MT I

Melanotan-I
Shipped with Ice Packs
In Stock

Description

Multi-Target Compounds (MT-CPDs)

MT-CPDs are compounds with activity against five or more biological targets, as defined in high-throughput screening studies. Below is a structured analysis of their properties and research findings.

Dataset Composition

The following table summarizes MT-CPD and ST-CPD distributions from mixed and biochemical assays :

Dataset TypePD ThresholdMT-CPDsST-CPDs
Mixed≥52,85815,839
Biochemical≥51,2426,629
Biochemical≥33,46811,793

PD = Promiscuity degree (number of targets a compound affects).

Machine Learning Insights

Recent studies reveal that:

  • Feature Subsets: MT-CPDs share structural features with ST-CPDs, enabling cross-prediction using machine learning models .

  • Diagnostic Tools: Explainable ML identifies critical molecular descriptors (e.g., aromatic rings, halogens) driving multi-target activity .

Meitnerium (Mt): Lack of Compounds

Meitnerium, a transactinide element, has no synthesized or characterized compounds due to its extreme radioactivity and short half-lives (e.g., Mt-278: 4.5 s; Mt-282: 67 s unconfirmed) .

Theoretical Predictions

PropertyTheoretical Value
Electron Configuration[Rn] 5f¹⁴ 6d⁷ 7s² .
Predicted Oxidation States+3, +6, +1 (analogous to iridium) .
Chemical ChallengesSublimation experiments exceed 500°C, unsuitable for current detection methods .

Bond Enthalpy Data

No experimental bond enthalpies exist for meitnerium compounds. Theoretical tables for diatomic species (e.g., MtH, MtO) remain empty .

Recommendations for Further Research

  1. MT-CPDs: Prioritize structural analysis of feature subsets driving multi-target activity .

  2. Meitnerium: Focus on improving particle accelerator techniques to isolate longer-lived isotopes (e.g., Mt-282) for chemical studies .

Product Specs

Description
Melanotan-I is a peptide composed of the amino acid sequence Ser-Tyr-Ser-Nle-Glu-His-D-Phe-Arg-Trp-Gly-Lys-Pro-Val, possessing a molecular weight of 1647.4 Daltons.
Physical Appearance
Sterile Filtered off-White lyophilized powder.
Formulation
The protein was lyophilized without any additives to a concentration of 1mg/ml.
Solubility
Reconstitute the lyophilized Melanotan-I in sterile 1% acetic acid at a concentration not less than 100 µg/ml. Further dilutions can be made using other aqueous solutions.
Stability
Lyophilized Melanotan-I remains stable at room temperature for up to 3 weeks. However, for extended storage, it's recommended to store it desiccated below -18°C. After reconstitution, MT-I can be stored at 4°C for 2-7 days. For longer-term storage, freeze at -18°C. To ensure optimal stability during long-term storage, add a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 98.0% as determined by RP-HPLC analysis.
Synonyms
Melanotan-I, MT-I, Melanotan-1.

Q&A

What is Metallothionein-I and how does it differ from other MT isoforms?

Metallothionein-I is one of four mammalian MT isoforms (MT-I through MT-IV), characterized by its high cysteine content and metal-binding capacity. MT-I and MT-II are often studied together (MT-I+II) as they share significant structural and functional similarities, particularly in the brain. Unlike MT-III and MT-IV which have more restricted tissue expression patterns, MT-I is more widely expressed across multiple tissues and is particularly responsive to stress conditions .

The primary distinguishing feature of MT-I is its inducibility in response to various stimuli including brain injury, inflammation, and oxidative stress. From a methodological perspective, researchers should note that studying MT-I in isolation from MT-II can be challenging due to their similar expression patterns and functional redundancy, which is why many studies examine them collectively as MT-I+II .

What are the primary functions of MT-I in the central nervous system?

