Synonym |
EGFR |
Species |
Human |
Protein Accession |
P00533 |
Purity |
Greater than 95% as determined by SDS-PAGE |
Endotoxin Level |
< 1.0 EU per 1 μg of protein |
Biological Activity |
The ED50 for this effect is typically 0.15-0.6 ng/mL |
Expression System |
Human cells |
Fusion Tag |
His tag at N-terminus |
Predicted Molecular Mass |
74.9 kDa |
Formulation |
Lyophilized from a 0.2 μm filtered solution of 20mM PB, 150mM NaCl, pH 7.4 |
Reconstitution |
Centrifuge the vial prior to opening. Reconstitute in PBS to a concentration of 0.1 mg/mL |
Storage & Stability |
Store at -20°C. Please avoid freeze-thaw cycles. |
FAQ
What is the significance of EGF Receptor (985-996) in human biology?
The EGF Receptor (EGFR) is a
transmembrane protein that plays a crucial role in cellular processes such as cell growth,
differentiation, and repair. EGFR is part of the receptor tyrosine kinase (RTK) family and is activated
by binding epidermal growth factor (EGF) ligands. Upon ligand binding, EGFR undergoes a conformational
change that results in dimerization and autophosphorylation on specific tyrosine residues within its
cytoplasmic domain. This autophosphorylation creates docking sites for various downstream signaling
molecules, thereby initiating multiple signaling cascades, including the MAPK, AKT, and JAK/STAT
pathways, which regulate diverse cellular outcomes. The peptide segment EGF Receptor (985-996) is
located in the C-terminal tail of the receptor and is important for signal transduction and interaction
with downstream effectors. Aberrant EGFR signaling, such as overexpression or mutations, is implicated
in the pathogenesis of multiple cancers, notably non-small cell lung cancer, glioblastoma, and
colorectal cancer. Therefore, EGFR is a significant molecular target for anti-cancer therapies,
including monoclonal antibodies like cetuximab and tyrosine kinase inhibitors like gefitinib and
erlotinib, which have been developed to disrupt aberrant EGFR signaling. Understanding the structure,
function, and regulatory mechanisms of the EGF Receptor, particularly critical regions like (985-996),
is essential in cancer research and therapeutics and contributes to the broader understanding of cell
signaling and homeostasis.
How does EGF Receptor (985-996) contribute to disease
processes?
EGF Receptor (985-996) contributes to disease processes primarily through its
involvement in the aberrant signaling pathways responsible for cell proliferation and survival. In many
cancers, EGFR is either overexpressed, mutated, or has altered activation status leading to uncontrolled
cell growth and division. The segment between amino acids 985 and 996 in the EGFR is critical as it
contains residues involved in the interaction with downstream signaling molecules. This region affects
how signaling proteins bind to the receptor and propagate signals, such as those regulating cell cycle
progression and apoptosis. In cancer cells, for example, this unchecked signaling leads to enhanced
survival and evasion of normal cellular regulatory mechanisms, contributing to tumor initiation and
progression. Mutations within the EGFR, including those affecting (985-996), can lead to constitutive
activation of the receptor even in the absence of ligand, perpetuating oncogenic signaling. Moreover,
this segment can influence drug resistance, as mutations or modifications can alter how cancer therapies
target the EGFR. Understanding this region's role in modulating receptor-ligand interactions and
downstream signaling is also critical in understanding diseases like glioblastoma and head and neck
cancers, where classic EGFR mutation patterns have been identified. It aids in developing new
therapeutic strategies to effectively target these altered pathways, providing improved insight into how
resistance to existing EGFR inhibitors might develop and how such resistance might be overcome. The
study of EGF Receptor (985-996) thus remains a focal point of research aimed at decoding the molecular
underpinnings of EGFR-induced pathology.
What research techniques are commonly used to study EGF
Receptor (985-996)?
Research techniques used to study the EGF Receptor (985-996) span both
traditional biochemical methods and advanced molecular biology tools. Initially, standard techniques
such as site-directed mutagenesis allow researchers to create specific modifications within this region
to study the functional consequences on receptor activity and downstream signaling. These mutations are
often studied in the context of recombinant protein expression systems, allowing for expressing and
purifying modified EGFR to analyze biochemical properties under controlled laboratory conditions.
Structural biology techniques, including X-ray crystallography and nuclear magnetic resonance (NMR)
spectroscopy, provide detailed insights into the molecular conformation of EGFR, including regions like
(985-996). These techniques are crucial for visualizing how changes in the amino acid sequence may
impact the protein structure and function. In studying protein interactions, technologies such as
co-immunoprecipitation, Förster resonance energy transfer (FRET), and surface plasmon resonance (SPR)
can elucidate how mutations or modifications in the 985-996 segment affect binding to downstream
signaling proteins. Furthermore, mass spectrometry is an essential tool for identifying
post-translational modifications in this region, which can regulate receptor activity. Computational
modeling and molecular dynamics simulations also provide a complementary approach by predicting the
effects of alterations in the 985-996 segment on EGFR dynamics and its interaction landscape. Finally,
cell-based assays using CRISPR-Cas9 gene editing further allow for precise manipulation of the
endogenous EGFR gene, including regions like 985-996, in cell lines to study functional outcomes in a
native cellular environment. These diverse and multifaceted approaches are integrated to produce a
comprehensive understanding of how the EGF Receptor, especially critical regions such as (985-996),
functions within normal physiology and disease states.
Can EGF Receptor (985-996) serve as a
therapeutic target?
The EGF Receptor, particularly integral regions like (985-996), is a
promising therapeutic target for multiple reasons. Firstly, the receptor is pivotal in driving the
signaling cascades that lead to uncontrolled cell proliferation, survival, and metastasis in many
cancers, such as non-small cell lung cancer and glioblastoma. Targeting the EGF Receptor, and
specifically regions that are critical for its activity and interactions, can inhibit these pathological
signaling pathways. There are already numerous therapies, including small molecule tyrosine kinase
inhibitors and monoclonal antibodies, that target the EGFR, illustrating its importance as a therapeutic
focus. These therapies aim to block the receptor's ligand-binding domain, prevent downstream signaling,
or induce receptor internalization and degradation. Given the importance of the 985-996 region in these
processes, understanding its role could potentially lead to more precise interventions. Any mutations or
post-translational modifications in this region that affect drug interaction or effectiveness could
become useful biomarkers for guiding treatment regimens. Furthermore, therapies could be designed to
mimic or inhibit specific protein-protein interactions crucial for EGFR's pathological roles. As
resistance to current EGFR therapies can arise, often involving alterations within or affecting the
segment (985-996), new drugs targeting this region may help overcome resistance. Indeed, the protein
structure insights of this region can lead to designing more effective inhibitors that either covalently
bind or allosterically modulate its activity. Over the years, the understanding of EGFR roles and its
crucial regions will likely support the development of next-generation therapeutic strategies that are
more selective and have fewer side effects by specifically targeting receptor variants or modifications
often seen in cancers. This makes EGF Receptor (985-996) an invaluable focus in the continuum of cancer
research and drug development.
What are the challenges in targeting EGF Receptor (985-996) for
therapy?
Targeting the EGF Receptor, and specifically the segment (985-996), poses several
challenges in therapeutic development. One of the foremost difficulties is the complexity of EGFR
signaling itself. EGFR is not a lone actor; it partakes in a highly intricate network of signaling
pathways where it interacts with different molecules in precise temporal and spatial manners. Any
inaccuracies in targeting such a network can lead to off-target effects or insufficient therapeutic
responses. Complexity is further compounded by EGFR's ability to form heterodimers with other ErbB
family members, modulating different downstream signaling pathways, which can influence therapeutic
outcomes. The plasticity of EGFR signaling in cancer progression introduces another layer of challenge.
Tumors often become resistant to therapy through secondary mutations or by adopting compensatory
pathways that bypass EGFR signaling altogether. Specifically, alterations in the 985-996 region could
modulate how the receptor interacts with certain drugs or downstream signaling components, necessitating
the constant evolution of therapeutic strategies. Furthermore, not all patients exhibit the same EGFR
mutations or expression patterns, and thus, a therapeutic effective in one context may not be
universally applicable. Developing strategies that can effectively inhibit or modify interactions at the
EGF Receptor (985-996) without indiscriminately affecting other cellular signaling processes is a
sophisticated task. Moreover, monitoring and managing acquired resistance through secondary mutations in
and around the 985-996 segment during treatment remains a critical concern. Resistant mutant variants of
EGFR can become selected during treatment with current inhibitors, rendering them ineffective and
requiring alternative therapeutic interventions. Lastly, achieving specificity in targeting the unique
conformational states of mutated EGFR versions, especially within this region, requires detailed
structural insights and innovative drug design approaches. These challenges highlight the breadth of
research required to successfully target this segment for effective therapies, necessitating a careful
and comprehensive approach bridging molecular biology, structural genomics, and clinical oncology.