48 results on '"Unwin RJ"'
Search Results
2. Purinergic signalling in the kidney: In physiology and disease.
- Author
-
Monaghan MT, Bailey MA, and Unwin RJ
- Subjects
- Animals, Humans, Kidney drug effects, Kidney Diseases drug therapy, Purinergic Agonists administration & dosage, Purinergic Antagonists administration & dosage, Signal Transduction drug effects, Adenosine Triphosphate metabolism, Kidney metabolism, Kidney Diseases metabolism, Receptors, Purinergic metabolism, Signal Transduction physiology
- Abstract
Historically, the control of renal vascular and tubular function has, for the most part, concentrated on neural and endocrine regulation. However, in addition to these extrinsic factors, it is now appreciated that several complex humoral control systems exist within the kidney that can act in an autocrine and/or paracrine fashion. These paracrine systems complement neuroendocrine regulation by dynamically fine-tuning renal vascular and tubular function to buffer rapid changes in nephron perfusion and flow rate of tubular fluid. One of the most pervasive is the extracellular nucleotide/P2 receptor system, which is central to many of the intrinsic regulatory feedback loops within the kidney such as renal haemodynamic autoregulation and tubuloglomerular feedback (TGF). Although physiological actions of extracellular adenine nucleotides were reported almost 100 years ago, the conceptual framework for purinergic regulation of renal function owes much to the work of Geoffrey Burnstock. In this review, we reflect on our >20-year collaboration with Professor Burnstock and highlight the research that is still unlocking the potential of the renal purinergic system to understand and treat kidney disease., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
3. Acute saccharin infusion has no effect on renal glucose handling in normal rats in vivo.
- Author
-
Jacquillet G, Debnam ES, Unwin RJ, and Marks J
- Subjects
- Animals, Kidney metabolism, Male, Rats, Rats, Wistar, Saccharin administration & dosage, Saccharin adverse effects, Sodium-Glucose Transporter 1 metabolism, Sodium-Glucose Transporter 2 metabolism, Sweetening Agents administration & dosage, Sweetening Agents adverse effects, Glucose metabolism, Kidney drug effects, Renal Reabsorption, Saccharin pharmacology, Sweetening Agents pharmacology
- Abstract
Artificial sweeteners are extensively used by the food industry to replace sugar in food and beverages and are widely considered to be a healthy alternative. However, recent data suggest that artificial sweeteners may impact intestinal glucose absorption and that they might lead to glucose intolerance. Moreover, chronic consumption of artificial sweeteners has also been linked to detrimental changes in renal function. Using an in vivo approach, our study aimed to determine if short-term infusion of the artificial sweetener saccharin can alter renal function and renal glucose absorption. We show that saccharin infusion does not induce any major change in GFR or urine flow rate at either the whole kidney or single nephron level, suggesting that any reported change in renal function with artificial sweeteners must depend on chronic consumption. As expected for a nondiabetic animal, glucose excretion was low; however, saccharin infusion caused a small, but significant, decrease in fractional glucose excretion. In contrast to the whole kidney data, our micropuncture results did not show any significant difference in fractional glucose reabsorption in either the proximal or distal tubules, indicating that saccharin does not influence renal glucose handling in vivo under euglycemic conditions. In keeping with this finding, protein levels of the renal glucose transporters SGLT1 and SGLT2 were also unchanged. In addition, saccharin infusion in rats undergoing a glucose tolerance test failed to induce a robust change in renal glucose excretion or renal glucose transporter expression. In conclusion, our results demonstrate that saccharin does not induce acute physiologically relevant changes in renal function or renal glucose handling., (© 2018 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of The Physiological Society and the American Physiological Society.)
- Published
- 2018
- Full Text
- View/download PDF
4. Hyperglycemia-induced Renal P2X7 Receptor Activation Enhances Diabetes-related Injury.
- Author
-
Menzies RI, Booth JWR, Mullins JJ, Bailey MA, Tam FWK, Norman JT, and Unwin RJ
- Subjects
- Adenosine Triphosphate analogs & derivatives, Adenosine Triphosphate pharmacology, Animals, Cells, Cultured, Chemokine CCL2 metabolism, Diabetic Nephropathies pathology, Humans, Kidney pathology, Macrophages drug effects, Macrophages metabolism, Male, Mesangial Cells metabolism, Mice, Inbred C57BL, Mice, Knockout, Purinergic P2X Receptor Antagonists pharmacology, Pyridines pharmacology, Rats, Wistar, Receptors, Purinergic P2X7 genetics, Tetrazoles pharmacology, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Type 1 metabolism, Diabetic Nephropathies metabolism, Hyperglycemia metabolism, Kidney metabolism, Receptors, Purinergic P2X7 metabolism
- Abstract
Diabetes is a leading cause of renal disease. Glomerular mesangial expansion and fibrosis are hallmarks of diabetic nephropathy and this is thought to be promoted by infiltration of circulating macrophages. Monocyte chemoattractant protein-1 (MCP-1) has been shown to attract macrophages in kidney diseases. P2X7 receptors (P2X7R) are highly expressed on macrophages and are essential components of pro-inflammatory signaling in multiple tissues. Here we show that in diabetic patients, renal P2X7R expression is associated with severe mesangial expansion, impaired glomerular filtration (≤40ml/min/1.73sq.m.), and increased interstitial fibrosis. P2X7R activation enhanced the release of MCP-1 in human mesangial cells cultured under high glucose conditions. In mice, P2X7R-deficiency prevented glomerular macrophage attraction and collagen IV deposition; however, the more severe interstitial inflammation and fibrosis often seen in human diabetic kidney diseases was not modelled. Finally, we demonstrate that a P2X7R inhibitor (AZ11657312) can reduce renal macrophage accrual following the establishment of hyperglycemia in a model of diabetic nephropathy. Collectively these data suggest that P2X7R activation may contribute to the high prevalence of kidney disease found in diabetics., (Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
5. Postprandial adjustments in renal phosphate excretion do not involve a gut-derived phosphaturic factor.
- Author
-
Lee GJ, Mossa-Al Hashimi L, Debnam ES, Unwin RJ, and Marks J
- Subjects
- Animals, Biological Transport physiology, Homeostasis physiology, Male, Parathyroid Hormone metabolism, Rats, Rats, Sprague-Dawley, Symporters metabolism, Intestinal Mucosa metabolism, Kidney metabolism, Kidney physiology, Phosphates metabolism, Postprandial Period physiology, Sodium-Phosphate Cotransporter Proteins, Type IIa metabolism
- Abstract
New Findings: What is the central question of this study? Does a previously hypothesized signalling mechanism, believed to detect postprandial increases in intestinal phosphate and that can stimulate the kidneys to rapidly excrete phosphate, operate under physiological conditions? What is the main finding and its importance? Contrary to earlier reports, rapid signalling between the small intestine and kidney mediated by a gut-derived phosphaturic factor in response to a physiological intestinal phosphate load is not supported by the present findings; moreover, hyperphosphataemia and increased parathyroid hormone concentrations are likely to be the underlying factors responsible for the phosphaturia following a supraphysiological intestinal phosphate load. To date, the role of the small intestine in the regulation of postprandial phosphate homeostasis has remained unclear and controversial. Previous studies have proposed the presence of a gut-derived phosphaturic factor that acts independently of changes in plasma phosphate concentration or parathyroid hormone (PTH) concentration; however, these early studies used duodenal luminal phosphate concentrations in the molar range, and therefore, the physiological relevance of this is uncertain. In the present study, we used both in vivo and in vitro approaches to investigate the presence of this putative 'intestinal phosphatonin'. Instillation of 1.3 m phosphate into the duodenum rapidly induced phosphaturia, but in contrast to previous reports, this was associated with significant hyperphosphataemia and elevated PTH concentration; however, there was not the expected decrease in abundance of the renal sodium-phosphate cotransporter NaPi-IIa. Instillation of a physiological (10 mm) phosphate load had no effect on plasma phosphate concentration, PTH concentration or phosphate excretion. Moreover, phosphate uptake by opossum kidney cells was unaffected after incubation with serosal fluid collected from intestinal segments perfused with different concentrations of phosphate. Taken together, these findings do not support the concept of a gut-derived phosphaturic factor that can mediate rapid signalling between the gut and kidney, leading to increased urinary phosphate excretion, as part of normal phosphate homeostasis., (© 2017 The Authors. Experimental Physiology © 2017 The Physiological Society.)
- Published
- 2017
- Full Text
- View/download PDF
6. Purinergic signaling in kidney disease.
- Author
-
Menzies RI, Tam FW, Unwin RJ, and Bailey MA
- Subjects
- Animals, Humans, Kidney drug effects, Kidney pathology, Kidney physiopathology, Kidney Diseases drug therapy, Kidney Diseases pathology, Kidney Diseases physiopathology, Ligands, Purinergic P2X Receptor Antagonists therapeutic use, Receptors, Purinergic P1 drug effects, Receptors, Purinergic P2 drug effects, Receptors, Purinergic P2X7 drug effects, Receptors, Purinergic P2X7 metabolism, Adenosine metabolism, Kidney metabolism, Kidney Diseases metabolism, Purine Nucleotides metabolism, Receptors, Purinergic P1 metabolism, Receptors, Purinergic P2 metabolism, Signal Transduction drug effects
- Abstract
Nucleotides are key subunits for nucleic acids and provide energy for intracellular metabolism. They can also be released from cells to act physiologically as extracellular messengers or pathologically as danger signals. Extracellular nucleotides stimulate membrane receptors in the P2 and P1 family. P2X are ATP-activated cation channels; P2Y and P1 are G-protein coupled receptors activated by ATP, ADP, UTP, and UDP in the case of P2 or adenosine for P1. Renal P2 receptors influence both vascular contractility and tubular function. Renal cells also express ectonucleotidases that rapidly hydrolyze extracellular nucleotides. These enzymes integrate this multireceptor purinergic-signaling complex by determining the nucleotide milieu to titrate receptor activation. Purinergic signaling also regulates immune cell function by modulating the synthesis and release of various cytokines such as IL1-β and IL-18 as part of inflammasome activation. Abnormal or excessive stimulation of this intricate paracrine system can be pro- or anti-inflammatory, and is also linked to necrosis and apoptosis. Kidney tissue injury causes a localized increase in ATP concentration, and sustained activation of P2 receptors can lead to renal glomerular, tubular, and vascular cell damage. Purinergic receptors also regulate the activity and proliferation of fibroblasts, promoting both inflammation and fibrosis in chronic disease. In this short review we summarize some of the recent findings related to purinergic signaling in the kidney. We focus predominantly on the P2X7 receptor, discussing why antagonists have so far disappointed in clinical trials and how advances in our understanding of purinergic signaling might help to reposition these compounds as potential treatments for renal disease., (Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
7. Renal P2 receptors and hypertension.
- Author
-
Menzies RI, Unwin RJ, and Bailey MA
- Subjects
- Animals, Biological Transport physiology, Humans, Blood Pressure physiology, Hypertension metabolism, Kidney metabolism, Receptors, Purinergic P2 metabolism, Signal Transduction physiology
- Abstract
The regulation of extracellular fluid volume is a key component of blood pressure homeostasis. Long-term blood pressure is stabilized by the acute pressure natriuresis response by which changes in renal perfusion pressure evoke corresponding changes in renal sodium excretion. A wealth of experimental evidence suggests that a defect in the pressure natriuresis response contributes to the development and maintenance of hypertension. The mechanisms underlying the relationship between renal perfusion pressure and sodium excretion are incompletely understood. Increased blood flow through the vasa recta increases renal interstitial hydrostatic pressure, thereby reducing the driving force for transepithelial sodium reabsorption. Paracrine signalling also contributes to the overall natriuretic response by inhibiting tubular sodium reabsorption in several nephron segments. In this brief review, we discuss the role of purinergic signalling in the renal control of blood pressure. ATP is released from renal tubule and vascular cells in response to increased flow and can activate P2 receptor subtypes expressed in both epithelial and vascular endothelial/smooth muscle cells. In concert, these effects integrate the vascular and tubular responses to increased perfusion pressure and targeting P2 receptors, particularly P2X7, may prove beneficial for treatment of hypertension., (© 2014 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
8. Exaggerated renal fibrosis in P2X4 receptor-deficient mice following unilateral ureteric obstruction.
- Author
-
Kim MJ, Turner CM, Hewitt R, Smith J, Bhangal G, Pusey CD, Unwin RJ, and Tam FW
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Collagen Type I metabolism, Connective Tissue Growth Factor genetics, Disease Models, Animal, Fibrosis pathology, Immunoenzyme Techniques, Kidney metabolism, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Nephritis, Interstitial genetics, Nephritis, Interstitial metabolism, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Transforming Growth Factor beta genetics, Ureteral Obstruction genetics, Ureteral Obstruction metabolism, Kidney pathology, Nephritis, Interstitial physiopathology, Receptors, Purinergic P2X4 physiology, Ureteral Obstruction physiopathology
- Abstract
Background: The ATP-sensitive P2X7 receptor (P2X7R) has been shown to contribute to renal injury in nephrotoxic nephritis, a rodent model of acute glomerulonephritis, and in unilateral ureteric obstruction (UUO), a rodent model of chronic interstitial inflammation and fibrosis. Renal tubular cells, endothelial cells and macrophages also express the closely related P2X4 receptor (P2X4R), which is chromosomally co-located with P2X7R and has 40% homology; it is also pro-inflammatory and has been shown to interact with P2X7R to modulate its pro-apoptotic and pro-inflammatory effects. Therefore, we chose to explore the function of P2X4R in the UUO model of renal injury using knockout mice. We hypothesized that UUO-induced tubulointerstitial damage and fibrosis would also be attenuated in P2X4R(-/-) mice., Method: P2X4R(-/-) and wild-type (WT) mice were subjected to either UUO or sham operation. Kidney samples taken on Days 7 and 14 were evaluated for renal inflammation and fibrosis, and expression of pro-fibrotic factors., Results: To our surprise, the obstructed kidney in P2X4R(-/-) mice showed more severe renal injury, more collagen deposition (picrosirius red staining, increase of 53%; P < 0.05) and more type I collagen staining (increase of 107%; P < 0.01), as well as increased mRNA for TGF-β (increase of 102%, P < 0.0005) and CTGF (increase of 157%; P < 0.05) by Day 14, compared with the UUO WT mice., Conclusion: These findings showed that lack of P2X4R expression leads to increased renal fibrosis, and increased expression of TGF-β and CTGF in the UUO model., (© The Author 2014. Published by Oxford University Press on behalf of ERA-EDTA.)
- Published
- 2014
- Full Text
- View/download PDF
9. P2 purinoceptors: Renal pathophysiology and therapeutic potential.
- Author
-
Booth JW, Tam FW, and Unwin RJ
- Subjects
- Glomerulonephritis drug therapy, Glomerulonephritis etiology, Glomerulonephritis physiopathology, Humans, Kidney Diseases etiology, Polycystic Kidney Diseases drug therapy, Polycystic Kidney Diseases etiology, Polycystic Kidney Diseases physiopathology, Kidney physiopathology, Kidney Diseases drug therapy, Kidney Diseases physiopathology, Purinergic P2 Receptor Antagonists therapeutic use, Receptors, Purinergic P2 physiology
- Abstract
P2 purinoceptors, categorized into P2X and P2Y receptors, bind extracellular ATP and related di- and tri-phosphate nucleotides and are expressed throughout the kidney. P2X receptors are non-selective cation channels and P2Y receptors are metabotropic G protein-coupled receptors. Both families may couple to a range of second messenger systems and provoke outcomes including cell proliferation, cytokine secretion, membrane channel regulation and cell death. The cellular response to ATP release may vary widely and depends on both the pattern of local receptor expression and the action of ectonucleotidases altering agonist availability, creating a finely tuned network. P2 signaling participates in disparate physiological processes, including control of water and solute transport and autoregulation of renal blood flow. Given the ubiquity, complexity and diversity of the P2 network, it is not surprising that P2 signaling also contributes to mechanisms of renal disease. This review summarizes the current evidence for P2 receptor involvement in a range of kidney diseases, and highlights areas that may lead to potential therapeutic advances. Particular attention is paid to the pro-inflammatory P2X7 receptor, currently at the heart of renal P2 pathophysiology and for which selective receptor antagonists are now available.
- Published
- 2012
- Full Text
- View/download PDF
10. The calcineurin inhibitor tacrolimus activates the renal sodium chloride cotransporter to cause hypertension.
- Author
-
Hoorn EJ, Walsh SB, McCormick JA, Fürstenberg A, Yang CL, Roeschel T, Paliege A, Howie AJ, Conley J, Bachmann S, Unwin RJ, and Ellison DH
- Subjects
- Analysis of Variance, Animals, Bendroflumethiazide, Calcium urine, Cell Line, Chlorides urine, Creatinine urine, Enzyme-Linked Immunosorbent Assay, Fluorescent Antibody Technique, Humans, Hydrochlorothiazide, Immunoblotting, Immunohistochemistry, Mice, Mice, Knockout, Sodium Chloride Symporters genetics, Tacrolimus pharmacology, Calcineurin Inhibitors, Hypertension chemically induced, Kidney metabolism, Sodium Chloride Symporters metabolism, Tacrolimus adverse effects
- Abstract
Calcineurin inhibitors (CNIs) are immunosuppressive drugs that are used widely to prevent rejection of transplanted organs and to treat autoimmune disease. Hypertension and renal tubule dysfunction, including hyperkalemia, hypercalciuria and acidosis, often complicate their use. These side effects resemble familial hyperkalemic hypertension, a genetic disease characterized by overactivity of the renal sodium chloride cotransporter (NCC) and caused by mutations in genes encoding WNK kinases. We hypothesized that CNIs induce hypertension by stimulating NCC. In wild-type mice, the CNI tacrolimus caused salt-sensitive hypertension and increased the abundance of phosphorylated NCC and the NCC-regulatory kinases WNK3, WNK4 and SPAK. We demonstrated the functional importance of NCC in this response by showing that tacrolimus did not affect blood pressure in NCC-knockout mice, whereas the hypertensive response to tacrolimus was exaggerated in mice overexpressing NCC. Moreover, hydrochlorothiazide, an NCC-blocking drug, reversed tacrolimus-induced hypertension. These observations were extended to humans by showing that kidney transplant recipients treated with tacrolimus had a greater fractional chloride excretion in response to bendroflumethiazide, another NCC-blocking drug, than individuals not treated with tacrolimus; renal NCC abundance was also greater. Together, these findings indicate that tacrolimus-induced chronic hypertension is mediated largely by NCC activation, and suggest that inexpensive and well-tolerated thiazide diuretics may be especially effective in preventing the complications of CNI treatment.
- Published
- 2011
- Full Text
- View/download PDF
11. Multiphoton imaging of the functioning kidney.
- Author
-
Hall AM, Crawford C, Unwin RJ, Duchen MR, and Peppiatt-Wildman CM
- Subjects
- Animals, Fluorescent Dyes, In Vitro Techniques, Ischemia physiopathology, Kidney physiology, Male, Mitochondria physiology, NAD metabolism, Rats, Rats, Sprague-Dawley, Reactive Oxygen Species metabolism, Reperfusion, Kidney anatomy & histology, Microscopy, Fluorescence, Multiphoton
- Abstract
Translating discoveries made in isolated renal cells and tubules to the in vivo situation requires the assessment of cellular function in intact live organs. Multiphoton imaging is a form of fluorescence microscopy that is ideally suited to working with whole tissues and organs, but adequately loading cells with fluorescence dyes in vivo remains a challenge. We found that recirculation of fluorescence dyes in the rat isolated perfused kidney (IPK) resulted in levels of intracellular loading that would be difficult to achieve in vivo. This technique allowed the imaging of tubular cell structure and function with multiphoton microscopy in an intact, functioning organ. We used this approach to follow processes in real time, including (1) relative rates of reactive oxygen species (ROS) production in different tubule types, (2) filtration and tubular uptake of low-molecular-weight dextrans and proteins, and (3) the effects of ischemia-reperfusion injury on mitochondrial function and cell structure. This study demonstrates that multiphoton microscopy of the isolated perfused kidney is a powerful technique for detailed imaging of cell structure and function in an intact organ., (Copyright © 2011 by the American Society of Nephrology)
- Published
- 2011
- Full Text
- View/download PDF
12. Pathophysiology and management of hypokalemia: a clinical perspective.
- Author
-
Unwin RJ, Luft FC, and Shirley DG
- Subjects
- Humans, Hypokalemia physiopathology, Hypokalemia therapy, Kidney physiology, Water-Electrolyte Balance physiology
- Abstract
Potassium (K(+)) ions are the predominant intracellular cations. K(+) homeostasis depends on external balance (dietary intake [typically 100 mmol per day] versus excretion [95% via the kidney; 5% via the colon]) and internal balance (the distribution of K(+) between intracellular and extracellular fluid compartments). The uneven distribution of K(+) across cell membranes means that a mere 1% shift in its distribution can cause a 50% change in plasma K(+) concentration. Hormonal mechanisms (involving insulin, β-adrenergic agonists and aldosterone) modulate K(+) distribution by promoting rapid transfer of K(+) across the plasma membrane. Extrarenal K(+) losses from the body are usually small, but can be marked in individuals with chronic diarrhea, severe burns or prolonged sweating. Under normal circumstances, the kidney's distal nephron secretes K(+) and determines final urinary excretion. In patients with hypokalemia (plasma K(+) concentration <3.5 mmol/l), after the exclusion of extrarenal causes, alterations in sodium ion delivery to the distal nephron, mineralocorticoid status, or a specific inherited or acquired defect in distal nephron function (each of which affects distal nephron K(+) secretion), should be considered. Clinical management of hypokalemia should establish the underlying cause and alleviate the primary disorder. This Review aims to inform clinicians about the pathophysiology and appropriate treatment for hypokalemia.
- Published
- 2011
- Full Text
- View/download PDF
13. Renal stone disease: a commentary on the nature and significance of Randall's plaque.
- Author
-
Evan AP, Unwin RJ, and Williams JC Jr
- Subjects
- Crystallization, Disease Progression, Humans, Kidney pathology, Kidney Calculi diagnosis, Kidney Calculi etiology, Kidney Calculi therapy, Risk Factors, Calcium Oxalate metabolism, Calcium Phosphates metabolism, Kidney metabolism, Kidney Calculi metabolism
- Published
- 2011
- Full Text
- View/download PDF
14. Potential role of serine proteases in modulating renal sodium transport in vivo.
- Author
-
Jacquillet G, Rubera I, and Unwin RJ
- Subjects
- Animals, Biological Transport physiology, Fibrinolysin urine, Humans, Models, Biological, Nephrotic Syndrome metabolism, Nephrotic Syndrome physiopathology, Nephrotic Syndrome urine, Plasminogen, Kidney metabolism, Nephrons metabolism, Serine Proteases metabolism, Sodium metabolism
- Abstract
The maintenance of sodium (Na+) homeostasis is an essential function of the kidney. It is achieved by a variety of transport processes localized all along the highly specialised segments of the nephron. Impairment of these transport mechanisms, and thereby Na+ handling, is associated with disturbed Na+ and water balance, leading to hypertension and oedema. This review focuses on the novel regulation of sodium reabsorption by serine proteases acting along the entire nephron., (Copyright © 2011 S. Karger AG, Basel.)
- Published
- 2011
- Full Text
- View/download PDF
15. Phosphate homeostasis and the renal-gastrointestinal axis.
- Author
-
Marks J, Debnam ES, and Unwin RJ
- Subjects
- Animals, Epithelial Cells metabolism, Homeostasis, Humans, Hyperphosphatemia etiology, Hyperphosphatemia metabolism, Hyperphosphatemia prevention & control, Intestinal Mucosa metabolism, Kidney Failure, Chronic complications, Kidney Failure, Chronic metabolism, Phosphates blood, Intestinal Absorption, Intestine, Small metabolism, Kidney metabolism, Phosphates metabolism, Sodium-Phosphate Cotransporter Proteins metabolism
- Abstract
Transport of phosphate across intestinal and renal epithelia is essential for normal phosphate balance, yet we know less about the mechanisms and regulation of intestinal phosphate absorption than we do about phosphate handling by the kidney. Recent studies have provided strong evidence that the sodium-phosphate cotransporter NaPi-IIb is responsible for sodium-dependent phosphate absorption by the small intestine, and it might be that this protein can link changes in dietary phosphate to altered renal phosphate excretion to maintain phosphate balance. Evidence is also emerging that specific regions of the small intestine adapt differently to acute or chronic changes in dietary phosphate load and that phosphatonins inhibit both renal and intestinal phosphate transport. This review summarizes our current understanding of the mechanisms and control of intestinal phosphate absorption and how it may be related to renal phosphate reabsorption; it also considers the ways in which the gut could be targeted to prevent, or limit, hyperphosphatemia in chronic and end-stage renal failure.
- Published
- 2010
- Full Text
- View/download PDF
16. Regulation of renal function by the gastrointestinal tract: potential role of gut-derived peptides and hormones.
- Author
-
Michell AR, Debnam ES, and Unwin RJ
- Subjects
- Animals, Electrolytes metabolism, Humans, Gastrointestinal Hormones physiology, Gastrointestinal Tract physiology, Kidney physiology, Peptide Hormones physiology
- Abstract
The concept of a regulatory link between the gastrointestinal tract and kidneys is not new. The idea that dietary intake and composition can affect renal function is perhaps self-evident, but defining this relationship, especially in terms of sensors and effectors, is proving more difficult. That the gastrointestinal tract can exert some control over renal function was strengthened by the early observation that oral ingestion of a sodium chloride load has a greater natriuretic effect than when the same amount is given intravenously. This effect was subsequently shown to be independent of changes in aldosterone and atrial natriuretic peptide, although not necessarily angiotensin-II. However, the nature of any intestinal natriuretic peptide remains uncertain, despite suggestions that various gut-derived hormones, including guanylin and uroguanylin, may be involved. There is also an emerging concept of gastrointestinal taste and taste-like receptor mechanisms present throughout the gastrointestinal tract, which may govern the excretion of other key electrolytes, including potassium and phosphate. The evidence for gut sensors of nutrients such as proteins, amino acids, glucose, and acid is now becoming more established. Thus, we can anticipate the existence and eventual characterization of several gut ion sensors.
- Published
- 2008
- Full Text
- View/download PDF
17. Immunolocalization of ectonucleotidases along the rat nephron.
- Author
-
Vekaria RM, Shirley DG, Sévigny J, and Unwin RJ
- Subjects
- 5'-Nucleotidase immunology, Adenosine Triphosphatases immunology, Animals, Antigens, CD immunology, Apyrase immunology, COS Cells, Chlorocebus aethiops, Humans, Immunohistochemistry, Male, Nephrons metabolism, Phosphoric Diester Hydrolases immunology, Pyrophosphatases immunology, Rats, Rats, Sprague-Dawley, 5'-Nucleotidase metabolism, Adenosine Triphosphatases metabolism, Antigens, CD metabolism, Apyrase metabolism, Kidney enzymology, Phosphoric Diester Hydrolases metabolism, Pyrophosphatases metabolism
- Abstract
Evidence is accumulating that extracellular nucleotides act as autocrine/paracrine agents in most tissues, including the kidneys. Several families of surface-located enzymes, collectively known as ectonucleotidases, can degrade nucleotides. Using immunohistochemistry, we have examined the segmental distribution of five ectonucleotidases along the rat nephron. Perfusion-fixed kidneys were obtained from anesthetized male Sprague-Dawley rats. Cryostat sections of cortical and medullary regions were incubated with antibodies specific to the following enzymes: ectonucleoside triphosphate diphosphohydrolase (NTPDase) 1, NTPDase2, NTPDase3, ectonucleotide pyrophosphatase phosphodiesterase 3 (NPP3), and ecto-5'-nucleotidase. Sections were then costained with Phaseolus vulgaris erythroagglutinin (for identification of proximal tubules) and antibodies against Tamm-Horsfall protein (for identification of thick ascending limb), calbindin-D(28k) (for identification of distal tubule), and aquaporin-2 (for identification of collecting duct). The tyramide signal amplification method was used when the ectonucleotidase and marker antibody were raised in the same species. The glomerulus expressed NTPDase1 and NPP3. The proximal tubule showed prominent expression of NPP3 and ecto-5'-nucleotidase in most, but not all, segments. NTPDase2 and NTPDase3, but not NPP3 or ecto-5'-nucleotidase, were expressed in the thick ascending limb and distal tubule. NTPDase3, with some low-level expression of ecto-5'-nucleotidase, was also found in cortical and outer medullary collecting ducts. Inner medullary collecting ducts displayed low-level staining for NTPDase1, NTPDase2, NTPDase3, and ecto-5'-nucleotidase. We conclude that these ectonucleotidases are differentially expressed along the nephron and may play a key role in activation of purinoceptors by nucleotides and nucleosides.
- Published
- 2006
- Full Text
- View/download PDF
18. Renal tubular acidosis: developments in our understanding of the molecular basis.
- Author
-
Laing CM, Toye AM, Capasso G, and Unwin RJ
- Subjects
- Acidosis, Renal Tubular classification, Acidosis, Renal Tubular genetics, Adult, Animals, Anion Exchange Protein 1, Erythrocyte genetics, Anion Exchange Protein 1, Erythrocyte physiology, Carbonic Anhydrase II physiology, Child, Humans, Proton-Translocating ATPases physiology, Acidosis, Renal Tubular physiopathology, Kidney physiopathology
- Abstract
Renal tubular acidosis is a metabolic acidosis due to impaired acid excretion by the kidney. Hyperchloraemic acidosis with a normal anion gap and normal (or near normal) glomerular filtration rate, and in the absence of diarrhoea, defines this disorder. However, systemic acidosis is not always evident and renal tubular acidosis can present with hypokalaemia, medullary nephrocalcinosis and recurrent calcium phosphate stone disease, as well as growth retardation and rickets in children, or short stature and osteomalacia in adults. Renal dysfunction in renal tubular acidosis is not always confined to acid excretion and can be part of a more generalised renal tubule defect, as in the renal Fanconi syndrome. Isolated renal tubular acidosis is more usually acquired, due to drugs, autoimmune disease, post-obstructive uropathy or any cause of medullary nephrocalcinosis. Less commonly, it is inherited and may be associated with deafness, osteopetrosis or ocular abnormalities. The clinical classification of renal tubular acidosis has been correlated with our current physiological model of how the nephron excretes acid, and this has facilitated genetic studies that have identified mutations in several genes encoding acid and base ion transporters. In vitro functional studies of these mutant proteins in cell expression systems have helped to elucidate the molecular mechanisms underlying renal tubular acidosis, which ultimately may lead to new therapeutic options in what is still treatment only by giving an oral alkali.
- Published
- 2005
- Full Text
- View/download PDF
19. The P2X7 ATP receptor in the kidney: a matter of life or death?
- Author
-
Hillman KA, Burnstock G, and Unwin RJ
- Subjects
- Animals, Chronic Disease, Gene Expression Regulation, Developmental, Humans, Inflammation, Kidney growth & development, Kidney Glomerulus physiology, Mice, Polycystic Kidney Diseases physiopathology, Rats, Receptors, Purinergic P2X7, Signal Transduction, Up-Regulation, Kidney physiology, Kidney Diseases physiopathology, Receptors, Purinergic P2 biosynthesis, Receptors, Purinergic P2 physiology
- Abstract
P2X7 is an intriguing membrane receptor for the extracellular nucleotide ATP, which functions as a ligand-gated ion channel; it can activate cell membrane permeabilization and also has a wide range of downstream signaling pathways, including mediation of inflammatory responses and modulation of cell turnover. Despite recent identification of P2X7 receptor protein in the renal tract, the biological and potential pathological functions of this receptor and its signaling cascades in the kidney are not yet fully understood. P2X7 receptor protein is expressed in normal kidney development, predominantly in the condensing mesenchyme, and later in the maturing and adult derivatives of the ureteric bud. Glomerular expression of the molecule is scarce in normal kidney, but is upregulated in chronic and inflammatory conditions, suggesting a role in the inflammatory response or in repair and remodeling in these settings. P2X7 receptor expression in the adult collecting ducts of murine kidney, as well as the collecting duct cysts in autosomal recessive polycystic kidney disease, has been described and agonists of the receptor can modulate the development of renal cysts in an in vitro model of cyst formation derived from the cpk/cpk mouse. Further investigation of the function of the P2X7 receptor in normal and abnormal kidneys might lead to novel therapeutic targets in a wide range of renal diseases.
- Published
- 2005
- Full Text
- View/download PDF
20. Uric acid and the kidney: urate transport, stone disease and progressive renal failure.
- Author
-
Capasso G, Jaeger P, Robertson WG, and Unwin RJ
- Subjects
- Animals, Biological Transport, Carrier Proteins physiology, Humans, Hyperuricemia complications, Organic Anion Transport Protein 1 physiology, Organic Anion Transporters physiology, Organic Cation Transport Proteins, Kidney metabolism, Kidney Calculi etiology, Renal Insufficiency etiology, Uric Acid metabolism
- Abstract
In this brief review and update, we try to cover recent developments in our understanding of uric acid transport by the kidney, the contribution of uric acid to renal stone disease, its potential role in progressive renal failure and, most recently, the novel and as yet unexplained link between the urinary glycoprotein Tamm-Horsfall protein (uromodulin) and hyperuricaemia and two inherited forms of renal disease with chronic renal failure.
- Published
- 2005
- Full Text
- View/download PDF
21. An overview of divalent cation and citrate handling by the kidney.
- Author
-
Unwin RJ, Capasso G, and Shirley DG
- Subjects
- Humans, Cations, Divalent metabolism, Citrates metabolism, Kidney metabolism
- Abstract
Urinary calcium, magnesium and citrate levels are important in promoting or inhibiting renal stone formation. Here we review current information on the tubular handling of these ions. Most filtered calcium is reabsorbed in the proximal tubule and the thick ascending limb (TAL) of the loop of Henle, largely paracellularly; most of the remainder is reabsorbed in the distal tubule, transcellularly. Calcium reabsorption in the TAL and distal tubule is stimulated by parathyroid hormone and vitamin D; other factors influencing its renal handling include extracellular volume status and acid-base balance. Little filtered magnesium is reabsorbed in the proximal tubule; the bulk is reabsorbed paracellularly in the TAL, while most of the remainder is reabsorbed transcellularly in the distal tubule. Dietary intake, peptide hormones and chronic potassium depletion can all influence magnesium reabsorption in the TAL and distal tubule. Most filtered citrate is taken up across the apical membrane of the proximal tubule via a sodium-dicarboxylate co-transporter (NaDC-1). It also enters proximal tubular cells across the basolateral membrane; citrate contributes to the cells' oxidative metabolism. Citrate excretion is affected by acid-base balance, acetazolamide treatment, chronic potassium depletion and urinary excretion of calcium and magnesium. Where possible, we have indicated the mechanisms of these complex interactions., (Copyright (c) 2004 S. Karger AG, Basel.)
- Published
- 2004
- Full Text
- View/download PDF
22. The pattern of distribution of selected ATP-sensitive P2 receptor subtypes in normal rat kidney: an immunohistological study.
- Author
-
Turner CM, Vonend O, Chan C, Burnstock G, and Unwin RJ
- Subjects
- Animals, Cell Membrane metabolism, Epithelial Cells cytology, Epithelial Cells metabolism, Fibroblasts cytology, Fibroblasts metabolism, Glomerular Mesangium cytology, Glomerular Mesangium metabolism, Immunohistochemistry, Kidney cytology, Kidney Tubules, Proximal cytology, Kidney Tubules, Proximal metabolism, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, Rats, Rats, Sprague-Dawley, Receptors, Purinergic P2X, Receptors, Purinergic P2X2, Receptors, Purinergic P2X4, Receptors, Purinergic P2Y1, Adenosine Triphosphate metabolism, Kidney metabolism, Receptors, Purinergic P2 metabolism
- Abstract
Using immunohistological techniques and available polyclonal antibodies, we have identified several ATP-sensitive P2 receptor subtypes in specific structures of the normal rat kidney. Of the P2 receptor subtypes examined, P2X1, P2X2 and P2Y1 receptors were found in the smooth muscle layer of intrarenal vessels. The P2Y1 receptor was also found on glomerular mesangial cells, the brush border membrane of the proximal straight tubule and on peritubular fibroblasts. In the cortex, P2Y4 receptors were found on the tubule epithelium of the proximal convoluted tubule, and P2Y2 receptors on glomerular epithelial cells (podocytes). P2X4 and P2X6 receptors were present throughout the renal tubule epithelium from the proximal tubule to the collecting duct. P2X5 receptors were expressed on medullary collecting duct cells and the apical membrane of the S3 segment of the proximal tubule. Possible functions of these receptor subtypes in normal rat kidney are discussed., (Copyright 2003 S. Karger AG, Basel)
- Published
- 2003
- Full Text
- View/download PDF
23. P2X(7) receptors are expressed during mouse nephrogenesis and in collecting duct cysts of the cpk/cpk mouse.
- Author
-
Hillman KA, Johnson TM, Winyard PJ, Burnstock G, Unwin RJ, and Woolf AS
- Subjects
- Aging, Animals, Blotting, Western, Immunohistochemistry, Kidney metabolism, Kidney Tubules, Collecting growth & development, Kidney Tubules, Collecting pathology, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Microscopy, Confocal, Polycystic Kidney Diseases genetics, Polycystic Kidney Diseases pathology, Receptors, Purinergic P2 immunology, Receptors, Purinergic P2X7, Kidney embryology, Kidney growth & development, Kidney Tubules, Collecting metabolism, Polycystic Kidney Diseases metabolism, Receptors, Purinergic P2 metabolism
- Abstract
Background: Purinergic receptors are cell-surface molecules that bind extracellular nucleotides, notably ATP. The P2X family includes seven nonselective ion channels with one member, P2X(7), implicated in cytolytic pore formation and cell death., Materials and Methods: We sought P2X(7) expression in mouse nephrogenesis and cpk/cpk renal cyst growth, conditions in which both proliferation and apoptosis are prominent., Results: P2X(7) immunolocalized to condensed metanephric mesenchyme: both proliferation and apoptosis were detected in this compartment, assessed by proliferating cell nuclear antigen expression and propidium iodide-stained pyknotic nuclei respectively. Later in nephrogenesis, P2X(7) was detected in collecting ducts, a pattern persisting to maturity. A mesenchymal to epithelial shift of P2X(7) expression was also documented in ureter development. In cpk/cpk kidneys, P2X(7)-expressing collecting duct cysts dominated histology from two weeks until four weeks after birth, when animals die from uremia. In polycystic kidneys pyknotic nuclei were rarely identified in P2X(7)-expressing epithelia, but were detected between cysts, consistent with a non-apoptotic role for P2X(7) in cyst enlargement., Conclusion: P2X(7) is expressed during normal nephrogenesis and in a model of congenital polycystic kidney disease. Further experiments are necessary to define possible functions of P2X(7) in these settings., (Copyright 2002 S. Karger AG, Basel)
- Published
- 2002
- Full Text
- View/download PDF
24. Band 3 Walton, a C-terminal deletion associated with distal renal tubular acidosis, is expressed in the red cell membrane but retained internally in kidney cells.
- Author
-
Toye AM, Bruce LJ, Unwin RJ, Wrong O, and Tanner MJ
- Subjects
- Adult, Amino Acid Sequence, Animals, Anion Exchange Protein 1, Erythrocyte chemistry, Anions, Base Sequence, Biological Transport, Carbonic Anhydrases metabolism, Female, Fluorescent Antibody Technique, Heterozygote, Humans, Male, Molecular Sequence Data, Oocytes metabolism, Protein Isoforms genetics, Transfection, Xenopus, Acidosis, Renal Tubular genetics, Anion Exchange Protein 1, Erythrocyte genetics, Cell Membrane metabolism, Erythrocyte Membrane metabolism, Gene Expression, Kidney metabolism, Sequence Deletion
- Abstract
Human band 3 Walton is an AE1 mutation that results in the deletion of the 11 COOH-terminal amino acids of the protein and is associated with dominant distal renal tubular acidosis. The properties of band 3 Walton expressed with normal band 3 in the heterozygous mutant erythrocytes and the kidney isoform expressed in Xenopus oocytes and in the Madin-Darby canine kidney cell line were examined. The mutant erythrocytes have normal hematology but have reduced band 3 Walton content. Transport studies showed that erythrocyte band 3 Walton has normal sulfate transport activity, and kidney band 3 Walton has normal chloride transport activity when expressed in Xenopus oocytes. The mutant protein is clearly able to reach the cell surface of erythrocytes and oocytes. In contrast, while normal kidney band 3 was expressed at the cell surface in the kidney cell line, the Walton mutant protein was retained intracellularly within the kidney cells. The results demonstrate that band 3 Walton is targeted differently in erythrocytes and kidney cells and indicate that the COOH-terminal tail of band 3 is required to allow movement to the cell surface in kidney cells. It is proposed here that the mutant band 3 gives rise to dominant distal renal tubular acidosis by inhibiting the movement of normal band 3 to the cell surface. It is suggested that this results from the association of the normal and mutant proteins in band 3 hetero-oligomers, which causes the intracellular retention of normal band 3 with the mutant protein.
- Published
- 2002
- Full Text
- View/download PDF
25. P2 receptors in the kidney.
- Author
-
Bailey MA, Hillman KA, and Unwin RJ
- Subjects
- Adenosine Triphosphate metabolism, Adenosine Triphosphate physiology, Animals, Humans, Protein Sorting Signals physiology, Purinergic P2 Receptor Agonists, Renal Circulation physiology, Kidney metabolism, Receptors, Purinergic P2 metabolism
- Abstract
Our understanding of the actions of extracellular ATP in controlling kidney function via stimulation of P2 receptors is still at an early stage. Recently, several groups, including our own, have begun to address this subject: in this brief review, we discuss some of these effects and speculate on likely function of extracellular nucleotides in the kidney.
- Published
- 2000
- Full Text
- View/download PDF
26. Effects of changes in dietary intake of sodium and potassium and of metabolic acidosis on 11beta-hydroxysteroid dehydrogenase activities in rat kidney.
- Author
-
Thompson A, Bailey MA, Michael AE, and Unwin RJ
- Subjects
- 11-beta-Hydroxysteroid Dehydrogenases, Acidosis physiopathology, Aldosterone blood, Animals, In Vitro Techniques, Kidney enzymology, Kidney metabolism, Male, NAD pharmacology, NADP pharmacology, Potassium blood, Potassium urine, Rats, Rats, Sprague-Dawley, Sodium blood, Sodium urine, Ammonium Chloride pharmacology, Hydroxysteroid Dehydrogenases metabolism, Kidney drug effects, Potassium, Dietary administration & dosage, Sodium, Dietary administration & dosage
- Abstract
Background/aim: Glucocorticoid activity is modulated by NADP(+)- and NAD(+)-dependent isoforms of the enzyme 11beta-hydroxysteroid dehydrogenase (11betaHSD) which convert glucocorticoids to their inactive metabolites. The NAD(+)-dependent isoform, 11betaHSD2, is present in the distal nephron where it confers aldosterone specificity on mineralocorticoid receptors. The objective of this study was to establish whether renal 11betaHSD activities are affected by changes in sodium and potassium balance and by metabolic acidosis., Methods: Renal 11betaHSD activities were measured ex vivo from rats fed normal and high- and low-potassium diets and a low-sodium diet or given 1.5% NH(4)Cl to drink., Results: Rats maintained on high-potassium and low-sodium diets exhibited 59% (p < 0.01) and 28% (p < 0.05) decreases, respectively, in NAD(+)-dependent renal 11betaHSD activity (relative to rats fed control diet) with no changes in NADP(+)-dependent cortisol oxidation. Short-term (3 day) and longer-term (10 day) metabolic acidosis also decreased NAD(+)-dependent 11betaHSD activity by 50 and 52%, respectively, without affecting NADP(+)-dependent cortisol oxidation. The low-potassium diet had no detectable effect on renal 11betaHSD activities., Conclusion: These results suggest that adaptations to a high-potassium or a low-sodium diet and to metabolic acidosis involve decreases in renal 11betaHSD2 activity, enhancing the access of glucocorticoids to renal corticosteroid receptors., (Copyright 2000 S. Karger AG, Basel)
- Published
- 2000
- Full Text
- View/download PDF
27. Inhibition of renal 11beta-hydroxysteroid dehydrogenase in vivo by carbenoxolone in the rat and its relationship to sodium excretion.
- Author
-
Sewell KJ, Shirley DG, Michael AE, Thompson A, Norgate DP, and Unwin RJ
- Subjects
- 11-beta-Hydroxysteroid Dehydrogenases, Animals, Cortisone metabolism, Dose-Response Relationship, Drug, Hydrocortisone metabolism, Kidney drug effects, Male, NAD metabolism, NADP metabolism, Potassium urine, Rats, Rats, Sprague-Dawley, Tritium metabolism, Carbenoxolone pharmacology, Enzyme Inhibitors pharmacology, Hydroxysteroid Dehydrogenases antagonists & inhibitors, Kidney enzymology, Sodium urine
- Abstract
1. The type 2 isoform of 11beta-hydroxysteroid dehydrogenase, an enzyme which converts cortisol or corticosterone to inactive 11-ketosteroid metabolites, is thought to be responsible for preventing access of endogenous glucocorticoids to mineralocorticoid receptors in the distal nephron; although direct in vivo evidence for this is still lacking. We have examined whether graded inhibition of renal 11beta-hydroxysteroid dehydrogenase activities in vivo results in corresponding changes in urinary electrolyte excretion due to exposure of mineralocorticoid receptors to circulating endogenous glucocorticoids.2. Anaesthetized rats were infused intravenously with vehicle alone or with one of three doses of carbenoxolone: 0.06, 0.6 or 6 mg/h. After measurement of renal electrolyte excretion, the kidneys were snap-frozen in liquid nitrogen and 11beta-hydroxysteroid dehydrogenase activities were measured directly by enzyme assay in the presence of NAD+ or NADP+.3. A dose-dependent inhibition of renal 11beta-hydroxysteroid dehydrogenase activities was observed: the low, intermediate and high doses of carbenoxolone causing approximately 50%, 80% and >90% inhibition respectively. Only with the high dose was an effect on renal function observed (decreased fractional Na+ excretion and urinary Na+/K+ ratio).4. The poor correlation between the extent of inhibition of renal 11beta-hydroxysteroid dehydrogenase and altered urinary Na+ excretion, apparent at the lower doses of carbenoxolone, suggests either that 11beta-hydroxysteroid dehydrogenase has considerable functional reserve, or that it may not be the only mechanism determining mineralocorticoid receptor specificity in the distal nephron.
- Published
- 1998
28. Conventional and confocal epi-reflection and fluorescence microscopy of the rat kidney in vivo.
- Author
-
Boyde A, Capasso G, and Unwin RJ
- Subjects
- Animals, Fluorescein-5-isothiocyanate analogs & derivatives, Fluoresceins, Insulin analogs & derivatives, Intracellular Fluid metabolism, Ion Transport, Kidney blood supply, Kidney physiology, Male, Microscopy, Confocal instrumentation, Microscopy, Fluorescence instrumentation, Microscopy, Video instrumentation, Microscopy, Video methods, Models, Biological, Rats, Rats, Sprague-Dawley, Renal Circulation, Kidney anatomy & histology, Microscopy, Confocal methods, Microscopy, Fluorescence methods
- Abstract
To visualize superficial and accessible renal tubule cells functioning in situ and to relate what we can 'see' to what we know of their function from more invasive in vivo or less direct in vitro studies means applying and adapting recent advances in epifluorescence and confocal microscopy to improve image resolution and to combine this with the use of fluorescent labels to monitor the handling of specific molecules by the proximal and distal renal tubule cells in vivo. Doing this in living tissue is novel, especially in the kidney. Application of confocal microscopy to the imaging of living tissue, as opposed to isolated cells, has not been widely reported. The kidney surface has been imaged before using the confocal microscope and in preliminary studies we have extended this by using a different confocal system with and without fluorescence. While the studies published up to now have been morphological, comparing standard renal (structural) histology of surface glomeruli and renal tubules with the corresponding in vivo confocal images, more dynamic, real-time studies have been limited. Individual red blood cells can be seen flowing around the peritubule capillary network and nucleated white blood cells can also be distinguished. Tubule cells, endothelial cells, the proximal tubule cell brush border and cell mitochondria can be visualized. Filtration and secretion can be observed, and the early and late parts of the proximal tubule distinguished, and the distal tubule recognized. Localization of fluorescently labeled insulin to the luminal brush border and progressive uptake of label and distribution within proximal tubule cells toward the basolateral (blood side) membrane can be demonstrated. The possibility of monitoring hemodynamic changes and tracking the filtration, uptake, secretion and absorption of fluorescently tagged molecules, as well as intracellular fluorescence, e.g. calcium or pH, is an exciting prospect and is ripe for detailed exploration.
- Published
- 1998
- Full Text
- View/download PDF
29. Potential functional roles of extracellular ATP in kidney and urinary tract.
- Author
-
Chan CM, Unwin RJ, and Burnstock G
- Subjects
- Animals, Humans, Kidney chemistry, Adenosine Triphosphate metabolism, Extracellular Space metabolism, Kidney metabolism, Receptors, Purinergic metabolism
- Abstract
P2 receptors are sensitive to adenosine triphosphate (ATP) and uridine triphosphate and can be divided into two major subtypes: P2X and P2Y receptors. They are specific membrane-bound receptors which when activated by extracellular ATP initiate a variety of biological effects. The P2X receptors are ligand-gated channels, whereas the P2Y receptors are coupled to G proteins linked to second-messenger systems. In this review, the potential functional roles of extracellular ATP in the kidney and lower urinary tract are discussed and briefly explored in the context of some specific renal and urinary tract abnormalities.
- Published
- 1998
- Full Text
- View/download PDF
30. Localization of P2X1 purinoceptors by autoradiography and immunohistochemistry in rat kidneys.
- Author
-
Chan CM, Unwin RJ, Bardini M, Oglesby IB, Ford AP, Townsend-Nicholson A, and Burnstock G
- Subjects
- Animals, Autoradiography, Immunohistochemistry methods, Male, Rats, Rats, Sprague-Dawley, Receptors, Purinergic P2X, Staining and Labeling, Tissue Distribution, Kidney metabolism, Receptors, Purinergic P2 metabolism
- Abstract
P2 receptors have been identified in rat kidney by autoradiography, using the radioligand [3H] alpha, beta-methylene ATP, and by immunohistochemistry, using a polyclonal antibody to the P2X1 purinoceptor. They have been localized to the vascular smooth muscle of intrarenal arteries, including arcuate and interlobular arteries, and afferent arterioles, but not glomeruli, postglomerular efferent arterioles, or renal tubules. We conclude that at least some of the P2 receptors present on vascular smooth muscle are of the P2X1 subtype. The functional significance of these findings in the vascular control of the kidney is discussed.
- Published
- 1998
- Full Text
- View/download PDF
31. Hyperglycemia and intestinal and renal glucose transport: implications for diabetic renal injury.
- Author
-
Debnam ES and Unwin RJ
- Subjects
- Adaptation, Physiological, Animals, Biological Transport physiology, Humans, Kidney Tubules, Proximal metabolism, Monosaccharide Transport Proteins metabolism, Rabbits, Sodium metabolism, Diabetes Complications, Diabetic Nephropathies etiology, Glucose metabolism, Hyperglycemia complications, Intestine, Small metabolism, Kidney metabolism
- Published
- 1996
- Full Text
- View/download PDF
32. Effect of vasopressin on renal lithium reabsorption: a micropuncture and microperfusion study.
- Author
-
Walter SJ, Shirley DG, and Unwin RJ
- Subjects
- Absorption, Animals, Loop of Henle metabolism, Male, Perfusion, Punctures, Rats, Rats, Brattleboro, Renal Agents pharmacology, Deamino Arginine Vasopressin pharmacology, Kidney metabolism, Lithium pharmacokinetics
- Abstract
Micropuncture techniques were used to investigate the nephron site(s) responsible for the vasopressin-induced reductions in lithium clearance and fractional lithium excretion (FELi) in anesthetized Brattleboro rats lacking endogenous vasopressin. In rats treated intravenously with the vasopressin analogue 1-desamino-8-D-arginine vasopressin (DDAVP; 40 pg/min), FELi was significantly lower than in untreated animals (0.23 +/- 0.01 vs. 0.28 +/- 0.02, P < 0.05). Free-flow micropuncture showed that fractional lithium delivery (FDLi) to late proximal convolutions was identical in the two groups, whereas at the early distal tubule both FDLi (0.28 +/- 0.02 vs. 0.33 +/- 0.01, P < 0.05) and the tubular fluid-to-plasma lithium concentration ratio (1.18 +/- 0.04 vs. 1.36 +/- 0.06, P < 0.05) were lower in the DDAVP-treated group. No differences between the groups with respect to lithium handling beyond the early distal tubule were observed. In rats subjected to in vivo microperfusion of loops of Henle, intravenous infusion of DDAVP (40 pg/min) increased fractional lithium reabsorption in the loop from 0.56 +/- 0.03 to 0.66 +/- 0.04 (P < 0.05) and from 0.39 +/- 0.02 to 0.45 +/- 0.02 (P < 0.05) at perfusion rates of 10 and 15 nl/min, respectively. We conclude that DDAVP stimulates lithium reabsorption in the loop of Henle and suggest that this results from an increased transepithelial potential difference in the thick ascending limb.
- Published
- 1996
- Full Text
- View/download PDF
33. Time-course and relationship of the early changes in renal sodium excretion and aldosterone secretion during dietary sodium restriction in normal man.
- Author
-
Carmichael DJ, Sutters MS, Unwin RJ, Gordon D, Few J, James VH, Wadsworth J, and Peart WS
- Subjects
- Adult, Arginine Vasopressin blood, Atrial Natriuretic Factor blood, Humans, Male, Reference Values, Saliva metabolism, Time Factors, Aldosterone metabolism, Diet, Sodium-Restricted, Kidney metabolism, Sodium urine
- Abstract
1. The fall in renal sodium excretion after dietary sodium restriction is prompt and reproducible. The importance of increased aldosterone secretion during the early phase (within 48 h) of this response is unclear. Using two indirect measures of aldosterone secretion (in urine and saliva), we have tried to relate changes in excretion and concentration of this hormone to renal sodium excretion during the abrupt transition from a normal (approximately 150 mmol/day) or high (260 mmol/day) to a low (5-25 mmol/day) sodium intake in 11 and seven male volunteers, respectively. 2. All subjects showed reduced renal sodium excretion within 36 h of dietary restriction, but the times at which increases in renal aldosterone excretion, saliva aldosterone concentration and plasma renin activity became statistically significant varied widely (8-72 h, 2.5- greater than 62.5 h and less than 4- greater than 38 h for renal aldosterone secretion, saliva aldosterone concentration and plasma renin activity, respectively). Circadian fluctuations in saliva aldosterone concentration were apparent and increased in amplitude during sodium restriction. 3. Urine flow rate tended to increase on the first day of sodium restriction and this reached statistical significance in the group initially on a high sodium intake (64.0 +/- 8.8 to 84.3 +/- 11.2 ml/h, P less than 0.01); although the pattern of urine flow did correlate with plasma arginine vasopressin concentration (r = -0.49, P less than 0.01), there was no significant decrease in mean plasma arginine vasopressin concentration [1.15 (0.92-1.44) to 0.90 (0.72-1.12) pmol/l, P = 0.08; geometric mean and 95% confidence limits].(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1990
- Full Text
- View/download PDF
34. Brain-gut peptides, renal function and cell growth.
- Author
-
Unwin RJ, Ganz MD, and Sterzel RB
- Subjects
- Animals, Cell Division, Cyclic AMP metabolism, Gastrointestinal Hormones physiology, Humans, Kidney cytology, Kidney growth & development, Kidney physiology, Neuropeptides physiology
- Published
- 1990
- Full Text
- View/download PDF
35. Effects of vasoactive intestinal polypeptide infusions on renal function in patients with liver disease.
- Author
-
Calam J, Unwin RJ, Dorudi S, Taylor C, Springer CJ, and Peart WS
- Subjects
- Blood Cell Count, Female, Hemodynamics drug effects, Humans, Infusions, Intravenous, Kidney drug effects, Kidney Function Tests, Liver Cirrhosis, Alcoholic physiopathology, Male, Middle Aged, Vasoactive Intestinal Peptide administration & dosage, Kidney physiopathology, Liver Diseases physiopathology, Vasoactive Intestinal Peptide pharmacology
- Abstract
1. The effect of intravenous vasoactive intestinal peptide (VIP, 6 pmol/kg per min) on renal function in six patients with cirrhosis of the liver was examined. 2. VIP caused generalized vasodilation and increased plasma renin activity, but diminished the glomerular filtration rate by about one third. 3. The excretion of water, sodium, potassium and calcium also fell significantly. 4. These results differ from our findings in normal man in whom VIP diminished water and electrolyte secretion largely by increasing tubular reabsorption. 5. It is concluded that the elevated VIP levels present in patients with severe liver disease may affect renal function, but that the presence of liver disease may affect renal responses to VIP.
- Published
- 1988
- Full Text
- View/download PDF
36. Effects of vasoactive intestinal polypeptide on renal function in man.
- Author
-
Calam J, Dimaline R, Peart WS, Singh J, and Unwin RJ
- Subjects
- Adult, Electrolytes blood, Electrolytes urine, Hemodynamics drug effects, Humans, Kidney physiology, Male, Renin blood, Vasoactive Intestinal Peptide blood, Kidney drug effects, Vasoactive Intestinal Peptide pharmacology
- Abstract
Six healthy males received vasoactive intestinal polypeptide (VIP; 6 pmol kg-1 min-1) by intravenous infusion for 90 min, with 60 min control periods before and after. Plasma VIP levels rose by about 100 pmol l-1 during the infusion. VIP produced changes in heart rate and blood pressure consistent with generalized vasodilatation, but there were no significant changes in effective renal plasma flow or glomerular filtration rate. Both plasma solids and haematocrit rose by about 5%, suggesting that haemoconcentration had occurred during VIP infusion. Urine flow fell to about a third, and the fractional excretion of sodium, potassium, chloride and calcium fell to between half and two-thirds of control values. Fractional excretion of phosphate did not fall significantly. Plasma renin activity rose about 3-fold during VIP infusion.
- Published
- 1983
- Full Text
- View/download PDF
37. Renal effects of gastrin C-terminal tetrapeptide (as pentagastrin) and cholecystokinin octapeptide in conscious rabbit and man.
- Author
-
Calam J, Gordon D, Peart WS, Taylor SA, and Unwin RJ
- Subjects
- Adult, Animals, Blood Pressure drug effects, Electrolytes blood, Electrolytes urine, Female, Heart Rate drug effects, Humans, Kidney physiology, Male, Rabbits, Hemodynamics drug effects, Kidney drug effects, Pentagastrin pharmacology, Sincalide pharmacology
- Abstract
Pentagastrin and cholecystokinin octapeptide (CCK8) were infused i.v. at three different doses in two sets of 4 conscious rabbits following a repeated measurements design (130, 1,300 and 13,000 pmol kg-1 min-1 pentagastrin; 5, 50 and 450 pmol kg-1 min-1 CCK8). In man, two different doses of pentagastrin (13 and 65 pmol kg-1 min-1) were infused in two groups of 6 subjects, and CCK8 (2 pmol kg-1 min-1) in a third group. According to published human postprandial levels, plasma CCK8-like immunoreactivity concentrations were supraphysiological at all doses infused. In the rabbit, pentagastrin produced a dose-related fall in urine flow and free water clearance, but no significant change in systemic and renal haemodynamics, electrolyte excretion and measured plasma constituents; however, in human subjects, pentagastrin increased renal sodium excretion and reduced potassium excretion but did not change glomerular filtration rate. In the rabbit, CCK8 produced a dose-related fall in plasma renin activity, plasma calcium concentration and mean arterial blood pressure; dose-dependent increases in effective renal plasma flow, glomerular filtration rate and renal sodium excretion. In man, changes in sodium and potassium excretion similar to pentagastrin were observed; there were no significant changes in plasma renin activity, plasma calcium concentration, blood pressure, effective renal plasma flow or glomerular filtration rate. The pharmacological renal effects of pentagastrin in conscious water-loaded rabbits resemble vasopressin. In contrast, CCK8's most striking effect was vasodilatation and was unusual in inhibiting rather than stimulating renin release. In man the net changes in urine composition found during infusion of these peptides are similar to those produced by the potassium-sparing diuretics, amiloride and triamterene. However the generally weak renal effects observed, even at pharmacological doses, indicate that these peptides are unlikely to influence renal function under normal physiological conditions.
- Published
- 1987
- Full Text
- View/download PDF
38. Effects of vasoactive intestinal polypeptide (VIP) on renal function and plasma renin activity in the conscious rabbit.
- Author
-
Dimaline R, Peart WS, and Unwin RJ
- Subjects
- Animals, Chlorides urine, Hematocrit, Hemodynamics drug effects, Kidney physiology, Male, Potassium urine, Rabbits, Sodium urine, Kidney drug effects, Renin blood, Vasoactive Intestinal Peptide pharmacology
- Abstract
Conscious rabbits received either vasoactive intestinal polypeptide (VIP) at a dose of 1, 10 or 25 pmol kg-1 min-1 or vehicle alone (control) through an ear vein for 2 h. Experimental design followed a randomized Latin square arrangement. VIP led to a decrease in effective renal plasma flow and glomerular filtration rate (P less than 0.01) during infusion of the middle and high doses. Mean arterial blood pressure rose slightly (P less than 0.05) and filtration fraction increased (P less than 0.01) during infusion of the middle dose. The high dose produced a rise in heart rate, a fall in plasma sodium, potassium and phosphate concentrations and a rise in plasma solids (P less than 0.01). In spite of the renal haemodynamic effects and changes in plasma composition during infusion of the high dose, fractional excretion of sodium, potassium and chloride doubled (P less than 0.05), suggesting a direct action of VIP on renal tubular function. Plasma renin activity increased between 2- and 3-fold (P less than 0.01). The mechanism of the renin response is uncertain. These results, together with the reported presence of VIP-like material in the renal cortex, may indicate a role for VIP in the regulation of renal function, including renin release.
- Published
- 1983
- Full Text
- View/download PDF
39. Renal function during bovine neurotensin infusion in man.
- Author
-
Unwin RJ, Calam J, Peart WS, Hanson C, Lee YC, and Bloom SR
- Subjects
- Adult, Calcium urine, Chlorides urine, Diet, Sodium-Restricted, Hemodynamics drug effects, Humans, Kidney blood supply, Kidney drug effects, Male, Natriuresis drug effects, Neurotensin blood, Potassium urine, Renin blood, Sodium administration & dosage, Kidney physiology, Neurotensin pharmacology
- Abstract
The peptide hormone neurotensin (NT) is found mainly in gut endocrine cells of the ileum, but has also been identified as a putative neurotransmitter in the central and peripheral nervous systems. It may have a dual role as a circulating gastrointestinal hormone and peripheral neurotransmitter. Its predominant effects are to reduce oesophageal sphincter tone, inhibit gastric secretion and emptying and inhibit intestinal motility, but stimulate intestinal and pancreatic exocrine secretion; NT-like immunoreactivity has been found in kidney and therefore NT may influence renal function. When infused i.v. in rabbits it causes antinatriuresis. We have studied its renal effects in 11 healthy males by i.v. infusion under conditions of altered dietary sodium. Postprandial circulating neurotensin levels were reproduced by infusion. There were no consistent systemic or renal haemodynamic effects. Plasma electrolytes and renin did not change. Only renal chloride excretion changed significantly, falling by ca. 30%, and recovering after infusion. There is no evidence for a specific renal tubular chloride transport mechanism, but coupled cotransport, Na+:K+:2CI-, may be hormonally regulated. NT might stimulate this process and contribute to the renal response to changes in dietary composition, especially sodium intake.
- Published
- 1987
- Full Text
- View/download PDF
40. Renal electrolyte excretion and renin release during calcium and parathormone infusions in conscious rabbits.
- Author
-
Peart WS, Roddis SA, and Unwin RJ
- Subjects
- Animals, Calcium metabolism, Chlorides metabolism, Hematocrit, Hemodynamics drug effects, Kidney blood supply, Potassium metabolism, Rabbits, Renin blood, Sodium metabolism, Calcium Chloride pharmacology, Kidney drug effects, Parathyroid Hormone pharmacology
- Abstract
Following a random block experimental design in each case, three repeated measurement studies were carried out in three different groups of conscious rabbits, to investigate the renal effects of increasing doses of intravenous calcium chloride (CaCl2) and bovine parathyroid hormone (PTH). In the first study, each rabbit received either CaCl2 (0.15, 0.3, 0.5 or 1.0 mg kg-1 min-1) or vehicle alone (control) for 160 min. In the second study, rabbits were given either PTH (0.15 microgram kg-1 min-1), CaCl2 (1.0 mg kg-1 min-1), PTH plus CaCl2 (0.15 microgram kg-1 min-1 and 1.0 mg kg-1 min-1, respectively) or vehicle alone; PTH was infused for just over 60 min. In the third study, a much smaller dose (0.05 mg kg-1 min-1) of CaCl2 was infused for 100 min. CaCl2 infusion produced a striking fall in fractional excretion of sodium of at least 50% (P less than 0.01), but this was not dose related, being almost maximal at the smaller doses infused. Although this effect was evident in the absence of any changes in total plasma calcium concentration at the lower doses of CaCl2, renal calcium excretion was increased between 2- and 20-fold (P less than 0.01) at all doses infused. Fractional excretion of chloride doubled at the two higher doses of CaCl2 (P less than 0.01), but potassium excretion was unchanged. There were no consistent alterations in mean arterial blood pressure, effective renal plasma flow, glomerular filtration rate or plasma renin activity (PRA); total plasma calcium concentration was consistently elevated only during infusion of the high dose by just under 1 mmol l-1. PTH infusion had no measured effect on fractional excretion of sodium or renal calcium excretion, but doubled fractional potassium excretion (P less than 0.05). Heart rate and PRA increased (P less than 0.01 and less than 0.05, respectively), the latter by 50%, but systemic pressure and renal haemodynamics were not significantly affected. By contrast, PTH infused with CaCl2 produced a 4-fold rise in fractional sodium excretion and although renal calcium excretion remained increased, it was reduced by ca. 80% when compared with renal calcium excretion during infusion of CaCl2 alone. Infusion of PTH alone increased PRA, but when PTH and CaCl2 were infused together, PRA did not change.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1986
- Full Text
- View/download PDF
41. Renal function during vasoactive intestinal peptide (VIP) infusions in normal man and patients with liver disease.
- Author
-
Calam J, Unwin RJ, Singh J, Dorudi S, and Peart WS
- Subjects
- Adult, Aldosterone blood, Blood Pressure drug effects, Electrolytes urine, Glomerular Filtration Rate drug effects, Heart Rate drug effects, Humans, Kidney physiology, Kidney physiopathology, Male, Middle Aged, Reference Values, Renal Circulation drug effects, Renin blood, Kidney drug effects, Liver Diseases physiopathology, Vasoactive Intestinal Peptide blood
- Abstract
VIP containing nerves are present in the kidney and plasma VIP levels are elevated in cardiac failure and severe liver disease. We studied the effects of intravenous VIP; 6 pmol kg-1 min-1 on 6 normal subjects and 3 patients with liver disease. In normal subjects VIP produced flushing and significant rises in heart rate and pulse pressure but the clearance rates of paraaminohippurate and creatinine did not change significantly. Urine flow fell to about 1/3 and the rate of excretion of electrolytes (except phosphate) fell to about a half of control values. Plasma renin activity rose about 3-fold and there were significant rises in haematocrit and the plasma concentrations of solids, calcium and phosphate. The patients with liver disease responded similarly. Elevated plasma VIP could contribute to salt and water retention in disease states.
- Published
- 1984
- Full Text
- View/download PDF
42. Renal adaptation and gut hormone release during sodium restriction in ileostomized man.
- Author
-
Unwin RJ, Moss S, Peart WS, and Wadsworth J
- Subjects
- Adult, Aged, Aldosterone metabolism, Arginine Vasopressin blood, Diuresis, Female, Humans, Ileum analysis, Kallikreins metabolism, Male, Middle Aged, Pancreatic Polypeptide metabolism, Potassium metabolism, Renin blood, Sodium metabolism, Adaptation, Physiological, Diet, Sodium-Restricted, Gastrointestinal Hormones metabolism, Ileostomy, Kidney metabolism
- Abstract
The renal excretion of water, electrolytes, aldosterone and kallikrein was monitored in 12 ileostomized patients before and during sodium deprivation. Changes in plasma renin activity (PRA), plasma aldosterone and plasma arginine vasopressin (AVP) concentrations were measured, together with aldosterone in ileal fluid. The pattern of gut peptide release in response to a test meal was also examined to assess whether a circulating gut peptide might be involved in the renal adaptation to sodium restriction, and compared with healthy normal subjects who were under no dietary constraint. In each patient renal sodium excretion fell within 8-12 h of sodium deprivation and was associated with a prompt and significant rise in PRA; much later increases in plasma aldosterone concentration and renal aldosterone excretion occurred, and were established by day 2 of sodium restriction. No consistent change in renal kallikrein excretion was found. Ileal sodium loss was little changed by sodium deprivation, but ileal potassium concentration rose steadily and became significantly correlated with PRA, and to a lesser extent with renal aldosterone excretion. Of the gut peptides measured in plasma, only the insulin profile was altered by sodium deprivation, with an increase in the test meal response; insulin has previously been shown to have a significant antinatriuretic action at physiological concentrations. Plasma levels of pancreatic polypeptide and motilin were increased in ileostomized patients when compared with normal subjects, but were unaffected by the change to a low sodium diet. An early increase in urine flow and water diuresis occurred during sodium deprivation, following a cyclical pattern with peaks each evening.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1985
- Full Text
- View/download PDF
43. Brain dysfunction in tubular and tubulointerstitial kidney diseases
- Author
-
Viggiano D, Bruchfeld A, Carriazo S, de Donato A, Endlich N, Ferreira AC, Figurek A, Fouque D, Franssen CFM, Giannakou K, Goumenos D, Hoorn EJ, Nitsch D, Arduan AO, Pešić V, Rastenyté D, Soler MJ, Rroji M, Trepiccione F, Unwin RJ, Wagner CA, Wiecek A, Zacchia M, Zoccali C, Capasso G, CONNECT Action (Cognitive Decline in Nephro-Neurology European Cooperative Target)., Viggiano, D, Bruchfeld, A, Carriazo, S, de Donato, A, Endlich, N, Ferreira, Ac, Figurek, A, Fouque, D, Franssen, Cfm, Giannakou, K, Goumenos, D, Hoorn, Ej, Nitsch, D, Arduan, Ao, Pešić, V, Rastenyté, D, Soler, Mj, Rroji, M, Trepiccione, F, Unwin, Rj, Wagner, Ca, Wiecek, A, Zacchia, M, Zoccali, C, Capasso, G, CONNECT Action (Cognitive Decline in Nephro-Neurology European Cooperative, Target)., NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), CarMeN, laboratoire, University of the Study of Campania Luigi Vanvitelli, Karolinska University Hospital [Stockholm], Linköping University (LIU), IIS‑Fundación Jiménez Diaz‑Autonoma University [Madrid, Spain], University of Medicine Greifswald, Universidade Nova de Lisboa = NOVA University Lisbon (NOVA), Universität Zürich [Zürich] = University of Zurich (UZH), Cardiovasculaire, métabolisme, diabétologie et nutrition (CarMeN), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Hospices Civils de Lyon (HCL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Hospices Civils de Lyon (HCL), Centre Hospitalier Lyon Sud [CHU - HCL] (CHLS), University of Groningen [Groningen], European University of Cyprus, General University Hospital of Patras, Erasmus University Medical Center [Rotterdam] (Erasmus MC), London School of Hygiene and Tropical Medicine (LSHTM), University of Belgrade [Belgrade], Lithuanian University of health Sciences [Kaunas], Vall d’Hebron Research Institute (VHIR), University Hospital Center 'Mother Tereza' [Tirana, Albania] (UHCMT), University College of London [London] (UCL), Medical University of Silesia (SUM), Renal Research Institute [New York, NY, USA] (2RI), CONNECT Action (Cognitive Decline in Nephro-Neurology European Cooperative Target): Giovambattista Capasso, Alexandre Andrade, Maie Bachmann, Inga Bumblyte, Adrian Constantin Covic, Pilar Delgado, Nicole Endlich, Andreas Engvig, Denis Fouque, Casper Franssen, Sebastian Frische, Liliana Garneata, Loreto Gesualdo, Konstantinos Giannakou, Dimitrios Goumenos, Ayşe Tuğba Kartal, Laila-Yasmin Mani, Hans-Peter Marti, Christopher Mayer, Rikke Nielsen, Vesna Pšić, Merita Rroji Molla, Giorgos Sakkas, Goce Spasovski, Kate I Stevens, Evgueniy Vazelov, Davide Viggiano, Lefteris Zacharia, Ana Carina Ferreira, Jolanta Malyszko, Ewout Hoorn, Andreja Figurek, Robert Unwin, Carsten A Wagner, Christoph Wanner, Annette Bruchfeld, Marion Pépin, Andrzej Wieçek, Dorothea Nitsch, Ivo Fridolin, Gaye Hafez, Maria José Soler, Michelangela Barbieri, Bojan Batinić, Laura Carrasco, Sol Carriazo, Ron Gansevoort, Gianvito Martino, Francesco Mattace Raso, Ionut Nistor, Alberto Ortiz, Giuseppe Paolisso, Daiva Rastenytė, Gabriel Stefan, Gioacchino Tedeschi, Ziad A Massy, Boris Bikbov, Karl Hans Endlich, Olivier Godefroy, Jean-Marc Chillon, Anastassia Kossioni, Justina Kurganaite, Norberto Perico, Giuseppe Remuzzi, Tomasz Grodzicki, Francesco Trepiccione, Carmine Zoccali, Mustafa Arici, Peter Blankestijn, Kai-Uwe Eckardt, Danilo Fliser, Eugenio Gutiérrez Jiménez, Maximilian König, Ivan Rychlik, Michela Deleidi, George Reusz, and Internal Medicine
- Subjects
ACIDOSIS ,[SDV]Life Sciences [q-bio] ,Review ,Disease ,electrolyte ,030204 cardiovascular system & hematology ,urologic and male genital diseases ,0302 clinical medicine ,Child ,610 Medicine & health ,MUTATION ,0303 health sciences ,Kidney ,Proteinuria ,Reabsorption ,female genital diseases and pregnancy complications ,3. Good health ,[SDV] Life Sciences [q-bio] ,BARTTER-SYNDROME ,medicine.anatomical_structure ,Nephrology ,Child, Preschool ,GITELMANS-SYNDROME ,Kidney Diseases ,medicine.symptom ,Glomerular Filtration Rate ,medicine.medical_specialty ,brain ,chronic kidney disease ,cognitive function ,tubulointerstitial ,Urology ,Renal function ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,medicine ,Humans ,Renal Insufficiency, Chronic ,AcademicSubjects/MED00340 ,NEPHRITIS ,030304 developmental biology ,Rheumatology and Autoimmunity ,Transplantation ,Reumatologi och inflammation ,HYPONATREMIA ,business.industry ,urogenital system ,AQP2 ,medicine.disease ,Nephrogenic diabetes insipidus ,GENE ,KLOTHO ,MODEL ,Nephritis, Interstitial ,business ,Tubulointerstitial Disease ,Kidney disease - Abstract
Funding: This article is published as part of a supplement financially supported by the COST Action CA19127-Cognitive Decline in Nephro-Neurology: European Cooperative Target (CONNECT). Kidney function has two important elements: glomerular filtration and tubular function (secretion and reabsorption). A persistent decrease in glomerular filtration rate (GFR), with or without proteinuria, is diagnostic of chronic kidney disease (CKD). While glomerular injury or disease is a major cause of CKD and usually associated with proteinuria, predominant tubular injury, with or without tubulointerstitial disease, is typically non-proteinuric. CKD has been linked with cognitive impairment, but it is unclear how much this depends on a decreased GFR, altered tubular function or the presence of proteinuria. Since CKD is often accompanied by tubular and interstitial dysfunction, we explore here for the first time the potential role of the tubular and tubulointerstitial compartments in cognitive dysfunction. To help address this issue we selected a group of primary tubular diseases with preserved GFR in which to review the evidence for any association with brain dysfunction. Cognition, mood, neurosensory and motor disturbances are not well characterized in tubular diseases, possibly because they are subclinical and less prominent than other clinical manifestations. The available literature suggests that brain dysfunction in tubular and tubulointerstitial diseases is usually mild and is more often seen in disorders of water handling. Brain dysfunction may occur when severe electrolyte and water disorders in young children persist over a long period of time before the diagnosis is made. We have chosen Bartter and Gitelman syndromes and nephrogenic diabetes insipidus as examples to highlight this topic. We discuss current published findings, some unanswered questions and propose topics for future research. publishersversion published
- Published
- 2022
- Full Text
- View/download PDF
44. Renal tubular acidosis: developments in our understanding of the molecular basis
- Author
-
Chris Laing, Giovambattista Capasso, Ashley M. Toye, Robert J. Unwin, Laing, C. M., Toye, A. M., Capasso, Giovambattista, and Unwin, Rj
- Subjects
Adult ,medicine.medical_specialty ,Anion exchanger ,Anion gap ,Nephron ,Kidney ,urologic and male genital diseases ,Carbonic Anhydrase II ,Biochemistry ,Renal tubular acidosis ,Anion Exchange Protein 1, Erythrocyte ,Internal medicine ,medicine ,Animals ,Humans ,Nephrocalcinosi ,Child ,Acidosis ,Osteomalacia ,Chemistry ,Metabolic acidosis ,Acidosis, Renal Tubular ,Cell Biology ,medicine.disease ,Proton pump ,Proton-Translocating ATPases ,Endocrinology ,medicine.anatomical_structure ,medicine.symptom ,Nephrocalcinosis ,Renal tubular acidosi ,Intercalated cell - Abstract
Renal tubular acidosis is a metabolic acidosis due to impaired acid excretion by the kidney. Hyperchloraemic acidosis with a normal anion gap and normal (or near normal) glomerular filtration rate, and in the absence of diarrhoea, defines this disorder. However, systemic acidosis is not always evident and renal tubular acidosis can present with hypokalaemia, medullary nephrocalcinosis and recurrent calcium phosphate stone disease, as well as growth retardation and rickets in children, or short stature and osteomalacia in adults. Renal dysfunction in renal tubular acidosis is not always confined to acid excretion and can be part of a more generalised renal tubule defect, as in the renal Fanconi syndrome. Isolated renal tubular acidosis is more usually acquired, due to drugs, autoimmune disease, post-obstructive uropathy or any cause of medullary nephrocalcinosis. Less commonly, it is inherited and may be associated with deafness, osteopetrosis or ocular abnormalities. The clinical classification of renal tubular acidosis has been correlated with our current physiological model of how the nephron excretes acid, and this has facilitated genetic studies that have identified mutations in several genes encoding acid and base ion transporters. In vitro functional studies of these mutant proteins in cell expression systems have helped to elucidate the molecular mechanisms underlying renal tubular acidosis, which ultimately may lead to new therapeutic options in what is still treatment only by giving an oral alkali. © 2005 Elsevier Ltd. All rights reserved.
- Published
- 2005
- Full Text
- View/download PDF
45. Conventional and Confocal Epi-Reflection and Fluorescence Microscopy of the Rat Kidney in vivo
- Author
-
Giovambattista Capasso, Robert J. Unwin, Alan Boyde, Boyde, A, Capasso, Giovambattista, and Unwin, Rj
- Subjects
Intracellular Fluid ,Male ,Pathology ,medicine.medical_specialty ,Brush border ,Kidney, in vivo ,Physiology ,Confocal ,Biology ,Kidney ,Models, Biological ,Fluorescence ,Renal Circulation ,law.invention ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Confocal microscopy ,law ,In vivo ,Genetics ,Fluorescence microscope ,medicine ,Animals ,Insulin ,Fluorescein ,Ion Transport ,Microscopy, Confocal ,Microscopy, Video ,Confocal laser scanning microscope ,General Medicine ,Fluoresceins ,Rats ,medicine.anatomical_structure ,Tubule ,Microscopy, Fluorescence ,chemistry ,Nephrology ,Biophysics ,Fluorescein-5-isothiocyanate - Abstract
To visualize superficial and accessible renal tubule cells functioning in situ and to relate what we can ‘see’ to what we know of their function from more invasive in vivo or less direct in vitro studies means applying and adapting recent advances in epifluorescence and confocal microscopy to improve image resolution and to combine this with the use of fluorescent labels to monitor the handling of specific molecules by the proximal and distal renal tubule cells in vivo. Doing this in living tissue is novel, especially in the kidney. Application of confocal microscopy to the imaging of living tissue, as opposed to isolated cells, has not been widely reported. The kidney surface has been imaged before using the confocal microscope and in preliminary studies we have extended this by using a different confocal system with and without fluorescence. While the studies published up to now have been morphological, comparing standard renal (structural) histology of surface glomeruli and renal tubules with the corresponding in vivo confocal images, more dynamic, real-time studies have been limited. Individual red blood cells can be seen flowing around the peritubule capillary network and nucleated white blood cells can also be distinguished. Tubule cells, endothelial cells, the proximal tubule cell brush border and cell mitochondria can be visualized. Filtration and secretion can be observed, and the early and late parts of the proximal tubule distinguished, and the distal tubule recognized. Localization of fluorescently labeled insulin to the luminal brush border and progressive uptake of label and distribution within proximal tubule cells toward the basolateral (blood side) membrane can be demonstrated. The possibility of monitoring hemodynamic changes and tracking the filtration, uptake, secretion and absorption of fluorescently tagged molecules, as well as intracellular fluorescence, e.g. calcium or pH, is an exciting prospect and is ripe for detailed exploration.
- Published
- 1998
- Full Text
- View/download PDF
46. Combined proteomic and metabonomic studies in three genetic forms of the renal Fanconi syndrome
- Author
-
Jeremy K. Nicholson, Giovambattista Capasso, Annalisa Vilasi, Pedro R. Cutillas, Elaine Holmes, Anthony D. Maher, Severine Zirah, Robert J. Unwin, Anthony W. G. Norden, Second University of Naples-Caserta, University of Naples Federico II, Ludwig Institute for Cancer Research, Biomolecular Medicine, Department of Surgery and Cancer Faculty of Medicine, Imperial College London, Center of Mass Spectrometry, Proteomics, and Bioinformatics, Institute of Food Science, ConsiglioNazionale Delle Ricerche, Avellino, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Vilasi, A, Cutillas, Pr, Maher, Ad, Zirah, Sf, Capasso, Giovambattista, Norden, Aw, Holmes, E, Nicholson, Jk, and Unwin, Rj
- Subjects
Male ,Proteomics ,Pathology ,Magnetic Resonance Spectroscopy ,Physiology ,Dent Disease ,Urine ,Kidney ,0302 clinical medicine ,Trypsin ,ComputingMilieux_MISCELLANEOUS ,0303 health sciences ,Proteinuria ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Aminoaciduria ,Female ,medicine.symptom ,Adult ,medicine.medical_specialty ,Spectrometry, Mass, Electrospray Ionization ,Urinary system ,Biology ,03 medical and health sciences ,Tubulopathy ,Chloride Channels ,[CHIM.ANAL]Chemical Sciences/Analytical chemistry ,Internal medicine ,medicine ,Humans ,Ifosfamide ,Antineoplastic Agents, Alkylating ,030304 developmental biology ,Mass spectrometry ,nutritional and metabolic diseases ,medicine.disease ,Fanconi Syndrome ,DNA Fingerprinting ,NMR ,Endocrinology ,Metabolism ,Oculocerebrorenal Syndrome ,Multivariate Analysis ,Renal Fanconi Syndrome ,Peptides ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Kidney disease - Abstract
The renal Fanconi syndrome is a defect of proximal tubular function causing aminoaciduria and low-molecular-weight proteinuria. Dent's disease and Lowe syndrome are defined X-linked forms of Fanconi syndrome; there is also an autosomal dominant idiopathic form (ADIF), phenotypically similar to Dent's disease though its gene defect is still unknown. To assess whether their respective gene products are ultimately involved in a common reabsorptive pathway for proteins and low-molecular-mass endogenous metabolites, we compared renal Fanconi urinary proteomes and metabonomes with normal (control) urine using mass spectrometry and1H-NMR spectroscopy, respectively. Urine from patients with low-molecular-weight proteinuria secondary to ifosfamide treatment (tubular proteinuria; TP) was also analyzed for comparison. All four of the disorders studied had characteristic proteomic and metabonomic profiles. Uromodulin was the most abundant protein in normal urine, whereas Fanconi urine was dominated by albumin.1H-NMR spectroscopic data showed differences in the metabolic profiles of Fanconi urine vs. normal urine, due mainly to aminoaciduria. There were differences in the urinary metabolite and protein compositions between the three genetic forms of Fanconi syndrome: cluster analysis grouped the Lowe and Dent's urinary proteomes and metabonomes together, whereas ADIF and TP clustered together separately. Our findings demonstrate a distinctive “polypeptide and metabolite fingerprint” that can characterize the renal Fanconi syndrome; they also suggest that more subtle and cause-specific differences may exist between the different forms of Fanconi syndrome that might provide novel insights into the underlying mechanisms and cellular pathways affected.
- Published
- 2007
- Full Text
- View/download PDF
47. Uric acid and the kidney: Urate transport, stone disease and progressive renal failure
- Author
-
R Unwin, William G. Robertson, Ph. Jaeger, Giovambattista Capasso, Capasso, Giovambattista, Jager, P., Robertson, Wg, and Unwin, Rj
- Subjects
medicine.medical_specialty ,Renal failure ,Tamm–Horsfall protein ,Organic Cation Transport Proteins ,Urinary system ,Tamm-Horsfall protein ,Urology ,Organic Anion Transporters ,Hyperuricemia ,Urate transport ,urologic and male genital diseases ,Kidney ,Nephropathy ,chemistry.chemical_compound ,Kidney Calculi ,Organic Anion Transport Protein 1 ,Internal medicine ,Drug Discovery ,Nephrolithiasi ,Uromodulin ,medicine ,Animals ,Humans ,Renal Insufficiency ,Pharmacology ,biology ,Kidney metabolism ,Biological Transport ,medicine.disease ,Endocrinology ,medicine.anatomical_structure ,chemistry ,biology.protein ,Uric acid ,Carrier Proteins ,Kidney disease - Abstract
In this brief review and update, we try to cover recent developments in our understanding of uric acid transport by the kidney, the contribution of uric acid to renal stone disease, its potential role in progressive renal failure and, most recently, the novel and as yet unexplained link between the urinary glycoprotein Tamm-Horsfall protein (uromodulin) and hyperuricaemia and two inherited forms of renal disease with chronic renal failure. © 2005 Bentham Science Publishers Ltd.
- Published
- 2005
48. Urinary acidification and distal renal tubular acidosis
- Author
-
Rj, Unwin, Dg, Shirley, Giovambattista Capasso, Unwin, Rj, Shirley, Dg, and Capasso, Giovambattista
- Subjects
Metabolic acidosi ,viruses ,Cl-HCO ,H-ATPase ,Acidosis, Renal Tubular ,biochemical phenomena, metabolism, and nutrition ,Kidney ,exchanger ,Distal tubular acidosi ,Reference Values ,Humans ,Kidney Tubules, Distal ,Acids - Abstract
Historically, renal tubular acidosis (RTA) has been classified on a clinical basis, without any reference to the underlying disorder. Here we review the normal mechanisms of renal acidification and we identify disorders of specific transporters (genetic, disease-related or drug-induced) that lead to the main categories of distal RTA. We also describe the approach to diagnosis and the current treatment of distal RTA.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.