Skip to main content

Principles of clinical genetics for rheumatologists: clinical indications and interpretation of broad-based genetic testing

Abstract

Advances in DNA sequencing technologies, especially next-generation sequencing (NGS), which is the basis for whole-exome sequencing (WES) and whole-genome sequencing (WGS), have profoundly transformed immune-mediated rheumatic disease diagnosis. Recently, substantial cost reductions have facilitated access to these diagnostic tools, expanded the capacity of molecular diagnostics and enabled the pursuit of precision medicine in rheumatology. Understanding the fundamental principles of genetics and diversity in genetic variant classification is a crucial milestone in rheumatology. However, despite the growing availability of DNA sequencing platforms, a significant number of autoinflammatory diseases (AIDs), neuromuscular disorders, hereditary collagen diseases, and monogenic bone diseases remain unsolved, and variants of uncertain significance (VUS) pose a formidable challenge to addressing these unmet needs in the coming decades. This article aims to provide an overview of the clinical indications and interpretation of comprehensive genetic testing in the medical field, addressing the related complexities and implications.

Background

Advances in DNA sequencing technologies precipitated by next-generation sequencing (NGS) have led to a revolution in the diagnosis of immune-mediated rheumatic diseases. This progress has not only facilitated the adoption of whole-exome sequencing (WES) in clinical practice but also made whole-genome sequencing (WGS) feasible for molecular investigation. With the success of the Human Genome Project, various molecular diagnostic tools have emerged, and the field has experienced exponential growth, enabling the pursuit of precision medicine [1]. Moreover, NGS has become more affordable, and therefore readily available, for both basic researchers and clinicians in recent years [2]. Among myopathies, genetic testing has redefined previous diagnoses of polymyositis due to the increased capability of identifying metabolic myopathies or muscular dystrophies [3]. Similarly, NGS has also allowed for the elucidation of congenital bone diseases and the identification of heritable connective tissue disorders [4, 5]. As access to genetic testing has progressively increased, rheumatologists have been able to expand the spectrum of 485 monogenic inborn errors of immunity (IEIs), especially primary immunoregulatory diseases (PIRDs) [6,7,8]. For instance, the genotype-first approach allowed for the identification of VEXAS syndrome (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic syndrome), a late-onset IEI [9], which, in turn, was found to be more prevalent than expected [10]. Emerging monogenic diseases are expanding the phenotypic spectrum of rheumatic diseases, disrupting conventional paradigms and underscoring the relevance of noncoding genetic variations [1].

Genetics analysis has resulted in the reclassification of some subgroups of rheumatic diseases according to similarities in molecular pathway activation [11]. Furthermore, the post-NGS era has driven our interest in molecular diagnosis as an avenue for targeted therapy, family counseling, a deeper understanding of the pathophysiology of rheumatic diseases, and the exclusion of other mimicking conditions. Despite the progress in molecular diagnostics, many practicing physicians often struggle to stay up to date on various platforms, especially regarding genetic test interpretation of uncertain or ambiguous cases. In addition, the molecular diagnosis of 60–70% of autoinflammatory diseases (AIDs), neuromuscular disorders, heritable connective tissue diseases and monogenic bone diseases has not yet been elucidated, and variants of uncertain significance (VUS) pose a challenge to these unmet demands in the coming decades [12, 13]. This article aims to provide an overview of the clinical indications and interpretations of comprehensive genetic testing for rheumatologists.

Back to basics: rediscovering the genetic universe

Basic genetic concepts are fundamental for broadly understanding the main clinical indications, interpreting the results and determining differences in genetic sequencing tests. The human genome is composed of 3 billion base pairs (bp) of DNA in 22 pairs of autosomal chromosomes and 1 pair of sex chromosomes that are responsible for our entire structural and functional framework encoded by approximately 20,000 genes. Our genetic material is composed of a specific sequence of purine (adenine, guanine) or pyrimidine (thymine, cytosine) nitrogenous bases that undergo histone modifications (mainly acetylation and methylation), which regulate gene expression [14].

The transcription process initiates the cascade of events guiding targeted protein production. Ribonucleic acid (RNA) polymerase assumes a crucial role, first identifying the exact location for the transcriptional complex to assemble and then catalyzing the synthesis of RNA from DNA, culminating in single-stranded messenger RNA (mRNA) transcription [15]. The mRNA may be subsequently regulated in multiple stages by posttranscriptional events that may modulate gene expression [16]. Each gene encodes a specific protein, but only the exonic regions contain the sequences necessary for the translation process. The introns are removed through spliceosome-mediated cleavage shortly after transcription to form mature mRNAs [17]. Several other subtypes of RNA, such as microRNAs (miRNAs), inhibitory RNAs (long noncoding RNAs [lncRNAs]), and small interfering RNAs (siRNAs), can also modulate protein synthesis at the nuclear level [15, 16].

Proteins are synthesized when a mature mRNA transcript is transported to the cytoplasm and undergoes translation by ribosomes in the endoplasmic reticulum. The base sequence of these coding regions is deciphered by the ribosomal machinery in informational units of three bases, known as codons. Each codon (Fig. 1) either encodes a specific amino acid (Fig. 2) or performs a regulatory function, such as initiating or terminating protein chain synthesis. Aminoacyl transport RNA (tRNA), often referred to as charged tRNA, is an RNA molecule that carries a specific amino acid and possesses an anticodon sequence that is complementary to the mRNA codon [15, 16]. Figure 3 succinctly illustrates this pathophysiological process.

