SAMHD1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction

The SAMHD1 antibody is a research tool designed to detect the SAM domain and HD domain-containing protein 1, a critical enzyme involved in immune regulation, antiviral defense, and DNA repair. SAMHD1 is renowned for its role in restricting HIV replication by depleting intracellular deoxynucleotide triphosphates (dNTPs), which are essential for reverse transcription. This antibody is widely used in molecular biology to study SAMHD1 expression, localization, and interactions in various cell types and disease contexts, including HIV infection, Aicardi–Goutières syndrome (AGS), and acute myeloid leukemia (AML) .

Sources and Types of SAMHD1 Antibodies

Multiple commercial and research-grade SAMHD1 antibodies are available, differing in specificity, host species, and applications. Key products include:

AntibodyHost/IsotypeReactivityApplicationsSource
Abcam OTI1A1Mouse/IgG2bHuman, Dog, Monkey, Rat, MouseIHC-P, WB, ICC/IF, Flow Cyt
Abcam OTI1F9Mouse/IgG2bHuman, MouseWB, IHC, IF/ICC, Flow Cyt
Proteintech 12586-1-APRabbit/IgGHuman, MouseWB, IHC, IF, FC, IP, ELISA
Cell Signaling #49158Rabbit/IgGHumanWB
R&D Systems MAB8120Mouse/IgGHumanWB, Simple Western

Key Features and Applications

a. Western Blot (WB):

  • Proteintech’s Rabbit IgG antibody (12586-1-AP) detects a 72 kDa band in cell lysates (HepG2, K-562) and tissues (human heart, brain) .

  • Cell Signaling’s Rabbit antibody (#49158) shows specificity in human cell lines (HepG2, Daudi) .

b. Immunohistochemistry (IHC):

  • Abcam’s OTI1A1 is validated for paraffin-embedded tissues, including human lung and colon cancer samples .

c. Flow Cytometry (Intra):

  • OTI1A1 and OTI1F9 (Abcam) enable intracellular staining in transfected or native cells .

d. Immunoprecipitation (IP):

  • Proteintech’s antibody (12586-1-AP) successfully pulls down SAMHD1 from K-562 lysates .

Research Findings

a. HIV and Viral Research:
SAMHD1 antibodies are critical in studying viral restriction mechanisms. For example, Vpx-mediated degradation of SAMHD1 was analyzed using Western blot with Abcam’s OTI1A1, demonstrating its role in HIV replication .

b. AML and Cancer:
Low SAMHD1 expression in AML blasts, detected via IHC, correlates with favorable outcomes in high-dose cytarabine therapy . Proteintech’s antibody (12586-1-AP) is cited in studies linking SAMHD1 to chemotherapy resistance .

c. Aicardi–Goutières Syndrome (AGS):
Mutations in SAMHD1, identified using Rabbit IgG antibodies (Proteintech), lead to aberrant IFN production and autoinflammation .

d. Innate Immunity:
SAMHD1’s suppression of NF-κB and IFN-I pathways was confirmed using Abcam’s OTI1F9 in macrophage models .

Therapeutic Implications

SAMHD1 antibodies are pivotal in developing targeted therapies:

  • Inhibitors of SAMHD1 are under investigation to enhance cytarabine efficacy in AML .

  • Antibodies like OTI1A1 (Abcam) aid in validating SAMHD1 as a biomarker for AGS and HIV .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Product dispatch typically occurs within 1-3 business days of order receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
CHBL2 antibody; DCIP antibody; Dendritic cell derived IFNG induced protein antibody; Dendritic cell-derived IFNG-induced protein antibody; Deoxynucleoside triphosphate triphosphohydrolase SAMHD1 antibody; dNTPase antibody; HD domain containing 1 antibody; HDDC1 antibody; Mg11 antibody; Monocyte protein 5 antibody; MOP 5 antibody; MOP-5 antibody; MOP5 antibody; OTTHUMP00000030889 antibody; SAM domain and HD domain 1 antibody; SAM domain and HD domain containing protein 1 antibody; SAM domain and HD domain-containing protein 1 antibody; SAMH1_HUMAN antibody; Samhd1 antibody; SBBI88 antibody
Target Names
SAMHD1
Uniprot No.

