Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Int J Nephrol Renovasc Dis ; 13: 19-26, 2020.
Article in English | MEDLINE | ID: mdl-32110087

ABSTRACT

AIM OF STUDY: To investigate podocyte density in aging diabetic Ins2± and Ins2±, A1AR-/- mouse models in C57Bl/6 background. METHODS: Ins2± mice and especially Ins2±, adenosine A1 receptor knockout mice (Ins2±, A1AR-/-) are mouse models with a phenotype of diabetic nephropathy. Aged mice (at ~40 weeks) were assessed for glomerular filtration barrier function by measuring albuminuria, glomerular filtration, glomerular damage by electron microscopy, and podocyte numbers by Wilms Tumor protein (WT-1) staining. RESULTS: Compared to healthy wild-type mice, both diabetic mouse models developed diabetic nephropathy, including hyperfiltration (p<0.01) and albuminuria (p<0.05). Typical diabetic structural glomerular and podocyte damage was visualized by electron microscopy. Podocyte count per glomerular area (podocyte density) was significantly decreased in both diabetic mouse models (p<0.01). In contrast, no significant correlation was detected between albuminuria and absolute podocyte count per glomerulus. CONCLUSION: The amount of albuminuria as marker of diabetic nephropathy does not correlate with the podocytes density; however, a relative podocyte deficiency became evident with an increase in glomerular area in the diabetic animals, suggesting a relative podocytopenia.

3.
JCI Insight ; 3(23)2018 12 06.
Article in English | MEDLINE | ID: mdl-30518688

ABSTRACT

Methylmalonic acidemia (MMA), an organic acidemia characterized by metabolic instability and multiorgan complications, is most frequently caused by mutations in methylmalonyl-CoA mutase (MUT). To define the metabolic adaptations in MMA in acute and chronic settings, we studied a mouse model generated by transgenic expression of Mut in the muscle. Mut-/-;TgINS-MCK-Mut mice accurately replicate the hepatorenal mitochondriopathy and growth failure seen in severely affected patients and were used to characterize the response to fasting. The hepatic transcriptome in MMA mice was characterized by the chronic activation of stress-related pathways and an aberrant fasting response when compared with controls. A key metabolic regulator, Fgf21, emerged as a significantly dysregulated transcript in mice and was subsequently studied in a large patient cohort. The concentration of plasma FGF21 in MMA patients correlated with disease subtype, growth indices, and markers of mitochondrial dysfunction but was not affected by renal disease. Restoration of liver Mut activity, by transgenesis and liver-directed gene therapy in mice or liver transplantation in patients, drastically reduced plasma FGF21 and was associated with improved outcomes. Our studies identify mitocellular hormesis as a hepatic adaptation to metabolic stress in MMA and define FGF21 as a highly predictive disease biomarker.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Fibroblast Growth Factors/metabolism , Hormesis , Methylmalonyl-CoA Mutase/metabolism , Stress, Physiological , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Biomarkers/blood , Disease Models, Animal , Female , Fibroblast Growth Factors/blood , Genetic Therapy , Humans , Kidney Diseases/metabolism , Liver/metabolism , Liver/pathology , Liver Transplantation , Male , Methylmalonyl-CoA Mutase/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Phenotype , Transcriptome
4.
Clin Exp Nephrol ; 22(5): 1052-1060, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29516300

ABSTRACT

BACKGROUND: Cirrhosis of the liver is often associated with an impairment of renal function that is usually not associated with consistent structural abnormalities of the renal parenchyma, but is thought to be the functional consequence of arterial underfilling and reduced arterial blood pressure. METHOD: We have used the cirrhosis model of chronic bile duct ligation (BDL) to assess the response of renal blood flow to a change of blood pressure. We have measured renal haemodynamics in BDL rats. RESULT: Three weeks after BDL, rats showed elevated levels of total bilirubin, AST, and ALT as well as reduced arterial blood pressure. Creatinine clearance was significantly reduced, and plasma creatinine and urea nitrogen were elevated. Renal blood flow at baseline blood pressure was significantly lower in the BDL group than in the sham group. Clamp-induced reductions of renal perfusion pressure caused significantly greater changes of renal blood flow in BDL than control rats. The autoregulatory index over a comparable blood pressure range averaged 0.28 ± 0.35 in control rats and 1.26 ± 0.6 in BDL rats (p = 0.0004) indicating impairment of renal autoregulation in liver cirrhosis. CONCLUSION: Tubuloglomerular feedback (TGF) responses were significantly attenuated in BDL rats, especially in the subnormal flow range. Impairment of renal blood flow autoregulation, to some extent mediated by reduced TGF-mediated vasodilatation, may contribute to the renal vascular constrictor state in liver cirrhosis by preventing the full dilatory response to the blood pressure reduction.


