UMOD Human

Uromodulin Human
Shipped with Ice Packs
In Stock

Description

Uromodulin Human Native protein produced from Human Urine, is a glycosylated polypeptide chain containing having a total Mw of 80 kDa on SDS-PAGE.

Product Specs

Introduction
Uromodulin, the most abundant protein in normal urine, is secreted after proteolytic cleavage of its glycosyl phosphatidylinosital-anchored counterpart on the luminal surface of the loop of Henle. It acts as a natural inhibitor of calcium crystallization in renal fluids and protects against urinary tract infections by binding to uropathogenic bacteria. Defects in Uromodulin are linked to autosomal dominant renal disorders like medullary cystic kidney disease-2 (MCKD2) and familial juvenile hyperuricemic nephropathy (FJHN), characterized by early-onset hyperuricemia, gout, and progressive renal failure. While multiple transcript variants may exist, only two are fully characterized. UMOD also regulates cytokine activity by binding to interleukin-1 (IL-1), interleukin-2 (IL-2), and tumor necrosis factor (TNF) with high affinity.
Description
Uromodulin Human Native protein, sourced from human urine, is a glycosylated polypeptide chain with a molecular weight of 80 kDa as determined by SDS-PAGE.
Physical Appearance
White lyophilized powder, filtered for purity.
Formulation
The UMOD protein was lyophilized at a concentration of 0.5 mg/ml from a 0.4 μm filtered solution in deionized water.
Solubility
To create a working stock solution of approximately 0.5 mg/mL, add deionized water to the lyophilized pellet and allow it to dissolve completely. Note: This product is not sterile. Filter it through an appropriate sterile filter before use in cell culture.
Stability
Lyophilized UMOD remains stable at room temperature for 3 weeks but should ideally be stored at 2-8°C. Do not freeze.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms
Tamm-Horsfall urinary glycoprotein, THP, FJHN, HNFJ, THGP, MCKD2, ADMCKD2, UMOD, Uromodulin.
Source
Human Urine.

Q&A

What is the UMOD gene and what protein does it encode?

The UMOD gene encodes uromodulin (also known as Tamm-Horsfall protein), which is the most abundant protein in normal human urine. Uromodulin is expressed almost exclusively in the thick ascending loop of Henle (TAL) of the kidney and undergoes proteolytic cleavage of its ectodomain from its glycosyl phosphatidylinositol-anchored counterpart on the luminal cell surface before being secreted into urine . The protein structure includes three epidermal growth factor-like (EGF-like) domains, a cysteine-rich D8C domain, and a zona pellucida domain .

To investigate UMOD gene structure and function, researchers commonly employ methods including:

  • Sanger sequencing for identification of point mutations

  • Next-generation sequencing for comprehensive gene analysis

  • Structure-function analysis through homology modeling

  • Functional expression studies in cell culture systems

Where is uromodulin primarily expressed in the human body?

Uromodulin expression is highly tissue-specific, being almost exclusively expressed in the thick ascending limb of the loop of Henle in mammalian kidneys . Importantly, recent immunofluorescence studies in human kidney biopsy samples have demonstrated that UMOD is also expressed in the primary cilia of renal tubules . This distinct localization pattern makes uromodulin an important tissue marker for renal physiological studies.

Methodologies for detecting UMOD expression include:

  • Immunohistochemistry and immunofluorescence in renal tissue

  • In situ hybridization for UMOD mRNA detection

  • Quantitative real-time PCR for expression level quantification

  • Western blotting for protein detection in tissue lysates

  • Confocal microscopy for colocalization with cellular compartment markers

What are the known physiological functions of uromodulin?

Uromodulin serves multiple physiological functions in the kidney:

  • Acts as a constitutive inhibitor of calcium crystallization in renal fluids, potentially preventing kidney stone formation

  • Provides defense against urinary tract infections caused by uropathogenic bacteria

  • Participates in ion transport regulation in the loop of Henle, potentially through interaction with the NKCC2 cotransporter (SLC12A1)

  • Contributes to blood pressure homeostasis through effects on sodium reabsorption and plasma volume

  • Forms a protective gel on the luminal surface of renal tubular cells

Experimental methods to study uromodulin functions include:

  • UMOD knockout mouse models

  • In vitro crystallization assays

  • Bacterial adhesion studies

  • Electrophysiological measurements of ion transport

  • Proteomic analyses of protein-protein interactions

What diseases are associated with mutations in the UMOD gene?