Target Background

Function

SAMHD1 (Sterile Alpha Motif and HD Domain-containing protein 1) is a protein with dual functionality. It acts as a host restriction factor, participating in antiviral defense responses, and as a regulator of DNA end resection at stalled replication forks. SAMHD1 exhibits deoxynucleoside triphosphate (dNTPase) activity, crucial for its antiviral properties. This activity reduces cellular dNTP levels, hindering retroviral reverse transcription and thus blocking early-stage viral replication, particularly in dendritic and myeloid cells. This mechanism is effective against viruses like HIV-1 but not HIV-2, due to the counteracting effect of the HIV-2 viral protein Vpx. Beyond its antiviral role, SAMHD1's dNTPase activity regulates DNA precursor pools. Phosphorylation at threonine 592 (Thr-592) acts as a molecular switch, modulating SAMHD1's functions. Phosphorylation inhibits dNTPase activity and antiviral capabilities while promoting DNA end resection at stalled replication forks. During the S phase of the cell cycle, at stalled replication forks, SAMHD1 promotes the resection of gapped or reversed forks by stimulating MRE11 exonuclease activity, activating the ATR-CHK1 pathway, and enabling replication restart. This degradation of nascent DNA at stalled forks is essential for preventing type I interferon induction and subsequent chronic inflammation. Importantly, its role in promoting DNA end resection is independent of its dNTPase activity. Furthermore, SAMHD1 enhances immunoglobulin hypermutation in B-lymphocytes by promoting transversion mutations.

Gene References Into Functions

The following studies further elucidate SAMHD1's functions and regulation:

