HSD17B8 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Role of HSD17B8

HSD17B8 belongs to the short-chain dehydrogenase/reductase (SDR) superfamily and functions as a 17-beta-hydroxysteroid dehydrogenase . It oxidizes active estrogens and androgens (e.g., E2, testosterone) into less active forms (e.g., E1, androstenedione) . Its interaction with PTEN and role in ERK/MAPK signaling highlight its importance in regulating cell proliferation and tumor suppression .

Cancer Biology

  • Breast Cancer Prognosis: High HSD17B8 expression correlates with improved survival in ER+ breast cancer patients . Antibodies confirmed reduced HSD17B8 protein levels in tumor tissues versus normal .

  • Cell Cycle Regulation: Knocking down HSD17B8 in MCF-7 cells increased phosphorylated ERK1/2 and arrested cells in G2/M phase .

Hormonal Pathways

  • Estrogen Metabolism: HSD17B8 antibodies validated its enzymatic activity in converting E2 to E1, which modulates ERα signaling .

  • PTEN Interaction: Co-immunoprecipitation studies revealed PTEN physically binds HSD17B8, inhibiting its dehydrogenase activity and suppressing tumor growth .

Validation Data from Key Studies

StudyMethodKey ResultCitation
MCF-7 Cell AnalysisWB, Flow CytometryHSD17B8 knockdown reduced ERK activation and increased G2/M arrest
Tissue MicroarrayIHCLower HSD17B8 protein levels in breast cancer vs. normal tissues
ERα RegulationChIP, Co-IPERα recruits C/EBPβ to HSD17B8 promoter upon E2 stimulation

Challenges and Considerations

  • Molecular Weight Discrepancy: Observed molecular weight (~34 kDa) differs from calculated (27 kDa), likely due to post-translational modifications .

  • Species Cross-Reactivity: Most antibodies target human HSD17B8, with limited validation in rodents .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery times may vary depending on the purchase method and location. For specific delivery details, please consult with your local distributors.
Synonyms
17 beta HSD 8 antibody; 17 beta hydroxysteroid dehydrogenase 8 antibody; 17-beta-HSD 8 antibody; 17-beta-hydroxysteroid dehydrogenase 8 antibody; 17-beta-hydroxysteroid dehydrogenase VIII antibody; 3-oxoacyl-[acyl-carrier-protein] reductase antibody; 3-oxoacyl-acyl-carrier protein reductase, E. coli, homolog of antibody; Beta ketoacyl [acyl carrier protein] reductase like antibody; D6S2245E antibody; DHB8_HUMAN antibody; dJ1033B10.9 antibody; Estradiol 17 beta dehydrogenase 8 antibody; Estradiol 17-beta-dehydrogenase 8 antibody; Estrogen 17 oxidoreductase antibody; FABG antibody; FabG, E. coli, homolog of antibody; FabG-like antibody; FABGL antibody; H2 KE6 antibody; H2-KE6 antibody; HKE6 antibody; HSD17B8 antibody; Hydroxysteroid (17 beta) dehydrogenase 8 antibody; Ke-6 antibody; KE6 antibody; KE6, mouse homolog of antibody; Protein Ke6 antibody; Really interesting new gene 2 protein antibody; RING2 antibody; SDR30C1 antibody; Short chain dehydrogenase/reductase family 30C member 1 antibody; Testosterone 17 beta dehydrogenase 8 antibody; Testosterone 17-beta-dehydrogenase 8 antibody
Target Names
HSD17B8
Uniprot No.

