HERV-K

Endogenous retrovirus K Envelope Human Recombinant
Shipped with Ice Packs
In Stock

Description

Virological Classification and Genomic Features

HERV-K belongs to the Betaretrovirus genus and represents the youngest, most intact endogenous retrovirus family in humans . Key classification details:

Taxonomic RankClassification
RealmRiboviria
PhylumArtverviricota
ClassRevtraviricetes
OrderOrtervirales
FamilyRetroviridae
GenusBetaretrovirus (unranked subgroup)

The HERV-K (HML-2) subgroup contains 91 intact proviruses with preserved open reading frames (ORFs) for gag, pro, pol, and env genes . Alternative splicing produces two regulatory proteins:

  • Rec: Nuclear RNA export protein (similar to HIV Rev)

  • Np9: Oncoprotein interacting with cellular signaling pathways

Cancer Associations and Mechanisms

HERV-K exhibits tumor-specific expression across multiple malignancies:

Table 1: HERV-K in Human Cancers

Cancer TypeKey FindingsClinical Correlation
Breast Cancer- 70% express HERV-K env
- Basal subtype shows 1.7× higher expression
Reduced survival (HR=2.1, p<0.001)
Melanoma- Viral particles observed
- Required for metastatic transition
4× increased cell lysis with CAR T-cells
Pancreatic Cancer- 80% tumor positivity
- shRNA knockdown reduces metastasis by 89%
RAS-ERK pathway activation
Ovarian Cancer- Env expression in 62% tumors
- Induces EMT via ERK1/2
Correlation with platinum resistance (p=0.03)

Mechanistic studies reveal:

  • Epithelial-Mesenchymal Transition (EMT): HERV-K env upregulates Snail/Slug transcription factors (≥2.5×) and decreases E-cadherin

  • Oncogenic Signaling: Activates RAS-ERK (≥3× p-ERK increase) and PI3K-AKT pathways

  • Immune Evasion: Env protein suppresses CD3+ T-cell proliferation by 40%

Biomarker Potential

  • Serum Detection:

    • Pancreatic cancer: 82% sensitivity for HERV-K RNA vs. 12% in controls (p<0.001)

    • Prostate cancer: env mRNA levels differentiate Gleason ≥7 tumors (AUC=0.87)

  • Tissue Markers:

    • Breast cancer: HERV-K RT predicts recurrence (HR=3.2, 95% CI 1.8-5.6)

    • Melanoma: Env expression correlates with Clark level (ρ=0.71)

Immunotherapy Development

CAR T-Cell Trials:

  • Anti-HERV-K CAR T-cells achieve 95% target cell lysis in vitro

  • Mouse xenografts show 10× tumor volume reduction vs. controls

Antibody Therapies:

  • 6H5 monoclonal antibody reduces breast cancer growth by 78% in models

  • HERV-K vaccination prevents renal cell carcinoma metastasis in mice (p=0.008)

Neurodegenerative Disorders

  • ALS: Anti-HERV-K antibodies present in 89% patients vs. 7% controls

  • HIV Interaction: HERV-K+ T-cells increase 3.5× during uncontrolled HIV replication

Autoimmune Conditions

DiseaseHERV-K FeatureClinical Impact
Rheumatoid ArthritisIgG anti-gag antibodies (2.4× increase) Correlates with CRP levels (r=0.62)
Multiple SclerosisEnv superantigen production Contradictory cohort studies

Evolutionary Insights

  • Primate Speciation: Rhesus macaques contain 145 HML-2-like proviruses with MER11 recombination events (~20 MYA)

  • Human-Specific Elements: 36 proviruses show <1% LTR divergence, suggesting recent activity

Research Challenges and Future Directions

  1. Technical Limitations:

    • Current assays cannot distinguish exogenous vs. endogenous viral particles

    • Epigenetic regulation mechanisms remain partially characterized

  2. Therapeutic Considerations:

    • Potential off-target effects from shared human/retroviral epitopes

    • Need for subtype-specific targeting (HML-2 vs. other HERVs)

