Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Int J Infect Dis ; 145: 107074, 2024 May 09.
Article En | MEDLINE | ID: mdl-38734057

OBJECTIVES: The prevalence of respiratory infectious diseases has changed in the post-COVID-19 epidemic era, and mycoplasma pneumoniae (MP) infection in children has attracted wide attention. METHODS: Children hospitalized for pneumonia in Wuhan, China, in 2023 were enrolled. Respiratory secretions were obtained for the targeted next-generation sequencing (tNGS) including mutation of MP. Pulmonary inflammation was divided into bronchopneumonia and pulmonary consolidation/atelectasis according to lung computed tomography imaging. RESULTS: Of the 667 pediatric pneumonia, 478 were MP positive (72%). The positive rate of MP detected by tNGS increased from April, and MP had become the primary pathogen of pneumonia in children in 2023. The 23S rRNA mutations were all A2063G, accounting for 85% of detected MP. The clinical symptoms of the mutant and wild-type strains were similar, with half of them experiencing atelectasis and lung consolidation. Early bronchoscopic lavage combined with azithromycin in pediatric pulmonary consolidation was an effective therapy strategy, which could be an alternative selection to MP pneumonia treatment. CONCLUSIONS: A2063G mutant strain MP was the primary pathogen of mycoplasma pneumoniae in children recently, which was often complicated by extra-pulmonary symptoms and complications.

2.
J Vasc Access ; : 11297298241245066, 2024 Apr 06.
Article En | MEDLINE | ID: mdl-38581270

Peripherally inserted central catheters are widely used in patients with extensive burns, with the guidelines recommending insertion through unburned skin. This case report describes a patient who was burned over 88% of their surface area and suffered severe inhalation injury. For him, the popliteal vein was the only vein on unburned skin available for catheter catheterization. Based on evidence, we successfully placed a peripherally inserted central catheter through the popliteal vein under ultrasound while the patient was in the prone position and avoided associated complications.

3.
Int J Biol Macromol ; 238: 124111, 2023 May 31.
Article En | MEDLINE | ID: mdl-36948330

Okra (Abelmoschus esculentus (L.) Moench) is rich in various bioactive ingredients and used as a medicinal plant in traditional medicine. In the present study, to find the polysaccharide with anti-lipotoxicity effects from okra and clarify its structure, a pectin OP-1 was purified from okra, which had a backbone containing →4)-α-GalpA-(1 â†’ residues, and 1,5-Ara linked the main chain through the O-3 of the residue →3,4)-α-GalpA-(1→, and the C-6 of residue 1, 4-α-GalpA replaced by methyl ester. In vitro experiments showed that OP-1 pretreatment alleviated oleic acid (OA)-induced lipid accumulation, ROS generation, apoptosis, transaminase leakage, and inflammatory cytokine secretion in HepG2 cells, resulting in reduced lipotoxicity. Further molecular results revealed that OP-1 increased Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation and affected the expression of AMPK downstream targets, including inhibit SREBP1c and FAS, as well as activate CPT-1A. Impressively, AMPK inhibitor dorsomorphin (Compound C) blocked the effects of OP-1 against lipotoxicity. The effects of OP-1 on lipid metabolism were also diminished by dorsomorphin. Our results demonstrated that OP-1 possesses a potent function in preventing lipotoxicity via regulating AMPK-mediated lipid metabolism and provide a novel insight into the future utilization of okra polysaccharide.


Abelmoschus , Pectins , Pectins/pharmacology , Abelmoschus/chemistry , AMP-Activated Protein Kinases , Polysaccharides/pharmacology , Polysaccharides/chemistry , Antioxidants/chemistry
4.
Chem Biol Interact ; 368: 110250, 2022 Dec 01.
Article En | MEDLINE | ID: mdl-36347319

