Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Public Health ; 129(12): 1579-84, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26298588

ABSTRACT

OBJECTIVES: Many studies have shown that insomnia is a common problem among university students, but there are wide variations in the prevalence of insomnia. In this systematic review, we aimed to explore the prevalence of insomnia among university students using scientific and conclusive methods. STUDY DESIGN: A systematic review is designed to analyze the studies reporting on prevalence of insomnia among university students. METHODS: Systemic searches were conducted in PubMed, BioMed Central, EBSCO, ScienceDirect, Ovid LWW and Medline databases between January 2000 and July 2014, The Meta analyst software was used to calculate the prevalence rate of each study, the pooled means of prevalence rates and 95% CIs across studies were then calculated and presented. RESULTS: Seven articles that met the inclusion and exclusion criteria were selected. The overall sample size in the current review was 16,478, with a minimum of 219 and a maximum of 10,322. The prevalence rates of the seven studies ranged between 9.4% (95%CI 8.8-10.0%) and 38.2% (95% CI 35.4-41.1%). Overall, the total students studied with a weighted mean prevalence of 18.5% (95% CI 11.2-28.8%), considerably higher than rates of 7.4% (95% CI 5.8-9.0%) reported in general population. CONCLUSIONS: This review emphasized that insomnia prevalence in university students is considerably higher than that in general population, suggested that more attention should be paid to insomnia in university students.


Subject(s)
Sleep Initiation and Maintenance Disorders/epidemiology , Students/statistics & numerical data , Adult , China/epidemiology , Female , Humans , Male , Prevalence , Universities , Young Adult
2.
Zygote ; 14(4): 341-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17266792

ABSTRACT

A major challenge in the widespread application of hES (human embryonic stem) cells in clinical therapy and basic scientific research is the development of efficient cryopreservation protocols. Conventional slow-cooling protocols utilizing standard cryoprotectant concentrations i.e. 10% (v/v) DMSO, yield extremely low survival rates of less than 5% as reported by previous studies. This study characterized cell death in frozen-thawed hES colonies that were cryopreserved under standard conditions. Surprisingly, our results showed that immediately after post-thaw washing, the overwhelming majority of hES cells were viable (approximately 98%), as assessed by the trypan blue exclusion test. However, when the freshly thawed hES colonies were placed in a 37 degrees C incubator, there was a gradual reduction in cell viability over time. The kinetics of cell death was drastically slowed down by keeping the freshly thawed hES colonies at 4 degrees C, with more than 90% of cells remaining viable after 90 min of incubation at 4 degrees C. This effect was reversible upon re-exposing the cells to physiological temperatures. The vast majority of low temperature-exposed hES colonies gradually underwent cell death upon incubation for a further 90 min at 37 degrees C. Hence, our observations would strongly suggest involvement of a self-induced apoptotic mechanism, as opposed to cellular necrosis arising from cryoinjury.


Subject(s)
Cryopreservation/methods , Embryonic Stem Cells , Apoptosis , Cell Adhesion , Cell Death , Cell Survival , Cells, Cultured , Cold Temperature , Embryonic Stem Cells/cytology , Humans , In Vitro Techniques , Kinetics
3.
Chem Rev ; 105(5): 1603-62, 2005 May.
Article in English | MEDLINE | ID: mdl-15884785
4.
Biochem Biophys Res Commun ; 322(4): 1384-91, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15336987

ABSTRACT

Most of the membrane trafficking phenomena including those involving the interactions between endosomes and lysosomes are regulated by changes in intracellular Ca2+ (Cai). These processes are disturbed in some types of mucolipidoses and other lysosomal storage disorders, such as mucolipidosis IV (MLIV), a neurological disorder that usually presents during the first year of life with blindness, cognitive impairment, and psychomotor delays. It is caused by mutations in MCOLN1, the gene encoding mucolipin-1 (MLN1), which we have recently established to represent a Ca2+-permeable cation channel that is transiently modulated by changes in Cai. The cells of MLIV patients contain enlarged lysosomes that are likely associated with abnormal sorting and trafficking of these and related organelles. We studied fibroblasts from MLIV patients and found disturbed Ca2+ signaling and large acidic organelles such as late endosomes and lysosomes (LEL) with altered cellular localization in these cells. The fusion between LEL vesicles in these cells was defective. This is a Ca2+-dependent process related to signaling pathways involved in regulation of Ca2+ homeostasis and trafficking. The MLN1 channels could play a key role in Ca2+ release from LEL vesicles, which triggers the fusion and trafficking of these organelles. The characterization of this MLN1-mediated Ca2+-dependent process should provide new insights into the pathophysiological mechanisms that lead to the development of MLIV and other mucolipidoses associated with similar disturbances in membrane trafficking.


Subject(s)
Calcium Signaling , Membrane Proteins/physiology , Mucolipidoses/metabolism , Animals , Biological Transport , Calcium/metabolism , Endosomes/metabolism , Humans , Intracellular Membranes/metabolism , Lysosomes/metabolism , Membrane Fusion , Patch-Clamp Techniques , TRPM Cation Channels , Transient Receptor Potential Channels , Xenopus laevis
5.
Bone ; 27(1): 21-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10865205

ABSTRACT

The existence in osteoblasts of the G-protein-coupled extracellular calcium (Ca(o)(2+))-sensing receptor (CaR) that was originally cloned from parathyroid and kidney remains controversial. In our recent studies, we utilized multiple detection methods to demonstrate the expression of CaR transcripts and protein in several osteoblastic cell lines, including murine MC3T3-E1 cells. Although we and others have shown that high Ca(o)(2+) and other polycationic CaR agonists modulate the function of MC3T3-E1 cells, none of these actions has been unequivocally shown to be mediated by the CaR. Previous investigations using neurons and lens epithelial cells have shown that activation of the CaR stimulates Ca(2+)-activated K(+) channels. Because osteoblastic cells express a similar type of channel, we have examined the effects of specific "calcimimetic" CaR activators on the activity of a Ca(2+)-activated K(+) channel in MC3T3-E1 cells as a way of showing that the CaR is not only expressed in those cells but is functionally active. Patch-clamp analysis in the cell-attached mode showed that raising Ca(o)(2+) from 0.75 to 2.75 mmol/L elicited about a fourfold increase in the open state probability (P(o)) of an outward K(+) channel with a conductance of approximately 92 pS. The selective calcimimetic CaR activator, NPS R-467 (0.5 micromol/L), evoked a similar activation of the channel, while its less active stereoisomer, NPSS-467 (0.5 micromol/L), did not. Thus, the CaR is not only expressed in MC3T3-E1 cells, but is also functionally coupled to the activity of a Ca(2+)-activated K(+) channel. This receptor, therefore, could transduce local or systemic changes in Ca(o)(2+) into changes in the activity of this ion channel and related physiological processes in these and perhaps other osteoblastic cells.


Subject(s)
Osteoblasts/physiology , Potassium Channels/physiology , Receptors, Cell Surface/physiology , Animals , Cell Line , Ion Channel Gating/physiology , Mice , Patch-Clamp Techniques , Receptors, Calcium-Sensing , Signal Transduction
6.
J Neurosci ; 19(20): 8876-84, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10516307

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is thought to be caused in part by the age-related accumulation of amyloid beta-protein (Abeta). The presence of neuritic plaques containing abundant Abeta-derived amyloid fibrils in AD brain tissue supports the concept that fibril accumulation per se underlies neuronal dysfunction in AD. Recent observations have begun to challenge this assumption by suggesting that earlier Abeta assemblies formed during the process of fibrillogenesis may also play a role in AD pathogenesis. Here, we present the novel finding that protofibrils (PF), metastable intermediates in amyloid fibril formation, can alter the electrical activity of neurons and cause neuronal loss. Both low molecular weight Abeta (LMW Abeta) and PF reproducibly induced toxicity in mixed brain cultures in a time- and concentration-dependent manner. No increase in fibril formation during the course of the experiments was observed by either Congo red binding or electron microscopy, suggesting that the neurotoxicity of LMW Abeta and PF cannot be explained by conversion to fibrils. Importantly, protofibrils, but not LMW Abeta, produced a rapid increase in EPSPs, action potentials, and membrane depolarizations. These data suggest that PF have inherent biological activity similar to that of mature fibrils. Our results raise the possibility that the preclinical and early clinical progression of AD is driven in part by the accumulation of specific Abeta assembly intermediates formed during the process of fibrillogenesis.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Neurons/drug effects , Neurons/physiology , Neurotoxins/pharmacology , Amyloid beta-Peptides/analysis , Animals , Cerebral Cortex/pathology , Coloring Agents , Congo Red , Culture Media/chemistry , Electrophysiology , Microscopy, Electron , Molecular Weight , Protein Conformation , Rats , Time Factors
7.
Glia ; 26(1): 64-72, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10088673

ABSTRACT

An extracellular calcium (Ca2+o)-sensing receptor (CaR) plays crucial roles in maintaining systemic calcium homeostasis. The CaR is also expressed in other cells uninvolved in systemic mineral ion homeostasis, including keratinocytes, fibroblasts, and neurons. In brain the CaR is widely distributed, being particularly abundant in neurons in subfornical organ, cingulate cortex, hippocampus, and cerebellum. It is also present in fiber tracts in rat brain, presumably in oligodendroglia and in cultured rat oligodendrocytes, suggesting that the CaR modulates the function of nonneuronal cells within brain. In this report, we show functional CaR expression in a human astrocytoma cell line (U87). Reverse transcription-polymerase chain reaction (RT-PCR) amplified a product from U87 cell RNA exhibiting >98% homology with the human CaR. Northern blot revealed a 5.5 kb transcript, similar to the principal transcript in human parathyroid, and a smaller 2.4 kb transcript. U87 cells expressed CaR protein as assessed by immunocytochemistry and Western blot using an affinity-purified, anti-CaR antiserum. Patch clamp analysis in the cell-attached mode revealed that raising Ca2+o from 0.75 to 1.75 or 2.75 mM produced approximately threefold increases in the open state probability (Po) of an outward K+ channel with a conductance of approximately 88 pS. A specific "calcimimetic" CaR activator, R-467 (0.5 microM), activated this K+ channel similarly, while its less active stereoisomer, S-467, did not. Thus U87 astrocytoma cells express both CaR mRNA and protein, and the receptor activates an outward K+ channel previously suggested to be involved in membrane polarization and cellular excitability.


Subject(s)
Astrocytoma/physiopathology , Calcium Channels/physiology , Potassium Channels/physiology , Transcription, Genetic , Animals , Blotting, Northern , Brain Neoplasms/physiopathology , Calcium/pharmacology , Calcium/physiology , Calcium Channels/genetics , Humans , Ion Channel Gating , Membrane Potentials/drug effects , Membrane Potentials/physiology , Oligodendroglia/physiology , Patch-Clamp Techniques , Potassium Channels/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stereoisomerism , Tumor Cells, Cultured
8.
Brain Res ; 851(1-2): 116-24, 1999 Dec 18.
Article in English | MEDLINE | ID: mdl-10642834

ABSTRACT

A receptor for extracellular calcium ions (Ca2+o), cloned from parathyroid gland, serves a critical function in Ca2+o homeostasis by regulating PTH release via "sensing" of its physiological agonist, Ca2+o. Its cloning from rat striatum revealed that the extracellular calcium-sensing receptor (CaR) could be involved in sensing ambient Ca2+o within the brain, where Ca2+ plays key roles in virtually all aspects of central nervous system (CNS) function. The CaR is expressed in neurons, oligodendrocytes, microglia and the human astrocytoma cell line, U87 where its functions include control of cellular proliferation and modulation of ion channels, such as outward K+ channels and nonselective cation channels (NCC). In this report, we have shown that the CaR is expressed in U373 cells as assessed by RT-PCR using CaR-specific primers followed by sequencing of the amplified products, by Northern blot analysis using a CaR-specific probe as well as by Western analysis utilizing a specific polyclonal anti-CaR antiserum. Furthermore, agents known to activate the cloned CaR induce increases in cellular proliferation and the open probability of an NCC. Thus our study strongly suggests that elevated levels of Ca2+o, acting via the CaR, activate an NCC that could contribute to the associated CaR-induced stimulation of proliferation.


Subject(s)
Astrocytoma/metabolism , Calcium/metabolism , Glioblastoma/metabolism , Ion Pumps/metabolism , Receptors, Cell Surface/physiology , Animals , Calcium/pharmacology , Cell Division/physiology , Humans , Ion Pumps/drug effects , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Middle Aged , Rats , Receptors, Calcium-Sensing , Receptors, Cell Surface/agonists , Tumor Cells, Cultured
9.
Glia ; 24(4): 449-58, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9814825

ABSTRACT

A G protein-coupled, extracellular calcium (Ca(0)2+)-sensing receptor (CaR) cloned from parathyroid, kidney, and brain plays crucial roles in systemic calcium metabolism. In brain, the CaR is located in nerve terminals as well as in fiber tracts, where it may be expressed in glia. Moreover, there is Ca2+- and K+-dependent communication between axons and oligodendroglia. To investigate further the potential role of the CaR in oligodendroglia, we studied expression of CaR mRNA and protein as well as the effects of CaR agonists on cellular proliferation and Ca2+-activated K+ channel activity in immature rat oligodendrocytes in primary culture. Reverse transcriptase polymerase chain reaction and sequencing of CaR transcripts from oligodendrocytes revealed >99% sequence identity with the rat kidney CaR. Northern analysis demonstrated 7.5 and 4.1 kb transcripts in oligodendrocytes, similar to those in rat parathyroid and kidney, while Western analysis and immunocytochemistry with CaR-specific antisera showed the presence of CaR protein. Immunocytochemically, the CaR was colocalized with galactocerebroside in the cultured oligodendrocytes. Raising Ca(0)2+ from 1.8 to 4.8 mM or addition of the polycationic CaR agonist neomycin (300 microM) modestly but significantly increased [3H]-thymidine incorporation into oligodendrocytes. Elevating Ca(0)2+ from 0.75 to 3.0 mM or addition of 100 microM neomycin also produced 2-2.5-fold increases in the open state probability (Po) of an outward K+ channel with a unitary conductance of 88+/-5 pS. Taken together, our data show that the CaR is expressed in immature oligodendrocytes and may be functionally linked to cellular proliferation and an outward K+ channel potentially contributing to local ionic homeostasis in the vicinity of oligodendroglia.


Subject(s)
Calcium/metabolism , Oligodendroglia/metabolism , Potassium Channels/metabolism , Receptors, Cell Surface/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Division , Extracellular Space/metabolism , Fluorescent Antibody Technique, Indirect , Gene Expression , Neomycin/pharmacology , Polymerase Chain Reaction/methods , Rats , Rats, Wistar , Receptors, Calcium-Sensing , Receptors, Cell Surface/genetics
10.
Am J Physiol ; 273(4): C1315-23, 1997 10.
Article in English | MEDLINE | ID: mdl-9357776

ABSTRACT

The Ca2+-sensing receptor (CaR) is activated at physiological levels of external Ca2+ (Ca(o)) but is expressed in a number of tissues that do not have well-established roles in the control of Ca(o), including several regions of the brain and the intestine. Polyamines are endogenous polyvalent cations that can act as agonists for the CaR, as shown by our current studies of human embryonic kidney (HEK-293) cells transfected with the human CaR. Cellular parameters altered by polyamines included cytosolic free Ca2+ (Ca(i)), inositol phosphate production, and the activity of a nonselective cation channel. Spermine stimulated Ca(i) transients in CaR-transfected HEK cells, with a concentration producing a half-maximal response (EC50) of approximately 500 microM in the presence of 0.5 mM Ca2+, whereas sustained increases in Ca(i) had an EC50 of approximately 200 microM. The order of potency was spermine > spermidine >> putrescine. Elevation of Ca(o) shifted the EC50 for spermine sharply to the left, with substantial stimulation below 100 microM. Addition of subthreshold concentrations of spermine increased the sensitivity of CaR-expressing HEK cells to Ca(o). Parathyroid hormone secretion from bovine parathyroid cells was inhibited by 50% in the presence of 200 microM spermine, a response similar to that elicited by 2.0 mM Ca(o). These data suggest that polyamines could be effective agonists for the CaR, and several tissues, including the brain, may use the CaR as a target for the actions of spermine and other endogenous polycationic agonists.


Subject(s)
Parathyroid Glands/physiology , Parathyroid Hormone/metabolism , Receptors, Cell Surface/physiology , Spermidine/pharmacology , Spermine/pharmacology , Animals , Calcium/pharmacology , Cattle , Cell Line , Cells, Cultured , Humans , Kidney , Membrane Potentials/drug effects , Membrane Potentials/physiology , Parathyroid Glands/cytology , Parathyroid Glands/drug effects , Patch-Clamp Techniques , Receptors, Calcium-Sensing , Receptors, Cell Surface/biosynthesis , Recombinant Proteins/metabolism , Transfection
11.
Endocrinology ; 136(5): 2238-43, 1995 May.
Article in English | MEDLINE | ID: mdl-7720673

ABSTRACT

The modulation of K+ channels by Ca2+ may have important functional implications in parathyroid cells, since in most endocrine cells they control membrane voltage regulating Ca2+ influx and hormone secretion. To characterize specific channel mechanisms regulating membrane voltage in parathyroid cells, the patch-clamp technique was used to determine the activities of K+ channels at different levels of intracellular Ca2+ concentration (Ca2+i) associated with changes in extracellular Ca2+ concentration (Ca2+o). This study shows that the membranes of dispersed bovine parathyroid cells contain a K+ channel that is activated by elevated Ca2+o through an indirect mechanism (i.e. exposure of the entire cell to high Ca2+o activates the channel despite a low Ca2+ concentration within the pipette solution on the external side of the channel under study). This K+ channel has a unitary conductance of 191 pS and is highly selective for K+, similar to the so-called maxi type of Ca(2+)-activated K+ channel previously defined in a number of other cell types. Like the latter channel, the activity of this channel in excised patches from parathyroid cells is markedly increased when an EGTA-containing buffer on the cytoplasmic face of the membrane is replaced with one containing 0.5 microM Ca2+. Changes in Ca2+ on the intracellular side of the membrane also shift the level of voltage necessary for half-maximal activation of the channel from 103 mV at 0.1 microM Ca2+ to 79 mV and 54 mV at 0.25 and 0.5 microM Ca2+, respectively. When similar studies were carried out using cell-attached patches on parathyroid cells exposed to 0.5, 1.5, or 2.0 mM Ca2+o, the values for half-maximal activation were approximately 105, 56, and 29 mV, respectively. The latter result suggests that in intact parathyroid cells, the channel is exposed to Ca2+i concentrations of about 0.15-0.2, 0.4 and 0.6-0.7 microM at these three extracellular Ca2+ concentrations, values that are in excellent agreement with those previously measured using Ca(2+)-sensitive fluorescent dyes. Thus, parathyroid cells express a maxi type of Ca(2+)-activated K+ channel that is indirectly regulated by Ca2+o, presumably through concomitant changes in Ca2+i. The latter may limit the extent of the cellular depolarization produced in response to elevated Ca2+o in this cell type.


Subject(s)
Calcium/metabolism , Parathyroid Glands/physiology , Potassium Channels/physiology , Animals , Calcium/pharmacology , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Parathyroid Glands/drug effects , Patch-Clamp Techniques , Potassium Channels/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...