Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 42(4): 831-41, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22531910

ABSTRACT

T-cell activation and the subsequent transformation of activated T cells into T-cell blasts require profound changes in cell volume. However, the impact of cell volume regulation for T-cell immunology has not been characterized. Here we studied the role of the cell-volume regulating osmolyte transporter Taut for T-cell activation in Taut-deficient mice. T-cell mediated recall responses were severely impaired in taut(-/-) mice as shown with B16 melanoma rejection and hapten-induced contact hypersensitivity. CD4(+) and CD8(+) T cells were unequivocally located within peripheral lymph nodes of unprimed taut(-/-) mice but significantly decreased in taut(-/-) compared with taut(+/+) mice following in vivo activation. Further analysis revealed that Taut is critical for rescuing T cells from activation-induced cell death in vitro and in vivo as shown with TCR, superantigen, and antigen-specific activation. Consequently, reduction of CD4(+) and CD8(+) T cells in taut(-/-) mice upon antigen challenge resulted in impaired in vivo generation of T-cell memory. These findings disclose for the first time that volume regulation in T cells is an element in the regulation of adaptive immune responses and that the osmolyte transporter Taut is crucial for T-cell survival and T-cell mediated immune reactions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Membrane Transport Proteins/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Death/immunology , Cell Line, Tumor , Cell Size/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Immunologic Memory/drug effects , Immunologic Memory/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Superantigens/immunology , Superantigens/pharmacology
2.
Infect Immun ; 78(4): 1642-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20100858

ABSTRACT

Deletion of the taurine transporter gene (taut) results in lowered levels of taurine, the most abundant amino acid in mammals. Here, we show that taut-/- mice have lost their ability to self-heal blood-stage infections with Plasmodium chabaudi malaria. All taut-/- mice succumb to infections during crisis, while about 90% of the control taut(+/+) mice survive. The latter retain unchanged taurine levels even at peak parasitemia. Deletion of taut, however, results in the lowering of circulating taurine levels from 540 to 264 micromol/liter, and infections cause additional lowering to 192 micromol/liter. Peak parasitemia levels in taut-/- mice are approximately 60% higher than those in taut(+/+) mice, an elevation that is associated with increased systemic tumor necrosis factor alpha (TNF-alpha) and interleukin-1beta (IL-1beta) levels, as well as with liver injuries. The latter manifest as increased systemic ammonia levels, a perturbed capacity to entrap injected particles, and increased expression of genes encoding TNF-alpha, IL-1beta, IL-6, inducible nitric oxide synthase (iNOS), NF-kappaB, and vitamin D receptor (VDR). Autopsy reveals multiorgan failure as the cause of death for malaria-infected taut-/- mice. Our data indicate that taut-controlled taurine homeostasis is essential for resistance to P. chabaudi malaria. Taurine deficiency due to taut deletion, however, impairs the eryptosis of P. chabaudi-parasitized erythrocytes and expedites increases in systemic TNF-alpha, IL-1beta, and ammonia levels, presumably contributing to multiorgan failure in P. chabaudi-infected taut-/- mice.


Subject(s)
Malaria/immunology , Malaria/mortality , Membrane Glycoproteins/deficiency , Membrane Transport Proteins/deficiency , Plasmodium chabaudi/immunology , Plasmodium chabaudi/pathogenicity , Sequence Deletion , Ammonia/blood , Animals , Female , Interleukin-1beta/blood , Liver/pathology , Malaria/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Organ Failure , Parasitemia , Survival Analysis , Taurine/blood , Tumor Necrosis Factor-alpha/blood
3.
Exp Dermatol ; 17(12): 1031-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18557924

ABSTRACT

Compatible organic osmolytes, such as betaine, myo-inositol and taurine, are involved in cell protection. Human dermal fibroblasts accumulate these osmolytes and express mRNA specific for their transporting systems betaine-/gamma-amino-n-butyric acid (GABA) transporter (BGT-1), sodium-dependent myo-inositol transporter (SMIT) and taurine transporter (TAUT). Taurine uptake was about sixfold higher than that of betaine and myo-inositol. Compared with normoosmotic (305 mOsm/l) control, hyperosmotic exposure (405 mOsm/l) led to a twofold induction of osmolyte uptake. Ultraviolet A (UVA) upregulated osmolyte transporter mRNA levels and increased osmolyte uptake. Taurine inhibited UVA-induced interleukin-6 (Il-6) mRNA expression by 40%. Furthermore, Il-6 accumulation in the supernatants of UVA-irradiated dermal fibroblasts was much slower when cells were preincubated with taurine. These data indicate that taurine accumulation seems to be part of the fibroblast response to UVA radiation and may protect against UVA-induced Il-6 overexpression.


Subject(s)
Fibroblasts/metabolism , Organic Chemicals/metabolism , Ultraviolet Rays , Betaine/pharmacokinetics , Betaine/pharmacology , Biological Transport/drug effects , Biological Transport/radiation effects , Carrier Proteins/genetics , Cells, Cultured , Dermis/drug effects , Dermis/metabolism , Dermis/radiation effects , Electrolytes/metabolism , Fibroblasts/drug effects , Fibroblasts/radiation effects , GABA Plasma Membrane Transport Proteins , Gene Expression/drug effects , Gene Expression/radiation effects , Heat-Shock Proteins/genetics , Humans , Inositol/pharmacokinetics , Inositol/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Osmolar Concentration , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Saline Solution, Hypertonic/pharmacology , Symporters/genetics , Taurine/pharmacokinetics , Taurine/pharmacology
4.
Biol Chem ; 388(12): 1345-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18020950

ABSTRACT

We have previously shown that compatible organic osmolytes, such as betaine, myo-inositol and taurine, are part of the stress response of normal human keratinocytes (NHKs) to ultraviolet B (UVB) radiation. In this regard, we tested human HaCaT keratinocytes as a surrogate cell line for NHK. HaCaT cells osmo-dependently express mRNA specific for transport proteins for betaine (BGT-1), myo-inositol (SMIT) and taurine (TAUT). Compared to normoosmotic (305 mosmol/l) controls, which strongly constitutively expressed BGT-1 mRNA, strong induction of SMIT and TAUT mRNA as well as low induction of BGT-1 mRNA expression was observed between 3 and 9 h after hyperosmotic exposure (405 mosmol/l). This expression correlated with an increased osmolyte uptake. Conversely, hypoosmotic (205 mosmol/l) stimulation led to a significant efflux of osmolytes. Exposure to UVB (290-315 nm) radiation induced cell shrinkage which was followed by an upregulation of osmolyte transporter mRNA levels and osmolyte uptake. These results demonstrate that human HaCaT keratinocytes possess an osmolyte strategy including UVB-induced cell shrinkage and following increased osmolyte uptake. However, several differences in osmolyte transporter expression and uptake were noted between NHK and HaCaT cells, indicating that the use of HaCaT cells as a surrogate cell line for NHK has limitations.


Subject(s)
Carrier Proteins/biosynthesis , Cell Size/radiation effects , Keratinocytes/radiation effects , RNA, Messenger/biosynthesis , Betaine/metabolism , Blotting, Northern , Carrier Proteins/genetics , Cell Line , GABA Plasma Membrane Transport Proteins , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Humans , Inositol/metabolism , Intracellular Fluid/metabolism , Keratinocytes/metabolism , Keratinocytes/ultrastructure , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Osmolar Concentration , RNA, Messenger/genetics , Symporters/biosynthesis , Symporters/genetics , Taurine/metabolism , Ultraviolet Rays , Water/metabolism
5.
Am J Physiol Gastrointest Liver Physiol ; 293(6): G1134-46, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17916651

ABSTRACT

Inverse acinar regulation of Mrp2 and 3 represents an adaptive response to hepatocellular cholestatic injury. We studied whether obstructive cholestasis (bile duct ligation) and LPS treatment affect the zonal expression of Bsep (Abcb11), Mrp4 (Abcc4), Ntcp (Slc10a1), and Oatp isoforms (Slco1a1, Slco1a4, and slco1b2) in rat liver, as analyzed by semiquantitative immunofluorescence. Contribution of TNF-alpha and IL-1beta to transporter zonation in obstructive cholestasis was studied by cytokine inactivation. In normal liver Bsep, Mrp4, Ntcp, and Oatp1a1 were homogeneously distributed in the acinus, whereas Oatp1a4 and Oatp1b2 expression increased from zone 1 to 3. Glutamine synthetase-positive pericentral hepatocytes exhibited markedly lower Oatp1a4 expression than the remaining zone 3 hepatocytes. In cholestatic liver Bsep and Ntcp immunofluorescence in periportal hepatocytes significantly decreased to 66 +/- 4% (P < 0.01) and 67 +/- 7% (P < 0.05), whereas it was not altered in pericentral hepatocytes. Oatp1a4 was significantly induced in hepatocytes with a primarily low expression, i.e., in periportal hepatocytes and in glutamine synthetase-positive pericentral hepatocytes. Likewise, Oatp1b2 was upregulated in periportal hepatocytes. Mrp4 zonal induction was homogeneous. Inactivation of TNF-alpha and IL-1beta prevented periportal downregulation of Bsep. Recruitment of neutrophils and polymorphonuclear cells mainly occurred in the periportal zone. Likewise, IL-1beta induction was largely found periportally. No significant transporter zonation was seen following LPS treatment. In conclusion, zonal downregulation of Bsep in obstructive cholestasis is associated with portal inflammation and is mediated by TNF-alpha and IL-1beta. Periportal downregulation of Ntcp and induction of Oatp1a4 and Oatp1b2 may represent adaptive mechanisms to reduce cholestatic injury in hepatocytes with profound downregulation of Bsep and Mrp2.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bile Ducts/metabolism , Cholestasis, Extrahepatic/metabolism , Interleukin-1/metabolism , Liver/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/metabolism , Symporters/metabolism , Tumor Necrosis Factor-alpha/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Animals , Down-Regulation , Male , Rats , Rats, Sprague-Dawley , Solute Carrier Organic Anion Transporter Family Member 1B3 , Tissue Distribution
6.
Methods Enzymol ; 428: 439-58, 2007.
Article in English | MEDLINE | ID: mdl-17875433

ABSTRACT

This chapter reports present knowledge on the properties of mice with disrupted gene coding for the taurine transporter (taut-/- mice). Study of those mice unraveled some of the roles of taurine and its membrane transport for the development and maintenance of normal organ functions and morphology. When compared with wild-type controls, taut-/- mice have decreased taurine levels in skeletal and heart muscle by about 98%, in brain, kidney, plasma, and retina by 80 to 90%, and in liver by about 70%. taut-/- mice exhibit a lower body mass as well as a strongly reduced exercise capacity compared with taut+/- and wild-type mice. Furthermore, taut-/- mice show a variety of pathological features, for example, subtle derangement of renal osmoregulation, changes in neuroreceptor expression, and loss of long-term potentiation in the striatum, and they develop clinically relevant age-dependent disorders, for example, visual, auditory, and olfactory dysfunctions, unspecific hepatitis, and liver fibrosis. Taurine-deficient animal models such as acutely dietary-manipulated foxes and cats, pharmacologically induced taurine-deficient rats, and taurine transporter knockout mouse are powerful tools allowing identification of the mechanisms and complexities of diseases mediated by impaired taurine transport and taurine depletion (Chapman et al., 1993; Heller-Stilb et al., 2002; Huxtable, 1992; Lake, 1993; Moise et al., 1991; Novotny et al., 1991; Pion et al., 1987; Timbrell et al., 1995; Warskulat et al., 2004, 2006b). Taurine, which is the most abundant amino acid in many tissues, is normally found in intracellular concentrations of 10 to 70 mmol/kg in mammalian heart, brain, skeletal muscle, liver, and retina (Chapman et al., 1993; Green et al., 1991; Huxable, 1992; Timbrell et al., 1995). These high taurine levels are maintained by an ubiquitous expression of Na(+)-dependent taurine transporter (TAUT) in the plasma membrane (Burg, 1995; Kwon and Handler, 1995; Lang et al., 1998; Liu et al., 1992; Ramamoorthy et al., 1994; Schloss et al., 1994; Smith et al., 1992; Uchida et al., 1992; Vinnakota et al., 1997; Yancey et al., 1975). Taurine is not incorporated into proteins. It is involved in cell volume regulation, neuromodulation, antioxidant defense, protein stabilization, stress responses, and via formation of taurine-chloramine in immunomodulation (Chapman et al., 1993; Green et al., 1991; Huxtable, 1992; Timbrell et al., 1995). On the basis of its functions, taurine may protect cells against various types of injury (Chapman et al., 1993; Green et al., 1991; Huxtable, 1992; Kurz et al., 1998; Park et al., 1995; Stapleton et al., 1998; Timbrell et al., 1995; Welch and Brown, 1996; Wettstein and Häussinger, 1997). In order to examine the multiple taurine functions, murine models have several intrinsic advantages for in vivo research compared to other animal models, including lower cost, maintenance, and rapid reproduction rate. Further, experimental reagents for cellular and molecular studies are widely available for the mouse. In particular, mice can be easily genetically manipulated by making transgene and knockout mice. This chapter focuses on the phenotype of the TAUT-deficient murine model (taut-/-; Heller-Stilb et al., 2002), which may help researchers elucidate the diverse roles of taurine in development and maintenance of normal organ functions and morphology.


Subject(s)
Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Taurine/physiology , Animals , Brain/physiopathology , Ear, Inner/physiopathology , Female , Kidney/physiopathology , Liver/pathology , Liver Diseases/etiology , Male , Membrane Glycoproteins/deficiency , Membrane Transport Proteins/deficiency , Mice , Mice, Knockout , Mice, Transgenic , Motor Activity , Olfactory Bulb/physiopathology , Phenotype , Physical Conditioning, Animal , Retina/physiopathology , Retinal Degeneration/etiology , Synaptic Transmission/physiology
7.
J Immunol ; 179(6): 3604-12, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785795

ABSTRACT

Organic osmolytes, such as taurine, are involved in cell volume homeostasis and cell protection. Epidermal keratinocytes possess an osmolyte strategy, i.e., they take up taurine upon hyperosmotic stress and express the corresponding transporter TAUT. UVB irradiation also triggers taurine uptake and TAUT expression in this cell type. We therefore asked whether taurine plays a role in photoprotection. By using a TAUT-deficient mouse model, lack of taurine in the skin was found to cause a significantly higher sensitivity to UVB-induced immunosuppression. This was not due to an increased generation or decreased repair of UVB-induced DNA photoproducts in the skin of these animals. Instead, decreased skin taurine levels were associated with an increased formation of the soluble immunosuppressive molecule platelet-activating factor (PAF) from the membranes of UVB-irradiated epidermal cells. Blocking PAF activity in taut-deficient mice with a PAF receptor antagonist abrogated their increased sensitivity to UVB-induced immunosuppression. Moreover, taut -/- mice were more sensitive to PAF-mediated immunosuppression than taut +/+ mice. These data suggest that taurine uptake by epidermal cells prevents undue PAF formation, and thereby photoimmunosuppression. Thus, similar to nucleotide excision repair, taurine uptake is critically involved in photoprotection of the skin.


Subject(s)
Immunosuppression Therapy , Membrane Glycoproteins/physiology , Membrane Transport Proteins/physiology , Skin/immunology , Skin/radiation effects , Taurine/physiology , Ultraviolet Rays/adverse effects , Animals , Cells, Cultured , DNA Repair/radiation effects , Female , Genetic Predisposition to Disease , Interleukin-10/antagonists & inhibitors , Interleukin-10/biosynthesis , Interleukin-10/radiation effects , Langerhans Cells/radiation effects , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmotic Pressure/radiation effects , Platelet Activating Factor/metabolism , Platelet Activating Factor/radiation effects , Platelet Membrane Glycoproteins/antagonists & inhibitors , Pyrimidine Dimers/biosynthesis , Pyrimidine Dimers/radiation effects , Receptors, G-Protein-Coupled/antagonists & inhibitors , Skin/cytology , Skin/metabolism , Taurine/deficiency , Taurine/metabolism
8.
Arch Biochem Biophys ; 462(2): 202-9, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17459327

ABSTRACT

Apoptosis is characterized by cell shrinkage, nuclear condensation, DNA-fragmentation and apoptotic body formation. Compatible organic osmolytes, e.g. taurine, modulate the cellular response to anisotonicity and may protect from apoptosis. Taurine transporter knockout mice (taut-/- mice) show strongly decreased taurine levels in a variety of tissues. They develop clinically important age-dependent diseases and some of them are characterized by apoptosis. Increased photoreceptor apoptosis leads to blindness of taut-/- mice at an early age. The taurine transporter may not be essential for the differentiation of photoreceptor cells, but many mature cells do not survive without an intact taurine transporter. The olfactory epithelium of taut-/- mice also exhibits structural and functional abnormalities. When compared with wild-types, taut-/- mice have a significantly higher proliferative activity of immature olfactory receptor neurons and an increased number of apoptotic cells. This is accompanied by electrophysiological findings indicating a reduced olfactory sensitivity. Furthermore, taut-/- and taut+/- mice develop moderate unspecific hepatitis and liver fibrosis beyond 1 year of age where hepatocyte apoptosis and activation of the CD95 system are pronounced.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Liver Diseases/metabolism , Membrane Glycoproteins/deficiency , Membrane Transport Proteins/deficiency , Muscular Diseases/metabolism , Taurine/metabolism , fas Receptor/metabolism , Animals , Liver Diseases/pathology , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Muscular Diseases/pathology
9.
Biol Chem ; 387(10-11): 1449-54, 2006.
Article in English | MEDLINE | ID: mdl-17081118

ABSTRACT

Taurine is the most abundant free amino acid in heart muscle and protects against heart failure. In the present study, the consequences of hereditary taurine deficiency on cardiac gene expression were examined in 2- and 15-16-month-old taurine transporter knockout (taut(-/-)) mice using a mouse-specific DNA microarray. This oligonucleotide-based microarray contains probes for 251 genes with relevance for heart function. Of these, 163 probes exhibited a reproducible hybridization signal and were analyzed. alpha-Actin type 1 mRNA levels were 70% lower in the heart of young and older taut(-/-) mice compared to wild-type controls. Interestingly, the hearts of taut(-/-) mice showed a switch from alpha-actin 1 to alpha-actin 2 expression, as confirmed by real-time PCR and Western blot analysis. In addition, mRNA levels of biomarkers for pressure overload and hypertension were upregulated in taut(-/-) hearts, i.e., atrial natriuretic factor (+848%), brain natriuretic peptide (+90%), cardiac ankyrin repeat protein (+118%), and procollagen 1a1, 1a2 and 3a1 (+40% at least). These results point to a stress situation in the heart of taut(-/-) mice under laboratory conditions, and it can be speculated that taut(-/-) hearts may be even more susceptible to failure in the wild when under exogenous stress.


Subject(s)
Actins/metabolism , Cardiomegaly/metabolism , Myocardium/metabolism , Taurine/metabolism , Up-Regulation , Animals , Biomarkers , Blood Pressure , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Isomerism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Taurine/deficiency , Taurine/genetics
10.
FASEB J ; 20(3): 574-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16421246

ABSTRACT

Taurine is an abundant organic osmolyte with antioxidant and immunomodulatory properties. Its role in the pathogenesis of chronic liver disease is unknown. The liver phenotype was studied in taurine transporter knockout (taut-/-) mice. Hepatic taurine levels were ~21, 15 and 6 mumol/g liver wet weight in adult wild-type, heterozygous (taut+/-) and homozygous (taut-/-) mice, respectively. Immunoelectronmicroscopy revealed an almost complete depletion of taurine in Kupffer and sinusoidal endothelial cells, but not in parenchymal cells of (taut-/-) mice. Compared with wild-type mice, (taut-/-) and (taut+/-) mice developed moderate unspecific hepatitis and liver fibrosis with increased frequency of neoplastic lesions beyond 1 year of age. Liver disease in (taut-/-) mice was characterized by hepatocyte apoptosis, activation of the CD95 system, elevated plasma TNF-alpha levels, hepatic stellate cell and oval cell proliferation, and severe mitochondrial abnormalities in liver parenchymal cells. Mitochondrial dysfunction was suggested by a significantly lower respiratory control ratio in isolated mitochondria from (taut-/-) mice. Taut knockout had no effect on taurine-conjugated bile acids in bile; however, the relative amount of cholate-conjugates acid was decreased at the expense of 7-keto-cholate-conjugates. In conclusion, taurine deficiency due to defective taurine transport triggers chronic liver disease, which may involve mitochondrial dysfunction.


Subject(s)
Hepatitis/etiology , Liver Cirrhosis, Experimental/etiology , Liver Neoplasms, Experimental/etiology , Membrane Glycoproteins/deficiency , Membrane Transport Proteins/deficiency , Taurine/physiology , Animals , Apoptosis , Bile Acids and Salts/metabolism , Cytokines/biosynthesis , Endothelial Cells/chemistry , Endothelial Cells/pathology , Female , Genotype , Hepatitis/genetics , Hepatitis/pathology , Hepatocytes/chemistry , Hepatocytes/pathology , Kupffer Cells/chemistry , Kupffer Cells/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/physiology , Mice , Mice, Knockout , Mitochondria, Liver/ultrastructure , Oxidative Stress , Phagocytosis , Tumor Necrosis Factor-alpha/analysis , fas Receptor/metabolism
11.
Pflugers Arch ; 451(5): 668-77, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16249932

ABSTRACT

Cellular taurine uptake or release counteracts alterations of cell volume. Na+-coupled taurine transporter TAUT mediates concentrative cellular uptake of taurine. Inhibition of vasopressin secretion by hypotonicity may involve taurine release from glial cells of supraoptic nucleus. We compared renal function of mice lacking TAUT (taut-/-) and wild-type littermates (taut+/+). We observed renal taurine loss and subsequent hypotaurinemia in taut-/- mice. With free access to water, plasma and urine osmolality, urinary flow rate as well as urinary excretion and plasma concentrations of Na+ and K+ were similar in taut-/- and taut+/+ mice, whereas plasma concentrations of urea were enhanced in taut-/- mice. An oral water load (1 ml/16 g body weight) induced a similar diuresis in both genotypes. Repeating the oral water load immediately after normalization of urine flow rate, however, resulted in delayed diuresis and higher urinary vasopressin/creatinine ratios in taut-/- mice. In comparison, the repeated diuretic response to vasopressin V2 receptor blockade was not different between genotypes. Water deprivation for 36 h led to similar antidiuresis and increases of urinary osmolality in both genotypes. Upon free access to water after deprivation, taut-/- mice continued to concentrate urine up to 6 days, while taut+/+ mice rapidly returned to normal urinary osmolality. Urinary vasopressin/creatinine ratios and plasma aldosterone concentrations were not different under basal conditions but were significantly higher in taut-/- mice than in taut+/+ mice at 6 days after water deprivation. In conclusion, taut-/- mice suffer from renal taurine loss and impaired ability to lower urine osmolality and to increase urinary water excretion. The latter defect could reside extrarenally and result from a role of taurine in the suppression of vasopressin release which may be attenuated in taut-/- mice.


Subject(s)
Kidney/metabolism , Membrane Glycoproteins/physiology , Membrane Transport Proteins/physiology , Water-Electrolyte Balance/physiology , Water/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists , Female , Fluid Therapy , Kidney Function Tests , Male , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Water Deprivation/physiology
12.
Anat Embryol (Berl) ; 210(5-6): 363-72, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16222546

ABSTRACT

The aim of this study was to determine whether the knockout of the taurine-transporter gene in the mouse affects the densities of GABA(A), kainate, AMPA and NMDA receptors in the brain. The caudate-putamen, the hippocampus and its subregions, and the cerebellum of six homozygous taurine-transporter gene knockout mice and six wild-type (WT) animals were examined by means of quantitative receptor autoradiography. Saturation studies were carried out for all four receptor types in order to find possible intergroup differences in Bmax and K(D) values. Taurine-transporter gene knockout animals showed significantly higher GABA(A) receptor densities in the molecular layer of the hippocampal dentate gyrus and in the cerebellum than did WT animals. The densities of kainate receptors were significantly higher in the caudate-putamen, the CA1 and hilus regions of the hippocampus and in the cerebellum of knockout animals. The caudate-putamen and cerebellum of these mice also contained significantly higher AMPA receptor densities. However, there were no significant differences between knockout and WT animals concerning the densities of NMDA receptors. Reduced brain taurine levels are associated with increased GABA(A), kainate and AMPA receptor densities in some of the regions we examined.


Subject(s)
Brain/metabolism , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Receptors, AMPA/metabolism , Receptors, GABA-A/metabolism , Receptors, Kainic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Autoradiography , Brain Chemistry , Brain Mapping , Cerebellum/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Membrane Glycoproteins/physiology , Membrane Transport Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Protein Binding , Putamen/metabolism , Taurine/metabolism
13.
Hepatology ; 41(5): 1160-72, 2005 May.
Article in English | MEDLINE | ID: mdl-15841457

ABSTRACT

Mutations of the bile salt export pump (BSEP) or the multidrug resistance P-glycoprotein 3 (MDR3) are linked to impaired bile salt homeostasis and lead to progressive familial intrahepatic cholestasis (PFIC)-2 and -3, respectively. The regulation of bile salt transporters in PFIC is not known. Expression of hepatobiliary transporters in livers of ten patients with a PFIC phenotype was studied by quantitative reverse transcription polymerase chain reaction, Western blotting, and immunofluorescence microscopy. PFIC was diagnosed by clinical and laboratory findings. All patients could be assigned to PFIC-2 or PFIC-3 by the use of BSEP- and MDR3-specific antibodies and by MDR3 gene-sequencing. Whereas in all PFIC-2 patients, BSEP immunoreactivity was absent from the canalicular membrane, in three PFIC-3 livers, canalicular MDR3 immunoreactivity was detectable. Serum bile salts were elevated to 276 +/- 233 and to 221 +/- 109 micromol/L in PFIC-2 and PFIC-3, respectively. Organic anion transporting polypeptide OATP1B1, OATP1B3, and MRP2 mRNA and protein levels were reduced, whereas sodium taurocholate cotransporting polypeptide (NTCP) was only reduced at the protein level, suggesting a posttranscriptional NTCP regulation. Whereas MRP3 mRNA and protein were not significantly altered, MRP4 messenger RNA and protein were significantly increased in PFIC. In conclusion, PFIC-2 may be reliably diagnosed by immunofluorescence, whereas the diagnosis of PFIC-3 requires gene-sequencing. Several mechanisms may contribute to elevated plasma bile salts in PFIC: reduced bile salt uptake via NTCP, OATP1B1, and OATP1B3, decreased BSEP-dependent secretion into bile, and increased transport back into plasma by MRP4. Upregulation of MRP4, but not of MRP3, might represent an important escape mechanism for bile salt extrusion in PFIC.


Subject(s)
Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/physiopathology , Liver/physiology , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adult , Aged , Bile Acids and Salts/metabolism , Female , Gene Expression/physiology , Humans , Liver-Specific Organic Anion Transporter 1/genetics , Liver-Specific Organic Anion Transporter 1/metabolism , Male , Membrane Transport Proteins/metabolism , Middle Aged , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , RNA, Messenger/analysis , Solute Carrier Organic Anion Transporter Family Member 1B3 , Symporters , gamma-Glutamyltransferase/metabolism
14.
Gastroenterology ; 128(2): 433-48, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15685554

ABSTRACT

BACKGROUND AND AIMS: Gap junctional communication was studied in quiescent and activated hepatic stellate cells. METHODS: Connexin expression and intercellular dye transfer were studied in rat hepatic stellate cells in culture and in vivo. RESULTS: Protein expression of connexin 43 was up-regulated in activated hepatic stellate cells in vivo and in vitro and was mainly localized on the cell surface, whereas connexin 26 was found intracellularly. In contrast to hepatocytes, hepatic stellate cells do not express connexin 32. Confluent hepatic stellate cells in culture communicate via gap junctions, resulting in lucifer yellow transfer and propagation of intracellular calcium signals. Phorbol ester induces a protein kinase C-dependent hyperphosphorylation and degradation of connexin 43 and inhibits intercellular communication on a short-term time scale. At the long-term level, vitamin D(3) , lipopolysaccharide, thyroid hormone T(3), dexamethasone, platelet-derived growth factor, endothelin 1, and interleukin 1beta up-regulate connexin 43 protein and messenger RNA expression and enhance intercellular communication. Slight down-regulation of connexin 43 is observed in response to vitamin A. Connexin 43 induction by endothelin 1 is inhibited by both endothelin A and endothelin B receptor antagonists. In coculture systems, hepatic stellate cells communicate with each other, which is suggestive of a syncytial organization, but no communication was found between hepatic stellate cells and other liver cell types. As shown by immunohistochemistry and electron microscopy, gap junctions are formed between activated hepatic stellate cells in vivo. CONCLUSIONS: Gap junctional communication occurs between hepatic stellate cells, is enhanced after activation, and underlies complex regulation by cytokines, hormones, and vitamins.


Subject(s)
Cell Communication/physiology , Connexin 43/genetics , Connexins/genetics , Gap Junctions/physiology , Liver/physiology , Animals , Carbon Tetrachloride , Carbon Tetrachloride Poisoning/pathology , Cells, Cultured , Gene Expression Regulation , Liver/cytology , Liver/pathology , Male , Rats , Rats, Wistar , Gap Junction beta-1 Protein
15.
J Invest Dermatol ; 123(3): 516-21, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15304091

ABSTRACT

Compatible organic osmolytes, such as betaine, myoinositol, and taurine, are involved in cell volume homeostasis as well as in cell protection, for example, against oxidative stress. This so-called osmolyte strategy requires the expression of specific osmolyte transporting systems such as the betaine/gamma-amino-n-butyric acid (GABA) transporter, the sodium-dependent myoinositol transporter and the taurine transporter (TAUT). In contrast to liver, kidney, and neural cells, nothing is known about osmolytes in the skin. Here we report that primary normal human keratinocytes (NHK) express mRNA specific for the betaine/GABA transporter, for the sodium-dependent myoinositol transporter and for the TAUT. In comparison to normoosmotic (305 mosmol per L) controls, a 3-5-fold induction of mRNA expression for the betaine/GABA-, the sodium-dependent myoinositol- and the TAUT was observed within 6-24 h after hyperosmotic exposure (405 mosmol per L). Expression of osmolyte transporters was associated with an increased uptake of radiolabeled osmolytes. Conversely, hypoosmotic (205 mosmol per L) stimulation induced significant efflux of these osmolytes. Exposure to ultraviolet B (290-315 nm) or ultraviolet A (340-400 nm) radiation, which are major sources of oxidative stress in skin, significantly stimulated osmolyte uptake. Increased osmolyte uptake was associated with upregulation of mRNA steady-state levels for osmolyte transporters in irradiated cells. These studies demonstrate that NHK possess an osmolyte strategy, which is important for their capacity to maintain cell volume homeostasis and seems to be part of their response to UV radiation.


Subject(s)
Keratinocytes/physiology , Membrane Transport Proteins , Water-Electrolyte Balance/physiology , Betaine/pharmacokinetics , Carrier Proteins/genetics , Cells, Cultured , GABA Plasma Membrane Transport Proteins , Gastrointestinal Agents/pharmacokinetics , Heat-Shock Proteins/genetics , Homeostasis/physiology , Humans , Inositol/pharmacokinetics , Keratinocytes/cytology , Keratinocytes/radiation effects , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , RNA, Messenger/analysis , Symporters/genetics , Taurine/pharmacokinetics , Ultraviolet Rays
16.
Biol Chem ; 385(3-4): 331-9, 2004.
Article in English | MEDLINE | ID: mdl-15134348

ABSTRACT

Lipopolysaccharide (LPS) induces hepatocellular down-regulation and endocytic retrieval of multidrug resistance protein 2 (Mrp2, Abcc2). Basolateral Mrp isoforms may compensate for the intracellular metabolic changes in cholestasis. Therefore, the effect of LPS on the zonal localization of Mrp2 and Mrp3 and the expression of Mrp3, Mrp4, Mrp5, and Mrp6 mRNA were investigated in rat liver. In normal rat liver Mrp3 was found in pericentral hepatocytes also expressing glutamine synthetase. In LPS-treated rat liver the decrease in Mrp2 protein was most pronounced in pericentral hepatocytes, with only minor down-regulation in periportal hepatocytes. Conversely, induction of Mrp3 was found in pericentral hepatocytes with a low expression of Mrp2. Furthermore, we found a strong induction of Mrp5 mRNA. Likewise, Mrp6 mRNA was up-regulated, however Mrp6 protein expression was not significantly altered. It is concluded that Mrp3 is inversely regulated to Mrp2 in a zonal pattern and may compensate for the LPS-induced loss of Mrp2 in the perivenous area. Induction of pericentral Mrp3 and up-regulation of Mrp5 mRNA may play an important role in the hepatocellular clearance of cholephilic substances and cyclic nucleotides accumulating after LPS treatment.


Subject(s)
Lipopolysaccharides/pharmacology , Liver/metabolism , Multidrug Resistance-Associated Proteins/biosynthesis , Animals , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Liver/drug effects , Male , Multidrug Resistance-Associated Proteins/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar
17.
Urology ; 63(2): 392-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14972509

ABSTRACT

OBJECTIVES: The Fas-Fas ligand system is an important regulator of apoptosis and is involved in tumor development. Invasive cancers downregulate Fas expression to evade antitumor immune responses. Fas is a transcriptional target of p53, which is often mutated in bladder cancers. Therefore, Fas expression and its relation to p53 mutation was investigated. METHODS: Expression of Fas protein and p53 status was studied by immunohistochemistry in 83 bladder cancer specimens. In addition, mRNA levels for soluble (decoy) and membrane-bound forms of Fas were compared between 10 bladder cancer cell lines and primary uroepithelial cells by quantitative TaqMan polymerase chain reaction. Mutational analysis of the death domain of the Fas gene was performed in all cell lines. RESULTS: Organ-confined tumors maintained specific Fas staining at the cell membrane and often also in the cytoplasm. In higher stage carcinomas, Fas expression became restricted to a smaller fraction of cells or was lacking entirely. The correlation of Fas staining with tumor stage was highly significant but no correlation to tumor grade or survival was found. Furthermore, no statistically significant relationship was observed with either the presence or lack of mutated p53 accumulation. Membrane-bound Fas mRNA was decreased in most, and soluble Fas was increased in all transitional cell carcinoma lines compared with primary uroepithelial cells. No mutations in the death domain were detected. CONCLUSIONS: Fas downregulation occurring in advanced bladder cancer is unrelated to p53 mutations. The results of immunohistochemistry and mRNA studies of soluble and membrane-bound Fas in transitional cell carcinoma lines support the hypothesis of immune evasion in advanced bladder cancer.


Subject(s)
Carcinoma, Transitional Cell/genetics , Gene Expression Regulation, Neoplastic , Genes, p53 , Neoplasm Proteins/biosynthesis , Tumor Escape/genetics , Urinary Bladder Neoplasms/genetics , fas Receptor/biosynthesis , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor/metabolism , Cell Membrane/metabolism , Down-Regulation , Humans , Life Tables , Neoplasm Proteins/genetics , Neoplasm Staging , Polymorphism, Single-Stranded Conformational , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Solubility , Survival Analysis , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , fas Receptor/genetics
18.
FASEB J ; 18(3): 577-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14734644

ABSTRACT

Taurine is the most abundant free amino acid in heart and skeletal muscle. In the present study, the effects of hereditary taurine deficiency on muscle function were examined in taurine transporter knockout (taut-/-) mice. These mice show an almost complete depletion of heart and skeletal muscle taurine levels. Treadmill experiments demonstrated that total exercise capacity of taut-/- mice was reduced by >80% compared with wild-type controls. The decreased performance of taut-/- mice correlated with increased lactate levels in serum during exercise. Surprisingly, cardiac function of taut-/- mice as assessed by magnetic resonance imaging, echocardiography, and isolated heart studies showed a largely normal phenotype under both control and stimulated conditions. However, analysis of taut-/- skeletal muscle revealed electromyographic abnormalities. (1)H nuclear magnetic resonance spectroscopy of tissue extracts showed that in the heart of taut-/- mice the lack of taurine was compensated by the up-regulation of various organic solutes. In contrast, a deficit of >10 mM in total organic osmolyte concentration was found in skeletal muscle. The present study identifies taurine transport as a crucial factor for the maintenance of skeletal muscle function and total exercise capacity, while cardiac muscle apparently can compensate for the loss of taurine.


Subject(s)
Carrier Proteins/physiology , Exercise Tolerance/physiology , Membrane Transport Proteins , Muscle, Skeletal/metabolism , Myocardium/metabolism , Taurine/physiology , Animals , Carbon Dioxide/metabolism , Cardiac Pacing, Artificial , Carrier Proteins/genetics , Cations/metabolism , Dobutamine , Echocardiography , Electromyography , Energy Metabolism , Female , Lactates/blood , Male , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/pathology , Neural Conduction , Nuclear Magnetic Resonance, Biomolecular , Organ Specificity , Oxygen Consumption , Phenotype , Ventricular Function, Left
19.
Cell Physiol Biochem ; 13(6): 337-46, 2003.
Article in English | MEDLINE | ID: mdl-14631140

ABSTRACT

In nucleated cells cellular taurine is released prior to DNA fragmentation and the breakdown of phosphatidylserine asymmetry within the plasma membrane. Similar to what is seen in nucleated cells, phosphatidylserine asymmetry is also abolished in erythrocytes exposed to osmotic shock or oxidative stress. The present study has been performed to explore the sensitivity of erythrocytes from a taurine transporter knockout mouse (taut-/-) against osmotic shock and oxidative stress. Erythrocyte cell volume was estimated from forward scatter and breakdown of phosphatidylserine asymmetry was identified by determination of annexin binding using FACS analysis. Erythrocytes from taut-/- mice were compared to erythrocytes from wild type littermates (taut+/+). Plasma concentration and erythrocyte content of taurine was significantly lower in taut-/- than in taut+/+ mice, but the intraerythrocyte taurine concentration did not exceed the plasma concentration. Hyperosmotic shock (exposure to 700 mOsm) and oxidative stress (exposure to 0.1 mM tert-butyl-hydroperoxide) significantly decreased the cell volume and increased the number of annexin binding sites of erythrocytes from both, taut-/- and taut+/+ mice. However, decrease of cell volume and increase of annexin binding was significantly blunted in erythrocytes from taut-/- mice as compared to their taut+/+ littermates. Stimulation of erythropoiesis by prior hemorrhage did not abrogate the difference between taut+/+ and taut-/- erythrocytes. The present observations point to a decreased sensitivity of mature erythrocytes from taut-/- mice to osmotic shock and oxidative stress, rendering them more resistant to apoptosis.


Subject(s)
Apoptosis , Erythrocytes/cytology , Erythrocytes/metabolism , Membrane Glycoproteins/deficiency , Membrane Transport Proteins , Amino Acids/blood , Animals , Carrier Proteins/metabolism , Cell Size , Erythropoiesis , Female , Hemorrhage , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Osmotic Pressure , Oxidative Stress , Phosphatidylserines/metabolism , Sodium/metabolism
20.
Pancreas ; 27(1): 67-74, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12826908

ABSTRACT

INTRODUCTION: Proliferation and matrix synthesis of activated pancreatic stellate cells (PSCs) participate in the development of chronic pancreatitis. Besides other substances, endothelin-1 (ET-1) may influence the activation process of PSCs. Until now, ET-1 has not been studied in this particular cell type. AIMS: To characterize PSCs in rat pancreas with respect to expression of ET(A)-receptors, production of ET-1, and physiological effects induced by ET-1 during PSC activation. METHODOLOGY: Immunocytochemical and ELISA techniques and cDNA microarray analysis were used. Physiologic effects were characterized by single cell measurements of free cytosolic Ca2+-concentration and of PSC contractility on collagen lattices. RESULTS: Activation of PSCs in vitro, as assessed by alpha-smooth muscle actin expression, was accompanied by the de novo expression of ET(A)-receptors and synthesis of ET-1 mRNA and protein. Cytosolic Ca2+-concentration was increased upon ET-1 stimulation in activated but not in quiescent PSCs. Contractility of activated PSCs was significantly reduced by the selective ET(A)-receptor antagonist BQ123 but not by the ET(B)-receptor antagonist IRL-1038. CONCLUSIONS: The results suggest that ET-1 may act as a paracrine and autocrine factor for activated PSCs and may mediate contractions of activated, but not quiescent, PSCs.


Subject(s)
Endothelin-1/biosynthesis , Endothelin-1/pharmacology , Pancreas/cytology , Pancreas/drug effects , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Endothelin A Receptor Antagonists , Endothelin B Receptor Antagonists , Endothelin-1/genetics , Endothelins/pharmacology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Male , Oligonucleotide Array Sequence Analysis , Pancreas/metabolism , Peptide Fragments/pharmacology , Peptides, Cyclic/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...