HBG1 Human

Hemoglobin Gamma A Human Recombinant
Shipped with Ice Packs
In Stock

Description

HBG1 Human Recombinant produced in E. coli is a single polypeptide chain containing 170 amino acids (1-147) and having a molecular mass of 18kDa. HBG1 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
The gamma globin genes, HBG1 and HBG2, are typically expressed in the fetal liver, spleen, and bone marrow. These genes encode gamma chains, which combine with alpha chains to form fetal hemoglobin (HbF). HbF is the primary hemoglobin type in fetuses and is usually replaced by adult hemoglobin (HbA) after birth. However, gamma chain production can persist into adulthood in certain conditions, such as beta-thalassemias. The two gamma chain types differ at amino acid position 136: HBG2 (G-gamma) contains glycine, while HBG1 (A-gamma) contains alanine. HBG2 is the predominant form at birth.
Description
Recombinant human HBG1, expressed in E. coli, is a single polypeptide chain consisting of 170 amino acids (residues 1-147). It has a molecular weight of 18 kDa. The protein includes a 23-amino acid His-tag fused at the N-terminus to facilitate purification, which is achieved using proprietary chromatographic methods.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The HBG1 protein is supplied in a solution at a concentration of 0.25 mg/ml. The solution contains 20 mM Tris-HCl buffer (pH 8.0), 0.15 M NaCl, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To further enhance stability during long-term storage, adding a carrier protein like HSA or BSA to a final concentration of 0.1% is advisable. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 85% using SDS-PAGE analysis.
Synonyms
HBGA, HBGR, HSGGL1, PRO2979, Hemoglobin subunit gamma-1, Gamma-1-globin, Hb F Agamma, Hemoglobin gamma-1 chain, Hemoglobin gamma-A chain, HBG1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMGHFTEE DKATITSLWG KVNVEDAGGE TLGRLLVVYP WTQRFFDSFG NLSSASAIMG NPKVKAHGKK VLTSLGDATK HLDDLKGTFA QLSELHCDKL HVDPENFKLL GNVLVTVLAI HFGKEFTPEV QASWQKMVTA VASALSSRYH.

Q&A

What is HBG1 and what is its role in hemoglobin production?

HBG1 (Hemoglobin Subunit Gamma 1) is one of two gamma globin genes that encode the gamma chains of fetal hemoglobin (HbF). HBG1 specifically encodes the A-gamma chain, which pairs with two alpha globin chains to form fetal hemoglobin (HbF) . HbF represents the main hemoglobin fraction during fetal development, with HBG1 and its paralog HBG2 being highly expressed during this period .

The primary function of HBG1 is to contribute to oxygen transport during fetal development. The gamma chains have a higher affinity for oxygen compared to beta chains found in adult hemoglobin, facilitating efficient oxygen transfer from maternal to fetal circulation. After birth, HBG1 expression gradually decreases as part of the hemoglobin switching process, where HbF is replaced by adult hemoglobin (HbA) .

This developmental regulation is orchestrated by multiple transcription factors, including BCL11A, which acts as a repressor of gamma globin expression in adult erythroid cells . The switch from fetal to adult hemoglobin is normally completed within the first 6 months of life, resulting in HbF levels below 2% in adults .

What distinguishes HBG1 from HBG2?

HBG1 and HBG2 are paralogous genes located in the β-globin locus on chromosome 11 that encode the gamma chains of fetal hemoglobin. Though they share high sequence similarity, they have several important differences:

FeatureHBG1HBG2
Protein productA-gamma chainG-gamma chain
Amino acid at position 136AlanineGlycine
Chromosomal positionDownstream in β-globin clusterUpstream of HBG1
Expression patternPredominantly fetalPredominantly fetal

HBG1 expresses the A-gamma chain while HBG2 expresses the G-gamma chain, differing at amino acid position 136 (alanine in A-gamma, glycine in G-gamma) . Despite their similarity, HBG1 and HBG2 can be differentially regulated and may respond differently to various therapeutic interventions. Both genes contain conserved regulatory elements in their promoter regions, including binding sites for transcription factors like BCL11A that silence gamma globin expression in adult erythroid cells .