  1. SAMHD1 is specifically targeted by PP2A-B55alpha holoenzymes during mitotic exit. (PMID: 29884836)
  2. SAMHD1 modulates dNTP pools, influencing the efficacy of nucleotide analogue reverse transcription inhibitors against HIV-1. (PMID: 28220857)
  3. SAMHD1 does not localize to dot-like structures upon DNA double-strand break induction in HeLa cells. (PMID: 29614270)
  4. SAMHD1 plays a significant role in the pathogenesis of T-cell prolymphocytic leukemia (T-PLL). (PMID: 29352181)
  5. SAMHD1 downregulates innate immune responses to viral infections and inflammatory stimuli. (PMID: 29610295)
  6. SAMHD1 prevents chronic inflammation by limiting the release of single-stranded DNA from stalled replication forks. (PMID: 29670289)
  7. Low SAMHD1 expression is associated with HIV-1 infection. (PMID: 29084722)
  8. Multiple domains of SAMHD1 are essential for Vpx-induced degradation. (PMID: 29963825)
  9. SAMHD1 regulates HIV-1 latency. (PMID: 29793958)
  10. CD81 interaction with SAMHD1 controls the metabolic rate of HIV-1 replication. (PMID: 28871089)
  11. Immune activation during HIV-1 infection influences SAMHD1 expression and degradation. (PMID: 27922067)
  12. Additional cellular regulatory mechanisms modulate SAMHD1 enzymatic activity. (PMID: 27511536)
  13. IRF3 phosphorylation and nuclear translocation upregulate SAMHD1 expression. (PMID: 27411355)
  14. SAMHD1 facilitates CtIP accrual to promote DNA end resection in homologous recombination-mediated DNA double-strand break repair. (PMID: 28834754)
  15. Vpx overcomes a dNTP concentration-independent restriction for lentiviruses in resting CD4 T cells. (PMID: 28228523)
  16. The RXL motif is crucial for SAMHD1 tetramer formation, dNTPase activity, and HIV-1 restriction. (PMID: 29321329)
  17. SAMHD1 restricts HBV replication through its dNTPase activity. (PMID: 27229711)
  18. SAMHD1 is a potential therapeutic target for various malignancies. (PMID: 28436707)
  19. SAMHD1 overexpression contributes to resistance to PARP inhibitors and Ara-C in glioblastoma cells. (PMID: 29274141)
  20. Three cysteine residues in SAMHD1 form a redox switch, reversibly inhibiting tetramerization and dNTPase activity. (PMID: 28398823)
  21. SAMHD1's regulation of the type I IFN response is genetically separable from its HIV-1 restriction function. (PMID: 28229507)
  22. miR-181a mediates interferon-induced SAMHD1 expression in astrocytes and microglia. (PMID: 27219130)
  23. SAMHD1 affects sensitivity to nucleoside inhibitors in HIV-1 permissive cells. (PMID: 28359840)
  24. p21 modulates SAMHD1 activity and antiviral function. (PMID: 28931685)
  25. SAMHD1 exhibits dual roles as an inhibitor of uncontrolled proliferation and a resistance factor limiting anticancer treatment efficacy. (PMID: 28502830)
  26. Studies reveal nucleotide substrate specificity and a novel inhibitory mechanism for SAMHD1 dNTPase activity. (PMID: 28046007)
  27. Allosteric regulation of SAMHD1 has been identified through molecular dynamics simulations. (PMID: 28321930)
  28. Raf/MEK/ERK kinase cascade activation upregulates CDK1, leading to SAMHD1 phosphorylation and antiviral activity deactivation. (PMID: 28122869)
  29. SAMHD1 inhibits cutaneous T-cell lymphoma cell growth. (PMID: 27929746)
  30. miR-181 regulates SAMHD1 protein expression in neoplastic CD4+ T-cells. (PMID: 27889686)
  31. Cyclin binding contributes to SAMHD1 expression and stability in dividing cells. (PMID: 27815502)
  32. SAMHD1's RNase function is implicated in nucleic acid-triggered inflammatory responses and its anti-retroviral function. (PMID: 27387229)
  33. A phosphomimetic mutation affects SAMHD1 tetramer dissociation rates but not allosteric activation equilibrium. (PMID: 27566548)
  34. Mutually exclusive oligomeric state requirements for ssNA binding and dNTP hydrolase activity modulate SAMHD1 functions. (PMID: 27775344)
  35. SAMHD1 inhibits HBV replication at the reverse transcription step, likely through dNTP depletion. (PMID: 27179347)
  36. SAMHD1's dNTPase activity affects cell proliferation, cell cycle distribution, and apoptosis. (PMID: 27183329)
  37. Neurologic phenotypes associated with SAMHD1 mutations in Aicardi-Goutieres Syndrome. (PMID: 27643693)
  38. Cytokines influence HIV-1 reservoir establishment by driving proliferation and increasing CD4+ lymphocyte susceptibility through SAMHD1 inactivation. (PMID: 26923586)
  39. Heterozygous cancer-associated SAMHD1 mutations increase mutation rates in cancer cells. (PMID: 27071091)
  40. SAMHD1 restricts HIV-1 infection in non-cycling cells by limiting dNTP pools. (PMID: 26733158)
  41. Association of SAMHD1 gene mutations with cerebrovascular disease. (PMID: 26504826)
  42. SAMHD1 may facilitate THP-1 cell differentiation. (PMID: 26606981)
  43. Factors beyond Vpx-SAMHD1 binding influence Vpx-mediated SAMHD1 degradation. (PMID: 26779819)
  44. SAMHD1 possesses a dNTP-independent function contributing to lentiviral restriction. (PMID: 26655245)
  45. dNTP metabolism in cancer development and SAMHD1's role in regulating this process. (PMID: 26416562)
  46. SAMHD1 phosphorylation ablates restriction and tetramer formation without affecting triphosphohydrolase kinetics. (PMID: 26431200)
  47. Vpx promotes SAMHD1 degradation in memory CD4+ T-lymphocytes, increasing viremia and immunodeficiency. (PMID: 25996507)
  48. Increased SAMHD1 in human astrocytes contributes to HIV restriction. (PMID: 25890101)
  49. SAMHD1's RNase activity controls retrovirus replication but not non-retro RNA viruses. (PMID: 26032178)
  50. SAMHD1 regulates all dNTP pools, and mitochondrial dGK recycles deoxyguanosine from dGTP degraded by nuclear SAMHD1. (PMID: 26342080)
Database Links

