LXN Mouse

Latexin Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Hematopoietic Stem Cell Regulation

  • Key Finding: Lxn⁻/⁻ mice exhibit a 56% increase in long-term hematopoietic stem cells (LT-HSCs) and progenitors (CMP, GMP, CLP) compared to wild-type (WT) mice .

  • Mechanism: LXN deficiency disrupts carboxypeptidase-mediated regulation, enhancing stem cell proliferation.

Inflammatory Bowel Disease (IBD)

  • DSS-Induced Colitis Model:

    • Lxn⁻/⁻ mice show accelerated weight loss, severe mucosal damage, and elevated pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) .

    • Histological Severity: Complete crypt structure loss in Lxn⁻/⁻ colons by day 7 of DSS treatment vs. partial loss in WT .

    • Therapeutic Resistance: Retinoic acid (RA) fails to ameliorate colitis in Lxn⁻/⁻ mice, confirming LXN’s role in RA-mediated anti-inflammatory pathways .

Cancer and Immune Evasion

  • Subcutaneous Tumor Growth:

    • Lxn⁻/⁻ mice exhibit faster tumor progression (colon and lung cancer models) with increased M2 macrophages (F4/80⁺CD11b⁺CD206⁺) and reduced CD8⁺/CD4⁺ T cells .

    • Bone Marrow Transplantation: Mice receiving Lxn⁻/⁻ hematopoietic cells develop heavier tumor loads and higher PD-L2⁺ macrophage infiltration .

NF-κB Pathway Modulation

  • Overexpression of LXN suppresses TNF-α-induced NF-κB activation, reducing IL-1β, IL-6, and ICAM1 expression in human intestinal epithelial cells (HIECs) .

  • LXN binds HECTD1 and IκBα, inhibiting ubiquitination and degradation of IκBα, thereby blocking NF-κB nuclear translocation .

JAK1/STAT3/PD-L2 Axis in Macrophages

  • Lxn⁻/⁻ macrophages exhibit hyperactivated STAT3, upregulating PD-L2 expression and suppressing T cell cytotoxicity .

  • Rescue Experiments: Adoptive transfer of WT macrophages restores T cell function in Lxn⁻/⁻ mice .

Disease Associations

ConditionLXN RoleReference
ColitisSuppresses NF-κB/STAT3 pathways
Hematologic CancersRegulates HSC quiescence
Solid TumorsInhibits M2 polarization

Research Applications and Limitations

  • Strengths:

    • Recombinant LXN enables in vitro study of carboxypeptidase inhibition .

    • Lxn⁻/⁻ models clarify immune-metabolic cross-talk in colitis and cancer .

  • Limitations:

    • Most studies focus on female mice; sex-specific effects remain unexplored .

    • RA-LXN dependency in colitis therapy requires validation in human trials .

Product Specs

Introduction
Latexin, also known as LXN, belongs to the protease inhibitor I47 (latexin) family. It acts as a potent, hardly reversible, and non-competitive inhibitor of carboxypeptidases CPA1, CPA2, and CPA4. Additionally, LXN is involved in inflammatory processes. Notably, LXN has been associated with conditions like endocervicitis and listeriosis.
Description
Recombinant LXN Mouse, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 245 amino acids (1-222 a.a). It has a molecular weight of 27.9 kDa. The protein consists of LXN fused to a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
LXN protein solution at a concentration of 1 mg/ml in Phosphate Buffered Saline (pH 7.4) containing 30% glycerol and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), store the vial at 4°C. For extended storage, freeze the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms
Latexin, Endogenous carboxypeptidase inhibitor, ECI, Tissue carboxypeptidase inhibitor, TCI.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMEIPPTH YAASRAASVA ENCINYQQGT PHKLFLVQTV QQASKEDIPG RGHKYHLKFS VEEIIQKQVT VNCTAEVLYP QMGQGSAPEV NFTFEGEIGK NPDEEDNTFY QSLMSLKRPL EAQDIPDNFG NVSPQMKPVQ HLAWVACGYV MWQNSTEDTW YKMLKIQTVK QVQRNDDFIE LDYTILLHDI ASQEIIPWQM QVLWHPQYGT KVKHNSRLPK EGQAE.

Q&A

What is the LXN gene and what are its primary functions in hematopoiesis?

