Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
BMC Pediatr ; 24(1): 209, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38521927

ABSTRACT

BACKGROUND: Tumor lysis syndrome (TLS) is a hematologic oncological emergency characterized by metabolic and electrolyte imbalances. On breakdown of tumor cells, enormous amounts of potassium, phosphate, and nucleic acids are released into systemic circulation. TLS mainly occurs during chemotherapy. However, there are rare incidences of spontaneous tumor lysis syndrome (STLS) prior to commencement of therapy. CASE PRESENTATION: In the case being reported, the child had just undergone a biopsy. As the incision was being closed, there was a sudden onset of high fever, arrhythmia, severe hyperkalemia, hypocalcemia, and acidosis. Following timely symptomatic treatment and continuous renal replacement therapy(CRRT), the child's laboratory results improved, and organ function was restored to normal. The final pathological diagnosis confirmed Burkitt lymphoma. The boy is currently on maintenance chemotherapy. CONCLUSIONS: TLS is a potentially life-threatening complication in hematologic oncology. Several important conclusions can be drawn from this case, reminding clinicians to: (1) be fully aware of the risk factors of TLS and evaluate the level of risk; (2) pay attention to the possibility of STLS during operation, if surgical procedures are necessary and operate with minimal trauma and in the shortest time possibly; (3) take preoperative prophylaxis actively for high-risk TLS patients, including aggressive fluid management and rational use of diuretics and uric-acid-lowering drugs. In addition, this case confirms the effectiveness of CRRT for severe STLS.


Subject(s)
Burkitt Lymphoma , Tumor Lysis Syndrome , Water-Electrolyte Imbalance , Male , Child , Humans , Burkitt Lymphoma/complications , Burkitt Lymphoma/diagnosis , Burkitt Lymphoma/therapy , Tumor Lysis Syndrome/diagnosis , Tumor Lysis Syndrome/etiology , Tumor Lysis Syndrome/therapy , Risk Factors , Biopsy/adverse effects
2.
Pediatr Surg Int ; 39(1): 181, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37061578

ABSTRACT

OBJECTIVE: The present research aimed to analyze the application of indocyanine green (ICG) fluorescence contrast technique in the resection of hepatoblastoma (HB) in children, and to discuss the use of ICG in the surgery of HB and the value of guidance. METHODS: We retrospectively analyzed the data of 23 children with HB resected using ICG fluorescence contrast technique at the Children's Hospital of Nanjing Medical University from June 2020 to September 2022, including 16 boys and 7 girls, aged 5 days to 80 months. The patients were administered with an ICG injection of 0.1 mg/kg around 24-48 h before surgery. The surgical margin was detected by real-time fluorescence imaging and confirmed by postoperative pathology. RESULTS: All primary lesions showed bright fluorescence in 23 HB cases. 22 had clear borders with normal liver tissue, while one neonatal case showed no difference between tumor and background. 13 anatomic resection and 10 non-anatomic resection were performed with ICG fluorescence navigation. The surface of the residual liver was scattered with multiple tumor fluorescence, which was then locally enucleated according to the fluorescence. 22 isolated specimens were dissected and fluorescently visualized. Pathology identified deformed, vacuolated and densely arranged hepatocytes resembling pseudo-envelope changes without tumor residual, due to the compression of the tissue at the site of circumferential imaging. CONCLUSION: The ring ICG fluorescence imaging of HB indicates the tumor resection boundary effectively, especially in multiple lesions cases.


