TGFBI Human, 182 a.a.

Transforming Growth Factor Beta-Induced (182 a.a.) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Composition

  • Source: E. coli-expressed recombinant protein .

  • Length: 182 amino acids (502–683 residues of the 683-residue full-length TGFBI).

  • Molecular Weight: 19.9 kDa (non-glycosylated; SDS-PAGE migration may appear higher due to charge effects).

  • Key Domains:

    • Fasciclin Domain: Four internal repeated domains conserved in species (critical for extracellular matrix binding).

    • RGD Motif: Integrin-binding site involved in cell adhesion .

PropertySpecification
Amino Acid SequenceMGTVMDVLKG DNRFSMLVAA IQSAGLTETL NREGVYTVFA PTNEAFRALP PRERSRLLGD AKELANILKY HIGDEILVSG GIGALVRLKS LQGDKLEVSL KNNVVSVNKE PVAEPDIMAT NGVVHVITNV LQPPANRPQE RGDELADSAL EIFKQASAFS RASQRSVRLA PVYQKLLERM KH .
Purity>95% (SDS-PAGE) .

Research Findings: Corneal Dystrophies

TGFBI mutations are linked to autosomal dominant corneal dystrophies (CDs), including lattice corneal dystrophy (LCD) and granular corneal dystrophy (GCD). The 182 a.a. fragment is critical for studying mutant protein aggregation.

Key Mutations and Aggregation

  • Arg124Cys (R124C): Common in LCD, leading to amyloid deposits .

  • Arg555Gln (R555Q): Linked to GCD type 1 .

  • Arg124His (R124H): Associated with GCD type 2 .

MutationPhenotypeAggregation Kinetics
R124CAmyloid lattice deposits in corneaRapid Aβ aggregation (12 hrs)
R555QGranular deposits in Bowman’s layerSlower aggregation (24 hrs)
Wild-TypeNo aggregationBasal aggregation (24 hrs)

Mechanistic Insights:

  • Amyloid Formation: R124C mutants promote amyloid-β (Aβ) aggregation in vitro, exacerbated by osmolytes like TMAO .

  • Therapeutic Target: TMAO suppresses R124C-induced aggregation, suggesting potential for chemical interventions .

Role in Diabetes and Metabolic Regulation

TGFBI influences pancreatic islet function and glucose homeostasis:

Experimental Models

  • TGFBI Knockout (KO) Mice:

    • Islet Survival: Reduced β-cell survival post-streptozotocin (STZ) injury .

    • Glucose Tolerance: Impaired fasting blood glucose levels .

  • TGFBI Overexpression:

    • Islet Regeneration: Enhanced β-cell proliferation and insulin release .

ModelOutcome
TGFBI KOIncreased diabetes susceptibility (STZ-induced) .
TGFBI TransgenicImproved glucose tolerance and islet regeneration .

Signaling Pathways:

  • AKT/mTORC1: TGFBI activates AKT1S1, RPS6, and EIF4EBP1, linking to insulin sensitivity .

Epigenetic and Oncogenic Roles

TGFBI is implicated in cancer through epigenetic silencing:

Burkitt Lymphoma (BL)

  • Epigenetic Alterations: Promoter hypermethylation in EBV-positive BL cells .

  • Oncogenic Drivers: Aflatoxin B1 (AFB1) and EBV latent membrane protein 1 (LMP1) downregulate TGFBI, promoting tumorigenesis .

FactorEffect on TGFBICancer Association
EBV InfectionLMP1 suppresses TGFBI expression .Burkitt lymphoma progression .
AFB1 ExposureSynergizes with EBV to reduce TGFBI .BL risk in aflatoxin-endemic regions .

Product Specs

Introduction
Transforming growth factor (TGF)-beta 1 induces the extracellular matrix protein known as TGFBI. This protein plays a role in various cellular processes, including cell growth, differentiation, wound healing, and adhesion. Notably, specific missense mutations in the TGFBI gene have been linked to autosomal dominant corneal dystrophies in humans. The TGFBI gene encodes a protein composed of 683 amino acids, featuring an RGD motif and four internal repeating domains with highly conserved sequences found across multiple species, known as Fasciclin domains.
Description
Recombinant Human TGFBI, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 182 amino acids (502-683). It has a molecular mass of 19.9 kDa. However, its apparent molecular weight on SDS-PAGE might be higher. The purification process of this recombinant human TGFBI protein involves proprietary chromatographic methods.
Physical Appearance
Sterile, filtered liquid solution.
Formulation
TGFBI 182 a.a. recombinant Human is supplied in a solution containing 20mM Tris-HCl with a pH of 8, 1mM EDTA, 0.1mM PMSF, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, freezing at -20°C is recommended. Adding a carrier protein such as 0.1% HSA or BSA is advisable for long-term storage. It's crucial to avoid repeated freeze-thaw cycles.
Purity
The purity of the product is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
Transforming growth factor-beta-induced protein ig-h3, Beta ig-h3, Kerato-epithelin, RGD-containing collagen-associated protein, RGD-CAP, TGFBI, BIGH3, CSD, CDB1, CDG2, CSD1, CSD2, CSD3, EBMD, LCD1, CDGG1.
Source
Escherichia Coli.
Amino Acid Sequence
MGTVMDVLKG DNRFSMLVAA IQSAGLTETL NREGVYTVFA PTNEAFRALP PRERSRLLGD AKELANILKY HIGDEILVSG GIGALVRLKS LQGDKLEVSL KNNVVSVNKE PVAEPDIMAT NGVVHVITNV LQPPANRPQE RGDELADSAL EIFKQASAFS RASQRSVRLA PVYQKLLERM KH.

Q&A

What is TGFBI Human, 182 a.a. and how does it differ from the full-length TGFBI protein?

TGFBI Human, 182 a.a. is a recombinant protein fragment containing amino acids 502-683 of the full-length human TGFBI (Transforming growth factor-beta-induced protein ig-h3). While the complete TGFBI protein consists of 683 amino acids with multiple functional domains, this 182 a.a. fragment represents the C-terminal portion with a molecular mass of approximately 19.9 kDa . This recombinant version is produced in E. coli as a single, non-glycosylated polypeptide, which differs from the naturally occurring full-length TGFBI that undergoes post-translational modifications in mammalian cells . The 182 a.a. fragment is particularly useful for studying specific C-terminal interactions without interference from other domains present in the complete protein.

What are the key structural domains and functional motifs present in the TGFBI Human, 182 a.a. fragment?

The 182 a.a. fragment of TGFBI (amino acids 502-683) contains significant functional domains including part of the fourth FAS1 domain and the C-terminal region. Based on sequence analysis and functional studies, this fragment likely contains:

  • Portions of the fourth fasciclin-like domain which mediates interactions with integrins, particularly α3β1, αVβ3 and αVβ5

  • The C-terminal region which may contain the RGD motif that facilitates binding to various integrins

  • Structural elements that contribute to extracellular matrix interactions

The amino acid sequence provided (MGTVMDVLKG DNRFSMLVAA IQSAGLTETL NREGVYTVFA PTNEAFRALP PRERSRLLGD AKELANILKY HIGDEILVSG GIGALVRLKS LQGDKLEVSL KNN) represents part of this fragment and contains regions that contribute to TGFBI's ability to interact with cellular components and the extracellular matrix .

What are the optimal storage and handling conditions for maintaining the stability of TGFBI Human, 182 a.a. in research applications?

For optimal stability of TGFBI Human, 182 a.a., researchers should implement the following evidence-based protocols:

  • Short-term storage (2-4 weeks): Store at 4°C in the original formulation (20mM Tris-HCl pH-8, 1mM EDTA, 0.1mM PMSF and 20% glycerol)

  • Long-term storage: Store at -20°C with the addition of a carrier protein (0.1% HSA or BSA) to prevent protein adsorption to storage vessel surfaces and maintain stability

  • Avoid repeated freeze-thaw cycles: Aliquot the protein before freezing to minimize degradation from repeated temperature changes

  • Working solution preparation: When preparing working dilutions, use buffers that maintain a pH between 7.5-8.5 to preserve protein structure and function

  • Handling precautions: Use sterile techniques and non-metallic utensils when aliquoting to prevent contamination and potential metal-induced oxidation

These conditions have been experimentally determined to maximize protein stability while preserving functional domains critical for experimental applications.

What are the recommended experimental approaches for studying TGFBI-integrin interactions using the 182 a.a. fragment?

To effectively study TGFBI-integrin interactions using the 182 a.a. fragment, researchers should consider these methodological approaches:

  • Solid-phase binding assays:

    • Immobilize purified integrins (particularly α3β1, αVβ3, and αVβ5) on microplates

    • Add varying concentrations of labeled TGFBI 182 a.a.

    • Quantify binding through ELISA-based detection systems

    • Include appropriate controls with known integrin ligands and blocking antibodies

  • Cell adhesion assays:

    • Coat cell culture surfaces with TGFBI 182 a.a. at 2-10 μg/mL

    • Evaluate adhesion of cells expressing different integrin subtypes

    • Employ function-blocking antibodies against specific integrins to confirm interaction specificity

    • Compare results with full-length TGFBI to identify domain-specific effects

  • Surface Plasmon Resonance (SPR):

    • Immobilize TGFBI 182 a.a. on sensor chips

    • Flow purified integrins at various concentrations

    • Determine binding kinetics (kon, koff) and equilibrium constants (KD)

    • Compare with known integrin binding domains

  • Co-immunoprecipitation assays:

    • Use anti-TGFBI antibodies to pull down protein complexes from cells treated with TGFBI 182 a.a.

    • Identify integrin associations through Western blotting

    • Validate with reciprocal co-IP using anti-integrin antibodies

This multi-faceted approach provides comprehensive data on binding specificity, affinity, and functional relevance of the TGFBI 182 a.a. interactions with cellular integrin receptors .

How does TGFBI Human, 182 a.a. modulate the AKT/mTORC1 signaling pathway, and what experimental approaches best elucidate these mechanisms?

TGFBI modulates the AKT/mTORC1 signaling pathway through specific molecular interactions that can be investigated using targeted experimental approaches. Based on phosphoprotein array analyses, TGFBI stimulation affects multiple components in this pathway:

  • Key molecular targets affected by TGFBI:

    • AKT1S1 (PRAS40): Links AKT and mTORC1 pathways

    • AKT: Ser473 phosphorylation increases upon TGFBI stimulation

    • RPS6: Increased phosphorylation indicates mTORC1 activation

    • EIF4EBP1: Phosphorylation altered in response to TGFBI

  • Recommended experimental approaches:

    • Phosphorylation kinetics assay: Treat cells with TGFBI 182 a.a. (10-50 μg/mL) for various time points (5 min to 24 h) and analyze phosphorylation patterns by Western blotting

    • Chemical inhibitor studies: Pre-treat cells with AKT inhibitors (MK-2206, 0.5-10 μM) prior to TGFBI stimulation to confirm pathway specificity

    • siRNA knockdown validation: Selectively downregulate AKT1S1, RPS6, or EIF4EBP1 using targeted siRNAs to establish their necessity in TGFBI-mediated effects

    • Proximity ligation assays: Detect in situ protein-protein interactions between TGFBI, integrins, and downstream signaling molecules

  • Data interpretation framework:

    • Compare signaling responses between full-length TGFBI and the 182 a.a. fragment

    • Analyze cell type-specific responses (particularly in islet cells vs. cancer cells)

    • Correlate signaling activation with functional outcomes (cell survival, proliferation, etc.)

This methodological approach provides a comprehensive understanding of how TGFBI 182 a.a. engages and modulates the AKT/mTORC1 pathway, which appears critical for its effects on cell survival and function .

What experimental models are most appropriate for investigating TGFBI's role in diabetes risk, and how can the 182 a.a. fragment be utilized in these studies?

Based on genetic evidence linking TGFBI to diabetes risk, several experimental models can effectively investigate its mechanistic role, with specific protocols for utilizing the 182 a.a. fragment:

  • Pancreatic islet models:

    • Ex vivo islet culture: Isolate pancreatic islets from wild-type and TGFBI KO mice; treat with TGFBI 182 a.a. (10-50 μg/mL) in serum-free conditions

    • Assessment metrics: Measure islet survival (using TUNEL assay), insulin secretion (glucose-stimulated insulin secretion assay), and AKT/mTORC1 pathway activation

    • Stress challenges: Expose islets to inflammatory cytokines (IL-1β, TNF-α, IFN-γ) or glucolipotoxicity with/without TGFBI 182 a.a. supplementation

  • In vivo models:

    • STZ-induced diabetes: Administer multiple low-dose streptozotocin (40 mg/kg for 5 consecutive days) to wild-type mice with concurrent TGFBI 182 a.a. treatment

    • Islet transplantation studies: Transplant a suboptimal mass of islets treated ex vivo with TGFBI 182 a.a. into diabetic recipients

    • Monitoring parameters: Blood glucose, glucose tolerance tests, insulin sensitivity, and histological assessment of β-cell mass/proliferation

  • Molecular investigations:

    • Domain-specific functions: Compare TGFBI 182 a.a. effects with full-length protein to identify functional domains critical for islet preservation

    • Receptor identification: Use blocking antibodies against various integrins to determine which mediates TGFBI's effects on islet cells

    • Signaling cascade analysis: Focus on AKT1S1, RPS6, and EIF4EBP1 phosphorylation status, as these have been implicated in TGFBI signaling in islets

  • Translational relevance:

    • Correlate experimental findings with human genetic data linking TGFBI SNPs to diabetes risk

    • Develop potential therapeutic strategies based on TGFBI 182 a.a. administration or enhancement of endogenous TGFBI signaling

These methodologies enable comprehensive investigation of TGFBI's role in islet biology and diabetes pathophysiology, with particular focus on whether the 182 a.a. fragment retains the protective functions observed with the full-length protein .

How can researchers effectively investigate the seemingly contradictory roles of TGFBI in cancer progression using the 182 a.a. fragment?

To address the paradoxical roles of TGFBI in cancer biology, researchers should implement a systematic experimental approach using the 182 a.a. fragment:

  • Context-dependent signaling analysis:

    • Compare TGFBI 182 a.a. effects across multiple cancer cell lines (ovarian, pancreatic, etc.)

    • Measure proliferation, migration, invasion, and apoptosis responses using standardized assays

    • Analyze dose-dependent responses (1-100 μg/mL) to identify potential biphasic effects

    • Map activated signaling networks using phosphoproteomic approaches in each cellular context

  • Proteolytic processing investigation:

    • Determine if the 182 a.a. fragment undergoes further proteolytic processing in the tumor microenvironment

    • Compare intact fragment versus potential cleavage products generated by tumor-associated proteases

    • Assess whether proteolytic processing converts TGFBI from tumor-promoting to tumor-suppressive functions

  • Integrin receptor profiling:

    • Characterize integrin expression patterns across cancer cell lines using flow cytometry

    • Correlate integrin expression profiles with TGFBI 182 a.a. response patterns

    • Use integrin-blocking antibodies to determine which specific integrins mediate tumor-promoting versus tumor-suppressive effects

  • Experimental design matrix:

Experimental ConditionHigh Integrin-Expressing CellsLow Integrin-Expressing Cells
TGFBI 182 a.a. (10 μg/mL)Measure: proliferation, migration, signalingMeasure: same parameters
TGFBI 182 a.a. + integrin blockersDetermine integrin dependencyConfirm specificity
TGFBI 182 a.a. + protease inhibitorsAssess role of proteolytic processingCompare with control
TGFBI 182 a.a. in hypoxic conditionsMimic tumor microenvironmentEvaluate context dependency
  • Clinical correlation:

    • Analyze TGFBI expression in patient tumor samples in relation to clinical outcomes

    • Correlate findings with experimental data using the 182 a.a. fragment to identify translational relevance

This methodological framework enables systematic investigation of TGFBI's dual functions in cancer, helping researchers decipher when and how it promotes or suppresses tumor progression .

What methodological approaches should be employed to study the interaction between TGFBI 182 a.a. and β3 integrin in ovarian cancer therapy resistance?

To investigate the role of TGFBI-β3 integrin interactions in ovarian cancer therapy resistance, researchers should implement the following methodological approaches:

  • Cell Line Models and Resistance Profiling:

    • Establish paired topotecan-sensitive and resistant ovarian cancer cell lines

    • Quantify TGFBI and β3 integrin expression using qPCR and Western blot

    • Compare expression levels between sensitive and resistant lines

    • Perform immunofluorescence co-localization studies to visualize TGFBI-β3 integrin interactions

  • Functional Modulation Studies:

    • TGFBI knockdown/overexpression: Generate stable cell lines with modulated TGFBI expression using lentiviral vectors

    • β3 integrin blockade: Use function-blocking antibodies (LM609) or small molecule inhibitors (cilengitide)

    • Recombinant protein treatment: Treat cells with TGFBI 182 a.a. (5-50 μg/mL) with/without β3 integrin blockade

    • Measure outcomes: Assess chemosensitivity using dose-response curves (IC50 determination), apoptosis assays, and cell cycle analysis

  • Mechanistic Pathway Analysis:

    • Perform phosphoproteomic analysis following TGFBI 182 a.a. treatment focusing on:

      • FAK/Src activation status

      • AKT/mTOR pathway components

      • DNA damage response proteins

    • Use pathway inhibitors to validate key nodes in resistance signaling

    • Employ chromatin immunoprecipitation to identify transcriptional changes mediated by TGFBI-β3 integrin signaling

  • Tumor Microenvironment Considerations:

    • Co-culture resistant cancer cells with stromal components

    • Analyze TGFBI production by cancer-associated fibroblasts

    • Evaluate the impact of extracellular matrix proteins on TGFBI-mediated resistance

    • Use 3D spheroid models to better approximate in vivo conditions

  • Translational Validation:

    • Analyze TGFBI and β3 integrin expression in patient samples before and after therapy

    • Correlate expression with treatment response and progression-free survival

    • Develop predictive models based on TGFBI/β3 integrin expression patterns

These methodologies provide a comprehensive framework for investigating how TGFBI 182.a.a interactions with β3 integrin contribute to chemotherapy resistance in ovarian cancer, potentially identifying novel therapeutic vulnerabilities .

What are the most effective methods for detecting TGFBI mutations in research and clinical samples, and can these be adapted for studies using the 182 a.a. fragment?

For effective detection of TGFBI mutations in both research and clinical contexts, several validated methodologies can be employed:

  • Real-Time PCR with Melting Curve Analysis:

    • Optimal for rapid detection of common TGFBI mutations

    • Design specific fluorescent probes for mutation hotspots

    • Distinguish genotypes through distinct melting peaks at different temperatures

    • Complete analysis in approximately 54 minutes

    • Shows 100% concordance with conventional DNA sequencing

  • Next-Generation Sequencing (NGS) Approaches:

    • Panel design: Include full TGFBI coding regions, focusing on exons 4 and 12 where mutations commonly occur

    • Library preparation: Use amplicon-based or hybrid capture methods

    • Analysis pipeline: Employ specialized bioinformatic tools for variant calling

    • Validation: Confirm novel variants with Sanger sequencing

  • Digital Droplet PCR for Low-Frequency Mutation Detection:

    • Partition DNA samples into thousands of droplets

    • Perform PCR amplification in each droplet

    • Quantify rare mutations with high sensitivity (0.1%)

    • Particularly useful for detecting mosaic mutations

  • Application to 182 a.a. Fragment Studies:

    • Design primers to specifically amplify the region corresponding to the 182 a.a. fragment (amino acids 502-683)

    • Create a mutation panel focused on C-terminal mutations affecting protein function

    • Correlate genotypic findings with functional effects using recombinant mutant versions of the 182 a.a. fragment

  • Methodological Workflow for Comprehensive Analysis:

StepTechniquePurposeTurnaround Time
1Real-time PCR/Melting CurveRapid screening of common mutations1-2 hours
2Sanger SequencingConfirmation of mutations identified1-2 days
3NGS PanelComprehensive mutation profiling3-7 days
4Functional ValidationTesting effect of mutations using recombinant proteins1-2 weeks

This comprehensive approach ensures both rapid detection of common mutations and thorough characterization of novel variants, with direct applications to research using the TGFBI 182 a.a. fragment .

How can researchers effectively differentiate between the functions of the 182 a.a. fragment and the full-length TGFBI protein in experimental systems?

To systematically differentiate between the functions of the 182 a.a. fragment and full-length TGFBI protein, researchers should implement the following comparative methodological approaches:

  • Domain-Specific Functional Assays:

    • Binding studies: Compare ECM component binding profiles of both proteins using solid-phase binding assays

    • Integrin interaction analysis: Conduct parallel receptor binding assays to identify differences in integrin recognition patterns

    • Structural studies: Employ circular dichroism spectroscopy to compare secondary/tertiary structural elements

  • Comparative Signaling Analysis:

    • Treat cells with equimolar concentrations of 182 a.a. fragment versus full-length TGFBI

    • Perform time-course Western blotting for phosphorylation of key signaling molecules:

      • AKT and AKT1S1

      • RPS6 and EIF4EBP1

      • FAK and downstream effectors

    • Use phosphoproteomic arrays to identify unique signaling signatures

  • Cell-Type Specific Response Mapping:

    • Test both proteins across multiple cell types (islets, epithelial cells, fibroblasts)

    • Measure key functional outcomes (survival, proliferation, migration)

    • Construct response heat maps to visualize differential effects

  • Competitive Inhibition Experiments:

    • Pre-treat cells with one form before adding the other

    • Determine if the 182 a.a. fragment can compete with full-length TGFBI for binding sites

    • Identify potential antagonistic or synergistic relationships

  • Structural Analysis Protocol:

TechniqueApplicationKey Information Obtained
Limited proteolysisBoth proteins exposed to controlled proteolytic digestionDifferences in accessible cleavage sites
Surface plasmon resonanceBinding kinetics analysisDifferential association/dissociation rates with partners
Intracellular traffickingFluorescently labeled proteins added to cellsDifferences in cellular uptake and localization
Molecular modelingIn silico structural analysisPrediction of exposed binding surfaces unique to each form
  • Genetic Complementation Studies:

    • Introduce either the 182 a.a. fragment or full-length TGFBI into TGFBI-knockout cells

    • Determine which functions can be rescued by the fragment versus the complete protein

    • Analyze phenotypic outcomes in detail

This systematic approach enables researchers to create comprehensive functional maps distinguishing the roles of the truncated 182 a.a. fragment from the complete TGFBI protein, critical for understanding domain-specific functions and potential therapeutic applications .

What are the key considerations for designing experiments to investigate the potential therapeutic applications of TGFBI 182 a.a. in diabetes prevention or treatment?

When designing experiments to evaluate TGFBI 182 a.a. as a potential therapeutic agent for diabetes, researchers should implement these methodologically rigorous approaches:

  • Preclinical Model Selection:

    • Spontaneous diabetes models: NOD mice (T1D), db/db or ob/ob mice (T2D)

    • Induced diabetes models: Multiple low-dose STZ (40 mg/kg for 5 days) for T1D-like condition; high-fat diet with STZ for T2D-like condition

    • Humanized models: Immunodeficient mice engrafted with human immune cells and islets

    • Critical controls: Include TGFBI knockout mice as negative controls and TGFBI transgenic mice as positive controls

  • Therapeutic Administration Protocols:

    • Preventive regimen: Begin TGFBI 182 a.a. administration before diabetes onset

    • Treatment regimen: Initiate after diabetes development

    • Dosing optimization: Test multiple doses (0.1-10 mg/kg) and schedules (daily vs. intermittent)

    • Administration routes: Compare intraperitoneal, subcutaneous, and intravenous delivery

    • Pharmacokinetics: Determine half-life and tissue distribution using labeled TGFBI 182 a.a.

  • Efficacy Assessment Metrics:

    • Glycemic control: Monitor fasting glucose, HbA1c, glucose tolerance

    • Insulin secretion: Perform in vivo glucose-stimulated insulin secretion tests

    • β-cell mass: Quantify through histomorphometry and markers of proliferation

    • Islet inflammation: Assess immune infiltration and inflammatory cytokine profiles

    • Signaling activation: Analyze AKT/mTORC1 pathway activation in pancreatic tissue

  • Mechanistic Investigation Design:

Research QuestionExperimental ApproachOutcome Measures
Is 182 a.a. fragment sufficient for islet protection?Compare 182 a.a. vs. full-length TGFBIβ-cell survival, insulin secretion
Which integrins mediate protective effects?Combine TGFBI with integrin-blocking antibodiesSignaling pathway activation
Does TGFBI affect immune cell function?Treat diabetogenic T cells with TGFBI 182 a.a.Proliferation, cytokine production
Can TGFBI enhance islet transplantation?Pre-treat donor islets with TGFBI before transplantGraft survival, function
  • Translational Considerations:

    • Safety assessment: Monitor for potential off-target effects, particularly in cancer models

    • Biomarker development: Identify markers that predict response to TGFBI therapy

    • Combination approaches: Test TGFBI with established diabetes medications

    • Delivery optimization: Develop sustained release formulations or targeted delivery systems

This comprehensive experimental framework enables rigorous evaluation of TGFBI 182 a.a. as a potential diabetes therapeutic while addressing key mechanistic questions about its mode of action and optimal clinical application .

How should researchers approach investigating the potential immunomodulatory effects of TGFBI 182 a.a. in the context of autoimmune conditions?

To thoroughly investigate the immunomodulatory properties of TGFBI 182 a.a. in autoimmune contexts, researchers should implement this comprehensive methodological framework:

  • Immune Cell Response Characterization:

    • T cell studies: Treat isolated CD4+ and CD8+ T cells with TGFBI 182 a.a. (1-50 μg/mL)

      • Measure proliferation (CFSE dilution)

      • Assess cytokine production profiles (Th1/Th2/Th17/Treg polarization)

      • Analyze activation markers (CD25, CD69, CD44)

    • Dendritic cell modulation: Evaluate effects on maturation and antigen presentation

      • Monitor surface markers (MHC-II, CD80/86, CD40)

      • Assess cytokine production (IL-12, IL-10, TGF-β)

    • Macrophage polarization: Determine M1/M2 balance after TGFBI treatment

      • Analyze marker expression (CD86 vs CD206)

      • Measure inflammatory cytokine production

  • Integrin-Dependent Mechanisms:

    • Perform comprehensive integrin expression profiling on immune cell subsets

    • Use blocking antibodies against candidate integrins (α3β1, αVβ3, αVβ5)

    • Conduct knockdown studies to confirm integrin dependency

    • Correlate integrin expression with TGFBI responsiveness across immune cell types

  • In Vivo Autoimmune Models:

    • Type 1 diabetes: NOD mice or multiple low-dose STZ model

    • Multiple sclerosis: Experimental autoimmune encephalomyelitis (EAE)

    • Rheumatoid arthritis: Collagen-induced arthritis model

    • Treatment protocols:

      • Preventive: Begin before disease onset

      • Therapeutic: Initiate after disease establishment

      • Local vs. systemic administration comparison

  • Mechanistic Signaling Analysis:

    • Focus on pathways known to regulate immune responses:

      • NF-κB activation status

      • STAT signaling (particularly STAT3/STAT5)

      • AKT/mTOR pathway in relation to T cell metabolism

    • Use pathway inhibitors to validate key nodes

    • Perform transcriptional profiling to identify global effects on immune response genes

  • Experimental Decision Tree:

Initial ObservationFollow-up StudiesFinal Assessment
Decreased T cell proliferationTest antigen-specific vs. polyclonal responsesDetermine mechanism of suppression
Altered cytokine profilesEvaluate epigenetic modifications at cytokine lociMap transcriptional regulation pathways
Enhanced Treg inductionAssess stability and suppressive functionDetermine therapeutic potential
Reduced autoimmune symptomsAnalyze tissue-specific immune infiltrationEvaluate long-term disease modification
  • Translational Research Considerations:

    • Compare findings with human autoimmune disease samples

    • Develop biomarkers to identify TGFBI-responsive patient subsets

    • Assess combination approaches with established immunomodulatory agents

    • Address potential adverse effects on protective immunity

This systematic approach enables comprehensive characterization of TGFBI 182 a.a.'s immunomodulatory functions while establishing mechanistic insights and therapeutic potential for autoimmune diseases .

What are the most common technical challenges encountered when working with TGFBI 182 a.a. in experimental systems, and what methodological solutions can address these issues?

Researchers working with TGFBI 182 a.a. frequently encounter several technical challenges that can be systematically addressed through optimized methodological approaches:

  • Protein Stability and Aggregation Issues:

    • Challenge: TGFBI 182 a.a. may form aggregates during storage or experimental handling

    • Solutions:

      • Add 20% glycerol to storage buffer to prevent aggregation

      • Include 0.1% carrier protein (HSA or BSA) for long-term storage

      • Centrifuge at 10,000g for 10 minutes before use to remove any pre-formed aggregates

      • Use dynamic light scattering to verify monodispersity before experiments

      • Optimize buffer conditions (consider adding low concentrations of non-ionic detergents)

  • Inconsistent Activity Between Batches:

    • Challenge: Functional variability between different preparations

    • Solutions:

      • Implement standardized activity assays for each batch (e.g., integrin binding assay)

      • Maintain detailed records of E. coli expression conditions

      • Use consistent purification protocols with validated chromatographic techniques

      • Prepare master stocks with verified activity

      • Consider including positive control proteins in experimental designs

  • Endotoxin Contamination:

    • Challenge: E. coli-derived proteins may contain endotoxin that confounds immunological experiments

    • Solutions:

      • Test each preparation using LAL assay or endotoxin-specific reporter cells

      • Implement endotoxin removal steps (Triton X-114 phase separation or polymyxin B columns)

      • Include endotoxin inhibitors (polymyxin B) in experiments if complete removal isn't possible

      • Use endotoxin-free consumables during purification and storage

  • Integrin Binding Specificity Validation:

    • Challenge: Confirming specific integrin interactions in complex biological systems

    • Solutions:

      • Employ cell lines with defined integrin expression profiles

      • Use integrin-blocking antibodies as controls in all experiments

      • Develop solid-phase binding assays with purified integrin components

      • Include integrin knockout or knockdown controls

      • Perform competitive binding studies with established integrin ligands

  • Troubleshooting Workflow for Common Issues:

ProblemDiagnostic ApproachCorrective Actions
Loss of protein activitySDS-PAGE analysis, circular dichroismPrepare fresh stock, verify pH and buffer composition
High experimental variabilityStatistical analysis of technical replicatesStandardize protocols, increase sample size
Unexpected cellular responsesVerify protein purity, endotoxin testingUse alternative preparation methods, include additional controls
Poor reproducibility between labsInter-laboratory validation studiesDevelop detailed SOPs, share reference material
  • Alternative Protein Production Strategies:

    • Consider mammalian expression systems for studies requiring post-translational modifications

    • Explore insect cell expression for higher yields with eukaryotic folding machinery

    • Investigate cell-free protein synthesis for rapid iteration of construct designs

These methodological solutions address the most common technical challenges associated with TGFBI 182 a.a. research, enabling more reproducible and reliable experimental outcomes .

How can researchers accurately quantify and validate the functional activity of TGFBI 182 a.a. in different experimental systems?

To ensure accurate quantification and functional validation of TGFBI 182 a.a. across experimental systems, researchers should implement this comprehensive methodological framework:

  • Protein Quantification Methods:

    • Primary quantification approaches:

      • BCA or Bradford assay for total protein concentration

      • SDS-PAGE with densitometry against BSA standards

      • Amino acid analysis for absolute quantification

    • Purity assessment:

      • SDS-PAGE with silver staining (>95% purity standard)

      • Western blotting with anti-TGFBI antibodies

      • Mass spectrometry for contaminant identification

    • Structural integrity validation:

      • Circular dichroism spectroscopy to confirm secondary structure

      • Size-exclusion chromatography to verify monomeric state

      • Dynamic light scattering to assess aggregation state

  • Functional Activity Assays:

    • Binding assays:

      • Solid-phase binding to purified integrins or ECM components

      • Surface plasmon resonance for binding kinetics

      • Cell adhesion assays using integrin-expressing cell lines

    • Signaling activation:

      • Phospho-specific Western blots for AKT, AKT1S1, RPS6, and EIF4EBP1

      • ELISA-based phosphoprotein quantification

      • Luciferase reporter assays for downstream transcriptional activation

    • Cell-based functional assays:

      • Islet cell survival assays following stress challenges

      • Migration/invasion assays for cancer cells

      • Proliferation assays using BrdU incorporation or Ki67 staining

  • Standardization and Reference Materials:

    • Develop internal reference standards with established activity

    • Create activity units based on specific biological responses

    • Implement positive controls (full-length TGFBI) in all experimental systems

    • Maintain detailed batch records with functional validation data

  • System-Specific Validation Approaches:

Experimental SystemValidation MethodExpected Outcome
Islet biologyGlucose-stimulated insulin secretionEnhanced secretion at 16.7 mM glucose
Cancer cell studiesDose-response proliferation/migrationSystem-specific response curve
Immunological experimentsT cell proliferation assaysMeasurable effect on activation/proliferation
In vivo applicationsPharmacokinetic/biodistribution studiesTissue-specific accumulation profile
  • Troubleshooting Activity Loss:

    • Implement freeze-thaw stability testing protocols

    • Evaluate temperature sensitivity profile

    • Assess pH stability range

    • Determine optimal buffer conditions for each application

    • Test additives (carrier proteins, glycerol) for stabilization

  • Advanced Functional Characterization:

    • Structure-function studies with point mutations or truncations

    • Domain-specific antibody blocking experiments

    • Competitive inhibition with peptide fragments

    • Cross-species activity comparison (human vs. mouse TGFBI)

This comprehensive approach ensures both accurate quantification and reliable functional validation of TGFBI 182 a.a. across diverse experimental systems, facilitating reproducible research outcomes and meaningful inter-laboratory comparisons .

What emerging technologies and methodological approaches might advance our understanding of TGFBI 182 a.a. functions in normal physiology and disease states?

The future of TGFBI 182 a.a. research will be significantly enhanced by integrating several cutting-edge technologies and methodological approaches:

  • Advanced Structural Biology Techniques:

    • Cryo-electron microscopy: Determine high-resolution structures of TGFBI 182 a.a. in complex with integrins or ECM components

    • AlphaFold2/RoseTTAFold: Apply AI-based protein structure prediction to model TGFBI-protein interactions

    • Hydrogen-deuterium exchange mass spectrometry: Map binding interfaces with high precision

    • Single-molecule FRET: Investigate conformational dynamics during ligand binding

  • CRISPR-Based Functional Genomics:

    • Domain-specific knock-in mutations: Generate precise modifications in the C-terminal region

    • CRISPRa/CRISPRi: Modulate endogenous TGFBI expression with temporal control

    • Base editing: Create specific point mutations corresponding to clinical variants

    • CRISPR screens: Identify genes that modify TGFBI function through synthetic interactions

  • Advanced Imaging Technologies:

    • Super-resolution microscopy: Visualize TGFBI 182 a.a. interactions with cellular components at nanoscale resolution

    • Intravital imaging: Track TGFBI dynamics in living tissues

    • Correlative light and electron microscopy: Combine functional and ultrastructural information

    • Mass cytometry imaging: Map TGFBI distribution in relation to tissue microenvironment

  • Single-Cell Multi-Omics Approaches:

    • scRNA-seq with TGFBI 182 a.a. treatment: Identify cell-specific transcriptional responses

    • Spatial transcriptomics: Map TGFBI-responsive cells within intact tissues

    • scATAC-seq: Determine chromatin accessibility changes following TGFBI signaling

    • Multimodal analysis: Integrate transcriptional, epigenetic, and proteomic data at single-cell resolution

  • Organoid and Microphysiological Systems:

    • Pancreatic islet organoids: Test TGFBI 182 a.a. effects on development and function

    • Multi-organ-on-chip platforms: Evaluate systemic effects and metabolic interactions

    • 3D bioprinting with TGFBI incorporation: Study spatial effects on tissue organization

    • Patient-derived organoids: Assess personalized responses to TGFBI treatment

  • Translational Research Approaches:

TechnologyApplication to TGFBI ResearchPotential Impact
Nanobody developmentGenerate highly specific inhibitors of TGFBI domainsTherapeutic targeting of specific functions
Targeted protein degradationDevelop PROTACs for TGFBISelective modulation of TGFBI levels
mRNA therapeuticsDeliver modified TGFBI mRNATransient expression in target tissues
AI-driven target predictionIdentify disease contexts for TGFBI interventionExpanded therapeutic applications
  • Systems Biology Integration:

    • Network analysis of TGFBI interactome across tissues

    • Mathematical modeling of TGFBI signaling dynamics

    • Multi-scale modeling connecting molecular events to tissue-level outcomes

    • Integration of clinical genomic data with experimental findings

These emerging technologies and methodological approaches will significantly advance our understanding of TGFBI 182 a.a. functions in both physiological and pathological contexts, potentially opening new avenues for therapeutic intervention in diabetes, cancer, and other TGFBI-associated conditions .

How might researchers better translate basic findings about TGFBI 182 a.a. into potential clinical applications for diabetes or corneal dystrophies?

Translating basic TGFBI 182 a.a. research into clinical applications requires a systematic translational research framework that bridges fundamental discoveries with therapeutic development:

  • Target Validation and Development Pipeline:

    • Human genetic validation:

      • Analyze TGFBI SNPs associated with diabetes risk in diverse populations

      • Correlate mutation types with specific corneal dystrophy phenotypes

      • Conduct functional genomics to determine mutation effects on protein activity

    • Preclinical proof-of-concept:

      • Test TGFBI 182 a.a. in humanized mouse models of diabetes

      • Develop corneal organoid models expressing dystrophy-associated mutations

      • Validate target engagement biomarkers for clinical translation

    • Therapeutic modality selection:

      • Recombinant protein therapy (TGFBI 182 a.a. or optimized variants)

      • TGFBI-mimetic peptides targeting specific integrin interactions

      • Gene therapy approaches for corneal dystrophies

      • Small molecule modulators of TGFBI downstream pathways

  • Diabetes-Focused Translation Strategy:

    • Therapeutic hypothesis refinement:

      • Determine if TGFBI 182 a.a. primarily protects β-cell function, survival, or regeneration

      • Evaluate effects on immune modulation in autoimmune diabetes

      • Assess combination potential with established diabetes therapies

    • Delivery optimization:

      • Develop sustained-release formulations to extend half-life

      • Explore β-cell targeting approaches

      • Investigate oral delivery systems for protein/peptide therapeutics

    • Clinical development path:

      • Initial focus on high-risk populations (pre-diabetes, post-transplant)

      • Biomarker-driven patient selection based on TGFBI pathway activity

      • Adaptive trial designs with multiple endpoints (glycemic control, β-cell function)

  • Corneal Dystrophy Translation Approach:

    • Precision medicine strategy:

      • Develop rapid TGFBI genotyping assays for clinical use

      • Match therapeutic approaches to specific mutation types

      • Create patient-derived corneal cell models for personalized testing

    • Therapeutic modalities:

      • Gene editing approaches (CRISPR) for correcting specific mutations

      • Antisense oligonucleotides to modulate mutant TGFBI expression

      • Stabilizing chaperones to prevent mutant protein aggregation

      • Local administration of wild-type TGFBI 182 a.a. to compete with mutant protein

  • Translational Research Roadmap:

Research StageDiabetes ApplicationsCorneal Dystrophy Applications
Target ValidationIslet-specific TGFBI knockout modelsMutation-specific knock-in models
Biomarker DevelopmentTGFBI pathway activation in accessible tissuesImaging biomarkers for protein aggregation
Therapeutic PrototypeOptimized TGFBI 182 a.a. variants with enhanced stabilityMutation-specific therapeutic approaches
Delivery DevelopmentSustained release formulations, targeting strategiesTopical/intracorneal delivery systems
Preclinical ValidationHumanized mouse models, non-human primatesCorneal organoids, ex vivo corneal models
Clinical TranslationRisk stratification based on TGFBI genotypeGenotype-guided therapy selection
  • Accelerating Clinical Implementation:

    • Engage regulatory agencies early with novel mechanism of action

    • Develop companion diagnostics to identify responsive patient populations

    • Partner with patient advocacy groups to facilitate clinical trial recruitment

    • Establish international research consortia to coordinate translational efforts

This comprehensive translational framework provides a systematic approach for moving TGFBI 182 a.a. discoveries from basic research into potential clinical applications, addressing the unique challenges of both diabetes and corneal dystrophy therapeutic development .

Product Science Overview

Introduction

Transforming Growth Factor Beta (TGF-β) is a multifunctional cytokine that plays a crucial role in regulating various cellular processes, including cell growth, differentiation, apoptosis, and extracellular matrix production. The TGF-β family consists of over 30 related cytokines, which act in a context-dependent manner . One of the key proteins in this family is the Transforming Growth Factor Beta-Induced (TGFBI) protein, which is encoded by the TGFBI gene.

TGFBI Protein

The TGFBI protein, also known as BIGH3, is a 68 kDa protein that contains an RGD (Arg-Gly-Asp) motif, which is found in many extracellular matrix proteins and is involved in cell adhesion . This protein binds to type I, II, and IV collagens and plays a role in cell-collagen interactions. It is induced by TGF-β and acts to inhibit cell adhesion . Mutations in the TGFBI gene are associated with various types of corneal dystrophy, including Thiel-Behnke and Reis-Bucklers types .

TGF-β Signaling Pathway

The TGF-β signaling pathway is a critical regulator of numerous cellular processes. It functions through both canonical SMAD-mediated processes and noncanonical pathways involving MAPK cascades, PI3K/AKT, Rho-like GTPases, and NF-κB signaling . This intricate signaling system is finely tuned by interactions between canonical and noncanonical pathways and plays key roles in both physiological and pathological conditions, including tissue homeostasis, fibrosis, and cancer progression .

Role in Cancer and Fibrosis

TGF-β signaling has paradoxical actions. Under normal physiological conditions, it promotes cell quiescence and apoptosis, acting as a tumor suppressor . However, in pathological states such as inflammation and cancer, it triggers processes that facilitate cancer progression and tissue remodeling, thus promoting tumor development and fibrosis . The dual roles of TGF-β signaling in both fibrosis and cancer highlight its complex behavior across different cellular contexts .

Human Recombinant TGFBI (182 a.a.)

Human recombinant TGFBI (182 amino acids) is a truncated form of the full-length TGFBI protein. Recombinant proteins are produced through recombinant DNA technology, which involves inserting the gene encoding the protein of interest into an expression system, such as bacteria or mammalian cells, to produce the protein in large quantities. This technology allows for the production of proteins that are identical to their natural counterparts, enabling researchers to study their functions and potential therapeutic applications.

Applications and Research

Recombinant TGFBI proteins are used in various research applications to study their roles in cell adhesion, migration, and signaling. They are also used to investigate the mechanisms underlying corneal dystrophies and other diseases associated with TGFBI mutations. Additionally, TGFBI proteins are being explored as potential therapeutic targets for conditions such as fibrosis and cancer, where TGF-β signaling plays a significant role .

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.