BRAF Human

B-Raf Proto-Oncogene Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to BRAF Human

The BRAF gene (v-Raf murine sarcoma viral oncogene homolog B1) encodes a serine/threonine kinase critical to the MAPK/ERK signaling pathway. Located on chromosome 7q34, BRAF regulates cell proliferation, differentiation, and survival. Mutations in this gene are among the most common oncogenic drivers in human cancers, while germline mutations cause rare developmental syndromes .

Protein Structure

BRAF comprises three conserved regions:

RegionFunctionKey Features
CR1AutoinhibitionRas-binding domain (RBD), zinc finger motif for membrane docking
CR2Regulatory hingePhosphorylated residues (e.g., S445) repel CR1, enabling kinase activation
CR3Catalytic domainPhosphorylates MEK1/2, initiating ERK signaling

Activation Mechanism

  • Ras-GTP Binding: Binds to Ras-GTP via CR1, releasing CR1 inhibition .

  • Phosphorylation: Constitutive phosphorylation of S445 in CR2 maintains kinase readiness .

  • Dimerization: Forms dimers for full catalytic activity .

Germline Mutations

Linked to RASopathies:

SyndromeBRAF MutationsKey Features
CardiofaciocutaneousP.G466V, P.G469VFacial dysmorphia, cardiac defects, intellectual disability
Noonan/CostelloP.V600E, P.G468AShort stature, heart defects, developmental delay

Somatic Mutations

V600E Mutation (c.1799T>A):

Cancer TypePrevalenceClinical Impact
Melanoma35–66% Targeted by RAF inhibitors (vemurafenib, dabrafenib)
Colorectal12% Resistant to anti-EGFR therapy; treated with encorafenib + cetuximab
Thyroid44% Papillary carcinoma; BRAF inhibitors in trials

BRAF Mutation Classes and Therapeutic Implications

ClassMechanismTreatment SensitivityExamples
IConstitutive monomerRAF inhibitors (vemurafenib)V600E, V600K
IIConstitutive dimerDimer-specific inhibitorsK601E, L597Q
IIIKinase-impairedMEK inhibitors (trametinib)D594A, G466V

Combination Therapies

  • Melanoma: Dabrafenib + trametinib improves survival vs. monotherapy .

  • Colorectal: Encorafenib + cetuximab + FOLFOX6 (BREAKWATER trial) achieves 60.9% ORR vs. 40% SOC .

Pan-Cancer Approaches

  • Basket Trials: Testing BRAF inhibitors across tumor types (e.g., thyroid, NSCLC) .

  • Next-Gen Targets: MEK inhibitors for Class III mutants; dimer inhibitors for Class II .

Research Frontiers and Challenges

  • Resistance Mechanisms: Upregulation of ERK1/2 or alternative RTK signaling limits BRAF inhibitor efficacy .

  • Senescence in Nevi: BRAF V600E induces reversible cell-cycle arrest in benign melanocytes, revealing tumor suppression mechanisms .

  • Liquid Biopsies: Detecting BRAF mutations in circulating DNA for early diagnosis .

Product Specs

Introduction
B-Raf Proto-Oncogene, also called BRAF, belongs to the raf/mil family of serine/threonine protein kinases. It plays a role in the MAP kinase/ERKs signaling pathway, which influences cell division, differentiation, and secretion. BRAF mutations are linked to cardiofaciocutaneous syndrome, a condition marked by heart defects, intellectual disability, and distinctive facial features. Additionally, BRAF mutations are implicated in various cancers, such as non-Hodgkin lymphoma, colorectal cancer, malignant melanoma, thyroid carcinoma, non-small cell lung carcinoma, and lung adenocarcinoma.
Description
Recombinant Human BRAF, produced in E. coli, is a non-glycosylated polypeptide chain consisting of 360 amino acids (432-766a.a). It has a molecular weight of 40.6 kDa. The protein includes a 25 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The BRAF protein solution is provided at a concentration of 0.25 mg/ml and is formulated in a buffer containing 20mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product should be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
Purity is determined to be greater than 80.0% by SDS-PAGE analysis.
Synonyms

Proto-Oncogene B-Raf, BRAF1, RAFB1, NS7, EC 2.7.11.1, B-RAF1, P94.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEFSEDRN RMKTLGRRDS SDDWEIPDGQ ITVGQRIGSG SFGTVYKGKW HGDVAVKMLN VTAPTPQQLQ AFKNEVGVLR KTRHVNILLF MGYSTKPQLA IVTQWCEGSS LYHHLHIIET KFEMIKLIDI ARQTAQGMDY LHAKSIIHRD LKSNNIFLHE DLTVKIGDFG LATVKSRWSG SHQFEQLSGS ILWMAPEVIR MQDKNPYSFQ SDVYAFGIVL YELMTGQLPY SNINNRDQII FMVGRGYLSP DLSKVRSNCP KAMKRLMAEC LKKKRDERPL FPQILASIEL LARSLPKIHR SASEPSLNRA GFQTEDFSLY ACASPKTPIQ AGGYGAFPVH.

Q&A

What is the BRAF gene and what is its primary biological function?

The BRAF gene (v-raf murine sarcoma viral homolog B1) encodes a serine/threonine kinase that functions as a critical component of the RAS/MAPK signaling pathway. It plays a significant role in transmitting chemical signals from outside the cell to its nucleus, contributing to essential cellular processes including growth, differentiation, survival, proliferation, and apoptosis . The BRAF protein consists of 766 amino acids and has a molecular weight of approximately 84,436 daltons. The gene is located on chromosome 7 at position 34, specifically from base pair 140,080,750 to base pair 140,271,032 . As part of the tightly regulated RAS/RAF/MEK/ERK/MAPK pathway, BRAF assumes an antiapoptotic role necessary to regulate cell death and serves as a critical effector in the transmission of mitogenic signals .

How does BRAF signaling operate in normal cellular contexts?

In normal cellular contexts, BRAF functions as a key signaling effector in the canonical NMDA-R–CaMKII–SynGap–Ras–BRaf–MEK–ERK transduction cascade at synapses . This signaling pathway is tightly regulated through a series of phosphorylation events. When activated, BRAF phosphorylates MEK (MAPK/ERK kinase), which subsequently phosphorylates ERK (extracellular signal-regulated kinase), leading to various cellular responses. Under normal conditions, BRAF switches on and off appropriately to deliver proteins necessary for cell cycle regulation and growth . This signaling is particularly important in neuronal tissues, where BRAF is highly expressed and contributes to synaptic plasticity and function .

What experimental models are most effective for studying BRAF function?

Research indicates that multiple experimental models are effective for studying BRAF function:

  • Lentivirus-based replacement approaches: These permit replacement of endogenous proteins with recombinant mutant proteins at functionally equivalent amounts. This approach has been validated in rat, mouse, and human neurons, making it particularly valuable for producing large numbers of animal models .

  • Human embryonic stem cell (hESC)-induced neurons: These have been successfully used to study BRAF mutations and their effects on phosphorylated p42/44 MAPK levels, providing insights into how BRAF mutations affect human neuronal function .

  • Multiple patch-clamp recordings: This technique allows for high-throughput, high-resolution analysis of BRAF function in neurons, enabling researchers to quantify the effects of BRAF mutations on AMPA-R-mediated synaptic transmission .

  • Cancer cell lines: Various cancer cell lines harboring BRAF mutations provide valuable models for studying BRAF's role in oncogenesis and testing targeted therapies.

What are the major types of BRAF mutations identified in human diseases?

BRAF mutations are diverse and can be categorized into different classes based on their downstream signaling effects. The most common BRAF mutation is V600E, which represents approximately 80% of all BRAF mutations . This mutation results from a T1799A nucleotide change, leading to a valine to glutamic acid substitution at position 600. Other clinically significant mutations include:

  • G469E mutation: Associated with decreased BRAF signaling and reduced AMPA responses in neurons

  • K482M mutation: Also linked to decreased BRAF signaling

  • K499E mutation: Associated with increased BRAF signaling

Approximately 40 BRAF mutations have been associated with the neurodevelopmental disorder cardio-facio-cutaneous syndrome (CFC) . Different types of BRAF alterations can include:

  • Missense mutations (introduction of an incorrect amino acid)

  • Gain-of-function mutations

  • Loss-of-function mutations

  • Copy number alterations (CNAs) including hemizygous deletions, gains, and high-level amplifications

How do BRAF mutations contribute to cancer development and progression?

BRAF mutations contribute to cancer development and progression primarily by dysregulating the MAPK signaling pathway, leading to uncontrolled cell proliferation. The V600E mutation is particularly oncogenic as it constitutively activates BRAF, resulting in continuous downstream signaling independent of upstream regulation .