  3. Emerging Opportunities:

    • CRISPR-based silencing reduces melanoma growth by 65%

    • Combination therapies with immune checkpoint inhibitors

Product Specs

Introduction
HERVK is associated with several tumors as a passenger virus, and anti-retroviral therapy targeting amyotrophic lateral sclerosis (ALS) has shown promise in reducing ALS symptoms. Numerous viral sequences reside in the human genome but remain dormant. However, these viruses can be activated under pathological conditions. Human Endogenous Retrovirus-K is expressed in the neurons of a subset of ALS patients. The envelope protein of HERVK can lead to neuronal degeneration, and transgenic animals expressing this protein develop an ALS-like syndrome, potentially triggered by nucleolar dysfunction in motor neurons. The reactivation of HERVK is regulated by the transcription factor TDP-43. Consequently, therapeutic strategies targeting this virus could potentially modify the disease course.
Description
This recombinant HERV-K protein, derived from E. coli, is a truncated version fused to a C-terminal Six-histidine tag. It has a molecular weight of 51.5 kDa and an isoelectric point of 9.06.
Purity
The protein purity is greater than 90% as determined by SDS-PAGE.
Physical Appearance
The product is a sterile-filtered, clear solution.
Formulation
The protein is supplied in 10 mM Tris-HCl buffer at pH 7.2.
Stability
While HERV-K remains stable at 4°C for up to one week, storage below -18°C is recommended. Avoid repeated freeze-thaw cycles.
Applications
The use of this product for detecting Human Endogenous Retrovirus K in individuals has not been validated.
Source

Escherichia Coli.

Amino Acid Sequence

MWTVPSFTND SYQVYNVFST NSFQLLTVKR TPHEAWRVPL TTKTNKTKGL PDCPKKPTNG PFIVTSILWD NCNAPKAVVL QTLAMGIVID WAPKGHYWQD CSSKNTLCSE FIYSLDYIEH GWQSYTMRQR VSPYPFKWMD TGIAPPRPKI IHPFFTPEHP ELWKLAAALS GIKIWNTTYQ LLRTKTKTPT FNITLISEWV IPIRSCVKPP YMLLVGNIIM MPDAQTIECH NCKLFTCIDA TFNPTTSILL VRAREGVWIP VSLHRPWESS PSIHIVNEVL KDILKRTKRF IFTLIAVLAG LLAVTATAAT AGVAIRSSVQ TAHYVEACQK NSSRLWNSQA QIDQKLANQI NDLRQSVTWL GDRVMNLQHR MQLQCDWNTS DYCITPYAYN QDQHSWENVS RHLKAWDDNL TLDISQLKEQIFEASQAHLS TVPGSHIFEG ITKQLPDFNP FKWLKPVRGS LLLLALLILV CLCCLLLVCRCL.

Q&A

What is HERV-K and how prevalent is it in the human genome?

HERV-K is a family of human endogenous retroviruses that comprises nearly 8% of the human genome, derived from ancient integrations of retroviruses into the germline . The HERV-K group (also known as HML-2 elements) exists in approximately 30 full-length copies and around 2,000 solitary long terminal repeats (LTRs) . Unlike most endogenous retroviral families, certain HERV-K elements have maintained functional retroviral capabilities, including the ability to encode enzymatic proteins, produce viral particles, and express autoimmune antigens . The retention of these functions makes HERV-K particularly significant for research into viral evolution and potential pathogenic mechanisms.

How are HERV-K elements classified and what is their evolutionary history?

HERV-K is phylogenetically considered a supergroup of viruses, with the HML-2 subtype being the most active and extensively studied . These elements have spread throughout primate evolution, with some integrations specifically occurring in the human lineage . Evolutionary analyses have revealed that while most HERV-K elements integrated prior to the divergence of hominoids from Old World monkey lineages, certain HERV-K elements show evidence of recent integration, as demonstrated by the minimal sequence divergence between LTRs flanking elements like HERV-K10 .

