0% found this document useful (0 votes)
15 views35 pages

GREGoR - Accelerating Genomics For Rare Diseases - Nature

The GREGoR Consortium aims to enhance the understanding and diagnosis of rare diseases, which affect 1 in 20 individuals globally, by utilizing advanced genomics technologies. Despite recent advancements, over half of rare disease cases remain undiagnosed, prompting GREGoR to standardize and evaluate new genomic approaches across thousands of challenging cases. The consortium has generated extensive datasets from over 7,500 individuals, facilitating global research efforts to improve genetic diagnosis in rare diseases.

Uploaded by

hamadaosama1989
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
15 views35 pages

GREGoR - Accelerating Genomics For Rare Diseases - Nature

The GREGoR Consortium aims to enhance the understanding and diagnosis of rare diseases, which affect 1 in 20 individuals globally, by utilizing advanced genomics technologies. Despite recent advancements, over half of rare disease cases remain undiagnosed, prompting GREGoR to standardize and evaluate new genomic approaches across thousands of challenging cases. The consortium has generated extensive datasets from over 7,500 individuals, facilitating global research efforts to improve genetic diagnosis in rare diseases.

Uploaded by

hamadaosama1989
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd

11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

nature perspectives article

Perspective Published: 12 November 2025

GREGoR: accelerating genomics for rare diseases


Moez Dawood , Ben Heavner, Marsha M. Wheeler, Rachel A. Ungar, Jonathan LoTempio, Laurens
Wiel, Seth Berger, Jonathan A. Bernstein, Jessica X. Chong, Emmanuèle C. Délot, Evan E. Eichler,
James R. Lupski, Ali Shojaie, Michael E. Talkowski, Alex H. Wagner, Chia-Lin Wei, Christopher
Wellington, Matthew T. Wheeler, GREGoR Partner Members, Claudia M. B. Carvalho, Richard A.
Gibbs, Casey A. Gifford, Susanne May, Danny E. Miller, Genomics Research to Elucidate the Genetics
of Rare Diseases (GREGoR) Consortium Show authors

Nature 647, 331–342 (2025)

1900 Accesses 17 Altmetric Metrics

Abstract

Rare diseases are collectively common, affecting approximately 1 in 20 individuals


worldwide. In recent years, rapid progress has been made in rare disease diagnostics due
to advances in next-generation sequencing, development of new computational and
functional genomics approaches to prioritize genes and variants and increased global
sharing of clinical and genetic data. However, more than half of individuals suspected to
have a rare disease lack a genetic diagnosis. The Genomics Research to Elucidate the
Genetics of Rare Diseases (GREGoR) Consortium was initiated to study thousands of
challenging rare disease cases and families and apply, standardize and evaluate
emerging genomics technologies and analytics to accelerate their adoption in clinical
practice. Furthermore, all data generated, currently representing over 7,500 individuals
from over 3,000 families, are rapidly made available to researchers worldwide through
the Analysis, Visualization and Informatics Lab-space (AnVIL) to catalyse global efforts
to develop approaches for genetic diagnoses in rare diseases. Most of these families
[Link] 1/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

have undergone previous clinical genetic testing but remained unsolved, with most
being exome-negative. Here we describe the collaborative research framework, datasets
and discoveries comprising GREGoR that will provide foundational resources and
substrates for the future of rare disease genomics.

This is a preview of subscription content, access via your institution

Access options

Access through your institution

Access Nature and 54 other Nature Portfolio


journals
Get Nature+, our best-value online-access subscription

$32.99 / 30 days
cancel any time

Learn more

Buy this article Subscribe to this journal


Receive 51 print issues and online access

Purchase on SpringerLink
Instant access to full article PDF
$199.00 per year
only $3.90 per issue

[Link] 2/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

Buy now
Prices Learnduring
may be subject to local taxes which are calculated more checkout

Additional access options:

Log in
Learn about institutional subscriptions
Read our FAQs
Contact customer support

Similar content being viewed by others

Genomic reanalysis of a Solving patients with rare Rare disease gene


pan-European rare-disease diseases through association discovery in
resource yields new programmatic reanalysis the 100,000 Genomes
diagnoses of genome-phenome data Project

Article Open access Article Open access Article Open access


17 January 2025 01 June 2021 26 February 2025

References

1. Baxter, S. M. et al. Centers for Mendelian Genomics: a decade of facilitating gene


discovery. Genet. Med. 24, 784–797 (2022).

2. Taylor, J. C. et al. Factors influencing success of clinical genome sequencing across a


broad spectrum of disorders. Nat. Genet. 47, 717–726 (2015).

[Link] 3/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

3. Wright, C. F. et al. Genomic diagnosis of rare pediatric disease in the United Kingdom
and Ireland. N. Engl. J. Med. 388, 1559–1571 (2023).

4. Rimmer, A. et al. Integrating mapping-, assembly- and haplotype-based approaches


for calling variants in clinical sequencing applications. Nat. Genet. 46, 912–918
(2014).

5. Monaco, L. et al. Research on rare diseases: ten years of progress and challenges at
IRDiRC. Nat. Rev. Drug Discov. 21, 319–320 (2022).

6. Yang, Y. et al. Molecular findings among patients referred for clinical whole-exome
sequencing. JAMA 312, 1870–1879 (2014).

7. Farnaes, L. et al. Rapid whole-genome sequencing decreases infant morbidity and


cost of hospitalization. npj Genom. Med. 3, 10 (2018).

8. Posey, J. E. et al. Resolution of disease phenotypes resulting from multilocus


genomic variation. N. Engl. J. Med. 376, 21–31 (2017). Multilocus genomic diagnoses
occur in nearly 5% of solved exome cases, underscoring the complexity and need
for comprehensive interpretation in rare disease phenotypes.

9. Turro, E. et al. Whole-genome sequencing of patients with rare diseases in a national


health system. Nature 583, 96–102 (2020).

[Link] 4/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

10. Bamshad, M. J. et al. The Centers for Mendelian Genomics: a new large-scale
initiative to identify the genes underlying rare Mendelian conditions. Am. J. Med.
Genet. A 158A, 1523–1525 (2012).

11. Posey, J. E. et al. Insights into genetics, human biology and disease gleaned from
family based genomic studies. Genet. Med. 21, 798–812 (2019).

12. Chong, J. X. et al. The genetic basis of Mendelian phenotypes: discoveries,


challenges, and opportunities. Am. J. Hum. Genet. 97, 199–215 (2015).

13. Surl, D. et al. Clinician-driven reanalysis of exome sequencing data from patients
with inherited retinal diseases. JAMA Netw. Open 7, e2414198 (2024).

14. Seaby, E. G. et al. A gene pathogenicity tool ‘GenePy’ identifies missed biallelic
diagnoses in the 100,000 Genomes Project. Genet. Med. 26, 101073 (2024).

15. Liu, P. et al. Reanalysis of clinical exome sequencing data. N. Engl. J. Med. 380, 2478–
2480 (2019). Systematic reanalysis of clinical exome data substantially
increased diagnostic yield and was driven by the newest novel disease gene
discoveries.

16. Berger, S. I. et al. Increased diagnostic yield from negative whole genome-slice
panels using automated reanalysis. Clin. Genet. 104, 377–383 (2023).

[Link] 5/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

17. Wenger, A. M., Guturu, H., Bernstein, J. A. & Bejerano, G. Systematic reanalysis of
clinical exome data yields additional diagnoses: implications for providers. Genet.
Med. 19, 209–214 (2017).

