Skip to main content
Clinical Kidney Journal logoLink to Clinical Kidney Journal
. 2025 Jul 31;18(9):sfaf246. doi: 10.1093/ckj/sfaf246

CRISPR and gene editing for kidney diseases: where are we?

Viola D'Ambrosio 1, Chen Huimei 2, Nicole Vo 3, Keith Siew 4,5, Rhys D R Evans 6,7, Benjamin Freedman 8,9, Francesco Pesce 10,11,
PMCID: PMC12415518  PMID: 40927382

ABSTRACT

Genome editing technologies, particularly clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9, have transformed biomedical research by enabling precise genetic modifications. Due to its efficiency, cost-effectiveness and versatility, CRISPR has been widely applied across various stages of research, from fundamental biological investigations in preclinical models to potential therapeutic interventions. In nephrology, CRISPR represents a groundbreaking tool for elucidating the molecular mechanisms underlying kidney diseases and developing innovative therapeutic approaches. This review synthesizes the latest advancements in CRISPR-based gene editing within nephrology, highlighting its applications in genetic kidney disorders, polygenic nephropathies and functional genomic studies. Preclinical studies utilizing CRISPR-engineered kidney organoids and animal models have provided crucial insights into disease pathophysiology, offering platforms for drug discovery and precision medicine. Additionally, CRISPR-based functional screens have identified novel disease-associated pathways, particularly in diabetic nephropathy and glomerular disorders. Beyond experimental research, the therapeutic potential of CRISPR in nephrology is emerging, with recent advances in base editing and prime editing demonstrating the feasibility of correcting pathogenic mutations in conditions such as Alport syndrome and autosomal dominant polycystic kidney disease. Moreover, CRISPR plays a pivotal role in xenotransplantation, with gene-edited porcine kidneys addressing key immunological and virological barriers. Despite its promise, clinical translation faces challenges, including delivery efficiency, off-target effects and ethical considerations. This review provides an overview of the current state and future directions of CRISPR-based gene editing in nephrology, underscoring its transformative potential in advancing kidney disease research and therapeutics.

Keywords: CRISPR, gene editing, gene therapy, organoids, xenotransplantation

INTRODUCTION

CRISPR technology: how it started

Clustered regularly interspaced short palindromic repeats (CRISPR) were first identified in 1987 by a group of Japanese researchers as repetitive DNA sequences in Escherichia coli [1] (see Box 1 for definitions of technical terms). These DNA sequences were palindromic, meaning they could be read the same backward and forward, ≈30 base pairs (bp) long and separated by spacers that were not palindromic and ≈36 bp long [2, 3]. In 1990, Francisco Mojica, identified the same palindromic repeats in Archaea, discovered that the spacers matched viral DNA and hypothesized the role of these spacers in bacterial immunity against viral infections [4]. Some years later, in 2007, Philippe Horvath proved that CRISPR was a sort of adaptive immune response against viruses [5]. Essentially, it was discovered that CRISPR sequences play a crucial role in microbial adaptive immunity, enabling microbes to recognize and eliminate specific foreign DNA sequences from invading viruses [6, 7]. A gene adjacent to the palindromic repeats is responsible for the cutting. This gene encodes for the Cas9 protein, an endonuclease responsible for cutting viral DNA sequences and protecting the bacteria from infection [8]. Building on these foundational discoveries, Jennifer Doudna and Emmanuelle Charpentier introduced the fully resolved CRISPR-Cas9 system in 2012 [9] by identifying the missing component of this complex system: transactivating CRISPR-RNA (tracrRNA), the RNA sequence that guides the CRISPR system [10]. They later synthesized a single guide RNA sequence (sgRNA) [11] to substitute for tracrRNA. With the complete system composed of palindromic repeats, spacers, sequence encoding for Cas9 and sgRNA, the CRISPR-Cas9 system was later translated into mammalian cells for gene-editing purposes. Gene editing is a process that involves modifying a cell's DNA sequence and its genetic information.

The advent of CRISPR-Cas9 ushered in a transformative era in gene editing [1]. This groundbreaking tool precisely targets DNA sequences with the Cas9 protein, which induces double-stranded DNA breaks, thereby enabling efficient and targeted genomic modifications [12]. Unlike earlier gene-editing technologies such as meganucleases, zinc-finger nucleases and transcription activator-like effector nucleases—which required labour-intensive and costly protein engineering for each target sequence—CRISPR's RNA-guided approach dramatically simplifies the process [13]. By merely altering the sgRNA sequence, researchers can efficiently direct Cas9 to almost any genomic target, akin to programming a Global Positioning System to navigate to a desired destination [13].

CRISPR technology: advantages

Due to its unparalleled precision, efficiency and cost-effectiveness, CRISPR has rapidly become an indispensable tool in biomedical research [1]. Its applications span from preclinical studies involving cell and animal models to its emerging therapeutic potential in clinical practice [14]. When CRISPR induces double-stranded DNA breaks, cells utilize two main pathways for repair: non-homologous end joining (NHEJ) and homologous recombination (HR) [15]. NHEJ, although faster and more prevalent, is error prone and can result in insertions or deletions at the repair site [15]. Conversely, HR is a more precise mechanism, leveraging a DNA template—either naturally present or externally supplied—for accurate repair [15]. HR facilitates the correction of point mutations, deletions and other genetic alterations underlying many diseases. To further enhance its versatility, various CRISPR variants, such as CasMINI and Cas14, have been developed [16–18]. These innovations extend CRISPR's capabilities to include small-target editing and compact delivery systems, broadening the range of applications across diverse fields of research and therapy.

CRISPR-based gene-editing therapies are now transitioning from concept to clinical reality. In 2019, the U.S. Food and Drug Administration (FDA) approved a CRISPR-based cell therapy for sickle cell disease and beta-thalassemia, marking a significant milestone in the field [19–21]. These therapies primarily function by inducing targeted gene disruption through the NHEJ repair mechanism ex vivo, prior to cell administration. In the future, as the technology matures, CRISPR may expand its therapeutic reach to a broader spectrum of diseases, including immune disorders and rare genetic conditions.

In the field of nephrology, CRISPR presents unprecedented opportunities for advancing kidney disease research. By enabling precise genomic modifications, CRISPR offers a powerful tool to elucidate the molecular mechanisms underlying kidney diseases and to develop innovative therapeutic strategies. This review synthesizes the latest evidence on the application of CRISPR-Cas and related gene-editing technologies in nephrology, emphasizing their transformative potential in advancing the diagnosis and treatment of kidney diseases (Fig. 1).