The classification system for HERV-K includes:

HERV-K SubgroupKey CharacteristicsEvolutionary Age
HML-2Most active, best studied, some complete ORFsMost recent, including human-specific integrations
Other HML groupsVarying degrees of defectivenessEarlier integrations, generally shared across primates

What molecular techniques are most effective for detecting HERV-K expression?

Research demonstrates that multiple molecular techniques can effectively quantify HERV-K expression. Droplet digital PCR (ddPCR) has been successfully employed to measure the HERV-K env gene in serum samples from patients with neurological conditions . Additionally, quantitative PCR (qPCR) has proven effective for analyzing HERV-K env expression in tissue samples, particularly in disease-affected brain regions . For protein-level detection, confocal immunofluorescence microscopy using antibodies against HERV-K reverse transcriptase protein provides visual confirmation of expression and cellular localization .

These methods should be selected based on the specific research question:

  • Use ddPCR for highly sensitive quantification in liquid biopsies

  • Apply qPCR for relative expression analysis in tissue samples

  • Implement immunofluorescence for spatial localization studies

How is HERV-K implicated in cancer development and progression?

HERV-K expression has been associated with various malignancies, including breast cancer and both acute and chronic leukemias . In pediatric acute lymphoblastic leukemia (ALL), studies have specifically investigated the expression patterns of HERV-K np9, a spliced product of the env gene implicated in cancer development . The oncogenic potential of HERV-K appears linked to several mechanisms:

  • HERV-K np9, derived from the env gene, may directly contribute to malignant transformation

  • Activation of HERV-K elements can trigger inflammatory pathways that promote cancer development

  • HERV-K proteins may interact with cellular regulatory proteins, disrupting normal cellular function

Research indicates that the immunocompromised state resulting from leukemia or its treatment may increase susceptibility to HERV-K reactivation, potentially creating a pathological feedback loop .

What evidence links HERV-K to neurodegenerative diseases?

Multiple studies have established connections between HERV-K expression and neurodegenerative conditions. In behavioral variant frontotemporal dementia (bvFTD), HERV-K env levels are significantly elevated in patient serum compared to controls (P = 3.5 × 10^-6), with an AUC value of 0.867, indicating strong diagnostic potential . Additionally, HERV-K env transcripts are specifically elevated in the superior frontal cortex of bvFTD patients with TDP-43 pathology .

Similar patterns have been observed in amyotrophic lateral sclerosis (ALS), where TDP-43 pathology coexists with HERV-K activation . In neuronal cell models, overexpression of TDP-43 induces HERV-K env transcription, suggesting a mechanistic link between protein aggregation pathology and retroviral element activation .

How does HERV-K interact with the immune system in autoimmune disorders?

HERV-K has been implicated in several autoimmune conditions, notably type I diabetes. Research has identified a HERV-K env-encoded superantigen associated with insulin-dependent diabetes mellitus (IDDM) . This autoantigen is detected in IDDM patients, raising the possibility that genetic susceptibility to developing the disease could be linked to variations in HERV-K expression or presence .

The mechanisms through which HERV-K contributes to autoimmunity likely include:

  • Molecular mimicry between HERV-K proteins and self-antigens

  • Superantigen-mediated non-specific T-cell activation

  • Alteration of immune tolerance through aberrant HERV-K expression during development

What structural insights exist for HERV-K reverse transcriptase and how can they inform inhibitor design?

Recent crystallographic studies have determined the X-ray structure of HERV-K reverse transcriptase (RT) in a ternary complex with double-stranded DNA substrate . This structural analysis has revealed important insights:

  • HERV-K RT shows structural similarities to diverse RT families

  • There is striking similarity to the HIV-1 RT asymmetric heterodimer

  • The structure explains the poor inhibition by 3TC (lamivudine) and lack of inhibition by nonnucleoside inhibitors like nevirapine and efavirenz

These insights provide crucial information for the rational design of selective HERV-K RT inhibitors. The structural differences between HERV-K RT and other viral RTs offer opportunities for developing compounds that specifically target HERV-K without affecting other polymerases, which is essential for both research tools and potential therapeutic applications .

