Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Ethnopharmacol ; 269: 113720, 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33358858

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Jiajian Guishen Formula (JJGSF), which is a prescription of Traditional Chinese Medicine (TCM), has been reported to be useful in the treatment of premature ovarian insufficiency (POI). AIM OF THE STUDY: To investigate the therapeutic effects of JJGSF on the treatment of POI induced by 4-vinylcyclohexene diep-oxide (VCD), an endocrine-disrupting chemical (EDC), and to elucidate the potential mechanism. MATERIALS AND METHODS: Female 8-week-old ICR mice (N = 72) were randomized into six groups, containing the Model group, Control group, three JJGSF groups, and Progynova group which was served as a positive control. After model establishment by VCD, the Progynova group were given a daily intragastric administration of Progynova, and the three JJGSF groups (high dose group, medium dose group and low dose group) received a daily intragastric administration of JJGSF at doses of 9, 4.5 and 2.25 g/kg for four weeks. The general growth of the mice was observed and the estrous cycles were examined. The serum hormone concentrations were measured by enzyme-linked immunosorbent assay (ELISA). To explore the potential mechanism of effect, the protein expressions of H3K9me3, HP1, and HMGA1/HMGA2 related to senescence-associated heterochromatic foci (SAHF), were determined by Immunofluorescence and Western blot analysis, respectively. RESULTS: After treating with JJGSF, the estrous cycles were improved significantly. The level of estrogen (E2) and anti-müllerian hormone (AMH) was increased and the ratio of follicle-stimulating hormone (FSH) to luteinizing hormone (LH) in serum was decreased significantly. Furthermore, a significant down-regulation of HMGA1/HMGA2 on protein level, a reduction distribution of HP1 and H3K9me3 in ovarian, and a lower fraction of SAHF-positive cells were observed after the administration with JJGSF, additionally effects showed a positive correlation with dosages. CONCLUSIONS: JJGSF could treat POI by the mechanism of inhibiting SAHF.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Heterochromatin/drug effects , Primary Ovarian Insufficiency/drug therapy , Aging , Animals , Anti-Mullerian Hormone/metabolism , Cellular Senescence/drug effects , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Cyclohexenes/toxicity , Cytokines/blood , Disease Models, Animal , Down-Regulation/drug effects , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Endocrine Disruptors/toxicity , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogens/metabolism , Estrous Cycle/drug effects , Female , Follicle Stimulating Hormone/blood , HMGA1a Protein/genetics , HMGA1a Protein/metabolism , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Histones/metabolism , Luteinizing Hormone/blood , Medicine, Chinese Traditional , Mice, Inbred ICR , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Vinyl Compounds/toxicity
2.
Nat Commun ; 11(1): 4063, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792525

ABSTRACT

The neuroendocrine hypothalamus is the central regulator of vital physiological homeostasis and behavior. However, the cellular and molecular properties of hypothalamic neural progenitors remain unexplored. Here, hypothalamic radial glial (hRG) and hypothalamic mantle zone radial glial (hmRG) cells are found to be neural progenitors in the developing mammalian hypothalamus. The hmRG cells originate from hRG cells and produce neurons. During the early development of hypothalamus, neurogenesis occurs in radial columns and is initiated from hRG cells. The radial glial fibers are oriented toward the locations of hypothalamic subregions which act as a scaffold for neuronal migration. Furthermore, we use single-cell RNA sequencing to reveal progenitor subtypes in human developing hypothalamus and characterize specific progenitor genes, such as TTYH1, HMGA2, and FAM107A. We also demonstrate that HMGA2 is involved in E2F1 pathway, regulating the proliferation of progenitor cells by targeting on the downstream MYBL2. Different neuronal subtypes start to differentiate and express specific genes of hypothalamic nucleus at gestational week 10. Finally, we reveal the developmental conservation of nuclear structures and marker genes in mouse and human hypothalamus. Our identification of cellular and molecular properties of neural progenitors provides a basic understanding of neurogenesis and regional formation of the non-laminated hypothalamus.