Fig. 1
figure 1

Classification of codons according to the genetic code. A adenine, Ala alanine, Arg arginine, Asn asparagine, Asp aspartic acid, C cytosine, Cys cysteine, G guanine, Glu glutamic acid, Gln glutamine, Gly glycine, His histidine, Ile isoleucine, Leu leucine, Lys lysine, Met methionine, Phe phenylalanine, Pro proline, Ser serine, START start codon, STOP stop codon, Thr threonine, Trp tryptophan, Tyr tyrosine, U uracil, Val valine. Created in BioRender.com

Fig. 2
figure 2

Classification of essential and nonessential amino acids. The figure illustrates nonpolar, polar, and electrically charged amino acids, such as acidic amino acids (negatively charged) and basic amino acids (positively charged). Created in BioRender.com

Fig. 3
figure 3

Chromosomes are structures that contain genes, and their genetic material is tightly coiled. Nitrogenous bases (cytosine, guanine, thymine, and adenine) form the fundamental components of each codon (a sequence of three nitrogenous bases) that can undergo epigenetic modifications, such as histone acetylation (exposure of genetic material to transcription factors) or methylation (nitrogenous bases encrypted to external factors). Various factors, such as siRNAs, miRNAs, and IncRNAs, can either enhance gene expression or inhibit gene production. Each gene encodes a single specific protein and undergoes multiple transcription stages in which a set of ribonucleoproteins, known as the spliceosome, degrades the noncoding regions (introns) and retains only the coding regions (exons) during mRNA synthesis. Created in BioRender.com. A adenine, C cytosine, G guanine, miRNA microRNA (regulates gene expression), lncRNA long noncoding RNA (regulates gene expression), mRNA messenger RNA (encodes proteins), siRNA small interfering RNA (silencing gene expression), T thymine

In a gene locus, each DNA copy carries a distinct sequence referred to as an allele. At autosomal loci, there are two alleles, each inherited from a different parent. Autosomal recessive diseases require alterations in both alleles, occurring either in homozygosity (both alleles carrying the same variant) or compound heterozygosity (each allele carrying different variants). Conversely, autosomal dominant diseases manifest with alterations in a single allele carrying a heterozygous variant. In females, who have two X chromosomes, all loci within the X chromosome have two alleles. In contrast, in males, who have one X chromosome and one Y chromosome, each X and Y chromosome locus has only one allele. X-linked diseases may manifest in males when alterations occur in the unique X chromosome allele (hemizygosity) or, occasionally, in females when one of the X chromosome alleles is altered (heterozygosity) [18].

De novo genetic variants do not exist in parents and are frequently associated with autosomal dominant disorders when one mutated allele is sufficient to induce the clinical pathological phenotype. Approximately 80% of de novo variants originate on the paternal allele and are associated with an advanced paternal age at conception [18]. All types of genetic variants can be categorized as constitutional (previously referred to as “germline”), when they are present in all cells, or as somatic, when they occur either shortly after the initial zygotic divisions (early-onset mosaicism) or in adulthood (late-onset mosaicism) [19].

The Goldilocks effect describes the paradigm of achieving the ‘just right’ balance of gene-encoded protein function, especially with respect to wild-type variants [20]. Hypermorphic variants, known as gain-of-function variants, increase protein function. Hypomorphic variants are associated with different mechanisms, including partial function caused by haploinsufficiency (when a single variant reduces the overall protein production by approximately half without interfering with healthy allele transcription), dominant negative effects (when a single variant interferes with healthy allele function), and complete loss of function (often associated with autosomal recessive inheritance) [21].

There are several challenges in pedigree analysis and interpretation of the genetic basis of a condition within a family. First, incomplete penetrance, in which not every individual harboring the same pathogenic variant will consistently exhibit a clearly defined clinical phenotype, can occur. Second, clinical expressivity is variable, as several diseases exhibit substantial diversity in the manifestation of clinical features associated with a particular genetic variant.

Classification of gene variants

In recent decades, some genetic nomenclature has caused confusion due to misleading concepts. Traditionally, a “mutation” was defined as a permanent alteration in the nucleotide sequence, typically occurring at a frequency of less than 1%, while a “polymorphism” was defined as a variant with a frequency greater than 1%. To enhance clarity and align with recommendations from the American College of Medical Genetics and Genomics (ACMG), replacing these terms with more standardized and precise terminology is advisable. The ACMG recommends employing the term “variant” alongside the following modifiers: (i) pathogenic, (ii) likely pathogenic, (iii) VUS, (iv) likely benign, or (v) benign. This revised terminology better allows for clear and consistent communication in medical genetics, ensuring that the significance of genetic variations is conveyed accurately [22].

Variations may also be divided in the manner that they occur. Structurally, they can be classified as silent, missense, nonsense, frameshift, splicing, or reversion genetic variants. A silent variant is a variant in which a single nitrogenous base is substituted for another, resulting in a change in the codon but not affecting the encoded amino acid. Consequently, the protein remains unaltered. These variants can occur because different sequences of nitrogenous bases can encode the same amino acid. These variants are usually not pathogenic and are commonly referred to as “synonymous” [23].

Missense variants occur when a single nucleotide in the DNA code changes, replacing one amino acid with another within a protein. These variants may maintain normal protein expression or result in protein dysfunction or instability, potentially causing disease [24]. While most missense variants are often classified as VUS, in silico tools are valuable in predicting pathogenicity. Additionally, functional studies, posttranslational modifications, protein‒protein interactions, and protein 3D features in database software contribute to a comprehensive assessment [25].

A nonsense variant occurs when a single nitrogenous base change transforms an amino acid codon into a stop codon leading a protein to terminate or end its translation earlier than expected. This change halts protein synthesis, typically resulting in an unstable and dysfunctional truncated protein, usually with loss-of-function characteristics. The position of nonsense variants plays an important role in protein function and predicting phenotype severity [26].

INDEL (insertion-deletion) are variants caused by insertions or deletions of nitrogenous bases. If a base pair insertion or deletion occurs in a multiple of three starting from the first “wobble” nucleotide, which represents the creation or disappearance of balanced codons, an “in-frame” variant is created; therefore, the protein sequence is altered exclusively at this position. However, if the number of inserted or deleted nucleotides is not a multiple of three or does not start at the beginning of a codon, the reading frame will be disrupted from that point on, causing a “frameshift” that completely changes the sequence and often creates a profoundly altered, unstable, dysfunctional protein. In these cases, a stop codon typically occurs within fewer than 100 nucleotides, resulting in prematurely truncated protein products [27].