Target Background

Function
HSD17B8 antibody is essential for the solubility and assembly of the heterotetrameric 3-ketoacyl-[acyl carrier protein] (ACP) reductase functional complex (KAR or KAR1) that constitutes a part of the mitochondrial fatty acid synthase (mtFAS). As the alpha-subunit of the KAR complex, HSD17B8 serves as a scaffold protein. Its presence is critical for the stability of carbonyl reductase type-4 (CBR4, beta-subunit of the KAR complex) and for its 3-ketoacyl-ACP reductase activity. This contributes to mitochondrial fatty acid biosynthesis. HSD17B8 also catalyzes the NAD-dependent conversion of (3R)-3-hydroxyacyl-CoA into 3-ketoacyl-CoA (3-oxoacyl-CoA) without any chain length preference. This enzymatic activity, however, is not required for the KAR function. HSD17B8 exhibits a stronger preference for (3R)-3-hydroxyacyl-CoA over (3S)-3-hydroxyacyl-CoA and demonstrates enzymatic activity only in the presence of NAD(+). In collaboration with enoyl-CoA hydratase 1 within mitochondria, HSD17B8 forms an alternative route to the auxiliary enzyme pathways involved in the breakdown of Z-PUFA (cis polyunsaturated fatty acid) enoyl-esters. Additionally, HSD17B8 acts as a NAD-dependent 17-beta-hydroxysteroid dehydrogenase, exhibiting the highest activity towards estradiol (17beta-estradiol or E2). Its activity towards testosterone and dihydrotestosterone (17beta-hydroxy-5alpha-androstan-3-one) is considerably lower. Primarily an oxidative enzyme, HSD17B8 can switch to a reductive mode under appropriate physiological conditions, catalyzing the reduction of estrone (E1) to form biologically active 17beta-estradiol.
Gene References Into Functions
  1. Aromatase, 17beta-HSD type 7, and 17beta-HSD type 12, but not 17beta-HSD type 1, are commonly expressed in human breast cancer. PMID: 16930994
  2. Transcription of the HSD17B8 gene in the liver is regulated by C/EBPbeta. PMID: 17583490
  3. The expression of the human Ke 6 gene, 17beta-hydroxysteroid dehydrogenase type 8, in E. coli and the substrate specificity of the expressed protein were examined. The tissue distribution of mRNA expression of the human Ke 6 gene was also investigated. PMID: 17978863
  4. ERalpha is involved in the transcriptional regulation of the HSD17B8 gene in response to E2 through its interaction with C/EBPbeta. PMID: 18852215
  5. Hs17beta-HSD8 and HsCBR4 exhibit a strong genetic interaction in vivo in yeast. These genes, when expressed together, rescue the respiratory deficiency and restore the lipoic acid content of oar1Delta cells. PMID: 19571038
  6. HSD17B8 mRNA is expressed in human skin, at lower levels in the face compared to the arm or hip. HSD17B8 levels remain unaffected by topical 17-beta-estradiol treatment. PMID: 18794456
Database Links

HGNC: 3554

OMIM: 601417

KEGG: hsa:7923

STRING: 9606.ENSP00000363794

UniGene: Hs.415058

Protein Families
Short-chain dehydrogenases/reductases (SDR) family
Subcellular Location
Mitochondrion matrix.
Tissue Specificity
Widely expressed, particularly abundant in prostate, placenta and kidney. Expressed at protein level in various tissues like brain, cerebellum, heart, lung, kidney, ovary, testis, adrenals and prostate.

Q&A

What is HSD17B8 and what is its primary function in cellular metabolism?

HSD17B8 is an NAD-dependent 17-beta-hydroxysteroid dehydrogenase that demonstrates highest catalytic activity toward estradiol. Its primary function involves converting estradiol (E2) to estrone (E1) through oxidative activity . Additionally, when complexed with CBR4, it exhibits NADH-dependent 3-ketoacyl-acyl carrier protein reductase activity, suggesting a potential role in mitochondrial fatty acid biosynthesis .

Methodologically, researchers investigating HSD17B8 function should consider:

  • Measuring enzyme activity using NAD as a cofactor

  • Evaluating both steroid conversion (E2→E1) and potential fatty acid metabolic functions

  • Examining protein-protein interactions, particularly with CBR4 and PTEN

How should researchers select the appropriate HSD17B8 antibody for their experimental design?

Selection criteria should be based on:

ApplicationRecommended Antibody TypeDilution RangeSpecial Considerations
Western BlotPolyclonal (e.g., 16752-1-AP)1:500-1:2000Expected MW: 27 kDa (calculated), 34 kDa (observed)
ImmunohistochemistryPolyclonal antibodies1:50-1:500Antigen retrieval with TE buffer pH 9.0 recommended
ImmunofluorescenceABIN7256494 (polyclonal)Application-dependentValidated for human, mouse, rat samples

For optimal results, researchers should:

  • Validate antibody specificity using positive controls (HeLa cells, human testis tissue)

  • Consider cross-reactivity requirements based on species (human, mouse, rat)

  • Select antibodies targeting specific domains based on research questions (e.g., AA 28-127 vs. full-length protein)

How does HSD17B8 influence breast cancer proliferation, and what experimental approaches are optimal for studying this relationship?

Recent findings demonstrate that HSD17B8 plays a crucial role in breast cancer cell proliferation through estrogen metabolism regulation. HSD17B8 converts estradiol (E2) to estrone (E1), with these hormones having opposing effects on cancer cells .

Key experimental findings:

  • E1 stimulates breast cancer cell proliferation while E2 has inhibitory effects

  • HSD17B8 knockdown significantly suppresses growth of MCF-7 cells

  • E2 treatment and HSD17B8 knockdown arrest tumor cells in G2/M phase

Recommended experimental approaches:

  • siRNA-mediated knockdown of HSD17B8 in ER+ breast cancer cell lines

  • Flow cytometry for cell cycle analysis following E1/E2 treatment

  • Western blot analysis of phosphorylated ERK levels as a downstream indicator

  • Colony formation assays to assess long-term proliferative effects

What is the relationship between PTEN and HSD17B8, and how can researchers effectively study this interaction?

The PTEN-HSD17B8 interaction represents a novel regulatory mechanism in cell proliferation. Research indicates that:

  • PTEN physically interacts with HSD17B8 to inhibit its enzymatic conversion of E2 to E1

  • Loss of PTEN results in released HSD17B8 activity, leading to decreased E2 levels

  • This regulatory relationship affects ERK/MAPK activation pathways

Methodological approaches for studying this interaction:

  • Co-immunoprecipitation assays to confirm physical interaction

  • In vitro enzyme activity assays with purified proteins

  • Proximity ligation assays in intact cells

  • Comparative analysis of E1:E2 ratios in PTEN-deficient versus normal cells

  • Combined knockdown experiments to establish epistatic relationships

How can researchers effectively measure and interpret changes in HSD17B8 enzymatic activity in experimental models?

Quantifying HSD17B8 activity requires specialized approaches:

MethodApplicationTechnical Considerations
LC/MS metabolomicsMeasuring E1:E2 ratios in cell/tissue extractsRequires specialized equipment, high sensitivity
In vitro conversion assaysDirect measurement of enzymatic activityUses purified protein or cell lysates with NAD+ cofactor
Western blottingProtein expression correlationNot direct activity measurement; use antibodies like 16752-1-AP
Gene expression analysisTranscriptional regulationDoes not necessarily correlate with enzymatic activity

For robust results, researchers should:

  • Include appropriate controls (NAD+ vs. NADH dependency)

  • Correlate expression levels with activity measurements

  • Consider the heterotetramer formation with CBR4 when studying fatty acid metabolism functions

  • Account for post-translational modifications that may affect enzyme activity

What are the critical parameters for successful Western blotting of HSD17B8?

Optimized Western blot protocols for HSD17B8 detection should address:

  • Sample preparation: Effective lysis in tissues with high expression (liver, small intestine, testis)

  • Expected molecular weight: 27 kDa (calculated) vs. 34 kDa (observed)

  • Antibody selection: Polyclonal antibodies (e.g., 16752-1-AP) at 1:500-1:2000 dilution

  • Blocking conditions: Optimize based on antibody manufacturer recommendations

  • Positive controls: HeLa cells, mouse liver tissue, human testis tissue

Technical validation approaches:

  • Knockdown/knockout controls to confirm band specificity

  • Use of recombinant HSD17B8 as positive control

  • Comparison of multiple antibodies targeting different epitopes

What approaches should researchers use to resolve contradictory findings in HSD17B8 functional studies?

