Synonym |
(D-Phe12,Nle21Ψ38)-CRF (12-41) |
Species |
human, rat |
Protein Accession |
P30988 |
Purity |
> 95% |
Endotoxin Level |
< 1.0 EU per μg |
Biological Activity |
Not specified |
Expression System |
Not specified |
Fusion Tag |
Not specified |
Predicted Molecular Mass |
Approximately 3545.1 Da |
Formulation |
Lyophilized from a 0.2 μm filtered solution in Acetonitrile and TFA |
Reconstitution |
Centrifuge vial before opening. Reconstitute in sterile distilled water to a concentration of
0.1-1.0 mg/ml. |
Storage & Stability |
Store lyophilized protein at -20°C. Aliquot reconstituted protein and store at -20°C to -80°C.
|
FAQ
What is (D-Phe12,Nle21-38)-CRF (12-41) used for in scientific research?
(D-Phe12,Nle21-38)-CRF
(12-41) is a peptide that serves as a corticotropin-releasing factor (CRF) antagonist, extensively used
in scientific research to study its effects on the hypothalamic-pituitary-adrenal (HPA) axis and its
broader implications on stress-related physiological responses. In both humans and rats, CRF plays a
crucial role in the stress response by stimulating the release of adrenocorticotropic hormone (ACTH),
which subsequently influences cortisol release—a primary stress hormone. By inhibiting the actions of
endogenous CRF, (D-Phe12,Nle21-38)-CRF (12-41) helps researchers better understand the intricate
mechanisms of stress response and anxiety disorders. One major area of interest in research utilizing
this peptide revolves around its potential therapeutic applications. Researchers investigate whether
modulating the HPA axis via CRF antagonists can alleviate symptoms of chronic stress and associated
disorders such as anxiety and depression. This peptide provides a valuable tool for dissecting the CRF
receptor-mediated pathways involved in these conditions, offering insight into potential new treatment
strategies. Furthermore, (D-Phe12,Nle21-38)-CRF (12-41) is pivotal in the exploration of stress-related
eating disorders and addiction. Stress has been implicated as a key factor that exacerbates addictive
behaviors and eating disorders, and CRF plays a central role in mediating these effects. The blockade of
CRF receptors helps to elucidate the potential neurobiological links between stress hormones and
pathological states, thereby aiding the development of targeted interventions. In addition to these
mental and behavioral disorders, (D-Phe12,Nle21-38)-CRF (12-41) is used to explore its effects on immune
function, as stress hormones are known to have significant impacts on immune system regulation. Stress
can suppress immune function, and by understanding the specific pathways through which CRF influences
immune responses, researchers aim to address stress-related immune issues. In sum,
(D-Phe12,Nle21-38)-CRF (12-41) is an invaluable research tool in elucidating the relationships between
stress physiology, CRF receptor activity, and a variety of physical and psychological health
outcomes.
How does (D-Phe12,Nle21-38)-CRF (12-41) interact with CRF
receptors?
(D-Phe12,Nle21-38)-CRF (12-41) acts as an antagonist to corticotropin-releasing factor
(CRF) receptors, primarily targeting CRF type 1 receptors, also known as CRF1 receptors. CRF receptors
are part of the G protein-coupled receptor family, which play pivotal roles in mediating the stress
response initiated by endogenous CRF. When CRF binds to these receptors, it triggers a series of
intracellular signaling cascades that lead to the activation of the hypothalamic-pituitary-adrenal (HPA)
axis, culminating in the release of adrenocorticotropic hormone (ACTH) and subsequent cortisol
production. This cascade is integral to the body's physiological response to stress.
(D-Phe12,Nle21-38)-CRF (12-41) interferes with this process by competitively binding to the CRF1
receptors, effectively blocking CRF from activating its receptor-mediated signaling pathways. This
blockade is beneficial in examining the downstream effects that would otherwise be triggered by CRF
activation. By inhibiting CRF-induced receptor activity, scientists can study the peptide's effects on
various physiological and behavioral outcomes, particularly those influenced by stress, such as
anxiety-like behavior, mood disorders, and addictive behaviors. The peptide's interaction with the CRF
receptors is characterized by its ability to attenuate the release of ACTH, thereby reducing cortisol
levels and modulating the stress response. This antagonistic action is particularly useful in research
focused on understanding the maladaptive responses to chronic stress and the development of
stress-related disorders. By blocking CRF receptors, researchers can delineate the specific pathways
through which CRF contributes to pathologies such as anxiety, depression, and substance use disorders.