It is well known that the excessive accumulation of lipid in hepatocytes is one of the important causes of non-alcoholic fatty liver disease (NAFLD). The purpose of this study was to explore the effects of isosilybin on lipid metabolism in free fatty acids (FFAs) or TO901317-induced HepG2 cells. Cells were treated with FFAs (oleic acid: palmitic acid, 2:1) or TO901317 to induce steatosis in vitro. Intracellular triglyceride (TG) content was quantified using commercial assay kits. The mRNA and protein expression of genes involved in fatty acid uptake, synthesis and oxidation were analyzed by RT-qPCR and western blotting. Selected biological pathways regulated by isosilybin treatment were determined by GO and KEGG analysis. The results showed that isosilybin significantly reduced TG levels in FFAs- and TO901317-induced HepG2 cells. Further studies showed that isosilybin treatment decreased the mRNA and protein expression of lipid synthesis genes Srebp-1c, Pnpla3, Acc and Fas, as well as the mRNA expression of fatty acid uptake gene CD36, whereas increased the mRNA levels of lipid oxidation genes Pparα, Acox1 and Cpt1α, as well as the mRNA expression of lipid export gene Mttp, in FFAs-induced HepG2 cells. Moreover, TO901317 was employed to induce endogenous lipid synthesis and steatosis, and the expression of Srebp-1c and its target genes in TO901317-induced hepatocytes was basically similar to that in FFAs-induced hepatocytes following isosilybin treatment. We also observed the increased level of phosphorylated AMP kinase (AMPK) after isosilybin treatment, while this effect was reversed after further treatment with AMPK inhibitor, compound C. The results of GO and KEGG analysis indicated that the pathways of fatty acid and TG metabolism were regulated by isosilybin. Interestingly, we found that treatment with the diastereoisomer A of isosilybin increased TG level, while exposure to the diastereoisomer B of isosilybin decreased TG level in FFAs-induced HepG2 cells. The above results suggest that isosilybin can inhibit lipid synthesis and activate lipid oxidation through AMPK signaling pathway, thereby improving steatosis of hepatocytes, and isosilybin B is the basis of its active substance.


Lipogenesis , Non-alcoholic Fatty Liver Disease , Humans , Sterol Regulatory Element Binding Protein 1/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/metabolism , Lipid Metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Hep G2 Cells , Palmitic Acid/pharmacology , Fatty Acids, Nonesterified/metabolism , Fatty Acids , RNA, Messenger/metabolism
5.
Front Pediatr ; 8: 405, 2020.
Article En | MEDLINE | ID: mdl-32850529

The association of preterm or low birth weight (LBW) with the risk of metabolic syndrome is still unclear. This study aimed to assess the association between preterm or LBW and metabolic syndrome risk according to study or participants' characteristics. PubMed, Web of Science, and EMBASE were searched for epidemiologic studies on the association published up to April 30, 2020. Pooled odds ratio (ORs) and weighted mean differences (WMDs) with 95% confidence intervals (CIs) were calculated using the random-effects model. Low birth weight was associated with an increased risk of metabolic syndrome (OR, 1.37; 95% CI, 1.17-1.61). In the subgroup analysis by study design, the pooled ORs for LBW and metabolic syndrome in the cohort and cross-sectional studies were 1.79 and 1.22. In the subgroup analysis by sex, LBW was found to be associated with an increased risk of metabolic syndrome in pooled studies including both men and women or studies including only women. The association between premature birth and risk of metabolic syndrome was significant in cohort studies (OR, 1.72; 95% CI, 1.12-2.65). Also, LBW or preterm was significantly associated with a higher Homeostasis Model Assessment of Insulin Resistance (WMD, 0.28; 95% CI, 0.19-0.36). Low birth weight and preterm might be risk factors for metabolic syndrome.

6.
Pharmacogenomics ; 21(12): 863-870, 2020 08.
Article En | MEDLINE | ID: mdl-32559398

Aim: This study was conducted to investigate the effects of VKORC1, CYP2C9, CYP4F2 and EPHX1 and nongenetic factors on warfarin maintenance dose in a very elderly, frail Han-Chinese population. Materials & methods: 16 variants of VKORC1, CYP2C9, CYP4F2 and EPHX1 were genotyped. Univariate analysis and multivariable regression model were performed for the associations of gene variants and warfarin maintenance dose. Results & conclusion:EPHX1 rs2260863 nonvariant CC homozygotes required significantly lower daily warfarin dose than GC heterozygotes. In the multivariable model, VKORC1 rs9923231, CYP2C9 rs1057910, EPHX1 rs2260863, CYP4F2 rs2189784 and body surface area altogether explained 26.9% of dosing variability. This study revealed the main impact of genetic factors on warfarin response in this special population.


Asian People/genetics , Epoxide Hydrolases/genetics , Frail Elderly , Maintenance Chemotherapy/methods , Polymorphism, Single Nucleotide/genetics , Warfarin/administration & dosage , Aged , Aged, 80 and over , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Female , Humans , Maintenance Chemotherapy/adverse effects , Male , Warfarin/adverse effects
7.
Sheng Wu Gong Cheng Xue Bao ; 36(4): 763-771, 2020 Apr 25.
Article Zh | MEDLINE | ID: mdl-32347070

The recombinant adenoviruses expressing miR-22 (Ad-miR-22) was constructed and the effect of Ad-miR-22 on insulin signal pathway and glucose uptake in HepG2 cells was analyzed. MiR-22 gene was amplified by PCR from human hepatocytes and cloned into the pAdTrack-CMV vector to generate the shuttle plasmid pAdT-22. The positive colonies were confirmed by PCR and sequencing. The resultant shuttle plasmid was linearized with Pme I, followed by co-transformation into competent BJ5183 cells containing an adenoviral backbone plasmid (pAdEasy-1) to create the recombinant plasmid pAd-miR-22. After digested with Pac I, the linearized pAd-miR-22 was transfected into 293A packaging cell line to generate recombinant adenoviruses Ad-miR-22. HepG2 cells were infected with Ad-miR-22 or control Ad-GFP (adenoviruses expressing green fluorescent protein), and then the miR-22 expression levels were analyzed by qPCR. The result shows that adenovirus-mediated overexpression of miR-22 significantly decreased insulin-induced glucose uptake in HepG2 cells. Moreover, overexpression of miR-22 markedly decreased insulin-induced phosphorylation of GSK-3ß. miR-22 also increased the mRNA levels of gluconeogenic genes in HepG2 cells. Furthermore, Western blotting results indicate that the protein expression of SIRT1 decreased in Ad-miR-22 infected HepG2 cells as compared with Ad-GFP infected HepG2 cells. In summary, overexpressing of miR-22 significantly increased gluconeogenesis while decreased glucose uptake in HepG2 cells. The effect of miR-22 on glucose metabolism may be mediated by SIRT1.


Adenoviridae , Glucose , MicroRNAs , Adenoviridae/genetics , Glucose/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hep G2 Cells , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction/genetics , Transfection
8.
Chin Med J (Engl) ; 132(13): 1533-1540, 2019 Jul 05.
Article En | MEDLINE | ID: mdl-31205075

BACKGROUND: Advanced technology has become a valuable tool in etiological studies of intellectual disability/global developmental delay (ID/GDD). The present study investigated the role of genetic analysis to confirm the etiology in ID/GDD patients where the cause of the disease was uncertain in central China. METHODS: We evaluated 1051 ID/GDD children aged 6 months to 18 years from March 2009 to April 2017. Data concerning basic clinical manifestations were collected, and the method of etiology confirmation was recorded. Genome-wide copy number variations (CNVs) detection and high-throughput sequencing of exons in the targeted regions was performed to identify genetically-based etiologies. We compared the incidence of different methods used to confirm ID/GDD etiology among groups with differing degrees of ID/GDD using the Chi-square or Fisher exact probability test. RESULTS: We recruited 1051 children with mild (367, 34.9%), moderate (301, 28.6%), severe (310, 29.5%), and profoundly severe (73, 6.9%) ID/GDD. The main causes of ID/GDD in the children assessed were perinatal factors, such as acquired brain injury, as well as single gene imbalance and chromosomal gene mutation. We identified karyotype and/or CNVs variation in 46/96 (47.9%) of cases in severe ID/GDD patients, which was significantly higher than those with mild and moderate ID/GDD of 34/96 (35.4%) and 15/96 (15.6%), respectively. A total of 331/536 (61.8%) patients with clear etiology have undergone genetic analysis while 262/515 (50.9%) patients with unclear etiology have undergone genetic analysis (χ = 12.645, P < 0.001). Gene structure variation via karyotype analysis and CNV detection increased the proportion of children with confirmed etiology from 51.0% to 56.3%, and second-generation high-throughput sequencing dramatically increased this to 78.9%. Ten novel mutations were detected, recessive mutations in X-linked genes (ATPase copper transporting alpha and bromodomain and WD repeat domain containing 3) and dominant de novo heterozygous mutations in X-linked genes (cyclin-dependent kinase like 5, protocadherin 19, IQ motif and Sec7 domain 2, and methyl-CpG binding protein 2) were reported in the study. CONCLUSIONS: The present study indicates that genetic analysis is an effective method to increase the proportion of confirmed etiology in ID/GDD children and is highly recommended, especially in ID/GDD children with uncertain etiology.


Intellectual Disability/genetics , Adolescent , Child , Child, Preschool , China , DNA Copy Number Variations/genetics , Female , Genetic Testing , Humans , Infant , Infant, Newborn , Male , Mutation/genetics
9.
Endocrinology ; 160(6): 1363-1376, 2019 06 01.
Article En | MEDLINE | ID: mdl-30977789

Bioactive free IGF-I is critically important for growth. The bioavailability of IGF-I is modulated by the IGF-binding proteins (IGFBPs) and their proteases, such as pregnancy-associated plasma protein-A2 (PAPP-A2). We have created a mouse model with a specific mutation in PAPPA2 identified in a human with PAPP-A2 deficiency. The human mutation was introduced to the mouse genome via a knock-in strategy, creating knock-in mice with detectable protein levels of Papp-a2 but without protease activities. We found that the Pappa2 mutation led to significant reductions in body length (10%), body weight (10% and 20% in males and females, respectively), and relative lean mass in mice. Micro-CT analyses of Pappa2 knock-in femurs from adult mice showed inhibited periosteal bone expansion leading to more slender bones in both male and female mice. Furthermore, in the Pappa2 knock-in mice, insulin resistance correlated with decreased serum free IGF-I and increased intact IGFBP-3 concentrations. Interestingly, mice heterozygous for the knock-in mutation demonstrated a growth rate for body weight and length as well as a biochemical phenotype that was intermediate between wild-type and homozygous mice. This study models a human PAPPA2 mutation in mice. The mouse phenotype closely resembles that of the human patients, and it provides further evidence that the regulation of IGF-I bioavailability by PAPP-A2 is critical for human growth and for glucose and bone metabolism.


Blood Glucose/metabolism , Body Weight/genetics , Insulin Resistance/physiology , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/metabolism , Pregnancy-Associated Plasma Protein-A/genetics , Animals , Bone Density/physiology , Female , Femur/diagnostic imaging , Femur/metabolism , Male , Mice , Mice, Transgenic , Mutation , Pregnancy-Associated Plasma Protein-A/metabolism , X-Ray Microtomography
10.
J Endocr Soc ; 2(7): 646-656, 2018 Jul 01.
Article En | MEDLINE | ID: mdl-29942928

CONTEXT: Pregnancy-associated plasma protein A2 (PAPPA2) is a protease that cleaves IGF-binding protein (IGFBP)-3 and IGFBP-5, liberating free IGF-I. Five patients from two families with genetic mutations in PAPPA2 presented with growth retardation, elevated total IGF-I, and IGFBP-3 but decreased free IGF-I. OBJECTIVE: To determine whether plasma transfusion or recombinant human (rh)PAPPA2 could increase free IGF-I in patients with PAPPA2 deficiency or idiopathic short stature (ISS). DESIGN: Single patient interventional study combined with in vitro experimentation. SETTING: Academic medical center. PATIENTS: Three siblings with PAPPA2 deficiency and four patients with ISS. INTERVENTIONS: An adult female with PAPPA2 deficiency received a 20 mL/kg plasma transfusion. PAPPA2, intact IGFBP-3, and free and total IGF-I levels were monitored during 2 weeks. rhPAPPA2 was added to serum from patients with PAPPA2 deficiency and ISS in vitro for 4 hours. Intact IGFBP-3 and free IGF-I levels were assayed via ELISA. MAIN OUTCOME MEASURES: Free IGF-I concentrations. RESULTS: Plasma transfusion resulted in a 2.5-fold increase of free IGF-I levels on day 1 posttransfusion with a return to baseline during a 2-week period. In vitro studies demonstrated a dose-dependent increase in free IGF-I and decrease in intact IGFBP-3 after the addition of rhPAPPA2. The increase in free IGF-I was more pronounced in patients with PAPPA2 deficiency compared with those with ISS. CONCLUSIONS: PAPPA2 plays a key role in regulation of IGF-I bioavailability. rhPAPPA2 is a promising therapy to increase free IGF-I levels both in patients with PAPPA2 deficiency as well as in patients with ISS.

11.
Horm Res Paediatr ; 88(5): 364-370, 2017.
Article En | MEDLINE | ID: mdl-28675896

BACKGROUND: Short stature can be caused by mutations in a multitude of different genes. 3-M syndrome is a rare growth disorder marked by severe pre- and postnatal growth retardation along with subtle dysmorphic features. There have only been 2 prior reports of mutations in CCDC8 causing 3-M syndrome. METHODS: Two patients presenting with mild short stature underwent whole exome sequencing. The mutation was confirmed via Sanger sequencing. We compare the clinical characteristics of our 2 patients to patients previously reported with mutations in the same gene. RESULTS: Exome sequencing identified a homozygous frameshift mutation in CCDC8 in both patients. They presented with a much milder phenotype than previously described patients with the same mutation. CONCLUSION: In this study, we report a case of 2 sisters with relatively mild short stature who were found via exome sequencing to carry a previously reported homozygous mutation in CCDC8. These patients expand the anthropometric phenotype of 3-M syndrome and demonstrate the power of exome sequencing in the diagnosis of children with short stature. 3-M syndrome should be considered in children with mild skeletal abnormalities, normal/high growth hormone-IGF axis parameters, and normal intelligence.


Carrier Proteins/genetics , Dwarfism/diagnosis , Growth Disorders/diagnosis , Muscle Hypotonia/diagnosis , Mutation , Siblings , Spine/abnormalities , Child , Child, Preschool , Dwarfism/genetics , Female , Growth Disorders/genetics , Humans , Infant , Muscle Hypotonia/genetics , Severity of Illness Index
12.
J Huazhong Univ Sci Technolog Med Sci ; 35(6): 904-909, 2015 Dec.
Article En | MEDLINE | ID: mdl-26670444

Activated protein C (APC), a natural anticoagulant, has been reported to exert direct vasculoprotective, neural protective, anti-inflammatory, and proneurogenic activities in the central nervous system. This study was aimed to explore the neuroprotective effects and potential mechanisms of APC on the neurovascular unit of neonatal rats with intrauterine infection-induced white matter injury. Intraperitoneal injection of 300 µg/kg lipopolysaccharide (LPS) was administered consecutively to pregnant Sprague-Dawley rats at embryonic days 19 and 20 to establish the rat model of intrauterine infection- induced white matter injury. Control rats were injected with an equivalent amount of sterile saline on the same time. APC at the dosage of 0.2 mg/kg was intraperitoneally injected to neonatal rats immediately after birth. Brain tissues were collected at postnatal day 7 and stained with hematoxylin and eosin (H&E). Immunohistochemistry was used to evaluate myelin basic protein (MBP) expression in the periventricular white matter region. Blood-brain barrier (BBB) permeability and brain water content were measured using Evens Blue dye and wet/dry weight method. Double immunofluorescence staining and real-time quantitative PCR were performed to detect microglial activation and the expression of protease activated receptor 1 (PAR1). Typical pathological changes of white matter injury were observed in rat brains exposed to LPS, and MBP expression in the periventricular region was significantly decreased. BBB was disrupted and the brain water content was increased. Microglia were largely activated and the mRNA and protein levels of PAR1 were elevated. APC administration ameliorated the pathological lesions of the white matter and increased MBP expression. BBB permeability and brain water content were reduced. Microglia activation was inhibited and the PAR1 mRNA and protein expression levels were both down-regulated. Our results suggested that APC exerted neuroprotective effects on multiple components of the neurovascular unit in neonatal rats with intrauterine infection- induced white matter injury, and the underlying mechanisms might involve decreased expression of PAR1.


Animals, Newborn , Cerebrovascular Circulation , Protein C/metabolism , Animals , Blood-Brain Barrier , Brain Edema/metabolism , Female , Male , Rats , Rats, Sprague-Dawley
13.
Clin Sci (Lond) ; 129(4): 385-94, 2015 Aug.
Article En | MEDLINE | ID: mdl-25875376

Intrauterine exposure to hyperglycaemia may increase the risk of later-life metabolic disorders. Although the underlying mechanism is not fully understood, epigenetic dysregulation in fetal programming has been implicated. With regard to energy homoeostasis, PGC-1α (peroxisome-proliferator-activated receptor γ co-activator-1α, encoded by the PPARGC1A gene) plays a regulatory role in several biochemical processes. We hypothesized that maternal gestational glucose levels would positively correlate with DNA methylation of the PPARGC1A promoter in placental tissue. We undertook a cross-sectional study of 58 mothers who underwent uncomplicated Caesarean delivery in a university hospital. Maternal gestational glucose concentration was determined after a 75-g OGTT (oral glucose tolerance test) at 24-28 weeks of gestation. Placenta tissue and cord blood were collected immediately after delivery. Genomic DNA was extracted and thereafter bisulfite conversion was performed. After PCR amplification, the DNA methylation of the PPARGC1A promoter was quantified using a pyrosequencing technique. The protein level of PGC-1α was evaluated by Western blotting. For all participants as a whole, including the GDM (gestational diabetes mellitus) and normoglycaemia groups, the maternal gestational glucose level was positively correlated with placental DNA methylation, and negatively correlated with cord blood DNA methylation of the PPARGC1A promoter in a CpG site-specific manner. In the GDM group alone, the placental CpG site-specific methylation of the PPARGC1A promoter strongly correlated with gestational 2-h post-OGTT glycaemia. Epigenetic alteration of the PPAGRC1A promoter may be one of the potential mechanisms underlying the metabolic programming in offspring exposed to intrauterine hyperglycaemia.


Blood Glucose/metabolism , DNA Methylation , Diabetes, Gestational/blood , Diabetes, Gestational/genetics , Epigenesis, Genetic , Placenta/metabolism , Promoter Regions, Genetic , Transcription Factors/genetics , Adult , Biomarkers/metabolism , Case-Control Studies , CpG Islands , Cross-Sectional Studies , Diabetes, Gestational/diagnosis , Energy Metabolism , Female , Gestational Age , Humans , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pregnancy , Prenatal Exposure Delayed Effects , Transcription Factors/metabolism
14.
Int J Mol Med ; 35(5): 1199-212, 2015 May.
Article En | MEDLINE | ID: mdl-25777531

Maternal intrauterine inflammation or infection is an important risk factor for neonatal cerebral white matter injury (WMI) and future neurological deficits. Activated protein C (APC), a natural anticoagulant, has been shown to exhibit anti-inflammatory, anti-apoptotic, profibrinolytic and cytoprotective activities. Recent studies have demonstrated that the novel prothrombinase, fibrinogen-like protein 2 (fgl2), contributes to the pathogenesis of a number of inflammatory diseases through the generation of fibrin. Thus, we hypothesized that APC may regulate coagulant and inflammatory processes and improve brain injury in an experimental rat model of intrauterine inflammation-induced WMI. The animal model was established by the administration of an intraperitoneal injection of lipopolysaccharide (LPS) to pregnant Sprague-Dawley rats on embryonic day (E)17 and E18. APC was administered intraperitoneally 30 min after the second LPS injection. The expression of fgl2 and the pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1ß expression in the placentas and fetal brains was determined on E19. Nerve cell death, the brain water content and protease-activated receptor 1 (PAR1) and nuclear factor κB (NF-κB) p65 expression was detected in the fetal brains. WMI in the neonatal rat brains was evaluated by hematoxylin and eosin (H&E) staining and immunohistochemistry for myelin basic protein (MBP). The results revealed that APC markedly reduced the LPS-induced increase in fgl2 expression and fibrin deposition, as well as the production of the pro-inflammatory cytokines, TNF-α, IL-6 and IL-1ß, in the placentas and fetal brains. In addition, APC attenuated cerebral apoptosis and brain edema, downregulated PAR1 and NF-κB p65 expression in the fetal brains, and improved hypomyelination and structural disturbances in the periventricular area of the neonatal rat brains. Our observations provide evidence that APC attenuates fetal neuroinflammation and the associated secondary WMI in the developing brain by inhibiting the expression of fgl2 and pro-inflammatory mediators, suggesting that APC may be a potential therapeutic approach for intrauterine inflammation-induced neonatal brain injury.


Cytokines/genetics , Fibrinogen/genetics , Gene Expression Regulation , Leukoencephalopathies/etiology , Neuroprotective Agents/pharmacology , Protein C/pharmacology , Animals , Apoptosis/drug effects , Brain Edema/drug therapy , Brain Edema/etiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Down-Regulation , Female , Inflammation/complications , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Placenta Diseases/genetics , Placenta Diseases/metabolism , Placenta Diseases/pathology , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, PAR-1/metabolism , Transcription Factor RelA/metabolism
15.
Pediatr Res ; 77(5): 625-32, 2015 May.
Article En | MEDLINE | ID: mdl-25675425

BACKGROUND: Intrauterine growth restriction (IUGR) followed by postnatal accelerated growth (CG-IUGR) is associated with long-term adverse metabolic consequences, and an involvement of epigenetic dysregulation has been implicated. Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a key orchestrator in energy homeostasis. We hypothesized that CG-IUGR programed an insulin-resistant phenotype through the alteration in DNA methylation and transcriptional activity of PGC-1α. METHODS: A CG-IUGR rat model was adopted using maternal gestational nutritional restriction followed by infantile overnutrition achieved by reducing the litter size. The DNA methylation was determined by pyrosequencing. The mRNA expression and mitochondrial content were assessed by real-time PCR. The insulin-signaling protein expression was evaluated by western blotting. RESULTS: Compared with controls, the CG-IUGR rats showed an increase in the DNA methylation of specific CpG sites in PGC-1α, and a decrease in the transcriptional activity of PGC-1α, mitochondrial content, protein level of PI3K and phosphorylated-Akt2 in liver and muscle tissues. The methylation of specific CpG sites in PGC-1α was positively correlated with fasting insulin concentration. CONCLUSION: IUGR followed by infantile overnutrition programs an insulin-resistant phenotype, possibly through the alteration in DNA methylation and transcriptional activity of PGC-1α. The genetic and epigenetic modifications of PGC-1α provide a potential mechanism linking early-life nutrition insult to long-term metabolic disease susceptibilities.


Animal Nutrition Sciences , DNA Methylation , Epigenesis, Genetic , Fetal Growth Retardation/physiopathology , Insulin Resistance , Insulin/metabolism , Transcription Factors/metabolism , Animals , CpG Islands , DNA, Mitochondrial/metabolism , Disease Models, Animal , Fatty Acids/chemistry , Homeostasis , Liver/metabolism , Male , Muscles/metabolism , Oxygen/chemistry , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Phosphorylation , Rats , Sequence Analysis, DNA , Temperature , Triglycerides/metabolism
16.
Zhonghua Er Ke Za Zhi ; 52(2): 112-6, 2014 Feb.
Article Zh | MEDLINE | ID: mdl-24739722

OBJECTIVE: To explore clinical, radiographical and genetic characteristics of classical Hutchinson-Gilford progeria syndrome (HGPS). METHOD: Data of a case of HGPS diagnosed at Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology was analyzed and related literature was reviewed. RESULT: At the age of 8 months, the affected-infant presented with characteristic manifestation such as short stature, low weight, frontal bossing, alopecia, prominent scalp veins, micrognathia with a vertical midline groove in the chin, sclerodermatous skin, knee joints contracture with a horse-riding stance, and limited range of movement of ankle joints. Blood test showed blood platelet count (416-490) ×10(9)/L. Lower extremities MRI showed reduced subcutaneous fat. LMNA gene analysis showed that the affected-infant carried typical heterozygous mutation: c. 1824C>T (p. G608G), while his parents were normal. At the age of 13 months, X-rays showed short distal phalanges and clavicles with acro-osteolysis. After following up for 15 months, his appearance of progeria became more apparent. As far as we know, there are only 2 cases of classical HGPS confirmed by gene analysis in China. CONCLUSION: Classical HGPS should be considered when infants appeared with sclerodermatous skin. Genetic analysis could help to diagnose classical HGPS as early as possible and avoid unnecessary investigations. In addition, affected-infants need to be long term followed-up and provided genetic counseling.


Lamin Type A/genetics , Mutation/genetics , Progeria/diagnosis , Skin Diseases/diagnosis , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/pathology , DNA Mutational Analysis , Diagnosis, Differential , Hand/diagnostic imaging , Hand/pathology , Humans , Infant , Lower Extremity/diagnostic imaging , Lower Extremity/pathology , Male , Osteolysis, Essential/pathology , Progeria/genetics , Progeria/pathology , Retrospective Studies , Skin Diseases/genetics , Skin Diseases/pathology , Tomography, X-Ray Computed
17.
Gene ; 522(1): 102-6, 2013 Jun 10.
Article En | MEDLINE | ID: mdl-23562786

A 2.75-year-old Chinese boy presented with typical clinical features of pseudoachondroplasia, including disproportionate short-limb short stature, brachydactyly, genu varus and waddling gait. Radiologically, tubular bones were short with widened metaphyses, irregular and small epiphyses; anterior tonguing or beaking of vertebral bodies were characteristic. DNA sequencing analysis of the COMP gene revealed a heterozygous mutation (c.1511G>A, p.Cys504Tyr) in the patient but his parents were unaffected without this genetic change. The missense mutation (c.1511G>A) was not found in 100 healthy controls and has not been reported previously. Our findings expand the spectrum of known mutations in COMP leading to pseudoachondroplasia.


Achondroplasia/genetics , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Mutation, Missense , Achondroplasia/diagnosis , Asian People , Cartilage Oligomeric Matrix Protein , Child, Preschool , Genetic Predisposition to Disease , Heterozygote , Humans , Male , Matrilin Proteins
18.
Pediatr Res ; 73(3): 263-7, 2013 Mar.
Article En | MEDLINE | ID: mdl-23222907

BACKGROUND: Suppressor of cytokine signaling-1 and -3 (SOCS-1 and SOCS-3) are two important negative regulators in the insulin-signaling pathway, and their overexpression may aggravate insulin resistance. Subjects with insulin resistance are often obese and have increased expressions of SOCS-1 and SOCS-3. We speculated that SOCS-1 and SOCS-3 may be involved in abnormal deposition of adipose tissues during insulin resistance. METHODS: A catch-up growth intrauterine growth retardation (CG-IUGR) rat model with insulin resistance was established; mRNA and protein expression of SOCS-1, SOCS-3, the CCAAT/enhancer binding protein (C/EBPα), and peroxisome proliferator-activated receptor (PPARγ) in adipose tissue were measured by real-time PCR and western blot; plasmids carrying small hairpin RNAs (shRNAs) targeting the SOCS-1 and SOCS-3 genes were constructed and transfected into preadipocytes, which were then induced to mature. At 72 h after differentiation was induced, the expressions of C/EBPα and PPARγ, two important molecules promoting the differentiation of preadipocytes, were detected. RESULTS: Expressions of SOCS-1, SOCS-3, C/EBPα, and PPARγ were markedly increased in adipose tissues of CG-IUGR rats, whereas the expressions of C/EBPα and PPARγ were significantly reduced after gene silencing of SOCS-1 or SOCS-3 in adipocytes. CONCLUSION: Overexpression of SOCS-1 and SOCS-3 may enhance the expression of C/EBPα and PPARγ, resulting in abnormal deposition of adipose tissues during insulin resistance.


