Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Diabetes ; 73(5): 701-712, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38320268

ABSTRACT

Bile acids (BAs) are pleiotropic regulators of metabolism. Elevated levels of hepatic and circulating BAs improve energy metabolism in peripheral organs, but the precise mechanisms underlying the metabolic benefits and harm still need to be fully understood. In the current study, we identified orosomucoid 2 (ORM2) as a liver-secreted hormone (i.e., hepatokine) induced by BAs and investigated its role in BA-induced metabolic improvements in mouse models of diet-induced obesity. Contrary to our expectation, under a high-fat diet (HFD), our Orm2 knockout (Orm2-KO) exhibited a lean phenotype compared with C57BL/6J control, partly due to the increased energy expenditure. However, when challenged with a HFD supplemented with cholic acid, Orm2-KO eliminated the antiobesity effect of BAs, indicating that ORM2 governs BA-induced metabolic improvements. Moreover, hepatic ORM2 overexpression partially replicated BA effects by enhancing insulin sensitivity. Mechanistically, ORM2 suppressed interferon-γ/STAT1 activities in inguinal white adipose tissue depots, forming the basis for anti-inflammatory effects of BAs and improving glucose homeostasis. In conclusion, our study provides new insights into the molecular mechanisms of BA-induced liver-adipose cross talk through ORM2 induction.


Subject(s)
Bile Acids and Salts , Orosomucoid , Mice , Animals , Bile Acids and Salts/metabolism , Orosomucoid/metabolism , Orosomucoid/pharmacology , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Liver/metabolism , Diet, High-Fat/adverse effects
2.
Mol Cells ; 46(9): 527-534, 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37691258

ABSTRACT

Liver ischemia-reperfusion injury (IRI) is the main cause of organ dysfunction and failure after liver surgeries including organ transplantation. The mechanism of liver IRI is complex and numerous signals are involved but cellular metabolic disturbances, oxidative stress, and inflammation are considered the major contributors to liver IRI. In addition, the activation of inflammatory signals exacerbates liver IRI by recruiting macrophages, dendritic cells, and neutrophils, and activating NK cells, NKT cells, and cytotoxic T cells. Technological advances enable us to understand the role of specific immune cells during liver IRI. Accordingly, therapeutic strategies to prevent or treat liver IRI have been proposed but no definitive and effective therapies exist yet. This review summarizes the current update on the immune cell functions and discusses therapeutic potentials in liver IRI. A better understanding of this complex and highly dynamic process may allow for the development of innovative therapeutic approaches and optimize patient outcomes.


Subject(s)
Liver , Reperfusion Injury , Humans , Inflammation , Killer Cells, Natural , Macrophages
3.
Mol Metab ; 72: 101717, 2023 06.
Article in English | MEDLINE | ID: mdl-37004989

ABSTRACT

OBJECTIVE: Mitophagy removes damaged mitochondria to maintain cellular homeostasis. Aryl hydrocarbon receptor (AhR) expression in the liver plays a crucial role in supporting normal liver functions, but its impact on mitochondrial function is unclear. Here, we identified a new role of AhR in the regulation of mitophagy to control hepatic energy homeostasis. METHODS: In this study, we utilized primary hepatocytes from AhR knockout (KO) mice and AhR knockdown AML12 hepatocytes. An endogenous AhR ligand, kynurenine (Kyn), was used to activate AhR in AML12 hepatocytes. Mitochondrial function and mitophagy process were comprehensively assessed by MitoSOX and mt-Keima fluorescence imaging, Seahorse XF-based oxygen consumption rate measurement, and Mitoplate S-1 mitochondrial substrate utilization analysis. RESULTS: Transcriptomic analysis indicated that mitochondria-related gene sets were dysregulated in AhR KO liver. In both primary mouse hepatocytes and AML12 hepatocyte cell lines, AhR inhibition strongly suppressed mitochondrial respiration rate and substrate utilization. AhR inhibition also blunted the fasting response of several essential autophagy genes and the mitophagy process. We further identified BCL2 interacting protein 3 (BNIP3), a mitophagy receptor that senses nutrient stress, as an AhR target gene. AhR is directly recruited to the Bnip3 genomic locus, and Bnip3 transcription was enhanced by AhR endogenous ligand treatment in wild-type liver and abolished entirely in AhR KO liver. Mechanistically, overexpression of Bnip3 in AhR knockdown cells mitigated the production of mitochondrial reactive oxygen species (ROS) and restored functional mitophagy. CONCLUSIONS: AhR regulation of the mitophagy receptor BNIP3 coordinates hepatic mitochondrial function. Loss of AhR induces mitochondrial ROS production and impairs mitochondrial respiration. These findings provide new insight into how endogenous AhR governs hepatic mitochondrial homeostasis.


Subject(s)
Mitochondria , Receptors, Aryl Hydrocarbon , Mice , Animals , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Ligands , Mitochondria/metabolism , Liver/metabolism , Mice, Knockout , Homeostasis
4.
Int J Cancer ; 150(10): 1690-1705, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35020952

ABSTRACT

Hepatocellular carcinoma (HCC) is the second most common cancer worldwide, demonstrating aggressiveness and mortality more frequently in men than in women. Despite reports regarding the inhibitory ability of estrogen receptor alpha (ERα, ESR1) in certain cancer progression, targets and the basis of underlying gender disparity in HCC worsening remain elusive. Here, we report the ability of ERα to transcriptionally inhibit G protein subunit alpha 12 (Gα12) responsible for HCC worsening. First, using human samples and public database, the expression of ERα and Gα12 in HCC was examined. Then, quantitative real-time PCR, chromatin immunoprecipitation-assay, luciferase assay and immunoblottings of liver cancer cell lines confirmed the inhibitory ability of ERα on Gα12 and HCC progression. Gα12 promoted mesenchymal characteristics and amoeboidal movement, which was antagonized by ERα overexpression. Additionally, we found microRNA-141 and microRNA-200a as downstream targets of the Gα12 signaling axis for cancer malignancy regulation under the control of ERα. As for in-depth mechanism, PTP4A1 was found to be directly inhibited by microRNA-141 and microRNA-200a. Moreover, we found the inhibitory effect of ERα on amoeboidal movement by analyzing the morphology and blebbing of liver cancer cells and the active form of MLC levels. The identified targets and ESR1 levels are inversely correlated with human specimens, as well as with sex-biased survival rates of HCC patients. Collectively, ERα-dependent repression of Gα12 and consequent changes in the Gα12 signaling may explain the gender disparity in HCC, providing pharmacological clues for the control of metastatic HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Male , MicroRNAs/metabolism
5.
Int J Cancer ; 147(9): 2550-2563, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32449166

ABSTRACT

Activation of sterol regulatory element-binding protein 1 (SREBP-1), a master lipogenic transcription factor, is associated with cancer metabolism and metabolic disorders. Neddylation, the process of adding NEDD8 to its substrate, contributes to diverse biological processes. Here, we identified SREBP-1 as a substrate for neddylation by UBC12 and explored its impact on tumor aggressiveness. In cell-based assays, SREBP-1 neddylation prolonged SREBP-1 stability with a decrease in ubiquitination. Consequently, NEDD8 overexpression facilitated proliferation, migration, and invasion of SK-Hep1 liver tumor cells. MLN4924 (an inhibitor of the NEDD8-activating enzyme-E1) treatment or UBC12 knockdown prevented SREBP-1 neddylation and tumor cell phenotype change. This effect was corroborated in an in vivo xenograft model. In human specimens, SREBP-1, UBC12, and NEDD8 were all upregulated in hepatocellular carcinoma (HCC) compared to nontumorous regions. Moreover, SREBP-1 levels positively correlated with UBC12. In GEO database analyses, SREBP-1 levels were greater in metastatic HCC samples accompanying UBC12 upregulation. In HCC analysis, tumoral SREBP-1 and UBC12 levels discriminated overall patient survival rates. Additionally, MLN4924 treatment destabilized SREBP-1 in MDA-MB-231 breast cancer cells and in the tumor cell xenograft. SREBP-1 and UBC12 were also highly expressed in human breast cancer tissues. Moreover, most breast cancers with lymph node metastasis displayed predominant SREBP-1 and UBC12 expressions, which compromised overall patient survival rates. In summary, SREBP-1 is neddylated by UBC12, which may contribute to HCC and breast cancer aggressiveness through SREBP-1 stabilization, and these events can be intervented by MLN4924 therapy. Our findings may also provide potential reliable prognostic markers for tumor metastasis.


Subject(s)
Breast Neoplasms/mortality , Carcinoma, Hepatocellular/mortality , Liver Neoplasms/mortality , Sterol Regulatory Element Binding Protein 1/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Animals , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cyclopentanes/pharmacology , Cyclopentanes/therapeutic use , Female , Humans , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Lymphatic Metastasis/pathology , Mice , NEDD8 Protein/metabolism , Prognosis , Protein Stability/drug effects , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Sterol Regulatory Element Binding Protein 1/analysis , Survival Rate , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/analysis , Ubiquitination/drug effects , Up-Regulation , Xenograft Model Antitumor Assays
6.
Hepatology ; 71(3): 1055-1069, 2020 03.
Article in English | MEDLINE | ID: mdl-31355949

ABSTRACT

BACKGROUND AND AIMS: Liver receptor homolog-1 (LRH-1; NR5A2) is a nuclear receptor that regulates metabolic homeostasis in the liver. Previous studies identified phosphatidylcholines as potential endogenous agonist ligands for LRH-1. In the liver, distinct subsets of phosphatidylcholine species are generated by two different pathways: choline addition to phosphatidic acid through the Kennedy pathway and trimethylation of phosphatidylethanolamine through phosphatidylethanolamine N-methyl transferase (PEMT). APPROACH AND RESULTS: Here, we report that a PEMT-LRH-1 pathway specifically couples methyl metabolism and mitochondrial activities in hepatocytes. We show that the loss of Lrh-1 reduces mitochondrial number, basal respiration, beta-oxidation, and adenosine triphosphate production in hepatocytes and decreases expression of mitochondrial biogenesis and beta-oxidation genes. In contrast, activation of LRH-1 by its phosphatidylcholine agonists exerts opposite effects. While disruption of the Kennedy pathway does not affect the LRH-1-mediated regulation of mitochondrial activities, genetic or pharmaceutical inhibition of the PEMT pathway recapitulates the effects of Lrh-1 knockdown on mitochondria. Furthermore, we show that S-adenosyl methionine, a cofactor required for PEMT, is sufficient to induce Lrh-1 transactivation and consequently mitochondrial biogenesis. CONCLUSIONS: A PEMT-LRH-1 axis regulates mitochondrial biogenesis and beta-oxidation in hepatocytes.


Subject(s)
Hepatocytes/metabolism , Mitochondria/physiology , Phosphatidylethanolamine N-Methyltransferase/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Hep G2 Cells , Humans , Male , Mice , Oxidation-Reduction , S-Adenosylmethionine/metabolism , S-Adenosylmethionine/pharmacology
7.
Sci Transl Med ; 11(513)2019 10 09.
Article in English | MEDLINE | ID: mdl-31597753

ABSTRACT

The impact of liver disease on whole-body glucose homeostasis is largely attributed to dysregulated release of secretory proteins in response to metabolic stress. The molecular cues linking liver to whole-body glucose metabolism remain elusive. We found that expression of G protein α-13 (Gα13) was decreased in the liver of mice and humans with diabetes. Liver-specific deletion of the Gna13 gene in mice resulted in systemic glucose intolerance. Comparative secretome analysis identified inter-α-trypsin inhibitor heavy chain 1 (ITIH1) as a protein secreted by liver that was responsible for systemic insulin resistance in Gna13-deficient mice. Liver expression of ITIH1 positively correlated with surrogate markers for diabetes in patients with impaired glucose tolerance or overt diabetes. Mechanistically, a decrease in hepatic Gα13 caused ITIH1 oversecretion by liver through induction of O-GlcNAc transferase expression, facilitating ITIH1 deposition on the hyaluronan surrounding mouse adipose tissue and skeletal muscle. Neutralization of secreted ITIH1 ameliorated glucose intolerance in obese mice. Our findings demonstrate systemic insulin resistance in mice resulting from liver-secreted ITIH1 downstream of Gα13 and its reversal by ITIH1 neutralization.


Subject(s)
Alpha-Globulins/metabolism , Insulin Resistance/physiology , Liver/metabolism , Alpha-Globulins/genetics , Animals , Antibodies, Neutralizing/metabolism , Cell Line , Cells, Cultured , Chromatography, Liquid , Glucose Intolerance/metabolism , Glucose Tolerance Test , HEK293 Cells , Hepatocytes/metabolism , Humans , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Tandem Mass Spectrometry
8.
Arch Pharm Res ; 42(1): 48-62, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30610616

ABSTRACT

Hepatocellular carcinoma (HCC) is a tumor with poor prognosis and frequently aggressive. The development of HCC is associated with fibrosis and cirrhosis, which mainly results from nonalcoholic fatty liver disease, excessive alcohol consumption, and viral infections. Non-coding RNAs (ncRNAs) are RNAs transcribed from the genome, but are not translated into proteins. Recently, ncRNAs emerged as key contributors to tumor development and progression because of their abilities to regulate various targets and modulate cell proliferation, differentiation, apoptosis, and development. In this review, we summarize the frequently activated pathways in HCC and discuss the pathological implications of ncRNAs in the context of human liver disease progression, in particular HCC development and progression. This review aims to summarize the role of ncRNA dysregulation in the diseases and discuss the diagnostic and therapeutic potentials of ncRNAs.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Disease Progression , Liver Diseases/metabolism , Liver Neoplasms/metabolism , RNA, Untranslated/physiology , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Liver Diseases/pathology , Liver Neoplasms/pathology , RNA, Long Noncoding/physiology , Signal Transduction/physiology
9.
Gut ; 68(4): 708-720, 2019 04.
Article in English | MEDLINE | ID: mdl-29475852

ABSTRACT

OBJECTIVE: Alcoholic liver disease (ALD) is a leading cause of death among chronic liver diseases. However, its pathogenesis has not been completely established. MicroRNAs (miRNAs) are key contributors to liver diseases progression. This study investigated hepatocyte-abundant miRNAs dysregulated by ALD, its impact on hepatocyte injury and the underlying basis. DESIGN: Alcoholic hepatitis (AH) human and animal liver samples and hepatocytes were used to assess miR-148a levels. Pre-miR-148a was delivered specifically to hepatocytes in vivo using lentivirus. Immunoblottings, luciferase reporter assays, chromatin immunoprecipitation and immunofluorescence assays were carried out in cell models. RESULTS: The miRNA profile and PCR analyses enabled us to find substantial decrease of miR-148a in the liver of patients with AH. In mice subjected to Lieber-DeCarli alcohol diet or binge alcohol drinking, miR-148a levels were also markedly reduced. In cultured hepatocytes and mouse livers, alcohol exposure inhibited forkhead box protein O1 (FoxO1) expression, which correlated with miR-148a levels and significantly decreased in human AH specimens. FoxO1 was identified as a transcription factor for MIR148A transactivation. MiR-148a directly inhibited thioredoxin-interacting protein (TXNIP) expression. Consequently, treatment of hepatocytes with ethanol resulted in TXNIP overexpression, activating NLRP3 inflammasome and caspase-1-mediated pyroptosis. These events were reversed by miR-148a mimic or TXNIP small-interfering RNA transfection. Hepatocyte-specific delivery of miR-148a to mice abrogated alcohol-induced TXNIP overexpression and inflammasome activation, attenuating liver injury. CONCLUSION: Alcohol decreases miR-148a expression in hepatocytes through FoxO1, facilitating TXNIP overexpression and NLRP3 inflammasome activation, which induces hepatocyte pyroptosis. Our findings provide information on novel targets for reducing incidence and progression of ALD.


Subject(s)
Carrier Proteins/metabolism , Hepatitis, Alcoholic/metabolism , Hepatocytes/metabolism , Inflammasomes/metabolism , Pyroptosis , Thioredoxins/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Disease Progression , Fluorescent Antibody Technique , Humans , Immunoblotting , Mice , MicroRNAs , Polymerase Chain Reaction
10.
J Med Chem ; 61(20): 9266-9286, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30252468

ABSTRACT

Ocular diseases featuring pathologic neovascularization are the leading cause of blindness, and anti-VEGF agents have been conventionally used to treat these diseases. Recently, regulating factors upstream of VEGF, such as HIF-1α, have emerged as a desirable therapeutic approach because the use of anti-VEGF agents is currently being reconsidered due to the VEGF action as a trophic factor. Here, we report a novel scaffold discovered through the complete structure-activity relationship of ring-truncated deguelin analogs in HIF-1α inhibition. Interestingly, analog 6i possessing a 2-fluorobenzene moiety instead of a dimethoxybenzene moiety exhibited excellent HIF-1α inhibitory activity, with an IC50 value of 100 nM. In particular, the further ring-truncated analog 34f, which showed enhanced HIF-1α inhibitory activity compared to analog 2 previously reported by us, inhibited in vitro angiogenesis and effectively suppressed hypoxia-mediated retinal neovascularization. Importantly, the heteroatom-substituted benzene ring as a key structural feature of analog 34f was identified as a novel scaffold for HIF-1α inhibitors that can be used in lieu of a chromene ring.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Drug Design , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Retinal Neovascularization/drug therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Benzene/chemistry , Benzene/pharmacology , Benzene/therapeutic use , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mice , Solubility , Structure-Activity Relationship , Water/chemistry
11.
Oncoscience ; 2(10): 777-8, 2015.
Article in English | MEDLINE | ID: mdl-26682240
12.
Oncotarget ; 6(6): 3918-31, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25714015

ABSTRACT

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths. Protoporphyrin IX (PPIX) has been used for photodynamic therapy. Mesenchymal cancer cells adapt to tumor microenvironments for growth and metastasis possibly in association with miRNA dysregulation. In view of the effect of PPIX on cancer-related genes, and its potential to inhibit tumor growth and migration/invasion, this study investigated whether PPIX enables mesenchymal liver tumor to restore dysregulated miRNAs, and if so, whether it sensitizes the cancer cells to chemotherapy. In addition, we explored new target(s) of the miRNA(s) that contribute to the anti-cancer effects. Of the ten miRNAs predicted by the 3'-UTR of HIF-1α mRNA, PPIX treatment increased miR-199a-5p, leading to the inhibition of E2F3 expression which is upregulated in mesenchymal liver tumor. miR-199a-5p levels were downregulated in HCC with E2F3 overexpression. An approach modulating epithelial-mesenchymal transition provided the expected changes in miR-199a-5p and E2F3 in vivo. PPIX prevented tumor cell growth and migration/invasion, and had a synergistic anti-cancer effect when combined with chemotherapeutics. In a xenograft model, PPIX treatment decreased overall growth and average tumor volume, which paralleled E2F3 inhibition. Overall, PPIX inhibited growth advantage and migratory ability of cancer cells and sensitized mesenchymal liver tumor cells to chemotherapeutics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , E2F3 Transcription Factor/antagonists & inhibitors , Liver Neoplasms/drug therapy , Mesenchymal Stem Cells/drug effects , MicroRNAs/biosynthesis , Photosensitizing Agents/pharmacology , Protoporphyrins/pharmacology , Animals , Base Sequence , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Drug Synergism , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/metabolism , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Data , Protoporphyrins/administration & dosage , Random Allocation , Transfection , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
13.
Oncotarget ; 5(9): 2792-806, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24798342

ABSTRACT

Hepatocellular carcinoma (HCC) is classified as a poor prognostic tumor, and becomes frequently aggressive. MicroRNAs emerge as key contributors to tumor progression. This study investigated whether miR-148a dysregulation differentiates poor prognosis of HCC, exploring new targets of miR-148a. miR-148a dysregulation discriminated not only the overall survival and recurrence free survival rates of HCC, but the microvascular invasion. In the human HCC samples, ubiquitin specific protease 4 (USP4) and sphingosine 1-phosphate receptor 1 (S1P1) were up-regulated as the new targets of miR-148a. USP4 and S1P1 were up-regulated in mesenchymal-type liver-tumor cells with miR-148a dysregulation, facilitating migration and proliferation of tumor cells. The inverse relationship between miR-148a and the identified targets was verified in a tumor xenograft model. In the analysis of human samples, the expression of USP4, but not S1P1, correlated with the decrease of miR-148a. In a heterotropic patient-derived HCC xenograft model, USP4 was also overexpressed in G1 and G2 tumors when miR-148a was dysregulated, reflecting the closer link between miR-148a and USP4 for a shift in the expansion phase of tumorgraft. In conclusion, miR-148a dysregulation affects the poor prognosis of HCC. Of the identified targets of miR-148a, USP4 overexpression may contribute to HCC progression towards more aggressive feature.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Apoptosis , Blotting, Western , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Adhesion , Cell Movement , Cell Proliferation , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , MicroRNAs/metabolism , Middle Aged , Neoplasm Grading , Neoplasm Staging , Nerve Tissue Proteins/genetics , Prognosis , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Proteases , Xenograft Model Antitumor Assays
14.
Arch Pharm Res ; 35(9): 1673-83, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23054725

ABSTRACT

Ductus arteriosus is a normal connecting blood vessel between the pulmonary artery and aorta in the fetus. However, if the ductus arteriosus is not closed and maintained as the open state even after 72 h of the birth, this is called a patent ductus arteriosus (PDA). Intravenous indomethacin is the conventional treatment for PDA in immature infants, but remains controversial in mature infants. The purpose of this study was to compare intravenous indomethacin and oral ibuprofen with regard to efficacy and safety for treatment of PDA. 78 neonates treated for PDA were included and classified into immature (n = 49) and mature (n = 29) groups. Ductal closure occurred in immature infants treated with indomethacin (74.1%) and ibuprofen (90.9%). Ductal closure occurred in mature infants treated with indomethacin (66.7%) and ibuprofen (92.9%). Platelet counts were increased in immature infants treated with ibuprofen (p = 0.027). Hyponatremia occurred in immature infants treated with ibuprofen (p = 0.002) and in both groups of mature infants (p = 0.001 for both groups). Serum creatinine values were lowered in mature infants treated with ibuprofen (p = 0.032). Bleeding occurred in 5 immature infants treated with indomethacin. Administration of furosemide for urine output was more frequent in the mature groups than in the immature group. In conclusion, oral ibuprofen was as effective as intravenous indomethacin in the immature groups and more effective in the mature groups. Adverse effects of oral ibuprofen were less severe than intravenous indomethacin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Ductus Arteriosus, Patent/drug therapy , Ibuprofen/therapeutic use , Indomethacin/therapeutic use , Infant, Premature , Respiratory Distress Syndrome, Newborn/prevention & control , Administration, Oral , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Comorbidity , Ductus Arteriosus, Patent/blood , Ductus Arteriosus, Patent/epidemiology , Ductus Arteriosus, Patent/physiopathology , Female , Humans , Hyponatremia/epidemiology , Hyponatremia/prevention & control , Ibuprofen/administration & dosage , Ibuprofen/adverse effects , Incidence , Indomethacin/administration & dosage , Indomethacin/adverse effects , Infant, Newborn , Injections, Intravenous , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/epidemiology , Intracranial Hemorrhages/prevention & control , Male , Off-Label Use , Renal Insufficiency/drug therapy , Renal Insufficiency/epidemiology , Renal Insufficiency/prevention & control , Republic of Korea/epidemiology , Respiratory Distress Syndrome, Newborn/epidemiology , Respiratory Distress Syndrome, Newborn/etiology , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...