Splicing variants are caused by alterations in the genetic rearrangement between exons and introns during mRNA formation. These variants lead to the production of modified mature mRNAs. Variants occurring at the first and second positions before the beginning of a new exon or at the first and second positions after the end of an exon, the so-called canonical splicing sites, are most frequently linked to splicing alterations. In most canonical splice site variants, the mRNA is either destroyed or translated into unstable and/or dysfunctional proteins [28, 29].

Reversion variants occur in exceptional cases of somatic mosaicism, triggered by the restoration of an inherited pathogenic variant to a normal state. In reversion mosaicism, the reversion variant serves to partially or fully reinstate the effect of the primary disease-causing variant. The most common and simplest type of reversion is a true back variant, which refers to the reversal of the constitutional variant site to the wild-type sequence [30]. The primary genetic variant types and their impact on the gene products are detailed in Table 1.

Table 1 Primary genetic variant types. Herein, the wild-type gene has a short nucleotide sequence that encodes a protein with an amino acid sequence that reads “I am rheumatologist”. Different variants disrupt this sequence in different ways. The underlined letters correspond to variants of either nucleotides within the codon regions or amino acids

A variety of publicly or commercially available in silico pathogenicity prediction tools can help determine the chance that a sequence variant is harmful. These tools employ diverse computational algorithms to assess the variant’s impact on both nucleotide and amino acid sequences in the protein. It may predict the effect of genetic variants on the structure or function of a protein without conducting functional tests. Several software tools have been designed to assess different variant types [22]. Meta-predictors that utilize machine learning algorithms and integrate different sources of data, such as REVEL (rare exome variant ensemble learner) and BeyesDel, have gained prominence [22, 38, 39]. Caution is advised when using these prediction tools, and one should refrain from relying solely on them to make clinical decisions. The term “damaging” does not necessarily imply “pathogenic”, since a variant that damages a gene may not be inherently harmful to an individual’s health [40].

An increasing number of variants are being deposited in population databases. These databases play an essential role in classifying gene variants, assessing their pathogenicity, and aggregating diverse sources of validated articles to predict their population risk of appearance. Table 2 provides examples of key databases that can be valuable in the assessment of gene variants [41,42,43,44,45]. Clinicians can also use these different tools to interpret genetic variations and their connections with phenotypes.

Table 2 Population, disease, and sequence databases. These databases are valuable tools for querying identified gene variants, as they can be continually updated with new insights related to population frequency, functional validation studies, and key references from relevant research

Types of genetic tests

Clinical genetic testing has become increasingly accessible and cost-effective, providing a variety of techniques with unique advantages and limitations. Sanger sequencing (SS), NGS targeted gene panels, WES, WGS, and chromosomal analyses (CAs) are the primary options for achieving a genetic diagnosis [46, 47]. Physicians should choose the appropriate methodology according to the specific clinical context and other factors, such as the scope of analysis, cost constraints, and the nature of the genetic condition under investigation. Table 3 lists the characteristics, including the strengths and limitations, of each method [12, 48].

Table 3 Currently available genetic tests

Sanger sequencing (SS)

In the 1970s, a method originally described by Frederick Sanger was the gold standard for identifying single-gene disorders [49]. SS involves a manual analysis process in which nucleotide pairs are examined exon by exon, which limits automation. SS involves the construction of specific DNA primers for each region of interest that direct in vitro DNA replication by DNA polymerase. Chain-terminating dideoxynucleotides are randomly incorporated in this process, generating DNA fragments of different sizes that are subsequently analyzed using gel electrophoresis and, more recently, capillary electrophoresis to detect genetic variants [50]. SS is highly accurate but is limited to specific genes and may be too time-consuming and costly for broader analysis. Notably, this method may fail to detect copy number variations (CNVs), such as microdeletions or microduplications, or somatic variants characterized by a low variant allele frequency (VAF) [48].

Next-generation sequencing targeted gene panels

Unlike in SS, multiple genes can be simultaneously analyzed in NGS using an automated approach. When several potential monogenic causes fit a well-established phenotype, the use of NGS becomes necessary, because it is cost effective and faster. Currently, there are targeted panels, which detect fewer genes, and expanded panels; the choice of panel depends on the specific cause under investigation [51]. There are several NGS platforms, and each platform has unique steps for sample preparation, library elaboration and sequencing. DNA extraction from a biological sample followed by fragmentation generally constitutes the initial step. Next, the genomic regions of interest are isolated and enriched (a step often referred to as capture); for targeted gene panels, a limited subset of genes, varying from a few to hundreds, may be enriched. Linkers are affixed to the termini of DNA fragments, and these fragments are tethered to a solid support, commonly a bead, where they are typically amplified through an emulsion polymerase chain reaction (PCR) method. In this process, the information derived from nitrogenous bases is transformed into binary sequences, and the outcome is computationally analyzed, often resulting in a substantial volume of data [37]. Figure 4 provides a concise overview of the key differences between SS and NGS methods [37, 50].

Fig. 4
figure 4

General methods of DNA sequencing encompass both traditional Sanger sequencing (a) and next-generation sequencing (b). a Sanger sequencing involves PCR amplification of genomic DNA fragments containing gene coding regions, followed by sequencing using labeled di-deoxy nucleotides. The amplified fragments are separated by capillary electrophoresis to generate a chromatogram, facilitating sequence determination. b In contrast, next-generation sequencing techniques, such as shotgun sequencing, begin with fragmentation of genomic DNA by sonication or enzymatic methods. Linkers are added to DNA fragments, which are then immobilized on solid supports like beads. Emulsion PCR amplifies these fragments, with labeled deoxy nucleotides flowing into reaction chambers containing polymerase and reaction buffers. Incorporation of each nucleotide emits detectable light or ions, allowing real-time sequencing. This approach enables high-throughput sequencing via cyclic-array methods, where millions of immobilized PCR colonies (“polonies”) facilitate parallel processing of sequencing reactions. Imaging-based detection of fluorescent labels during enzymatic extensions enables simultaneous acquisition of sequencing data across all features, resulting in contiguous sequencing reads for each array feature. Figure modified from Shendure and Torgerson et al. [37, 50]

Some advantages of targeted gene panels include the ability to study multiple regions and genes of the genome (more than can be studied with SS) at a relatively low cost (generally lower than that of WES or WGS). Moreover, mosaicisms with lower VAFs and CNVs, such as microdeletions and microduplications, may be detected. However, pseudogenes may lead to misinterpretation of variant results [48].

