RAD51D (1-328) Human

RAD51D (1-328 a.a.) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles in DNA Repair

RAD51D (1-328) is indispensable for HR-mediated DSB repair and replication fork stability . Its roles include:

  • BCDX2 Complex Formation: Partners with RAD51B, RAD51C, and XRCC2 to stabilize RAD51 filaments and facilitate homology search .

  • Telomere Maintenance: Protects telomeres from attrition and fusion, preventing chromosomal instability .

  • Error-Free Repair: Mitigates replication stress by resolving stalled forks .

Mechanistic Insight: RAD51D deficiency shifts DSB repair toward error-prone non-homologous end joining (NHEJ), leading to genomic scars like large deletions .

Clinical Relevance and Mutational Patterns

Germline RAD51D mutations are linked to inherited cancer syndromes, particularly breast-ovarian cancer (BROVCA4) . Key findings include:

Common Mutations in High-Risk Populations

A founder mutation, c.270_271dupTA; p.(Lys91Ilefs*13), is prevalent in Asian populations. This frameshift creates a truncated protein (91 aa) with:

  • Loss of Function: Impaired RAD51 recruitment and HR deficiency .

  • Variable Penetrance: Incomplete HRD positivity (67% HRD+ in tumors) .

Table 2: RAD51D Mutations and Clinical Impacts

MutationProtein EffectClinical ImpactHRD ScoreReferences
c.270_271dupTA; p.Lys91Ilefs*13Truncated (91 aa)Increased cancer risk, HRD+ in some cases67% HRD+
c.556C>T; p.Arg186*Premature stopLoss of function, HRD-Negative
c.801delC; p.Trp268Glyfs*42Frameshift, truncatedLoss of function, HRD-Negative

Prevalence in Ovarian Cancer

In Chinese ovarian cancer cohorts:

  • Mutation Rate: 2.9% (11/380 patients) .

  • BILOF Dominance: 72.7% of mutations were bi-allelic loss-of-function (BILOF), correlating with improved chemotherapy response .

Table 3: RAD51D in Ovarian Cancer Studies

Study PopulationMutation RateBILOF CasesPFS ImprovementReferences
Chinese FACT cohort2.9% (11/380)72.7% (8/11)HR 0.375 (P=0.025)

Homologous Recombination Deficiency (HRD)

  • HRD Assays: Tumors with RAD51D mutations show variable HRD scores, complicating therapeutic predictions .

  • Nonsense-Mediated Decay (NMD): The c.270_271dupTA mutation triggers NMD, reducing RAD51D protein levels .

Chemotherapy Sensitivity

RAD51D-mutated tumors exhibit enhanced sensitivity to platinum-based chemotherapy, particularly in BRCA1/2 wild-type patients .

Therapeutic Implications

  • PARP Inhibitors: RAD51D-deficient tumors may benefit from PARP inhibitors, though HRD status must be confirmed .

  • Biomarker Potential: RAD51D expression and HRD scores could guide personalized treatment strategies .

Product Specs

Introduction
RAD51D belongs to the RAD51 protein family, which shares significant similarity with bacterial RecA and Saccharomyces cerevisiae Rad51. These proteins play a crucial role in DNA repair and homologous recombination. Specifically, RAD51D interacts with other RAD51 family members to form a complex that facilitates homologous pairing between single- and double-stranded DNA, contributing to the early stages of DNA recombinational repair.
Description
RAD51D (1-328) Human Recombinant is a protein produced in E. coli. It is a single polypeptide chain consisting of 351 amino acids, with a molecular weight of 37.4kDa. A 23 amino acid His-tag is fused to the N-terminus of the protein. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized through filtration.
Formulation
The RAD51D (1-328) solution is provided at a concentration of 0.25mg/ml. It is formulated in a buffer containing 20mM Tris-HCl (pH 8.0), 0.1M NaCl, 10% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the solution can be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To ensure long-term stability, adding a carrier protein like HSA or BSA (0.1%) is advisable. Avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of the RAD51D (1-328) protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
BROVCA4, R51H3, RAD51L3, TRAD, DNA repair protein RAD51 homolog 4, RAD51 homolog D, RAD51-like protein 3, RAD51D.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMGVLRVG LCPGLTEEMI QLLRSHRIKT VVDLVSADLE EVAQKCGLSY KALVALRRVL LAQFSAFPVN GADLYEELKT STAILSTGIG SLDKLLDAGL YTGEVTEIVG GPGSGKTQVC LCMAANVAHG LQQNVLYVDS NGGLTASRLL QLLQAKTQDE EEQAEALRRI QVVHAFDIFQ MLDVLQELRG TVAQQVTGSS GTVKVVVVDS VTAVVSPLLG GQQREGLALM MQLARELKTL ARDLGMAVVV TNHITRDRDS GRLKPALGRS WSFVPSTRIL LDTIEGAGAS GGRRMACLAK SSRQPTGFQE MVDIGTWGTS EQSATLQGDQ T.

