CagA Pylori

Helicobacter Pylori Cytotoxin-Associated Gene A Recombinant
Shipped with Ice Packs
In Stock

Description

CagA Pylori recombinant antigen is produced in E. coli expressing the H. pylori Cytotoxin-Associated Gene A having the Mw of 34 kDa. ADK is fused to a His-tag at C-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction

Helicobacter pylori is a type of bacteria that can infect the stomach. It is a gram-negative bacterium, meaning it has a specific structure to its cell wall. H. pylori infection is common, affecting roughly half of the global population. When present, it can cause inflammation of the stomach lining (gastritis), potentially leading to more severe conditions like peptic ulcers and even gastric cancer. Due to its association with cancer, the World Health Organization classifies H. pylori as a Group 1 carcinogen.

One of the ways H. pylori causes damage is through a virulence factor called Cytotoxin-associated gene A (CagA). This gene is part of a larger set of genes known as the cag pathogenicity island (PAI). The Cag PAI allows H. pylori to inject the CagA protein directly into the cells lining the stomach.

Once inside these cells, CagA disrupts normal cell functions. This interference can lead to several issues, including:

  • Uncontrolled cell growth (hyperproliferation)
  • Programmed cell death (apoptosis)
  • Changes in the cell's internal structure (cytoskeleton), affecting its shape and function

Additionally, CagA's presence triggers the stomach lining cells to release signaling molecules called cytokines, such as tumor necrosis factor (TNF) and interleukins (IL) 1, 6, and 8. These cytokines amplify the inflammatory response, attracting immune cells to the site. One specific cytokine, IL-8, is known to contribute to the production of harmful molecules called free radicals. These free radicals can damage the DNA of surrounding cells, increasing the risk of mutations that might lead to cancer.

Description

CagA Pylori recombinant antigen is a laboratory-produced version of the CagA protein from H. pylori. It is generated using E. coli bacteria as a production factory. The protein has a molecular weight of 34 kDa. For purification and research purposes, a His-tag is attached to the C-terminus of the protein. The purification process involves specialized chromatographic techniques to isolate the CagA protein.

Physical Appearance
A clear, colorless liquid that has been sterilized by filtration.
Formulation

CagA Pylori recombinant protein is supplied in a solution of 1xPBS with a pH of 7.4.

Stability

For short-term storage (up to one week), CagA Pylori can be kept at refrigerated temperature (4°C). However, for long-term preservation, it is crucial to store it in a frozen state at -18°C or below. Repeated freezing and thawing cycles can degrade the protein and should be avoided.

Purity

Analysis using a technique called PAGE (polyacrylamide gel electrophoresis) with Coomassie staining reveals that the purity of the CagA Pylori recombinant protein is greater than 95%. This method separates proteins based on size, allowing for the assessment of sample purity.

Source

Escherichia Coli.

Purification Method

Purified by proprietary chromatographic technique.

Q&A

What is CagA and what is its significance in H. pylori infection?

CagA (Cytotoxin-associated gene A) is a protein encoded by the cagA gene in Helicobacter pylori. It represents one of the most clinically significant bacterial virulence factors identified to date. When H. pylori colonizes the gastric mucosa, CagA is delivered into gastric epithelial cells via the bacterial type IV secretion system, where it functions as a non-physiological scaffold/hub protein by interacting with multiple host signaling molecules. Chronic infection with H. pylori cagA-positive strains is recognized as the strongest risk factor for gastric cancer development . The significance of CagA lies in its ability to manipulate intracellular signaling pathways, promoting epithelial cell transformations that can ultimately lead to neoplastic changes in gastric tissue. Understanding CagA is therefore crucial for comprehending H. pylori pathogenesis and developing targeted interventions.

How does CagA function at the molecular level in host cells?

Upon delivery into gastric epithelial cells, CagA undergoes tyrosine phosphorylation at Glutamic acid-Proline-Isoleucine-Tyrosine-Alanine (EPIYA) motifs. Following phosphorylation, CagA interacts with various host proteins in both phosphorylation-dependent and -independent manners. Key interactions include:

  • Phosphorylated CagA binds to and aberrantly activates the pro-oncogenic phosphatase SHP2, initiating signaling cascades that promote cell proliferation and motility .

  • Unphosphorylated CagA interacts with the polarity-regulating kinase PAR1/MARK through the CagA multimerization (CM) motif, disrupting cellular polarity and tight junction function .

  • CagA also interferes with Wnt-dependent planar cell polarity (Wnt/PCP) signaling by binding to the C-terminal cytoplasmic tails of VANGL1/2, causing their mislocalization from the plasma membrane to the cytoplasm .

These molecular interactions collectively contribute to the oncogenic potential of CagA by dysregulating cellular processes including proliferation, polarity, migration, and differentiation.

What experimental models are most commonly used to study CagA effects?

Researchers employ multiple experimental systems to investigate CagA biology:

  • Cell Culture Models: Human gastric epithelial cell lines (e.g., AGS, MKN28) are frequently used to study CagA delivery, phosphorylation, and downstream signaling events.

  • Transgenic Animal Models: Transgenic mice expressing CagA have confirmed the protein's oncogenic potential, demonstrating that CagA expression alone can induce gastrointestinal and hematopoietic neoplasms .

  • Developmental Models: Xenopus laevis (African clawed frog) embryos have been utilized to study how CagA disrupts Wnt/PCP-dependent morphogenetic processes, showing impaired gastrulation, neural tube formation, and axis elongation when CagA is ectopically expressed .

  • Bacterial Genetic Manipulation: Isogenic mutant strains of H. pylori with varying cagA copy numbers have been generated to assess how CagA expression levels affect host cell toxicity .

Each model system offers unique advantages for investigating different aspects of CagA biology, from molecular interactions to whole-organism pathological effects.

What structural polymorphisms exist in CagA proteins worldwide?

CagA exhibits significant structural polymorphism globally, which may underlie geographic differences in gastric cancer incidence. Major structural variants include:

  • Western CagA: Typically contains EPIYA-A, EPIYA-B, and 1-3 repeats of EPIYA-C segments. Western CagA usually possesses 2-4 repeats of the CM(W) motif, with one CM(W) following each EPIYA-C segment .

  • East Asian CagA: Contains EPIYA-A, EPIYA-B, and EPIYA-D segments (instead of EPIYA-C). East Asian CagA does not contain a CM motif in its EPIYA-D segment but carries a single CM(E) immediately downstream of EPIYA-D .

  • Amerindian Variants: Some indigenous populations in the Amazon rainforest carry unique CagA variants with Amerindian type-I CM (CM(AmI)) and Amerindian type-II CM (CM(AmII)) motifs .

The binding affinity of these CagA variants to host proteins like PAR1b varies substantially. A Western CagA with 4 CM(W) motifs shows a PAR1b-binding affinity more than 30-fold higher than that of a Western CagA with a single CM(W). Additionally, a single CM(E) demonstrates binding affinity to PAR1b comparable to that of two tandem CM(W) motifs . These structural differences directly impact CagA's pathogenic potential.

How do researchers determine the EPIYA motif patterns in clinical isolates?

Determining EPIYA motif patterns in clinical isolates typically involves a multi-step approach:

  • PCR Amplification: Researchers design primers targeting conserved regions flanking the EPIYA-containing C-terminal region of CagA.

  • DNA Sequencing: Sanger sequencing of the amplified products allows identification of the specific EPIYA segments (A, B, C, or D) and their arrangement.

  • Restriction Fragment Length Polymorphism (RFLP): Some studies use restriction enzymes that cut at specific sites within different EPIYA motifs to distinguish between variants.

  • Western Blotting: The molecular weight of CagA proteins varies based on the number of EPIYA repeats, allowing rough estimation of repeat numbers via immunoblotting.

  • Whole Genome Sequencing: Increasingly, researchers are using next-generation sequencing to characterize the entire cagA gene and its genomic context.

These methods can be combined to provide comprehensive characterization of EPIYA motif patterns, which is crucial for understanding the epidemiological associations between specific CagA variants and disease outcomes.

How does CagA copy number variation affect virulence and host cell responses?

Recent research has revealed that H. pylori can carry multiple tandem copies of cagA that can change dynamically, challenging the previous understanding that virtually all H. pylori strains carry only one or no copies of cagA . This variation has significant implications:

  • Increased CagA Production: Isolates harboring more cagA copies produce proportionally more CagA protein, enhancing toxicity to host cells .

  • Geographic Distribution: Analysis of 314 H. pylori clinical strains showed that 7.5% of strains from the United States carried multiple cagA copies, while none of the South Korean strains did, suggesting potential regional differences in virulence .

  • Dynamic Modulation: H. pylori can undergo recombination events that alter cagA copy number, representing a novel molecular mechanism by which the bacterium can modulate its virulence .

  • Host Cell Impact: Greater CagA concentrations resulting from multiple cagA copies lead to enhanced SHP2 binding, increased cellular elongation (hummingbird phenotype), and more pronounced disruption of tight junctions .

The ability to quantify cagA copy number using techniques like real-time PCR relative to a single-copy reference gene (e.g., ureA) provides researchers with a tool to assess this virulence mechanism in clinical isolates .

What is the epidemiological evidence linking CagA-positive H. pylori and gastric cancer?

Substantial epidemiological evidence supports the link between CagA-positive H. pylori and gastric cancer:

These epidemiological findings strongly support the classification of CagA as a bacterial oncoprotein and highlight the importance of CagA status in risk stratification for gastric cancer.

What is the "Hit-and-Run" carcinogenesis model for CagA and how is it experimentally supported?

The "Hit-and-Run" carcinogenesis model for CagA proposes that CagA initiates cellular transformations that lead to cancer, but the protein itself is no longer required for maintaining the established cancer phenotype . This model is supported by several lines of experimental evidence:

  • Transgenic Animal Models: Transgenic mice expressing CagA develop gastrointestinal and hematopoietic neoplasms, confirming CagA's oncogenic potential in vivo .

  • Cell Culture Studies: Prolonged exposure to CagA induces cellular changes that persist after CagA removal, including genomic instability, acquisition of cancer stem cell-like properties, and dysregulation of signaling pathways.

  • Clinical Observations: In human gastric cancer tissues, CagA is often absent or expressed at low levels despite the cancer having originated from H. pylori-infected tissue.

  • Genetic Alterations: CagA has been shown to induce BRCAness (a phenotype resembling BRCA1/2 deficiency), leading to genomic instability that may underlie bacterial gastric carcinogenesis .

  • Epigenetic Modifications: CagA can induce epigenetic changes including DNA methylation patterns that persist after bacterial eradication.

The "Hit-and-Run" model explains why successful H. pylori eradication therapies may not prevent gastric cancer in patients who have already undergone certain CagA-induced pre-neoplastic changes, emphasizing the importance of early intervention.

How does CagA polymorphism correlate with regional variations in gastric disease incidence?

CagA polymorphism shows strong correlations with regional variations in gastric disease incidence:

  • East Asian vs. Western Variants: East Asian CagA (with EPIYA-D motifs) is associated with higher SHP2 binding affinity and greater oncogenic potential compared to Western CagA (with EPIYA-C motifs). This correlates with the higher incidence of gastric cancer in East Asian countries compared to Western nations .

  • CM Motif Variations: The CM motif, which binds PAR1/MARK kinases, shows polymorphism between Western CagA (CM(W)) and East Asian CagA (CM(E)). A single CM(E) has binding affinity comparable to two tandem CM(W) motifs, potentially explaining enhanced pathogenicity of East Asian strains .

  • Amerindian Variants: CagA proteins with Amerindian CM motifs (CM(AmI) and CM(AmII)) show extremely weak binding to PAR1b, if any, which may be associated with unique disease patterns in these populations .

  • Copy Number Variations: Research has shown geographic differences in cagA copy numbers, with 7.5% of U.S. strains carrying multiple copies compared to none of the South Korean strains studied. This variation may contribute to regional differences in H. pylori pathogenicity .

Understanding these polymorphisms provides insight into the molecular basis of geographic variations in gastric cancer incidence and may guide region-specific prevention strategies.

How does CagA disrupt the Wnt/PCP signaling pathway and what are the implications?

Recent research has revealed that CagA disrupts Wnt-dependent planar cell polarity (Wnt/PCP) signaling, which coordinates collective cell behaviors and orients cells within the plane of an epithelium. The mechanism and implications include:

  • Molecular Interaction: The N-terminus of CagA directly interacts with the C-terminal cytoplasmic tails of VANGL1/2, which are critical components of Wnt/PCP signaling .

  • Protein Mislocalization: This interaction induces mislocalization of VANGL1/2 from the plasma membrane to the cytoplasm, impairing Wnt/PCP signaling .

  • Developmental Disruption: Ectopic expression of CagA in Xenopus laevis embryos impairs gastrulation, neural tube formation, and axis elongation – processes driven by convergent extension movements that depend on the Wnt/PCP pathway .

  • Stem Cell Proliferation: In mice expressing CagA specifically in the stomach epithelium, pyloric glands were elongated due to hyperproliferation of cells at the gland base, where Lgr5+ stem and progenitor cells reside .

  • Differentiation Impact: CagA expression was associated with fewer differentiated enteroendocrine cells, suggesting that CagA-mediated Wnt/PCP disruption affects cell fate determination .

These findings suggest that CagA may contribute to gastric cancer development by subverting a Wnt/PCP-dependent mechanism that normally restrains pyloric gland stem cell proliferation and promotes appropriate cellular differentiation.

What cellular signaling pathways are dysregulated by CagA and how do they contribute to oncogenesis?

CagA dysregulates multiple cellular signaling pathways that collectively contribute to oncogenesis:

  • SHP2-ERK Pathway: Phosphorylated CagA binds to and activates the tyrosine phosphatase SHP2, leading to sustained activation of the ERK MAPK pathway, which promotes cell proliferation and survival .

  • JAK/STAT Pathway: CagA activates the transcription factor STAT3, promoting inflammatory responses and anti-apoptotic gene expression.

  • β-catenin Pathway: CagA disrupts E-cadherin/β-catenin complexes at adherens junctions, leading to nuclear translocation of β-catenin and activation of genes involved in proliferation and epithelial-mesenchymal transition (EMT).

  • NF-κB Pathway: CagA can activate NF-κB signaling, inducing inflammatory cytokine production and pro-survival signals.

  • PI3K/Akt Pathway: CagA activates the PI3K/Akt pathway, promoting cell survival and resistance to apoptosis.

  • Wnt/PCP Pathway: CagA disrupts the Wnt/PCP pathway by causing mislocalization of VANGL1/2, affecting stem cell proliferation and differentiation .

  • PAR1/MARK Pathway: Through CM motif binding to PAR1/MARK, CagA disrupts cell polarity and tight junction function, contributing to epithelial barrier disruption .

These dysregulated pathways synergistically contribute to the hallmarks of cancer, including sustained proliferation, resistance to cell death, invasion, and metastasis.

How do researchers experimentally distinguish between phosphorylation-dependent and -independent effects of CagA?

Distinguishing between phosphorylation-dependent and -independent effects of CagA involves several sophisticated experimental approaches:

  • Site-Directed Mutagenesis: Researchers create CagA variants with mutations in the tyrosine residues of EPIYA motifs (typically Y→F substitutions), rendering them unable to be phosphorylated while maintaining protein structure.

  • Phosphorylation-Specific Antibodies: Antibodies that specifically recognize phosphorylated EPIYA motifs allow detection of CagA phosphorylation status in experimental settings.

  • SRC Kinase Inhibitors: Since SRC family kinases are primarily responsible for CagA phosphorylation, inhibitors like PP2 can be used to block phosphorylation without affecting CagA delivery.

  • Binding Partner Analysis: Co-immunoprecipitation experiments with known phosphorylation-dependent (e.g., SHP2) and -independent (e.g., PAR1/MARK) binding partners help distinguish pathway activation.

  • Phenotypic Assays: Certain cellular phenotypes are specifically associated with either phosphorylation-dependent (e.g., cell elongation/hummingbird phenotype) or -independent (e.g., tight junction disruption) effects.

  • Domain Deletion Constructs: Constructs expressing only N-terminal or C-terminal portions of CagA can help isolate regions responsible for specific effects.

Through these approaches, researchers have established that CagA's phosphorylation-dependent effects primarily involve SHP2 activation and downstream ERK signaling, while phosphorylation-independent effects include PAR1/MARK binding, tight junction disruption, and Wnt/PCP pathway interference.

What are the current best practices for quantifying CagA copy number in H. pylori strains?

Recent research has revealed that H. pylori can carry multiple tandem copies of cagA, necessitating reliable quantification methods. Current best practices include:

  • Quantitative Real-Time PCR (qPCR): This approach compares the amplification of cagA relative to a single-copy reference gene such as ureA. The amplification efficiencies of both genes should be validated as approximately equal for accurate quantification .

  • PCR Detection of Arrangements: A combination of PCR strategies can detect different genomic arrangements of cagA:

    • PCR using primers flanking the entire cagA region

    • PCR using only forward primers or only reverse primers to detect inverted repeats

    • PCR using both forward and reverse primers to detect either inverted or tandem repeats

  • Southern Blot Analysis: This technique can provide direct visualization of cagA copy number and arrangement in the genome, though it is more labor-intensive than PCR-based methods.

  • Whole Genome Sequencing: Next-generation sequencing followed by bioinformatic analysis can identify cagA copy number and reveal homologous areas (CHAs) involved in recombination events .

  • Droplet Digital PCR (ddPCR): This emerging technology provides absolute quantification without the need for a standard curve, offering potentially greater precision in copy number determination.

When applying these methods, researchers should be aware that standard PCR and sequencing approaches may not distinguish between more than two copies of cagA, as interior copies might not be amplified with primers that anneal outside the repeat region .

What cell culture models best recapitulate the effects of CagA on gastric epithelial cells?

Several cell culture models are used to study CagA effects, each with specific advantages:

  • AGS Cells: This human gastric adenocarcinoma cell line is widely used because it readily displays the "hummingbird phenotype" (cell elongation) upon infection with CagA-positive H. pylori, making it ideal for studying cytoskeletal effects and SHP2 pathway activation.

  • MKN28 Cells: These well-differentiated gastric epithelial cells form tight junctions, making them suitable for studying CagA's effects on epithelial barrier function and cell polarity.

  • MDCK Cells: While not of gastric origin, Madin-Darby Canine Kidney cells are useful for studying polarity disruption by CagA due to their well-characterized apical-basolateral polarity.

  • Primary Gastric Epithelial Cells: These provide the most physiologically relevant model but are challenging to isolate and maintain in culture.

  • Gastric Organoids: Three-dimensional organoid cultures derived from gastric stem cells represent an advanced model that recapitulates the architecture and cellular diversity of gastric glands, allowing for studies of stem cell dynamics, differentiation, and tissue-level responses to CagA.

  • Co-culture Systems: Models incorporating both epithelial cells and immune cells provide insights into CagA's effects on host immune responses and inflammation.

For comprehensive studies, researchers often employ multiple cell models to validate findings across different systems and to leverage the specific advantages of each model.

How can researchers effectively detect and analyze EPIYA phosphorylation in experimental settings?

Detection and analysis of EPIYA phosphorylation in experimental settings employ several complementary techniques:

These approaches collectively provide a comprehensive understanding of EPIYA phosphorylation patterns, kinetics, and functional consequences in diverse experimental settings.

How might understanding CagA mechanisms inform novel therapeutic approaches for H. pylori-associated diseases?

Understanding CagA mechanisms opens several promising therapeutic avenues:

  • Small Molecule Inhibitors: Compounds that specifically disrupt CagA interactions with critical host proteins (e.g., SHP2, PAR1/MARK, or VANGL1/2) could block pathogenic effects while avoiding the problems of antibiotic resistance.

  • Peptide-Based Therapeutics: Peptides mimicking binding domains of CagA targets could competitively inhibit CagA-host protein interactions. For example, peptides containing the CM-binding domain of PAR1 might prevent CagA-induced polarity disruption.

  • Targeted Vaccination: Vaccines specifically targeting CagA, particularly conserved functional domains, might provide protection against the most virulent H. pylori strains while avoiding disruption of the potentially beneficial components of the microbiome.

  • Recombination Inhibitors: Compounds that inhibit recombination events could prevent the development of multiple cagA copies, potentially reducing virulence .

  • Wnt/PCP Pathway Modulation: Therapies targeting components of the Wnt/PCP pathway might counteract CagA-induced dysregulation of gastric epithelial stem cell proliferation and differentiation .

  • Precision Medicine Approaches: Identifying patients infected with particularly virulent CagA variants (e.g., East Asian-type or multiple-copy cagA) could inform more aggressive eradication strategies and closer surveillance for pre-cancerous lesions.

These approaches represent potential paradigm shifts from current treatment strategies that rely primarily on antibiotics and acid suppression.

What are the current challenges in studying CagA strain variations in clinical settings?

Researchers face several challenges when studying CagA strain variations in clinical settings:

  • Heterogeneity of Infection: Patients may be simultaneously infected with multiple H. pylori strains carrying different CagA variants, complicating analysis and interpretation.

  • Sampling Limitations: Endoscopic biopsies sample only a small portion of the gastric mucosa, potentially missing important strain variations present in unsampled regions.

  • Culture Bias: Traditional culture methods may select for specific H. pylori subpopulations, leading to underrepresentation of certain strains.

  • Dynamic CagA Copy Number: The recently discovered ability of H. pylori to dynamically alter cagA copy number through recombination complicates strain characterization, as this property may change during culture or infection .

  • Limited Sample Availability: Restrictions on endoscopic procedures mean that longitudinal samples from the same patient are often unavailable, making it difficult to study strain evolution over time.

  • Standardization Issues: Lack of standardized methods for CagA typing and quantification makes cross-study comparisons challenging.

  • Host Factors Influence: Host genetic and immunological factors significantly impact H. pylori adaptation and CagA expression, introducing variables that are difficult to control in clinical studies.

Addressing these challenges will require improved sampling techniques, culture-independent analysis methods, standardized typing protocols, and integrated approaches that consider both bacterial and host factors simultaneously.

What recent technological advances have enabled new insights into CagA biology?

Several technological advances have revolutionized CagA research in recent years:

  • CRISPR/Cas9 Genome Editing: This technology has enabled precise modification of host genes to study their interactions with CagA, as well as engineering of H. pylori strains with specific CagA variants.

  • Single-Cell Analysis: Techniques like single-cell RNA-sequencing reveal heterogeneous responses to CagA within cell populations and identify previously unrecognized cell subsets particularly vulnerable to CagA effects.

  • Advanced Imaging: Super-resolution microscopy and live-cell imaging have provided unprecedented insights into CagA localization, trafficking, and dynamic interactions with host proteins.

  • Organoid Technology: Three-dimensional gastric organoids derived from human stem cells provide physiologically relevant models for studying long-term CagA effects on tissue architecture and stem cell dynamics .

  • Proximity Labeling: Techniques like BioID and APEX2 allow identification of the CagA interactome in living cells, revealing transient and weak interactions that might be missed by traditional co-immunoprecipitation.

  • Bacterial Genetics Tools: Improved genetic manipulation techniques for H. pylori have facilitated the creation of isogenic strains differing only in specific CagA properties, enabling precise analysis of structure-function relationships .

  • Multi-omics Approaches: Integration of genomics, transcriptomics, proteomics, and metabolomics data provides systems-level understanding of how CagA reprograms host cells.

These technological advances continue to deepen our understanding of CagA biology and open new avenues for therapeutic intervention in H. pylori-associated diseases.

Product Science Overview

Cytotoxin-Associated Gene A (CagA)

CagA is a protein encoded by the cagA gene, which is part of the cag pathogenicity island (cag PAI). This island consists of approximately 40 kb of chromosomal DNA and includes up to 32 open reading frames (ORFs) that encode the effector protein CagA and components of the bacterial type IV secretion system (T4SS) . The T4SS forms a syringe-like structure that delivers CagA into gastric epithelial cells .

Mechanism of Action

Once inside the host cells, CagA undergoes tyrosine phosphorylation and activates a series of intracellular signal transduction reactions. This leads to severe tissue inflammation and damage . CagA has been shown to play a critical role in gastric inflammation and the development of gastric cancer . It negatively regulates autophagy by downregulating BECN1, a key protein in the autophagy pathway . This inhibition of autophagy promotes the production of pro-inflammatory mediators, contributing to the pathogenicity of H. pylori .

Recombinant CagA

Recombinant CagA refers to the CagA protein that has been produced using recombinant DNA technology. This involves cloning the cagA gene into an expression vector, which is then introduced into a host organism (such as E. coli) to produce the protein. Recombinant CagA is used in research to study its structure, function, and role in H. pylori pathogenicity.

Clinical Implications

Infection with CagA-positive H. pylori strains is associated with a higher risk of developing severe gastric diseases compared to CagA-negative strains . Nearly 70% of all H. pylori strains isolated worldwide possess the cag PAI, with higher prevalence in East Asian isolates . Understanding the role of CagA in H. pylori infection can help in developing targeted therapies and preventive measures for gastric diseases.

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.