Download PDF
Perspective  |  Open Access  |  20 Sep 2022

Genome-edited rabbit, a prospective alternative model for neurological diseases

Views: 1847 |  Downloads: 1028 |  Cited:   1
Ageing Neur Dis 2022;2:16.
10.20517/and.2022.15 |  © The Author(s) 2022.
Author Information
Article Notes
Cite This Article

Abstract

Animal models have great importance in the research of human neurodegenerative diseases due to their value in symptom mimicking, mechanism investigation, and preclinical tests. Although non-human primate and large animal models have good performance in disease modeling due to their high maintenance cost and critical ethical standards, rodent models are commonly used. Rodent models have been successfully applied in modeling many neurological diseases; however, their genetic background, neuroanatomical features, and nervous system development are different from those of humans. Moreover, the short lifespan and small body size of rodent models also limit the monitoring of disease progression and observation of clinical symptoms in studying neuronal disorders that are late-onset or have a long course of progression. In comparison with rodents, rabbits are phylogenetically closer to humans and have closer similarities to humans in brain development, thus are an alternate animal model for human neurological diseases.

Keywords

Neurodegenerative diseases, genome editing, animal model, rabbit, Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis

INTRODUCTION

Neurodegenerative diseases (NDDs) are associated with progressive neuron losses, most of which are linked with genetic disorders[1]. At present, neurological disorders are considered as one of the major causes of mortality and disability worldwide[2]. Unfortunately, many NDDs are late-onset and hard to detect in the early stage; for instance, Parkinson’s disease (PD) and Alzheimer’s disease (AD) have high morbidity in elderly patients, while amyotrophic lateral sclerosis (ALS) does not exhibit clear symptoms at the early stage of the disease. Moreover, due to the irreversibility of neuronal death, no effective therapeutic approaches are available at present. Therefore, a thorough investigation of these diseases is essential for the development of disease-specific and effective prognostic, diagnostic, and therapeutic strategies. Model organisms are essential platforms for the above research; cell lines and animal models are frequently used. Although cell models can be used for the investigation of pathological pathways and molecular mechanisms of disease pathogenesis[3], due to the limitation in modeling organogenesis and human physiology[4], they cannot mimic histological, morphological, and behavioral changes in human diseases. Therefore, animal disease models that partially recapitulate the aspects of human diseases are essential. Additionally, animal models have irreplaceable value in preclinical tests; they are also important for the development of prognostic and therapeutic strategies.

In comparison with non-mammalian animals such as zebrafish and Drosophila melanogaster, mammalian models have greater similarities to humans in genetics, metabolism, and physiology and can therefore mimic some of the biological and clinical features of human disease[5,6]. In practical research, large animal models are often used because of the biological characteristics of these animals for modeling human NDDs[7]. With the development of genome editing tools and somatic cell nuclear transfer (SCNT) techniques, genetically modified large animal models can be effectively produced, which can promote the utilization of these models[8]. Unfortunately, maintenance costs, space requirements, and ethical standards are still problems for the use of large animal models in biomedical research. Compared with large animal models, rodents have a small body size, low maintenance cost, and can be easily handled, making them cost-efficient models. Moreover, rodents have a relatively high genetic identity and physiological similarity to humans, and genetic modification capabilities can facilitate the modeling of genetic disorders[9]. Therefore, currently, rodents are the main experimental animal for biomedical research and disease modeling. From 1950 to 2010, approximately 80% of animal-based biomedical studies were performed on rodents (59% on mice and 18% on rats)[10].

Rodents have been applied in modeling many neurological diseases and have adequate precision in mimicking the pathology and physiology in some cases. However, due to factors such as lifespan, genetic differences from humans, and small body size, rodent models have some limitations in studying neuronal disorders[9]. For example, they cannot replicate the exact pathological hallmarks in some human diseases because of physiological and genetic differences. PD mouse models (α-synuclein transgene or knockout of LRRK2, PRKN, and PINK1) do not show degeneration of nigrostriatal dopaminergic neurons[11] and striatal neurons remain viable, which is different from the pathological features in human disease[11]. The absence of intranuclear inclusion body formation in neuronal cells of ALS mice overexpressing mutant hSODl is inconsistent with the phenotype of human ALS disease. Thus, finding an alternative animal species for modeling is needed to produce better models for diseases that cannot be recapitulated in mice.

Rabbits are docile and easy to handle; their short reproductive cycle and high reproductive performance can guarantee an abundant sample size for experiments; and the efficiency of model production and the low demand for rearing and surgical operation equipment make rabbits easy to maintain and handle[12]. Moreover, rabbits have an intermediate lifespan (longer than rodents but shorter than large animals such as non-human primates), and, compared with rodents, rabbits are phylogenetically closer to humans[13] and are more similar to humans in brain development[14]; therefore, they may have better precision in disease modeling. With the development of targeted genome editing tools, producing targeted genome-edited rabbit models for human neuronal disorders has become attainable.

ADVANTAGES OF RABBITS AS ANIMAL MODELS OF NEUROLOGICAL DISEASES

Rabbits are phylogenetically closer to humans than mice

Genetic similarity to humans is linked with the identity of protein structure and function, and high genetic similarity could increase the precision of disease modeling. Compared with rodents, rabbits are phylogenetically closer to primates[13], suggesting they may have better precision in disease modeling. Notably, some human genes do not have orthologs in mice: approximately 1% of human genes cannot find orthologs in mice’s genomes. For instance, caspase 10, a gene that is linked to neurodegeneration via the extrinsic apoptosis pathway[15], is absent in mice but has orthologs in rabbits[11]. However, the phylogenetic similarity between rabbits and humans does not guarantee rabbits would be a better model for all human diseases; the performance of disease modeling is still dependent on the type of mutant gene, and the mechanism involved in the pathogenesis should also be considered.

The development of the central nervous system of rabbits has greater similarity to humans compared with rodents

Neurological features are critical for NDD modeling, and the CNS development of rabbits is highly similar to that of humans compared with that of rodents. Specifically, the phase of brain development and myelination in rabbits is more similar to humans than that of rodents, since such a process happens during the perinatal period in humans and rabbits but postnatally in rodents[14]. Moreover, rabbits have a higher brain volume and cerebral surface area than mice. The time point of morphological configuration of major CNS structures of rabbits is closer to humans. The development of structures such as primitive streak, neural tube closure, and primary brain vesicles in rabbits is chronologically similar to that in humans[14]. In addition, the development of white matter in rabbits is closer to that of humans[14], and rabbits have a higher white matter ratio than mice (approximately 20% vs. 10%)[16,17] [Table 1]. Moreover, rabbits have larger brain volume, cortex surface area, and number of neurons compared with rodents [Table 1], suggesting that rabbits may exhibit better cognitive, learning, and memory abilities.

Table 1

Major differences in size and structures between rabbits and mice

HumanRabbitMice
Brain volume[18]1300-1400 g10-13 g0.4-0.5 g
Spinal cord length[18]43 to 45 cm18 cm7.5
Gray-white matter ratio[16,17]40:6080:2090:10
Duration to reach adult brain volume[19,20]20 years4 months2 months
Glia-neuron ratio (GNR)[21]1.660.32-0.490.29-0.42
Number of neurons[22-24]86,000 million494.2 million71 million

Rabbits can be trained to learn basic skills (e.g., recall signals) through positive reinforcement[25]. Rabbits also have both short- and long-term memory[26,27] and can exhibit memory losses when mimicking NNDs such as AD[27]. Specifically, in an AD rabbit model constructed by drug induction, the results of novel object recognition (NOR) and object location memory (OLM) tests suggest that the model can track cognitive impairment[28]. In other studies, the results of conditional and unconditional response tests also suggest that the AD rabbit model has reduced learning ability[29,30].

Additionally, axon degeneration is a common pathological feature of NDDs, and neurons with longer projections have a higher vulnerability to axon degeneration, which can be easily affected in NDDs[31]. Some mice models of motor neuron diseases exhibit molecular pathological features in neurons but only exhibit mild or even no behavioral symptoms[32,33]; vulnerability to axon degeneration might be the explanation for this phenomenon, since the axon length in rodents is shorter than that in larger animals. Collectively, rabbit models might have better accuracy in mimicking human neuronal diseases compared to rodents.

Rabbits have a relatively large body size for handling and sampling

In addition to the genetic and neurological features, rabbits also have a bigger body size compared with rodents, which can facilitate better animal handling and symptom observation. The relatively large body size of the rabbits is also associated with larger organ size and blood volume (45-75 mL per kg body weight versus 1.5-2.5 mL)[10,34], which can be beneficial for diagnostic investment, surgical operation, and sampling for pathological analysis. Cerebrospinal fluid (CSF) and blood biomarker analyses are commonly used in the diagnosis of NDDs such as AD and FTD[35-37]. Such assays are hard to perform in mice due to the poor sample size, especially for experiments that need continuous monitoring; in contrast, for larger animals such as rabbits, an adequate amount of sample can be collected with minimal harm to the animal.

In addition, medical imaging approaches are usually needed for the prognosis and diagnosis of NDDs[38,39]. However, it is hard to perform high-definition medical imaging on small animals due to the limitation of the equipment. High-resolution magnetic resonance imaging (MRI) in mice requires a scanner with an ultra-high field strength of 7 T or higher[40], which is inaccessible for most researchers. Compared with rodents, rabbits have larger CNS [Table 1]; thus, a normal MRI scanner with a 3.0 T field strength is adequate for CNS imaging in rabbits[41]. The large body size of rabbits can also benefit electromyography tests, which are commonly used in the diagnosis of neuromuscular diseases such as ALS. Additionally, the scale of the central neuron system also affects the maneuverability of tissue sampling and the intraparenchymal or epidural injection of therapeutic vectors such as AAV in future translational medical research.

Collectively, the larger body size of rabbits makes it easier to handle and sample compared with small animals, which largely facilitate phenotype observation and surgical operation.

The lifespan of rabbits is long enough for the observation of disease progression

The onset of neurodegenerative disease and the speed of progression are affected by both genetic and environmental factors[42]. For pathological mutations that induce late onset and slow progression, the lifespan of animal models should also be considered, since the effect of aging can interfere with the observation of clinical symptoms[9]. Generally, NDDs are progressive diseases that last from years to decades. For instance, the median survival time of ALS patients is 20-48 months[43], while that of AD patients can reach up to 30 years[44]. Moreover, late-onset NDDs such as AD and PD develop late in life; both diseases usually begin at age 60 or older in human patients[45]. However, the normal lifespan of mice is 12-36 months[10], which means that for diseases that begin late or have a slow rate of progression, mice may not fully exhibit the whole course of the disease. For mutations that can only induce late-onset symptoms or slow progressive disease, mice models may not exhibit observable symptoms in their lifetime without extra administration, such as drug stimulation[46]. Furthermore, due to the short lifespan of mice, it is hard to identify whether a symptom (e.g., vision loss) is caused by pathological neuronal death or age-related reasons[9]. In the adult phase, 2.6 mice days is equivalent to one human year, while, in the post-senescence phase, 2.069 mice days is equivalent to one human year[10]. Such fast senescence processes can largely limit the progression of disease and interfere with the observation of disease-related clinical symptoms. In contrast, the maximum lifespan of laboratory rabbits can reach up to 10 years under proper conditions, and one human year is equivalent to 18.25 and 50.34 rabbit days in the adult and post-senescence phases, respectively[34]. Thus, for most NDDs, the lifespan of rabbits is long enough for the observation of disease progression.

PRODUCTION OF GENOME-MODIFIED RABBIT DISEASE MODELS VIA CRISPR-CAS SYSTEM

The production of disease models that recapitulate the pathological features of human disease is an important approach to investigating the pathogenesis of the disease. Artificially induced disease models can exhibit clinical features of some NDDs. For instance, hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, rotenone, and paraquat are commonly used in the induction of Parkinson’s disease[47]. However, many NDDs are caused by pathological mutation of the disease-related gene, and an induction model cannot fully recapitulate the whole pathological pathway of diseases caused by genetic disorders[1]. Therefore, to elucidate the whole pathogenesis process of neuron degeneration, the production of animal models that carry pathological mutations that mimic human disease is necessary.

With the development of gene-editing tools, efficient and accurate genome modification has become achievable. To date, various genome-edited rabbits have been constructed, as shown in Table 2. In 2013, the CRISPR-Cas9 system was harnessed for efficient targeted genome editing in eukaryotic cells[99,100]. Moreover, with the further development of research on CRISPR-Cas systems, the CRISPR-Cas systems and their derivates can facilitate targeted gene knockout (KO), knockin (KI), activation, suppression, and single-base substitution. Presently, various genome editing tools based on CRISPR-Cas systems are widely used in multiple species, including non-human primates, large non-primate animals, rodents, and rabbits[101-103].

Table 2

Summary of genetically modified rabbits

SystemGenesModificationApplicationRefs.
ZFNIgMKOImmunodeficiency[48]
ZFNAPOC3KOLipid metabolism and
atherosclerosis
[49]
ZFNAPOEKOLipid metabolism and
atherosclerosis
[50]
ZFNCETPKOLipid metabolism and
atherosclerosis
[51]
TALENsRAG1; RAG2KOImmunodeficiency[52]
TALENsFAHKOHereditary tyrosinemia type 1[53]
CRISPR/Cas9FBN1KOMarfanoid progeroid lipodystrophy syndrome[54]
CRISPR/Cas9DMDKODuchenne muscular dystrophy[55]
CRISPR/Cas9ANO5KOMuscular dystrophy[56]
CRISPR/Cas9α-CrystallinKOCongenital cataracts[57]
CRISPR/Cas9GJA8KOCongenital cataracts[58]
CRISPR/Cas9LDLRKOLipid metabolism and
atherosclerosis
[59]
CRISPR/Cas9MSTNKOMuscle hypertrophy[60,61]
CRISPR/Cas9SRYKOSex reversal syndromes and
hermaphroditism syndromes
[53,54]
CRISPR/Cas9PHEXKOX-linked hypophosphatemia[62]
CRISPR/Cas9LMNAKOPremature aging syndrome[63]
CRISPR/Cpf1WRNKOWerner syndrome[64]
CRISPR/Cas9TYRKOOculocutaneous albinism[65,66]
CRISPR/Cas9DMP1KOMineralization defects[67]
CRISPR/Cas9GADD45GKOCongenital cleft palate[68]
CRISPR/Cas9HOXC13KOHair and nail ectodermal dysplasia[69]
CRISPR/Cas9GCKKOMaturity-onset diabetes of the young type 2[70]
CRISPR/Cas9HBB2KOβ-thalassemia[71]
CRISPR/Cas9WASKOWiskott-Aldrich syndrome[72]
CRISPR/Cas9CBSKOCongenital hyper-homocysteinemia[73]
CRISPR/Cas9LDLR; APOEKOLipid metabolism and
atherosclerosis
[74]
CRISPR/Cas9APOC3KOLipid metabolism and
atherosclerosis
[75]
CRISPR/Cas9CFTRKOCystic fibrosis[76]
CRISPR/Cas9CFTRKO ΔF508Cystic fibrosis[77]
CRISPR/Cas9CLPGKOMuscular hypertrophy syndrome[69]
CRISPR/Cas9FGF5KOLong hair[78]
CRISPR/Cas9IL2RGKOX-linked severe combined
immunodeficiency
[79]
CRISPR/Cas9MC1RKOBlock the synthesis of eumelanin and create a novel pale-yellow coat color[80]
CRISPR/Cas9XIST P1KOX-chromosome inactivation[73]
CRISPR/Cas9MSTNKOMuscle hypertrophy[74]
CRISPR/Cas9PCSK9p.S386ALipid metabolism and
atherosclerosis
[75]
CRISPR/Cas9ATP7Bp. R778LWilson Disease[81]
CRISPR/Cas9TYRp. T373KOculocutaneous albinism[82]
CRISPR/Cas9TYRKOOculocutaneous albinism[83]
CRISPR/Cas9RAG; RAG2; TIKI1; ALB; IL2RGMultiplex gene KOImmunodeficiency[84-86]
CRISPR/Cas9FUT1; FUT2; SEC1KOFucosyltransferases enzymes activity[87]
CRISPR/Cas9ROSA 26KISafe harbor gene[88,89]
BE3MSTNp.Q93stopMuscle hypertrophy[90]
TYRp.Q68stopOculocutaneous albinism
ABE7.10DMDp.T279ADuchenne muscular dystrophy
eAID-BE4maxTYRp.R299HOculocutaneous albinism[91]
YFE-BE4maxTYRp.Q68StopOculocutaneous albinism[92]
nNme2-CBEFGF5p.Q79StopLong hair[93]
eA3G-BETYRp.Q48stopOculocutaneous albinism[94]
NG-ABEmaxHOXC13p.Q271RHair and nail ectodermal dysplasia[95]
BE4maxFGF5Start Codon DisruptionLong hair[96]
ABE8.17TYRp.T325AOculocutaneous albinism[97]
LMNAp.L530PEmery-Dreifuss muscular dystrophy
PE3HEXAp.Y427fsTay-Sachs disease[98]

The first CRISPR-Cas-mediated gene KO in rabbits was successfully generated in 2014[101] [Table 2]; however, full-length gene KO can only recapitulate diseases caused by loss of function. To mimic diseases caused by gain-of-function mutation due to point mutation, more accurate gene manipulation is needed. Furthermore, more than 50,000 disease-causing mutations in humans are point mutations; therefore, a novel system that can mediate single base substitution is needed. Since 2017, the development of cytosine and adenine base editing systems can facilitate efficient C to T and A to G base substitutions, which can facilitate precise gene manipulation[104]. Such systems were identified as having ideal editing efficiency in rabbits [Table 2]; the efficiency of cytidine base editor (CBE) and adenine base editor (ABE) in rabbits after co-microinjection of base editor mRNA and sgRNA are 53%-88% and 44%-100%, respectively[90]. The following refinement of base editors has overcome or reduced the limitations of PAM sequences and the incidence of bystander activities[92,105]. At this stage, base editing systems are capable of inducing disease causative missense and nonsense mutations in rabbits to generate disease models.

Although base editing systems can induce four transversion mutations, it is impossible for such systems to induce the other eight transversion mutations. Moreover, the generation of bystander mutations cannot be completely avoided when there are multiple C or A in the editing window. Importantly, conventional gene editing systems cannot induce efficient single base or oligonucleotide insertions and deletions. Therefore, it is hard to generate disease models with fragment shift mutations. Fortunately, the development of prime editing systems solved such problems in 2019. The system, which is based on the target binding capacity of the CRISPR-Cas9 system and the retro-transcription activity of retrotrancripsase, can facilitate the whole genome “search and replace” activity in organisms. Prime editor was successfully used in generating a Tay-Sachs disease (TSD) rabbit model in 2021 [Table 2], which is a model of neurological disease generated by prime editor-mediated four base insertion[98].

PROSPECTS AND LIMITATIONS FOR EVALUATING RABBIT DISEASE MODELS

The observation of the clinical phenotypes of diseases is important for the evaluation of animal models. However, unlike the well-developed testing platforms for rodent models, currently, the evaluation criteria for rabbit NDD models are not well established.

In general, the diagnosis, prognosis, and autopsy criteria in human NDDs can be used in animal models. Such investigations can provide data that are comparable to human clinical reports and have better referential value. Indeed, commercialized analysis platforms, such as serological testing, enzyme-linked immunosorbent assay, MRI, electromyography, and histological analysis, are versatile and authentic tools for the assessment of both humans and animals including rabbits. However, it is impossible to apply the whole set of human diagnostic criteria to animals. For example, the investigation methods for behavioral and cognitive analyses in humans are hard to apply in animal models. Standardized and species-specific behavioral analysis platforms can support the assessment of animal disease models. For rodents, systematic behavioral analysis systems are well established and standardized; systems such as multivariate concentric square field and cylinder test are used to investigate traits such as sensory-motor function[106]. In contrast, the behavioral and cognitive analysis platforms for rabbits are not well developed at present, and further development of these systems is necessary for the future use of rabbits in neurological disease modeling.

CONCLUSION

Collectively, rabbits are more similar to humans in brain development, with more genetic similarities than rodents, and longer lifespan and larger body size, suggesting that rabbits can perform well in human neurological disease modeling in addition to traditional non-human primates, large animals, and rodent models. Therefore, it is expected that, in the near future, with the further development of genome editing technology and the establishment of phenotype assessment platforms for rabbit models, the value of rabbits in the research of neurological diseases can be maximized, not only for the understanding of pathological mechanisms but also for innovation of therapeutic approaches.

DECLARATIONS

Authors’ contributions

Made substantial contributions to conception and design of the study and performed data analysis and interpretation: Zhang Z, Song Y

Performed data acquisition, as well as provided administrative, technical, and material support: Li Z, Lai L

Availability of data and materials

Not applicable.

Financial support and sponsorship

This study was financially supported by the National Key Research and Development Program of China Stem Cell and Translational Research (2019YFA0110700).

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2022.

REFERENCES

1. Bertram L, Tanzi RE. The genetic epidemiology of neurodegenerative disease. J Clin Invest 2005;115:1449-57.

2. Chin JH, Vora N. The global burden of neurologic diseases. Neurology 2014;83:349-51.

3. Mirabelli P, Coppola L, Salvatore M. Cancer cell lines are useful model systems for medical research. Cancers (Basel) 2019;11:1098.

4. Heydari Z, Moeinvaziri F, Agarwal T, et al. Organoids: a novel modality in disease modeling. Biodes Manuf 2021;4:689-716.

5. Johan Arief MF, Choo BKM, Yap JL, Kumari Y, Shaikh MF. A systematic review on non-mammalian models in epilepsy research. Front Pharmacol 2018;9:655.

6. Götz J, Streffer JR, David D, et al. Transgenic animal models of Alzheimer’s disease and related disorders: histopathology, behavior and therapy. Mol Psychiatry 2004;9:664-83.

7. Murray SJ, Mitchell NL. The translational benefits of sheep as large animal models of human neurological disorders. Front Vet Sci 2022;9:831838.

8. Zhou X, Xin J, Fan N, et al. Generation of CRISPR/Cas9-mediated gene-targeted pigs via somatic cell nuclear transfer. Cell Mol Life Sci 2015;72:1175-84.

9. Fisher EMC, Bannerman DM. Mouse models of neurodegeneration: know your question, know your mouse. Sci Transl Med 2019;11:eaaq1818.

10. Dutta S, Sengupta P. Men and mice: Relating their ages. Life Sci 2016;152:244-8.

11. Eckhart L, Ballaun C, Hermann M, et al. Identification of novel mammalian caspases reveals an important role of gene loss in shaping the human caspase repertoire. Mol Biol Evol 2008;25:831-41.

12. Mapara M, Thomas BS, Bhat KM. Rabbit as an animal model for experimental research. Dent Res J (Isfahan) 2012;9:111-8.

13. Graur D, Duret L, Gouy M. Phylogenetic position of the order Lagomorpha (rabbits, hares and allies). Nature 1996;379:333-5.

14. de Almeida da Anunciação AR, Favaron PO, de Morais-Pinto L, et al. Central nervous system development in rabbits (Oryctolagus cuniculus L. 1758). Anat Rec (Hoboken) 2021;304:1313-28.

15. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res 2019;29:347-64.

16. Selçuk ML, Tıpırdamaz S. A morphological and stereological study on brain, cerebral hemispheres and cerebellum of New Zealand rabbits. Anat Histol Embryol 2020;49:90-6.

17. Krafft PR, Bailey EL, Lekic T, et al. Etiology of stroke and choice of models. Int J Stroke 2012;7:398-406.

18. Butt M, Bolon B. Fundamental neuropathology for pathologists and toxicologists: principles and techniques. John Wiley & Sons; 2011. p. 551-4.

19. Harel S, Watanabe K, Linke I, Schain RJ. Growth and development of the rabbit brain. Biol Neonate 1972;21:381-99.

20. Yin P, Li S, Li XJ, Yang W. New pathogenic insights from large animal models of neurodegenerative diseases. Protein Cell 2022;13:707-20.

21. Bahney J, von Bartheld CS. The cellular composition and glia-neuron ratio in the spinal cord of a human and a nonhuman primate: comparison with other species and brain regions. Anat Rec (Hoboken) 2018;301:697-710.

22. Herculano-Houzel S. The remarkable, yet not extraordinary, human brain as a scaled-up primate brain and its associated cost. Proc Natl Acad Sci U S A 2012;109 Suppl 1:10661-8.

23. Herculano-Houzel S, Ribeiro P, Campos L, et al. Updated neuronal scaling rules for the brains of Glires (rodents/lagomorphs). Brain Behav Evol 2011;78:302-14.

24. Herculano-Houzel S, Mota B, Lent R. Cellular scaling rules for rodent brains. Proc Natl Acad Sci U S A 2006;103:12138-43.

25. Bradbury AG, Dickens GJ. Appropriate handling of pet rabbits: a literature review. J Small Anim Pract 2016;57:503-9.

26. Schreurs BG, Gusev PA, Tomsic D, Alkon DL, Shi T. Intracellular correlates of acquisition and long-term memory of classical conditioning in purkinje cell dendrites in slices of rabbit cerebellar lobule HVI. J Neurosci 1998;18:5498-507.

27. Schreurs BG. Cholesterol and copper affect learning and memory in the rabbit. Int J Alzheimers Dis 2013;2013:518780.

28. Weiss C, Bertolino N, Procissi D, et al. Diet-induced Alzheimer’s-like syndrome in the rabbit. Alzheimers Dement (N Y) 2022;8:e12241.

29. Woodruff-Pak DS, Agelan A, Del Valle L. A rabbit model of Alzheimer’s disease: valid at neuropathological, cognitive, and therapeutic levels. J Alzheimers Dis 2007;11:371-83.

30. Woodruff-Pak DS, Vogel RW 3rd, Wenk GL. Galantamine: effect on nicotinic receptor binding, acetylcholinesterase inhibition, and learning. Proc Natl Acad Sci U S A 2001;98:2089-94.

31. Kneynsberg A, Collier TJ, Manfredsson FP, Kanaan NM. Quantitative and semi-quantitative measurements of axonal degeneration in tissue and primary neuron cultures. J Neurosci Methods 2016;266:32-41.

32. Tudor EL, Galtrey CM, Perkinton MS, et al. Amyotrophic lateral sclerosis mutant vesicle-associated membrane protein-associated protein-B transgenic mice develop TAR-DNA-binding protein-43 pathology. Neuroscience 2010;167:774-85.

33. Devon RS, Orban PC, Gerrow K, et al. Als2-deficient mice exhibit disturbances in endosome trafficking associated with motor behavioral abnormalities. Proc Natl Acad Sci U S A 2006;103:9595-600.

34. Dutta S, Sengupta P. Rabbits and men: relating their ages. J Basic Clin Physiol Pharmacol 2018;29:427-35.

35. Blennow K, Zetterberg H. Biomarkers for Alzheimer’s disease: current status and prospects for the future. J Intern Med 2018;284:643-63.

36. Bridel C, van Wieringen WN, Zetterberg H, et al. the NFL Group. Diagnostic value of cerebrospinal fluid neurofilament light protein in neurology: a systematic review and meta-analysis. JAMA Neurol 2019;76:1035-48.

37. Heller C, Foiani MS, Moore K, et al. GENFI. Plasma glial fibrillary acidic protein is raised in progranulin-associated frontotemporal dementia. J Neurol Neurosurg Psychiatry 2020;91:263-70.

38. Matsuda H. MRI morphometry in Alzheimer’s disease. Ageing Res Rev 2016;30:17-24.

39. Fyfe I. A clinically useful MRI marker of PD? Nat Rev Neurol 2019;15:2-3.

40. Mandino F, Cerri DH, Garin CM, et al. Animal functional magnetic resonance imaging: trends and path toward standardization. Front Neuroinform 2019;13:78.

41. Müllhaupt D, Augsburger H, Schwarz A, et al. Magnetic resonance imaging anatomy of the rabbit brain at 3 T. Acta Vet Scand 2015;57:47.

42. Coppedè F, Mancuso M, Siciliano G, Migliore L, Murri L. Genes and the environment in neurodegeneration. Biosci Rep 2006;26:341-67.

43. Chiò A, Logroscino G, Hardiman O, et al. Eurals Consortium. Prognostic factors in ALS: a critical review. Amyotroph Lateral Scler 2009;10:310-23.

44. Armstrong RA. Factors determining disease duration in Alzheimer’s disease: a postmortem study of 103 cases using the Kaplan-Meier estimator and Cox regression. Biomed Res Int 2014;2014:623487.

45. Pagano G, Ferrara N, Brooks DJ, Pavese N. Age at onset and Parkinson disease phenotype. Neurology 2016;86:1400-7.

46. Nair RR, Corrochano S, Gasco S, et al. Uses for humanised mouse models in precision medicine for neurodegenerative disease. Mamm Genome 2019;30:173-91.

47. Bové J, Prou D, Perier C, Przedborski S. Toxin-induced models of Parkinson’s disease. NeuroRx 2005;2:484-94.

48. Flisikowska T, Thorey IS, Offner S, et al. Efficient immunoglobulin gene disruption and targeted replacement in rabbit using zinc finger nucleases. PLoS One 2011;6:e21045.

49. Yang D, Zhang J, Xu J, et al. Production of apolipoprotein C-III knockout rabbits using zinc finger nucleases. J Vis Exp 2013:e50957.

50. Ji D, Zhao G, Songstad A, Cui X, Weinstein EJ. Efficient creation of an APOE knockout rabbit. Transgenic Res 2015;24:227-35.

51. Zhang J, Niimi M, Yang D, et al. Deficiency of cholesteryl ester transfer protein protects against atherosclerosis in rabbits. Arterioscler Thromb Vasc Biol 2017;37:1068-75.

52. Song J, Zhong J, Guo X, et al. Generation of RAG 1- and 2-deficient rabbits by embryo microinjection of TALENs. Cell Res 2013;23:1059-62.

53. Song Y, Liu T, Wang Y, et al. Mutation of the Sp1 binding site in the 5’ flanking region of SRY causes sex reversal in rabbits. Oncotarget 2017;8:38176-83.

54. Song Y, Xu Y, Liang M, et al. CRISPR/Cas9-mediated mosaic mutation of SRY gene induces hermaphroditism in rabbits. Biosci Rep 2018;38:BSR20171490.

55. Sui T, Lau YS, Liu D, et al. A novel rabbit model of Duchenne muscular dystrophy generated by CRISPR/Cas9. Dis Model Mech 2018;11:dmm032201.

56. Sui T, Xu L, Lau YS, et al. Development of muscular dystrophy in a CRISPR-engineered mutant rabbit model with frame-disrupting ANO5 mutations. Cell Death Dis 2018;9:609.

57. Yuan L, Yao H, Xu Y, et al. CRISPR/Cas9-mediated mutation of αA-crystallin gene induces congenital cataracts in rabbits. Invest Ophthalmol Vis Sci 2017;58:BIO34-41.

58. Yuan L, Sui T, Chen M, et al. CRISPR/Cas9-mediated GJA8 knockout in rabbits recapitulates human congenital cataracts. Sci Rep 2016;6:22024.

59. Lu R, Yuan T, Wang Y, et al. Spontaneous severe hypercholesterolemia and atherosclerosis lesions in rabbits with deficiency of low-density lipoprotein receptor (LDLR) on exon 7. EBioMedicine 2018;36:29-38.

60. Guo R, Wan Y, Xu D, et al. Generation and evaluation of Myostatin knock-out rabbits and goats using CRISPR/Cas9 system. Sci Rep 2016;6:29855.

61. Lv Q, Yuan L, Deng J, et al. Efficient generation of myostatin gene mutated rabbit by CRISPR/Cas9. Sci Rep 2016;6:25029.

62. Sui T, Yuan L, Liu H, et al. CRISPR/Cas9-mediated mutation of PHEX in rabbit recapitulates human X-linked hypophosphatemia (XLH). Hum Mol Genet 2016;25:2661-71.

63. Sui T, Liu D, Liu T, et al. LMNA-mutated rabbits: a model of premature aging syndrome with muscular dystrophy and dilated cardiomyopathy. Aging Dis 2019;10:102-15.

64. Wu H, Liu Q, Shi H, et al. Engineering CRISPR/Cpf1 with tRNA promotes genome editing capability in mammalian systems. Cell Mol Life Sci 2018;75:3593-607.

65. Honda A, Hirose M, Sankai T, et al. Single-step generation of rabbits carrying a targeted allele of the tyrosinase gene using CRISPR/Cas9. Exp Anim 2015;64:31-7.

66. Song Y, Xu Y, Deng J, et al. CRISPR/Cas9-mediated mutation of tyrosinase (Tyr) 3’ UTR induce graying in rabbit. Sci Rep 2017;7:1569.

67. Liu T, Wang J, Xie X, et al. DMP1 Ablation in the rabbit results in mineralization defects and abnormalities in haversian canal/osteon microarchitecture. J Bone Miner Res 2019;34:1115-28.

68. Lu Y, Liang M, Zhang Q, et al. Mutations of GADD45G in rabbits cause cleft lip by the disorder of proliferation, apoptosis and epithelial-mesenchymal transition (EMT). Biochim Biophys Acta Mol Basis Dis 2019;1865:2356-67.

69. Wan Y, Guo R, Deng M, et al. Efficient generation of CLPG1-edited rabbits using the CRISPR/Cas9 system. Reprod Domest Anim 2019;54:538-44.

70. Song Y, Sui T, Zhang Y, et al. Genetic deletion of a short fragment of glucokinase in rabbit by CRISPR/Cas9 leading to hyperglycemia and other typical features seen in MODY-2. Cell Mol Life Sci 2020;77:3265-77.

71. Yang Y, Kang X, Hu S, et al. CRISPR/Cas9-mediated β-globin gene knockout in rabbits recapitulates human β-thalassemia. J Biol Chem 2021;296:100464.

72. Zhou J, Yan Q, Tang C, et al. Development of a rabbit model of Wiskott-Aldrich syndrome. FASEB J 2021;35:e21226.

73. Yao B, Liang M, Liu H, et al. The minimal promoter (P1) of Xist is non-essential for X chromosome inactivation. RNA Biol 2020;17:623-9.

74. Zhang T, Lu Y, Song S, et al. “Double-muscling” and pelvic tilt phenomena in rabbits with the cystine-knot motif deficiency of myostatin on exon 3. Biosci Rep 2019;39:BSR20190207.

75. Yan K, Zhang T, Zha Y, Liang J, Cheng Y. Construction of point mutation rabbits using CRISPR/Cas9. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021;50:229-38.

76. Xu J, Livraghi-Butrico A, Hou X, et al. Phenotypes of CF rabbits generated by CRISPR/Cas9-mediated disruption of the CFTR gene. JCI Insight 2021;6:139813.

77. Yang D, Liang X, Pallas B, et al. Production of CFTR-ΔF508 rabbits. Front Genet 2020;11:627666.

78. Xu Y, Liu H, Pan H, et al. CRISPR/Cas9-mediated disruption of fibroblast growth factor 5 in rabbits results in a systemic long hair phenotype by prolonging anagen. Genes (Basel) 2020;11:297.

79. Hashikawa Y, Hayashi R, Tajima M, et al. Generation of knockout rabbits with X-linked severe combined immunodeficiency (X-SCID) using CRISPR/Cas9. Sci Rep 2020;10:9957.

80. Xiao N, Li H, Shafique L, et al. A novel pale-yellow coat color of rabbits generated via MC1R mutation with CRISPR/Cas9 system. Front Genet 2019;10:875.

81. Jiang W, Liu L, Chang Q, et al. Production of Wilson disease model rabbits with homology-directed precision point mutations in the ATP7B gene using the CRISPR/Cas9 system. Sci Rep 2018;8:1332.

82. Song Y, Zhang Y, Chen M, et al. Functional validation of the albinism-associated tyrosinase T373K SNP by CRISPR/Cas9-mediated homology-directed repair (HDR) in rabbits. EBioMedicine 2018;36:517-25.

83. Song Y, Yuan L, Wang Y, et al. Efficient dual sgRNA-directed large gene deletion in rabbit with CRISPR/Cas9 system. Cell Mol Life Sci 2016;73:2959-68.

84. Song J, Wang G, Hoenerhoff MJ, et al. Bacterial and pneumocystis infections in the lungs of gene-knockout rabbits with severe combined immunodeficiency. Front Immunol 2018;9:429.

85. Song J, Yang D, Ruan J, Zhang J, Chen YE, Xu J. Production of immunodeficient rabbits by multiplex embryo transfer and multiplex gene targeting. Sci Rep 2017;7:12202.

86. Yan Q, Zhang Q, Yang H, et al. Generation of multi-gene knockout rabbits using the Cas9/gRNA system. Cell Regen 2014;3:12.

87. Liu H, Sui T, Liu D, et al. Multiple homologous genes knockout (KO) by CRISPR/Cas9 system in rabbit. Gene 2018;647:261-7.

88. Yang D, Song J, Zhang J, et al. Identification and characterization of rabbit ROSA26 for gene knock-in and stable reporter gene expression. Sci Rep 2016;6:25161.

89. Song J, Yang D, Xu J, Zhu T, Chen YE, Zhang J. RS-1 enhances CRISPR/Cas9- and TALEN-mediated knock-in efficiency. Nat Commun 2016;7:10548.

90. Liu Z, Chen M, Chen S, et al. Highly efficient RNA-guided base editing in rabbit. Nat Commun 2018;9:2717.

91. Liu Z, Shan H, Chen S, et al. Improved base editor for efficient editing in GC contexts in rabbits with an optimized AID-Cas9 fusion. FASEB J 2019;33:9210-9.

92. Liu Z, Chen S, Shan H, et al. Efficient base editing with high precision in rabbits using YFE-BE4max. Cell Death Dis 2020;11:36.

93. Liu Z, Chen S, Jia Y, et al. Efficient and high-fidelity base editor with expanded PAM compatibility for cytidine dinucleotide. Sci China Life Sci 2021;64:1355-67.

94. Liu Z, Chen S, Shan H, et al. Precise base editing with CC context-specificity using engineered human APOBEC3G-nCas9 fusions. BMC Biol 2020;18:111.

95. Liu Z, Shan H, Chen S, et al. Highly efficient base editing with expanded targeting scope using SpCas9-NG in rabbits. FASEB J 2020;34:588-96.

96. Chen S, Xie W, Liu Z, et al. CRISPR start-loss: a novel and practical alternative for gene silencing through base-editing-induced start codon mutations. Mol Ther Nucleic Acids 2020;21:1062-73.

97. Zhao D, Qian Y, Li J, Li Z, Lai L. Highly efficient A-to-G base editing by ABE8.17 in rabbits. Mol Ther Nucleic Acids 2022;27:1156-63.

98. Qian Y, Zhao D, Sui T, et al. Efficient and precise generation of Tay-Sachs disease model in rabbit by prime editing system. Cell Discov 2021;7:50.

99. Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013;339:819-23.

100. Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science 2013;339:823-6.

101. Yang D, Xu J, Zhu T, et al. Effective gene targeting in rabbits using RNA-guided Cas9 nucleases. J Mol Cell Biol 2014;6:97-9.

102. Kang Y, Chu C, Wang F, Niu Y. CRISPR/Cas9-mediated genome editing in nonhuman primates. Dis Model Mech 2019;12:dmm039982.

103. Tu Z, Yang W, Yan S, Guo X, Li XJ. CRISPR/Cas9: a powerful genetic engineering tool for establishing large animal models of neurodegenerative diseases. Mol Neurodegener 2015;10:35.

104. Gaudelli NM, Komor AC, Rees HA, et al. Publisher correction: programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 2018;559:E8.

105. Liu Z, Shan H, Chen S, et al. Efficient base editing with expanded targeting scope using an engineered Spy-mac Cas9 variant. Cell Discov 2019;5:58.

106. Hånell A, Marklund N. Structured evaluation of rodent behavioral tests used in drug discovery research. Front Behav Neurosci 2014;8:252.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Zhang Z, Song Y, Lai L, Li Z. Genome-edited rabbit, a prospective alternative model for neurological diseases. Ageing Neur Dis 2022;2:16. http://dx.doi.org/10.20517/and.2022.15

AMA Style

Zhang Z, Song Y, Lai L, Li Z. Genome-edited rabbit, a prospective alternative model for neurological diseases. Ageing and Neurodegenerative Diseases. 2022; 2(4): 16. http://dx.doi.org/10.20517/and.2022.15

Chicago/Turabian Style

Zhang, Zhongtian, Yuning Song, Liangxue Lai, Zhanjun Li. 2022. "Genome-edited rabbit, a prospective alternative model for neurological diseases" Ageing and Neurodegenerative Diseases. 2, no.4: 16. http://dx.doi.org/10.20517/and.2022.15

ACS Style

Zhang, Z.; Song Y.; Lai L.; Li Z. Genome-edited rabbit, a prospective alternative model for neurological diseases. Ageing. Neur. Dis. 2022, 2, 16. http://dx.doi.org/10.20517/and.2022.15

About This Article

Special Issue

© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
1847
Downloads
1028
Citations
1
Comments
0
17

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 25 clicks
Like This Article 17 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Ageing and Neurodegenerative Diseases
ISSN 2769-5301 (Online)

Portico

All published articles will be preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will be preserved here permanently:

https://www.portico.org/publishers/oae/