• Title/Summary/Keyword: Taurine uptake

Search Result 19, Processing Time 0.02 seconds

The Change of Taurine Transport in Osteocytes by Oxidative Stress, Hypertonicity and Calcium Channel Blockers

  • Kang, Young-Sook;Kim, Soon-Joo
    • Biomolecules & Therapeutics
    • /
    • v.16 no.3
    • /
    • pp.219-225
    • /
    • 2008
  • Taurine is the most abundant amino acid in many tissues and is found to be enhancing the bone tissue formation or inhibits the bone loss. Although it is reported that taurine reduces the alveolar bone loss through inhibiting the bone resorption, its functions of taurine and expression of taurine transporter (TauT) in bone have not been identified yet. The purpose of this study is to clarify the uptake mechanism of taurine in osteoblast using mouse osteoblast cell lines. In this study, mouse stromal ST2 cells and mouse osteoblast-like MC3T3-E1 cells as osteoblast cell lines were used. The activity of taurine uptake was assessed by measuring the uptake of [$^3H$]taurine in the presence or absence of inhibitors. TauT mRNA was detected in ST2 and MC3T3-E1 cells. [$^3H$]Taurine uptake by these cells was dependent on the presence of extracellular calcium ion. The [$^3H$]taurine uptake in ST2 cells treated with 4 mM calcium was increased by 1.7-fold of the control which was a significant change. In contrast, in $Ca^{++}$-free condition and L-type calcium channel blockers (CCBs), taurine transport to osteocyte was significantly inhibited. In oxidative stress conditions, [$^3H$]taurine uptake was decreased by TNF-$\alpha$ and $H_2O_2$. Under the hyperosmotic conditions, taurine uptake was increased, but inhibited by CCBs in hyperosmotic condition. These results suggest that, in mouse osteoblast cell lines, taurine uptake by TauT was increased by the presence of extracellular calcium, whereas decreased by CCBs and oxidative stresses, such as TNF-$\alpha$ and $H_2O_2$.

Characterization of Taurine Transport in Conditionally Immortalized Rat Brain Capillary Endothelial Cell Lines (조건적 불사화 흰쥐 뇌 모세혈관 내피세포주에서 Taurine 수송 특성)

  • 강영숙;이나영
    • Biomolecules & Therapeutics
    • /
    • v.10 no.2
    • /
    • pp.99-103
    • /
    • 2002
  • Taurine has a neuroprotective action from oxidative stress in neural cell. In the present study, we studied taurine transport under basal and stressed conditions in conditionally immortalized rat brain capillary endothelial cell line (TR-BBB13) in vitro. The uptake of[$^3{H}$]taurine in the TR-BBB13 was increased by time-dependently and dependent on both Na$^{+}$ and Cl/ sup -/. Furthermore, $\beta$-alanine strongly inhibited the uptake of [TEX>$^3{H}$]taurine in the TR-BBB13. To study the effcts of oxidative stress on taurine transport, we used diethyl maleate (DEM) and lipopolysccharide (LPS). Diethyl maleate (DEM, $300\Mu\textrm{M}$) significantly reduced uptake of [TEX>$^3{H}$]taurine by time-dependently until 8 hr exposure in TR-BBB 13. But, the [TEX>$^3{H}$]taurine uptake was not changed by lipopolysccharide (LPS, 10 ng/ml) in TR-BBB13.3.

The Regulation of Taurine Transport through the Blood-Placental Barrier under Oxidative Stress

  • Kang, Young-Sook;Yoon, Yoo-Geum
    • Journal of Pharmaceutical Investigation
    • /
    • v.38 no.5
    • /
    • pp.295-302
    • /
    • 2008
  • In the present study, we examined the changes of uptake and efflux of taurine under various conditions inducing oxidative stress using rat conditionally immortalized syncytiotrophoblast cell line, TR-TBT cell, as blood-placental barrier in vitro model. In addition, we identified the characteristics of taurine transport in TR-TBT cells including general features, besides effect of calcium ion on taurine transport. Taurine uptake showed time, $Na^+$ and $Cl^-$ dependency, and was decreased by PKC activator in TR-TBT cells. Also, calcium free condition decreased taurine uptake and evoked taurine efflux in the cells. Oxidative stress induced the change of taurine transport in TR-TBT cells, but the changes were different depending on the types of stimulation inducing oxidative stress. The taurine uptake was increased by TNF-$\alpha$, LPS and DEM stimulation but decreased by $H_2O_2$ and NO stimulation. Also, the taurine efflux was regulated by TNF-$\alpha$ stimulation. In conclusion, the taurine transport through the blood-placental barrier was regulated in oxidative stress conditions, and these results demonstrated that oxidative stress affected the taurine supplies to fetus and taurine level of fetus.

The Change of Taurine Transport in Variable Stress States through the Inner Blood-Retinal Barrier using In Vitro Model

  • Kang, Young-Sook;Lee, Na-Young;Chung, Yeon-Yee
    • Biomolecules & Therapeutics
    • /
    • v.17 no.2
    • /
    • pp.175-180
    • /
    • 2009
  • Taurine is the most abundant free amino acid in the retina and transported into retina via taurine transporter (TauT) at the inner blood-retinal barrier (iBRB). In the present study, we investigated whether the taurine transport at the iBRB is regulated by oxidative stress or disease-like state in a conditionally immortalized rat retinal capillary endothelial cell line (TR-iBRB) used as an in vitro model of iBRB. First, [$^3H$]taurine uptake and efflux by TR-iBRB were regulated in the presence of extracellular $Ca^{2+}$. [$^3H$]Taurine uptake was inhibited and efflux was enhanced under $Ca^{2+}$ free condition in the cells. In addition, oxidative stress inducing agents such as tumor necrosis factor-$\alpha$ (TNF-$\alpha$), lipopolysaccharide (LPS), diethyl maleate (DEM) and glutamate increased [$^3H$]taurine uptake and decreased [$^3H$]taurine efflux in TR-iBRB cells. Whereas, 3-morpholinosydnonimine (SIN-1), which is known to NO donor decreased [$^3H$]taurine uptake. Lastly, TR-iBRB cells exposed to high glucose (25 mM) medium and the [$^3H$]taurine uptake was reduced about 20% at the condition. Also, [$^3H$]taurine uptake was decreased by cytochalasin B, which is known to glucose transport inhibitor. In conclusion, taurine transport in TR-iBRB cells is regulated diversely at extracellular $Ca^{2+}$, oxidative stress and hyperglycemic condition. It suggested that taurine would play a role as a retinal protector in diverse disease states.

Characterization of Absorption Process of Taurine Across Rat Small Intestine

  • Kim, Kyung-Soon
    • Archives of Pharmacal Research
    • /
    • v.6 no.2
    • /
    • pp.109-114
    • /
    • 1983
  • A mechanism of taurine transfer across the rat small intestine was elucidated by using the in situ recirculation perfusion or loop method. Taurine uptake was saturable, Km= 39.9 mM, and energy dependent, and required sodium. The close structural analogues, aminomethane sulfonic acid, .gamma.-amino-butyric acid, hypotaurine, and .betha.-alanine, reduced significantly taurine uptake when present in 10-fold excess. The .alpha.-amino acid, glycine, did not inhibit uptake. Hence, all of these findings lead to a conclusion that a carrier-mediated transport system for taurine exists in the small intestine.

  • PDF

Sodium Dependent Taurine Transport into the Choroid Plexus, the Blood-Cerebrospinal Fluid Barrier

  • Chung, Suk-Jae;Ramanathan, Vikram;Brett, Claire M.;Giacomini, Kathleen M.
    • Journal of Pharmaceutical Investigation
    • /
    • v.25 no.3
    • /
    • pp.7-20
    • /
    • 1995
  • Taurine, a ${\beta}-amino$ acid, plays an important role as a neuromodulator and is necessary for the normal development of the brain. Since de novo synthesis of taurine in the brain is minimal and in vivo studies suggest that taurine dose not cross the blood-brain barrier, we examined whether the choroid plexus, the blood-cerebrospinal fluid (CSF) barrier, plays a role in taurine transport in the central nervous system. The uptake of $[^3H]-taurine$ into ATP depleted choroid plexus from rabbit was substantially greater in the presence of an inwardly directed $Na^+$ gradient taurine accumulation was negligible. A transient in side-negative potential gradient enhanced the $Na^+-driven$ uptake of taurine into the tissue slices, suggesting that the transport process is electrogenic, $Na^+-driven$ taurine uptake was saturable with an estimated $V_{max}$ of $111\;{\pm}\;20.2\;nmole/g/15\;min$ and a $K_M\;of\;99.8{\pm}29.9\;{\mu}M$. The estimated coupling ratio of $Na^+$ and taurine was $1.80\;{\pm}\;0.122.$ $Na^+-dependent$ taurine uptake was significantly inhibited by ${\beta}-amino$ acids, but not by ${\alpha}-amino$ acids, indicating that the transporter is selective for ${\beta}-amino$ acids. Since it is known that the physiological concentration of taurine in the CSF is lower than that in the plasma, the active transport system we characterized may face the brush border (i.e., CSF facing) side of the choroid plexus and actively transport taurine out of the CSF. Therefore, we examined in vivo elimination of taurine from the CSF in the rat to determine whether elimination kinetics of taurine from the CSF is consistent with the in vitro study. Using a stereotaxic device, cannulaes were placed into the lateral ventricle and the cisterna magna of the rat. Radio-labelled taurine and inulin (a marker of CSF flow) were injected into the lateral ventricle, and the concentrations of the labelled compounds in the CSF were monitored for upto 3 hrs in the cisterna magna. The apparent clearance of taurine from CSF was greater than the estimated CSF flow (p<0.005) indicating that there is a clearance process in addition to the CSF flow. Taurine distribution into the choroid plexus was at least 10 fold higher than that found in other brain areas (e. g., cerebellum, olfactory bulb and cortex). When unlabelled taurine was co-administered with radio-labelled taurine, the apparent clearance of taurine was reduced (p<0.0l), suggesting a saturable disposition of taurine from CSF. Distribution of taurine into the choroid plexus, cerebellum, olfactory bulb and cortex was similarly diminished, indicating that the saturable uptake of taurine into these tissues is responsible for the non-linear disposition. A pharmacokinetic model involving first order elimination and saturable distribution described these data adequately. The Michaelis-Menten rate constant estimated from in vivo elimination study is similar to that obtained in the in vitro uptake experiment. Collectively, our results demonstrate that taurine is transported in the choroid plexus via a $Na^+-dependent,saturable$ and apparently ${\beta}-amino$ acid selective mechanism. This process may be functionally relevant to taurine homeostasis in the brain.

  • PDF

Pharmacokinetics and Blood-Brain Barrier Permeability of Taurine in Spontaneously Hypertensive Rats and Normotensive Rats (자연발생 고혈압 흰쥐와 정상흰쥐 데서 타우린의 체내동태 및 뇌투과성)

  • 강영숙;임지현;김안근
    • Biomolecules & Therapeutics
    • /
    • v.8 no.2
    • /
    • pp.194-198
    • /
    • 2000
  • Taurine, 2-aminoethanesulfonic acid is widely distributed in animal tissues and has a variety of bio-logical activities. A recent worldwide study demonstrated beneficial effects of taurine on aging and age-associated disorders. In general, taurine levels in the brain decease when an animal is subjected to pathologic conditions such as ischemia-anoxia and seizure. But the taurine levles tend to increase in the brain in hypertensive state. In the present study, the blood-brain barrier (BBB) transport of [$^3$H]taurine was compared between spontaneously hypertensive rats (SHR) and normotensive Sprague-Dawley rats (SD) using intravenous injection technique in vivo. We also obtained pharmacokinetic parameters of plasma volume maker, [$^{14}$ C] sucrose and [$^3$H]taurine after inject to rats simulatenously. BBB permeability surface area product (PS) value of [$^3$H]taurine in SHR (16$\pm$2.9$\times$10$^{-3}$ ml/min/g) was significantly higher than that in SD (7.4$\pm$0.8$\times$10$^{-3}$ ml/min/g). There is also significant difference for brain uptake of [$^3$H]taurine between SHR (0.195$\pm$0.031%ID/g) and SD (0.058$\pm$0.003% ID/g). This is due to difference of area under the plasma concentration-time curve (AUC) and that of total clearance (Class) between SHR and SD. No significant difference was indicated from other organ uptakes such as lung, heart, liver SHR and SD. But also kidney uptake was much higher in SHR. In conclusion, [$^3$H]taurine in plasma was slowly eliminated in SHR than in SD and uptake of [$^3$H]taurine in SHR is much higher than that of SD. This results suggest increased taurine level in the brain in hypertension state have an any effect on the brain uptake of taurine.

  • PDF

Ion dependent cellular uptake of taurine in mouse osteoblast cell lines

  • Naomi Ishido;Emi Nakashima;Kang, Young-Sook
    • Proceedings of the Korean Society of Applied Pharmacology
    • /
    • 2003.11a
    • /
    • pp.109-109
    • /
    • 2003
  • Taurine is present in a variety of tissue and exhibits many important physiological functions in many tissues. Although it is known that many tissues mediate taurine transport, its functions of taurine transport in bone have not been identified yet. In the present study, we investigated the expression of taurine transporter (TauT) and taurine uptake using mouse stromal ST2 cells and osteoblast-like MC3T3-El cells, which is bone related cells. Detection of TauT mRNA expression in these cells were performed by reverse transcription polymerase chain reaction (RT-PCR). The activity of TauT was assessed by measuring the uptake of [$^3$H]taurine in the presence or absence of inhibitors. TauT mRNA was detected in these cells. [$^3$H]Taurine uptake was dependent upon the presence of extracellular sodium, chloride and calcium ions, and inhibited by cold-taurine and ${\beta}$-alanine. These results suggest that taurine has biological functions in bone and some effect on the bone cells.

  • PDF

Regulation of Taurine Transporter Activity by Glucocorticoid Hormone

  • Kim, Ha-Won;Shim, Mi-Ja;Kim, Won-Bae;Kim, Byong-Kak
    • BMB Reports
    • /
    • v.28 no.6
    • /
    • pp.527-532
    • /
    • 1995
  • Human taurine transporter has 12 transmembrane domains and its molecular weight is 69.6 kDa. The long cytoplasmic carboxy and amino termini might function as regulatory attachment sites for other proteins. Six potential protein kinase C phosphorylation sites have been reported in human taurine transporter. In this report, we studied the effects of phorbol 12-myristate 13-acetate (PMA) and glucocorticoid hormone on taurine transportation in the RAW 264.7, mouse macrophage cell line. When the cells were incubated with $[^{3}H]taurine$ in the presence or absence of $Na^+$ ion for 40 min at $37^{\circ}C$, the [$[^{3}H]taurine$ uptake rate was 780-times higher in the $Na^{+}-containing$ buffer than in the $Na^{+}-deficient$ buffer, indicating that this cell line expresses taurine transporter protein on the cell surface. THP1, a human promonocyte cell line, also showed a similar property. The $[^{3}H]taurine$ uptake rate was not influenced by the inflammatory inducing cytokines such as interleukin-1, gamma-interferon or interleukin-1+gamma-interferon, but was decreased by the PMA in the RAW 264.7 cell line. This suggests that activation of protein kinase C inhibits taurine transporter activity directly or indirectly. The inhibition of $[^{3}H]taurine$ uptake by PMA was time-dependent. Maximal inhibition occurred in one hr stimulation with PMA Increasing the treatment time beyond one h reduced the $[^{3}H]taurine$ uptake inhibition due to the depletion or inactivation of protein kinase C. The cell line also showed concentration-dependent $[^{3}H]taurine$ uptake under PMA stimulation. The phorbol-ester caused 23% inhibition at the concentration of 1 ${\mu}m$ PMA. The inhibition was significant even at a concentration as low as 10 nM PMA The reduced $[^{3}H]taurine$ uptake could be recovered by treatment with glucocorticosteroid hormone. Dexamethasone led to recover of the reduced taurine uptake induced by phorbol-ester, recovering maximally after one hr. This may suggest that macrophage cells require higher taurine concentration in a stressed state, for the secretion of glucocorticoid hormone is increased by hypothalamo-pituitary-adrenocortical (HPA) axis activation in the blood stream.

  • PDF

$\beta$-Alanine Induced Down-Regulation of the Taurine Transporter Activity in the Human Colon Carcinoma Cell Line (HT-29) (인체 소장상피세포주 모델(HT-29)에서 $\beta$-알라닌이 타우린수송체 활성에 미치는 영향)

  • 박태선;윤미영;정한나;이해미
    • Journal of the Korean Society of Food Science and Nutrition
    • /
    • v.30 no.2
    • /
    • pp.314-319
    • /
    • 2001
  • In the present study, effects of $\beta$-alanine, a known taurine antagonist for its structural similarity, on the adaptive regulation and kinetic behavior of the taurine transporter were investigated in the HT-29, human colon carcinoma cell line. Pretreatment of the cell with $\beta$-alanine(10mM) for varying periods from 3 to 30 hrs significantly reduced the taurine uptake compared to the value for control cells. This decrease in the taurine transporter activity was dependent on the incubation time with $\beta$-alanine, and the maximal down-regulation of the transporter activity was observed in cells pretreated with $\beta$-alanine for 24 hrs (25% of the control value, p<0.01). The taurine transporter appears to bind exclusively with $\beta$-alanine in the HT-29 cells since the same concentration of $\alpha$-alanine added in the culture medium for 24 hrs did not influence the taurine uptake. Kinetic analyses of the taurine transporter activity was performed in the HT-29 cell line with varying taurine concentration (5~60$\mu$M) in the uptake medium. Active taurine uptake was significantly lower in $\beta$-alanine pretreated cells compared to the value for control cells in the range of taurine concentration used in the experiment (p<0.001). The cells pretreated with $\beta$-alanine showed a 50% lower maximal velocity (Vmax, 1.7$\pm$2.0 nmole.mg $protein^{-1}$.$30min^{-1}$), and a 99% higher Michaelis constant (Km, 40.3$\pm$7.6$\mu$M) than the control values (3.3$\pm$1.9 nmole.mg $protein^{-1}$.$30min^{-1}$, and 20.3$\pm$2.1$\mu$M, respectively). These results on kinetic data suggest that $\beta$-alanine induced down-regulation of the taurine transporter activity was associated with decreases in both maximal velocity and affinity of the transporter.

  • PDF