Recombinant Human Parathyroid hormone protein (PTH), partial (Active)

Shipped with Ice Packs
In Stock

Description

Definition and Scope

Recombinant Human Parathyroid Hormone protein (PTH), partial (Active) refers to truncated forms of PTH produced via genetic engineering, retaining biological activity. Common variants include:

  • PTH (1-34): The N-terminal 34-amino acid fragment, which replicates the full hormone’s activity in bone and kidney regulation .

  • PTH (1-84): The full mature hormone (84 amino acids after post-translational processing), used in hypoparathyroidism treatment .

These fragments are synthesized in microbial systems like Escherichia coli or yeast, with >95% purity and low endotoxin levels (<1 EU/μg) .

Amino Acid Sequence

  • PTH (1-34): Sequence: M S V S E I Q L M H N L G K H L N S M E R V E W L R K K L Q D V H N F V A L G A P L A P R D A G .

  • Key domains:

    • N-terminal (1-34): Binds PTH1R receptor to activate cAMP/PKA and PLC/PKC pathways .

    • Mid-region (38-106): Contains nuclear localization signals for intracrine signaling .

Receptor Interaction

PTH activates the PTH1R receptor via a two-site mechanism:

  1. Site 1 (AA 12–34): Binds extracellular domain for affinity.

  2. Site 2 (AA 1–12): Triggers conformational changes in transmembrane helices for signaling .

Production Methods

ParameterPTH (1-34)PTH (1-84)
Expression SystemE. coli , Hansenula polymorpha E. coli
Purity>95% >95%
YieldHigh (optimized fermentation) Moderate

Yeast systems like H. polymorpha reduce production costs by 50% compared to bacterial systems .

Mechanisms of Action

  • Bone: Stimulates osteoclast-mediated resorption via RANKL/TRANCE upregulation, releasing calcium into the bloodstream .

  • Kidney: Reduces calcium excretion and enhances phosphate excretion .

Functional Assays

  • cAMP induction: ED₅₀ ≤50 ng/ml in MC3T3/E1 osteoblasts .

  • Glycogen synthesis: Activates 2-deoxy-D-glucose transport in osteoblastic cells .

Hypoparathyroidism

  • REPLACE Trial (rhPTH[1-84]): 53% of patients achieved ≥50% reduction in calcium/vitamin D supplements vs. 2% on placebo (P<0.0001) .

  • Safety: Adverse events (e.g., hypocalcemia, nausea) were comparable to placebo .

Osteoporosis

  • PTH (1-34) (Teriparatide): Increases bone mineral density by 9–13% over 24 months .

Pharmacological Data

ParameterPTH (1-34)PTH (1-84)
Half-life1 hour (subcutaneous) 3 hours (subcutaneous)
Dosing20 μg/day 50–100 μg/day
Cost~€11,000/24 months Higher (complex production)

Research and Development Frontiers

  • Small-molecule agonists: Compounds like PCO371 target PTH1R’s Proline 415 residue for oral bioavailability .

  • Gene therapy: Exploring sustained PTH secretion via viral vectors .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered phosphate-buffered saline (PBS), pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Avoid repeated freeze-thaw cycles. Store reconstituted aliquots at 4°C for up to one week.
Reconstitution
Prior to opening, briefly centrifuge the vial to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting to -20°C/-80°C. Our standard protocol utilizes 50% glycerol; this may serve as a reference for your application.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the inherent stability of the protein. Generally, the liquid form has a shelf life of 6 months at -20°C/-80°C, while the lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
hPTH; Parathormone; Parathyrin; Parathyroid hormone 1; Parathyroid hormone; Prepro PTH; Preproparathyroid hormone; PTH; PTH1; PTH1 receptor; PTH1R; PTHR; PTHR1; PTHY_HUMAN
Datasheet & Coa
Please contact us to get it.
Expression Region
32-65aa
Mol. Weight
4.1 kDa
Protein Length
Partial
Purity
>97% as determined by SDS-PAGE.
Research Area
Signal Transduction
Source
E.coli
Species
Homo sapiens (Human)
Target Names
PTH
Uniprot No.

Target Background

Function

Parathyroid hormone (PTH) elevates calcium levels by increasing bone resorption and reducing renal calcium excretion. It also stimulates [1-14C]-2-deoxy-D-glucose (2DG) transport and glycogen synthesis in osteoblastic cells.