Whole-exome sequencing

WES covers the coding regions of the genome (exons), making it an automated method that generates a substantial volume of data for bioinformatics analysis. When the clinical phenotype is nonspecific for a single causative gene, a defined group of genes or even a unique metabolic pathway, WES has been shown to be a pivotal strategy for investigating a wide spectrum of genetic disorders. The steps of WES steps are very similar to those of NGS targeted gene panels, as discussed above. The isolation and fragmentation of genomic DNA are followed by the addition of oligonucleotide adapters. Fragmented adapter-ligated DNA libraries necessitate an additional positive selection capture step to prevent off-target sequencing of noncoding genome regions. The ideal result is equal capture of all exome regions; however, enrichment tends to be uneven [52]. As most known monogenic defects are located within coding regions, WES is a valuable and relatively accessible diagnostic tool that is more affordable than WGS. Nevertheless, due to the large amount of data, VUS frequently emerge, posing a challenge in interpretation. WES allows for the study of large CNVs and high-grade mosaicism, despite its limitations in identifying relevant intronic variants, low-grade mosaicisms and small CNVs [53].

Whole-genome sequencing

WGS covers most of the genome, encompassing coding and noncoding regions. However, its widespread availability is limited by its high cost and the relatively low number of known intronic pathogenic variants; therefore, this application is currently indicated for clinical research purposes [54]. The WGS methodology closely resembles that of WES, with the notable exception of the absence of an exome enrichment step. The process involves DNA fragmentation, attachment of linker sequences, and subsequent massively parallel sequencing. WGS technologies can be categorized on the basis of their capacity to read short sequences (< 1 kilobase) versus long sequences (> 1 kilobase). Long-read sequencing, despite its relative clinical unavailability, shows promise for mitigating DNA fragmentation, offering deeper reads without sacrificing any nucleotide bases during the process [55, 56]. WGS is more effective at detecting CNVs than WES is and has the potential to identify novel disease-causing gene variants. However, due to the “big data” analysis involved, storing raw data for future reanalysis is a challenge. Moreover, the high cost, coupled with the significant volume of VUS and unknown intronic variants, currently presents substantial hurdles to larger routine laboratory use [56].

Chromosomal analyses

Various techniques can be employed to assess CAs, such as karyotyping, microarray analysis, and fluorescence in situ hybridization (FISH). While karyotyping can reveal large deletions, duplications, translocations and inversions, it is limited in the identification of microdeletions, microduplications, or smaller rearrangements that may be detectable only by microarray or FISH [57]. CA techniques enable the identification of chromosomal losses and gains and are recommended as first-tier approaches, particularly for syndromic phenotypes characterized by dysmorphic features, congenital malformations, failure to thrive or neurodevelopmental disorders [58]. The generally fast turnaround time and lower cost of these methods compared to WES or WGS represent significant advantages. However, confirming CNVs of uncertain significance can be challenging, and the detection of unbalanced chromosomal rearrangements smaller than 100 kb may be challenging [59].

Practical approach to order genetic tests

Early genetic sequencing in selected patients, whether through preestablished multigene panels or WES, is indicated for diagnosing monogenic diseases. Given the extensive clinical spectrum of genetic diseases of interest in rheumatology, it is challenging to establish universal guidelines and warning signs. The Jeffrey Modell Foundation has devised several warning indicators for IEIs that can aid in matching a specific phenotype with its related molecular diagnosis (Table 4) [60, 61].

Table 4 IEI warning signs from the Jeffrey Modell Foundation: clues for diagnosis

In the pre-NGS era, a molecular assay targeting an individual gene typically represented the conclusive phase of the diagnostic process, after clinical, laboratory, and histological assessments aimed at delineating the most likely diagnosis. As NGS becomes more integrated into routine clinical diagnostics, sequencing methods are frequently employed at an earlier stage, immediately following a thorough clinical evaluation [3].

In addition, as the costs of genetic tests have decreased, there has been a continuous increase in the identification of new variants responsible for novel Mendelian diseases [62]. Targeted gene panels focusing on heritable extracellular matrix diseases and thoracic aortic aneurysms played a pivotal role in elucidating the genetic underpinnings of autosomal dominant diseases, such as Marfan syndrome, vascular Ehlers–Danlos syndrome, and Loeys–Dietz syndrome, that were previously considered extremely rare and were revealed to be notably prevalent. Molecular diagnostics serve as a valuable tool for distinguishing these conditions from vasculitis mimickers, aiding in accurate diagnosis and preventing unnecessary immunosuppression [63]. Moreover, with the increased availability of genetic panels, polymyositis has become an increasingly rare entity due to the recognition of inherited neuromuscular diseases [64].

The identification of monogenic bone diseases can be an integral part of investigating unexplained reductions or increases in bone mineral density, bone mineralization or bone turnover. Genetic sequencing is fundamental for corroborating the clinical diagnoses of patients with osteogenesis imperfecta, juvenile Paget disease, or fibrodysplasia ossificans progressiva [65].

Notably, it is imperative that any genetic sequencing results are consistently interpreted in light of the clinical phenotype and the identified molecular pathway. Figure 5 presents a suggested algorithm for molecular investigation. Numerous parameters must be considered in the classification and interpretation of a variant. Zygosity is a crucial factor, as monoallelic variants can cause autosomal dominant conditions, whereas biallelic variants contribute to recessive disorders. The assessment of pathogenicity hinges upon several facets, including population frequencies of the variants in genome databases, computational and in silico predictions, functional data, segregation analysis within family pedigrees, allelic data, functional insights, and patient phenotype. It is essential to emphasize that these data should not be interpreted in isolation and must always be evaluated within the context of the relevant metabolic pathway [22].

