Premade Retrovirus Particles
Product DetailsApplicationCase StudyFAQ
Product Details
Gamma-retrovirus (γ-retrovirus) is a genus in the Retroviridae family. Two types of γ-retroviruses are widely used in gene therapy which include MoMLV (Moloney Murine Leukemia Virus) and MSCV (Murine Stem Cell Virus). γ-retroviruses can only transduce dividing cells and can promote the efficient transfer of genes into a variety of cell types.
The retroviral products are available from Creative Biogene are derived from MMLV or MSCV. These particles are developed for expressing fluorescent or bioluminescent reporters which can be used for both in vitro and in vivo applications.
Key Advantages of Our Premade Retrovirus Particles
- Broad cell tropism: MoMLV and MSCV can transduce diverse mitotic cell types.
- High efficiency: Achieve highly efficient stable gene expression in dividing cells.
- Long-term expression: Enable stable genomic integration for lifelong gene expression.
- Large cargo capacity: Can package up to 8-10 kb gene inserts.
Premade Retrovirus Particles List
Application
MMLV and MSCV retroviral vectors enable highly efficient stable gene transfer into dividing cells, making them invaluable tools for diverse applications including overexpression and knockdown studies, cell line engineering, stem cell reprogramming, optogenetics, gene therapy, and gene editing. Their ability to efficiently deliver genes into a broad range of mitotic cell types and achieve permanent genomic integration underlies their utility across these areas.
Here are some key applications of retroviruses for gene delivery demonstrated using our retroviral products:
- Fluorescent labeling and cell tracking: For stable expression of fluorescent reporters like GFP and mCherry to label and track cells long-term.
- Gene function and cell biology studies: To express genes and selection markers in cell lines for studies of gene function and cell biology.
- Bioluminescent reporting: As bioluminescent reporters when fused to luciferase to enable live imaging of cells.
- Gene overexpression and knockdown: For ectopic expression of cDNAs and shRNAs to examine gene overexpression or knockdown phenotypes.
- Neuronal transduction and optogenetics: To deliver fluorescent reporters and optogenetic tools like Channelrhodopsin into neurons.
- Generating stable cell lines: For expression of drug selection markers to generate stable cell lines.
Case Study
Case Study 1
Researchers utilized optogenetic methods to characterize and manipulate a subset of adult-born dentate granule cells (abDGCs) in the mouse hippocampus, finding they constitute high-firing neurons that increase population sparsity during a critical maturation window of 4-7 weeks. Suppressing abDGC spiking led to reduced hippocampal sparsity and impaired novelty recognition memory. To manipulate abDGC activity, the team introduced the excitatory opsin Channelrhodopsin-2 into abDGCs using retroviral and transgenic mouse tools.
Figure 1. To target abDGCs, the researchers injected wild-type C57Bl6/J mice with a retrovirus from Creative Biogene (MMLV-pMX-T2A-Cre-mCherry) to deliver Cre recombinase into abDGCs. (McHugh S B, et al., 2022).
Case Study 2
The study engineered patient T cells to express folate receptor-α targeted bispecific engagers, finding they could activate endogenous tumor-infiltrating lymphocytes and mediate ovarian cancer elimination. Cytokine modulation of engager T cell differentiation influenced therapeutic efficacy, with less differentiated cells showing optimal tumor accumulation and antitumor activity. These results demonstrate the adoptive transfer of engager-secreting T cells as a promising immunotherapeutic approach for ovarian cancer.
Figure 2. FR-Bh T cells target FRα+ tumor cells and initiate antitumor immune responses against patients with OC specimens. The researchers activated human or mouse T cells using CD3ε and CD28 antibodies before transducing them with Moloney murine leukemia virus (MMLV)-based retroviruses encoding the bispecific engagers. (McGray A J R, et al., 2023).
FAQ
A: A retrovirus is a virus that inserts a copy of its RNA genome into the DNA of a host cell that it invades, thus changing the genome of that cell. The retroviral RNA genome is reverse-transcribed into DNA by a virally encoded reverse transcriptase enzyme. The DNA copy of the retroviral genome is then integrated into the host cell genome by the viral integrase enzyme.
Q: What are the differences between engineered γ retroviruses and lentiviruses?
A: (1) γ retroviruses can only transduce dividing cells whereas lentiviruses can transduce both dividing and non-dividing cells due to the ability of lentiviruses to traverse the intact nuclear membrane.
(2) The genome size packaging capacity is smaller for γ retroviruses (~8 kb) compared to lentiviruses (~10 kb).
(3) Lentiviruses exhibit less genotoxicity and insertional mutagenesis than γ retroviruses.
(4) Lentiviral vectors have more complex genome structures and require additional viral proteins for optimal gene delivery.
(5) γ retroviral vectors are produced at higher titers than lentiviral vectors
Q: What are the application scenarios for retroviruses?
A: (1) As gene delivery vectors for gene therapy, including for genetic diseases, cancer, and regenerative medicine.
(2) As oncolytic viruses for cancer virotherapy.
(3) For the generation of induced pluripotent stem cells by delivering reprogramming factors.
(4) For stable gene transfer and expression in dividing cells like stem cells and lymphocytes.
(5) As viral vectors for gene editing by delivering CRISPR/Cas9 components.
(6) As animal models of infection to study viral pathogenesis.
Q: What advantages do retroviruses have over adenoviruses, lentiviruses, and adeno-associated viruses?
A: (1) Higher gene transfer efficiency in dividing cells than adenoviruses.
(2) Greater packaging capacity than AAVs.
(3) Higher and more stable gene expression than lentiviruses in dividing cells.
(4) Can accommodate larger transgene inserts than AAVs.
(5) Can integrate into host genome for stable long-term expression, unlike adenoviruses.
(6) Simpler vector genome than lentiviruses.
(7) Higher and more scalable vector production titers than lentiviruses.
* For research use only. Not intended for any clinical use.