Pleiotrophin Human, His

Pleiotrophin Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Biological Functions

Pleiotrophin Human, His mimics the activity of endogenous PTN, a multifunctional cytokine involved in development, tissue repair, and disease.

Neuronal Development and Plasticity

  • Neurite outgrowth: PTN promotes axonal growth in embryonic rat, mouse, and chicken brain cells when coated on culture plates (3–8 µg/mL) .

  • Synaptic plasticity: Inhibits long-term potentiation in the hippocampus and regulates perineuronal nets .

  • Neuroprotection: Expressed post-ischemia or neuronal injury; supports oligodendrocyte progenitor survival via PTPRZ1 receptor .

Hematopoietic Stem Cell (HSC) Expansion

PTN enhances HSC regeneration in vitro and in vivo:

ParameterControl (TSF alone)PTN-Treated (100–500 ng/mL)
CD34+CD38−Lin− HSC countsBaseline4x increase in CFC content
SCID-repopulating capacityLow3x higher engraftment at 4 weeks
MechanismN/API3K/Notch signaling activation

Angiogenesis and Tumor Growth

  • Angiogenic factor: Promotes endothelial cell migration and tumor vascularization .

  • Cancer progression: Overexpressed in myeloma, breast, and pancreatic cancers; enhances metastasis via syndecan-3 and ALK receptors .

Metabolic Regulation

  • Glucose/lipid homeostasis:

    • Reduces hepatic steatosis and insulin resistance in Ptn-deficient mice .

    • Enhances β-cell proliferation in pancreatic islets during regeneration .

  • Muscle function: Maintains oxidative metabolism in skeletal muscle .

**3.1 In Vitro Studies

ApplicationProtocolOutcome
Neurite outgrowth assaysPre-coat plates with 3–8 µg/mL PTNEnhanced axonal extension in neurons
HSC expansionCulture CD34+ cells with 100–500 ng/mL PTNIncreased SCID-repopulating capacity
Macrophage migrationPTN-coated surfaces + Mac-1 integrinErk1/2 phosphorylation and cell spreading

**3.2 In Vivo Models

  • Hematopoietic recovery: Systemic PTN administration in irradiated mice expands bone marrow HSCs .

  • Cardiac repair: PTN overexpression in endothelial cells accelerates myocardial regeneration .

Therapeutic Potential

DiseasePTN RoleMechanism
Hematologic disordersHSC engraftment enhancerPI3K/Notch signaling activation
Diabetesβ-cell regeneration promoterInsulin gene expression induction
CancerTarget for anti-angiogenic therapySyndecan-3/ALK receptor antagonism

Diagnostic Biomarkers

  • Osteoarthritis: Elevated synovial fluid PTN levels correlate with early disease progression .

  • Aging: Circulating PTN levels increase with age, linked to metabolic decline .

Research Challenges and Future Directions

  • Receptor specificity: PTN binds multiple receptors (PTPRZ1, syndecan-3, ALK), complicating therapeutic targeting .

  • Toxicity concerns: Overexpression linked to tumor progression necessitates dose optimization .

  • Delivery systems: Development of PTN-mimetic peptides or gene therapy vectors for localized administration.

Product Specs

Introduction
Pleiotrophin, also known as Osteoblast-Specific Factor-1 (OSF-1), is a protein comprised of 136 amino acid residues. Its structure is notable for a high proportion of cationic amino acids (24%), with lysine clusters located at both the N-terminal and C-terminal ends. Gene expression analysis using Northern blotting revealed that the OSF-1 gene is expressed in mouse calvarial osteoblast-enriched cells and brain tissues. However, it was not detected in other tissues such as the thymus, spleen, kidney, liver, lung, testis, or heart. Pleiotrophin demonstrates the ability to stimulate adhesion, migration, proliferation, and differentiation in human osteoprogenitor cells. This embryonic growth and differentiation factor is not only present in certain types of cancer but also found in adults experiencing inflammatory diseases. Notably, in osteoarthritis, pleiotrophin is particularly prominent in the early stages, and its concentration in the synovial fluid could potentially serve as a marker for disease progression. Pleiotrophin might play a role in cartilage repair within the context of osteoarthritis, especially during its initial phases.
Description
Recombinant Human Pleiotrophin, a His-Tagged Fusion Protein, is produced in E. coli. This 17.3 kDa protein comprises 136 amino acid residues of human OSF-1 and an additional 16 amino acid residues, including a HisTag and a thrombin cleavage site.
Physical Appearance
White lyophilized powder after filtration.
Formulation
The protein solution, at a concentration of 0.5 mg/ml in 0.1M phosphate buffer with 0.1M NaCl at pH 7.2, is filtered and then lyophilized.
Solubility
To reconstitute, add PBS at pH 7.2 and allow the lyophilized pellet to dissolve completely.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to minimize repeated freezing and thawing. The reconstituted protein remains stable at 4°C for a limited period and shows no significant change after two weeks at this temperature.
Purity
Purity is determined to be greater than 95% using SDS-PAGE analysis.
Synonyms
PTN, Heparin Affin Regulatory Protein, HARP, Heparin-binding growth factor-8, HBGF-8, Osteoblast-Specific Factor-1, OSF-1, Heparin-binding growth-associated molecule, HB-GAM, HBNF-1 Heparin-binding brain mitogen, Heparin-binding neurite outgrowth-promoting factor 1, HBBM, NEGF1.
Source
Escherichia Coli.
Amino Acid Sequence
MKHHHHHHHM LVPRGSGKKE KPEKKVKKSD CGEWQWSVCV PTSGDCGLGT REGTRTGAEC KQTMKTQRCK IPCNWKKQFG AECKYQFQAW GECDLNTALK TRTGSLKRAL HNAECQKTVT ISKPCGKLTK PKPQAESKKK KKEGKKQEKM LD.

