Description: Fully humanized mouse with in situ replacement of murine heavy and kappa light chain V(D)J loci with human counterparts, retaining murine constant regions .
Key Features:
Applications:
Description: Combines human heavy chain variable regions with a single human kappa light chain VJ locus, enabling efficient assembly of bispecific antibodies .
Key Features:
Applications:
Description: Generates heavy-chain-only antibodies (HCAbs) with modified constant regions, enabling single-domain antibodies (sdAbs) or nanobodies .
Key Features:
Applications:
RenNano-derived HCAbs demonstrate exceptional diversity and binding kinetics:
RenNano mice show robust responses to diverse antigens:
Parameter | RenNano | Wild-Type |
---|---|---|
Spleen B Cell % | ~50% | ~50% |
F follicular B % | ~70% | ~70% |
MZ B Cell % | ~20% | ~20% |
Data from |
RenMab™: Monoclonal antibodies (e.g., anti-PD-1, anti-VEGF) .
RenNano®: Nanobodies for imaging or targeted toxin delivery .
Biocytogen has licensed RenMice platforms to major pharmaceutical companies:
While not directly related to the antibody platforms, studies on hypertension in (mRen-2)27 rats highlight complexities in renin-angiotensin system (RAS) interactions. Key findings include:
Kinetic Similarity: Mouse Ren-2 and rat renin showed comparable K<sub>cat</sub>/ K<sub>m</sub> values when acting on rat angiotensinogen (0.04 vs. 0.05 L·μmol⁻¹·s⁻¹) .
Hypertension Drivers: Tissue RAS activation, not kinetic efficiency, likely explains hypertension in transgenic models .
Mice exhibit low circulating angiotensinogen, making substrate availability a rate-limiting factor for angiotensin II production . This contrasts with humans and rats, where renin is the primary regulator .
Several REN mouse models have been developed for cardiovascular research, with the most significant being the Ren-2 gene knockout mice and the newer humanized REN mouse platforms. The targeted inactivation of the Ren-2 gene in strain 129 mice produces viable and healthy subjects that allow for direct evaluation of the Ren-1d gene's ability to regulate blood pressure in the absence of Ren-2 enzyme expression . This model is particularly valuable for studying the physiological significance of the duplicated renin gene found in wild mouse strains and some inbred laboratory strains. Additionally, more advanced platforms like RenMab, RenLite, and RenNano humanized mice have been developed for specialized antibody discovery applications targeting the renin-angiotensin system .
Each humanized REN mouse platform has been engineered with specific research applications in mind:
Mouse Model | Genetic Modification | Key Features | Primary Research Applications |
---|---|---|---|
RenMab™ | Complete replacement of murine variable regions with full human heavy and kappa light chain V(D)J loci | Normal immune system comparable to wild-type mice, greater diversity of antibody genes | Discovery of fully human therapeutic antibodies |
RenLite® | Replacement of murine heavy chain variable regions with human ones, plus replacement of one κ light chain VJ locus | Diverse epitopes and high affinities, more efficient downstream assembly of complex drug molecules | Bispecific antibodies and bispecific antibody-drug conjugates (bsADCs) |
RenNano® | Complete human heavy chain variable genes with modified mouse constant regions | Generates heavy-chain-only antibodies (HCAbs) with great diversity and nM-level binding affinity | Single-domain antibodies (sdAbs), bispecific/multispecific antibodies, CAR cell therapies |
RenTCR-mimic™ | Further modified to express human leukocyte antigen (HLA) genes | High specificity and affinity against intracellular tumor antigens | T cell engagers, bispecific/multispecific antibodies, CAR-T therapies |
These platforms enable researchers to address various aspects of renin biology through antibody-based approaches .
For precise quantification of renin expression in mouse models, the recommended approach is using a high-sensitivity enzyme-linked immunosorbent assay (ELISA) specifically designed for mouse REN. The Mouse REN (Renin) ELISA Kit offers outstanding precision with a detection range of 7.81-500 pg/mL and sensitivity of 4.52 pg/mL . For optimal results, the following methodology is recommended:
Sample collection: Obtain serum, plasma, or other biological fluids (25 μL sample volume is typically sufficient)
Assay procedure: Follow the sandwich ELISA protocol, which typically requires approximately 3.5 hours
Quality control: Ensure specificity by confirming no significant cross-reactivity between Mouse REN and analogues
Data analysis: Calculate concentration using the standard curve generated during the assay
This approach enables accurate measurement of renin levels crucial for investigating blood pressure regulation, vascular function, and electrolyte balance in mouse models .
