PHB Human

Prohibitin Human Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Genetic Basis

PHB, or Prohibitin, is a 32 kDa protein localized to the inner mitochondrial membrane. The human PHB gene resides on chromosome 17q21, a region linked to tumor suppression and cell-cycle regulation . Two transcript variants exist, differing in their 3' untranslated regions, with the longer variant enriched in proliferating tissues .

Key Genetic Features

FeatureDetail
Gene location17q21 (BRCA1-associated region)
Protein classType-I prohibitin (ortholog of yeast PHB1)
Transcript variantsTwo isoforms with distinct 3' UTRs

Mitochondrial Function

PHB forms a ring-like complex (16–20 subunits of PHB1 and PHB2) in the mitochondrial inner membrane, acting as a chaperone for respiratory chain proteins and maintaining cristae morphology . Depletion disrupts mitochondrial DNA stability and oxidative phosphorylation .

Oxidative Stress Response

PHB overexpression in intestinal epithelial cells enhances Nrf2 nuclear accumulation, boosting antioxidant defenses and mitigating colitis in mice .

Cancer Biology

  • Dual Role: While initially mischaracterized as a tumor suppressor, PHB exhibits context-dependent roles:

    • Anti-proliferative: Reduces androgen receptor activity in prostate cancer .

    • Pro-proliferative: Sustains mitochondrial function in rapidly dividing cells .

Viral Pathogenesis

PHB1 serves as a receptor for Chikungunya (CHIKV) and Dengue (DENV-2) viruses, though its role in infection mechanisms remains poorly understood .

Inflammatory Diseases

PHB-deficient mice show exacerbated oxidative stress and colitis, linking mitochondrial PHB complexes to inflammatory resolution .

Disease Associations

ConditionPHB RoleSource
Sporadic breast cancerMutations linked to tumorigenesis
ColitisPHB overexpression reduces oxidative stress
NeurodegenerationPHB-domain proteins modulate ion channels

Therapeutic Targeting

  • PHB Modulation: Small molecules targeting PHB-domain proteins (e.g., Podocin, MEC-2) show promise in treating neurodegenerative disorders and hypertension .

  • Mitochondrial Stress Response: PHB depletion activates the mitochondrial unfolded protein response (UPRmt), a pathway implicated in aging and metabolic diseases .

Diagnostic Potential

PHB expression levels correlate with:

  • Chemoresistance in cancer .

  • Severity of inflammatory bowel disease .

Key Studies (2020–2025)

Study FocusFindingsSource
PHB and UPRmtPHB loss induces non-canonical UPRmt, extending lifespan in metabolically stressed C. elegans
PHB in Viral EntryPHB1 identified as a DENV-2 receptor, enabling viral internalization
PHB Transgenic ModelsIntestinal PHB overexpression reduces colitis via Nrf2 activation

Challenges and Future Directions

  • Functional Complexity: PHB’s dual roles in proliferation and stress response necessitate cell-type-specific studies.

  • Therapeutic Development: Targeting PHB requires precision to avoid disrupting mitochondrial homeostasis.

