Fact-checked by Grok 2 weeks ago

Genetically modified mouse

A genetically modified mouse is a in which the has been deliberately altered through the insertion, deletion, or replacement of specific DNA sequences using technology, enabling precise control over to mimic genetic conditions or study biological processes. These modifications typically involve techniques such as pronuclear microinjection of foreign DNA into fertilized eggs for creating transgenic mice, which express an added gene, or targeted gene disruption via homologous recombination to produce knockout mice, where a specific gene is inactivated. The development of such models began in the 1970s, with pioneering work in 1974 demonstrating the integration of viral DNA into mouse embryos, leading to the first stably heritable transgenic mice in the early 1980s. Genetically modified mice have revolutionized biomedical research by serving as versatile tools for investigating functions, mechanisms, and therapeutic interventions, with notable examples including the engineered in 1983 to model cancer through oncogene insertion. Applications span fields like , , and infectious s, where models such as human ACE2-expressing mice have been used to study viral pathogens like SARS-CoV-2. Advances in CRISPR-Cas9 technology since the have further streamlined the creation of these models, enhancing precision and reducing time compared to earlier methods. Key ethical and practical considerations include maintaining genetic stability across generations and adhering to institutional guidelines for , ensuring these models contribute reliably to scientific progress without undue harm.

Overview

Definition and Characteristics

A is a of the species Mus musculus in which the has been deliberately altered using to insert, delete, or modify specific , primarily for purposes. These alterations enable researchers to study , regulatory mechanisms, and biological processes in a controlled mammalian model. The first were created in 1974 through the insertion of viral DNA into early-stage embryos. Genetically modified mice are typically derived from inbred strains such as , which provide genetic uniformity and reproducibility in experiments. Modifications can be , affecting heritable changes passed to , or , limited to specific tissues in the individual animal. Common examples include transgenic models, where genes are overexpressed to mimic gain-of-function scenarios, and knockout models, where genes are inactivated to create loss-of-function effects. The mouse genome consists of approximately 2.6 billion base pairs and encodes around 20,000 protein-coding genes, facilitating precise targeting of modifications. Mice exhibit a short generation time of 3-4 months, from birth to producing the next generation, along with high reproductive rates and ease of maintenance in laboratory settings compared to larger mammals. They are preferred over other genetically modified animals due to their small size, which reduces housing costs and enables large-scale studies, as well as an established infrastructure for breeding and genetic analysis. Additionally, mice share about 85% sequence identity in protein-coding regions with humans and exhibit conserved synteny across approximately 90% of their genomes, making them highly relevant for modeling human biology.

Importance in Research

Genetically modified mice play a pivotal role in elucidating by allowing researchers to precisely manipulate specific , thereby observing resulting phenotypes, molecular pathways, and interactions within a whole living . Techniques such as knockouts eliminate a to reveal its roles, while transgenics introduce or overexpress to study regulatory sequences and disease mechanisms, such as in models of immune deficiencies or tumor development. This precision has advanced understanding of complex biological processes, including developmental pathways like those involving Hox and Wnt , and multifactorial conditions like and . Their contributions extend to landmark discoveries in biomedical science, notably enabling the 2007 Nobel Prize in Physiology or Medicine awarded to Mario R. Capecchi, Sir Martin J. Evans, and for developing principles of targeted modification in mice using embryonic stem cells. This breakthrough facilitated the creation of "knockout" mice, transforming research on roles in and and underpinning advancements in fields like , , and . Compared to other models, genetically modified mice offer key advantages as ethical alternatives to experimentation, with their small size, short (19-21 days ), and low maintenance costs making them more practical and economical than larger animals like dogs or non- . Sharing approximately 95% genetic with s, they effectively mimic and disease processes, serving as a vital complement to cell cultures or computational simulations by providing systemic insights into gene-environment interactions. The economic significance of genetically modified mouse research is profound, with the global mice model market valued at $1.53 billion in 2024 and projected to grow to $2.74 billion by 2030, reflecting substantial annual investments in this foundational tool for and basic science. Infrastructure like , a leading provider of mouse strains and services, bolsters this ecosystem with an operating budget of $628 million in 2023 and generates over $1.2 billion in regional economic impact through employment, vendor support, and research facilitation.

History

Early Pioneering Work

The foundational concepts for genetically modified mice emerged from earlier advances in manipulation, particularly in amphibians during the . British developmental biologist demonstrated that nuclei from differentiated cells of tadpoles could be transplanted into enucleated eggs, resulting in viable, fertile adult s that were genetically identical to the donor, thus proving the totipotency of nuclei and inspiring similar efforts in mammals. This work laid the groundwork for mammalian experiments by showing that genetic material could be introduced and reprogrammed in early embryonic stages. In the late 1960s and early 1970s, pioneering researchers began adapting these techniques to . Beatrice Mintz developed embryo aggregation methods, fusing early-stage embryos from different genetic backgrounds to create chimeric "allophenic" , which demonstrated the developmental potential of combined cell populations and provided a model for studying in mosaics. Simultaneously, Brinster advanced pronuclear techniques, initially using them to introduce mRNA into fertilized eggs, enabling the study of function and setting the stage for DNA delivery despite initial limitations in efficiency. A landmark achievement occurred in 1974 when Rudolf Jaenisch and Beatrice Mintz injected SV40 viral DNA into preimplantation mouse blastocysts, producing healthy adult mice with integrated viral sequences in their somatic tissues, marking the first creation of a genetically modified mammal. However, this integration was not heritable, as the DNA was not transmitted to the germline. Throughout the 1970s, researchers faced significant challenges, including low rates of DNA integration—often below 10%—and unpredictable insertion sites, leading to a reliance on viral vectors like SV40 for delivery and the refinement of microinjection to improve uptake in embryos. These early efforts, though inefficient, established critical proof-of-principle for foreign DNA incorporation in mammals.

Major Milestones and Advancements

In 1982, Ralph Brinster and Richard Palmiter achieved the first stable transmission of a foreign in mice through pronuclear of a metallothionein- (MT-hGH) fusion , demonstrating that injected DNA could integrate into the host genome and be passed to offspring. This breakthrough enabled the production of transgenic mice expressing the human , resulting in dramatically enhanced growth rates—up to twice the size of non-transgenic littermates—earning them the nickname "supermice" and establishing pronuclear injection as a foundational technique for transgenesis. These early experiments highlighted the potential for overexpressing specific to study physiological effects, paving the way for broader applications in . The late 1980s marked a pivotal advancement with the creation of the first knockout mice using in embryonic stem (ES) cells, independently developed by , , and . In 1989, Evans' group reported the first targeted disruption of the HPRT in ES cells, which were then used to generate chimeric mice transmitting the mutation through the germline, allowing precise inactivation of specific to model loss-of-function phenotypes. This gene-targeting method revolutionized mammalian by enabling the study of function , far surpassing earlier random insertion techniques. Their contributions were recognized with the 2007 in Physiology or Medicine for "discoveries of principles for introducing specific modifications in mice by the use of embryonic stem cells." During the 1990s and 2000s, specialized transgenic models proliferated, including the "," patented by in 1988 for its insertion of an activated (such as or ) that predisposed mice to tumor development, providing an early platform for . Concurrently, humanized mice emerged with the introduction of (HLA) transgenes, such as HLA-A2 and , starting in the early 1990s, which allowed modeling of human immune responses by expressing human MHC molecules in murine backgrounds to study and T-cell interactions. The Cre-loxP recombination system, adapted from bacteriophage P1 in the early 1990s, saw a significant rise in adoption for conditional knockouts after , enabling tissue- and time-specific gene deletion to circumvent embryonic lethality and refine phenotypic analysis. This inducible approach, often combined with tamoxifen-activated Cre drivers, facilitated over 10,000 Cre mouse lines by the mid-2010s, transforming studies of complex diseases like neurodegeneration and cancer by isolating gene effects in adult tissues. By the 2010s and into the 2020s, genetically modified mice integrated advanced tools like and , enhancing circuit-level and molecular resolution in models of neural and immune disorders. Transgenic lines expressing under cell-type-specific promoters, developed from 2013 onward, allowed precise optical control of neuronal activity , revealing roles in and disease. Complementing this, single-cell sequencing applied to GM mouse tissues since the mid-2010s has mapped heterogeneous responses in disease models, such as enteric nervous system development, enabling identification of rare cell states and therapeutic targets up to 2025.