Subject(s)
Hypothalamus/cytology , Hypothalamus/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Cluster Analysis , Female , Genes, Tumor Suppressor , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Humans , In Situ Hybridization , Mammals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neurogenesis/genetics , Neurogenesis/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pregnancy
3.
Int J Oral Sci ; 12(1): 10, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32231210

ABSTRACT

Drug-induced gingival overgrowth (DIGO) is recognized as a side effect of nifedipine (NIF); however, the underlying molecular mechanisms remain unknown. In this study, we found that overexpressed miR-4651 inhibits cell proliferation and induces G0/G1-phase arrest in gingival mesenchymal stem cells (GMSCs) with or without NIF treatment. Furthermore, sequential window acquisition of all theoretical mass spectra (SWATH-MS) analysis, bioinformatics analysis, and dual-luciferase report assay results confirmed that high-mobility group AT-hook 2 (HMGA2) is the downstream target gene of miR-4651. Overexpression of HMGA2 enhanced GMSC proliferation and accelerated the cell cycle with or without NIF treatment. The present study demonstrates that miR-4651 inhibits the proliferation of GMSCs and arrests the cell cycle at the G0/G1 phase by upregulating cyclin D and CDK2 while downregulating cyclin E through inhibition of HMGA2 under NIF stimulation. These findings reveal a novel mechanism regulating DIGO progression and suggest the potential of miR-4651 and HMGA2 as therapeutic targets.


Subject(s)
Cell Proliferation , Gingiva , HMGA2 Protein/genetics , Mesenchymal Stem Cells , MicroRNAs/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HMGA2 Protein/metabolism , Humans , MicroRNAs/metabolism , Nifedipine/pharmacology
4.
EBioMedicine ; 44: 194-208, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31129098

ABSTRACT

BACKGROUND: Brain metastases are a major cause of death in patients with metastatic breast cancer. While surgical resection and radiation therapy are effective treatment modalities, the majority of patients will succumb from disease progression. We have developed a novel therapy for brain metastases that delivers athermal radiofrequency electromagnetic fields that are amplitude-modulated at breast cancer specific frequencies (BCF). METHODS: 27.12 MHz amplitude-modulated BCF were administered to a patient with a breast cancer brain metastasis by placing a spoon-shaped antenna on the anterior part of the tongue for three one-hour treatments every day. In preclinical models, a BCF dose, equivalent to that delivered to the patient's brain, was administered to animals implanted with either brain metastasis patient derived xenografts (PDXs) or brain-tropic cell lines. We also examined the efficacy of combining radiation therapy with BCF treatment. Additionally, the mechanistic underpinnings associated with cancer inhibition was identified using an agnostic approach. FINDINGS: Animal studies demonstrated a significant decrease in growth and metastases of brain-tropic cell lines. Moreover, BCF treatment of PDXs established from patients with brain metastases showed strong suppression of their growth ability. Importantly, BCF treatment led to significant and durable regression of brain metastasis of a patient with triple negative breast cancer. The tumour inhibitory effect was mediated by Ca2+ influx in cancer cells through CACNA1H T-type voltage-gated calcium channels, which, acting as the cellular antenna for BCF, activated CAMKII/p38 MAPK signalling and inhibited cancer stem cells through suppression of ß-catenin/HMGA2 signalling. Furthermore, BCF treatment downregulated exosomal miR-1246 level, which in turn decreased angiogenesis in brain environment. Therefore, targeted growth inhibition of breast cancer metastases was achieved through CACNA1H. INTERPRETATION: We demonstrate that BCF, as a single agent or in combination with radiation, is a novel treatment approach to the treatment of brain metastases. This paradigm shifting modality warrants further clinical trials for this unmet medical need.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium Channels, T-Type/metabolism , Calcium/metabolism , Magnetic Field Therapy , Animals , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Electromagnetic Fields , Female , Gene Expression Profiling , HMGA2 Protein , Humans , Immunohistochemistry , MAP Kinase Signaling System , Magnetic Field Therapy/methods , Mice , Models, Biological , Neoplastic Stem Cells/metabolism
5.
Int J Mol Sci ; 20(7)2019 Mar 27.
Article in English | MEDLINE | ID: mdl-30934671

ABSTRACT

We tested the hypothesis that Let-7d-3p contributes to cardiac cell protection during hypoxic challenge. Myoblast H9c2 cells and primary neonatal rat ventricular cardiomyocytes (NRVM) were transfected with five selected miRNA mimics. Both cell lines were subjected to 0.2% oxygen hypoxia. The protective effects of these miRNAs were determined by assessment of cell metabolic activity by CCK8 assay and measurement of lactate dehydrogenase (LDH) release as a marker of cell injury. Apoptosis and autophagy flux were assessed by Annexin V/7-AAD double staining and the ratio of LC3 II/I with Baf-A1 treatment, an autophagy flux inhibitor, respectively. Luciferase-reporter assay, RT-qPCR and Western blots were performed to identify the changes of relevant gene targets. Among five miRNA mimic transfections, Let-7d-3p increased CCK8 activity, and decreased LDH release in both H9c2 and NRVM during hypoxia. Apoptosis was significantly reduced in H9c2 cells transfected with Let-7d-3p mimic. Autophagy and autophagy flux were not affected. In silico, mRNAs of HMGA2, YY1, KLF9, KLF12, and MEX3C are predicted targets for Let-7d-3p. Luciferase-reporter assay confirmed that Let-7d-3p bound directly to the 3'-UTR region of HMGA2, MEX3C, and YY1, the down-regulations of these mRNAs were verified in both H9c2 and NRVM. The protein expression of HMGA2, but not others, was downregulated in H9c2 and NRVM. It is known that HMGA2 is a strong apoptosis trigger through the blocking of DNA repair. Thus, we speculate that the anti-apoptotic effects of Let-7d-3p mimic during hypoxia challenge are due to direct targeting of HMGA2.


Subject(s)
Cardiotonic Agents/metabolism , HMGA2 Protein/metabolism , MicroRNAs/metabolism , Myocardium/metabolism , Animals , Apoptosis , Autophagy , Base Sequence , Cell Line , Hypoxia/metabolism , Hypoxia/pathology , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Reproducibility of Results , YY1 Transcription Factor/metabolism
6.
PLoS One ; 8(4): e61931, 2013.
Article in English | MEDLINE | ID: mdl-23626755

ABSTRACT

Macrophage infiltration into adipose tissue is associated with obesity and the crosstalk between adipocytes and infiltrated macrophages has been investigated as an important pathological phenomenon during adipose tissue inflammation. Here, we sought to identify adipocyte mRNAs that are regulated by interaction with infiltrated macrophages in vivo. An anti-inflammatory vitamin, vitamin B6, suppressed macrophage infiltration into white adipose tissue and altered mRNA expression. We identified >3500 genes whose expression is significantly altered during the development of obesity in db/db mice, and compared them to the adipose tissue mRNA expression profile of mice supplemented with vitamin B6. We identified PTX3 and MMP3 as candidate genes regulated by macrophage infiltration. PTX3 and MMP3 mRNA expression in 3T3-L1 adipocytes was up-regulated by activated RAW264.7 cells and these mRNA levels were positively correlated with macrophage number in adipose tissue in vivo. Next, we screened adipose genes down-regulated by the interaction with macrophages, and isolated RASSF6 (Ras association domain family 6). RASSF6 mRNA in adipocytes was decreased by culture medium conditioned by activated RAW264.7 cells, and RASSF6 mRNA level was negatively correlated with macrophage number in adipose tissue, suggesting that adipocyte RASSF6 mRNA expression is down-regulated by infiltrated macrophages in vivo. Finally, this study also showed that decreased RASSF6 expression up-regulates mRNA expression of several genes, such as CD44 and high mobility group protein HMGA2. These data provide novel insights into the biological significance of interactions between adipocytes and macrophages in adipose tissue during the development of obesity.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , Macrophages/metabolism , Monomeric GTP-Binding Proteins/metabolism , Obesity/metabolism , RNA, Messenger/metabolism , Tumor Suppressor Proteins/metabolism , Adipocytes/drug effects , Adipocytes/pathology , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Animals , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Cell Communication/drug effects , Cell Line , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Macrophages/drug effects , Macrophages/pathology , Male , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Transgenic , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Obesity/genetics , Obesity/pathology , RNA, Messenger/genetics , Signal Transduction/drug effects , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Vitamin B 6/pharmacology
7.
Mol Cell Biol ; 23(24): 9104-16, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645522

ABSTRACT

The HMGA2 protein belongs to the HMGA family of architectural transcription factors, which play an important role in chromatin organization. HMGA proteins are overexpressed in several experimental and human tumors and have been implicated in the process of neoplastic transformation. Hmga2 knockout results in the pygmy phenotype in mice and in a decreased growth rate of embryonic fibroblasts, thus indicating a role for HMGA2 in cell proliferation. Here we show that HMGA2 associates with the E1A-regulated transcriptional repressor p120(E4F), interfering with p120(E4F) binding to the cyclin A promoter. Ectopic expression of HMGA2 results in the activation of the cyclin A promoter and induction of the endogenous cyclin A gene. In addition, chromatin immunoprecipitation experiments show that HMGA2 associates with the cyclin A promoter only when the gene is transcriptionally activated. These data identify the cyclin A gene as a cellular target for HMGA2 and, for the first time, suggest a mechanism for HMGA2-dependent cell cycle regulation.


Subject(s)
Cyclin A/genetics , HMGA2 Protein/metabolism , Adenovirus E4 Proteins/chemistry , Adenovirus E4 Proteins/genetics , Adenovirus E4 Proteins/metabolism , Animals , Base Sequence , Binding Sites , CHO Cells , Cell Cycle , Cell Line , Cell Transformation, Neoplastic , Cricetinae , DNA, Complementary/genetics , HMGA2 Protein/genetics , Humans , Mice , Models, Biological , NIH 3T3 Cells , Promoter Regions, Genetic , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcriptional Activation , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL