Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Am J Physiol Renal Physiol ; 313(6): F1223-F1231, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28877884

ABSTRACT

Cystic epithelia acquire mesenchymal-like features in polycystic kidney disease (PKD). In this phenotypic alteration, it is well known that transforming growth factor (TGF)-ß/Smad3 signaling is involved; however, there is emerging new data on Smad3 phosphoisoforms: Smad3 phosphorylated at linker regions (pSmad3L), COOH-terminal regions (pSmad3C), and both (pSmad3L/C). pSmad3L/C has a pathological role in colorectal cancer. Mesenchymal phenotype-specific cell responses in the TGF-ß/Smad3 pathway are implicated in carcinomas. In this study, we confirmed mesenchymal features and examined Smad3 phosphoisoforms in the cpk mouse, a model of autosomal recessive PKD. Kidney sections were stained with antibodies against mesenchymal markers and domain-specific phospho-Smad3. TGF-ß, pSmad3L, pSmad3C, JNK, cyclin-dependent kinase (CDK) 4, and c-Myc were evaluated by Western blotting. Cophosphorylation of pSmad3L/C was assessed by immunoprecipitation. α-Smooth muscle actin, which indicates mesenchymal features, was expressed higher in cpk mice. pSmad3L expression was increased in cpk mice and was predominantly localized in the nuclei of tubular epithelial cells in cysts; however, pSmad3C was equally expressed in both cpk and control mice. Levels of pSmad3L, JNK, CDK4, and c-Myc protein in nuclei were significantly higher in cpk mice than in controls. Immunoprecipitation showed that Smad3 was cophosphorylated (pSmad3L/C) in cpk mice. Smad3 knockout/cpk double-mutant mice revealed amelioration of cpk abnormalities. These findings suggest that upregulating c-Myc through the JNK/CDK4-dependent pSmad3L pathway may be key to the pathophysiology in cpk mice. In conclusion, a qualitative rather than a quantitative abnormality of the TGF-ß/Smad3 pathway is involved in PKD and may be a target for disease-specific intervention.


Subject(s)
Epithelial Cells/metabolism , Kidney/metabolism , Polycystic Kidney, Autosomal Recessive/metabolism , Smad3 Protein/metabolism , Animals , Cyclin-Dependent Kinase 4/metabolism , Disease Models, Animal , Epithelial Cells/pathology , Genetic Predisposition to Disease , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphorylation , Polycystic Kidney, Autosomal Recessive/genetics , Polycystic Kidney, Autosomal Recessive/pathology , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Smad3 Protein/deficiency , Smad3 Protein/genetics
2.
J Neurosci Methods ; 219(1): 162-8, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-23912024

ABSTRACT

BACKGROUND: Although different gait analysis methods such as Walking Track Analysis exist, they cannot be used to demonstrate the physical condition of mice with specific gait disorder characteristic. Therefore, we developed a new method for the gait analysis of such mice to accurately assess hind limb angle based on the pelvic axis. NEW METHOD: We established and verified a gait analysis method capable of pelvic axis-based limb angle measurement by video-recording the gait of a control mice group (C57BL/6J(B6)) and three ataxic mice (ataxic B6-wob/t, Parkinson's disease model (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine treated (MPTP)), and cerebellum hypoplasia (cytosine-ß-d-arabinofuranoside treated)) from the ventral side. RESULTS: The assessed hind limb angles of B6-wob/t and MPTP-treated mice were significantly wider than B6 mice (p<0.01). Moreover, we could draw separating lines with slopes of minus one that could separate the data of each group in the scatter plot of the normalized hind limb step width and angle. COMPARISON WITH EXISTING METHODS: We found no significance when we applied the already existing nose-tail method for the analysis of the hind limb angles of B6 and B6-wob/t mice. In the nose-tail method, since the whole body axis of the trunk varies while the trunk of the mouse is laterally bent changing the hind limb angle, B6 and B6-wob/t mice could not be differentiated. However, the two mice groups could be differentiated by the pelvic axis-based gait analysis method. CONCLUSION: The pelvic axis-based gait analysis method is promising and valid for mice with gait disorder.