Furthermore, (D-Phe12,Nle21-38)-CRF (12-41)'s interaction with CRF receptors extends to its impact on
neuroinflammation and immune responses, highlighting its role in broader physiological processes
influenced by stress-related peptides. The peptide's ability to selectively inhibit CRF1 receptor
activity without interacting with other receptor subtypes provides a nuanced approach to studying
CRF-related networks. Last but not least, the specificity of (D-Phe12,Nle21-38)-CRF (12-41) for CRF1
receptors makes it an excellent research tool for studying receptor-specific mechanisms, allowing for
the investigation of targeted drug therapies that could block pathological CRF signaling without
affecting other neuroendocrine functions.
What are some potential applications of
(D-Phe12,Nle21-38)-CRF (12-41) in biomedical research?
(D-Phe12,Nle21-38)-CRF (12-41) is employed
in biomedical research to explore its diverse potential applications across various domains,
particularly focusing on stress-related disorders, psychiatric conditions, and immune system regulation.
The primary application revolves around its capacity as a CRF antagonist to modulate the
hypothalamic-pituitary-adrenal (HPA) axis, providing profound insights into stress physiology and the
potential for therapeutic intervention. One significant area of interest is its use in studying and
potentially treating anxiety and depression. Researchers gain insight into the development and
progression of these conditions by using (D-Phe12,Nle21-38)-CRF (12-41) to block CRF receptors and
analyze changes in stress hormone levels and corresponding behaviors. This peptide helps to elucidate
the relationship between chronic stress exposure, altered HPA axis function, and the onset of mood
disorders. Understanding these mechanisms lays the groundwork for developing CRF-targeted
pharmaceuticals that can alleviate symptoms of anxiety and depression by regulating the body's response
to stress. Another promising application is in addiction research, particularly concerning the role of
stress in exacerbating substance use disorders. By antagonizing CRF receptors, (D-Phe12,Nle21-38)-CRF
(12-41) helps to clarify how stress contributes to addiction maintenance and relapse. This research
further explores how CRF-driven stress pathways can be manipulated to develop effective treatments for
preventing relapse and reducing cravings. In the context of immune function, (D-Phe12,Nle21-38)-CRF
(12-41) serves as an essential tool for understanding stress-related immunosuppression. Given the
considerable influence stress hormones have over immune system processes, studies utilizing CRF
antagonists provide valuable data on how stress management can enhance immune responses. This research
opens the door for new treatments targeting autoimmune disorders and inflammatory diseases, addressing
the detrimental effects of chronic stress on immune health. In addition to these areas,
(D-Phe12,Nle21-38)-CRF (12-41) is relevant in neurobiology and psychiatric studies, where the focus is
on the broader implications of CRF signaling in conditions such as post-traumatic stress disorder (PTSD)
and eating disorders. By using the peptide to delineate CRF's role in these complex conditions,
scientists aim to delineate the molecular pathways that contribute to their pathology and explore
potential novel therapeutic targets. Finally, the specific targeting of CRF receptors using
(D-Phe12,Nle21-38)-CRF (12-41) holds promise in personalized medicine, offering strategies for tailoring
interventions based on individual stress response profiles.
How is (D-Phe12,Nle21-38)-CRF (12-41)
synthesized for research purposes?
The synthesis of (D-Phe12,Nle21-38)-CRF (12-41) for research
purposes involves a process called solid-phase peptide synthesis (SPPS), a widely utilized method for
creating synthetic peptides with high specificity and purity. Typically, SPPS is conducted on a polymer
resin support, where the growing peptide chain is anchored through its carboxyl end, allowing sequential
addition of protected amino acids to build the desired peptide sequence. The synthesis begins with the
coupling of the first amino acid, protected by a temporary protecting group at its amino end, to the
resin. Following this, the temporary protecting group is removed, revealing a free amino group ready for
the next coupling reaction with the subsequent amino acid. This coupling-deprotection cycle is repeated
for each amino acid in the sequence of (D-Phe12,Nle21-38)-CRF (12-41), ensuring specific peptide bond
formation between each residue. (D-Phe12,Nle21-38)-CRF (12-41) includes modified amino acids such as
D-Phenylalanine (D-Phe) and Norleucine (Nle), which are incorporated into the sequence as needed. These
non-standard amino acids can be synthesized beforehand or purchased as building blocks, equipped with
compatible protecting groups that allow them to integrate seamlessly with SPPS processes. The specific
sequence is assembled step by step to ensure that the integrity of the peptide structure is maintained.