What are the optimal cell and animal models for studying HERV-K pathogenesis?

While the search results don't explicitly detail optimal models, we can infer from the methodologies described that several systems are valuable for HERV-K research:

  • Cellular models:

    • Neuronal cell lines with inducible TDP-43 expression have successfully demonstrated HERV-K env transcriptional activation

    • Lymphocytic cell lines are appropriate for studying HERV-K in hematological malignancies

  • Primary tissue analysis:

    • Disease-affected brain regions, particularly superior frontal cortex, for neurodegenerative conditions

    • Peripheral blood mononuclear cells from leukemia patients

  • Serum biomarker studies:

    • Patient serum analysis by ddPCR has proven effective for quantifying circulating HERV-K env

While not explicitly mentioned in the search results, transgenic animal models expressing human HERV-K elements would likely provide valuable insights into pathogenesis.

How can HERV-K activation be experimentally induced and measured in laboratory settings?

Based on the research, several approaches can be employed to induce and measure HERV-K activation:

  • Induction methods:

    • Viral infection: Studies have investigated associations between HERV-K expression and infections with herpes simplex virus (HSV), human parvovirus B19, and polyomavirus BK

    • TDP-43 overexpression: In neuronal cell lines, this has been shown to induce HERV-K env transcription

    • Immunosuppressive conditions: The immunocompromised state associated with diseases like leukemia or its treatment may trigger HERV-K reactivation

  • Measurement techniques:

    • Molecular quantification: ddPCR and qPCR for nucleic acid detection

    • Protein detection: Confocal immunofluorescence microscopy with HERV-K-specific antibodies

    • Functional assays: Reverse transcriptase activity assays to assess enzymatic function

Which inhibitors show efficacy against HERV-K reverse transcriptase activity?

Research on HERV-K RT inhibition has revealed varying degrees of efficacy among different compounds:

Inhibitor TypeExamplesEfficacy Against HERV-K RTNotes
Nucleotide analogsVarious antiretroviralsVariable range of inhibitionStructure explains differential efficacy
Nonnucleoside analogsNevirapine, EfavirenzNo inhibitionStructural differences explain lack of activity
Lamivudine (3TC)-Poor inhibitionStructure clarifies reasons for limited efficacy

The crystal structure of HERV-K RT provides crucial insights into these efficacy patterns and suggests opportunities for developing selective HERV-K RT inhibitors . This structural information can guide rational drug design approaches targeting unique features of the HERV-K RT.

What clinical trials have explored targeting HERV-K in disease contexts?

While the search results do not specifically detail clinical trials, they do mention that "several clinical studies of antiviral RT inhibitors" have been inspired by HERV associations with diseases . These studies have likely focused on conditions where HERV-K expression is linked to pathology, such as certain cancers, autoimmune disorders, and neurodegenerative diseases.

The structural characterization of HERV-K RT is described as enabling "the design of selective HERV-K RT tools for drug target validation" , suggesting that more targeted clinical approaches may be forthcoming. Current research appears to be in the phase of establishing HERV-K as a valid therapeutic target before moving to large-scale clinical trials.

How might epigenetic approaches be utilized to modulate HERV-K expression?

Although the search results don't directly address epigenetic approaches, they do mention that many HERV elements are "defective or silenced through epigenetic changes" . This observation suggests that HERV-K expression is naturally regulated by epigenetic mechanisms, providing potential targets for therapeutic intervention.

Epigenetic approaches that might be explored include:

  • DNA methylation modulators targeting HERV-K LTRs

  • Histone modification enzymes that affect chromatin accessibility at HERV-K loci

  • Non-coding RNA strategies to induce transcriptional gene silencing of HERV-K elements

These approaches could provide more specific and potentially safer alternatives to broad-spectrum antiretroviral drugs, particularly in chronic conditions where long-term treatment is necessary.

What are the primary technical challenges in distinguishing pathological from physiological HERV-K expression?

Specific technical challenges include:

  • Establishing appropriate tissue-specific and developmental-stage-specific baseline expression levels

  • Distinguishing between expression of different HERV-K subtypes with potentially distinct functions

  • Determining causality versus correlation in disease associations

  • Accounting for individual genetic variation in HERV-K copy number and polymorphisms

Future research should focus on developing standardized quantification methodologies and reference ranges for different tissues and conditions.

How can researchers differentiate between actual HERV-K protein expression and antibody cross-reactivity?

While not explicitly addressed in the search results, this represents an important methodological consideration. Best practices would include:

  • Using multiple antibodies targeting different HERV-K epitopes

  • Validating antibody specificity through techniques like:

    • Western blotting with recombinant HERV-K proteins

    • Peptide competition assays

    • Genetic knockdown of HERV-K expression

  • Correlating protein detection with nucleic acid measurements

  • Including appropriate negative controls lacking HERV-K expression

Careful validation of detection methods is particularly important given the sequence similarity between different HERV families and the potential for cross-reactivity.

What emerging technologies show promise for advancing HERV-K research?

Based on current research trends, several emerging technologies are likely to advance HERV-K research:

  • Structural biology approaches: X-ray crystallography has already provided valuable insights into HERV-K RT , and cryo-electron microscopy could further illuminate complex structures.

  • Single-cell technologies: Single-cell RNA sequencing could reveal cell-type-specific expression patterns and heterogeneity within tissues.

  • CRISPR-based tools: Precise genome editing could enable functional studies of specific HERV-K elements without affecting others.

  • Long-read sequencing: Technologies like Oxford Nanopore or PacBio sequencing could better characterize full-length HERV-K integrations and their genomic contexts.

  • Spatial transcriptomics: These methods could map HERV-K expression within tissue architecture, particularly valuable for understanding neurodegenerative diseases.

Product Science Overview

Introduction to Human Endogenous Retroviruses (HERVs)

Human endogenous retroviruses (HERVs) are remnants of ancient viral infections that have integrated into the human genome. These viral sequences are passed down through generations and now constitute approximately 8% of the human genome . HERVs are classified based on the tRNA primer binding site used to initiate reverse transcription, and they are generally defective in viral replication due to accumulated mutations and deletions .

HERV-K: A Unique Subgroup

Among the various HERV families, HERV-K (HML-2) is one of the most recently integrated and biologically active groups. HERV-K retains open reading frames (ORFs) capable of encoding functional proteins, including the Gag, Pol, and Env proteins . The envelope (Env) protein of HERV-K is particularly noteworthy as it plays a crucial role in viral entry and has been implicated in various physiological and pathological processes .

The Envelope Protein (Env) of HERV-K

The Env protein of HERV-K is a glycoprotein that facilitates the virus’s entry into host cells by mediating the fusion of the viral and cellular membranes. This protein is composed of two subunits: the surface (SU) subunit, which is responsible for receptor binding, and the transmembrane ™ subunit, which mediates membrane fusion . The Env protein is also known for its immunosuppressive properties, which can modulate the host immune response .

Recombinant HERV-K Env Protein

The recombinant HERV-K Env protein is produced using recombinant DNA technology, where the gene encoding the Env protein is cloned and expressed in a suitable host system, such as bacteria or mammalian cells. This recombinant protein can be used for various research purposes, including studying the protein’s structure and function, developing diagnostic tools, and exploring its potential as a therapeutic target .

Biological and Clinical Significance

HERV-K Env protein has been associated with several diseases, including cancer and autoimmune disorders. Reactivation of HERV-K expression has been observed in various cancers, such as melanoma, breast cancer, and ovarian cancer . The Env protein’s immunosuppressive properties may contribute to tumor immune evasion, making it a potential target for cancer immunotherapy .

In addition to its role in cancer, HERV-K Env protein has been implicated in neurological disorders, such as amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS). The protein’s expression in the central nervous system and its potential to induce neuroinflammation highlight its relevance in these diseases .

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.