Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Respirol Case Rep ; 11(4): e01119, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36910132

ABSTRACT

Melioidosis is a rare but often fatal tropical infection caused by gram-negative bacteria Burkholderia pseudomallei. It most commonly manifests as pneumonia and rarely presents as pericarditis. Melioidosis can be difficult to diagnose because of its diverse clinical manifestation and close resemblance to bacteria of the genus Pseudomonas. We report a rare case of melioidosis presenting as pericarditis and pneumonia in a 61-year-old male patient with poorly controlled diabetes mellitus. He was initially misdiagnosed with Pseudomonas aeruginosa infection and later treated empirically as tuberculosis pericarditis for 2 months, before reaching the diagnosis of melioidosis.

2.
J Orthop Translat ; 35: 99-112, 2022 Jul.
Article in English | MEDLINE | ID: mdl-36262374

ABSTRACT

Background: Metabolic disruption commonly follows Anterior Cruciate Ligament Reconstruction (ACLR) surgery. Brief exposure to low amplitude and frequency pulsed electromagnetic fields (PEMFs) has been shown to promote in vitro and in vivo murine myogeneses via the activation of a calcium-mitochondrial axis conferring systemic metabolic adaptations. This randomized-controlled pilot trial sought to detect local changes in muscle structure and function using MRI, and systemic changes in metabolism using plasma biomarker analyses resulting from ACLR, with or without accompanying PEMF therapy. Methods: 20 patients requiring ACLR were randomized into two groups either undergoing PEMF or sham exposure for 16 weeks following surgery. The operated thighs of 10 patients were exposed weekly to PEMFs (1 â€‹mT for 10 â€‹min) for 4 months following surgery. Another 10 patients were subjected to sham exposure and served as controls to allow assessment of the metabolic repercussions of ACLR and PEMF therapy. Blood samples were collected prior to surgery and at 16 weeks for plasma analyses. Magnetic resonance data were acquired at 1 and 16 weeks post-surgery using a Siemens 3T Tim Trio system. Phosphorus (31P) Magnetic Resonance Spectroscopy (MRS) was utilized to monitor changes in high-energy phosphate metabolism (inorganic phosphate (Pi), adenosine triphosphate (ATP) and phosphocreatine (PCr)) as well as markers of membrane synthesis and breakdown (phosphomonoesters (PME) and phosphodiester (PDE)). Quantitative Magnetization Transfer (qMT) imaging was used to elucidate changes in the underlying tissue structure, with T1-weighted and 2-point Dixon imaging used to calculate muscle volumes and muscle fat content. Results: Improvements in markers of high-energy phosphate metabolism including reductions in ΔPi/ATP, Pi/PCr and (Pi â€‹+ â€‹PCr)/ATP, and membrane kinetics, including reductions in PDE/ATP were detected in the PEMF-treated cohort relative to the control cohort at study termination. These were associated with reductions in the plasma levels of certain ceramides and lysophosphatidylcholine species. The plasma levels of biomarkers predictive of muscle regeneration and degeneration, including osteopontin and TNNT1, respectively, were improved, whilst changes in follistatin failed to achieve statistical significance. Liquid chromatography with tandem mass spectrometry revealed reductions in small molecule biomarkers of metabolic disruption, including cysteine, homocysteine, and methionine in the PEMF-treated cohort relative to the control cohort at study termination. Differences in measurements of force, muscle and fat volumes did not achieve statistical significance between the cohorts after 16 weeks post-ACLR. Conclusion: The detected changes suggest improvements in systemic metabolism in the post-surgical PEMF-treated cohort that accords with previous preclinical murine studies. PEMF-based therapies may potentially serve as a manner to ameliorate post-surgery metabolic disruptions and warrant future examination in more adequately powered clinical trials. The Translational Potential of this Article: Some degree of physical immobilisation must inevitably follow orthopaedic surgical intervention. The clinical paradox of such a scenario is that the regenerative potential of the muscle mitochondrial pool is silenced. The unmet need was hence a manner to maintain mitochondrial activation when movement is restricted and without producing potentially damaging mechanical stress. PEMF-based therapies may satisfy the requirement of non-invasively activating the requisite mitochondrial respiration when mobility is restricted for improved metabolic and regenerative recovery.

