In this study, the expression levels of FoxAs (i.e., FoxA1, FoxA2 and FoxA3) were detected in intrahepatic CCA tissues and the functions of FoxAs were studied in CCA cell lines. FoxA1 and FoxA2 were mainly localized in the nuclei of normal bile duct (NBD) cells and some of the cancer cells. Low expression of FoxA1 in CCA tissues (72%) was significantly correlated with poor prognosis. FoxA3 expression of CCA cells was localized in the nucleus and cytoplasm, whereas it was slightly detected in NBDs. High expression of FoxA3 in cancer tissues (61%) was significantly related to high metastasis status. These findings suggest the opposing roles of FoxA1 and FoxA3 in CCA. Moreover, the FoxA1-over-expressing CCA cell line exhibited a significant reduction in proliferative and invasive activities compared to control cells. Knockdown of FoxA3 in CCA cells resulted in a significant decrease in proliferative and invasive activities compared with control cells. Taken together, in CCA, FoxA1 is down-regulated and has tumor suppressive roles, whereas FoxA3 is up-regulated and has oncogenic roles.


Subject(s)
Bile Duct Neoplasms/pathology , Biomarkers, Tumor/metabolism , Cholangiocarcinoma/pathology , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-gamma/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Disease Progression , Female , Follow-Up Studies , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Humans , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Survival Rate , Tumor Cells, Cultured
13.
Dig Dis Sci ; 65(7): 2009-2023, 2020 07.
Article in English | MEDLINE | ID: mdl-31722057

ABSTRACT

BACKGROUND AND AIMS: Bioartificial livers (BALs) have attracted much attention as potential supportive therapies for liver diseases. A serum-free microcarrier culture strategy for the in vitro high-density expansion of human-induced hepatocyte-like cells (hiHeps) suitable for BALs was studied in this article. METHODS: hiHeps were transdifferentiated from human fibroblasts by the lentiviral overexpression of FOXA3, HNF1A, and HNF4A. Cells were cultured on microcarriers, their proliferation was evaluated by cell count and CCK-8 assays, and their function was evaluated by detecting liver function parameters in the supernatant, including urea secretion, albumin synthesis, and lactate dehydrogenase levels. The expressions of hepatocyte function-associated genes of hiHeps were measured by qRT-PCR in 2D and 3D conditions. The expression of related proteins during fibronectin promotes cell adhesion, and proliferation on microcarrier was detected by western blotting. RESULTS: During microcarrier culture, the optimal culture conditions during the adherence period were the use of half-volume high-density inoculation, Cytodex 3 at a concentration of 3 mg/mL, a cell seeding density of 2.0 × 105 cells/mL, and a stirring speed of 45 rpm. The final cell density in self-developed, chemically defined serum-free medium (SFM) reached 2.53 × 106 cells/mL, and the maximum increase in expansion was 12.61-fold. In addition, we found that fibronectin (FN) can promote hiHep attachment and proliferation on Cytodex 3 microcarriers and that this pro-proliferative effect was mediated by the integrin-ß1/FAK/ERK/CyclinD1 signaling pathway. Finally, the growth and function of hiHeps on Cytodex 3 in SFM were close to those of hiHeps on Cytodex 3 in hepatocyte maintenance medium (HMM), and cells maintained their morphology and function after harvest on microcarriers. CONCLUSIONS: Serum-free microcarrier culture has important implications for the expansion of a sufficient number of hiHeps prior to the clinical application of BALs.


Subject(s)
Cell Culture Techniques/methods , Cell Proliferation , Cell Transdifferentiation , Hepatocytes/cytology , Liver, Artificial , Albumins/biosynthesis , Cell Adhesion , Cellular Reprogramming Techniques/methods , Culture Media, Serum-Free , Cyclin D1/metabolism , Dextrans , Fibroblasts/cytology , Fibroblasts/metabolism , Fibronectins/metabolism , Focal Adhesion Kinase 1/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/metabolism , Hepatocytes/physiology , Humans , Integrin beta1/metabolism , L-Lactate Dehydrogenase/metabolism , MAP Kinase Signaling System , Reverse Transcriptase Polymerase Chain Reaction , Urea/metabolism
14.
Stem Cells ; 37(6): 803-812, 2019 06.
Article in English | MEDLINE | ID: mdl-30805989

ABSTRACT

Remodeling of the gene regulatory network in cells is believed to be a prerequisite for their lineage reprogramming. However, its key regulatory factors are not yet elucidated. In this article, we investigate the role of PIWI proteins and provide evidence that one of them, MIWI2, is elicited during transdifferentiation of fibroblasts into hepatocyte-like cells. In coincidence with the peak expression of MIWI2, we identified the appearance of a unique intermediate epigenetic state characterized by a specific Piwi-interacting RNA (piRNA) profile consisting of 219 novel sequences. Knockout of MIWI2 greatly improved the formation of the induced hepatocytes, whereas overexpression of exogenous MIWI2 completely abolished the stimulated effect. A bioinformatics analysis of piRNA interaction network, followed by experimental validation, revealed the Notch signaling pathway as one of the immediate effectors of MIWI2. Altogether, our results show for the first time that temporal expression of MIWI2 contributes negatively to cell plasticity not only in germline, but also in developed cells, such as mouse fibroblasts. Stem Cells 2019;37:803-812.


Subject(s)
Argonaute Proteins/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Fibroblasts/metabolism , Hepatocytes/metabolism , RNA, Small Interfering/genetics , Albumins/genetics , Albumins/metabolism , Animals , Argonaute Proteins/deficiency , CRISPR-Cas Systems , Cell Lineage/genetics , Cell Transdifferentiation/genetics , Fibroblasts/cytology , Gene Regulatory Networks , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/metabolism , Hepatocytes/cytology , Lentivirus/genetics , Lentivirus/metabolism , Mice , Mice, Knockout , RNA, Small Interfering/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Transduction, Genetic
15.
Thorax ; 74(1): 18-32, 2019 01.
Article in English | MEDLINE | ID: mdl-29991510

ABSTRACT

RATIONALE: Goblet cell hyperplasia (GCH) is one of the cardinal features of chronic obstructive pulmonary disease (COPD) and contributes to airways obstruction. Rhinovirus (RV), which causes acute exacerbations in patients with COPD, also causes prolonged airways obstruction. Previously, we showed that RV enhances mucin gene expression and increases goblet cell number in a COPD mouse model. This study examines whether RV causes sustained GCH in relevant models of COPD. METHODS: Mucociliary-differentiated COPD and normal airway epithelial cell cultures and mice with normal or COPD phenotype were infected with RV or sham and examined for GCH by immunofluorescence and/or mucin gene expression. In some experiments, RV-infected COPD cells and mice with COPD phenotype were treated with γ-secretase inhibitor or interleukin-13 neutralising antibody and assessed for GCH. To determine the contribution of NOTCH1/3 in RV-induced GCH, COPD cells transduced with NOTCH1/3 shRNA were used. RESULTS: RV-infected COPD, but not normal cell cultures, showed sustained GCH and increased mucin genes expression. Microarray analysis indicated increased expression of NOTCH1, NOTCH3 and HEY1 only in RV-infected COPD cells. Blocking NOTCH3, but not NOTCH1, attenuated RV-induced GCH in vitro. Inhibition of NOTCH signalling by γ-secretase inhibitor, but not neutralising antibody to IL-13, abrogated RV-induced GCH and mucin gene expression. CONCLUSIONS: RV induces sustained GCH via NOTCH3 particularly in COPD cells or mice with COPD phenotype. This may be one of the mechanisms that may contribute to RV-induced prolonged airways obstruction in COPD.


Subject(s)
Goblet Cells/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Receptor, Notch3/genetics , Respiratory Mucosa/pathology , Rhinovirus , Actins/metabolism , Amyloid Precursor Protein Secretases/pharmacology , Animals , Antibodies, Neutralizing/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , Disease Models, Animal , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Female , Gene Expression/drug effects , Gene Silencing , Goblet Cells/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Humans , Hyperplasia/metabolism , Hyperplasia/virology , Interleukin-13/immunology , Mice , Mucin 5AC/genetics , Mucin-5B/genetics , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch3/metabolism , Respiratory Mucosa/metabolism , Signal Transduction/drug effects
16.
Gene ; 685: 202-210, 2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30415009

ABSTRACT

Despite advances in early diagnosis and treatment, cancer still remains the major reason of mortality worldwide. The forkhead box A (FOXA) family is reported to participate in diverse human diseases. However, little is known about their expression and prognostic values in human lung cancer. Herein, we conducted a detailed cancer vs. normal analysis. The mRNA expression levels of FOXA family in numerous kind of cancers, including lung cancer, were analyzed using the Oncomine and GEPIA database. We observed that the mRNA expression levels of FOXA1, and FOXA3 were all increased while FOXA2 were decreased in most cancers compared with normal tissues, especially in lung cancer. Moreover, the expression levels of FOXA1, and FOXA3 are also highly expressed, while FOXA2 were decreased in almost all cancer cell lines, particularly in lung cancer cell lines, analyzing by Cancer Cell Line Encyclopedia (CCLE) and EMBL-EBI databases. Furthermore, the LinkedOmics database was used to evaluate the prognostic values, indicating that higher expression of FOXA1, FOXA3 indicated a poor overall survival (OS), while increased FOXA2 revealed a better OS in lung cancer. To conclusion, FOXA family showed significant expression differences between cancer and normal tissues, especially lung cancer, and FOXA1, FOXA3 could be promising prognostic biomarkers for lung cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Cell Line, Tumor , Computational Biology/methods , Databases, Genetic , Gene Expression Profiling , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-gamma/metabolism , Humans , Lung Neoplasms/metabolism , Multigene Family , Prognosis , RNA, Messenger/genetics , Transcription, Genetic
17.
Acta Pharmacol Sin ; 40(5): 620-629, 2019 May.
Article in English | MEDLINE | ID: mdl-30315254

ABSTRACT

Human liver or hepatocyte transplantation is limited by a severe shortage of donor organs. Direct reprogramming of other adult cells into hepatic cells may offer a solution to this problem. In a previous study, we have generated hepatocyte-like cells from mouse fibroblasts using only one transcription factor (TF) plus a chemical cocktail. Here, we show that human urine-derived epithelial-like cells (hUCs) can also be transdifferentiated into human hepatocyte-like cells (hiHeps) using one TF (Foxa3, Hnf1α, or Hnf4α) plus the same chemical cocktail CRVPTD (C, CHIR99021; R, RepSox; V, VPA; P, Parnate; T, TTNPB; and D, Dznep). These hiHeps express multiple hepatocyte-specific genes and display functions characteristic of mature hepatocytes. With the introduction of the large T antigen, these hiHeps can be expanded in vitro and can restore liver function in mice with concanavalin-A-induced acute liver failure. Our study provides a strategy to generate functional hepatocyte-like cells from hUCs by using a single TF plus a chemical cocktail.


Subject(s)
Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Epithelial Cells/cytology , Hepatocytes/cytology , Liver Failure, Acute/therapy , Urine/cytology , Animals , Concanavalin A , Epithelial Cells/metabolism , HEK293 Cells , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/transplantation , Humans , Liver Failure, Acute/chemically induced , Male , Mice , Transfection , Young Adult
18.
Methods Mol Biol ; 1905: 93-101, 2019.
Article in English | MEDLINE | ID: mdl-30536093

ABSTRACT

Primary hepatocytes are widely used in regenerative medicine, drug metabolism analysis, and in vitro drug screens. To overcome the shortage of liver donors, several strategies, such as differentiation of pluripotent stem cells and transdifferentiation from somatic cells, were developed to generate hepatocytes from alternative sources. Here, we describe in detail lenti-virus-based procedure for direct conversion of human fibroblasts to hepatocytes (hiHep cells) in vitro. A detailed protocol for preparation of human fibroblasts from scar tissues is also provided. Based on this protocol, FOXA3, HNF1A, and HNF4A are introduced into SV40-large-T-antigen-expressing human scar fibroblasts by lenti-virus. It usually takes about 5-7 days to get epithelial hiHep colonies. SV40-large-T-antigen-expressing hiHep (hiHepLT) cells are proliferative and can be expanded to a large number for potential uses.