When designing experiments to study Ren genes' role in blood pressure regulation, researchers should implement a multi-faceted approach:
Baseline characterization: Establish baseline blood pressure measurements in both wild-type and Ren-modified mice using either tail-cuff plethysmography (for non-invasive screening) or telemetry (for continuous monitoring)
Challenge testing: Subject mice to physiological challenges that stress the renin-angiotensin system:
Salt loading and salt restriction to evaluate adaptive responses
Angiotensin II infusion to test downstream pathway responsiveness
Dehydration/rehydration cycles to assess dynamic regulation
Comparative analysis: Include both Ren-2 null mice and control mice to directly evaluate the ability of Ren-1d gene to regulate blood pressure in the absence of Ren-2 expression
Biochemical profiling: Measure both active renin and prorenin concentrations in plasma, as Ren-2 null mice show increased active renin but decreased prorenin levels
Tissue expression mapping: Perform histopathological analysis of renin-expressing tissues to identify potential compensatory mechanisms
This comprehensive experimental design allows for robust investigation of the functional equivalence between different Ren genes and their compensatory mechanisms in blood pressure regulation.
Researchers can strategically utilize REN mouse models for therapeutic development through several advanced approaches:
Target validation: Use Ren-2 null mice to validate the specific contributions of renin isoforms to pathophysiological processes, enabling precise targeting in drug development
Antibody development pipeline: Implement a systematic approach using humanized REN mouse platforms:
Initial immunization and screening using RenMab mice to generate diverse, fully human antibodies against renin targets
Further refinement through RenLite mice for development of bispecific antibodies that can simultaneously target multiple components of the renin-angiotensin system
Utilization of RenNano mice to develop single-domain antibodies with superior tissue penetration for targeting previously inaccessible renin-expressing tissues
Therapeutic screening: Test candidate therapeutics in the Ren-2 null model to assess efficacy in a system where blood pressure regulation depends solely on Ren-1d expression
Combination therapies: Develop multi-modal approaches that target different components of the renin-angiotensin-aldosterone system simultaneously
This integrated approach enables comprehensive therapeutic development targeting the renin-angiotensin system from initial discovery through preclinical validation.
Advanced spatial mapping of renin expression requires sophisticated techniques that preserve both molecular and spatial information:
Multiplexed error-robust fluorescence in situ hybridization (MERFISH): This cutting-edge technique allows visualization and annotation of spatial locations of renin-expressing cells with single-cell resolution. The technique can be integrated with brain coordinate frameworks (like the Allen Mouse Brain Common Coordinate Framework) to precisely map expression patterns
Single-cell RNA sequencing (scRNA-seq) combined with anatomical microdissection: This approach begins with anatomically defined, guided tissue microdissections followed by scRNA-seq to create high-resolution transcriptomic profiles of renin-expressing cells
Integration of transcriptomic and spatial data: By combining scRNA-seq data with MERFISH spatial data, researchers can develop comprehensive atlases that reveal both the molecular identity and spatial distribution of renin-expressing cells
Hierarchical classification: Analysis can be structured into nested levels (e.g., classes, subclasses, supertypes, and clusters) to fully characterize the cellular diversity of renin-expressing populations
These advanced spatial mapping techniques enable unprecedented insights into the heterogeneity and regional specialization of renin-expressing cells within different tissues.
Transcription factors play a crucial role in determining cell-type specification and maintenance in renin-expressing cells. Recent comprehensive transcriptomic and spatial atlases of mouse cells have revealed:
Combinatorial transcription factor codes: Specific combinations of transcription factors define cell types across all parts of the brain and other tissues. For renin-expressing cells, these combinatorial codes determine their identity and functional properties
Regional heterogeneity: Renin-expressing cells show varying transcription factor expression patterns depending on their anatomical location, contributing to their functional specialization in different tissues
Regulatory networks: Transcription factors operate within complex regulatory networks that control the expression of renin and other components of the renin-angiotensin system
Developmental programming: Specific transcription factor cascades guide the development and maturation of renin-expressing cells during organogenesis
Understanding these transcription factor networks provides opportunities for targeted manipulation of renin expression in specific cell populations, offering potential therapeutic avenues for hypertension and other cardiovascular disorders.
When faced with contradictory renin expression data between different mouse models, researchers should employ a systematic analytical approach:
Consider genetic background effects: Different mouse strains may have varying compensatory mechanisms that affect renin expression. For instance, mice with the Ren-2 gene inactivated demonstrate different renin dynamics than single-renin gene models
Evaluate methodology-induced variations:
Analyze differential tissue expression: Renin expression varies significantly across tissues, and the Ren-1d gene has a different tissue expression profile than Ren-2
Account for active renin versus prorenin ratios: Ren-2 null mice show increased active renin but decreased prorenin levels in plasma, which might lead to seemingly contradictory results depending on what form is being measured
Validate with multiple techniques: Combine ELISA quantification with transcriptomic analysis and spatial mapping techniques like MERFISH to develop a comprehensive understanding of renin expression patterns
This structured approach helps reconcile apparent contradictions and develops a more nuanced understanding of renin biology across different mouse models.
Complex datasets from REN mouse experiments require sophisticated statistical approaches:
For transcriptomic data analysis:
Implement iterative clustering analysis for single-cell transcriptomic data to identify cell types and states
Perform pairwise cluster comparisons to identify differentially expressed genes (DEGs) associated with renin function
Apply dimensionality reduction techniques (e.g., t-SNE, UMAP) to visualize relationships between cell populations
For spatial data integration:
For phenotypic and physiological measurements:
Implement mixed-effects models to account for within-subject correlation in longitudinal blood pressure data
Use ANOVA with appropriate post-hoc tests when comparing multiple experimental groups
Apply non-parametric alternatives when data violate normality assumptions
For validation and reproducibility:
These statistical approaches ensure robust, reproducible findings from complex REN mouse experiments while minimizing false positives and appropriately handling the multilayered nature of modern biomedical data.
Single-cell technologies offer unprecedented opportunities to advance renin-angiotensin system research:
Single-cell multi-omics integration: Combining single-cell transcriptomics with proteomics and epigenomics will provide comprehensive molecular profiles of renin-expressing cells. This integration can reveal how transcriptional, post-transcriptional, and epigenetic mechanisms collectively regulate renin expression and function
Spatial transcriptomics advancement: Further development of MERFISH and other spatial technologies will enable three-dimensional mapping of renin-expressing cells with subcellular resolution, revealing the precise tissue architecture and cellular neighborhoods that influence renin production
Temporal dynamics analysis: Applying single-cell RNA velocity analysis to REN mouse data can reveal developmental trajectories and dynamic state transitions of renin-expressing cells under various physiological and pathological conditions
Single-cell CRISPR screens: Implementing pooled CRISPR screens at single-cell resolution in REN mouse models will enable systematic identification of genes that regulate renin expression and function
Computational modeling: Integrating single-cell data with physiological measurements will enable the development of multi-scale computational models that predict how cellular-level changes in renin expression propagate to systemic effects on blood pressure regulation
These advanced single-cell approaches will provide unprecedented insights into the cellular and molecular heterogeneity underlying the renin-angiotensin system, potentially revealing new therapeutic targets for cardiovascular and renal diseases.
Several cutting-edge genetic engineering approaches show promise for enhancing REN mouse models:
CRISPR-Cas9 base editing and prime editing: These precise genome editing technologies allow for seamless introduction of specific mutations in renin genes without double-strand breaks, enabling the creation of more refined disease models that mimic human renin polymorphisms
Conditional and inducible renin expression systems: Advanced genetic switches can enable temporal and spatial control of renin expression, allowing researchers to study acute versus chronic effects of renin dysregulation in specific tissues
Humanized renin-angiotensin system models: Creating mice with fully humanized renin-angiotensin components would better recapitulate human biology and improve translational relevance of findings
Reporter systems for live imaging: Integrating fluorescent or bioluminescent reporters into endogenous renin loci would enable real-time, non-invasive monitoring of renin expression dynamics in living animals
Tissue-specific humanization: Development of tissue-selective REN humanized mice that express human renin only in relevant tissues (kidney juxtaglomerular cells, brain, adrenal gland) would provide more precise models for studying tissue-specific renin functions
These emerging genetic engineering approaches hold tremendous potential for creating next-generation REN mouse models that more accurately reflect human renin biology and pathophysiology, accelerating therapeutic discovery for hypertension and related cardiovascular disorders.
Renin, also known as REN or angiotensinogenase, is a circulating enzyme that plays a crucial role in the body’s renin-angiotensin system (RAS). This system is essential for regulating blood pressure and fluid balance in the body. The recombinant form of renin from mice is often used in research to study its functions and potential therapeutic applications.
The gene encoding renin is localized on mouse chromosome 1. In recombinant studies, renin is typically expressed in HEK 293 cells, which are human embryonic kidney cells. This expression system is chosen because it allows for proper folding and post-translational modifications of the protein, ensuring that the recombinant renin closely mimics the natural enzyme .
Renin is a protease enzyme with a calculated molecular weight of approximately 43.2 kDa. However, due to glycosylation, the observed molecular weight can range between 45-55 kDa when analyzed by SDS-PAGE under reducing conditions. The enzyme is often tagged with a 6-His tag at the C-terminus to facilitate purification .
Renin’s primary function is to activate the renin-angiotensin system by cleaving angiotensinogen, a protein produced by the liver, to yield angiotensin I. Angiotensin I is then converted into angiotensin II by the angiotensin-converting enzyme (ACE), primarily within the capillaries of the lungs. Angiotensin II is a potent vasoconstrictor that increases blood pressure and stimulates the release of aldosterone, which promotes sodium retention by the kidneys .
Renin plays an essential role in the elevation of arterial blood pressure and increased sodium retention by the kidney. It is a key regulator of blood pressure and electrolyte balance, making it a critical target for antihypertensive therapies. Dysregulation of renin activity can lead to conditions such as hypertension and congestive heart failure .
Recombinant mouse renin is widely used in research to study the mechanisms of blood pressure regulation and to develop new treatments for hypertension and related cardiovascular diseases. It is also used in the development of renin inhibitors, which are a class of antihypertensive drugs that directly inhibit the activity of renin .