Figure 1:

Figure 1:

Schematic diagram of CRISPR gene editing for preclinical and clinical application.

GENE-EDITING APPLICABILITY IN NEPHROLOGY

Therapeutic potential in kidney diseases

Chronic kidney disease (CKD) represents a significant global health challenge, ranking as the ninth leading cause of mortality according to the World Health Organization (WHO) in 2021 [22]. Affecting more than 10% of the global population, the prevalence of CKD is anticipated to increase due to increasing life expectancy and the growing prevalence of chronic comorbidities such as diabetes and hypertension. Kidney diseases span a diverse range of genetic, polygenic and acquired conditions, many of which are associated with well-characterized molecular pathways and genetic alterations [23].

Monogenic kidney diseases

Congenital nephrotic syndrome (CNS) is an example of a group of genetic kidney disorders, presenting early in life with symptoms including massive proteinuria, hypoalbuminaemia, severe oedema and hyperlipidaemia [24]. At least three key pathogenic genes are implicated in CNS: NPHS1 (encoding nephrin), NPHS2 (encoding podocin) and an unidentified gene located in the 11q21-22 region [25]. Furthermore, monogenic kidney disorders such as autosomal dominant polycystic kidney disease (ADPKD), Alport syndrome and nephronophthisis are caused by mutations in genes including PKD1, PKD2, COL4A and NPHP3. Advances in genome-wide association studies (GWAS) and genetic analyses have identified >600 nephropathy-associated genes, providing an invaluable resource for understanding the genetic contributors to kidney diseases [26]. CRISPR-mediated gene editing provides the potential for precise correction of such mutations, offering a promising approach to halt or reverse disease progression.

Polygenic/multifactorial kidney diseases

While diabetes and hypertension remain the most reported causes of CKD, recent sequencing technologies have revealed its polygenic and multifactorial nature [27, 28]. Polygenic nephropathy refers to genetic predispositions influenced by multiple loci and interacting pathways. Key genes such as UMOD (encoding uromodulin), APOL1 (encoding apolipoprotein L1) and HNF1B (encoding hepatocyte nuclear factor 1 beta) have been identified as significant contributors to kidney disease susceptibility [29]. Variants in APOL1, for instance, are strongly linked to an increased risk of focal segmental glomerulosclerosis (FSGS) and hypertensive nephropathy, particularly in individuals of African ancestry [30]. APOL1 risk alleles could conceivably be disrupted using CRISPR-Cas9, as a means of genome-editing therapy. Demonstrating direct links between genetic mutations and their phenotypes requires robust functional experimental studies. CRISPR-Cas systems provide a highly effective platform for such studies, enabling precise gene modifications to investigate molecular pathways underlying kidney disease pathogenesis. This approach not only advances our understanding of disease mechanisms but also identifies potential therapeutic targets, particularly for conditions characterized by inflammation, fibrosis and metabolic dysregulation.

Diagnostic and screening potential in kidney diseases

Beyond the treatment of genetic mutations, CRISPR technology plays a role in gene screening and pathway identification. CRISPR-based functional screens have been instrumental in identifying possible regulatory networks in diabetic nephropathy, providing insights into how hyperglycaemia exacerbates kidney damage. These screens can pinpoint novel mediators of kidney injury, paving the way for the development of new therapeutic interventions. Furthermore, the nephron, comprising specialized cell types such as podocytes, tubular epithelial cells and mesangial cells, highlights the complexity of kidney function and disease. Precision tools like CRISPR are essential for studying the contributions of individual cell types to disease phenotypes. When combined with CRISPR, kidney organoids derived from human induced pluripotent stem cells (PSCs) offer a powerful platform for modelling disease-specific mutations and phenotypes in physiologically relevant contexts. These organoids enable the study of complex kidney diseases, facilitating the identification of molecular mechanisms and the evaluation of potential therapeutic strategies.

Given the multifactorial and multicellular involvement in kidney diseases, alongside their well-defined genetic background, CRISPR and gene-editing technologies present tremendous potential in nephrology. By addressing the genetic and molecular complexities of kidney diseases, CRISPR not only enhances our understanding of pathophysiology but also provides a robust framework for therapeutic development. Its ability to model, investigate and potentially correct disease-causing mutations highlights its transformative role in advancing kidney research and improving clinical outcomes.

PRECLINICAL APPLICATION OF CRISPR-CAS

Functional studies and gene-edited cell lines

GWASs have identified several genes that are associated with kidney diseases. However, in order to validate this association, experimental functional studies are required to prove that the mutation actually causes the disease and how it does so at a molecular level. CRISPR-Cas technology can answer these questions. For instance, it can be used to experimentally knock out or introduce mutations into candidate genes and then compare the phenotype to wild-type controls. Mandai et al. [31] created a knock-in human embryonic kidney cell line with an intronic polymorphism in the STK39 gene that encodes for STE20/SPS-1-related proline/alanine-rich kinase (SPAK) via CRISPR-Cas. This intronic polymorphism was identified through GWASs as one of the susceptibility genes for hypertension, and this association was also confirmed by a meta-analysis, although the biological function of the polymorphism was not determined. Mandai et al. thus demonstrated that by introducing this polymorphism, cells had higher STK39 mRNA and SPAK expression and phosphorylation, leading to an increase in the sodium–potassium–chloride cotransporter (NKCC1) that is mainly expressed in the afferent arteriole's smooth muscle cells and in the outer and inner medullary collecting duct [31, 32]. CRISPR-Cas thus allowed identification of the biochemical function of a disease-associated polymorphism.

Again, Porath et al. [33] generated a GANAB-mutated cell line using CRISPR-Cas9 to prove GANAB gene product involvement in ADPKD. GANAB is a gene that encodes for glucosidase II subunit α (GIIα) and was found to be associated with ADPKD thanks to whole-exome sequencing of unresolved ADPKD cases (ADPKD cases negative for mutations in PKD1 and PKD2) identifying a missense mutation in the gene. Using CRISPR-Cas9 technology, Porath et al. [33] proved that GIIα is necessary for the correct maturation and localization of polycystin 1 and 2 (PC1 and PC2) and therefore contributed to the genetic understanding of ADPKD.

Gene-edited organoids

Organoids are heterocellular structures that mimic bodily tissues and organs to study biology and disease. Human kidney organoids can be derived from induced PSCs. The hallmark of these organoids is the appearance of podocytes, proximal tubules and distal tubules in contiguous segments. In current practice, induced PSCs are typically edited in the undifferentiated state to generate stable cell lines, which are subsequently differentiated into organoids to study the effects of mutations [34, 35].