Methods of Genetic Modification

Transgenic Approaches

Transgenic approaches represent classical methods for generating genetically modified mice by introducing exogenous DNA into the genome, primarily to achieve gain-of-function through gene overexpression. These techniques typically involve random integration of transgenes, enabling the study of gene function in vivo. The first successful production of transgenic mice occurred in 1980 via pronuclear microinjection, marking a foundational advancement in mammalian transgenesis. Pronuclear microinjection entails the direct injection of linear DNA constructs into the pronuclei of fertilized mouse zygotes, allowing random into the genome during early embryonic development. This results in multiple copy insertions at unpredictable sites, with integration efficiencies typically ranging from 1% to 5% of injected embryos yielding transgenic founders. Following injection, embryos are transferred to pseudopregnant surrogate mothers, and founder mice are screened for transgene presence via Southern blotting or . The approach's simplicity has made it a for creating overexpression models, though it requires substantial embryo numbers due to low success rates. Another established transgenic method involves transposon systems, such as or piggyBac, which facilitate efficient integration of transgenes via cut-and-paste or copy-and-paste mechanisms. These systems, revived or developed in the 1990s and 2000s, achieve integration efficiencies of 20-40% in some protocols, higher than traditional , and allow for large-scale production of transgenic lines with reduced mosaicism. Transposon vectors are electroporated or injected into zygotes, with the enzyme promoting excision and insertion at TA dinucleotide sites (for piggyBac). This approach is particularly useful for screens and has been applied to generate reporter lines and disease models, though it can cause semi-random integrations leading to variable expression. Viral vector methods utilize retroviruses or lentiviruses to deliver transgenes for stable genomic integration, offering higher efficiency than , often exceeding 50% in some protocols. Retroviral vectors, employed since the early , insert DNA up to 7-8 kb but carry risks of by disrupting endogenous genes. Lentiviral vectors, an evolution of this technique, enable integration into non-dividing cells like oocytes, facilitating transmission with reduced pathogenicity through self-inactivating designs. These methods are particularly useful for large-scale transgenesis but demand careful vector pseudotyping to avoid immune responses . Promoter-driven expression systems enhance control over activity by incorporating specific regulatory elements, such as tissue-specific or inducible promoters, to direct expression patterns. For instance, the Tet-On/Off system uses a tetracycline-responsive promoter to modulate via administration, allowing reversible activation or repression in transgenic mice. The locus serves as a "safe harbor" site for targeted integration via or site-specific recombinases, minimizing disruptions and ensuring ubiquitous, stable expression across generations. Representative applications include fluorescent reporter lines, such as (GFP) overexpression models developed in the 1990s, which enable non-invasive tracking of cells and tissues in live animals. Despite their utility, transgenic approaches are limited by position effects, where the integration site's chromatin environment causes variable or silenced expression across lines. Additionally, founder mice often exhibit mosaicism, with incomplete transgene transmission to all cells, necessitating breeding to homozygous lines for consistent phenotypes. These challenges underscore the need for screening multiple founders to identify optimal lines for research.

Gene Targeting Techniques

Gene targeting techniques in mice primarily rely on to achieve precise modifications at specific genomic loci, enabling loss-of-function or gain-of-function alterations in embryonic stem (ES) cells. This approach, distinct from random integration methods, allows researchers to disrupt, replace, or insert sequences with high specificity, facilitating the creation of genetically modified mouse models for studying . ES cells, derived from the of mouse blastocysts typically from the 129 strain, are cultured and maintain pluripotency, enabling their genetic manipulation and subsequent contribution to all tissues, including the . These cells are transfected via with linear DNA targeting vectors, followed by selection to identify successfully modified clones. Once targeted ES cells are isolated, they are injected into wild-type blastocysts to form chimeric embryos, which are implanted into pseudopregnant females; high-contribution chimeras are then bred to transmit the modification through the , yielding heterozygous mice. Homologous recombination involves the exchange of genetic information between the targeting vector and the endogenous chromosomal locus, mediated by cellular repair machinery. The vector typically consists of a linear DNA fragment with two homology arms (1-10 kb each) flanking the desired modification, which align with the target sequence to promote double-strand break repair and replacement of the endogenous DNA. To enhance detection of rare targeting events, positive-negative selection is employed: the vector includes a neomycin resistance (neoR) cassette for positive selection of cells with vector integration, and a herpes simplex virus thymidine kinase (HSV-TK) gene for negative selection using ganciclovir, which kills cells with random non-homologous integrations while sparing those with precise recombination that excises the TK gene. This strategy, developed in the late 1980s, dramatically enriches for targeted clones. For knockouts (KO), homologous recombination disrupts gene function by deleting or inserting sequences that abolish protein expression, such as replacing critical exons with a selectable marker. A classic example is the disruption of the hypoxanthine phosphoribosyltransferase () gene, modeling Lesch-Nyhan syndrome, achieved by inserting a neoR cassette into an exon. Conditional knockouts extend this by incorporating loxP sites—short 34-bp recognition sequences—flanking essential gene regions; upon breeding with Cre recombinase-expressing mice, tissue- or time-specific recombination deletes the flanked sequence, allowing study of gene function without embryonic lethality. Knockins (KI) enable precise insertion of exogenous sequences, such as point mutations or reporter s, into the endogenous locus to mimic human disease alleles while preserving native regulatory elements. For instance, KI models of insert human amyloid precursor protein () mutations like (K670N/M671L) and (E693G) into the murine App via homology arms encompassing the mutation sites, resulting in amyloid-beta pathology without overexpression artifacts. The efficiency of in ES cells is inherently low, typically occurring in 1 in 10^3 to 10^6 transfected cells, depending on factors like arm length and isogenic DNA matching between vector and host genome, which can improve rates up to 100-fold. Despite this, the method's specificity has enabled the targeted mutation of several thousand mouse genes since its inception. The foundational development of these techniques occurred in the and , with establishing totipotent ES cell lines from mouse blastocysts in 1981, providing the cellular platform for targeting. advanced vector design and selection strategies, demonstrating the first targeted in mouse ES cells in 1987 by correcting the HPRT locus. contributed early insights into recombination mechanisms, and by 1989, the first germline-transmitting KO mice were generated, revolutionizing mammalian . This work culminated in the 2007 in or for Evans, Capecchi, and Smithies.