Q&A

What is RAD51D and what is its primary function in human cells?

RAD51D is one of five RAD51 paralog members (alongside RAD51B, RAD51C, XRCC2, and XRCC3) that plays a crucial role in DNA double-strand break (DSB) repair through homologous recombination (HR). It assists in the recruitment of RAD51, which functions as a DNA recombinase during DNA replication and is essential for maintaining genomic stability . Within cellular pathways, RAD51D forms a specific subcomplex with RAD51B, RAD51C, and XRCC2, commonly referred to as the BCDX2 complex, which is regulated by the BRCA1-PALB2-BRCA2 effector complex upstream of RAD51 . This molecular cooperation is fundamental to error-free DNA repair and chromosome integrity maintenance during cellular replication and in response to DNA damage. Understanding this primary function provides the essential foundation for more advanced investigations into RAD51D's role in pathological conditions.

How does RAD51D (1-328) differ from other RAD51 paralogs in structure and function?

RAD51D (1-328) represents a specific region of the human RAD51D protein that contains functionally important domains for its DNA repair activities. While all RAD51 paralogs share some structural similarities, RAD51D exhibits unique properties in its DNA binding capabilities and protein-protein interactions that distinguish it from RAD51B, RAD51C, XRCC2, and XRCC3 . Structurally, RAD51D contains domains necessary for its participation in the BCDX2 complex formation, which is critical for its function in homologous recombination. Unlike some other paralogs, RAD51D has been specifically implicated in ovarian cancer susceptibility, with studies showing approximately a six-fold increase in ovarian cancer risk in individuals with germline RAD51D variants . Its role appears to be particularly important in gynecological malignancies, suggesting tissue-specific functions that are not equally shared among all paralogs. Research into RAD51D's specific interaction networks may further elucidate its unique contributions to cellular processes beyond the common functions shared with other RAD51 family members.

What is the prevalence of RAD51D mutations in cancer populations?

The prevalence of RAD51D mutations varies across different cancer types and populations. In a comprehensive study of Chinese ovarian cancer patients, 2.9% (11/380) harbored RAD51D mutations, making it a significant non-BRCA contributor to homologous recombination deficiency (HRD) in this population . Among these mutations, the majority (72.7% or 8/11) were bi-allelic loss-of-function (BILOF) mutations, suggesting complete inactivation of the protein in these cases . Interestingly, epidemiological studies have noted that RAD51D mutations are mutually exclusive with BRCA1/2 mutations at the personnel level, indicating separate pathways of oncogenesis . The frequency of RAD51D mutations positions it as the third most commonly mutated homologous recombination repair gene after BRCA1 and BRCA2 in some populations, with approximately half the mutation frequency of BRCA2 . The clinical significance of these mutations extends beyond prevalence alone, as they correlate with treatment response and may serve as biomarkers for targeted therapies such as PARP inhibitors.

How should researchers design experiments to assess RAD51D function in DNA repair?

Designing robust experiments to assess RAD51D function requires a multifaceted approach combining molecular, cellular, and functional assays. When investigating RAD51D's role in DNA repair, researchers should first consider using CRISPR-Cas9 gene editing to create isogenic cell lines with wild-type and mutant RAD51D variants, including specific point mutations identified in clinical samples . Cell-based assays measuring homologous recombination efficiency, such as DR-GFP (Direct Repeat Green Fluorescent Protein) reporter assays, provide quantitative measurements of HR repair capacity in the presence of different RAD51D variants. For protein-level analysis, co-immunoprecipitation experiments can elucidate RAD51D's interactions with other members of the BCDX2 complex and the broader HR machinery . The formation of RAD51 foci after DNA damage induction (using agents such as ionizing radiation or cisplatin) can be monitored by immunofluorescence microscopy to assess RAD51D's role in RAD51 recruitment. For more comprehensive analysis, researchers should combine these approaches with next-generation sequencing to identify genome-wide effects of RAD51D dysfunction on mutation patterns and chromosomal aberrations. Such integrative approaches provide more complete insights into RAD51D's functional importance in maintaining genomic stability.

What are the recommended methods for detecting and characterizing RAD51D variants in clinical samples?

Detection and characterization of RAD51D variants in clinical samples require specialized methodologies that balance sensitivity, specificity, and clinical applicability. The current gold standard involves next-generation sequencing (NGS) approaches, either as part of multi-gene panels targeting homologous recombination repair genes or through whole exome sequencing when broader genomic evaluation is desired . For germline variant detection, peripheral blood samples provide reliable DNA sources, while tumor samples are essential for somatic mutation identification. Importantly, researchers should implement bioinformatic pipelines capable of detecting various mutation types, including single nucleotide variants, small insertions/deletions, copy number variations, and most critically for RAD51D, splice site mutations that may affect RNA processing . The case report of a RAD51D c.904-2A>T splicing variant demonstrates the importance of comprehensively evaluating intronic regions within ±2 bases from exon-intron boundaries . Functional characterization should follow variant detection, incorporating RNA analysis to confirm splicing alterations, protein expression studies to assess stability, and ideally, functional assays measuring homologous recombination efficiency. Classification of variants should adhere to American College of Medical Genetics and Association for Molecular Pathology (ACMG-AMP) guidelines, with particular attention to computational predictions for splice site variants .

How can researchers accurately measure homologous recombination deficiency (HRD) in relation to RAD51D status?

Accurately measuring homologous recombination deficiency in relation to RAD51D status requires a multi-parameter approach that captures the genomic consequences of defective HR repair. Current methodologies employ HRD scoring systems that quantify genomic scars characteristic of defective homologous recombination, including loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST) . In research settings, these parameters are often combined into composite HRD scores, as exemplified in the EOC-HRD study which demonstrated that RAD51D mutations, particularly bi-allelic loss-of-function mutations, correlate with elevated HRD scores . When establishing experimental systems, researchers should include positive controls (such as BRCA1/2 mutated samples) and negative controls to calibrate HRD score thresholds. Functional assays that directly measure HR capacity, such as RAD51 foci formation after DNA damage induction, provide complementary information to genomic scar analysis . For comprehensive assessment, researchers should integrate these approaches with transcriptomic and proteomic analyses to capture compensatory mechanisms that may modulate the HRD phenotype despite RAD51D mutations. This integrated approach provides more nuanced insights into the relationship between RAD51D status and homologous recombination deficiency than any single measurement alone.

How do RAD51D (1-328) mutations influence treatment response and clinical outcomes?

RAD51D mutations significantly impact treatment response and clinical outcomes, particularly in the context of DNA-damaging therapies. Research indicates that tumors harboring RAD51D mutations demonstrate increased sensitivity to platinum-based chemotherapy regimens, which function by creating DNA crosslinks that are particularly lethal to cells with homologous recombination deficiencies . In the EOC-HRD FACT cohort study, RAD51D mutations served as strong predictors of first-line chemotherapy efficacy, with mutated cases showing improved progression-free survival compared to wild-type cases . This treatment sensitivity extends beyond conventional chemotherapy to targeted therapies such as poly (ADP-ribose) polymerase (PARP) inhibitors, which exploit synthetic lethality in HR-deficient cells . The clinical implications are substantial, as identification of RAD51D mutations may guide treatment selection and predict therapeutic outcomes. Interestingly, the type of mutation appears relevant, with bi-allelic loss-of-function mutations potentially conferring greater sensitivity than monoallelic alterations . Future research should address whether different domains within the RAD51D protein (such as the 1-328 region) have differential impacts on treatment response when mutated, and whether these effects vary across cancer types. Longitudinal studies tracking the acquisition of resistance mechanisms in RAD51D-mutated tumors would provide valuable insights into optimizing treatment strategies for these patients.

What is the molecular mechanism by which RAD51D variants contribute to cancer development?

The molecular mechanisms underlying RAD51D-mediated carcinogenesis involve complex disruptions to DNA repair pathways that ultimately lead to genomic instability. RAD51D functions within the BCDX2 complex to facilitate homologous recombination repair, and pathogenic variants typically lead to impaired DNA double-strand break repair capacity . When RAD51D function is compromised through germline or somatic mutations, cells accumulate unrepaired DNA damage and chromosomal aberrations over time. This genomic instability creates a permissive environment for additional oncogenic mutations and structural variations that drive malignant transformation . At the molecular level, loss of functional RAD51D impairs the recruitment of RAD51 to DNA damage sites, disrupting the formation of nucleoprotein filaments essential for strand invasion during homologous recombination . The resultant shift toward error-prone repair mechanisms, such as non-homologous end joining (NHEJ), further accelerates mutational accumulation. Interestingly, the case report of leiomyosarcoma with a RAD51D splicing variant suggests that the oncogenic impact extends beyond epithelial malignancies typically associated with HR deficiency . The interaction with other genetic alterations, such as TP53 mutations which may affect RAD51 expression regulation, creates complex molecular landscapes that influence the specific cancer phenotypes associated with RAD51D variants . This complexity necessitates comprehensive molecular profiling to fully understand the oncogenic mechanisms in individual cases.

What are the tissue-specific effects of RAD51D dysfunction in human physiology?

RAD51D dysfunction exhibits intriguing tissue-specific effects that may explain its differential impact on cancer risk across organ systems. Research has consistently demonstrated that germline RAD51D variants confer the highest risk for ovarian cancer, with an estimated six-fold increase compared to the general population . This ovarian tissue tropism suggests specialized functions or vulnerabilities related to RAD51D in the fallopian tube and ovarian epithelium. Several hypotheses may explain this tissue specificity: the hormonal microenvironment of reproductive tissues may influence DNA repair processes; tissue-specific transcriptional regulation of RAD51D or its partners could create vulnerabilities; or the high proliferative demands of certain epithelia may exacerbate the consequences of HR deficiency . Beyond ovarian cancer, emerging evidence suggests associations with breast cancer risk regardless of subtype, indicating broader implications in estrogen-responsive tissues . The reported case of peritoneal leiomyosarcoma harboring a germline RAD51D splicing variant represents the first documented association with non-epithelial sarcomas, potentially expanding our understanding of RAD51D's tissue-specific effects to mesenchymal tissues . This suggests that RAD51D's role may not be confined to epithelial cancers but could extend to various tissue types under specific genetic or environmental contexts. Future research employing tissue-specific conditional knockout models could further elucidate the differential dependencies on RAD51D-mediated DNA repair across human tissues.

How should RAD51D variants be interpreted and classified in clinical genetic testing?

Interpretation and classification of RAD51D variants in clinical genetic testing require rigorous application of established guidelines while incorporating emerging research findings. According to the American College of Medical Genetics and Genomics (ACMG) and Association for Molecular Pathology (AMP) standards, variants affecting splice sites (particularly those within ±2 bases from exon-intron boundaries) provide strong evidence for pathogenicity as they typically lead to loss of function through impaired splicing . As exemplified by the RAD51D c.904-2A>T case, computational predictions for splicing disruption can provide additional evidence for variant classification . Population frequency data are essential for interpretation, with pathogenic variants typically occurring at very low frequencies in population databases such as gnomAD, HGVD, and ToMMo (see Table 1 for allele frequencies) .

Table 1: Allele Frequencies of RAD51D c.904-2A>T in Population Databases

DatabasePopulationAllele numberAllele frequency
HGVDJapanese2,4204.13 × 10^-4
GEM-J WGAJapanese15,1626.60 × 10^-4
ToMMoJapanese16,7606.60 × 10^-4
KRGDBKorean2,9223.00 × 10^-4
gnomADEast Asian18,3945.44 × 10^-5
gnomADGlobal251,4883.98 × 10^-6
TOPMedGlobal264,6904.00 × 10^-6

Clinical correlation with personal and family cancer history provides additional evidence for variant classification, although the absence of family history does not exclude pathogenicity, as demonstrated in several documented cases (see Table 2) . Importantly, genetic counseling should be offered to relatives of individuals with pathogenic RAD51D variants, as exemplified by the case where two daughters of a patient with leiomyosarcoma were found to carry the same pathogenic variant . This approach enables appropriate risk management and surveillance, potentially extending to considerations for non-epithelial tumors based on emerging evidence.

What are the recommendations for surveillance and risk management in individuals with RAD51D mutations?

Surveillance and risk management recommendations for individuals with pathogenic RAD51D variants must balance cancer risk reduction with quality of life considerations. Current evidence suggests that RAD51D mutation carriers have an estimated 11-13% cumulative risk of tubo-ovarian cancer by age 80, placing them at moderate risk compared to the general population . For these individuals, gynecologic surveillance should include transvaginal ultrasound and CA-125 measurements, typically beginning at age 30-35 years or 5-10 years earlier than the youngest diagnosis in the family . Risk-reducing salpingo-oophorectomy may be considered after completion of childbearing or around age 45-50, though optimal timing remains under investigation. Breast cancer surveillance recommendations are evolving as data accumulate on the association between RAD51D variants and breast cancer risk, with current approaches including annual mammography and consideration of breast MRI beginning at age 40 or younger based on family history . The novel association with leiomyosarcoma raises questions about whether additional surveillance measures should be considered, though current evidence is insufficient to recommend specific protocols for non-epithelial tumors . As with all cancer predisposition syndromes, management should be individualized through multi-disciplinary consultation, incorporating personal and family history, variant-specific risk assessments, and patient preferences. Future research should focus on developing tailored screening protocols that account for the specific spectrum of cancers associated with RAD51D mutations.

How can RAD51D status be leveraged for precision medicine approaches in cancer treatment?

RAD51D status represents a promising biomarker for precision medicine approaches in cancer treatment, particularly in the context of DNA-damaging therapies and targeted agents. Research demonstrates that tumors with RAD51D mutations exhibit elevated homologous recombination deficiency (HRD) scores, suggesting impaired DNA repair capacity that can be therapeutically exploited . This mechanistic vulnerability translates to clinical benefit, as evidenced by improved progression-free survival in RAD51D-mutated ovarian cancer patients receiving platinum-based chemotherapy . Beyond conventional chemotherapy, RAD51D status has significant implications for targeted therapy selection, particularly poly (ADP-ribose) polymerase (PARP) inhibitors . These agents, which trap PARP on DNA and prevent single-strand break repair, create synthetic lethality in HR-deficient cells, providing a rational therapeutic approach for RAD51D-mutated tumors . Clinical trials incorporating RAD51D testing alongside other HRR genes are ongoing, suggesting that a broader "HRR3" gene panel (including BRCA1, BRCA2, and RAD51D) may identify additional patients likely to benefit from PARP inhibition . To maximize the utility of RAD51D as a biomarker, researchers should consider both germline and somatic testing, distinguish between monoallelic and biallelic alterations (as biallelic mutations may confer greater sensitivity), and integrate RAD51D status with other genomic biomarkers . The future of RAD51D-informed precision medicine likely includes combination strategies that target multiple vulnerabilities simultaneously, potentially overcoming resistance mechanisms that emerge during treatment.

What are the emerging technologies that could advance RAD51D research?

Emerging technologies are poised to revolutionize RAD51D research across multiple dimensions, from structural biology to clinical applications. CRISPR-based technologies beyond gene editing, such as base editors and prime editors, offer unprecedented precision for modeling specific RAD51D variants without introducing double-strand breaks that might confound experimental interpretation . These approaches could create isogenic cell models that faithfully recapitulate patient-derived variants. Advances in cryo-electron microscopy and AlphaFold-based protein structure prediction are illuminating the three-dimensional architecture of RAD51D and its complexes, providing structural insights into how variants disrupt protein function . Single-cell sequencing technologies allow researchers to assess the heterogeneity of RAD51D expression and function within tumors, potentially revealing subpopulations with differential therapy responses. For clinical applications, liquid biopsy approaches that detect circulating tumor DNA harboring RAD51D mutations offer minimally invasive methods for monitoring treatment response and resistance development . Advanced computational methods integrating machine learning could improve variant classification and treatment response prediction based on RAD51D status and broader genomic contexts. Long-read sequencing technologies facilitate the detection of complex structural variations affecting RAD51D that might be missed by conventional short-read approaches. Together, these technological advances promise to deepen our understanding of RAD51D biology while enhancing its clinical utility as a biomarker and therapeutic target.

What are the unresolved questions regarding RAD51D's role in DNA repair and cancer biology?

Despite significant advances in understanding RAD51D, several fundamental questions remain unresolved regarding its precise mechanistic functions and clinical implications. At the molecular level, the exact structural determinants within RAD51D that mediate its interactions with other components of the BCDX2 complex and the broader homologous recombination machinery remain incompletely characterized . How these interactions are dynamically regulated during the cell cycle and in response to different DNA damage types requires further investigation. The potential moonlighting functions of RAD51D beyond canonical homologous recombination repair, such as roles in replication fork protection or mitochondrial DNA maintenance, represent exciting areas for exploration . From a clinical perspective, the full spectrum of cancers associated with RAD51D variants remains to be defined, with emerging evidence suggesting implications beyond ovarian and breast cancers to potentially include non-epithelial tumors such as leiomyosarcoma . The mechanisms underlying this tissue specificity remain enigmatic and could involve tissue-specific expression patterns, cofactor availability, or functional redundancy with other repair pathways. Another critical unanswered question involves resistance mechanisms to PARP inhibitors and platinum agents in RAD51D-mutated tumors, which could involve reversion mutations, compensatory pathway activation, or complex epigenetic adaptations . Understanding these resistance mechanisms will be essential for developing strategies to overcome or prevent therapy resistance. Finally, the potential interactions between RAD51D and environmental exposures, including radiation or chemical mutagens, merit investigation as potential modifiers of cancer risk in variant carriers.

How might combinatorial approaches targeting RAD51D and related pathways improve cancer treatment outcomes?

Combinatorial therapeutic approaches targeting RAD51D-deficient tumors represent a promising frontier for improving cancer treatment outcomes. The synthetic lethality concept underlying PARP inhibitor efficacy in homologous recombination-deficient cells provides a framework for identifying additional synergistic combinations . Several strategic approaches warrant investigation: first, combining PARP inhibitors with agents that prevent replication fork protection (such as ATR inhibitors) could enhance lethality in RAD51D-mutated cells by simultaneously targeting multiple DNA damage response mechanisms . Second, immunotherapy combinations merit exploration, as genomic instability resulting from RAD51D dysfunction may increase neoantigen load and tumor mutational burden, potentially enhancing immune recognition . Third, epigenetic modifiers that prevent RAD51D reexpression or activation of alternative repair pathways could combat acquired resistance to DNA-damaging therapies . Fourth, cell cycle checkpoint inhibitors (particularly WEE1 or CHK1 inhibitors) could force RAD51D-deficient cells through mitosis with unrepaired DNA damage, increasing genomic catastrophe . For optimal clinical translation, these combinations should be evaluated in preclinical models that faithfully recapitulate RAD51D mutations observed in patients, with particular attention to distinguishing monoallelic from biallelic loss-of-function . Biomarker strategies incorporating RAD51D status alongside other homologous recombination repair genes and functional HRD assays will be essential for patient selection and treatment monitoring . As these combinatorial approaches advance to clinical testing, careful assessment of overlapping toxicities and development of intermittent dosing schedules may optimize the therapeutic window while maintaining quality of life.

Product Science Overview

Structure and Expression

The RAD51D (1-328 a.a.) (Human Recombinant) protein is a human full-length protein expressed in Escherichia coli. It consists of 328 amino acids and has a molecular weight of approximately 37.4 kDa . The recombinant version of this protein is often tagged with a His-tag at the N-terminus to facilitate purification and detection .

Function and Mechanism

RAD51D is involved in the HRR pathway, which is essential for repairing double-stranded DNA breaks. It binds to single-stranded DNA (ssDNA) and exhibits DNA-dependent ATPase activity. RAD51D is a part of the BCDX2 complex, which also includes RAD51B, RAD51C, and XRCC2. This complex acts in the BRCA1-BRCA2-dependent HR pathway, playing a role downstream of BRCA2 recruitment and upstream of RAD51 recruitment .

The BCDX2 complex predominantly binds to the intersection of the four duplex arms of the Holliday junction and the junction of replication forks. It has been reported to bind single-stranded DNA, single-stranded gaps in duplex DNA, and specifically to nicks in duplex DNA. Additionally, RAD51D is involved in telomere maintenance .

Clinical Significance

Mutations in the RAD51D gene are associated with a high risk of ovarian cancer. Studies have shown that mutations in RAD51D, along with BRIP1 and RAD51C, significantly increase the susceptibility to ovarian cancer. These mutations are considered high-risk factors for ovarian cancer, second only to BRCA1 and BRCA2 mutations .

Applications

The recombinant RAD51D (1-328 a.a.) protein is used in various research applications, including:

  • SDS-PAGE: To analyze the protein’s purity and molecular weight.
  • Mass Spectrometry (MS): For detailed protein characterization.
  • Functional Studies: To investigate the protein’s role in DNA repair and its interaction with other proteins in the HRR pathway .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.