Fig. 5
figure 5

Suggested algorithm for molecular investigation. GOF gain-of-function, LOF loss-of-function, NGS next-generation sequencing, WES whole-exome sequencing, WGS whole-genome sequencing; *metabolic myopathies, muscular dystrophies, congenital bone diseases, heritable connective tissue disorders, or inborn errors of immunity can be considered in the phenotyping

Clinicians should not categorically attribute a VUS as the primary cause of a condition solely based on its apparent clinical relevance, as variant pathogenicity classifications evolve over time, encompassing shifts toward both increased and reduced pathogenicity [66]. Only a small proportion of VUS are likely to ultimately demonstrate pathogenicity upon subsequent evaluation. Unfortunately, the clarification of this uncertainty is often a protracted process. In silico predictors, proximity to previously described hotspots, functional investigations, and parental segregation studies can provide valuable assistance in the clinical decision-making process [67].

One of the most efficacious approaches for obtaining insights into the clinical importance of a VUS or for discerning compound heterozygosity in the trans configuration is familial segregation analysis [68]. Segregation analysis, which studies the inheritance pattern of a variant within a family, may be a valuable approach to determine the pathogenicity of variants [69]. Segregation analysis can also aid in the identification of de novo variants, providing stronger evidence for pathogenicity. Additionally, it can facilitate the reclassification of rare variants as benign or likely benign. Hence, this step is pivotal in refining the precise interpretation of genetic findings [70].

Limitations of genetic analysis

Somatic mosaicism may be an explanation for negative genetic analysis results. In these cases, the tissue or cell containing the cryptic variant may have not been assessed, or the VAF in peripheral blood may be so low that even WES or WGS may lack the requisite sensitivity to detected it. Various NGS panels have endeavored to achieve the detection of variants with progressively lower VAFs, exemplified by the search for UBA1 (ubiquitin like modifier activating enzyme 1) in the context of VEXAS syndrome diagnosis [71] or for the LEMD3 (LEM domain containing 3), KRAS (kirsten rat sarcoma viral oncogene homolog), MAP2K1 (mitogen-activated protein kinase kinase 1) and SMAD3 (SMAD family member 3) genes in melorheostosis [72].

Structural variants (SVs) are a category of genetic alterations exceeding 50 bp in length, with some extending up to several megabases (Mb). This category encompasses various changes, including CNVs, deletions, duplications, insertions, inversions, mobile element insertions (transposons), translocations, and complex rearrangements. Short-read sequencing technologies may not detect some SVs due to their limited precision and accuracy. Emerging techniques employing long-read sequencing (100–300 bp long) are being implemented to bypass the genetic material sonication process and prevent nucleotide base losses using nanopore sequencing [13, 73].

While nucleotide repeats form the basis of 3% of the human genome, certain repeat sequences known as short tandem repeats are associated with specific diseases and may remain undetectable using conventional tools. Another limitation of standard platforms arises from pseudogenes, which are segments of DNA structurally resembling genes but lacking the capacity for protein encoding, introducing several biases in regular analysis [53].

Diseases or variants that have never been previously documented in databases can complicate the management of these conditions, which often manifest as ultrarare diseases. One strategy to overcome this challenge is to periodically reanalyze the genetic sequence. Several groups have shown that reanalyzing raw genomic data can boost diagnostic yields by 5–26% for WES and 4–11% for WGS [13, 74]. Despite the promising advances beyond broad-spectrum genetic sequencing methodologies that will be made in the coming decades, some cases are exceedingly intricate and transcend the scope of the general medical practitioner. In these cases, the pursuit of interdisciplinary collaboration, knowledge exchange with specialists, and referral to molecular diagnostic reference centers seems more appropriate.

Conclusions and future perspectives

The increasing accessibility of genetic tests enhances diagnostic yield and elucidates the molecular underpinnings of rheumatic diseases [11]. Advances in these techniques have reshaped our understanding of the pathophysiology of rheumatic disorders, especially AIDs, neuromuscular disorders, hereditary extracellular matrix diseases, and monogenic bone diseases.

However, as we rely more on these tools, new challenges emerge, such as interpreting VUS, detecting mosaicism, and identifying SVs. Promising solutions include complementing short-read genome sequencing with RNA sequencing, long-read genome sequencing, metabolomics, proteomics, and DNA methylation profiling. Furthermore, novel functional tests are essential for validating novel genetic variant results [13, 73]. Machine learning can serve as a strategy for gathering extensive datasets encompassing various types of biomarkers to complement genetic sequencing [75, 76].

A significant hindrance in diagnosing rare patients is the cost, given that some genetic tests are still research-based and not yet integrated into healthcare systems. Efforts are required to increase the accuracy and affordability of high-throughput technologies, bridging the diagnostic gap for undiagnosed patients. A delicate balance is imperative when considering the cost-effectiveness of molecular diagnoses and personalized targeted therapies as we progress toward precision medicine [77, 78].

Data availability

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Abbreviations

A:

Adenine

AID:

Autoinflammatory disease

ACMG:

American College of Medical Genetics and Genomics

bp:

Base pairs

C:

Cytosine

CA:

Chromosomal analysis

CNV:

Copy number variation

COPD:

Chronic obstructive pulmonary disease

DMARD:

Disease-modifying antirheumatic drug

DNA:

Deoxyribonucleic acid

EBV:

Epstein–Barr virus

FISH:

Fluorescence in situ hybridization

G:

Guanine

GOF:

Gain-of-function

IBD:

Inflammatory bowel disease

IEI:

Inborn error of immunity

IncRNA:

Long noncoding RNA

ILD:

Interstitial lung disease

JIA:

Juvenile idiopathic arthritis

LOF:

Loss-of-function

Mb:

Megabase

mRNA:

Messenger RNA

miRNA:

MicroRNA

NGS:

Next-generation sequencing

NMSC:

Nonmelanoma skin cancer

PCR:

Polymerase chain reaction

PID:

Primary immunodeficiency disease

PIRD:

Primary immunoregulatory disease

RNA:

Ribonucleic acid

siRNA:

Small interfering RNA

SS:

Sanger sequencing

T:

Thymine

UBA1:

Ubiquitin like modifier activating enzyme 1

VAF:

Variant allele frequency

VEXAS syndrome:

Vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic syndrome

VUS:

Variant of undetermined significance

WES:

Whole-exome sequencing

WGS:

Whole-genome sequencing

References

  1. Donlin LT, Park SH, Giannopoulou E, Ivovic A, Park-Min KH, Siegel RM, et al. Insights into rheumatic diseases from next-generation sequencing. Nat Rev Rheumatol 2019;15:327–39.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Wiley GB, Kelly JA, Gaffney PM. Use of next-generation DNA sequencing to analyze genetic variants in rheumatic disease. Arthritis Res Ther. 2014;16(490).

  3. Thompson R, Spendiff S, Roos A, Bourque PR, Warman Chardon J, Kirschner J, et al. Advances in the diagnosis of inherited neuromuscular diseases and implications for therapy development. Lancet Neurol 2020;19:522–32.

    Article  CAS  PubMed  Google Scholar 

  4. Veatch OJ, Steinle J, Hossain WA, Butler MG. Clinical genetics evaluation and testing of connective tissue disorders: a cross-sectional study. BMC Med Genomics. 2022;15(1).

  5. Mäkitie RE, Costantini A, Kämpe A, Alm JJ, Mäkitie O. New insights into monogenic causes of osteoporosis. Front Endocrinol (Lausanne). 2019;10.

  6. Bousfiha A, Moundir A, Tangye SG, Picard C, Jeddane L, Al-Herz W, et al. The 2022 update of IUIS phenotypical classification for human inborn errors of immunity. J Clin Immunol 2022;42:1508–20.

    Article  PubMed  Google Scholar 

  7. Notarangelo LD, Bacchetta R, Casanova JL, Su HC. Human inborn errors of immunity: an expanding universe. Sci Immunol. 2020;5(49).

  8. Chan AY, Torgerson TR. Primary immune regulatory disorders: a growing universe of immune dysregulation. Curr Opin Allergy Clin Immunol. 2020;20(6):582–90.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Beck DB, Ferrada MA, Sikora KA, Ombrello AK, Collins JC, Pei W, et al. Somatic mutations in UBA1 and severe adult-onset autoinflammatory disease. NEJM 2020;383:2628–38.

    Article  CAS  PubMed  Google Scholar 

  10. Beck DB, Bodian DL, Shah V, Mirshahi UL, Kim J, Ding Y, et al. Estimated prevalence and clinical manifestations of UBA1 variants associated with VEXAS syndrome in a clinical population. JAMA 2023;329:318–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Eyre S, Orozco G, Worthington J. The genetics revolution in rheumatology: large scale genomic arrays and genetic mapping. Nat Rev Rheumatol. 2017;13(7):421–32.

    Article  CAS  PubMed  Google Scholar 

  12. Schnappauf O, Aksentijevich I. Current and future advances in genetic testing in systemic autoinflammatory diseases. Rheumatology (United Kingdom). 2019;58:VI44–55.

    Article  CAS  Google Scholar 

  13. Marwaha S, Knowles JW, Ashley EA. A guide for the diagnosis of rare and undiagnosed disease: beyond the exome. Genome Med. 2022;14(1).

  14. Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011;21(3):381–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Koellner CM, Mensink KA, Highsmith WE. Basic concepts in human molecular genetics. In: Molecular Pathology: the Molecular Basis of Human Disease. Elsevier Inc.; 2018. pp. 99–120.

    Chapter  Google Scholar 

  16. Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Irimia M, Roy SW. Origin of spliceosomal introns and alternative splicing. Cold Spring Harb Perspect Biol. 2014;6(6).

  18. Acuna-Hidalgo R, Veltman JA, Hoischen A. New insights into the generation and role of de novo mutations in health and disease. Genome Biol. BioMed Central Ltd.. 2016;17.

  19. Hoffman HM, Broderick L. Editorial: it just takes one: somatic mosaicism in autoinflammatory disease. Arthritis Rheumatol. 2017;69(2):253–56.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Ma CS, Tangye SG. Inborn errors of immunity: the Goldilocks effect-susceptibility to disease due to a little too much or a little too little. Clin Exp Immunol. 2023;212(2):93–95.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Vorsteveld EE, Hoischen A, van der Made CI. Next-generation sequencing in the field of primary immunodeficiencies: current yield, challenges, and future perspectives. Clin Rev Allergy Immunol. 2021;61(2):212–25.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 2015;17:405–24.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Brookes AJ. Single Nucleotide Polymorphism (SNP). In: Encyclopedia of Life Sciences. 2005.

    Google Scholar 

  24. Quinodoz M, Peter VG, Cisarova K, Royer-Bertrand B, Stenson PD, Cooper DN, et al. Analysis of missense variants in the human genome reveals widespread gene-specific clustering and improves prediction of pathogenicity. Am J Hum Genet 2022;109:457–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Iqbal S, Pérez-Palma E, Jespersen JB, May P, Hoksza D, Heyne HO, et al. Comprehensive characterization of amino acid positions in protein structures reveals molecular effect of missense variants. Proc Natl Acad Sci U S A 2020;117:28201–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Torella A, Zanobio M, Zeuli R, Del Vecchio Blanco F, Savarese M, Giugliano T, et al. The position of nonsense mutations can predict the phenotype severity: a survey on the DMD gene. PLoS One 2020;15:e0237803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Roth JR. Frameshift mutations. 1974; Available from: www.annualreviews.org

  28. Marasco LE, Kornblihtt AR. The physiology of alternative splicing. Nat Rev Mol Cell Biol. 2023;24(4):242–54.

    Article  CAS  PubMed  Google Scholar 

  29. Lord J, Baralle D. Splicing in the diagnosis of rare disease: advances and challenges. Front Genet. 2021;12:689892.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Miyazawa H, Wada T. Reversion mosaicism in primary immunodeficiency diseases. Front Immunol. 2021;12.

  31. Liu Y, Ramot Y, Torrelo A, Paller AS, Si N, Babay S, et al. Mutations in proteasome subunit β type 8 cause chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature with evidence of genetic and phenotypic heterogeneity. Arthritis Rheum 2012;64:895–907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Touitou I. The spectrum of Familial Mediterranean Fever (FMF) mutations. Eur J Hum Genet. 2001;9(7):473–83.

    Article  PubMed  Google Scholar 

  33. Cuisset L, Drenth JP, Simon A, Vincent MF, Van Der Velde Visser S, Van Der Meer WM, et al. Molecular analysis of MVK mutations and enzymatic activity in hyper-IgD and periodic fever syndrome. Eur J Hum Genet 2001.

  34. Barron KS, Aksentijevich I, Deuitch NT, Stone DL, Hoffmann P, Videgar-Laird R, et al. The spectrum of the deficiency of Adenosine Deaminase 2: an observational analysis of a 60 patient cohort. Front Immunol. 2022;12.

  35. Lee Y, Wessel AW, Xu J, Reinke JG, Lee E, Kim SM, et al. Genetically programmed alternative splicing of NEMO mediates an autoinflammatory disease phenotype. J Clin Investig. 2022;132(6).

  36. Davis BR, Dicola MJ, Prokopishyn NL, Rosenberg JB, Moratto D, Muul LM, et al. Unprecedented diversity of genotypic revertants in lymphocytes of a patient with Wiskott-Aldrich syndrome. Blood 2008;111:5064–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Torgerson T, Ochs H. Genetics of primary immune deficiencies. In: Stiehm’s Immune Deficiencies. Elsevier Inc.; 2014. pp. 73–81.

    Chapter  Google Scholar 

  38. Houdayer C, Caux-Moncoutier V, Krieger S, Barrois M, Bonnet F, Bourdon V, et al. Guidelines for splicing analysis in molecular diagnosis derived from a set of 327 combined in silico/in vitro studies on BRCA1 and BRCA2 variants. Hum Mutat 2012;33:1228–38.

    Article  CAS  PubMed  Google Scholar 

  39. Quaio CRDC, Ceroni JRM, Pereira MA, Teixeira ACB, Yamada RY, Cintra VP, et al. The hospital Israelita Albert Einstein standards for constitutional sequence variants classification: version 2023. Hum Genomics. 2023;17(1).

  40. Eilbeck K, Quinlan A, Yandell M. Settling the score: variant prioritization and Mendelian disease. Nat Rev Genet. 2017;18(10):599–612.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gudmundsson S, Singer-Berk M, Watts NA, Phu W, Goodrich JK, Solomonson M, et al. Variant interpretation using population databases: lessons from gnomAD. Hum Mutat 2022;43:1012–30.

    Article  PubMed  Google Scholar 

  42. Touitou I, Lesage S, McDermott M, Cuisset L, Hoffman H, Dode C, et al. Infevers: an evolving mutation database for auto-inflammatory syndromes. Hum Mutat 2004;24:194–98.

    Article  CAS  PubMed  Google Scholar 

  43. Amberger JS, Bocchini CA, Schiettecatte F, Scott AF, Hamosh A.OMIM.org: online Mendelian Inheritance in Man (OMIM®), an Online catalog of human genes and genetic disorders. Nucleic Acids Res. 2015;43(D1):D789–98.

    Article  PubMed  Google Scholar 

  44. Landrum MJ, Chitipiralla S, Brown GR, Chen C, Gu B, Hart J, et al. ClinVar: improvements to accessing data. Nucleic Acids Res 2020;48:D835–44.

    Article  CAS  PubMed  Google Scholar 

  45. Stenson PD, Mort M, Ball EV, Chapman M, Evans K, Azevedo L, et al. The Human Gene Mutation Database (HGMD®): optimizing its use in a clinical diagnostic or research setting. Hum Genet 2020;139:1197–207.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Pareek CS, Smoczynski R, Tretyn A. Sequencing technologies and genome sequencing. J Appl Genet. 2011;52(4):413–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shendure J, Balasubramanian S, Church GM, Gilbert W, Rogers J, Schloss JA, et al. DNA sequencing at 40: past, present and future. Nature. 2017;550(7676).

  48. Chinn IK, Chan AY, Chen K, Chou J, Dorsey MJ, Hajjar J, et al. Diagnostic interpretation of genetic studies in patients with primary immunodeficiency diseases: a working group report of the Primary Immunodeficiency Diseases Committee of the American Academy of Allergy, Asthma & Immunology. J Allergy Clin Immunol 2020;145:46–69.

    Article  CAS  PubMed  Google Scholar 

  49. Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors (DNA polymerase/nucleotide sequences/bacteriophage 4×174). 1977;74(12):5463–67.

  50. Shendure J, Ji H. Next-generation DNA sequencing. Nat Biotechnol. 2008;26(10):1135–45.

    Article  CAS  PubMed  Google Scholar 

  51. Yohe S, Thyagarajan B. Review of clinical next-generation sequencing. Arch Pathol Lab Med. 2017;141(11):1544–57.

    Article  CAS  PubMed  Google Scholar 

  52. Petersen BS, Fredrich B, Hoeppner MP, Ellinghaus D, Franke A. Opportunities and challenges of whole-genome and -exome sequencing. BMC Genet. 2017;18(1).

