Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
Intern Med ; 63(1): 17-23, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36642527

RESUMEN

Uromodulin, also known as the Tamm-Horsfall protein, is predominantly expressed in epithelial cells of the kidney. It is secreted mainly in the urine, although small amounts are also found in serum. Uromodulin plays an important role in maintaining renal homeostasis, particularly in salt/water transport mechanisms and is associated with salt-sensitive hypertension. It also regulates urinary tract infections, kidney stones, and the immune response in the kidneys or extrarenal organs. Uromodulin has been shown to be associated with the renal function, age, nephron volume, and metabolic abnormalities and has been proposed as a novel biomarker for the tubular function or injury. These findings suggest that uromodulin is a key molecule underlying the mechanisms or therapeutic approaches of chronic kidney disease, particularly nephrosclerosis and diabetic nephropathy, which are causes of end-stage renal disease. This review focuses on the current understanding of the role of uromodulin from a biological, physiological, and pathological standpoint.


Asunto(s)
Cálculos Renales , Insuficiencia Renal Crónica , Humanos , Uromodulina/metabolismo , Riñón/metabolismo , Insuficiencia Renal Crónica/etiología , Homeostasis
3.
Sci Rep ; 13(1): 17815, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37857738

RESUMEN

Tamm-Horsfall protein (THP) is a highly N-glycosylated protein from epithelial cells of the ascending limb of Henle loop. It is secreted into the urine as part of the innate immune response against uropathogenic pathogens. As women are more likely to suffer from urinary tract infections, biomedical studies were conducted to investigate sex-differences in THP excretion, as well as differences in the THP N-glycosylation pattern. A total of 238 volunteers (92 men, 146 women, 69 with hormonal contraceptives) participated in this study, providing urine samples. Women showed a clear tendency to have higher THP concentration and excretion rates than men (p < 0.16). Regular intake of hormonal contraceptives had no significant influence on urinary THP concentration compared to no regular intake. The individual N-glycosylation pattern of THP in urine samples from randomly selected individuals (10 female, 10 male) was investigated after enzymatic release and MS analysis of the oligosaccharides. Female subjects tended to have an increased proportion of oligomannose type N-glycans and non-fucosylated glycans, whereas men had an increased proportion of fucosylated complex-type glycans. The higher level of oligomannose-type glycans in THP from women might be explained by a self-defence mechanism to overcome the higher infections pressure by the female anatomical properties.


Asunto(s)
Asa de la Nefrona , Polisacáridos , Humanos , Masculino , Femenino , Uromodulina/metabolismo , Glicosilación , Asa de la Nefrona/metabolismo , Polisacáridos/metabolismo , Anticonceptivos
4.
EMBO Mol Med ; 15(12): e18242, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-37885358

RESUMEN

Missense mutations in the uromodulin (UMOD) gene cause autosomal dominant tubulointerstitial kidney disease (ADTKD), one of the most common monogenic kidney diseases. The unknown impact of the allelic and gene dosage effects and fate of mutant uromodulin leaves open the gap between postulated gain-of-function mutations, end-organ damage and disease progression in ADTKD. Based on two prevalent missense UMOD mutations with divergent disease progression, we generated UmodC171Y and UmodR186S knock-in mice that showed strong allelic and gene dosage effects on uromodulin aggregates and activation of ER stress and unfolded protein and immune responses, leading to variable kidney damage. Deletion of the wild-type Umod allele in heterozygous UmodR186S mice increased the formation of uromodulin aggregates and ER stress. Studies in kidney tubular cells confirmed differences in uromodulin aggregates, with activation of mutation-specific quality control and clearance mechanisms. Enhancement of autophagy by starvation and mTORC1 inhibition decreased uromodulin aggregates. These studies substantiate the role of toxic aggregates as driving progression of ADTKD-UMOD, relevant for therapeutic strategies to improve clearance of mutant uromodulin.