Subject(s)
Hepatoblastoma , Liver Neoplasms , Male , Child , Female , Infant, Newborn , Humans , Hepatoblastoma/diagnostic imaging , Hepatoblastoma/surgery , Indocyanine Green , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/surgery , Liver Neoplasms/pathology , Retrospective Studies , Optical Imaging/methods , Fluorescence , Margins of Excision
3.
Int J Mol Sci ; 23(19)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36232686

ABSTRACT

Polycystic ovarian syndrome (PCOS) is a reproductive, endocrine, and metabolic disorder. Circulating markers of oxidative stress are abnormal in women with PCOS. There is a close relationship between oxidative stress and insulin resistance (IR). However, little information is available about oxidative stress in the skeletal muscles of those affected by PCOS. In this study, PCOS was induced in prepubertal C57BL/6J mice by injection with dehydroepiandrosterone. Oxidative stress biomarkers were then measured in both serum and skeletal muscles. The underlying mechanisms were investigated in C2C12 myotubes treated with testosterone (T). We discovered increased oxidative biomarkers, increased ROS production, and damaged insulin sensitivity in the skeletal muscles of mice with PCOS. High levels of T caused mitochondrial dysfunction and increased ROS levels through the androgen receptor (AR)-nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) signaling pathway in C2C12 cells. Treatment of C2C12 cells with an antioxidant N-acetylcysteine (NAC) decreased T-induced ROS production, improved mitochondrial function, and reversed IR. Administration of NAC to mice with PCOS improved insulin sensitivity in the skeletal muscles of the animals. Hyperandrogenism caused mitochondrial dysfunction and redox imbalance in the skeletal muscles of mice with PCOS. We discovered that oxidative stress contributed to skeletal muscle IR in PCOS. Reducing ROS levels may improve the insulin sensitivity of skeletal muscles in patients with PCOS.


Subject(s)
Insulin Resistance , Polycystic Ovary Syndrome , Acetylcysteine/metabolism , Animals , Antioxidants/metabolism , Biomarkers/metabolism , Dehydroepiandrosterone , Female , Humans , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , NADP/metabolism , Oxidative Stress , Oxidoreductases/metabolism , Polycystic Ovary Syndrome/metabolism , Reactive Oxygen Species/metabolism , Receptors, Androgen/metabolism , Testosterone/metabolism
4.
Biol Reprod ; 103(6): 1199-1208, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32813010

ABSTRACT

The development and maturity of follicles are regulated by sex hormones and growth factors. It has been proven that peri-ovarian adipose tissue (POAT) plays an important role in folliculogenesis and fertility in the female ICR and KM mice. The aim of the present study was to further investigate whether the removal of bilateral POAT affected follicular development and lipid metabolism in the female C57BL/6 J mice. Female C57BL/6 J mice at 6-week old were sham-operated (Sham) or removed bilateral POAT (Surgery). After 2 weeks, the mice were subjected to the body composition analysis and indirect calorimetry measurement. Our results show that the Surgery mice exhibited abnormal follicular development, including increased follicular dysplasia and atresia, decreased serum sex hormone levels, and abnormal expression of follicular development-related genes. Correspondingly, the endometrial thickness of the Surgery mice was less than the Sham mice. In addition, the Surgery mice had abnormal lipid metabolism, including reduced fat mass, increased energy expenditure, and up-regulated gene and protein expression involved in lipolysis. These data confirmed the importance of POAT in the follicular development in the female reproduction and suggested the contribution of POAT to the whole-body lipid metabolism.


Subject(s)
Adipose Tissue/physiology , Ovarian Follicle/growth & development , Animals , Body Composition , Body Weight , Estrogens/blood , Female , Follicle Stimulating Hormone/blood , Gonadotropins/pharmacology , Luteinizing Hormone/blood , Mice , Mice, Inbred C57BL , Progesterone/blood
5.
J Cell Physiol ; 234(7): 10576-10587, 2019 07.
Article in English | MEDLINE | ID: mdl-30443988

ABSTRACT

It has been suggested that circular RNAs play critical roles in natural growth and disease development. Nevertheless, whether the circular RNAs were related in Hirschsprung's disease (HSCR) remains unknown. Thus, we discovered the cir-CCDC66 was downregulated in HSCR compared with the normal gut tissues. The cir-CCDC66 reduction might inhibit cells' proliferation and migration in vitro. Then, we found that DCX transcript was putative cir-CCDC66 competing endogenous RNA. Furthermore, the function of cir-CCDC66 as a sponge for miR-488-3p to regulate DCX RNA expression was demonstrated by immunoprecipitation and luciferase reporter assays. In conclusion, this is the first report revealing that cir-CCDC66 modulates DCX expression through sponging miR-488-3p and thus participates in the onset of HSCR.


Subject(s)
Eye Proteins/genetics , Hirschsprung Disease/genetics , MicroRNAs/genetics , Microtubule-Associated Proteins/genetics , Neuropeptides/genetics , RNA, Circular/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Doublecortin Domain Proteins , Doublecortin Protein , Female , Gene Expression Regulation/genetics , HEK293 Cells , Hirschsprung Disease/pathology , Humans , Infant , Male , RNA-Binding Proteins/genetics
6.
Mol Reprod Dev ; 86(4): 370-378, 2019 04.
Article in English | MEDLINE | ID: mdl-30633842

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common endocrinopathy and an important metabolic disorder in women of reproductive age. Insulin resistance (IR) is one of its most important clinical features in patients with PCOS. Androgen excess-induced mitochondrial dysfunction contributes to skeletal muscle IR in dehydroepiandrosterone (DHEA)-induced PCOS mice. The effect of androgen excess on the skeletal muscle, however, is incompletely characterized. A nontargeted metabolomics approach was thus applied to analyze the metabolites in skeletal muscle of DHEA-induced PCOS mice. Data from metabolomic analysis revealed the significant changes in 32 metabolites and the marked impact of five metabolic pathways. ATP production was also found to be significantly reduced in skeletal muscle of DHEA mice. Combined with the quantification of type I and II myofibers and lipid measurement in the skeletal muscle of the mice, the results from the present study supported the role of mitochondrial impairment rather than lipid accumulation in the pathogenesis of skeletal muscle IR in DHEA-induced PCOS mice. In summary, we show here for the first time the profile of the metabolites in the skeletal muscle of DHEA-induced PCOS mice which exhibit IR. The work would help better understand the pathology of skeletal muscle IR in PCOS.


Subject(s)
Dehydroepiandrosterone/adverse effects , Metabolomics , Muscle, Skeletal/metabolism , Polycystic Ovary Syndrome/metabolism , Animals , Dehydroepiandrosterone/pharmacology , Disease Models, Animal , Female , Humans , Mice , Muscle, Skeletal/pathology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/pathology
7.
J Cell Mol Med ; 22(9): 4045-4055, 2018 09.
Article in English | MEDLINE | ID: mdl-29956457

ABSTRACT

Researches over the past decade suggest that lipopolysaccharide is a dominant driver of gastrointestinal motility and could damage the enteric neuron of rat or porcine. However, it remains poorly defined whether LPS participates in Hirschsprung's disease (HSCR). Here, we discovered that LPS increased in HSCR tissues. Furthermore, LPS treatment suppressed the proliferation and differentiation of neural precursor cells (NPCs) or proliferation and migration of human 293T cells. ADAR2 (adenosine deaminase acting on RNA2)-mediated post-transcriptional adenosine-to-inosine RNA editing promotes cancer progression. We show that increased LPS activates ADAR2 and subsequently regulates the A-to-I RNA editing which suppresses the miR-142 expression. RNA sequencing combined with qRT-PCR suggested that ADAR2 restrain cell migration and proliferation via pri-miR-142 editing and STAU1 up-regulation. In conclusion, the findings illustrate that LPS participates in HSCR through the LPS-ADAR2-miR-142-STAU1 axis.


Subject(s)
Adenosine Deaminase/genetics , Cytoskeletal Proteins/genetics , Hirschsprung Disease/genetics , Lipopolysaccharides/metabolism , MicroRNAs/genetics , Neural Stem Cells/metabolism , RNA-Binding Proteins/genetics , Adenosine Deaminase/metabolism , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Cytoskeletal Proteins/metabolism , Female , HEK293 Cells , Hirschsprung Disease/metabolism , Hirschsprung Disease/pathology , Humans , Infant , Intestines/pathology , Male , Mice , Mice, Inbred ICR , MicroRNAs/metabolism , Neural Stem Cells/pathology , RNA Editing , RNA-Binding Proteins/metabolism , Signal Transduction
9.
Biol Reprod ; 95(4): 79, 2016 10.
Article in English | MEDLINE | ID: mdl-27557647

ABSTRACT

Patients with polycystic ovary syndrome (PCOS) can suffer from psychological disorders, among which depression is the most commonly diagnosed. However, the pathogenesis is still unclear. The aims of the present study were to investigate the behaviors of dehydroepiandrosterone (DHEA)-induced PCOS mice, the effects of high-fat diet (HFD) on mouse behaviors, and the underlying mechanism. Prepubertal C57BL/6 mice (25 days of age) were divided into four groups and injected daily with the vehicle sesame oil or DHEA on the normal chow or a 60% HFD for 20 consecutive days. Depression-like behavior of the mice was examined using a forced swim test, tail suspension test, and open field test. Thereafter, the animals were killed and four brain regions were collected. The brain levels of monoamines and their metabolites, including norepinephrine, serotonin, 5-hydroxy-3-indolacetic acid, dopamine, and 3,4-hydroxyphenylacetic acid, were analyzed by HPLC. Our data show that DHEA-treated mice exhibited depression-like behavior according to the results from behavioral assessment. The brain contents of monoamines and/or their metabolites decreased in DHEA-treated mice compared with controls. HFD did not seem to markedly affect the behavioral responses, the brain monoamines, or their metabolites in the mice. These findings suggest that DHEA treatment induced depression-like behavior in PCOS mice, possibly through down-regulation of brain monoamines and/or their metabolites, which implies the contribution of hyperandrogenism to the psychological symptoms of women with PCOS.


Subject(s)
Behavior, Animal/drug effects , Dehydroepiandrosterone/toxicity , Depression/etiology , Polycystic Ovary Syndrome/psychology , Animals , Behavior, Animal/physiology , Biogenic Monoamines/metabolism , Brain/drug effects , Brain/metabolism , Depression/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Down-Regulation/drug effects , Female , Humans , Mice , Mice, Inbred C57BL , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/etiology , Tissue Distribution
10.
Discov Oncol ; 15(1): 174, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38761221

ABSTRACT

A boy aged 55 months was diagnosed with stage IV Neuroblastoma (NB) of the right adrenal gland 2 years ago. Preoperative chemotherapy was given and he was then treated with retroperitoneal tumor resection and lymph node dissection. After surgery, the children were transferred to the Hemato-Oncology Department for chemotherapy according to the high-risk group NB, with outpatient follow-up every 6 months. In the second postoperative year, abdominal computed tomography (CT) scan revealed a rounded hypodense area in the upper part of the right posterior lobe of the liver, with marked inhomogeneous enhancement in the venous phase after enhancement, which was surgically resected, and postoperative pathology confirmed inflammatory myofibroblastic tumor (IMT) of liver. The patient was not given any special treatment after surgery. In this study, whole transcriptome sequencing was performed on the postoperative specimen of adrenal NB and the specimen of IMT of liver. This unusual case emphasizes the need for close monitoring of second tumor development in NB survivors even in the absence of known predisposing factors.

11.
Clin Kidney J ; 17(1): sfad277, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38223339

ABSTRACT

Background: Denys-Drash syndrome (DDS) is a rare disease characterized with pseudohermaphroditism, nephroblastoma (also known as Wilms tumor), and diffuse mesangial sclerosis. The therapy for DDS is largely supportive, i.e. surgery and chemotherapy for Wilms tumor and renal replacement therapy. Due to the limited understanding of the pathogenesis, precision therapy for DDS is yet to be explored. We sought to explore the cellular components and interactions in kidney tissues from an infant with DDS. Methods: Whole-exome sequencing was performed to examine the mutations associated with DDS. Single-cell RNA sequencing (scRNA-seq) was performed to explore the heterogenicity of kidney tissue samples. Results: A 6-month-old infant with bilateral Wilms tumors and genital ambiguity was diagnosed as having DDS. Whole exome sequencing revealed a novel de novo mutation (p.F185fs*118) in exon 1 of WT1. scRNA-seq was performed in tissue samples from bilateral Wilms tumors and the normal kidney from this infant. Fibroblasts, myocytes, epithelial cells, endothelial cells, and mononuclear phagocytes (MPs) ranked at the top of the 31 135 total cells. Fibroblasts and myocytes were dominant in the Wilms tumor samples. In contrast, most epithelial cells and endothelial cells were found in normal kidney tissues. CD44 and TUBA1A were significantly changed in myocyte subclusters, which may contribute to chemotherapy drug resistance. Macrophages intensively interacted with cancerous cells, including fibroblasts, epithelial cells, and myocytes. Conclusions: A novel mutation (p.F185fs*118) in exon 1 of WT1 was identified in an infant with DDS. scRNA-Seq revealed the heterogenicity of cellular components in Wilms tumors and kidney tissues, shedding light on the pathogenesis of DDS.

12.
Gene ; 908: 148292, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38369247

ABSTRACT

Hepatoblastoma (HB) is the most common malignant tumor in children under 5 years old, but its pathogenesis remains unclear. Nur77 has been reported to be an important regulator for cancer progression in various cancer types. This study found that Nur77 was downregulated in HB tumors, compared with paracancer tissue. Knockout or overexpression of Nur77 in HB tumor cell line HepG2 and HuH6 could significantly enhance or inhibit the proliferation, migration and invasion of tumor cells both in vitro and in vivo. Further studies illustrated that Nur77 regulated the proliferation of tumor cells by affecting the expression of ß-catenin. Nur77 agonist Csn-B effectively enhanced the therapeutic effect of cisplatin on HB tumors both in vitro and in vivo. This study confirms that Nur77 may act as an oncogene in HB tumors and mediate the progression of HB by inhibiting the expression of ß-catenin, which provides a new targeted therapy for the clinical treatment of HB patients; meanwhile, the combination of Nur77 agonist and cisplatin treatment may improve the chemotherapeutic efficacy of HB patients, which provides a new idea for the improvement of the clinical prognosis of HB patients.


Subject(s)
Hepatoblastoma , Liver Neoplasms , Child , Humans , Child, Preschool , Hepatoblastoma/drug therapy , Hepatoblastoma/genetics , Cisplatin/pharmacology , Cisplatin/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Cell Line, Tumor , Cell Proliferation
13.
Cell Death Discov ; 9(1): 326, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37658042

ABSTRACT

Prostate cancer is one of the most common malignancies in men, which has been considered a public health threat. KIF15 is a kind of driver protein, and its abnormal expression is closely related to the occurrence and development of malignant tumors. The purpose of the study was to explore the significance and role of KIF15 in prostate cancer and to show some potential value for prostate cancer. Immunohistochemistry analysis showed that KIF15 was highly expressed in prostate cancer tissues, which was also positively correlated with T Infiltrate. The loss-of-function and gain-of-function assays based on prostate cancer cells indicated that the change in KIF15 expression could significantly affect cell proliferation, tumorigenesis, migration, and cell apoptosis. The inhibition of prostate cancer development by KIF15 knockdown was also assured in vivo. The Human Apoptosis Antibody Array showed that CD40L, cytoC, DR6, and p21 were up-regulated upon KIF15 knockdown, while IGF-I and Survivin were down-regulated. Moreover, the involvement of the PI3K/Akt pathway in the KIF15-mediated regulation of prostate cancer was preliminarily proved. In summary, KIF15 was identified to play an important role in the development or biological progress of prostate cancer and is considered to possess the potential to be used as a therapeutic target.

14.
Bioengineered ; 13(1): 1858-1871, 2022 01.
Article in English | MEDLINE | ID: mdl-35012429

ABSTRACT

Glioma is one of the leading causes of tumor-related deaths worldwide, but its potential mechanism remains unclear. This study aimed to explore the biological role and potential mechanism of argininosuccinate synthase 1 (ASS1) in glioma. The relative expression levels of ASS1 in glioma specimens and cell lines were calculated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blotting. The biological functions of ASS1 were demonstrated using the 5-ethynyl-2'-deoxyuridine (EdU) assay, transwell assay, and in vivo experiments. In addition, methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP), and luciferase reporter assays were performed to explore the molecular mechanism of ASS1 in glioma. ASS1 expression levels were found to be downregulated in glioma specimens and cell lines. Functionally, we confirmed that ASS1 inhibited glioma cell proliferation, migration, invasion, and growth both. Furthermore, we found that ASS1 was a target of N(6)-adenosine-methyltransferase-14 (METTL14)-mediated N6-methyladenosine (m6A) modification. Overexpression of METTL14 markedly elevated ASS1 mRNA m6A modification and suppressed ASS1 mRNA expression. We also revealed that METTL14-mediated ASS1 mRNA degradation relied on the YTH m6A RNA-binding protein 2 (YTHDF2)-dependent pathway. We confirmed that decreased ASS1 expression promoted the cell proliferation, migration, and invasion in glioma, and that the METTL14/ASS1/YTHDF2 regulatory axis may be an effective therapeutic target for glioma.


Subject(s)
Adenosine/analogs & derivatives , Argininosuccinate Synthase/genetics , Brain Neoplasms/pathology , Glioma/pathology , Methyltransferases/genetics , RNA-Binding Proteins/genetics , Adenosine/metabolism , Animals , Argininosuccinate Synthase/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Case-Control Studies , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Humans , Male , Methyltransferases/metabolism , Mice , Neoplasm Transplantation , Prognosis , RNA-Binding Proteins/metabolism , Survival Analysis
15.
Front Genet ; 12: 820936, 2021.
Article in English | MEDLINE | ID: mdl-35116058

ABSTRACT

Circular RNAs (circRNAs) are non-coding RNAs with covalent closed-loop structures and are widely distributed in eukaryotes, conserved and stable as well as tissue-specific. Malignant solid tumors pose a serious health risk to children and are one of the leading causes of pediatric mortality. Studies have shown that circRNAs play an important regulatory role in the development of childhood malignant solid tumors, hence are potential biomarkers and therapeutic targets for tumors. This paper reviews the biological characteristics and functions of circRNAs as well as the research progress related to childhood malignant solid tumors.

16.
Front Neurol ; 12: 633637, 2021.
Article in English | MEDLINE | ID: mdl-34163418

ABSTRACT

Objective: The genetic aetiology of epileptic encephalopathy (EE) is growing rapidly based on next generation sequencing (NGS) results. In this single-centre study, we aimed to investigate a cohort of Chinese children with early infantile epileptic encephalopathy (EIEE). Methods: NGS was performed on 50 children with unexplained EIEE. The clinical profiles of children with pathogenic variants were characterised and analysed in detail. Conservation analysis and homology modelling were performed to predict the impact of STXBP1 variant on the protein structure. Results: Pathogenic variants were identified in 17 (34%) of 50 children. Sixteen variants including STXBP1 (n = 2), CDKL5 (n = 2), PAFAH1B1, SCN1A (n = 9), SCN2A, and KCNQ2 were de novo, and one (PIGN) was a compound heterozygous variant. The phenotypes of the identified genes were broadened. PIGN phenotypic spectrum may include EIEE. The STXBP1 variants were predicted to affect protein stability. Significance: NGS is a useful diagnostic tool for EIEE and contributes to expanding the EIEE-associated genotypes. Early diagnosis may lead to precise therapeutic interventions and can improve the developmental outcome.

