Synonym |
Cyclo(D-Ala-Val) |
Species |
Human |
Protein Accession |
NA |
Purity |
Greater than 95% by HPLC analysis |
Endotoxin Level |
Less than 1 EU/μg |
Biological Activity |
NA |
Expression System |
Chemical Synthesis |
Fusion Tag |
NA |
Predicted Molecular Mass |
NA |
Formulation |
Supplied as a lyophilized powder |
Reconstitution |
Centrifuge vial before opening. Reconstitute in water to a concentration of 0.1-1.0 mg/ml |
Storage & Stability |
Stable at -20°C for up to 12 months. Store at 4°C after reconstitution for up to two weeks. For
long term storage, aliquot and store at -20°C |
FAQ
What is Cyclo(D-Ala-Val) and what makes it unique compared to other cyclic
peptides?
Cyclo(D-Ala-Val) is a cyclic dipeptide composed of the two amino acids D-alanine
(D-Ala) and valine (Val). As a cyclic peptide, it is distinct from linear peptides due to its closed
ring structure, which is formed by the peptide bond between the carboxyl group of one amino acid and the
amino group of another. This cyclic configuration provides several advantages over linear forms, notably
increased stability against enzymatic degradation and modulation of biological activity. The stability
arises because the circular structure is less susceptible to proteolytic enzymes that typically
recognize open peptide bonds, allowing Cyclo(D-Ala-Val) to maintain its integrity longer than linear
peptides when exposed to biological environments.
The formation of a dipeptide linking D-alanine,
which is an enantiomer of the more common L-alanine, contributes to the unique properties of
Cyclo(D-Ala-Val). The use of D-amino acids in the synthesis can significantly enhance the stability of
the peptide as it can prevent recognition and degradation by standard proteases that are specifically
tuned to target L-amino acids. This makes Cyclo(D-Ala-Val) a particularly interesting molecule in the
field of pharmaceutical research, where stability can be a critical factor for therapeutic
effectiveness.
Beyond stability, the activity profile of Cyclo(D-Ala-Val) is of great research
interest. Its compact and cyclic nature can enable unique interactions with biological molecules,
potentially offering pharmacological activities such as antimicrobial, anticancer, or anti-inflammatory
effects. These effects are still an active area of research, leading to inquiries about
Cyclo(D-Ala-Val)'s potential role in therapeutic applications.
The combination of its stability,
use of D-amino acids, and cyclic structure offers a promising platform for drug design. Cyclo(D-Ala-Val)
serves not only as a unique biochemical tool but also as a template for synthetic modifications aimed at
targeting specific biological pathways. Researchers continue to study this cyclic dipeptide to unlock
new insights into its mechanisms and potential applications.
How can Cyclo(D-Ala-Val) contribute
to antimicrobial research and development?
Cyclo(D-Ala-Val) has gained attention within
antimicrobial research due to its potential inhibitory effects against pathogenic microorganisms,
representing a viable candidate in the ongoing battle against resistant bacterial strains. One of the
compelling aspects of Cyclo(D-Ala-Val) is its structural properties that may allow it to function as an
antimicrobial agent. Its cyclic conformation is believed to contribute to its stability and interaction
capabilities with microbial membranes or essential proteins, which could result in the disruption of
pathogenic organisms' survival mechanisms.
The exploration of Cyclo(D-Ala-Val) within
antimicrobial research has several focal points. Firstly, its potential efficacy against both
Gram-positive and Gram-negative bacteria is significant. These two major classes of bacteria have
distinct cell wall compositions, and the ability of Cyclo(D-Ala-Val) to interact with or disrupt both
types could signify a broad-spectrum antimicrobial property. Researchers are particularly interested in
understanding how this cyclic peptide might permeate or disrupt bacterial membranes or possibly
interfere with essential bacterial protein functions.