CRISPR-Cas technology has been instrumental in exploring gene functions and modelling kidney diseases using in vitro kidney organoids [34, 36] (Table 1). Kidney organoids derived from human iPS cells have been used to create genetic disease models. For polycystic kidney disease, introducing loss-of-function mutations in PKD1 and PKD2 via CRISPR-Cas9 produced cysts from kidney tubules upon differentiation, indicating that the pathognomonic disease phenotype could be recapitulated in a simplified system in vitro [34]. Additional studies identified a critical role for microenvironmental factors in cystogenesis as well as glucose absorption in PKD organoid cystogenesis [36, 37]. Cystogenesis could also be introduced via a cytosine base editor to create nonsense mutations [38]. Tubuloids derived from adult kidney also exhibit a cystic phenotype after CRISPR editing to disrupt PKD1 or PKD2 [39].

Table 1:

List of genes that were modified using CRISPR gene editing in preclinical kidney organoid models of disease.

Kidney disease Target gene Target cell type Technical method Function/phenotype References
Polycystic kidney PKD1 or PKD2 Tubule CRISPR-Cas9 or cytosine base editor knock-out ↑ cystogenesis,
↓ polycystin-1 levels,
↓ ciliary polycystin-2,
↓ ECM compaction
[34, 36, 84, 38]
PKHD1 Tubule CRISPR-Cas9 knock-out ↑ cystogenesis [85]
Nephrotic syndrome PODXL Podocyte CRISPR-Cas9 knock-out ↓ junctional migration,
↓ cell–cell spacing,
↓ microvillus formation
[34, 40]
NPHS1 Podocyte CRISPR-Cas9 knock-in ↓ slit diaphragm protein expression/localization [86]
FSGS APOL1 Podocyte, endothelial CRISPR-Cas9 knock-in Dedifferentiation of podocytes, loss of endothelial cells [42]
Ciliopathy KIF3A or KIF3B Tubule CRISPR-Cas9 knock-out ↓ nephrogenesis, ↑ cystogenesis [87]
Fabry syndrome GLA Podocyte CRISPR-Cas9 knock-out ↓ lysosomal aspartylglucosaminidase enzyme activity [88–90]
Alport syndrome COL4A3 or COL4A5 Podocyte CRISPR-Cas9 knock-in, variant correction Reserve podocyte number [50]
COL4A5 Podocyte CRISPR-Cas9 knock-out Parietal cell hyperplasia, mesangial sclerosis and interstitial fibrosis [91]

Additionally, CRISPR-Cas has been used to investigate phenotypes related to podocyte dysfunction and glomerular disease. Knockout of PODXL, encoding podocalyxin, a key protein in podocyte junction organization, resulted in apical diffusion of slit diaphragm proteins within podocyte clusters in organoids [34, 40]. Findings in kidney organoids were validated in mouse models, where podocalyxin-deficient mice succumbed to kidney failure postnatally, aligning with clinical cases of congenital nephrotic syndrome caused by PODXL mutations. One interesting gene target for CRISPR is APOL1, for which human-specific risk variants exist that predispose individuals for glomerular disease [41]. It has been shown that APOL1 can be induced in kidney organoids by treatment with interferon-gamma and is associated with changes in gene expression consistent with dedifferentiation of kidney podocytes, as well as the loss of endothelial cells via pyroptosis [42, 43]. Future work may utilize such systems to develop CRISPR treatments that disrupt APOL1 risk alleles, thus protecting the kidneys from deleterious effects.

Gene-edited animals

Animal models of human diseases are fundamental to advance research and to investigate the underlying pathophysiology of several diseases. Unfortunately, there aren't many animals that mimic human anatomy, physiology and pathology accurately. Currently the two best animal models are mice and rats, with rats being physiologically more similar to humans but more difficult to manipulate genetically than mice.

Besides cell cultures, CRISPR-Cas can be used to create gene-edited animals, not only for a single gene but for multiple genes simultaneously [44]. For example, Xu et al. [45] created a mouse model using CRISPR-Cas to tag the odd-skipped related 1 (Osr1) protein, a protein involved in the early stages of nephrogenesis. Heterozygous mutations are associated with decreased kidney size and function [46], whereas homozygous mutations lead to lethal congenital kidney malformations [47]. Tagging Osr1 through CRISPR-Cas clarified the biological function of Osr1 in the early stage of kidney development [45].

The pathogenic role of APOL1, a gene associated with FSGS in African Americans, was clarified in CRISPR-Cas9-edited zebrafish embryos [48]. Similarly, the link between Sec61 translocon alpha 1 subunit (SEC6A1A) and autosomal dominant tubulointerstitial kidney disease was confirmed in CRISPR-Cas9-edited zebrafish [49].

CLINICAL APPLICATION OF CRISPR-CAS

Potential gene-editing therapy for genetic kidney diseases

Another exciting potential application of CRISPR-Cas and gene-editing technology is the treatment of genetic diseases. These modifications can be done in somatic cells or in germinal cells, the latter ensuring that the modification can be inherited. As a therapeutic approach, it has been reported in preclinical studies for specific genetic kidney diseases such as Alport syndrome. The study conducted by Daga et al. [50] demonstrated a 40% reversion of variants with a small percentage of off-target indels in urine-derived human podocyte lineage–carrying mutations in the X-linked COL4A5 and autosomal COL4A3 gene.

In the previously mentioned study by Vishy et al. [38], the cytosine base editor–related cystogenesis in ADPKD organoids was fully reversed when homozygous mutants were converted to heterozygotes using an adenine base editor, suggesting that partial restoration of gene function may be sufficient to prevent disease progression. Recently, another tool for genetic editing, called adenine base editor (ABE), was successfully used for ADPKD [51] and primary hyperoxaluria type 1 [52]. Wang et al. [51] reported the successful use of ABE to correct PKD1 mutations in human induced PSCs from peripheral blood mononuclear cells (PBMCs) of a heterozygous ADPKD patient. Kidney organoids developed from the corrected cells did not develop cysts upon treatment with forskolin. Lastly, Chen et al. [52] delivered specific ABEs through AAVs in vivo in a mouse model of primary hyperoxaluria type 1, restoring the production of hepatic alanine glyoxylate aminotransferase.

Besides the treatment of genetic diseases, Jerke et al. [53] demonstrated the applicability of CRISPR-Cas9 for autoimmune diseases such as anti-neutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis. Interestingly, they showed that introducing a mutation in the gene encoding for proteinase 3 (one of the two main targets of autoimmunity in ANCA-associated vasculitis) in human CD34+ haematopoietic stem and progenitor cells, selectively reduced the abundance and activity of the protein. This is a potential breakthrough for ANCA-associated vasculitis and a promising avenue for immunosuppressant-free tolerance induction in autoimmune diseases.

The latest breakthrough involves using CRISPR technology to potentially treat diseases caused by enzymatic deficiencies. A recent article by Musunuru et al. [54] showed promising results for a customized base-editing therapy in a neonate affected by a rare metabolic disorder (carbamoyl-phosphate synthetase 1 deficiency). This opens a new avenue for CRISPR in metabolic nephropathies caused by enzymatic deficiencies such as adenine phosphoribosyltransferase enzyme deficiency, primary hyperoxalurias, Fabry disease or glycogen storage diseases.

Xenotransplantation

Kidney transplantation is the best therapeutic option for most patients with end-stage kidney disease (ESKD). Since the first successful human kidney transplantation [55] performed by Murray in 1954 in Boston, Massachusetts, USA, tremendous improvements have been made from the surgical, immunological and nephrological perspectives to ensure better short- and long-term outcomes, survival and quality of life of kidney transplant recipients [56]. Despite the increasing global incidence of kidney transplantation [57, 58] and the ever-growing transplantation of expanded criteria donor organs [59, 60], demand still exceeds supply. This has forced the medical, surgical and scientific community to look for an alternative, such as xenotransplantation (transplantation of living cells, tissues or organs from one species to another). The domestic pig (Sus scrofa f. domestica) presents a potential source of non-human tissue because it is easy to breed, its litter size is relatively large and its organs are anatomically similar to those of humans [61, 62]. Unsurprisingly, this has raised several immunological, xenozoonosis-related, physiological and ethical concerns. CRISPR technology could serve as an exciting and promising tool to bridge some of these gaps. To obviate the immunological barriers, CRISPR and gene editing can be used to reduce the immunogenicity of porcine kidneys and therefore modulate the recipient's immune response. As previously mentioned, one of the biggest limitations of xenotransplantation is the hyperimmunogenicity of xenografts. This is partly due to the presence of antigens on porcine cells that cause a violent immunological response known as hyperacute rejection (HAR) in the recipient [63, 64]. HAR is a type of humoral rejection mediated by pre-existing recipient antibodies that recognize epitopes expressed on xenograft endothelial cells [65]. This leads to complement activation and destruction of xenograft vascular integrity, with its immediate failure [66]. It normally happens minutes to hours after xenotransplantation. The antigens involved in xenograft HAR include galactose-α1,3-galactose (α-Gal), an epitope synthesized by the α-1,3-galactosyltransferase (GGTA1) enzyme encoded by the porcine GGTA1 gene [67]; cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH), encoded by the porcine CMAH gene [68]; and β-1,4-N-acetylgalactosaminyltransferase 2 (β4GalNT2) encoded by the porcine β4GalNT2 gene [69]. CRISPR gene editing has been used to engineer knock-out (KO) pigs for the three surface antigens (GGTA1, CMAH and β4GalNT2) that are involved in HAR, and this has shown decreased immunoreactivity of human antibodies (IgM and IgG) towards KO PBMCs [70]. Gene-editing technology has also been used to reduce the risk of acute humoral rejection (AHR) and acute cellular rejection (ACR). For AHR, gene-editing technology has been used to modify the class I swine major histocompatibility complex (SLA) [71], reduce the toxicity of natural killer cells [72], inhibit complement activity and inhibit macrophage phagocytosis [63]. For ACR, CRISPR-Cas technology has been used to reduce T-lymphocyte activity [73] (although this approach was unsuccessful) and to reduce the expression of both class I and class II [74] SLA molecules. However, there is a need for further preclinical studies.

Another avenue worth exploring to reduce the hyperimmunogenicity of xenografts is the use of CRISPR to harvest human kidneys in pigs. More specifically, this could be possible thanks to a technology called interspecies blastocyst complementation that consists of the injection of PSCs of a species (human, for example) into the blastocyst of another species. This has been previously done in rats and mice for pancreas, heart and eyes, but could be expanded to kidneys [75].

Besides immunogenicity, another problem for pig xenotransplantation is the risk of xenozoonosis, which is the transmission of infectious agents between two species via a xenograft. Porcine endogenous retroviruses are viruses that are integrated into the pig genome and that should be eliminated before xenotransplantation. This could be done via CRISPR [76].

Significant advances have been made over recent years in the clinical use of gene-edited pig kidneys in human recipients, when initial xenotransplants were undertaken in brain dead donors [77–80]. Within the last year, the first genetically edited pig kidney was transplanted into a live human recipient [81, 82] at the Massachusetts General Hospital, Boston, MA, USA. The recipient was a 62-year-old man with ESKD undergoing haemodialysis after previous graft loss with a medical history of type 2 diabetes and hypertension. The recipient had primary graft function, experienced early T cell–mediated rejection, which was treated with augmented immunosuppression, but died with a functioning graft at day 52 post-transplant from a cardiac event.

Future opportunities and challenges

While CRISPR technology offers promising avenues for the treatment of kidney diseases, its clinical translation faces several significant challenges spanning delivery systems, technical limitations, safety, ethics and regulatory frameworks. The structural and functional complexity of the kidney presents unique obstacles for effective delivery systems. Urine-directed delivery leverages the accessibility of the urinary system, with mechanisms such as glomerular filtration aiding in the targeted localization of CRISPR components to kidney cells. This method shows potential for targeting tubular epithelial cells by delivering components directly through the urinary tract. Tubular-specific delivery methods employ molecular carriers or nanoparticles engineered to selectively bind to receptors expressed on tubular cells. Strategies such as ligand-conjugated nanoparticles and kidney-specific promoters are actively being explored to improve delivery efficiency and specificity. While promising, these methods remain in the experimental phase and require further refinement to ensure safety and feasibility in clinical applications.

Similar to other complex biological systems, the diverse cellular architecture of the kidney complicates precise gene editing. Efficiently targeting specific cell populations, addressing repair pathway biases (which favour non-homologous end joining over homologous recombination) and minimizing off-target effects are critical areas requiring optimization. Additionally, maintaining the stability of CRISPR components during systemic delivery and ensuring consistent therapeutic outcomes are key challenges that must be addressed to facilitate the transition from preclinical success to clinical application (Fig. 2).