17.
Oncotarget ; 9(15): 11905-11921, 2018 Feb 23.
Article in English | MEDLINE | ID: mdl-29552281

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common endocrinopathy in women of reproductive age and also an important metabolic disorder associated with insulin resistance (IR). Hyperandrogenism is a key feature of PCOS. However, whether hyperandrogenism can cause IR in PCOS remains largely unknown. The mammalian target of rapamycin complex 1 (mTORC1) and its regulated autophagy are closely associated with IR. In the present study, we investigated the role of mTORC1-autophagy pathway in skeletal muscle IR in a dehydroepiandrosterone (DHEA)-induced PCOS mouse model. DHEA-treated mice exhibited whole-body and skeletal muscle IR, along with the activated mTORC1, repressed autophagy, impaired mitochondria, and reduced plasma membrane glucose transporter 4 (GLUT4) expression in skeletal muscle of the mice. In cultured C2C12 myotubes, treatment with high dose testosterone activated mTORC1, reduced autophagy, impaired mitochondria, decreased insulin-stimulated glucose uptake, and induced IR. Inhibition of mTORC1 or induction of autophagy restored mitochondrial function, up-regulated insulin-stimulated glucose uptake, and increased insulin sensitivity. On the contrary, inhibition of autophagy exacerbated testosterone-induced impairment. Our findings suggest that the mTORC1-autophagy pathway might contribute to androgen excess-induced skeletal muscle IR in prepubertal female mice by impairing mitochondrial function and reducing insulin-stimulated glucose uptake. These data would help understanding the role of hyperandrogenism and the underlying mechanism in the pathogenesis of skeletal muscle IR in PCOS.

18.
Cell Cycle ; 17(4): 459-467, 2018.
Article in English | MEDLINE | ID: mdl-29429387

ABSTRACT

Recently studies reported that long non-coding RNAs (lncRNAs) may take part in a lot of congenital diseases, meanwhile, Hirschsprung's disease (HSCR) is a major congenital digestive tract malformation. Nevertheless whether lncRNAs participate in the occurrence of HSCR and how it contributes to this disease are still unknown. LOC100507600 was selected from our gene expression microarray data obtained from bowel tissues from HSCR patients and negative controls. Subsequently, we used qRT-PCR to prove the result in 64 pairs of HSCR disease bowel stenosis tissues and negative controls. Transwell assay, CCK-8 assay and flow cytometry were employed to explore whether cellular functions change after knocking down the LOC100507600 in SH-SY5Y cell and human 293T cell. Dual-luciferase reporter assay was used to confirm the competitive relationship between BMI1 and LOC100507600 through their association with hsa-miR128-1-3p. Protein extraction and Western blotting were used to further confirm the relationship between LOC100507600 and BMI1. We found that LOC100507600 was obvious reduced in tissues from HSCR patients with noteworthy correlation with BMI1. Furthermore, Downregulation of LOC100507600 repressed cell migration and proliferation and didn't affect cell apoptosis or cycle. Dual-luciferase reporter assay, qRT-PCR and Western blotting assay verified that LOC100507600 serves as a competitive endogenous RNA of miR128-1-3p and down-regulates BMI1 expression by sponging miR128-1-3p in HSCR. In sum, our study researches the potential diagnostic value of LOC100507600 in HSCR and deduces that LOC100507600 can contributes to HSCR as a competitive endogenous RNA to regulate BMI1 expression by sponging miR128-1-3p.


