Synonym |
(Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide (human) |
Species |
Human |
Protein Accession |
P25116 |
Purity |
≥ 95% |
Endotoxin Level |
< 1.0 EU per µg |
Biological Activity |
N/A |
Expression System |
Chemical synthesis |
Fusion Tag |
None |
Predicted Molecular Mass |
834.0 Da |
Formulation |
Lyophilized from a 0.2 μm filtered solution of 20% acetonitrile and 0.1% TFA |
Reconstitution |
It is recommended to reconstitute the lyophilized product with sterile water at a concentration
of 0.1 mg/ml, which can then be further diluted in aqueous solutions as needed. |
Storage & Stability |
Store lyophilized product at -20°C. Store reconstituted product at -80°C. |
FAQ
What is (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide (human) used for in research?
(Phe1,Ser2,Tyr6)-PAR-1
(1-6) amide (human) is predominantly used in research as a tool for studying proteinase-activated
receptor 1 (PAR-1) functions and pathways. PAR-1 is a crucial receptor in the human system, intricately
involved in numerous physiological and pathophysiological processes, including thrombosis, inflammation,
and cancer progression. Researchers are particularly interested in how this receptor is activated by
thrombin, a key protein involved in blood coagulation. The peptide form, (Phe1,Ser2,Tyr6)-PAR-1 (1-6)
amide, allows investigators to specifically stimulate or inhibit PAR-1 to dissect its roles across
various cellular and physiological contexts. Furthermore, because PAR-1 is expressed in multiple cell
types and tissues, such as platelets, endothelial cells, and neurons, the compound serves as a versatile
probe for examining diverse biological hypotheses. This multi-faceted application underscores the
compound's significant utility in understanding cell signaling, receptor-ligand interactions, and the
broader frameworks of cardiovascular and cerebral pathologies.
Moreover, research on this
receptor has illuminated its function in restenosis — the re-narrowing of blood vessels after
angioplasty. This becomes critically relevant given the ever-increasing global incidence of
cardiovascular diseases. By employing (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide, scientists can better
understand vascular smooth muscle cell proliferation, platelet aggregation, and the signaling pathways
driving these phenomena. These insights equip researchers with the capacity to strategize innovative
therapeutic interventions that target these mechanisms more precisely. Additionally, studies have
explored PAR-1’s interactions with various G-proteins, leading to the activation of downstream effectors
that influence cellular responses such as migration, apoptosis, and cell survival. Given the peptide's
role in elucidating these pathways, it also represents a pivotal component for developing potential drug
candidates that may ameliorate clinical conditions ranging from arterial thrombosis to certain cancer
subtypes. Overall, the utilization of (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide continues to advance our
understanding of complex biological processes and holds the promise for impactful translational research
applications in the near future.
How does (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide (human) interact
with PAR-1 receptors?
(Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide (human) interacts with PAR-1 receptors
by mimicking the natural activation sequence of the receptor. PAR-1, as a member of a family of
G-protein-coupled receptors, is activated by cleavage through proteolytic enzymes such as thrombin. This
cleavage exposes a new N-terminus, which acts as a tethered ligand, binding intramolecularly to initiate
receptor activation. The (Phe1,Ser2,Tyr6) sequence constitutes part of this tethered ligand, meaning
that the peptide serves as a mimic that can directly activate the receptor independent of proteolytic
cleavage.
Upon interaction with PAR-1 receptors, (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide triggers the
intracellular signaling cascades typically associated with receptor activation. This allows researchers
to observe physiological outcomes in various cellular models, consonant with those occurring during
normal PAR-1 activation. Interactions often lead to the mobilization of intracellular calcium, peptide
activation of transformation pathways, and the harboring of selective downstream effectors. Studies have
identified that these signaling pathways frequently involve proteins such as mitogen-activated protein
kinases (MAPKs), phosphoinositide 3-kinase (PI3K), and Rho-type GTPases.
The peptide's
interaction capabilities extend to probing receptor affinity and functionality. Using
(Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide, researchers can measure binding kinetics and receptor occupancy
across varying conditions, which is crucial in drug discovery applications where antagonist or agonist
characterization might be pivotal. The interactions might also assist in unveiling the receptor's
structural nuances, providing key insights into molecular dynamics and potential allosteric sites. For
experimental applications, the compound can be employed at varying concentrations to produce either full
or partial activation of PAR-1, offering a degree of control and specificity that is highly valuable in
mechanistic studies.