Modern Genome Editing Tools

The CRISPR-Cas9 system, adapted from bacterial adaptive immunity, emerged as a transformative tool for in the early 2010s, enabling precise and efficient modifications in mouse genomes without relying on embryonic stem cell-based methods. Initially demonstrated for programmable DNA cleavage in 2012, it was rapidly applied to mammalian cells and, by 2013, used to generate the first knockout mice through direct injection into zygotes. In a landmark study, Wang et al. reported the one-step creation of mice with mutations in multiple genes, including Tet1, Tet2, and Tet3, achieving biallelic disruptions in up to 80% of founder animals. At its core, CRISPR-Cas9 employs a single (gRNA) to direct the endonuclease to a specific genomic locus, where it induces a double-strand break (DSB) adjacent to a (PAM) sequence, typically NGG. Cellular repair of the DSB occurs primarily through (NHEJ), which introduces insertions or deletions (indels) to disrupt function for knockouts, or (HDR) in the presence of a donor template for precise knock-ins. This mechanism allows for versatile editing directly in one-cell embryos, bypassing the need for laborious in embryonic stem cells. Key advantages of CRISPR-Cas9 in genetic modification include its high efficiency, often yielding 50-80% rates in targeted loci, and its capacity for , where multiple gRNAs enable simultaneous editing of several genes in a single injection. For instance, this approach facilitates rapid generation of compound mutants, reducing timelines from years to months compared to traditional methods. Additionally, zygotic injection supports editing, producing germline-transmissible modifications with minimal mosaicism when optimized. Subsequent variants have expanded CRISPR's precision and scope in mice. Cas12a (formerly Cpf1) offers alternative PAM requirements (T-rich), enabling targeting of sites inaccessible to Cas9, and has been used to generate knock-in mice with efficiencies up to 40% via . Base editors, fusing Cas9 nickase to deaminases, enable single-nucleotide conversions (e.g., C-to-T) without DSBs, minimizing indels and achieving up to 70% editing in mouse embryos for modeling point mutations. Prime editing, introduced in 2019, further refines this by using a fused to a pegRNA for insertions, deletions, or base changes with over 50% efficiency in mice, as demonstrated in models of genetic disorders. In the 2020s, advancements have integrated with high-throughput and computational tools for mouse research. In vivo screens, using pooled gRNA libraries delivered via adeno-associated viruses, have identified gene functions in native tissues, with efficiencies enabling screens of thousands of genes in adult mice. Humanized organoids engrafted into immunodeficient mice, edited via , have modeled patient-specific responses, achieving targeted knockouts in up to 90% of cells. AI-optimized gRNA design algorithms have improved prediction of off-target effects, enhancing the specificity of mouse model generation. Representative examples illustrate CRISPR's impact, such as the rapid production of / double-knockout mice via multiplexed injection, yielding intestinal tumors in founders within weeks to cooperation. These tools have thus accelerated the creation of complex genotypes, enhancing the utility of in dissecting genetic interactions.

Applications

Disease Modeling

Genetically modified serve as critical tools for replicating pathologies, enabling researchers to investigate mechanisms, progression, and genetic interactions in a controlled environment. These models, often referred to as genetically engineered mouse models (GEMMs), incorporate specific genetic alterations to mimic states, providing insights into and that are challenging to obtain from alone. By expressing -associated genes or disrupting murine equivalents, GEMMs facilitate the of initiation, maintenance, and tissue-specific effects, with applications spanning multiple categories. In , GEMMs have been instrumental in modeling tumor initiation, progression, and . The , developed in 1984 by introducing the SV40 T-antigen under the control of the mammary tumor virus (MMTV) promoter, was one of the first transgenic models to spontaneously develop mammary tumors, establishing a foundation for studying -driven . Subsequent models, such as the MMTV-PyMT transgenic , express the polyomavirus middle T antigen in mammary , recapitulating multistage progression including ductal , , and invasive with lung , closely mirroring disease and timeline. For , the Kras^{G12D} conditional model activates oncogenic Kras mutations in lung epithelial cells, leading to formation and progression to upon combination with tumor suppressors like Lkb1 loss, which induces aggressive, metastatic disease akin to KRAS-mutant cancers. These models highlight how GEMMs elucidate cooperative genetic events in tumorigenesis. Neurological disorders benefit from GEMMs that replicate , neuronal loss, and behavioral deficits observed in human patients. In , the APP/PS1 knock-in model expresses human amyloid precursor protein (APP) with the Swedish mutation and (PS1) with the deltaE9 mutation, resulting in early-onset -beta plaque deposition, synaptic dysfunction, and cognitive impairments starting at 6-8 months, providing a platform to study pathology and . For , overexpression of human under the PDGF-beta promoter leads to Lewy body-like inclusions, degeneration in the , and motor deficits, mimicking alpha-synucleinopathy progression. is modeled by the R6/2 transgenic mouse, which expresses the first of the human gene with an expanded repeat (approximately 150 repeats), causing progressive motor dysfunction, striatal atrophy, and ubiquitinated inclusions by 4-6 weeks, allowing investigation of polyglutamine toxicity. Metabolic diseases are effectively modeled through targeted disruptions that induce hallmark physiological changes. The , generated by homozygous of the gene, exhibits hyperphagia, severe , , and from birth, serving as a cornerstone for understanding signaling in and obesity-related complications. Similarly, () mice develop spontaneous and accelerated on a chow diet, with extensive aortic plaque formation by 3-6 months, replicating human lipid-driven vascular pathology and enabling studies of plaque progression and . For infectious diseases, humanized immune system mice address the limitations of standard mice by engrafting human hematopoietic cells into immunodeficient backgrounds. The NOD-scid IL2rgamma null (NSG) strain, lacking mature lymphocytes and natural killer cells, supports robust human immune reconstitution when combined with mutations in the signal regulatory protein alpha (SIRPα) gene, which reduces macrophage-mediated rejection of human cells. These NSG-SIRPα models sustain human T cells, B cells, and myeloid lineages, allowing chronic HIV-1 infection with CD4+ T cell depletion, viremia, and immune activation similar to human AIDS progression; they also support hepatitis C virus replication in human hepatocytes, facilitating studies of viral persistence and liver pathology. Despite their utility, GEMMs face fidelity challenges due to species differences, such as the absence of certain immune genes (e.g., orthologs) in mice, which limits accurate modeling of human-specific immune responses in diseases like or . Additionally, constitutive mutations often cause embryonic lethality or compensatory adaptations, necessitating conditional models using Cre-loxP recombination to induce alterations post-development in specific tissues, thereby improving physiological relevance. Recent advancements integrate patient tumor into GEMMs, enhancing precision by engineering mice with individualized mutations identified via sequencing. For instance, /Cas9-edited GEMMs incorporating patient-derived oncogenic variants allow recapitulation of tumor heterogeneity and evolution, bridging genomic data with in vivo disease dynamics for personalized mechanistic studies.

Biomedical and Pharmaceutical Research

Genetically modified mice play a pivotal role in preclinical testing for drug efficacy and , enabling researchers to evaluate therapeutic responses in models that mimic . For instance, (KO) models have demonstrated the effectiveness of EGFR inhibitors in treating lung adenocarcinoma; mice engineered to express EGFR mutants develop tumors highly sensitive to these inhibitors, validating their mechanism before clinical trials. Similarly, humanized liver chimeric mice, such as PXB-mice, accurately predict by incorporating human hepatocytes, allowing assessment of drug clearance and that differs from wild-type mice. In , (CYP) humanized mice enhance the prediction of human-specific and adverse effects, addressing discrepancies between murine and human pathways. These models, expressing human CYP enzymes like and , replicate human drug-induced toxicities more reliably than standard mice, facilitating safer candidate selection by identifying reactive metabolites that cause or other organ damage. For example, P450-humanized mice have been used to study drug-drug interactions and predict idiosyncratic toxicities, improving the accuracy of risk assessments in pharmaceutical development. Genetically modified mice have accelerated development and evaluation, particularly during the . Transgenic mice expressing human (hACE2), such as K18-hACE2 models, were instrumental in 2020 for testing vaccine candidates; these mice supported SARS-CoV-2 infection and replication, enabling efficacy assessments of mRNA vaccines that provided long-term protection against respiratory challenges. In immunotherapy, PD-1 mice have been essential for testing checkpoint inhibitors, revealing enhanced anti-tumor immunity through increased T-cell activation and reduced myeloid suppression, which informed clinical strategies for cancers like . Notable success stories underscore the impact of these models in drug validation. , a breakthrough for chronic (CML), was preclinically confirmed using BCR-ABL transgenic mouse models; these mice recapitulated the disease and showed tumor regression upon imatinib treatment, directly supporting its approval and transforming CML therapy. Many drugs that show promise in preclinical models fail in clinical trials due to species differences in , immune responses, and progression. To address this, approaches combining patient-derived xenografts (PDX) with genetically engineered models (GEMMs) are emerging, integrating patient-specific tumors into engineered backgrounds to better capture heterogeneity and resistance mechanisms for more predictive drug screening. As of , AI-driven phenotyping is accelerating drug screens in genetically modified mice by automating behavioral and physiological analysis, reducing manual effort and enabling high-throughput evaluation of treatment effects. These tools, leveraging for video-based tracking and detection, have shortened discovery timelines and supported FDA initiatives to minimize while enhancing precision in efficacy predictions.

Other Scientific Uses

Genetically modified mice have proven invaluable in , particularly through optogenetic approaches that enable precise mapping of neural circuits. Transgenic mice expressing channelrhodopsin-2 (ChR2) under cell-type-specific promoters allow light-activated stimulation of targeted neurons, facilitating the dissection of circuit functions . For instance, Cre-dependent ChR2 mouse lines have been used to stimulate specific cortical layers or subcortical projections, revealing patterns and synaptic dynamics that underpin and . models further elucidate behavioral roles; mice lacking stathmin, a enriched in the , exhibit impaired , demonstrating reduced freezing responses to conditioned stimuli and highlighting stathmin's role in modulating anxiety and learned fear via altered dynamics in amygdalar neurons.00875-5) In , Hox gene knockouts have illuminated the molecular basis of formation. Targeted disruption of paralogous groups, such as Hox10 and Hox11, results in profound skeletal transformations, including homeotic shifts where adopt thoracic identities or sacral elements mimic lumbar ones, underscoring the genes' redundant yet essential roles in specifying regional identities along the anterior-posterior axis. These models reveal how Hox dosage and combinatorial expression dictate vertebral morphology and limb positioning, providing foundational insights into evolution and congenital anomalies. Environmental science benefits from GM mice engineered for pollutant sensitivity, notably aryl hydrocarbon receptor (AhR) transgenics. Constitutively active AhR mouse lines mimic chronic exposure, exhibiting heightened sensitivity to environmental toxins like 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), with accelerated hepatocarcinogenesis and altered expression, enabling studies on dioxin-mediated endocrine disruption and immunotoxicity. In , reporter mouse lines with fluorescent or enzymatic tags driven by ubiquitous or tissue-specific promoters support of genetic perturbations. For example, Cre reporter strains expressing β-galactosidase or GFP allow rapid visualization of recombination efficiency across regions, accelerating the validation of tools and drug candidates . Similarly, (TERT)-overexpressing mice, often termed "supermice," display extended longevity, with median lifespan increases of up to 40% and delayed aging phenotypes like improved glucose tolerance, without elevating cancer risk.01191-4) GM mice also provide proofs-of-concept for agricultural applications in . Transgenic strategies first validated in mice, such as pronuclear injection for overexpression, informed the development of enhanced-milk or disease-resistant models, demonstrating feasibility for traits like increased muscle mass or before scaling to larger . Emerging uses include , where NASA-funded studies employ chimeric or mice to model . These models exposed to simulated galactic cosmic rays assess and repair mechanisms, revealing differential tumor susceptibility and informing countermeasures for astronaut health during long-duration missions in the 2020s.

Ethical and Regulatory Aspects

Animal Welfare Concerns

Genetically modified mice used in disease modeling often experience significant pain and distress due to the phenotypes engineered to mimic human conditions. For instance, in the SOD1-G93A transgenic mouse model of (), animals develop progressive hind limb weakness starting at 3-4 months of age, leading to full and a moribund state by approximately 5 months, resulting in severe suffering from mobility loss and . Similarly, models inducing tumors or neurodegeneration, such as those for cancer or , can cause from tumor growth or neuronal damage, necessitating welfare refinements like administration of analgesics or implementation of early humane endpoints to minimize prolonged distress. The 3Rs principle—Replacement, Reduction, and Refinement—plays a central role in addressing in genetically modified mouse research, though its application reveals trade-offs. While these mice enable by providing precise models that decrease the overall number of animals needed compared to less targeted approaches, they can introduce novel challenges requiring Refinement, such as monitoring for unexpected issues or optimizing to alleviate . efforts focus on non-animal alternatives where possible, but the inherent in many GM models underscores ongoing ethical debates about balancing scientific gains with animal well-being. Phenotypic variability in genetically modified mice can lead to unintended welfare effects beyond the targeted modification, complicating ethical oversight. For example, ( -/-) mice exhibit increased anxiety-like behaviors and depressive phenotypes, including heightened baseline anxiety and reduced exploratory activity, which may arise from compensatory changes in brain serotonin systems and contribute to . Such off-target effects highlight the need for comprehensive to identify and mitigate risks not directly related to the goal. Animal welfare advocacy groups, including the , actively promote alternatives to use, emphasizing the development of models and computational simulations to reduce reliance on mice prone to . Studies on impacts indicate that a substantial proportion of genetically modified mice—often classified under moderate to severe severity categories—experience compromised well-being due to genetic alterations, with reports noting frequent occurrences of pain, distress, or behavioral abnormalities in production and experimental lines. By 2025, welfare scoring systems and harm-benefit analyses have become mandatory in many laboratories, particularly under , requiring prospective evaluation of potential harms to against expected scientific benefits to ensure ethical justification. These frameworks, including multidimensional assessments of behavioral and physiological indicators, facilitate standardized monitoring and refinement to protect animal well-being throughout the research lifecycle. In the United States, oversight of genetically modified mouse research primarily falls under the Public Health Service (PHS) Policy on Humane Care and Use of Laboratory Animals, enforced by the National Institutes of Health's Office of Laboratory Animal Welfare (OLAW), established in 2000 to ensure compliance with animal welfare standards in federally funded research. There is no specific federal law regulating genetically modified animals as a distinct category, but all institutions receiving PHS funding must establish Institutional Animal Care and Use Committees (IACUCs) to review and approve protocols involving live vertebrates, including the creation and use of transgenic or knockout mice, with requirements for minimizing pain and distress. Additionally, the NIH Guidelines for Research Involving Recombinant or Synthetic Nucleic Acid Molecules govern the biosafety of transgenic rodent creation, classifying it under containment levels (e.g., Biosafety Level 1 for most standard strains) to prevent unintended release or environmental impact. In the , Directive 2010/63/EU on the protection of animals used for scientific purposes establishes a comprehensive framework for research, requiring prospective project authorization by competent authorities that includes a detailed and harm-benefit . The directive mandates severity of procedures (non-recovery, mild, moderate, severe) and prohibits those expected to cause severe or long-lasting pain, suffering, or distress without overriding scientific justification, particularly relevant for models involving genetic alterations that may induce phenotypes with moderate to severe effects, such as cancer or neurological disorders in mice. Member states implement these rules through national legislation, with additional guidance from the European Commission's Framework for genetically altered animals, emphasizing containment, genetic stability monitoring, and post-authorization inspections. Internationally, efforts by the (WHO) and the () promote harmonized biosafety standards for laboratory-contained genetically modified organisms, including mice, through guidelines that focus on , containment practices, and information sharing to facilitate cross-border collaboration while protecting human health and the environment. The , adopted in 2000 under the , regulates the transboundary movement, handling, and use of living modified organisms (LMOs) to prevent adverse effects on , but laboratory-contained research with GM mice is generally exempt as it does not involve intentional environmental release. Intellectual property frameworks have significantly influenced the sharing and commercialization of genetically modified mouse strains, exemplified by the Harvard (U.S. Patent No. 4,736,866), granted in as the first U.S. for a animal engineered to carry an activated for , which licensed exclusive rights to and raised concerns about restricting academic access to proprietary strains. This precedent, upheld in various jurisdictions despite ethical debates, has led to policies encouraging material transfer agreements and repositories like the to balance innovation with collaborative research. As of 2025, stricter measures for -based genetic modifications in animals stem from updated U.S. dual-use research of concern (DURC) policies, revised via a May 2025 to enhance oversight of experiments with potential misuse risks, influenced by post-2020 concerns over technologies that could amplify pathogen transmission or ecological disruptions. These policies require federal funding agencies to screen applications in model organisms like mice for dual-use potential, mandating risk mitigation plans and institutional committee reviews to address off-target effects and failures.

Future Directions

Emerging Technologies

Recent advancements in genetically modified mouse models have integrated organoid and chimera technologies to enhance human-relevant disease modeling and organ generation. Human-mouse chimeras, created by injecting human induced pluripotent stem (hiPS) cells into mouse blastocysts, have demonstrated the potential for human cells to contribute to mouse embryonic development, as shown in a 2017 study where human embryonic stem cells integrated into both embryonic and extraembryonic lineages upon inhibition. complementation further enables the generation of functional organs, such as livers, by injecting human pluripotent stem cells into mouse blastocysts lacking specific organogenic genes, allowing host developmental cues to direct human cell differentiation into complex tissues. These approaches build on CRISPR-based modifications in mice to create niches for human organoids, offering scalable platforms for studying human and transplantation potential. Multi-omics techniques are transforming the analysis of genetically engineered models (GEMMs) by providing high-resolution insights into mechanisms. Single-cell RNA sequencing (scRNA-seq) applied to GEMMs of pancreatic ductal has revealed cellular heterogeneity, identifying distinct molecular subtypes of cancer cells, fibroblasts, and macrophages that drive tumor progression. complements this by mapping , as demonstrated in α-synucleinopathy models where it uncovered neuron-specific transcriptional changes around pathological aggregates, linking to neurodegenerative processes. Integrating these with CRISPR-modified GEMMs enables precise dissection of genetic perturbations at single-cell and tissue levels, accelerating the identification of therapeutic targets. Artificial intelligence and are streamlining analysis and management in GM mouse research. algorithms, such as those evaluated for predicting phenotypes from genomic and environmental data, have shown superior accuracy over traditional statistical methods in forecasting traits in mouse models, aiding in the prioritization of genotypes for . Robotic systems for automated and , including centralized platforms that handle cage changes and , have increased colony productivity by up to 50% in multi-strain facilities, reducing and labor while maintaining welfare standards. These tools, often integrated with workflows, enable high-throughput validation of modifications. Adeno-associated virus (AAV) vectors are advancing gene editing in adult GM mice, offering targeted delivery beyond embryonic stages. AAV-mediated delivery in adult mice has achieved efficient in the via systemic injection, with serotypes like AAV9 enabling blood-brain barrier crossing for neurological disease correction. In hemophilia models, AAV-ZFN systems edited the locus to express therapeutic proteins, restoring clotting function long-term without off-target effects. This approach extends GM mouse utility to post-developmental therapies, simulating human clinical scenarios. By 2025, microbiome-modified strains are pushing boundaries in precision neuroscience. Microbiome-modified strains, such as germ-free mice colonized with specific bacterial consortia, have elucidated gut-brain axis dynamics, showing strain-specific behavioral changes in autism spectrum disorder models that highlight microbiota's role in neural modulation. These innovations promise unprecedented sensitivity in tracking GM mouse phenotypes for complex trait studies.

Challenges and Opportunities

Despite significant advancements in genetically modified (GM) mouse technology, notable challenges persist due to inherent biological and technical limitations. One primary hurdle is the genetic divergence between mice and s, particularly in the , where differences in immune cell composition, receptor diversity, and response pathways can limit the applicability of mouse models to human diseases. For instance, mice lack certain human-specific immune components, such as diverse T-cell receptors and mucosal immune responses, which complicates the study of human immunobiology . Additionally, generating custom GM mouse lines remains costly, with estimates for full , including , , and validation, often exceeding $50,000 per line due to expenses in targeting, CRISPR/Cas9 implementation, and breeding. Off-target effects in genome tools like /Cas9 further pose risks, as unintended mutations at non-target sites can alter gene function and confound experimental outcomes, with early studies reporting off-target cleavages at loci with up to 4 mismatches in mouse embryos. Translational gaps exacerbate these issues, as only a small fraction of therapies validated in models successfully advance to clinical applications. Recent analyses indicate that while approximately 50% of animal-tested therapies progress to initial studies, just 5% achieve regulatory approval, highlighting a success rate of 10-20% or lower for transitioning from preclinical data to clinical trials, often due to species-specific physiological differences. Variability across strains adds another layer of complexity, as genetic differences—such as structural variants and allelic polymorphisms—can lead to divergent phenotypes and immune responses, reducing and generalizability in . Opportunities abound to address these challenges and expand the utility of GM mice. Advances in enable the creation of patient-specific models by integrating induced pluripotent stem cells (iPSCs) derived from human patient genomes into GM mouse frameworks, such as humanized mice engrafted with iPSC-derived tumors, allowing tailored disease modeling and drug testing. Global repositories like the International Mouse Phenotyping Consortium (IMPC) facilitate broader access, having phenotyped over 8,700 lines by 2025, with projections nearing 11,000 by 2027, providing a vast resource of standardized GM strains for collaborative research. Sustainability efforts offer promising paths to minimize animal use, with emerging technologies like digital twins—virtual replicas of biological systems powered by —and organ-on-chip platforms simulating multi-organ interactions , potentially reducing reliance on live GM mice for and testing. Prospectively, ethical applications of GM mice extend beyond ; for , gene drives in modified mice can propagate disease-resistance traits, such as anti-tick modifications to curb transmission in wild populations. In space exploration, GM models like "mighty mice" engineered with myostatin inhibition help study microgravity-induced muscle and bone loss, informing countermeasures for human astronauts.

References

  1. [1]
    Knockout Mice Fact Sheet
    Aug 17, 2020 · A knockout mouse is a lab mouse where a gene is inactivated by replacing or disrupting it with artificial DNA.
  2. [2]
    Definition of transgenic mice - NCI Dictionary of Cancer Terms
    Mice that have had DNA from another source put into their DNA. The foreign DNA is put into the nucleus of a fertilized mouse egg.
  3. [3]
    Genetically Altered Mice: A Revolutionary Research Resource - NCBI
    Transgenic mice are made by using glass micropipettes to inject a solution that contains DNA from a chosen source into the nucleus of a fertilized mouse egg. At ...
  4. [4]
    Scientists revolutionize the creation of genetically altered mice to ...
    May 2, 2013 · Scientists create models in mice by altering specific genes that have been associated with a given disease. The models allow for the study of ...
  5. [5]
    OncoMouse | National Museum of American History
    In early 1983 Harvard University scientists Philip Leder and Timothy Stewart created OncoMice by using a fine glass needle to inject known cancer genes into ...
  6. [6]
    Genetically modified mouse models to help fight COVID-19 - Nature
    Oct 26, 2020 · Several genetically engineered mouse model (GEMMs) that express the hACE2 gene were created over a decade ago for use in SARS virus studies.
  7. [7]
    Designing and generating a mouse model: frequently asked questions
    Mar 26, 2021 · A genetically engineered mouse in which a specific gene is disrupted or deleted is called a knockout mouse. By studying what went wrong in a ...
  8. [8]
    Breeding - Transgenic Mouse Facility
    The fundamental consideration in breeding genetically modified mice is that any given transgene, mutation, or other genetic modification can interact with a ( ...
  9. [9]
    Genetically Engineered Mouse Core | Abramson Cancer Center ...
    The Genetically Altered Mouse Core also carries out embryo re-derivation, embryo and sperm cryopreservation, in vitro fertilization (IVF), and centralized ...<|control11|><|separator|>
  10. [10]
    Science Milestone: The first transgenic mice | Drug Discovery News
    Rudolf Jaenisch and Beatrice Mintz obtained the first transgenic mice in 1974. Jaenisch reported germline transmission two years later.
  11. [11]
    000664 - B6 Strain Details - The Jackson Laboratory
    Jan 3, 2019 · C57BL/6J is the most widely used inbred strain and the first to have its genome sequenced. Although this strain is refractory to many tumors ...
  12. [12]
    Generating mouse models for biomedical research: technological ...
    Jan 8, 2019 · Knock-down mouse: a genetically altered mouse in which gene expression is lowered or silenced by using RNAi to degrade the mRNA of that gene.
  13. [13]
    Why Mouse Matters - National Human Genome Research Institute
    Jul 23, 2010 · On average, the protein-coding regions of the mouse and human genomes are 85 percent identical; some genes are 99 percent identical while others ...
  14. [14]
    Why mice? - Understanding Animal Research
    Oct 20, 2025 · The small size of mice means that they are easier and more cost-effective to house than larger animals. This allows large-scale studies to ...
  15. [15]
    Why do we use mice in research? - The Francis Crick Institute
    Sep 10, 2025 · Mice are used because they are genetically similar to humans, have comparable biological processes, are easy to keep, have a short generation ...Missing: preferred | Show results with:preferred
  16. [16]
    Comparative transcriptomics in human and mouse - PMC - NIH
    Mar 12, 2019 · Humans and mice share a very similar genetic background, and around 90% of both genomes can be partitioned into regions of conserved synteny.
  17. [17]
    The Nobel Prize in Physiology or Medicine 2007 - NobelPrize.org
    The Nobel Prize in Physiology or Medicine 2007 was awarded jointly to Mario R. Capecchi, Sir Martin J. Evans and Oliver Smithies for their discoveries of ...
  18. [18]
    The Impact of Rodents on Advances in Biomedical Research - PMC
    Rodents are preferred for biomedical research due to their similarity to humans, small size, ease of maintenance, short life cycle, and genetic resources. They ...
  19. [19]
    Why mice are used in animal research | EARA
    Mice and humans are genetically very similar, with almost all mouse genes sharing the same functions as our genes. · This 'humanising' process is done by ...Missing: preferred | Show results with:preferred
  20. [20]
    Why do scientists use mice in medical research?
    Jun 1, 2016 · Mice are used due to similar physiology, genetic diversity, genome manipulation, and as a whole living organism for disease research.Missing: preferred | Show results with:preferred
  21. [21]
    Mice Model Market Growth, Drivers, and Opportunities
    Global mice model market valued at $1.53B in 2024, reached $1.70B in 2025, and is projected to grow at a robust 10.0% CAGR, hitting $2.74B by 2030.
  22. [22]
    Fast Facts - The Jackson Laboratory
    In 2022, JAX researchers were supported by $65.3 million in peer-reviewed research funding (annual direct costs). JAX has had a National Cancer Institute ...
  23. [23]
    [PDF] CALIFORNIA ECONOMIC IMPACT REPORT THE JACKSON ...
    Overall, in 2023, JAX had an economic impact of more than $1.2 billion. Data taken from January 1 to December 31, 2023. Page 2. Vendor and employeelocations ...
  24. [24]
    The cloning of a frog | Development | The Company of Biologists
    Jun 15, 2013 · In 1962, a paper examining the developmental capacity of nuclei taken from intestinal epithelial cells of Xenopus laevis tadpoles was published.
  25. [25]
    Profile of John Gurdon and Shinya Yamanaka, 2012 Nobel ... - PNAS
    In 1962, in a series of experiments inspired by Briggs and King (1), Gurdon demonstrated that the nucleus of a frog somatic cell could be reprogrammed to behave ...
  26. [26]
    Genetic Mosaicism in Adult Mice of Quadriparental Lineage - Science
    Genetic mosaic mice can be produced by aggregating, during cleavage stages, the blastomeres of two embryos of different genotype into a single cluster, ...
  27. [27]
    Germ-Line Transformation of Mice - PMC - PubMed Central
    During the late 1970s, these methods were adapted for microinjection of mRNA, and then DNA, into mouse eggs (6, 7). In late 1980, the first report describing ...
  28. [28]
    Simian Virus 40 DNA Sequences in DNA of Healthy Adult ... - PNAS
    Simian Virus 40 DNA Sequences in DNA of Healthy Adult Mice Derived from Preimplantation Blastocysts Injected with Viral DNA. Rudolf Jaenisch and Beatrice Mintz ...
  29. [29]
    3. Genetically Altered Mice: A Revolutionary Research Resource
    HOW TO MAKE A TRANSGENIC MOUSE. The revolution began in 1974 when Rudolph Jaenisch and Beatrice Mintz infected mouse embryos with SV40 virus and showed that ...
  30. [30]
    Dramatic growth of mice that develop from eggs microinjected with ...
    Dec 16, 1982 · Of 21 mice that developed from these eggs, seven carried the fusion gene and six of these grew significantly larger than their littermates.Missing: supermouse | Show results with:supermouse
  31. [31]
    INTRODUCTION OF GENES INTO THE GERM LINE OF ANIMALS
    Jun 1, 2016 · In the experiments described by Brinster and Palmiter, a gene composed of the mouse metallothionein I (MT) promoter/regulator fused to the ...
  32. [32]
    The Nobel Prize in Physiology or Medicine 2007 - Press release
    Oct 8, 2007 · Evans had used the ES cells to generate mice that carried new genetic material. Two ideas come together – homologous recombination in ES cells.
  33. [33]
    The Nobel Prize in Physiology or Medicine 2007 - NobelPrize.org
    Mario Capecchi and Oliver Smithies, independently of each other, discovered how homologous recombination between segments of DNA molecules can be used to target ...
  34. [34]
    US4736866A - Transgenic non-human mammals - Google Patents
    A transgenic non-human eukaryotic animal whose germ cells and somatic cells contain an activated oncogene sequence introduced into the animal.<|separator|>
  35. [35]
    The development of human immune system mice and their use to ...
    For these reasons, HLA transgenic mice were among the first “humanized” animal models of autoimmune diseases [[2], [3], [4], [5]]. These models have become even ...
  36. [36]
    Conditional Gene Targeting: Dissecting the Cellular Mechanisms of ...
    Dec 29, 2010 · Cre/lox conditional gene targeting requires a mouse that has been pre-engineered with a loxP-flanked gene (or gene segment), generated with ...
  37. [37]
    Generating mouse models for biomedical research: technological ...
    Jan 8, 2019 · Over the past decade, new methods and procedures have been developed to generate genetically engineered mouse models of human disease.
  38. [38]
    Development of transgenic animals for optogenetic manipulation of ...
    Oct 15, 2013 · Here we review the rapidly growing toolbox of transgenic mice and rats that exhibit functional expression of engineered opsins for neuronal ...
  39. [39]
    Applications of Single-Cell Sequencing Technology to the Enteric ...
    Mar 15, 2022 · We review recent papers applying single-cell sequencing tools to the nascent neural crest and to the developing and mature enteric nervous system.Missing: optogenetics 2020s
  40. [40]
    Genetic transformation of mouse embryos by microinjection of ...
    These results demonstrate that genes can be introduced into the mouse genome by direct insertion into the nuclei of early embryos.
  41. [41]
  42. [42]
  43. [43]
    Gene Targeting Using Homologous Recombination in Embryonic ...
    Apr 11, 2016 · By now, several thousand genes have been mutated using homologous recombination-based methods in embryonic stem (ES) cells (Capecchi, 2005), and ...
  44. [44]
    Novel App knock-in mouse model shows key features of amyloid ...
    Jun 11, 2022 · The AppSAA knock-in mouse model was engineered by insertion of 6 mutations into the genomic App locus via homologous recombination. Three amino ...
  45. [45]
    Single App knock-in mouse models of Alzheimer's disease - PubMed
    We generated knock-in mice that harbor Swedish and Beyreuther/Iberian mutations with and without the Arctic mutation in the APP gene.
  46. [46]
  47. [47]
    Multiplexed base editing through Cas12a variant-mediated cytosine ...
    Nov 2, 2022 · We develop Cas12a-mediated cytosine base editor (CBE) and adenine base editor (ABE) systems with elevated efficiencies and expanded targeting scope.
  48. [48]
    Genetically engineered mouse models in oncology research ... - NIH
    Dec 27, 2016 · GEMMs have successfully been used to validate candidate cancer genes and drug targets, assess therapy efficacy, dissect the impact of the tumor ...
  49. [49]
    Humanized Mice for the Study of Infectious Diseases - PMC - NIH
    Recently, it was shown that NSG-BLT mice infected with HIV-1 generate human CD8 T cell responses that closely resemble cellular immune responses observed in ...Missing: SIRPα | Show results with:SIRPα
  50. [50]
    Non-germline genetically engineered mouse models for ... - NIH
    This Review introduces new approaches to modelling cancer, with emphasis on a growing collection of non-germline GEMMs (nGEMMs). These offer flexibility, speed ...
  51. [51]
    SEMMs: Somatically Engineered Mouse Models. A New Tool for In ...
    Apr 22, 2021 · These models recapitulate the phenotype of KrasG12D driven tumors with cooperating Trp53, Lkb1 and Arid1A lesions in a single workhorse mouse by ...
  52. [52]
    Prediction of human pharmacokinetics for low‐clearance ...
    Jun 3, 2021 · Chimeric mice with humanized livers (PXB-mice) were generated from urokinase-type plasminogen activator-cDNA/severe combined immunodeficiency ...
  53. [53]
    P450-Humanized and Human Liver Chimeric Mouse Models for ...
    Thus, drugs are often metabolized differently in mice and in humans. Reactive drug metabolites drive toxicity, and their presence or absence is determined by ...
  54. [54]
    Humanized mouse lines and their application for prediction of ...
    Humanized mouse lines and their application for prediction of human drug metabolism and toxicological risk assessment ... human CYP-mediated drug metabolism ...
  55. [55]
    A single-dose mRNA vaccine provides a long-term protection for ...
    Feb 3, 2021 · ... hACE2 transgenic mice from SARS-CoV-2 ... Since the beginning of the outbreak, the development of COVID-19 vaccines has proceeded at record speed.
  56. [56]
    Targeted deletion of PD-1 in myeloid cells induces anti-tumor immunity
    To examine whether PD-1 might have an active role in tumor-induced stress myelopoiesis, we used PD-1 deficient (PD-1−/−) mice. PD-1 deletion, which resulted in ...Results · Figure 1: Pd-1 And Pd-L1 Are... · Figure 2: Pd-1 And Pd-L1 Is...
  57. [57]
    The development of imatinib as a therapeutic agent for chronic ...
    The human BCR-ABL+ leukemia cell line KU812 injected into nude mice was used to study the mechanisms underlying in vivo resistance to imatinib.33 If imatinib ...
  58. [58]
    Why 90% of clinical drug development fails and how to improve it?
    Although many successful strategies are correctly implemented to overcome the four possible reasons of 90% of clinical development failures, the success rate of ...
  59. [59]
    AI-driven drug discovery picks up as FDA pushes to reduce animal ...
    Sep 2, 2025 · Within the next three to five years, using AI and cutting back on animal testing could reduce timelines and costs by at least half, according to ...Missing: phenotyping modified
  60. [60]
    A Constitutively Active Dioxin/Aryl Hydrocarbon Receptor Promotes ...
    Transgenic mice of this strain [constitutively active AhR (CA-AhR mice)] show increased expression of AhR-dependent genes such as CYP1A1 in several organs ...
  61. [61]
    Cre Reporting & Characterization System for Whole Mouse Brain
    Using these reporters and a high-throughput in situ hybridization platform, we are systematically profiling Cre-directed gene expression throughout the mouse ...Results · Gene Targeting In Es Cells... · Transgenic Mice...
  62. [62]
    Genetic engineering of animals: Ethical issues, including welfare ...
    Genetically engineered farm animals can be created to enhance food quality (9). For example, pigs have been genetically engineered to express the Δ12 fatty acid ...Missing: proofs- | Show results with:proofs-
  63. [63]
    Chimeric Mouse Models for Space Radiation Risk Investigations
    Preliminary data from histopathological analysis suggest chimeric mouse models are suitable for investigations of space radiation risks, as the humanized liver ...Missing: genetically resistance
  64. [64]
    Welfare Issues of Genetically Modified Animals | ILAR Journal
    Conditions such as Alzheimer's disease, amyotrophic lateral sclerosis (ALS 1 ), and Parkinson's disease are subjects of intense efforts to develop new models.
  65. [65]
    3R measures in facilities for the production of genetically modified ...
    The 3R concept is the basis for bringing this demand into practice: Replace animal experiments with alternatives where possible, Reduce the number of animals ...Missing: principle | Show results with:principle
  66. [66]
    [PDF] Putting animal welfare principles and 3Rs into action - EFPIA
    This method eliminates the generation and breeding of genetically modified rodents and has therefore a huge impact on decreasing animal numbers.
  67. [67]
    Abnormal behavioral phenotypes of serotonin transporter knockout ...
    Behavioral phenotyping function in knock-outs revealed genetic background-related abnormalities, including increased anxiety-like behaviors, reduced aggression,Missing: unintended effects welfare
  68. [68]
    deficient mice - PMC - PubMed Central
    Serotonin transporter (SERT) knockout (−/−) mice have an altered phenotype in adulthood, including high baseline anxiety and depressive-like behaviors, ...
  69. [69]
    Alternatives to animal experiments
    Explore innovative alternatives to animal testing that protect animals from cruelty. Support humane science and help take suffering out of research ...Missing: modified | Show results with:modified
  70. [70]
    [PDF] Refinement and reduction in production of genetically modified mice
    (iii) Effects of genetic modiŽ cation on animal well-being. Genetic modifi cation can compromise animal welfare by causing or predisposing animals to pain, ...
  71. [71]
    Animals in science - Environment - European Commission
    Information on the NTS will provide insight into project objectives, expected outcomes, and benefits, explain likely harms to the animals and provide details ...Missing: modified | Show results with:modified
  72. [72]
    Mission impossible accomplished? A European cross-national ...
    Feb 20, 2024 · The objectives of this study are to analyze the operationalization of HBA in EU member states and investigate the consistency of HBA's implementation.<|control11|><|separator|>
  73. [73]
    FAQs for Research on Genetically Modified (Transgenic) Animals
    The creation of transgenic rodents falls under one of three sections of the NIH Guidelines depending on the containment level required to house the rodents.Missing: OLAW | Show results with:OLAW
  74. [74]
    Framework for the genetically altered animals under Directive 2010 ...
    Jun 8, 2022 · The following is intended as guidance to assist the Member States and others affected by Directive 2010/63/EU on the protection of animals ...Missing: modified | Show results with:modified
  75. [75]
    Safety Assessment of Transgenic Organisms in the Environment ...
    Jul 27, 2023 · The OECD consensus documents identify information of relevance to the environmental risk/safety assessment of genetically engineered organisms.Missing: laboratory | Show results with:laboratory
  76. [76]
    [PDF] United States Government Policy for Oversight of Dual Use ...
    May 6, 2024 · The intent of research oversight is to increase the awareness of researchers, research institutions, and federal funding agencies about the ...Missing: CRISPR editing
  77. [77]
    Human embryonic stem cells contribute to embryonic and ... - Nature
    Nov 3, 2017 · Human embryonic stem cells contribute to embryonic and extraembryonic lineages in mouse embryos upon inhibition of apoptosis.
  78. [78]
    Producing human livers from human stem cells via blastocyst ...
    This review discusses the latest innovations in blastocyst complementation and highlights the progress made in creating organs for transplant.
  79. [79]
    In Vivo Generation of Organs by Blastocyst Complementation - NIH
    Blastocyst complementation was first demonstrated when wild type mouse embryonic stem cells (mESCs) were injected into Rag2 mutant blastocysts that grew into ...
  80. [80]
    Cellular heterogeneity during mouse pancreatic ductal ... - NIH
    Single-cell RNA sequencing of mouse models of pancreatic ductal adenocarcinoma reveals the molecular subtypes of cancer cells, fibroblasts, and macrophages.
  81. [81]
    Spatial transcriptomics reveals molecular dysfunction associated ...
    Mar 26, 2024 · In the current study, we use spatial transcriptomics to capture whole transcriptome signatures from cortical neurons with α-synuclein pathology ...
  82. [82]
    An evaluation of machine-learning for predicting phenotype
    In this paper, we compare for phenotype prediction a state-of-the-art classical statistical genetics method and a mixed-model approach BLUP (used extensively ...
  83. [83]
    Impact of Automated Genotyping and Increased Breeding ... - Frontiers
    We investigated the use of commercial automated genotyping and centralized breeding management on overall breeding colony productivity in a colony of multiple ...
  84. [84]
    Adeno-associated virus for gene therapy of human diseases
    Apr 3, 2024 · Adeno-associated virus (AAV) has emerged as a pivotal delivery tool in clinical gene therapy owing to its minimal pathogenicity and ability to establish long- ...
  85. [85]
    In vivo genome editing of the albumin locus as a platform for protein ...
    Oct 8, 2015 · AAV- and ZFN-mediated targeting of the albumin locus corrects disease phenotype in mouse models of hemophilia A and B. Robust expression from ...<|separator|>
  86. [86]
    Mouse strain-specific responses along the gut-brain axis upon fecal ...
    Our results reveal that gut microbiota alone induce changes in ASD-like behavior, and highlight the importance of mouse strain selection when investigating ...Missing: modified | Show results with:modified
  87. [87]
    Humanized immune system mouse models - PubMed Central - NIH
    However, critical differences in the genetics and immune systems of mice and those of humans have precluded studies in mice of uniquely human immune responses.
  88. [88]
    Mouse models of human disease: An evolutionary perspective - NIH
    But there are many differences between the mouse and human immune systems, such that much research on immunological diseases in mice is not transferable to ...
  89. [89]
    Cost of Custom Mouse Model Generation: KO vs. KI vs. Conditional
    Apr 29, 2025 · The cost of generating a KO model primarily involves the design and synthesis of the targeting vector, embryonic stem (ES) cell culture and ...
  90. [90]
    Off-target effects in CRISPR/Cas9 gene editing - PMC - NIH
    The off-target effects occur when Cas9 acts on untargeted genomic sites and creates cleavages that may lead to adverse outcomes. The off-target sites are often ...
  91. [91]
    Off- and on-target effects of genome editing in mouse embryos - NIH
    Off-target effects are minimal and manageable in mouse embryos. Early reports found off-target mutations at loci containing mismatches of less than 4 ...
  92. [92]
    Analysis of animal-to-human translation shows that only 5% of ... - NIH
    Jun 13, 2024 · The overall proportion of therapies progressing from animal studies was 50% to human studies, 40% to RCTs, and 5% to regulatory approval.
  93. [93]
    Mouse genomic variation and its effect on phenotypes and gene ...
    Sep 14, 2011 · We report genome sequences of 17 inbred strains of laboratory mice and identify almost ten times more variants than previously known.
  94. [94]
    Resolving genetic diversity in mouse strains - The Jackson Laboratory
    Apr 5, 2023 · Structural variation between mouse strains. Mice have their own reference genome, known as GRCm39, based on the sequence of C57BL/6J, a strain ...
  95. [95]
    Autologous humanized mouse models of iPSC-derived tumors ... - NIH
    Jan 14, 2022 · In this study, we generated genetically defined tumor cell lines from primary and iPSC-derived cells that, when combined with immune ...
  96. [96]
    Systematic ocular phenotyping of 8707 knockout mouse lines ...
    Jan 20, 2025 · BMC Genomics volume 26, Article number: 48 (2025) ... 8,707 knockout mouse lines phenotyped by the International Mouse Phenotyping Consortium ...
  97. [97]
    Commentary: The International Mouse Phenotyping Consortium
    Sep 10, 2024 · By 2027 when the current round of funding expires, the IMPC will have produced and phenotyped 11,846 knockout mouse lines representing ~ 60% of ...
  98. [98]
    Artificial intelligence in preclinical research: enhancing digital twins ...
    AI revolutionizes preclinical research, offering alternatives to animal testing. Techniques like ML, DL, OoC, and DTs enable precise drug safety simulations.
  99. [99]
    enhancing digital twins and organ-on-chip to reduce animal testing
    Apr 21, 2025 · This review examines the transformative impact of AI in preclinical research, highlighting its advancements, challenges, and the critical steps needed
  100. [100]
    Mice Against Ticks: an experimental community-guided effort ... - NIH
    Mar 25, 2019 · Mice Against Ticks is a community-guided ecological engineering project that aims to prevent tick-borne disease by using CRISPR-based genome editing.
  101. [101]
    “Mighty Mice” in Space Could Aid Muscle & Bone Loss on Earth
    Sep 10, 2020 · The team found that the wild type mice exposed to microgravity lost significant muscle and bone mass unless treated with the drug to inhibit ...