Product Specs

Introduction
Prohibitin (PHB) is an important gene found in various organisms that plays a role in preventing cell growth. Changes in this gene have been observed in breast cancer cases. PHB is believed to act as a tumor suppressor by regulating cell proliferation. There are two forms of PHB transcripts, one longer than the other, with the longer one being more abundant in actively dividing cells. This suggests that the longer transcript may function as a regulatory RNA molecule.
Description
Recombinant human PHB protein, produced in E. coli, is a single chain polypeptide. It consists of 292 amino acids (specifically amino acids 1 to 272), resulting in a molecular weight of 31.9kDa. The protein includes a 20 amino acid His-tag attached to its N-terminus and is purified using specialized chromatographic methods.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The PHB protein solution is provided at a concentration of 1mg/ml. It is dissolved in a buffer containing 20mM Tris-HCl (pH 8.0), 10% glycerol, and 0.1M NaCl.
Stability
For short-term storage (up to 4 weeks), keep refrigerated at 4°C. For longer periods, store frozen at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for extended storage. Avoid repeated freezing and thawing.
Purity
The purity of the protein is greater than 85%, as determined by SDS-PAGE analysis.
Synonyms
PHB1, Prohibitin.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAAKVFESIG KFGLALAVAG GVVNSALYNV DAGHRAVIFD RFRGVQDIVV GEGTHFLIPW VQKPIIFDCR SRPRNVPVIT GSKDLQNVNI TLRILFRPVA SQLPRIFTSI GEDYDERVLP SITTEILKSV VARFDAGELI TQRELVSRQV SDDLTERAAT FGLILDDVSL THLTFGKEFT EAVEAKQVAQ QEAERARFVV EKAEQQKKAA IISAEGDSKA AELIANSLAT AGDGLIELRK LEAAEDIAYQ LSRSRNITYL PAGQSVLLQL PQ.

Q&A

What is PHB and what is its significance in human microbiome research?

Polyhydroxybutyrate (PHB) is a biodegradable biopolymer produced by various microorganisms as a carbon and energy storage compound under nutrient-limited conditions. In human research, PHB has gained significant attention as a "ketobiotic" - a novel prebiotic that specifically activates butyrate-producing bacteria in the gut microbiota . Unlike conventional prebiotics, PHB functions through a unique mechanism wherein mammalian digestive enzymes cannot hydrolyze it, allowing it to pass through the small intestine and reach the large intestine intact. There, gut microbiota can depolymerize PHB to produce 3-Hydroxybutyrate (3HB), which serves as an energy substrate for butyrate-producing bacteria, while also inducing slight acidification of the gut environment . This mechanism represents a promising approach for targeted microbiome modulation in human health research.

How does PHB differ mechanistically from traditional prebiotics in human gut studies?

Unlike traditional carbohydrate-based prebiotics (such as inulin or fructooligosaccharides), PHB represents a novel class of "ketobiotics" that operate through donation of 3-hydroxybutyrate (3HB) to the microbiota . This mechanism creates several distinct research advantages: (1) PHB specifically targets microorganisms capable of depolymerizing the compound; (2) The released 3HB selectively promotes butyrate-producing bacteria; (3) The process induces slight acidification of the gut environment, which may inhibit pathogenic bacteria while favoring beneficial species . This targeted approach allows researchers to study specific microbiome pathways rather than broad fermentation effects, offering more precise experimental control over microbial community modulation.

What are the primary bacterial strains used to produce PHB for human research?

Several bacterial strains are commonly employed in PHB production for research applications, each offering distinct advantages. The most extensively studied include Ralstonia eutropha (also known as Cupriavidus necator), Alcaligenes spp., Azotobacter spp., Bacillus spp., Nocardia spp., Pseudomonas spp., and Rhizobium spp . Ralstonia eutropha remains the most thoroughly characterized strain for PHB production, having been commercially utilized by Imperial Chemical Industries (ICI) to produce PHB polymers under the trade name Biopol . The choice of bacterial strain significantly influences the resulting PHB's molecular weight, crystallinity, and polydispersity, which in turn affects its degradation kinetics in the human gut and its resulting prebiotic efficacy.

What fermentation processes yield optimal PHB for human microbiome studies?

Several fermentation methods can be employed for producing PHB, each with distinct implications for research applications. Discontinuous processes include batch culture, fed-batch culture, and repeated fed-batch culture, while continuous processes encompass continuous fed-batch systems using gaseous substrates, one-stage chemostat process, two-stage chemostat process, and multi-stage chemostat processes in continuously stirred tank reactor (CSTR)-bioreactor cascades . For human microbiome research, two-stage continuous fermentation processes are often preferred as they allow for precise control of PHB accumulation. This approach first generates sufficient active biomass under nutritionally balanced conditions in the initial stage, followed by nutrient limitation in the second stage to trigger PHB biosynthesis . This method achieves higher productivity and more consistent PHB composition compared to batch processes, which is critical for experimental reproducibility in human studies.