BRAF mutations are implicated in various cancers, including:

  • Malignant melanoma

  • Colorectal cancer

  • Ovarian cancer

  • Thyroid cancer

In breast cancer, BRAF expression and copy number alterations (CNAs) have been shown to predict unfavorable outcomes. Research from the METABRIC dataset indicates that BRAF CNAs are significantly associated with tumor grade and hormone receptor status . The table below summarizes these associations:

CharacteristicHemizygous deletion (n = 225)Neutral (n = 1662)Gain (n = 242)High-level amplification (n = 44)p value
Grade I/II87 (8.5%)836 (81.6%)94 (9.2%)8 (0.8%)< 0.001
Grade III124 (11.8%)744 (71.1%)144 (13.8%)35 (3.3%)< 0.001
ER Positive161 (10.0%)1275 (79.2%)153 (9.5%)20 (1.2%)< 0.001
ER Negative58 (11.9%)320 (65.6%)87 (17.8%)23 (4.7%)< 0.001
PR Positive102 (9.8%)833 (80.1%)93 (8.9%)12 (1.2%)< 0.001
PR Negative100 (10.6%)687 (73.1%)127 (13.5%)26 (2.8%)< 0.001

These data show that BRAF alterations, particularly gains and high-level amplifications, are more common in higher-grade tumors and hormone receptor-negative breast cancers .

How can researchers accurately detect and characterize BRAF mutations in clinical samples?

Researchers can employ several methodologies to detect and characterize BRAF mutations:

  • Next-Generation Sequencing (NGS): Provides comprehensive analysis of the BRAF gene, capable of detecting both common and rare mutations.

  • Microarray gene expression profiling: Used to analyze BRAF mRNA expression levels, with data typically presented as log2 intensity values. The METABRIC dataset shows BRAF mRNA expression ranging from 5.2-7.6 log intensity, with a mean of 5.93 ± 0.3 .

  • Copy Number Alteration (CNA) analysis: Detects hemizygous deletions, gains, and high-level amplifications. In the METABRIC dataset, BRAF CNAs were classified as: -1 (hemizygous deletion, 10.4%), 0 (neutral/no change, 76.5%), 1 (gain, 11.1%), and 2 (high-level amplification, 2.0%) .

  • Immunohistochemistry (IHC): Particularly useful for detecting V600E mutations using mutation-specific antibodies.

  • Droplet Digital PCR (ddPCR): Provides highly sensitive detection of specific BRAF mutations, particularly valuable for liquid biopsy applications.

For optimal results, researchers should select methods based on their specific research questions, sample types, and required sensitivity levels.

What are the most effective strategies for studying BRAF signaling in neuronal systems?

Research has established several effective strategies for studying BRAF signaling in neuronal systems:

  • Lentivirus-based gene replacement method: This approach allows for replacement of endogenous BRAF with recombinant mutant proteins at functionally equivalent levels. The technique utilizes an H1 promoter to drive shRNA-mediated knockdown of endogenous BRAF while simultaneously expressing a GFP-tagged human BRAF under an EF1α promoter . This method has been validated in rat, mouse, and human neurons.

  • Multiple patch-clamp recordings: This technique enables simultaneous recording from multiple neurons, allowing researchers to quantify the effects of BRAF mutations on synaptic transmission. Studies have utilized this approach to analyze over 6,000 pairs of rat neurons, demonstrating that both loss-of-function and gain-of-function BRAF mutations induce a wide range of increases or decreases in AMPA-R-mediated transmission at synapses .

  • Behavioral assays: Fear conditioning and Y-maze learning tests can be used to correlate BRAF signaling alterations with cognitive function. Research has shown that the level of BRAF mutation-induced alterations in synaptic transmission correlates with the degree of learning deficits in animals and cognitive impairments in human patients .

  • Analysis of phosphorylated p42/44 MAPK levels: This downstream marker of BRAF activity can be used to assess the functional consequences of BRAF mutations in neuronal systems .

How can researchers effectively model BRAF mutations in experimental systems?

Researchers can model BRAF mutations through several experimental approaches:

  • Lentiviral expression systems: As demonstrated in the research, lentiviral constructs can be designed to simultaneously knock down endogenous BRAF and express mutant forms at physiologically relevant levels . This approach allows for precise control of expression levels, which is critical for accurately modeling BRAF mutations.

  • CRISPR/Cas9 genome editing: This technology enables the introduction of specific BRAF mutations into cellular and animal models, creating more physiologically relevant systems for studying mutation effects.

  • Patient-derived xenografts (PDXs): Tumor samples from patients with BRAF mutations can be implanted into immunodeficient mice, preserving the genetic and histological characteristics of the original tumor.

  • Human ESC-induced neurons: These provide a valuable system for studying how BRAF mutations affect human neuronal function. Research has successfully used this approach to examine how different BRAF mutations affect MAPK signaling in human neurons .

  • Conditional knockout/knock-in mouse models: These allow for tissue-specific and temporally controlled expression of BRAF mutations, providing insights into how these mutations affect development and function in specific contexts.

What statistical approaches are most appropriate for analyzing BRAF expression data in clinical samples?

Based on the research data, several statistical approaches are appropriate for analyzing BRAF expression data:

When analyzing BRAF expression data, researchers should consider converting continuous gene expression data to categorical data for certain analyses, such as survival analysis. In the METABRIC dataset, for example, BRAF mRNA expression was categorized as "low" (≤5.93 log intensity) or "high" (>5.93 log intensity) .

What are the current therapeutic approaches targeting BRAF mutations?

Current therapeutic approaches targeting BRAF mutations include:

  • BRAF inhibitors: Three BRAF inhibitors have been approved for clinical use . These drugs specifically target mutant BRAF proteins, particularly those with the V600E mutation, and have shown effectiveness in treating BRAF-mutant cancers, especially melanoma.

  • Combination therapies: Five combinations of a BRAF inhibitor with another agent have been approved for managing certain cancers . These combination approaches often pair BRAF inhibitors with MEK inhibitors to prevent resistance development and enhance efficacy.

  • Pan-cancer basket trials: These trials enroll patients based on the presence of specific BRAF mutations rather than tumor type, allowing for evaluation of targeted therapies across multiple cancer types .

  • Targeted inhibitor combination therapies: These are now approved for patients with advanced or metastatic BRAF V600E-mutated tumors .

Research has shown that these targeted approaches have been effective against tumors with BRAF V600 mutations . In laboratory studies, inhibition of BRAF V600E has been shown to induce cell death in select melanoma cell lines .

What mechanisms underlie resistance to BRAF-targeted therapies?

While not explicitly detailed in the provided search results, based on the general understanding of BRAF signaling and targeted therapies, resistance mechanisms likely include:

  • Secondary mutations in BRAF: These can prevent drug binding while maintaining kinase activity.

  • Activation of alternative RAF isoforms: When BRAF is inhibited, CRAF or ARAF may become activated to maintain downstream MAPK signaling.

  • RAS mutations: These can bypass BRAF inhibition by activating other RAF isoforms.

  • MEK mutations: These can reactivate the MAPK pathway downstream of BRAF.

  • Receptor tyrosine kinase upregulation: Increased expression or activation of upstream receptors can overcome BRAF inhibition.

  • Alternative pathway activation: Activation of PI3K/AKT or other parallel signaling pathways can compensate for BRAF inhibition.

Understanding these resistance mechanisms is critical for developing more effective therapeutic strategies and rational drug combinations.

How can BRAF mutation status inform clinical decision-making and precision medicine approaches?

BRAF mutation status is increasingly important for clinical decision-making:

  • Biomarker-driven therapeutic selection: BRAF mutations, particularly V600E, serve as biomarkers for selecting patients who are likely to respond to BRAF inhibitors. Developments in targeted therapeutics have led to biomarker-driven clinical trials, beginning with targeted BRAF inhibitor monotherapy in melanoma, followed by exploratory pan-cancer basket trials .

  • Prognostic stratification: In breast cancer, BRAF expression levels and copy number alterations have prognostic significance. The research shows that BRAF alterations are associated with tumor grade and hormone receptor status, potentially helping to identify patients at higher risk of poor outcomes .

  • Therapeutic monitoring: BRAF testing can be used to monitor treatment response and detect the emergence of resistance.

  • Cross-cancer applications: Understanding the spectrum and targetability of BRAF alterations is now fundamental to the practice of diagnostic and therapeutic oncology .

Gene testing for BRAF mutations is reliable and inexpensive, and is likely to become a standard of care in some cancers . As genetic testing and targeted therapies continue to advance, the potential for improved management of diseases related to BRAF mutations is promising .

How do different classes of BRAF mutations differentially affect downstream signaling pathways?

Research has identified distinct classes of BRAF mutations with varying effects on downstream signaling:

  • Gain-of-function mutations (e.g., V600E, K499E): These mutations increase BRAF signaling, leading to heightened phosphorylation of downstream targets like MEK and ERK. In neuronal systems, these mutations increase AMPA-R-mediated transmission at synapses .

  • Loss-of-function mutations (e.g., G469E, K482M): These mutations impair BRAF signaling, reducing phosphorylation of downstream targets. In neurons, these mutations decrease AMPA-R-mediated transmission .

Research indicates that all BRAF mutations impair its signaling to some degree, but the extent of impairment varies. Mutations at sites crucial for interaction with ATP and substrate proteins cause predominant deficits in BRAF signaling and cognition . These differential effects have important implications for both disease manifestation and therapeutic approaches.