Gene References Into Functions

Selected Research Highlights on Parathyroid Hormone (PTH):

  1. In individuals with normal renal function, PTH levels showed a potential association with metabolic syndrome in women, while vitamin D levels were possibly linked to metabolic syndrome in men. PMID: 29348466
  2. In pregnant women with adequate dietary calcium intake, serum 25-hydroxyvitamin D levels are crucial for maintaining normal serum PTH levels (study conducted on Caucasian women in Ireland). PMID: 30018262
  3. Fetuin A, vitamin D, and intact PTH levels were associated with sarcopenia, with intact PTH specifically indicating sarcopenic left ventricular hypertrophy in elderly individuals. PMID: 28112206
  4. Dkk-1 and PTH levels in psoriatic arthritis were lower than in healthy female controls, unlike rheumatoid arthritis where they are elevated. PMID: 28634697
  5. Existing evidence on the genetic association of serum PTH levels across various pathological conditions is limited, with published studies providing weak support (review and meta-analysis). PMID: 29794776
  6. Clinical data from patients with chronic kidney disease suggest a potential inverse association between leptin and PTH, although not consistently observed across all studies. [REVIEW] PMID: 28730419
  7. Parathyroid hormone regulates bone and kidney homeostasis through GNAS and Gq-G11 heterotrimeric G proteins. (Review) PMID: 28363951
  8. Patients undergoing parathyroidectomy for primary hyperparathyroidism exhibit similar intraoperative PTH degradation kinetics regardless of renal function. PMID: 29128188
  9. PTH levels are significantly higher in untreated sustained hypertension patients compared to those with white-coat hypertension or normotensive individuals. PMID: 29176783
  10. PTH treatment (continuous or pulsatile) did not show a positive effect on healing callus formation, suggesting a lack of benefit in the early stages of fracture healing. PMID: 24039091
  11. Physiological concentrations of PTH reduced the uptake of labelled 25-hydroxyvitamin D3 in muscle fibers and myotubes. PMID: 28104493
  12. Baseline PTH level was not associated with changes in frailty status in men. PMID: 28609827
  13. Elevated PTH induces the transition of endothelial cells to chondrogenic cells via endothelial-mesenchymal transition, potentially mediated by beta-catenin nuclear translocation. PMID: 27582099
  14. A cysteine mutation at the 25th residue of hPTH(1-34) may lead to a high bone mass phenotype. PMID: 27300576
  15. Common genetic variants near genes involved in vitamin D metabolism and calcium/renal phosphate transport were associated with variations in circulating PTH concentrations. PMID: 27927781
  16. PTH pretreatment inhibited TGF-beta1 and high glucose-induced Smad2/3 phosphorylation and subsequent upregulation of fibronectin and type IV collagen. PMID: 27530924
  17. FGFR1c and PTHR signaling pathways converge on NHERF1 to inhibit PTH- and FGF23-sensitive phosphate transport and downregulate NPT2A. PMID: 27432882
  18. The rs6254GA polymorphism independently affects the development and severity of bone mineral density complications in patients with asymptomatic hyperparathyroidism. PMID: 27756092
  19. In patients on dual antiplatelet therapy, higher PTH levels correlate with increased ADP-mediated platelet reactivity and suboptimal response to clopidogrel. PMID: 27086085
  20. Parathyroid hormone gene rs6256 variants are not associated with colorectal cancer susceptibility. PMID: 25124570
  21. PTH levels were significantly higher in patients with aldosterone-producing adenomas. PMID: 26304960
  22. No significant association was found between endogenous PTH and knee osteoarthritis, although a trend towards negative association was observed in women. PMID: 25879737
  23. This study investigated the prevalence of adenomatous colon polyps in subjects with diabetes mellitus and chronic kidney disease. PMID: 27993873
  24. PTH, along with other heart failure markers, may provide valuable diagnostic and staging information for heart failure syndromes. PMID: 27546695
  25. Patients experiencing a significant decrease in serum PTH after one year of hemodialysis are at increased risk of short-term cardiovascular death. PMID: 26880460
  26. This study investigated the functions of sclerostin using a human dermal fibroblast cell model. PMID: 26851122
  27. Recombinant human klotho inhibits PTH-mediated 1-OH expression in vitro and in vivo. PMID: 26287968
  28. NO-mediated vasorelaxation partially contributes to PTH's anabolic effects on cortical bone. PMID: 26834008
  29. PTH levels were not independently associated with hypertension risk overall, but some evidence suggests a potential association in Black individuals. PMID: 26867053
  30. This article examines the impact of renin-angiotensin system inhibitors on intact PTH levels in continuous ambulatory peritoneal dialysis patients. PMID: 25271253
  31. PTH does not influence the metabolic components of obesity, but a positive correlation exists between 25(OH)D and HDL-C. PMID: 26451492
  32. PTH's effects on bone remodeling involve not only osteoblasts/osteocytes but also bone marrow T-lymphocytes, which regulate stromal cell differentiation/lifespan and responsiveness to PTH. [REVIEW] PMID: 26662934
  33. Serum secreted iPTH levels cannot be reliably predicted by total parathyroid gland mass or blood supply. PMID: 26997379
  34. A homozygous [Cys25]PTH(1-84) mutation impairing PTH/PTHrP receptor activation defines a novel form of hypoparathyroidism. PMID: 25891861
  35. Specific PTH gene promoter region variants are associated with altered 25(OH)D levels and vitamin D deficiency in certain populations. PMID: 26339419
  36. Elevated PTH, unlike vitamin D, is independently associated with COPD severity and may serve as a superior biomarker for COPD. PMID: 26398210
  37. Abnormal diurnal PTH patterns are linked to sustained mild hypercalcemia in non-dialyzed chronic kidney disease patients. PMID: 27012036
  38. EphrinB2/EphB4 signaling within the osteoblast lineage is essential for late-stage osteoblast differentiation. PMID: 23165727
  39. Serum 25(OH)D levels were inversely associated with serum PTH and bone mineral density. PMID: 23045165
  40. Elevated serum PTH concentrations may contribute to colorectal cancer development, indicated by higher rates of adenomas (particularly with dysplasia) in women. PMID: 26021763
  41. iPTH levels were higher in ESRD patients with the VDR BsmI Bb genotype compared to those with the bb genotype, both overall and in Caucasians. PMID: 25007156
  42. The presence of PTH and other hormonal resistances suggests the need to investigate underlying genetic disorders contributing to metabolic disorders. {REVIEW} PMID: 25913526
  43. Intraoperative PTH monitoring during parathyroidectomy is a useful marker for assessing the adequacy and safety of surgery and treatment success. PMID: 25241609
  44. Serum PTH levels are independently associated with bone mineral density (BMD) in both vitamin D-sufficient and -insufficient women, but not in men. High serum PTH is detrimental to BMD in postmenopausal women. PMID: 25242259
  45. Higher serum PTH concentrations were associated with increased left ventricular mass and a higher risk of incident heart failure in a diverse population without prevalent cardiovascular disease. PMID: 25468653
  46. In elderly individuals, calcium was independently associated with increased arterial stiffness, while PTH was independently associated with intra-arterial peripheral and calculated central blood pressures. PMID: 25562577
  47. iPTH measurement may be valuable for assessing mineral bone disorder in chronic kidney disease patients. PMID: 26299086
  48. This study details the in vitro characterization of amyloid fibrils generated from human parathyroid hormone (hPTH(1-84)). PMID: 25554227
  49. A salt bridge between Arg-20 on PTH and Asp-137 on the PTH1 receptor is crucial for full receptor affinity. PMID: 25218037
  50. A combination of readily accessible determinants can explain a significant portion of PTH variance and the occurrence of secondary hyperparathyroidism. PMID: 24202062
Database Links

HGNC: 9606

OMIM: 146200

KEGG: hsa:5741

STRING: 9606.ENSP00000282091

UniGene: Hs.37045

Involvement In Disease
Hypoparathyroidism, familial isolated (FIH)
Protein Families
Parathyroid hormone family
Subcellular Location
Secreted.

Q&A

What are the primary functional characteristics of recombinant PTH in experimental systems?

Recombinant PTH demonstrates several key biological activities that can be measured in experimental systems:

  • Calcium homeostasis regulation: PTH elevates calcium levels through two primary mechanisms - dissolving bone mineral salts and preventing renal calcium excretion .

  • Cellular signaling induction: It triggers cAMP accumulation in osteoblast cell lines, with an ED50 (effective dose for 50% response) of ≤50 ng/ml in MC3T3/E1 mouse pre-osteoblast cells .

  • Metabolic activity: PTH stimulates [1-14C]-2-deoxy-D-glucose (2DG) transport and glycogen synthesis in osteoblastic cells .

When designing functional assays, researchers should include appropriate positive and negative controls and utilize cell types that express PTH receptors to accurately assess biological activity.

How should researchers validate the quality and activity of recombinant PTH preparations before experimental use?

Validation of recombinant PTH should follow a multi-parameter approach:

  • Purity assessment: SDS-PAGE analysis under reducing conditions (using a 15% gel) to confirm molecular weight and purity (should exceed 95%) .

  • Endotoxin testing: Ensure levels below 1 EU/μg to prevent non-specific inflammatory responses in biological assays .

  • Functional validation: Measure cAMP accumulation in responsive cell lines such as MC3T3/E1 mouse pre-osteoblastic cells. A properly active preparation should demonstrate dose-dependent induction with an ED50 ≤50 ng/ml .

  • Mass spectrometry: Confirm protein sequence integrity and identify any potential post-translational modifications or degradation products .

This comprehensive validation approach ensures experimental reproducibility and prevents attribution of non-specific effects to PTH activity.

What are the critical considerations for designing in vitro experiments with recombinant PTH?

When designing in vitro experiments with recombinant PTH, researchers should address several methodological factors:

  • Receptor expression: Select cell lines with verified expression of PTH receptors. MC3T3/E1 pre-osteoblasts are well-characterized models that respond to PTH stimulation via cAMP signaling .

  • Dose-response relationships: Establish complete dose-response curves starting from concentrations below 10 ng/ml to above 100 ng/ml, as the ED50 for cAMP accumulation is typically ≤50 ng/ml .

  • Timing considerations: PTH exhibits both anabolic and catabolic effects depending on exposure duration - intermittent exposure typically promotes bone formation while continuous exposure favors resorption.

  • Buffer composition: Calcium and phosphate concentrations in experimental media significantly influence PTH activity and should be controlled and reported precisely.

  • Storage and handling: PTH activity decreases with repeated freeze-thaw cycles and prolonged storage at inappropriate temperatures. Aliquoting and storage at -80°C is recommended for maintaining activity.

How do researchers effectively measure PTH-induced signaling cascades in experimental systems?

Measuring PTH-induced signaling requires specific methodological approaches targeting multiple pathway components:

  • cAMP pathway measurement: As the primary signaling mechanism, cAMP accumulation should be quantified using ELISA-based detection systems or live-cell FRET-based sensors for temporal dynamics .

  • PKA activity assessment: Phosphorylation of PKA substrates (including CREB) can be measured via phospho-specific antibodies in western blotting or immunofluorescence.

  • Calcium mobilization: Since PTH alters calcium homeostasis, intracellular calcium flux measurements using fluorescent indicators (Fura-2, Fluo-4) provide insights into immediate signaling events.

  • Gene expression analysis: Quantitative PCR for PTH-responsive genes (RANKL, OPG, SOST) should be performed at multiple time points (1-48 hours) to capture both immediate and delayed transcriptional responses.

  • Protein phosphorylation patterns: Phospho-proteomics can reveal broader signaling networks activated by PTH beyond the canonical cAMP pathway.

This multi-parameter approach provides a comprehensive view of PTH signaling rather than relying on single readouts that may miss important biological effects.

What methodological approaches should be used to assess PTH effects on calcium homeostasis in experimental models?

To properly assess PTH effects on calcium homeostasis, researchers should employ a systematic approach:

  • Serum/media calcium measurement: Total and ionized calcium concentrations should be measured using calcium-selective electrodes or colorimetric assays calibrated with appropriate standards .

  • Phosphate determination: Since PTH regulates calcium-phosphate balance, concurrent measurement of phosphate levels is essential using molybdate-based colorimetric assays .

  • Urinary calcium excretion: In animal models, 24-hour urine collection with calcium quantification reveals PTH effects on renal calcium handling .

  • Bone mineral density assessment: micro-CT analysis provides quantitative data on bone mineral content and microarchitecture in response to PTH treatment.

  • Histomorphometry: Bone histology with dynamic labeling (calcein, tetracycline) allows assessment of bone formation and resorption parameters.

  • Marker proteins: Measure serum levels of formation markers (P1NP, osteocalcin) and resorption markers (CTX, TRACP5b) to assess bone turnover dynamics.

When designing these experiments, researchers should carefully control for factors that independently affect calcium metabolism, including vitamin D status, dietary calcium intake, and circadian variations in calcium levels .

What are the methodological approaches for studying PTH-related paradoxical effects on bone metabolism?

PTH demonstrates complex, seemingly contradictory effects on bone metabolism that require specialized experimental approaches to investigate:

  • Intermittent versus continuous administration: Implement precisely controlled administration protocols using programmable infusion pumps in animal models or timed media changes in cell culture. Intermittent PTH (daily injections or pulsed exposure) typically promotes bone formation, while continuous exposure favors bone resorption .

  • Temporal signaling analysis: Utilize live-cell imaging with pathway-specific fluorescent reporters to track signaling dynamics over time (minutes to hours) following PTH exposure.

  • Cell-specific responses: Implement co-culture systems of osteoblasts, osteocytes, and osteoclasts to map the intercellular communication networks mediating differential responses to PTH.

  • Downstream mediator blocking: Systematically inhibit specific pathways (PKA, PKC, β-arrestin) while monitoring bone formation/resorption markers to identify the molecular switches controlling anabolic versus catabolic effects.

  • Transcriptomic temporal profiling: Perform RNA-seq analysis at multiple timepoints following PTH administration to identify gene expression patterns that differentiate anabolic from catabolic responses.

These approaches collectively address the mechanistic basis of the paradoxical effects rather than simply documenting their existence.

How can researchers differentiate between PTH and PTHrP biological activities in experimental systems?

Differentiating between PTH and PTHrP (PTH-related protein) activities requires methodological precision:

  • Receptor selectivity assays: Utilize cells expressing only PTHR1 versus those with both PTHR1 and PTHR2 to distinguish binding preferences. While both hormones activate PTHR1, they may produce different conformational changes and downstream signaling patterns.

  • Temporal signaling differences: Employ real-time cAMP biosensors to map the kinetics of receptor activation and signal duration, as PTH typically produces more sustained cAMP responses than PTHrP.

  • Domain-specific mutants: Use recombinant proteins with strategic mutations in N-terminal (activation) and C-terminal (binding) domains to map structure-function relationships specific to each hormone.

  • Tissue-specific responses: Compare responses in cells from different tissues (kidney, bone, cartilage) that may have different complements of signaling adaptors downstream of PTHR1.

  • Competitive binding assays: Perform radioligand displacement studies with labeled PTH and unlabeled PTHrP (and vice versa) to quantify binding affinities and kinetics.

This comprehensive approach reveals the mechanistic differences between these related but distinct hormones.

What methodological considerations are important when using recombinant PTH as a research tool for studying hypoparathyroidism?

When using recombinant PTH to study hypoparathyroidism, researchers should implement the following methodological approaches:

This approach ensures translational relevance of findings to clinical hypoparathyroidism management.

What are the methodological considerations for designing clinical trials of recombinant PTH for hypoparathyroidism?

Designing rigorous clinical trials for recombinant PTH requires methodological precision:

  • Patient stratification: Categorize participants based on hypoparathyroidism etiology (surgical, autoimmune, genetic), disease duration, and baseline requirement for calcium/vitamin D supplementation .

  • Endpoint selection:

    • Primary: Proportion of patients achieving stable serum calcium while reducing calcium/vitamin D supplementation by ≥50%

    • Secondary: Urinary calcium excretion, bone mineral density, quality of life measures, symptom scores using validated instruments like HPT-SD

  • Dosing protocols: Implement titration schedules starting at low doses (e.g., 25 mcg/day) with stepwise increases (to 50, 75, or 100 mcg/day) based on serum calcium response .

  • Control group design: The REPLACE study demonstrated effective trial design with a 2:1 randomization ratio (rhPTH[1-84] vs. placebo) providing 80% power to detect differences with an alpha error of 0.05 .

  • Safety monitoring: Implement systematic assessment for potential adverse events including:

    • Hypercalcemia and hypocalcemia

    • Hypercalciuria

    • Antibody development

    • Hypertension

This structured approach enhances the scientific validity and clinical relevance of trial outcomes.

How should researchers interpret conflicting data on PTH efficacy from different experimental systems?

When faced with conflicting data on PTH efficacy across different experimental systems, researchers should implement a systematic analytical approach:

  • Model system differences:

    • Species variations in PTH receptor structure and downstream signaling

    • Cell/tissue-specific receptor expression patterns

    • Differences in calcium homeostasis regulatory mechanisms between models

  • Protocol variations assessment:

    • Dosing regimens (intermittent vs. continuous)

    • Duration of treatment (acute vs. chronic)

    • Concurrent treatments (vitamin D, calcium supplements)

  • Endpoint measurement standardization:

    • Methodology for calcium measurement (total vs. ionized)

    • Timing of measurements relative to PTH administration

    • Techniques for assessing bone parameters

  • Statistical approach harmonization:

    • Power calculations to determine if negative findings are truly negative or potentially underpowered

    • Appropriate statistical tests for the data distribution patterns

    • Multiple testing corrections for high-dimensional datasets

  • Integration framework:

    • Weigh evidence based on methodological rigor rather than simply tallying positive versus negative studies

    • Consider translational proximity to human physiology

    • Evaluate reproducibility across independent research groups

This systematic approach transforms apparently conflicting data into a coherent understanding of context-dependent PTH effects.

What methodologies should researchers employ to assess long-term safety of recombinant PTH administration?

Comprehensive assessment of long-term PTH safety requires multi-faceted methodological approaches:

  • Bone safety assessment:

    • Sequential bone mineral density measurements (DXA, 6-12 month intervals)

    • Bone turnover markers (P1NP, CTX) evaluated monthly initially, then quarterly

    • Bone biopsy with histomorphometry in selected cases to assess microarchitecture

  • Calcium homeostasis monitoring:

    • Regular measurement of serum and urinary calcium to detect hypercalcemia and hypercalciuria

    • Renal function assessment (eGFR, creatinine) at least quarterly

    • Kidney ultrasound to detect nephrocalcinosis in patients with persistent hypercalciuria

  • Immunogenicity evaluation:

    • Anti-PTH antibody screening at baseline, 3 months, 6 months, and annually thereafter

    • Neutralizing antibody functional assessment if binding antibodies are detected

  • Cardiovascular monitoring:

    • Regular blood pressure measurements

    • Electrocardiogram to assess QT interval at baseline and during dose adjustments

    • Cardiovascular event tracking (standardized definitions)

  • Cancer surveillance:

    • Monitoring for osteosarcoma and other malignancies through regular clinical examination

    • Implementation of structured adverse event reporting systems

    • Long-term registry studies extending beyond clinical trial duration

This comprehensive approach enables detection of safety signals that may not emerge during shorter-term studies.

What methodological approaches can overcome stability challenges when working with recombinant PTH?

Addressing stability challenges with recombinant PTH requires specific technical strategies:

  • Storage optimization:

    • Store lyophilized PTH at -20°C to -80°C

    • Divide reconstituted protein into single-use aliquots to avoid freeze-thaw cycles

    • Use low-protein-binding tubes to prevent adsorption losses

  • Buffer formulation:

    • Include carrier proteins (0.1-0.5% BSA or HSA) to prevent adsorption to containers

    • Add antioxidants (e.g., methionine, 1-5 mM) to protect against oxidative degradation

    • Control pH tightly (range 4.5-5.5) to minimize deamidation and aggregation

  • Activity verification:

    • Implement functional testing (cAMP induction) before critical experiments

    • Develop and validate ELISAs specific for active conformation

    • Establish relative potency assays against reference standards

  • Degradation monitoring:

    • Use size-exclusion chromatography to detect aggregation

    • Employ reversed-phase HPLC to identify chemical modifications

    • Implement routine stability-indicating methods on stored samples

These methodological approaches ensure consistent biological activity across experiments and prevent false negative results due to protein degradation.

How should researchers design comparative studies between different recombinant PTH formulations?

Designing rigorous comparative studies between different recombinant PTH formulations requires methodological precision:

  • Structural characterization:

    • Amino acid sequence verification (mass spectrometry)

    • Secondary structure analysis (circular dichroism, FTIR)

    • Aggregation assessment (size-exclusion chromatography, dynamic light scattering)

  • Functional equivalence testing:

    • Receptor binding assays with identical experimental conditions

    • Dose-response curves for cAMP induction across multiple cell lines

    • Calcium mobilization in responsive cell types

  • Standardized bioassays:

    • Establish a reference standard and express potency of test formulations relative to this standard

    • Perform parallel line bioassays with statistical analysis of relative potency

    • Include multiple independent batches of each formulation

  • Pharmacokinetic assessment:

    • Compare absorption, distribution, and elimination profiles

    • Use identical sampling protocols and analytical methods

    • Apply non-compartmental and compartmental analysis approaches

  • Statistical design considerations:

    • Power analysis based on expected variability in key parameters

    • Crossover designs where appropriate to reduce inter-subject variability

    • Blinding of sample analysis to prevent bias

This comprehensive approach enables scientifically valid comparisons that account for both analytical and biological variability.

What methodological approaches best characterize the differential effects of PTH fragments versus full-length protein?

Investigating differential effects of PTH fragments versus full-length protein requires specialized methodological approaches:

  • Structure-activity relationship mapping:

    • Systematic testing of N-terminal, mid-region, and C-terminal fragments

    • Creation of chimeric constructs to identify domain-specific functions

    • Point mutations at key receptor interaction sites

  • Receptor conformation analysis:

    • FRET-based sensors to detect distinct receptor conformational changes

    • Biased signaling assessment (G-protein vs. β-arrestin pathways)

    • Receptor internalization and recycling kinetics

  • Signaling specificity determination:

    • Phosphoproteomic analysis to identify differential activation patterns

    • Temporal resolution of signaling cascades (seconds to hours)

    • Pathway inhibitor studies to establish causality in divergent effects

  • Physiological response comparison:

    • Fragment-specific effects on calcium mobilization vs. phosphate handling

    • Differential gene expression patterns in target tissues

    • Distinct effects on bone formation versus resorption

  • Pharmacokinetic/pharmacodynamic modeling:

    • Compartmental modeling of fragment distribution

    • Correlation of plasma levels with biomarker responses

    • Duration of action and offset kinetics comparison

This systematic approach reveals mechanistic insights beyond simple potency comparisons, illuminating how structural features translate to specific biological outcomes.

What methodological approaches will advance understanding of PTH receptor signaling complexity?

Future research into PTH receptor signaling should employ these innovative methodological approaches:

  • Single-cell signaling analysis:

    • FRET/BRET biosensors for real-time visualization of signaling in individual cells

    • Single-cell RNA-seq to capture heterogeneity in transcriptional responses

    • Correlation of signaling dynamics with cell fate decisions

  • Structural biology integration:

    • Cryo-EM structures of PTH-receptor complexes in different activation states

    • Molecular dynamics simulations of receptor conformational changes

    • Structure-guided design of conformation-selective antibodies as research tools

  • Biased signaling investigation:

    • Development of G-protein vs. β-arrestin pathway-specific readouts

    • High-throughput screening for pathway-selective PTH analogs

    • Knock-in models with signaling-biased receptors to determine physiological relevance

  • Spatial signaling organization:

    • Super-resolution microscopy to map signaling complex formation

    • Optogenetic approaches to activate signaling with spatial precision

    • Compartment-specific biosensors to differentiate membrane vs. endosomal signaling

  • Systems biology frameworks:

    • Mathematical modeling of integrated signaling networks

    • Sensitivity analysis to identify critical control points

    • Physiological response prediction from signaling signatures

These approaches will transform our understanding from linear pathways to dynamic, context-dependent signaling networks.

What innovative experimental approaches could overcome current limitations in PTH research?

Several innovative experimental approaches show promise for addressing current limitations in PTH research:

  • Engineered tissue models:

    • Organ-on-chip systems integrating bone, kidney, and intestinal components

    • 3D-bioprinted bone constructs with defined cellular composition

    • Microfluidic systems enabling precise temporal control of PTH exposure

  • Genome editing strategies:

    • CRISPR-Cas9 modification of endogenous PTH receptors

    • Knock-in reporter lines for real-time pathway visualization

    • Creation of humanized animal models expressing human PTH receptors

  • Advanced in vivo imaging:

    • Intravital microscopy for cellular responses to PTH in living bone

    • PET tracers for non-invasive assessment of PTH receptor occupancy

    • Label-free imaging of mineral dynamics in response to PTH

  • Computational approaches:

    • AI-driven analysis of multiparametric datasets

    • Virtual screening for novel PTH analogs with desired properties

    • Physiologically-based pharmacokinetic modeling for translation between species

  • Biomarker development:

    • Proteomic identification of PTH-responsive circulating proteins

    • Exosome analysis for tissue-specific PTH response markers

    • Metabolomic signatures of PTH activity

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.