After synthesis, the peptide-resin conjugate undergoes a cleavage step, in which the finished peptide is
detached from the resin through a process that simultaneously removes side-chain protecting groups. This
cleavage is typically executed using a strong acid, such as trifluoroacetic acid (TFA), which
effectively cleaves the bonds attaching the peptide to the resin, leaving the linear peptide chain with
a free carboxyl terminal end. Post-cleavage, the crude peptide is subjected to purification processes,
most often performed using high-performance liquid chromatography (HPLC). HPLC facilitates the
separation of the target peptide from any impurities, such as incomplete peptide chains or resin-related
by-products. The purified peptide's identity and purity are validated through techniques such as mass
spectrometry and analytical HPLC, ensuring that the final product matches the theoretical molecular
weight and composition of (D-Phe12,Nle21-38)-CRF (12-41). The rigor of these synthesis steps ensures
that the synthesized peptide is of high purity and biological activity, critical for accurate and
reliable research studies. Furthermore, when synthesized accurately, (D-Phe12,Nle21-38)-CRF (12-41)
serves as a potent tool for researchers to explore valuable insights into CRF receptor-mediated pathways
and their implications in health and disease.
Can (D-Phe12,Nle21-38)-CRF (12-41) be used in
clinical trials or as a therapeutic drug?
Currently, (D-Phe12,Nle21-38)-CRF (12-41) is
predominantly utilized in preclinical research settings due to its role as a corticotropin-releasing
factor (CRF) antagonist, helping researchers investigate stress-related pathways and potential
therapeutic targets for stress-induced conditions. The transition from research-focused applications to
clinical trials or therapeutic use requires several rigorous validation steps and regulatory approvals,
which this peptide has not yet addressed to enable its use as a therapeutic entity. While
(D-Phe12,Nle21-38)-CRF (12-41) has shown promising results in modulating the stress response and
influencing physiological and behavioral responses in animal models, its application as a therapeutic
drug in humans requires comprehensive clinical evaluation. Before any peptide or compound can be
considered for clinical trials, it must undergo extensive preclinical testing to evaluate its safety,
efficacy, and pharmacokinetics. These trials involve in vitro studies and animal model assessments to
identify potential therapeutic windows and any toxicological concerns. Once a peptide passes these
preclinical evaluations and demonstrates a favorable benefit-risk profile, it may proceed through
various phases of human clinical trials. These trials are characterized by a phased approach: Phase I
focuses on safety and tolerability in a small group of healthy volunteers; Phase II evaluates efficacy
and optimal dosing in a larger patient population; and Phase III involves extensive testing across
diverse subject populations to validate clinical effectiveness and monitor adverse reactions. Despite
its extensive use in understanding CRF-related mechanisms and potential implications for human health,
(D-Phe12,Nle21-38)-CRF (12-41) has not yet transitioned through these clinical trial phases to be
approved for therapeutic use in humans. Regulatory agencies, such as the U.S. Food and Drug
Administration (FDA) or the European Medicines Agency (EMA), have established stringent guidelines and
requirements that any potential therapeutic candidate must meet before gaining approval for use in human
populations. Moreover, the ethical and practical challenges of translating findings from animal models,
including differing physiological pathways between species, underscore the additional complexities that
accompany turning research peptides into safe and effective drugs. Researchers remain optimistic about
the future potential of (D-Phe12,Nle21-38)-CRF (12-41) or similar compounds as therapeutic candidates,
given their substantial preclinical data advocating their benefits in modulating the HPA axis and stress
response. However, without adequate clinical data, this peptide continues to be a subject of interest
primarily for experimental research endeavors rather than a clinical intervention.
What is the
importance of using (D-Phe12,Nle21-38)-CRF (12-41) in stress-related
research?
(D-Phe12,Nle21-38)-CRF (12-41) holds a pivotal role in stress-related research due to
its ability to effectively antagonize corticotropin-releasing factor (CRF) receptors, primarily CRF type
1 receptors (CRF1). The relevance of using this peptide stems from our growing understanding of the
complexity and impact of the body's stress response, largely orchestrated by the
hypothalamic-pituitary-adrenal (HPA) axis, where CRF is a central component. This peptide offers a
mechanism to delineate the specific pathways through which CRF contributes to stress physiology and
related disorders, facilitating a deeper understanding of stress dynamics. The importance of
(D-Phe12,Nle21-38)-CRF (12-41) is underscored by its role in helping researchers explore the
neurobiological and psychological effects of stress. Stress transduces its effects via CRF and
subsequent activation of the HPA axis, leading to the release of adrenocorticotropic hormone (ACTH) and
thus cortisol, a well-known stress hormone. By blocking CRF receptors, (D-Phe12,Nle21-38)-CRF (12-41)
offers a powerful tool to help researchers investigate how alterations in the stress response can become
pathogenic, contributing to conditions like anxiety, depression, and post-traumatic stress disorder
(PTSD). It allows scientists to study how stress-related neurochemical pathways affect brain regions
involved in mood regulation, cognition, and memory. This antagonist also has significant utility in
exploring the links between chronic stress and its systemic effects, such as those on eating behavior,
addiction, and immune function. Chronic stress has implications for eating disorders and substance
abuse, through reinforcing maladaptive behaviors that (D-Phe12,Nle21-38)-CRF (12-41) can help elucidate
by disrupting CRF's impact. Furthermore, by inhibiting CRF's action, it becomes possible to study the
effects of stress on immune functioning and inflammation, thereby reinforcing the connection between
physical health and neuroendocrine stress pathways. Additionally, the quest to develop novel
pharmacological interventions for stress-related disorders has been accelerated by research involving
(D-Phe12,Nle21-38)-CRF (12-41). Understanding its mechanistic impact on stress pathways informs the
development of CRF receptor antagonists as promising therapeutic leads potentially suited for
ameliorating stress-induced psychopathologies and enhancing resilience against stressors. As a result,
this peptide has become integral for both fundamental research aimed at understanding stress hormonal
regulation and applied research looking to translate findings into viable clinical interventions.
Expanding research using (D-Phe12,Nle21-38)-CRF (12-41) continues to address not only the biochemical
pathways mediating stress but also involves examining how genetic, environmental, and biological factors
converge to influence stress susceptibility and resilience. The insights generated are invaluable in the
pursuit of effective treatments and preventative strategies to combat the detrimental health outcomes
associated with chronic stress exposure.
What are the structural characteristics of
(D-Phe12,Nle21-38)-CRF (12-41) that distinguish it from other CRF
antagonists?
(D-Phe12,Nle21-38)-CRF (12-41) is defined by its distinguished structural
characteristics which provide both specificity and effectiveness as a corticotropin-releasing factor
(CRF) antagonist. This peptide features modifications that enhance its utility in research, particularly
in its interaction with CRF1 receptors. Notably, its structural uniqueness involves the incorporation of
non-standard amino acids such as D-Phenylalanine (D-Phe) and Norleucine (Nle), which confer enhanced
receptor binding affinity and resistance to enzymatic degradation. These modifications are designed to
improve the peptide's pharmacological properties, particularly its bioavailability and inhibitory
potential. The sequence alterations, for instance, the incorporation of D-Phe, facilitate distinct
binding properties that differ from natural peptides, contributing to the enhanced receptor selectivity
that is pivotal in research contexts. The presence of D-amino acids, like D-Phe, generally decreases the
peptide's susceptibility to proteolytic enzymes that would otherwise degrade natural L-form peptides
rapidly, thereby providing sustained activity that is hugely beneficial in experimental settings. Nle,
another distinctive amino acid in (D-Phe12,Nle21-38)-CRF (12-41), substitutes the conventional
methionine, diminishing the likelihood of undesired oxidative reactions which might occur in the
presence of methionine under experimental conditions. Such modifications enhance the peptide's
stability, allowing it to maintain its functional integrity under diverse assay conditions, which is
crucial for acquiring consistent and reliable experimental data. In terms of secondary structure, these
alterations can also influence the conformation of the peptide, potentially affecting its binding
orientation to the CRF1 receptor. This binding efficiency and specificity are crucial for effectively
blocking CRF-mediated signal transduction, allowing researchers to better understand the implications of
CRF receptor inhibition in stress-related pathways. Additionally, (D-Phe12,Nle21-38)-CRF (12-41)
distinguishes itself from other CRF antagonists due to its truncated form, spanning the 12th to the 41st
residues of the native CRF peptide. By retaining specific functional motifs essential for receptor
binding and activity blockade, while excluding portions irrelevant to its antagonistic function, the
peptide achieves targeted inhibitory action with minimized off-target effects. This truncation strategy
aligns with the need for higher selectivity and reduced risk of cross-reactivity, potentially
influencing other cellular systems. These structural characteristics collectively render
(D-Phe12,Nle21-38)-CRF (12-41) a salient tool in scientific research, providing clarity and precision in
dissecting CRF's role in the body’s stress responses. Through specificity, stability, and resistance to
proteolytic breakdown, researchers leverage these attributes to advance our comprehension of CRF-related
pathways and their therapeutic implications.