SFTPB Human

Surfactant Protein B Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to SFTPB Human

SFTPB Human refers to the human gene encoding surfactant protein B (SP-B), a critical component of pulmonary surfactant. This amphipathic protein ensures alveolar stability by reducing surface tension in lung air sacs, preventing collapse during exhalation. Mutations in SFTPB are linked to severe respiratory disorders, while its dysregulation is implicated in lung diseases and cancer .

Key Processes Involving SP-B

  1. Lamellar Body Formation: SP-B structures lipid-protein complexes into stacked membranes, enabling surfactant storage and secretion .

  2. Surfactant Secretion: SP-B ensures efficient release of surfactant into alveoli, facilitating gas exchange .

  3. Immune Regulation: Interacts with surfactant proteins A and D (SP-A, SP-D) to modulate innate immunity .

Pathogenic Mutations and Phenotypes

Mutation TypeExample MutationClinical OutcomeSource
Frameshiftp.Pro133GlnfsTer95Neonatal respiratory distress syndrome
Missensep.Cys121GlyImpaired lamellar body formation
Splice-Sitec.913-1G>ASP-B deficiency (fatal in infancy)

Over 30 SFTPB mutations cause surfactant dysfunction, leading to:

  • SP-B Deficiency: Neonatal respiratory failure due to surfactant insufficiency .

  • Cystic Fibrosis (CF) Interactions: SNPs like rs7316 and rs2077079 associate with mild CF severity via intergenic interactions with SFTPA2 and SFTPD .

Emerging Strategies

ApproachDetails
iPSC-Derived OrganoidsCorrection of SFTPB mutations via lentiviral gene therapy restores lamellar bodies and surfactant secretion .
Synthetic Peptides21-residue peptides mimicking SP-B reduce surface tension in vitro .
Genetic ScreeningSFTPB variant catalogs (e.g., LOVD database) aid in early diagnosis .

Pro-SFTPB and Disease Risk

Biomarker StatusLung Cancer Risk (OR)Source
Nondetectable5.88 (95% CI: 1.24–27.48)
High Detectable Levels1.41 per log-unit increase

Plasma pro-SFTPB levels exhibit a J-shaped association with lung cancer risk, suggesting utility in early detection . Elevated SP-B is also linked to acute respiratory distress syndrome and smoking-related lung damage .

Product Specs

Introduction
Surfactant protein B (SP-B) is crucial for lung function and maintaining stability after birth. It is a hydrophobic surfactant protein, unlike the other surfactant proteins that are hydrophilic. Pulmonary surfactant, a lipoprotein complex comprised of 90% lipids and 10% proteins (including plasma proteins and apolipoproteins SPA, SPB, SPC, and SPD), helps to maintain alveolar stability by reducing surface tension at the air-liquid interface within the peripheral air spaces. SP-B enhances the spreading rate and stability of surfactant monolayers in vitro, increasing the collapse pressure of palmitic acid to roughly 70 millinewtons per meter.
Description
Recombinant Human SFTPB is a single glycosylated polypeptide chain consisting of 363 amino acids (25-381 a.a.) with a molecular weight of 40.4 kDa (calculated). It is fused to a 6 a.a. tag on the C-terminal.
Physical Appearance
White lyophilized powder after filtration.
Formulation
SFTPB is filtered (0.4 μm) and lyophilized from a 0.5 mg/ml solution containing PBS and 5% trehalose (w/v) at pH 7.4.
Solubility
To prepare a working stock solution of about 0.5 mg/ml, add deionized water and allow the lyophilized pellet to dissolve completely.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to prevent repeated freezing and thawing. Reconstituted protein remains stable at 4°C for a limited time and shows no change after two weeks at 4°C.
Purity
Purity is greater than 95.0% as determined by SDS-PAGE.
Synonyms

Pulmonary surfactant-associated protein B, SP-B, 18 kDa pulmonary-surfactant protein, 6 kDa protein, Pulmonary surfactant-associated proteolipid SPL(Phe), SFTPB, SFTP3.

Source

HEK293 Cells.

Amino Acid Sequence

WTTSSLACAQ GPEFWCQSLE QALQCRALGH CLQEVWGHVG ADDLCQECED IVHILNKMAK EAIFQDTMRK FLEQECNVLP LKLLMPQCNQ VLDDYFPLVI DYFQNQTDSN GICMHLGLCK SRQPEPEQEP GMSDPLPKPL RDPLPDPLLD KLVLPVLPGA LQARPGPHTQ DLSEQQFPIP LPYCWLCRAL IKRIQAMIPK GALAVAVAQV CRVVPLVAGG ICQCLAERYS VILLDTLLGR MLPQLVCRLV LRCSMDDSAG PRSPTGEWLPR DSECHLCMSV TTQAGNSSEQ AIPQAMLQAC VGSWLDREKC KQFVEQHTPQ LLTLVPRGWD AHTTCQALGV CGTMSSPLQC IHSPDLHHHHHH.

Q&A

What is the SFTPB gene and what is its normal function in human lung physiology?

The SFTPB gene (GeneID: 6439, Locus tag: HGNC 10801; MIM 178640) is a 9.7-kb genomic region that encodes surfactant protein-B (SP-B), a critical 79-amino-acid hydrophobic protein essential for pulmonary surfactant function . SP-B is one of four proteins in surfactant, a mixture of phospholipids and proteins that lines lung tissue to facilitate breathing . The protein plays a crucial role in reducing surface tension in lung alveoli, preventing their collapse after exhalation and enabling efficient oxygen delivery to the body .

SP-B specifically helps spread surfactant across the lung tissue surface, directly contributing to the surface tension-lowering properties that maintain alveolar patency at end-expiration . This function is absolutely essential for successful fetal-neonatal pulmonary transition and continued respiratory function throughout life .

How is SFTPB expression regulated during lung development?

SFTPB expression is tightly regulated during lung development, with the transcription factor Nkx2 homeobox 1 (NKX2-1) playing a crucial role in this process. NKX2-1 is expressed in the primordial lung bud and activates surfactant-related genes including SFTPB . The regulatory elements of SFTPB respond to developmental cues, ensuring proper temporal and spatial expression patterns during alveolar development.

Research approaches to studying SFTPB regulation include:

  • Chromatin immunoprecipitation (ChIP) assays to identify transcription factor binding sites

  • Promoter analysis using reporter gene constructs

  • RNA sequencing to track developmental expression patterns

  • Conditional gene knockout models to assess regulatory relationships

Understanding the precise mechanisms of SFTPB regulation remains an active area of research, particularly as it relates to developmental timing and cell-specific expression in alveolar type II cells.

What are the processing steps required for SP-B protein maturation?

SP-B undergoes complex post-translational processing before becoming functionally mature. The protein is initially synthesized as a larger precursor that requires several proteolytic cleavage steps. These processing events occur in specialized cellular structures known as lamellar bodies within alveolar type II cells .

The processing pathway involves:

  • Synthesis of proSP-B in the endoplasmic reticulum

  • Initial glycosylation and folding

  • Transport to the Golgi apparatus

  • Proteolytic cleavage of N-terminal and C-terminal propeptides

  • Packaging into lamellar bodies

  • Final maturation steps within lamellar bodies

  • Secretion of mature SP-B with surfactant phospholipids

Research methodologies to study SP-B processing include pulse-chase experiments, subcellular fractionation, and immunoblotting with antibodies specific to different processing intermediates. Understanding these steps is critical as disruptions in SP-B processing can lead to severe respiratory pathology even when the gene itself is intact .

How does SP-B interact with phospholipids in surfactant to reduce surface tension?

SP-B functions through direct biophysical interactions with surfactant phospholipids. The protein's amphipathic structure allows it to:

  • Insert into phospholipid layers, facilitating rapid spreading at the air-liquid interface

  • Promote formation of surface-active phospholipid films

  • Enhance adsorption of phospholipids to the air-liquid interface

  • Stabilize compressed phospholipid films during respiration cycles

  • Participate in the formation and stability of tubular myelin, an extracellular surfactant structure

Research approaches to study these interactions include:

  • Surface balance measurements of surfactant surface tension properties

  • Fluorescence microscopy of labeled SP-B in model membrane systems

  • Atomic force microscopy of surfactant films

  • Molecular dynamics simulations of SP-B-phospholipid interactions

These protein-lipid interactions are essential for the surface tension reduction that prevents alveolar collapse during respiration .

What types of genetic variants have been identified in the SFTPB gene?

Comprehensive resequencing studies of SFTPB have revealed significant genetic diversity within this relatively small gene. A population-based cohort study (n = 1,116) identified:

  • 81 single nucleotide polymorphisms (SNPs)

  • 5 small insertion/deletion variants (indels)

  • An excess of low-frequency variations

  • Weak linkage disequilibrium (LD) across the gene

  • High haplotype diversity

A notable feature of SFTPB's genetic architecture is the presence of a recombination hotspot that spans the gene, contributing to the weak linkage disequilibrium observed . This genetic diversity has important implications for association studies, as it suggests that comprehensive resequencing approaches may be more effective than tag-SNP strategies for identifying disease-associated variants.

Variant TypeNumber IdentifiedFrequency Characteristics
SNPs81Predominantly low-frequency
Indels5Rare
Exonic variantsVariableNo common disruptive variants identified

What are the molecular mechanisms by which SFTPB mutations cause surfactant dysfunction?

More than 30 mutations in the SFTPB gene have been identified that cause surfactant dysfunction . These mutations disrupt normal SP-B production and function through several mechanisms:

  • Complete loss of SP-B production: Some mutations result in a complete absence of mature SP-B protein, which is invariably fatal without lung transplantation.

  • Partial reduction in SP-B levels: Studies indicate that reduction of SP-B expression by more than 75% can disrupt surfactant function and cause respiratory distress .

  • Impaired processing of SP-B: Mutations may allow production of the proprotein but prevent proper processing to the mature form.

  • Disruption of lamellar body formation: SP-B is essential for normal lamellar body formation, and its absence leads to structural abnormalities in these organelles .

  • Secondary effects on SP-C processing: The lack of normal lamellar bodies due to SP-B deficiency leads to abnormal processing of surfactant protein-C (SP-C), resulting in reduced mature SP-C and accumulation of unprocessed forms .

The combined dysfunction of both SP-B and SP-C contributes to the severity of respiratory distress observed in patients with SFTPB mutations. Research approaches to study these mechanisms include transgenic mouse models, patient-derived cell cultures, and analysis of bronchoalveolar lavage fluid from affected individuals.

How can lung organoids be used to model SFTPB deficiency and potential treatments?

Lung organoids represent a cutting-edge approach to modeling SFTPB deficiency in vitro. These three-dimensional multicellular structures can be derived from patient-specific induced pluripotent stem cells (iPSCs) and differentiated to contain cell populations representative of both proximal and distal lung regions .

The methodology for using lung organoids to study SFTPB involves:

  • Generation of patient-specific iPSCs: Cells from patients with SFTPB mutations are reprogrammed to pluripotency.

  • Differentiation into lung organoids: Through careful manipulation of developmental signaling pathways, iPSCs are directed to form lung organoid structures containing various lung cell types including alveolar type II cells.

  • Genetic correction approaches: The mutated SFTPB gene can be corrected using techniques such as lentiviral delivery of wild-type SFTPB.

  • Functional assessment: Organoids can be analyzed for:

    • SFTPB mRNA expression during differentiation

    • SFTPB protein production in mature organoids

    • Formation of normal lamellar bodies

    • Secretion of surfactant into culture medium

    • Ultrastructural analysis of surfactant components

Research has demonstrated that lentiviral delivery of wild-type SFTPB to deficient iPSCs can restore SP-B expression, normal lamellar body formation, and surfactant secretion in derived organoids . This approach provides a valuable platform for testing potential therapeutic interventions before moving to in vivo models.

What genetic engineering approaches show promise for correcting SFTPB deficiency?

Several genetic engineering strategies have demonstrated potential for correcting SFTPB deficiency in experimental models:

  • Lentiviral gene delivery: Introducing wild-type SFTPB via lentiviral vectors has successfully corrected deficiency in patient-derived organoid models . This approach allows for stable integration and expression of the functional gene.

  • CRISPR/Cas9 gene editing: Precise correction of specific SFTPB mutations using CRISPR/Cas9 technology offers the potential for permanent repair of the endogenous gene without introducing foreign DNA.

  • mRNA therapy: Delivery of SFTPB mRNA encapsulated in lipid nanoparticles could provide temporary protein expression while avoiding genomic integration.

  • AAV-mediated gene therapy: Adeno-associated viral vectors could deliver functional SFTPB genes to affected lung tissue with potentially lower immunogenicity than lentiviral approaches.

Research challenges include achieving sufficient transduction efficiency in alveolar type II cells, maintaining long-term expression, and ensuring appropriate regulation of the corrected gene. The lung organoid model provides an excellent platform for initial testing of these approaches before advancing to animal models or clinical applications.

What methodologies are currently used to diagnose SFTPB deficiency in clinical settings?

Diagnosis of SFTPB deficiency requires a multifaceted approach combining clinical, radiological, and molecular techniques:

  • Clinical assessment: Evaluation of respiratory distress in newborns that is refractory to conventional treatments.

  • Imaging studies: Chest radiographs and high-resolution CT scans showing diffuse ground-glass opacities consistent with surfactant dysfunction.

  • Bronchoalveolar lavage (BAL) analysis:

    • Protein analysis of BAL fluid for absence of mature SP-B

    • Assessment of phospholipid composition and surface tension properties

    • Evaluation of unprocessed SP-C forms that accumulate in SP-B deficiency

  • Genetic testing:

    • Targeted sequencing of SFTPB coding regions

    • Analysis for common mutations based on ethnicity

    • Comprehensive gene resequencing for novel variants

    • Expression analysis from lung biopsy if available

  • Histopathological examination: Lung biopsy showing characteristic findings of alveolar proteinosis, type II cell hyperplasia, and abnormal lamellar bodies on electron microscopy.

Given the genetic complexity of SFTPB with over 30 identified disease-causing mutations, comprehensive resequencing is recommended over panel-based approaches that might miss novel or rare variants .

What are the current challenges in interpreting SFTPB genetic variants of uncertain significance?

The interpretation of SFTPB genetic variants presents several challenges for researchers and clinicians:

  • High genetic diversity: The excess of low-frequency variants and high haplotype diversity in SFTPB complicates variant interpretation .

  • Weak linkage disequilibrium: Due to a recombination hotspot spanning SFTPB, variants are less likely to be inherited together, making it difficult to use tagging SNPs for association studies .

  • Variable penetrance: While complete loss-of-function mutations are fully penetrant, the clinical significance of partial reductions in SP-B levels is less clear.

  • Functional assessment limitations: Current in silico prediction tools may not accurately predict the functional impact of all SFTPB variants. Notably, homology-based software tools have failed to identify definitively damaging exonic variants in population studies .

  • Complex genotype-phenotype relationships: The relationship between specific variants and clinical phenotypes is not fully established for many SFTPB variants.

Research approaches to address these challenges include:

  • Development of high-throughput functional assays for variant classification

  • Creation of variant databases specific to surfactant dysfunction disorders

  • Improved computational models for predicting variant effects on SP-B processing and function

  • Integration of multi-omics data to better understand variant impact

Complete resequencing of SFTPB is recommended as the optimal approach for genetic association studies, particularly in genetically diverse populations .

How might single-cell RNA sequencing advance our understanding of SFTPB regulation in different lung cell populations?

Single-cell RNA sequencing (scRNA-seq) technologies offer unprecedented opportunities to advance SFTPB research:

  • Cell-type specific expression patterns: While SFTPB is primarily expressed in alveolar type II cells, scRNA-seq could reveal previously undetected expression in other lung cell populations or identify specific subsets of type II cells with unique expression patterns.

  • Developmental trajectory mapping: scRNA-seq can map the ontogeny of SFTPB-expressing cells during lung development, providing insights into the timing and regulation of expression.

  • Disease-associated transcriptional changes: By comparing scRNA-seq data from healthy and diseased lung tissue, researchers can identify dysregulated pathways that affect SFTPB expression in various pulmonary conditions.

  • Regulatory network identification: Integration of scRNA-seq with ATAC-seq or ChIP-seq data can reveal cell-type specific regulatory elements and transcription factor networks governing SFTPB expression.

  • Therapeutic response monitoring: Following genetic or pharmacological interventions, scRNA-seq can track changes in SFTPB expression and associated pathways at cellular resolution.

Research methodologies combining scRNA-seq with spatial transcriptomics would be particularly valuable, as they could reveal how SFTPB expression varies across different regions of the developing and mature lung.

What are the prospects for in utero interventions for SFTPB deficiency identified through prenatal diagnosis?

The development of in utero interventions for prenatally diagnosed SFTPB deficiency represents a frontier in fetal therapy research:

  • Amniotic delivery of surfactant: Direct delivery of exogenous surfactant to amniotic fluid might promote lung maturation, though challenges include maintaining therapeutic concentrations and ensuring alveolar uptake.

  • Fetal gene therapy: Viral vector-mediated gene delivery to the developing lung could potentially correct SFTPB deficiency before birth, leveraging the smaller size and immunological tolerance of the fetal system.

  • In utero stem cell transplantation: Introduction of healthy stem cells capable of engrafting in the developing lung and differentiating into SFTPB-producing cells represents a potential curative approach.

  • Maternal corticosteroid optimization: While not correcting the underlying defect, optimized corticosteroid regimens might maximize development of the remaining functional lung tissue.

  • Ex utero intrapartum treatment (EXIT) procedures: Advanced surgical techniques could allow partial delivery with maintenance of placental circulation while interventions such as surfactant administration or genetic treatments are performed.

Research challenges include:

  • Development of safe delivery systems that specifically target the fetal lung

  • Achieving sufficient transduction efficiency to support respiratory function

  • Balancing intervention timing with fetal development stages

  • Ethical considerations surrounding experimental fetal interventions

Early diagnosis through improved genetic testing workflows will be essential for the success of any in utero intervention strategy.

Product Science Overview

Introduction

Surfactant Protein B (SP-B) is a crucial component of the pulmonary surfactant system, which is essential for normal respiratory function. The human recombinant form of SP-B is a synthetic version produced through recombinant DNA technology, which allows for the study and therapeutic use of this protein.

Structure and Function

SP-B is an amphipathic protein, meaning it contains both hydrophilic (water-attracting) and hydrophobic (water-repelling) regions. This unique structure enables SP-B to interact with the lipid components of the pulmonary surfactant, a complex mixture of lipids and proteins that reduces surface tension in the lungs . By lowering surface tension, SP-B helps maintain alveolar stability, preventing the collapse of alveoli during exhalation and ensuring efficient gas exchange .

Genetic Background

The gene encoding SP-B is known as SFTPB (Surfactant Protein B gene). Mutations in this gene can lead to surfactant metabolism dysfunction, which is associated with severe respiratory conditions such as hereditary pulmonary alveolar proteinosis and neonatal respiratory distress syndrome . These conditions highlight the critical role of SP-B in lung function and homeostasis.

Production of Human Recombinant SP-B

Human recombinant SP-B is produced using recombinant DNA technology, which involves inserting the SFTPB gene into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of SP-B, which can be purified and used for research or therapeutic purposes .

Clinical Significance

The therapeutic potential of human recombinant SP-B is significant, particularly for treating respiratory conditions caused by surfactant deficiency. For example, recombinant SP-B can be administered to premature infants with underdeveloped lungs to improve their respiratory function and reduce the risk of respiratory distress syndrome . Additionally, research into SP-B and its interactions with other surfactant proteins and lipids continues to provide valuable insights into lung biology and disease mechanisms.

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.