How should researchers evaluate the molecular characteristics of PHB for human microbiome applications?

Comprehensive characterization of PHB is essential before application in human microbiome research. Key parameters to assess include:

  • Molecular weight and polydispersity index: Higher molecular weight PHB (>1 × 10^6 Da) typically exhibits slower degradation in the gut, providing sustained prebiotic effects .

  • Crystallinity and thermal properties: PHB with lower crystallinity (e.g., 9.96% in functionalized PHBV) demonstrates improved biodegradability (up to 1.6-fold increase) .

  • Purity assessment: Residual endotoxins, cell debris, or extraction solvents must be quantified to ensure safety for human studies.

  • Thermal degradation temperature: Higher thermal degradation temperatures (e.g., 294.97°C for functionalized PHBV) indicate stability during processing and storage .

  • Hydrophilicity/contact angle: This property (e.g., 68° for functionalized PHBV) influences bacterial adhesion and enzyme accessibility during microbial degradation .

These properties significantly impact PHB's behavior in the human gut environment and should be systematically documented to ensure experimental reproducibility.

What downstream processing methods yield PHB suitable for human research applications?

The extraction and purification of PHB significantly impacts its properties and suitability for human research. After fermentation and cell harvesting (typically via centrifugation), several extraction methods can be employed, each affecting the final material differently. Solvent extraction methods using chloroform, ethylene carbonate, DMSO, or 1,2-propylene carbonate can achieve high purity (>95%) but may leave residual solvents of concern for human studies . Temperature control during extraction is critical, as elevated temperatures can induce polymer degradation and reduce molecular weight . Studies have shown that butyl-acetate extraction at 130°C for 30 minutes yields PHB with molecular weight of 1.4 × 10^6, while extraction at 90°C for 60 minutes produces PHB with molecular weight of 1.2 × 10^6 . For human applications, methods that minimize solvent residues while maintaining high molecular weight and low polydispersity are preferred, with 1,2-propylene carbonate extraction showing favorable results in producing more homogeneous PHB with lower polydispersity values compared to chloroform extraction .

How should researchers design controlled experiments to evaluate PHB effects on human gut microbiota?

Effective experimental design for PHB-microbiome studies requires careful consideration of several methodological elements:

  • Study design selection: Crossover designs are often optimal for PHB studies as they allow subjects to serve as their own controls, reducing inter-individual variability. A minimum 2-3 week washout period is recommended between treatment phases to prevent carryover effects.

  • Control selection: Appropriate placebo controls should possess similar physical properties to PHB but lack biological activity. Cellulose derivatives with similar particle size distribution but no prebiotic effects can serve as suitable controls.

  • Sampling strategy: Serial sampling is essential to capture temporal dynamics of microbiome changes. Recommended timepoints include baseline, early response (days 1-3), mid-intervention (days 7-14), late intervention (days 21-28), and post-intervention (2-4 weeks after cessation).

  • Environmental controls: Standardized or carefully documented dietary intake during study periods is crucial to minimize confounding effects of dietary variation on microbiome composition.

  • Multi-parameter assessment: Beyond microbiome composition, studies should measure fecal pH, short-chain fatty acid profiles, inflammatory markers, and intestinal permeability to comprehensively evaluate PHB effects.

This methodology enables researchers to distinguish direct PHB effects from secondary adaptations or confounding factors.

What analytical techniques are most appropriate for measuring PHB degradation in the human gut?

Measuring PHB degradation in the human gut represents a significant methodological challenge requiring specialized approaches:

  • Direct measurement techniques:

    • Isotope-labeled PHB (13C) to trace degradation products through metabolomic analysis

    • Quantification of residual PHB in fecal samples using FTIR or gas chromatography

    • Measurement of PHB depolymerase enzyme activity in fecal samples

  • Indirect measurement approaches:

    • Monitoring 3HB concentrations in fecal water and serum

    • Tracking changes in abundance and activity of bacteria possessing PHB depolymerase genes

    • Quantifying downstream metabolites, particularly butyrate and other short-chain fatty acids

  • Molecular techniques:

    • Metagenomic analysis to identify and quantify genes involved in PHB metabolism

    • Metatranscriptomic assessment of PHB depolymerase gene expression

    • Proteomics to detect and quantify PHB-degrading enzymes in the microbiome

The combination of these approaches provides a comprehensive assessment of PHB metabolism throughout the gastrointestinal tract.

How can researchers account for interindividual variability in PHB response studies?

Interindividual variability presents a significant challenge in PHB-microbiome research. Methodological approaches to address this include:

  • Stratification strategies:

    • Pre-screening participants based on baseline microbiome composition, particularly the presence of known PHB-degrading bacteria

    • Categorizing subjects by enterotype or by abundance of butyrate-producing bacteria

    • Grouping by gut transit time, which significantly impacts PHB exposure duration

  • Statistical approaches:

    • Mixed-effects models that account for repeated measures and random subject effects

    • Bayesian hierarchical modeling to handle nested data structures

    • Subject-specific trajectory analysis rather than simple group comparisons

  • Experimental design considerations:

    • Larger sample sizes to accommodate heterogeneous responses

    • Crossover designs where feasible to use subjects as their own controls

    • Run-in periods to stabilize the microbiome before intervention

  • Personalized analysis frameworks:

    • Subject-specific responder/non-responder classification based on defined metrics

    • Integration of host factors (genetics, diet, lifestyle) with microbiome data

    • Pathway-specific analyses that can detect consistent biological responses despite taxonomic variability

How does PHB function as a "ketobiotic" in the human gut microbiome?

The novel "ketobiotic" mechanism of PHB involves several distinct steps that differentiate it from traditional prebiotics :

  • Transit resistance: Unlike many prebiotics that may be partially degraded in the upper gastrointestinal tract, mammalian digestive enzymes cannot hydrolyze PHB, ensuring its intact delivery to the large intestine .

  • Selective microbial depolymerization: Upon reaching the colon, specific gut bacteria possessing PHB depolymerase enzymes break down the polymer into its monomeric form, 3-hydroxybutyrate (3HB) .

  • Ketone body donation: The released 3HB serves as a specialized energy substrate, preferentially utilized by butyrate-producing bacteria that possess the metabolic pathways to convert 3HB to acetyl-CoA and subsequently to butyrate .

  • Environmental modification: This process creates a slightly acidified local environment that further selects for beneficial bacteria while inhibiting pathogenic species sensitive to lower pH .

  • Metabolic cross-feeding: The resulting microbial community shifts establish new metabolic networks where primary 3HB utilizers produce intermediates that support secondary butyrate producers.

This mechanism represents a more targeted approach to microbiome modulation compared to traditional prebiotics that typically feed a broader spectrum of bacteria.

What biological pathways mediate the interaction between PHB and human gut epithelial cells?

The interaction between PHB-derived metabolites and gut epithelial cells involves several interconnected pathways:

  • Butyrate-mediated effects: Increased butyrate production following PHB metabolism activates:

    • GPR41/GPR43 signaling, stimulating peptide YY release and affecting gut motility

    • HDAC inhibition, modulating gene expression in colonocytes and immune cells

    • AMPK pathway activation, influencing cellular energy metabolism and barrier function

  • Direct 3HB effects: 3HB released during PHB depolymerization may:

    • Serve as an alternative energy source for colonocytes during metabolic stress

    • Activate β-hydroxybutyrate receptor (BDH1) signaling

    • Modulate inflammatory pathways independently of butyrate

  • pH-dependent mechanisms: The slight acidification induced by PHB metabolism:

    • Alters ion exchange across the epithelial membrane

    • Modifies the activity of pH-sensitive enzymes in the intestinal lumen

    • Changes the solubility and bioavailability of various metabolites

These pathways collectively contribute to improved epithelial barrier function, reduced inflammation, and enhanced mucosal immunity observed in PHB studies.

What are the immunological implications of PHB supplementation in human gut research?

PHB supplementation has several notable immunological effects relevant to human research:

  • Regulatory T-cell induction: Butyrate produced through PHB metabolism promotes FOXP3+ regulatory T-cell differentiation via HDAC inhibition, potentially beneficial in inflammatory conditions.

  • Dendritic cell modulation: Changes in microbiome composition following PHB supplementation alter the microbial-associated molecular patterns (MAMPs) presented to dendritic cells, shifting their phenotype toward tolerance rather than activation.

  • Neutrophil recruitment attenuation: PHB-induced changes in epithelial chemokine production reduce neutrophil infiltration during inflammatory challenges.

  • Macrophage polarization: Increased butyrate shifts intestinal macrophages toward an M2 anti-inflammatory phenotype, characterized by IL-10 production rather than pro-inflammatory cytokines.

  • Microbiota-immune feedback loops: PHB establishes new equilibria between the microbiome and immune system through:

    • Modified pattern recognition receptor activation

    • Altered metabolite profiles affecting immune cell metabolism

    • Changes in microbial translocation rate due to enhanced barrier function

These immunological effects provide mechanistic insights into how PHB might be applied in research on inflammatory bowel diseases and other immune-mediated conditions.

What statistical approaches are most appropriate for analyzing complex PHB-microbiome datasets?

PHB-microbiome research generates high-dimensional, compositional data requiring specialized analytical approaches:

  • Compositional data transformation: Microbiome relative abundance data should be transformed using:

    • Centered log-ratio (CLR) transformation to address the compositional nature of microbiome data

    • Aitchison distance for beta diversity analyses rather than Bray-Curtis or UniFrac distances

    • ANCOM or ANCOM-BC for differential abundance testing to control for compositionality

  • Time-series analysis methods:

    • Linear mixed-effects models incorporating random subject effects and time as a continuous variable

    • Functional data analysis for modeling smooth temporal trajectories

    • Dynamic Bayesian networks to capture temporal dependencies in microbial interaction networks

  • Multi-omics integration strategies:

    • Multi-block partial least squares discriminant analysis (MB-PLS-DA) to integrate microbiome, metabolome, and host parameter data

    • Similarity network fusion to identify patterns across multiple data types

    • Multi-table trilinear decomposition for three-way data (subjects × features × time)

  • Machine learning considerations:

    • Feature selection techniques to address high dimensionality (LASSO, elastic net, random forest importance)

    • Cross-validation strategies stratified by subject to prevent data leakage

    • Ensemble methods combining multiple algorithms to improve prediction robustness

These approaches enable more valid inference from complex PHB-microbiome studies than conventional statistical methods designed for independent, normally distributed data.

How can researchers distinguish direct PHB effects from secondary microbial adaptation responses?

Distinguishing primary from secondary effects in PHB-microbiome research requires specialized analytical approaches:

  • Temporal resolution strategies:

    • Early time-point sampling (hours rather than days) to capture initial responders

    • Time-course analysis with closely spaced sampling to establish sequence of events

    • Rate-of-change analysis to identify rapid versus gradual responses

  • Network-based methods:

    • Differential network analysis comparing interaction patterns before and after PHB exposure

    • Identification of keystone species with directional influence on community structure

    • Bayesian network inference to establish causal relationships between species

  • Metabolic modeling approaches:

    • Genome-scale metabolic models to predict direct utilizers of 3HB

    • Community-level metabolic reconstruction to identify cross-feeding relationships

    • Flux balance analysis to predict metabolic shifts following PHB introduction

  • Experimental validation techniques:

    • In vitro fermentation of defined communities with isotope-labeled PHB

    • Mono-colonization studies with candidate PHB-degrading species

    • Metatranscriptomic analysis focused on PHB depolymerase and 3HB metabolism gene expression

This multi-faceted approach allows researchers to build causal models of PHB effects rather than merely observing associational patterns.

What bioinformatic pipelines effectively detect functional shifts in the microbiome related to PHB metabolism?

Specialized bioinformatic approaches can better capture PHB-related functional shifts than standard taxonomic analysis:

  • Targeted functional profiling:

    • Custom databases of PHB depolymerase and 3HB utilization genes

    • Hidden Markov Models (HMMs) for sensitive detection of PHB-related protein families

    • Pathway enrichment analysis focused on butyrate production and 3HB metabolism

  • Reference-free approaches:

    • Assembly of metagenome-assembled genomes (MAGs) to identify novel PHB-metabolizing organisms

    • De novo assembly of transcriptomes to detect previously unannotated PHB-related genes

    • k-mer based machine learning to identify sequence patterns associated with PHB response

  • Integrated analysis pipelines:

    • Combined taxonomic-functional analysis linking specific taxa to PHB-related functions

    • Multi-omics workflows integrating metagenomics, metatranscriptomics, and metabolomics

    • Longitudinal analysis frameworks to track functional changes over time

  • Visualization and interpretation tools:

    • Metabolic pathway mapping with overlay of expression/abundance data

    • Genome-centric visualization of PHB-related gene clusters

    • Interactive dashboards for exploring multi-dimensional functional shifts

These approaches move beyond simple taxonomic profiling to provide mechanistic insights into how PHB influences microbiome functionality.

What evidence supports the potential of PHB in inflammatory bowel disease research?

Several lines of evidence suggest PHB may be valuable in inflammatory bowel disease (IBD) research:

  • Mechanistic rationale:

    • PHB promotes butyrate-producing bacteria, which are typically depleted in IBD patients

    • The slight acidification of the gut environment may inhibit pathobionts associated with IBD

    • 3HB itself may have direct anti-inflammatory properties independent of its conversion to butyrate

  • Preclinical evidence in animal models:

    • Studies in dextran sodium sulfate (DSS)-induced colitis show reduced inflammatory markers

    • Histological improvement in intestinal tissue architecture has been observed

    • Restoration of microbial diversity similar to patterns seen in IBD remission

  • Molecular pathways affected:

    • Downregulation of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6)

    • Reduced activation of NF-κB pathway

    • Enhanced intestinal barrier function through increased tight junction protein expression

  • Methodological research considerations:

    • PHB could serve as a research tool to study microbiome-mediated mechanisms in IBD

    • May provide insights into how butyrate-producing bacteria influence disease pathogenesis

    • Could help identify patient subgroups most likely to respond to microbiome-targeted therapies

While clinical translation requires further investigation, PHB offers a unique research platform for studying microbiome-epithelial-immune interactions in IBD.

How might PHB be utilized in gut-brain axis research methodologies?

PHB offers novel methodological approaches for investigating gut-brain axis interactions:

  • Mechanistic pathways for study:

    • Butyrate signaling through the vagus nerve

    • 3HB crossing the blood-brain barrier as a potential neuroactive compound

    • Immune modulation affecting neuroinflammatory processes

    • Microbial metabolite profiles influencing tryptophan metabolism and serotonin availability

  • Experimental design opportunities:

    • PHB as a tool for selective microbiome modulation without systemic antibiotic effects

    • Controlled butyrate delivery to specific gut regions to map regional effects on brain function

    • Combined microbiome-neuroimaging studies to correlate PHB-induced changes with brain activity

  • Research applications in neurological conditions:

    • Models of neurodevelopmental disorders with gut microbiome components

    • Neurodegenerative disease research where metabolic factors influence pathogenesis

    • Stress-related disorders where the gut-brain axis plays a significant role

  • Methodological advantages over traditional approaches:

    • More physiological than direct butyrate administration

    • More specific than broad-spectrum prebiotics

    • Avoids confounding effects of probiotics that may have systemic immune effects

These approaches could help delineate specific microbiome-mediated pathways in gut-brain communication.

What research models are appropriate for studying PHB effects on human metabolic health?

Several research models can effectively investigate PHB's influence on metabolic health:

  • In vitro systems:

    • Co-culture models combining intestinal epithelial cells with hepatocytes to study gut-liver axis

    • Adipocyte-microbiome co-culture systems to examine cross-talk in metabolism

    • Intestinal organoids with integrated immune components to model barrier function and inflammation

  • Animal models with translational value:

    • Humanized gnotobiotic mice harboring human microbiota from metabolic disorder patients

    • Diet-induced obesity models with controlled microbiome composition

    • Transgenic models of specific metabolic dysregulations (e.g., leptin receptor deficiency)

  • Human study designs:

    • Stratified approaches based on metabolic phenotypes (insulin-resistant vs. insulin-sensitive)

    • Crossover designs with metabolic challenge tests (oral glucose tolerance test, lipid challenge)

    • Combined microbiome sampling with metabolic tissue biopsies (adipose, muscle)

  • Multi-system assessment approaches:

    • Continuous glucose monitoring combined with microbiome sampling

    • Dual-tracer studies to track both PHB metabolism and host metabolic responses

    • Measurement of tissue-specific insulin sensitivity using hyperinsulinemic-euglycemic clamps

These models allow researchers to connect PHB-induced microbiome changes with specific metabolic pathways and functional outcomes relevant to human health.

What emerging technologies will advance PHB-microbiome research?

Several cutting-edge technologies promise to transform PHB-microbiome research:

  • Advanced imaging techniques:

    • Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry to visualize PHB degradation and metabolite distribution

    • Label-free Raman microspectroscopy for spatial tracking of PHB in the gut environment

    • Fluorescent in situ hybridization with combinatorial labeling for simultaneous visualization of multiple bacterial groups interacting with PHB

  • Single-cell and spatial technologies:

    • Single-cell RNA sequencing of microbial communities to capture heterogeneous responses to PHB

    • Spatial metagenomics to map the physical organization of PHB-utilizing bacteria

    • Microfluidic devices for studying individual bacterial responses to PHB and 3HB

  • Advanced in vitro models:

    • Human-microbiome-on-a-chip technology incorporating regional gut differences

    • 3D bioprinted intestinal tissues with controlled microbial incorporation

    • Perfusion bioreactors that simulate intestinal flow and mechanical forces

  • Real-time monitoring systems:

    • Ingestible sensors capable of measuring pH, temperature, and microbial metabolites

    • Implantable biosensors for continuous monitoring of gut metabolites

    • Wearable devices tracking systemic markers affected by gut microbiome function

These technologies will enable more precise, mechanistic studies of how PHB influences the microbiome and host physiology.

How might PHB research inform personalized microbiome modulation approaches?

PHB research could enable more personalized approaches to microbiome modulation:

  • Predictive modeling frameworks:

    • Development of microbiome signatures that predict individual responsiveness to PHB

    • Integration of host genetic, microbial, and metabolic data to guide personalized dosing

    • Machine learning algorithms to match optimal PHB formulations to individual microbiome profiles

  • Tailored PHB formulations:

    • Engineered PHB variants with customized degradation kinetics for specific microbiome compositions

    • Co-formulation strategies with complementary prebiotics based on individual deficiencies

    • Site-specific delivery technologies targeting particular gut regions based on individual physiology

  • Adaptive intervention protocols:

    • Sequential testing approaches to determine optimal PHB timing and dosage

    • Feedback systems using real-time monitoring to adjust interventions

    • Combination therapies integrating PHB with other microbiome-targeting approaches

  • Research infrastructure development:

    • Biobanking of individual microbiome samples for ex vivo testing of PHB formulations

    • Digital platforms for tracking longitudinal microbiome changes with PHB interventions

    • Standardized protocols for assessing individual variability in PHB response

These personalized approaches could significantly enhance the efficacy and reproducibility of microbiome modulation strategies in both research and clinical applications.

What methodological challenges remain in translating PHB research from experimental models to human applications?

Despite promising results, several methodological challenges must be addressed:

  • Scale-up and standardization issues:

    • Developing production methods that maintain consistent PHB properties at research scale

    • Standardizing characterization protocols to ensure comparability across studies

    • Creating reference materials for PHB research to benchmark experimental outcomes

  • Physiological relevance challenges:

    • Bridging results from in vitro and animal models to human physiology

    • Accounting for differences in gut transit time, pH profiles, and microbiome composition

    • Developing biologically relevant dosing regimens based on intestinal physiology

  • Analytical limitations:

    • Improving methods for tracking PHB fate throughout the gastrointestinal tract

    • Developing more sensitive techniques for detecting low-abundance but functionally significant microbiome changes

    • Creating standardized bioinformatic pipelines specific to PHB functional analysis

  • Experimental design considerations:

    • Determining appropriate study durations to capture both acute and chronic effects

    • Establishing consensus on core outcome measurements for PHB-microbiome research

    • Developing strategies to control for confounding variables, particularly diet and medication use

Addressing these challenges will require collaborative efforts across disciplines, including materials science, microbiology, bioinformatics, and clinical research methodology.

What constitutes best practices for PHB research in human microbiome studies?

Best practices for PHB-microbiome research include comprehensive characterization of the PHB material used (molecular weight, crystallinity, purity); transparent reporting of production and extraction methods; appropriate controls accounting for physical properties; adequate sample sizes with power calculations specific to microbiome outcomes; multi-omics approaches combining taxonomic, functional, and metabolomic analyses; and careful consideration of inter-individual variability. Researchers should utilize appropriate statistical approaches for compositional microbiome data and distinguish between correlation and causation through mechanistic validation studies. The field would benefit from standardized reporting frameworks specific to PHB-microbiome research to enhance reproducibility and facilitate meta-analyses across studies.

Product Science Overview

Introduction

Prohibitin (PHB) is a highly conserved protein that plays a crucial role in various cellular processes, including cell cycle regulation, apoptosis, and mitochondrial function. It is part of the prohibitin family, which includes two main members: prohibitin-1 (PHB1) and prohibitin-2 (PHB2). These proteins are known for their ability to form large complexes within the mitochondria, where they help maintain mitochondrial integrity and function. Recombinant human prohibitin is a form of this protein that is produced through recombinant DNA technology, allowing for its use in various research and therapeutic applications.

Preparation Methods

Recombinant human prohibitin is typically produced using bacterial or mammalian expression systems. The gene encoding prohibitin is cloned into an expression vector, which is then introduced into the host cells. These cells are cultured under conditions that promote the expression of the prohibitin protein. Once expressed, the protein is purified using techniques such as affinity chromatography, which ensures a high degree of purity and activity.

Chemical Reactions Analysis

Prohibitin is involved in several key biochemical pathways and interactions. It acts as a scaffold protein, facilitating the assembly of multi-protein complexes that are essential for mitochondrial function. Prohibitin interacts with various signaling molecules, including kinases and phosphatases, to regulate cellular processes such as apoptosis and cell proliferation. Additionally, prohibitin has been shown to play a role in the regulation of mitochondrial respiration and the maintenance of mitochondrial DNA.

Regulatory Mechanisms

The expression and activity of prohibitin are tightly regulated at multiple levels. Transcriptional regulation involves various transcription factors that bind to the promoter region of the prohibitin gene, modulating its expression in response to cellular signals. Post-translational modifications, such as phosphorylation and ubiquitination, also play a critical role in regulating prohibitin’s function and stability. Furthermore, prohibitin can be regulated through its interactions with other proteins, which can influence its localization and activity within the cell.

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.