Figure 2:

Figure 2:

CRISPR gene editing range of possible opportunities and challenges in kidney diseases.

CRISPR technology also raises significant concerns in the domains of safety, ethics and regulation. Unintended off-target effects could lead to harmful genetic modifications, while CRISPR-induced cellular stress may impair cell viability or hinder editing success. Some cells may recover from editing but others may undergo apoptosis or senescence, limiting therapeutic efficacy. Ethical concerns include the potential for CRISPR to be used for non-therapeutic human enhancement, such as improving physical or cognitive traits, which could exacerbate social inequities. Germline editing introduces additional risks, such as permanent genetic changes that could affect future generations, raising complex ethical questions. In 2018, an international group of scientists called for a moratorium on germline editing research, underscoring the need for a globally accepted ethical framework. The Somatic Cell Genome Editing Consortium of the National Institutes of Health was also founded in 2018, to improve the technology and assess the biological effects of genome editing [83]. In 2020, Doudna and Charpentier were awarded the Nobel Prize in Chemistry for the invention of CRISPR gene editing. With so much attention being paid to gene editing around the globe, this technology is sure to produce many significant advances in the years to come, including novel gene therapies for kidney disorders.

CONCLUSIONS

In summary, CRISPR-Cas technology is a revolutionary tool for research with a high potential for clinical translation. In nephrology, CRISPR-Cas has mainly been used for preclinical models, whether they be cell lines, gene-edited organoids or animals. These preclinical models shed some light onto the pathophysiology of certain diseases, such as monogenic and polygenic kidney disorders, and helped us understand the intracellular pathways involved. However, during the last decade the clinical application of this technology has expanded. From gene-editing therapies for genetic kidney diseases to induction of tolerance in autoimmune diseases and xenotransplantation, CRISPR technology is already revolutionizing the field of nephrology.

Contributor Information

Viola D'Ambrosio, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy.

Chen Huimei, Centre for Computational Biology (CCB) and Programme in Cardiovascular and Metabolic Disorders (CVMD), Duke-NUS Medical School, Singapore.

Nicole Vo, Department of Medicine, Division of Nephrology, Institute for Stem Cell and Regenerative Medicine  and Kidney Research Institute, University of Washington School of Medicine, Seattle, WA, USA.

Keith Siew, London Tubular Centre, Department of Renal Medicine, University College London, London, UK; Centre for Kidney and Bladder Health, University College London, London, UK.

Rhys D R Evans, London Tubular Centre, Department of Renal Medicine, University College London, London, UK; Centre for Kidney and Bladder Health, University College London, London, UK.

Benjamin Freedman, Department of Medicine, Division of Nephrology, Institute for Stem Cell and Regenerative Medicine  and Kidney Research Institute, University of Washington School of Medicine, Seattle, WA, USA; Plurexa LLC, Seattle, WA, USA.

Francesco Pesce, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Division of Renal Medicine, Ospedale Isola Tiberina-Gemelli Isola, Rome, Italy.

FUNDING

This study is supported by the Ministry of Education of Singapore for AcRF Tier 2 funding (T2EP30221-0013), by the National Institutes of Health (U01AI176460, U2CTR004867) and the Lara Nowak-Macklin Research Fund (to B.S.F.).

AUTHORS’ CONTRIBUTIONS

VDA and FP conceptualized the manuscript. VDA and CH prepared the initial draft. BF supervised the manuscript. All authors contributed to reviewing and editinging.

DATA AVAILABILITY STATEMENT

No new data were generated or analysed in support of this research.

CONFLICT OF INTEREST STATEMENT

VDA received consultancy fees from Allena Pharmaceuticals. FP received honoraria from AstraZeneca, GKS and Otsuka. BSF received support for the present manuscript from NIH. BSF received grants of contracts from Cystinosis Research Foundation, royalties or licences from STEMCELL Technologies and received consulting fees from Plurexa LLC. BSF holds ownership interests in Plurexa and patent from Partners Healthcare. KS, RE, NV and CH have no conflict of interest.

REFERENCES

  • 1. Pacesa  M, Pelea  O, Jinek  M. Past, present, and future of CRISPR genome editing technologies. Cell  2024;187:1076–100. 10.1016/j.cell.2024.01.042 [DOI] [PubMed] [Google Scholar]
  • 2. Isaev  AB, Musharova  OS, Severinov  KV. Microbial arsenal of antiviral defenses – part I. Biochemistry (Moscow)  2021;86:319–37. 10.1134/S0006297921030081 [DOI] [PubMed] [Google Scholar]
  • 3. Isaev  AB, Musharova  OS, Severinov  KV. Microbial arsenal of antiviral defenses. Part II. Biochemistry (Moscow)  2021;86:449–70. 10.1134/S0006297921040064 [DOI] [PubMed] [Google Scholar]
  • 4. Mojica  FJ, Juez  G, Rodríguez-Valera  F. Transcription at different salinities of Haloferax mediterranei sequences adjacent to partially modified PstI sites. Mol Microbiol  1993;9:613–21. 10.1111/j.1365-2958.1993.tb01721.x [DOI] [PubMed] [Google Scholar]
  • 5. Barrangou  R, Fremaux  C, Deveau  H  et al.  CRISPR provides acquired resistance against viruses in prokaryotes. Science  2007;315:1709–12. 10.1126/science.1138140 [DOI] [PubMed] [Google Scholar]
  • 6. Terns  RM, Terns  MP. CRISPR-based technologies: prokaryotic defense weapons repurposed. Trends Genet  2014;30:111–8. 10.1016/j.tig.2014.01.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Gasiunas  G, Barrangou  R, Horvath  P  et al.  Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc Natl Acad Sci USA  2012;109:E2579–86. 10.1073/pnas.1208507109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Bolotin  A, Quinquis  B, Sorokin  A  et al.  Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology  2005;151:2551–61. 10.1099/mic.0.28048-0 [DOI] [PubMed] [Google Scholar]
  • 9. Ledford  H, Callaway  E. Pioneers of revolutionary CRISPR gene editing win chemistry Nobel. Nature  2020;586:346–7. 10.1038/d41586-020-02765-9 [DOI] [PubMed] [Google Scholar]
  • 10. Deltcheva  E, Chylinski  K, Sharma  CM  et al.  CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature  2011;471:602–7. 10.1038/nature09886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Jinek  M, Chylinski  K, Fonfara  I  et al.  A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science  2012;337:816–21. 10.1126/science.1225829 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Manghwar  H, Li  B, Ding  X  et al.  CRISPR/Cas systems in genome editing: methodologies and tools for sgRNA design, off-target evaluation, and strategies to mitigate off-target effects. Adv Sci  2020;7:1902312. 10.1002/advs.201902312 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Gaj  T, Gersbach  CA, Barbas  CF. ZFN, TALEN and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol  2013;31:397–405. 10.1016/j.tibtech.2013.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Laurent  M, Geoffroy  M, Pavani  G  et al.  CRISPR-based gene therapies: from preclinical to clinical treatments. Cells  2024;13:800. 10.3390/cells13100800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Su  T, Liu  F, Gu  P  et al.  A CRISPR-Cas9 assisted non-homologous end-joining strategy for one-step engineering of bacterial genome. Sci Rep  2016;6:37895. 10.1038/srep37895 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Xu  X, Chemparathy  A, Zeng  L  et al.  Engineered miniature CRISPR-Cas system for mammalian genome regulation and editing. Mol Cell  2021;81:4333–4345.e4. 10.1016/j.molcel.2021.08.008 [DOI] [PubMed] [Google Scholar]
  • 17. Komor  AC, Kim  YB, Packer  MS  et al.  Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature  2016;533:420–4. 10.1038/nature17946 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Anzalone  AV, Randolph  PB, Davis  JR  et al.  Search-and-replace genome editing without double-strand breaks or donor DNA. Nature  2019;576:149–57. 10.1038/s41586-019-1711-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Parums  DV. Editorial: First regulatory approvals for CRISPR-Cas9 therapeutic gene editing for sickle cell disease and transfusion-dependent β-thalassemia. Med Sci Monit  2024;30:e944204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Frangoul  H, Altshuler  D, Cappellini  MD  et al.  CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. N Engl J Med  2021;384:252–60. 10.1056/NEJMoa2031054 [DOI] [PubMed] [Google Scholar]
  • 21. Sharma  A, Boelens  J-J, Cancio  M  et al.  CRISPR-Cas9 editing of the HBG1 and HBG2 promoters to treat sickle cell disease. N Engl J Med  2023;389:820–32. 10.1056/NEJMoa2215643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Kovesdy  CP. Epidemiology of chronic kidney disease: an update 2022. Kidney Int Suppl  2022;12:7–11. 10.1016/j.kisu.2021.11.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Groopman  EE, Marasa  M, Cameron-Christie  S  et al.  Diagnostic utility of exome sequencing for kidney disease. N Engl J Med  2019;380:142–51. 10.1056/NEJMoa1806891 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Boyer  O, Schaefer  F, Haffner  D  et al.  Management of congenital nephrotic syndrome: consensus recommendations of the ERKNet-ESPN Working Group. Nat Rev Nephrol  2021;17:277–89. 10.1038/s41581-020-00384-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Koziell  A, Grech  V, Hussain  S  et al.  Genotype/phenotype correlations of NPHS1 and NPHS2 mutations in nephrotic syndrome advocate a functional inter-relationship in glomerular filtration. Hum Mol Genet  2002;11:379–88. 10.1093/hmg/11.4.379 [DOI] [PubMed] [Google Scholar]
  • 26. Wuttke  M, Köttgen  A. Insights into kidney diseases from genome-wide association studies. Nat Rev Nephrol  2016;12:549–62. 10.1038/nrneph.2016.107 [DOI] [PubMed] [Google Scholar]
  • 27. Ercegović  V, Džimbeg  M, Gelemanović  A. Genetic susceptibility of type 2 diabetes and metabolic syndrome. Diabetology  2025;6:11. 10.3390/diabetology6020011 [DOI] [Google Scholar]
  • 28. Tremblay  J, Hamet  P. Environmental and genetic contributions to diabetes. Metabolism  2019;100:153952. 10.1016/j.metabol.2019.153952 [DOI] [PubMed] [Google Scholar]
  • 29. Sawaf  H, Gudura  TT, Dorobisz  S  et al.  Genetic susceptibility to chronic kidney disease: links, risks and management. Int J Nephrol Renovasc Dis  2023;16:1–15. 10.2147/IJNRD.S363041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kallash  M, Wang  Y, Smith  A  et al.  Rapid progression of focal segmental glomerulosclerosis in patients with high-risk APOL1 genotypes. Clin J Am Soc Nephrol  2023;18:344–55. 10.2215/CJN.0000000000000069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Mandai  S, Mori  T, Sohara  E  et al.  Generation of hypertension-associated STK39 polymorphism knockin cell lines with the clustered regularly interspaced short palindromic repeats/Cas9 system. Hypertension  2015;66:1199–206. 10.1161/HYPERTENSIONAHA.115.05872 [DOI] [PubMed] [Google Scholar]
  • 32. Koumangoye  R, Bastarache  L, Delpire  E. NKCC1: newly found as a human disease-causing ion transporter. Function  2020;2:zqaa028. 10.1093/function/zqaa028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Porath  B, Gainullin  VG, Cornec-Le Gall  E  et al.  Mutations in GANAB, encoding the glucosidase IIα subunit, cause autosomal-dominant polycystic kidney and liver disease. Am Hum Genet  2016;98:1193–207. 10.1016/j.ajhg.2016.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Freedman  BS, Brooks  CR, Lam  AQ  et al.  Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat Commun  2015;6:8715. 10.1038/ncomms9715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Peters  DT, Cowan  CA, Musunuru  K. Genome editing in human pluripotent stem cells. In: StemBook. Cambridge, MA: Harvard Stem Cell Institute, 2008. http://www.ncbi.nlm.nih.gov/books/NBK143767/  (8 March 2025, date last accessed). [PubMed] [Google Scholar]
  • 36. Cruz  NM, Song  X, Czerniecki  SM  et al.  Organoid cystogenesis reveals a critical role of microenvironment in human polycystic kidney disease. Nat Mater  2017;16:1112–9. 10.1038/nmat4994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Li  SR, Gulieva  RE, Helms  L  et al.  Glucose absorption drives cystogenesis in a human organoid-on-chip model of polycystic kidney disease. Nat Commun  2022;13:7918. 10.1038/s41467-022-35537-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Vishy  CE, Thomas  C, Vincent  T  et al.  Genetics of cystogenesis in base-edited human organoids reveal therapeutic strategies for polycystic kidney disease. Cell Stem Cell  2024;31:537–53.e5. 10.1016/j.stem.2024.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Xu  Y, Kuppe  C, Perales-Patón  J  et al.  Adult human kidney organoids originate from CD24+ cells and represent an advanced model for adult polycystic kidney disease. Nat Genet  2022;54:1690–701. 10.1038/s41588-022-01202-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Kim  YK, Refaeli  I, Brooks  CR  et al.  Gene-edited human kidney organoids reveal mechanisms of disease in podocyte development. Stem Cells  2017;35:2366–78. 10.1002/stem.2707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Gbadegesin  RA, Ulasi  I, Ajayi  S  et al.  APOL1 bi- and monoallelic variants and chronic kidney disease in West Africans. N Engl J Med  2025;392:228–38. 10.1056/NEJMoa2404211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Liu  E, Radmanesh  B, Chung  BH  et al.  Profiling APOL1 nephropathy risk variants in genome-edited kidney organoids with single-cell transcriptomics. Kidney360  2020;1:203–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Juliar  BA, Stanaway  IB, Sano  F  et al.  Interferon-γ induces combined pyroptotic angiopathy and APOL1 expression in human kidney disease. Cell Rep  2024;43:114310. 10.1016/j.celrep.2024.114310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Wang  H, Yang  H, Shivalila  CS  et al.  One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell  2013;153:910–8. 10.1016/j.cell.2013.04.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Xu  J, Liu  H, Chai  OH  et al.  Osr1 interacts synergistically with Wt1 to regulate kidney organogenesis. PLoS One  2016;11:e0159597. 10.1371/journal.pone.0159597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Zhang  Z, Iglesias  D, Eliopoulos  N  et al.  A variant OSR1 allele which disturbs OSR1 mRNA expression in renal progenitor cells is associated with reduction of newborn kidney size and function. Hum Mol Genet  2011;20:4167–74. 10.1093/hmg/ddr341 [DOI] [PubMed] [Google Scholar]
  • 47. Lozić  B, Krželj  V, Kuzmić-Prusac  I  et al.  The OSR1 rs12329305 polymorphism contributes to the development of congenital malformations in cases of stillborn/neonatal death. Med Sci Monit  2014;20:1531–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Anderson  BR, Howell  DN, Soldano  K  et al.  In vivo modeling implicates APOL1 in nephropathy: evidence for dominant negative effects and epistasis under anemic stress. PLoS Genet  2015;11:e1005349. 10.1371/journal.pgen.1005349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Bolar  NA, Golzio  C, Živná  M  et al.  Heterozygous loss-of-function SEC61A1 mutations cause autosomal-dominant tubulo-interstitial and glomerulocystic kidney disease with anemia. Am Hum Genet  2016;99:174–87. 10.1016/j.ajhg.2016.05.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Daga  S, Donati  F, Capitani  K  et al.  New frontiers to cure Alport syndrome: COL4A3 and COL4A5 gene editing in podocyte-lineage cells. Eur J Hum Genet  2020;28:480–90. 10.1038/s41431-019-0537-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wang  J, Qiu  Y, Zhang  L  et al.  Adenine base editor corrected ADPKD point mutations in hiPSCs and kidney organoids. Adv Biotechnol  2024;2:20. 10.1007/s44307-024-00026-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Chen  Z, Zhang  D, Zheng  R  et al.  In vivo base editing rescues primary hyperoxaluria type 1 in rats. Kidney Int  2024;105:496–507. 10.1016/j.kint.2023.11.029 [DOI] [PubMed] [Google Scholar]
  • 53. Jerke  U, Eulenberg-Gustavus  C, Wagner  DL  et al.  CRISPR-Cas9 mediated proteinase 3 autoantigen deletion as a treatment strategy for anti-neutrophil cytoplasmic autoantibody-associated vasculitis. Kidney Int  2025;108:145–9. [DOI] [PubMed] [Google Scholar]
  • 54. Musunuru  K, Grandinette  SA, Wang  X  et al.  Patient-specific in vivo gene editing to treat a rare genetic disease. N Engl J Med  2025;392:2235–43. 10.1056/NEJMoa2504747 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Harrison  JH, Merrill  JP, Murray  JE. Renal homotransplantation in identical twins. Surg Forum  1956;6:432–6. [PubMed] [Google Scholar]
  • 56. Tantisattamo  E, Maggiore  U, Piccoli  GB. History of kidney transplantation: a journey of progression and evolution for success. J Nephrol  2022;35:1783–6. 10.1007/s40620-022-01453-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Boenink  R, Kramer  A, Tuinhout  RE  et al.  Trends in kidney transplantation rate across Europe: study from the ERA Registry. Nephrol Dial Transplant  2023;38:1528–39. 10.1093/ndt/gfac333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Stel  VS, Boenink  R, Astley  ME  et al.  A comparison of the epidemiology of kidney replacement therapy between Europe and the United States: 2021 data of the ERA Registry and the USRDS. Nephrol Dial Transplant  2024;39:1593–603. 10.1093/ndt/gfae040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Argani  H. Expanded criteria donors. Exp Clin Transplant  2022;20:13–9. 10.6002/ect.DonorSymp.2022.L13 [DOI] [PubMed] [Google Scholar]
  • 60. Filiopoulos  V, Boletis  JN. Renal transplantation with expanded criteria donors: which is the optimal immunosuppression?  World J Transplant  2016;6:103–14. 10.5500/wjt.v6.i1.103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Xi  J, Zheng  W, Chen  M  et al.  Genetically engineered pigs for xenotransplantation: hopes and challenges. Front Cell Dev Biol  2023;10:1093534. 10.3389/fcell.2022.1093534 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Cooper  DKC, Cozzi  E. The immense potential of xenotransplantation. Eur J Transplant  2023;1:163–6. 10.57603/EJT-272 [DOI] [Google Scholar]
  • 63. Ryczek  N, Hryhorowicz  M, Zeyland  J  et al.  CRISPR/Cas technology in pig-to-human xenotransplantation research. Int J Mol Sci  2021;22:3196. 10.3390/ijms22063196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Cooper  DKC, Ekser  B, Ramsoondar  J  et al.  The role of genetically engineered pigs in xenotransplantation research. J Pathol  2016;238:288–99. 10.1002/path.4635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Cooper  DKC, Ekser  B, Tector  AJ. Immunobiological barriers to xenotransplantation. Int J Surg  2015;23:211–6. 10.1016/j.ijsu.2015.06.068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Ngo  BT-T, Beiras-Fernandez  A, Hammer  C  et al.  Hyperacute rejection in the xenogenic transplanted rat liver is triggered by the complement system only in the presence of leukocytes and free radical species. Xenotransplantation  2013;20:177–87. 10.1111/xen.12035 [DOI] [PubMed] [Google Scholar]
  • 67. Lin  SS, Hanaway  MJ, Gonzalez-Stawinski  GV  et al.  The role of anti-Galα1-3Gal antibodies in acute vascular rejection and accommodation of xenografts. Transplantation  2000;70:1667–74. 10.1097/00007890-200012270-00002 [DOI] [PubMed] [Google Scholar]
  • 68. Varki  A. Loss of N-glycolylneuraminic acid in humans: mechanisms, consequences, and implications for hominid evolution. Am J Phys Anthropol  2001;116:54–69. 10.1002/ajpa.10018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Byrne  G, Ahmad-Villiers  S, Du  Z  et al.  B4GALNT2 and xenotransplantation: a newly appreciated xenogeneic antigen. Xenotransplantation  2018;25:e12394. 10.1111/xen.12394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Estrada  JL, Martens  G, Li  P  et al.  Evaluation of human and non-human primate antibody binding to pig cells lacking GGTA1/CMAH/β4GalNT2 genes. Xenotransplantation  2015;22:194–202. 10.1111/xen.12161 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Wang  Y, Du  Y, Zhou  X  et al.  Efficient generation of B2m-null pigs via injection of zygote with TALENs. Sci Rep  2016;6:38854. 10.1038/srep38854 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Long  EO, Kim  HS, Liu  D  et al.  Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol  2013;31:227–58. 10.1146/annurev-immunol-020711-075005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Phelps  CJ, Ball  SF, Vaught  TD  et al.  Production and characterization of transgenic pigs expressing porcine CTLA4-Ig. Xenotransplantation  2009;16:477–85. 10.1111/j.1399-3089.2009.00533.x [DOI] [PubMed] [Google Scholar]
  • 74. Reyes  LM, Estrada  JL, Wang  ZY  et al.  Creating class I MHC-null pigs using guide RNA and the Cas9 endonuclease. J Immunol  2014;193:5751–7. 10.4049/jimmunol.1402059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Cruz  NM, Freedman  BS. CRISPR gene editing in the kidney. Am J Kidney Dis  2018;71:874–83. 10.1053/j.ajkd.2018.02.347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Niu  D, Wei  H-J, Lin  L  et al.  Inactivation of porcine endogenous retrovirus in pigs using CRISPR-Cas9. Science  2017;357:1303–7. 10.1126/science.aan4187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Montgomery  RA, Stern  JM, Lonze  BE  et al.  Results of two cases of pig-to-human kidney xenotransplantation. N Engl J Med  2022;386:1889–98. 10.1056/NEJMoa2120238 [DOI] [PubMed] [Google Scholar]
  • 78. Porrett  PM, Orandi  BJ, Kumar  V  et al.  First clinical-grade porcine kidney xenotransplant using a human decedent model. Am J Transplant  2022;22:1037–53. 10.1111/ajt.16930 [DOI] [PubMed] [Google Scholar]
  • 79. Locke  JE, Kumar  V, Anderson  D  et al.  Normal graft function after pig-to-human kidney xenotransplant. JAMA Surg  2023;158:1106–8. 10.1001/jamasurg.2023.2774 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Judd  E, Kumar  V, Porrett  PM  et al.  Physiologic homeostasis after pig-to-human kidney xenotransplantation. Kidney Int  2024;105:971–9. 10.1016/j.kint.2024.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Kawai  T, Williams  WW, Elias  N  et al.  Xenotransplantation of a porcine kidney for end-stage kidney disease. N Engl J Med  2025;392:1933–40. [DOI] [PubMed] [Google Scholar]
  • 82. Mallapaty  S, Kozlov  M. First pig kidney transplant in a person: what it means for the future. Nature  2024;628:13–4. 10.1038/d41586-024-00879-y [DOI] [PubMed] [Google Scholar]
  • 83. Saha  K, Sontheimer  EJ, Brooks  PJ  et al.  The NIH Somatic Cell Genome Editing program. Nature  2021;592:195–204. 10.1038/s41586-021-03191-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Freedman  BS, Lam  AQ, Sundsbak  JL  et al.  Reduced ciliary polycystin-2 in induced pluripotent stem cells from polycystic kidney disease patients with PKD1 mutations. J Am Soc Nephrol  2013;24:1571–86. 10.1681/ASN.2012111089 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Hiratsuka  K, Miyoshi  T, Kroll  KT  et al.  Organoid-on-a-chip model of human ARPKD reveals mechanosensing pathomechanisms for drug discovery. Sci Adv  2022;8:eabq0866. 10.1126/sciadv.abq0866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Ohmori  T, De  S, Tanigawa  S  et al.  Impaired NEPHRIN localization in kidney organoids derived from nephrotic patient iPS cells. Sci Rep  2021;11:3982. 10.1038/s41598-021-83501-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Cruz  NM, Reddy  R, McFaline-Figueroa  JL  et al.  Modelling ciliopathy phenotypes in human tissues derived from pluripotent stem cells with genetically ablated cilia. Nat Biomed Eng  2022;6:463–75. 10.1038/s41551-022-00880-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Kim  JW, Kim  HW, Nam  SA  et al.  Human kidney organoids reveal the role of glutathione in Fabry disease. Exp Mol Med  2021;53:1580–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Cui  S, Fang  X, Lee  H  et al.  Modeling of Fabry disease nephropathy using patient derived human induced pluripotent stem cells and kidney organoid system. J Transl Med  2023;21:138. 10.1186/s12967-023-03992-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Cui  S, Shin  YJ, Fang  X  et al.  CRISPR/Cas9-mediated A4GALT suppression rescues Fabry disease phenotypes in a kidney organoid model. Transl Res  2023;258:35–46. 10.1016/j.trsl.2023.02.005 [DOI] [PubMed] [Google Scholar]
  • 91. Namba  M, Kobayashi  T, Kohno  M  et al.  Creation of X-linked Alport syndrome rat model with Col4a5 deficiency. Sci Rep  2021;11:20836. 10.1038/s41598-021-00354-y [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No new data were generated or analysed in support of this research.


Articles from Clinical Kidney Journal are provided here courtesy of Oxford University Press

RESOURCES