Subject(s)
Ataxia/physiopathology , Gait/physiology , Pelvis/physiology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , Biomechanical Phenomena , Cytarabine/pharmacology , Diet , Dopamine Agents/pharmacology , Hindlimb/anatomy & histology , Hindlimb/physiology , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Video Recording
3.
J Diabetes ; 3(4): 301-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21801331

ABSTRACT

BACKGROUND: The growth-associated increase in the blood glucose level of animals with Type 2 diabetes is inhibited by moderate hyperbaric exposure at 1.25 atmospheres absolute (ata) with 36% oxygen, presumably due to an increase in oxidative metabolism. However, there are no data available regarding the effect of moderate hyperbaric oxygen (HBO) on diabetes-induced cataracts. METHODS: Four-week-old mice with Type 2 diabetes and cataracts were exposed to 1.25 ata with 36% oxygen, 6 h daily, for 12 weeks, followed by normal conditions at 1 ata with 21% oxygen for 16 weeks (cataract + hyperbaric group). Levels of blood glucose and derivatives of reactive oxygen metabolites (dROMs), used as an index of oxidative stress, and the turbidities of the lenses from these mice at 4, 8, 12, 16, and 32 weeks of age were compared with those of control and diabetic (cataract group) mice not exposed to HBO. RESULTS: Non-fasting and fasting blood glucose levels were lower in the cataract + hyperbaric group at 12, 16, and 32 weeks of age than in the age-matched cataract group. The levels of dROMs were lower in the cataract + hyperbaric group at 16 and 32 weeks of age than in the age-matched cataract group. The turbidities of the peripheral and central regions of the lenses were lower in the cataract + hyperbaric group at 12, 16, and 32 weeks of age than in the age-matched cataract group. CONCLUSIONS: Hyperbaric exposure at 1.25 ata with 36% oxygen delays cataract development and progression in mice with Type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Cataract/etiology , Cataract/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Hyperbaric Oxygenation , Animals , Lens, Crystalline/pathology , Male , Mice , Oxidative Stress
4.
Am J Physiol Renal Physiol ; 300(2): F511-20, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21084407

ABSTRACT

In polycystic kidney disease (PKD), cyst lining cells show polarity abnormalities. Recent studies have demonstrated loss of cell contact in cyst cells, suggesting induction of epithelial-to-mesenchymal transition (EMT). Recently, EMT has been implicated in the pathogenesis of PKD. To explore further evidence of EMT in PKD, we examined age- and segment-specific expression of adhesion molecules and mesenchymal markers in PCK rats, an orthologous model of human autosomal-recessive PKD. Kidneys from 5 male PCK and 5 control rats each at 0 days, 1, 3, 10, and 14 wk, and 4 mo of age were serially sectioned and stained with segment-specific markers and antibodies against E-cadherin, Snail1, ß-catenin, and N-cadherin. mRNAs for E-cadherin and Snail1 were quantified by real-time PCR. Vimentin, fibronectin, and α-smooth muscle actin (α-SMA) expressions were assessed as mesenchymal markers. E-cadherin expression pattern was correlated with the disease pathology in that tubule segments showing the highest expression in control had much severer cyst formation in PCK rats. In PCK rats, E-cadherin and ß-catenin in cystic tubules was attenuated and localized to lateral areas of cell-cell contact, whereas nuclear expression of Snail1 increased in parallel with cyst enlargement. Some epithelial cells in large cysts derived from these segments, especially in adjacent fibrotic areas, showed positive immunoreactivity for vimentin and fibronectin. In conclusion, these findings suggest that epithelial cells in cysts acquire mesenchymal features in response to cyst enlargement and participate in progressive renal fibrosis. Our study clarified the nephron segment-specific cyst profile related to EMT in PCK rats. EMT may play a key role in polycystic kidney disease.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Polycystic Kidney, Autosomal Recessive/genetics , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Kidney/metabolism , Kidney/pathology , Kidney Tubules/chemistry , Kidney Tubules/metabolism , Male , Polycystic Kidney Diseases/metabolism , Polycystic Kidney, Autosomal Recessive/metabolism , Polycystic Kidney, Autosomal Recessive/pathology , Rats
5.
Kidney Blood Press Res ; 32(4): 239-49, 2009.
Article in English | MEDLINE | ID: mdl-19752573

