Services


In vitro Hepatocyte Transfection Services:

In vitro hepatocyte transfection is a method used to introduce foreign DNA or RNA into liver cells (hepatocytes) under controlled conditions outside a living organism. This technique has important applications in studying liver biology, drug metabolism, toxicity testing, and disease modeling.

In vitro Hepatocyte Transfection Services would typically provide the following:

  1. Cell Sourcing:
    • Primary hepatocytes from various species (e.g., human, rat, mouse).
    • Immortalized hepatocyte cell lines.
  2. Transfection Methods:
    • Liposome-mediated Transfection: Uses lipid vesicles (liposomes) to deliver genetic material into cells.
    • Electroporation: Uses an electric field to introduce nucleic acids into cells.
    • Viral Vectors: Uses modified viruses to deliver genetic material efficiently into hepatocytes.
    • Other Non-viral Methods: Such as calcium phosphate precipitation, DEAE-dextran, and nanoparticle-based delivery.
  3. Optimization Services:
    • Determining the best transfection method for specific experiments.
    • Optimization of transfection efficiency and cell viability.
  4. Post-transfection Analysis:
    • Gene expression analysis (qPCR, Western blotting, immunocytochemistry).
    • Functional assays to determine the effects of the transfected gene.
    • Drug metabolism and toxicity assays, especially if studying drug targets or drug-metabolizing enzymes.
  5. Custom Vector Construction:
    • Cloning of specific genes or RNA sequences into suitable vectors for transfection.
    • Incorporation of reporter genes (like GFP) for easy tracking of transfection efficiency.
  6. Scale-Up Services:
    • Expansion of transfected hepatocytes for larger experiments or screenings.
  7. Disease Modeling:
    • Introducing specific mutations known to cause liver diseases to model the disease in vitro.
    • CRISPR/Cas9-mediated genome editing to study gene function or create disease models.
  8. Quality Control:
    • Validation of cell viability, purity, and functionality.
    • Verification of transfection efficiency and expression of the target gene.
  9. Data Analysis and Reporting:
    • Providing detailed reports of the procedures, results, and interpretations.
    • Recommendations for further experiments or optimizations.

When looking for hepatocyte transfection services, it’s essential to ensure that the service provider has a track record of successful hepatocyte cultures and transfections. Given the delicate nature of primary hepatocytes and the challenges associated with maintaining their differentiated state in culture, expertise in this area is crucial. Always check for client testimonials, publications, and case studies to gauge the competency and reliability of the service provider.

In vivo Liver Transfection Services:

In vivo liver transfection is the introduction of foreign DNA or RNA directly into the liver of a living organism, aiming to express or knockdown genes specifically in hepatocytes (liver cells). It offers a more physiologically relevant setting than in vitro transfection, and it’s crucial for studying liver biology, potential therapeutic interventions, and disease modeling in a more systemic context.

In vivo Liver Transfection Services would typically offer the following:

  1. Targeted Delivery Methods:
    • Hydrodynamic Injection: A method that involves rapid, high-volume injection of a solution containing the DNA into the bloodstream, leading to a transient increase in liver vascular pressure and facilitating gene uptake by hepatocytes.
    • Viral Vectors: Utilizing liver-tropic viruses or engineered viral vectors, such as adenoviruses, lentiviruses, or adeno-associated viruses (AAV), to deliver genes specifically to the liver.
    • Lipid or Nanoparticle-based Vectors: Designed for targeted delivery to hepatocytes when introduced into the circulation.
  2. Optimization Services:
    • Testing different delivery methods or conditions for maximum transfection efficiency with minimal off-target effects or toxicity.
    • Dosage optimization for the chosen delivery vector.
  3. Custom Vector Construction:
    • Cloning of specific genes or RNA sequences into suitable vectors for transfection.
    • Incorporation of reporter genes for tracking transfection efficiency or gene expression in vivo.
  4. Disease Modeling:
    • Creating liver-specific knockouts or overexpressing specific genes to model liver diseases in animals.
    • Utilizing CRISPR/Cas9-mediated genome editing for targeted mutations.
  5. Monitoring and Analysis:
    • In vivo Imaging: For real-time monitoring of gene expression or tracking using fluorescent or bioluminescent markers.
    • Histological Analysis: Examining liver tissue sections post-transfection for cellular and molecular changes.
    • Functional Assays: Studying the liver’s metabolic, synthetic, or detoxification functions post-transfection.
    • Safety and Toxicity Assessment: Monitoring the liver and overall health status of the transfected animal over time.
  6. Data Analysis and Reporting:
    • Providing detailed reports of the procedures, results, and interpretations.
    • Recommendations for scaling, further experiments, or optimizations.
  7. Regulatory Compliance:
    • Ensuring that all in vivo experiments are conducted following established ethical guidelines and obtaining necessary permissions or licenses for animal experiments.
  8. Therapeutic Applications:
    • Some services might offer the development or testing of potential gene therapies targeted to the liver, including the formulation, delivery, and subsequent efficacy and safety assessments.

When considering in vivo liver transfection services, the service provider’s expertise in animal handling, ethical considerations, and familiarity with the challenges of in vivo gene delivery is vital. It’s also essential to look for a service provider that can handle the complexities of in vivo experiments, from the nuances of the chosen animal model to the intricacies of data interpretation.

Viral Vector Production and Purification:

Viral vectors are popular tools in molecular biology and gene therapy because of their efficiency in delivering genes to target cells. The production and purification of viral vectors are crucial steps to ensure that they are safe, effective, and suitable for research or therapeutic purposes.

Viral Vector Production:

  1. Choice of Viral Vector: Commonly used viral vectors include adenoviruses, lentiviruses, retroviruses, and adeno-associated viruses (AAV). The choice depends on the desired application (e.g., gene therapy, vaccination, or basic research), the target tissue, and the desired duration of gene expression.
  2. Plasmid Construction: Specific genes of interest, along with necessary regulatory elements, are cloned into plasmids designed for viral production.
  3. Cell Line Selection: Producer cell lines are chosen based on the type of viral vector. For instance, HEK293 or HEK293T cells are often used for adenovirus, lentivirus, and AAV production.
  4. Transfection: The producer cell line is transfected with the viral plasmid(s) and other necessary helper plasmids, depending on the viral system.
  5. Viral Vector Harvest: After a specified period, the medium containing the viral particles is harvested.

Viral Vector Purification:

  1. Cell Debris Removal: The harvested medium is first centrifuged to remove cells and large cellular debris.
  2. Precipitation: This method uses solutions like polyethylene glycol (PEG) to concentrate viral particles.
  3. Ultracentrifugation: Viral particles are further purified and concentrated using density gradient ultracentrifugation. This method separates viral particles from other cellular components based on density.
  4. Chromatography: Techniques such as ion-exchange chromatography, size-exclusion chromatography, or affinity chromatography can be employed to purify viral vectors further.
  5. Filtration: Ultrafiltration or nanofiltration can be used for final purification, concentration, and buffer exchange.

Quality Control:

  1. Titer Determination: This measures the number of viral particles or infectious units in the preparation. Common methods include plaque assays, RT-qPCR (for RNA viruses), or qPCR (for DNA viruses).
  2. Purity Assessment: SDS-PAGE and Western blotting can be used to assess the purity and confirm the presence of viral proteins.
  3. Functional Assays: Depending on the application, it might be essential to test the viral vector’s ability to transduce target cells and express the gene of interest.
  4. Safety Testing: It’s crucial to check for replication-competent viruses, especially if the viral system is designed to be replication-deficient. This is particularly vital for therapeutic applications.
  5. Sterility Testing: Ensuring the absence of bacterial or fungal contamination.
  6. Endotoxin Level: Especially important for in vivo applications, this test ensures that endotoxin levels are within acceptable limits to prevent undue immune reactions when the viral vector is administered.

Given the complexities and exacting standards required in viral vector production and purification, many researchers rely on specialized facilities or commercial vendors. These organizations possess the equipment, expertise, and quality control measures necessary to produce high-quality viral vectors suitable for both research and clinical applications.