Latexin (Lxn) is a negative stem cell regulatory gene identified based on genetic diversity. It plays a crucial role in regulating hematopoietic stem cell (HSC) function and maintaining homeostatic hematopoiesis. Research using Lxn knockout mouse models demonstrates that Lxn acts as a negative regulator of hematopoiesis, with its inactivation leading to expansion of the entire hematopoietic hierarchy. Mechanistically, Lxn affects gene expression related to cell-matrix and cell-cell interactions, with Thrombospondin 1 (Thbs1) identified as a potential downstream target .

How are LXN knockout mice generated and characterized?

LXN knockout (Lxn−/−) mice are typically generated on the C57BL/6 background. Since the Lxn gene lies within the mitochondrial elongation factor G (Gfm1) gene, careful targeting is required to avoid affecting Gfm1 expression. Specifically, only exons 2-4 of the Lxn gene are targeted for deletion to minimize any potential effect on Gfm1. Complete deletion of the LXN protein should be confirmed by Western blot analysis of various tissues including bone marrow, spleen, liver, and brain, while ensuring that GFM1 protein expression remains unaffected .

What are the key phenotypic characteristics of LXN knockout mice?

LXN knockout mice exhibit several distinctive hematopoietic phenotypes:

ParameterWild-typeLxn−/−Change
Peripheral blood counts (WBC, neutrophils, etc.)BaselineIncreasedSignificant
Bone marrow cellularityBaselineIncreasedSignificant
HSC/HPC populationsBaselineExpanded~56% increase in LSK cells
Clonogenic potential (CAFC day 35)BaselineEnhanced3-fold higher
Competitive repopulationBaselineEnhancedImproved long-term reconstitution

Additionally, Lxn−/− mice show slight but significant increases in percentages of macrophages, granulocytes (Mac-1/Gr-1+), and B lymphocytes (B220+) .

How should researchers design experiments to study long-term effects of LXN inactivation?

For studying long-term effects of LXN inactivation, researchers should implement a comprehensive experimental design strategy:

  • Establish age-matched cohorts of Lxn−/− and wild-type mice for longitudinal studies (up to 28 months)

  • Regularly monitor complete blood counts and bone marrow composition

  • Implement competitive repopulation assays with equal numbers of Lxn−/− and wild-type LSK cells

  • Analyze donor-derived cells at multiple timepoints (4, 8, 12, and 16 weeks)

  • Perform secondary transplantation to evaluate self-renewal capacity

  • Subject separate cohorts to hematopoietic stress conditions (e.g., 5-FU treatment)

All experimental designs should follow the 3Rs principles (Replacement, Refinement, Reduction) while ensuring sufficient statistical power through adequate sample sizes . Remember that unlike chemical reagents, mice are biological entities that show phenotypic variability even within inbred strains .

What molecular pathways are altered in LXN-deficient hematopoietic stem cells?

Gene expression profiling of Lxn-null HSCs reveals several altered pathways:

  • Cell-matrix interaction pathways: Gene set enrichment analysis (GSEA) shows significant alterations in cell-matrix and cell-cell interactions

  • Thrombospondin 1 (Thbs1) pathway: Thbs1 is dramatically downregulated in Lxn-null HSCs, and enforced expression of Thbs1 can restore the Lxn inactivation-mediated HSC phenotypes

  • Survival pathways: Loss of Lxn enhances HSC survival in a cell-intrinsic manner

Among 3,561 differentially expressed genes in Lxn−/− HSCs, approximately one-third (1,235) are upregulated while two-thirds (2,326) are downregulated .

How can researchers differentiate between cell-intrinsic and cell-extrinsic effects of LXN inactivation?

Distinguishing between cell-intrinsic and cell-extrinsic effects requires specific methodological approaches:

  • Competitive transplantation assays: Transplanting equal numbers of Lxn−/− and wild-type HSCs into the same recipient creates a controlled environment where both cell types are exposed to identical extrinsic factors. The observed 3-fold higher donor-derived LSK cells from Lxn−/− mice provides strong evidence for cell-intrinsic mechanisms .

  • In vitro culture systems: Using purified Lxn−/− and wild-type HSCs in standardized culture conditions helps assess intrinsic differences in clonogenic potential through CAFC and CFC assays .

  • Reciprocal transplantation experiments:

    • Wild-type HSCs → Lxn−/− recipients

    • Lxn−/− HSCs → Wild-type recipients

This approach helps distinguish the contribution of the bone marrow niche (cell-extrinsic) from the intrinsic properties of HSCs.

What are the implications of LXN research for understanding hematological malignancies?

The relationship between LXN and hematological malignancies presents an interesting paradox:

  • Tumor suppressor potential: Lxn is downregulated in leukemia, lymphoma, and several other cancers (Li et al., 2011; Liu et al., 2012; Mitsunaga et al., 2012; Abd Elmageed et al., 2013; Muthusamy et al., 2013; Ni et al., 2014) .

  • HSC expansion without malignant transformation: Despite expanded HSC populations, aged Lxn−/− mice (28 months) do not spontaneously develop hematological malignancies. They show no significant differences in complete or differentiated blood cell counts compared to age-matched wild-type mice, with only slight but significant increases in myeloid lineage and decreases in B lymphocytes .

  • Therapeutic potential: Understanding how Lxn regulates HSC expansion without malignant transformation could inform approaches for expanding HSCs ex vivo for transplantation, potentially leading to "safe and effective approaches to manipulate HSCs for clinical benefit" .

How do aging and stress conditions affect LXN knockout phenotypes?

The effects of aging and stress on LXN knockout phenotypes reveal important insights:

  • Aging effects (28-month-old mice):

    • No significant difference in complete or differentiated blood cell counts compared to age-matched wild-type mice

    • Nearly 2-fold more LT-HSCs in both frequency and absolute numbers

    • No significant differences in other HSC sub-populations

    • No apparent pathological changes in blood, bone marrow, spleen, and liver

  • Stress response:

    • Lxn−/− mice show "much faster recovery in hematopoietic stem/progenitor cells from 5-FU-induced stress"

    • Enhanced survival and long-term engraftment of hematopoietic stem cells

    • Improved resistance to hematopoietic stress

This differential response indicates that Lxn plays a context-dependent role in hematopoiesis, with particularly important functions under stress conditions.

What controls and experimental conditions are essential when working with LXN knockout models?

When designing experiments with LXN knockout mice, several controls and experimental conditions are essential:

  • Genetic background controls:

    • Use age-matched, sex-matched wild-type mice of the same genetic background (C57BL/6)

    • Consider potential genetic drift by periodically refreshing breeding colonies

    • Use littermate controls whenever possible to minimize environmental variations

  • Environmental standardization:

    • Maintain consistent housing conditions (temperature, humidity, light cycles)

    • Control for microbiome differences through standardized diet and housing

    • Document any environmental changes that might affect experimental outcomes

  • Experimental consistency:

    • Implement blinded analysis protocols to prevent bias

    • Use consistent tissue collection and processing methods

    • Include appropriate positive and negative controls in all functional assays

    • Consider sex as a biological variable in all analyses

What specific assays are recommended for functional characterization of LXN knockout HSCs?

For comprehensive functional characterization of LXN knockout HSCs, researchers should employ multiple complementary assays:

  • In vitro functional assays:

    • Cobblestone area-forming cell (CAFC) assay: Measures primitive HSCs (CAFC day 35)

    • Colony-forming cell (CFC) assay: Assesses HPC clonogenic potential

  • In vivo functional assays:

    • Competitive repopulation assay: Equal numbers of Lxn−/− and wild-type LSK cells transplanted into lethally irradiated recipients

    • Analysis of donor-derived peripheral blood cells at 4, 8, 12, and 16 weeks post-transplantation

    • Assessment of multilineage reconstitution (myeloid, B-cell, and T-cell lineages)

    • Secondary transplantation to evaluate self-renewal capacity

  • Molecular characterization:

    • Gene expression profiling through RNA-sequencing

    • Validation of key findings through qRT-PCR and protein analysis

    • Functional validation through genetic manipulation of key targets (e.g., Thbs1)

How should researchers address potential confounding factors in LXN mouse studies?

Mouse studies require careful control of confounding factors for reproducible results:

  • Mouse strain selection:

    • Know your mouse strain characteristics thoroughly

    • Consider strain-specific baseline hematopoietic parameters

    • Remember that mouse strains can be "as variable as dog breeds"

  • Experimental design principles:

    • Follow the 3Rs framework (Replacement, Refinement, Reduction)

    • Power analysis to determine appropriate sample sizes

    • Account for developmental timing and age effects

  • Environmental factors:

    • Mice are "sensitive to small environmental insults"

    • