Fact-checked by Grok 2 weeks ago

RAG1

RAG1, also known as recombination activating gene 1, is a protein encoded by the RAG1 gene located on chromosome 11p12 in humans, forming part of the RAG protein complex alongside RAG2 that is essential for initiating V(D)J recombination in developing B and T lymphocytes. This process involves the precise rearrangement of variable (V), diversity (D), and joining (J) gene segments in the DNA, enabling the production of diverse antigen receptors that allow the adaptive immune system to recognize and respond to a wide array of pathogens. The RAG1 protein contains a catalytic domain with an RNase H fold structure that recognizes specific recombination signal sequences (RSSs) flanking the V, D, and J segments, introducing double-strand DNA breaks necessary for recombination, while RAG2 stabilizes the complex and ensures cleavage occurs only at appropriate sites during lymphocyte development. Discovered in 1989 through studies on V(D)J recombination mechanisms, RAG1 and RAG2 were identified as key enzymes expressed exclusively in immature B and T cells, highlighting their specialized role in adaptive immunity. The genes show high conservation across vertebrates, with human RAG1 sharing about 90% amino acid identity with its mouse counterpart, underscoring evolutionary importance for jawed vertebrate immunity originating from an ancient transposon insertion approximately 450 million years ago. In healthy individuals, functional RAG1 activity is critical for generating a functional repertoire of T-cell receptors (TCRs) and B-cell immunoglobulins, without which the immune system cannot mount effective antigen-specific responses. Mutations in the RAG1 gene are a major cause of primary immunodeficiencies, particularly (SCID), where complete loss of function leads to the absence of mature T and B cells, rendering infants highly susceptible to infections and often fatal within the first year of life without intervention such as or emerging gene therapies. Hypomorphic (partial loss-of-function) mutations can result in less severe conditions, including —characterized by erythroderma, , and —or combined immunodeficiencies with granulomas and , affecting cellular and humoral immunity into later childhood. Over 70 distinct RAG1 mutations have been linked to these disorders, emphasizing the gene's dosage sensitivity and the spectrum of phenotypes from complete to leaky SCID.

Discovery and Structure

Discovery

The RAG1 gene was identified in 1989 by David G. Schatz, Marjorie A. Oettinger, and David Baltimore at the Whitehead Institute for Biomedical Research, through a genetic screen aimed at isolating factors that could induce V(D)J recombination—a process essential for assembling immunoglobulin and T-cell receptor genes—in non-lymphoid cells such as fibroblasts. Using a transient transfection assay with a plasmid containing a substrate for V(D)J recombination, they cloned a lymphoid-specific cDNA, designated recombination activating gene 1 (RAG1), which weakly stimulated recombination signal cleavage when overexpressed alone. Building on this, in 1990, the same researchers discovered an adjacent gene, RAG2, which synergistically activates V(D)J recombination with RAG1. Key experiments demonstrated that co-transfection of RAG1 and RAG2 into non-lymphoid cells like 3T3 fibroblasts enabled efficient cleavage and rearrangement at recombination signal sequences (RSSs), mimicking the activity observed in immature lymphoid cells; neither gene alone was sufficient for robust recombination. This finding established RAG1 and RAG2 as core components required for initiating V(D)J recombination, a lymphocyte-specific process that generates antibody and T-cell receptor diversity. In 1992, the human RAG1 gene was mapped to the short arm of chromosome 11 at band p12 through fluorescence in situ hybridization (FISH) analysis, confirming its location near other immune-related loci. A major milestone came in 1992 with the generation of RAG1-deficient mice via targeted disruption of the gene, which resulted in animals lacking mature B and T lymphocytes due to a complete block in V(D)J recombination at early stages of lymphoid development; these mice exhibited small lymphoid organs and severe combined immunodeficiency (SCID)-like phenotypes. This genetic validation underscored RAG1's indispensable role in adaptive immunity.

Gene and Protein Structure

The human RAG1 gene is located on the short arm of at position 11p12 and spans approximately 83 kilobases (kb). The gene consists of two s, with only the second being protein-coding, and it encodes a protein of 1,040 with a predicted molecular weight of approximately 125 kDa. The RAG1 protein features distinct structural domains that contribute to its role in DNA recognition and catalysis. The N-terminal region includes zinc finger motifs, such as (residues 289–334) and a (residues 354–383), which are involved in DNA binding and protein interactions. The nonamer-binding domain (NBD, residues ~384–390) recognizes the nonamer sequence of recombination signal sequences (RSS). The core domain (residues 384–1,008) encompasses the catalytic region, including the RAG1 homeodomain and the DDE motif (aspartate D600, aspartate D708, glutamate E962), which forms the for endonuclease activity. Structural studies of RAG1 have advanced through cryo-electron microscopy (cryo-EM), revealing its architecture within the synaptic complex with RSS. Landmark cryo-EM structures from 2015 captured the zebrafish RAG1-RAG2 synaptic complex at resolutions up to 3.4 Å, showing a dimeric assembly with DNA bound in a closed conformation and base flipping at cleavage sites. More recent cryo-EM analyses in 2025 of nearly full-length mouse RAG1-RAG2 complexed with RSS at resolutions around 3.2–3.8 Å highlighted evolutionary adaptations that suppress transposition while enabling V(D)J recombination initiation. Post-translational modifications regulate RAG1 stability and activity. Autoubiquitination at lysine 233 (K233) in the N-terminal region, mediated by RAG1's intrinsic E3 ubiquitin ligase activity in the domain, enhances the DNA cleavage activity of the RAG1/RAG2 complex. Phosphorylation sites, including serine 528 (S528) targeted by (AMPK), modulate RAG1 function, while multiple SQ/TQ motifs (up to ten) serve as potential substrates for and kinases, influencing complex stability during DNA processing.

Biological Function

V(D)J Recombination Mechanism

V(D)J recombination is initiated by the protein, which functions as the catalytic core of the RAG recombinase complex, recognizing specific recombination signal sequences (RSSs) that flank the variable (V), diversity (D), and joining (J) gene segments in immunoglobulin and T-cell receptor loci. Each RSS consists of a conserved heptamer (CACAGTG), adjacent to the coding segment, followed by a spacer of either 12 or 23 base pairs, and a nonamer (ACAAAAACC) at the distal end; RAG1 primarily interacts with the nonamer and heptamer sequences through its core domain, facilitating specific binding via base flipping and hydrogen bonding with key residues such as Lys912 and Arg916. This recognition adheres to the 12/23 rule, ensuring that efficient recombination occurs only between RSSs with spacers of different lengths, preventing non-productive rearrangements. The cleavage process begins with synapsis, where RAG1, in complex with RAG2, brings together a pair of compatible RSSs (one 12-RSS and one 23-RSS) to form a synaptic complex, positioning the heptamer-coding junctions for cleavage. Cleavage proceeds in two enzymatic steps: first, nicking at the 5' end of the heptamer-coding junction introduces a single-strand break, generating a 3'-hydroxyl group on the coding flank; second, transesterification occurs as this 3'-hydroxyl attacks the opposing on the , forming a covalently sealed structure on the coding ends while producing blunt double-strand breaks at the signal ends. These blunt signal ends consist of the excised RSS sequences, which are 5'-phosphorylated and compatible for direct ligation. RAG1 acts as a transposase-like endonuclease, sharing structural and mechanistic similarities with bacterial transposases and retroviral integrases, including the use of a catalytic DDE triad composed of Asp600, Asp708, and Glu962 residues that coordinate divalent metal ions for phosphodiester bond hydrolysis and transesterification. This triad forms a single active site contributed in trans by one RAG1 subunit to the RSS-bound subunit, enabling precise DNA cleavage at the RSS borders. Following cleavage, the post-cleavage complex holds the hairpin coding ends and blunt signal ends, which must be resolved by the (NHEJ) pathway to complete recombination; coding ends are processed by endonuclease to open hairpins and add nucleotides via , while signal ends are directly ligated with high fidelity using , , XRCC4, and ligase IV. Release from the RAG complex requires phosphorylation by or to displace RAG proteins, ensuring preferential use of error-prone NHEJ for coding joins that generate junctional diversity. To prevent genomic instability, V(D)J recombination is tightly regulated to the G0/G1 phase of the cell cycle, primarily through cell cycle-dependent degradation of RAG2 at the G1-to-S transition via cyclin A-CDK2 phosphorylation at Thr490, which limits RAG1 activity to periods when NHEJ predominates over alternative repair pathways. This restriction ensures that double-strand breaks are repaired faithfully before DNA replication, avoiding aberrant translocations during S/G2/M phases.

Interactions with Other Proteins

RAG1 primarily functions in V(D)J recombination through its essential partnership with RAG2, forming a heterotetrameric RAG1/RAG2 complex that recognizes and cleaves recombination signal sequences (RSSs). RAG2 stabilizes RAG1's binding to DNA and confers specificity for the 12/23 rule, ensuring paired cleavage of RSSs with 12- and 23-base pair spacers. This interaction occurs via the core domains of both proteins, with RAG1's non-core N-terminal region further modulating complex stability and activity. Accessory proteins enhance the efficiency of RAG1-mediated cleavage and complex formation. High-mobility group proteins HMG1 and HMG2 bind to the minor groove of RSS DNA, inducing bends that facilitate between 12-RSS and 23-RSS substrates within the RAG1/RAG2 complex. Post-cleavage, the non-homologous end-joining (NHEJ) factors , Ku70, and Ku80 associate with the RAG1/RAG2-bound signal ends, stabilizing the synaptic complex and promoting repair while preventing aberrant joining. Regulatory interactions fine-tune RAG1 activity to maintain genomic stability during recombination. The kinase plays a key role in sensing RAG-induced double-strand breaks and coordinating their repair to inhibit off-target cleavage. The assembly of the RAG1/RAG2 complex follows an ordered process, with RAG1 initially binding RSS DNA before recruiting RAG2, which releases RAG1's autoinhibitory constraints to enable cleavage. This sequential binding, combined with HMG1/2-mediated DNA distortion, ensures precise regulation of the recombination signal.

Clinical Significance

Associated Diseases

Mutations in the RAG1 gene cause a spectrum of primary immunodeficiencies, primarily through disruption of V(D)J recombination, leading to impaired T and B cell development. The most severe form is autosomal recessive severe combined immunodeficiency (SCID), characterized by the absence of mature T and B lymphocytes, resulting in profound susceptibility to infections, failure to thrive, and early mortality without intervention. This T⁻B⁻NK⁺ SCID phenotype arises from null mutations that abolish RAG1 recombinase activity. Omenn syndrome, a variant of leaky SCID, results from hypomorphic RAG1 mutations that retain partial activity, allowing limited, oligoclonal T cell expansion but leading to immune dysregulation. Patients present with erythroderma, , elevated IgE levels, , and recurrent infections due to autoreactive T cells and impaired function. Hypomorphic RAG1 mutations can also cause delayed-onset combined immunodeficiencies, such as combined immunodeficiency with granulomas and/or (CID-G/AI), which manifest later in childhood or with granulomatous , autoimmune cytopenias, and chronic infections. Missense mutations associated with CID-G/AI are often located in the coding flank-sensitive region of RAG1. Over 200 pathogenic mutations in the RAG1 and RAG2 genes have been documented, with approximately 70% being missense variants, many affecting the core domain essential for recombinase activity. A notable example is the c.1304A>G (p.M435V) mutation in the core domain, which retains about 24% recombination activity and correlates with or CID-G/AI phenotypes. Functional assays from 2014, analyzing 79 variants, established strong genotype-phenotype correlations, showing that recombination activity levels predict clinical severity: near-zero activity for classic SCID, 1-10% for , and higher residual activity (>20%) for milder, delayed forms. Animal models, such as RAG1 knockout mice (Rag1⁻/⁻), recapitulate human SCID by exhibiting arrested B and T cell development, small lymphoid organs, and absence of mature lymphocytes, providing insights into disease pathophysiology and therapeutic testing.

Diagnostic and Therapeutic Approaches

Diagnosis of RAG1-related immunodeficiencies typically begins with clinical suspicion based on recurrent infections and failure to thrive in infancy, followed by immunological evaluation including flow cytometry to assess lymphocyte subsets, revealing characteristic T-B-NK+ severe combined immunodeficiency (SCID) patterns with absent or low T and B cells but normal natural killer cells. Genetic confirmation is achieved through next-generation sequencing (NGS) panels targeting primary immunodeficiency (PID) genes, which identify biallelic RAG1 mutations in approximately 4% of SCID cases. Functional assays, such as flow cytometry-based reporter systems measuring V(D)J recombination activity in patient-derived fibroblasts or cell lines, further validate the pathogenicity of identified variants by quantifying recombination efficiency. Newborn screening for SCID, implemented nationwide in the United States since 2010 following its addition to the Recommended Uniform Screening Panel, uses excision circle (TREC) assays on dried blood spots to detect low T-cell production, with RAG1/RAG2 deficiencies accounting for about 28.6% of identified SCID cases in programs like California's. Positive screens prompt confirmatory testing, enabling early diagnosis and intervention before infections occur. The primary curative treatment for RAG1-deficient SCID is allogeneic (HSCT), which restores adaptive immunity with overall survival rates of 65-95%, exceeding 90% when performed early (before 3.5 months of age) using HLA-matched donors. Emerging gene therapies, including lentiviral vector-mediated RAG1 gene addition to autologous hematopoietic stem cells, have shown immune reconstitution in preclinical models and are under evaluation in phase I/II human trials (e.g., NCT04797260), with initial safety data reported post-2023. As of August 2025, the trial is ongoing and recruiting, with recent preclinical studies demonstrating restored T-cell development in patient-derived cells using artificial thymic organoids. Therapeutic challenges include managing hypomorphic RAG1 variants, which cause partial and may require tailored HSCT conditioning or dosing to achieve sufficient RAG1 expression without toxicity. Gene editing approaches face risks of off-target double-strand breaks, potentially increasing genotoxicity and necessitating precise delivery systems like CRISPR-Cas9 with enhancers.

Evolutionary Aspects

Conservation and Evolution

The RAG1 gene originated from the domestication of a Transib transposon that integrated into the genome of the common ancestor of jawed vertebrates approximately 500 million years ago, during the emergence of adaptive immunity. This event involved the co-option of a RAG1-like transposase for catalyzing V(D)J recombination, marking a pivotal innovation in vertebrate evolution. RAG1 is absent in jawless vertebrates, such as lampreys and hagfish, which instead employ a proto-lymphocyte-based system lacking RAG-mediated recombination. Recent studies have identified RAG-like (RAGL) transposases in , providing "missing links" that illustrate the molecular events converting ancestral transposases into the RAG1–RAG2 complex in vertebrates. Sequence conservation of RAG1 is pronounced in its core domain (spanning 384–1043 in humans), which exhibits 65% identity and 77% similarity between distantly related vertebrates like and humans, reflecting its essential role in DNA cleavage and recombination. This high conservation extends across mammals, birds, reptiles, amphibians, and bony fish, with identity levels ranging from 70–90% within these groups for key functional motifs, including the DDE . In contrast, the N-terminal non-core region ( 1–383) displays greater sequence variability, with lower identity (around 40–60%) across species, allowing for lineage-specific regulatory adaptations while preserving overall protein architecture. The evolutionary assembly of the RAG locus involved the acquisition of an ancestral RAG2-like by the Transib transposon encoding RAG1, forming a tightly linked gene pair without evidence of subsequent duplication between RAG1 and RAG2; this cluster has been maintained intact across all lineages. No complete losses of RAG1 occur in major clades, though partial divergence is observed in some teleost fish due to whole-genome duplications, resulting in paralogous copies with retained core function. Functionally, ancestral RAG1 acted as a mobile facilitating DNA , but in modern s, it has diverged to function primarily as a site-specific endonuclease, suppressing while enhancing recombination fidelity at RSS sites.

Applications in Phylogenetics

RAG1 serves as a valuable in , particularly for resolving deep evolutionary s among vertebrates due to its single-copy nature and conserved structure. The gene's approximately 1 kb region is commonly amplified using universal primers, enabling broad applicability across diverse taxa such as tetrapods. For instance, a comprehensive phylogeny of tetrapods was constructed using nearly complete RAG1 sequences from species spanning all major clades, providing robust estimates of diversification patterns and divergence times. In various studies, RAG1 sequences have been combined with RAG2 or (mtDNA) to enhance phylogenetic resolution, particularly in addressing complex relationships within and lineages. During the , RAG1 data played a key role in clarifying the phylogeny of , a diverse of spiny-rayed fishes, by testing alternative sister-group hypotheses for tetraodontiforms and supporting monophyly of major orders through Bayesian and analyses. Similarly, in phylogenetics, RAG1 has been integrated with mtDNA to reconcile mito-nuclear discordance, as seen in analyses of horned lizards (Phrynosoma), where nuclear loci like RAG1 revealed patterns of incomplete lineage sorting or ancient that conflicted with mitochondrial trees. The primary advantages of RAG1 in phylogenetic applications stem from its slow-evolving core domains, which are suitable for inferring ancient splits without issues, and its lack of paralogous copies, reducing ambiguity in assignment compared to multigene families. These features have made it a standard nuclear locus for complementing faster-evolving mtDNA markers in . However, limitations include the relatively short amplifiable sequences (typically ~1 kb), which may limit resolution at shallow divergences, and sparse data availability for non- taxa due to RAG1's absence outside vertebrates. Recent phylogenomic analyses have begun integrating RAG1 with whole-genome data to overcome these constraints and improve accuracy in time-calibrated trees.

References

  1. [1]
    RAG1 gene: MedlinePlus Genetics
    Feb 1, 2017 · The RAG1 gene provides instructions for making a member of a group of proteins called the RAG complex. This complex is active in immune system cells ( ...
  2. [2]
    5896 - Gene ResultRAG1 recombination activating 1 [ (human)] - NCBI
    Aug 19, 2025 · Study reports that RAG1 associates with chromatin at thousands of active promoters and enhancers in the genome of developing lymphocytes. The ...
  3. [3]
    179615 - RECOMBINATION-ACTIVATING GENE 1; RAG1 - OMIM
    RAG1 contains the RNase H fold catalytic domain and regions that make direct contact with RSSs and is responsible for the enzymatic activity of the RAG complex.
  4. [4]
    The V(D)J recombination activating gene, RAG-1 - PubMed - NIH
    Dec 22, 1989 · The V(D)J recombination activating gene, RAG-1. Cell. 1989 Dec 22 ... D G Schatz , M A Oettinger, D Baltimore. Affiliation. 1 Whitehead ...
  5. [5]
    RAG-1 and RAG-2, Adjacent Genes That Synergistically Activate V ...
    The recombination activating gene, RAG-1, which is a gene expressed exclusively in maturing lymphoid cells, was previously isolated. RAG-1 inefficiently induced ...
  6. [6]
    Sequence and chromosome assignment to 11p13-p12 of ... - PubMed
    Mapping analysis of RAG-1 and RAG-2 genes on human chromosomes by fluorescence in situ hybridization indicated that the genes are located on chromosome 11p13- ...Missing: localization | Show results with:localization
  7. [7]
    RAG-1-deficient mice have no mature B and T lymphocytes - PubMed
    RAG-1-deficient mice have small lymphoid organs that do not contain mature B and T lymphocytes. The arrest of B and T cell differentiation occurs at an early ...Missing: 1993 | Show results with:1993
  8. [8]
  9. [9]
    RAG1 Gene - GeneCards | RAG1 Protein | RAG1 Antibody
    RAG1 (Recombination Activating 1) is a Protein Coding gene. Diseases associated with RAG1 include Severe Combined Immunodeficiency, Autosomal Recessive, T Cell ...
  10. [10]
    RAG1 - V(D)J recombination-activating protein 1 | UniProtKB - UniProt
    Nov 30, 2010 · Catalytic component of the RAG complex, a multiprotein complex that mediates the DNA cleavage phase during V(D)J recombination.Missing: exons | Show results with:exons
  11. [11]
    Mapping and Quantitation of the Interaction between the ...
    Mini-RAG1 consists primarily of the catalytic center and the residues N-terminal to it, but it lacks a zinc finger region in RAG1 previously implicated in ...
  12. [12]
    How RAG1/2 evolved from ancestral transposases to initiate V(D)J ...
    To investigate how RAG1/2 suppresses transposition and regulates DNA cleavage, we determined cryo-electron microscopy (cryo-EM) structures of nearly full-length ...
  13. [13]
    Role of RAG1 autoubiquitination in V(D)J recombination - PNAS
    Here we show that ubiquitination of RAG1, which can be promoted by RAG1's own ubiquitin ligase activity, is a powerful activator of the DNA cutting and ...
  14. [14]
    Metabolic sensor AMPK directly phosphorylates RAG1 protein and ...
    May 28, 2013 · The S528A mutation completely abolished AMPK phosphorylation (Fig. 2A), suggesting that in vitro S528 is the AMPK phosphorylation site in RAG1.
  15. [15]
    Repair of Chromosomal RAG-Mediated DNA Breaks by Mutant RAG ...
    RAG2 possesses three SQ/TQ motifs, and RAG1 has ten, any of which could be phosphorylated by ATM and/or DNA-PKcs to modulate RAG function during the repair ...
  16. [16]
    Molecular Mechanism of V(D)J Recombination from Synaptic RAG1 ...
    We report cryo-electron microscopy structures of synaptic RAG complexes at up to 3.4 Å resolution, which reveal a closed conformation with base flipping.Missing: 2019-2023 | Show results with:2019-2023
  17. [17]
    The DDE Motif in RAG-1 Is Contributed in trans to a Single Active ...
    Recently, a DDE triad of amino acids essential for the cleavage steps of V(D)J recombination has been identified in RAG-1 (Asp-600, Asp-708, and Glu-962); no ...
  18. [18]
    V(D)J Recombination: Recent Insights in Formation of the ...
    Apr 29, 2022 · Here, we will describe how the cell ensures a proper transition from the RAG-bound post-cleavage complex to the NHEJ repair complex. We also ...
  19. [19]
    COUPLING OF V(D)J RECOMBINATION TO THE CELL CYCLE ...
    V(D)J gene segment recombination is linked to the cell cycle by the periodic phosphorylation and destruction of the RAG-2 protein at the G1-to-S cell cycle ...
  20. [20]
    Distinct Roles of RAG1 and RAG2 in Binding the V(D)J ... - NIH
    The RAG1 and RAG2 proteins initiate V(D)J recombination by introducing double-strand breaks at the border between a recombination signal sequence (RSS) and ...
  21. [21]
    The recombinase activating genes: architects of immune diversity ...
    The RAG1 gene is situated on chromosome 11p13 of the human genome and encodes the RAG1 protein which comprises 1.043 amino acids (aa) (6–8). The RAG1 protein ...
  22. [22]
    The RAG1 Homeodomain Recruits HMG1 and HMG2 To Facilitate ...
    Several steps of the V(D)J recombination reaction can be reconstituted in vitro with only RAG1/2 plus the high-mobility-group protein HMG1 or HMG2. Here we show ...
  23. [23]
    Evidence for Ku70/Ku80 association with full-length RAG1 - PMC
    Genetic and biochemical evidence suggest that the RAG proteins play an active role in guiding the repair of DNA breaks introduced during V(D)J recombination to ...Missing: ATM PARP1
  24. [24]
    RAG1 and RAG2 Form a Stable Postcleavage Synaptic Complex ...
    A stable complex is formed postcleavage between synapsed recombination signals. This complex includes the proteins RAG1, RAG2, HMG-1 or the closely related HMG ...
  25. [25]
    RAG-Mediated DNA Double Stranded Breaks: Response & Repair
    During V(D)J recombination, Ku70 and Ku80 are essential for both signal and coding joint formation, whereas DNA-PKcs is essential only for coding joint ...Missing: HMG1/ | Show results with:HMG1/
  26. [26]
    ATM and 53BP1 regulate alternative end joining–mediated V(D)J ...
    ATM and Parp1 separately support the residual recombination fidelity of coding ends by A-EJ. ATM also suppresses excessive distal V-J recombination and ...
  27. [27]
    Autoinhibition of DNA cleavage mediated by RAG1 and RAG2 ... - NIH
    Dec 13, 2010 · Gene assembly of the variable domain of antigen receptors is initiated by DNA cleavage by the RAG1–RAG2 protein complex at sites flanking V, ...
  28. [28]
    Stimulation of V(D)J cleavage by high mobility group proteins
    We show that the DNA‐bending proteins HMG1 and HMG2 stimulate cleavage and RAG protein binding at the 23 bp spacer signal.
  29. [29]
    RAG Deficiency: Two Genes, Many Diseases - PMC - PubMed Central
    RAG gene defects in humans are associated with a surprisingly broad spectrum of clinical and immunological phenotypes.
  30. [30]
  31. [31]
    An Immunodeficiency Disease with RAG Mutations and Granulomas
    May 8, 2008 · The Omenn syndrome can have a fatal outcome despite stem-cell transplantation. We describe a type of primary immunodeficiency disease with RAG1 ...
  32. [32]
    Flow Cytometry for Diagnosis of Primary Immune Deficiencies—A ...
    Sep 11, 2019 · This review focuses on use of flow cytometry in disease-specific diagnosis of PIDDs in the context of a developing country.
  33. [33]
    RAG1/RAG2 Deficiency - Immunodeficiency Search
    RAG1 or RAG2 deficiency causes a form of autosomal recessive TB-NK+ SCID. It accounts for approximately 4% of all SCID cases.
  34. [34]
    RAG deficiencies: Recent advances in disease pathogenesis and ...
    The extended phenotype associated with hypomorphic RAG mutations includes also combined immune deficiency (CID), which is often associated with granulomas and/ ...
  35. [35]
    The Clinical and Genetic Spectrum of 82 Patients With RAG ...
    Jun 9, 2020 · Sixty-seven (81.7%) patients carried RAG1 and 15 patients (18.3%) carried RAG2 biallelic variants. We estimate that the minimal annual incidence ...
  36. [36]
    Multiomics dissection of human RAG deficiency reveals distinctive ...
    Jan 10, 2025 · By analyzing numerous naturally occurring RAG variants, we expanded on the genotype-phenotype correlation, confirming that CID and LS phenotypes ...
  37. [37]
    SCID Newborn Screening: What we've learned - PMC
    In 2010, severe combined immunodeficiency (SCID) was added to the national list of conditions for which newborn screening (NBS) should be conducted, called the ...
  38. [38]
    History and Current Status of Newborn Screening for Severe ...
    SCID was the first immunodeficiency disease to be added to the Recommended Uniform Screening Panel of Core Conditions in the United States in 2010, and is now ...
  39. [39]
    Gene therapy strategies for RAG1 deficiency: Challenges and ...
    Mutations in the recombination activating genes (RAG) cause various forms of immune deficiency. Hematopoietic stem cell transplantation (HSCT) is the only cure ...
  40. [40]
    Partial correction of immunodeficiency by lentiviral vector gene ...
    Nov 13, 2023 · Lentiviral vector (LV) gene therapy has been shown to obtain effective RAG1 expression and immune reconstitution in Rag1-/- mice using a MND-c. ...
  41. [41]
    Phase I/II Clinical Trial Stem Cell Gene Therapy in RAG1-Deficient ...
    Currently, the only curative treatment option for RAG-deficient SCID is allogeneic hematopoietic stem cell transplantation (HSCT). Despite improvements in HSCT ...
  42. [42]
    RAG1 lentiviral gene therapy restores T cell development ... - PubMed
    Aug 19, 2025 · Our phase I/II clinical trial (NCT04797260) is currently evaluating lentiviral RAG1 gene addition in autologous hematopoietic stem and ...
  43. [43]
    Partial correction of immunodeficiency by lentiviral vector gene ...
    Nov 12, 2023 · Our work shows that GT could partially alleviate the combined immunodeficiency of hypomorphic RAG1 patients and that extensive efficacy and safety studies
  44. [44]
    The origins of the RAG genes – from transposition to V(D)J ...
    This review will focus on the evolutionary origin of the Rag1/2 proteins, and how they evolved from a mobile DNA element into a sophisticated and tightly ...
  45. [45]
  46. [46]
    Collaboration of RAG2 with RAG1-like proteins during the evolution ...
    Apr 7, 2016 · We propose a two-step model in which a Transib element acquired an ancestral RAG2 gene, leading to a “RAG transposon” containing both RAG1 and ...
  47. [47]
    The Recombination Activation Gene 1 (RAG1) of Rainbow Trout ...
    When compared with other previously reported RAG1 sequences, the predicted amino acid translation (1073 aa) displayed a minimum of 78% similarity for the ...
  48. [48]
    Elucidating the domain architecture and functions of non-core RAG1
    May 20, 2011 · (A) Sequence conservation ... A WW-like module in the RAG1 N-terminal domain contributes to previously unidentified protein-protein interactions.
  49. [49]
    Evolution of the RAG1-RAG2 locus: both proteins came from the ...
    Apr 28, 2015 · The evolutionary origin of RAG1 from a Transib element was previously established, but a transposon encoding both RAG1 and RAG2 had never been ...Missing: conservation | Show results with:conservation
  50. [50]
    The Development of Three Long Universal Nuclear Protein-Coding ...
    For example, RAG1, a representative NPCL marker, has been successfully used to make phylogenetic inferences within all major osteichthyan groups. However, such ...
  51. [51]
    Calibration Choice, Rate Smoothing, and the Pattern of Tetrapod ...
    A phylogeny of tetrapods is inferred from nearly complete sequences of the nuclear RAG-1 gene sampled across 88 taxa encompassing all major clades.
  52. [52]
    A molecular test of alternative hypotheses of tetraodontiform ...
    The single-copy nuclear (scn) gene RAG1 was selected for use in this study because of its demonstrated utility in reconstructing phylogenies for different ...
  53. [53]
    Base composition, selection, and phylogenetic significance of indels ...
    Dec 16, 2009 · The Recombination Activating Proteins, RAG1 and RAG2, play a crucial role in the immune response in vertebrates. Among the nuclear markers ...
  54. [54]
    Time-calibrated phylogeny of neotropical freshwater fishes - Frontiers
    Dec 6, 2024 · A phylogenetic analysis of the major groups of catfishes (Teleostei: siluriformes) using rag1 and rag2 nuclear gene sequences. Mol ...