Subject(s)
Cell Culture Techniques/methods , Cicatrix/pathology , Fibroblasts/cytology , Hepatocytes/cytology , Lentivirus/genetics , Antigens, Polyomavirus Transforming/genetics , Cell Lineage , Cell Proliferation , Cell Transdifferentiation , Cellular Reprogramming , Cicatrix/genetics , Cicatrix/metabolism , Fibroblasts/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/metabolism , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Humans , Simian virus 40/genetics
19.
Methods Mol Biol ; 1905: 103-115, 2019.
Article in English | MEDLINE | ID: mdl-30536094

ABSTRACT

In vivo conversion of fibroblasts into hepatocyte-like cells provides one potential approach for the treatment of liver fibrosis. In our previous study, we showed in vivo conversion of myofibroblasts into induced hepatocytes (iHeps) by forced expression of four transcription factors in genetic fate-tracing mouse model of chronic liver disease. These in vivo-generated iHeps showed similar expression profile with endogenous hepatocytes (eHeps) and also exhibited similar functional characteristics, such as albumin secretion, urea synthesis, cytochrome activity, and drug responsiveness. Furthermore, the targeted expression of our reprogramming factors in myofibroblasts attenuated liver fibrosis. Our study suggests that in vivo reprogramming may open new perspectives for the treatment of diseases such as liver fibrosis.


Subject(s)
Dependovirus/genetics , Fibroblasts/cytology , Hepatocytes/cytology , Liver Cirrhosis/therapy , Transcription Factors/genetics , Animals , Carbon Tetrachloride/adverse effects , Cell Lineage , Cellular Reprogramming , Disease Models, Animal , Fibroblasts/metabolism , GATA4 Transcription Factor/genetics , Genetic Vectors/administration & dosage , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/metabolism , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice , Pyridines/adverse effects
20.
Cell Res ; 29(2): 124-135, 2019 02.
Article in English | MEDLINE | ID: mdl-30560924

ABSTRACT

Normal cells become cancer cells after a malignant transformation, but whether cancer cells can be reversed to normal status remains elusive. Here, we report that the combination of hepatocyte nuclear factor 1A (HNF1A), HNF4A and forkhead box protein A3 (FOXA3) synergistically reprograms hepatocellular carcinoma (HCC) cells to hepatocyte-like cells (reprogrammed hepatocytes, rHeps). Our results show that rHeps lose the malignant phenotypes of cancer cells and retrieve hepatocyte-specific characteristics including hepatocyte-like morphology; global expression pattern of genes and specific biomarkers of hepatocytes; and the unique hepatic functions of albumin (ALB) secretion, glycogen synthesis, low-density lipoprotein (LDL) uptake, urea production, cytochrome P450 enzymes induction and drug metabolism. Intratumoral injection of these three factors efficiently shrank patient-derived tumor xenografts and reprogrammed HCC cells in vivo. Most importantly, transplantation of rHeps in the liver of fumarylacetoacetate hydrolase-deficient (Fah-/-) mice led to the reconstruction of hepatic lobules and the restoration of hepatic function. Mechanistically, exogenous expression of HNF1A, HNF4A and FOXA3 in HCC cells initiated the endogenous expression of numerous hepatocyte nuclear factors, which promoted the conversion of HCC cells to hepatocyte-like cells. Collectively, our results indicate the successful conversion of hepatoma cells to hepatocyte-like cells, not only extending our current knowledge of cell reprogramming but also providing a route towards a novel therapeutic strategy for cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cellular Reprogramming/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 3-gamma/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cytochrome P-450 Enzyme System/metabolism , Genetic Vectors , HEK293 Cells , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 4/genetics , Heterografts , Humans , Liver Neoplasms/pathology , Mice , Mice, Knockout , Mice, Nude , Phenotype , Serum Albumin, Human/analysis , Transfection , Tumor Burden/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...