MT-I functions as a multipurpose neuroprotectant in the central nervous system through several key mechanisms:

  • Metal ion homeostasis - Primarily binding zinc and copper ions

  • Free radical scavenging - Protecting against oxidative stress

  • Immunomodulation - Inhibiting macrophages, T lymphocytes and their inflammatory mediators

  • Anti-apoptotic effects - Preventing neuronal cell death

  • Promotion of cell survival and proliferation - Enhancing cell cycle progression and mitosis

When designing experiments to study these functions, researchers should consider using multiple complementary approaches to assess each function separately. For instance, metal-binding capacity can be evaluated through atomic absorption spectroscopy, while anti-inflammatory functions might require immunological assays examining cytokine profiles and immune cell activation states .

How is MT-I expression regulated at the transcriptional level?

MT-I expression is regulated by multiple transcriptional mechanisms, with metal response elements (MREs) playing a key role in its induction. Following brain injury, hepatic MT-I mRNA levels significantly increase within 24 hours, indicating rapid transcriptional activation . This regulation involves several elements:

  • Metal-responsive transcription factor-1 (MTF-1)

  • Antioxidant response elements (AREs)

  • Glucocorticoid response elements (GREs)

  • STAT (Signal Transducers and Activators of Transcription) binding sites

For studying transcriptional regulation of MT-I, quantitative reverse-transcriptase PCR (RT-PCR) has been effectively used to measure mRNA levels in various tissues following physiological challenges. When designing such experiments, researchers should include appropriate housekeeping genes as controls and consider the temporal dynamics of expression, as MT-I mRNA and protein levels may peak at different timepoints .

What are the optimal methods for measuring MT-I protein levels in tissue samples?

Based on established research protocols, the optimal methods for measuring MT-I protein levels include:

  • Enzyme-linked immunosorbent assay (ELISA) using validated antibodies like UC1MT

  • Western blotting with isoform-specific antibodies

  • Immunohistochemistry for tissue localization

  • Mass spectrometry for precise quantification and post-translational modification analysis

When implementing ELISA for MT-I detection, validation with appropriate controls is critical. Researchers have successfully used displacement curves constructed with tissues from MT-I/II knockout (MT-I/II−/−) mice to validate ELISA specificity . This approach helps distinguish genuine MT-I signal from background or cross-reactivity.

For temporal studies, it's important to note that protein expression may lag behind mRNA induction. For example, after brain injury, hepatic MT-I/II protein levels weren't significantly increased until 3 days post-injury, despite mRNA increases within 24 hours. Maximum protein levels were observed at 7 days post-injury, highlighting the importance of extended timepoint analysis .

How can researchers effectively differentiate between MT-I and MT-II in experimental settings?

Differentiating between MT-I and MT-II presents significant challenges due to their structural similarity and often overlapping expression patterns. Methodological approaches to address this include:

  • Isoform-specific RT-PCR primers targeting unique regions of MT-I and MT-II mRNAs

  • High-resolution chromatography techniques coupled with mass spectrometry

  • Isoform-specific antibodies (though true specificity should be validated)

  • Genetic approaches using isoform-specific knockout models

When designing experiments requiring isoform specificity, researchers should employ multiple complementary techniques and include appropriate controls. For instance, tissues from MT-I/II knockout mice serve as excellent negative controls for validating antibody specificity . Additionally, researchers should be conscious that many studies report combined MT-I+II data due to these technical challenges, which should be considered when interpreting literature findings.

What genetically modified mouse models are available for studying MT-I function?

Key mouse models available for MT-I research include:

  • MT-I/II double knockout mice (MT-I/II−/−) - Allow for studying the combined effects of MT-I and MT-II deficiency

  • MT-I overexpression transgenic mice - Enable investigation of protective effects of elevated MT-I levels

  • Cell-specific or inducible MT-I/II expression models - Permit targeted studies of MT-I/II function in specific tissues or developmental stages

These models have been instrumental in demonstrating the importance of MT-I+II for coping with brain pathology. Experiments using MT-I/II knockout mice have shown altered zinc handling after brain injury, including failure to normalize liver zinc levels at 7 days post-injury, suggesting that MT-I/II is responsible for sequestering elevated zinc to the liver following brain trauma .

When using these models, researchers should consider potential compensatory mechanisms that may develop, particularly in constitutive knockout models, and include appropriate wild-type controls matched for genetic background .

How does MT-I contribute to zinc homeostasis following brain injury?

MT-I plays a crucial role in zinc homeostasis after brain injury through a dynamic process involving multiple organs:

  • Following brain injury, MT-I expression is induced in both brain and liver tissues

  • Hepatic zinc content initially decreases at 1 and 3 days post-injury (DPI)

  • By 7 DPI, zinc levels return to normal in wild-type mice but remain depleted in MT-I/II knockout mice

  • This suggests MT-I/II is responsible for sequestering and normalizing elevated levels of zinc to the liver

This zinc sequestration mechanism appears to be a protective response, potentially preventing zinc-mediated toxicity in the injured brain. For researchers investigating this phenomenon, atomic absorption spectroscopy provides an effective method for measuring tissue zinc content. When designing such studies, it's important to include multiple timepoints (acute, subacute, and chronic phases) and compare wild-type with MT-I/II knockout animals to fully characterize the temporal dynamics of zinc redistribution .

What methodologies are recommended for studying MT-I metal-binding properties in vitro?

For investigating MT-I metal-binding properties, several complementary approaches are recommended:

  • Isothermal titration calorimetry (ITC) - For determining binding affinities and thermodynamic parameters

  • Atomic absorption spectroscopy - For quantifying metal content

  • Circular dichroism (CD) spectroscopy - For monitoring conformational changes upon metal binding

  • Fluorescence spectroscopy - For studying binding kinetics using zinc-specific fluorophores

  • X-ray absorption spectroscopy - For detailed coordination environment analysis

When conducting these studies, researchers should consider using recombinant MT-I protein to ensure homogeneity and control metallation state. It's also advisable to perform experiments under anaerobic conditions when possible, as the thiol groups in MT-I are susceptible to oxidation, which can significantly alter metal-binding properties.

A critical methodological consideration is pH control, as protonation of thiol groups affects metal binding. Experiments should be conducted at physiologically relevant pH (7.4) and with appropriate metal chelators as controls to validate specific binding .

What cellular mechanisms underlie MT-I's neuroprotective effects?

MT-I exerts neuroprotection through multiple cellular mechanisms:

  • Anti-inflammatory effects - MT-I+II inhibit macrophages, T lymphocytes and their formation of pro-inflammatory mediators including interleukins, tumor necrosis factor-alpha, matrix metalloproteinases, and reactive oxygen species

  • Enhanced cell survival - MT-I+II promote cell cycle progression, mitosis, and activate anti-apoptotic pathways

  • Neuronal apoptosis inhibition - MT-I+II interfere with pro-apoptotic signaling cascades

  • Metal ion homeostasis - By binding excess zinc and copper, MT-I prevents metal-induced neurotoxicity

  • Free radical scavenging - MT-I directly neutralizes reactive oxygen species

Methodologically, researchers investigating these mechanisms should employ pathway-specific assays. For studying anti-inflammatory effects, flow cytometry analysis of immune cell populations, multiplex cytokine assays, and RNA-seq of inflammatory gene signatures are recommended. Cell survival mechanisms can be assessed through BrdU incorporation assays, cell cycle analysis, and apoptosis detection methods including caspase activity assays and TUNEL staining .

How does exogenous administration of MT-I compare to endogenous expression for neuroprotection?

Research has demonstrated that both endogenous MT-I+II and exogenously administered MT-I or MT-II can provide neuroprotection, suggesting both intra- and extracellular mechanisms of action:

  • Endogenous MT-I overexpression in transgenic models has confirmed protective effects against brain pathology

  • Exogenous MT-I or MT-II administered intraperitoneally promotes similar neuroprotective effects as endogenous MT-I+II

  • This dual efficacy indicates MT-I+II functions through both intracellular and extracellular mechanisms

For researchers designing MT-I administration studies, several methodological considerations are important:

  • Dose-response relationships should be established for different routes of administration

  • Pharmacokinetics and tissue distribution should be assessed using labeled MT-I

  • Blood-brain barrier penetration or alternative mechanisms of central action should be evaluated

  • Timing of administration relative to injury is critical, with earlier intervention typically showing greater efficacy

How do MT-I expression patterns differ between acute injury and chronic neurodegenerative conditions?

MT-I expression exhibits distinct patterns in acute versus chronic neurological conditions:

Condition TypeMT-I Expression PatternCellular SourcesTemporal Profile
Acute Brain InjuryRapid inductionPrimarily astrocytes, also hepatocytesPeaks within days, may persist for weeks
Chronic NeurodegenerationVariable, often dysregulatedMultiple cell types including microgliaFluctuating levels, correlation with disease progression

In acute injury models such as traumatic brain injury, MT-I expression shows a clear temporal pattern with hepatic MT-I mRNA levels significantly increasing within 24 hours and protein levels peaking at 7 days post-injury . This pattern suggests a coordinated systemic response.

For researchers investigating these differential patterns, longitudinal studies with multiple timepoints are essential. Tissue-specific analysis should include both central (brain regions) and peripheral (liver) sources of MT-I. Single-cell RNA sequencing can provide valuable insights into cell-type-specific expression patterns, particularly in heterogeneous neural tissues .

What are the challenges in translating MT-I research from animal models to clinical applications?

Translating MT-I research to clinical applications faces several methodological and conceptual challenges:

  • Interspecies differences in MT regulation and function

  • Delivery methods for MT-I as a potential therapeutic

  • Blood-brain barrier penetration limitations

  • Difficulty in monitoring MT-I function and efficacy in human subjects

  • Developing human-specific MT-I modulators with acceptable pharmacokinetic profiles

Despite these challenges, MT-I and MT-II compounds have demonstrated good tolerability in preclinical studies, suggesting potential for therapeutic development . For researchers working on translational aspects, several approaches may help address these challenges:

  • Humanized mouse models expressing human MT variants

  • Advanced drug delivery systems targeting MT-I to the CNS

  • Identification of small molecule MT-I inducers with favorable pharmacokinetics

  • Development of biomarkers that correlate with MT-I activity in humans

  • Comparative studies examining MT-I function across species to identify conserved mechanisms

How can contradictory findings about MT-I function be reconciled in experimental design?

Contradictory findings regarding MT-I function may arise from several methodological variables:

  • Inability to distinguish between MT-I and MT-II specific effects

  • Differences in injury/disease models and severity

  • Temporal considerations - sampling at different timepoints post-intervention

  • Variations in genetic backgrounds of experimental animals

  • Differences in metal content of diet or housing conditions affecting baseline MT status

To reconcile contradictory findings, researchers should implement comprehensive experimental designs that:

  • Include both MT-I/II knockout and MT-I overexpressing models

  • Perform detailed time-course studies with multiple sampling points

  • Control for environmental variables affecting metal homeostasis

  • Directly compare multiple injury/disease models using identical MT-I assessment methods

  • Utilize multi-omics approaches (transcriptomics, proteomics, metallomics) to capture the complexity of MT-I responses

What role might MT-I play in multi-target drug development approaches?

MT-I research intersects with multi-target drug development through several pathways:

  • Understanding MT-I as a naturally occurring multi-functional protein provides insights into designing multi-target compounds (MT-CPDs)

  • MT-I's ability to modulate multiple pathways (anti-inflammatory, anti-apoptotic, antioxidant) offers a template for multi-target drug design

  • Structural studies of MT-I interactions with various proteins can inform pharmacophore development

Multi-target compounds (MT-CPDs) are distinguished from single-target compounds (ST-CPDs) by their ability to specifically interact with multiple targets. Machine learning studies have provided evidence for target combination-dependent structural characteristics that differentiate MT-CPDs from ST-CPDs . These insights could guide the design of new compounds with desired multi-target activity, including those mimicking MT-I's beneficial properties.

Researchers exploring this intersection should consider computational approaches including:

  • Molecular modeling of MT-I interactions with various binding partners

  • Machine learning algorithms to identify structural features that enable multi-target engagement

  • Network pharmacology approaches to predict the impact of MT-I modulation on disease pathways

What are the current technical limitations in measuring MT-I in clinical samples?

Current technical challenges in clinical MT-I assessment include:

  • Limited sensitivity of commercially available antibodies for distinguishing MT-I from other MT isoforms

  • Lack of standardized reference ranges for MT-I in human tissues and biofluids

  • Pre-analytical variables affecting MT-I stability in clinical samples

  • Absence of validated high-throughput assays suitable for clinical laboratories

To address these limitations, researchers should consider developing:

  • Next-generation MS-based assays with isoform-specific peptide detection

  • Aptamer-based detection methods with improved specificity

  • Standardized sample collection and processing protocols that preserve MT-I integrity

  • Novel biomarkers that correlate with MT-I activity rather than just protein levels

When designing clinical studies, researchers should incorporate method validation steps including recovery experiments, matrix effect evaluation, and comparison with existing methodologies when possible .

What are the most promising future research directions for MT-I in neuroscience?

Based on current evidence, several promising research directions for MT-I in neuroscience emerge:

  • Development of MT-I mimetic compounds with enhanced blood-brain barrier penetration

  • Exploration of cell-specific MT-I functions using conditional knockout or expression models

  • Investigation of MT-I in the context of neurodevelopmental disorders

  • Elucidation of MT-I's role in glial-neuronal interactions during brain repair

  • Application of systems biology approaches to understand MT-I within the broader context of metalloproteins and zinc homeostasis

For researchers pursuing these directions, integration of multiple methodological approaches is recommended. Single-cell technologies combined with spatial transcriptomics can reveal cell-specific MT-I functions in complex neural tissues. CRISPR-based approaches allow for precise manipulation of MT-I expression in specific cell populations. Advanced imaging techniques, including metal-specific probes, can track MT-I-dependent metal redistribution in real-time .

Product Science Overview

Chemical Structure and Mechanism of Action

Melanotan-I is a linear peptide consisting of 13 amino acids. It has a similar structure to α-MSH, with two key differences: the fourth amino acid is norleucine, and the seventh amino acid is D-phenylalanine . These modifications make Melanotan-I more potent and resistant to enzymatic breakdown compared to its natural counterpart.

The peptide works by binding to melanocortin receptors, particularly the melanocortin 1 receptor (MC1R), which is primarily found in melanocytes, the cells responsible for melanin production. Upon binding to MC1R, Melanotan-I stimulates the production of eumelanin, a type of melanin that provides a darker pigmentation and offers protection against ultraviolet (UV) radiation .

Clinical Applications and Benefits

Melanotan-I has been investigated for several clinical applications, including:

  1. Skin Tanning: The primary use of Melanotan-I is to induce skin tanning. It has been shown to significantly enhance tanning and reduce the risk of sunburn by increasing melanin production .
  2. Photoprotection: By increasing melanin levels, Melanotan-I provides a protective effect against UV radiation, reducing the risk of skin damage and potentially lowering the incidence of skin cancers .
  3. Treatment of Skin Disorders: Melanotan-I has been studied for its potential in treating various skin disorders, such as vitiligo, a condition characterized by the loss of skin pigmentation .
Safety and Side Effects

While Melanotan-I has shown promise in clinical studies, its safety profile is still under investigation. Some of the reported side effects include:

  • Mild administration-related symptoms, such as nausea and flushing .
  • Potential long-term risks, which require further research to fully understand .

It is important to note that Melanotan-I should not be confused with Melanotan-II, another synthetic peptide with similar properties but a different receptor specificity and side effect profile .

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.