The high sequence similarity between these genes presents technical challenges for researchers, requiring specialized approaches to distinguish between them, including long-range PCR followed by nested PCR and sequencing techniques .

What is Hereditary Persistence of Fetal Hemoglobin (HPFH) and how is it related to HBG1?

Hereditary Persistence of Fetal Hemoglobin (HPFH) is a condition characterized by continued expression of fetal hemoglobin (HbF) into adulthood. HPFH is generally considered a benign condition but has significant clinical implications when co-inherited with hemoglobinopathies like sickle cell disease or beta-thalassemia, as elevated HbF levels can ameliorate the clinical severity of these conditions .

HPFH can be classified into two main categories:

  • Deletional HPFH: Caused by large deletions in the beta-globin gene cluster

  • Non-deletional HPFH: Caused by point mutations in the promoter regions of HBG1 and/or HBG2

HBG1-related HPFH typically involves specific mutations in the promoter region of the HBG1 gene that disrupt binding sites for transcriptional repressors like BCL11A . These mutations prevent the normal silencing of gamma globin expression after birth, resulting in continued production of fetal hemoglobin into adulthood.

Promoter variants in either HBG1 or HBG2 can result in non-deletional HPFH, which is clinically benign but can ameliorate disease severity in sickle cell disease and thalassemia . Understanding the molecular mechanisms of HPFH has provided valuable insights for developing therapeutic strategies that aim to reactivate gamma globin expression in hemoglobinopathy patients.

What are the clinical impacts of HBG1 variants?

Variants in HBG1 can have diverse clinical effects ranging from benign to pathological, particularly in neonates. Over 100 gamma globin variants have been described, with varying clinical significance :

Variant CategoryEffectClinical Significance
Promoter variantsAltered gene expressionCan cause HPFH; may ameliorate hemoglobinopathies
Unstable variantsReduced protein stabilityMay cause hemolytic anemia/hyperbilirubinemia in neonates
Altered oxygen affinity variantsChanged oxygen bindingMay cause erythrocytosis (high affinity) or cyanosis (low affinity)
M-hemoglobin variantsAltered heme structureMay cause methemoglobinemia in neonates
Clinically silent variantsNo significant effectNo clinical manifestations

Importantly, clinical symptoms related to gamma globin variants typically resolve after the first six months of life due to the natural hemoglobin switch from fetal to adult hemoglobin expression . This means that conditions like neonatal hemolytic anemia, cyanosis, or methemoglobinemia caused by HBG1 variants generally improve without specific intervention as HBG1 expression naturally decreases.

How is HBG1 expression regulated during development?

HBG1 expression follows a developmentally regulated pattern known as hemoglobin switching. This complex process involves multiple regulatory mechanisms:

  • Transcription factor networks:

    • BCL11A acts as a key repressor of gamma globin expression in adult erythroid cells

    • ZBTB7A (also known as LRF) cooperates with BCL11A to silence gamma globin genes

    • KLF1 indirectly represses gamma globin by activating BCL11A

  • Epigenetic regulation:

    • DNA methylation increases at the HBG1 promoter during development

    • Histone modifications shift from active (H3K4me3, H3K27ac) to repressive (H3K27me3, H3K9me3) marks

    • Chromatin accessibility at the HBG1 locus decreases during development

  • Chromatin organization:

    • The locus control region (LCR) forms dynamic interactions with globin gene promoters

    • These interactions shift from HBG1/HBG2 to HBB during development

    • CTCF-mediated chromatin boundaries help organize the β-globin locus

  • Post-transcriptional regulation:

    • MicroRNAs play roles in fine-tuning hemoglobin production

    • RNA stability mechanisms contribute to proper gamma globin expression

The developmental silencing of HBG1 is a multifactorial process involving the coordinated action of these mechanisms. Understanding these regulatory pathways has provided targets for therapeutic intervention, as disrupting repressive mechanisms can lead to reactivation of HBG1 expression in adult erythroid cells, which may benefit patients with hemoglobinopathies.

What genome editing approaches are being developed for HBG1 manipulation?

Genome editing of HBG1 and HBG2 has emerged as a promising therapeutic strategy for hemoglobinopathies. Current approaches include:

  • CRISPR/Cas9-mediated disruption of repressor binding sites:

    • Targeting BCL11A binding sites in the HBG1/HBG2 promoters

    • Studies have shown that Cas9 editing of γ-globin gene promoters in hematopoietic stem cells can increase red cell HbF by up to 40%

    • No deleterious effects on hematopoiesis or off-target mutations were detected 16 weeks after xenotransplantation of edited HSCs

  • Technical advancements:

    • Engineered Cas9 containing 3 nuclear localization sequences edits human hematopoietic stem and progenitor cells more efficiently than conventional Cas9 with 2 nuclear localization sequences

    • Targeted delivery methods using electroporation of ribonucleoprotein complexes show high efficiency and reduced off-target effects

  • Safety monitoring:

    • CIRCLE-seq (circularization for in vitro reporting of cleavage effects by sequencing) allows comprehensive detection of potential off-target sites

    • Long-term xenotransplantation studies confirm that on-target editing does not impair CD34+ cell regeneration or differentiation into multiple lineages

These approaches aim to recapitulate naturally occurring HPFH mutations or create novel disruptions in repressor binding sites. The goal is to increase HbF production to therapeutically relevant levels that can ameliorate the clinical manifestations of hemoglobinopathies while ensuring safety and efficacy.

How do mutations in HBG1 promoter regions affect fetal hemoglobin production?

Mutations in the HBG1 promoter can significantly alter fetal hemoglobin production by disrupting binding sites for transcription factors that normally repress gamma globin expression. The HBG1 promoter contains several critical regulatory elements:

Regulatory ElementBinding FactorsEffect of Mutations
CACCC boxKLF1, ZBTB7ADisruption increases HBG1 expression
CCAAT boxNF-Y, CP1Mutations alter basal transcription
BCL11A binding siteBCL11AMutations prevent repression
TATA boxTBP and othersAffects basic transcription machinery

Key HPFH-associated mutations in the HBG1 promoter include -175 T>C, -202 C>G, and -196 C>T, among others . These mutations can:

  • Prevent binding of repressors like BCL11A and ZBTB7A

  • Enhance binding of activators of gamma globin expression

  • Alter chromatin structure to favor an open, transcriptionally active state

  • Disrupt long-range chromatin interactions that normally repress gamma globin genes

Understanding these mechanisms has been facilitated by single-cell genome editing functional assays that enable specific mutations to be recapitulated individually and in combination, providing insights into how multiple regulatory elements collectively contribute to HbF expression .

How can single-cell analysis improve our understanding of HBG1 regulation?

Single-cell analysis has revolutionized our understanding of HBG1 regulation by revealing previously unappreciated heterogeneity in gene expression and regulatory mechanisms:

  • Resolving cellular heterogeneity:

    • Single-cell RNA sequencing (scRNA-seq) reveals that HBG1 expression varies significantly even within seemingly homogeneous erythroid populations

    • This heterogeneity may explain variations in response to HbF-inducing treatments

  • Linking genotype to phenotype at single-cell resolution:

    • Single-cell genome editing functional assays enable specific mutations to be recapitulated individually and in combination

    • This allows assessment of how multiple mutation-harboring functional elements collectively contribute to HbF expression

  • Multi-omics approaches provide comprehensive insights:

    • Single-cell ATAC-seq for chromatin accessibility

    • Single-cell ChIP-seq for histone modifications

    • Single-cell Hi-C for 3D chromatin organization

Recent studies have employed single-cell approaches to create unified models of human hemoglobin switching . These approaches have provided insights into how transcription factors like BCL11A and ZBTB7A cooperatively regulate HBG1, the dynamics of chromatin accessibility changes during switching, and the interplay between different regulatory mechanisms.

In conjunction with quantitative modeling and chromatin capture analyses, single-cell technologies illustrate how distinct regulatory mechanisms can synergistically modulate HbF expression , advancing our understanding of hemoglobin regulation and potentially leading to more effective therapeutic strategies.

What technical challenges exist in studying HBG1, and how can they be overcome?

Studying HBG1 presents several technical challenges that researchers must navigate:

  • High sequence similarity with HBG2:

    • The high homology between HBG1 and HBG2 makes specific targeting difficult

    • Long-range PCR followed by nested PCR and bidirectional sequencing is required for accurate analysis

    • Custom primers and sequencing strategies must be designed to distinguish between these genes

  • Experimental model limitations:

    • Animal models may not accurately reflect human hemoglobin switching

    • Cell culture systems often fail to recapitulate normal developmental regulation

    • Patient samples show high variability due to genetic backgrounds

  • Detection and quantification challenges:

    • Distinguishing A-gamma (HBG1) from G-gamma (HBG2) protein requires specialized techniques

    • Accurately quantifying HbF levels in mixed populations requires standardized approaches

To overcome these challenges, researchers have developed several strategies:

ChallengeMethodological SolutionReference
Gene specificityLong-range PCR with nested primers
Distinguishing proteinsHigh-performance liquid chromatography
Functional analysisSingle-cell genome editing assays
Off-target effectsCIRCLE-seq for comprehensive detection
Clinical relevanceXenotransplantation studies

Advanced techniques like CRISPR/Cas9 with customized guide RNAs have also improved targeting specificity. For example, engineered Cas9 containing 3 nuclear localization sequences has been shown to edit human hematopoietic stem and progenitor cells more efficiently and consistently than conventional Cas9 , representing an important technical advance.

What are the interactions between HBG1 and other genes in the β-globin locus?

The β-globin locus is a complex genomic region where HBG1 functions through intricate interactions with other genes and regulatory elements:

  • Structure of the β-globin locus:

    • Located on chromosome 11p15.5

    • Contains five functional β-like globin genes: HBE (ε), HBG2 (Gγ), HBG1 (Aγ), HBD (δ), and HBB (β)

    • Also contains the HBBP1 pseudogene, which recent research suggests may have functional roles rather than being non-functional

    • The locus control region (LCR) lies upstream and contains multiple DNase I hypersensitive sites

  • Long-range chromatin interactions:

    • The LCR forms dynamic interactions with globin gene promoters

    • These interactions change during development, shifting from embryonic/fetal to adult globin genes

    • Chromatin capture analyses provide insights into how distinct regulatory mechanisms synergistically modulate HbF expression

  • Competitive interactions:

    • The β-globin genes compete for interaction with the LCR enhancers

    • Gene competition is influenced by promoter strengths, distance from the LCR, chromatin state, and transcription factor binding

    • The gamma genes (HBG1 and HBG2) can suppress HBB expression through competition when activated

  • Coordinated regulation:

    • CTCF binding sites throughout the locus help organize chromatin domains

    • Multiple transcription factors, including BCL11A, ZBTB7A, and KLF1, regulate the entire locus

    • Balanced production of alpha and beta-like globin chains is crucial for proper hemoglobin assembly

Understanding these complex interactions is essential for developing therapeutic approaches that aim to reactivate HBG1 expression in adult erythroid cells for treating hemoglobinopathies .

How is HBG1 induction being leveraged for hemoglobinopathy treatment?

The induction of HBG1 expression to increase fetal hemoglobin (HbF) levels represents a promising therapeutic strategy for hemoglobinopathies like sickle cell disease and β-thalassemia:

  • Genome editing approaches:

    • CRISPR/Cas9-mediated disruption of the γ-globin gene promoters in hematopoietic stem cells can increase red cell HbF by up to 40%

    • Targeting BCL11A binding sites in HBG1/HBG2 promoters shows no deleterious effects on hematopoiesis or off-target mutations

    • These approaches aim to recapitulate naturally occurring HPFH mutations that lead to elevated HbF levels

  • Pharmacological approaches:

    • DNA methyltransferase inhibitors like 5-azacytidine

    • Histone deacetylase inhibitors

    • Hydroxyurea, which remains the most widely used HbF inducer in clinical practice

  • Combination strategies:

    • Targeting multiple regulatory elements simultaneously

    • Combining genome editing with pharmacological agents

    • Multiplex editing approaches addressing several repressive mechanisms

  • Clinical translation challenges:

    • Optimizing delivery methods for gene editing technologies

    • Ensuring long-term expression and safety

    • Addressing patient-to-patient variability in response

The levels of HbF induction achieved through genome editing approaches (up to 40%) are potentially therapeutic, as clinical studies have shown that HbF levels of 20-30% can significantly ameliorate symptoms in sickle cell disease and β-thalassemia. Xenotransplantation studies show that edited hematopoietic stem cells maintain their differentiation capacity into multiple lineages, suggesting a favorable safety profile .

What recent advances in HBG1 research are most promising for clinical applications?

Recent advances in HBG1 research have opened new avenues for clinical applications:

  • Technical improvements in genome editing:

    • Engineered Cas9 containing 3 nuclear localization sequences edits human hematopoietic stem and progenitor cells more efficiently and consistently than conventional Cas9

    • Improved delivery methods using electroporation of ribonucleoprotein complexes show high efficiency and reduced off-target effects

    • Advanced off-target detection methods like CIRCLE-seq provide comprehensive safety assessment

  • Single-cell functional genomics:

    • Single-cell genome editing functional assays enable specific mutations to be recapitulated individually and in combination

    • This allows for precise assessment of how multiple regulatory elements collectively contribute to HbF expression

    • These approaches provide a unified model of human hemoglobin switching

  • Comprehensive regulatory network understanding:

    • Identification of novel factors in the gamma globin regulatory network

    • Better understanding of how BCL11A, ZBTB7A, and other factors cooperatively regulate HBG1

    • Insights into chromatin dynamics and long-range interactions

  • Safety and efficacy data:

    • No deleterious effects on hematopoiesis or off-target mutations were detected 16 weeks after xenotransplantation of edited HSCs

    • On-target editing did not impair CD34+ cell regeneration or differentiation into erythroid, T, B, or myeloid cell lineages

These advances collectively point toward the clinical feasibility of HBG1-targeted therapies. The combination of improved technical approaches, deeper mechanistic understanding, and promising safety profiles suggests that therapeutic strategies targeting HBG1 may soon translate to clinical applications for patients with hemoglobinopathies.

Product Science Overview

Structure and Function

Hemoglobin Gamma A (HBG1) encodes the gamma-A chain of fetal hemoglobin (HbF). Fetal hemoglobin is composed of two alpha chains and two gamma chains (α2γ2). The gamma chains are crucial for the high oxygen affinity of fetal hemoglobin, which allows efficient oxygen transfer from the mother to the fetus .

The two types of gamma chains, gamma-A (HBG1) and gamma-G (HBG2), differ at a single amino acid position: glycine is found in the gamma-G chain, while alanine is found in the gamma-A chain . This slight difference does not significantly affect the overall function of fetal hemoglobin.

Expression and Regulation

The expression of gamma globin genes is tightly regulated and predominantly occurs during fetal development. After birth, the production of gamma globin decreases, and adult hemoglobin (HbA), composed of two alpha and two beta chains (α2β2), becomes the dominant form . However, in certain conditions such as beta-thalassemia and sickle cell disease, the expression of gamma globin can persist into adulthood .

The regulation of gamma globin gene expression involves complex interactions between various transcription factors, enhancers, and chromatin remodeling complexes. The beta-globin locus control region (LCR) plays a crucial role in the regulation of the entire beta-globin gene cluster, including the gamma globin genes .

Recombinant Hemoglobin Gamma A

Recombinant Hemoglobin Gamma A is produced using genetic engineering techniques, where the HBG1 gene is cloned and expressed in suitable host cells, such as bacteria or yeast. This allows for the large-scale production of the gamma-A chain, which can be used for research and therapeutic purposes.

Recombinant hemoglobin is valuable for studying the structure and function of hemoglobin, as well as for developing treatments for hemoglobinopathies. By understanding the properties of recombinant hemoglobin, researchers can gain insights into the mechanisms of diseases like sickle cell anemia and beta-thalassemia and develop potential therapies .

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.