Asunto(s)
Enfermedades Renales , Riñón , Animales , Ratones , Alelos , Progresión de la Enfermedad , Riñón/metabolismo , Enfermedades Renales/genética , Mutación , Uromodulina/genética , Uromodulina/metabolismo
5.
Hypertension ; 79(11): 2419-2429, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36378920

RESUMEN

The exclusive expression of uromodulin in the kidneys has made it an intriguing protein in kidney and cardiovascular research. Genome-wide association studies discovered variants of uromodulin that are associated with chronic kidney diseases and hypertension. Urinary and circulating uromodulin levels reflect kidney and cardiovascular health as well as overall mortality. More recently, Mendelian randomization studies have shown that genetically driven levels of uromodulin have a causal and adverse effect on kidney function. On a mechanistic level, salt sensitivity is an important factor in the pathophysiology of hypertension, and uromodulin is involved in salt reabsorption via the NKCC2 (Na+-K+-2Cl- cotransporter) on epithelial cells of the ascending limb of loop of Henle. In this review, we provide an overview of the multifaceted physiology and pathophysiology of uromodulin including recent advances in its genetics; cellular trafficking; and mechanistic and clinical studies undertaken to understand the complex relationship between uromodulin, blood pressure, and kidney function. We focus on tubular sodium reabsorption as one of the best understood and pathophysiologically and clinically most important roles of uromodulin, which can lead to therapeutic interventions.


Asunto(s)
Estudio de Asociación del Genoma Completo , Hipertensión , Humanos , Uromodulina/genética , Uromodulina/metabolismo , Hipertensión/genética , Hipertensión/metabolismo , Presión Sanguínea/fisiología , Riñón/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/metabolismo , Cloruro de Sodio/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/genética
6.
Semin Nephrol ; 42(3): 151277, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-36411194

RESUMEN

Tamm-Horsfall protein (THP) is produced exclusively by the kidney, where it is released into both the urine and the circulation. Although the primary form of circulating THP is nonpolymerizing, urinary THP exists as a mix of polymerizing and nonpolymerizing forms. Urinary THP has been shown to play roles in such disparate processes as prevention of urinary tract infections and kidney stone formation, along with the regulation of multiple ion channels within the kidney. The generation of THP knockout mouse models has allowed the investigation of these phenomena and shown a prospective role for circulating THP in ischemia-reperfusion acute kidney injury as well as sepsis. Recent studies have suggested that THP is protective in ischemic injury owing to its inhibition of oxidative stress via the calcium channel transient receptor potential cation channel, subfamily M, member 2 t(TRPM2), and protection in sepsis is at least partially due to THP's promotion of macrophage function.


Asunto(s)
Lesión Renal Aguda , Canales Catiónicos TRPM , Animales , Ratones , Humanos , Uromodulina/metabolismo , Riñón/metabolismo , Lesión Renal Aguda/metabolismo , Ratones Noqueados , Estrés Oxidativo , Canales Catiónicos TRPM/metabolismo
7.
Hypertension ; 79(11): 2409-2418, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35959659

RESUMEN

Uromodulin (or Tamm-Horsfall protein) is a glycoprotein uniquely produced in the kidney by tubular cells of the thick ascending limb of the loop of Henle and early distal tubules. This protein exhibits bidirectional secretion in the urine and in the renal interstitium and circulation. The role of this protein in maintaining renal and systemic homeostasis is becoming increasingly appreciated. Furthermore, perturbations of its functions may play a role in various diseases affecting the kidney and distant organs. In this review, we will discuss important advances in understanding its biology, highlighting the recent discoveries of its secretion and differential precursor processing that generates 2 forms: (1) a highly polymerizing form that is apically excreted in the urine and generates filaments and (2) a nonpolymerizing form that retains a polymerization inhibitory pro-peptide and is released basolaterally in the kidney interstitium and circulation, but can also be found in the urine. We will also discuss factors regulating its production and release, taking into account its intricate physiology, and propose best practices to report its levels. We also discuss breaking advances in its role in hypertension, acute kidney injury and progression to chronic disease, immunomodulation and regulating renal and systemic oxidative stress. We anticipate that this work will be a great resource for researchers and clinicians. This review will highlight the importance of defining what regulates the 2 forms of uromodulin, so that modulation of uromodulin levels and function could become a novel tool in our therapeutic armamentarium against kidney disease.