Contradictory results have been reported regarding HSD17B8 function, particularly in metabolic contexts . To address these contradictions, researchers should:

  • Consider tissue-specific effects:

    • Analyze expression patterns across different tissues

    • Perform conditional knockout studies rather than global deletion

  • Evaluate experimental model differences:

    • Compare in vitro vs. in vivo systems

    • Assess acute vs. chronic manipulation of HSD17B8 levels

    • Consider compensatory mechanisms in knockout models

  • Examine dual enzymatic functions:

    • Separately assess steroid metabolism vs. fatty acid metabolism roles

    • Investigate context-dependent protein complex formation (e.g., with CBR4)

  • Control for background genetic differences:

    • Use isogenic cell lines for comparative studies

    • Consider strain background in mouse models

How can researchers effectively validate knockdown or inhibition of HSD17B8 in their experimental systems?

Validation of HSD17B8 manipulation requires multi-level confirmation:

Validation LevelTechniquesNotes
GenomicPCR, sequencingFor CRISPR/Cas9 edited lines
TranscriptionalqRT-PCR, RNA-seqConfirm mRNA reduction
ProteinWestern blot, immunofluorescenceUsing validated antibodies (e.g., 16752-1-AP)
FunctionalE2→E1 conversion assayDirect measurement of enzymatic activity
PhenotypicCell proliferation, cell cycle analysisAssess downstream effects based on research context

For siRNA approaches:

  • Use multiple siRNA sequences to control for off-target effects

  • Implement dose-response studies to determine optimal knockdown conditions

  • Establish time course for protein depletion after siRNA treatment

  • Include appropriate negative controls (scrambled siRNA)

Beyond estrogen metabolism, what other metabolic roles of HSD17B8 warrant investigation?

Current evidence suggests broader metabolic functions that deserve further research:

  • Mitochondrial fatty acid biosynthesis: The heteroteramer with CBR4 exhibits NADH-dependent 3-ketoacyl-acyl carrier protein reductase activity

  • Potential involvement in liver metabolism and non-alcoholic fatty liver disease pathways

  • Possible roles in metabolic syndrome and insulin resistance related to steroid hormone balance

Methodological considerations for these studies:

  • Mitochondrial isolation techniques for functional assays

  • Lipidomic analysis to profile fatty acid changes

  • Metabolic flux analysis using isotope-labeled precursors

  • Tissue-specific conditional knockout models focusing on liver, adipose tissue, and muscle

How can researchers address the technical challenges in studying protein-protein interactions involving HSD17B8?

HSD17B8 forms functional complexes with multiple proteins, including CBR4 and PTEN . Effective study of these interactions requires:

  • In vitro interaction analysis:

    • Recombinant protein co-immunoprecipitation

    • Surface plasmon resonance to determine binding kinetics

    • Isothermal titration calorimetry for thermodynamic parameters

  • Structural studies:

    • X-ray crystallography of protein complexes

    • Cryo-EM for larger assemblies

    • Hydrogen-deuterium exchange mass spectrometry for interaction interfaces

  • Cellular confirmation techniques:

    • Förster resonance energy transfer (FRET)

    • Bimolecular fluorescence complementation

    • Proximity ligation assays in fixed cells

  • Functional validation:

    • Mutagenesis of predicted interaction domains

    • Activity assays with reconstituted complexes

    • Cellular phenotypes with interaction-deficient mutants

These approaches provide complementary data to fully characterize the interactions that mediate HSD17B8's diverse cellular functions.

What are the most promising therapeutic applications emerging from HSD17B8 research?

Recent findings highlight several potential therapeutic directions:

  • Breast cancer treatment: Inhibiting HSD17B8 may suppress ER+ breast cancer cell proliferation by altering the E1:E2 ratio

  • Metabolic disorders: Modulating HSD17B8 activity could potentially address aspects of fatty liver disease

  • Cell cycle regulation: HSD17B8 manipulation affects G2/M phase arrest, suggesting applications in cancer therapy approaches

Future research should focus on:

  • Development of specific inhibitors of HSD17B8 enzymatic activity

  • Tissue-specific delivery systems to target HSD17B8 in cancer cells

  • Combination approaches with existing therapies like anti-estrogens

  • Clinical correlations between HSD17B8 expression/activity and patient outcomes

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.