18. Weisburd, B. et al. Diagnosing missed cases of spinal muscular atrophy in genome,
exome, and panel sequencing data sets. Genet. Med. 27, 101336 (2025).

19. Guo, M. H. et al. Inferring compound heterozygosity from large-scale exome


sequencing data. Nat. Genet. 56, 152–161 (2024). This work presents a method to
infer phasing of rare variant pairs in short-read exomes using gnomAD,
enabling improved diagnosis of recessive Mendelian conditions.

20. Gudmundsson, S. et al. Variant interpretation using population databases: lessons


from gnomAD. Hum. Mutat. 43, 1012–1030 (2022).

21. Mitani, T. et al. High prevalence of multilocus pathogenic variation in


neurodevelopmental disorders in the Turkish population. Am. J. Hum. Genet. 108,
1981–2005 (2021).

22. Lemire, G. et al. Exome copy number variant detection, analysis, and classification
in a large cohort of families with undiagnosed rare genetic disease. Am. J. Hum.
Genet. 111, 863–876 (2024).

23. Du, H. et al. HMZDupFinder: a robust computational approach for detecting


intragenic homozygous duplications from exome sequencing data. Nucleic Acids
Res. 52, e18 (2024).

[Link] 6/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

24. Babadi, M. et al. GATK-gCNV enables the discovery of rare copy number variants
from exome sequencing data. Nat. Genet. 55, 1589–1597 (2023).

25. Du, H. et al. VizCNV: An integrated platform for concurrent phased BAF and CNV
analysis with trio genome sequencing data. Preprint at bioRxiv
[Link] (2024).

26. Wojcik, M. H. et al. Beyond the exome: what’s next in diagnostic testing for
Mendelian conditions. Am. J. Hum. Genet. 110, 1229–1248 (2023). Offers a roadmap
for diagnostic escalation beyond exome sequencing, including RNA-seq,
genome sequencing, and long-read technologies, essential for solving
unsolved Mendelian cases.

27. Sudmant, P. H. et al. An integrated map of structural variation in 2,504 human


genomes. Nature 526, 75–81 (2015).

28. Collins, R. L. et al. A structural variation reference for medical and population
genetics. Nature 581, 444–451 (2020).

29. Byrska-Bishop, M. et al. High-coverage whole-genome sequencing of the expanded


1000 Genomes Project cohort including 602 trios. Cell 185, 3426–3440 (2022).

30. Collins, R. L. et al. Defining the diverse spectrum of inversions, complex structural
variation, and chromothripsis in the morbid human genome. Genome Biol. 18, 36
(2017).

[Link] 7/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

31. Wojcik, M. H. et al. Genome sequencing for diagnosing rare diseases. N. Engl. J. Med.
390, 1985–1997 (2024). Genome sequencing provided unique diagnoses in 8% of
previously unsolved cases.

32. Karczewski, K. J. et al. The mutational constraint spectrum quantified from


variation in 141,456 humans. Nature 581, 434–443 (2020).

33. Vitsios, D., Dhindsa, R. S., Middleton, L., Gussow, A. B. & Petrovski, S. Prioritizing
non-coding regions based on human genomic constraint and sequence context
with deep learning. Nat. Commun. 12, 1504 (2021).

34. Chen, S. et al. A genomic mutational constraint map using variation in 76,156
human genomes. Nature 625, 92–100 (2024).

35. Zhang, X. et al. Genetic constraint at single amino acid resolution in protein
domains improves missense variant prioritisation and gene discovery. Genome
Med. 16, 88 (2024).

36. Chao, K. R. et al. The landscape of regional missense mutational intolerance


quantified from 125,748 exomes. Preprint at bioRxiv
[Link] (2024).

37. Saad, A. K. et al. Biallelic in-frame deletion in TRAPPC4 in a family with


developmental delay and cerebellar atrophy. Brain J. Neurol. 143, e83 (2020).

[Link] 8/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

38. Dawood, M. et al. A biallelic frameshift indel in PPP1R35 as a cause of primary


microcephaly. Am. J. Med. Genet. A 191, 794–804 (2023).

39. Dardas, Z. et al. NODAL variants are associated with a continuum of laterality
defects from simple D-transposition of the great arteries to heterotaxy. Genome
Med. 16, 53 (2024).

40. Miller, D. E. et al. Targeted long-read sequencing identifies a retrotransposon


insertion as a cause of altered GNAS Exon A/B methylation in a family with
autosomal dominant pseudohypoparathyroidism type 1b (PHP1B). J. Bone Miner.
Res. 37, 1711–1719 (2022).

41. Mori, T. et al. CFAP47 is implicated in X-linked polycystic kidney disease. Kidney Int.
Rep. 9, 3580–3591 (2024).

42. Bruels, C. C. et al. Diagnostic capabilities of nanopore long-read sequencing in


muscular dystrophy. Ann. Clin. Transl. Neurol. 9, 1302–1309 (2022).

43. Chen, X. et al. Genome-wide profiling of highly similar paralogous genes using HiFi
sequencing. Nat. Commun. 16, 2340 (2025).

44. Negi, S. et al. Advancing long-read nanopore genome assembly and accurate
variant calling for rare disease detection. Am. J. Hum. Genet. 112, 428–449 (2025).

45. Mahmoud, M. et al. Closing the gap: solving complex medically relevant genes at
scale. Preprint at medRxiv [Link] (2024).

[Link] 9/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

46. Dias, K.-R. et al. Narrowing the diagnostic gap: genomes, episignatures, long-read
sequencing, and health economic analyses in an exome-negative intellectual
disability cohort. Genet. Med. 26, 101076 (2024).

47. Bilgrav Saether, K. et al. Leveraging the T2T assembly to resolve rare and
pathogenic inversions in reference genome gaps. Genome Res. 34, 1785–1797
(2024).

48. Fu, Y. et al. MethPhaser: methylation-based long-read haplotype phasing of human


genomes. Nat. Commun. 15, 5327 (2024).

49. LaFlamme, C. W. et al. Diagnostic utility of DNA methylation analysis in genetically


unsolved pediatric epilepsies and CHD2 episignature refinement. Nat. Commun. 15,
6524 (2024).

50. Zheng, X. et al. STIX: long-reads based accurate structural variation annotation at
population scale. Preprint at bioRxiv [Link]
(2024).

51. Smolka, M. et al. Detection of mosaic and population-level structural variants with
Sniffles2. Nat. Biotechnol. 42, 1571–1580 (2024).

52. Sedlazeck, F. J. et al. Accurate detection of complex structural variations using


single-molecule sequencing. Nat. Methods 15, 461–468 (2018).

53. Stergachis, A. B., Debo, B. M., Haugen, E., Churchman, L. S. & Stamatoyannopoulos,
J. A. Single-molecule regulatory architectures captured by chromatin fiber
[Link] 10/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

sequencing. Science 368, 1449–1454 (2020).

54. Jha, A. et al. DNA-m6A calling and integrated long-read epigenetic and genetic
analysis with fibertools. Genome Res. 34, 1976–1986 (2024).

55. Vollger, M. R. et al. Synchronized long-read genome, methylome, epigenome and


transcriptome profiling resolve a Mendelian condition. Nat. Genet. 57, 469–479
(2025). By integrating long-read genome, methylome, epigenome, and
transcriptome data, this study demonstrates how complex, multi-mechanism
rare diseases can be mechanistically resolved in a single assay.

56. Grasberger, H. et al. STR mutations on chromosome 15q cause thyrotropin


resistance by activating a primate-specific enhancer of MIR7-2/MIR1179. Nat. Genet.
56, 877–888 (2024).

57. Carvalho, C. M. B. et al. Interchromosomal template-switching as a novel molecular


mechanism for imprinting perturbations associated with Temple syndrome.
Genome Med. 11, 25 (2019).

58. Jensen, T. D. et al. Integration of transcriptomics and long-read genomics


prioritizes structural variants in rare disease. Genome Res. 35, 914–928 (2025).

59. Pais, L. S. et al. seqr: a web-based analysis and collaboration tool for rare disease
genomics. Hum. Mutat. 43, 698–707 (2022).

[Link] 11/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

60. Lansdon, L. A. et al. Factors affecting migration to GRCh38 in laboratories


performing clinical next-generation sequencing. J. Mol. Diagn. 23, 651–657 (2021).

61. Li, H. et al. Exome variant discrepancies due to reference-genome differences. Am. J.
Hum. Genet. 108, 1239–1250 (2021). Discrepancies between GRCh37 and GRCh38
reference genomes affect variant calling in around 200 genes including
Mendelian disease genes.

62. Ungar, R. A. et al. Impact of genome build on RNA-seq interpretation and


diagnostics. Am. J. Hum. Genet. 111, 1282–1300 (2024). Genome build choice
significantly alters RNA-seq interpretation across >2800 genes, directly
impacting transcriptomics-guided rare disease diagnostics.

63. Behera, S. et al. FixItFelix: improving genomic analysis by fixing reference errors.
Genome Biol. 24, 31 (2023).

64. Wagner, J. et al. Curated variation benchmarks for challenging medically relevant
autosomal genes. Nat. Biotechnol. 40, 672–680 (2022).

65. Liao, W.-W. et al. A draft human pangenome reference. Nature 617, 312–324 (2023).

[Link] 12/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

66. Behera, S. et al. Comprehensive genome analysis and variant detection at scale
using DRAGEN. Nat. Biotechnol. 43, 1177–1191 (2025). The DRAGEN pipeline is a
high-accuracy, scalable variant detection method that leverages multigenome
mapping and machine learning to identify all major variant types—including
single-nucleotide variants, copy-number variants, structural variants and
short tandem repeats—across diverse populations.

67. Chin, C.-S. et al. A pan-genome approach to decipher variants in the highly complex
tandem repeat of LPA. Preprint at bioRxiv
[Link] (2022).

68. Samocha, K. E. et al. A framework for the interpretation of de novo mutation in


human disease. Nat. Genet. 46, 944–950 (2014). A framework to evaluate de novo
mutations improves gene discovery in rare diseases by distinguishing
pathogenic mutations from background variation.

69. Lupski, J. R., Belmont, J. W., Boerwinkle, E. & Gibbs, R. A. Clan genomics and the
complex architecture of human disease. Cell 147, 32–43 (2011). Describes the
foundational framework for emphasizing the role of recent, rare, and private
variants in disease risk and highlights the importance of family-centric rare
disease analysis.

70. Teran, N. A. et al. Nonsense-mediated decay is highly stable across individuals and
tissues. Am. J. Hum. Genet. 108, 1401–1408 (2021).

[Link] 13/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

71. Coban-Akdemir, Z. et al. Identifying genes whose mutant transcripts cause


dominant disease traits by potential gain-of-function alleles. Am. J. Hum. Genet. 103,
171–187 (2018).

72. Valencia, A. M. et al. Landscape of mSWI/SNF chromatin remodeling complex


perturbations in neurodevelopmental disorders. Nat. Genet. 55, 1400–1412 (2023).

73. Paine, I. et al. Paralog studies augment gene discovery: DDX and DHX genes. Am. J.
Hum. Genet. 105, 302–316 (2019).

74. Gillentine, M. A. et al. Rare deleterious mutations of HNRNP genes result in shared
neurodevelopmental disorders. Genome Med. 13, 63 (2021).

75. Amberger, J. S., Bocchini, C. A., Scott, A. F. & Hamosh, A. [Link]: leveraging
knowledge across phenotype-gene relationships. Nucleic Acids Res. 47, D1038–
D1043 (2019).

76. DiStefano, M. T. et al. The gene curation coalition: a global effort to harmonize
gene-disease evidence resources. Genet. Med. 24, 1732–1742 (2022).

77. Ochoa, S. et al. A deep intronic splice-altering AIRE variant causes APECED
syndrome through antisense oligonucleotide-targetable pseudoexon inclusion. Sci.
Transl. Med. 16, eadk0845 (2024).

78. Wu, N. et al. TBX6 null variants and a common hypomorphic allele in congenital
scoliosis. N. Engl. J. Med. 372, 341–350 (2015).

[Link] 14/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

79. Lord, J. et al. Non-coding variants are a rare cause of recessive developmental
disorders in trans with coding variants. Genet. Med. 26, 101249 (2024).

80. Mao, K. et al. FOXI3 pathogenic variants cause one form of craniofacial
microsomia. Nat. Commun. 14, 2026 (2023).

81. Ganesh, V. S. et al. Neurodevelopmental disorder caused by deletion of CHASERR, a


lncRNA gene. N. Engl. J. Med. 391, 1511–1518 (2024). Discovery of a
neurodevelopmental disorder caused by CHASERR long non-coding deletion
reveals regulatory non-coding elements as critical contributors to rare disease
pathogenesis.

82. Greene, D. et al. Mutations in the U4 snRNA gene RNU4-2 cause one of the most
prevalent monogenic neurodevelopmental disorders. Nat. Med. 30, 2165–2169
(2024).

83. Chen, Y. et al. De novo variants in the RNU4-2 snRNA cause a frequent
neurodevelopmental syndrome. Nature 632, 832–840 (2024).

84. Greene, D. et al. Mutations in the small nuclear RNA gene RNU2-2 cause a severe
neurodevelopmental disorder with prominent epilepsy. Nat. Genet. 57, 1367–1373
(2025).

85. Nava, C. et al. Dominant variants in major spliceosome U4 and U5 small nuclear
RNA genes cause neurodevelopmental disorders through splicing disruption. Nat.
Genet. 57, 1374–1388 (2025).

[Link] 15/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

86. Bozkurt-Yozgatli, T. et al. Multilocus pathogenic variants contribute to intrafamilial


clinical heterogeneity: a retrospective study of sibling pairs with
neurodevelopmental disorders. BMC Med. Genom. 17, 85 (2024).

87. Liu, P. et al. An organismal CNV mutator phenotype restricted to early human
development. Cell 168, 830–842 (2017).

88. Du, H. et al. The multiple de novo copy number variant (MdnCNV) phenomenon
presents with peri-zygotic DNA mutational signatures and multilocus pathogenic
variation. Genome Med. 14, 122 (2022).

89. Logsdon, G. A. et al. Complex genetic variation in nearly complete human


genomes. Nature 644, 430–441 (2025).

90. Ebert, P. et al. Haplotype-resolved diverse human genomes and integrated analysis
of structural variation. Science 372, eabf7117 (2021).

91. Grochowski, C. M. et al. Inverted triplications formed by iterative template switches


generate structural variant diversity at genomic disorder loci. Cell Genom. 4,
100590 (2024).

92. Dardas, Z. et al. Genomic balancing act: deciphering DNA rearrangements in the
complex chromosomal aberration involving 5p15.2, 2q31.1, and 18q21.32. Eur. J.
Hum. Genet. 33, 231–238 (2025).

93. Pehlivan, D. et al. Structural variant allelic heterogeneity in MECP2 duplication


syndrome provides insight into clinical severity and variability of disease
[Link] 16/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

expression. Genome Med. 16, 146 (2024).

94. Jakubosky, D. et al. Discovery and quality analysis of a comprehensive set of


structural variants and short tandem repeats. Nat. Commun. 11, 2928 (2020).

95. Jakubosky, D. et al. Properties of structural variants and short tandem repeats
associated with gene expression and complex traits. Nat. Commun. 11, 2927 (2020).

96. Dolzhenko, E. et al. Characterization and visualization of tandem repeats at


genome scale. Nat. Biotechnol. 42, 1606–1614 (2024).

97. English, A. C. et al. Analysis and benchmarking of small and large genomic variants
across tandem repeats. Nat. Biotechnol. 43, 431–442 (2025).

98. Dolzhenko, E. et al. REViewer: haplotype-resolved visualization of read alignments


in and around tandem repeats. Genome Med. 14, 84 (2022).

99. Behera, S. et al. Identification of allele-specific KIV-2 repeats and impact on Lp(a)
measurements for cardiovascular disease risk. BMC Med. Genom. 17, 255 (2024).

100. Weisburd, B., Tiao, G. & Rehm, H. L. Insights from a genome-wide truth set of
tandem repeat variation. Preprint at bioRxiv
[Link] (2023).

101. Cui, Y. et al. A genome-wide spectrum of tandem repeat expansions in 338,963


humans. Cell 187, 2336–2341 (2024). This study establishes a biobank-scale,

[Link] 17/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

population reference of tandem repeat expansions across ancestries from


short-read genome sequencing.

102. Weisburd, B. et al. Defining a tandem repeat catalog and variation clusters for
genome-wide analyses and population databases. Preprint at bioRxiv
[Link] (2024).

103. Wang, Q. et al. Landscape of multi-nucleotide variants in 125,748 human exomes


and 15,708 genomes. Nat. Commun. 11, 2539 (2020).

104. Srinivasan, S. et al. Misannotated multi-nucleotide variants in public cancer


genomics datasets lead to inaccurate mutation calls with significant implications.
Cancer Res. 81, 282–288 (2021).

105. Campbell, I. M. et al. Multiallelic positions in the human genome: challenges for
genetic analyses. Hum. Mutat. 37, 231–234 (2016).

106. Lindeboom, R. G. H., Vermeulen, M., Lehner, B. & Supek, F. The impact of
nonsense-mediated mRNA decay on genetic disease, gene editing and cancer
immunotherapy. Nat. Genet. 51, 1645–1651 (2019).

107. Lindeboom, R. G. H., Supek, F. & Lehner, B. The rules and impact of nonsense-
mediated mRNA decay in human cancers. Nat. Genet. 48, 1112–1118 (2016).

[Link] 18/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

108. Torene, R. I. et al. Systematic analysis of variants escaping nonsense-mediated


decay uncovers candidate Mendelian diseases. Am. J. Hum. Genet. 111, 70–81
(2024).

109. Potter, A. S. et al. Rare variant in MRC2 associated with familial supraventricular
tachycardia and Wolff-Parkinson-White syndrome. Circ. Genomic Precis. Med. 17,
e004614 (2024).

110. Gudmundsson, S. et al. Exploring penetrance of clinically relevant variants in over


800,000 humans from the Genome Aggregation Database. Preprint at bioRxiv
[Link] (2024).

111. Rehm, H. L. et al. The landscape of reported VUS in multi-gene panel and genomic
testing: time for a change. Genet. Med. 25, 100947 (2023). This multi-laboratory
analysis reveals the high prevalence and clinical burden of VUSs in genetic
testing from panel testing and advocates for refined reporting practices to
reduce VUSs.

112. Dawood, M. et al. Using multiplexed functional data to reduce variant


classification inequities in underrepresented populations. Genome Med. 16, 143
(2024). This study defines variant classification disparities in several biobanks
and demonstrates how multiplexed functional data can reduce variant
classification disparities across ancestries, offering a scalable strategy to
make genomic medicine more equitable.

113. Young, J. L. et al. Beyond race: recruitment of diverse participants in clinical


genomics research for rare disease. Front. Genet. 13, 949422 (2022).

[Link] 19/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

114. Wojcik, M. H. et al. Rare diseases, common barriers: disparities in pediatric clinical
genetics outcomes. Pediatr. Res. 93, 110–117 (2023).

115. Serrano, J. G. et al. Advancing understanding of inequities in rare disease


genomics. Clin. Ther. 45, 745–753 (2023).

116. D’Angelo, C. S. et al. Barriers and considerations for diagnosing rare diseases in
indigenous populations. Front. Pediatr. 8, 579924 (2020).

117. Savage, S. K. et al. Using a chat-based informed consent tool in large-scale genomic
research. J. Am. Med. Inform. Assoc. 31, 472–478 (2024). A chat-based consent tool
successfully scaled participant enrollment for large rare disease genomics
studies, reducing staff burden while maintaining participant understanding.

118. Abou Tayoun, A. N. & Rehm, H. L. Genetic variation in the Middle East—an
opportunity to advance the human genetics field. Genome Med. 12, 116 (2020).

119. AlAbdi, L. et al. Diagnostic implications of pitfalls in causal variant identification


based on 4577 molecularly characterized families. Nat. Commun. 14, 5269 (2023).

120. AlAbdi, L. et al. Beyond the exome: utility of long-read whole genome sequencing
in exome-negative autosomal recessive diseases. Genome Med. 15, 114 (2023).

121. AlAbdi, L. et al. Arab founder variants: Contributions to clinical genomics and
precision medicine. Med 6, 100528 (2025).

[Link] 20/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

122. Sulem, P. et al. Identification of a large set of rare complete human knockouts. Nat.
Genet. 47, 448–452 (2015).

123. Oddsson, A. et al. Deficit of homozygosity among 1.52 million individuals and
genetic causes of recessive lethality. Nat. Commun. 14, 3453 (2023).

124. Wenger, T. L. et al. SeqFirst: building equity access to a precise genetic diagnosis in
critically ill newborns. Am. J. Hum. Genet. 112, 508–522 (2025).

125. Stark, Z. et al. A call to action to scale up research and clinical genomic data
sharing. Nat. Rev. Genet. 26, 141–147 (2025).

126. Rehm, H. L. Time to make rare disease diagnosis accessible to all. Nat. Med. 28,
241–242 (2022).

127. Wilkinson, M. D. et al. The FAIR guiding principles for scientific data management
and stewardship. Sci. Data 3, 160018 (2016).

128. Bamshad, M. J., Nickerson, D. A. & Chong, J. X. Mendelian gene discovery: fast and
furious with no end in sight. Am. J. Hum. Genet. 105, 448–455 (2019).

129. Wagner, A. H. et al. The GA4GH variation representation specification: a


computational framework for variation representation and federated
identification. Cell Genom. 1, 100027 (2021).

[Link] 21/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

130. Pawliczek, P. et al. ClinGen allele registry links information about genetic variants.
Hum. Mutat. 39, 1690–1701 (2018).

131. Köhler, S. et al. The Human Phenotype Ontology in 2021. Nucleic Acids Res. 49,
D1207–D1217 (2021).

132. Stegmann, J. D. et al. Bi-allelic variants in CELSR3 are implicated in central nervous
system and urinary tract anomalies. npj Genom. Med. 9, 18 (2024).

133. Herman, I. et al. Quantitative dissection of multilocus pathogenic variation in an


Egyptian infant with severe neurodevelopmental disorder resulting from multiple
molecular diagnoses. Am. J. Med. Genet. A 188, 735–750 (2022).

134. Calame, D. G. et al. Monoallelic variation in DHX9, the gene encoding the DExH-
box helicase DHX9, underlies neurodevelopment disorders and Charcot-Marie-
Tooth disease. Am. J. Hum. Genet. 110, 1394–1413 (2023).

135. Jolly, A. et al. Rare variant enrichment analysis supports GREB1L as a contributory
driver gene in the etiology of Mayer-Rokitansky-Küster-Hauser syndrome. HGG
Adv. 4, 100188 (2023).

136. Lima, A. R. et al. Phenotypic and mutational spectrum of ROR2-related Robinow


syndrome. Hum. Mutat. 43, 900–918 (2022).

[Link] 22/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

137. Zhang, C. et al. Novel pathogenic variants and quantitative phenotypic analyses of
Robinow syndrome: WNT signaling perturbation and phenotypic variability. HGG
Adv. 3, 100074 (2022).

138. Garcia, B. T. et al. Improving automated deep phenotyping through large


language models using retrieval-augmented generation. Genome Med. 17, 91
(2025).

139. Gustafson, J. A. et al. High-coverage nanopore sequencing of samples from the


1000 Genomes Project to build a comprehensive catalog of human genetic
variation. Genome Res. 34, 2061–2073 (2024). High-coverage long-read ONT
sequencing of 1000 Genomes Project samples enables improved detection of
structural variants and epigenetic changes.

140. Harrison, P. W. et al. Ensembl 2024. Nucleic Acids Res. 52, D891–D899 (2024).

141. Liu, X., Li, C., Mou, C., Dong, Y. & Tu, Y. dbNSFP v4: a comprehensive database of
transcript-specific functional predictions and annotations for human
nonsynonymous and splice-site SNVs. Genome Med. 12, 103 (2020).

142. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic
variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164
(2010).

143. Pagel, K. A. et al. Integrated informatics analysis of cancer-related variants. JCO


Clin. Cancer Inform. 4, 310–317 (2020).

[Link] 23/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

144. Rodrigues, E. D. S. et al. Variant-level matching for diagnosis and discovery:


challenges and opportunities. Hum. Mutat. 43, 782–790 (2022).

145. Seaby, E. G. et al. A panel-agnostic strategy ‘HiPPo’ improves diagnostic efficiency


in the UK Genomic Medicine Service. Healthcare 11, 3179 (2023).

146. Chong, J. X. et al. Considerations for reporting variants in novel candidate genes
identified during clinical genomic testing. Genet. Med. 26, 101199 (2024).

147. Rai, A. et al. Genomic rare variant mechanisms for congenital cardiac laterality
defect: a digenic model approach. Am. J. Hum. Genet. 112, 1664–1680 (2025).

148. Töpf, A. et al. Digenic inheritance involving a muscle-specific protein kinase and
the giant titin protein causes a skeletal muscle myopathy. Nat. Genet. 56, 395–407
(2024).

149. Gifford, C. A. et al. Oligogenic inheritance of a human heart disease involving a


genetic modifier. Science 364, 865–870 (2019).

150. Arriaga, T. M. et al. Transcriptome-wide outlier approach identifies individuals


with minor spliceopathies. Am. J. Hum. Genet. 112, 2458–2475 (2025).

Acknowledgements

We thank all of the patient participants and their families; and the expansive set of
collaborators, including clinical providers, analysts and rare disease researchers.
Support for title page creation and format was provided by AuthorArranger, a tool
developed at the National Cancer Institute. This work was supported by the NIH NHGRI
[Link] 24/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

GREGoR Consortium (U01HG011758, U01HG011755, U01HG011762, U01HG011745,


U01HG011744, U24HG011746).
Author information

Authors and Affiliations


Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
Moez Dawood, James R. Lupski, Richard A. Gibbs, Fritz J. Sedlazeck, Sairam Behera, Harsha Doddapaneni, Jianhong Hu, Shalini N. Jhangiani, Medhat
Mahmoud, Donna Muzny, Piyush Panchal, Aniko Sabo, Evette Scott, Kimberly Walker, Bo Yuan & Xinchang Zheng

Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
Moez Dawood, James R. Lupski, Richard A. Gibbs, Fritz J. Sedlazeck, Jennifer E. Posey, Hatoon Al Ali, Elizabeth G. Atkinson, Shaghayegh T. Beheshti, Zeynep H.
Coban-Akdemir, Karen J. Coveler, Zain Dardas, Haowei Du, Ruizhi Duan, Jawid Fatih, Brandon Garcia, Nikhita Gogate, Christopher M. Grochowski, Minal
Jamsandekar, Angad Jolly, Parneet Kaur, Ahmed K. Saad, Richard A. Lewis, Yidan Li, Pengfei Liu, Chloe Munderloh, Donna Muzny, Shruti Pande, Edgar Andres
Rivera-Munoz, Vernon Reid Sutton, Lauren Westerfield & Bo Yuan

Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
Moez Dawood

Department of Biostatistics, University of Washington, Seattle, WA, USA


Ben Heavner, Marsha M. Wheeler, Ali Shojaie, Susanne May, Emily Bonkowski, Sarah Conner, Matthew P. Conomos, Stephanie M. Gogarten, Sarah C.
Nelson, Sheryl Payne, Jaime Prosser, Guanghao Qi, Adrienne M. Stilp, Catherine C. Tong & Quenna Wong

Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA


Rachel A. Ungar, Laurens Wiel, Casey A. Gifford, Stephen B. Montgomery, Taylor M. Arriaga, Euan A. Ashley, Denver Bradley, Clarisa Chavez Martinez, Ivy
Evergreen, William Greenleaf, Tanner D. Jensen, Christopher Jin, Aimee Juan, Jessica Kain, Anshul Kundaje, Samuel Lancaster, Michael P. Snyder, Suchitra
Sudarshan & Frank Wong

Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA


Rachel A. Ungar, Laurens Wiel, Stephen B. Montgomery, Page Goddard, Lauren Meador, Alexander Miller, Thuy-mi P. Nguyen, Jonathan Nguyen, Jeren D.
Olsen, Evin M. Padhi, Paul Petrowski, Astaria D. Podesta, Wanqiong Qiao, Stuart A. Scott, Kevin S. Smith, Brigitte Stark, Ziming Weng & Yao Yang

Stanford Center for Biomedical Ethics, School of Medicine, Stanford University, Stanford, CA, USA
Rachel A. Ungar

Institute for Clinical and Translational Science, University of California, Irvine, Irvine, CA, USA
Jonathan LoTempio, Emmanuèle C. Délot, Eric Vilain, Light Auriga, Charles Hadley King & Georgia Pitsava

Division of Cardiovascular Medicine, School of Medicine, Stanford University, Stanford, CA, USA
Laurens Wiel, Matthew T. Wheeler, Euan A. Ashley, Themistocles Assimes, Jennefer Carter, Sara Emami, John Gorzynski, Rodrigo Guarischi-Sousa, David
Jimenez-Morales, Shruti Marwaha, Hector Rodrigo Mendez, Thomas Quertermous, Chloe M. Reuter, Raquel L. Summers, Philip Tsao, Isabella Voutos, Juliana M.
Walrod, Jiye Yu & Jimmy Zhen