  53. Corominas J, Smeekens SP, Nelen MR, Yntema HG, Kamsteeg EJ, Pfundt R, et al. Clinical exome sequencing—mistakes and caveats. Hum Mutat 2022;43:1041–55.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Prokop JW, May T, Strong K, Bilinovich SM, Bupp C, Rajasekaran S, et al. Genome sequencing in the clinic: the past, present, and future of genomic medicine. Physiol Genomics. 2018;50:563–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Goodwin S, McPherson JD, McCombie WR. Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet. 2016;17(6):333–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lappalainen T, Scott AJ, Brandt M, Hall IM. Genomic analysis in the age of human genome sequencing. Cell. 2019;177(1):70–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Qaisar U, Tayyeb A, Bhat TA. Techniques of chromosomal studies. In: Chromosome Structure and Aberrations. Springer India; 2017. pp. 307–30.

    Chapter  Google Scholar 

  58. Sharkey FH, Maher E, Fitzpatrick DR. Chromosome analysis: what and when to request. Arch Dis Child. 2005;90(12):1264–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ponnuraj KT. Cytogenetic techniques in diagnosing genetic disorders. Chapter 3. In: Advances in the Study of Genetic Disorders. 2011.

    Google Scholar 

  60. McCusker C, Upton J, Warrington R. Primary immunodeficiency. Allergy Asthma Clin Immunol. 2018;14.

  61. Sogkas G, Witte T. The link between rheumatic disorders and inborn errors of immunity. EBioMedicine. 2023;90.

  62. Ostberg NP, Zafar MA, Ziganshin BA, Elefteriades JA. The genetics of thoracic aortic aneurysms and dissection: a clinical perspective. Biomolecules. 2020;10(2).

  63. Miller CL, Kontorovich AR, Hao K, Ma L, Iyegbe C, Björkegren JLM, et al. Precision medicine approaches to vascular disease: JACC Focus Seminar 2/5. J Am Coll Cardiol 2021;77:2531–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Amato AA, Griggs RC. Unicorns, dragons, polymyositis, and other mythological beasts. Neurology. 2003;61(3):288–90.

    Article  PubMed  Google Scholar 

  65. Faruqi T, Dhawan N, Bahl J, Gupta V, Vohra S, Tu K, et al. Molecular, phenotypic aspects and therapeutic horizons of rare genetic bone disorders. Biomed Res Int. 2014;2014.

  66. Sullivan KE. The scary world of variants of uncertain significance (VUS): a hitchhiker’s guide to interpretation. J Allergy Clin Immunol. 2021;147(2):492–94.

    Article  CAS  PubMed  Google Scholar 

  67. Burke W, Parens E, Chung WK, Berger SM, Appelbaum PS. The challenge of genetic variants of uncertain clinical significance: a narrative review. Ann Intern Med. 2022;175(7):994–1000.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Buie RW, Rañola JMO, Chen AT, Shirts BH.An algorithm for optimal testing in co-segregation analysis. Hum Mutat. 2022;43(5):547–56.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Belman S, Parsons MT, Hons B, Spurdle AB, Goldgar DE, Feng BJ. Considerations in assessing germline variant pathogenicity using cosegregation analysis. Genet Med. 2020.

  70. Quaio CRD, Angioli C, Ceroni JRM, Cervato MC, Thurow HS, Moreira CM, Trindade ACG, et al. Parental segregation study reveals rare benign and likely benign variants in a Brazilian cohort of rare diseases. Sci Rep. 2022;12(1).

  71. Aluri J, Cooper MA. Somatic mosaicism in inborn errors of immunity: current knowledge, challenges, and future perspectives. Semin Immunol. 2023;67.

  72. Kang H, Jha S, Ivovic A, Fratzl-Zelman N, Deng Z, Mitra A, et al. Somatic SMAD3-activating mutations cause melorheostosis by up-regulating the TGF-β/SMAD pathway. J Exp Med. 2020;217(5).

  73. Wortmann SB, Oud MM, Alders M, Coene KLM, van der Crabben SN, Feichtinger RG, et al. How to proceed after “negative” exome: a review on genetic diagnostics, limitations, challenges, and emerging new multiomics techniques. J Inherit Metabol Dis. John Wiley and Sons Inc. 2022;45:663–81.

    Article  CAS  Google Scholar 

  74. Murdock DR, Rosenfeld JA, Lee B. Annual review of medicine what has the undiagnosed diseases network taught us about the clinical applications of genomic testing? Annu Rev Med. 2022;73:575–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Reel PS, Reel S, Pearson E, Trucco E, Jefferson E. Using machine learning approaches for multi-omics data analysis: a review. Biotechnol Adv. 2021;49.

  76. Maceachern SJ, Forkert ND. Machine learning for precision medicine. Genome. 2021;64(4):416–25.

    Article  PubMed  Google Scholar 

  77. Carrasco-Ramiro F, Peiró-Pastor R, Aguado B. Human genomics projects and precision medicine. Gene Ther. 2017;24(9):551–61.

    Article  CAS  PubMed  Google Scholar 

  78. Olivier M, Asmis R, Hawkins GA, Howard TD, Cox LA. The need for multi-omics biomarker signatures in precision medicine. Int J Mol Sci. 2019;20(19).

Download references

Acknowledgements

The authors would like to thank the Sociedade Brasileira de Reumatologia (SBR).

Funding

There is no funding to be declared.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to this manuscript. All the authors read and approved the final manuscript.

Corresponding author

Correspondence to Sandro Félix Perazzio.

Ethics declarations

Ethics approval and consent to participate

This manuscript includes rare disease reviews performed by a panel of experts; therefore, there is no pertinent research requiring ethics approval. Consent for publication: All the authors agree with the content of the manuscript.

Consent for publication

All the authors are aware of the full content of the manuscript and provided consent for its submission to Advances in Rheumatology.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

do Nascimento, R., Quaio, C., Chung, C. et al. Principles of clinical genetics for rheumatologists: clinical indications and interpretation of broad-based genetic testing. Adv Rheumatol 64, 59 (2024). https://doi.org/10.1186/s42358-024-00400-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s42358-024-00400-z

Keywords