Furthermore, by analyzing the interaction of this peptide with PAR-1,
scientific endeavors can pivotalize around the roles of this receptor in diverse biological systems.
Research leveraging its mechanisms addresses pivotal questions about coagulation and thrombosis,
immunological responses, cancer metastasis, and development of receptor-targeted therapeutic agents.
Therefore, the interaction between (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide and PAR-1 receptors becomes a
focal point for a diverse array of exploratory and translational research studies, parsing out both
fundamental scientific insights and avenues for clinical innovation.
What methods and models are
used in researching (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide (human)?
To research
(Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide, scientists employ a multitude of methods and models that coalesce to
paint a comprehensive picture of its biological implications and mechanisms of action. In vitro,
cell-based assays represent a foundational approach, leveraging cultured cell lines that express PAR-1
to observe direct effects of the peptide. With varying cell types including endothelial cells, smooth
muscle cells, and neurons, these assays help delineate PAR-1-specific responses at subcellular,
cellular, and multicellular levels.
Functional assays, such as cell migration and adhesion assays
or calcium ion flux assays, aid in measuring tangible outcomes of PAR-1 activation by the peptide. These
quantifiable endpoints allow for the assessment of cellular behaviors influenced by the compound's
activity. Technologies such as fluorescence microscopy and flow cytometry are frequently adapted in
these experiments to offer insights into changes in cell morphology and signaling protein activation
patterns. Western blotting and quantitative PCR further enable exploration of downstream signaling
effectors, providing details on changes in protein expression and gene transcription
levels.
Further, computational modeling and molecular dynamics simulations are leveraged to study
the interaction between (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide and PAR-1 receptors. These approaches allow
investigation into the structural dynamics of receptor-peptide complexes, giving valuable insights into
binding sites, affinity, and potential allosteric modulations. Such models inform the development of
novel pharmacological agents by elucidating the structural determinants of receptor
activation.
Animal models serve as a bridge between cellular data and human physiology, allowing
researchers to validate findings from in vitro studies in a living system. Rodent models, for instance,
are invaluable for studying the pathophysiological impact of PAR-1 activation in systems like the
cardiovascular or nervous systems, where the peptide's effects on disease models can be closely
observed. In vivo imaging techniques such as fluorescence or bioluminescence imaging may be used to
track the biodistribution and pharmacodynamics of the compound.
Finally, collaborative efforts
integrating omics technologies into PAR-1 research provide burgeoning opportunities for holistic
understanding. Proteomics and transcriptomics enable the capture of vast molecular changes following
peptide treatment, identifying cellular pathways and networks influenced by its interaction with PAR-1.
Data analytics and bioinformatics become essential in interpreting such complex datasets, leading to
strategized hypotheses for further exploration.
With such a diverse toolkit, the research methods
and models applied to (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide study are expansive, setting the stage for
innovative discoveries spanning cell biology, therapeutic research, and biomolecular engineering.
Through these efforts, scientists can cultivate an intricate understanding of the peptide's role in
human biology, pushing the frontier of targeted receptor-based therapies forward.
What are the
potential therapeutic applications of (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide in
medicine?
(Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide holds considerable potential in the therapeutic
landscape, primarily due to its targeted modulation of the PAR-1 receptor, involved in several critical
physiological and pathophysiological processes. The peptide's utility primarily extends to
cardiovascular interventions, given PAR-1's central role in thrombin signaling, platelet function, and
maintaining vascular integrity. By modulating this receptor, researchers aim to develop novel
therapeutic strategies to combat arterial thrombosis, myocardial infarction, and other thromboembolic
diseases. Of particular interest is the development of selective PAR-1 antagonists and agonists that can
finely tune hemostatic processes without the risk profiles presented by broader anticoagulant
therapies.
In oncology, the regulatory circuit involving PAR-1 in tumor progression and
metastasis opens another promising avenue. Experimental paradigms suggest that (Phe1,Ser2,Tyr6)-PAR-1
(1-6) amide might modify tumor microenvironment interactions, particularly influencing cancer cell
proliferation, migration, and invasion. Through these interactions, there is potential to inhibit cancer
metastasis, providing a therapeutic edge alongside established oncological regimens. The targeted nature
of PAR-1 modulation could offer a solution with fewer off-target effects compared to current systemic
chemotherapy strategies.
