Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 293(42): 16488-16502, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30139743

RESUMEN

Up to 15% of the population have mild to moderate chronic hypomagnesemia, which is associated with type 2 diabetes mellitus, hypertension, metabolic syndrome, and chronic kidney disease. The kidney is the key organ for magnesium homeostasis, but our understanding of renal magnesium regulation is very limited. Uromodulin (UMOD) is the most abundant urinary protein in humans, and here we report that UMOD has a role in renal magnesium homeostasis. Umod-knockout (Umod-/-) mice excreted more urinary magnesium than WT mice and displayed up-regulation of genes promoting magnesium absorption. The majority of magnesium is absorbed in the thick ascending limb. However, both mouse strains responded similarly to the diuretic agent furosemide, indicating appropriate function of the thick ascending limb in the Umod-/- mice. Magnesium absorption is fine-tuned in the distal convoluted tubule (DCT) via the apical magnesium channel transient receptor potential melastatin 6 (TRPM6). We observed decreased apical Trpm6 staining in the DCT of Umod-/- mice. Applying biotinylation assays and whole-cell patch-clamp recordings, we found that UMOD enhances TRPM6 cell-surface abundance and current density from the extracellular space. UMOD physically interacted with TRPM6 and thereby impaired dynamin-dependent TRPM6 endocytosis. WT mice fed a low-magnesium diet had an increased urinary UMOD secretion compared with the same mice on a regular diet. Our results suggest that increased urinary UMOD secretion in low-magnesium states reduces TRPM6 endocytosis and thereby up-regulates TRPM6 cell-surface abundance to defend against further urinary magnesium losses.


Asunto(s)
Homeostasis , Riñón/química , Magnesio/metabolismo , Canales Catiónicos TRPM/metabolismo , Uromodulina/fisiología , Animales , Endocitosis , Furosemida/farmacología , Humanos , Túbulos Renales Distales/metabolismo , Magnesio/orina , Ratones , Ratones Noqueados , Uromodulina/genética
2.
Curr Opin Nephrol Hypertens ; 28(5): 481-489, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31205055

RESUMEN

PURPOSE OF REVIEW: Uromodulin (UMOD), also known as Tamm-Horsfall protein, is the most abundant protein in human urine. UMOD has multiple functions such as protection against urinary tract infections and nephrolithiasis. This review outlines recent progress made in UMOD's role in renal physiology, tubular transport, and mineral metabolism. RECENT FINDINGS: UMOD is mostly secreted in the thick ascending limb (TAL) and to a lesser degree in the distal convoluted tubule (DCT). UMOD secretion is regulated by the calcium-sensing receptor. UMOD upregulates ion channels [e.g., renal outer medullary potassium channel, transient receptor potential cation channel subfamily V member 5, and transient receptor potential melastatin 6 (TRPM6)] and cotransporters [e.g., Na,K,2Cl cotransporter (NKCC2) and sodium-chloride cotransporter (NCC)] in the TAL and DCT. Higher serum UMOD concentrations have been associated with higher renal function and preserved renal reserve. Higher serum UMOD has also been linked to a lower risk of cardiovascular disease and diabetes mellitus. SUMMARY: With better serum UMOD detection assays the extent of different functions for UMOD is still expanding. Urinary UMOD regulates different tubular ion channels and cotransporters. Variations of urinary UMOD secretion can so contribute to common disorders such as hypertension or nephrolithiasis.


Asunto(s)
Minerales/metabolismo , Uromodulina/fisiología , Animales , Calcio/metabolismo , Humanos , Hipertensión/etiología , Hipertensión/metabolismo , Transporte Iónico , Túbulos Renales/metabolismo , Magnesio/metabolismo , Insuficiencia Renal Crónica/etiología , Uromodulina/sangre
3.
J Am Soc Nephrol ; 29(4): 1326-1335, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29511113

RESUMEN

Many biomarkers have been epidemiologically linked with CKD; however, the possibility that such associations are due to reverse causation or confounding limits the utility of these biomarkers. To overcome this limitation, we used a Mendelian randomization (MR) approach to identify causal mediators of CKD. We performed MR by first identifying genetic determinants of 227 serum protein biomarkers assayed in 4147 participants of the Outcome Reduction with Initial Glargine Intervention (ORIGIN) trial who had early or prediabetes, and assessing the effects of these biomarkers on CKD in the CKD genetics consortium (n=117,165; 12,385 cases) using the inverse-variance weighted (fixed-effects) method. We then estimated the relationship between the serum concentration of each biomarker identified and incident CKD in ORIGIN participants. MR identified uromodulin (UMOD) and human EGF receptor 2 (HER2) as novel, causal mediators of CKD (UMOD: odds ratio [OR], 1.30 per SD; 95% confidence interval [95% CI], 1.25 to 1.35; P<5×10-20; HER2: OR, 1.30 per SD; 95% CI, 1.14 to 1.48; P=8.0×10-5). Consistent with these findings, blood HER2 concentration associated with CKD events in ORIGIN participants (OR, 1.07 per SD; 95% CI, 1.01 to 1.13; P=0.01). Additional exploratory MR analyses identified angiotensin-converting enzyme (ACE) as a regulator of HER2 levels (ß=0.13 per SD; 95% CI, 0.08 to 0.16; P=2.5×10-7). This finding was corroborated by an inverse relationship between ACE inhibitor use and HER2 levels. Thus, UMOD and HER2 are independent causal mediators of CKD in humans, and serum HER2 levels are regulated in part by ACE. These biomarkers are potential therapeutic targets for CKD prevention.


Asunto(s)
Estado Prediabético/sangre , Receptor ErbB-2/sangre , Insuficiencia Renal Crónica/etiología , Uromodulina/sangre , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Antihipertensivos/farmacología , Biomarcadores , Causalidad , Femenino , Estudios de Seguimiento , Genes erbB-2 , Humanos , Riñón/anatomía & histología , Donadores Vivos , Masculino , Análisis de la Aleatorización Mendeliana , Persona de Mediana Edad , Nefrectomía , Tamaño de los Órganos , Peptidil-Dipeptidasa A/fisiología , Polimorfismo de Nucleótido Simple , Estado Prediabético/genética , Receptor ErbB-2/genética , Receptor ErbB-2/fisiología , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/epidemiología , Insuficiencia Renal Crónica/genética , Uromodulina/genética , Uromodulina/fisiología
4.
Curr Opin Nephrol Hypertens ; 26(2): 136-142, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27898524

RESUMEN

PURPOSE OF REVIEW: Although uromodulin or Tamm-Horsfall protein was discovered over 60 years ago, its functional role in humans remains unclear. This review highlights new studies elucidating the clinical correlates of uromodulin, its association with kidney function decline, nephrolithiasis and urinary host defense. RECENT FINDINGS: Uromodulin is evolutionarily conserved and has multiple functional roles. In large population studies, higher levels of uromodulin are associated with higher estimated glomerular filtration rate (eGFR) and kidney size, possibly indicating greater kidney functional reserve. Greater uromodulin excretion is associated with markers of volume overload such as fractional excretion of uric acid, sodium and chloride, indicating a possible role in salt and water retention. Recent evidence also suggests that higher uromodulin levels are associated with lower risk of eGFR decline, death and possibly a lower risk of acute kidney injury. Higher levels of uromodulin are associated with lower risk of urinary tract infections in older adults. Serum uromodulin levels are positively associated with eGFR, although its functional role remains unclear. SUMMARY: Over the last decade, we have begun to understand the functional role of uromodulin in health and disease. Large prospective studies in generalizable populations are needed to confirm these preliminary results, evaluate the clinical utility of measuring uromodulin and examine whether levels of this biomarker can be altered for therapeutic benefit.


Asunto(s)
Tasa de Filtración Glomerular/fisiología , Riñón/anatomía & histología , Riñón/fisiología , Uromodulina/sangre , Lesión Renal Aguda/sangre , Biomarcadores/sangre , Humanos , Cálculos Renales/sangre , Tamaño de los Órganos/fisiología , Factores Protectores , Infecciones Urinarias/sangre , Uromodulina/fisiología
5.
Pediatr Nephrol ; 29(7): 1151-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23880785

RESUMEN

The most abundant urinary protein, Tamm-Horsfall protein, later renamed uromodulin, is expressed exclusively by the thick ascending limb cells of the kidney and released into urine from the apical cell membrane. Uromodulin is believed to protect against urinary tract infections and stones, but its other physiologic functions have remained obscure until recently. Renewed interest in uromodulin has been brought about by the identification of uromodulin mutations as causes of a discrete group of diseases that are distinct from nephronophthisis. The three overlapping clinical uromodulin-associated kidney diseases (UAKD) are medullary cystic disease type 2, familial juvenile hyperuricemic nephropathy and glomerulocystic kidney disease. Previously thought of as "adult diseases", it is now recognized that they may also present in childhood and even in infancy. Common characteristics of all three diseases are autosomal dominant inheritance, unremarkable urine sediment and slow progression to end-stage renal disease (ESRD). They are frequently associated with hyperuricemia and gout. These diseases appear to result from failure of the mutant uromodulin to be incorporated into the apical cilium, thereby placing UAKD in the category of "ciliopathies". In addition to causing specific UAKD, certain uromodulin gene polymorphisms have been linked to ESRD in general, suggesting that uromodulin plays a modulatory role in kidney disease progression.


Asunto(s)
Gota/etiología , Hiperuricemia/etiología , Enfermedades Renales/etiología , Riñón Poliquístico Autosómico Dominante/etiología , Uromodulina/deficiencia , Uromodulina/fisiología , Animales , Enfermedades del Sistema Nervioso Central/etiología , Esmalte Dental/anomalías , Diabetes Mellitus Tipo 2/etiología , Humanos , Enfermedades Renales Quísticas/etiología , Mutación , Insuficiencia Renal Crónica/etiología , Uromodulina/química , Uromodulina/genética
6.
J Am Soc Nephrol ; 23(5): 834-41, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22323644

RESUMEN

Kidney-specific induction of heme oxygenase-1 (HO-1) attenuates the development of angiotensin II (Ang II) -dependent hypertension, but the relative contribution of vascular versus tubular induction of HO-1 is unknown. To determine the specific contribution of thick ascending loop of Henle (TALH) -derived HO-1, we generated a transgenic mouse in which the uromodulin promoter controlled expression of human HO-1. Quantitative RT-PCR and confocal microscopy confirmed successful localization of the HO-1 transgene to TALH tubule segments. Medullary HO activity, but not cortical HO activity, was significantly higher in transgenic mice than control mice. Enhanced TALH HO-1 attenuated the hypertension induced by Ang II delivered by an osmotic minipump for 10 days (139 ± 3 versus 153 ±2 mmHg in the transgenic and control mice, respectively; P<0.05). The lower blood pressure in transgenic mice associated with a 60% decrease in medullary NKCC2 transporter expression determined by Western blot. Transgenic mice also exhibited a 36% decrease in ouabain-sensitive sodium reabsorption and a significantly attenuated response to furosemide in isolated TALH segments. In summary, these results show that increased levels of HO-1 in the TALH can lower blood pressure by a mechanism that may include alterations in NKCC2-dependent sodium reabsorption.


Asunto(s)
Angiotensina II/fisiología , Hemo-Oxigenasa 1/fisiología , Hipertensión/prevención & control , Asa de la Nefrona/enzimología , Animales , Presión Sanguínea/efectos de los fármacos , Furosemida/farmacología , Hipertensión/etiología , Ratones , Ratones Transgénicos , Ouabaína/farmacología , Rubidio/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/análisis , Simportadores de Cloruro de Sodio-Potasio/análisis , Miembro 1 de la Familia de Transportadores de Soluto 12 , Uromodulina/análisis , Uromodulina/fisiología
7.
Am J Kidney Dis ; 59(3): 452-61, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22277744

RESUMEN

Uromodulin, also known as Tamm-Horsfall protein, is a glycoprotein expressed exclusively by renal tubular cells lining the thick ascending limb of the loop of Henle. Although the physiologic functions of this protein remain elusive, significant progress has been made during the last decade that highlights the importance of uromodulin in the pathophysiology of various diseases, such as medullary cystic kidney disease, urinary tract infections, and nephrolithiasis. Meanwhile, there is renewed interest in the role of uromodulin in kidney injury, both acute and chronic. In this article, we review the existing evidence that supports a role for uromodulin in acute kidney injury, chronic kidney disease, and renal inflammation. Contrary to the conventional view of uromodulin as an instigator in kidney injury, new data from uromodulin knockout mice show a protective role for this protein in acute kidney injury, possibly through downregulating interstitial inflammation. In chronic kidney disease, uromodulin excretion, when adjusted for kidney function, is increased; the significance of this is unclear. Although it has been suggested that uromodulin exacerbates progressive kidney injury, we propose that the elevation in uromodulin secretion is instead reactive to injury and reflects an increase of uromodulin in the renal parenchyma, where it slows the injury process.


Asunto(s)
Lesión Renal Aguda/etiología , Uromodulina/fisiología , Animales , Humanos , Inflamación/etiología , Masculino , Persona de Mediana Edad
8.
Molecules ; 16(3): 2119-34, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21372759

RESUMEN

The molecular basis of polymorphonuclear neutrophil (PMN) phagocytosis-enhancing activity (PEA) by human purified urinary Tamm-Horsfall glyco- protein (THP) has not been elucidated. In this study, we found human THP bound to lactoferrin (LF) and cathepsin G (CG) expressed on the surface of PMN, identified by a proteomic study with MALDI-TOF- LC/LC/mass spectrometric analysis. Pre-incubation of 10% SDS-PAGE electrophoresed PMN lysates with monoclonal anti-LF or anti-CG antibody reduced the binding with THP. To elucidate the signaling pathway of THP on PMN activation, we found THP enhanced ERK1/2 phosphorylation, reduced p38 MAP kinase phosphorylation, but had no effect on DNA binding of the five NF-kB family members in PMN. To further clarify whether the carbohydrate-side chains or protein-core structure in THP molecule is responsible for THP-PEA, THP was cleaved by different degrading enzymes with carbohydrate specificity (neuraminidase and ß-galactosidase), protein specificity (V8 protease and proteinase K) or glycoconjugate specificity (carboxylpeptidase Y and O-sialoglycoprotein endopeptidase). We clearly demonstrated that the intact protein-core structure in THP molecule was more important for THP-PEA than carbohydrate-side chains. Putting these results together, we conclude that THP adheres to surface-expressed LF and CG on PMN and transduces signaling via the MAP kinase pathway to enhance PMN phagocytosis.


Asunto(s)
Catepsina G/metabolismo , Lactoferrina/metabolismo , Sistema de Señalización de MAP Quinasas , Neutrófilos/inmunología , Fagocitosis/fisiología , Uromodulina/fisiología , Humanos , Fosforilación , Unión Proteica , Transducción de Señal , Uromodulina/metabolismo
9.
Cell Physiol Biochem ; 26(3): 311-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20798515

RESUMEN

The glycosylated protein uromodulin is exclusively found in the thick ascending limb cells (TAL) of the kidney, where it is produced on mass and apically targeted, eventually being secreted into the urine. Recently, there has been a renewed interest in this protein due to its ability to interact with the immune system, implicating this protein as a renal inflammatory molecule. Here we investigated the potential role of membrane bound uromodulin on neutrophil adhesion and trans-epithelial migration. The renal tubular epithelial cell line, LLC-PK1, stably transfected with human uromodulin was used to investigate the influence of uromodulin on neutrophil adherence and migration. Uromodulin expression resulted in a significant increase of neutrophil adherence and trans-epithelial migration, in both the apical to basolateral and the basolateral to apical direction. Although uromodulin is GPI anchored and targeted to the apical membrane, we could also observe expression in the basal and lateral membranes domains, which may be responsible for basolateral to apical migration. Furthermore we show that uromodulin binds both the heavy and light chain of IgG, and that IgG enhances neutrophil migration. This study demonstrates that uromodulin can facilitate neutrophil trans-epithelial migration and that this migration can be amplified by co-factors such as IgG.


Asunto(s)
Movimiento Celular , Riñón/inmunología , Neutrófilos/inmunología , Uromodulina/fisiología , Animales , Adhesión Celular , Células Epiteliales/inmunología , Humanos , Inmunoglobulina G/metabolismo , Riñón/citología , Células LLC-PK1 , Neutrófilos/citología , Porcinos , Transfección , Uromodulina/genética , Uromodulina/metabolismo
10.
Urol Res ; 38(6): 429-38, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20658131

RESUMEN

Nephrocalcinosis is the deposition of calcium salts in renal parenchyma and can be intratubular or interstitial. Animal model studies indicate that intratubular nephrocalcinosis is a result of increased urinary supersaturation. Urinary supersaturation with respect to calcium oxalate (CaOx) and calcium phosphate (CaP) are generally achieved at different locations in the renal tubules. As a result experimental induction of hyperoxaluria in animals with CaP deposits does not lead to growth of CaOx over CaP. Interstitial nephrocalcinosis has been seen in mice with lack of crystallization modulators Tamm-Horsfall protein and osteopontin. Sodium phosphate co-transporter or sodiumhydrogen exchanger regulator factor-1 null mice also produced interstitial nephrocalcinosis. Crystals plug the tubules by aggregating and attaching to the luminal cell surface. Structural features of the renal tubules also play a role in crystal retention. The crystals plugging the terminal collecting ducts when exposed to the metastable pelvic urine may promote the formation of stone.


Asunto(s)
Modelos Animales de Enfermedad , Nefrocalcinosis/etiología , Animales , Oxalato de Calcio/metabolismo , Fosfatos de Calcio/metabolismo , Riñón/metabolismo , Ratones , Osteopontina/fisiología , Ratas , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Porcinos , Uromodulina/fisiología
11.
Urol Res ; 38(6): 439-52, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21063698

RESUMEN

Extensive evidence now supports the role of papillary interstitial deposits-Randall's plaques-in the formation of stones in the idiopathic, calcium oxalate stone former. These plaques begin as deposits of apatite in the basement membranes of the thin limbs of Henle's loop, but can grow to become extensive deposits beneath the epithelium covering the papillary surface. Erosion of this covering epithelium allows deposition of calcium oxalate onto this plaque material, and the transition of mineral type and organic material from plaque to stone has been investigated. The fraction of the papilla surface that is covered with Randall's plaque correlates with stone number in these patients, as well as with urine calcium excretion, and plaque coverage also correlates inversely with urine volume and pH. Two animal models--the NHERF-1 and THP-null mice--have been shown to develop sites of interstitial apatite plaque in the renal papilla. In these animal models, the sites of interstitial plaque in the inner medulla are similar to that found in human idiopathic calcium oxalate stone formers, except that the deposits in the mouse models are not localized solely to the basement membrane of the thin limbs of Henle's loop, as in humans. This may be due to the different morphology of the human versus mouse papillary region. Both mouse models appear to be important to characterize further in order to determine how well they mimic human kidney stone disease.


Asunto(s)
Oxalato de Calcio/metabolismo , Modelos Animales de Enfermedad , Cálculos Renales/patología , Fosfoproteínas/fisiología , Intercambiadores de Sodio-Hidrógeno/fisiología , Uromodulina/fisiología , Animales , Cristalización , Humanos , Cálculos Renales/etiología , Cálculos Renales/metabolismo , Ratones , Microscopía Electrónica de Transmisión
12.
Kidney Blood Press Res ; 33(6): 456-75, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21109754

RESUMEN

Uromodulin (UMOD) is a glycoprotein expressed on the luminal surface of the apical membrane of renal tubular epithelial cells forming the thick ascending limb of Henle. Here, UMOD forms filamentous structures probably ensuring water impermeability and the countercurrent gradient. The multidomain structure, cellular topology of UMOD and clinical consequences associated with UMOD dysfunction, however, suggest that it may be involved in other biological processes such as receptor-mediated endocytosis, mechanosensation of urinary flow, Wnt-signaling, cell cycle regulation and planar cell polarity. A specific, but as yet unidentified, protease(s) releases UMOD into the urine, where it probably contributes to colloid osmotic pressure, retards passage of positively charged electrolytes, prevents urinary tract infection and modulates formation of supersaturated salts and their crystals. UMOD expression, biosynthesis and excretion are regulated in a complex manner, and dysregulation is found in a wide range of pathological conditions. It is strongly reduced or absent in cases with mutations in UMOD, renin, HNF1B and other genetic disorders causing autosomal dominant hyperuricemic nephropathy. In contrast, elevated UMOD excretion may be associated with, and thus predictive of, chronic kidney disease. UMOD analysis is therefore of importance in all conditions with renal involvement and may be useful in the proper classification of renal diseases.


Asunto(s)
Hiperuricemia/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Uromodulina/fisiología , Animales , Humanos , Hiperuricemia/clasificación , Hiperuricemia/fisiopatología , Enfermedades Renales/clasificación , Mutación
13.
Kidney Blood Press Res ; 33(5): 393-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20948228

RESUMEN

Uromodulin (Tamm-Horsfall protein) is produced in the kidney by cells of the thick ascending limb and distal tubule. Recent genetic studies suggest a role of uromodulin in chronic kidney disease. Mutations in the UMOD gene cause uromodulin storage disease. They code for amino acid substitutions that lead to misfolding of the molecule and its retention in the endoplasmic reticulum. Single nucleotide polymorphisms in the region of the UMOD gene have been shown to be associated with chronic kidney disease and reduced glomerular filtration rate. These polymorphisms affect uromodulin concentration in the urine, and lower genetically determined urinary uromodulin concentrations seem to protect against renal disease. Chronic kidney disease is associated with higher serum levels of uromodulin. From animal experiments and human studies it is hypothesized that uromodulin entering the renal interstitium either by basolateral secretion or urinary back-leakage in damaged tubuli interacts with and stimulates cells of the immune system and thereby causes inflammation and progression of chronic kidney disease.


Asunto(s)
Enfermedades Renales/etiología , Uromodulina/fisiología , Enfermedad Crónica , Humanos , Enfermedades Renales/patología , Polimorfismo Genético/fisiología , Transporte de Proteínas , Uromodulina/análisis , Uromodulina/genética , Uromodulina/metabolismo
14.
Science ; 369(6506): 1005-1010, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32616672

RESUMEN

Uromodulin is the most abundant protein in human urine, and it forms filaments that antagonize the adhesion of uropathogens; however, the filament structure and mechanism of protection remain poorly understood. We used cryo-electron tomography to show that the uromodulin filament consists of a zigzag-shaped backbone with laterally protruding arms. N-glycosylation mapping and biophysical assays revealed that uromodulin acts as a multivalent ligand for the bacterial type 1 pilus adhesin, presenting specific epitopes on the regularly spaced arms. Imaging of uromodulin-uropathogen interactions in vitro and in patient urine showed that uromodulin filaments associate with uropathogens and mediate bacterial aggregation, which likely prevents adhesion and allows clearance by micturition. These results provide a framework for understanding uromodulin in urinary tract infections and in its more enigmatic roles in physiology and disease.


Asunto(s)
Infecciones Urinarias/metabolismo , Uromodulina/química , Uromodulina/fisiología , Adhesinas Bacterianas/química , Microscopía por Crioelectrón , Glicosilación , Humanos , Ligandos
15.
Nat Rev Nephrol ; 13(9): 525-544, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28781372

RESUMEN

Uromodulin (also known as Tamm-Horsfall protein) is exclusively produced in the kidney and is the most abundant protein in normal urine. The function of uromodulin remains elusive, but the available data suggest that this protein might regulate salt transport, protect against urinary tract infection and kidney stones, and have roles in kidney injury and innate immunity. Interest in uromodulin was boosted by genetic studies that reported involvement of the UMOD gene, which encodes uromodulin, in a spectrum of rare and common kidney diseases. Rare mutations in UMOD cause autosomal dominant tubulointerstitial kidney disease (ADTKD), which leads to chronic kidney disease (CKD). Moreover, genome-wide association studies have identified common variants in UMOD that are strongly associated with risk of CKD and also with hypertension and kidney stones in the general population. These findings have opened up a new field of kidney research. In this Review we summarize biochemical, physiological, genetic and pathological insights into the roles of uromodulin; the mechanisms by which UMOD mutations cause ADTKD, and the association of common UMOD variants with complex disorders.


Asunto(s)
Enfermedades Renales/etiología , Uromodulina/fisiología , Animales , Humanos , Enfermedades Raras/etiología , Uromodulina/genética
16.
G Ital Nefrol ; 32 Suppl 642015.
Artículo en Italiano | MEDLINE | ID: mdl-26479057

RESUMEN

Uromodulin is the most abundant protein excreted in the urine under physiological conditions. It is exclusively expressed in the kidney by epithelial cells lining the thick ascending limb of Henles loop. It is mainly localized at the apical plasma membrane of tubular cells and released through a proteolytic cleavage. Although its function is still elusive it is proposed to have a protective role against urinary tract infection and kidney stone formation, in ion transport and in kidney innate immunity. Mutations in the gene UMOD encoding uromodulin lead to rare autosomal dominant diseases, collectively referred to as uromodulin-associated kidney disease, that are characterized by progressive tubulo-interstitial damage, impaired urinary concentrating ability, hyperuricemia, and progressive renal failure. Recently, genome-wide association studies identified uromodulin as a risk factor for chronic kidney disease and hypertension. Risk variants in the UMOD gene are common in all studied populations and are associated with higher expression and urinary level of the protein.


Asunto(s)
Enfermedades Renales/etiología , Uromodulina/fisiología , Animales , Células Cultivadas , Humanos , Hipertensión/etiología , Mutación , Insuficiencia Renal Crónica/etiología , Uromodulina/química , Uromodulina/genética
17.
PLoS One ; 10(8): e0134764, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26241473

RESUMEN

Enhanced sodium excretion is associated with intrarenal oxidative stress. The present study evaluated whether oxidative stress caused by high sodium (HS) may be involved in calcium oxalate crystal formation. Male rats were fed a sodium-depleted diet. Normal-sodium and HS diets were achieved by providing drinking water containing 0.3% and 3% NaCl, respectively. Rats were fed a sodium-depleted diet with 5% hydroxyl-L-proline (HP) for 7 and 42 days to induce hyperoxaluria and/or calcium oxalate deposition. Compared to normal sodium, HS slightly increased calcium excretion despite diuresis; however, the result did not reach statistical significance. HS did not affect the hyperoxaluria, hypocalciuria or supersaturation caused by HP; however, it increased calcium oxalate crystal deposition soon after 7 days of co-treatment. Massive calcium oxalate formation and calcium crystal excretion in HS+HP rats were seen after 42 days of treatment. HP-mediated hypocitraturia was further exacerbated by HS. Moreover, HS aggravated HP-induced renal injury and tubular damage via increased apoptosis and oxidative stress. Increased urinary malondialdehyde excretion, in situ superoxide production, NAD(P)H oxidase and xanthine oxidase expression and activity, and decreased antioxidant enzyme expression or activity in the HS+HP kidney indicated exaggerated oxidative stress. Interestingly, this redox imbalance was associated with reduced renal osteopontin and Tamm-Horsfall protein expression (via increased excretion) and sodium-dependent dicarboxylate cotransporter NaDC-1 upregulation. Collectively, our results demonstrate that a HS diet induces massive crystal formation in the hyperoxaluric kidney; this is not due to increased urinary calcium excretion but is related to oxidative injury and loss of anticrystallization defense.


Asunto(s)
Oxalato de Calcio/química , Hiperoxaluria/metabolismo , Cálculos Renales/etiología , Túbulos Renales/metabolismo , Natriuresis/fisiología , Estrés Oxidativo/efectos de los fármacos , Sodio en la Dieta/farmacología , Animales , Antioxidantes/metabolismo , Biomarcadores , Citratos/orina , Creatinina/orina , Cristalización , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/fisiología , Dieta Hiposódica , Diuresis/efectos de los fármacos , Inducción Enzimática , Regulación de la Expresión Génica , Hidroxiprolina/toxicidad , Hiperoxaluria/inducido químicamente , Hiperoxaluria/genética , Cálculos Renales/metabolismo , Cálculos Renales/orina , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Dependiente/fisiología , Osteopontina/genética , Osteopontina/fisiología , Ratas , Ratas Wistar , Sodio en la Dieta/administración & dosificación , Sodio en la Dieta/toxicidad , Superóxidos/metabolismo , Simportadores/genética , Simportadores/fisiología , Uromodulina/genética , Uromodulina/fisiología
18.
Gene ; 531(2): 363-9, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23988501

RESUMEN

BACKGROUND: Familial juvenile hyperuricemic nephropathy (FJHN) is an autosomal dominant disorder characterized by hyperuricemia and progressive chronic kidney disease. Uromodulin gene (UMOD) mutations, leading to abnormalities of uromodulin intracellular trafficking contribute to the progress of the disease. METHODS: We did UMOD screening in three Chinese FJHN families. We thus constructed mutant uromodulin express plasmids by site-mutagenesis from wild type uromodulin vector and transfected them into HEK293 (human embryonic kidney) cells. And then we detected uromodulin expression by western blot and observed intracellular distribution by immunofluorescence. RESULTS: We found three heterozygous mutations. Mutation Val109Glu (c.326T/A; p.Val109Glu) and mutation Pro236Gln (c.707C/A; p.Pro236Gln) were newly indentified mutations in two distinct families (family F1 and family F3). Another previously reported UMOD mutation Cys248Trp (c.744C/G; p.Cys248Trp) was detected in family F2. Phenotypes varied both within the same family and between different families. Uromodulin expression is abnormal in the patient biopsy. Functional analysis of mutation showed that mutant types of uromodulin were secreted into the supernatant medium much less when compared with wild type. In mutant type uromodulin transfected cells, intracellular uromodulin localized less in the Golgi apparatus and more in endoplasmic reticulum(ER). CONCLUSIONS: Our results suggested that the novel uromodulin mutations found in the Chinese families lead to misfolded protein, which was retained in the endoplasmic reticulum, finally contributed to the phenotype of FJHN.


Asunto(s)
Gota/genética , Gota/metabolismo , Hiperuricemia/genética , Hiperuricemia/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Mutación , Uromodulina/genética , Uromodulina/metabolismo , Adolescente , Adulto , Pueblo Asiatico/genética , Análisis Mutacional de ADN , Familia , Femenino , Células HEK293 , Humanos , Espacio Intracelular/metabolismo , Masculino , Persona de Mediana Edad , Mutación/fisiología , Linaje , Pliegue de Proteína , Transporte de Proteínas/genética , Uromodulina/fisiología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA