Myo Human w/o H

Myoglobin (Heme free) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Myoglobin heme free Human Recombinant produced in E.Coli is a non-glycosylated polypeptide chain having a molecular mass of 11.67 kDa. The Myoglobin heme free contains N-terminal T7 tag and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Myoglobin, a member of the globin superfamily found in skeletal and cardiac muscles, functions as a haemoprotein. It plays a crucial role in intracellular oxygen storage and facilitates oxygen diffusion across cells. Structurally, myoglobin is a single-chain globular protein composed of 153 amino acids. It features a heme prosthetic group, an iron-containing porphyrin, at its core, around which the remaining apoprotein folds. With 8 alpha helices and a hydrophobic core, myoglobin exhibits a molecular weight of 16.7 kDa. As the primary oxygen-carrying pigment in muscle tissues, myoglobin's oxygen binding differs from the cooperative binding observed in hemoglobin, which is exclusive to multimeric/oligomeric proteins. Instead, myoglobin's oxygen binding remains unaffected by the surrounding tissue's oxygen pressure. Characterized by a hyperbolic oxygen dissociation curve, myoglobin is often described as possessing an "instant binding tenacity" for oxygen. This characteristic, along with high myoglobin concentrations in their muscle cells, enables various organisms to hold their breath for extended periods. Furthermore, myoglobin contributes to the red pigmentation of meat. The meat's color is partially determined by the iron atom's charge within myoglobin and the oxygen bound to it. Primarily found in Type I, Type II A, and Type II B muscle fibers, myoglobin is generally not present in smooth muscle. Damaged muscle tissue, particularly in conditions like rhabdomyolysis, releases myoglobin due to its high concentration within these tissues. Although filtered by the kidneys, the released myoglobin exhibits toxicity to the renal tubular epithelium, potentially leading to acute renal failure.
Description
Recombinant Human Myoglobin, heme-free, is produced in E. coli. This non-glycosylated polypeptide chain has a molecular mass of 11.67 kDa. The protein is purified using proprietary chromatographic techniques and contains an N-terminal T7 tag.
Physical Appearance
Clear, sterile solution.
Formulation
The sterile solution is formulated in phosphate-buffered saline (pH 8.0) with 50mM phosphate-borate.
Stability
Myoglobin heme free can be stored at 15°C for up to 2 weeks but is best stored at 4°C. For long term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid freezing.
Purity
Purity is determined to be greater than 95.0% by SDS-PAGE analysis.
Synonyms
Myoglobin, MB, PVALB, MGC13548.
Source
Escherichia Coli.

Q&A

What is myo-inositol and what are its primary biochemical functions in human metabolism?

Myo-inositol is a naturally occurring cyclohexanehexol isomer that serves as a crucial precursor for phosphatidylinositol and inositol phosphates, which function as essential second messengers in cellular signal transduction systems. It plays a fundamental role in insulin signaling through inositolphosphoglycans that mediate glucose utilization across multiple tissue types . In human physiology, myo-inositol participates in numerous metabolic pathways including insulin sensitization, ovarian function, and oocyte development, making it particularly relevant in endocrinological research . The compound's participation in cell membrane formation and maintenance further highlights its biological importance in maintaining cellular homeostasis and proper physiological function. Research indicates that altered myo-inositol metabolism may contribute to various pathophysiological states, particularly those involving insulin resistance.

How does myo-inositol differ from D-chiro-inositol in terms of tissue distribution and physiological effects?

Myo-inositol and D-chiro-inositol, while both being inositol isomers, demonstrate significant differences in their tissue distribution and physiological effects that are critical for researchers to understand when designing studies. Myo-inositol is the most abundant naturally occurring inositol isomer in the human body and serves as a precursor to D-chiro-inositol through the action of an epimerase enzyme that is stimulated by insulin . Importantly, these isomers exhibit tissue-specific distributions and functions, with myo-inositol having preferential activity in ovarian tissue where it promotes follicular development and oocyte maturation . D-chiro-inositol, conversely, appears more active in liver and muscle tissues where it primarily enhances glycogen synthesis and glucose utilization . This tissue-specific activity explains why supplementation with myo-inositol alone is beneficial for classical PCOS patients with hyperandrogenism, whereas administration of D-chiro-inositol or combined formulations may yield different metabolic and reproductive outcomes . Understanding these differential physiological roles is essential for researchers when selecting appropriate inositol formulations for experimental protocols.

What is the current understanding of myo-inositol's mechanism of action in insulin signaling pathways?

Myo-inositol significantly influences insulin signaling through its conversion into inositolphosphoglycan mediators that act as second messengers in the insulin transduction pathway. These glycan mediators activate key enzymes involved in glucose metabolism, including pyruvate dehydrogenase and glycogen synthase, thereby enhancing cellular glucose uptake and utilization . Research demonstrates that myo-inositol supplementation can improve insulin receptor sensitivity, which is particularly relevant in conditions characterized by insulin resistance such as PCOS . In hyperandrogenic PCOS phenotypes, myo-inositol supplementation at doses of approximately 4,000mg/day has been shown to reduce testosterone levels by over 50%, suggesting a direct or indirect effect on steroidogenic pathways in ovarian tissue . The compound appears to restore normal insulin signaling by replenishing depleted inositolphosphoglycan pools, thereby addressing a fundamental metabolic defect in insulin-resistant states. This mechanism explains why myo-inositol supplementation may be particularly beneficial in classical PCOS phenotypes where insulin resistance drives hyperandrogenism and reproductive dysfunction.

What are the optimal protocols for quantifying myo-inositol concentrations in human biological samples?

Accurate quantification of myo-inositol in biological samples requires sophisticated analytical methodologies that can distinguish between closely related isomers. Mass spectrometry coupled with either liquid or gas chromatography represents the gold standard for myo-inositol analysis, providing high sensitivity and specificity in complex biological matrices . Researchers should implement careful sample preparation protocols, including protein precipitation and derivatization steps, to ensure reliable detection of myo-inositol across various tissue types and bodily fluids. When designing analytical protocols, consideration must be given to potential matrix interference from structurally similar compounds that might compromise measurement accuracy. Standardization using isotopically labeled internal standards is essential for controlling variability in extraction efficiency and instrument response. Additionally, researchers should validate their analytical methods according to established guidelines, determining parameters such as lower limits of quantification, linear range, precision, and accuracy before undertaking large-scale sample analysis.

How should pharmacokinetic/pharmacodynamic models be applied when studying myo-inositol interventions?

Pharmacokinetic/pharmacodynamic (PK/PD) modeling provides crucial insights into myo-inositol's absorption, distribution, and biological effects that are essential for optimizing study designs. Drawing from approaches used in other therapeutic areas, such as the MYO-029 antibody research, researchers should implement multi-compartmental PK models that account for both linear and potential non-linear clearance mechanisms of myo-inositol . Tissue distribution studies using radiolabeled compounds can elucidate myo-inositol's biodistribution across various organs and determine tissue-specific retention times, similar to methods employed in the 125I-labeled MYO-029 mouse studies . When analyzing dose-response relationships, effect compartment models or indirect response models may better characterize the delayed onset of myo-inositol's therapeutic effects, particularly on hormonal parameters. Model discrimination should be based on objective function values, graphical analysis of residuals, and visual predictive checks as demonstrated in comprehensive PK/PD research . Researchers should consider collecting serial blood samples at strategic timepoints following myo-inositol administration to accurately characterize absorption and elimination phases, while simultaneously measuring relevant biomarkers to establish exposure-response relationships.

What considerations are critical when designing clinical studies evaluating myo-inositol efficacy in PCOS patients?

Clinical studies evaluating myo-inositol in PCOS require careful phenotypic stratification of participants due to the heterogeneous nature of the syndrome and the compound's differential effects across patient subgroups. Researchers must classify PCOS patients according to established criteria (Rotterdam, Androgen Excess Society, or NIH) and further stratify by androgen status, as myo-inositol appears particularly beneficial in hyperandrogenic classical PCOS phenotypes but potentially detrimental in hypo-androgenic variants . Study designs should implement appropriate washout periods for confounding medications, particularly insulin sensitizers, hormonal contraceptives, and supplements that might influence the same pathways. Power calculations must account for the expected differential response across phenotypes, potentially requiring larger sample sizes than typically used in published studies . Outcome measures should extend beyond ovulation rates to include comprehensive metabolic and hormonal profiling, including detailed androgen panels, insulin sensitivity indices, and inflammatory markers. Additionally, researchers should carefully document concurrent supplementation, as evidence indicates that combining myo-inositol with substances like DHEA produces counterproductive effects due to their opposing actions on androgen levels .

How can researchers address the contradictory results observed in myo-inositol clinical trials?

The contradictory results observed across myo-inositol studies stem from several methodological factors that researchers must systematically address in future investigations. First, the tremendous variation in myo-inositol to D-chiro-inositol ratios used in different studies—ranging from 0.4:1 to 104:1—makes cross-study comparisons virtually impossible and necessitates the establishment of standardized formulations based on physiological ratios . Second, inadequate phenotypic characterization of study participants, particularly regarding androgen status, contributes significantly to inconsistent outcomes since myo-inositol demonstrates opposing effects in hyperandrogenic versus hypo-androgenic patients . Third, variability in dosing regimens, treatment duration, and outcome measures further confounds result interpretation and requires harmonization through consensus guidelines. Researchers should implement factorial study designs that systematically evaluate different dosages and isomer combinations across well-defined PCOS phenotypes. Additionally, studies should include comprehensive baseline characterization of participants' endocrine profiles and employ standardized analytical methods for measuring both myo-inositol levels and outcome parameters. Meta-analyses should be conducted with careful attention to subgroup effects based on phenotype and treatment protocol rather than pooling heterogeneous studies.

What are the implications of combining myo-inositol with other supplements or medications in research protocols?

The interaction of myo-inositol with other supplements or medications introduces significant confounding variables that researchers must carefully control in study designs. A particularly important consideration involves the contraindicated combination of myo-inositol with dehydroepiandrosterone (DHEA), as these compounds exert opposing effects on androgen levels . While DHEA supplementation aims to increase testosterone levels, particularly in patients with premature ovarian aging or hypo-androgenic conditions, myo-inositol acts to reduce testosterone levels, creating a physiological conflict that may neutralize therapeutic benefits when administered concurrently . Similarly, interactions between myo-inositol and insulin-sensitizing medications like metformin require investigation, as they may produce additive, synergistic, or potentially redundant effects on insulin signaling pathways. Researchers should implement factorial designs that systematically evaluate such combinations, with comprehensive monitoring of relevant biomarkers to detect potential interactions. Additionally, standardized reporting of concomitant supplements and medications should be mandated in clinical trials, as the widespread use of over-the-counter supplements may introduce unrecognized confounding effects. Pharmacokinetic studies examining potential alterations in myo-inositol absorption or metabolism when co-administered with other compounds would provide valuable insights for optimizing combination therapies.

How should dose-response relationships be established for myo-inositol across different clinical populations?

Establishing precise dose-response relationships for myo-inositol requires sophisticated pharmacological approaches that account for phenotypic heterogeneity and potential non-linear responses across different clinical populations. Researchers should implement adaptive trial designs that allow for sequential dose adjustment based on preliminary efficacy and safety signals, rather than fixed-dose protocols that predominate current literature . For classical PCOS patients with hyperandrogenism, preliminary evidence suggests effectiveness at doses of approximately 4,000mg/day, but systematic dose-ranging studies examining both lower and higher doses are lacking . When studying myo-inositol in combination with D-chiro-inositol, researchers should systematically evaluate multiple ratio configurations while maintaining control over total inositol dose, as current studies employ widely varying ratios from 0.4:1 to 104:1 without clear pharmacological justification . Pharmacokinetic studies should determine whether myo-inositol demonstrates linear or saturable absorption across the dosing range, which would influence optimal dosing regimens. Additionally, research protocols should include comprehensive biomarker assessment at multiple timepoints to establish temporal relationships between myo-inositol administration, plasma concentrations, and biochemical responses. Finally, genomic and metabolomic profiling of study participants may identify predictive biomarkers of dose response, allowing for more personalized dosing recommendations in future clinical applications.

What novel methodologies can enhance the study of myo-inositol's effects on reproductive outcomes?

Advanced methodologies for studying myo-inositol's reproductive effects should integrate emerging technologies that provide deeper insights into underlying mechanisms beyond conventional clinical endpoints. Single-cell transcriptomic analysis of granulosa cells and cumulus cells obtained during assisted reproductive procedures could elucidate myo-inositol's molecular effects on folliculogenesis at unprecedented resolution . Researchers should implement three-dimensional ultrasound with power Doppler to quantitatively assess ovarian vascularization and follicular development in response to myo-inositol therapy, providing functional anatomical correlates to hormonal parameters. Metabolomic profiling of follicular fluid could identify signature metabolite patterns associated with successful outcomes following myo-inositol treatment, potentially revealing new biomarkers of treatment response . In vitro fertilization protocols offer unique opportunities to assess myo-inositol's direct effects on oocyte quality through detailed morphological and molecular assessment of retrieved oocytes, with particular attention to meiotic spindle configurations and mitochondrial distribution. Additionally, researchers should consider implementing prospective studies with long-term follow-up to assess whether myo-inositol's effects on reproductive outcomes persist after discontinuation of therapy or whether continuous supplementation is necessary to maintain benefits. Finally, computational modeling integrating multiple data streams could help predict individual patient responses to myo-inositol intervention based on baseline characteristics.

What is the significance of gut microbiome analysis in myo-inositol research?

The emerging relationship between the gut microbiome and myo-inositol metabolism represents an important frontier in understanding individual variability in treatment response that researchers should incorporate into study designs. Recent advances in computational tools for gut microbiome analysis, such as Mayo Clinic's Gut Microbiome Wellness Index 2, demonstrate at least 80% accuracy in differentiating healthy individuals from those with disease states and could be applied to predict responsiveness to myo-inositol therapy . The gut microbiome may influence myo-inositol bioavailability through bacterial metabolism of dietary sources or modification of absorption kinetics, potentially explaining inter-individual variability in clinical response. Researchers should collect comprehensive microbiome data using shotgun metagenomic sequencing before and during myo-inositol administration to identify specific bacterial taxa or functional pathways that correlate with treatment outcomes. Particular attention should be paid to bacteria capable of producing phytase enzymes that liberate myo-inositol from phytate in the gut, potentially enhancing its bioavailability. Integrating microbiome data with metabolomic profiles may reveal how bacterial communities influence the conversion between inositol isomers, potentially identifying patients who might benefit from specific isomer ratios based on their microbiome composition. Additionally, fecal transplant studies in animal models could establish causal relationships between microbiome composition and myo-inositol metabolism, providing mechanistic insights for human applications.

How can genomic approaches enhance personalization of myo-inositol therapy in clinical research?

Genomic approaches offer unprecedented opportunities to identify genetic determinants of myo-inositol response, enabling truly personalized research protocols that maximize therapeutic efficacy. Researchers should implement genome-wide association studies in large cohorts of myo-inositol-treated patients to identify polymorphisms in genes involved in inositol metabolism, insulin signaling, and steroidogenesis that may predict treatment response . The examination of genetic variations in inositol-related pathways, similar to studies that identified IL17RB gene variants affecting treatment outcomes in alcohol addiction therapy, could reveal pharmacogenomic markers specific to myo-inositol efficacy . Whole exome sequencing approaches, modeled after Mayo Clinic's Tapestry study which analyzed protein-coding genes from over 100,000 diverse participants, could establish comprehensive genetic profiles associated with optimal myo-inositol response . RNA sequencing of target tissues before and after myo-inositol administration would elucidate transcriptomic changes underpinning therapeutic effects, potentially identifying novel biomarkers and molecular targets. Additionally, researchers should investigate epigenetic modifications induced by myo-inositol treatment, as these may contribute to sustained therapeutic effects beyond the supplementation period. Integration of genomic data with clinical, metabolomic, and microbiome datasets would enable the development of predictive algorithms for identifying optimal candidates for myo-inositol therapy and personalizing dosing regimens.

What standardization approaches are needed to improve reproducibility in myo-inositol research?

Improving reproducibility in myo-inositol research requires comprehensive standardization efforts across multiple domains of study design, implementation, and reporting that the scientific community must collectively address. Researchers should establish consensus guidelines for myo-inositol formulation standardization, including acceptable ranges for isomer ratios and minimum purity requirements for research-grade materials to eliminate the current wide variation in commercial preparations . Analytical methods for quantifying myo-inositol in biological samples require harmonization through international ring trials and certified reference materials development to ensure comparability of results across laboratories . Patient classification schemes specific to myo-inositol research should be developed, moving beyond general diagnostic criteria for conditions like PCOS to incorporate parameters known to influence myo-inositol response, particularly androgen status and insulin sensitivity metrics . Standardized reporting templates for myo-inositol clinical trials should be implemented, requiring disclosure of detailed supplement characteristics, concurrent medications, and comprehensive baseline patient phenotyping. Additionally, researchers should establish minimum datasets for publication, including key pharmacokinetic parameters and biomarkers of metabolic and hormonal response measured at standardized timepoints. Finally, development of shared data repositories would facilitate individual participant data meta-analyses that could overcome the limitations of small, heterogeneous studies and identify consistent patterns across diverse populations and methodologies.

What translational models can bridge preclinical and clinical research on myo-inositol's mechanisms?

Developing robust translational models is essential for elucidating myo-inositol's mechanisms of action and efficiently bridging the gap between preclinical discoveries and clinical applications. Researchers should implement patient-derived primary cell cultures from target tissues (ovarian theca and granulosa cells, adipocytes, skeletal muscle) to directly assess myo-inositol's molecular effects in relevant human cellular contexts . Three-dimensional organoid models of ovarian follicles would provide systems for studying myo-inositol's impact on steroidogenesis and oocyte development under controlled conditions that better recapitulate in vivo physiology than traditional cell lines. Humanized mouse models with tissue-specific insulin receptor modifications could help elucidate the differential tissue responses to myo-inositol supplementation observed clinically, particularly the opposing effects in ovarian versus peripheral tissues . Implementation of integrated PK/PD modeling approaches, similar to those used in the MYO-029 antibody development program, would facilitate translation of exposure-response relationships from preclinical species to humans, ensuring that clinical dosing achieves target engagement . Innovative imaging techniques such as positron emission tomography with radiolabeled myo-inositol could map tissue-specific distribution and metabolism in both animal models and humans, providing crucial insights into pharmacokinetics and target engagement. Additionally, ex vivo perfused human tissue models could assess acute metabolic responses to myo-inositol under physiologically relevant conditions while controlling for confounding variables present in whole-organism studies.

What are the key analytical challenges in measuring tissue-specific myo-inositol concentrations?

Accurate measurement of tissue-specific myo-inositol concentrations presents several analytical challenges that researchers must address to generate reliable data for mechanistic studies. The high structural similarity between inositol isomers necessitates highly selective analytical methods capable of distinguishing myo-inositol from D-chiro-inositol and other isomers without cross-reactivity, typically requiring specialized chromatographic separation coupled with mass spectrometric detection . Tissue extraction protocols must be optimized for each specific tissue type to ensure complete recovery of myo-inositol while minimizing artifactual changes in isomer ratios that might occur during sample processing. Researchers should implement methods similar to those used in the 125I-labeled MYO-029 biodistribution studies, where tissues were carefully perfused prior to analysis to eliminate blood contamination that could confound tissue-specific measurements . The relatively rapid metabolism and interconversion between inositol isomers necessitates careful timing of sample collection and immediate preservation to capture accurate in vivo concentrations. Matrix effects from lipids, proteins, and other biological compounds can significantly interfere with myo-inositol quantification, requiring extensive method validation across different tissue types. Additionally, researchers must develop standardized approaches for normalizing myo-inositol concentrations, weighing the relative merits of expressing results per unit weight, per cell number, or per protein content depending on the research question. Finally, the incorporation of isotopically labeled internal standards is essential for controlling pre-analytical variability and ensuring accurate quantification across diverse sample types.

How can researchers control for dietary myo-inositol intake in clinical studies?

Controlling for dietary myo-inositol represents a significant methodological challenge that researchers must address to isolate the effects of supplementation from background intake. Implementation of standardized food frequency questionnaires specifically validated for inositol-containing foods should be included in study protocols, with particular attention to major dietary sources such as fruits, beans, grains, and nuts . Researchers should consider incorporating controlled feeding periods before baseline assessments and at critical timepoints throughout intervention studies, providing meals with known and consistent myo-inositol content to minimize dietary variability. Analysis of 24-hour urine collections for myo-inositol and its metabolites could provide objective measures of total inositol exposure, complementing self-reported dietary data with biological markers of intake. In studies where dietary control is not feasible, statistical approaches should be developed to adjust for estimated background dietary intake using validated nutritional databases that include myo-inositol content of common foods. Stratification of randomization based on baseline dietary myo-inositol intake could help ensure balanced distribution of this potential confounder between treatment groups. Additionally, researchers should measure baseline serum or plasma myo-inositol concentrations prior to supplementation to account for pre-existing differences in endogenous levels that might influence treatment response. Finally, crossover study designs with adequate washout periods might help control for individual differences in dietary patterns, although the appropriate washout duration for myo-inositol remains to be established through dedicated pharmacokinetic studies.

What statistical approaches are most appropriate for analyzing dose-response relationships in myo-inositol research?

Analysis of myo-inositol dose-response relationships requires sophisticated statistical approaches that can accommodate non-linear responses, inter-individual variability, and multiple interacting factors influencing treatment outcomes. Researchers should implement mixed-effects modeling techniques that can simultaneously account for fixed factors (dose, duration, formulation) and random effects (inter-individual variability in pharmacokinetics and pharmacodynamics) when analyzing longitudinal data from dose-ranging studies . Bayesian hierarchical models offer advantages for small sample sizes typical in early-phase clinical research, allowing incorporation of prior knowledge about myo-inositol's biological effects while quantifying uncertainty in parameter estimates. When evaluating different myo-inositol:D-chiro-inositol ratios, response surface methodology provides a statistical framework for identifying optimal combinations across multiple outcome measures simultaneously, more efficiently than traditional factorial designs . Researchers should employ time-to-event analyses for outcomes such as ovulation or pregnancy that incorporate right-censoring rather than dichotomizing these continuous processes at arbitrary timepoints. For mechanistic studies exploring multiple biological pathways, structural equation modeling can elucidate direct and indirect effects of myo-inositol on clinical endpoints through various mediating factors. Additionally, researchers should implement sensitivity analyses that systematically evaluate the impact of potential confounders such as diet, concurrent medications, and baseline characteristics on observed dose-response relationships. Finally, adaptive trial designs with Bayesian decision rules could optimize resource allocation by modifying dosing arms based on accumulating efficacy and safety data, particularly valuable given the current uncertainty regarding optimal dosing regimens.

How can systems biology approaches enhance understanding of myo-inositol's biological effects?

Systems biology approaches offer powerful frameworks for comprehensively mapping myo-inositol's complex interactions across multiple biological pathways and hierarchical levels of organization. Researchers should implement multi-omics integration strategies that combine transcriptomics, proteomics, metabolomics, and lipidomics data from the same individuals before and after myo-inositol administration to construct comprehensive molecular response networks . Network pharmacology approaches can identify hub genes and proteins mediating myo-inositol's effects on insulin signaling, steroidogenesis, and inflammation, potentially revealing novel therapeutic targets or biomarkers. Constraint-based metabolic modeling using genome-scale reconstructions of human metabolism could predict tissue-specific metabolic flux changes induced by myo-inositol supplementation, generating testable hypotheses about mechanism of action. Researchers should develop dynamic mathematical models of inositol phosphate signaling pathways that incorporate differential tissue distribution and interconversion of inositol isomers to predict system-level responses to various supplementation regimens . Integration of clinical, molecular, and genetic data through machine learning algorithms could identify patient subgroups with distinct response patterns to myo-inositol intervention, enabling personalized therapeutic approaches. Additionally, comparative systems analyses between myo-inositol and established insulin sensitizers like metformin could elucidate shared and unique mechanism of action, potentially informing combination therapy approaches. Finally, quantitative systems pharmacology models that incorporate physiologically-based pharmacokinetics with pharmacodynamic effect models would provide a mathematical framework for translating preclinical findings to clinical applications and optimizing dosing regimens across different patient populations.

What are the best approaches for investigating myo-inositol's effects across different human tissue types?

Investigating myo-inositol's differential effects across human tissues requires innovative methodological approaches that can overcome the ethical and practical limitations of tissue sampling in clinical research. Researchers should implement tissue-specific organoid models derived from human pluripotent stem cells or adult stem cells to study myo-inositol's effects on key target tissues such as ovary, adipose, skeletal muscle, and liver under controlled conditions . Single-cell RNA sequencing of accessible tissues (blood, adipose) before and after myo-inositol administration could identify cell type-specific transcriptional signatures that might serve as surrogate markers for responses in less accessible tissues. Functional tissue slice cultures obtained from surgical specimens provide opportunities to study acute myo-inositol responses in intact tissue architecture with preserved intercellular communications and extracellular matrix. Non-invasive imaging modalities such as magnetic resonance spectroscopy could potentially quantify tissue-specific myo-inositol concentrations in vivo, allowing longitudinal assessment of biodistribution following supplementation. Researchers should consider implementing techniques from pharmacokinetic studies of other compounds, such as the radiolabeled tissue distribution analyses used for MYO-029, adapting these approaches for studying myo-inositol distribution across multiple tissues . Explant cultures of reproductive tissues (endometrium, placenta) obtained during routine clinical procedures offer opportunities to study myo-inositol's effects on specific reproductive functions. Additionally, innovative biobanking approaches that coordinate collection of multiple tissue types from the same donors undergoing surgical procedures could enable integrated multi-tissue analyses of myo-inositol's differential effects, particularly valuable for understanding the opposing actions observed in ovarian versus peripheral tissues.

How should researchers integrate patient-reported outcomes in myo-inositol clinical studies?

Integration of patient-reported outcomes (PROs) in myo-inositol research requires methodologically rigorous approaches that complement traditional biochemical and clinical endpoints while capturing dimensions of health important to patients. Researchers should implement validated PRO instruments specific to the condition being studied, such as the Polycystic Ovary Syndrome Questionnaire (PCOSQ) or modified Ferriman-Gallwey score for hirsutism in PCOS studies, rather than generic quality of life measures that may lack sensitivity to condition-specific improvements . Mixed-methods designs that combine quantitative PRO measures with qualitative interviews would provide complementary insights into patients' experiences with myo-inositol therapy, potentially identifying benefits or adverse effects not captured by standardized instruments. Ecological momentary assessment using smartphone applications could collect real-time symptom data during myo-inositol treatment, overcoming recall bias associated with retrospective questionnaires and capturing symptom fluctuations with greater temporal resolution. Researchers should develop and validate PRO measures specifically designed to assess symptoms known to respond to myo-inositol, such as insulin resistance manifestations, menstrual irregularity, and hyperandrogenic symptoms . Integration of PRO data with biomarker measurements through mediation analysis could elucidate mechanisms underlying symptom improvement and identify biological correlates of patient-experienced benefits. Additionally, researchers should implement responder analyses that define clinically meaningful improvements from the patient perspective rather than relying solely on statistical significance of mean changes in PRO scores. Finally, long-term follow-up of PROs after study completion would provide valuable insights into the durability of myo-inositol's effects on symptoms and quality of life beyond the typical short duration of controlled trials.

Product Science Overview

Structure and Function

Myoglobin consists of a single polypeptide chain of 153 amino acids and a heme prosthetic group. The heme group is responsible for the protein’s ability to bind oxygen. Myoglobin’s high affinity for oxygen allows it to effectively store and release oxygen as needed by muscle cells. This property is crucial for maintaining cellular respiration and energy production during physical exertion.

Heme-Free Myoglobin

Heme-free myoglobin refers to the protein without its heme group. The removal of the heme group can be achieved through various biochemical techniques, resulting in an apo-myoglobin form. This form is often used in research to study the structural and functional properties of the protein without the influence of the heme group. Heme-free myoglobin can also be reconstituted with synthetic or modified heme groups to investigate the effects of different heme structures on the protein’s function.

Human Recombinant Myoglobin

Recombinant DNA technology allows for the production of human myoglobin in various host systems, such as bacteria, yeast, or mammalian cells. This recombinant myoglobin is identical to the naturally occurring protein in humans and can be produced in large quantities for research and therapeutic purposes. The recombinant production of myoglobin ensures a consistent and pure supply of the protein, which is essential for detailed biochemical and biophysical studies.

Applications in Research and Medicine
  1. Structural Studies: Heme-free myoglobin is used to investigate the folding and stability of the protein. By studying the apo form, researchers can gain insights into the role of the heme group in the overall structure and function of myoglobin.
  2. Oxygen Binding Studies: Recombinant myoglobin, both with and without heme, is used to study oxygen binding kinetics and the effects of various ligands on oxygen affinity. These studies are important for understanding how myoglobin functions under different physiological conditions.
  3. Therapeutic Potential: Myoglobin and its derivatives have potential therapeutic applications in conditions where oxygen delivery to tissues is compromised, such as in ischemic diseases. Recombinant myoglobin can be engineered to enhance its oxygen-binding properties or to deliver therapeutic agents to specific tissues.

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.