UMOD Porcine

Uromodulin Porcine
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Function of UMOD

UMOD is a glycoprotein with a zona pellucida (ZP) module that enables polymerization into filamentous structures . Key functional insights include:

  • Structural Properties: Cryo-EM studies reveal UMOD forms helical filaments through its ZP domain, with N-glycosylation sites critical for stability .

  • Roles in Kidney Physiology:

    • Inhibits calcium crystallization in renal fluids .

    • Modulates sodium reabsorption via NKCC2 and ROMK transporters, linking it to blood pressure regulation .

    • Facilitates neutrophil migration and cytokine binding (e.g., IL-1, TNF) .

Pathogenic Mutations and Disease

Mutations in UMOD cause autosomal dominant tubulointerstitial kidney disease (ADTKD-UMOD), characterized by endoplasmic reticulum (ER) stress, misfolded protein accumulation, and progressive renal damage . For example:

  • Umod C93F and R186S mutations in mice lead to ER stress, activation of unfolded protein response (UPR) pathways, and tubular atrophy .

  • Allelic variants (e.g., C171Y vs. R186S) exhibit differential aggregation propensities, influencing disease severity .

Porcine Models in Biomedical Research

Pigs are increasingly used as translational models due to physiological similarities with humans . Key applications include:

Table 1: Advantages of Porcine Models in Renal and Genetic Studies

FeaturePorcine Model RelevanceHuman Relevance
Renal AnatomyComparable nephron structure and tubular function Direct applicability to human kidney disease mechanisms
Genetic ModificationCRISPR/Cas9 enables precise modeling of mutations Mimics human genetic disorders (e.g., ADTKD-UMOD)
Disease PhenotypesDevelop hypertension, fibrosis, and CKD Recapitulates human disease progression

Key Findings in Porcine Systems:

  • Regenerative Medicine: Porcine extracellular matrix (ECM) scaffolds promote tissue repair, relevant to UMOD’s role in renal epithelial organization .

  • Transgenic Models: Pigs with mutations in TP53 and KRAS demonstrate oncogenic pathways akin to humans, highlighting their utility in studying UMOD-related ER stress and fibrosis .

Table 2: Comparative UMOD Properties Across Species

PropertyHuman UMODMurine UMODPorcine Inference
Expression SiteThick ascending limb (TAL) cells TAL cells Likely similar expression pattern
GlycosylationHeavily N-glycosylated Conserved glycosylation sites Expected similarity
Disease MutationsADTKD-linked (e.g., C93F, R186S) ER stress, tubular atrophy Potential for analogous phenotypes

Mechanistic Overlaps:

  • ER Stress and Aggregation: Porcine models of protein misfolding (e.g., TP53 mutants) mirror UMOD-associated ER stress, suggesting pigs could model UMOD aggregation pathologies .

  • Therapeutic Testing: Pharmacological agents targeting autophagy (e.g., mTORC1 inhibitors) alleviate UMOD aggregates in mice, a strategy testable in swine .

Future Directions and Applications

  1. Generating UMOD-Mutant Porcine Lines: CRISPR-edited pigs with UMOD mutations could elucidate renal pathophysiology and test allele-specific therapies .

  2. Biomaterial Development: Porcine-derived UMOD polymers may inspire synthetic scaffolds for renal tissue engineering .

  3. Precision Medicine: Linking UMOD risk variants (e.g., rs12917707) to hypertension in pigs could validate NKCC2 inhibitors for salt-sensitive hypertension .

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 caused by uropathogenic bacteria. Defects in Uromodulin expression 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. Although multiple transcript variants might exist, only two have been fully characterized. UMOD regulates circulating cytokine activity by binding to interleukin-1 (IL-1), IL-2, and tumor necrosis factor (TNF) with high affinity.
Description
Porcine Uromodulin, a 97kDa glycoprotein, is synthesized in the thick ascending limb of Henle's loop and the early distal convoluted tubules within the nephron.
Physical Appearance
White, lyophilized powder that has been filtered for purity.
Formulation
The Uromodulin protein was lyophilized from a 0.4 µm filtered solution with a concentration of 0.5 mg/mL 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. Please note that this product is not sterile. Filter it through a sterile filter before using it in cell culture.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to prevent repeated freezing and thawing. The reconstituted protein remains stable at 4°C for a limited period and shows no changes after two weeks at this temperature.
Synonyms
Tamm-Horsfall urinary glycoprotein, THP, FJHN, HNFJ, THGP, MCKD2, ADMCKD2, UMOD, Uromodulin.
Source
Porcine Urine.

Q&A

What is UMOD and why are porcine models valuable for its study?

UMOD (uromodulin) is the gene encoding the most abundant protein in mammalian urine under physiological conditions. The protein is produced in the thick ascending limb of Henle's loop (TALH) in the kidney and undergoes proteolytic cleavage of its ectodomain before urinary excretion. Functionally, uromodulin acts as an inhibitor of calcium crystallization in renal fluids and provides defense against urinary tract infections .

Porcine models are particularly valuable for UMOD research due to their significant physiological and anatomical similarities to humans. Their kidney structure and function closely resembles human kidneys, making them ideal for studying renal disorders . Historical data demonstrates that porcine models correlate well with human responses in biomedical research, particularly for studying complex physiological processes involving proteins like UMOD .

Methodologically, porcine models provide advantages including:

  • Appropriate size for surgical interventions and repeated sampling

  • Similar drug metabolism and pharmacokinetics to humans

  • Comparable renal clearance mechanisms

  • Feasibility for longitudinal studies of progressive renal conditions

What techniques are recommended for isolating UMOD protein from porcine kidney samples?

Isolation of UMOD protein from porcine kidney samples requires a specialized multi-step process to maintain protein integrity:

  • Tissue collection and processing:

    • Focus on the outer medulla where TALH cells are concentrated

    • Immediate preservation in appropriate buffer solutions with protease inhibitors

    • Mechanical homogenization under controlled temperature conditions

  • Extraction protocol:

    • Differential centrifugation to separate cellular components

    • Mild detergent treatment to solubilize membrane-bound forms

    • Precise pH control during extraction to maintain native conformation

  • Purification workflow:

    • Salt precipitation techniques for initial separation

    • Affinity chromatography using specific anti-UMOD antibodies

    • Size-exclusion chromatography for final purification

  • Validation methods:

    • Western blotting with validated anti-UMOD antibodies

    • Mass spectrometry for confirmation of protein identity

    • Analysis using LC-MS/MS for comprehensive profiling

Studies demonstrate that porcine trypsin digestion followed by overnight incubation at 37°C optimizes peptide recovery for downstream mass spectrometry analysis . This approach enables detailed characterization of post-translational modifications that are critical for UMOD functionality.

How should researchers evaluate mitochondrial function in UMOD porcine models?

When evaluating mitochondrial function in UMOD porcine models, researchers should implement a comprehensive approach:

  • Morphological assessment:

    • Electron microscopy to quantify mitochondrial number and structure

    • Assessment of organelle distribution within TAL cells

    • Measurement of mitochondrial-to-cytoplasm ratio

  • Protein expression analysis:

    • Quantification of mitochondrial proteins using proteomics

    • Assessment of OXPHOS and citrate cycle pathway components

    • Evaluation of mitochondrial transcription factors (especially NRF1)

  • Function and dynamics measures:

    • Analysis of mitochondrial fission proteins (particularly FIS1)

    • Assessment of mitochondrial biogenesis pathways

    • Evaluation of energy homeostasis through the LKB1-AMPK pathway

  • Energy metabolism evaluation:

    • Oxygen consumption rate measurements

    • ATP production quantification

    • Assessment of reactive oxygen species generation

Research indicates that UMOD mutations can lead to secondary mitochondrial dysfunction, characterized by reduced mitochondrial protein abundance and impaired energy homeostasis. This dysfunction may result from disturbed post-translational processing of NRF1 and reduced abundance of FIS1, affecting organelle biogenesis and fission .

What are standard protocols for establishing porcine kidney cell cultures for UMOD research?

Establishing porcine kidney cell cultures for UMOD research requires attention to specialized methodologies:

  • Tissue acquisition and processing:

    • Careful dissection of kidney segments enriched for TAL cells

    • Enzymatic digestion with collagenase and DNase

    • Mechanical disaggregation and filtration to obtain single-cell suspensions

  • Cell isolation techniques:

    • Density gradient centrifugation to separate cell populations

    • Immunomagnetic selection using epithelial markers

    • Flow cytometry sorting for specific cell populations

  • Culture establishment and maintenance:

    • Selection of appropriate media supplemented with growth factors

    • Optimization of serum concentration for cell proliferation

    • Maintenance of physiological osmolality and pH

  • Validation of UMOD expression:

    • Immunocytochemistry to confirm UMOD-producing cells

    • RT-PCR to verify UMOD mRNA expression

    • Western blotting to detect UMOD protein synthesis

Recent advances include the development of porcine organoid systems, which provide three-dimensional culture models that better recapitulate the in vivo environment. While not specifically for kidney tissue, the methodology for generating porcine organoids has been established and can be adapted for renal applications .

How do researchers accurately quantify UMOD expression in porcine kidney tissues?

Accurate quantification of UMOD expression in porcine kidney tissues requires multiple complementary approaches:

  • Transcriptional analysis:

    • RT-qPCR with porcine-specific primers

    • RNA sequencing for comprehensive transcriptome profiling

    • In situ hybridization for spatial localization

  • Protein quantification:

    • Western blotting with validated antibodies

    • ELISA for quantitative measurement

    • Mass spectrometry-based proteomics for absolute quantification

  • Histological methods:

    • Immunohistochemistry for localization and semi-quantification

    • Immunofluorescence for co-localization studies

    • Laser capture microdissection for segment-specific analysis

  • Data normalization strategies:

    • Use of established housekeeping genes (GAPDH, β-actin)

    • Total protein normalization for Western blots

    • Inclusion of internal standards for mass spectrometry

When performing quantitative proteomics, LC-MS/MS analysis after trypsin digestion provides comprehensive protein profiling. This approach allows identification of differentially abundant proteins, including UMOD and related pathway components .

How can porcine models be optimized for studying ADTKD-UMOD pathophysiology?

Optimizing porcine models for Autosomal Dominant Tubulointerstitial Kidney Disease-UMOD (ADTKD-UMOD) research requires sophisticated approaches:

  • Genetic engineering strategies:

    • CRISPR/Cas9-mediated introduction of human disease mutations

    • Selection of mutations that mirror specific ADTKD-UMOD variants

    • Validation of mutation effects on protein processing

  • Comprehensive phenotyping:

    • Serial assessment of renal function parameters

    • Urinalysis for UMOD excretion patterns

    • Histopathological examination with specific focus on interstitial fibrosis

  • Molecular characterization:

    • Assessment of ER stress markers (BiP/HSPA5, phosphorylated eIF2α, ATF4, ATF6, CHOP/DDIT3)

    • Evaluation of hypoxia-inducible proteins (HYOU1, TXNDC5, ERO1L)

    • Analysis of mitochondrial protein abundance and function

  • Energy homeostasis evaluation:

    • Assessment of OXPHOS pathway components

    • Quantification of citrate cycle proteins

    • Analysis of LKB1-AMPK pathway activation

Research demonstrates that ADTKD-UMOD involves impaired maturation and secretion of mutant uromodulin in TAL cells, resulting in endoplasmic reticulum stress and unfolded protein response. This leads to secondary mitochondrial dysfunction with reduced abundance of multiple mitochondrial proteins, disturbed biogenesis, and impaired energy homeostasis .

What are the most effective methods for analyzing ER stress in UMOD mutant porcine models?

Analysis of endoplasmic reticulum (ER) stress in UMOD mutant porcine models requires sophisticated methodological approaches:

  • UPR pathway component analysis:

    • Quantification of key ER stress markers (BiP/HSPA5, phosphorylated eIF2α)

    • Assessment of transcription factors (ATF4, ATF6, CHOP/DDIT3)

    • Evaluation of XBP1 splicing as indicator of IRE1 activation

  • Imaging approaches:

    • Electron microscopy to visualize ER expansion and morphology

    • Immunofluorescence co-localization of UMOD with ER markers

    • Live-cell imaging of ER stress using fluorescent reporters

  • Functional assessments:

    • Evaluation of protein folding capacity

    • Analysis of calcium homeostasis

    • Assessment of ER-associated degradation (ERAD) pathway

  • Transcriptomic and proteomic profiling:

    • RNA sequencing to identify UPR gene expression signatures

    • Quantitative proteomics to map ER-resident protein changes

    • Phosphoproteomics to assess UPR signaling cascades

Studies in ADTKD-UMOD models show that mutant UMOD protein accumulates in the ER, triggering increased abundance of ER stress proteins including BiP/HSPA5, phosphorylated eIF2α, ATF4, ATF6, and CHOP/DDIT3. Additionally, hypoxia-inducible proteins with stress survival functions (HYOU1, TXNDC5, ERO1L) show increased abundance, representing critical markers for monitoring ER stress progression .

What techniques are most effective for creating disease-relevant transgenic porcine UMOD models?

Creating disease-relevant transgenic porcine UMOD models requires advanced genetic engineering approaches:

  • Precise gene editing technologies:

    • CRISPR/Cas9 system optimized for porcine applications

    • Homology-directed repair for precise mutation introduction

    • Base editing for specific nucleotide modifications

    • Prime editing for scarless DNA modifications

  • Delivery methods:

    • Microinjection into zygotes

    • Somatic cell nuclear transfer after editing donor cells

    • Lentiviral vector delivery systems

    • Electroporation of ribonucleoprotein complexes

  • Selection and screening strategies:

    • PCR-based genotyping with mutation-specific primers

    • Next-generation sequencing for comprehensive mutation verification

    • Functional validation of mutation effects on protein processing

    • Phenotypic screening for disease manifestations

  • Developmental considerations:

    • Embryo culture optimization

    • Recipient synchronization protocols

    • Early embryo screening techniques

    • Non-invasive monitoring of pregnancies

Porcine models are increasingly recognized as ideal animal models due to their physiological and anatomical similarities to humans. Established protocols for generating porcine pluripotent stem cells (PSCs) provide a foundation for sophisticated genetic modifications, enabling the creation of disease-specific models for UMOD-related disorders .

How can multi-omics approaches enhance understanding of UMOD function in porcine kidney models?

Multi-omics approaches provide comprehensive insights into UMOD function in porcine kidney models:

  • Integrated genomics and transcriptomics:

    • Whole genome sequencing to identify regulatory elements

    • RNA sequencing to map expression patterns

    • ChIP-seq to identify transcription factor binding sites

    • Single-cell transcriptomics to resolve cellular heterogeneity

  • Proteomics strategies:

    • Quantitative LC-MS/MS for global protein profiling

    • Phosphoproteomics to map signaling cascades

    • Glycoproteomics to characterize UMOD modifications

    • Spatial proteomics to map protein localization

  • Metabolomics integration:

    • Targeted analysis of energy metabolism components

    • Lipidomics to assess membrane composition changes

    • Flux analysis to track metabolic pathway activities

    • Integration with mitochondrial function assessments

  • Data integration frameworks:

    • Network analysis to identify regulatory hubs

    • Pathway enrichment to understand biological processes

    • Machine learning for pattern recognition

    • Systems biology modeling of UMOD-related pathways

Research demonstrates the value of proteomics in identifying differentially abundant proteins in UMOD-related disorders. This approach has revealed 212 differentially abundant proteins in TAL-enriched samples from ADTKD-UMOD models compared to controls, providing insights into UPR activation, mitochondrial dysfunction, and impaired energy homeostasis .

What methodologies are recommended for studying UMOD-associated inflammatory responses in porcine models?

Studying UMOD-associated inflammatory responses in porcine models requires specialized methodological approaches:

  • Inflammatory cytokine profiling:

    • Multiplex assays for cytokine quantification

    • RT-qPCR for cytokine gene expression

    • In situ hybridization for spatial localization

    • Single-cell analysis of cytokine production

  • Immune cell characterization:

    • Flow cytometry for immune cell phenotyping

    • Immunohistochemistry for tissue infiltration assessment

    • Laser capture microdissection for region-specific analysis

    • Co-culture systems with immune cells and renal epithelium

  • Signaling pathway analysis:

    • Phosphoproteomic assessment of inflammatory signaling

    • Reporter assays for NF-κB activation

    • Western blotting for MAPK pathway components

    • RNA sequencing for inflammatory gene signatures

  • Functional assays:

    • Neutrophil migration across renal epithelial barriers

    • Macrophage polarization in response to UMOD

    • Complement activation assessment

    • Evaluation of inflammasome activation

Research indicates that UMOD facilitates neutrophil migration across renal epithelia, suggesting an important role in inflammatory processes . Additionally, studies using porcine organoid systems have demonstrated their utility for examining host-microbe interactions, providing a framework that could be adapted for studying UMOD-mediated inflammatory responses .

Product Science Overview

Structure and Production

Uromodulin is a 97kDa glycoprotein . In its porcine form, it is derived from porcine urine and is typically available as a lyophilized (freeze-dried) powder . The protein is produced through proteolytic cleavage of its glycosyl phosphatidylinositol-anchored counterpart situated on the luminal cell surface of the loop of Henle .

Functions

Uromodulin plays several critical roles in renal physiology and systemic health:

  • Inhibition of Calcium Crystallization: It acts as a constitutive inhibitor of calcium crystallization in renal fluids, which helps prevent kidney stone formation .
  • Protection Against Urinary Tract Infections: Uromodulin provides a defense mechanism against urinary tract infections by preventing the adhesion of uropathogenic bacteria to the urinary tract .
  • Regulation of Cytokine Activity: It binds to cytokines such as IL-1, IL-2, and TNF with high affinity, thereby regulating their circulating activity .
Clinical Significance

Defects in uromodulin expression are associated with several autosomal dominant renal disorders, including:

  • Medullary Cystic Kidney Disease-2 (MCKD2): Characterized by juvenile onset of hyperuricemia, gout, and progressive renal failure .
  • Familial Juvenile Hyperuricemic Nephropathy (FJHN): Similar to MCKD2, this disorder also involves hyperuricemia and progressive renal failure .
Stability and Storage

Porcine uromodulin is stable when stored lyophilized at -20°C. After reconstitution, it should be aliquoted to avoid repeated freezing and thawing cycles. The reconstituted protein can be stored at 4°C for a limited period, typically showing no change after two weeks .

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 2025 TheBiotek. All Rights Reserved.