Future research should focus on more precisely characterizing how different BRAF mutations affect various downstream pathways beyond the canonical MEK-ERK cascade, including potential effects on other signaling networks that may contribute to disease phenotypes.

What is the role of BRAF in neurodevelopmental disorders, and how can this inform therapeutic approaches?

BRAF plays a critical role in neurodevelopment, as evidenced by its association with neurodevelopmental disorders:

  • Cardio-facio-cutaneous syndrome (CFC): Approximately 40 BRAF mutations are associated with this rare neurodevelopmental disorder . CFC is characterized by distinctive physical features and developmental challenges .

  • Synaptic function: Research has identified BRAF as a key effector in the NMDA-R–CaMKII–SynGap–Ras–BRaf–MEK–ERK transduction cascade at synapses . This signaling pathway is crucial for synaptic plasticity and learning.

  • Correlation with cognitive impairment: The level of BRAF mutation-induced alterations in transmission correlates with the degree of learning deficits in animals and the extent of cognitive impairments in human patients .

Understanding BRAF's role in neurodevelopment could inform therapeutic approaches through:

  • Targeted modulation of BRAF signaling: Depending on whether mutations cause gain or loss of function, therapies could be developed to normalize BRAF activity.

  • Critical developmental windows: Identifying when BRAF signaling is most critical for neurodevelopment could inform the timing of therapeutic interventions.

  • Pathway-specific interventions: Targeting specific downstream effectors of BRAF that are particularly important for neurodevelopment.

Future research should focus on more precisely defining the temporal and spatial requirements for BRAF signaling during neurodevelopment and identifying specific cellular processes affected by BRAF mutations in neurodevelopmental disorders.

How can integrative multi-omics approaches enhance our understanding of BRAF biology and pathology?

Integrative multi-omics approaches can significantly enhance our understanding of BRAF biology through:

  • Combined genomic, transcriptomic, and proteomic analyses: Integrating these data can provide a more comprehensive view of how BRAF mutations affect cellular function at multiple levels. The METABRIC dataset demonstrates the value of integrating genomic data (BRAF CNAs) with transcriptomic data (BRAF mRNA expression) and clinical outcomes .

  • Correlation of molecular alterations with phenotypes: Multi-omics approaches can help correlate BRAF alterations with disease phenotypes across different tissues and contexts. For example, research has shown that BRAF alterations correlate with tumor grade and hormone receptor status in breast cancer .

  • Identification of biomarkers and therapeutic targets: Integrating multiple types of molecular data can help identify novel biomarkers and potential therapeutic targets associated with BRAF mutations.

  • Systems biology approaches: These can help model how BRAF mutations affect entire cellular networks and pathways, providing insights into disease mechanisms beyond direct BRAF effectors.

Future research should focus on developing and applying integrative computational approaches to analyze multi-omics data related to BRAF, with the goal of identifying novel diagnostic biomarkers, prognostic indicators, and therapeutic targets.

Product Science Overview

Introduction

The B-Raf proto-oncogene, also known as BRAF, is a gene that encodes a protein belonging to the raf/mil family of serine/threonine protein kinases. This protein plays a crucial role in regulating the MAP kinase/ERK signaling pathway, which is involved in cell division, differentiation, and secretion .

Structure and Function

The BRAF protein is a serine/threonine kinase that is part of the RAF kinase family. It is composed of several domains, including a Ras-binding domain, a cysteine-rich domain, and a kinase domain. The protein is involved in transmitting signals from the cell membrane to the nucleus, thereby influencing cellular responses to external stimuli .

Biological Properties

BRAF is essential for normal cell growth and development. It is involved in the MAPK/ERK signaling pathway, which regulates various cellular processes such as proliferation, differentiation, and apoptosis. Mutations in the BRAF gene can lead to uncontrolled cell growth and cancer .

Mutations and Clinical Significance

Mutations in the BRAF gene are associated with several types of cancer, including melanoma, colorectal cancer, and thyroid carcinoma. The most common mutation, V600E, results in a substitution of valine (V) with glutamic acid (E) at position 600, leading to increased kinase activity and uncontrolled cell proliferation .

Recombinant BRAF Protein

Recombinant human BRAF protein is produced using various expression systems, including baculovirus-infected insect cells. This recombinant protein is used in research to study the function of BRAF and its role in cancer. It is also used in drug development to screen for inhibitors that target the BRAF kinase .

Applications in Research and Medicine

Recombinant BRAF protein is widely used in cancer research to understand the molecular mechanisms underlying BRAF-related cancers. It is also used in the development of targeted therapies, such as BRAF inhibitors, which have shown promise in treating cancers with BRAF mutations .

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.