Recombinant Mouse HIV Tat-specific factor 1 homolog (Htatsf1)

Shipped with Ice Packs
In Stock

Description

Functional Roles in Biological Processes

Htatsf1’s roles are inferred from human and rodent studies:

RNA Splicing and Transcription

  • Spliceosome Assembly: Part of the 17S U2 SnRNP complex, stabilizing branchpoint interactions during intron excision .

  • Transcriptional Elongation: Acts as a cofactor for HIV-1 Tat, enhancing viral RNA synthesis by promoting elongation .

  • Intron Retention: Linked to splicing of ribosomal protein mRNAs and pluripotency in stem cells .

DNA Repair

In human cells, HTATSF1 facilitates homologous recombination during DNA double-strand breaks by recruiting TOPBP1 . Mouse studies remain limited, but conserved mechanisms are plausible.

Environmental and Chemical Interactions

Rodent models (e.g., rat) reveal Htatsf1’s sensitivity to environmental toxins:

ChemicalEffect on Htatsf1Evidence
ArsenicAlters DNA methylationCTD PMID:25304211
Valproic AcidIncreases mRNA expressionCTD PMID:29154799
Bisphenol SElevates protein levelsCTD PMID:34186270

Detection and Quantification

The Mouse Htatsf1 ELISA Kit (AbbeXa) enables precise measurement of endogenous protein levels:

ParameterDetails
Test Range0.156–10 ng/ml
Detection MethodColorimetric
Sample TypesTissue homogenates, cell lysates, biological fluids
Storage4°C short-term, -20°C long-term

Antibody-Based Techniques

Antibodies like 20805-1-AP (Proteintech) cross-react with mouse Htatsf1 for multiple applications:

ApplicationDilutionSample Validated
Western Blot (WB)1:1000–1:4000HeLa, mouse brain, HepG2
Immunoprecipitation (IP)0.5–4.0 µgHepG2
Immunohistochemistry (IHC)1:20–1:200Human breast cancer, brain

HIV-1 Pathogenesis

Htatsf1’s interaction with HIV-1 Tat is critical for viral transcription. In human studies, Tat upregulates HTATSF1, enhancing viral RNA production . Mouse models may replicate this mechanism, aiding antiviral research.

Pluripotency and Stem Cell Biology

Htatsf1’s association with naïve pluripotency in humans suggests a role in stem cell maintenance . Rodent studies could explore its interplay with Nanog and KLF2.

Toxicology and Epigenetics

Chemical interactions (e.g., arsenic, valproic acid) highlight Htatsf1’s role in epigenetic regulation and environmental disease .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notice and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default glycerol concentration is 50%, but this can be adjusted as needed.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If a specific tag type is required, please inform us for preferential development.
Synonyms
Htatsf1; HIV Tat-specific factor 1 homolog
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-757
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Htatsf1
Target Protein Sequence
SGNNLSGND EFDEQLRMQE LYGGDPKEGD TQNEPSGEAH SLGQPPDDTP YEWDLDKKAW FPKITEDFIA TYQANYGFSS DGASSSTANV QDANTKAVEE PPQKEVPETP DSKRKGEKRK AESGWFHVEE DRNTNVYVSG LPPDITVDEF IQLMSKFGII MRDPQTEEFK VKLYKDDQGN LKGDGLCCYL KKESVELALK LLDEDEIRGY KLHVEVAKFQ LKGEYDASKK KKKCKDYKKK LSLQQKQLDW RPERRAGPNR LRHERVVILK NMFHPMDFED DPLVLNEIRE DLRVECSKFG QIRKLLLFDR HPDGVASVSF REPEEADHCI QTLDGRWFGG RQITAQAWDG TTDYQVEETS REREERLRGW EAFLNAPEAS RGLRRMDSIA GSERPGPSRM RHFSEHPSMS NMKAQEATTG MAFEETIDEN KFEKAEEGGE SEGDASEKDA KEGGSDGDHP EREGGEGCSK KENEEGCPER ALEPEEGNPQ TEAQENGPER EARKKSKMDY EKNGFSKESE DNDLGKESEG EDSLKKESED DDSEEESEED SSEKQSQDGS DKEIEENGVK KDVDQDVSDK EFPEDVEKES EENETDKSEF DEGSERVLDE EGSEREFEED SDEKEEEGDD DEEEVVYERV FDDDSDDIEE EEEADEKECE DADDKEEDND IDEKLFDDSD EKEDEEDTDG KKDDDASDKV FEDNSNEKLF DEEEGPNEKL FDDSDERGTV GNVKEDGSQS TDSSFALSSS DDDDDEV
Uniprot No.

Target Background

Function
This protein functions as a general transcription factor, playing a crucial role in transcriptional elongation. It may also mediate the reciprocal stimulatory effect of splicing on transcriptional elongation.
Gene References Into Functions
  1. Studies suggest sex influences HIV's effects on the brain. Increased behavioral deficits in males correlate with glial activation and synaptic damage, factors implicated in cognitive/motor impairments in HIV patients. [PMID: 24352707](https://www.ncbi.nlm.nih.gov/pubmed/24352707)
  2. CA150 FF domains recognize multiple sites within the Tat-SF1 protein, conforming to the consensus motif (D/E)(2/5)-F/W/Y-(D/E)(2/5). [PMID: 15485897](https://www.ncbi.nlm.nih.gov/pubmed/15485897)
Database Links

KEGG: mmu:72459

STRING: 10090.ENSMUSP00000086027

UniGene: Mm.2152

Protein Families
HTATSF1 family
Subcellular Location
Nucleus.

Q&A

What is Htatsf1 and what is its primary function in mouse models?

Htatsf1 (HIV TAT specific factor 1) in mice is a homolog of the human HTATSF1 gene that functions as a cofactor for the stimulation of transcriptional elongation by HIV-1 Tat. This protein facilitates HIV-1 Tat binding to the HIV-1 promoter through Tat-TAR interaction . In mouse models, Htatsf1 serves as a dual-function factor that couples transcription and splicing processes, enabling their reciprocal activation .

Mouse Htatsf1 shares significant structural homology with human HTATSF1, though it contains notable differences that affect its interaction with HIV-1 Tat. Specifically, mouse cyclin T1, which works with Htatsf1, lacks a critical cysteine residue needed to form a functional complex with Tat, resulting in limited direct transcriptional activity in mice .

How does the structure and expression pattern of Htatsf1 vary across mouse tissues?

Htatsf1 shows a complex expression pattern across multiple mouse tissues. According to MGI database information, Htatsf1 expression has been documented in:

System/Tissue TypeHtatsf1 Expression
Nervous systemPresent
Reproductive systemPresent
Hematopoietic systemPresent
Embryonic tissuesPresent (multiple components)
Renal/urinary systemPresent
Digestive/alimentary systemPresent
Cardiovascular systemPresent

The gene exhibits developmental regulation with expression patterns changing during embryonic development through adult stages .

What are the optimal methods for expressing recombinant Htatsf1 in experimental systems?

The expression of recombinant Htatsf1 requires careful consideration of several factors:

For in vitro expression:

  • Bacterial expression systems: While economical, these often struggle with proper folding of complex mammalian proteins like Htatsf1

  • Mammalian cell expression systems: HEK293 or CHO cells provide better post-translational modifications

  • Baculovirus expression systems: Offer a balance between proper folding and cost-effectiveness

For in vivo expression in mouse models:

  • Adenoviral vectors have proven effective for gene transfer of related HIV-1 proteins such as HIV-Tat. Similar approaches can be adapted for Htatsf1 expression .

  • For tissue-specific expression, researchers should consider promoter selection based on target tissue distribution patterns identified in mouse genome databases .

How can researchers effectively design knockdown experiments to study Htatsf1 function?

For effective Htatsf1 knockdown studies:

  • siRNA approach: Commercial Htatsf1-specific siRNA oligo duplexes are available with validated knockdown efficiency. Optimally designed siRNAs can achieve >70% knockdown when used at 10 nM concentration .

  • Experimental design considerations:

    • Include appropriate controls (scrambled siRNA duplexes)

    • Validate knockdown efficiency using quantitative RT-PCR

    • Ensure transfection efficiency >90% using fluorescent transfection controls

    • Perform time-course studies (24-72 hours post-transfection)

  • Measurement parameters:

    • Primary: mRNA expression levels by qRT-PCR

    • Secondary: Protein levels by Western blotting

    • Functional: Changes in HIV-1 transcriptional activity in relevant model systems

Knockdown MethodAdvantagesLimitationsValidation Method
siRNA (transient)Rapid results, easily titratableTemporary effectqRT-PCR, Western blot
shRNA (stable)Long-term studies possibleMore complex deliveryqRT-PCR, Western blot, functional assays
CRISPR/Cas9Complete knockout possibleOff-target effectsSequencing, protein absence confirmation

How does mouse Htatsf1 differ from human HTATSF1 in HIV-1 transcriptional studies?

Critical differences between mouse Htatsf1 and human HTATSF1 significantly impact their utility in HIV-1 research:

  • Transcriptional activity differences:
    Mouse Htatsf1 has limited direct transcriptional activity in HIV-1 models because mouse cyclin T1 lacks a critical cysteine residue needed to form a functional complex with Tat .

  • Alternative mechanistic pathways:
    While human HTATSF1 directly enhances HIV-1 transcription, mouse Htatsf1 works indirectly by inducing expression of cytokines like TNF-α that increase HIV-1 transcription via NF-κB dependent mechanisms .

  • Experimental implications:
    When using mouse models for HIV-1 research, researchers must account for these species-specific differences. Mouse models may not fully recapitulate human HTATSF1 function unless human HTATSF1 is introduced.

  • Structure-function relationships:
    Despite sequence differences, both proteins contain conserved RNA recognition motifs essential for RNA processing functions.

What are the methodological approaches for studying Htatsf1 interactions with HIV-Tat in mouse models?

To study Htatsf1-HIV-Tat interactions in mouse models:

  • Co-immunoprecipitation approaches:

    • Use epitope-tagged versions of both proteins (HA-tagged Htatsf1, FLAG-tagged Tat)

    • Perform reciprocal pull-downs to confirm interactions

    • Include RNase treatment controls to determine if interactions are RNA-dependent

  • Mouse model systems:

    • HIV-Tg26 transgenic mice express HIV-1 genes and can be used to study Htatsf1 interactions

    • Adenoviral vector delivery of HIV-Tat genes can be performed in wild-type or Htatsf1-modified mice

    • Newborn mice models can be particularly useful for investigating developmental aspects of these interactions

  • Transcriptional readout systems:

    • HIV-LTR reporter constructs can measure transcriptional activity

    • qRT-PCR analysis of HIV-1 gene expression

    • Analysis of downstream cytokine production (e.g., TNF-α)

How should researchers interpret contradictory results in Htatsf1 functional studies?

When confronting contradictory results in Htatsf1 studies, researchers should systematically evaluate:

  • Model system differences:

    • Cell type specificity: Htatsf1 may function differently across various cell types

    • Species differences: Results from mouse vs. human systems require careful interpretation

    • Developmental context: Htatsf1 function may vary across developmental stages

  • Technical variables:

    • Expression level effects: Over-expression vs. endogenous studies may yield different results

    • Protein tag interference: Tags may affect protein interactions or localization

    • Knockdown efficiency: Incomplete knockdown may lead to residual function

  • Reconciliation approaches:

    • Perform dose-response studies to determine threshold effects

    • Use multiple complementary techniques to validate findings

    • Consider temporal dynamics of Htatsf1 interactions

    • Evaluate potential compensatory mechanisms in chronic vs. acute manipulation studies

  • Statistical considerations:

    • Use appropriate statistical tests based on data distribution

    • Consider biological vs. statistical significance

    • Report effect sizes alongside p-values

    • Account for multiple comparisons in complex datasets

What are the approaches for quantifying Htatsf1 activity in HIV-1 transcription studies?

Quantification of Htatsf1 activity requires multi-level analysis:

  • Direct transcriptional activity measurement:

    • HIV-LTR luciferase reporter assays

    • Cell-free transcription systems with purified components

    • ChIP assays to measure promoter occupancy

  • RNA processing activity assessment:

    • RNA immunoprecipitation (RIP) to identify bound RNAs

    • Splicing reporter assays to measure splicing efficiency

    • RT-PCR analysis of splice variant ratios

  • Downstream effects quantification:

    • Measurement of HIV-1 gene expression levels

    • Analysis of cytokine induction (e.g., TNF-α, which mediates indirect effects)

    • Viral production in cell culture systems

  • Data normalization strategies:

    • Use multiple housekeeping genes for qRT-PCR normalization

    • Account for transfection efficiency in transient assays

    • Include activity controls (known activators/inhibitors) in each experiment

How can mouse Htatsf1 studies inform HIV-associated nephropathy (HIVAN) research?

Mouse models incorporating Htatsf1 and HIV-Tat have proven valuable for HIVAN research:

  • Established model systems:
    The HIV-Tg26 mouse line has been extensively used to study HIVAN. These mice carry a 7.4-kb HIV-1 construct lacking the 3-kb sequence overlapping the gag/pol region .

  • HIV-Tat and Htatsf1 role in kidney pathology:

    • Adenoviral vector-mediated expression of HIV-Tat in newborn HIV-Tg26 mice reproduces the full HIVAN phenotype

    • HIV-Tat, potentially through interaction with Htatsf1, plays a critical role in the pathogenesis of HIVAN by:
      a) Inducing renal expression of HIV-1 genes
      b) Working in synergy with heparin-binding growth factors
      c) Increasing dedifferentiation and proliferation of renal epithelial cells

  • Experimental design considerations:

    • Timing is critical: expression of HIV-Tat in newborn HIV-Tg26 mice precipitates HIVAN development during the first month of life

    • Homozygous HIV-Tg26 mice show more severe phenotypes but die early, while heterozygous mice survive longer and allow extended studies

    • Domain-specific effects: The activation and basic binding domains of Tat are sufficient to induce renal expression of HIV-genes, while the RGD motif is not essential

How can researchers design experiments to determine tissue-specific effects of Htatsf1 manipulation?

To investigate tissue-specific Htatsf1 functions:

  • Conditional expression/knockout systems:

    • Cre-loxP systems with tissue-specific promoters driving Cre expression

    • Tet-On/Off systems for temporal control of expression

    • Tissue-specific adenoviral delivery systems as demonstrated with HIV-Tat

  • Tissue panel analysis:

    • Comprehensive analysis across multiple tissues showing Htatsf1 expression, including nervous system, reproductive system, hematopoietic system, and embryonic tissues

    • Comparative expression analysis in normal vs. pathological states

  • Multi-omics approach:

    • Tissue-specific transcriptomics following Htatsf1 manipulation

    • Proteomics to identify tissue-specific interaction partners

    • Epigenomic analysis to determine tissue-specific chromatin contexts

  • Developmental considerations:

    • Timing of manipulation based on developmental expression patterns

    • Analysis of phenotypes across developmental stages

    • Consideration of compensatory mechanisms in long-term studies

What emerging technologies are advancing Htatsf1 functional studies?

Recent technological advances have expanded research capabilities:

  • CRISPR-based approaches:

    • CRISPRi for tunable repression of Htatsf1

    • CRISPRa for enhanced expression in specific contexts

    • Base editing for introducing specific mutations to study structure-function relationships

  • Protein engineering tools:

    • Optogenetic control of Htatsf1 activity

    • Chemical-induced proximity systems for temporal control

    • Domain-swapping experiments between mouse and human homologs

  • Single-cell technologies:

    • scRNA-seq to study cell-type specific effects of Htatsf1

    • Spatial transcriptomics to map Htatsf1 activity in complex tissues

    • Live-cell imaging with fluorescent protein fusions

  • Structural biology advances:

    • Cryo-EM studies of Htatsf1-containing complexes

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) for dynamic structural analysis

    • Integrative modeling approaches combining multiple structural data types

How can researchers address the species-specific limitations of mouse Htatsf1 in HIV research?

To overcome species-specific limitations:

  • Humanized systems:

    • Expression of human HTATSF1 in mouse cells/models

    • Co-expression of human cyclin T1 to restore Tat-mediated transcriptional activity

    • Humanized mouse models with human immune system components

  • Chimeric protein approaches:

    • Design of mouse/human chimeric Htatsf1 proteins

    • Domain-swapping experiments to identify critical functional regions

    • Mutagenesis of specific residues based on comparative sequence analysis

  • Alternative experimental systems:

    • Use of ex vivo human tissue cultures alongside mouse models

    • Comparative studies in multiple species

    • iPSC-derived cellular models from both species

  • Computational approaches:

    • Molecular modeling of protein-protein interfaces

    • Simulation of species-specific interaction differences

    • Systems biology models incorporating species-specific parameters

How should researchers troubleshoot poor expression or activity of recombinant Htatsf1?

When encountering expression or activity issues:

  • Expression optimization strategies:

    • Codon optimization for expression system

    • Testing multiple affinity tags (N-terminal vs. C-terminal)

    • Expression temperature and induction conditions optimization

    • Use of solubility-enhancing fusion partners

  • Purification challenges:

    • Optimization of lysis conditions to prevent aggregation

    • Addition of stabilizing agents during purification

    • Testing multiple chromatography approaches

    • Native vs. denaturing purification protocols

  • Activity assessment:

    • Verification of proper folding using circular dichroism

    • Analysis of post-translational modifications

    • RNA binding assays to confirm basic functionality

    • Comparison with positive controls (e.g., human HTATSF1)

  • Storage and handling:

    • Optimization of buffer composition for stability

    • Determination of appropriate storage conditions

    • Testing of freeze-thaw stability

    • Addition of stabilizing agents or carrier proteins

What are the best practices for validating siRNA-mediated Htatsf1 knockdown experiments?

For robust validation of Htatsf1 knockdown:

  • Essential controls:

    • Universal scrambled negative control siRNA duplexes

    • Positive control siRNAs (e.g., HPRT) with known knockdown efficiency

    • Fluorescent transfection controls to verify >90% transfection efficiency

  • Multi-level validation:

    • mRNA level: qRT-PCR with multiple primer sets

    • Protein level: Western blot with validated antibodies

    • Functional assays: HIV-1 transcription reporter systems

  • Technical considerations:

    • Testing multiple siRNA sequences (e.g., 3 unique 27mer siRNA duplexes)

    • Dose-response studies to determine optimal concentration

    • Time-course analysis to determine knockdown kinetics

    • Cell-type specific optimization of transfection conditions

  • Performance standards:

    • Expect at least 70% knockdown of target mRNA when used at 10 nM concentration

    • At least two of three siRNA duplexes should provide efficient knockdown

    • Consistent phenotypes across multiple siRNA sequences targeting different regions

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 2025 TheBiotek. All Rights Reserved.