SCGB1A1 Human

Uteroglobin Human Recombinant
Shipped with Ice Packs
In Stock

Description

Mechanisms of Action

SCGB1A1 exerts diverse biological effects through multiple pathways:

  • Anti-inflammatory Activity: Inhibits phospholipase A2 (PLA2), reducing proinflammatory prostaglandins and cytokines like IL-1β, IL-6, and TNF-α .

  • Immune Regulation: Modulates alveolar macrophage responses to microbial stimuli, suppressing chemotaxis and cytokine surges .

  • Tumor Suppression: Downregulation correlates with lung cancer progression and head/neck squamous cell carcinoma (HNSCC) .

Respiratory Diseases

  • Asthma and COPD:

    • Serum SCGB1A1 levels are reduced in asthma (1.6–2.4 ng/mL vs. 5.1 ng/mL in healthy controls) .

    • The A38G polymorphism (rs3741240) in SCGB1A1 is linked to asthma severity .

    • Biomass smoke exposure decreases SCGB1A1 levels, accelerating COPD progression .

Study CohortSerum SCGB1A1 (ng/mL)Correlation with FEV₁ (r-value)
Healthy Controls5.1 (4.5–7.2)0.7–0.8
Biomass Smoke COPD1.6 (1.3–2.4)0.7–0.8
Tobacco Smoke COPD1.8 (1.4–2.5)0.7–0.8

Therapeutic Potential

  • Biomarker Utility:

    • Predicts HNSCC prognosis and drug sensitivity .

    • Correlates with immune checkpoint regulation (e.g., PD-L1, CTLA-4) .

  • Recombinant Therapy: SCGB1A1’s acid/heat resistance and small size make it viable for targeted delivery via oncolytic viruses .

Research Advancements

  • In Vitro Studies: Exogenous SCGB1A1 reduces cytokine release from macrophages by 40–60% .

  • Genetic Models: Scgb1a1⁻/⁻ mice exhibit altered immune responses and club cell morphology .

  • Pan-Cancer Analysis: SCGB1A1 is downregulated in 85% of tumors compared to normal tissues .

Future Directions

Ongoing research focuses on:

  • SCGB1A1’s role in TGF-β signaling and extracellular matrix interactions .

  • Clinical trials evaluating recombinant SCGB1A1 for COPD and asthma .

Product Specs

Introduction
Uteroglobin (SCGB1A1), a member of the Secretoglobin (SCGBs) family, is a versatile protein with anti-inflammatory and anti-tumorigenic properties. It achieves this by binding to small hydrophobic molecules like phospholipids and prostaglandins. Uteroglobin plays a crucial role in various biological processes, including inflammation reduction, phospholipase A2 inhibition, and the sequestration of hydrophobic ligands. Primarily produced by Clara cells (non-ciliated, non-mucous secretory cells abundant in lung bronchioles) and other epithelia interacting with the external environment, SCGB1A1 not only sequesters pro-inflammatory mediators and carcinogens but also participates in inhibiting cell migration and invasion, platelet aggregation, and T cell differentiation. Notably, defects in the SCGB1A1 gene are linked to an increased susceptibility to asthma.
Description
Recombinant Human Uteroglobin, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 70 amino acids. With a molecular weight of 15.8 kDa, SCGB1A1 undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The Uteroglobin protein was lyophilized from a 0.2µm filtered solution concentrated in PBS with a pH of 7.4.
Solubility
For reconstitution of lyophilized SCGB1A1, sterile 18M-cm H₂O at a concentration of at least 100µg/ml is recommended. This solution can be further diluted with other aqueous solutions.
Stability
Lyophilized Uteroglobin remains stable at room temperature for up to 3 weeks; however, storage in desiccated conditions below -18°C is recommended. After reconstitution, SCGB1A1 should be stored at 4°C for 2-7 days. For long-term storage, adding a carrier protein (0.1% HSA or BSA) is advisable. It is important to avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 98.0% as determined by: (a) Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) analysis and (b) Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analysis.
Biological Activity
The ED₅₀, determined by the immobilized protein's ability to support the adhesion of A549 human lung carcinoma cells, is less than 5.0µg/ml. This corresponds to a specific activity exceeding 200 IU/mg.
Synonyms
Uteroglobin, Clara cell phospholipid-binding protein, CCPBP, Clara cells 10 kDa secretory protein, CC10, Secretoglobin family 1A member 1, Urinary protein 1, UP-1, UP1, Urine protein 1, SCGB1A1, CCSP, UGB, CC16.
Source
Escherichia Coli.
Amino Acid Sequence
EICPSFQRVI ETLLMDTPSS YEAAMELFSP DQDMREAGAQ LKKLVDTLPQ KPRESIIKLM EKIAQSSLCN.

Q&A

What is SCGB1A1 and where is it primarily expressed in humans?

SCGB1A1 (Secretoglobin Family 1A Member 1) is a small secreted protein primarily produced by club cells (formerly known as Clara cells) in the respiratory epithelium. It is abundantly expressed in the human airway epithelium and has been detected in various biological fluids including bronchoalveolar lavage (BAL) fluid, sputum, and serum . The protein plays a crucial role in maintaining airway homeostasis through its anti-inflammatory, immunomodulatory, and surfactant-related functions. Interestingly, SCGB1A1 gene expression has also been detected in human alveolar macrophages using real-time PCR, suggesting a broader cellular distribution than initially thought . The protein's expression is regulated by various factors, including transcription factors such as FOXA2, which has been shown to be essential for SCGB1A1 expression at baseline . Research indicates that SCGB1A1 is part of a complex regulatory network in the lungs that contributes to protection against respiratory disorders.

How do SCGB1A1 levels differ between healthy individuals and those with respiratory conditions?

Human SCGB1A1 protein has been consistently shown to be significantly reduced in bronchoalveolar lavage (BAL), sputum, and serum from individuals with asthma compared to healthy individuals . This reduction appears to occur at the transcriptional level, with SCGB1A1 mRNA expression significantly reduced by approximately 53% in epithelial cells from subjects with asthma compared to healthy controls . The mechanism behind this reduction involves the downregulation of FOXA2, a transcription factor essential for SCGB1A1 expression . Multiple studies have confirmed lower levels of SCGB1A1 in BAL, serum, and urine samples from individuals with asthma compared to healthy control subjects . A genetic polymorphism, A38G (rs3741240), located 38 bp downstream of the transcription start site of SCGB1A1, has been identified as a potential risk factor for the development and severity of asthma . The A allele of this polymorphism is associated with reduced levels of SCGB1A1 in BAL and circulation, suggesting that the decreased protein levels observed in asthmatic individuals may originate from reduced cellular production .

How does SCGB1A1 contribute to asthma pathophysiology?

SCGB1A1's role in asthma pathophysiology appears to be primarily protective, with its reduction contributing to disease development and progression. Studies in animal models have demonstrated that compared to wild-type mice, Scgb1a1 knockout mice exhibited increased airway hyperreactivity and inflammation when exposed to ovalbumin, confirming the anti-inflammatory role of SCGB1A1 in protection against asthma phenotypes . At the molecular level, the reduction of SCGB1A1 in asthmatic airways is mediated by the downregulation of FOXA2, an essential transcription factor for SCGB1A1 expression . This reduction can be triggered by T-helper cell type 2 (Th2) cytokines, specifically IL-4 and IL-13, which repress epithelial expression of both SCGB1A1 and FOXA2 . Additionally, human rhinovirus infection similarly reduces expression of these two genes, suggesting that FOXA2 may be the common regulator of SCGB1A1 . The causal relationship between reduced FOXA2 and SCGB1A1 repression was established by demonstrating that FOXA2 is required for SCGB1A1 expression at baseline, and FOXA2 overexpression can restore repressed SCGB1A1 expression in IL-13–treated or rhinovirus-infected cells .

How is SCGB1A1 expression regulated at the transcriptional level?

SCGB1A1 expression is regulated by a complex network of transcription factors, with FOXA2 playing a central role. Research has demonstrated that FOXA2 is required for SCGB1A1 expression at baseline conditions, establishing a direct regulatory relationship . When FOXA2 is overexpressed, it drives both the promoter activity and expression of SCGB1A1, highlighting its importance in transcriptional activation . This regulation becomes particularly relevant in disease states such as asthma, where both SCGB1A1 and FOXA2 are downregulated . The transcriptional repression of SCGB1A1 can be induced by T-helper cell type 2 (Th2) cytokines, specifically IL-4 and IL-13, which simultaneously repress epithelial expression of both SCGB1A1 and FOXA2 . Similarly, human rhinovirus infection reduces the expression of both genes, further supporting FOXA2 as the common regulator of SCGB1A1 . The functional significance of this regulation was demonstrated when FOXA2 overexpression was able to restore the repressed SCGB1A1 expression in IL-13–treated or rhinovirus-infected cells . Additionally, genetic factors influence SCGB1A1 expression, with the A38G (rs3741240) polymorphism 38 bp downstream of the transcription start site being associated with reduced SCGB1A1 levels and increased asthma risk .

What signaling pathways are affected by SCGB1A1 in immune and epithelial cells?

SCGB1A1 influences multiple signaling pathways in both immune and epithelial cells, contributing to its anti-inflammatory and immunomodulatory functions. In alveolar macrophages (AMs), SCGB1A1 sufficiency is associated with the upregulation of 37 biological pathways in early adult mice compared to weaning, with 30 of these pathways directly involved in antigen presentation, anti-viral immunity, and inflammation . Under SCGB1A1 deficiency, these pathways are significantly downregulated compared to age-matched SCGB1A1-sufficient counterparts, suggesting that SCGB1A1 shapes AM-mediated inflammation and immune responses . In vitro experiments have demonstrated that exogenous supplementation of SCGB1A1 protein significantly reduces AM responses to microbial stimuli, effectively blunting the release of cytokines and chemokines including IL-1β, IL-6, IL-8, MIP-1α, TNF-α, and MCP-1 . These effects were observed when AMs were stimulated with various Toll-like receptor (TLR) agonists, including TLR2 (heat-killed Listeria monocytogenes), TLR4 (Lipopolysaccharide from Escherichia coli K12), and TLR5 (Salmonella typhimurium Flagellin) . For example, IL-1β levels in LPS-stimulated AMs were reduced from 52.660 ± 1.719 pg/mL to 31.490 ± 1.588 pg/mL (p < 0.001) with SCGB1A1 supplementation . In cancer contexts, SCGB1A1 regulates multiple key cancer-related signaling pathways, including extracellular matrix receptor interaction, transforming growth factor-β, and tumor metabolism signaling pathways .

What are the current methods for measuring SCGB1A1 levels in clinical samples?

Researchers employ several methods to quantify SCGB1A1 levels in various clinical samples, each with specific advantages depending on the research question. For protein detection in biological fluids, enzyme-linked immunosorbent assays (ELISAs) are commonly used to measure SCGB1A1 in bronchoalveolar lavage (BAL) fluid, sputum, serum, and urine samples from patients with respiratory conditions such as asthma . Western blotting provides another protein detection approach, particularly useful for semi-quantitative analysis in tissue homogenates. At the transcript level, real-time quantitative PCR (RT-qPCR) is employed to measure SCGB1A1 mRNA expression in brushed human airway epithelial cells and alveolar macrophages, as demonstrated in studies comparing expression between asthmatic and healthy individuals . For broader transcriptomic analyses, researchers utilize RNA-sequencing on purified cell populations, such as alveolar macrophages isolated from mouse models with different Scgb1a1 expression profiles . This approach allows for high-throughput gene expression analysis and pathway identification. Importantly, the selection of clinical samples should be carefully considered, as some studies focusing on gene expression profiles from brushed human bronchial epithelial cells are preferred over BAL cells or biopsies due to significant contamination from non-epithelial cell types in the latter .

How can researchers effectively study SCGB1A1 function in vitro and in vivo?

Researchers employ complementary in vitro and in vivo approaches to comprehensively investigate SCGB1A1 function. For in vitro studies, cultured human airway epithelial cells are commonly used to examine the regulation of SCGB1A1 expression under various conditions, such as treatment with T-helper cell type 2 cytokines (IL-4 and IL-13) or rhinovirus infection . These systems allow researchers to establish causal relationships, as demonstrated when FOXA2 overexpression restored repressed SCGB1A1 expression in IL-13–treated or rhinovirus-infected cells . Alveolar macrophage cultures provide another valuable in vitro model, where flow-sorted AMs can be plated and incubated with Toll-like receptor (TLR) agonists in the presence or absence of recombinant SCGB1A1 protein to assess its effects on inflammatory responses . For in vivo investigations, Scgb1a1 knockout mice serve as a powerful tool to examine the physiological and pathological consequences of SCGB1A1 deficiency . These models have revealed that compared to wild-type mice, Scgb1a1 knockout mice exhibit increased airway hyperreactivity and inflammation when exposed to ovalbumin, confirming SCGB1A1's protective role against asthma phenotypes . Age-dependent studies in these models further demonstrate how SCGB1A1 deficiency affects alveolar macrophage development and function over time, with significant transcriptomic changes observed particularly at 12 weeks of age .

How does SCGB1A1 influence alveolar macrophage development and function?

SCGB1A1 plays a crucial role in shaping alveolar macrophage (AM) development and function across different life stages. Research using murine models has revealed age-dependent effects of SCGB1A1 on AM phenotype and gene expression . While AMs from SCGB1A1-deficient and wild-type mice appear phenotypically similar at early time points (4 and 8 weeks), a unique population with CD11c high and Siglec-F low expression begins to emerge in SCGB1A1-knockout mice by 12 weeks . By 40 weeks, this population represents approximately 7% of cells in knockout mice compared to just 1% in age-matched wild-type controls . Transcriptomic analyses have revealed that SCGB1A1 sufficiency is associated with the upregulation of 37 biological pathways in early adult mice compared to weaning, with 30 of these pathways directly involved in antigen presentation, anti-viral immunity, and inflammation . Under SCGB1A1 deficiency, these pathways are significantly downregulated compared to age-matched SCGB1A1-sufficient counterparts . Furthermore, AMs from SCGB1A1-deficient mice show an early activation of inflammatory pathways compared to those from SCGB1A1-sufficient mice, suggesting that SCGB1A1 helps regulate the timing and magnitude of inflammatory responses .

What is the potential for SCGB1A1 as a therapeutic target in respiratory diseases?

SCGB1A1 holds significant promise as a therapeutic target in respiratory diseases, particularly asthma, due to its demonstrated anti-inflammatory properties and reduced expression in disease states. Research has shown that SCGB1A1 is significantly reduced in asthmatic airways, likely by the action of T-helper cell type 2 cytokines and/or rhinovirus infection . This reduction contributes to enhanced airway inflammation and hyperreactivity, as demonstrated in Scgb1a1 knockout mice, which exhibit exacerbated asthma phenotypes when exposed to ovalbumin compared to wild-type mice . The therapeutic potential of SCGB1A1 is further supported by in vitro studies showing that exogenous supplementation of SCGB1A1 protein significantly reduces inflammatory responses in alveolar macrophages stimulated with microbial products . Specifically, SCGB1A1 supplementation effectively blunts the release of multiple pro-inflammatory cytokines and chemokines, including IL-1β, IL-6, IL-8, MIP-1α, TNF-α, and MCP-1 . These findings suggest that SCGB1A1 replacement or augmentation strategies could potentially mitigate excessive inflammatory responses in asthma and other respiratory conditions. Additionally, the understanding that SCGB1A1 expression is regulated by FOXA2 offers another potential therapeutic avenue, as interventions aimed at increasing FOXA2 expression or activity could indirectly boost SCGB1A1 levels .

What are the key knowledge gaps that need to be addressed in SCGB1A1 research?

Several critical knowledge gaps need to be addressed to fully understand SCGB1A1 biology and its therapeutic potential. First, while SCGB1A1 reduction in asthma has been well-documented, the specific mechanisms by which this reduction contributes to disease pathogenesis beyond increased inflammation require further elucidation . Second, the precise signaling pathways through which SCGB1A1 exerts its anti-inflammatory effects in different cell types, including alveolar macrophages and airway epithelial cells, need more comprehensive characterization . Third, while genetic polymorphisms in SCGB1A1 have been associated with asthma risk, the functional consequences of these variants on protein expression, structure, and activity remain incompletely understood . Fourth, the regulation of SCGB1A1 expression by factors beyond FOXA2, particularly in non-respiratory tissues where SCGB1A1 may also be expressed, warrants investigation . Fifth, the role of SCGB1A1 in other respiratory conditions beyond asthma, such as chronic obstructive pulmonary disease (COPD) and respiratory infections, needs further exploration . Sixth, while SCGB1A1 shows promise as a biomarker in head and neck squamous cell carcinoma, its potential utility in other cancer types remains largely unexplored . Finally, the developmental aspects of SCGB1A1 function, particularly how it shapes immune cell development and respiratory health from early life through aging, represent an important area for future research .

What novel methodologies might advance our understanding of SCGB1A1 function?

Advancing our understanding of SCGB1A1 function will require the application of novel methodologies across multiple research domains. Single-cell RNA sequencing technologies would provide unprecedented insights into the heterogeneity of SCGB1A1 expression across different cell populations in healthy and diseased lungs, potentially revealing new cellular sources and targets of this protein . CRISPR-Cas9 gene editing approaches could enable precise manipulation of SCGB1A1 and its regulatory elements in relevant cell types, facilitating detailed mechanistic studies of its function and regulation . The development of conditional and inducible Scgb1a1 knockout models would allow for temporal and tissue-specific deletion of SCGB1A1, helping to dissect its role in different developmental stages and disease contexts . Proteomics approaches, including proximity labeling techniques, could identify novel protein-protein interactions involving SCGB1A1, shedding light on its molecular mechanisms of action . Advanced imaging techniques, such as intravital microscopy combined with fluorescently tagged SCGB1A1, would enable real-time visualization of SCGB1A1 distribution and function in living tissues . The development of small molecule modulators of SCGB1A1 expression or activity would provide valuable tools for both mechanistic studies and potential therapeutic applications . Finally, systems biology approaches integrating transcriptomic, proteomic, and metabolomic data from SCGB1A1-manipulated systems would help construct comprehensive models of SCGB1A1's role in health and disease, potentially revealing unexpected functions and therapeutic opportunities .

Product Science Overview

Structure and Expression

Uteroglobin is a small, non-glycosylated protein that forms a homodimeric structure with a molecular weight of approximately 16.1 kDa . It consists of 142 amino acid residues and is produced and secreted by non-ciliated, non-mucous Clara cells, which are predominant in the epithelial surfaces of pulmonary airways . These cells are also found in other non-ciliated epithelia .

Biological Functions

Uteroglobin has several important biological functions:

  1. Anti-inflammatory Effects: By binding to phospholipids and prostaglandins, Uteroglobin can sequester pro-inflammatory mediators, thereby reducing inflammation .
  2. Anti-tumorigenic Effects: It has been shown to inhibit cell migration and invasion, which are critical processes in cancer metastasis .
  3. Immune Modulation: Uteroglobin can influence immune responses by affecting T cell differentiation and platelet aggregation .
Recombinant Production

Recombinant Human Uteroglobin is produced using Escherichia coli (E. coli) expression systems . The recombinant protein is purified to a high degree, with a purity of ≥ 98% as determined by SDS-PAGE gel and HPLC analyses . The endotoxin level is maintained at less than 0.1 ng/µg of protein, ensuring its suitability for research applications .

Applications in Research

Due to its multifunctional properties, Uteroglobin is of significant interest in various fields of research, including:

  • Cancer Research: Its anti-tumorigenic effects make it a valuable tool for studying cancer cell behavior and potential therapeutic interventions .
  • Inflammation Studies: Researchers utilize Uteroglobin to investigate mechanisms of inflammation and develop anti-inflammatory therapies .
  • Immune System Research: Its role in modulating immune responses is crucial for understanding immune-related diseases and developing immunotherapies .

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.