Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35165191

ABSTRACT

FOXP1 syndrome caused by haploinsufficiency of the forkhead box protein P1 (FOXP1) gene is a neurodevelopmental disorder that manifests motor dysfunction, intellectual disability, autism, and language impairment. In this study, we used a Foxp1+/- mouse model to address whether cognitive and motor deficits in FOXP1 syndrome are associated with mitochondrial dysfunction and oxidative stress. Here, we show that genes with a role in mitochondrial biogenesis and dynamics (e.g., Foxo1, Pgc-1α, Tfam, Opa1, and Drp1) were dysregulated in the striatum of Foxp1+/- mice at different postnatal stages. Furthermore, these animals exhibit a reduced mitochondrial membrane potential and complex I activity, as well as decreased expression of the antioxidants superoxide dismutase 2 (Sod2) and glutathione (GSH), resulting in increased oxidative stress and lipid peroxidation. These features can explain the reduced neurite branching, learning and memory, endurance, and motor coordination that we observed in these animals. Taken together, we provide strong evidence of mitochondrial dysfunction in Foxp1+/- mice, suggesting that insufficient energy supply and excessive oxidative stress underlie the cognitive and motor impairment in FOXP1 deficiency.


Subject(s)
Forkhead Transcription Factors/genetics , Intellectual Disability/genetics , Motor Disorders/genetics , Repressor Proteins/genetics , Animals , Autism Spectrum Disorder/genetics , Autistic Disorder/metabolism , Cognition/physiology , Disease Models, Animal , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/metabolism , Haploinsufficiency/genetics , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Motor Activity/genetics , Motor Disorders/metabolism , Neurodevelopmental Disorders/metabolism , Neurogenesis , Oxidative Stress/physiology , Repressor Proteins/deficiency , Repressor Proteins/metabolism
2.
J Cell Mol Med ; 28(8): e18294, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38652109

ABSTRACT

Forkhead box protein 1 (FOXP1) serves as a tumour promoter or suppressor depending on different cancers, but its effect in oesophageal squamous cell carcinoma has not been fully elucidated. This study investigated the role of FOXP1 in oesophageal squamous cell carcinoma through bioinformatics analysis and experimental verification. We determined through public databases that FOXP1 expresses low in oesophageal squamous cell carcinoma compared with normal tissues, while high expression of FOXP1 indicates a better prognosis. We identified potential target genes regulated by FOXP1, and explored the potential biological processes and signalling pathways involved in FOXP1 in oesophageal squamous cell carcinoma through GO and KEGG enrichment, gene co-expression analysis, and protein interaction network construction. We also analysed the correlation between FOXP1 and tumour immune infiltration levels. We further validated the inhibitory effect of FOXP1 on the proliferation of oesophageal squamous cell carcinoma cells through CCK-8, colony formation and subcutaneous tumour formation assays. This study revealed the anticarcinogenic effect of FOXP1 in oesophageal squamous cell carcinoma, which may serve as a novel biological target for the treatment of tumour.


Subject(s)
Cell Proliferation , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Forkhead Transcription Factors , Gene Expression Regulation, Neoplastic , Repressor Proteins , Humans , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Cell Line, Tumor , Animals , Repressor Proteins/metabolism , Repressor Proteins/genetics , Computational Biology/methods , Mice , Prognosis , Protein Interaction Maps/genetics , Signal Transduction , Gene Regulatory Networks , Mice, Nude
3.
J Biol Chem ; 299(6): 104795, 2023 06.
Article in English | MEDLINE | ID: mdl-37150320

ABSTRACT

In recent years, lactate has been recognized as an important circulating energy substrate rather than only a dead-end metabolic waste product generated during glucose oxidation at low levels of oxygen. The term "aerobic glycolysis" has been coined to denote increased glucose uptake and lactate production despite normal oxygen levels and functional mitochondria. Hence, in "aerobic glycolysis," lactate production is a metabolic choice, whereas in "anaerobic glycolysis," it is a metabolic necessity based on inadequate levels of oxygen. Interestingly, lactate can be taken up by cells and oxidized to pyruvate and thus constitutes a source of pyruvate that is independent of insulin. Here, we show that the transcription factor Foxp1 regulates glucose uptake and lactate production in adipocytes and myocytes. Overexpression of Foxp1 leads to increased glucose uptake and lactate production. In addition, protein levels of several enzymes in the glycolytic pathway are upregulated, such as hexokinase 2, phosphofructokinase, aldolase, and lactate dehydrogenase. Using chromatin immunoprecipitation and real-time quantitative PCR assays, we demonstrate that Foxp1 directly interacts with promoter consensus cis-elements that regulate expression of several of these target genes. Conversely, knockdown of Foxp1 suppresses these enzyme levels and lowers glucose uptake and lactate production. Moreover, mice with a targeted deletion of Foxp1 in muscle display systemic glucose intolerance with decreased muscle glucose uptake. In primary human adipocytes with induced expression of Foxp1, we find increased glycolysis and glycolytic capacity. Our results indicate Foxp1 may play an important role as a regulator of aerobic glycolysis in adipose tissue and muscle.


Subject(s)
Adipocytes , Forkhead Transcription Factors , Glycolysis , Muscle Cells , Transcription Factors , Animals , Mice , Adipocytes/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucose/metabolism , Glycolysis/genetics , Lactic Acid/metabolism , Muscle Cells/metabolism , Pyruvates , Transcription Factors/metabolism , Rats , Cell Line , Transcriptome
4.
Diabet Med ; : e15386, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38887963

ABSTRACT

AIM: Impaired wound healing in patients with diabetes can develop into nonhealing ulcerations. Because bone marrow mesenchymal stem cells (BMSCs) exosomes can promote wound healing, this study aims to investigate the mechanism of BMSCs-isolated exosomal miR-221-3p in angiogenesis and diabetic wound healing. METHODS: To mimic diabetes in vitro, human umbilical vein endothelial cells (HUVECs) were subjected to high glucose (HG). Exosomes were derived from BMSCs and identified by transmission electron microscopy (TEM), western blot analysis and dynamic light scattering (DLS). The ability to differentiate BMSCs was assessed via Oil red O staining, alkaline phosphatase (ALP) staining and alizarin red staining. The ability to internalise PKH26-labelled exosomes was assessed using confocal microscopy. Migration, cell viability and angiogenesis were tested by scratch, MTT and tube formation assays separately. The miRNA and protein levels were analysed by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) or western blotting. The relationship among miR-221-3p, FOXP1 and SPRY1 was determined using the dual-luciferase reporter, ChIP and RIP assays. RESULTS: Exosomal miR-221-3p was successfully isolated from BMSCs and delivered into HUVECs. HG was found to suppress the angiogenesis, cell viability and migration of HUVECs and exosomal miR-221-3p separated from BMSCs inhibited the above phenomenon. FOXP1 could transcriptionally upregulate SPRY1, and the silencing of FOXP1 reversed the HG-stimulated angiogenesis inhibition, cell viability and migration in HUVECs via the downregulation of SPRY1. Meanwhile, miR-221-3p directly targeted FOXP1 and the overexpression of FOXP1 reversed the positive effect of exosomal miR-221-3p on HUVEC angiogenesis. CONCLUSION: Exosomal miR-221-3p isolated from BMSCs promoted angiogenesis in diabetic wounds through the mediation of the FOXP1/SPRY1 axis. Furthermore, the findings of this study can provide new insights into probing strategies against diabetes.

5.
Am J Med Genet A ; : e63713, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38924631

ABSTRACT

Haploinsufficiency of FOXP1 gene is responsible for a neurodevelopmental disorder presenting with intellectual disability (ID), autism spectrum disorder (ASD), hypotonia, mild dysmorphic features, and multiple congenital anomalies. Joint contractures are not listed as a major feature of FOXP1-related disorder. We report five unrelated individuals, each harboring likely gene disruptive de novo FOXP1 variants or whole gene microdeletion, who showed multiple joint contractures affecting at least two proximal and/or distal joints. Consistent with the phenotype of FOXP1-related disorder, all five patients showed developmental delay with moderate-to-severe speech delay, ID, ASD, and facial dysmorphic features. FOXP1 is implicated in neuronal differentiation and in organizing motor axon projections, thus providing a potential developmental basis for the joint contractures. The combination of joint contractures and neurodevelopmental disorders supports the clinical suspicion of FOXP1-related phenotype.

6.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891897

ABSTRACT

Heterozygous mutations in the FOXP1 gene (OMIM#605515) are responsible for a well-characterized neurodevelopmental syndrome known as "intellectual developmental disorder with language impairment with or without autistic features" (OMIM#613670) or FOXP1 syndrome for short. The main features of the condition are global developmental delay/intellectual disability; speech impairment in all individuals, regardless of their level of cognitive abilities; behavioral abnormalities; congenital anomalies, including subtle dysmorphic features; and strabismus, brain, cardiac, and urogenital abnormalities. Here, we present two siblings with a de novo heterozygous FOXP1 variant, namely, a four-year-old boy and 14-month-old girl. Both children have significantly delayed early psychomotor development, hypotonia, and very similar, slightly dysmorphic facial features. A lack of expressive speech was the leading symptom in the case of the four-year-old boy. We performed whole-exome sequencing on the male patient, which identified a pathogenic heterozygous c.1541G>A (p.Arg514His) FOXP1 mutation. His sister's targeted mutation analysis also showed the same heterozygous FOXP1 variant. Segregation analysis revealed the de novo origin of the mutation, suggesting the presence of parental gonadal mosaicism. To the best of our knowledge, this is the first report of gonadal mosaicism in FOXP1-related neurodevelopmental disorders in the medical literature.


Subject(s)
Forkhead Transcription Factors , Mosaicism , Neurodevelopmental Disorders , Repressor Proteins , Humans , Forkhead Transcription Factors/genetics , Male , Female , Child, Preschool , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/diagnosis , Infant , Repressor Proteins/genetics , Mutation , Exome Sequencing , Heterozygote
7.
Stem Cells ; 40(9): 843-856, 2022 09 26.
Article in English | MEDLINE | ID: mdl-35759955

ABSTRACT

Quiescent hair follicle stem cells (HFSCs) reside in specialized bulge niche where they undergo activation and differentiation upon sensing niche-dependent signals during hair follicle (HF) homeostasis and wound repair. The underlying mechanism of HFSCs and bulge niche maintenance is poorly understood. Our previous study has reported that a transcription factor, forkhead box P1 (Foxp1), functions to maintain the quiescence of HFSCs. Here, we further discovered that forkhead box P4 (Foxp4), a close family member of Foxp1, had similar expression profiles in various components of HFs and formed a complex with Foxp1 in vitro and in vivo. The HF-specific deficiency of Foxp4 resulted in the precocious activation of HFSCs during hair cycles. In contrast to single Foxp1 or Foxp4 conditional knockout (cKO) mice, Foxp1/4 double cKO exerted an additive effect in the spectrum and severity of phenotypes in HFSC activation, hair cycling acceleration and hair loss, coupled with remarkable downregulation of fibroblast growth factor 18 (Fgf18) and bone morphogenetic protein 6 (Bmp6) expression in bulge cells. In addition, the double KO of Foxp1/4 induced the apoptosis of K6-positive (K6+) inner bulge cells, a well-established stem cell (SC) niche, thus resulting in the destruction of the bulge SC niche and recurrent hair loss. Our investigation reveals the synergistic role of Foxp1/4 in sustaining K6+ niche cells for the quiescence of HFSCs.


Subject(s)
Bone Morphogenetic Protein 6 , Stem Cell Niche , Alopecia/metabolism , Animals , Apoptosis/genetics , Bone Morphogenetic Protein 6/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Hair Follicle , Mice , Repressor Proteins/metabolism
8.
Mol Ther ; 30(4): 1692-1705, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35124178

ABSTRACT

Preeclampsia (PE) is associated with maternal and fetal perinatal morbidity and mortality, which brings tremendous suffering and imposes an economic burden worldwide. The failure of uterine spiral artery remodeling may be related to the abnormal function of trophoblasts and lead to the occurrence and progression of PE. Aberrant expression of long non-coding RNAs (lncRNAs) is involved in the failure of uterine spiral artery remodeling. However, the regulation of lncRNA expression in PE is poorly characterized. Here, we reported that hypoxia-induced microRNA (miR)-218 inhibited the expression of lncRNA TUG1 by targeting FOXP1. Further RNA sequencing and mechanism analysis revealed that silencing of TUG1 increased the expression of DNA demethylase TET3 and proliferation-related DUSP family, including DUSP2, DUSP4, and DUSP5, via binding to SUV39H1 in the nucleus. Moreover, TUG1 modulated the DUSP family in vitro through a TET3-mediated epigenetic mechanism. Taken together, our results unmask a new regulatory network mediated by TUG1 as an essential determinant of the pathogenesis of PE, which regulates cell growth and possibly the occurrence and development of other diseases.


Subject(s)
MicroRNAs , Pre-Eclampsia , RNA, Long Noncoding , Arteries/metabolism , Arteries/pathology , Cell Proliferation/genetics , Female , Forkhead Transcription Factors/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Repressor Proteins
9.
J Assist Reprod Genet ; 40(7): 1559-1572, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37052757

ABSTRACT

PURPOSE: As a member of the C19MC family, miR-526b-5p is mainly expressed in the placental tissue and is a well-known tumor suppressor microRNA. However, its effect on the function of trophoblasts and its role in the development of recurrent spontaneous abortion (RSA) remains unclear. METHODS: Transcriptome sequencing, quantitative real-time polymerase chain reaction (RT-qPCR), Western blot, 5-ethynyl-2'-deoxyuridine (Edu) proliferation analysis, cell counting kit-8 (CCK8) assay, Transwell assays, and wound healing were used to detect the proliferation, migration, and invasion capacity of trophoblasts. Target genes of miR-526b-5p were obtained by the dual luciferase reporter system. The promoter-reporter system and ChIP-qPCR were used to prove that c-Myc positively regulated the expression of Foxp1 RESULTS: The miR-526b-5p levels were significantly higher in patients with RSA than in controls. High expression of miR-526b-5p inhibited the proliferation, migration, and invasion of trophoblast cell line. By contrast, low expression of miR-526b-5p promoted the proliferation and migration of trophoblast cell line. Target genes of miR-526b-5p were c-Myc and Foxp1. c-Myc positively regulated the expression of Foxp1 by binding to the Foxp1 promoter location -146/-135. Finally, miR-526b-5p impeded the proliferation, migration, and invasion of trophoblasts by negatively regulating c-Myc by rescue experiments. CONCLUSION: Thus, miR-526b-5p affected the proliferation, migration, and invasion of trophoblasts by targeting c-Myc and Foxp1. Low expression of c-Myc further deactivated the positive transcriptional regulation of c-Myc on Foxp1, which may be the mechanism of RSA. This study provides potential therapeutic targets and clues for the diagnosis and treatment of RSA.


Subject(s)
Abortion, Habitual , Forkhead Transcription Factors , MicroRNAs , Proto-Oncogene Proteins c-myc , Female , Humans , Pregnancy , Abortion, Habitual/genetics , Abortion, Habitual/metabolism , Cell Proliferation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Placenta/metabolism , Transcription Factors/metabolism , Trophoblasts/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
10.
Lasers Med Sci ; 38(1): 106, 2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37074483

ABSTRACT

The current study was performed to investigate the treatment of tumors with gold nanoparticles, laser, and photodynamic therapy (PDT) by using an immunohistochemistry method and to investigate the expression of FOXP1 in infected mice with mammary adenocarcinoma whether it can be used as an indicator to estimate the recovery of tissues from cancer disease. Twenty-five albino female mice were used in this research; they were divided into five groups, four groups were infected with mammary adenocarcinoma, and then three of them were treated with gold nanoparticles, laser, and PDT, respectively, while the fourth group was left without any treatment and represents the positive control, and the fifth group (normal mice) represents the negative control. Tissue sections were taken from different groups of mice in order to estimate FOXP1 expression in infected mice by using an immunohistochemistry assay. FOXP1 expression was higher in the tumor and kidney tissues of the mice treated with PDT than that in mice treated with either gold nanoparticles or laser alone. Also, in the group of mice treated with laser, FOXP1 expression was higher than the expression in mice which were treated with gold nanoparticles but lower than that in mice which were treated with PDT. FOXP1 can be used as a biomarker for the prognosis outcome of breast and other solid tumors, as well as it considers a key tumor suppressor. PDT is the best choice to treat cancer in comparison to using either gold nanoparticles or the laser separately.


Subject(s)
Adenocarcinoma , Metal Nanoparticles , Photochemotherapy , Animals , Mice , Photochemotherapy/methods , Gold , Immunohistochemistry , Lasers , Adenocarcinoma/drug therapy , Photosensitizing Agents/therapeutic use , Repressor Proteins , Forkhead Transcription Factors
11.
Cytokine ; 152: 155812, 2022 04.
Article in English | MEDLINE | ID: mdl-35180562

ABSTRACT

OBJECTIVE: The purpose of this design was to explore the specific role and related mechanism of long noncoding RNA (lncRNA) regulators of reprogramming (ROR) in viral myocarditis (VMC). METHODS: AC16 cells were infected with coxsackievirus B3 (CVB3) to establish a VMC cell model in vitro. The release of interleukin (IL)-1ß and IL-18 was evaluated by enzyme-linked immunosorbent assay (ELISA). Gene expression was calculated using quantitative real-time (qRT)-PCR. Cell pyroptosis was determined by flow cytometry and Western blot assays. Cell counting Kit-8 (CCK-8) detected cell viability. The molecular associations were verified by employing RNA immunoprecipitation (RIP), RNA pulldown and chromatin immunoprecipitation (ChIP) assays. RESULTS: The lncRNA ROR was more highly expressed in CVB3 virus-infected AC16 cells than in controls. Knockdown of ROR markedly rescued cell viability and reduced the release of IL-1ß and IL-18, cell pyroptosis and pyroptotic proteins such as NLRP3, ASC and cleaved caspase 1. Mechanistically, ROR destroyed the mRNA stability of Forkhead Box P Factor 1 (FOXP1) by binding polypyrimidine tract binding protein 1 (PTBP1). FOXP1 repressed the transcription of NLRP3 by directly interacting with its promoter. Importantly, coinhibition of FOXP1 impeded the protective role of ROR silencing in CVB3-infected AC16 cells. CONCLUSION: In conclusion, these findings elucidated that ROR knockdown inhibited CVB3-induced cardiomyocyte inflammation and NLRP3-mediated pyroptosis by regulating the PTBP1/FOXP1 axis, implying that ROR might be a new inducer in CVB3-infected VMC.


Subject(s)
Myocarditis , RNA, Long Noncoding , Forkhead Transcription Factors/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Interleukin-18/metabolism , Myocarditis/metabolism , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Polypyrimidine Tract-Binding Protein/genetics , Pyroptosis/genetics , RNA, Long Noncoding/genetics , Repressor Proteins/metabolism
12.
Respir Res ; 23(1): 281, 2022 Oct 11.
Article in English | MEDLINE | ID: mdl-36221131

ABSTRACT

BACKGROUND: Genes involved in lung development may become dysregulated in adult life and contribute to the pathogenesis of lung diseases. Multiple genes regulate lung development, including Forkhead box protein P1-4 (FoxP1-4). METHODS: We examined the association between variants in the FoxP1-4 genes and lung function using data from a GWAS that included close to 400,000 individuals and 20 million SNPs. RESULTS: More than 100 variants in the FoxP1 gene, but none in the FoxP2-4 genes, are associated with lung function. The sentinel variant in the FoxP1 gene associated with FEV1 was rs1499894 (C > T), while the sentinel variant in the FoxP1 gene associated with FVC was rs35480566 (A > G). Those with the T allele instead of the C allele for rs1499894, or the G allele instead of the A allele for rs35480566 had increased FoxP1 mRNA levels in transcriptomic data, higher FEV1 and FVC, and reduced odds of being diagnosed with idiopathic pulmonary fibrosis. Further, knockdown of FoxP1 in lung epithelial cells by RNA interference led to increased mRNA levels for matrix metalloproteinases 1, 2, 3 and pro-inflammatory cytokines IL-6 & IL-8, as well as reduced cell viability after exposure to cigarette smoke-all processes implicated in the pathogenesis of COPD and IPF. CONCLUSIONS: Our results suggest that the protein encoded by the FoxP1 gene may protect against the development of COPD and IPF. A causal role for FoxP1 in the pathogenesis of COPD and IPF may warrant further investigation, and FoxP1 may be a novel therapeutic target for these lung disorders.


Subject(s)
Idiopathic Pulmonary Fibrosis , Pulmonary Disease, Chronic Obstructive , Adult , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Inflammation Mediators , Interleukin-6 , Interleukin-8 , Lung/metabolism , Matrix Metalloproteinases , RNA, Messenger , Repressor Proteins/genetics
13.
Mol Cell Biochem ; 477(3): 951-963, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35098439

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most prevalent subtype of non-Hodgkin lymphoma and is a very aggressive malignancy with tumor growing rapidly in organs like lymph nodes. The pathogenesis of DLBCL is not clear and the prognosis of DLBCL requires improvement. Here, we investigated the mechanisms of DLBCL, with the focus on lncRNA PVT1/miR-34b-5p/Foxp1 axis. Human DLBCL tissues from diagnosed DLBCL patients and four human DLBCL cell lines, one normal human B lymphoblastoid cell line were used. qRT-PCR and western blotting were employed to measure expression levels of lncRNA PVT1, Foxp1, miR-34b-5p, ß-catenin, and proliferation-related proteins. MTT assay and colony formation assay were performed to determine cell proliferation. Flow cytometry was used to examine cell apoptosis. ChIP and Dual-luciferase assay were utilized to validate interactions of Foxp1/promoters, PVT1/miR-34b-5p and miR-34b-5p/Foxp1. Mouse tumor xenograft model was used to determine the effect of sh-PVT1 on tumor growth in vivo. In this study, we found PVT1 and Foxp1 were elevated in DLBCL tissues and cells while miR-34b-5p was decreased. Knockdown of PVT1, overexpression of miR-34b-5p, or Foxp1 knockdown repressed DLBCL cell proliferation but enhanced cell apoptosis. PVT1 directly bound miR-34b-5p to disinhibit Foxp1/ß-catenin signaling. Foxp1 regulated CDK4, CyclinD1, and p53 expression via binding with their promoters. Knockdown of Foxp1 partially reversed the effects of miR-34b-5p inhibitor on DLBCL cell proliferation and apoptosis. Inhibition of PVT1 through shRNA suppressed DLBCL tumor growth in vivo. All in all, lncRNA PVT1 promotes DLBCL progression via acting as a miR-34b-5p sponge to disinhibit Foxp1/ß-catenin signaling.


Subject(s)
Forkhead Transcription Factors/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , Repressor Proteins/metabolism , Signal Transduction , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Repressor Proteins/genetics
14.
Cell Biol Int ; 46(2): 265-277, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34816539

ABSTRACT

Forkhead box protein P1 (Foxp1) is a kind of tumor suppressor gene, and the role of Foxp1 in the macrophages of myocardial infarction (MI) has not been studied yet. Here, we verified the role of the transcription factor high mobility group box 1 (HMGB1) and its target gene Foxp1 in the inflammatory response. In this study, the key genes HMGB1 and Foxp1 in the macrophages of mouse MI model were screened out through single-cell transcriptome analysis of GSE136088 (GEO database). In vitro experiment indicated that hypoxia induced the inflammatory response in RAW264.7 macrophages, promoted the secretion of inflammatory factors (tumor necrosis factor α [TNF-α], interleukin 6 [IL-6], and IL-1ß) and the activation of NLRP3 inflammasome (NLRP3, ASC, and pro-caspase-1). Meanwhile, HMGB1 increased while Foxp1 decreased in hypoxia-treated RAW264.7 macrophages. HMGB1 bound to the upstream promoter region of Foxp1 as demonstrated by the dual-luciferase reporter assay, chromatin immunoprecipitation (ChIP)-quantitative polymerase chain reaction (qPCR) and agarose gel electrophoresis. As a transcription factor, HMGB1 regulated Foxp1 expression. The secretion of inflammatory factors and the expression of NLRP3 inflammasome protein were changed when the expression of HMGB1 and Foxp1 was regulated in the hypoxia-treated RAW264.7 macrophages. This study verified that HMGB1 could aggravate the hypoxia-treated inflammatory response of macrophages through downregulating Foxp1, which not only provides evidence to support the role of HMGB1/Foxp1 in macrophages but also offers another angle for the treatment of MI.


Subject(s)
HMGB1 Protein , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Hypoxia/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Repressor Proteins/metabolism
15.
Graefes Arch Clin Exp Ophthalmol ; 260(12): 3857-3867, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35695913

ABSTRACT

BACKGROUND: Diabetic retinopathy (DR) is still the fastest growing cause of blindness in working aged adults, and its typical characteristics are endothelial cell dysfunction and pericytes loss. Transcription factor fork head box P1 (FOXP1) is a member of FOX family involved in diabetes progression and is expressed in endothelial cells. The purpose of this study was to investigate the role and mechanism of FOXP1 in DR. METHODS: The vitreous of DR patients and non-DR patients were collected, and the expression of FOXP1 was detected by real-time polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). Human umbilical vein endothelial cells (HUVECs) cultured in high glucose simulated DR environment, and the expressions of FOXP1, vascular endothelial growth factor (VEGF), and pigment epithelium derived factor (PEDF) were detected by RT-qPCR and western blot (WB) after transfection of small interfering RNA (siRNA) to knock out FOXP1. At the same time, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay (MTT), 5-ethynyl-2'-deoxyuridine assay (EDU), flow cytometry, Transwell assay, and tube-forming experiment were performed to determine cell proliferation, migration, and tube-forming ability. RESULTS: We found that FOXP1 was highly expressed in the vitreous of DR patients and HUVECs under high glucose condition. After FOXP1 was decreased, the activation of VEGF expression and inhibition of PEDF expression in HUVECs induced by high glucose were reversed; meanwhile, cell proliferation, migration, and tube formation decreased, and apoptosis was promoted. CONCLUSION: Generally, FOXP1 is highly expressed in the vitreous of DR patients, and its silence prevented VEGF/PEDF signaling pathway stimulated by high glucose and also reduced the proliferation, migration, and tube formation of endothelial cell, thus improving vascular endothelial dysfunction caused by DR. The results indicate that FOXP1 may be a therapeutic target of DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Adult , Humans , Middle Aged , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Transcription Factors , Endothelial Cells/metabolism , Cell Proliferation , Vascular Endothelial Growth Factors , Glucose/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Repressor Proteins , Forkhead Transcription Factors/genetics
16.
Endocr J ; 69(9): 1067-1078, 2022 Sep 28.
Article in English | MEDLINE | ID: mdl-35545535

ABSTRACT

Gestational diabetes mellitus (GDM) is a health risk for pregnant women and infants. Emerging evidence suggests that the deregulation of circular RNAs (circRNAs) is associated with the progression of this disorder. The objective of this study was to investigate the role of circ_FOXP1 in GDM. Cell models of GDM were established by treating human trophoblast cells with high glucose (HG). The expression of circ_FOXP1, miR-508-3p and SMAD family member 2 (SMAD2) mRNA was detected by quantitative real-time PCR (qPCR). Cell proliferation was assessed by EdU assay and MTT assay, and cell cycle and cell apoptosis were determined by flow cytometry assay. The protein levels of proliferation- and apoptosis-related markers and SMAD2 were measured by western blot. The relationship between miR-508-3p and circ_FOXP1 or SMAD2 was validated by dual-luciferase reporter assay or pull-down assay. The expression of circ_FOXP1 was downregulated in HG-treated HTR-8/SVneo cells. Circ_FOXP1 overexpression promoted HG-inhibited HTR-8/SVneo cell proliferation and suppressed HG-induced HTR-8/SVneo cell cycle arrest and apoptosis. Circ_FOXP1 positively regulated the expression of SMAD2 by targeting miR-508-3p. MiR-508-3p was overexpressed in HG-treated HTR-8/SVneo cells, and its overexpression reversed the effects of circ_FOXP1 overexpression. MiR-508-3p inhibition also alleviated HG-induced HTR-8/SVneo cell injuries, while the knockdown of SMAD2 abolished these effects. Collectively, circ_FOXP1 promotes the growth and survival of HG-treated human trophoblast cells through the miR-508-3p/SMAD2 pathway, hinting that circ_FOXP1 was involved in GDM progression.


Subject(s)
Diabetes, Gestational , MicroRNAs , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucose/metabolism , Humans , Infant , MicroRNAs/genetics , MicroRNAs/metabolism , Pregnancy , RNA, Circular/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trophoblasts/metabolism
17.
Proc Natl Acad Sci U S A ; 116(44): 22237-22245, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611379

ABSTRACT

Gastrointestinal dysfunctions in individuals with autism spectrum disorder are poorly understood, although they are common among this group of patients. FOXP1 haploinsufficiency is characterized by autistic behavior, language impairment, and intellectual disability, but feeding difficulties and gastrointestinal problems have also been reported. Whether these are primary impairments, the result of altered eating behavior, or side effects of psychotropic medication remains unclear. To address this question, we investigated Foxp1+/- mice reflecting FOXP1 haploinsufficiency. These animals show decreased body weight and altered feeding behavior with reduced food and water intake. A pronounced muscular atrophy was detected in the esophagus and colon, caused by reduced muscle cell proliferation. Nitric oxide-induced relaxation of the lower esophageal sphincter was impaired and achalasia was confirmed in vivo by manometry. Foxp1 targets (Nexn, Rbms3, and Wls) identified in the brain were dysregulated in the adult Foxp1+/- esophagus. Total gastrointestinal transit was significantly prolonged due to impaired colonic contractility. Our results have uncovered a previously unknown dysfunction (achalasia and impaired gut motility) that explains the gastrointestinal disturbances in patients with FOXP1 syndrome, with potential wider relevance for autism.


Subject(s)
Autistic Disorder/genetics , Esophageal Achalasia/genetics , Forkhead Transcription Factors/genetics , Gastrointestinal Transit , Repressor Proteins/genetics , Animals , Autistic Disorder/physiopathology , Brain/metabolism , Cell Proliferation , Colon/metabolism , Colon/pathology , Colon/physiopathology , Esophageal Achalasia/physiopathology , Esophagus/metabolism , Esophagus/pathology , Esophagus/physiopathology , Feeding Behavior , Female , Forkhead Transcription Factors/metabolism , Heterozygote , Male , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Syndrome , Trans-Activators/genetics , Trans-Activators/metabolism
18.
J Cell Mol Med ; 25(4): 2000-2012, 2021 02.
Article in English | MEDLINE | ID: mdl-33372387

ABSTRACT

Endometriosis is a common multi-factorial gynaecological disease. Recent studies have revealed that long non-coding RNAs (lncRNAs) are involved in the pathogenesis of endometriosis. In the present study, the expression profiles of lncRNAs in 6 pairs of endometriosis ectopic endometrium (ecEM) and eutopic endometrium (euEM) tissues were analysed by RNA sequencing. From the profiles, LINC01116 was found to be up-regulated in ecEM tissues compared to euEM tissues and was verified by quantitative real-time PCR (qRT-PCR). Then, functional experiments demonstrated that LINC01116 promoted the proliferation and migration of ectopic primary endometrial stromal cells (ESCs), while miR-9-5p exerted the opposite effects. Dual-luciferase reporter assays verified that LINC01116 directly sponged miR-9-5p and relieved the suppression of its target, Forkhead box protein P1 (FOXP1). Rescue experiments further demonstrated that LINC01116 could promote proliferation and migration of ESCs by targeting FOXP1 via sponging miR-9-5p. Overall, our study illuminates that LINC01116 promotes the progression of endometriosis through the miR-9-5p/FOXP1 axis. This finding provides a novel therapeutic target for patients with endometriosis.


Subject(s)
Endometriosis/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation , MicroRNAs/genetics , RNA Interference , Repressor Proteins/genetics , Stromal Cells/metabolism , Adult , Cell Movement , Cell Proliferation , Computational Biology/methods , Disease Susceptibility , Endometriosis/metabolism , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Transcriptome , Young Adult
19.
Biochem Biophys Res Commun ; 557: 221-227, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33887587

ABSTRACT

Although substantial progress has been made in early detection and treatment of GC, this disease remains a major burden worldwide. CircRNAs have potential as prognostic and diagnostic biomarkers in tumorigenesis. Therefore, we aimed to clarify the role and mechanism of circACC1 in GC cell proliferation. The expression levels of circACC1, miR-29c-3p and FOXP1 were validated in GC tissue samples and adjacent tissue samples. The impact of circACC1 and miR-29c-3p on overall survival was evaluated in GC specimens. A functional study was performed on MKN-45 and BGC823 cells transfected with different vectors. Cell proliferation was assayed by CCK-8 and colony formation assays. The interactions among circACC1, miR-29c-3p and FOXP1 were tested by RNA immunoprecipitation and luciferase reporter assays. This study demonstrated that circACC1 is upregulated in GC tissues, and its upregulation predicts poorer OS in GC patients. Upregulation of circACC1 promoted GC cell proliferation, as indicated by CCK-8 and colony formation assays. A mechanistic study revealed that the pro-oncogenic effect of circACC1 was mainly caused by binding to miR-29c-3p, thus regulating expression of its downstream target FOXP1. The circACC1/miR-29c-3p/FOXP1 network plays a key role in gastric cancer by regulating cell proliferation.


Subject(s)
Cell Proliferation/genetics , Forkhead Transcription Factors/metabolism , RNA, Circular/metabolism , Repressor Proteins/metabolism , Stomach Neoplasms/metabolism , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Prognosis , RNA, Circular/genetics , Repressor Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Up-Regulation
20.
Biotechnol Lett ; 43(2): 353-367, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33128129

ABSTRACT

miRNAs have been pointed to play critical role in the development of congenital heart disease (CHD). miRNA-375-3p (miR-375-3p) was involved in cardiac dysfunction and cardiogenesis. However, no prior study had established a therapeutic role of miR-375-3p in CHD. We intended to investigate the effect and mechanism of miR-375-3p on apoptosis in hypoxic cardiomyocytes in vitro. Expression of miR-375-3p, forkhead box P1 (FOXP1) and Bcl2 like protein 2 (Bcl2l2) was detected using real-time quantitative PCR and western blot. Apoptosis was measured with MTT assay, flow cytometry and caspase-3 activity assay. The potential target binding between miR-375-3p and FOXP1/Bcl2l2 was predicted on DianaTools, and was validated by luciferase reporter assay and RNA pull-down assay. As a result, miR-375-3p was upregulated and FOXP1/Bcl2l2 was downregulated in maternal serum of women with fetal CHD and hypoxia-induced rat cardiomyocyte h9c2 cells. Hypoxia induced apoptosis rate elevation, caspase-3 activity promotion and viability inhibition in h9c2 cells; overexpression of miR-375-3p promoted, whereas knockdown of miR-375-3p antagonized hypoxia-induced effects in h9c2 cells. In addition, miR-375-3p was validated to negatively regulate FOXP1 and Bcl2l2 expression through target binding, and silencing of FOXP1 and Bcl2l2 could independently abate the anti-apoptosis role of miR-375-3p knockdown in hypoxic h9c2 cells. Collectively, blocking miR-375-3p suppressed hypoxia-evoked apoptosis of cardiomyocytes by targeting and upregulating FOXP1 and Bcl2l2. Our results might suggest maternal serum miR-375-3p as a potential biomarker for prenatal detection of fetal CHD.


Subject(s)
Forkhead Transcription Factors/blood , Heart Defects, Congenital/blood , MicroRNAs/blood , Proto-Oncogene Proteins c-bcl-2/blood , Repressor Proteins/blood , Animals , Apoptosis/genetics , Biomarkers/blood , Caspase 3/genetics , Cell Hypoxia/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL