Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
PLoS One ; 17(11): e0277339, 2022.
Article in English | MEDLINE | ID: mdl-36413532

ABSTRACT

BACKGROUND: The adaptation process for first-year medical students is an important problem because it significantly affects educational activities. The previous study showed that 63% of students had difficulties adapting to the learning process in their first year at medical school. Therefore, students need the most suitable learning style to support the educational process, such as Problem-based learning (PBL). This method can improve critical thinking skills, problem-solving and self-directed learning. Although PBL has been adopted in medical education, the effectiveness of PBL in first-year medical students is still not yet clear. The purpose of this meta-analysis is to verify whether the PBL approach has a positive effect in improving knowledge, problem-solving and self-directed learning in first-year medical students compared with the conventional method. METHODS: We searched PubMed, ScienceDirect, Cochrane, and Google Scholar databases until June 5, 2021. Search terms included problem-based learning, effectiveness, effectivity, and medical student. We excluded studies with the final-year medical student populations. All analyses in our study were carried out using Review Manager version 5.3 (RevMan Cochrane, London, UK). RESULT: Seven eligible studies (622 patients) were included. The pooled analysis demonstrated no significant difference between PBL with conventional learning method in critical thinking/knowledge assessment (p = 0.29), problem-solving aspect (p = 0.47), and self-directed learning aspect (p = 0.34). CONCLUSION: The present study concluded that the PBL approach in first-year medical students appeared to be ineffective in improving critical thinking/knowledge, problem-solving, and self-directed compared with the conventional teaching method.


Subject(s)
Problem-Based Learning , Students, Medical , Humans , Problem-Based Learning/methods , Thinking , Problem Solving , Learning
3.
Am J Physiol Renal Physiol ; 311(2): F343-51, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27279487

ABSTRACT

Na(+)/H(+) exchanger (NHE)3, a major Na(+) transporter in the luminal membrane of the proximal tubule, is subject to ANG II regulation in renal Na(+)/fluid absorption and blood pressure control. We have previously shown that inositol 1,4,5-trisphosphate receptor-binding protein released with inositol 1,4,5-trisphosphate (IRBIT) mediates ANG II-induced exocytosis of NHE3 in cultured proximal tubule epithelial cells. In searching for scaffold protein(s) that coordinates with IRBIT in NHE3 trafficking, we found that NHE regulatory factor (NHERF)1, NHE3, and IRBIT proteins were coexpressed in the same macrocomplexes and that loss of ANG II type 1 receptors decreased their expression in the renal brush-border membrane. We found that NHERF1 was required for ANG II-mediated forward trafficking and activation of NHE3 in cultured cells. ANG II induced a concomitant increase of NHERF1 interactions with NHE3 and IRBIT, which were abolished when the NHERF1 PDZ1 domain was removed. Overexpression of a yellow fluorescent protein-NHERF1 construct that lacks PDZ1, but not PDZ2, failed to exaggerate the ANG II-dependent increase of NHE3 expression in the apical membrane. Moreover, exogenous expression of PDZ1 exerted a dominant negative effect on NHE3 activation by ANG II. We further demonstrated that IRBIT was indispensable for the ANG II-provoked increase in NHERF1-NHE3 interactions and that phosphorylation of IRBIT at Ser(68) was necessary for the assembly of the NHEF1-IRBIT-NHE3 complex. Taken together, our findings suggest that NHERF1 mediates ANG II-induced activation of renal NHE3, which requires coordination between IRBIT and the NHERF1 PDZ1 domain in binding and transporting NHE3.


Subject(s)
Adenosylhomocysteinase/metabolism , Angiotensin II/pharmacology , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/agonists , Sodium-Hydrogen Exchangers/metabolism , Animals , Biotinylation , Cell Line , Hydrogen-Ion Concentration , Mice , Mice, Knockout , Microvilli/metabolism , Plasmids/genetics , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Sodium/metabolism , Sodium-Hydrogen Exchanger 3
4.
PLoS One ; 10(9): e0137513, 2015.
Article in English | MEDLINE | ID: mdl-26352431

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a cytokine that has broad effects on immune system and inflammatory response. A growing body of evidence implicates the role of MIF in tumor growth and metastasis. Lysophosphatidic acid (LPA), a bioactive lipid mediator, regulates colon cancer cell proliferation, invasion, and survival through LPA2 receptor. Loss of LPA2 results in decreased expression of MIF in a rodent model of colon cancer, but the mechanism of MIF regulation by LPA is yet to be determined. In this study, we show that LPA transcriptionally regulates MIF expression in colon cancer cells. MIF knockdown decreased LPA-mediated proliferation of HCT116 human adenocarcinoma cells without altering the basal proliferation rates. Conversely, extracellular recombinant MIF stimulated cell proliferation, suggesting that the effect of MIF may in part be mediated through activation of surface receptor. We have shown recently that LPA increases hypoxia-inducible factor 1α (HIF1α) expression. We found that MIF regulation by LPA was ablated by knockdown of HIF1α, indicating that MIF is a transcriptional target of HIF1α. Conversely, knockdown of MIF ablated an increase in HIF1α expression in LPA-treated cells, suggesting a reciprocal relationship between HIF1α and MIF. LPA stimulated co-immunoprecipitation of HIF1α and MIF, indicating that their association is necessary for stabilization of HIF1α. It has been shown previously that CSN9 signalosome subunit 5 (CSN5) interacts with HIF1α to stabilize HIF1α under aerobic conditions. We found that LPA did not alter expression of CSN5, but stimulated its interaction with HIF1α and MIF. Depletion of CSN5 mitigated the association between HIF1α and MIF, indicating that CSN5 acts as a physical link. We suggest that HIF1α, MIF, and CSN5 form a ternary complex whose formation is necessary to prevent degradation of HIF1α under aerobic conditions.


Subject(s)
Gene Expression/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intramolecular Oxidoreductases/metabolism , Lysophospholipids/pharmacology , Macrophage Migration-Inhibitory Factors/metabolism , Peptide Hydrolases/metabolism , Signal Transduction/drug effects , COP9 Signalosome Complex , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Peptide Hydrolases/genetics
5.
Am J Physiol Cell Physiol ; 309(1): C14-21, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25855080

ABSTRACT

Na(+)/H(+) exchange by Na(+)/H(+) exchanger 3 (NHE3) is a major route of sodium absorption in the intestine and kidney. We have shown previously that lysophosphatidic acid (LPA), a small phospholipid produced ubiquitously by all types of cells, stimulates NHE3 via LPA5 receptor. Stimulation of NHE3 activity by LPA involves LPA5 transactivating EGF receptor (EGFR) in the apical membrane. EGFR activates proline-rich tyrosine kinase 2 (Pyk2) and ERK, both of which are necessary for NHE3 regulation. However, Pyk2 and ERK are regulated by EGFR via independent pathways and appear to converge on an unidentified intermediate that ultimately targets NHE3. The p90 ribosomal S6 kinase (RSK) family of Ser/Thr protein kinases is a known effector of EGFR and ERK. Hence, we hypothesized that RSK may be the convergent effector of Pyk2 and ERK although it is not known whether Pyk2 regulates RSK. In this study, we show that Pyk2 is necessary for the maintenance of phosphoinositide-dependent kinase 1 (PDK1) autophosphorylation, and knockdown of Pyk2 or PDK1 mitigated LPA-induced phosphorylation of RSK and stimulation of NHE3 activity. Additionally, we show that RSK2, but not RSK1, is responsible for NHE3 regulation. RSK2 interacts with NHE3 at the apical membrane domain, where it phosphorylates NHE3. Alteration of S663 of NHE3 ablated LPA-induced phosphorylation of NHE3 and stimulation of the transport activity. Our study identifies RSK2 as a new kinase that regulates NHE3 activity by direct phosphorylation.


Subject(s)
Cell Membrane/enzymology , Lysophospholipids/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sodium-Hydrogen Exchangers/drug effects , 3-Phosphoinositide-Dependent Protein Kinases/genetics , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Animals , Caco-2 Cells , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , Humans , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Mutation , Phosphorylation , RNA Interference , Receptor Cross-Talk , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Transfection
6.
Cell Signal ; 27(5): 961-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25683913

ABSTRACT

Lysophosphatidic acid (LPA) is a simple phospholipid with potent mitogenic effects on various cells including colon cancer cells. LPA stimulates proliferation of colon cancer cells by activation of ß-catenin or Krüppel-like factor 5 (KLF5), but the functional relationship between these two transcription factors is not clear. Hence, we sought to investigate the mechanism of ß-catenin activation by LPA and the role of KLF5 in the regulation of ß-catenin by LPA. We found that LPA and Wnt3 additively activated the ß-catenin/TCF (T cell factor) reporter activity in HCT116 cells. In addition to phosphorylating glycogen synthase kinase 3ß (GSK-3ß) at Ser9, LPA resulted in phosphorylation of ß-catenin at Ser552 and Ser675. Mutation of Ser552 and Ser675 ablated LPA-induced ß-catenin/TCF transcriptional activity. Knockdown of KLF5 significantly attenuated activation of ß-catenin/TCF reporter activity by LPA but not by Wnt3. However, nuclear accumulation of ß-catenin by LPA was not altered by knockdown of KLF5. ß-catenin, TCF, and KLF5 were present in a 250-300kDa macro-complex, and their presence was enhanced by LPA. LPA simulated the interaction of ß-catenin with TCF4, and depletion of KLF5 decreased ß-catenin-TCF4 association and the transcriptional activity. In summary, LPA activates ß-catenin by multiple pathways involving phosphorylation of GSK-3 and ß-catenin, and enhancing ß-catenin interaction with TCF4. KLF5 plays a critical role in ß-catenin activation by increasing the ß-catenin-TCF4 interaction.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Colonic Neoplasms/metabolism , Kruppel-Like Transcription Factors/metabolism , Lysophospholipids/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/pathology , Colon/metabolism , Colon/pathology , Colonic Neoplasms/pathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Phosphorylation , Transcription Factor 4
7.
J Biol Chem ; 289(26): 18360-72, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24831004

ABSTRACT

Na(+)/H(+) exchanger NHE3 expressed in the intestine and kidney plays a major role in NaCl and HCO3 (-) absorption that is closely linked to fluid absorption and blood pressure regulation. The Nedd4 family of E3 ubiquitin ligases interacts with a number of transporters and channels via PY motifs. A comparison of NHE3 sequences revealed the presence of PY motifs in NHE3s from human and several non-human primates but not in non-primate NHE3s. In this study we evaluated the differences between human and non-primate NHE3s in ubiquitination and interaction with Nedd4-2. We found that Nedd4-2 ubiquitinated human NHE3 (hNHE3) and altered its expression and activity. Surprisingly, rat NHE3 co-immunoprecipitated Nedd4-2, but its expression and activity were not altered by silencing of Nedd4-2. Ubiquitination by Nedd4-2 rendered hNHE3 to undergo internalization at a significantly greater rate than non-primate NHE3s without altering protein stability. Insertion of a PY motif in rabbit NHE3 recapitulated the interaction with Nedd4-2 and enhanced internalization. Thus, we propose a new model where disruption of Nedd4-2 interaction elevates hNHE3 expression and activity.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Sodium-Hydrogen Exchangers/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/genetics , Gene Expression Regulation , Humans , Mammals/classification , Mammals/genetics , Molecular Sequence Data , Nedd4 Ubiquitin Protein Ligases , Opossums , Phylogeny , Protein Binding , Rabbits , Rats , Sequence Alignment , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
8.
J Biol Chem ; 288(35): 25244-25253, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23880760

ABSTRACT

Hypoxia-inducible factor 1α (HIF-1α) and p53 are pivotal regulators of tumor growth. Lysophosphatidic acid (LPA) is a lipid mediator that functions as a mitogen by acting through LPA receptors. We have shown previously that LPA stimulates HIF-1α expression in colon cancer cells. To determine the mechanism of HIF-1α induction by LPA, we compared the effect of LPA on HIF-1α in several colon cancer cell lines. LPA transcriptionally induced HIF-1α in colon cancer cells. HIF-1α induction was observed in cells expressing WT p53, where LPA decreased p53 expression. However, LPA failed to induce HIF-1α when the p53 gene was mutated. A decrease in p53 expression was dependent on induction of p53-specific E3 ubiquitin ligase Mdm2 by LPA. Krüppel-like factor 5 (KLF5) is an effector of LPA-induced proliferation of colon cancer cells. Because HIF-1α was necessary for LPA-induced growth of colon cancer cells, we determined the relationship between KLF5 and HIF-1α by a loss-of-function approach. Silencing of KLF5 inhibited LPA-induced HIF-1α induction, suggesting that KLF5 is an upstream regulator of HIF-1α. KLF5 and p53 binding to the Hif1α promoter was assessed by ChIP assay. LPA increased the occupancy of the Hif1α promoter by KLF5, while decreasing p53 binding. Transfection of HCT116 cells with KLF5 or p53 attenuated the binding of the other transcription factor. These results identify KLF5 as a transactivator of HIF-1α and show that LPA regulates HIF-1α by dynamically modulating its interaction with KLF5 and p53.


Subject(s)
Colonic Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Transcription Factors/metabolism , Lysophospholipids/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Silencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kruppel-Like Transcription Factors/genetics , Lysophospholipids/genetics , Mutation , Protein Binding , Response Elements , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics
9.
Am J Physiol Gastrointest Liver Physiol ; 303(2): G180-8, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22556145

ABSTRACT

The Caco-2 cell line represents absorptive polarized intestinal epithelial cells that express multiple forms of Na(+)/H(+) exchanger (NHE) in their plasma membranes. Caco-2 cells express the major apical NHE isoform NHE3, but low NHE3 expression together with inefficient transfection often hamper intended studies. In this study, we examined whether SK-CO15 cells could be used to study NHE3 regulation. SK-CO15 cells grown on Transwell inserts developed polarized epithelial cells with microvilli. The transfection efficiency of SK-CO15 cells was markedly higher compared with Caco-2 cells, an advantage in gene transfer and knockout. SK-CO15 cells expressed NHE1, NHE2, and NHE3. NHE3 expression was significantly greater in these cells than Caco-2, and NHE3 comprised more than half of total NHE activity. Apical expression of NHE3 in SK-CO15 cells was confirmed by confocal immunofluorescence and surface biotinylation. NHE regulatory factors NHERF1 and NHERF2, which are important for regulation of NHE3 activity, were expressed in these cells. Stimulatory response of NHE3 in SK-CO15 cells was assessed by dexamethasone and lysophosphatidic acid (LPA). Treatment with dexamethasone for 24-48 h increased NHE3 expression and activity. Similarly to Caco-2 cells, SK-CO15 cells lacked the expression of the LPA receptor LPA(5,) but exogenous expression of LPA(5) resulted in acute stimulation of NHE3. Forskolin acutely inhibited NHE3 activity in SK-CO15 cells, further attesting the validity of these cells. We conclude that SK-CO15 cells with the amenity for transfection and high endogenous NHE3 expression are a new and better cell model for NHE3 regulatory investigation than widely used Caco-2 cells.


Subject(s)
Intestinal Mucosa/physiology , Sodium-Hydrogen Exchangers/biosynthesis , Cation Transport Proteins/biosynthesis , Cell Line , Cell Polarity/drug effects , Cell Polarity/physiology , Colforsin/pharmacology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Humans , Intestinal Mucosa/drug effects , Lysophospholipids/pharmacology , Phosphoproteins/biosynthesis , Receptors, Lysophosphatidic Acid/biosynthesis , Receptors, Lysophosphatidic Acid/genetics , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Transfection
10.
Cell Motil Cytoskeleton ; 52(2): 66-81, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12112149

ABSTRACT

The distribution of two proteins in Naegleria gruberi, N-gammaTRP (Naegleria gamma-tubulin-related protein) and N-PRP (Naegleria pericentrin-related protein), was examined during the de novo formation of basal bodies and flagella that occurs during the differentiation of N. gruberi. After the initiation of differentiation, N-gammaTRP and N-PRP began to concentrate at the same site within cells. The percentage of cells with a concentrated region of N-gammaTRP and N-PRP was maximal (68%) at 40 min when the synthesis of tubulin had just started but no assembled microtubules were visible. When concentrated tubulin became visible (60 min), the region of concentrated N-gammaTRP and N-PRP was co-localized with the tubulin spot and then flagella began to elongate from the region of concentrated tubulin. When cells had elongated flagella, the concentrated N-gammaTRP and N-PRP were translocated to the opposite end of the flagellated cells and disappeared. The transient concentration of N-gammaTRP coincided with the transient formation of an F-actin spot at which N-gammaTRP and alpha-tubulin mRNA were co-localized. The concentration of N-gammaTRP and formation of the F-actin spot occurred without the formation of microtubules but were inhibited by cytochalasin D. These observations suggest that the regional concentration of N-gammaTRP and N-PRP is mediated by actin filaments and might provide a site of microtubule nucleation for the assembly of newly synthesized tubulins into basal bodies and flagella.


Subject(s)
Antigens/analysis , Centrioles/chemistry , Flagella/chemistry , Naegleria/growth & development , Protozoan Proteins/analysis , Tubulin/analysis , Actins/metabolism , Animals , Antigens/physiology , Cell Differentiation , Cytochalasin D/metabolism , Flagella/metabolism , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/classification , Naegleria/cytology , Naegleria/genetics , Protozoan Proteins/physiology , RNA, Messenger/metabolism , Tubulin/chemistry , Tubulin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...