Novel PNLDC1 mutations underlie nonobstructive azoospermia in humans and mice.

  • Abstract
  • Literature Map
  • Similar Papers
Abstract
Translate article icon Translate Article Star icon
Take notes icon Take Notes

PIWI-interacting RNAs (piRNAs) are small regulatory RNAs (21-35 nucleotides) exclusively expressed in germ cells, where they play a critical role in transposable element repression and post-meiotic gene regulation. The poly(A)-specific RNase-like domain-containing 1 (PNLDC1) protein is essential for piRNA maturation, specifically in 3'-end trimming. Disruption of PNLDC1 has been implicated in nonobstructive azoospermia (NOA) and male infertility. Through whole-exome sequencing, we identified a compound heterozygous mutation (MT1 c.449G > A, p.Trp150* and MT2 c.821A > G, p.His274Ala) in a Chinese NOA patient (P9241) and a homozygous nonsense mutation (MT3 c.1288C > T, p.Arg430*) in a Pakistani NOA patient (II:2) born to a consanguineous couple. Mutant PNLDC1 mRNA was detected, but not the corresponding protein, was detected in the testes of P9241. In contrast, truncated PNLDC1 protein was observed in HEK293T cells transfected with a plasmid harboring mutation MT3. To investigate the functional consequences, we generated a Pnldc1KI/KI mouse model mimicking the MT3 using CRISPR/Cas9 genome editing, which exhibited infertility due to spermiogenesis arrest, phenocopying the NOA condition in patient II:2. Notably, Pnldc1KI/KI testes showed significant derepression of the retrotransposon LINE1 and increased spermatid apoptosis. These findings provide strong functional evidence that PNLDC1 mutations disrupt piRNA biogenesis, impair spermatogenesis, and underlie NOA in both humans and mice.

Similar Papers
  • Research Article
  • 10.1093/humrep/deaf097.324
P-015 Pathogenic mutations in DMRTB1 cause human male infertility with non-obstructive azoospermia by impairing transcription regulation in spermatogenesis
  • Jun 1, 2025
  • Human Reproduction
  • T Hu + 5 more

Study question What is the role of DMRTB1 in the etiology of non-obstructive azoospermia (NOA) and spermatogenesis in humans? Summary answer DMRTB1 pathogenic mutation leads to spermatogenesis by disrupting transcriptional regulation, ultimately resulting in NOA in humans. What is known already DMRTB1 is a member of the DM domain family. Dmrtb1 knockout mice showed male infertility, which indicated that Dmrtb1 is associated with spermatogenesis. The specific role of DMRTB1 in regulating spermatogenesis is unclear, and clinical evidence for the relationship between DMRTB1 and the pathogenesis of human male infertility is still unknown. Study design, size, duration We performed DMRTB1 mutation screening by whole-exome sequencing(WES) from 626 infertile men with NOA, and 392 fertile individuals (as control). We conducted pedigree analysis and a series of bioinformatics analyses on two patients identified as carrying DMRTB1 mutations. DMRTB1-specific antibodies were used to detect the expression of DMRTB1 in patients, and Immunoprecipitation-Mass spectrometry(IP-MS) was performed in mouse testes for subsequent mechanism exploration. The entire study was conducted over 2 years. Participants/materials, setting, methods WES was performed in participants of this cohort, followed by Sanger sequencing validation. Bioinformatics predictions were performed to evaluate the pathogenicity of candidate disease-causing variants. Hematoxylin and eosinstaining, and immunofluorescence assays (IF) were performed to detect the spermatogenesis of testis. Dual-luciferase assay, IP-MS, co-immunoprecipitation (CO-IP), western blotting (WB), and IF were performed to identify the mutations' impact on function in-vivo and in-vitro. Main results and the role of chance Following a large-scale sequencing screening from clinical centers, our study identified two infertile men with NOA who were carriers of different DMRTB1 mutations (P1:c.446_449dupGAGC[p.A151Sfs*58]; P2:c.154G>A[p.A52T] and c.793C>T [p.P265S] ) from unrelated families. All are co-segregating with the disease according to a recessive pattern of inheritance. Functional evidence based on in-vivo and in-vitro has shown that these mutations cause abnormal expression of DMRTB1 protein and impair the transcription regulation of the promoter of PIWIL2 and PIWIL1. Overall, our results support the hypothesis that DMRTB1 expression was affected by pathogenic mutations identified in patients and it inhibits PIWIL1 and PIWIL2 transcription, thereby arresting normal spermatogenesis. Limitations, reasons for caution This is a preliminary report suggesting that defects in DMRTB1 can lead to NOA in humans. The pathogenic mechanism could be further validated in the knock-in mouse model in the future. Wider implications of the findings Our study of DMRTB1 in humans provides new information on the transcription regulatory mechanism of spermatogenesis, suggesting that these pathogenic mutations in DMRTB1 are etiological factors in patients with NOA and providing a valuable clue for genetic counselling on the DMRTB1 mutants associated with infertility in human males. Trial registration number No

  • Addendum
  • Cite Count Icon 2
  • 10.1093/humrep/deaa179
Corrigendum. Sequencing of a 'mouse azoospermia' gene panel in azoospermic men: identification of RNF212 and STAG3 mutations as novel genetic causes of meiotic arrest.
  • Jul 27, 2020
  • Human Reproduction
  • A Riera-Escamilla + 12 more

Corrigendum. Sequencing of a 'mouse azoospermia' gene panel in azoospermic men: identification of RNF212 and STAG3 mutations as novel genetic causes of meiotic arrest.

  • Research Article
  • Cite Count Icon 72
  • 10.1093/humrep/dez042
Sequencing of a 'mouse azoospermia' gene panel in azoospermic men: identification of RNF212 and STAG3 mutations as novel genetic causes of meiotic arrest.
  • May 24, 2019
  • Human Reproduction
  • A Riera-Escamilla + 12 more

What is the diagnostic potential of next generation sequencing (NGS) based on a 'mouse azoospermia' gene panel in human non-obstructive azoospermia (NOA)? The diagnostic performance of sequencing a gene panel based on genes associated with mouse azoospermia was relatively successful in idiopathic NOA patients and allowed the discovery of two novel genes involved in NOA due to meiotic arrest. NOA is a largely heterogeneous clinical entity, which includes different histological pictures. In a large proportion of NOA, the aetiology remains unknown (idiopathic NOA) and yet, unknown genetic factors are likely to play be involved. The mouse is the most broadly used mammalian model for studying human disease because of its usefulness for genetic manipulation and its genetic and physiological similarities to man. Mouse azoospermia models are available in the Mouse Genome Informatics database (MGI: http://www.informatics.jax.org/). The first step was to design of a 'mouse azoospermia' gene panel through the consultation of MGI. The second step was NGS analysis of 175 genes in a group of highly selected NOA patients (n = 33). The third step was characterization of the discovered gene defects in human testis tissue, through meiotic studies using surplus testicular biopsy material from the carriers of the RNF212 and STAG3 pathogenic variants. The final step was RNF212 and STAG3 expression analysis in a collection of testis biopsies. From a total of 1300 infertile patients, 33 idiopathic NOA patients were analysed in this study, including 31 unrelated men and 2 brothers from a consanguineous family. The testis histology of the 31 unrelated NOA patients was as follows: 20 Sertoli cell-only syndrome (SCOS), 11 spermatogenic arrest (6 spermatogonial arrest and 5 spermatocytic arrest). The two brothers were affected by spermatocytic arrest. DNA extracted from blood was used for NGS on Illumina NextSeq500 platform. Generated sequence data was filtered for rare and potentially pathogenic variants. Functional studies in surplus testicular tissue from the carriers included the investigation of meiotic entry, XY body formation and metaphases by performing fluorescent immunohistochemical staining and immunocytochemistry. mRNA expression analysis through RT-qPCR of RNF212 and STAG3 was carried out in a collection of testis biopsies with different histology. Our approach was relatively successful, leading to the genetic diagnosis of one sporadic NOA patient and two NOA brothers. This relatively high diagnostic performance is likely to be related to the stringent patient selection criteria i.e. all known causes of azoospermia were excluded and to the relatively high number of patients with rare testis histology (spermatocytic arrest). All three mutation carriers presented meiotic arrest, leading to the genetic diagnosis of three out of seven cases with this specific testicular phenotype. For the first time, we report biallelic variants in STAG3, in one sporadic patient, and a homozygous RNF212 variant, in the two brothers, as the genetic cause of NOA. Meiotic studies allowed the detection of the functional consequences of the mutations and provided information on the role of STAG3 and RNF212 in human male meiosis. All genes, with the exception of 5 out of 175, included in the panel cause azoospermia in mice only in the homozygous or hemizygous state. Consequently, apart from the five known dominant genes, heterozygous variants (except compound heterozygosity) in the remaining genes were not taken into consideration as causes of NOA. We identified the genetic cause in approximately half of the patients with spermatocytic arrest. The low number of analysed patients can be considered as a limitation, but it is a very rare testis phenotype. Due to the low frequency of this specific phenotype among infertile men, our finding may be considered of low clinical impact. However, at an individual level, it does have relevance for prognostic purposes prior testicular sperm extraction. Our study represents an additional step towards elucidating the genetic bases of early spermatogenic failure, since we discovered two new genes involved in human male meiotic arrest. We propose the inclusion of RNF212 and STAG3 in a future male infertility diagnostic gene panel. Based on the associated testis phenotype, the identification of pathogenic mutations in these genes also confers a negative predictive value for testicular sperm retrieval. Our meiotic studies provide novel insights into the role of these proteins in human male meiosis. Mutations in STAG3 were first described as a cause of female infertility and ovarian cancer, and Rnf212 knock out in mice leads to male and female infertility. Hence, our results stimulate further research on shared genetic factors causing infertility in both sexes and indicate that genetic counselling should involve not only male but also female relatives of NOA patients. This work was funded by the Spanish Ministry of Health Instituto Carlos III-FIS (grant number: FIS/FEDER-PI14/01250; PI17/01822) awarded to CK and AR-E, and by the European Commission, Reproductive Biology Early Research Training (REPROTRAIN, EU-FP7-PEOPLE-2011-ITN289880), awarded to CK, WB, and AE-M. The authors have no conflict of interest.

  • Research Article
  • Cite Count Icon 8
  • 10.1093/hropen/hoad022
Biallelic mutations in RNA-binding protein ADAD2 cause spermiogenic failure and non-obstructive azoospermia in humans.
  • May 18, 2023
  • Human Reproduction Open
  • Baolu Shi + 18 more

What are some pathogenic mutations for non-obstructive azoospermia (NOA) and their effects on spermatogenesis? Biallelic missense and frameshift mutations in ADAD2 disrupt the differentiation of round spermatids to spermatozoa causing azoospermia in humans and mice. NOA is the most severe cause of male infertility characterized by an absence of sperm in the ejaculate due to impairment of spermatogenesis. In mice, the lack of the RNA-binding protein ADAD2 leads to a complete absence of sperm in epididymides due to failure of spemiogenesis, but the spermatogenic effects of ADAD2 mutations in human NOA-associated infertility require functional verification. Six infertile male patients from three unrelated families were diagnosed with NOA at local hospitals in Pakistan based on infertility history, sex hormone levels, two semen analyses and scrotal ultrasound. Testicular biopsies were performed in two of the six patients. Adad2 mutant mice (Adad2Mut/Mut) carrying mutations similar to those found in NOA patients were generated using the CRISPR/Cas9 genome editing tool. Reproductive phenotypes of Adad2Mut/Mut mice were verified at 2 months of age. Round spermatids from the littermates of wild-type (WT) and Adad2Mut/Mut mice were randomly selected and injected into stimulated WT oocytes. This round spermatid injection (ROSI) procedure was conducted with three biological replicates and >400 ROSI-derived zygotes were evaluated. The fertility of the ROSI-derived progeny was evaluated for three months in four Adad2WT/Mut male mice and six Adad2WT/Mut female mice. A total of 120 Adad2Mut/Mut, Adad2WT/Mut, and WT mice were used in this study. The entire study was conducted over 3 years. Whole-exome sequencing was performed to detect potentially pathogenic mutations in the six NOA-affected patients. The pathogenicity of the identified ADAD2 mutations was assessed and validated in human testicular tissues and in mouse models recapitulating the mutations in the NOA patients using quantitative PCR, western blotting, hematoxylin-eosin staining, Periodic acid-Schiff staining, and immunofluorescence. Round spermatids of WT and Adad2Mut/Mut mice were collected by fluorescence-activated cell sorting and injected into stimulated WT oocytes. The development of ROSI-derived offspring was evaluated in the embryonic and postnatal stages. Three recessive mutations were identified in ADAD2 (MT1: c.G829T, p.G277C; MT2: c.G1192A, p.D398N; MT3: c.917_918del, p.Q306Rfs*43) in patients from three unrelated Pakistani families. MT1 and MT2 dramatically reduced the testicular expression of ADAD2, likely causing spermiogenesis failure in the NOA patients. Immunofluorescence analysis of the Adad2Mut/Mut male mice with the corresponding MT3 mutation showed instability and premature degradation of the ADAD2 protein, resulting in the spermiogenesis deficiency phenotype. Through ROSI, the Adad2Mut/Mut mice could produce pups with comparable embryonic development (46.7% in Adad2Mut/Mut versus 50% in WT) and birth rates (21.45 ± 10.43% in Adad2Mut/Mut versus 27.5 ± 3.536% in WT, P = 0.5044) to WT mice. The Adad2WT/Mut progeny from ROSI (17 pups in total via three ROSI replicates) did not show overt developmental defects and had normal fertility. N/A. This is a preliminary report suggesting that ROSI can be an effective treatment for infertile Adad2Mut/Mut mice. Further assisted reproductive attempts need to be carefully examined in humans during clinical trials. Our work provides functional evidence that mutations in the ADAD2 gene are deleterious and cause consistent spermiogenic defects in both humans and mice. In addition, preliminary results show that ROSI can help Adad2Mut/Mut to produce biological progeny. These findings provide valuable clues for genetic counselling on the ADAD2 mutants-associated infertility in human males. This work was supported by the National Natural Science Foundation of China (32000587, U21A20204, and 32061143006), and the National Key Research and Developmental Program of China (2019YFA0802600 and 2021YFC2700202). This work was also supported by Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China. The authors declare no competing interests.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 121
  • 10.1371/journal.pgen.1003645
The Wilms Tumor Gene, Wt1, Is Critical for Mouse Spermatogenesis via Regulation of Sertoli Cell Polarity and Is Associated with Non-Obstructive Azoospermia in Humans
  • Aug 1, 2013
  • PLoS Genetics
  • Xiao Na Wang + 23 more

Azoospermia is one of the major reproductive disorders which cause male infertility in humans; however, the etiology of this disease is largely unknown. In the present study, six missense mutations of WT1 gene were detected in 529 human patients with non-obstructive azoospermia (NOA), indicating a strong association between WT1 mutation and NOA. The Wilms tumor gene, Wt1, is specifically expressed in Sertoli cells (SCs) which support spermatogenesis. To examine the functions of this gene in spermatogenesis, Wt1 was deleted in adult testis using Wt1flox and Cre-ERTM mice strains. We found that inactivation of Wt1 resulted in massive germ cell death and only SCs were present in most of the seminiferous tubules which was very similar to NOA in humans. In investigating the potential mechanism for this, histological studies revealed that the blood–testis barrier (BTB) was disrupted in Wt1 deficient testes. In vitro studies demonstrated that Wt1 was essential for cell polarity maintenance in SCs. Further studies found that the expression of cell polarity associated genes (Par6b and E-cadherin) and Wnt signaling genes (Wnt4, Wnt11) were downregulated in Wt1 deficient SCs, and that the expression of Par6b and E-cadherin was regulated by Wnt4. Our findings suggest that Wt1 is important in spermatogenesis by regulating the polarity of SCs via Wnt signaling pathway and that WT1 mutation is one of the genetic causes of NOA in humans.

  • Research Article
  • 10.1038/s10038-025-01350-0
Bi-allelic KCTD19 variants associated with meiotic arrest and non-obstructive azoospermia in humans.
  • May 23, 2025
  • Journal of human genetics
  • Shuai Xu + 15 more

Non-obstructive azoospermia (NOA) represents the severe form of male infertility, affecting approximately 1% of men during their reproductive years. It is marked by the absence of sperm production caused by testicular dysfunction and has many genetic origins. However, the genetic factors underlying most NOA cases are still unclear. Meiosis, a crucial process ensuring accurate chromosome segregation and generating genetic diversity in gametes, is susceptible to genetic disruptions that may result in NOA. In this study, whole exome sequencing (WES) was conducted on 969 NOA patients, identifying six compound heterozygous KCTD19 variants in three Chinese pedigrees. KCTD19 has been demonstrated to interact with ZFP541 and HDAC1, thereby participating in the modulation of chromatin remodeling and transcriptional programs during meiosis in mice. Herein, our findings expand the phenotypic and mutational spectrum of KCTD19 in male infertility and provide further insights into its role during meiosis. This research underscores the importance of KCTD19 in meiotic progression and male fertility, highlighting the need for further investigation into the molecular mechanisms underlying gametogenic failure in NOA.

  • Research Article
  • Cite Count Icon 65
  • 10.1073/pnas.1513682113
Major spliceosome defects cause male infertility and are associated with nonobstructive azoospermia in humans
  • Mar 28, 2016
  • Proceedings of the National Academy of Sciences
  • Hao Wu + 12 more

Processing of pre-mRNA into mRNA is an important regulatory mechanism in eukaryotes that is mediated by the spliceosome, a huge and dynamic ribonucleoprotein complex. Splicing defects are implicated in a spectrum of human disease, but the underlying mechanistic links remain largely unresolved. Using a genome-wide association approach, we have recently identified single nucleotide polymorphisms in humans that associate with nonobstructive azoospermia (NOA), a common cause of male infertility. Here, using genetic manipulation of corresponding candidate loci in Drosophila, we show that the spliceosome component SNRPA1/U2A is essential for male fertility. Loss of U2A in germ cells of the Drosophila testis does not affect germline stem cells, but does result in the accumulation of mitotic spermatogonia that fail to differentiate into spermatocytes and mature sperm. Lack of U2A causes insufficient splicing of mRNAs required for the transition of germ cells from proliferation to differentiation. We show that germ cell-specific disruption of other components of the major spliceosome manifests with the same phenotype, demonstrating that mRNA processing is required for the differentiation of spermatogonia. This requirement is conserved, and expression of human SNRPA1 fully restores spermatogenesis in U2A mutant flies. We further report that several missense mutations in human SNRPA1 that inhibit the assembly of the major spliceosome dominantly disrupt spermatogonial differentiation in Drosophila. Collectively, our findings uncover a conserved and specific requirement for the major spliceosome during the transition from spermatogonial proliferation to differentiation in the male testis, suggesting that spliceosome defects affecting the differentiation of human spermatogonia contribute to NOA.

  • Research Article
  • Cite Count Icon 5
  • 10.3389/fgene.2022.1017302
A homozygous missense variant in DND1 causes non-obstructive azoospermia in humans.
  • Sep 30, 2022
  • Frontiers in genetics
  • Xuefeng Xie + 18 more

Non-obstructive azoospermia (NOA) is a severe factor of male infertility; it affects approximately 1% of the global male population and accounts for 40% of male infertility cases. However, the majority of NOA cases remain idiopathic. This is the first study using whole-exome sequencing (WES) to identify a novel missense mutation in the DND1 gene (c.212A>C, p. E71A) from a Pakistani family, that includes three males with NOA. This mutation is predicted to cause DND1 protein misfolding and weaken the DND1 interaction with NANOS2, a significant regulator in primordial germ cell development. Our study identified a DND1 pathogenic mutation in NOA patients and highlighted its critical role in male fertility in humans.

  • Peer Review Report
  • 10.1111/cge.13475/v2/decision1
Decision letter for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"
  • Nov 8, 2018
  • Reiner A Veitia

Decision letter for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"

  • Peer Review Report
  • 10.1111/cge.13475/v2/response1
Author response for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"
  • Oct 24, 2018
  • Ya-Xin Zhang + 9 more

Author response for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"

  • Peer Review Report
  • 10.1111/cge.13475/v1/review1
Review for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"
  • Sep 25, 2018

Review for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"

  • Peer Review Report
  • 10.1111/cge.13475/v1/decision1
Decision letter for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"
  • Oct 10, 2018
  • Reiner A Veitia

Decision letter for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"

  • Peer Review Report
  • 10.1111/cge.13475/v1/review2
Review for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"
  • Oct 1, 2018
  • Toshinobu Miyamoto

Review for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"

  • Peer Review Report
  • 10.1111/cge.13475/v2/review1
Review for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"
  • Nov 8, 2018

Review for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"

  • Peer Review Report
  • 10.1111/cge.13475/v1/review3
Review for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"
  • Oct 5, 2018

Review for "<i>XRCC2</i> mutation causes premature ovarian insufficiency as well as non-obstructive azoospermia in humans"

More from: Human molecular genetics
  • New
  • Addendum
  • 10.1093/hmg/ddaf170
Correction to: The role of senescence and prosurvival signaling in controlling the oncogenic activity of FGFR2 mutants associated with cancer and birth defects.
  • Oct 31, 2025
  • Human molecular genetics

  • New
  • Research Article
  • 10.1093/hmg/ddaf163
Haploinsufficiency of GRHL2 is associated with orofacial clefting in humans.
  • Oct 31, 2025
  • Human molecular genetics
  • Sarah W Curtis + 20 more

  • Research Article
  • 10.1093/hmg/ddaf160
Impact of somatic XIST deletions on ongoing XIST expression and inactive X silencing and heterochromatin.
  • Oct 23, 2025
  • Human molecular genetics
  • Karanveer S Bhangu + 3 more

  • Research Article
  • 10.1093/hmg/ddaf150
Genetic dissection of Huntington's disease modification by variation at RRM2B.
  • Oct 15, 2025
  • Human molecular genetics
  • Kiuk Lee + 17 more

  • Research Article
  • 10.1093/hmg/ddaf156
Mosaic expression of SLC35A2 pathogenetic variants impairs neuronal migration and dendritogenesis in the developing cortex.
  • Oct 13, 2025
  • Human molecular genetics
  • Antonio Falace + 15 more

  • Research Article
  • 10.1093/hmg/ddaf159
Compromised lipid metabolism, mitochondria respiration and neuroprotective effects in iPSC-derived astrocytes from a Smith-Lemli-Opitz syndrome patient.
  • Oct 12, 2025
  • Human molecular genetics
  • Keiji Kawatani + 9 more

  • Research Article
  • 10.1093/hmg/ddaf157
Q373fs variant of RBM20 affects splicing and expression of cardiac-related genes and cardiac function: human sudden death case and mouse experiments.
  • Oct 12, 2025
  • Human molecular genetics
  • Aya Miura + 13 more

  • Research Article
  • 10.1093/hmg/ddaf131
Genetic heterogeneity and homogeneity among orofacial cleft subtypes: genome-wide association studies in the cleft collective.
  • Oct 11, 2025
  • Human molecular genetics
  • Kyle Dack + 14 more

  • Research Article
  • 10.1093/hmg/ddaf154
Differential expression of a disease-associated MRE11 variant reveals distinct phenotypic outcomes.
  • Oct 11, 2025
  • Human molecular genetics
  • Mckenna B Defoer + 8 more

  • Research Article
  • 10.1093/hmg/ddaf151
Genetic variants associated with gout identified through a genome-wide study in the UK biobank (N = 150 542).
  • Oct 11, 2025
  • Human molecular genetics
  • Yiwen Tao + 8 more

Save Icon
Up Arrow
Open/Close
  • Ask R Discovery Star icon
  • Chat PDF Star icon

AI summaries and top papers from 250M+ research sources.

Search IconWhat is the difference between bacteria and viruses?
Open In New Tab Icon
Search IconWhat is the function of the immune system?
Open In New Tab Icon
Search IconCan diabetes be passed down from one generation to the next?
Open In New Tab Icon