
Tuberous sclerosis complex (TSC) is a dominant hereditary disease characterized by a form of hamartoma and benign tumors involving multiple organs and systems (Islam and Roach, 2015). TSC shows highly spontaneous mutagenicity with an incidence of 1:6,000–1:10,000 worldwide, seriously affecting the life quality of patients in an age-related and sex-dependent manner (DiMario et al., 2015; Salussolia et al., 2019). The devastating complications caused by pathological changes in the nervous system, heart, kidney, and lungs lead to poor prognosis and even death. Although there are currently accepted diagnostic standards for TSC (Krueger and Northrup, 2013; Portocarrero et al., 2018), the detailed molecular pathogenesis of TSC remains to be elucidated for effective therapeutic purposes.
TSC is closely related to two tumor suppressor genes, TSC1 and TSC2 (Yu et al., 2014; Curatolo et al., 2015). Hamartin and tuberin proteins, encoded by TSC1 and TSC2 genes, respectively, form a heterodimer complex playing a crucial role in regulating cell growth, differentiation, and proliferation through the inhibition of the mechanistic target of rapamycin (mTOR) pathway (Cao et al., 2017; Saxton and Sabatini, 2017). Mutations in either TSC1 or TSC2 promote an overreaction of the mTOR pathway, leading to the occurrence of TSC (Lam et al., 2017; Randle, 2017). Numerous pathogenic mutations of TSC1 and TSC2 genes have been identified in patients, which not only enrich the TSC1 and TSC2 gene mutation spectrum but also show the genetic heterogeneity of TSC (Au et al., 2007; Martin et al., 2017). While the TSC2 mutations exhibit an arbitrary distribution, the TSC1 mutations are frequently reported in the 15th and 17th exons, indicating the existence of mutation hot spots in the TSC1 gene (van Slegtenhorst et al., 1999; Rubilar et al., 2017).
By conventional and conditional knockout of the TSC1 or TSC2 gene, researchers have successfully established and characterized various genetically modified mouse models of TSC (Kobayashi et al., 1999; Onda et al., 1999; Kobayashi et al., 2001; Kwiatkowski et al., 2002). These mouse models have been extensively used and provide valuable insights into the understanding of the pathogenesis and treatment of TSC. However, no mouse model is close to replicate the clinical features, such as cardiac rhabdomyosarcoma (CRM) and subependymal nodules, observed in patients with TSC, which in turn compromise the findings from these mouse models (Kwiatkowski, 2010). The significant differences in physiology between rodents and humans highlight the demand for the study of TSC using larger animal models closer to humans. Currently, pigs are extensively used in biomedical studies in that they have many similarities to humans concerning anatomy, physiology, and clinical relevance (Hai et al., 2017; Yao et al., 2019). More importantly, the CRISPR/Cas9 system along with somatic cell nuclear transfer (SCNT) technology enables the rapid generation of a variety of genetically engineered pigs mimicking human diseases (Chen et al., 2015; Yan et al., 2018). In addition, the short duration of pregnancy, large litter size, and ethical and economic acceptance make pigs a more preferable model than non-human primates for human disease research.
However, no pig model carrying the germline TSC1 mutation, which could be essential to dissect the molecular pathological mechanism of TSC, is yet available. Homozygosity of TSC1 mutation causes embryonic lethality in mice (Kobayashi et al., 2001), indicating that the function of hamartin is essential for mammalian development. Therefore, in the present study, we aim to generate monoallelic TSC1-modified pigs by CRISPR/Cas9-mediated gene targeting and initially characterize their phenotypes.
The TSC1 targeting sgRNA was designed using online tools (https://zlab.bio/guide-design-resources) based on the genomic sequence of the 15th exon of TSC1 of Bama miniature pigs determined by Sanger sequencing. The precise target site is shown in Fig. 1A. To establish TSC1-modified Bama miniature pig fetal fibroblast (PFF) cell lines, we simultaneously transfected a plasmid encoding the Cas9 endonuclease and sgRNA for TSC1 and a neomycin-resistant plasmid (pCMV-tdTomato) into early passages of PFFs derived from a male Bama miniature pig fetus. A total of 40 resistant colonies were harvested after G418 screening for ten days. PCR amplicons spanning the TSC1 target region were ligated into T-vectors for Sanger sequencing. As the homozygous TSC1 mutations caused embryonic lethality in mice (Kobayashi et al., 2001), we screened the TSC1-modified cell colonies that contained a wild-type (WT) TSC1 allele. Of the 40 colonies analyzed, eight were identified as heterozygous ones as each harbored a WT TSC1 allele and a modified TSC1 allele. These cell colonies were pooled and used as cell donors for the SCNT procedure. Twelve live-born male piglets were generated from two recipients (Fig. 1B). The genotypes of cloned piglets were analyzed by PCR sequencing using DNA isolated from their ear tissues. The results revealed that all cloned piglets, except #4 and #9, were heterozygous TSC1 mutants (Fig. 1C). Piglets #4 and #9 were identified as WT, probably owing to contamination of donor cells with a few WT PFFs, which were used as controls in this study.
Albeit the CRISPR/Cas9 system shows high efficiency in pig gene targeting, it might trigger DNA cleavage at undesired sites showing homology to the sgRNA sequence. Therefore, we screened the pig genome sequence using an online tool (http://www.rgenome.net/cas-offinder/) to evaluate the potential off-target sites (OTSs) of the TSC1 sgRNA. In this way, a total of 33 OTSs were computationally identified. We randomly chose 20 OTSs (Table S1) for experimental validation. The PCR amplicons spanning the OTSs from TSC1+/− piglets and WT controls were Sanger sequenced and aligned. No indel mutation was detected in the sequencing reads, indicating that the TSC1+/− piglets are unlikely to have off-target mutagenesis at these sites.
TSC1 gene variations result in overreaction of the mTOR pathway and lead to occurrence of TSC (Lam et al., 2017; Randle, 2017). Among the analyzed samples, TSC1+/− piglet #5 died shortly after birth but showed no external developmental abnormalities in weight, body shape, and skin appearance. To determine whether mTOR signaling was altered in piglet #5, we first conducted a quantitative reverse transcription PCR (qRT-PCR) analysis to assess TSC1 gene expression in the heart and kidney tissues because no commercial antibody against porcine TSC1 is available. Piglet #9 (WT) was used as control. As expected, the TSC1 gene was significantly downregulated in both the TSC1+/− piglet heart and kidney compared with the WT control (Fig. 2A). We then analyzed mTORC1 activity in porcine hearts and kidneys using a readout of the phosphorylated target ribosomal protein S6, a direct substrate of mTORC1 (Ben-Sahra and Manning, 2017). TSC1+/− piglet #5 displayed a significantly higher ratio of phosphor-S6/S6 than piglet #9, namely, a 4- to 5-fold increase relative to the WT piglet, indicating the hyperactivation of mTORC1 signaling due to the monoallelic modification of TSC1 (Fig. 2B). Consistent with the Western blot results, p-S6 expression was significantly elevated in the heart and kidney tissues of the TSC1+/− piglet compared with the WT piglet, as revealed by immunohistochemistry (IHC) staining (Fig. 2C). To examine whether hyperactivation of mTORC1 promotes excessive cell growth and proliferation, pig spleen paraffin sections were subjected to Ki67 staining. The results indicated that the proliferation of spleen cells was significantly enhanced in the TSC1+/− neonatal piglet compared with the WT control (Fig. 2D).
We further assessed TSC1 and p-S6 protein expression in the heart and kidney tissues derived from #3, #7, and #8 adult TSC1+/− pigs. Pig #4 was used as age-matched WT control. The results of qRT-PCR, Western blot, and IHC showed that the TSC1 gene was downregulated and p-S6 was significantly enhanced in the TSC1+/− pigs compared with the WT pig, suggesting that mTOR signaling is sustainably activated upon the absence of a TSC1 allele (Fig. 3A–C). Ki67 staining showed that adult TSC1+/− pigs had appreciable proliferation spleen cells compared with the age-matched WT control (Fig. 3D). Together, these results indicate that the heterozygous TSC1 mutation contributes to the overreaction of the mTOR signaling pathway in pigs.
Pathological changes in the central nervous system, including cortical or subcortical and subependymal nodules, can lead to severe complications and even death in patients with TSC (Lu et al., 2018). To determine whether losing a TSC1 allele causes neurological system damage in pigs, we examined the brain of piglet #5 and used piglet #9 as the WT control. No nodule could be seen on the brain surface of either the TSC1-mutant or the WT piglet. However, compared with piglet #9, the brain of piglet #5 showed severer edema, wider gyrus, shallower sulci, and more transparent meninges on the brain surface (Fig. 4A and B), which are similar to the clinical features of patients with TSC. The brains of piglets #5 and #9 were further examined by using magnetic resonance imaging (MRI) T2-weighted imaging. High-density shadows varying in size were seen in the lateral ventricles on both sides of theTSC1+/− newborn piglet brain with asymmetrical distribution but not in the control (Fig. 4C and D). Moreover, there are many irregularly distributed shadows in the cerebellum and medulla oblongata of the TSC1+/− piglet (Fig. 4E–h). We inferred that the TSC1+/− newborn piglet developed subependymal nodules in the brain owing to the lack of one of the two WT TSC1 alleles.
Cardiac rhabdomyoma occurs in 47–67% of patients with TSC, usually with the most massive tumor volume in the neonatal period and which shrinks to disappear with age (Shen et al., 2015; Aw et al., 2017). Therefore, heart tissue sections of piglets #5 and #9 were examined by hematoxylin and eosin (H&E) staining to assess the presence of cardiac rhabdomyoma. Unlike the WT control, theTSC1+/− newborn piglet heart tissue exhibited lesions with a clear boundary formed by hyperplasia of large vacuolar cells. The nodular cells displayed diverse morphologies and abundant eosinophilic cytoplasm (Fig. 5A). Compared with the adjacent cells, the nodular cells showed significantly increased glycogen content, as indicated by periodic acid-Schiff (PAS) staining (Fig. 5B). IHC analysis revealed that the striated muscle markers, including myoglobin, desmin, and vimentin, were strongly expressed in the nodular cells (Fig. 5C). However, Ki67 was barely detected in the nodular cells, indicating that these cells had no mitotic activity, and the lesions were more likely to be hamartomas than neoplasms (Fig. 5C). Taken together, we inferred that the monoallelic mutation in the TSC1 gene led to TSC in pigs as they developed subependymal nodules and cardiac rhabdomyoma, which are the two major clinical manifestations used to diagnose patients with TSC. Notably, the neonatal TSC1+/− newborn piglet did not develop any renal lesions, liver lesions, or epilepsy seen in TSC1+/− mice. Moreover, we did not observe pathological manifestations in the remaining TSC1+/− piglets even when they were at 16 months of age, partly owing to the genetic heterogeneity of TSC.
Animal models are valuable tools to elucidate the pathological mechanism of TSC and the development of highly specific and efficacious treatments. However, the most widely used mouse models bearing mutations in an allele of TSC1 or TSC2 have limitations as they do not sufficiently recapitulate the pathologic lesions seen in patients with TSC (Kobayashi et al., 2001; Kwiatkowski, 2010; Guo et al., 2016). Pigs have advantages over rodents in modeling human diseases because they have more similarities with humans in anatomy, physiology, and clinical relevance. However, the utilization of genetically engineered pigs as TSC models is still a vacancy. Here, we successfully establish Bama miniature pigs with monoallelic modification of TSC1 using the CRISPR/Cas9 system combined with SCNT technology. The mTORC1 signaling pathway is hyperactivated in both neonatal and adult TSC1+/− Bama miniature pigs. Moreover, the neonatal TSC1+/− Bama miniature pig exhibits CRM and subependymal nodules, which are the two major clinical features for TSC diagnostic criteria, indicating that the loss of aTSC1 allele leads to the occurrence of TSC in pigs.
Mutations in either TSC1 or TSC2 gene cause similar disease phenotypes by disrupting the interaction between their encoded proteins, hamartin and tuberin. Numerous mutations scattered in the genomic regions of these two genes have been extensively associated with TSC phenotypes. A large proportion of TSC1 gene mutations are detected in exons 15 and 17, whereas unanimous hot spot mutations in the TSC2 gene are yet to be determined. Most TSC1 variations are small insertions or deletions, which are also the most common modification forms induced by the CRISPR/Cas9 system via its preference for the error-prone nonhomologous end joining mechanism. The CRISPR/Cas9 technology has been widely applied in pig gene targeting because of its high mutagenesis efficiency. Therefore, the CRISPR/Cas9 system was used to target the 15th exon of TSC1 in this study. Consistent with previous studies (Fang et al., 2018), the CRISPR/Cas9 system showed high gene targeting efficiency in PFFs as eight monoallelic TSC1-modified colonies were obtained in one transfection experiment. A total of 12 TSC1+/− piglets with three kinds of indels in the targeted region were produced by just one round of SCNT, supporting that the CRISPR/Cas9 system in combination with SCNT can significantly facilitate the efficient generation of gene-modified pigs.
The ablation of TSC1 in the brain affects cellular differentiation, migration, and proliferation of the central nervous system, causing characteristic brain lesions in patients. CRM is frequently diagnosed in infants and children associated with TSC1 mutations (Uzun et al., 2007). Our TSC1+/− neonatal piglets showed CRM clearly on histological sections and noncalcified nodules in the subependymal region, medulla oblongata, and cerebellum as per MRI scanning. These phenotypes caused by TSC1 mutations in pigs resemble those in human patients with TSC, reflecting that the function of hamartin is highly conserved in both species. Unfortunately, the TSC1+/− neonatal piglet was in poor overall condition and died soon after birth for uncertain reasons. We suspected that CRM located on the ventricular muscle and/or the widespread subependymal nodules in the brain was a possible cause of its death. We also noticed that the TSC1+/− newborn piglets had an edematous brain surface, widened dorsal brain, and shallow sulci, indicating that the TSC1 gene affects brain development. Further investigation is required. No clinical symptoms associated with patients with TSC were seen in the remaining genetically modified TSC1 pigs even at the age of 16 months, partly because of the genetic heterogeneity of TSC, which was well documented (Martin et al., 2017). As TSC2+/− mice showed more severity in tumor development than TSC1+/− mice, we speculated that the establishment of TSC2-modified pigs would be a choice in future studies to obtain TSC animal models with apparent manifestations. Surprisingly, although the mTOR signal was sustainably upregulated in the TSC1+/− piglets, we have not seen any renal lesions, liver lesions, or epilepsy, which were present in TSC1-knockout mouse models. These discrepancies may raise some crucial issues in terms of pathology and phenotype differences between mice and large animal models of TSC disease.
Our study demonstrates that pig models can more faithfully recapitulate clinical features observed in patients with TSC, indicating that pigs are a valuable tool to model TSC and develop therapeutic strategies. However, there are some limitations in using large animal models because of the great technical difficulty, the expensive animal facility, and the limited sample size. While the sex-dependent difference in TSC incidence has been confirmed in humans and mouse models, we did not consider gender effects on phenotype as only male TSC1+/− piglets were produced in this study. Moreover, a more extensive survey is required because it may take longer than 16 months for the TSC1+/− pigs to develop TSC lesions.
In conclusion, monoallelic TSC1-modified Bama miniature pigs exhibited characteristic TSC features, including CRM and subependymal nodules, which were not observed in TSC1- or TSC2-knockout mouse models. Our TSC1-knockout pigs will serve as useful preclinical animal models to elucidate the pathogenic mechanism of TSC and develop clinical diagnosis and effective treatment of TSC.
Both WT adult Bama miniature pigs and Landrace pigs were purchased from Zhengda Co., Ltd. (Huaiyin, China) and raised in a large animal facility affiliated to Nanjing Medical University. Standard feeding procedures were applied to all animals. Isoflurane anesthesia was used in euthanasia and embryo transplantation to minimize the suffering of pigs. We conducted all animal experiments in accordance with the requirements approved by the Institutional Animal Care and Use Committee of Nanjing Medical University.
sgRNA for the porcine TSC1 gene was designed using online software (https://zlab.bio/guide-design-resources) and synthesized by GenScript (Nanjing, China) (TSC1 sgRNA:5′-CACCGTACGACCACCTTTTTGAGG-3′, 5′-AAACCCTCAAAAAGGTGGTCGTAC-3′). These oligos were mixed and incubated at 37°C for 30 min, heated at 95°C for 5 min, and then cooled down to 25°C at a rate of 5°C/min. The annealed oligos were then subcloned into Bbs I (Thermo Fisher Scientific, Waltham, USA) digested pX330 plasmid to generate the TSC1-targeting vector.
Approximately, 1.5 × 106 early passages of Bama miniature pig PFFs were suspended in 100 μL of Nucleofector solution (Amaxa Biosystems/Lonza, Cologne, Germany) with 5 μg of TSC1 Cas9-sgRNA targeting plasmid and 1 μg of the neomycin-resistant plasmid (pCMV-tdTomato). Electroporation was performed using nuclear infection procedure u-023 following the manufacturer's instructions. Two days later, G418 (Gibco, Grand Island, USA) was added to the post-transfection cells at a concentration of 800 μg/mL and maintained for ten days thereafter. The G418-resistant single-cell colonies were picked and inoculated into 24-well plates. When a confluence of 80% or more was reached, colonies were passaged to 12-well plates. Partial cells of each cell colony were used for PCR genotyping, and the rest of the cells were used as donors for SCNT. The primers used for the TSC1-targeted region amplification were as follows: forward: 5′-TTCGACTCTCCCTTCTACCG-3′; reverse: 5′-ATTAGCCTGTCCAGCACCTC-3′. PCR was performed with standard protocols as described previously by Chen et al. (2018). The PCR amplicons were ligated into a pMD18-T vector (Takara Clontech, Tokyo, Japan). A total of 15–20 individual clones for each amplicon were subjected to Sanger sequencing.
The oocytes were obtained from ovaries of 6-month gilts, which were collected and cultured for maturation (approximately 42–44 h). The enucleation of the mature oocytes was conducted as previously described by Zhang et al. (2018). A single donor cell was injected into the perivitelline cytoplasm of an enucleated oocyte. After electrofusion, the reconstructed embryos were cultured at 38.5°C for 24 h. A total of 250–300 reconstructed embryos were used for embryo transplantation. Pregnancy examination and delivery procedures were performed as described previously by Fang et al. (2018).
The ear tissues of newborn piglets were used to isolate genomic DNA using the QIAamp DNA Mini Kit (QIAGEN, Dusseldorf, Germany). PCR procedures described previously by Chen et al. (2018) were used to genotype the cloned piglets using primers for the TSC1 gene (forward: 5′-TTCGACTCTCCCTTCTACCG-3′, reverse: 5′-ATTAGCCTGTCCAGCACCTC-3′). Total RNA was isolated using the TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions and reverse transcribed into cDNA using the HiScript®II QRT SuperMix (Vazyme, Nanjing, China). The target genes were amplified in three replicates using the SYBR Green PCR Master Mix (Vazyme, Nanjing, China). The primers specific for porcine TSC1 were 5′-TAAAGGGCGGCAGTGAATGT-3′ and 5′-GCATTTGCTTCTAGGCGCTC-3′. The porcine GAPDH gene (forward: 5′-GCAAAGTGGACATTGTCGCCATCA-3′; reverse: 5′-TCCTGGAAGATGGTGATGGCCTTT-3′) was used as an internal control. The qRT-PCR procedures were as follows: 95°C for 5 s, followed by 40 cycles at 95°C for 10 s and 60°C for 30 s. We used the melting curve analysis to evaluate the amplification specificity of qRT-PCR and used the 2−ΔΔCt method (Pfaffl, 2001) to measure the relative gene expression levels.
Porcine tissues including the heart, kidney, and spleen derived from two WT (#4 and #9) and four TSC1+/− piglets (#3, #5, #7, #8) were fixed in 4% paraformaldehyde (PFA) over 24 h, dehydrated, and then embedded in paraffin blocks. The 5-μm sections were deparaffinized and rehydrated for H&E staining and PAS reaction as previously described byFu and Campbell-Thompson (2017) and Chen et al. (2018). For immunohistochemistry analysis, tissue sections were heated at 120°C for 3 min in 10 mM citric acid buffer (pH 6.0) to retrieve the antigen. To block endogenous peroxidase activity, tissue sections were rinsed with 3% H2O2 in methanol for 10 min after washing in PBST three times, then cleaned with PBST, and subsequently blocked with 10% goat serum for 1 h at room temperature. Tissue sections were then incubated with diluted primary antibodies overnight at 4°C. The following primary antibodies were used: anti–phosphor-S6 (S235/236) (#2211; CST, Boston, MA, USA), anti-Ki67 (ab15580; Abcam, Cambridge, MA, USA), anti-desmin (BM4101; BOSTER, Wuhan, China), anti-myoglobin (DF7358; Affinity Biosciences, Wuhan, China), and anti-vimentin (GB14167; Servicebio, Wuhan, China). The tissue sections were incubated with a goat anti-rabbit secondary antibody (sc-2004; Santa Cruz Biotechnology, Santa Cruz, CA, USA) for 1 h at room temperature, then washed, and stained with diaminobenzidine/imidazole. Immunohistochemistry images were acquired using an Olympus FSX100 microscope (Olympus, Tokyo, Japan).
A total of 20–30 μg of heart or kidney tissue extract was used for Western blot analysis. Total proteins were separated by 12% SDS-PAGE gel electrophoresis and transferred onto polyvinylidene fluoride membranes (Bio-Rad, Hercules, CA, USA) by electroblotting. The membranes were incubated at 4°C overnight with anti-phosphor-S6 (S235/236) or anti-S6 (#2217; CST, Boston, MA, USA) primary antibody. Goat anti-rabbit IgG horseradish peroxidase-conjugated secondary antibody (dilution: 1:2000; CWBiotech, Shanghai, China) was incubated with the membranes at 25°C for 1 h. The SuperSignal West Pico Chemiluminescent Substrate (Thermo Fisher Scientific, Waltham, MA, USA) was used for protein detection. Images were captured using a ChemiDoc Touch Imaging System (Bio-Rad, Hercules, CA, USA).
The whole porcine brains derived from two WT (#4 and #9) and four TSC1+/− piglets (#3, #5, #7, #8) were taken out from the cranial cavity and then fixed in 4% PFA for more than 48 h. The Biospec 7T/20 USR magnetic resonance system (Burker, Germany) was used for coronal scanning. The scanning parameters were as follows: RARE sequence T2WI: TR 3679.2 ms, TE 33.0 ms; layer thickness, 1.5 mm; and layer gap, 1.5 mm.
All data were presented as mean ± SD. Data from two groups were analyzed by a two-tailed unpaired Student's t-test. A P-value of 0.05 is set as the threshold value of statistical significance.
Xiaoxue Li: Methodology, Investigation, Validation, Writing - original draft. Tingdong Hu: Investigation, Methodology. Jiying Liu: Methodology, Investigation, Data curation. Bin Fang: Methodology, Investigation. Xue Geng: Methodology, Investigation. Qiang Xiong: Methodology, Investigation. Lining Zhang: Methodology, Investigation.Yong Jin: Methodology, Investigation. Xiaorui Liu: Methodology, Investigation. Lin Li: Methodology, Investigation. Ying Wang: Methodology, Investigation. Rongfeng Li: Methodology, Investigation. Xiaochun Bai: Supervision, Methodology. Haiyuan Yang: Supervision, Funding acquisition, Investigation, Project administration, Writing - original draft, Writing - review & editing. Yifan Dai: Conceptualization, Funding acquisition, Supervision, Writing - review & editing.
We thank all members of Jiangsu Key Laboratory of Xenotransplantation for their helpful suggestions regarding the manuscript. This work was supported by grants from the National Natural Science Foundation of China (31701283, 81970164), the National Key R&D Program of China (2017YFC1103701, 2017YFC1103702), the Jiangsu Key Laboratory of Xenotransplantation (BM2012116), the Sanming Project of Medicine in Shenzhen, the Fund for High Level Medical Discipline Construction of Shenzhen (2016031638), and the Shenzhen Foundation of Science and Technology (JCYJ20160229204849975, GCZX2015043017281705). The authors declared that they have no conflict of interest.
[1] |
Au, K.S., Williams, A.T., Roach, E.S., Batchelor, L., Sparagana, S.P., Delgado, M.R., Wheless, J.W., Baumgartner, J.E., Roa, B.B., Wilson, C.M., Smith-Knuppel, T.K., Cheung, M.Y., Whittemore, V.H., King, T.M., Northrup, H., 2007. Genotype/phenotype correlation in 325 individuals referred for a diagnosis of tuberous sclerosis complex in the United States. Genet. Med. 9, 88-100.
|
[2] |
Aw, F., Goyer, I., Raboisson, M.J., Boutin, C., Major, P., Dahdah, N., 2017. Accelerated cardiac rhabdomyoma regression with everolimus in infants with tuberous sclerosis complex. Pediatr. Cardiol. 38, 394-400.
|
[3] |
Ben-Sahra, I., Manning, B.D., 2017. mTORC1 signaling and the metabolic control of cell growth. Curr. Opin. Cell Biol. 45, 72-82.
|
[4] |
Cao, J., Tyburczy, M.E., Moss, J., Darling, T.N., Widlund, H.R., Kwiatkowski, D.J., 2017. Tuberous sclerosis complex inactivation disrupts melanogenesis via mTORC1 activation. J. Clin. Invest. 127, 349-364.
|
[5] |
Chen, F., Wang, Y., Yuan, Y., Zhang, W., Ren, Z., Jin, Y., Liu, X., Xiong, Q., Chen, Q., Zhang, M., Li, X., Zhao, L., Li, Z., Wu, Z., Zhang, Y., Hu, F., Huang, J., Li, R., Dai, Y., 2015. Generation of B cell-deficient pigs by highly efficient CRISPR/Cas9-mediated gene targeting. J. Genet. Genomics 42, 437-444.
|
[6] |
Chen, Q., Fang, B., Wang, Y., Li, C., Li, X., Wang, R., Xiong, Q., Zhang, L., Jin, Y., Zhang, M., Liu, X., Li, L., Mou, L., Li, R., Yang, H., Dai, Y., 2018. Overexpressing dominant-negative FGFR2-IIIb impedes lung branching morphogenesis in pigs. J. Genet. Genomics 45, 147-154.
|
[7] |
Curatolo, P., Moavero, R., Roberto, D., Graziola, F., 2015. Genotype/phenotype correlations in tuberous sclerosis complex. Semin. Pediatr. Neurol. 22, 259-273.
|
[8] |
DiMario, F.J., Jr., Sahin, M., Ebrahimi-Fakhari, D., 2015. Tuberous sclerosis complex. Pediatr. Clin. North Am. 62, 633-648.
|
[9] |
Fang, B., Ren, X., Wang, Y., Li, Z., Zhao, L., Zhang, M., Li, C., Zhang, Z., Chen, L., Li, X., Liu, J., Xiong, Q., Zhang, L., Jin, Y., Liu, X., Li, L., Wei, H., Yang, H., Li, R., Dai, Y., 2018. Apolipoprotein E deficiency accelerates atherosclerosis development in miniature pigs. Dis. Mod. Mech. 11, dmm036632.
|
[10] |
Fu, D.A., Campbell-Thompson, M., 2017. Periodic acid-schiff staining with diastase. Methods Mol. Bio. 1639, 145-149.
|
[11] |
Guo, Y., Dreier, J.R., Cao, J., Du, H., Granter, S.R., Kwiatkowski, D.J., 2016. Analysis of a mouse skin model of tuberous sclerosis complex. PLoS One 11, e0167384.
|
[12] |
Hai, T., Guo, W., Yao, J., Cao, C., Luo, A., Qi, M., Wang, X., Wang, X., Huang, J., Zhang, Y., Zhang, H., Wang, D., Shang, H., Hong, Q., Zhang, R., Jia, Q., Zheng, Q., Qin, G., Li, Y., Zhang, T., Jin, W., Chen, Z.Y., Wang, H., Zhou, Q., Meng, A., Wei, H., Yang, S., Zhao, J., 2017. Creation of miniature pig model of human Waardenburg syndrome type 2A by ENU mutagenesis. Hum. Genet. 136, 1463-1475.
|
[13] |
Islam, M.P., Roach, E.S., 2015. Tuberous sclerosis complex. Handb. Clin. Neurol. 132, 97-109.
|
[14] |
Kobayashi, T., Minowa, O., Kuno, J., Mitani, H., Hino, O., Noda, T., 1999. Renal carcinogenesis, hepatic hemangiomatosis, and embryonic lethality caused by a germline Tsc2 mutation in mice. Cancer Res. 59, 1206-1211.
|
[15] |
Kobayashi, T., Minowa, O., Sugitani, Y., Takai, S., Mitani, H., Kobayashi, E., Noda, T., Hino, O., 2001. A germ-line Tsc1 mutation causes tumor development and embryonic lethality that are similar, but not identical to, those caused by Tsc2 mutation in mice. Proc. Natl. Acad. Sci. U. S. A. 98, 8762-8767.
|
[16] |
Krueger, D.A., Northrup, H., 2013. Tuberous sclerosis complex surveillance and management: recommendations of the 2012 International Tuberous Sclerosis Complex Consensus Conference. Pediatr. Neurol. 49, 255-265.
|
[17] |
Kwiatkowski, D.J., 2010. Animal models of lymphangioleiomyomatosis (LAM) and tuberous sclerosis complex (TSC). Lymphat. Res. Biol. 8, 51-57.
|
[18] |
Kwiatkowski, D.J., Zhang, H., Bandura, J.L., Heiberger, K.M., Glogauer, M., el-Hashemite, N., Onda, H., 2002. A mouse model of TSC1 reveals sex-dependent lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1 null cells. Hum. Mol. Genet. 11, 525-534.
|
[19] |
Lam, H.C., Nijmeh, J., Henske, E.P., 2017. New developments in the genetics and pathogenesis of tumours in tuberous sclerosis complex. J. Pathol. 241, 219-225.
|
[20] |
Lu, D.S., Karas, P.J., Krueger, D.A., Weiner, H.L., 2018. Central nervous system manifestations of tuberous sclerosis complex. Am. J. Med. Genet. C. Semin. Med. Genet. 178, 291-298.
|
[21] |
Martin, K.R., Zhou, W., Bowman, M.J., Shih, J., Au, K.S., Dittenhafer-Reed, K.E., Sisson, K.A., Koeman, J., Weisenberger, D.J., Cottingham, S.L., DeRoos, S.T., Devinsky, O., Winn, M.E., Cherniack, A.D., Shen, H., Northrup, H., Krueger, D.A., MacKeigan, J.P., 2017. The genomic landscape of tuberous sclerosis complex. Nat. Commun. 8, 15816.
|
[22] |
Onda, H., Lueck, A., Marks, P.W., Warren, H.B., Kwiatkowski, D.J., 1999. Tsc2(+/-) mice develop tumors in multiple sites that express gelsolin and are influenced by genetic background. J. Clin. Invest. 104, 687-695.
|
[23] |
Pfaffl, M.W., 2001. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 29, e45.
|
[24] |
Portocarrero, L.K.L., Quental, K.N., Samorano, L.P., Oliveira, Z.N.P., Rivitti-Machado, M., 2018. Tuberous sclerosis complex: review based on new diagnostic criteria. An. Bras. Dermatol. 93, 323-331.
|
[25] |
Randle, S.C., 2017. Tuberous Sclerosis Complex: A Review. Pediatr. Ann. 46, e166-e171.
|
[26] |
Rubilar, C., Lopez, F., Troncoso, M., Barrios, A., Herrera, L., 2017. [Clinical and genetic study patients with tuberous sclerosis complex]. Rev. Chil. Pediatr. 88, 41-49.
|
[27] |
Salussolia, C.L., Klonowska, K., Kwiatkowski, D.J., Sahin, M., 2019. Genetic etiologies, diagnosis, and treatment of tuberous sclerosis complex. Annu. Rev. Genomics Hum. Genet. 20, 217-240.
|
[28] |
Saxton, R.A., Sabatini, D.M., 2017. mTOR signaling in growth, metabolism, and disease. Cell 168, 960-976.
|
[29] |
Shen, Q., Shen, J., Qiao, Z., Yao, Q., Huang, G., Hu, X., 2015. Cardiac rhabdomyomas associated with tuberous sclerosis complex in children. From presentation to outcome. Herz 40, 675-678.
|
[30] |
Uzun, O., Wilson, D.G., Vujanic, G.M., Parsons, J.M., De Giovanni, J.V., 2007. Cardiac tumours in children. Orphanet J. Rare Dis. 2, 11.
|
[31] |
van Slegtenhorst, M., Verhoef, S., Tempelaars, A., Bakker, L., Wang, Q., Wessels, M., Bakker, R., Nellist, M., Lindhout, D., Halley, D., van den Ouweland, A., 1999. Mutational spectrum of the TSC1 gene in a cohort of 225 tuberous sclerosis complex patients: no evidence for genotype-phenotype correlation. J. Med. Genet. 36, 285-289.
|
[32] |
Yan, S., Tu, Z., Liu, Z., Fan, N., Yang, H., Yang, S., Yang, W., Zhao, Y., Ouyang, Z., Lai, C., Yang, H., Li, L., Liu, Q., Shi, H., Xu, G., Zhao, H., Wei, H., Pei, Z., Li, S., Lai, L., Li, X.J., 2018. A Huntingtin knockin pig model recapitulates features of selective neurodegeneration in Huntington's disease. Cell 173, 989-1002.e13.
|
[33] |
Yao, J., Zeng, H., Zhang, M., Wei, Q., Wang, Y., Yang, H., Lu, Y., Li, R., Xiong, Q., Zhang, L., Chen, Z., Xing, G., Cao, X., Dai, Y., 2019. OSBPL2-disrupted pigs recapitulate dual features of human hearing loss and hypercholesterolaemia. J. Genet. Genomics 46, 379-387.
|
[34] |
Yu, Z., Zhang, X., Guo, H., Bai, Y., 2014. A novel TSC2 mutation in a Chinese family with tuberous sclerosis complex. J. Genet. 93, 169-172.
|
[35] |
Zhang, R., Wang, Y., Chen, L., Wang, R., Li, C., Li, X., Fang, B., Ren, X., Ruan, M., Liu, J., Xiong, Q., Zhang, L., Jin, Y., Zhang, M., Liu, X., Li, L., Chen, Q., Pan, D., Li, R., Cooper, D.K.C., Yang, H., Dai, Y., 2018. Reducing immunoreactivity of porcine bioprosthetic heart valves by genetically-deleting three major glycan antigens, GGTA1/β4GalNT2/CMAH. Acta Biomater. 72, 196-205.
|
[1] | Siyu Chen, Zhiquan Liu, Hao Yu, Liangxue Lai, Zhanjun Li. Efficient multinucleotide deletions using deaminase-Cas9 fusions in human cells[J]. Journal of Genetics and Genomics, 2022, 49(10): 927-933. doi: 10.1016/j.jgg.2022.03.007 |
[2] | Lin Zhu, Xiaoyan Yang, Juyi Li, Xiong Jia, Xiangli Bai, Ying Zhao, Wenzhuo Cheng, Meng Shu, Yan Zhu, Si Jin. Leptin gene-targeted editing in ob/ob mouse adipose tissue based on the CRISPR/Cas9 system[J]. Journal of Genetics and Genomics, 2021, 48(2): 134-146. doi: 10.1016/j.jgg.2021.01.008 |
[3] | Jinfu Zhang, Emmanuel M. Khazalwa, Hussein M. Abkallo, Yuan Zhou, Xiongwei Nie, Jinxue Ruan, Changzhi Zhao, Jieru Wang, Jing Xu, Xinyun Li, Shuhong Zhao, Erwei Zuo, Lucilla Steinaa, Shengsong Xie. The advancements, challenges, and future implications of the CRISPR/Cas9 system in swine research[J]. Journal of Genetics and Genomics, 2021, 48(5): 347-360. doi: 10.1016/j.jgg.2021.03.015 |
[4] | Yan Li, Wenjing Li, Jun Li. The CRISPR/Cas9 revolution continues: From base editing to prime editing in plant science[J]. Journal of Genetics and Genomics, 2021, 48(8): 661-670. doi: 10.1016/j.jgg.2021.05.001 |
[5] | Sukumar Biswas, Jiaqi Tian, Rong Li, Xiaofei Chen, Zhijing Luo, Mingjiao Chen, Xiangxiang Zhao, Dabing Zhang, Staffan Persson, Zheng Yuan, Jianxin Shi. Investigation of CRISPR/Cas9-induced SD1 rice mutants highlights the importance of molecular characterization in plant molecular breeding[J]. Journal of Genetics and Genomics, 2020, 47(5): 273-280. doi: 10.1016/j.jgg.2020.04.004 |
[6] | Jian Li, Zheng Wang, Guangming He, Ligeng Ma, Xing Wang Deng. CRISPR/Cas9-mediated disruption of TaNP1 genes results in complete male sterility in bread wheat[J]. Journal of Genetics and Genomics, 2020, 47(5): 263-272. doi: 10.1016/j.jgg.2020.05.004 |
[7] | Fenghua Zhang, Xianmei Li, Mudan He, Ding Ye, Feng Xiong, Golpour Amin, Zuoyan Zhu, Yonghua Sun. Efficient generation of zebrafish maternal-zygotic mutants through transplantation of ectopically induced and Cas9/gRNA targeted primordial germ cells[J]. Journal of Genetics and Genomics, 2020, 47(1): 37-47. doi: 10.1016/j.jgg.2019.12.004 |
[8] | Chang Ye, Zhuoxin Chen, Zhan Liu, Feng Wang, Xionglei He. Defining endogenous barcoding sites for CRISPR/Cas9-based cell lineage tracing in zebrafish[J]. Journal of Genetics and Genomics, 2020, 47(2): 85-91. doi: 10.1016/j.jgg.2019.11.012 |
[9] | Xiaohu Su, Wei Chen, Qingqing Cai, Puping Liang, Yaosheng Chen, Peiqing Cong, Junjiu Huang. Production of non-mosaic genome edited porcine embryos by injection of CRISPR/Cas9 into germinal vesicle oocytes[J]. Journal of Genetics and Genomics, 2019, 46(7): 335-342. doi: 10.1016/j.jgg.2019.07.002 |
[10] | Bei Yang, Xiaosa Li, Liqun Lei, Jia Chen. APOBEC: From mutator to editor[J]. Journal of Genetics and Genomics, 2017, 44(9): 423-437. doi: 10.1016/j.jgg.2017.04.009 |
[11] | Yufeng Hua, Chun Wang, Jian Huang, Kejian Wang. A simple and efficient method for CRISPR/Cas9-induced mutant screening[J]. Journal of Genetics and Genomics, 2017, 44(4): 207-213. doi: 10.1016/j.jgg.2017.03.005 |
[12] | Renjie Jiao, Caixia Gao. The CRISPR/Cas9 Genome Editing Revolution[J]. Journal of Genetics and Genomics, 2016, 43(5): 227-228. doi: 10.1016/j.jgg.2016.05.004 |
[13] | Chao Feng, Jing Yuan, Rui Wang, Yang Liu, James A. Birchler, Fangpu Han. Efficient Targeted Genome Modification in Maize Using CRISPR/Cas9 System[J]. Journal of Genetics and Genomics, 2016, 43(1): 37-43. doi: 10.1016/j.jgg.2015.10.002 |
[14] | Aurélie Lemoine, Gaëlle Chauveau-Le Friec, Francina Langa, Cédric Louvet. Generation of a Double KO Mouse by Simultaneous Targeting of the Neighboring Genes Tmem176a and Tmem176b Using CRISPR/Cas9: Key Steps from Design to Genotyping[J]. Journal of Genetics and Genomics, 2016, 43(5): 329-340. doi: 10.1016/j.jgg.2016.04.004 |
[15] | Yue Mei, Yan Wang, Huiqian Chen, Zhong Sheng Sun, Xing-Da Ju. Recent Progress in CRISPR/Cas9 Technology[J]. Journal of Genetics and Genomics, 2016, 43(2): 63-75. doi: 10.1016/j.jgg.2016.01.001 |
[16] | Chun Wang, Lan Shen, Yaping Fu, Changjie Yan, Kejian Wang. A Simple CRISPR/Cas9 System for Multiplex Genome Editing in Rice[J]. Journal of Genetics and Genomics, 2015, 42(12): 703-706. doi: 10.1016/j.jgg.2015.09.011 |
[17] | Andrew R. Bassett, Lesheng Kong, Ji-Long Liu. A Genome-Wide CRISPR Library for High-Throughput Genetic Screening in Drosophila Cells[J]. Journal of Genetics and Genomics, 2015, 42(6): 301-309. doi: 10.1016/j.jgg.2015.03.011 |
[18] | Andrew R. Bassett, Ji-Long Liu. CRISPR/Cas9 and Genome Editing in Drosophila[J]. Journal of Genetics and Genomics, 2014, 41(1): 7-19. doi: 10.1016/j.jgg.2013.12.004 |
[19] | Chuanxian Wei, Jiyong Liu, Zhongsheng Yu, Bo Zhang, Guanjun Gao, Renjie Jiao. TALEN or Cas9 – Rapid, Efficient and Specific Choices for Genome Modifications[J]. Journal of Genetics and Genomics, 2013, 40(6): 281-289. doi: 10.1016/j.jgg.2013.03.013 |
[20] | Ruifang Mi, Jianhui Ma, Dechang Zhang, Limin Li, Hongbing Zhang. Efficacy of combined inhibition of mTOR and ERK/MAPK pathways in treating a tuberous sclerosis complex cell model[J]. Journal of Genetics and Genomics, 2009, 36(6): 355-361. doi: 10.1016/S1673-8527(08)60124-1 |
1. | Li, X., Zhang, Y., Chang, J. et al. Mfsd2a attenuated hypoxic-ischemic brain damage via protection of the blood–brain barrier in mfat-1 transgenic mice. Cellular and Molecular Life Sciences, 2023, 80(3): 71. doi:10.1007/s00018-023-04716-9 | |
2. | Zhou, H., Yu, B., Sun, J. et al. Gut microbiota absence and transplantation affect diarrhea: an investigation in the germ-free piglet model. Animal Biotechnology, 2023. doi:10.1080/10495398.2023.2248200 | |
3. | Zhou, H., Yu, B., Sun, J. et al. Comparison of maternal and neonatal gut microbial community and function in a porcine model. Animal Biotechnology, 2023, 34(7): 2972-2978. doi:10.1080/10495398.2022.2126367 | |
4. | Ning, Y.. Schiff-linked prodrugs of pH-responsive nanoparticles with high drug loading and improved drug delivery. AIP Conference Proceedings, 2022. doi:10.1063/5.0093876 | |
5. | Zhang, J., Sun, X., Cao, C. Recent advances in genetically modified large-animal models of human diseases. BIO Integration, 2022, 3(4): 161-171. doi:10.15212/bioi-2022-0018 | |
6. | Wang, W., Yuan, Y., Li, L. et al. Establishment of PIN1 knockout porcine fetal fibroblast cell lines via CRISPR/Cas9 mediated gene-targeting | [PIN1基因敲除的长白猪胎儿成纤维细胞系的建立]. Journal of Army Medical University, 2022, 44(13): 1349-1355. doi:10.16016/j.2097-0927.202112217 | |
7. | Yum, S.-Y., Jang, G., Koo, O. Target-aid-mediated multiplex base editing in porcine fibroblasts. Animals, 2021, 11(12): 3570. doi:10.3390/ani11123570 |