Additionally, the use of Cyclo(D-Ala-Val)
in combating antibiotic-resistant strains is an emerging area of interest. The rise of antibiotic
resistance is a global healthcare challenge, necessitating new therapeutic options. Cyclo(D-Ala-Val)
offers an alternative mechanism whereby its non-traditional peptide structure and D-amino acid
incorporation could circumvent conventional bacterial resistance pathways. This capability makes
Cyclo(D-Ala-Val) a promising candidate for developing novel antimicrobial agents that can either
function independently or synergistically with existing antibiotics to enhance their effectiveness and
reduce the likelihood of resistance development.
Furthermore, beyond antibacterial activity,
Cyclo(D-Ala-Val) can be investigated for antifungal properties, which expands its potential
applicability across multiple categories of infectious agents. The likelihood of Cyclo(D-Ala-Val) or its
derivatives being developed into clinically usable antimicrobials will depend on ongoing studies that
evaluate its efficacy, safety profiles, and mechanism of action. Continuing research is crucial to
determine the full scope of its antimicrobial potential and the pathways through which it can be
optimized for therapeutic use.
What are the challenges in researching and developing
Cyclo(D-Ala-Val) for pharmaceutical applications?
Researching and developing Cyclo(D-Ala-Val) for
pharmaceutical applications presents several challenges, despite its potential benefits. The very
characteristics that make Cyclo(D-Ala-Val) attractive for development, such as its stability and unique
bioactivities, also present hurdles that must be overcome to translate lab-based findings into
clinically useful applications.
One significant challenge is the elucidation of its specific
mechanism of action. While initial studies may indicate broad antimicrobial, anticancer, or other
biological effects, understanding the exact molecular pathways through which Cyclo(D-Ala-Val) operates
is complex. This understanding is crucial for advancing the peptide through the drug development
pipeline, as it determines how the peptide can be optimized for potency, selectivity, and minimized
off-target effects. In uncontrolled environments like the human body, pinpointing the precise
interactions at the molecular level can be difficult due to the complex interplay of numerous biological
processes.
Another challenge is the potential for synthesis and scale-up difficulties. While
peptides can be synthesized using established methods, the process of scaling up production while
maintaining high purity and consistency is non-trivial. Large-scale synthesis must tackle issues such as
yield optimization, cost-effectiveness, and regulatory requirements. Ensuring that each batch is
consistent and free of contaminants is critical, especially when considering pharmaceutical applications
where even minor impurities can have significant impacts.
Regulatory hurdles also pose
challenges, as novel compounds like Cyclo(D-Ala-Val) typically fall under stringent scrutiny by
regulatory bodies. Comprehensive preclinical and clinical testing is necessary to demonstrate safety and
efficacy, which requires substantial time, financial investment, and support infrastructure. Navigating
the regulatory landscape involves detailed documentation and meeting specific criteria for drug
characterization, quality control, and clinical trial design.
Finally, there is the challenge of
market competition and positioning. Peptide-based therapies must compete with established small-molecule
drugs and newer treatments like biologics. To be viable in the marketplace, Cyclo(D-Ala-Val) must
demonstrate clear advantages over existing therapies, such as improved efficacy, fewer side effects, or
novel mechanisms of action that address unmet medical needs. The path from discovery to development
involves not only overcoming scientific hurdles but also strategic planning to ensure market entry and
acceptance.
How does the cyclic nature of Cyclo(D-Ala-Val) affect its biological activity
compared to linear peptides?
The cyclic nature of Cyclo(D-Ala-Val) imparts several distinct
characteristics that influence its biological activity compared to its linear peptide counterparts. A
fundamental advantage of the cyclic form is its enhanced structural stability. Linear peptides are prone
to conformational changes and are more susceptible to enzymatic degradation due to their free terminal
amino and carboxyl groups. In contrast, the cyclic nature of Cyclo(D-Ala-Val) provides a more rigid and
fixed three-dimensional structure, limiting the flexibility and making it more resistant to degradation
by peptidases and proteases commonly found in biological systems.
This stability is particularly
advantageous for maintaining biological activity once inside the body, where peptides face harsh
enzymatic environments. The resistance to enzymatic breakdown allows Cyclo(D-Ala-Val) to have a longer
half-life compared to linear peptides, potentially increasing its efficacy and reducing the frequency of
dosing in therapeutic applications. This characteristic is a significant consideration in drug design as
it can enhance patient compliance and therapeutic outcomes.
Moreover, the cyclic structure
confers the ability to engage in unique molecular interactions, which can influence binding affinity and
specificity to target molecules. This feature is due partly to the constrained conformation, which can
align functionally important side chains in a spatial arrangement optimal for binding to target proteins
or receptors. Such interactions may result in enhanced potency or selectivity, crucial for reducing
off-target effects and improving drug-like properties.
The cyclic nature also allows for
potential modulation of the peptide's activity through structural modifications. Chemists can design
variations by adding functional groups or substituting amino acids, which can further tailor its
interactions towards specific biological targets. Additionally, the incorporation of non-standard amino
acids, such as D-Ala, within the cyclic structure can introduce further resistance to metabolic
degradation and impact the interaction landscape.
Despite these advantages, the cyclic form's
rigidity can also be a limitation, as it may restrict the ability of Cyclo(D-Ala-Val) to fit into
binding pockets that require a degree of flexibility to adapt to various conformations. Thus, designing
cyclic peptides requires a balance between achieving necessary rigidity for stability and allowing for
enough flexibility to engage effectively with the intended target, which is a nuanced aspect of peptide
therapeutics design. Overall, the cyclic nature of Cyclo(D-Ala-Val) provides a beneficial framework for
biological activity, with implications for stability, selectivity, and interaction efficacy.
In
what ways can Cyclo(D-Ala-Val) be modified to enhance its therapeutic potential?
Cyclo(D-Ala-Val)
can undergo various modifications to enhance its therapeutic potential, optimizing its efficacy,
stability, and specificity for targeted applications. One of the primary avenues for modification
involves the incorporation of different amino acids or chemical groups to alter its pharmacokinetic and
pharmacodynamic properties. By substituting or adding amino acids, particularly D-amino acids or
non-natural amino acids, researchers can increase the peptide's resistance to enzymatic degradation and
extend its half-life in biological systems. These modifications are crucial for maintaining therapeutic
levels over extended periods, ultimately improving the drug's effectiveness and patient
adherence.
Another strategy is to conjugate Cyclo(D-Ala-Val) with other molecules to improve its
delivery and targeting capabilities. For instance, attaching polyethylene glycol (PEG) chains, a process
known as PEGylation, can enhance solubility and reduce immunogenicity, thereby improving the peptide's
bioavailability and circulatory stability. Additionally, linking Cyclo(D-Ala-Val) to targeting ligands,
such as antibodies or small molecules, can facilitate its delivery to specific cells or tissues,
enhancing its therapeutic index by concentrating the effect at the site of action while minimizing
systemic exposure and side effects.
Cyclo(D-Ala-Val) can also be modified to influence its
interaction with cellular membranes or specific receptors. Structurally, its cyclic nature can be
optimized by altering ring size or incorporating heterocycles to change conformation and binding
properties. This could enhance interaction with cell membranes for peptides intended to disrupt
microbial barriers or improve receptor binding affinity for peptides meant to modulate signaling
pathways. Such structural tuning is critical for achieving desired therapeutic outcomes, particularly in
cases where precise receptor engagement is necessary to exert the proper physiological
response.
Moreover, modifications can also be aimed at enhancing the peptide's ability to cross
biological barriers, such as the blood-brain barrier. This might involve optimizing lipophilicity or
adding specific sequences known to facilitate translocation across cell membranes. Achieving this can
expand the application range of Cyclo(D-Ala-Val) to include central nervous system disorders, broadening
its therapeutic impact.
Finally, Cyclo(D-Ala-Val) may benefit from prodrug strategies where it is
chemically modified to become active only within the target environment. These modifications allow the
parent peptide to remain inactive during systemic circulation, thereby minimizing possible side effects,
and then be activated by specific enzymes or conditions present at the target site. In essence, the
breadth of chemical biology techniques available for modifying Cyclo(D-Ala-Val) offers diverse pathways
for enhancing its therapeutic potential, making it a versatile template for drug design and development.