Furthermore, the compound's implications in inflammation and immune
function are a tantalizing field for therapeutic exploration. The roles of PAR-1 in mediating
inflammatory responses and its involvement in immune cell signaling are coming to the forefront,
suggesting applications in autoimmune diseases, sepsis, and inflammatory bowel diseases. The ability to
mediate receptor-specific pathways could allow selective dampening of inflammation, circumventing the
generalized immunosuppression effects seen with traditional corticosteroids or other
immunomodulators.
Neurological research also considers PAR-1 as a prospective target due to its
influence on neural signaling pathways. There may be therapeutic potential in neuroprotective
interventions, which address disorders involving neural injury or degenerative diseases such as
Alzheimer's or Parkinson's. By modulating the signaling cascades originating from PAR-1 activation, it's
conceivable to mitigate neuronal damage and promote neurogenesis or synaptic plasticity.
Lastly,
regenerative medicine may benefit from the insights provided by (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide,
particularly in tissue repair and wound healing processes where PAR-1 plays a significant role.
Controlled activation of this pathway could accelerate tissue regeneration, presenting opportunities to
improve outcomes in patients suffering from chronic wounds or surgical recovery challenges.
These
prospective therapeutic applications demonstrate the compound's versatility in addressing a breadth of
clinical challenges. Each area underscores the need for continued interdisciplinary efforts to translate
research findings into effective clinical interventions, aiming to revolutionize precise, receptor-based
therapeutic approaches for myriad health conditions.
What are the challenges in developing
therapies based on (Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide?
Developing therapies based on
(Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide involves addressing several significant challenges that impact the
feasibility and effectiveness of translating research findings into clinical interventions. One of the
primary challenges lies in the intricate molecular complexity of PAR-1 signaling. The multifaceted
interactions that PAR-1 has with various G-proteins and the differential outcomes based on cellular
context pose difficulties in predicting and controlling specific therapeutic effects. The receptor's
involvement in diverse physiological processes demands pinpoint precision in targeting specific pathways
while avoiding unintended side effects that might arise from broad-spectrum
modulation.
Additionally, achieving selective modulation of PAR-1 without impacting PARs in other
pathways remains a significant obstacle. The human body expresses multiple proteinase-activated
receptors, and PAR-1's close relationships with these other receptors often lead to compensatory
signaling or unintended cross-reactivity, which must be meticulously navigated in drug development. The
quest to develop agents that can discriminate between related receptors with high specificity, while
still exerting the desired therapeutic effect, involves sophisticated medicinal chemistry and a deep
understanding of receptor dynamics.
The bioavailability and pharmacokinetics of potential therapy
candidates are also significant hurdles. Peptides like (Phe1,Ser2,Tyr6)-PAR-1 (1-6) often suffer from
poor stability and bioavailability in physiological conditions due to rapid degradation by proteolytic
enzymes. Modifications to enhance stability without compromising functional efficacy are imperative;
however, they introduce additional layers of complexity to the drug design process. Drug delivery
systems, too, require innovation to ensure that these therapies reach target tissues effectively, which
often necessitates novel delivery vectors or formulation strategies.
Moreover, the risk of
adverse effects associated with off-target interactions or unexpected immune responses presents a
further challenge. Immune-modulating or anti-thrombotic therapies carry the risk of triggering adverse
immune responses or bleeding complications, respectively. Thorough preclinical testing frameworks are
necessary for evaluating safety and efficacy, which can be resource-intensive and time-consuming.
Balancing efficacy with safety profiles, while gaining regulatory approval, reflects one of the most
significant challenges development teams face, necessitating rigorous data collection and
analysis.
Economics and logistics, including the high costs associated with research and
development, production scalability, and market accessibility further complicate the path towards
therapeutic application. Funding scientific research through to clinical trials demands substantial
investment, with the inherent risk that compounds may fail during late-stage testing. Establishing
cost-effective manufacturing processes and ensuring consistency in large-scale production are ongoing
concerns that require innovative solutions to make eventual therapies widely accessible to
patients.
Addressing these formidable challenges requires a multi-disciplinary approach,
combining advanced drug discovery techniques, robust clinical trial designs, and strategic
collaborations between academia, industry, and regulatory bodies. With strategic investments of time and
resources, and the leveraging of cutting-edge technologies such as computational modeling, AI-driven
drug design, and personalized medicine approaches, research can mitigate these obstacles, ensuring that
(Phe1,Ser2,Tyr6)-PAR-1 (1-6) amide holds the promise to transform specific therapeutic sectors.