Mutations in the UMOD gene are associated with several autosomal dominant renal disorders collectively termed "Uromodulin-associated kidney disease" (UAKD) :

  • Medullary cystic kidney disease type 2 (MCKD2)

  • Familial juvenile hyperuricemic nephropathy (FJHN)

  • Glomerulocystic kidney disease (GCKD)

These disorders are characterized by juvenile-onset hyperuricemia, gout, and progressive renal failure. At least 46 different missense mutations in the UMOD gene have been described . Additionally, common variants in the UMOD gene are associated with risk of hypertension and chronic kidney disease (CKD) in the general population .

Research methods for investigating UMOD-associated diseases include:

  • Exome sequencing for mutation identification

  • Family segregation studies

  • Histopathological analysis of renal biopsies

  • Cellular models of UMOD mutations

  • Transgenic mice expressing pathogenic UMOD variants

How is uromodulin detected in biological samples?

Uromodulin can be detected in various biological samples using different methods:

  • In urine: ELISA (enzyme-linked immunosorbent assay) for quantification

  • In serum/plasma: High-sensitivity ELISA

  • In kidney tissue: Immunohistochemistry and immunofluorescence

  • Gene expression analysis: Quantitative PCR, microarrays, RNA-seq

  • Proteomics: Mass spectrometry for detailed characterization

Commercial ELISA kits, such as the Human Uromodulin (UMOD) ELISA Kit by Invitrogen, allow quantitation of UMOD in human serum, plasma, or cell culture medium . These assays use the sandwich ELISA method where a target-specific antibody is pre-coated in microplate wells. The intensity of the signal produced is directly proportional to the concentration of UMOD present in the original specimen .

What is the relationship between UMOD variants and blood pressure/hypertension?

Genome-wide association studies (GWAS) have identified genetic variants in the UMOD gene associated with blood pressure and hypertension. The variant rs4293393 in the UMOD gene promoter region has been particularly studied, with the ancestral T allele associated with increased risk of hypertension, while the derived C allele has a protective effect .

The proposed mechanism involves regulation of UMOD expression. The risk T allele leads to higher UMOD expression, which increases NKCC2 cotransporter activity in the loop of Henle, resulting in greater sodium reabsorption, plasma volume expansion, and consequently, elevated blood pressure .

Table 1: Association of UMOD variants with blood pressure and hypertension risk

UMOD VariantAlleleEffect on UMOD ExpressionEffect on Blood PressureHypertension Risk
rs4293393T (ancestral)IncreasedIncreasedIncreased
rs4293393C (derived)DecreasedDecreasedReduced
rs13333226G (minor)DecreasedDecreasedReduced
rs13333226A (major)IncreasedIncreasedIncreased

Methods to study this association include:

  • Case-control association studies

  • Prospective cohort studies

  • 24-hour ambulatory blood pressure measurements

  • Animal models with altered UMOD expression

  • Pharmacogenomic studies with diuretics

How do UMOD polymorphisms associate with kidney function and chronic kidney disease?

G-allele carriers at UMOD rs4293393 exhibit significantly higher estimated glomerular filtration rate (eGFR) values compared to non-carriers, and a lower risk of eGFR < 60 mL/min/1.73 m², which is the threshold for defining chronic kidney disease (CKD) .

In the Berlin Aging Study II (BASE-II), G-allele carriers showed:

  • AA genotype: eGFR 76.4 ml/min/1.73 m² (CI: 75.7-77.2)

  • AG genotype: eGFR 78.4 ml/min/1.73 m² (CI: 77.3-79.5)

  • GG genotype: eGFR 78.5 ml/min/1.73 m² (CI: 75.4-81.7); P = 0.010

Carriers of the AG genotype had a reduced risk of eGFR < 60 mL/min/1.73 m² (OR: 0.63, CI: 0.41-0.97, P = 0.033) .

It is postulated that lower UMOD expression (associated with the G allele) could reduce tubulointerstitial inflammation, decrease renal fibrosis, and prevent kidney function deterioration over time.

Methods to evaluate this association include:

  • Genetic association studies in different populations

  • Longitudinal kidney function monitoring

  • Urinary biomarkers of tubular damage

  • Advanced renal imaging (functional MRI)

  • Single-cell models to study cell-type specific effects

What experimental methods are most effective for studying UMOD expression and function in cellular and animal models?

For studying UMOD expression and function, various experimental methods are utilized:

Cellular models:

  • Human kidney cell lines (HEK293, HK-2)

  • Primary TAL cell cultures

  • Kidney organoids derived from iPSCs

  • Transient and stable expression systems

  • CRISPR/Cas9 gene editing to generate specific mutations

Animal models:

  • UMOD knockout mice

  • Transgenic mice overexpressing UMOD

  • Knock-in mice with specific UMOD mutations

  • Zebrafish for developmental studies

  • Models of nephrolithiasis and urinary tract infection

Analysis techniques:

  • High-resolution microscopy (confocal, super-resolution)

  • Electron microscopy for ultrastructure

  • Transcriptomic analysis (RNA-seq, scRNA-seq)

  • Proteomics and metabolomics

  • Functional studies of ion transport (patch-clamp, Ussing chambers)

Table 2: Comparison of experimental models for studying UMOD

ModelAdvantagesLimitationsPrimary Applications
Cell linesEasy manipulation, high reproducibilityDo not fully replicate in vivo physiologyMolecular mechanisms, protein interactions
Primary culturesHigher physiological relevanceLimited lifespan, variabilityProtein expression and trafficking studies
Organoids3D architecture, multiple cell typesIncomplete maturityRenal development, mutation effects
KO/KI miceSystemic evaluation, in vivo studiesSpecies differencesRenal physiology, disease pathogenesis
ZebrafishRapid development, transparencyLess complex renal systemDevelopmental studies, high-throughput screens

Is there evidence of evolutionary selection for UMOD variants in human populations?

There is evidence of evolutionary selection for UMOD variants in human populations. Studies have shown that:

  • The T allele of rs4293393 is the ancestral allele, present in all chimpanzee subspecies and other nonhuman primates analyzed .

  • Surprisingly, archaic hominids (Neanderthals and Denisovans) were homozygous for the derived C allele, whereas anatomically modern human samples spanning a period 3900-45,000 years ago only showed the ancestral T allele .

  • Haplotype analysis suggests that the derived C allele started increasing in frequency approximately 15,000 years ago, with limited differences among African and non-African populations .

This evolutionary dynamic might be related to pathogen-driven selection pressure, as uromodulin plays a role in defense against urinary tract infections. The variant that increases UMOD expression might provide greater protection against specific pathogens that emerged with changes in human lifestyle (such as agriculture and animal domestication).

Methods to study evolutionary selection:

  • Extended haplotype analysis

  • Selection tests based on allele frequency differentiation

  • Linkage disequilibrium analysis

  • Ancient DNA sequencing

  • Demographic and coalescent modeling

What are the molecular mechanisms linking UMOD with regulation of the NKCC2 cotransporter?

Studies have identified strong positive coexpression between UMOD and the Na-K-2Cl cotransporter (NKCC2, encoded by SLC12A1) in the thick ascending limb of Henle. This correlation was validated by qRT-PCR in 84 human samples, showing a highly significant correlation .

Proposed molecular mechanisms include:

  • Physical interaction: UMOD might directly interact with NKCC2 at the apical plasma membrane, stabilizing the cotransporter and increasing its activity.

  • Transcriptional regulation: UMOD and NKCC2 might be under the control of common transcription factors that regulate their coordinated expression.

  • Intracellular signaling: UMOD could modulate signaling pathways that regulate NKCC2 phosphorylation and activation.

  • Membrane microdomain organization: UMOD, as a GPI-anchored protein, might participate in the formation of lipid rafts that concentrate NKCC2 and its regulators.

Methods to study UMOD-NKCC2 interaction:

  • Co-immunoprecipitation for protein-protein interaction

  • Proximity Ligation Assay for spatial proximity (<40 nm)

  • FRET/BRET for direct interaction (<10 nm)

  • Confocal microscopy for colocalization

  • Electrophysiology for NKCC2 function

  • RNA-seq for expression correlation

How do UMOD mutations affect cellular localization and protein trafficking?

Pathogenic mutations in UMOD primarily affect cellular localization and protein trafficking:

  • Endoplasmic reticulum (ER) retention: Most UMOD mutations cause protein misfolding, resulting in ER retention and reduced urinary UMOD secretion.

  • Delayed ER-to-Golgi trafficking: Mutations alter protein conformation, delaying transport through the secretory pathway .

  • Intracellular aggregate formation: Misfolded UMOD proteins can form aggregates that activate the unfolded protein response (UPR) and increase the rate of apoptosis .

  • Reduced localization to primary cilia: Renal biopsies from patients with UMOD mutations show decreased UMOD expression in primary cilia of renal tubules .

Methods to study UMOD trafficking:

  • Live-cell real-time microscopy

  • Immunofluorescence with cellular compartment markers

  • Subcellular fractionation and Western blotting

  • Pulse-chase with radiolabeled proteins

  • Electron microscopy for cellular ultrastructure

What implications does UMOD expression in primary cilia have for renal disease pathogenesis?

The discovery that UMOD is expressed in primary cilia of renal tubules has important implications for renal disease pathogenesis:

  • Connection to ciliopathies: It suggests that UMOD-associated kidney diseases (UAKD) might be considered part of the ciliopathy spectrum, a group of disorders caused by primary cilia dysfunction .

  • Ciliary signaling: Primary cilia are sensory organelles that detect fluid flow and mediate several signaling pathways (Hedgehog, Wnt, etc.). UMOD might participate in these signaling pathways.

  • Mechanosensation: UMOD presence in cilia could influence fluid flow detection and transduction of mechanical signals into cellular responses.

  • Renal development: Ciliary signaling pathways are critical for normal kidney development, and alterations in UMOD might contribute to developmental anomalies.

Methods to study UMOD in primary cilia:

  • Immunofluorescence with confocal microscopy

  • Electron microscopy

  • Proteomic analysis of the ciliome

  • Ciliary function studies (e.g., intracellular Ca²⁺ measurement)

  • 3D models of ciliated renal epithelium

How can contradictory data on UMOD variant effects across different populations be reconciled?

Studies on UMOD variants sometimes show contradictory results across different populations:

  • Genetic heterogeneity: Differences in genetic background may modify UMOD variant effects through modifier genes.

  • Gene-environment interactions: Environmental factors such as diet (especially salt intake), climate, and endemic infections may modulate UMOD variant effects.

  • Linkage disequilibrium structure: The studied variants might be in linkage disequilibrium with different causal variants across populations.

  • Statistical power: Studies of different sizes may have different abilities to detect associations, especially for small effects.

  • Heterogeneous phenotypes: Differences in phenotype definition or measurement methods may contribute to discrepant results.

Methodologies to reconcile contradictory data:

  • Meta-analysis with subgroup analysis

  • Gene-environment interaction analysis

  • Replication studies in well-characterized populations

  • Extended haplotype analysis

  • Functional studies to validate biological mechanisms

  • Causal modeling to identify common pathways

What advanced imaging techniques are useful for studying UMOD subcellular localization?

Several advanced imaging techniques are used to study UMOD subcellular localization:

  • Confocal microscopy: Allows visualization of UMOD colocalization with specific cellular compartment markers (ER, Golgi, plasma membrane, cilia).

  • Super-resolution microscopy:

    • STED (Stimulated Emission Depletion): ~50 nm resolution

    • PALM/STORM (Photo-Activated Localization Microscopy/Stochastic Optical Reconstruction Microscopy): ~20 nm resolution

    • SIM (Structured Illumination Microscopy): ~100 nm resolution

  • Electron microscopy:

    • TEM (Transmission Electron Microscopy) with immunogold labeling

    • SEM (Scanning Electron Microscopy) for cell surfaces

    • Cryo-EM for molecular structures at near-atomic resolution

  • Live-cell real-time microscopy:

    • FRAP (Fluorescence Recovery After Photobleaching) for protein dynamics

    • Fluorescent fusion proteins (GFP, mCherry) for real-time trafficking

    • FRET sensors for protein-protein interactions

These techniques allow researchers to precisely localize UMOD within cellular compartments and study its dynamic behavior, providing insights into normal trafficking and pathological alterations in disease states.

What are the key methodological considerations for designing genetic association studies involving UMOD?

When designing genetic association studies involving UMOD, several methodological aspects should be considered:

  • Variant selection:

    • Candidate SNPs vs. genome-wide panels

    • Coverage of regulatory variants (promoter, enhancers)

    • Rare vs. common variants

    • Sequencing vs. genotyping

  • Phenotype definition:

    • Precise blood pressure measurements (24h ABPM)

    • Standardized kidney function assessment (eGFR, cystatin C)

    • Urinary and serum UMOD quantification

    • Intermediate phenotypes (sodium excretion, uric acid)

  • Study design:

    • Cross-sectional vs. longitudinal studies

    • Sample size and power calculations

    • Homogeneous vs. heterogeneous populations

    • Environmental controls (diet, medication)

  • Statistical analysis:

    • Correction for multiple testing

    • Population stratification

    • Haplotype vs. individual variant analysis

    • Modeling of gene-gene and gene-environment interactions

Table 3: Recommendations for genetic association studies with UMOD

AspectRecommendationRationale
Variants to studyInclude rs4293393, rs13333226 and haplotype tagsVariants with previous evidence of association
Sample size>5000 to detect modest effectsBased on effect sizes observed in previous GWAS
Phenotype measurementRepeated measurements, standardized protocolsReduces measurement error, increases power
CovariatesAge, sex, BMI, medication, ancestryFactors influencing kidney function and blood pressure
BiomarkersQuantify urinary and serum UMODIntermediate phenotypes connecting genotype with clinical outcome
ReplicationAt least one independent cohortReduces false positives

Product Science Overview

Gene and Protein Structure

The UMOD gene is located on chromosome 16 (16p12.3) in humans . The gene encodes a protein that is initially synthesized as a precursor with a signal peptide, which is cleaved to produce the mature form of the protein . The mature protein has a molecular weight of approximately 85 kDa but can form large aggregates in urine .

Expression and Localization

Uromodulin is produced almost exclusively by the epithelial cells lining the thick ascending limb (TAL) of the loop of Henle and the early distal convoluted tubule (DCT) in the kidney . It is released into both the urine and the interstitial space/circulation .

Biological Functions

Uromodulin plays several critical roles in maintaining urinary and systemic homeostasis . Some of its key functions include:

  • Modulating renal ion channel activity: Uromodulin influences the activity of various ion channels in the kidney, which is essential for maintaining electrolyte balance .
  • Salt and water balance: It helps regulate the reabsorption of salt and water in the kidney, contributing to overall fluid balance .
  • Immune response: Uromodulin has been shown to play a role in the immune response, particularly in modulating inflammation and protecting against urinary tract infections .
  • Mineral crystallization: It inhibits the formation of kidney stones by preventing the crystallization of minerals in the urine .
  • Bacterial adhesion: Uromodulin can bind to bacteria, preventing them from adhering to the urinary tract and causing infections .
Clinical Significance

Mutations in the UMOD gene are associated with a group of inherited kidney diseases, including autosomal dominant tubulointerstitial kidney disease (ADTKD) . Altered expression of uromodulin has also been linked to increased risks of urinary tract infections, kidney stones, hypertension, hyperuricemia, and both acute and chronic kidney diseases .

Research and Future Directions

Despite the extensive research on uromodulin, there are still many aspects of its regulation and function that remain unclear . Ongoing studies aim to better understand how uromodulin is regulated by external and intrinsic factors, how its deficiency leads to various pathophysiological conditions, and how it can be used as a biomarker or therapeutic target to improve patient outcomes .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.