HN1L Human

Hematological and Neurological Expressed 1-Like Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

HN1L (Hematological and Neurological Expressed 1-Like), encoded by the HN1L gene, is a conserved 190-amino acid protein expressed in human tissues such as skeletal muscle, prostate, colon, brain, and placenta . It shares structural homology with HN1, a protein involved in AKT/GSK3β signaling . Key features include:

  • NAADP binding: HN1L binds nicotinic acid adenine dinucleotide phosphate (NAADP) with nanomolar affinity, particularly in its C-terminal region .

  • Tissue specificity: Widely expressed, with elevated levels in cancerous tissues .

Cell Cycle Regulation

HN1L promotes cell cycle progression by:

  • Depleting Cyclin D: Facilitates G1-to-S phase transition, enhancing proliferation .

  • Anti-apoptotic effects: Regulates Bax, Bcl-2, and caspase-9 to inhibit apoptosis .

Calcium Signaling

As a NAADP-binding protein, HN1L mediates calcium release in T cells via interactions with ryanodine receptor 1 (RYR1) and the endoplasmic reticulum .

Breast Cancer

MechanismExperimental EvidenceClinical Relevance
HMGB1 regulationHN1L interacts with HSPA9 to upregulate HMGB1, promoting metastasis .High HN1L expression correlates with metastasis and poor prognosis .
In vivo inhibitionSilencing HN1L reduces lung metastasis in nude mice models .Potential therapeutic target .

Esophageal Squamous Cell Carcinoma (ESCC)

MechanismExperimental EvidenceClinical Relevance
PLK1 signalingHN1L enhances proliferation and metastasis via AP-2γ/PLK1 pathways .High expression linked to tumor invasion and chemoresistance .

Cross-Cancer Implications

HN1L is implicated in prostate, colon, and cervical cancers, where its silencing suppresses proliferation and migration .

Molecular Interactions and Pathways

Partner ProteinFunctional RoleCancer Type
HSPA9Regulates HMGB1 expression and metastasisBreast
PLK1Drives tumor progression and chemoresistanceESCC
RYR1Mediates NAADP-dependent calcium signalingT cells

Therapeutic Potential

HN1L’s dual role in promoting survival and driving oncogenesis positions it as a candidate for targeted therapies. Preclinical studies highlight:

  • Silencing strategies: siRNA-mediated knockdown inhibits metastasis in breast and ESCC models .

  • Combination therapies: Enhancing chemosensitivity in ESCC by reducing HN1L expression .

Product Specs

Introduction
Hematological and neurological expressed 1-like (HN1L) is a member of the HN1 family. It is believed to be involved in embryo development and is expressed at varying levels in various tissues, including the liver, kidney, prostate, testis, and uterus.
Description
Recombinant human HN1L, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 213 amino acids (1-190 a.a.) with a molecular weight of 22.5 kDa. It includes a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The HN1L protein solution (1 mg/ml) is supplied in 20mM Tris-HCl buffer (pH 8.0), containing 0.15M NaCl, 10% glycerol, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. Adding a carrier protein like 0.1% HSA or BSA is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
C16orf34, L11, Hematological and neurological expressed 1-like protein, HN1-like protein.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMFQVPDS EGGRAGSRAM KPPGGESSNL FGSPEEATPS SRPNRMASNI FGPTEEPQNI PKRTNPPGGK GSGIFDESTP VQTRQHLNPP GGKTSDIFGS PVTATSRLAH PNKPKDHVFL CEGEEPKSDL KAARSIPAGA EPGEKGSARK AGPAKEQEPM PTVDSHEPRL GPRPRSHNKV LNPPGGKSSI SFY.

Q&A

What is HN1L and what cellular functions does it perform?

HN1L (haematological and neurological expressed 1-like) is an evolutionarily conserved protein that plays critical roles in embryonic development and cellular growth regulation. It belongs to a small, poorly understood family of genes conserved across vertebrate species. Research indicates that HN1L is primarily localized in the cytoplasm and functions in regulating cell cycle progression, particularly promoting G1/S phase conversion and enhancing cellular activity . The protein is expressed in various tissues and has been implicated in multiple cellular processes including cell proliferation, migration, and invasion, particularly in the context of cancer development .

How is HN1L expression regulated in normal versus cancerous tissues?

In normal tissues, HN1L expression is tightly regulated and generally maintained at lower levels compared to cancerous tissues. Bioinformatic analyses using TCGA data demonstrate that HN1L is significantly overexpressed in breast cancer tissues compared to adjacent normal tissues . Similar patterns have been observed in prostate cancer, where HN1L expression is notably lower in normal RWPE-1 cells compared to cancerous androgen-responsive LNCaP and androgen-insensitive PC-3 cells . The regulatory mechanisms controlling this differential expression remain incompletely characterized, though they likely involve transcriptional regulation through pathways active in cellular proliferation and oncogenesis.

What is the evolutionary significance of HN1L?

HN1L is highly conserved across vertebrate species, suggesting fundamental biological importance. Molecular evolution analyses have placed HN1 and HN1L in larger conserved multigene protein families . This evolutionary conservation indicates that HN1L likely performs essential functions in cellular biology that have been maintained throughout vertebrate evolution. The high degree of conservation may also explain why perturbations in HN1L expression or function can have significant impacts on cellular processes and contribute to disease states such as cancer.

How does HN1L contribute to breast cancer metastasis?

HN1L plays a significant role in promoting breast cancer metastasis through multiple mechanisms:

  • Enhanced cell migration and invasion capability: Silencing HN1L significantly inhibits the invasion and metastasis of breast cancer cells (MDA-MB-231 and BT-549) in vitro .

  • Promotion of lung metastasis: In vivo studies using NOD-SCID mice implanted with control and HN1L knockdown MDA-MB-231 cells showed that HN1L knockdown resulted in approximately 70% decrease in lung metastases .

  • Molecular pathway alteration: HN1L interacts with HSPA9 and affects the expression of HMGB1, a protein known to be involved in tumor invasion and metastasis .

  • EMT modulation: Evidence suggests HN1L may influence the expression of epithelial-mesenchymal transition (EMT) markers, facilitating the invasive phenotype of cancer cells .

This multifaceted influence on metastatic processes makes HN1L an appealing potential therapeutic target for breast cancer treatment.

What molecular interactions drive HN1L's role in tumor progression?

The oncogenic functions of HN1L appear to be mediated through specific protein-protein interactions and downstream signaling effects:

  • HN1L-HSPA9 interaction: Co-immunoprecipitation and immunofluorescence assays have demonstrated that HN1L interacts with HSPA9 (a mitochondrial chaperone) in breast cancer cells .

  • HMGB1 regulation: This interaction affects the expression of HMGB1, which is overexpressed in various cancers and involved in tumor invasion and metastasis. Knockdown of HN1L leads to significant downregulation of HMGB1 expression .

  • Centrosome regulation: Related research on HN1 (which shares functional similarities with HN1L) shows interaction with γ-tubulin, affecting centrosome function and microtubule spindle assembly. HN1 depletion leads to increased γ-tubulin foci and disruption in microtubule spindle assembly .

  • Cell cycle regulation: HN1L influences cell cycle progression by affecting the expression of cell cycle regulators, including decreased levels of Cyclin D expression and reduced ratio of cells at the G1 phase .

These molecular interactions collectively contribute to HN1L's role in promoting cellular proliferation, survival, and metastatic capacity in cancer cells.

How does HN1L expression correlate with clinical outcomes in cancer patients?

Analysis of clinical data indicates significant correlations between HN1L expression and cancer patient outcomes:

  • Correlation with metastasis: HN1L overexpression positively correlates with M metastasis in breast cancer patients .

  • Prognosis indicator: Patients with high HN1L expression demonstrate poorer prognosis compared to those with lower expression levels, as analyzed through Kaplan-Meier survival curves .

  • Tissue-specific correlation: Immunohistochemistry studies of tissue paraffin blocks from 115 breast cancer patients revealed higher HN1L expression in cancerous tissues compared to normal tissues .

These clinical correlations suggest that HN1L expression could potentially serve as a prognostic biomarker for cancer progression and patient survival.

What are the recommended techniques for measuring HN1L expression in experimental and clinical samples?

Several complementary techniques have proven effective for measuring HN1L expression:

  • Bioinformatic analysis: Utilizing RNA-seq and RNAseqV2 data from databases such as TCGA, with standardization using the TMM (Trimmed Mean of M-values) method .

  • Western blotting: This technique is effective for protein-level detection, using anti-HN1L antibodies (such as Abcam, NM-144570) with GAPDH as a loading control. Densitometric analysis using software like Image J can quantify relative expression levels .

  • Immunohistochemistry (IHC): For tissue samples, particularly paraffin-embedded specimens, IHC can be performed using specific antibodies with DAB substrate staining. A standardized scoring system should be employed, considering both percentage of positive cells (0-4 scale) and staining intensity (0-3 scale) .

  • Immunofluorescence assay: Useful for determining cellular localization and co-localization with potential interaction partners .

  • qRT-PCR: For mRNA-level quantification, with appropriate housekeeping genes for normalization.

What approaches are most effective for modulating HN1L expression in experimental models?

Several genetic techniques have been successfully employed to modulate HN1L expression:

  • RNA interference (RNAi):

    • Short hairpin RNA (shRNA) constructs targeting HN1L mRNA

    • Delivery via lentiviral vectors for stable knockdown

    • Verification of knockdown efficiency via western blot and qRT-PCR

  • Overexpression systems:

    • Lentivirus vectors containing HN1L cDNA cloned into appropriate expression vectors

    • Flag-tagged constructs to facilitate detection

    • Transfection into target cells using appropriate transfection reagents

  • CRISPR-Cas9 gene editing:

    • For complete knockout studies or precise genomic modifications

    • Design of guide RNAs targeting specific exons

  • Rescue experiments:

    • Re-expression of HN1L in knockdown cells to confirm specificity of observed phenotypes

    • Expression of mutant forms to identify critical functional domains

These approaches should be selected based on the specific research question and experimental context.

What in vivo models are most appropriate for studying HN1L function in cancer?

Several animal models have proven valuable for investigating HN1L functions:

  • Xenograft mouse models:

    • NOD-SCID mice implanted with control and HN1L-knockdown cancer cells

    • Monitoring of tumor growth and metastasis development

    • Small living animal imaging technology for tracking metastatic spread

  • Lung metastasis models:

    • Particularly effective for breast cancer studies

    • Tail vein injection of cancer cells with modulated HN1L expression

    • Quantification of lung nodules and histological confirmation

  • Transgenic mouse models:

    • Tissue-specific overexpression or conditional knockout of HN1L

    • Useful for studying developmental and tissue-specific functions

  • Patient-derived xenografts (PDX):

    • More closely reflecting the heterogeneity and complexity of human tumors

    • Suitable for testing therapeutic approaches targeting HN1L

Selection of the appropriate model should consider the specific cancer type, research question, and desired endpoints.

How does HN1L influence cell cycle progression?

HN1L exerts significant effects on cell cycle regulation through several mechanisms:

  • G1/S phase transition promotion: HN1L upregulation promotes G1/S phase conversion, facilitating cell cycle progression and cellular proliferation .

  • Cyclin D regulation: HN1L upregulation decreases the levels of Cyclin D expression and reduces the ratio of cells at the G1 phase, suggesting complex effects on cell cycle regulatory proteins .

  • E2F, Rb, and P53 pathway activation: In 293T cells, HN1L mainly activates cell cycle-related signaling pathways such as E2F, Rb, and P53, which are critical regulators of cell proliferation and cell cycle progression .

  • Centrosome regulation: Related research on HN1 (functionally similar to HN1L) shows that it is present at centrosomes at different expression/stabilization levels during different phases of the cell cycle, suggesting a role in mitotic regulation .

These effects collectively contribute to HN1L's role in promoting cellular proliferation and potentially contributing to the unrestricted growth characteristic of cancer cells.

What are the key protein interactions of HN1L and their functional significance?

HN1L engages in several critical protein interactions that mediate its cellular functions:

  • HN1L-HSPA9 interaction:

    • Confirmed through co-immunoprecipitation and immunofluorescence assays

    • HSPA9 is a mitochondrial chaperone involved in cell senescence

    • This interaction affects HMGB1 expression, as knockdown of either HN1L or HSPA9 causes decreased HMGB1 protein levels

    • Functionally important for breast cancer cell invasion and metastasis

  • γ-tubulin interaction (shown for the related protein HN1):

    • Co-localization of HN1 with γ-tubulin foci in prostate cancer cells

    • Physical association validated by immunoprecipitation

    • Functional significance in centrosome regulation and microtubule spindle assembly

    • Depletion leads to increased γ-tubulin foci and disruption in microtubule spindle assembly

  • HMGB1 regulatory relationship:

    • HN1L silencing downregulates HMGB1 expression

    • HMGB1 overexpression rescues migration defects in HN1L-silenced cells

    • Suggests HMGB1 is a key downstream effector of HN1L in cancer metastasis

These interactions place HN1L at the intersection of multiple cellular processes including cell cycle regulation, senescence, and cytoskeletal organization.

How does HN1L affect gene expression patterns in cancer cells?

GeneChip analysis following HN1L silencing has revealed significant alterations in gene expression patterns:

  • Upregulated genes: Knockdown of HN1L led to significant accumulation of DDX58, suggesting HN1L may normally suppress this gene .

  • Downregulated genes: HN1L silencing decreased expression of SMAD2, PIM1, and notably HMGB1, indicating these are potentially positively regulated by HN1L .

  • EMT markers: Although not explicitly detailed in the provided research, the role of HN1L in promoting invasion and metastasis suggests potential effects on epithelial-mesenchymal transition (EMT) markers such as E-cadherin, N-cadherin, Slug, Snail, Vimentin, β-catenin, and Twist1, which were examined via western blotting in the research .

  • Cell senescence markers: The interaction between HN1L and HSPA9, combined with the observation that HMGB1 can function as a senescence marker, suggests HN1L may influence gene expression patterns related to cellular senescence .

The full spectrum of gene expression changes mediated by HN1L likely extends beyond these identified targets and warrants further investigation through comprehensive transcriptomic analyses.

What is the potential of HN1L as a therapeutic target in cancer?

HN1L presents several characteristics that make it an appealing therapeutic target:

  • Differential expression: HN1L is significantly overexpressed in breast cancer tissues compared to normal tissues, providing potential cancer specificity for therapeutic interventions .

  • Functional importance: Silencing HN1L inhibits invasion and metastasis both in vitro and in vivo, suggesting therapeutic efficacy could be achieved through HN1L inhibition .

  • Downstream effectors: The identification of HMGB1 as a key downstream effector provides additional options for targeting this pathway at multiple points .

  • Multiple cancer relevance: Evidence of HN1L's importance in both breast and prostate cancers suggests broader applicability across cancer types .

  • Pathway specificity: HN1L's specific interactions with proteins like HSPA9 could potentially allow for targeted therapeutic approaches with minimal off-target effects .

Researchers conclude that "HN1L may provide promising diagnostic and therapeutic options for breast cancer patients in the future" .

What methodological challenges exist in developing HN1L-targeted therapies?

Several significant challenges must be addressed in developing effective HN1L-targeted therapies:

  • Protein-protein interaction targeting: The HN1L-HSPA9 interaction represents a challenging target for small molecule development, as protein-protein interactions often lack well-defined binding pockets.

  • Normal tissue expression: HN1L's role in normal cellular processes and embryonic development suggests potential toxicity concerns for complete inhibition strategies .

  • Mechanistic gaps: The "detailed mechanism of the interaction between HN1L and HSPA9 affecting the expression of HMGB1 still needs further experimental studies" , indicating incomplete understanding of the pathway.

  • Delivery challenges: Effective targeting of HN1L in cancer cells may require innovative delivery approaches, particularly for nucleic acid-based therapeutics.

  • Biomarker development: Identification of patient populations most likely to benefit from HN1L-targeted therapies will require robust biomarker development and validation.

Addressing these challenges will require multidisciplinary approaches combining structural biology, medicinal chemistry, and advanced delivery technologies.

What are the most promising research directions for advancing our understanding of HN1L biology?

Several research directions hold particular promise for advancing HN1L science:

  • Structural characterization: Elucidating the three-dimensional structure of HN1L and its complexes with interacting partners like HSPA9 would facilitate rational drug design efforts.

  • Comprehensive interactome mapping: Expanding our understanding of HN1L's protein interaction network beyond the currently identified partners could reveal new functional roles and therapeutic opportunities.

  • Tissue-specific functions: Investigation of HN1L's role across different tissue types and cancer models would clarify the breadth of its biological significance.

  • Mechanistic detailing: Further investigation of "the detailed mechanism of the interaction between HN1L and HSPA9 affecting the expression of HMGB1" represents a critical knowledge gap.

  • Therapeutic proof-of-concept: Development and testing of preliminary HN1L-targeting approaches in preclinical models would establish feasibility for therapeutic development.

  • Clinical correlation studies: Larger and more diverse patient cohorts should be examined to validate the relationship between HN1L expression and clinical outcomes across cancer types.

These research directions would collectively advance both fundamental understanding of HN1L biology and its potential therapeutic applications.

Product Science Overview

Introduction

Hematological and Neurological Expressed 1-Like (HN1L), also known as C16orf34, CRAMP_1-like, or KIAA1426, is a protein encoded by the HN1L gene in humans. This protein has garnered attention due to its involvement in various biological processes and its potential as a therapeutic target in certain cancers.

Gene and Protein Structure

The HN1L gene is located on chromosome 16 and encodes a protein that is approximately 22.5 kDa in size. The recombinant form of this protein, often used in research, is typically expressed in E. coli and purified using conventional chromatography techniques. The recombinant protein is fused to a His-tag at the N-terminus to facilitate purification and detection .

Expression and Function

HN1L is expressed in a variety of tissues, including the liver, kidney, prostate, testis, and uterus, at varying levels . It is proposed to play a role in embryo development, although its exact functions in adult tissues are still being elucidated.

Role in Cancer

Recent studies have highlighted the significance of HN1L in cancer biology, particularly in triple-negative breast cancer (TNBC). TNBC is characterized by its aggressive clinical behavior and resistance to conventional chemotherapy. Research has shown that HN1L is upregulated in a significant proportion of breast cancer cases and is negatively correlated with relapse-free survival in TNBC patients .

HN1L has been identified as a novel therapeutic target for TNBC due to its role in regulating cancer stem cell self-renewal. Silencing HN1L in vitro and in vivo has been shown to suppress the breast cancer stem cell population, sensitize tumors to chemotherapy, and reduce metastasis . Mechanistically, HN1L is believed to function as a transcription factor that regulates the expression of STAT3 and related genes, promoting pathways that support cancer stem cell self-renewal .

Research and Applications

The recombinant form of HN1L is widely used in research to study its function and potential therapeutic applications. It is available from various suppliers and is typically used in studies involving protein-protein interactions, gene expression analysis, and cancer research .

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.