3.
J Obstet Gynaecol Res ; 48(7): 1997-2004, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35365933

ABSTRACT

Cervical varices are a rare condition characterized by recurrent antepartum hemorrhage and less than 20 cases were reported in the literature. It is usually associated with placenta previa. We herein describe four cases of cervical varices without placenta previa. Meticulous speculum examination, ultrasonography with Doppler and colposcopy are essential for establishing the diagnosis and assessing the extent of the cervical varix. We propose to classify it as the apparent external os type or ultrasonography-based endocervical type. Most cases presented in the literature were delivered by cesarean section. Nevertheless, one of our cases was a successful vaginal delivery. Our case illustrates that vaginal delivery is possible in isolated cervical varices. More case reports are needed to have a better understanding of this rare entity.


Subject(s)
Placenta Previa , Varicose Veins , Cervix Uteri/diagnostic imaging , Cesarean Section/adverse effects , Female , Humans , Placenta Previa/diagnostic imaging , Pregnancy , Uterine Hemorrhage/etiology , Varicose Veins/diagnostic imaging
5.
Front Mol Biosci ; 8: 699827, 2021.
Article in English | MEDLINE | ID: mdl-34513920

ABSTRACT

Angiotensin II (Ang II) plays an important role in regulating various physiological processes. However, little is known about the existence of intracellular Ang II (iAng II), whether iAng II would regulate the automaticity of early differentiating cardiomyocytes, and the underlying mechanism involved. Here, iAng II was detected by immunocytochemistry and ultra-high performance liquid chromatography combined with electrospray ionization triple quadrupole tandem mass spectrometry in mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) and neonatal rat ventricular myocytes. Expression of AT1R-YFP in mESC-CMs revealed that Ang II type 1 receptors were located on the surface membrane, while immunostaining of Ang II type 2 receptors (AT2R) revealed that AT2R were predominately located on the nucleus and the sarcoplasmic reticulum. While extracellular Ang II increased spontaneous action potentials (APs), dual patch clamping revealed that intracellular delivery of Ang II or AT2R activator C21 decreased spontaneous APs. Interestingly, iAng II was found to decrease the caffeine-induced increase in spontaneous APs and caffeine-induced calcium release, suggesting that iAng II decreased spontaneous APs via the AT2R- and ryanodine receptor-mediated pathways. This is the first study that provides evidence of the presence and function of iAng II in regulating the automaticity behavior of ESC-CMs and may therefore shed light on the role of iAng II in fate determination.

6.
Comput Math Methods Med ; 2020: 8861035, 2020.
Article in English | MEDLINE | ID: mdl-33144873

ABSTRACT

Prostate segmentation in multiparametric magnetic resonance imaging (mpMRI) can help to support prostate cancer diagnosis and therapy treatment. However, manual segmentation of the prostate is subjective and time-consuming. Many deep learning monomodal networks have been developed for automatic whole prostate segmentation from T2-weighted MR images. We aimed to investigate the added value of multimodal networks in segmenting the prostate into the peripheral zone (PZ) and central gland (CG). We optimized and evaluated monomodal DenseVNet, multimodal ScaleNet, and monomodal and multimodal HighRes3DNet, which yielded dice score coefficients (DSC) of 0.875, 0.848, 0.858, and 0.890 in WG, respectively. Multimodal HighRes3DNet and ScaleNet yielded higher DSC with statistical differences in PZ and CG only compared to monomodal DenseVNet, indicating that multimodal networks added value by generating better segmentation between PZ and CG regions but did not improve the WG segmentation. No significant difference was observed in the apex and base of WG segmentation between monomodal and multimodal networks, indicating that the segmentations at the apex and base were more affected by the general network architecture. The number of training data was also varied for DenseVNet and HighRes3DNet, from 20 to 120 in steps of 20. DenseVNet was able to yield DSC of higher than 0.65 even for special cases, such as TURP or abnormal prostate, whereas HighRes3DNet's performance fluctuated with no trend despite being the best network overall. Multimodal networks did not add value in segmenting special cases but generally reduced variations in segmentation compared to the same matched monomodal network.


Subject(s)
Algorithms , Image Interpretation, Computer-Assisted/statistics & numerical data , Multiparametric Magnetic Resonance Imaging/statistics & numerical data , Prostatic Neoplasms/diagnostic imaging , Computational Biology , Databases, Factual , Deep Learning , Humans , Machine Learning , Male , Mathematical Concepts , Neural Networks, Computer , Pattern Recognition, Automated , Prostatic Neoplasms/pathology
7.
Int J Cardiol ; 203: 169-81, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26512833

ABSTRACT

BACKGROUND: Cardiac pacemaking is a complex phenomenon that is not completely understood. Canonical transient receptor potential isoform 3 (TRPC3) channel is a cation channel that permeates both Ca(2+) and Na(+). TRPC3 was previously found to express in adult cardiomyocytes. However, its role in cardiac pacemaking is unexplored. Here we used mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) to investigate whether TRPC3 regulates the spontaneous automaticity and the underlying mechanism involved. METHODS AND RESULTS: Immunocytochemistry results showed that TRPC3 is expressed at the T-tubules of mESC-CMs. Whole-cell patch clamping showed that single mESC-CMs contain TRPC3 current. Confocal Ca(2+) imaging showed that the TRPC3-specific blocker Pyr3 decreased Ca(2+) transients and local Ca(2+) release (LCR) of mESC-CMs. Combined current and voltage clamp recordings from the same cell showed that reducing the TRPC3 current, either by Pyr3 or a dominant negative (loss-of-function) construct of TRPC3, decreased the pacemaker activity of mESC-CMs as reflected by a decrease in action potential rate, a depolarized maximum diastolic potential and a decrease in slope of phase 4 diastolic depolarization. Furthermore, decreasing the TRPC3 current diminished, while increasing the TRPC3 current augmented the sodium-calcium exchanger (NCX) current in mESC-CMs. Lastly, decrease in TRPC3 current decreased the phosphorylation of ryanodine receptor isoform 2 at Ser2809 and phospholamban at Thr17. CONCLUSIONS: TRPC3 positively regulates diastolic depolarization of spontaneous action potential by increasing LCR and NCX current and therefore is an important determinant in pacemaking of mESC-CMs.


Subject(s)
Action Potentials , Embryonic Stem Cells , Myocytes, Cardiac/physiology , TRPC Cation Channels/physiology , Animals , Mice
8.
PLoS One ; 10(7): e0133211, 2015.
Article in English | MEDLINE | ID: mdl-26208267

ABSTRACT

Cytosolic Ca2+ ([Ca2+]i) is an important signal that regulates cardiomyocyte differentiation during cardiogenesis. TRPV1 is a Ca2+-permeable channel that is expressed in cardiomyocytes. In the present study, we utilized mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) as a model to investigate the functional role of TRPV1 in cardiomyocyte differentiation. Induction of embryonic stem cells into cardiomyocytes was achieved using embryoid body (EB)-based differentiation method. Quantitative PCRs showed an increased TRPV1 expression during the differentiation process. In [Ca2+]i measurement study, application of TRPV1 agonists, capsaicin and camphor, elicited a [Ca2+]i rise in mESC-CMs, the effect of which was abolished by TRPV1-shRNA. In functional study, treatment of EBs with TRPV1 antagonists (capsazepine and SB366791) and TRPV1-shRNA reduced the size of the EBs and decreased the percentage of spontaneously beating EBs. TRPV1 antagonists and TRPV1-shRNA also suppressed the expression of cardiomyocyte marker genes, including cardiac actin, c-TnT, c-TnI, and α-MHC. Taken together, this study demonstrated an important functional role of TRPV1 channels in the differentiation of mESCs into cardiomyocytes.


Subject(s)
Cell Differentiation/genetics , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , TRPV Cation Channels/genetics , Animals , Biomarkers , Cells, Cultured , Gene Expression , Mice , RNA, Messenger/genetics , RNA, Small Interfering/genetics , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors
9.
Int J Cardiol ; 168(4): 3458-72, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-23706318

ABSTRACT

BACKGROUND: The mechanism of how reactive oxygen species (ROS) regulate cardiac differentiation in the long-run is unclear and the effect of pro-inflammatory cytokines secreted during myocardial infarction on the cardiac differentiation of embryonic stem cells (ESCs) is unknown. The aims of this study were 1) to investigate the effect of ROS on cardiac differentiation and the regulations of transcription factors in ESC differentiation cultures and 2) to investigate the effect of pro-inflammatory cytokines on the expression of cardiac structural genes and whether this effect is mediated through ROS signaling. METHODS: ESCs were differentiated using hanging drop method. Degree of cardiac differentiation was determined by the appearance of beating embryoid bodies (EBs) and by the expression of cardiac genes using real-time PCR and Western blot. Intracellular ROS level was examined by confocal imaging. RESULTS: H2O2-treated EBs were found to have enhanced cardiac differentiation in the long run as reflected by, firstly, an earlier appearance of beating EBs, and secondly, an upregulation in cardiac structural protein expression at both mRNA and protein levels. Also, ROS upregulated the expression of several cardiac-related transcription factors, and increased the post-translationally-activated transcription factors SRF and AP-1. IL-1ß, IL-10, IL-18 and TNF-α upregulated the expression of cardiac structural proteins and increased the ROS level in differentiating EBs. In addition, ROS scavenger reversed the cardiogenic effect of IL-10 and IL-18. CONCLUSIONS: These results demonstrated that ROS enhance cardiac differentiation of ESCs through upregulating the expression and activity of multiple cardiac-related transcription factors. IL-1ß, IL-10, IL-18 and TNF-α enhance cardiac differentiation and ROS may serve as the messenger in cardiogenic signaling from these cytokines.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Inflammation Mediators/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Cytokines/biosynthesis , Cytokines/metabolism , Mice , Myocardial Infarction/pathology , Transcription Factors/physiology
10.
J Cell Physiol ; 227(6): 2519-30, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21898397

ABSTRACT

Embryonic stem cells (ESCs) can self-renew indefinitely and differentiate into all cell lineages. Calcium is a universal second messenger which regulates a number of cellular pathways. Previous studies showed that store-operated calcium channels (SOCCs) but not voltage-operated calcium channels are present in mouse ESCs (mESCs). In this study, store-operated calcium entry (SOCE) was found to exist in mESCs using confocal microscopy. SOCC blockers lanthanum, 2-aminoethoxydiphenyl borate (2-APB) and SKF-96365 reduced mESC proliferation in a concentration-dependent manner, suggesting that SOCE is important for ESC proliferation. Pluripotent markers, Sox-2, Klf-4, and Nanog, were down-regulated by 2-APB, suggesting that self-renewal property of mESCs relies on SOCE. 17ß-estradiol (E2) enhanced mESC proliferation. This enhanced proliferation was associated with an increment of SOCE. Both stimulated proliferation and increased SOCE could be reversed by SOCC blockers suggesting that E2 mediates its stimulatory effect on proliferation via enhancing SOCE. Also, cyclosporin A and INCA-6, inhibitors of calcineurin [phosphatase that de-phosphorylates and activates nuclear factor of activated T-cells (NFAT)], reversed the proliferative effect of E2, indicating that NFAT is involved in E2-stimulated proliferation. Interestingly, E2 caused the nuclear translocation of NFATc4, and this could be reversed by 2-APB. These results suggested that NFATc4 is the downstream target of E2-induced SOCE. The present investigation provides the first line of evidence that SOCE and NFAT are crucial for ESCs to maintain their unique characteristics. In addition, the present investigation also provides novel information on the mechanisms of how E2, an important female sex hormone, affects ESC proliferation.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Cell Proliferation , Embryonic Stem Cells/metabolism , Estradiol/metabolism , NFATC Transcription Factors/metabolism , Pluripotent Stem Cells/metabolism , Animals , Biomarkers/metabolism , Calcineurin/metabolism , Calcineurin Inhibitors , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Signaling/drug effects , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/pharmacology , Homeodomain Proteins/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Microscopy, Confocal , NFATC Transcription Factors/genetics , Nanog Homeobox Protein , Pluripotent Stem Cells/drug effects , Recombinant Fusion Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Time Factors , Transfection
11.
J Pain ; 12(5): 600-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21296029

ABSTRACT

UNLABELLED: Irritable bowel syndrome (IBS), characterized mainly by abdominal pain, is a functional bowel disorder. The present study aimed to examine changes in the excitability and the activity of the voltage-gated K(+) channel in dorsal root ganglia (DRG) neurons innervating the colon of rats subjected to neonatal maternal separation (NMS). Colonic DRG neurons from NMS rats as identified by FAST DiI™ labeling showed an increased cell size compared with those from nonhandled (NH) rats. Whole cell current-clamp recordings showed that colonic DRG neurons from NMS rats displayed: 1) depolarized resting membrane potential; 2) increased input resistance; 3) a dramatic reduction in rheobase; and 4) a significant increase in the number of action potentials evoked at twice rheobase. Whole cell voltage-clamp recordings revealed that neurons from both groups exhibited transient A-type (I(A)) and delayed rectifier (I(K)) K(+) currents. Compared with NH rat neurons, the averaged density of I(K) was significantly reduced in NMS rat neurons. Furthermore, the Kv1.2 expression was significantly decreased in NMS rat colonic DRG neurons. These results suggest that NMS increases the excitability of colonic DRG neurons mainly by suppressing the I(K) current, which is likely accounted for by the downregulation of the Kv1.2 expression and somal hypertrophy. PERSPECTIVE: This study demonstrates the alteration of delayed rectifier K current and Kv1.2 expression in DRG neurons from IBS model rats, representing a molecular mechanism underlying visceral pain and sensitization in IBS, suggesting the potential of Kv1.2 as a therapeutic target for the treatment of IBS.


Subject(s)
Action Potentials/physiology , Colon/innervation , Ganglia, Spinal/physiology , Maternal Deprivation , Neurons/physiology , Potassium Channels, Voltage-Gated/metabolism , Animals , Colon/metabolism , Down-Regulation , Fluorescent Antibody Technique , Ganglia, Spinal/cytology , Male , Membrane Potentials/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
12.
Pflugers Arch ; 461(1): 191-202, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21110038

ABSTRACT

Embryonic stem cells (ESCs) can uniquely proliferate indefinitely and differentiate into all cell lineages. ESCs may therefore provide an unlimited supply of cells for cell-based therapies. Previous study reported the presence of hyperpolarization-activated inward currents in undifferentiated mouse (m) ESCs, but the functional role of this hyperpolarization-activated current in mESCs is unknown. In this study, the role of this current in maintaining the proliferative capacity and the cell cycle progression of ESCs was investigated. In D3 mESCs, this hyperpolarization-activated inward current can be blocked by HCN channel blocker ZD7288. Application of the HCN channel blockers, cesium (1-10 mM) or ZD7288 (0.1-30 µM), attenuated cell proliferation in a concentration-dependent manner. Both HCN blockers were found to be non-cytotoxic to mESCs as determined by cell viability test. Interestingly, ZD7288 at 10 and 30 µM was found to decrease the proportion of cells in G(0)/G(1) phase and increase the proportion of cells in S phase. This suggests that this hyperpolarization-activated current can affect the cell cycle progression in mESCs. In summary, the present investigation suggests that ESC proliferation and cell cycle progression can be regulated by this hyperpolarization-activated current.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Embryonic Stem Cells/cytology , Animals , Cell Survival/drug effects , Cesium/pharmacology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Cyclin B/biosynthesis , Embryonic Stem Cells/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membrane Potentials , Mice , Patch-Clamp Techniques , Pyrimidines/pharmacology
13.
Am J Physiol Cell Physiol ; 299(6): C1234-49, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20844252

ABSTRACT

Myocardial infarction has been the leading cause of morbidity and mortality in developed countries over the past few decades. The transplantation of cardiomyocytes offers a potential method of treatment. However, cardiomyocytes are in high demand and their supply is extremely limited. Embryonic stem cells (ESCs), which have been isolated from the inner cell mass of blastocysts, can self-renew and are pluripotent, meaning they have the ability to develop into any type of cell, including cardiomyocytes. This suggests that ESCs could be a good source of genuine cardiomyocytes for future therapeutic purposes. However, problems with the yield and purity of ESC-derived cardiomyocytes, among other hurdles for the therapeutic application of ESC-derived cardiomyocytes (e.g., potential immunorejection and tumor formation problems), need to be overcome before these cells can be used effectively for cell replacement therapy. ESC-derived cardiomyocytes consist of nodal, atrial, and ventricular cardiomyocytes. Specifically, for treatment of myocardial infarction, transplantation of a sufficient quantity of ventricular cardiomyocytes, rather than nodal or atrial cardiomyocytes, is preferred. Hence, it is important to find ways of increasing the yield and purity of specific types of cardiomyocytes. Atrial and ventricular cardiomyocytes have differential expression of genes (transcription factors, structural proteins, ion channels, etc.) and are functionally distinct. This paper presents a thorough review of differential gene expression in atrial and ventricular myocytes, their expression throughout development, and their regulation. An understanding of the molecular and functional differences between atrial and ventricular myocytes allows discussion of potential strategies for preferentially directing ESCs to differentiate into chamber-specific cells, or for fine tuning the ESC-derived cardiomyocytes into specific electrical and contractile phenotypes resembling chamber-specific cells.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression , Heart Atria/cytology , Heart Ventricles/cytology , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/transplantation , Animals , Humans , Ion Channels , Mice , Myocardial Contraction , Pluripotent Stem Cells/metabolism , Stem Cell Transplantation
14.
J Cell Physiol ; 224(1): 165-77, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20333647

ABSTRACT

Embryonic stem cells (ESCs) possess two unique characteristics: self-renewal and pluripotency. In this study, roles of voltage-gated potassium channels (K(v)) in maintaining mouse (m) ESC characteristics were investigated. Tetraethylammonium (TEA(+)), a K(v) blocker, attenuated cell proliferation in a concentration-dependent manner. Possible reasons for this attenuation, including cytotoxicity, cell cycle arrest and differentiation, were examined. Blocking K(v) did not change the viability of mESCs. Interestingly, K(v) inhibition increased the proportion of cells in G(0)/G(1) phase and decreased that in S phase. This change in cell cycle distribution can be attributed to cell cycle arrest or differentiation. Loss of pluripotency as determined at both molecular and functional levels was detected in mESCs with K(v) blockade, indicating that K(v) inhibition in undifferentiated mESCs directs cells to differentiate instead of to self-renew and progress through the cell cycle. Membrane potential measurement revealed that K(v) blockade led to depolarization, consistent with the role of K(v) as the key determinant of membrane potential. The present results suggest that membrane potential changes may act as a "switch" for ESCs to decide whether to proliferate or to differentiate: hyperpolarization at G(1) phase would favor ESCs to enter S phase while depolarization would favor ESCs to differentiate. Consistent with this notion, S-phase-synchronized mESCs were found to be more hyperpolarized than G(0)/G(1)-phase-synchronized mESCs. Moreover, when mESCs differentiated, the differentiation derivatives depolarized at the initial stage of differentiation. This investigation is the first study to provide evidence that K(v) and membrane potential affect the fate determination of ESCs.


Subject(s)
Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Animals , Biomarkers/metabolism , Cell Cycle , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Cell Survival , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Gene Expression Regulation, Developmental , Membrane Potentials , Mice , Pluripotent Stem Cells/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Tetraethylammonium/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...