ABSTRACT

BACKGROUND: Some reports have discussed the synergic effects of angiotensin II receptor blockers and calcium channel blockers on vascular injury or microalbuminuria. The present study examined the effects of combination treatment with olmesartan and azelnidipine on polycystic kidney disease in a mouse model (DBA/2-FG pcy mouse) and its mechanisms. METHODS: The mice were divided into the following groups: combination treatment (n = 21), olmesartan treatment alone (n = 23), azelnidipine treatment alone (n = 29) or untreated (n = 26). Mean blood pressure and kidney weight were measured at 4 and 8 weeks after the treatment. Renal expression of angiotensin II, gp91, nitrotyrosine and endothelial NO synthase (eNOS) were examined by immunostaining. In addition, extracellular signal-regulated kinase activation was evaluated by Western blotting. RESULTS: Olmesartan decreased the numbers of angiotensin II and gp91-positive cells, mainly macrophages, and cyst size at 4 weeks. However, only combination treatment suppressed cell infiltration, extracellular signal-regulated kinase activation and interstitial fibrosis with a significant change in the kidney weight/body weight ratio. The azelnidipine and combination treatment increased the numbers of interstitial eNOS-positive cells. CONCLUSION: The combination treatment protects against cyst enlargement in polycystic kidney disease by suppressing interstitial inflammation, fibrosis and oxidative stress by upregulating eNOS expression during disease course.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Azetidinecarboxylic Acid/analogs & derivatives , Calcium Channel Blockers/therapeutic use , Dihydropyridines/therapeutic use , Imidazoles/therapeutic use , Polycystic Kidney Diseases/drug therapy , Tetrazoles/therapeutic use , Angiotensin II/metabolism , Animals , Azetidinecarboxylic Acid/therapeutic use , Blotting, Western , Drug Synergism , Drug Therapy, Combination , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Immunohistochemistry , Kidney/pathology , Mice , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Polycystic Kidney Diseases/pathology
6.
Neurosci Res ; 57(4): 559-73, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17335925

ABSTRACT

Huntington's disease (HD) is caused by a mutation causing expanded polyglutamine tracts in the N-terminal fragment of huntingtin. A pathological hallmark of HD is the formation of aggregates in the striatal neurons. Here we report that ageing human huntingtin knock-in mice expressing mutant human huntingtin contained neuronal huntingtin aggregates, as revealed by immunohistochemical analysis. In heterozygous knock-in mice with 77 CAG repeats, aggregates of N-terminal fragments of huntingtin were specifically formed in nuclei and neuropils in the striatal projection neurons, and in neuropils in their projection regions. This aggregate formation progressed depending on age, became interacted with proteolytic or chaperone proteins, and occurred most prominently in the nucleus accumbens. These mutant mice demonstrated abnormal aggressive behavior. In homozygous knock-in mice, heavy deposits of intranuclear and neuropil aggregates were detected, which extended to other regions; and characteristic large perikaryal aggregates were also found in the affected neurons. However, cell death was not observed among the striatal and affected neurons of these mutant mice. Our results indicate that the polyglutamine aggregates do not necessarily correlate with neuronal death. These human huntingtin knock-in mice should be useful to provide an effective therapeutic approach against HD.


Subject(s)
Nerve Tissue Proteins/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Apoptosis/physiology , Behavior, Animal , Cell Nucleus/metabolism , Cell Nucleus/pathology , Corpus Striatum/cytology , Humans , Huntingtin Protein , In Situ Nick-End Labeling/methods , Mice , Mice, Transgenic , Motor Activity/genetics , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Neurons/pathology , Neuropil/metabolism , Protein Structure, Tertiary/physiology , Time Factors
7.
J Am Soc Nephrol ; 17(8): 2220-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16807403

ABSTRACT

Renal enlargement in polycystic kidney disease (PKD) is caused by the proliferation of mural epithelial cells and transepithelial fluid secretion into the cavities of innumerable cysts. Arginine vasopressin (AVP) stimulates the proliferation of human PKD cells in vitro via cAMP-dependent activation of the B-Raf/MEK (MAPK/ERK kinase/extracellular signal-regulated kinase (ERK) pathway. ERK activity is elevated in cells that line the cysts in animals with PKD, and AVP receptor antagonists reduce ERK activity and halt disease progression. For suppression of the effect of AVP physiologically, water intake was increased in PCK rats, a model of PKD, and the effect on renal morphology, cellular mechanism, and function was determined. The addition of 5% glucose in the drinking water increased fluid intake approximately 3.5-fold compared with rats that received tap water. In PCK rats, increased water intake for 10 wk reduced urinary AVP excretion (68.3%), and urine osmolality fell below 290 mOsmol/kg. High water intake was associated with reduced renal expression of AVP V2 receptors (41.0%), B-Raf (15.4%), phosphorylated ERK (38.1%), and proliferating cell nuclear antigen-positive renal cells (61.7%). High water intake reduced the kidney/body weight ratio 28.0% and improved renal function. Taken together, these data demonstrate that water intake that is sufficient to cause persistent water diuresis suppresses B-Raf/MEK/ERK activity and decreases cyst and renal volumes in PCK rats. It is suggested that limiting serum AVP levels by increased water intake may be beneficial to some patients with PKD.


Subject(s)
Polycystic Kidney Diseases/metabolism , Water/metabolism , Animals , Apoptosis , Cell Count , Cell Nucleus/metabolism , Disease Models, Animal , Disease Progression , Female , Immunohistochemistry , Male , Polycystic Kidney Diseases/pathology , Proliferating Cell Nuclear Antigen/metabolism , Random Allocation , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Sex Factors , Time Factors
8.
J Am Soc Nephrol ; 17(6): 1604-14, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16641154

ABSTRACT

The expression of mitogen-activated protein kinases (MAPK) in DBA/2-pcy/pcy (pcy) mice, a murine model of polycystic kidney disease was investigated. Proliferating cell nuclear antigen-positive cells were recognized in cyst epithelium from embryonic day 14.5 to 25 wk of age. Extracellular signal-regulated kinase (ERK) was expressed in the renal tubules of control and pcy mice, but stronger immunostaining was observed in cyst epithelium. Phosphorylated ERK was detected only in pcy mice and was localized predominantly in the cysts. p38 MAPK (p38) was no longer expressed after birth in controls but was detected in the cyst epithelium and in occasional tubular cells of pcy mice at all stages examined. c-Jun N-terminal kinase (JNK) was expressed in all tubular segments of controls after neonatal day 7, whereas in pcy kidneys, tubules became positive for JNK after 8 wk, and the cysts expressed little JNK. Administration of an oral MAP/ERK kinase inhibitor, PD184352, 400 mg/kg per d, to 10-wk-old pcy mice daily for the first week and then every third day for 6 additional weeks significantly decreased BP, kidney weight, serum creatinine level, and water intake and significantly increased urine osmolality. The cystic index and expression of phosphorylated ERK and ERK were significantly lower in PD184352-treated pcy mice. These results demonstrate that the expression of MAPK is dysregulated in cyst epithelium and that inhibition of ERK slowed the progression of renal disease in pcy mice.


Subject(s)
Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Gene Expression Regulation , Polycystic Kidney Diseases/drug therapy , Polycystic Kidney Diseases/enzymology , Animals , Apoptosis , Benzamides/pharmacology , Cell Proliferation , Disease Models, Animal , Disease Progression , Enzyme Activation , Mice , Phosphorylation , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Am Soc Nephrol ; 16(7): 2052-62, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15888569

ABSTRACT

Androgens have been implicated in mediating disease escalation in autosomal dominant polycystic kidney disease (ADPKD). Dihydrotestosterone (DHT), an agonist, and flutamide (FLT), an antagonist, were administered to Han:SPRD rats with ADPKD, and the role of androgen receptor (AR) abundance and activation on the enlargement and function of cystic kidneys was evaluated. Renal AR abundance determined by immunoblots in 8- to 10-wk-old Cy/+ male rats was naturally increased four-fold above that of littermate +/+ controls. In male Cy/+, castration decreased AR abundance below control +/+ by -89.4%, and AR expression within cyst mural epithelial cells was strikingly decreased. Castration of Cy/+ male rats also reduced the usual increases in kidney weight by -49.7%, kidney cyst area by -34.0%, and serum urea nitrogen by -72.8%; these indices were restored to precastration levels by DHT. In Cy/+ male rats, FLT administration reduced the increase in kidney weight by -27.6% and serum urea nitrogen by -53.7% and decreased the increment in AR expression by -84.2% in comparison with untreated +/+ controls. There was no effect of FLT in female rats. Immunoblot expression of phospho-extracellular signal-regulated kinase 1/2 (P-ERK) and B-Raf, key intermediates in the mitogen-activated protein kinase pathway that are abnormally elevated in Cy/+, was unaffected by castration and/or administration of DHT or FLT. AR was not expressed in renal epithelial cell nuclei of androgen-deficient rats but was displayed in most tubule and mural cyst cell nuclei of androgen-replete rats. In androgen-deficient Cy/+, 80.6% of renal epithelial cells that had entered the cell cycle (proliferating cell nuclear antigen positive) also expressed P-ERK. In androgen-replete rats, proliferating cell nuclear antigen-positive cells co-expressed AR (12.7%), P-ERK (36.4%), and P-ERK + AR (45.0%); 5.9% were probably stimulated by other mitogenic mechanisms. It is concluded that androgens potentiate renal cell proliferation and cyst enlargement through ERK1/2-dependent and ERK1/2-independent signaling mechanisms in Han:SPRD. It is suggested that the basal rate of cell proliferation is determined by ERK1/2 signaling to a major extent and that androgens have additive effects.


Subject(s)
Polycystic Kidney, Autosomal Dominant/physiopathology , Receptors, Androgen/metabolism , Androgen Antagonists/pharmacology , Animals , Cell Proliferation , Dihydrotestosterone/pharmacology , Disease Models, Animal , Disease Progression , Female , Flutamide/pharmacology , Male , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Polycystic Kidney, Autosomal Dominant/metabolism , Proto-Oncogene Proteins B-raf/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Androgen/drug effects , Sex Factors , Signal Transduction , Testosterone Congeners/pharmacology , eIF-2 Kinase/biosynthesis
10.
Clin Exp Pharmacol Physiol ; 32(4): 279-87, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15810992

ABSTRACT

1. The present study was carried out to explain the resistance of rats injected subcutaneously with risperidone, the atypical antipsychotic drug, for 21 consecutive days at 0.1 mg/kg per day (a dose equivalent to the one used for patients) to result in an excessive bodyweight despite the increase in diet-uptake in rats against risperidone-induced decrease in body temperature. 2. Rectal temperature measurements were made in 8-week-old male Sprague-Dawley rats maintained under standard laboratory conditions using a 12 h daylight cycle. A s.c. injection of risperidone (0.05 mg/kg) produced hypothermia in rats, which was observed during the daily injection for 21 consecutive days. 3. Sera, white and brown adipose tissues, skeletal muscle and liver were extracted from 8-week-old male Sprague-Dawley rats injected subcutaneously with risperidone (0.01 or 0.1 mg/kg per day) or a vehicle for 21 consecutive days. Serum levels of lipids, ketones and thyroid hormone were measured. The mRNA expression levels in these tissues and organs of the genes encoding the substances involved in heat production and/or lipid metabolism were investigated by using quantitative real-time polymerase chain reaction amplification. 4. Serum nonesterified fatty acid levels in risperidone 0.1 mg/kg per day s.c. injected rats were significantly lower than those in vehicle-injected ones. Serum beta-hydroxybutyrate levels in risperidone-injected rats tended to decrease compared with those in vehicle-injected ones. The serum level of neither triiodothyronine nor thyroxine was affected by risperidone s.c. injection at the doses examined, although their values were within normal limits. 5. Risperidone injection (0.1 mg/kg per day) for 21 consecutive days upregulated mRNA expressions in white adipose tissue of uncoupling protein 3 which dissipates energy as heat; peroxisome proliferator-activated receptor (PPAR) gamma coactivator 1alpha which activates mitochondrial biogenesis to expand the oxidative machinery; and PPARalpha which is necessary for the fat-depletion of adipocytes for thermogenesis. The mRNA of lipogenic enzymes (acetyl-CoA carboxylase alpha, fatty-acid synthase and glycerol-3-phosphate acyltransferase), hormone sensitive lipase and beta1-adrenoceptor were also enhanced in white adipose tissue by the injection of 0.1 mg/kg per day risperidone. 6. These findings suggest that the materials for heat generation in white adipose tissue would be readily supplied, which in turn would reduce a storage of lipids in white adipose tissue resulting in the lower rate of bodyweight gain of rats.


Subject(s)
Body Weight/drug effects , Risperidone/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Body Temperature/drug effects , Gene Expression/drug effects , Gene Expression Profiling , Lipids/blood , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Thyroid Hormones/blood , Up-Regulation/genetics
11.
Brain Res ; 1039(1-2): 116-29, 2005 Mar 28.
Article in English | MEDLINE | ID: mdl-15781053

ABSTRACT

The olfactory bulb (OB) is one of the few structures in the adult mammalian CNS that contains a continuous supply of newly generated neurons in the subventricular zone. Therefore, the balance between the supply of new cells and apoptosis in the OB might determine olfactory function. Lipopolysaccharide-induced tumor necrosis factor (TNF)-alpha triggers the apoptotic cascade mediated by the TNF/TNF receptor (TNFR) pathway. The present study therefore examines the effect of the propagated innate immune reaction triggered by peripheral lipopolysaccharide on the OB of C3H/HeN mice. Within 2 h of an intraperitoneal injection of lipopolysaccharide, mRNA expression levels of the genes encoding IkappaB, TNF-alpha, and TNFR type 1 in the mouse OB were significantly enhanced. Double immunofluorescence microscopy confirmed that almost all TNF-alpha-immunopositive cells in the OB of the TNF-injected mice were located in the subependymal zone and that they overlapped cells immunostained with antibody against glial fibrillary acidic protein, but not with the antibody against F4/80, an antigenic marker of microglia. The number of TUNEL-positive cells identified exclusively in the granule cell layer was significantly increased in mice injected with lipopolysaccharide and sacrificed at 24 h thereafter. These results suggest that peripheral lipopolysaccharide causes disequilibrium between the supply and disappearance of the cells in the OB, which might lead to olfactory dysfunction.


Subject(s)
Apoptosis/immunology , Lipopolysaccharides/immunology , Olfactory Bulb/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , I-kappa B Kinase , Immunohistochemistry , Injections, Intraperitoneal , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred C3H , Olfactory Bulb/cytology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/analysis , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/immunology , Tissue Distribution , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
J Mol Cell Cardiol ; 37(3): 767-74, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15350849

ABSTRACT

Cyclic nucleotides (cAMP and cGMP) phosphodiesterase (PDE) activities and expression are altered in the cardiac muscle of cardiomyopathic heart failure, and PDE inhibitors improve the abnormal muscle condition through changing the cyclic nucleotide concentration. These observations prompted us to investigate the role of calmodulin (CaM) in the regulation of cyclic nucleotide PDE activities, and moreover to study the modulation of the PDE isozymes in heart failure, using cardiac muscles of cardiomyopathic hamster. The CaM concentrations in the heart muscle of the normal control and cardiomyopathic hamsters (each of three to four hamsters) varied with cell fraction and with the age of the animal. The CaM concentrations in the soluble fraction obtained from cardiomyopathic hamster tissue were significantly increased at 25 and 32 weeks of age (2.02 +/- 0.62 microg/mg protein (mean +/- S.E.), and 3.21 +/- 0.95) compared with that obtained from the control (0.60 +/- 0.04) or cardiomyopathic (0.95 +/- 0.12) hamsters at 8 weeks of age. The solubilized PDE isolated from the hamster heart muscle (three or four hamsters in each age) by column chromatography on diethylaminoethyl (DEAE)-cellulose revealed three peaks of activity, which may correspond to the isozymes of PDE classified recently, namely PDE I, II, and III. These three peaks of activity, particularly peak III, seen in the soluble fraction of cardiomyopathic hamster heart declined in proportion to the age of the animal compared with that of the control hamster heart. In the cGMP-PDE assay system, the concentration of CaM inhibitor W-7 required for 50% inhibition (IC(50)) of PDE I, II, and III peak activities was 140, 29, and 46 microM, respectively, suggesting that PDE II is more sensitive to W-7. These results suggest that alteration in these isozyme activities accompanied with changes of CaM concentration may influence the cardiac muscle contractility in cardiomyopathic hamster via changes of cyclic nucleotide concentration.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/biosynthesis , Calmodulin/metabolism , Cardiomyopathies/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Age Factors , Animals , Cardiomyopathies/complications , Cardiomyopathies/pathology , Cricetinae , Gene Expression Regulation , Heart Failure/etiology , Heart Failure/pathology , Isoenzymes/biosynthesis , Myocardial Contraction , Myocardium/pathology
13.
Kidney Int ; 63(2): 427-37, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12631108

ABSTRACT

BACKGROUND: Abnormal proliferation of renal tubule epithelial cells is a central factor in the biogenesis and sustained expansion of cysts in autosomal-dominant polycystic kidney disease (ADPKD). Recent evidence from in vitro studies of human cyst wall epithelial cells has implicated a role for the mitogen-activated protein (MAP) kinase pathway in this aberrant proliferation. To determine the extent to which this signaling pathway is involved in cyst pathogenesis in vivo, we measured the expression of select components of the MAP kinase cascade in Han:SPRD rats with ADPKD at an early stage of the disease. METHODS: Kidneys of 8-week-old normal Han:SPRD rats (+/+) or rats heterozygous (Cy/+) for ADPKD were examined by Western blot analysis and immunohistochemistry to determine the expression of extracellular-regulated kinase (ERK), phosphorylated ERK (P-ERK), Raf-1 (MAPKKK), phosphorylated Raf-1 (P-Raf-1), B-Raf, Rap-1 and phosphorylated protein kinase A (P-PKA). RESULTS: P-ERK was expressed to a greater extent in Cy/+ kidneys (3.74 +/- 1.07 fold) than in normal kidneys, whereas ERK abundance was not different. P-Raf-1 levels were higher in Cy/+ than in +/+ kidneys (1.53 +/- 0.08 fold) consistent with upstream stimulation of receptor tyrosine kinase. B-Raf and Raf-1 abundances were greater in Cy/+ than in +/+ (1.74 +/- 0.25 and 1.27 +/- 0.08 fold, respectively). In Cy/+, immunohistochemistry showed increased P-ERK and B-Raf expression in the abnormal mural epithelial cells within cysts. These findings, together with the detection of P-PKA and the small G protein, Rap-1, in cyst epithelial cells, implicate a potential role for cyclic adenosine monophosphate (AMP) in the activation of ERK in ADPKD cells. CONCLUSIONS: We conclude that the MAP kinase pathway is activated to the level of ERK in the abnormal mural epithelial cells lining cysts in animals with a dominantly inherited type of polycystic kidney disease. We suggest that cAMP, acting through PKA, Rap-1 and B-Raf, may contribute to the activation of ERK in a way that complements receptor tyrosine kinase-mediated agonists in the promotion of cyst enlargement.


Subject(s)
Kidney/enzymology , Mitogen-Activated Protein Kinases/metabolism , Polycystic Kidney, Autosomal Dominant/enzymology , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Progression , Enzyme Activation , Immunoblotting , Immunohistochemistry , MAP Kinase Kinase Kinases/metabolism , Phosphorylation , Polycystic Kidney, Autosomal Dominant/physiopathology , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-raf/metabolism , Rats , Rats, Sprague-Dawley
14.
Hypertens Res ; 25(6): 939-43, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12484519

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary renal disorder in humans. Hypertension is one of the major complications, and its control might affect the renal survival and disease mortality. Suitable antihypertensive agents have been discussed based on clinical and animal studies, but no definitive conclusion has been reached. Generally, therefore, all antihypertensives are indiscriminately treated as if providing the same level of blood pressure control. In this study, the blood pressure control of two antihypertensives was investigated using a rat model of ADPKD in humans. Twenty-four male Hannover-Sprague Dawley (Han:SPRD) rats were divided into three groups: a group receiving amlodipine (6 mg/day), a group receiving benazepril (6 mg/day) and an untreated control group. Blood pressure, body weight, and urinary protein excretion were regularly measured up to week 52. Amlodipine and benazepril significantly decreased blood pressure and urinary protein excretion to the same degree. Moreover, a remarkably prolonged survival rate was observed in both groups (at week 52, the survival rate was 25% in controls, 50% in the amlodipine group, and 50% in the benazepril group). Examination at autopsy revealed that enlarged cysts were prevalent in the renal tissue of both experimental all three groups, suggesting that the cystic disease had reached the end-stage in all the animals. In conclusion, both amlodipine and benazepril significantly improved blood pressure control, urinary protein excretion, and survival rate, possibly due to their enhancement of renal survival.


Subject(s)
Amlodipine/therapeutic use , Antihypertensive Agents/therapeutic use , Benzazepines/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/physiopathology , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Kidney/pathology , Kidney Tubules/pathology , Male , Polycystic Kidney, Autosomal Dominant/mortality , Polycystic Kidney, Autosomal Dominant/pathology , Proteinuria/etiology , Proteinuria/urine , Rats , Rats, Sprague-Dawley , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL