Skip to main content
Erschienen in:

Open Access 10.06.2024 | Original Article

CRISPR/Cas9 Mediated Deletion of the Uox Gene Generates a Mouse Model of Hyperuricemia with Multiple Complications

verfasst von: Linzi Zeng, Shalaimaiti Shali, Yabiao Gao, Xingchen Du, Xiaoxia Zhu, Lin Li, Yuxiang Dai, Ping Zhou

Erschienen in: Journal of Cardiovascular Translational Research | Ausgabe 6/2024

Abstract

Hyperuricemia is a common metabolic disorder with severe complications. We aimed to develop a mouse model for spontaneous hyperuricemia. Uox-/- mouse model was generated on C57BL/6J background by deleting exon 2-4 of Uox using the CRISPR/Cas9 system. The prototypic Uox -/-mice had 5.5-fold increased serum uric acid (1351.04±276.58μmol/L) as compared to the wild type mice (P<0.0001), but died by 4 weeks. After allopurinol (3ug/g) intervention, they all survived > 8 weeks. The serum uric acid was 612.55±146.98μmol/L in the 8-week-old allopurinol-rescued Uox -/-mice, which manifested multiple complications including severe renal insufficiency, hypertension, left ventricular remodeling and systolic dysfunction, aortic endothelial dysfunction, hepatic steatosis and elevated liver enzymes, as well as hyperglycemia and hypercholesteremia. The present Uox-/- mice developed spontaneous hyperuricemia complicated with urate nephropathy, cardiovascular disease and cardiometabolic disorders, and may provide a novel tool to study hyperuricemia associated early-onset cardiovascular disorders in human.

Graphical Abstract

A mouse model of hyperuricemia with multiple complications constructed by knocking out of urate oxidase (Uox) using CRISPR/Cas9 technology. Uox-/-: homozygous; Uox+/-: heterozygous; SUA: serum uric acid; ALT: alanine aminotransferase; AST: aspartate aminotransferase.
Begleitmaterial
Hinweise
Associate Editor Junjie Xiao oversaw the review of this article

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12265-024-10526-6.
Linzi Zeng, Shalaimaiti Shali, and Yabiao Gao Contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALT
Alanine aminotransferase
AST
Aspartate aminotransferase
BP
Blood pressure
cDNA
Complementary DNA
HDL-C
High density lipoprotein cholesterol
HE
Hematoxylin-eosin
IQR
Interquartile ranges
LDL-C
Low-density lipoprotein cholesterol
NAFLD
Nonalcoholic fatty liver disease
PCNA
Proliferating cell nuclear antigen
PCR
Polymerase chain reaction
SUA
Serum uric acid
SEM
Standard error of mean
TC
Total cholesterol
TG
Triglyceride
UA
Uric acid (UA)
UOX
Urate oxidase
VG
Van Gieson
WGA
Wheat Germ Agglutinin
WT
Wild type (WT)

Introduction

Hyperuricemia, characterized by increased concentration of serum uric acid (SUA) above 420 μmol/L, has been an increasingly common metabolic disorder with a precipitous increase particularly among young adults [1, 2]. It is the major causal risk factor of gout and urate-nephropathy, and often entangles with a cluster of cardiometabolic disorders including hypertension, diabetes mellites, dyslipidemia and nonalcoholic fatty liver disease (NAFLD), which could jointly contribute to the development of cardiovascular disease [3]. However, the independent causal effect of hyperuricemia on these metabolic comorbidities and cardiovascular disease remains largely obscure [4]. Therefore, murine models to replicate early-onset hyperuricemia in humans is of paramount importance to better understand the progression trajectories and underlying mechanisms, and to develop effective medications.
Uric acid (UA) is an end product of purine metabolism that is specific to humans due to silencing of the gene encoding urate oxidase (Uox) or uricase during primate evolution. Unlike human, most mammals have active Uox that further degrades UA to allantoin, and therefore challenges exist in the establishment of efficient and sustainable animal model of hyperuricemia using pharmaceutical or dietary-induced approaches [5]. Given the major difference in Uox gene expression between human and mice, genetic modifications that target Uox either by homologous recombination in embryonic stem cells [6] or by using transcription activator-like effector nuclease mediated deletion have been used to generate Uox knockout (Uox-/-) mouse model with spontaneously high SUA concentrations [7]. However, both embryonic lethality and postnatal mortality are the major hurdles of these Uox deficient mice. Alternatively, CRISPR/Cas9 is an advanced technology that facilitates more precise genetic engineering. The uricase-deficient “Kunming-DY rats” with stable hyperuricemia and better survival is an example of such an approach [8]. Herein, we established a Uox-/- mouse model on C57BL/6J genetic background by deleting the exon 2-4 of Uox using CRISPR/Cas9 system. In this study, we present the hyperuricemic phenotypes regarding magnitude of serum UA elevation, urate nephropathy, cardiometabolic abnormalities, as well as cardiovascular and hepatic complications.

Materials and Methods

Construction of the Uox-/- Mouse Model

C57BL/6J Mice were raised in the Department of Laboratory Animal Science of Shanghai Medical college of Fudan University in a specific pathogen free environment at 22°C, with a humidity of 45%-55%, under 12-hour light-dark cycle, and with free approach to food and water.
The Uox gene is located on chromosome three, and had eight exons and six transcripts. Two gRNA fragments (gRNA1: 5’-GTTAACTCCAAACTATATAG-3’; gRNA2: 5’-GGTTACTGGATCATTGGTAC-3’) targeting at three exons (exon 2-4) of the Uox-201 transcript were generated by in vitro transcription, and then co-injected together with commercially available Cas9 protein into fertilized C57BL/6J mouse eggs. Subsequently, these eggs were transplanted into the oviducts of pseudopregnant female mice for embryonic development. Uox-/- mice (F0) were matched with Wild type (WT) mice to generate heterozygous Uox+/- mice (F1). Finally, homozygous Uox-/- (F2) offspring were generated by heterozygous mating (Graphical abstract).
Genotyping was conducted at one week after birth by polymerase chain reaction (PCR) amplification of complementary DNA (cDNA) obtained from tail tissue (Supplementary materials). The PCR products were separated by DNA electrophoresis at 313 base pairs (bp) for WT allele, at 462bp for homogeneous Uox-/- allele, while at both 313bp and 462bp for heterogeneous Uox+/- allele respectively, suggesting 3,295 bp would be deleted. (Fig. 1a).

Quantitative RT-PCR and Western Blotting

Total RNA was isolated from the liver tissue using Trizol reagent (Vazyme, Nanjing, China) and then reverse-transcribed into cDNA using a Hiscript III Reverse Transcriptase Kit (Vazyme, Nanjing, China). The sequencings of primers were 5’-GGACCTGACTGACTACCTCAT-3’ (β-actin-forward), 5’-GGACCTGACTGACTACCTCAT-3’ (β-actin-reverse), 5’-CCCAGGCTAAACTCTCAGGCT-3’ (Uox-forward), and 5’-TGTCAGGGAAACAGTCATTTCACA-3’ (Uox-reverse). Real-time quantitative PCR was performed using AceQ qPCR SYBR Green Master Mix (Vazyme, Nanjing, China) on a Fluorescent Quantitative PCR (Biored, California, USA) with the following parameters: 94°C five minutes, 94°C30 seconds, 60°C 30 seconds, 72°C one minute, 35 cycles. The threshold cycle (Ct) was determined and used to calculate ΔCT values. The ΔΔCt (2-ΔΔCt) was used to calculate relative mRNA expression, with each measurement performed in triplicate.
Uox protein expression was determined by Western blot using anti-Uox primary antibody (at 1:500; Santa Cruz Biotechnology, Dallas, TX) and anti-β-tubulin antibody (at 1:10000; ptoteintech, Wuhan, China). The membranes were then incubated with horseradish peroxidase-conjugated anti-mouse IgG (1:10000; CST, Boston, USA). Each measurement was performed in triplicate, and all results were normalized against β-tubulin.

Serum Biochemical Analysis

Blood sampling was performed from the outer canthus of anesthetized mice after overnight fasting. After one hour incubation at a room temperature, serum was obtained by spinning at 3,000×g and at 4°C for five minutes. SUA was measured using uric acid assay kit (Abcam, Cambridge, UK) following the protocol. Other serum biochemical indicators including, urea, creatinine, aspartate aminotransferase (AST), alanine aminotransferase (ALT), total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high density lipoprotein cholesterol (HDL-C) and fasting glucose were determined using an automatic biochemical analyzer (Hitachi, Tokyo, Japan).

Blood Pressure and Echocardiographic Measurement

The blood pressure (BP) measurements were conducted using the BP-2000 non-invasive BP analysis system (Visitech system, Drammen, Norway) with methods adhering to the provided instructions. Echocardiographic examination was performed using two-dimensional and M-mode imaging by the high-resolution real-time ultrasound pre-clinical imaging system (Fujifilm Visual Sonics, Toronto, Canada) in the parasternal long-axis view. Dimensional and functional parameters of the left ventricle were measured at the level of the papillary muscles.

Histological Analysis

Tissue sections of the kidney, heart, aorta and liver were subjected to hematoxylin-eosin (HE) staining, and visualized under fluorescence microscope in a light mode. Kidney sections were also stained by Masson’s trichrome. Besides, Wheat Germ Agglutinin (WGA) staining, Van Gieson (VG) staining and Oil Red O Staining were performed on the tissues from heart, aorta and liver, respectively. Moreover, immunohistochemical analyses were conducted using primary antibodies as follows: Uox (1:100; Santa Cruz Biotechnology, Dallas, TX), lymphocyte CD3 (1:1000, Proteintech, Wuhan, China), macrophage CD68 (1:1000, Proteintech, Wuhan, China), proliferating cell nuclear antigen (PCNA) (1;1000, Proteintech, Wuhan, China), ZO1 (1:300, Abmart, Shanghai, China) and Ve-cadherin (1:200, R&D Systems, Minneapolis, MN, USA).

Statistical Analysis

All quantitative values were presented in the form of mean ± standard error of mean (SEM) or median with the interquartile ranges (IQR) as appropriate, and the differences between groups were analyzed by either Student’s t-test or Mann–Whitney non-parametric tests. Categorical variables were presented as absolute values (percentages). Survival analysis was analyzed by Log-rank test. P < 0.05 was considered statistically significant.

Results

Generation of Uox-/- Mice with Spontaneous Hyperuricemia

Both mRNA and protein expressions of Uox were absent in the liver of Uox-/- mice , but not in the Uox+/- and WT mice (Fig. 1b-d). Notably, Uox-/- mice (3 weeks old) had an extremely high SUA concentration (1351.04±276.58μmol/L), which was 5.5-fold higher than that in WT mice (248.19±100.59μmol/L, P<0.0001) (Fig. 1e). Albeit significant reduction of Uox hepatic expression (P=0.0091, Fig. 1b-d), the Uox+/- mice (292.60±52.42μmol/L) had unchanged levels of SUA as compared to WT mice (P=0.364, Fig. 1e).
The birth rate of Uox-/- mice was at 21.35% (114 of 534), slightly lower than the expected Mendelian frequency, suggesting either embryonic lethality or neonatal death in the first week. Besides, they barely survived to four weeks (median: 3.0 weeks, IQR:1.7 to 3.6 weeks). However, daily gastric administration of allopurinol (3μg/g) [6] after genotyping not only reduced the concentrations of SUA by 54.75% (612.55±146.98μmol/L) to the similar levels in human hyperuricemia (P=0.0067, allopurinol-treated vs. not treated, Fig. 1f), but also improved their survivals up to 16 weeks (median: 12.7 weeks, IQR: 9.9-13.4 weeks, P<0.0001, Fig. 1g). Hence, the following phenotypic examinations were performed on the allopurinol-treated Uox-/- mice at the age of eight weeks.

Renal Dysfunction and Nephropathy

The glomerular filtration function was compromised in the Uox-/- mice, as indicated by elevated levels of blood urea (21.35±1.50 vs. 13.54±0.57 mmol/L, P<0.0005) and serum creatinine (24.20±1.45 vs. 10.68±0.72mmol/L, P<0.0001) compared to WT controls (Fig. 2a-b). Histological examination revealed severe kidney structural abnormalities. To begin, the tubular walls were thin, showing necrotic epithelial cells with evident karyopyknosis; the tubules were also dilated and filled with massive granular casts along with amyloid exudation; besides, severe interstitial bleeding in the renal medulla was also common (Fig. 2c). In addition, there was significant glomeruli enlargement with thronged erythrocytes and decreased capsular space; the mesangial expansion and increased eosinophilic matrix were also frequently detected (Fig. 2d). Moreover, the significant tubulointerstitial fibrosis and glomerulosclerosis were commonly observed (Fig. 2e), and so were the urate crystal deposits under polarized light microscope (Fig. 2f). Importantly, there were small amount of (CD3+) T lymphocytes (P=0.0050), and massive (CD68+) macrophages (P=0.0002) infiltration, indicating substantial corticomedullar inflammation (Fig. 2g-h).

Hypertension and Cardiometabolic Disturbance

The systolic BP was significantly higher in the Uox-/- mice (155.25±9.48mmHg) than in the WT controls (109.53±11.02mmHg, P< 0.0001), indicating that the Uox-/- mice developed early-onset hypertension (Fig. 3a). Also, biochemical analysis confirmed the deleterious effects of hyperuricemia on both glucose and lipid profiles. Compared with WT counterparts, the Uox-/- mice had significantly elevated fasting glucose (P=0.0146) and TC (P=0.0045) (Fig. 3b-c); besides, there was a trend towards elevated levels of TG and LDL-C, and decreased levels of HDL-C in the Uox-/- mice, although not statistically significant (all P>0.05, Fig. 3d-f).

Cardiovascular Complications

Echocardiography showed markedly reduced ejection fraction (P=0.0097), fractional shortening (P=0.0084), cardiac output (P=0.0071), and stroke volume (P=0.0008) in Uox-/- mice as compared to WT mice (Fig. 4a-b). Although, changes in echocardiographic dimensional parameters were not significant (Supplementary Fig. 1), histological examination revealed left ventricular wall thickening and decreased ventricular cavity in the Uox-/- mice. Microscopic changes included hypertrophic cardiomyocytes with cytoplasmic vacuolation and myofibrillar fragmentation (Fig. 4c and d).
In addition, there were multiple pathological changes in the aorta of Uox-/- mice. As compared to WT controls, there were more abundant aortic collagen fibers in the outer layer detected by VG staining (Fig. 4e). Moreover, the PCNA positive cells were less frequent (P<0.0001), indicating decreased proliferation of aortic endothelial cells (Fig. 4f); both ZO-1 (P<0.0001) and VE-cadherin (P=0.0270) expression were also significantly down-regulated in the aortic endothelial cells, suggesting that vascular endothelial barrier was compromised in the hyperuricemic mice (Fig. 4g and h).

Liver Injury

Hepatocyte injury in Uox-/- mice was evidenced by the elevated levels of serum AST (P=0.0146) and ALT (P=0.0145) enzymes (Fig. 5a-b). Pathohistological changes included significantly widened sinusoidal space, enlarged hepatocytes with lightly stained cytoplasm and nuclei, as well as cellular ballooning (Fig. 5c). Oil-red O staining detected highly accumulated lipid droplets in the liver of Uox-/- mice in comparison with their WT counterparts (P<0.0001, Fig. 5d).

Discussion

Despite many efforts to model hyperuricemia in human, a suitable mouse model that have stable hyperuricemia and normal lifespan has been particularly lack [9]. The present study introduced a novel Uox deficient mice with spontaneous hyperuricemia generated by CRISPR/Cas9 system on a pure C57BL/6J background. To improve postnatal survival, we also applied a strategy that combined genetic manipulation with allopurinol intervention, which enabled the SUA concentration not lethally high but maintain valid that mimics “human like” hyperuricemia. Moreover, this allopurinol-rescued Uox-/- mice presented various complications including kidney disease, metabolic syndrome, cardiovascular disease and NAFLD. The present mouse model could provide a candidate tool to study early-onset hyperuricemia and associated comorbidities.
Hyperuricemia is caused by hepatic overproduction and (or) renal and (or) intestinal underexcretion of SUA, which are regulated by genetic and environmental factors and their interactions [10]. Obviously, owing to the evolutionary inactivation of Uox in humans, genetic modification of rodent Uox is an essential and most effective way to replicate human hyperuricemia and urate biology, as compared to genetically engineered mice targeting other loci (for example, SLC2A9 and ABCG2), as well as environmentally induced models [9, 1113].
The first Uox knockout model was generated on hybrid genetic background mouse (C57BL/6J*129Sv) by Neomycin-cassette insertion into exon 3 of the Uox that resulted in resulted in an elevated SUA concentration of 650 μmol/L, 12 times higher than that in the WT controls [6]. Lately, the second Uox-/- mice was established on a C57BL/6J background by deleting 28 bp of exon 3 using TALEN technique, and the SUA levels were at 420 to 520 μmol/L, two to three times higher than in WT mice [7]. At present, ours represents the third Uox knockout mouse model so far, which has been generated on C57BL/6J background using CRISPR/Cas9 system for the first time. The exon 2 to exon 4 were targeted, which account for about 46% of the entire Uox protien coding gene. By deleting a bigger region (3296 bp) than the other models [6, 7], a frameshift mutation was expected with more significant phenotypic effect. In addition, deleting these three exons would result in less N-terminal amino acid residue. Beside, targeting other exons could affect the Dnase2b, since they coincide with the exons of Dnase2b. What’s more, off-target effect was highly unlikely given that their offspring presented the expected and stable biological features. Consequently, our Uox-/- mice reached an extremely high SUA concentration of 1351.04±276.58 μmol/L, which was 5.5-fold higher than that in WT mice (248.19±100.59μmol/L), while allopurinol-treated Uox-/- mice had moderately elevated SUA (612.55±146.98μmol/L), 2.5-time higher than that in WT mice. Due to lack of standard protocol for blood sampling and urate measurement, these reported SUA values varied widely without a clear definition of normal range in mice, and therefore were not directly comparable. Even though, the proportional increase in SUA suggested that present Uox-/- mice yield a strong phenotypic effect on SUA level.
Without a gradual adaption to the evolutionary changes in urate mediated biological system [9], acute disruption in the Uox gene may largely explain the substantial embryonic and postnatal mortality in mice. The percentages of the Uox-/- mice born from heterozygous mating were reported at 7.1% [6] and 15.9% [7], respectively, both far below the expected Mendelian frequency. By contrast, the birth rate in present Uox-/- mice is 21.4%, approximated to the expected rate of 25%, suggesting the advantage of CRISPR/Cas9 technique in reducing embryonic lethality. However, neonatal mortality at four weeks after birth was extremely higher (92%) in the present model than the previous reports (40%~65%) [6, 7]. The same genetic approach targeting at exon 2-4 of Uox on Sprague Dawley rats resulted in a 95% survival up to one year, but only mild hyperuricemia, suggesting different responses to Uox inactivation between animal species [8]. Clearly, the extremely high SUA is the fundamental cause of premature death in the present Uox-/-mice. With a 55% reduction in SUA by allopurinol intervention, they all could survive to eight weeks to be sexually matured.
The kidney is one of the important organs of urate metabolism, and studies have shown that patients with hyperuricemia are at increased risk of renal disease [3, 13]. As previous hyperuricemic mouse models [6, 7, 14, 15], our experimental results demonstrated severe renal insufficiency evidenced by remarkably increased levels of blood urea and serum creatinine, significant glomerular and tubular deformations accompanied by corticomedullar inflammation, interstitial fibrosis and uric acid crystal deposition. Given the fact that SUA concentration was even doubled in the Uox-/- mouse prototype, we assume that severe urate nephropathy and renal failure in the first four weeks of life may account for their poor survival.
Epidemiological and experimental studies have shown that hyperuricemia is associated with hypertension, coronary atherosclerosis and heart failure [1623]. However, the causal relationship between UA and cardiovascular disease remains controversial. Our experimental mouse model of hyperuricemia manifested hypertension along with distinct ventricular remodeling, reduced cardiac output and significant aortic endothelial dysfunction, which are consistent with the literature [2123]. Owing to the renal failure, the independent role of hyperuricemia in the development of hypertension remains uncertain. Likewise, both kidney disease and hypertension may contribute to cardiac dysfunction and atherosclerosis, reiterating the issue of inconclusive causal relationship between hyperuricemia and cardiovascular disease. Notwithstanding, such a complex links enables our Uox-/- mice as an appropriate model to study the progression trajectories of these comorbidities from birth to adulthood. Another appealing use is to detect whether new urate lowering drugs in addition to allopurinol could further ameliorate early-onset hypertension and cardiovascular complications as compared to allopurinol alone.
In addition to hypertension, our experimental mouse model of hyperuricemia manifested distinct hyperglycemia and dyslipidemia, all of which are typical to metabolic syndrome [18]. Onset sequence study showed that hyperuricemia is an earlier-onset metabolic disorder in relation to hypertension, hypertriglyceridemia, and diabetes mellitus, indicating a potential upstream role of hyperuricemia in the development of metabolic syndrome [1]. It was reported that high SUA could directly cause pancreatic β-cell apoptosis and dysfunction [24, 25]. The previous results from Uox-/- mice also supported hyperuricemia was probably a causal factor of islet dysfunction and therefore diabetes [7]. Whether hyperuricemia also induces insulin resistance in Uox-/- mice warrants further investigation.
Regarding lipid metabolism, our Uox-/- mice displayed so-called atherogenic lipid triad: markedly increased levels of TC, TG and LDL-C, as well as decreased levels of HDL-C, although not reaching a statistical significance except for TC. The underlying mechanism is unclear. One possible explanation is that knocking out of Uox may cause hepatocellular injury, as evidenced in our Uox-/- mice, thereby disturbing hepatic lipid metabolism [25]. On the other hand, excessive influxes of lipids could promote fat accumulation and NAFLD, which has been widely recognized as a cardiometabolic disorder [26]. NAFLD is speculated to cause hyperuricemia by inducing the expression of hepatic xanthine oxidase without changing Uox expression [27, 28]. Interestingly, an opposite trend was observed in our Uox-/- mice, in which NAFLD was evidenced by hepatic steatosis and hepatocellular ballooning, suggesting hyperuricemia as a potential driving cause of NAFLD. This was supported by previous experimental studies showing UA can induce hepatic steatosis in HepG 2 cells [29].
The present model has important shortfalls. To begin, we lack direct autopsy evidence for major cause of premature death in prototype Uox-/- mice. Moreover, only 50% of allopurinol-rescued Uox-/- mice survived to 12 weeks, limiting the use of current model in the long-term studies. However, the spontaneous high levels of SUA and multiple organ injuries actually replicate the clinical patients of early-onset hyperuricemia with severe complications. Another major limitation is that we failed to exam phenotypic sex disparities due to limited sample size [9]. Confounding factors due to sex bias should be considered when interpreting these study results. Most importantly, the use of allopurinol should be titrated in a dose and time dependent manner and dynamic observations of biological properties of our modified Uox-/- mice is necessary in the future.

Conclusions

In summary, this is a novel mouse model of hyperuricemia established in two steps: knocking out of Uox by CRISPR/Cas9 system (prototypic Uox-/- mice) and allopurinol administration to improve their survivals up to 16 weeks (modified Uox-/- mice). Given the significantly elevated SUA along with urate nephropathy, cardiovascular disease and metabolic syndrome, the present modified Uox-/- mice could be suitable candidate for short and mid-term studies on early-onset hyperuricemia with severe complications. The model is useful to improve our understanding the hyperuricemia mediated cardiovascular and cardiometabolic disorders, as well as to develop more efficient novel therapies.

Acknowledgements

We would like to thank Gempharmatech Co., Ltd. (Nanjing, China) for generating the Uox knockout mice.

Declarations

Ethical Approval

All institutional and national guidelines for the care and use of laboratory animals were followed, and the experimental protocols were approved by the Animal Care and Use Committee of Zhongshan Hospital Fudan University.

Conflict of Interest

The authors declare that they have no conflict of interest.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Chiang K-M, Tsay Y-C, Vincent Ng T-C, Yang H-C, Huang Y-T, Chen C-H, et al. Is hyperuricemia, an early-onset metabolic disorder, causally associated with cardiovascular disease events in Han Chinese? J Clin Med. 2019;8:1202.CrossRefPubMedPubMedCentral Chiang K-M, Tsay Y-C, Vincent Ng T-C, Yang H-C, Huang Y-T, Chen C-H, et al. Is hyperuricemia, an early-onset metabolic disorder, causally associated with cardiovascular disease events in Han Chinese? J Clin Med. 2019;8:1202.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Zhang M, Zhu X, Wu J, Huang Z, Zhao Z, Zhang X, et al. Prevalence of hyperuricemia among Chinese adults: findings from two nationally representative cross-sectional surveys in 2015-16 and 2018-19. Front Immunol. 2021;12:791983.CrossRefPubMed Zhang M, Zhu X, Wu J, Huang Z, Zhao Z, Zhang X, et al. Prevalence of hyperuricemia among Chinese adults: findings from two nationally representative cross-sectional surveys in 2015-16 and 2018-19. Front Immunol. 2021;12:791983.CrossRefPubMed
3.
Zurück zum Zitat Borghi C, Agabiti-Rosei E, Johnson RJ, Kielstein JT, Lurbe E, Mancia G, et al. Hyperuricaemia and gout in cardiovascular, metabolic and kidney disease. Eur J Intern Med. 2020;80:1–11.CrossRefPubMed Borghi C, Agabiti-Rosei E, Johnson RJ, Kielstein JT, Lurbe E, Mancia G, et al. Hyperuricaemia and gout in cardiovascular, metabolic and kidney disease. Eur J Intern Med. 2020;80:1–11.CrossRefPubMed
4.
Zurück zum Zitat Maruhashi T, Hisatome I, Kihara Y, Higashi Y. Hyperuricemia and endothelial function: from molecular background to clinical perspectives. Atherosclerosis. 2018;278:226–31.CrossRefPubMed Maruhashi T, Hisatome I, Kihara Y, Higashi Y. Hyperuricemia and endothelial function: from molecular background to clinical perspectives. Atherosclerosis. 2018;278:226–31.CrossRefPubMed
5.
Zurück zum Zitat Yeldandi AV, Patel YD, Liao M, Kao FT, Rao MS, Reddy JK, et al. Localization of the human urate oxidase gene (UOX) to 1p22. Cytogenet Cell Genet. 1992;61:121–2.CrossRefPubMed Yeldandi AV, Patel YD, Liao M, Kao FT, Rao MS, Reddy JK, et al. Localization of the human urate oxidase gene (UOX) to 1p22. Cytogenet Cell Genet. 1992;61:121–2.CrossRefPubMed
6.
Zurück zum Zitat Wu X, Wakamiya M, Vaishnav S, Geske R, Montgomery C Jr, Jones P, et al. Hyperuricemia and urate nephropathy in urate oxidase-deficient mice. Proc Natl Acad Sci U S A. 1994;91:742–6.CrossRefPubMedPubMedCentral Wu X, Wakamiya M, Vaishnav S, Geske R, Montgomery C Jr, Jones P, et al. Hyperuricemia and urate nephropathy in urate oxidase-deficient mice. Proc Natl Acad Sci U S A. 1994;91:742–6.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Lu J, Hou X, Yuan X, Cui L, Liu Z, Li X, et al. Knockout of the urate oxidase gene provides a stable mouse model of hyperuricemia associated with metabolic disorders. Kidney Int. 2018;93:69–80.CrossRefPubMed Lu J, Hou X, Yuan X, Cui L, Liu Z, Li X, et al. Knockout of the urate oxidase gene provides a stable mouse model of hyperuricemia associated with metabolic disorders. Kidney Int. 2018;93:69–80.CrossRefPubMed
9.
Zurück zum Zitat Lu J, Dalbeth N, Yin H, Li C, Merriman TR, Wei W-H. Mouse models for human hyperuricaemia: a critical review. Nat Rev Rheumatol. 2019;15:413–26.CrossRefPubMed Lu J, Dalbeth N, Yin H, Li C, Merriman TR, Wei W-H. Mouse models for human hyperuricaemia: a critical review. Nat Rev Rheumatol. 2019;15:413–26.CrossRefPubMed
10.
Zurück zum Zitat Ichida K, Matsuo H, Takada T, Nakayama A, Murakami K, Shimizu T, et al. Decreased extra-renal urate excretion is a common cause of hyperuricemia. Nat Commun. 2012;3:764.CrossRefPubMed Ichida K, Matsuo H, Takada T, Nakayama A, Murakami K, Shimizu T, et al. Decreased extra-renal urate excretion is a common cause of hyperuricemia. Nat Commun. 2012;3:764.CrossRefPubMed
11.
Zurück zum Zitat DeBosch BJ, Kluth O, Fujiwara H, Schürmann A, Moley K. Early-onset metabolic syndrome in mice lacking the intestinal uric acid transporter SLC2A9. Nat Commun. 2014;5:4642.CrossRefPubMed DeBosch BJ, Kluth O, Fujiwara H, Schürmann A, Moley K. Early-onset metabolic syndrome in mice lacking the intestinal uric acid transporter SLC2A9. Nat Commun. 2014;5:4642.CrossRefPubMed
12.
Zurück zum Zitat Takada T, Ichida K, Matsuo H, Nakayama A, Murakami K, Yamanashi Y, et al. ABCG2 Dysfunction Increases Serum Uric Acid by Decreased Intestinal Urate Excretion. Nucleosides, Nucleotides & Nucleic Acids. 2014;33:275–81.CrossRef Takada T, Ichida K, Matsuo H, Nakayama A, Murakami K, Yamanashi Y, et al. ABCG2 Dysfunction Increases Serum Uric Acid by Decreased Intestinal Urate Excretion. Nucleosides, Nucleotides & Nucleic Acids. 2014;33:275–81.CrossRef
13.
Zurück zum Zitat Borghi C, Rosei EA, Bardin T, Dawson J, Dominiczak A, Kielstein JT, et al. Serum uric acid and the risk of cardiovascular and renal disease. J Hypertens. 2015;33:1729–41.CrossRefPubMed Borghi C, Rosei EA, Bardin T, Dawson J, Dominiczak A, Kielstein JT, et al. Serum uric acid and the risk of cardiovascular and renal disease. J Hypertens. 2015;33:1729–41.CrossRefPubMed
14.
Zurück zum Zitat Preitner F, Bonny O, Laverrière A, Rotman S, Firsov D, Da Costa A, et al. Glut9 is a major regulator of urate homeostasis and its genetic inactivation induces hyperuricosuria and urate nephropathy. Proc Natl Acad Sci U S A. 2009;106:15501–6.CrossRefPubMedPubMedCentral Preitner F, Bonny O, Laverrière A, Rotman S, Firsov D, Da Costa A, et al. Glut9 is a major regulator of urate homeostasis and its genetic inactivation induces hyperuricosuria and urate nephropathy. Proc Natl Acad Sci U S A. 2009;106:15501–6.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Bakker PJ, Butter LM, Kors L, Teske GJD, Aten J, Sutterwala FS, et al. Nlrp3 is a key modulator of diet-induced nephropathy and renal cholesterol accumulation. Kidney Int. 2014;85:1112–22.CrossRefPubMed Bakker PJ, Butter LM, Kors L, Teske GJD, Aten J, Sutterwala FS, et al. Nlrp3 is a key modulator of diet-induced nephropathy and renal cholesterol accumulation. Kidney Int. 2014;85:1112–22.CrossRefPubMed
16.
Zurück zum Zitat Grayson PC, Kim SY, LaValley M, Choi HK. Hyperuricemia and incident hypertension: a systematic review and meta-analysis. Arthritis Care Res (Hoboken). 2011;63:102–10.CrossRefPubMed Grayson PC, Kim SY, LaValley M, Choi HK. Hyperuricemia and incident hypertension: a systematic review and meta-analysis. Arthritis Care Res (Hoboken). 2011;63:102–10.CrossRefPubMed
18.
Zurück zum Zitat Kanbay M, Segal M, Afsar B, Kang D-H, Rodriguez-Iturbe B, Johnson RJ. The role of uric acid in the pathogenesis of human cardiovascular disease. Heart. 2013;99:759–66.CrossRefPubMed Kanbay M, Segal M, Afsar B, Kang D-H, Rodriguez-Iturbe B, Johnson RJ. The role of uric acid in the pathogenesis of human cardiovascular disease. Heart. 2013;99:759–66.CrossRefPubMed
20.
Zurück zum Zitat Neogi T, George J, Rekhraj S, Struthers AD, Choi H, Terkeltaub RA. Are either or both hyperuricemia and xanthine oxidase directly toxic to the vasculature? A critical appraisal. Arthritis Rheum. 2012;64:327.CrossRefPubMedPubMedCentral Neogi T, George J, Rekhraj S, Struthers AD, Choi H, Terkeltaub RA. Are either or both hyperuricemia and xanthine oxidase directly toxic to the vasculature? A critical appraisal. Arthritis Rheum. 2012;64:327.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Park J-H, Jin YM, Hwang S, Cho D-H, Kang D-H, Jo I. Uric acid attenuates nitric oxide production by decreasing the interaction between endothelial nitric oxide synthase and calmodulin in human umbilical vein endothelial cells: a mechanism for uric acid-induced cardiovascular disease development. Nitric oxide. 2013;32:36–42.CrossRefPubMed Park J-H, Jin YM, Hwang S, Cho D-H, Kang D-H, Jo I. Uric acid attenuates nitric oxide production by decreasing the interaction between endothelial nitric oxide synthase and calmodulin in human umbilical vein endothelial cells: a mechanism for uric acid-induced cardiovascular disease development. Nitric oxide. 2013;32:36–42.CrossRefPubMed
22.
Zurück zum Zitat Puddu P, Puddu GM, Cravero E, Vizioli L, Muscari A. The relationships among hyperuricemia, endothelial dysfunction, and cardiovascular diseases: molecular mechanisms and clinical implications. J Cardiol. 2012;59:235–42.CrossRefPubMed Puddu P, Puddu GM, Cravero E, Vizioli L, Muscari A. The relationships among hyperuricemia, endothelial dysfunction, and cardiovascular diseases: molecular mechanisms and clinical implications. J Cardiol. 2012;59:235–42.CrossRefPubMed
23.
Zurück zum Zitat Ponticelli C, Podestà MA, Moroni G. Hyperuricemia as a trigger of immune response in hypertension and chronic kidney disease. Kidney Int. 2020;98:1149–59.CrossRefPubMed Ponticelli C, Podestà MA, Moroni G. Hyperuricemia as a trigger of immune response in hypertension and chronic kidney disease. Kidney Int. 2020;98:1149–59.CrossRefPubMed
24.
Zurück zum Zitat Jia L, Xing J, Ding Y, Shen Y, Shi X, Ren W, et al. Hyperuricemia causes pancreatic β-cell death and dysfunction through NF-κB signaling pathway. PLoS One. 2013;8:e78284.CrossRefPubMedPubMedCentral Jia L, Xing J, Ding Y, Shen Y, Shi X, Ren W, et al. Hyperuricemia causes pancreatic β-cell death and dysfunction through NF-κB signaling pathway. PLoS One. 2013;8:e78284.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Wan X, Xu C, Lin Y, Lu C, Li D, Sang J, et al. Uric acid regulates hepatic steatosis and insulin resistance through the NLRP3 inflammasome-dependent mechanism. J Hepatol. 2016;64:925–32.CrossRefPubMed Wan X, Xu C, Lin Y, Lu C, Li D, Sang J, et al. Uric acid regulates hepatic steatosis and insulin resistance through the NLRP3 inflammasome-dependent mechanism. J Hepatol. 2016;64:925–32.CrossRefPubMed
26.
Zurück zum Zitat Bhatia LS, Curzen NP, Calder PC, Byrne CD. Non-alcoholic fatty liver disease: a new and important cardiovascular risk factor? Eur Heart J. 2012;33:1190–200.CrossRefPubMed Bhatia LS, Curzen NP, Calder PC, Byrne CD. Non-alcoholic fatty liver disease: a new and important cardiovascular risk factor? Eur Heart J. 2012;33:1190–200.CrossRefPubMed
27.
Zurück zum Zitat Xu C, Wan X, Xu L, Weng H, Yan M, Miao M, et al. Xanthine oxidase in non-alcoholic fatty liver disease and hyperuricemia: one stone hits two birds. J Hepatol. 2015;62:1412–9.CrossRefPubMed Xu C, Wan X, Xu L, Weng H, Yan M, Miao M, et al. Xanthine oxidase in non-alcoholic fatty liver disease and hyperuricemia: one stone hits two birds. J Hepatol. 2015;62:1412–9.CrossRefPubMed
28.
Zurück zum Zitat Nakatsu Y, Seno Y, Kushiyama A, Sakoda H, Fujishiro M, Katasako A, et al. The xanthine oxidase inhibitor febuxostat suppresses development of nonalcoholic steatohepatitis in a rodent model. Am J Physiol Gastrointest Liver Physiol. 2015;309:G42–51.CrossRefPubMed Nakatsu Y, Seno Y, Kushiyama A, Sakoda H, Fujishiro M, Katasako A, et al. The xanthine oxidase inhibitor febuxostat suppresses development of nonalcoholic steatohepatitis in a rodent model. Am J Physiol Gastrointest Liver Physiol. 2015;309:G42–51.CrossRefPubMed
29.
Zurück zum Zitat Lanaspa MA, Sanchez-Lozada LG, Choi Y-J, Cicerchi C, Kanbay M, Roncal-Jimenez CA, et al. Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: potential role in fructose-dependent and-independent fatty liver. J Biol Chem. 2012;287:40732–44.CrossRefPubMedPubMedCentral Lanaspa MA, Sanchez-Lozada LG, Choi Y-J, Cicerchi C, Kanbay M, Roncal-Jimenez CA, et al. Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: potential role in fructose-dependent and-independent fatty liver. J Biol Chem. 2012;287:40732–44.CrossRefPubMedPubMedCentral
Metadaten
Titel
CRISPR/Cas9 Mediated Deletion of the Uox Gene Generates a Mouse Model of Hyperuricemia with Multiple Complications
verfasst von
Linzi Zeng
Shalaimaiti Shali
Yabiao Gao
Xingchen Du
Xiaoxia Zhu
Lin Li
Yuxiang Dai
Ping Zhou
Publikationsdatum
10.06.2024
Verlag
Springer US
Erschienen in
Journal of Cardiovascular Translational Research / Ausgabe 6/2024
Print ISSN: 1937-5387
Elektronische ISSN: 1937-5395
DOI
https://doi.org/10.1007/s12265-024-10526-6

Kompaktes Leitlinien-Wissen Innere Medizin (Link öffnet in neuem Fenster)

Mit medbee Pocketcards schnell und sicher entscheiden.
Leitlinien-Wissen kostenlos und immer griffbereit auf ihrem Desktop, Handy oder Tablet.

Neu im Fachgebiet Kardiologie

Kaffeegenuss sicher bei Vorhofflimmern

Menschen mit Vorhofflimmern fürchten oft, Kaffee könnte schlecht für ihr Herz sein. Solche Ängste sind offenbar unbegründet: Zwei Schweizer Untersuchungen deuten sogar auf eine reduzierte Rate von kardiovaskulären Ereignissen unter Kaffeetrinkern.

Mit jedem Defibrillationsversuch sinkt die Überlebenschance

Wie wirkt es sich auf die Prognose aus, wenn bei Herzstillstand einmal, zweimal oder gar 29 Mal geschockt werden muss? Laut einer aktuellen Studie besteht ein deutlicher Zusammenhang zwischen der Zahl der Defibrillationsversuche und den Überlebenschancen.

Mit Spenderherz von Triathlon zu Triathlon

Ein 50-Jähriger mit Spenderherz rennt, radelt und schwimmt bis zu 14 Stunden in der Woche – beim Hawaii-Triathlon erreicht er einen Platz im Mittelfeld. Sein neues Herz kann sich an die Belastung erstaunlich gut anpassen. Selbst mit 65 Jahren ist er noch ungewöhnlich fit.

Kein Unterschied bei inadäquaten Schocks zwischen ICD-Typen

Inadäquate Schockabgaben sind ein unerwünschter Effekt der Therapie mit implantierbaren Kardioverter-Defibrillatoren. Subkutanen Geräten haftet dabei der Ruf an, dafür besonders anfällig zu sein. Die PRAETORIAN-Forschungsgruppe ist dem nachgegangen.

EKG Essentials: EKG befunden mit System (Link öffnet in neuem Fenster)

In diesem CME-Kurs können Sie Ihr Wissen zur EKG-Befundung anhand von zwölf Video-Tutorials auffrischen und 10 CME-Punkte sammeln.
Praxisnah, relevant und mit vielen Tipps & Tricks vom Profi.

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.