Adipocytes/physiology , CCAAT-Enhancer-Binding Proteins/metabolism , Insulin Resistance/physiology , PPAR gamma/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Blotting, Western , Cell Differentiation/physiology , Cell Growth Processes/physiology , DNA Primers/genetics , Gene Silencing , Rats , Real-Time Polymerase Chain Reaction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics
19.
Pediatr Res ; 72(6): 550-9, 2012 Dec.
Article En | MEDLINE | ID: mdl-23007031

BACKGROUND: Individuals with intrauterine growth retardation (IUGR) who demonstrate a catch-up in body weight are prone to insulin resistance. High expressions of suppressor of cytokine signaling 3 (SOCS3) are thought to aggravate insulin resistance. We hypothesized that downregulating SOCS3 expression via small interfering RNA (siRNA) might have beneficial effects on insulin-resistant hepatocytes of catch-up growth IUGR rats (CG-IUGRs). METHODS: An IUGR rat model was employed via maternal nutritional restriction. After evaluating metabolic states of CG-IUGR offspring, effective SOCS3-specific siRNA (siSOCS3) was transfected into cultured hepatocytes using liposomes. mRNA levels of SOCS3, insulin receptor substrates (IRSs), phosphatidylinositol 3-kinase (PI3K), and Akt2, key gluconeogenesis genes, were assessed via real-time PCR. Protein expression and phosphorylation changes were evaluated via western blot. RESULTS: CG-IUGR hepatocytes showed increases in SOCS3 and gluconeogenic gene expressions, and decreases in IRS1 and PI3K expressions as compared with controls. After transfecting CG-IUGR hepatocytes with siSOCS3, protein levels of IRS1, PI3K, and phosphorylated Akt2 were higher as compared with those of untransfected CG-IUGR cells. Transcriptional suppression effects of gluconeogenesis genes were more obvious in siSOCS3-treated cells after insulin stimulation. CONCLUSION: Additional insights were provided to understand mechanisms of insulin resistance in CG-IUGR rats. Downregulating SOCS3 might improve insulin signaling transduction and ameliorate hepatic glucose metabolism in insulin-resistant CG-IUGR rats in vitro.


Down-Regulation , Fetal Growth Retardation/metabolism , Glucose/metabolism , Hepatocytes/metabolism , Insulin/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Animals , Gluconeogenesis/genetics , Lipid Metabolism , Rats , Suppressor of Cytokine Signaling 3 Protein
20.
Pediatr Res ; 69(6): 497-503, 2011 Jun.
Article En | MEDLINE | ID: mdl-21364493

Intrauterine growth retardation (IUGR) has been linked to metabolic syndrome including insulin resistance, and overexpression of suppressors of cytokine signaling (SOCSs) proteins is thought to be associated with increased whole-body insulin sensitivity. The insulin-resistant IUGR rat model was established by maternal food restriction (about 30% of the normal rats). The weight, length, and homeostasis model assessment of insulin resistance (HOMA-IR) of IUGR-born rats was higher than the control group. Insulin receptor substrate (IRS)-1 expression decreased, whereas SOCS-1 and SOCS-3 increased in the skeletal muscle of IUGR rats compared with the control group. The recombination plasmids PGPU6/GFP/Neo-SOCS-1small hairpin RNA (shRNA) and PGPU6/GFP/Neo-SOCS-3shRNA were transfected into skeletal muscle cells, and the shRNAs efficiently inhibited the expression of SOCS-1 and SOCS-3. Insulin-stimulated glucose transporter-4 (GLUT4) translocation was also dramatically increased. In conclusion, these data provide additional information on the mechanism of insulin resistance associated with IUGR. Down-regulation of SOCS-1 and SOCS-3 ameliorates the capacity of glucose transport and provides a potential gene therapy approach to managing metabolic syndrome.


Fetal Growth Retardation/metabolism , Glucose/metabolism , RNA Interference , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cells, Cultured , Female , Fetal Growth Retardation/genetics , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Male , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein
...