Division of Genetics and Metabolism, Children’s National Hospital, Washington, DC, USA
Seth Berger & Andrea J. Cohen

Center for Precision Medicine and Genomics Research, Children’s National Hospital, Washington, DC, USA
Seth Berger, Leandros Boukas & Arthur Ko

Department of Genomics and Precision Medicine, George Washington University, Washington, DC, USA
Seth Berger

[Link] 25/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

Department of Pediatrics, Division of Medical Genetics, Stanford University, Stanford, CA, USA
Jonathan A. Bernstein, Gill Bejerano, Devon Bonner, Caitlin Harrington, Laura Keehan, Dena R. Matalon, Elizabeth Porter, Oriane Rubio & Christina G. Tise

Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, USA
Jessica X. Chong, Michael J. Bamshad, Kailyn Anderson, Kati J. Buckingham, William W. Gordon, Jonas Gustafson, Colby T. Marvin, Olivia M.
Sommerland & Miranda P. Zalusky

Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
Jessica X. Chong, Danny E. Miller, Michael J. Bamshad, Sabrina Best, Silvia Casadei, Lea M. Starita & Andrew B. Stergachis

Department of Genome Sciences, University of Washington, Seattle, WA, USA


Evan E. Eichler, Chia-Lin Wei, Danny E. Miller, Peter Anderson, Sabrina Best, Silvia Casadei, Yong-Han Hank Cheng, Colleen P. Davis, Sophia B. Gibson, William T.
Harvey, Gail P. Jarvik, F. Kumara Mastrorosa, Sean R. McGee, Karynne Patterson, Matthew Richardson, Joshua D. Smith, Lea M. Starita, Andrew B.
Stergachis, Elliott G. Swanson, Jeffrey Weiss, Qian Yi & Christina Zakarian

Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA


Evan E. Eichler

Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA


James R. Lupski, Richard A. Lewis & Sebastian Ochoa

Department of Statistics, University of Washington, Seattle, WA, USA


Ali Shojaie

Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
Michael E. Talkowski, Heidi L. Rehm, Kaitlin E. Samocha, Anne O’Donnell-Luria, Christina Austin-Tse, Philip M. Boone, Harrison Brand, Celine de Esch, Michael
Duyzend, Arthur S. Lee, Monica Salani, Kaitlin Samocha, Alba Sanchis-Juan, Jui-Cheng Tai & Rachita Yadav

Program in Medical and Population Genetics, Broad Center for Mendelian Genomics, Broad Institute of MIT and Harvard, Cambridge,
MA, USA
Michael E. Talkowski, Heidi L. Rehm, Kaitlin E. Samocha, Anne O’Donnell-Luria, Siwaar Abouhala, K. D. Ahlquist, Mutaz Amin, Christina Austin-Tse, Samantha M.
Baxter, Benjamin Blankenmeister, Philip M. Boone, Harrison Brand, Colleen Carlston, Celine de Esch, Stephanie DiTroia, Michael Duyzend, Vijay Ganesh, Kiran
Garimella, Carmen Glaze, Emily Groopman, Sanna Gudmundsson, Stacey Hall, Yongqing Huang, Julia Klugherz, Katie Larsson, Arthur S. Lee, Gabrielle
Lemire, Jialan Ma, Daniel MacArthur, Brian Mangilog, Daniel Marten, Eva Martinez, Olfa Messaoud, Chloe Mighton, Mariana Moyses, Ashana Neale, Emily
O’Heir, Melanie C. O’Leary, Ikeoluwa Osei-Owusu, Lynn Pais, Alicia Pham, Lindsay Romo, Kathryn Russell, Monica Salani, Kaitlin Samocha, Alba Sanchis-
Juan, Jillian Serrano, Gulalai Shah, Moriel Singer-Berk, Mugdha Singh, Hana Snow, Kayla Socarras, Sarah L. Stenton, Jui-Cheng Tai, Grace VanNoy, Ben
Weisburd, Michael Wilson, Monica Wojcik, Isaac Wong & Rachita Yadav

Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
Michael E. Talkowski, Harrison Brand, Arthur S. Lee, Alba Sanchis-Juan, Jui-Cheng Tai & Rachita Yadav

Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
Michael E. Talkowski

Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA, USA
Michael E. Talkowski

Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
Alex H. Wagner

Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
Alex H. Wagner

Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA

[Link] 26/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature
Alex H. Wagner

Office of Genomic Data Science, National Human Genome Research Institute, Bethesda, MD, USA
Christopher Wellington

Pacific Northwest Research Institute, Seattle, WA, USA


Aimée Dudley & Claudia M. B. Carvalho

Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA


Casey A. Gifford & Matthew B. Neu

Basic Science and Engineering (BASE) Initiative, Betty Irene Moore Children’s Heart Center, Stanford Medicine Children’s Health,
Stanford, CA, USA
Casey A. Gifford

Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
Casey A. Gifford

Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA
Danny E. Miller

Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
Danny E. Miller

Department of Computer Science, Rice University, Houston, TX, USA


Fritz J. Sedlazeck

Department of Pediatrics, Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
Anne O’Donnell-Luria, Philip M. Boone, Colleen Carlston, Michael Duyzend, Daniel Marten, Mugdha Singh, Sarah L. Stenton & Monica Wojcik

Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
Jennifer E. Posey

Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
Jennifer E. Posey

Division of Genome Sciences, National Human Genome Research Institute, Bethesda, MD, USA
Lisa H. Chadwick, Sara Currin & Gabrielle C. Villard

Department of Pediatrics, Division of Genetic Medicine, Seattle Children’s Hospital, Seattle, WA, USA
Michael J. Bamshad

Department of Biomedical Data Science, Stanford University, Stanford, CA, USA


Stephen B. Montgomery, Emily Alsentzer & Euan A. Ashley

Illumina Artificial Intelligence Laboratory, Illumina, Foster City, CA, USA


Aashish Adhikari

Department of Ophthalmology, Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
Kinga M. Bujakowska

Medical Genomics Research, Illumina, San Diego, CA, USA


Ali Crawford

[Link] 27/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
Aimée Dudley & Jonas Gustafson

Ambry Genetics, Aliso Viejo, CA, USA


Kelly D. Farwell Hagman, Zain Dardas, Sami Belhadj, Krista Bluske, John Harting, Yun-Hua Hsiao, Rachid Karam & Bojan Losic

Department of Medicine, University of Chicago, Chicago, IL, USA


Yang I. Li

Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
Jill E. Moore

Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
Aaron R. Quinlan

Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
Bo Xia

Department of Neurosurgery, Baylor Research Institute, Baylor College of Medicine, Temple, TX, USA
S. Stephen Yi

Department of Medicine, School of Medicine—Temple, Baylor College of Medicine, Temple, TX, USA
S. Stephen Yi

Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The
University of Texas Health Science Center at Houston, Houston, TX, USA
Eric Boerwinkle, Tugce Bozkurt-Yozgatli, Zeynep H. Coban-Akdemir, Iman Egab, Archana Rai & Jiaoyang Xu

Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
Tugce Bozkurt-Yozgatli

Department of Pediatrics, Section of Pediatric Neurology and Developmental Neurosciences, Baylor College of Medicine, Houston,
TX, USA
Daniel G. Calame, Mira Gandhi, Jesse M. Levine & Davut Pehlivan

Department of Pediatrics, Division of Immunology, Allergy, and Retrovirology, Baylor College of Medicine, Houston, TX, USA
Ivan Chinn

Department of Neurology, The University of Texas Dell Medical School, Austin, TX, USA
Angad Jolly

Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA


Richard A. Lewis

Department of Medicine, Baylor College of Medicine, Houston, TX, USA


Richard A. Lewis

Department of Pediatrics, College of Medicine, Kuwait University, Safat, Kuwait


Dana Marafi

Section of Child Neurology, Department of Pediatrics, Adan Hospital, Ministry of Health, Hadiya, Kuwait
Dana Marafi

Kuwait Medical Genetics Centre, Ministry of Health, Sulaibikhat, Kuwait

[Link] 28/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature
Dana Marafi

Department of Pediatrics, Jichi Medical University, Shimotsuke, Tochigi, Japan


Tadahiro Mitani

Texas Children’s Hospital, Houston, TX, USA


Davut Pehlivan

Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
Davut Pehlivan

Baylor Genetics, Houston, TX, USA


Bo Yuan

Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA


Vijay Ganesh

Centre for Population Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
Daniel MacArthur

Centre for Population Genomics, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
Daniel MacArthur

Department of Pediatrics, Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA, USA
Monica Wojcik

Department of Computer Science, Stanford University, Stanford, CA, USA


Gill Bejerano, Ziwei Chen, Anshul Kundaje, Soumya Kundu & Riya Sinha

Department of Developmental Biology, School of Medicine, Stanford University, Stanford, CA, USA
Gill Bejerano

Institute of Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
Salil Deshpande

Department of Biomedical Data Science, School of Medicine, Stanford University, Stanford, CA, USA
Sohaib Hassan

Department of Pediatrics, University of California Irvine, Irvine, CA, USA


Miguel Almalvez & Ivan De Dios

Metabolic Disorders, Children’s Hospital Orange County (CHOC), Orange, CA, USA
Rebekah Barrick

Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
Ya Cui & Wei Li

Department of Pathology & Laboratory Medicine, Children’s National Hospital, Washington, DC, USA
Meghan Delaney

Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, USA
Changrui Xiao

Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA

[Link] 29/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature
Elizabeth E. Blue, Martha Horike-Pyne, Gail P. Jarvik, Annelise Y. Mah-Som, Adriana E. Sedeño Cortés & Andrew B. Stergachis

Cell & Molecular Biology, Center for Pediatric Neurological Disease Research, St Jude Children’s Research Hospital, Seattle, WA, USA
Heather C. Mefford

Consortia
GREGoR Partner Members
Aashish Adhikari, Kinga M. Bujakowska, Claudia M. B. Carvalho, Ali Crawford, Aimée
Dudley, Kelly D. Farwell Hagman, Yang I. Li, Jill E. Moore, Aaron R. Quinlan, Alex H.
Wagner, Bo Xia & S. Stephen Yi

Genomics Research to Elucidate the Genetics of Rare Diseases (GREGoR) Consortium


Moez Dawood, Ben Heavner, Marsha M. Wheeler, Rachel A. Ungar, Jonathan
LoTempio, Laurens Wiel, Claudia M. B. Carvalho, Richard A. Gibbs, Casey A.
Gifford, Susanne May, Danny E. Miller, Heidi L. Rehm, Kaitlin E. Samocha, Fritz J.
Sedlazeck, Eric Vilain, Anne O’Donnell-Luria, Jennifer E. Posey, Lisa H. Chadwick, Michael
J. Bamshad, Stephen B. Montgomery
U01HG011758

Jennifer E. Posey, Richard A. Gibbs, James R. Lupski, Hatoon Al Ali, Elizabeth G.


Atkinson, Sairam Behera, Shaghayegh T. Beheshti, Eric Boerwinkle, Tugce Bozkurt-
Yozgatli, Daniel G. Calame, Ivan Chinn, Zeynep H. Coban-Akdemir, Karen J. Coveler, Zain
Dardas, Moez Dawood, Harsha Doddapaneni, Haowei Du, Ruizhi Duan, Iman Egab, Jawid
Fatih, Mira Gandhi, Brandon Garcia, Nikhita Gogate, Christopher M.
Grochowski, Jianhong Hu, Minal Jamsandekar, Shalini N. Jhangiani, Angad Jolly, Parneet
Kaur, Ahmed K. Saad, Jesse M. Levine, Richard A. Lewis, Yidan Li, Pengfei Liu, Medhat
Mahmoud, Dana Marafi, Tadahiro Mitani, Chloe Munderloh, Donna Muzny, Sebastian
Ochoa, Piyush Panchal, Shruti Pande, Davut Pehlivan, Archana Rai, Edgar Andres Rivera-
Munoz, Aniko Sabo, Evette Scott, Fritz J. Sedlazeck, Vernon Reid Sutton, Kimberly
Walker, Lauren Westerfield, Jiaoyang Xu, Bo Yuan & Xinchang Zheng

U01HG011755

Anne O’Donnell-Luria, Heidi L. Rehm, Michael E. Talkowski, Siwaar Abouhala, K. D.


Ahlquist, Mutaz Amin, Christina Austin-Tse, Samantha M. Baxter, Benjamin
Blankenmeister, Philip M. Boone, Harrison Brand, Colleen Carlston, Celine de
[Link] 30/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

Esch, Stephanie DiTroia, Michael Duyzend, Vijay Ganesh, Kiran Garimella, Carmen
Glaze, Emily Groopman, Sanna Gudmundsson, Stacey Hall, Yongqing Huang, Julia
Klugherz, Katie Larsson, Arthur S. Lee, Gabrielle Lemire, Jialan Ma, Daniel
MacArthur, Brian Mangilog, Daniel Marten, Eva Martinez, Olfa Messaoud, Chloe
Mighton, Mariana Moyses, Ashana Neale, Emily O’Heir, Melanie C. O’Leary, Ikeoluwa
Osei-Owusu, Lynn Pais, Alicia Pham, Lindsay Romo, Kathryn Russell, Monica
Salani, Kaitlin Samocha, Alba Sanchis-Juan, Jillian Serrano, Gulalai Shah, Moriel Singer-
Berk, Mugdha Singh, Hana Snow, Kayla Socarras, Sarah L. Stenton, Jui-Cheng Tai, Grace
VanNoy, Ben Weisburd, Michael Wilson, Monica Wojcik, Isaac Wong & Rachita Yadav
U01HG011762

Stephen B. Montgomery, Jonathan A. Bernstein, Matthew T. Wheeler, Emily


Alsentzer, Taylor M. Arriaga, Euan A. Ashley, Themistocles Assimes, Gill Bejerano, Devon
Bonner, Denver Bradley, Jennefer Carter, Clarisa Chavez Martinez, Ziwei Chen, Salil
Deshpande, Sara Emami, Ivy Evergreen, Casey A. Gifford, Page Goddard, John
Gorzynski, William Greenleaf, Rodrigo Guarischi-Sousa, Caitlin Harrington, Sohaib
Hassan, Tanner D. Jensen, David Jimenez-Morales, Christopher Jin, Aimee Juan, Jessica
Kain, Laura Keehan, Anshul Kundaje, Soumya Kundu, Samuel Lancaster, Shruti
Marwaha, Dena R. Matalon, Lauren Meador, Hector Rodrigo Mendez, Alexander
Miller, Matthew B. Neu, Thuy-mi P. Nguyen, Jonathan Nguyen, Jeren D. Olsen, Evin M.
Padhi, Paul Petrowski, Astaria D. Podesta, Elizabeth Porter, Wanqiong Qiao, Thomas
Quertermous, Chloe M. Reuter, Oriane Rubio, Stuart A. Scott, Riya Sinha, Kevin S.
Smith, Michael P. Snyder, Brigitte Stark, Suchitra Sudarshan, Raquel L.
Summers, Christina G. Tise, Philip Tsao, Rachel A. Ungar, Isabella Voutos, Juliana M.
Walrod, Ziming Weng, Laurens Wiel, Frank Wong, Yao Yang, Jiye Yu & Jimmy Zhen

U01HG011745

Eric Vilain, Seth Berger, Emmanuèle C. Délot, Miguel Almalvez, Light Auriga, Rebekah
Barrick, Sami Belhadj, Krista Bluske, Leandros Boukas, Andrea J. Cohen, Ya Cui, Ivan De
Dios, Meghan Delaney, John Harting, Yun-Hua Hsiao, Rachid Karam, Charles Hadley
King, Arthur Ko, Wei Li, Bojan Losic, Jonathan LoTempio, Georgia Pitsava & Changrui
Xiao

[Link] 31/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

U01HG011744

Michael J. Bamshad, Chia-Lin Wei, Evan E. Eichler, Jessica X. Chong, Kailyn


Anderson, Peter Anderson, Sabrina Best, Elizabeth E. Blue, Kati J. Buckingham, Silvia
Casadei, Yong-Han Hank Cheng, Colleen P. Davis, Sophia B. Gibson, William W.
Gordon, Jonas Gustafson, William T. Harvey, Martha Horike-Pyne, Gail P. Jarvik, Annelise
Y. Mah-Som, Colby T. Marvin, F. Kumara Mastrorosa, Sean R. McGee, Heather C.
Mefford, Danny E. Miller, Karynne Patterson, Matthew Richardson, Adriana E. Sedeño
Cortés, Joshua D. Smith, Olivia M. Sommerland, Lea M. Starita, Andrew B.
Stergachis, Elliott G. Swanson, Jeffrey Weiss, Qian Yi, Christina Zakarian & Miranda P.
Zalusky

U24HG011746

Susanne May, Ali Shojaie, Emily Bonkowski, Sarah Conner, Matthew P.


Conomos, Stephanie M. Gogarten, Ben Heavner, Sarah C. Nelson, Sheryl Payne, Jaime
Prosser, Guanghao Qi, Adrienne M. Stilp, Catherine C. Tong, Marsha M.
Wheeler & Quenna Wong

NHGRI Program Management

Lisa H. Chadwick, Christopher Wellington, Sara Currin & Gabrielle C. Villard

Contributions
Conceptualization and methodology: M.D., B.H., M.M.W., R.A.U., J.L., L.W., S.B., J.A.B.,
J.X.C., E.C.D., E.E.E., J.R.L., A.S., M.E.T., A.H.W., C.-L.W., C.W., M.T.W., C.M.B.C., R.A.G.,
C.A.G., S.M., D.E.M., H.L.R., K.E.S., F.J.S., E.V., A.O.-L., J.E.P., L.H.C., M.J.B. and S.B.M.
Investigation: S.B., J.A.B., J.X.C., E.C.D., E.E.E., J.R.L., A.S., M.E.T., A.H.W., C.-L.W., C.W.,
M.T.W., C.M.B.C., R.A.G., C.A.G., S.M., D.E.M., H.L.R., K.E.S., F.J.S., E.V., A.O.-L., J.E.P.,
L.H.C., M.J.B. and S.B.M. Data curation: M.D., B.H., M.M.W. and S.B.M. Project
administration: S.B.M. Writing, reviewing and editing: M.D., B.H., M.M.W., R.A.U., J.L.,
L.W., C.M.B.C., R.A.G., C.A.G., S.M., D.E.M., H.L.R., K.E.S., F.J.S., E.V., A.O.-L., J.E.P., L.H.C.,
M.J.B. and S.B.M.

[Link] 32/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

Corresponding authors
Correspondence to Moez Dawood or Stephen B. Montgomery.
Ethics declarations

Competing interests
R.A.G. declares that Baylor Genetics is a Baylor College of Medicine affiliate that derives
revenue from genetic testing. BCM and Miraca Holdings have formed a joint venture
with shared ownership and governance of Baylor Genetics, which performs clinical
microarray analysis and other genomic studies (exome and genome sequencing) for
patient and family care. F.J.S. has received research support from Illumina, Pacific
Biosciences and Genentech. J.E.P. is an advisor to MaddieBio. S.B.M. is an advisor to
MyOme, PhiTech and Valinor Therapeutics. F.J.S. and D.E.M. have received research
support and/or consumables from ONT and have received travel funding to speak on
behalf of ONT. D.E.M. has received travel support from Pacific Biosciences. D.E.M. is on
an advisory board at ONT, a scientific advisory board at Basis Genetics and holds stock
options in both MyOme and Basis Genetics. M.J.B. is the chair of the scientific advisory
board of GeneDx and receives funding from the American Society of Human Genetics as
the editor-in-chief of HGG Advances. J.X.C. receives funding from the American Society
of Human Genetics as the Deputy Editor of HGG Advances. D.P. consults for Ionis
Pharmaceuticals. H.L.R. and K.E.S. have received rare-disease research funding from
Microsoft. H.L.R. has received research funding from Illumina and compensation as a
past member of the scientific advisory board of Genome Medical. A.O.-L. was a paid
consultant to Tome Biosciences, Ono Pharma USA, Addition Therapeutics, Congenica
and receives research funding from Pacific Biosciences. E.E.E. is a scientific advisory
board member of Variant Bio. M.P.S. is a cofounder and scientific advisor of Xthera,
Exposomics, Filtricine, Fodsel, iollo, InVu Health, January AI, Marble Therapeutics,
Mirvie, Next Thought AI, Orange Street Ventures, Personalis, Protos Biologics, Qbio,
RTHM and SensOmics. M.P.S. is a scientific advisor of Abbratech, Applied Cognition,
Enovone, Jupiter Therapeutics, M3 Helium, Mitrix, Neuvivo, Onza, Sigil Biosciences,

[Link] 33/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

Captify Inc, WndrHLTH, Yuvan Research and Ovul. A.A. and A.C. are employees and
shareholders of Illumina, Inc.
Peer review

Peer review information


Nature thanks Fowzan Alkuraya, Claudia Gonzaga-Jauregui and the other, anonymous,
reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Supplementary information

Supplementary Discussion
Supplementary functional genomics discussion.

Supplementary Table 1
Tracking GREGoR papers with molecular diagnoses. GREGoR has led or contributed to
83 papers studying molecular diagnoses in 365 genes with more than a third being novel
disease gene discoveries or phenotypic expansions. This table catalogues these genes
and their publications.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to
this article under a publishing agreement with the author(s) or other rightsholder(s);
author self-archiving of the accepted manuscript version of this article is solely
governed by the terms of such publishing agreement and applicable law.

[Link] 34/35
11/17/25, 11:11 AM GREGoR: accelerating genomics for rare diseases | Nature

Reprints and permissions


About this article

Cite this article


Dawood, M., Heavner, B., Wheeler, M.M. et al. GREGoR: accelerating genomics for rare diseases.
Nature 647, 331–342 (2025). [Link]

Received Accepted Published


15 December 2024 10 September 2025 12 November 2025

Version of record Issue date


12 November 2025 13 November 2025

DOI
[Link]

Subjects

Genome informatics • Medical genetics • Medical genomics • Personalized medicine

Nature (Nature) ISSN 1476-4687 (online) ISSN 0028-0836 (print)

[Link] 35/35

You might also like