HGNC: 15925

OMIM: 606754

KEGG: hsa:25939

STRING: 9606.ENSP00000262878

UniGene: Hs.580681

Involvement In Disease
Aicardi-Goutieres syndrome 5 (AGS5); Chilblain lupus 2 (CHBL2)
Protein Families
SAMHD1 family
Subcellular Location
Nucleus. Chromosome.
Tissue Specificity
Expressed in heart, skeletal muscle, spleen, liver, small intestine, placenta, lung and peripheral blood leukocytes. No expression is seen in brain and thymus.

Q&A

What is SAMHD1 and why are antibodies against it important for viral research?

SAMHD1 functions as a host restriction factor that inhibits replication of both retroviruses and DNA viruses by reducing intracellular dNTP pools through its deoxynucleoside triphosphate triphosphohydrolase (dNTPase) activity. SAMHD1 antibodies are crucial for studying:

  • Expression patterns during viral infections (upregulation was observed during HCMV infection)

  • Phosphorylation status, particularly at T592, which regulates its antiviral activity

  • Protein-protein interactions with components of innate immune signaling pathways

  • Subcellular localization before and after viral challenge

SAMHD1 has been demonstrated to inhibit not only HIV-1 replication in non-dividing myeloid cells and resting CD4+ T cells but also the replication of herpesviruses, enterovirus 71, hepatitis B virus, and hepatitis C virus . Additionally, SAMHD1 has been associated with Aicardi-Goutières syndrome, an autoinflammatory disorder .

What are the validated applications for SAMHD1 antibodies in molecular research?

SAMHD1 antibodies have been validated for multiple applications in molecular and cellular research:

ApplicationValidation MethodsResearch Examples
Western BlotDetecting ~72 kDa bands in Daudi and HepG2 cell lysatesMonitoring expression changes after viral infection
ImmunoprecipitationCo-IP with FLAG or HA-tagged proteinsStudying interactions with IKKα, IKKβ, and IKKγ
Simple WesternAutomated capillary-based immunoassayDetection in Daudi human Burkitt's lymphoma cell line
Immunocapture-MSMass spectrometry verificationSAMHD1 was enriched 641 times versus negative control

Researchers have successfully used these antibodies to demonstrate that SAMHD1 suppresses NF-κB activation by interacting with NF-κB1/2 and reducing phosphorylation of the NF-κB inhibitory protein IκBα .

How should researchers optimize Western blot protocols for SAMHD1 detection?

For optimal SAMHD1 detection via Western blot, consider the following methodological approach:

  • Sample preparation:

    • Use RIPA or NP-40 lysis buffers with protease and phosphatase inhibitors

    • Include positive controls (Daudi or HepG2 cell lysates) and negative controls (SAMHD1-knockdown samples)

  • Gel electrophoresis and transfer:

    • Run under reducing conditions using Immunoblot Buffer Group 1

    • Use 8-10% polyacrylamide gels to properly resolve the ~72 kDa SAMHD1 protein

  • Antibody incubation:

    • Optimal concentration: 1 μg/mL for primary antibody (e.g., MAB8120)

    • Follow with appropriate HRP-conjugated secondary antibody (e.g., HAF018)

  • Validation controls:

    • Perform siRNA knockdown validation (as shown in HCMV studies with ~70% knockdown efficiency)

    • Consider Vpx-mediated degradation as an alternative control method

Studies have demonstrated that these approaches can reliably detect SAMHD1 upregulation after viral infection, with significant increases observed in human foreskin fibroblasts, retinal pigment epithelial cells, and microvascular endothelial cells infected with HCMV .

What approaches should be used to study SAMHD1 phosphorylation states?

Phosphorylation, particularly at T592, critically regulates SAMHD1 function. Here's a comprehensive approach to studying SAMHD1 phosphorylation:

  • Antibody selection:

    • Use phospho-specific antibodies for T592 alongside total SAMHD1 antibodies

    • Include phosphatase-treated samples as negative controls

  • Time-course analysis:

    • Monitor phosphorylation at multiple timepoints after stimulation or infection

    • Research shows rapid phosphorylation of SAMHD1 at T592 after HCMV infection

  • Kinase identification:

    • Investigate cellular cyclin-dependent kinases, especially Cdk2, which phosphorylates SAMHD1

    • Consider viral kinases (e.g., pUL97 in HCMV) that may target SAMHD1

  • Functional correlation:

    • Correlate phosphorylation with subcellular localization (phospho-SAMHD1 relocalization to cytoplasm has been observed after HCMV infection)

    • Assess impact on tetramer formation, as phosphorylation at T592 destabilizes SAMHD1 tetramers

Research has demonstrated that only SAMHD1 dephosphorylated at T592 actively restricts HIV-1 and HBV, highlighting the importance of phosphorylation state in antiviral activity .

How can co-immunoprecipitation protocols be optimized for studying SAMHD1 protein interactions?

To effectively study SAMHD1's interactions with other proteins:

  • Experimental design:

    • Use both forward (anti-SAMHD1) and reverse (anti-interacting protein) IP approaches

    • Include appropriate IgG controls to assess non-specific binding

  • Expression systems:

    • Consider epitope-tagged proteins for cleaner pull-downs (HA-tagged SAMHD1 and FLAG-tagged interaction partners have been successful)

    • Validate in multiple cell types to confirm physiological relevance

  • Confirmation strategies:

    • Follow co-IP with direct in vitro pull-down assays to confirm direct interactions

    • Validate interactions through multiple methods (e.g., proximity ligation assay)

  • Detection optimization:

    • Use 10-20% input controls to normalize IP efficiency

    • Consider chemiluminescent or fluorescent detection for quantitative analysis

Studies have successfully used these approaches to demonstrate that SAMHD1 interacts with IKKα, IKKβ, and IKKγ, providing insight into its mechanism of NF-κB suppression .

How can researchers validate the specificity of SAMHD1 antibodies?

Rigorous validation of SAMHD1 antibodies is essential to ensure experimental reliability:

  • Multiple validation approaches:

    • Western blot analysis in positive control cell lines (Daudi, HepG2)

    • Immunocapture followed by mass spectrometry analysis (MS)

    • SAMHD1 knockdown/knockout controls

  • Mass spectrometry validation:

    • In published research, SAMHD1 was successfully enriched 641 times versus negative control (rabbit IgG) through immunocapture-MS

    • This approach confirms binding to the correct target and ensures antibody reproducibility

  • Genetic validation:

    • siRNA knockdown (achieving ~70% efficiency)

    • CRISPR/Cas9 knockout models

    • Vpx-mediated degradation of SAMHD1 (SIV VLPs loaded with Vpx)

  • Cross-reactivity testing:

    • Test across multiple cell types to ensure consistent detection

    • Peptide competition assays to confirm epitope specificity

According to the International Working Group for Antibody Validation standards, these approaches collectively provide robust validation of antibody specificity .

What are common challenges in detecting SAMHD1 in different cell types and how can they be addressed?

Researchers face several challenges when detecting SAMHD1 across cell types:

  • Expression level variations:

    • SAMHD1 expression varies by cell type and activation state

    • Higher expression observed in cells in G0 phase (cell cycle arrest) compared to proliferating cells

    • Solution: Use appropriate positive controls and adjust exposure times accordingly

  • Interferon effects:

    • Type I interferons may influence SAMHD1 expression

    • Solution: Consider using IFN receptor blocking antibodies to exclude IFN effects, as demonstrated in HCMV studies

  • Cell cycle considerations:

    • SAMHD1 expression correlates with cell cycle status

    • Solution: Include cell cycle markers (e.g., Ki-67) in parallel analyses

  • Post-translational modifications:

    • Phosphorylation may affect antibody recognition

    • Solution: Use multiple antibodies targeting different epitopes

  • Subcellular localization shifts:

    • SAMHD1 can relocalize from nucleus to cytoplasm after viral infection

    • Solution: Consider cellular fractionation or immunofluorescence approaches to track localization

Research has shown that SAMHD1 can be successfully detected across diverse cell types including fibroblasts, epithelial cells, endothelial cells, and monocytic cell lines when these factors are properly addressed .

How can SAMHD1 antibodies be used to investigate its role in NF-κB signaling pathways?

SAMHD1 has been discovered to suppress NF-κB activation through specific mechanisms. To investigate this function:

  • Phosphorylation analysis:

    • Monitor IκBα phosphorylation levels in the presence/absence of SAMHD1

    • Track IKKα/β/γ phosphorylation states using phospho-specific antibodies

  • Protein interaction studies:

    • Use co-immunoprecipitation to assess SAMHD1 interactions with:

      • NF-κB1/2 components

      • IKK complex components (IKKα, IKKβ, IKKγ)

    • Confirm direct interactions with in vitro pull-down assays

  • Functional readouts:

    • Assess NF-κB-dependent gene expression in SAMHD1-silenced versus control cells

    • Measure NF-κB nuclear translocation through subcellular fractionation and immunoblotting

    • Use NF-κB reporter assays to quantify activation levels

  • Stimulus-specific responses:

    • Compare effects after different stimuli (viral infections, inflammatory cytokines)

    • Analyze temporal dynamics of SAMHD1-mediated suppression

Research has demonstrated that SAMHD1 inhibits NF-κB activation by interacting with NF-κB1/2 and reducing phosphorylation of IκBα, providing a mechanism for its suppressive effect on innate immune responses .

What experimental approaches can be used to study the relationship between SAMHD1 and the cGAS-STING pathway?

To investigate SAMHD1's role in modulating the cGAS-STING pathway:

  • Cytosolic DNA quantification:

    • Measure cytosolic single-stranded DNA levels in SAMHD1-deficient versus wild-type cells

    • Correlate with cGAS activation and downstream signaling

  • Pathway component analysis:

    • Monitor cGAS-STING pathway activation markers in SAMHD1-silenced cells:

      • STING dimerization/phosphorylation

      • TBK1 and IRF3 phosphorylation

      • IFN-β production and ISG expression

  • DNA replication fork studies:

    • Investigate SAMHD1's role in regulating stalled DNA replication forks

    • Analyze correlation between replication stress and cytosolic DNA accumulation

  • Complementation experiments:

    • Perform rescue experiments with:

      • Wild-type SAMHD1

      • dNTPase-deficient mutants

      • Phosphorylation site mutants

Research has shown that SAMHD1 regulates stalled DNA replication forks and reduces cytosolic single-stranded DNA accumulation, thereby decreasing IFN-I production through the cGAS-STING pathway .

How can researchers design experiments to differentiate between dNTPase-dependent and independent functions of SAMHD1?

SAMHD1 exhibits both dNTPase-dependent and independent functions. To differentiate between these activities:

  • Mutant analysis approach:

    • Generate catalytically inactive SAMHD1 mutants (e.g., HD domain mutations)

    • Compare with phosphorylation site mutants (T592A/E)

    • Assess function in various assays:

      FunctiondNTPase-dependentdNTPase-independent
      HIV-1 restrictionYesNo
      NF-κB suppressionTo be determinedLikely yes
      IFN-I regulationPartialPartial
      DNA damage responseLikely yesLikely partial
  • dNTP manipulation studies:

    • Exogenously supply dNTPs to bypass dNTPase effects

    • Use other dNTPase enzymes as controls

    • Measure functional outcomes with normalized dNTP pools

  • Protein interaction mapping:

    • Identify interaction domains involved in various functions

    • Correlate with dNTPase activity requirements

    • Create domain-specific mutants

  • Viral restriction comparison:

    • Compare restriction of dNTP-dependent viruses (e.g., HIV-1) versus potentially dNTP-independent mechanisms (e.g., NF-κB suppression in HCMV infection)

SAMHD1's ability to suppress NF-κB activation and type I interferon induction may involve mechanisms distinct from its dNTPase activity, warranting careful experimental design to distinguish these functions .

How can SAMHD1 antibodies be employed to study its role in DNA damage response and cancer biology?

SAMHD1's emerging roles in DNA damage response and cancer biology can be investigated using:

  • DNA damage response analysis:

    • Track SAMHD1 localization to sites of DNA damage using immunofluorescence

    • Analyze SAMHD1 post-translational modifications after DNA damage induction

    • Study protein interactions with DNA repair machinery components

  • Cancer-specific investigations:

    • Analyze SAMHD1 expression patterns across cancer tissue microarrays

    • Correlate expression/mutations with clinical outcomes

    • Investigate synthetic lethal interactions in cancer contexts

  • Therapeutic response monitoring:

    • Assess how cancer treatments affect SAMHD1 expression/function

    • Study SAMHD1's impact on nucleoside analog metabolism in cancer therapy

    • Investigate potential as a biomarker for treatment response

  • Cell cycle regulation:

    • Examine SAMHD1 phosphorylation dynamics throughout the cell cycle

    • Correlate with dNTP pool regulation in normal versus cancer cells

SAMHD1's regulation of stalled DNA replication forks suggests important functions in genomic stability that may be relevant to cancer biology and therapeutic approaches .

What methodological approaches can detect novel post-translational modifications of SAMHD1?

Beyond phosphorylation, SAMHD1 likely undergoes additional post-translational modifications. To identify and characterize these:

  • Mass spectrometry-based discovery:

    • Immunoprecipitate SAMHD1 from cells under various conditions

    • Perform high-resolution MS/MS analysis

    • Use both data-dependent and targeted acquisition methods

  • Modification-specific enrichment:

    • Apply ubiquitin/SUMO enrichment strategies

    • Use acetyl-lysine antibodies for immunoprecipitation

    • Consider chemical approaches to enrich modified peptides

  • Site-directed mutagenesis:

    • Mutate predicted modification sites

    • Assess functional consequences on:

      • Antiviral activity

      • Protein stability

      • Subcellular localization

      • Protein interactions

  • Enzyme inhibitor studies:

    • Test effects of deacetylase, E3 ligase, or other PTM enzyme inhibitors

    • Monitor SAMHD1 function and modifications

While T592 phosphorylation is well-characterized , other modifications may provide additional regulatory mechanisms for SAMHD1's diverse functions in antiviral immunity and cellular homeostasis.

How can researchers investigate SAMHD1's differential roles across immune cell subsets?

To understand SAMHD1's cell type-specific functions in the immune system:

  • Expression profiling approach:

    • Analyze SAMHD1 levels across immune cell subsets using:

      • Flow cytometry with validated antibodies

      • Western blot of sorted populations

      • Single-cell RNA-seq correlation with protein expression

  • Functional comparative analysis:

    • Compare SAMHD1's antiviral activity in different immune cell types

    • Assess NF-κB suppression capacity across myeloid versus lymphoid cells

    • Examine cell type-specific protein interactions

  • Regulation mechanism investigation:

    • Study cell type-specific phosphorylation patterns

    • Analyze subcellular localization differences

    • Identify lineage-specific interaction partners

  • Conditional knockout models:

    • Generate immune cell subset-specific SAMHD1 knockout models

    • Assess functional consequences in infection and inflammation models

Research has demonstrated differential SAMHD1 activity in myeloid cells versus T cells in HIV restriction, suggesting that cell type-specific regulation is an important aspect of SAMHD1 biology .

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.