Subject(s)
Hirschsprung Disease/pathology , MicroRNAs/metabolism , Polycomb Repressive Complex 1/metabolism , RNA, Long Noncoding/metabolism , Antagomirs/metabolism , Apoptosis , Area Under Curve , Cell Line, Tumor , Cell Movement , Child , Female , HEK293 Cells , Hirschsprung Disease/diagnosis , Hirschsprung Disease/genetics , Humans , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/chemistry , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , ROC Curve
19.
Gene ; 658: 96-104, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29477871

ABSTRACT

Hirschsprung's disease (HSCR) is a complex disorder with multiple pathogenic gene mutations. Protocadherin alpha 9 (PCDHA9) was identified as a potential candidate gene for HSCR by whole-exome sequencing in a Chinese family. Sanger sequencing in 298 HSCR cases revealed two sporadic Chinese patients with a novel missence PCDHΑ9 mutation (NM_031857; c.1280C > T[p.Ala427Val]) and one sporadic Chinese patient with another novel missence PCDHΑ9 mutation (c.1425C > G[p.Phe475Leu]).The silico predictions and 3D modeling suggest the deleterious effect of identified mutations on protein function. Immunohistochemistry analysis showed PCDHΑ9 was predominantly expressed in the myenteric plexus of human colon tissues. For mouse embryos, PCDHΑ9 was expressed in the stomach but rarely seen in the intestine during E10.5-12.5, then obviously expressed in the intestinal mucosa at E13.5 and extensively expressed in intestinal muscularis and mucosa at E14.5. Moreover, the down-regulation of PCDHΑ9 in the SH-SY5Y cell line promoted the proliferation and migration rate but inhibited the apoptotic rate. In summary, PCDHΑ9 is potentially related to HSCR and the clustered protocadherins (Pcdhs) may involve in the enteric nervous system (ENS) ontogeny.


Subject(s)
Cadherins/genetics , Hirschsprung Disease/genetics , Mutation, Missense , Amino Acid Substitution , Animals , Cells, Cultured , DNA Mutational Analysis , Diseases in Twins/genetics , Embryo, Mammalian , Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Female , Humans , Male , Mice , Mice, Inbred ICR , Pedigree , Pregnancy , Siblings , Twins, Monozygotic
20.
Cell Prolif ; 51(5): e12489, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30062828

ABSTRACT

OBJECTIVES: Emerged evidence demonstrates that long non-coding RNAs (lncRNAs) may play quintessential regulatory roles in the cellular processes, tumourigenesis and the development of disease. Though focally amplified lncRNA on chromosome 1 (FAL1) has been identified to have crucial functions in many diseases, its biological mechanism in the development of Hirschsprung's disease (HSCR) still remains unknown. MATERIALS AND METHODS: The expression levels of FAL1 in HSCR aganglionic tissues and matched normal specimens were detected by quantitative real-time PCR (qRT-PCR). Cell proliferation and migration were detected by Cell Counting Kit-8 (CCK-8) assay, Ethynyl-deoxyuridine (EdU) assay and transwell assay relatively. Cell cycle and apoptosis were assessed using flow cytometer analysis. Moreover, the novel targets of FAL1 were confirmed with the help of bioinformatics analysis and dual-luciferase reporter assay. Western blot assay as well as RNA immunoprecipitation (RIP) assay was conducted to investigate the potential mechanism. RESULTS: FAL1 expression was markedly down-regulated in HSCR aganglionic tissues and decreased FAL1 expression was associated with the diagnosis of HSCR. Cell functional analyses indicated that FAL1 overexpressing notably promoted cell proliferation and migration, while down-regulation of FAL1 suppressed cell proliferation and migration. Additionally, Flow cytometry assay demonstrated that knockdown of FAL1 induced markedly cell cycle stalled in the G0/G1 phase. Furthermore, FAL1 could positively regulate AKT1 expression by competitively binding to miR-637. CONCLUSIONS: These results illuminated that FAL1 may work as a ceRNA to modulate AKT1 expression via competitively binding to miR-637 in HSCR, suggesting that it may be clinically valuable as a biomarker of HSCR.


Subject(s)
Down-Regulation/genetics , Hirschsprung Disease/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Long Noncoding/genetics , Apoptosis/genetics , Cell Cycle/genetics , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Computational Biology/methods , Female , HEK293 Cells , Humans , Infant , Male , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL