PPID (Peptidylprolyl isomerase D), also known as cyclophilin D (CypD), is a member of the cyclophilin family of peptidyl-prolyl cis-trans isomerases (PPIases). This enzyme catalyzes the isomerization of proline peptide bonds, facilitating protein folding and conformational changes . PPID is encoded by the PPID gene (NCBI Gene ID: 5481) and is expressed in mitochondria, where it regulates the mitochondrial permeability transition pore (mPTP), a critical mediator of cell death .
PPID antibodies are recombinant monoclonal or polyclonal reagents designed to detect endogenous PPID proteins in research applications. Key features include:
Property | Details |
---|---|
Target | Human PPID (UniProt: Q08752) |
Host Species | Rabbit |
Reactivity | Human, mouse, rat |
Molecular Weight | 41 kDa (calculated); 41 kDa (observed in WB) |
Applications | Western blot (WB), flow cytometry (FC), ELISA, immunoprecipitation (IP) |
Immunogen | PPID fusion protein (Ag3409) |
Storage Conditions | -80°C (PBS-only buffer) or -20°C (glycerol-containing buffer) |
These antibodies are validated for specificity, with minimal cross-reactivity .
PPID antibodies are critical tools for studying mitochondrial biology, protein folding, and disease mechanisms. Key applications include:
Investigating PPID’s role in mPTP regulation and apoptosis .
Analyzing interactions with Sec translocon components in bacterial protein translocation .
PPID forms a heterodimer with YfgM in E. coli, stabilizing the inner membrane and preventing PPID degradation. Deletion of yfgM leads to PPID truncation (PpiD’), impairing protein translocation .
PPID-affected horses exhibit α-synuclein (α-syn) aggregates in the pituitary pars intermedia. Antibodies against PPID identified cross-species seeding of human α-syn fibrils, suggesting utility in Parkinson’s disease research .
Researchers used PPID antibodies in sandwich ELISAs to develop stall-side ACTH tests, improving early diagnosis of pituitary pars intermedia dysfunction in horses .
PPID antibodies are pivotal in advancing studies on mitochondrial dysfunction, neurodegeneration, and bacterial proteostasis. Ongoing efforts focus on:
Melanoma Inhibitory Activity (MIA) protein is a clinically valuable biomarker in malignant melanoma research. It is a 107-amino acid protein (following cleavage of a 24-amino acid secretion signal from the 131-amino acid precursor) that serves as a diagnostic indicator for metastatic melanoma stages III and IV . MIA has significant clinical utility as elevated serum levels correlate with melanoma progression and metastasis. The protein is not expressed in normal skin and melanocytes but shows progressively increased expression in melanocytic tumors, making it an important differentiating marker in melanoma progression studies . As an extracellular protein with a unique structural fold, MIA represents an important target for understanding melanoma pathophysiology and developing potential therapeutic interventions.
Human MIA protein adopts an SH3 domain-like fold in solution, comprising two perpendicular, antiparallel, three- and five-stranded β-sheets . The protein's structural characterization revealed several unique features:
It is the first extracellular protein found to have an SH3 domain-like fold
Unlike typical SH3 domains, MIA exists as a single-domain protein
Its structure contains an additional antiparallel β-sheet not found in typical SH3 domains
It is stabilized by two disulfide bonds, contributing to its extracellular stability
Its three-dimensional structure was determined using multi-dimensional NMR spectroscopy with 1139 approximate inter-residue distance constraints, 22 dihedrals, and 12 hydrogen bond constraints
The unique structural features of MIA provide important insights into its function in the extracellular environment and its interactions with other extracellular matrix components.
MIA protein specifically interacts with fibronectin and potentially other extracellular matrix (ECM) components . This interaction has profound implications for melanoma cell behavior:
MIA inhibits attachment of melanoma cells to fibronectin and laminin
It appears to mask the binding sites of integrins to these ECM components
This masking effect promotes melanoma cell invasion and metastasis in vivo
The growth-inhibitory activity observed in vitro likely reflects MIA's ability to interfere with cell attachment to culture dishes
This fibronectin-binding property provides a mechanistic explanation for MIA's role in promoting melanoma progression through altered cell-matrix interactions rather than direct growth inhibition.
Production of recombinant human MIA (rhMIA) with histidine tags requires careful consideration of expression systems and purification strategies:
Expression System Selection: Bacterial expression in E. coli produces high yields but may compromise post-translational modifications and proper disulfide bond formation. For studies requiring native folding, mammalian or insect cell expression systems are preferred despite lower yields.
Optimal Construct Design:
N-terminal His-tags are generally preferred over C-terminal tags to avoid interference with C-terminal functional regions
A protease cleavage site (TEV or thrombin) should be incorporated between the His-tag and MIA sequence
Codon optimization for the expression host improves yield
Purification Protocol:
Initial capture using immobilized metal affinity chromatography (IMAC) with Ni-NTA resin
Tag removal using the appropriate protease if native protein is required
Secondary purification using size exclusion chromatography to ensure monodispersity
Final quality control using circular dichroism to confirm proper folding
Activity Verification: The biological activity of purified rhMIA should be verified using Boyden Chamber assays, which can confirm inhibition of melanoma cell invasion (approximately 45.6% ± 3.1 inhibition for properly folded rhMIA) .
Analysis of MIA-fibronectin interactions requires sophisticated biophysical and biochemical approaches:
Peptide Phage Display: This technique successfully identified MIA-binding peptides with sequences matching type III human fibronectin repeats, particularly FN14 . Key findings from phage display studies revealed:
Surface Plasmon Resonance (SPR):
Immobilize either fibronectin or MIA on a sensor chip
Measure binding kinetics (kon and koff rates)
Determine equilibrium dissociation constants (KD)
Perform competition assays with identified peptides to map binding epitopes
NMR Titration Studies:
Use 15N-labeled MIA and observe chemical shift perturbations upon fibronectin addition
Map the binding interface on the 3D structure of MIA
Identify critical residues for interaction
Mutagenesis Approaches:
Create point mutations in putative binding regions
Assess impact on binding affinity and biological activity
Develop structure-function relationships
Investigating MIA's contribution to melanoma metastasis requires integrated in vitro and in vivo methodologies:
Cell-Based Metastasis Assays:
Invasion Assays: Boyden chamber/transwell assays with ECM-coated membranes can quantify MIA's effect on invasive capacity (45.6% ± 3.1 inhibition reported with recombinant MIA)
Adhesion Assays: Measure cell attachment to fibronectin with and without MIA treatment
3D Spheroid Invasion Models: More physiologically relevant than 2D assays for studying invasion
Molecular Intervention Approaches:
CRISPR/Cas9-mediated MIA knockout in melanoma cell lines
Inducible expression systems to control MIA levels
Introduction of domain-specific MIA mutants to dissect functional regions
In Vivo Metastasis Models:
Tail vein injection models to assess lung colonization
Spontaneous metastasis models using orthotopic implantation
Intracardiac injection to study broad metastatic distribution
Quantification of circulating tumor cells correlated with MIA serum levels
Clinical Correlation Studies:
Analysis of MIA expression in primary tumors and matched metastases
Correlation of serum MIA levels with clinical outcomes
Integration with other biomarkers for improved prognostic value
Optimizing MIA as a clinical biomarker requires standardized approaches:
Standardization of MIA Detection:
ELISA remains the gold standard for serum MIA quantification
Reference ranges should be established for different patient populations
Quality control samples should be included in every assay run
Clinical Cutoff Determination:
Clinical Stage | Typical MIA Range (ng/mL) | Sensitivity (%) | Specificity (%) |
---|---|---|---|
Stage I/II | 4.5-9.0 | 55-63 | 89-93 |
Stage III | 9.0-15.0 | 71-78 | 89-93 |
Stage IV | >15.0 | 83-91 | 89-93 |
Integration with Other Biomarkers:
Combined use with S100B improves sensitivity and specificity
LDH levels provide complementary prognostic information
Multimarker panels should be developed for improved accuracy
Monitoring Protocol Development:
Baseline measurement before surgical intervention
Regular monitoring every 3-6 months to detect early recurrence
Investigation of rising MIA levels even in the absence of clinical symptoms
Understanding structure-function relationships requires multiple structural biology techniques:
Advanced NMR Studies:
X-ray Crystallography:
Co-crystallization with fibronectin fragments
Heavy atom derivatives for phase determination
Molecular replacement using the NMR structure as a starting model
Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS):
Maps solvent accessibility and conformational dynamics
Identifies regions protected upon ligand binding
Monitors structural changes under different conditions
Integrative Structural Biology:
Combining multiple techniques (NMR, X-ray, cryo-EM, SAXS)
Computational modeling to predict binding interfaces
Molecular dynamics simulations to explore conformational flexibility
AI and computational methods offer significant advantages for MIA research:
Structure Prediction and Refinement:
AlphaFold2 and RoseTTAFold for predicting MIA variants and complexes
MD refinement of predicted structures in explicit solvent
Ensemble refinement against experimental data
Virtual Screening for MIA Inhibitors:
Structure-based screening against the MIA-fibronectin interface
Pharmacophore modeling based on known binding peptides
Quantitative structure-activity relationship (QSAR) modeling
Machine Learning for Biomarker Optimization:
Pattern recognition in clinical MIA data to identify patient subgroups
Predictive models combining MIA with other clinical parameters
Natural language processing for mining MIA-related literature
Digital Medical Interview Applications:
While primarily focused on the medical domain, research into medical interview assistants (MIA) like the digital assistant described in search result could be leveraged to collect standardized melanoma patient histories
Integration of MIA biomarker knowledge into digital health systems for improved patient monitoring
Developing MIA-directed therapeutics requires specialized methodologies:
Peptide-Based Inhibitor Design:
Small Molecule Screening Strategy:
Fragment-based screening using NMR or thermal shift assays
Focused libraries targeting SH3-like domain interactions
Structure-based design leveraging the unique MIA fold
Antibody Development Approach:
Epitope mapping to identify accessible regions on MIA
Humanization of promising murine antibodies
Antibody-drug conjugates to deliver cytotoxic agents to MIA-expressing cells
RNA Therapeutics Pipeline:
siRNA design targeting MIA mRNA
Lipid nanoparticle formulation for delivery to melanoma cells
Antisense oligonucleotides to modulate MIA splicing or expression
While MIA was initially identified in melanoma, potential expanded research applications include:
Other Malignancies:
Expression in chondrosarcomas and other tumors of cartilaginous origin
Investigation in additional neural crest-derived tumors
Potential as a biomarker in tumors with similar invasion mechanisms
Cartilage Disorders:
Role in normal cartilage development and homeostasis
Contribution to osteoarthritis pathophysiology
Potential therapeutic target in cartilage regeneration
Developmental Biology:
Function in neural crest cell migration during embryogenesis
Impact on cell-matrix interactions during tissue formation
Potential role in wound healing and tissue remodeling
Autoimmune and Inflammatory Conditions:
Investigation of MIA's potential immunomodulatory functions
Role in tissue remodeling during chronic inflammation
Interaction with immune cell extracellular matrix receptors
Emerging technologies offer new opportunities for MIA research:
Single-Cell Approaches:
Single-cell RNA-seq to identify MIA-responsive cell populations
Spatial transcriptomics to map MIA expression in tissue context
CyTOF to correlate MIA levels with cellular phenotypes
Proteomics and Interactomics:
Proximity labeling (BioID, APEX) to identify MIA-proximal proteins
Crosslinking mass spectrometry to map interaction interfaces
Thermal proteome profiling to identify MIA-dependent protein stability changes
Advanced Imaging:
Super-resolution microscopy to visualize MIA-fibronectin interactions
Intravital imaging to track MIA-expressing cells in vivo
Correlative light and electron microscopy to link MIA localization with ultrastructure
Organoid and Microfluidic Systems:
Melanoma organoids to study MIA function in 3D context
Tumor-on-chip models incorporating extracellular matrix components
Vascularized models to study MIA's role in metastatic extravasation
Melanoma Inhibitory Activity (MIA) protein is a small, secreted protein that plays a significant role in the progression and metastasis of malignant melanoma. It is also known as melanoma-derived growth regulatory protein. The recombinant form of this protein, tagged with a His (histidine) tag, is widely used in research to study its structure, function, and potential therapeutic applications.
The human recombinant MIA protein with a His tag is typically expressed in systems such as HEK 293 cells or Escherichia coli. The His tag, usually consisting of six histidine residues, is added to the N-terminus of the protein to facilitate purification using metal affinity chromatography. The recombinant protein is often produced with high purity, exceeding 95%, and has a molecular weight of approximately 14.23 kDa .
MIA protein is involved in the regulation of cell adhesion and migration, which are critical processes in cancer metastasis. It interacts with extracellular matrix components such as fibronectin, disrupting cell-matrix interactions and promoting the detachment of melanoma cells from the primary tumor site . This detachment is a key step in the metastatic spread of melanoma cells to distant organs.
MIA is a valuable biomarker for malignant melanoma. Elevated levels of MIA in the blood are associated with advanced stages of melanoma, particularly metastatic stages III and IV . Monitoring MIA levels can aid in the diagnosis and prognosis of melanoma, as well as in assessing the response to therapy.
The recombinant human MIA protein with a His tag is used in various research applications, including: