Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Front Oncol ; 13: 1255112, 2023.
Article in English | MEDLINE | ID: mdl-37790757

ABSTRACT

Background: Ghrelin plays a critical role in regulating energy metabolism and homeostasis. The association between circulating ghrelin levels and gastric cancer has not been systematically analyzed. Objective: This work explored the association between circulating ghrelin levels and gastric cancer. Methods: The literature search for relevant articles published until November 2022 was performed using PubMed, Cochrane Library, EMBASE, and Web of Science with the keywords "ghrelin" and "gastric cancer". Standardized mean differences (SMD) with 95% confidence intervals were used to measure the effectiveness. We assessed pooled data by use of a random-effects model. Results: Of 5,302 identified studies, nine were included (N=3,196 participants). Circulating ghrelin levels were lower in gastric cancer patients (SMD=-0.255, 95%CI: -0.528 to 0.017, P < 0.00001), but with high heterogeneity (I2 = 88.8%). Conclusion: The circulating ghrelin levels in patients with gastric cancer were lower than in controls. However, there was heterogeneity among results; therefore, studies with larger sample sizes are recommended.

3.
Genes Dis ; 10(2): 521-530, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37223507

ABSTRACT

Severe bronchopulmonary dysplasia (BPD) is a chronic lung disorder that primarily affects premature babies with extremely low birth weight and involves in multiple organ system; no effective pharmacotherapy for this disease exists, and mortality remains high. Based on the evidence from previous preclinical studies and phase I clinical trials, this study aims to test the safety of intravenous application of a single dose of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) in patients with severe BPD. The Mesenchymal Stem cells for Bronchopulmonary Dysplasia Treatment (MSBDT) trial is a single center, open-label, dose-escalation phase I clinical trial. Severe BPD patients were enrolled in Children Hospital of Chongqing Medical University, Chongqing, China. The first six patients were treated with low-dose hUC-MSCs (1 × 106 cells/kg) and the next seven patients were treated with high-dose hUC-MSCs (5 × 106 cells/kg). This study is registered with ClinicalTrials.gov, number NCT03558334. No prespecified infusion-associated adverse events, immediate complication, respiratory or cardiovascular compromise were observed during infusion and 24 h after infusion. No significant changes in safety laboratory values were observed. One death event occurred in the low-dose group on study day 10, and one death event occurred in the high-dose group on study day 24, while, after review in detail, the two cases are not believed to be infusion-associated events. In conclusion, intravenous application of a single dose of hUC-MSCs was tolerated in thirteen patients with severe BPD.

4.
Front Immunol ; 12: 785549, 2021.
Article in English | MEDLINE | ID: mdl-34917093

ABSTRACT

Atopic dermatitis (AD) in early childhood is often the initial manifestation of allergic disease associated with high IgE. Accumulating evidences show that follicular helper T (Tfh) cells play a critical role in promoting B cell differentiation and IgE production, human regulatory B (Breg) cells participate in immunomodulatory processes and inhibition of allergic inflammation. However, the roles and interactions between IL-10-producing Breg cells and Tfh cells in childhood AD are unclear. In this study, we found that the percentage of CD19+IL-10+ Breg cells in children with extrinsic AD was significantly lower than that in age-matched healthy controls, and that it correlated negatively with enhanced CD4+CXCR5+PD-1+ICOS+ circulating Tfh cell responses and increased disease activity; however, there was no significant correlation with serum total IgE levels. A co-culture system revealed that Breg cells from patients with extrinsic AD cannot effectively inhibit differentiation of Tfh cells in an IL-10 dependent manner. Abnormal pSTAT3 signaling induced via Toll-like receptors (TLR), but not the B-cell receptor (BCR) signaling, might contribute to the defect of Breg cells in AD. Taken together, these observations demonstrate an important role for IL-10-producing Breg cells in inhibiting Tfh cell differentiation, and suggest that they may participate in the pathogenesis of AD.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Dermatitis, Atopic/immunology , T Follicular Helper Cells/immunology , B-Lymphocytes, Regulatory/metabolism , Case-Control Studies , Cell Communication/immunology , Cell Differentiation/immunology , Cell Separation , Cells, Cultured , Child , Child, Preschool , Coculture Techniques , Dermatitis, Atopic/blood , Female , Flow Cytometry , Healthy Volunteers , Humans , Infant , Interleukin-10/metabolism , Male , Primary Cell Culture , T Follicular Helper Cells/metabolism
5.
Trials ; 21(1): 125, 2020 Jan 31.
Article in English | MEDLINE | ID: mdl-32005282

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia (BPD) is a complex lung pathological lesion secondary to multiple factors and one of the most common chronic lung diseases. It has a poor prognosis, especially in preterm infants. However, effective therapies for this disease are lacking. Stem-cell therapy is a promising way to improve lung injury and abnormal alveolarization, and the human umbilical cord (hUC) is a good source of mesenchymal stem cells (MSCs), which have demonstrated efficacy in other diseases. We hypothesized that intravenously administered allogeneic hUC-MSCs are safe and effective for severe BPD. METHODS: The MSC-BPD trial is a randomized, single-center, open-label, dose-escalation, phase-II trial designed to investigate the safety and efficacy of hUC-MSCs in children with severe BPD. In this study, 72 patients will be enrolled and randomly divided into two intervention groups and one control group. Patients in the intervention groups will receive a low dose of hUC-MSCs (n = 24; 2.5 million cells/kg) or a high dose of hUC-MSCs (n = 24; 5 million cells/kg) in combination with traditional supportive treatments for BPD. The patients in the control group (n = 24) will be treated with traditional supportive treatments alone without hUC-MSCs. The primary outcome measures will be cumulative duration of oxygen therapy. Follow-up assessments will be performed at 1, 3, 6, 12, and 24 months post intervention, and the key outcome during follow-up will be changes on chest radiography. Statistical analyses will evaluate the efficacy of the hUC-MSC treatment. DISCUSSION: This will be the first randomized controlled trial to evaluate the safety and efficacy of intravenously administered hUC-MSCs in children with severe BPD. Its results should provide a new evidence-based therapy for severe BPD. TRIAL REGISTRATION: ClinicalTrials.gov, ID: NCT03601416. Registered on 26 July 2018.


Subject(s)
Bronchopulmonary Dysplasia/therapy , Cord Blood Stem Cell Transplantation/methods , Lung , Mesenchymal Stem Cells/physiology , Administration, Intravenous , Bronchopulmonary Dysplasia/diagnosis , Bronchopulmonary Dysplasia/physiopathology , Clinical Trials, Phase II as Topic , Female , Humans , Infant , Infant, Newborn , Infant, Premature/physiology , Lung/diagnostic imaging , Lung/physiopathology , Male , Outcome Assessment, Health Care/methods , Oxygen Inhalation Therapy/statistics & numerical data , Radiography, Thoracic/statistics & numerical data , Randomized Controlled Trials as Topic , Severity of Illness Index
6.
Biomed Pharmacother ; 97: 1349-1355, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29156524

ABSTRACT

BACKGROUND: KIF3A expression was decreased in asthmatic child patients and animal. Impaired KIF3A expression resulted in increased Th2 inflammation in mice and apoptosis in renal tubular epithelium and photoreceptor cells. This work aimed to investigate the role of KIF3A in epithelium apoptosis and bronchial inflammation in asthma. METHODS: After establishment of ovalbumin induced asthma, the mice were infected with KIF3A adenovirus through nasal cavity inhalation. KIF3A expression and apoptosis in epithelia of nasal mucosa and bronchia were determined using qRT-PCR, western blotting, immunohistochemistry and TUNEL staining. The mRNA expression of COX-2, IL-4, IL-5, IL-13, IL-6, IL-10 and TNF-α was also measured. In vitro, human bronchial epithelial cell line 16HBE 14o- was stimulated with IL-4, IL-13 and TNF-α, accompanied by KIF3A knockdown or overexpression using siRNA or KIF3A adenovirus respectively. Apoptosis, mRNA expression of CCL17, CCL26, IL-5 and IL-8, and protein expression of COX-2 and ß-catenin were determined using flow cytometry, qRT-PCR and western blotting. RESULTS: KIF3A expression was reduced in epithelia of nasal mucosa and bronchia of asthmatic mice, and overexpression of KIF3A ameliorated epithelial cell apoptosis and bronchial inflammation in asthmatic mice. In vitro, KIF3A knockdown significantly promoted epithelium apoptosis, facilitated the transcription of CCL17, CCL26, IL-5 and IL-8, and increased the protein levels of COX-2 and ß-catenin translocation, whereas overexpression of KIF3A exhibited the opposite effect. CONCLUSION: KIF3A plays an important role in epithelium apoptosis and bronchial inflammation in asthma, and may be a potential target for asthma treatment.


Subject(s)
Asthma/pathology , Inflammation/pathology , Kinesins/genetics , Respiratory Mucosa/pathology , Adenoviridae/genetics , Animals , Apoptosis/genetics , Asthma/genetics , Bronchi/pathology , Female , Flow Cytometry , Gene Expression Regulation , Gene Knockdown Techniques , Humans , In Situ Nick-End Labeling , Inflammation/genetics , Mice , Mice, Inbred BALB C , Ovalbumin , RNA, Messenger/metabolism
7.
Environ Toxicol Pharmacol ; 56: 136-144, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28934691

ABSTRACT

Perfluorooctanoic acid (PFOA) is a persistent organic pollutant. This study established an in ovo peroxisome proliferator-activated receptor alpha (PPAR alpha) silencing model in chicken embryo heart, and investigated the role of PPAR alpha in PFOA induced developmental cardiotoxicity. The in ovo silencing was achieved by introducing lentivirus expressing PPAR alpha siRNA into ED2 chicken embryo via microinjection (0.05ul/g egg weight). Transfection efficacy was confirmed by fluorescent microscopy and western blotting. To assess the developmental cardiotoxicity, cardiac function (heart rate) and morphology (right ventricular wall thickness) were measured in D1 hatchling chickens. 2mg/kg (egg weight) PFOA exposure at ED0 induced significant elevation of heart rate and thinning of right ventricular wall thickness in D1 hatchling chickens. PPAR alpha silencing did not prevent PFOA-induced elevation of heart rate; however, it did significantly increase the right ventricular wall thickness as compared to PFOA exposed animals. Meanwhile, PPAR alpha silencing did not abolish the protective effects exerted by exposure to 100mg/kg (egg weight) l-carnitine. In conclusion, PFOA-induced heart rate elevation is likely PPAR alpha independent, while the right ventricular wall thinning seems to be PPAR alpha dependent. The protective effects of l-carnitine do not require PPAR alpha.


Subject(s)
Caprylates/toxicity , Cardiotoxicity/prevention & control , Fluorocarbons/toxicity , Heart/drug effects , PPAR alpha/genetics , Animals , Cardiotoxicity/pathology , Cardiotoxicity/physiopathology , Carnitine/pharmacology , Chick Embryo , Gene Silencing , Heart/embryology , Heart/physiopathology , Heart Rate/drug effects , Lentivirus/genetics , Transfection
8.
Sci Rep ; 7: 39889, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28051154

ABSTRACT

The incidence and mortality of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) are still very high, but stem cells show some promise for its treatment. Here we found that intratracheal administration of human umbilical cord-mesenchymal stem cells (UC-MSCs) significantly improved survival and attenuated the lung inflammation in lipopolysaccharide (LPS)-induced ALI mice. We also used the proteins-chip and bioinformatics to analyze interactions between UC-MSCs treatment and immune-response alternations of ALI mice. Then we demonstrated that UC-MSCs could inhibit the inflammatory response of mouse macrophage in ALI mice, as well as enhance its IL-10 expression. We provide data to support the concept that the therapeutic capacity of UC-MSCs for ALI was primarily through paracrine secretion, particularly of prostaglandin-E2 (PGE2). Furthermore, we showed that UC-MSCs might secrete a panel of factors including GM-CSF, IL-6 and IL-13 to ameliorate ALI. Our study suggested that UC-MSCs could protect LPS-induced ALI model by immune regulation and paracrine factors, indicating that UC-MSCs should be a promising strategy for ALI/ARDS.


Subject(s)
Acute Lung Injury/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Acute Lung Injury/etiology , Acute Lung Injury/mortality , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cells, Cultured , Dinoprostone/metabolism , Disease Models, Animal , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-10/metabolism , Lipopolysaccharides/toxicity , Lung/metabolism , Lung/pathology , Macrophages/cytology , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Survival Rate , Umbilical Cord/cytology
9.
Genes Dis ; 4(4): 204-214, 2017 Dec.
Article in English | MEDLINE | ID: mdl-30258924

ABSTRACT

Alveolar epithelial cells (AECs) injury and failed reconstitution of the AECs barrier are both integral to alveolar flooding and subsequent pulmonary fibrosis (PF). Nevertheless, the exact mechanisms regulating the regeneration of AECs post-injury still remain unclear. SMARCA4 is a part of the large ATP-dependent chromatin remodelling complex SWI/SNF, which is essential for kidney and heart fibrosis. We investigates SMARCA4 function in lung fibrosis by establishing PF mice model with bleomycin firstly and found that the expression of SMARCA4 was mainly enhanced in alveolar type II (ATII) cells. Moreover, we established an alveolar epithelium-specific SMARCA4-deleted SP-C-rtTA/(tetO) 7 -Cre/SMARCA4 f/f mice (SOSM4 Δ/Δ ) model, as well as a new SMARCA4-deleted alveolar type II (ATII)-like mle-12 cell line. We found that the bleomycin-induced PF was more aggressive in SOSM4 Δ/Δ mice. Also, the proliferation of ATII cells was decreased with the loss of SMARCA4 in vivo and in vitro. In addition, we observed increased proliferation of ATII cells accompanied by abnormally high expression of SMARCA4 in human PF lung sections. These data uncovered the indispensable role of SMARCA4 in the proliferation of ATII cells, which might affect the progression of PF.

10.
Cell Biochem Biophys ; 71(2): 1089-95, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25344648

ABSTRACT

The objective of this study was to investigate the hepatoprotective effect of cod skin collagen peptides (CSCP), isolated from fishing industrial by-products, in vitro and in vivo. Effect of CSCP on cell proliferation of normal and H2O2-damaged Chang liver cells was determined by MTT assay in vitro. Two animal models, CCl4-induced and acetaminophenum-induced acute hepatotoxicity, were established to assess the hepatoprotective effect of CSCP. Liver weight index, serum ALT and AST, antioxidant enzymes, and lipid peroxidation product were used as the markers of liver toxicity. The cell viability in the H2O2-treated Chang liver cells was remarkably increased when pretreated with CSCP from 100 to 1,000 µg/ml in a dose-dependent manner. CSCP pretreatment also alleviated the CCL4-induced liver index loss, while no marked changes were found in acetaminophenum-treated mice. Furthermore, CSCP pulled down serum ALT and AST level, increased the activities of SOD and CAT, and decreased MDA in both murine models of acute liver toxicity. Pretreatment with CSCP protected liver tissue against oxidative injure in vivo and in vitro. The underlying mechanism might involve enhancement in the activities of antioxidant enzymes and reduction in the lipid peroxidation.


Subject(s)
Collagen/chemistry , Cytoprotection/drug effects , Gadiformes , Hepatocytes/drug effects , Liver/drug effects , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Alanine Transaminase/blood , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/blood , Cell Proliferation/drug effects , Hepatocytes/cytology , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Lipid Peroxidation/drug effects , Liver/cytology , Liver/enzymology , Liver/metabolism , Male , Mice
11.
Diabetol Metab Syndr ; 3: 31, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22082204

ABSTRACT

BACKGROUND: There are an increasing number of patients suffering from fatty liver caused by type 2 diabetes. We intended to study the preventive and therapeutic effect of L-carnitine (LC) on nonalcoholic fatty liver disease (NAFLD) in streptozotocin (STZ)-induced type 2 diabetic mice and to explore its possible mechanism. METHODS: Thirty male Kungming mice were randomly divided into five groups: control group, diabetic group, pre-treatment group (125 mg/kg BW), low-dose (125 mg/kg BW) therapeutic group and high-dose (250 mg/kg BW) therapeutic group. The morphology of hepatocytes was observed by light and electron microscopy. LC and ALC (acetyl L-carnitine) concentrations in the liver were determined by high-performance liquid chromatography (HPLC). Moreover, liver weight, insulin levels and free fatty acid (FFA) and triglyceride (TG) levels in the liver and plasma were measured. RESULTS: Average liver LC and ALC levels were 33.7% and 20% lower, respectively, in diabetic mice compared to control mice (P < 0.05). After preventive and therapeutic treatment with LC, less hepatocyte steatosis, clearer crista and fewer glycogen granules in the mitochondria were observed. Decreased liver weight, TG levels, and FFA concentrations (P < 0.05) in the liver were also observed after treatment with LC in diabetic mice. Moreover, liver LC and ALC levels increased upon treatment with LC, whereas the ratio of LC and ALC decreased significantly (P < 0.01). CONCLUSION: LC supplements ameliorated fatty liver in type 2 diabetic mice by increasing fatty acid oxidation and decreasing the LC/ALC ratio in the liver. Therefore, oral administration of LC protected mitochondrial function in liver.

SELECTION OF CITATIONS
SEARCH DETAIL
...