Q&A

What is Pleiotrophin and what are its primary biological functions?

Pleiotrophin (PTN) is an 18-kDa neurotrophic heparin-binding cytokine that was initially discovered in the early 1990s and has since been associated with numerous physiological events. The protein is encoded by the Ptn gene, which produces a basic protein of 168 amino acids that undergoes post-transcriptional modifications to yield the active 136 amino acid protein with a 32 amino acid signal peptide . PTN is highly expressed during embryonic development and early differentiation, with expression decreasing in adulthood except in the bone and nervous system .

The primary biological functions of PTN include:

  • Neural development during embryogenesis and the neonatal period

  • Tissue regeneration and bone repair

  • Angiogenesis and vascular remodeling

  • Modulation of inflammatory processes

  • Regulation of adipocyte differentiation and lipid metabolism

  • Hepatic lipid homeostasis

  • Pancreatic β-cell function and insulin secretion

PTN has been shown to bind to multiple receptors, particularly glycosaminoglycan (GAG)-containing proteoglycans, activating various intracellular kinases that control cellular functions .

How is PTN expression regulated in human tissues?

PTN expression is regulated through multiple mechanisms:

  • Developmental regulation: Expression is highest during embryonic development and early cell differentiation, with subsequent downregulation in most adult tissues except bone and the nervous system .

  • Growth factors and cytokines: PTN expression is enhanced by several growth factors and cytokines, including:

    • Androgens

    • Tumor necrosis factor alpha (TNFα)

    • Epidermal growth factor (EGF)

    • Platelet-derived growth factor (PDGF)

    • Basic fibroblast growth factor (FGF)

  • Vitamin D regulation: PTN expression is downregulated in a dose-dependent manner by 1α,25-Dihydroxyvitamin D(3) .

  • MicroRNA control: Several miRNAs regulate PTN expression:

    • miR-143 represses Ptn expression and enhances preadipocyte differentiation

    • miR-182 downregulates PTN levels in endometrial tissue

    • miR-137 and miR-384 are negatively correlated with PTN expression

    • miR-499 and miR-1709 regulate Ptn expression through DNA methylation changes

In adults, circulating PTN levels are significantly associated with advancing chronological age, confirming its continued expression in adult tissues .

What methodologies are recommended for detecting PTN in human tissue samples?

For reliable detection of PTN in human tissue samples, researchers should consider the following methodological approach:

  • Immunohistochemistry/Immunofluorescence:

    • Use antigen retrieval in citrate buffer (10mM sodium citrate, 0.05% Tween20, pH 6.0) for 30 min at 100°C

    • Block non-specific binding with appropriate blocking solution

    • Apply PTN-specific primary antibodies (1:200 rabbit polyclonal antibodies work well)

    • Use fluorescent-labeled secondary antibodies for visualization

    • Include DAPI nuclear counterstaining

    • Always perform control experiments including:

      • Omission of primary antibody

      • Omission of secondary antibody

      • Pre-absorption of primary antibody with excess recombinant PTN

  • Western Blotting:

    • Use denaturing conditions with appropriate sample preparation

    • Include positive controls (recombinant PTN) and tissue samples known to express PTN

    • Validate antibody specificity with competing peptides

  • PCR and qRT-PCR:

    • Design primers specific to human Ptn gene sequences

    • Include appropriate housekeeping genes for normalization

    • Consider the potential presence of splice variants

How can recombinant human PTN with histidine tags be effectively produced?

For researchers seeking to produce recombinant human PTN with histidine tags:

  • Expression System Selection:

    • Prokaryotic systems (E. coli): Suitable for high yield but may lack proper post-translational modifications

    • Eukaryotic systems (mammalian cells, insect cells): Provide proper folding and post-translational modifications

    • Consider CHO or HEK293 cells for mammalian expression with proper glycosylation

  • Vector Design:

    • Include the human PTN coding sequence (lacking the signal peptide for cytoplasmic expression)

    • Add a 6×His tag, preferably at the N-terminus to avoid interfering with C-terminal functional domains

    • Include appropriate purification and detection tags

    • Use inducible promoters for controlled expression

  • Purification Strategy:

    • Use immobilized metal affinity chromatography (IMAC) with Ni-NTA or Co-NTA columns

    • Employ a gradient elution with imidazole to minimize non-specific binding

    • Consider size exclusion chromatography as a second purification step

    • Dialyze against appropriate buffers to remove imidazole and other contaminants

  • Quality Control:

    • Confirm protein identity by mass spectrometry

    • Validate biological activity using cell-based assays

    • Assess endotoxin levels if intended for in vivo applications

    • Verify proper folding through circular dichroism spectroscopy

How does PTN influence pancreatic β-cell function and insulin secretion?

PTN plays a significant role in pancreatic development and β-cell function:

  • Developmental Role:

    • PTN is highly expressed during embryonic and fetal development in organs undergoing branching morphogenesis, including the pancreas

    • During pancreatic development (E11-13 in mice), PTN is localized in basement membranes of the pancreatic epithelium

    • Antisense inhibition of Ptn expression impairs differentiation of endocrine precursors and reduces glucagon and insulin expression

  • Adult β-cell Expression:

    • PTN maintains high expression levels in adult pancreatic β-cells of rats, mice, and humans

    • PTN is prominently expressed in Ins+ cells (immature pancreatic β-cells with multipotential islet endocrine cell lineage potential)

  • Effect on Insulin Secretion:

    • Treatment with recombinant PTN (1μg/ml) affects glucose-stimulated insulin secretion in isolated islets

    • Experimental protocol: Pre-incubation of islets in RPMI media containing 6mM glucose for 24h with or without PTN, followed by insulin secretion measurement

    • PTN administration in insulinoma cell lines induces β-cell expansion and enhances expression of insulin-related genes

  • Relationship to Metabolic Disorders:

    • Ptn-/- mice develop insulin resistance with altered glucose and lipid metabolism

    • During pregnancy, PTN deletion leads to glucose intolerance, reflecting a diabetogenic state

    • PTN expression in islets is down-regulated in diabetic animals

What experimental models are most appropriate for studying PTN function in metabolic disorders?

Researchers investigating PTN's role in metabolic disorders should consider these experimental models:

  • Genetic Models:

    • PTN Knockout Models: Ptn-/- mice show insulin resistance, decreased fat accumulation, enhanced lipolysis, and protection against hepatic steatosis on high-fat diets

    • PTN Transgenic Models: Tissue-specific PTN overexpression models provide insight into tissue-specific effects

    • Inducible Models: Consider temporal control of PTN expression to distinguish developmental from adult effects

  • Dietary Intervention Models:

    • High-Fat Diet (HFD): Ptn deletion protects against HFD-induced obesity, neuroinflammation, mitochondrial dysfunction, and protein aggregation

    • Pregnancy Models: Ptn-/- mice show altered metabolism during pregnancy with implications for gestational diabetes research

  • Cellular Models:

    • Pancreatic β-cell Lines: For studying insulin secretion and β-cell proliferation

    • Adipocyte Models: Primary preadipocytes or cell lines (e.g., 3T3-L1) for differentiation studies

    • Hepatocyte Models: Primary hepatocytes or HepG2 cells for lipid metabolism studies

    • Muscle Cell Models: C2C12 cells for studying oxidative metabolism

  • Ex Vivo Systems:

    • Isolated Islets: For glucose-stimulated insulin secretion studies

    • Adipose Tissue Explants: For lipolysis and browning studies

    • Precision-Cut Liver Slices: For hepatic metabolism studies

How do the structural characteristics of PTN influence its receptor interactions and signaling?

The structural aspects of PTN significantly influence its receptor interactions and downstream signaling:

  • Domain Structure and Function:

    • PTN consists of two β-sheet domains connected by a flexible linker

    • The N-terminal domain is involved in receptor binding

    • The C-terminal domain contributes to GAG binding

    • The flexible linker enables conformational changes upon binding

  • Receptor Interactions:

    • PTN binds to multiple receptors:

      • Protein tyrosine phosphatase receptor type Z (PTPRZ)

      • Anaplastic lymphoma kinase (ALK)

      • Syndecan family proteoglycans

      • Neuropilin-1

      • Integrin αvβ3

    • The receptor interactions are primarily mediated through electrostatic interactions

    • PTN-induced receptor oligomerization appears to be one mechanism by which PTN controls cellular functions

  • Signaling Pathways:

    • PTN activates multiple signaling cascades:

      • PI3K/Akt pathway: Important for cell survival and metabolism

      • MAPK/ERK pathway: Critical for cell proliferation

      • JAK/STAT pathway: Involved in cytokine responses

      • β-catenin pathway: Important for cell adhesion and gene transcription

  • Structure-Activity Relationships:

    • The heparin-binding domains of PTN are crucial for its biological activity

    • Mutations in specific lysine or arginine residues can dramatically alter binding and signaling

    • The His-tag position can affect receptor interactions when using recombinant proteins

What are the methodological approaches for analyzing PTN's effects on cellular metabolism?

When investigating PTN's effects on cellular metabolism, researchers should consider these methodological approaches:

  • Metabolic Flux Analysis:

    • Seahorse XF Analyzer: Measures oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) to assess mitochondrial respiration and glycolysis

    • Stable Isotope Tracing: Use 13C-labeled glucose, glutamine, or fatty acids to track metabolic pathway utilization

    • NMR Spectroscopy: For comprehensive metabolite profiling

  • Lipid Metabolism Assessment:

    • Lipogenesis Assays: Measure incorporation of labeled acetate or glucose into lipids

    • Lipolysis Assays: Quantify glycerol or free fatty acid release from adipocytes

    • Fatty Acid Oxidation: Measure 14C-labeled CO2 production from labeled fatty acids

    • Gene Expression Analysis: Assess expression of lipogenic genes (FASN, ACC, SCD1) and lipolytic genes (ATGL, HSL)

  • Glucose Metabolism Studies:

    • Glucose Uptake Assays: Using 2-deoxyglucose

    • Glucose-Stimulated Insulin Secretion: In isolated islets or β-cell lines

    • Glycogen Synthesis: Measuring incorporation of labeled glucose into glycogen

    • Glucose Production: Hepatic glucose output assays

  • Mitochondrial Function:

    • Citric Acid Cycle Activity: Measure enzyme activities and intermediates

    • Electron Transport System Assessment: Analyze complex activities and oxygen consumption

    • Mitochondrial Membrane Potential: Using fluorescent dyes (TMRM, JC-1)

    • ROS Production: Using specific fluorescent probes

  • Insulin Signaling:

    • Phosphorylation Status: Western blotting for phosphorylated IRS, Akt, and AS160

    • Glucose Transporter Translocation: GLUT4 trafficking in muscle and adipose cells

    • Insulin Resistance Models: HOMA-IR index calculations

How can researchers effectively investigate the role of PTN in tissue-specific metabolic regulation?

For researchers investigating PTN's tissue-specific metabolic functions:

  • Liver-Specific Studies:

    • Hepatic Lipid Content: Histological analysis (Oil Red O staining) and biochemical measurements

    • Lipogenic Enzyme Expression: qRT-PCR and Western blotting for FASN, ACC, and SCD1

    • Fatty Acid and Triglyceride Synthesis: Isotope incorporation studies

    • Hepatic Secretion Assays: VLDL secretion rates and apolipoprotein production

    • PPARα and NUR77 Activation: Binding assays and target gene expression

  • Adipose Tissue Studies:

    • Adipocyte Differentiation: Oil Red O staining and adipogenic gene expression

    • Lipolysis Assessment: Glycerol release and HSL/ATGL activation

    • Browning of White Adipose: UCP1 expression and mitochondrial content

    • Thermogenic Activity: Infrared imaging and core temperature measurements

    • Free Fatty Acid Release: Plasma FFA levels and tissue-specific release rates

  • Skeletal Muscle Studies:

    • Oxidative Metabolism: Citric acid cycle and electron transport system activity

    • Fiber Type Composition: Histochemical and immunofluorescence analysis

    • Vascularization: CD31 staining and angiogenic factor expression

    • Glucose Uptake: 2-deoxyglucose uptake assays

    • Insulin Sensitivity: Insulin-stimulated Akt phosphorylation

  • Pancreatic Studies:

    • β-Cell Mass and Proliferation: Morphometric analysis and BrdU incorporation

    • Islet Architecture: Immunofluorescence for insulin, glucagon, and somatostatin

    • Glucose-Stimulated Insulin Secretion: Static and perifusion assays

    • β-Cell Gene Expression: RNA-seq or qRT-PCR for insulin, PDX1, GLUT2, etc.

    • Islet Development: Branching morphogenesis in embryonic pancreas explants

How can researchers resolve contradictory findings in PTN research?

When faced with contradictory findings in PTN research, consider these methodological approaches:

  • Context-Dependent Effects:

    • Developmental Stage: PTN effects differ significantly between embryonic, neonatal, and adult tissues

    • Physiological State: Consider differences between normal, pregnant, and disease states

    • Species Differences: Human PTN may have different effects than murine PTN despite >90% sequence conservation

    • Tissue-Specific Responses: PTN actions in liver may contradict those in adipose tissue

  • Methodological Variations:

    • Recombinant Protein Sources: Different tags (His, GST) or expression systems may affect activity

    • Concentration Dependencies: PTN may have bell-shaped dose-response curves

    • Acute vs. Chronic Exposure: Short-term effects may differ from long-term adaptations

    • In Vitro vs. In Vivo Discrepancies: Isolated cell responses may not translate to whole organisms

  • Receptor Expression Patterns:

    • Receptor Profiling: Quantify expression levels of all PTN receptors in the experimental system

    • Receptor Competition: Consider interactions with other growth factors sharing receptors

    • Signaling Crosstalk: Map intersecting pathways that may amplify or suppress responses

  • Statistical Approaches:

    • Meta-Analysis: Pool data from multiple studies to identify consistent patterns

    • Multivariate Analysis: Consider how multiple variables interact to influence outcomes

    • Power Calculations: Ensure adequate sample sizes to detect true effects

What are the key considerations for experimental design when studying PTN's effects on energy metabolism?

For robust experimental design when studying PTN's effects on energy metabolism:

  • Control Considerations:

    • Wild-Type Controls: Age, sex, and background-matched for genetic models

    • Vehicle Controls: Ensure carrier solutions don't have metabolic effects

    • Time Controls: Account for circadian variations in metabolism

    • Diet Controls: Standardize feeding protocols and diet composition

  • Dosing Parameters:

    • Dose-Response Relationships: Test multiple concentrations (0.1-10 μg/ml typical range)

    • Timing of Administration: Consider acute vs. chronic effects

    • Route of Administration: Different for in vitro (culture media) vs. in vivo (injection routes)

    • Pharmacokinetics: Account for half-life and tissue distribution

  • Metabolic State Variables:

    • Nutritional State: Fasted vs. fed conditions dramatically affect results

    • Activity Level: Control for or measure physical activity

    • Thermoneutral vs. Cold Conditions: Particularly important for energy expenditure studies

    • Age Effects: Young animals have different metabolic profiles than aged ones

  • Comprehensive Phenotyping:

    • Whole-Body Metabolism: Indirect calorimetry, body composition, food intake

    • Glucose Homeostasis: GTT, ITT, hyperinsulinemic-euglycemic clamps

    • Tissue-Specific Metabolism: Ex vivo tissue function tests

    • Molecular Signaling: Phosphorylation cascades and transcriptional responses

How should PTN concentration data in biological samples be normalized and interpreted?

When analyzing PTN concentration data from biological samples:

  • Sample Collection and Processing:

    • Standardized Collection: Time of day, fasting status, sample handling

    • Processing Methods: Consistent extraction protocols for tissues

    • Storage Conditions: -80°C for long-term stability

    • Freeze-Thaw Cycles: Minimize and standardize across samples

  • Normalization Approaches:

    • Total Protein Normalization: Bradford or BCA assays for tissue homogenates

    • DNA Content: For cellular studies to account for cell number

    • Tissue Weight: For solid tissue samples (wet or dry weight)

    • Internal Standards: Spike-in controls for extraction efficiency

  • Analytical Methods:

    • ELISA: Commercial kits vs. lab-developed assays

    • Western Blotting: Semi-quantitative with appropriate loading controls

    • Mass Spectrometry: For absolute quantification

    • Method Validation: Linearity, recovery, precision, and accuracy

  • Data Interpretation:

    • Reference Ranges: Establish normal ranges for your experimental system

    • Fold Changes: Often more reliable than absolute values across studies

    • Biological Context: Correlate with physiological parameters

    • Multiple Timepoints: Capture dynamic changes rather than single points

What approaches are recommended for integrating PTN functional data with other -omics datasets?

For integrating PTN functional data with other -omics datasets:

  • Multi-Omics Integration Strategies:

    • Correlation Networks: Identify genes, proteins, or metabolites that correlate with PTN levels

    • Pathway Enrichment: Map affected pathways across different -omics layers

    • Causal Modeling: Use directed acyclic graphs to infer causal relationships

    • Machine Learning Approaches: Supervised and unsupervised learning to identify patterns

  • Transcriptomic Integration:

    • RNA-Seq After PTN Treatment: Identify directly and indirectly regulated genes

    • ChIP-Seq for Downstream Transcription Factors: Map the regulatory network

    • Single-Cell RNA-Seq: Identify cell-specific responses within heterogeneous tissues

    • Temporal Transcriptomics: Capture the dynamics of the response

  • Proteomic Integration:

    • Phosphoproteomics: Map PTN-induced signaling cascades

    • Interaction Proteomics: Identify protein complexes formed after PTN stimulation

    • Secretome Analysis: Identify secondary mediators released after PTN treatment

    • Protein Turnover Studies: Distinguish changes in synthesis vs. degradation

  • Metabolomic Integration:

    • Targeted Metabolomics: Focus on pathways known to be affected by PTN

    • Untargeted Metabolomics: Discover novel metabolic effects

    • Flux Analysis: Determine how PTN alters metabolic pathway activities

    • Lipid Profiling: Given PTN's effects on lipid metabolism

What is the current understanding of PTN's role in metabolic diseases?

PTN has emerging roles in several metabolic diseases:

  • Insulin Resistance and Type 2 Diabetes:

    • PTN knockout mice develop insulin resistance despite lower body fat and enhanced lipolysis

    • PTN expression changes in pancreatic islets during diabetic conditions

    • PTN is involved in β-cell mass regulation and insulin secretion

    • PTN deletion blocks HFD-induced hyperinsulinemia and insulin resistance

  • Non-Alcoholic Fatty Liver Disease (NAFLD):

    • PTN deletion protects against HFD-induced hepatic steatosis

    • PTN influences hepatic lipid accumulation and the expression of lipogenic enzymes

    • PTN is necessary for hepatic homeostasis during pregnancy

    • PTN regulates PPARα and NUR77 activation, affecting lipid and carbohydrate metabolism

  • Obesity and Adipose Tissue Dysfunction:

    • PTN expression increases in preadipocytes during confluence and decreases during differentiation

    • PTN inhibits preadipocyte differentiation when added exogenously

    • PTN deletion impairs fat accumulation and adipose tissue expandability

    • PTN deletion promotes browning of white adipose tissue and increases thermogenic activity

  • Aging-Related Metabolic Decline:

    • Circulating PTN levels are significantly associated with advancing chronological age

    • PTN may contribute to age-related changes in tissue regeneration and metabolism

    • PTN influences mitochondrial function and oxidative metabolism

How can researchers best design translational studies to investigate PTN as a therapeutic target?

For researchers designing translational studies on PTN as a therapeutic target:

  • Target Validation Approaches:

    • Genetic Association Studies: Examine PTN polymorphisms in metabolic disease cohorts

    • Expression Studies: Compare PTN levels in healthy vs. diseased human tissues

    • Receptor Profiling: Characterize PTN receptor expression in target tissues

    • Pathway Analysis: Confirm conservation of PTN signaling between models and humans

  • Intervention Strategies:

    • Recombinant PTN Administration: Dose, timing, and route optimization

    • PTN Neutralizing Antibodies: For conditions where PTN inhibition is desired

    • Small Molecule Modulators: Screen for compounds that enhance or inhibit PTN signaling

    • Receptor-Specific Targeting: Design agonists or antagonists for specific PTN receptors

  • Preclinical Model Selection:

    • Humanized Mouse Models: For better translation of findings

    • Large Animal Models: For pharmacokinetics and safety studies

    • Ex Vivo Human Tissue Studies: Using surgical or biopsy specimens

    • iPSC-Derived Human Cells: For personalized drug response testing

  • Biomarker Development:

    • Circulating PTN Levels: Standardize assays for clinical use

    • Pathway Activation Markers: Phosphorylation of downstream targets

    • Tissue Response Markers: Changes in target tissue metabolism or function

    • Genetic Predictors: Identify variants that predict response to PTN modulation

What methodological considerations are important for using PTN as a biomarker in metabolic disorders?

When developing PTN as a biomarker for metabolic disorders:

  • Assay Development and Validation:

    • Antibody Selection: Validate antibodies against recombinant PTN standards

    • Assay Platform: ELISA, multiplexed assays, or mass spectrometry

    • Sample Matrix Effects: Validate in the appropriate biological fluid (serum, plasma)

    • Reference Standards: Establish certified reference materials

  • Preanalytical Variables:

    • Circadian Variation: Standardize collection time

    • Fasting Status: Control for or document feeding state

    • Exercise Effects: Account for recent physical activity

    • Medication Effects: Document concurrent medications that may affect PTN levels

  • Clinical Validation:

    • Reference Ranges: Establish in healthy populations stratified by age and sex

    • Disease Association: Compare levels across disease states and severities

    • Longitudinal Studies: Track changes over disease progression

    • Intervention Response: Monitor changes with therapeutic interventions

  • Statistical Considerations:

    • ROC Analysis: Determine sensitivity and specificity for disease detection

    • Multivariate Models: Combine PTN with other biomarkers for improved prediction

    • Confounding Factors: Adjust for BMI, age, sex, and other variables

    • Risk Stratification: Develop cutoffs for clinical decision-making

What are the optimal conditions for working with recombinant PTN in experimental systems?

For optimal handling and use of recombinant PTN:

  • Storage and Stability:

    • Storage Temperature: -80°C for long-term storage of lyophilized protein

    • Working Solutions: Store at -20°C in single-use aliquots

    • Freeze-Thaw Cycles: Minimize; three or fewer cycles recommended

    • Buffer Composition: PBS with 0.1% BSA as a carrier protein improves stability

  • Reconstitution and Handling:

    • Reconstitution Solution: Sterile PBS or water; avoid DMSO or organic solvents

    • Concentration: Prepare stock solutions of 100-1000 μg/ml

    • Filtration: Use low protein-binding 0.22 μm filters if sterility is required

    • Temperature: Handle on ice when preparing solutions

  • Experimental Conditions:

    • Effective Concentrations: Typically 0.1-1 μg/ml for cell culture; may require higher doses (1-10 μg/ml) for some applications

    • Treatment Duration: Effects may vary between acute (minutes to hours) and chronic (days) exposure

    • Cell Culture Media: Serum may contain PTN or PTN-binding proteins; consider serum-free conditions

    • Binding Surfaces: PTN binds to glass and some plastics; pre-coat surfaces with BSA

  • Quality Control:

    • Activity Testing: Verify mitogenic activity on responsive cell lines

    • Endotoxin Testing: Critical for in vivo applications (<0.1 EU/μg protein)

    • Protein Integrity: Verify by SDS-PAGE before experiments

    • Biological Validation: Confirm receptor activation in your experimental system

How can researchers effectively design loss-of-function and gain-of-function studies for PTN?

For designing effective gain and loss-of-function studies:

  • Loss-of-Function Approaches:

    • Genetic Knockouts: Complete Ptn gene deletion in animal models

    • Conditional Knockouts: Tissue-specific and/or inducible Cre-loxP systems

    • RNA Interference: siRNA or shRNA targeting PTN mRNA

    • CRISPR/Cas9: Gene editing for precise mutations

    • Neutralizing Antibodies: Acute inhibition of extracellular PTN

    • Receptor Antagonists: Block downstream signaling

  • Gain-of-Function Approaches:

    • Recombinant Protein Administration: Direct addition of PTN protein

    • Viral Overexpression: Adenoviral, lentiviral, or AAV vectors for in vitro and in vivo expression

    • Transgenic Overexpression: Tissue-specific or inducible promoters

    • mRNA Delivery: For transient expression

    • Receptor Activation: PTN receptor agonists

  • Control Considerations:

    • Dose Titration: Establish dose-response relationships

    • Time Course Studies: Capture both immediate and delayed effects

    • Off-Target Effects: Include scrambled siRNAs, isotype antibodies

    • Rescue Experiments: Restore function to confirm specificity

    • Multiple Approaches: Combine genetic and pharmacological methods

  • Readout Selection:

    • Direct Targets: Immediate receptor activation and signaling

    • Secondary Responses: Transcriptional changes (6-24h)

    • Functional Outcomes: Metabolic, proliferative, or differentiation effects

    • Organ-Level Changes: Tissue remodeling or functional adaptation

    • Systemic Effects: Whole-body metabolic parameters

What technical challenges should researchers anticipate when studying PTN's interaction with its receptors?

When investigating PTN-receptor interactions, anticipate these technical challenges:

  • Receptor Complexity:

    • Multiple Receptors: PTN interacts with at least five different receptor types

    • Receptor Crosstalk: Interactions between different PTN receptors

    • Co-Receptors: Potential requirement for proteoglycans as co-receptors

    • Receptor Distribution: Tissue and cell-specific expression patterns

  • Binding Studies:

    • Affinity Measurements: Surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC)

    • Competition Assays: Displacement of labeled PTN by unlabeled PTN or competitors

    • Receptor Density: Account for variations in receptor expression levels

    • Non-Specific Binding: High background due to PTN's heparin-binding properties

  • Signaling Analysis:

    • Temporal Dynamics: Rapid and transient phosphorylation events

    • Pathway Complexity: Multiple parallel and intersecting pathways

    • Cell Type Specificity: Different signaling outcomes in different cell types

    • Signal Amplification: Secondary messengers and feedback loops

  • Visualization Techniques:

    • Co-Localization Studies: Requires specific antibodies for both PTN and receptors

    • FRET/BRET Approaches: For real-time interaction monitoring

    • Live Cell Imaging: Challenges with maintaining receptor functionality when tagged

    • Super-Resolution Microscopy: For detailed receptor clustering analysis

What are the current technological limitations in PTN research and potential future directions?

Current limitations and future directions in PTN research include:

  • Current Limitations:

    • Receptor Specificity: Difficulty in attributing effects to specific receptors

    • Isoform Characterization: Limited understanding of potential PTN isoforms

    • Temporal Resolution: Challenges in capturing rapid signaling dynamics

    • Tissue Heterogeneity: Cell-specific responses within complex tissues

    • Translational Gaps: Differences between model systems and human physiology

  • Emerging Technologies:

    • Single-Cell Analysis: Resolving cell-specific responses in heterogeneous tissues

    • CRISPR Screens: Systematic identification of PTN pathway components

    • Proximity Labeling: MS-based identification of PTN-interacting proteins

    • Organ-on-Chip Models: Better recapitulation of tissue complexity

    • In Vivo Imaging: Real-time monitoring of PTN activity

  • Future Directions:

    • Receptor-Specific Modulators: Development of agonists/antagonists for individual PTN receptors

    • Systems Biology Approaches: Integrative modeling of PTN networks

    • Epigenetic Regulation: Understanding how PTN expression is controlled

    • Precision Medicine: Targeting PTN pathways based on individual genetics

    • Therapeutic Development: PTN-based interventions for metabolic disorders

  • Methodological Advancements:

    • Engineered PTN Variants: Structure-function studies with domain-specific mutations

    • Optogenetic Control: Light-activated PTN signaling

    • Biosensors: Real-time monitoring of PTN activity

    • Computational Modeling: Prediction of PTN-receptor interactions and signaling outcomes

Product Science Overview

Biological Significance

Pleiotrophin plays a crucial role in various biological processes:

  • Neural Development: It stimulates neural development and provides trophic support, promoting positive recovery in neurodegenerative disorders .
  • Hematopoietic Stem Cell Maintenance: PTN is involved in the maintenance of hematopoietic stem cells .
  • Cell Differentiation: It aids in cell differentiation, contributing to the development and function of various tissues .

High serum levels of pleiotrophin are associated with a variety of solid tumors, including multiple myeloma . However, its expression is very limited in normal adult tissues .

Recombinant Pleiotrophin

Recombinant pleiotrophin is produced using Escherichia coli (E. coli) expression systems . The recombinant protein is often tagged with a His tag to facilitate purification and detection. The His tag is a sequence of histidine residues that binds to nickel ions, allowing for easy isolation of the protein using nickel-affinity chromatography.

Applications

Recombinant pleiotrophin is used in various research applications, including:

  • Functional Assays: To study its role in cell signaling and development .
  • ELISA: For quantifying pleiotrophin levels in biological samples .
  • Immunohistochemistry: To visualize pleiotrophin expression in tissue samples .
  • Western Blot: For detecting pleiotrophin in protein extracts .
Preparation and Storage

The lyophilized recombinant pleiotrophin is reconstituted in sterile, distilled water to a concentration of 0.1-1.0 mg/mL. It is recommended to store the reconstituted protein at 2°C to 8°C for up to one week. For extended storage, it should be further diluted in a buffer containing a carrier protein, such as 0.1% bovine serum albumin (BSA), and stored in working aliquots at -20°C to -80°C .

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.