Subject(s)
Disease Models, Animal , Homeostasis , Liver Failure , Animals , Bile Ducts , Liver , Male , Rats , Rats, Sprague-Dawley
5.
Pflugers Arch ; 469(2): 339-347, 2017 02.
Article in English | MEDLINE | ID: mdl-27975140

ABSTRACT

Administration of the nucleoside adenosine has been shown to induce hypothermia in a number of species, an effect mediated predominantly by the adenosine 1 receptor (A1AR) subtype. The present experiments were performed to explore the possibility that the rise of intracellular adenosine levels expected to accompany adenosine administration may contribute to the hypothermic effect of adenosine independent of A1AR activation. Since phosphorylation of adenosine by adenosine kinase (ADK) is causal in the maintenance of low intracellular adenosine, we have examined the effect of ADK inhibition on core body temperature (CBT). Our data show that inhibition of ADK by A-134974 causes a long-lasting deep hypothermia in wild-type mice. Since there was an about 4-fold increase of adenosine plasma levels, experiments were repeated in A1AR-/- mice. ADK inhibition caused deep hypothermia despite the absence of A1AR, although the effect was significantly reduced compared to WT. Furthermore, the dose-dependent hypothermia caused by adenosine administration in WT mice was found to be reduced, but not abolished in A1AR-/- mice. To assess the possible role of A2AR and A3AR activation in our experimental setting, we compared the effects of the agonists CPA (A1AR), CGS21680 (A2AR), and IB-MECA (A3AR) on CBT. Hypothermia induced by CPA was much greater than that caused by CGS21680 or IB-MECA indicating that A1AR activation is the major receptor-dependent pathway for adenosine-induced hypothermia under our experimental conditions. Induction of deep hypothermia by inhibition of ADK, maintenance of this effect in A1AR-/- mice, and maintenance of adenosine-induced hypothermia in A1AR-deficient mice suggest that a receptor-independent action of adenosine requiring intact function of adenosine kinase contributes importantly to the hypothermia induced by adenosine.


Subject(s)
Adenosine Kinase/antagonists & inhibitors , Adenosine/metabolism , Hypothermia/metabolism , Receptor, Adenosine A1/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Nucleosides/pharmacology , Phenethylamines/pharmacology
6.
Am J Physiol Heart Circ Physiol ; 310(7): H891-8, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26801314

ABSTRACT

Bombesin-like receptor 3 (BRS-3) is an orphan G protein-coupled receptor that regulates energy expenditure, food intake, and body weight. We examined the effects of BRS-3 deletion and activation on blood pressure and heart rate. In free-living, telemetered Brs3 null mice the resting heart rate was 10% lower than wild-type controls, while the resting mean arterial pressure was unchanged. During physical activity, the heart rate and blood pressure increased more in Brs3 null mice, reaching a similar heart rate and higher mean arterial pressure than control mice. When sympathetic input was blocked with propranolol, the heart rate of Brs3 null mice was unchanged, while the heart rate in control mice was reduced to the level of the null mice. The intrinsic heart rate, measured after both sympathetic and parasympathetic blockade, was similar in Brs3 null and control mice. Intravenous infusion of the BRS-3 agonist MK-5046 increased mean arterial pressure and heart rate in wild-type but not in Brs3 null mice, and this increase was blocked by pretreatment with clonidine, a sympatholytic, centrally acting α2-adrenergic agonist. In anesthetized mice, hypothalamic infusion of MK-5046 also increased both mean arterial pressure and heart rate. Taken together, these data demonstrate that BRS-3 contributes to resting cardiac sympathetic tone, but is not required for activity-induced increases in heart rate and blood pressure. The data suggest that BRS-3 activation increases heart rate and blood pressure via a central sympathetic mechanism.


Subject(s)
Blood Pressure , Heart Rate , Receptors, Bombesin/metabolism , Sympathetic Nervous System/physiology , Adrenergic Agents/pharmacology , Animals , Mice , Mice, Inbred C57BL , Receptors, Bombesin/genetics , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
7.
Am J Physiol Renal Physiol ; 309(1): F29-32, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25947344

ABSTRACT

Data variability is a costly complication of biomedical experimentation because the same experiment must be repeated a sufficient number of times so that the sample mean becomes a credible representation of the entire population. Since sampling is ideally done randomly in populations normalized for environmental and genetic backgrounds, data variability is viewed as a purely statistical issue reflecting the distribution in the population and captured as the standard deviation of the sampled data. The factors contributing to data variability are not analyzed by statistical methods; for want of a better explanation, data scatter is simply attributed to random noise and/or methodological limitations. In this commentary, evidence is discussed that documents an important role of interindividual biological diversity as a cause for data variability based on studies in which repeated sampling in the same individual permitted statistical comparisons between individuals in the same sample. Significant differences were found for proximal fluid reabsorption and plasma renin concentration between sample means of individuals of the same population. Furthermore, arterial blood pressure varied significantly between individual mice independently of strain and sex. Recognition of the extent of interindividual variability has important implications for data reproducibility, data collection, and data presentation in physiological research. Such nonrandom data variability may have different causes, but DNA modifications by genetic or epigenetic mechanisms could generate phenotype variants without being associated with disease symptoms. Exploration of the heritability of phenotypical diversity in physiology may be defined as "physiogenetics," and it would thus be the physiological corollary of pharmacogenetics and pharmacogenomics.


Subject(s)
Biomedical Research , Blood Pressure , Individuality , Renal Reabsorption , Renin/blood , Animals , Phenotype
8.
Annu Rev Physiol ; 77: 301-22, 2015.
Article in English | MEDLINE | ID: mdl-25668021

ABSTRACT

Tubuloglomerular feedback (TGF) describes the negative relationship between (a) NaCl concentration at the macula densa and (b) glomerular filtration rate or glomerular capillary pressure. TGF-induced vasoconstriction of the afferent arteriole results from the enhanced effect of several vasoconstrictors with an effect size sequence of adenosine = 20-HETE > angiotensin II > thromboxane = superoxide > renal nerves > ATP. TGF-mediated vasoconstriction is limited by the simultaneous release of several vasodilators with an effect size sequence of nitric oxide > carbon monoxide = kinins > adenosine. The sum of the constrictor effects exceeds that of the dilator effects by the magnitude of the TGF response. The validity of the additive model used in this analysis can be tested by determining the effect of combined inhibition of some or all agents contributing to TGF. Multiple independent contributors to TGF are consistent with the variability of TGF and of the factors contributing to TGF resetting.


Subject(s)
Feedback, Physiological/physiology , Kidney Glomerulus/physiology , Kidney Tubules/physiology , Signal Transduction/physiology , Vasoconstriction/physiology , Animals , Arterioles/physiology , Humans , Kidney/blood supply , Models, Animal , Vasoconstrictor Agents , Vasodilator Agents
9.
Clin J Am Soc Nephrol ; 10(4): 676-87, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25098598

ABSTRACT

The kidney filters vast quantities of Na at the glomerulus but excretes a very small fraction of this Na in the final urine. Although almost every nephron segment participates in the reabsorption of Na in the normal kidney, the proximal segments (from the glomerulus to the macula densa) and the distal segments (past the macula densa) play different roles. The proximal tubule and the thick ascending limb of the loop of Henle interact with the filtration apparatus to deliver Na to the distal nephron at a rather constant rate. This involves regulation of both filtration and reabsorption through the processes of glomerulotubular balance and tubuloglomerular feedback. The more distal segments, including the distal convoluted tubule (DCT), connecting tubule, and collecting duct, regulate Na reabsorption to match the excretion with dietary intake. The relative amounts of Na reabsorbed in the DCT, which mainly reabsorbs NaCl, and by more downstream segments that exchange Na for K are variable, allowing the simultaneous regulation of both Na and K excretion.


Subject(s)
Nephrons/metabolism , Renal Elimination , Renal Reabsorption , Sodium/metabolism , Animals , Biological Transport , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Glomerular Filtration Rate , Humans , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Natriuresis , Nephrons/physiopathology , Potassium/metabolism , Renin-Angiotensin System , Sodium, Dietary/metabolism , Water-Electrolyte Balance
10.
Am J Physiol Renal Physiol ; 307(8): F939-48, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25143457

ABSTRACT

Acute kidney injury (AKI) dramatically increases sepsis mortality, but AKI diagnosis is delayed when based on serum creatinine (SCr) changes, due in part, to decreased creatinine production. During experimental sepsis, we compared serum cystatin C (sCysC), SCr, and blood urea nitrogen (BUN) to inulin glomerular filtration rate (iGFR) before or 3-18 h after cecal ligation and puncture (CLP)-induced sepsis in CD-1 mice. sCysC had a faster increase and reached peak levels more rapidly than SCr in both sepsis and bilateral nephrectomy (BiNx) models. sCysC was a better surrogate of iGFR than SCr during sepsis. Combining sCysC with SCr values into a composite biomarker improved correlation with iGFR better than any biomarker alone or any other combination. We determined the renal contribution to sCysC handling with BiNx. sCysC and SCr were lower post-BiNx/CLP than post-BiNx alone, despite increased inflammatory and nonrenal organ damage biomarkers. Sepsis decreased CysC production in nephrectomized mice without changing body weight or CysC space. Sepsis decreased sCysC production and increased nonrenal clearance, similar to effects of sepsis on SCr. sCysC, SCr, and BUN were measured 6 h postsepsis to link AKI with mortality. Mice with above-median sCysC, BUN, or SCr values 6 h postsepsis died earlier than mice with below-median values, corresponding to a substantial AKI association with sepsis mortality in this model. sCysC performs similarly to SCr in classifying mice at risk for early mortality. We conclude that sCysC detects AKI early and better reflects iGFR in CLP-induced sepsis. This study shows that renal biomarkers need to be evaluated in specific contexts.


Subject(s)
Acute Kidney Injury/mortality , Biomarkers/blood , Creatinine/blood , Cystatin C/blood , Sepsis/mortality , Acute Kidney Injury/blood , Acute Kidney Injury/physiopathology , Animals , Blood Urea Nitrogen , Cecum/injuries , Glomerular Filtration Rate , Inulin , Ligation , Male , Mice , Nephrectomy , Punctures , Sepsis/complications
11.
F1000Res ; 2: 83, 2013.
Article in English | MEDLINE | ID: mdl-24358878

ABSTRACT

An increase of glomerular filtration rate (hyperfiltration) is an early functional change associated with type I or type II diabetes mellitus in patients and animal models. The causes underlying glomerular hyperfiltration are not entirely clear. There is evidence from studies in the streptozotocin model of diabetes in rats that an increase of proximal tubular reabsorption results in the withdrawal of a vasoconstrictor input exerted by the tubuloglomerular feedback (TGF) mechanism. In the present study, we have used micropuncture to assess single nephron function in wild type (WT) mice and in two strains of type I diabetic Ins2+/- mice in either a C57Bl/6 (Akita) or an A1AR-/- background (Akita/A1AR-/-) in which TGF is non-functional. Kidney glomerular filtration rate (GFR) of anesthetized mice was increased by 25% in Akita mice and by 52% in Akita/A1AR-/-, but did not differ between genotypes when corrected for kidney weight. Single nephron GFR (SNGFR) measured by end-proximal fluid collections averaged 11.8 ± 1 nl/min (n=17), 13.05 ± 1.1 nl/min (n=23; p=0.27), and 15.4 ± 0.84 nl/min (n=26; p=0.009 compared to WT; p=0.09 compared to Akita) in WT, Akita, and Akita/A1AR-/- mice respectively. Proximal tubular fluid reabsorption was not different between WT and diabetic mice and correlated with SNGFR in all genotypes. We conclude that glomerular hyperfiltration is a primary event in the Akita model of type I diabetes, perhaps driven by an increased filtering surface area, and that it is ameliorated by TGF to the extent that this regulatory system is functional.

12.
Am J Physiol Renal Physiol ; 305(9): F1352-64, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24049145

ABSTRACT

Deletions of claudin-2 (Cldn2) and aquaporin1 (AQP1) reduce proximal fluid reabsorption (PFR) by about 30% and 50%, respectively. Experiments were done to replicate these observations and to determine in AQP1/claudin-2 double knockout mice (DKO) if the effects of deletions of these established water pores are additive. PFR was determined in inactin/ketamine-anesthetized mice by free-flow micropuncture using single-nephron I(125)-iothalamate (io) clearance. Animal means of PFR [% of glomerular filtration rate (GFR)] derived from TF/Piothalamate ratios in 12 mice in each of four groups [wild type (WT), Cldn2(-/-), AQP1(-/-), and DKO) were 45.8 ± 0.85 (51 tubules), 35.4 ± 1 (54 tubules; P < 0.01 vs. WT), 36.8 ± 1 (63 tubules; P < 0.05 vs. WT), and 33.9 ± 1.4 (69 tubules; P < 0.01 vs. WT). Kidney and single-nephron GFRs (SNGFR) were significantly reduced in all mutant strains. The direct relationship between PFR and SNGFR was maintained in mutant mice, but the slope of this relationship was reduced in the absence of Cldn2 and/or AQP1. Transtubular osmotic pressure differences were not different between WT and Cldn2(-/-) mice, but markedly increased in DKO. In conclusion, the deletion of Cldn2, AQP1, or of both Cldn2 and AQP1 reduces PFR by 22.7%, 19.6%, and 26%, respectively. Our data are consistent with an up to 25% paracellular contribution to PFR. The reduced osmotic water permeability caused by absence of AQP1 augments luminal hypotonicity. Aided by a fall in filtered load, the capacity of non-AQP1-dependent transcellular reabsorption is sufficient to maintain PFR without AQP1 and claudin-2 at 75% of control.


Subject(s)
Aquaporin 1/physiology , Claudins/physiology , Kidney Tubules, Proximal/physiology , Animals , Gene Deletion , Kidney Function Tests , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmolar Concentration , RNA, Messenger/metabolism
13.
Proc Natl Acad Sci U S A ; 110(33): 13552-7, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23898205

ABSTRACT

Isolated methylmalonic acidemia (MMA), caused by deficiency of the mitochondrial enzyme methylmalonyl-CoA mutase (MUT), is often complicated by end stage renal disease that is resistant to conventional therapies, including liver transplantation. To establish a viable model of MMA renal disease, Mut was expressed in the liver of Mut(-/-) mice as a stable transgene under the control of an albumin (INS-Alb-Mut) promoter. Mut(-/-);Tg(INS-Alb-Mut) mice, although completely rescued from neonatal lethality that was displayed by Mut(-/-) mice, manifested a decreased glomerular filtration rate (GFR), chronic tubulointerstitial nephritis and ultrastructural changes in the proximal tubule mitochondria associated with aberrant tubular function, as demonstrated by single-nephron GFR studies. Microarray analysis of Mut(-/-);Tg(INS-Alb-Mut) kidneys identified numerous biomarkers, including lipocalin-2, which was then used to monitor the response of the GFR to antioxidant therapy in the mouse model. Renal biopsies and biomarker analysis from a large and diverse patient cohort (ClinicalTrials.gov identifier: NCT00078078) precisely replicated the findings in the animals, establishing Mut(-/-);Tg(INS-Alb-Mut) mice as a unique model of MMA renal disease. Our studies suggest proximal tubular mitochondrial dysfunction is a key pathogenic mechanism of MMA-associated kidney disease, identify lipocalin-2 as a biomarker of increased oxidative stress in the renal tubule, and demonstrate that antioxidants can attenuate the renal disease of MMA.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/enzymology , Antioxidants/pharmacology , Disease Models, Animal , Kidney Tubules, Proximal/physiopathology , Methylmalonyl-CoA Mutase/deficiency , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Antioxidants/therapeutic use , Biomarkers/metabolism , Blotting, Western , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Fluorescein-5-isothiocyanate , Genotype , Glomerular Filtration Rate/genetics , Humans , Immunohistochemistry , Methylmalonyl-CoA Mutase/genetics , Methylmalonyl-CoA Mutase/metabolism , Mice , Mice, Knockout , Microarray Analysis , Microscopy, Electron, Transmission , Nephritis, Interstitial/genetics , Real-Time Polymerase Chain Reaction , Transgenes/genetics , Ubiquinone/pharmacology
14.
Am J Physiol Renal Physiol ; 304(9): F1181-6, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23445620

ABSTRACT

Participation of connexin 40 (Cx40) in the regulation of renin secretion and in the tubuloglomerular feedback (TGF) component of renal autoregulation suggests that gap junctional coupling through Cx40 contributes to the function of the juxtaglomerular apparatus. In the present experiments, we determined the effect of targeted Cx40 deletion in C57BL/6 and FVB mice on TGF responsiveness. In C57BL/6 mice, stop-flow pressure (PSF) fell from 40.3 ± 2 to 34.5 ± 2 mmHg in wild-type (WT) and from 31 ± 1.06 to 26.6 ± 0.98 mmHg in Cx40-/- mice. PSF changes of 5.85 ± 0.67 mmHg in WT and of 4.3 ± 0.55 mmHg in Cx40-/- mice were not significantly different (P = 0.08). In FVB mice, PSF fell from 37.4 ± 1.5 to 31.6 ± 1.5 mmHg in WT and from 28.1 ± 1.6 to 25.4 ± 1.7 mmHg in Cx40-/-, with mean TGF responses being significantly greater in WT than Cx40-/- (5.5 ± 0.55 vs. 2.7 ± 0.84 mmHg; P = 0.002). In both genetic backgrounds, PSF values were significantly lower in Cx40-/- than WT mice at all flow rates. Arterial blood pressure in the animals prepared for micropuncture was not different between WT and Cx40-/- mice. We conclude that the TGF response magnitude in superficial cortical nephrons is reduced by 30-50% in mice without Cx40, but that with the exception of a small number of nephrons, residual TGF activity is maintained. Thus gap junctional coupling appears to modulate TGF, perhaps by determining the kinetics of signal transmission.


Subject(s)
Connexins/deficiency , Feedback, Physiological/physiology , Kidney Glomerulus/physiology , Kidney Tubules/physiology , Animals , Connexins/genetics , Connexins/physiology , Gap Junctions/physiology , Kidney Glomerulus/cytology , Kidney Tubules/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Punctures , Renin/physiology , Signal Transduction/physiology , Gap Junction alpha-5 Protein
15.
Am J Physiol Renal Physiol ; 304(4): F382-9, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23269643

ABSTRACT

A(1) adenosine receptors (A1AR) are required for the modulation of afferent arteriolar tone by changes in luminal NaCl concentration implying that extracellular adenosine concentrations need to change in synchrony with NaCl. The present experiments were performed in mice with a null mutation in the gene for the major equilibrative nucleoside transporter ENT1 to test whether interference with adenosine disposition by cellular uptake of adenosine may modify TGF characteristics. Responses of stop flow pressure (P(SF)) to maximum flow stimulation were measured in mice with either C57Bl/6 or SWR/J genetic backgrounds. Maximum flow stimulation reduced P(SF) in ENT1(-/-) compared with wild-type (WT) mice by 1.6 ± 0.4 mmHg (n = 28) and 5.8 ± 1.1 mmHg (n = 17; P < 0.001) in C57Bl/6 and by 1.4 ± 0.4 mmHg (n = 15) and 9 ± 1.5 mmHg (n = 9; P < 0.001) in SWR/J. Plasma concentrations of adenosine and inosine were markedly higher in ENT1(-/-) than WT mice (ado: 1,179 ± 78 and 225 ± 48 pmol/ml; ino: 179 ± 24 and 47.5 ± 9 pmol/ml). Renal mRNA expressions of the four adenosine receptors, ENT2, and adenosine deaminase were not significantly different between WT and ENT1(-/-) mice. No significant differences of glomerular filtration rate or mean arterial blood pressure were found while plasma renin concentration, and heart rates were significantly lower in ENT1(-/-) animals. In conclusion, TGF responsiveness is significantly attenuated in the absence of ENT1, pointing to a role of nucleoside transport in the NaCl-synchronous changes of extracellular adenosine levels in the juxtaglomerular apparatus interstitium.


Subject(s)
Equilibrative Nucleoside Transporter 1/physiology , Gene Deletion , Kidney Tubules/physiology , Adenosine/blood , Adenosine Deaminase/biosynthesis , Animals , Arterial Pressure/genetics , Equilibrative Nucleoside Transporter 1/genetics , Equilibrative-Nucleoside Transporter 2/biosynthesis , Female , Glomerular Filtration Rate/genetics , Heart Rate/genetics , Inosine/blood , Kidney Glomerulus/physiology , Male , Mice , Mice, Inbred C57BL , Receptors, Purinergic P1/biosynthesis , Renin/blood
16.
Pflugers Arch ; 465(1): 39-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22665048

ABSTRACT

The intratubular composition of fluid at the tubulovascular contact site of the juxtaglomerular apparatus serves as regulatory input for secretion and synthesis of renin. Experimental evidence, mostly from in vitro perfused preparations, indicates an inverse relation between luminal NaCl concentration and renin secretion. The cellular transduction mechanism is initiated by concentration-dependent NaCl uptake through the Na-K-2Cl cotransporter (NKCC2) with activation of NKCC2 causing inhibition and deactivation of NKCC2 causing stimulation of renin release. Changes in NKCC2 activity are coupled to alterations in the generation of paracrine factors that interact with granular cells. Among these factors, generation of PGE2 in a COX-2-dependent fashion appears to play a dominant role in the stimulatory arm of tubular control of renin release. [NaCl] is a determinant of local PG release over an appropriate concentration range, and blockade of COX-2 activity interferes with the NaCl dependency of renin secretion. The complex array of local paracrine controls also includes nNOS-mediated synthesis of nitric oxide, with NO playing the role of a modifier of the intracellular signaling pathway. A role of adenosine may be particularly important when [NaCl] is increased, and at least some of the available evidence is consistent with an important suppressive effect of adenosine at higher salt concentrations.


Subject(s)
Kidney Tubules/metabolism , Renin/biosynthesis , Animals , Cyclooxygenase 2/metabolism , Humans , Juxtaglomerular Apparatus/metabolism , Kidney Tubules/anatomy & histology , Kidney Tubules/physiology , Prostaglandins/metabolism , Renin/metabolism , Renin-Angiotensin System , Sodium-Potassium-Chloride Symporters/metabolism
17.
Ultrasonics ; 53(3): 727-38, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23218908

ABSTRACT

Ultrasound and Duplex ultrasonography in particular are routinely used to diagnose cardiovascular disease (CVD), which is the leading cause of morbidity and mortality worldwide. However, these techniques may not be able to characterize vascular tissue compositional changes due to CVD. This work describes an ultrasound-based hybrid imaging technique that can be used for vascular tissue characterization and the diagnosis of atherosclerosis. Ultrasound radiofrequency (RF) data were acquired and processed in time, frequency, and wavelet domains to extract six parameters including time integrated backscatter (T(IB)), time variance (T(var)), time entropy (T(E)), frequency integrated backscatter (F(IB)), wavelet root mean square value (W(rms)), and wavelet integrated backscatter (W(IB)). Each parameter was used to reconstruct an image co-registered to morphological B-scan. The combined set of hybrid images were used to characterize vascular tissue in vitro and in vivo using three mouse models including control (C57BL/6), and atherosclerotic apolipoprotein E-knockout (APOE-KO) and APOE/A(1) adenosine receptor double knockout (DKO) mice. The technique was tested using high-frequency ultrasound including single-element (center frequency=55 MHz) and commercial array (center frequency=40 MHz) systems providing superior spatial resolutions of 24 µm and 40 µm, respectively. Atherosclerotic vascular lesions in the APOE-KO mouse exhibited the highest values (contrast) of -10.11±1.92 dB, -12.13±2.13 dB, -7.54±1.45 dB, -5.10±1.06 dB, -5.25±0.94 dB, and -10.23±2.12 dB in T(IB), T(var), T(E), F(IB), W(rms), W(IB) hybrid images (n=10, p<0.05), respectively. Control segments of normal vascular tissue showed the lowest values of -20.20±2.71 dB, -22.54±4.54 dB, -14.94±2.05 dB, -9.64±1.34 dB, -10.20±1.27 dB, and -19.36±3.24 dB in same hybrid images (n=6, p<0.05). Results from both histology and optical images showed good agreement with ultrasound findings within a maximum error of 3.6% in lesion estimation. This study demonstrated the feasibility of a high-resolution hybrid imaging technique to diagnose atherosclerosis and characterize plaque components in mouse. In the future, it can be easily implemented on commercial ultrasound systems and eventually translated into clinics as a screening tool for atherosclerosis and the assessment of vulnerable plaques.


Subject(s)
Aortic Diseases/diagnostic imaging , Atherosclerosis/diagnostic imaging , Ultrasonography, Doppler/instrumentation , Animals , Aortic Diseases/pathology , Atherosclerosis/pathology , Disease Models, Animal , Image Processing, Computer-Assisted , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Processing, Computer-Assisted , Transducers
18.
Transplantation ; 94(9): 894-902, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23073466

ABSTRACT

BACKGROUND: Liver ischemia-reperfusion injury (IRI) is a known risk factor for the postoperative outcome of patients undergoing liver surgery/transplantation. Attempts to protect from organ damage require multidisciplinary strategies and are of emerging interest in view of patients with higher age and American Society of Anesthesiology status. Ischemic preconditioning has been successfully applied to prevent from IRI during liver resection/transplantation. Because even short periods of ischemia during preconditioning inevitably lead to hypoxia and formation of anti-inflammatory/cytoprotective acting adenosine, we reasoned that short nonischemic hypoxia also protects against hepatic IRI. METHODS: Mice underwent hypoxic preconditioning (HPC) by breathing 10% oxygen for 10 min followed by 10 min of 21% oxygen before left liver lobe ischemia (45 min) and reperfusion (4 hr). The interactions of hypoxia→adenosine→adenosine receptors were tested by pharmacologic antagonism at adenosine receptor (AR) sites in wild-type mice and in mice with genetic deletions at the A1, A2A, A2B, and A3 ARs. Hepatocellular damage, inflammation, and metabolic effects were quantified by enzyme activities, cytokines, liver myeloperoxidase, blood adenosine, and tissue AMP, respectively. RESULTS: Hepatoprotection by HPC was significant in wild-type and A1, A2A, and A3 AR knockout mice as quantified by lower alanine aminotransferase serum activities, cytokine levels, histologic damage scores, tissue myeloperoxidase concentrations, and preserved AMP concentrations. Protection by HPC was blunted in mice pretreated with the A2B AR antagonist MRS1754 or in A2B AR knockout mice. CONCLUSIONS: Because liver protective effects of HPC are negated when the A2B receptor is nonfunctional, the hypoxia→adenosine→A2B receptor pathway plays a critical role in the prevention of warm IRI in vivo. Hypoxic activation of this pathway warrants use of selective A2B AR agonists or even intermittent hypoxia (e.g., in deceased organ donors) to protect from liver IRI.


Subject(s)
Hypoxia/physiopathology , Ischemic Preconditioning , Liver/blood supply , Receptor, Adenosine A2B/physiology , Reperfusion Injury/prevention & control , Warm Ischemia , Acetamides/pharmacology , Adenosine/physiology , Animals , Hepatocytes/pathology , Hepatocytes/physiology , Liver/pathology , Liver/physiopathology , Liver Transplantation/pathology , Liver Transplantation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Purines/pharmacology , Receptor, Adenosine A2B/deficiency , Receptor, Adenosine A2B/drug effects , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction/physiology
19.
Am J Physiol Renal Physiol ; 303(8): F1166-75, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22896040

ABSTRACT

Adenosine 1 receptors (A1AR) have been shown in previous experiments to play a major role in the tubuloglomerular feedback (TGF) constrictor response of afferent arterioles (AA) to increased loop of Henle flow. Overexpression studies have pointed to a critical role of vascular A1AR, but it has remained unclear whether selective deletion of A1AR from smooth muscle cells is sufficient to abolish TGF responsiveness. To address this question, we have determined TGF response magnitude in mice in which vascular A1AR deletion was achieved using the loxP recombination approach with cre recombinase being controlled by a smooth muscle actin promoter (SmCre/A1ARff). Effective vascular deletion of A1AR was affirmed by absence of vasoconstrictor responses to adenosine or cyclohexyl adenosine (CHA) in microperfused AA. Elevation of loop of Henle flow from 0 to 30 nl/min caused a 22.1 ± 3.1% reduction of stop flow pressure in control mice and of 7.2 ± 1.5% in SmCre/A1ARff mice (P < 0.001). Maintenance of residual TGF activity despite absence of A1AR-mediated responses in AA suggests participation of extravascular A1AR in TGF. Support for this notion comes from the observation that deletion of A1ARff by nestin-driven cre causes an identical TGF response reduction (7.3 ± 2.4% in NestinCre/A1ARff vs. 20.3 ± 2.7% in controls), whereas AA responsiveness was reduced but not abolished. A1AR on AA smooth muscle cells are primarily responsible for TGF activation, but A1AR on extravascular cells, perhaps mesangial cells, appear to contribute to the TGF response.


Subject(s)
Arterioles/physiology , Blood Pressure/physiology , Glomerular Filtration Rate/physiology , Kidney/physiology , Receptor, Adenosine A1/genetics , Adenosine/pharmacology , Animals , Arterioles/drug effects , Blood Pressure/drug effects , Glomerular Filtration Rate/drug effects , Heart Rate/drug effects , Heart Rate/physiology , Kidney/blood supply , Kidney/drug effects , Mice , Mice, Transgenic , Receptor, Adenosine A1/metabolism , Vasoconstriction/drug effects , Vasoconstriction/physiology
20.
Proc Natl Acad Sci U S A ; 109(16): 6265-70, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22421436

ABSTRACT

Adenosine is a potent anticonvulsant acting on excitatory synapses through A1 receptors. Cellular release of ATP, and its subsequent extracellular enzymatic degradation to adenosine, could provide a powerful mechanism for astrocytes to control the activity of neural networks during high-intensity activity. Despite adenosine's importance, the cellular source of adenosine remains unclear. We report here that multiple enzymes degrade extracellular ATP in brain tissue, whereas only Nt5e degrades AMP to adenosine. However, endogenous A1 receptor activation during cortical seizures in vivo or heterosynaptic depression in situ is independent of Nt5e activity, and activation of astrocytic ATP release via Ca(2+) photolysis does not trigger synaptic depression. In contrast, selective activation of postsynaptic CA1 neurons leads to release of adenosine and synaptic depression. This study shows that adenosine-mediated synaptic depression is not a consequence of astrocytic ATP release, but is instead an autonomic feedback mechanism that suppresses excitatory transmission during prolonged activity.


Subject(s)
Adenosine/metabolism , Excitatory Postsynaptic Potentials/physiology , Feedback, Physiological/physiology , Neurons/metabolism , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Brain/physiopathology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Long-Term Synaptic Depression/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Receptor, Adenosine A1/metabolism , Seizures/metabolism , Seizures/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...