Asunto(s)
Lesión Renal Aguda , Hipertensión , Humanos , Uromodulina/metabolismo , Riñón/metabolismo , Lesión Renal Aguda/metabolismo , Hipertensión/metabolismo , Biología
8.
Int J Mol Sci ; 23(16)2022 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-36012675

RESUMEN

Uromodulin, a urinary protein synthesized and secreted from the thick ascending limb (TAL) of the loop of Henle, is associated with hypertension through the activation of sodium reabsorption in the TAL. Uromodulin is a potential target for hypertension treatment via natriuresis. However, its biological function in epithelial cells of the distal nephron segment, particularly the collecting duct, remains unknown. Herein, we examined the regulation of uromodulin production during water deprivation in vivo as well as the effect of uromodulin on the activity of the water channel aquaporin-2 (AQP2) in vitro and in vivo using transgenic mice. Water deprivation upregulated uromodulin production; immunofluorescence experiments revealed uromodulin adhesion on the apical surface of the collecting duct. Furthermore, the activation of AQP2 was attenuated in mice lacking uromodulin. Uromodulin enhanced the phosphorylation and apical trafficking of AQP2 in mouse collecting duct cells treated with the vasopressin analog dDAVP. The uromodulin-induced apical trafficking of AQP2 was attenuated via endocytosis inhibitor treatment, suggesting that uromodulin activates AQP2 through the suppression of endocytosis. This study provides novel insights into the cross-talk between TAL and the collecting duct, and indicates that the modulation of uromodulin is a promising approach for diuresis and hypertension treatment.


Asunto(s)
Acuaporina 2 , Hipertensión , Túbulos Renales Colectores , Uromodulina , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Hipertensión/metabolismo , Túbulos Renales Colectores/metabolismo , Ratones , Uromodulina/metabolismo , Privación de Agua
9.
Am J Physiol Renal Physiol ; 323(2): F212-F226, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35759740

RESUMEN

Sepsis is a significant cause of mortality in hospitalized patients. Concomitant development of acute kidney injury (AKI) increases sepsis mortality through unclear mechanisms. Although electrolyte disturbances and toxic metabolite buildup during AKI could be important, it is possible that the kidney produces a protective molecule lost during sepsis with AKI. We have previously demonstrated that systemic Tamm-Horsfall protein (THP; uromodulin), a kidney-derived protein with immunomodulatory properties, falls in AKI. Using a mouse sepsis model without severe kidney injury, we showed that the kidney increases circulating THP by enhancing the basolateral release of THP from medullary thick ascending limb cells. In patients with sepsis, changes in circulating THP were positively associated with a critical illness. THP was also found de novo in injured lungs. Genetic ablation of THP in mice led to increased mortality and bacterial burden during sepsis. Consistent with the increased bacterial burden, the presence of THP in vitro and in vivo led macrophages and monocytes to upregulate a transcriptional program promoting cell migration, phagocytosis, and chemotaxis, and treatment of macrophages with purified THP increases phagocytosis. Rescue of septic THP-/- mice with exogenous systemic THP improved survival. Together, these findings suggest that through releasing THP, the kidney modulates the immune response in sepsis by enhancing mononuclear phagocyte function, and systemic THP has therapeutic potential in sepsis.NEW & NOTEWORTHY Specific therapies to improve outcomes in sepsis with kidney injury have been limited by an unclear understanding of how kidney injury increases sepsis mortality. Here, we identified Tamm-Horsfall protein, known to protect in ischemic acute kidney injury, as protective in preclinical sepsis models. Tamm-Horsfall protein also increased in clinical sepsis without severe kidney injury and concentrated in injured organs. Further study could lead to novel sepsis therapeutics.


Asunto(s)
Lesión Renal Aguda , Sepsis , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/prevención & control , Animales , Modelos Animales de Enfermedad , Riñón/metabolismo , Sepsis/complicaciones , Sepsis/metabolismo , Uromodulina/genética , Uromodulina/metabolismo
10.
Dis Markers ; 2022: 2602717, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35251369

RESUMEN

Maintaining the balance of calcium (Ca2+) metabolism in the kidney is crucial in preventing the formation of kidney stones. Functionally, the microRNA (miRNA) participating in this process needs to be unveiled. We induced NRK-52E cell injury by oxalate treatment. The role of transient receptor potential cation channel subfamily V member 5 (TRPV5) in oxalate-induced cells was studied by TRPV5 overexpression transfection, qRT-PCR, Western blot, MTT, and crystal adhesion detection. After identifying uromodulin (UMOD) expression in injured cells, we confirmed the interaction between TRPV5 and UMOD by coimmunoprecipitation (CoIP) and cell-surface biotinylation assays. The validation of UMOD-regulating TRPV5 in viability, crystal adhesion, and Ca2+ concentration of oxalate-induced cells was performed. Bioinformatics analysis and luciferase assay were used to identify the miRNA-targeting UMOD. The role of the miR-103a-3p-regulating UMOD/TRPV5 axis was detected by rescue experiments. We constructed a rat model with treatment of ethylene glycol (EG) to investigate the miR-103a-3p/UMOD/TRPV5 axis in vivo by hematoxylin-eosin (H&E) staining, Western blot, and immunohistochemistry (IHC). Upregulation of TRPV5 protected NRK-52E cells from oxalate-induced injury by enhancing cell viability and inhibiting CaOx adhesion. UMOD was depleted in oxalate-induced cells and positively interacted with TRPV5. UMOD silencing reversed the effect of TRPV overexpression on oxalate-induced cells. miR-103a-3p targeted UMOD and was mediated in the regulation of the UMOD/TRPV5 axis in oxalate-induced cells. Downregulating miR-103a-3p mitigated EG-induced CaOx deposition in kidney tissues in vivo by activating the UMOD/TRPV5 axis. miR-103a-3p silencing ameliorated CaOx deposition in the rat kidney by activating the UMOD/TRPV5 axis.


Asunto(s)
Oxalato de Calcio/metabolismo , Cálculos Renales/prevención & control , Riñón/metabolismo , MicroARNs/metabolismo , Uromodulina/metabolismo , Animales , Biotinilación , Canales de Calcio/metabolismo , Modelos Animales de Enfermedad , Ratas , Canales Catiónicos TRPV/metabolismo , Regulación hacia Arriba
11.
Nat Struct Mol Biol ; 29(3): 190-193, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35273390

RESUMEN

Glycoprotein 2 (GP2) and uromodulin (UMOD) filaments protect against gastrointestinal and urinary tract infections by acting as decoys for bacterial fimbrial lectin FimH. By combining AlphaFold2 predictions with X-ray crystallography and cryo-EM, we show that these proteins contain a bipartite decoy module whose new fold presents the high-mannose glycan recognized by FimH. The structure rationalizes UMOD mutations associated with kidney diseases and visualizes a key epitope implicated in cast nephropathy.


Asunto(s)
Adhesinas Bacterianas , Fimbrias Bacterianas , Adhesinas Bacterianas/genética , Cristalografía por Rayos X , Proteínas Fimbrias/química , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/química , Fimbrias Bacterianas/metabolismo , Proteínas Ligadas a GPI , Humanos , Manosa/análisis , Uromodulina/análisis , Uromodulina/química , Uromodulina/metabolismo
12.
Pediatr Nephrol ; 37(2): 239-252, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33733301

RESUMEN

The thick ascending limb plays a central role in human kidney physiology, participating in sodium reabsorption, urine concentrating mechanisms, calcium and magnesium homeostasis, bicarbonate and ammonium homeostasis, and uromodulin synthesis. This review aims to illustrate the importance of these roles from a pathophysiological point of view by describing the interactions of the key proteins of this segment and by discussing how recently identified and long-known hereditary diseases affect this segment. The descriptions of two recently described salt-losing tubulopathies, transient antenatal Bartter syndrome and HELIX syndrome, which are caused by mutations in MAGED2 and CLDN10 genes, respectively, highlight the role of new players in the modulation of sodium reabsorption the thick ascending limb.


Asunto(s)
Síndrome de Bartter , Proteínas Adaptadoras Transductoras de Señales/genética , Antígenos de Neoplasias/genética , Síndrome de Bartter/genética , Femenino , Humanos , Riñón , Magnesio/metabolismo , Masculino , Embarazo , Sodio/metabolismo , Uromodulina/genética , Uromodulina/metabolismo
13.
Clin Sci (Lond) ; 135(24): 2749-2761, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34870708

RESUMEN

Uromodulin (UMOD) is the most abundant renal protein secreted into urine by the thick ascending limb (TAL) epithelial cells of the loop of Henle. Genetic studies have demonstrated an association between UMOD risk variants and hypertension. We aimed to dissect the role of dietary salt in renal UMOD excretion in normotension and chronic hypertension. Normotensive Wistar-Kyoto rats (WKY) and stroke-prone spontaneously hypertensive rats (SHRSP) (n=8/sex/strain) were maintained on 1% NaCl for 3 weeks. A subset of salt-loaded SHRSP was treated with nifedipine. Salt-loading in SHRSP increased blood pressure (ΔSBP 35 ± 5 mmHg, P<0.0001) and kidney injury markers such as kidney injury marker-1 (KIM-1; fold change, FC 3.4; P=0.003), neutrophil gelatinase-associated lipocalin (NGAL; FC, 2.0; P=0.012) and proteinuria. After salt-loading there was a reduction in urinary UMOD excretion in WKY and SHRSP by 26 and 55% respectively, compared with baseline. Nifedipine treatment reduced blood pressure (BP) in SHRSP, however, did not prevent salt-induced reduction in urinary UMOD excretion. In all experiments, changes in urinary UMOD excretion were dissociated from kidney UMOD protein and mRNA levels. Colocalization and ex-vivo studies showed that salt-loading increased intracellular UMOD retention in both WKY and SHRSP. Our study provides novel insights into the interplay among salt, UMOD, and BP. The role of UMOD as a cardiovascular risk marker deserves mechanistic reappraisal and further investigations based on our findings.


Asunto(s)
Riñón/fisiopatología , Cloruro de Sodio Dietético/efectos adversos , Uromodulina/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Femenino , Masculino , Nifedipino/farmacología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Uromodulina/orina
14.
Mol Reprod Dev ; 88(10): 686-693, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34590381

RESUMEN

Neural network-based models for protein structure prediction have recently reached near-experimental accuracy and are fast becoming a powerful tool in the arsenal of biologists. As suggested by initial studies using RoseTTAFold or the ColabFold implementation of AlphaFold2, a particularly interesting future development will be the optimization of these computational methods to also routinely yield high-confidence predictions of protein-protein interactions. Here I use AlphaFold2 and ColabFold to investigate the activation and polymerization of uromodulin (UMOD)/Tamm-Horsfall protein, a zona pellucida (ZP) module-containing protein whose precursor and filamentous structures have been previously determined experimentally by X-ray crystallography and cryo-EM, respectively. Despite having no knowledge of the UMOD polymer structure (coordinates for which were neither used for model training nor as template), AlphaFold2/ColabFold are able to recapitulate a crucial conformational change underlying UMOD polymerization, as well as the general organization of protein subunits within the resulting filament. This surprising result is achieved by simply deleting from the input sequence a stretch of residues that correspond to a polymerization-inhibiting C-terminal propeptide. By mimicking in silico the activating effect of propeptide dissociation triggered by site-specific proteolysis of the protein precursor, this example has implications for the assembly of egg coat proteins and the many other molecules that also contain a ZP module. Most importantly, it shows the potential of exploiting machine learning not only to accurately predict the structures of individual proteins or complexes, but also to carry out computational experiments replicating specific molecular events.


Asunto(s)
Polímeros , Zona Pelúcida , Secuencia de Aminoácidos , Aprendizaje Automático , Polímeros/análisis , Polímeros/metabolismo , Uromodulina/análisis , Uromodulina/química , Uromodulina/metabolismo , Zona Pelúcida/metabolismo
16.
J Cell Mol Med ; 25(9): 4316-4325, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33788378

RESUMEN

Uromodulin (UMOD) can bind complement factor H (cFH) and inhibit the activation of complement alternative pathway (AP) by enhancing the cofactor activity of cFH on degeneration of C3b. UMOD, an N-glycans-rich glycoprotein, is expressed in thick ascending limb of Henle's loop where the epithelia need to adapt to gradient change of pH and ion concentration. ELISA-based cofactor activity of cFH and erythrocytes haemolytic assay was used to measure the impact of native and de-glycosylated UMOD on the functions of cFH. The binding assay was performed under different pH and ion concentrations, using ELISA. The levels of sialic acid on UMOD, from healthy controls and patients with chronic kidney disease (CKD), were also detected by lectin-ELISA. It was shown that removal of glycans decreased the binding between UMOD and cFH and abolished the ability of enhancing C3b degradation. In acidic condition, the binding became stronger, but it reduced as sodium concentration increased. A significant decrease of α-2,3 sialic acids on UMOD was observed in CKD patients compared with that of healthy individuals. The sialic acids on UMOD, local pH and sodium concentration could impact the binding capacity between UMOD and cFH and thus regulate the activation of complement AP.


Asunto(s)
Factor H de Complemento/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Insuficiencia Renal Crónica/patología , Uromodulina/metabolismo , Adulto , Estudios de Casos y Controles , Femenino , Hemólisis , Humanos , Concentración de Iones de Hidrógeno , Masculino , Insuficiencia Renal Crónica/metabolismo
17.
Annu Rev Physiol ; 83: 477-501, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33566673

RESUMEN

Uromodulin, a protein exclusively produced by the kidney, is the most abundant urinary protein in physiological conditions. Already described several decades ago, uromodulin has gained the spotlight in recent years, since the discovery that mutations in its encoding gene UMOD cause a renal Mendelian disease (autosomal dominant tubulointerstitial kidney disease) and that common polymorphisms are associated with multifactorial disorders, such as chronic kidney disease, hypertension, and cardiovascular diseases. Moreover, variations in uromodulin levels in urine and/or blood reflect kidney functioning mass and are of prognostic value for renal function, cardiovascular events, and overall mortality. The clinical relevance of uromodulin reflects its multifunctional nature, playing a role in renal ion transport and immunomodulation, in protection against urinary tract infections and renal stones, and possibly as a systemic antioxidant. Here, we discuss the multifaceted roles of this protein in kidney physiology and its translational relevance.


Asunto(s)
Riñón/metabolismo , Uromodulina/metabolismo , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Humanos , Hipertensión/metabolismo , Hipertensión/patología , Riñón/patología , Cálculos Renales/metabolismo , Cálculos Renales/patología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Infecciones Urinarias/metabolismo , Infecciones Urinarias/patología
18.
J Cell Physiol ; 236(7): 5012-5021, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33400289

RESUMEN

Uromodulin (Umod) is the most abundant constituent of urine in humans and exclusively found in the kidney tubular epithelium. However, the specific role of Umod in renal tubulointerstitial injury is yet to be understood. The present study was conducted with aim of investigating the potential therapeutic mechanism of Umod in the regulation of renal tubulointerstitial injury. Protein expression of Umod in renal tubular epithelial cells was measured with the conduction of Western blot analysis. Enzyme-linked immunosorbent assay and immunofluorescence assay were performed to detect the complement activation products and the activation products of surface deposition. The expression of C1q, C2, C4, B factor, C3, C5, H factor, CD46, CD55, C3aR, and C5aR were determined with the use of reverse-transcription quantitative polymerase chain reaction and Western blot analyses. Subsequently, the unilateral ureteral obstruction (UUO) rat model was established. Renal tubulointerstitial injury was assessed with the application of hematoxylin-eosin staining and Masson staining in rats. UUO rats and normal rats were injected with si-NC or si-Umod and complement inhibitor. UUO rats were observed to have serious impairment of kidney tubule, renal tubular dilation, and epithelial atrophy, with downregulated Umod and activated complement pathway. Silencing of Umod resulted in the activation of complement system while promoting interstitial fibrosis in renal tubules. Moreover, addition of complement inhibitor significantly alleviated the renal tubule injury and fibrosis. Collectively, our study suggests that silencing of Umod mediates the complement pathway, exacerbating renal tubulointerstitial injury in rats, which provides insight into the development of novel therapeutic agents for renal tubulointerstitial injury.


Asunto(s)
Activación de Complemento/fisiología , Proteínas del Sistema Complemento/metabolismo , Túbulos Renales/patología , Obstrucción Ureteral/patología , Uromodulina/metabolismo , Animales , Línea Celular , Humanos , Túbulos Renales/lesiones , Masculino , Nefritis Intersticial/patología , Ratas , Ratas Sprague-Dawley , Uromodulina/genética
19.
J Am Soc Mass Spectrom ; 32(2): 436-443, 2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-33301684

RESUMEN

Uromodulin, also known as the Tamm-Horsfall protein or THP, is the most abundant protein excreted in human urine. It is associated with the progression of kidney diseases; therefore, changes in the glycosylation profile of this protein could serve as a potential biomarker for kidney health. The typical glycomics analysis approaches used to quantify uromodulin glycosylation involve time-consuming and tedious glycoprotein isolation and labeling steps, which limit their utility in clinical glycomics assays, where sample throughput is important. Herein, we introduce a radically simplified sample preparation workflow, with direct ESI-MS analysis, enabling the quantification of N-linked glycans that originate from uromodulin. The method omits any glycan labeling steps but includes steps to reduce the salt content of the samples, thereby minimizing ion suppression. The method is effective for quantifying subtle glycosylation differences of uromodulin samples derived from different biological states. As a proof of concept, glycosylation from samples that differ by pregnancy status were shown to be differentiable.


Asunto(s)
Polisacáridos/análisis , Espectrometría de Masa por Ionización de Electrospray/métodos , Uromodulina/metabolismo , Femenino , Fetuínas/metabolismo , Glicosilación , Humanos , Polisacáridos/metabolismo , Polisacáridos/orina , Embarazo , Reproducibilidad de los Resultados , Uromodulina/análisis , Uromodulina/orina
20.
Am J Physiol Renal Physiol ; 319(6): F1043-F1053, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33135481

RESUMEN

The genetic disease Gitelman syndrome, knockout mice, and pharmacological blockade with thiazide diuretics have revealed that reduced activity of the NaCl cotransporter (NCC) promotes renal Mg2+ wasting. NCC is expressed along the distal convoluted tubule (DCT), and its activity determines Mg2+ entry into DCT cells through transient receptor potential channel subfamily M member 6 (TRPM6). Several other genetic forms of hypomagnesemia lower the drive for Mg2+ entry by inhibiting activity of basolateral Na+-K+-ATPase, and reduced NCC activity may do the same. Lower intracellular Mg2+ may promote further Mg2+ loss by directly decreasing activity of Na+-K+-ATPase. Lower intracellular Mg2+ may also lower Na+-K+-ATPase indirectly by downregulating NCC. Lower NCC activity also induces atrophy of DCT cells, decreasing the available number of TRPM6 channels. Conversely, a mouse model with increased NCC activity was recently shown to display normal Mg2+ handling. Moreover, recent studies have identified calcineurin and uromodulin (UMOD) as regulators of both NCC and Mg2+ handling by the DCT. Calcineurin inhibitors paradoxically cause hypomagnesemia in a state of NCC activation, but this may be related to direct effects on TRPM6 gene expression. In Umod-/- mice, the cause of hypomagnesemia may be partly due to both decreased NCC expression and lower TRPM6 expression on the cell surface. This mini-review discusses these new findings and the possible role of altered Na+ flux through NCC and ultimately Na+-K+-ATPase in Mg2+ reabsorption by the DCT.


Asunto(s)
Síndrome de Gitelman/metabolismo , Túbulos Renales Distales/metabolismo , Magnesio/metabolismo , Eliminación Renal , Reabsorción Renal , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Calcineurina/metabolismo , Síndrome de Gitelman/genética , Síndrome de Gitelman/fisiopatología , Humanos , Túbulos Renales Distales/fisiopatología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Canales Catiónicos TRPM/metabolismo , Uromodulina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA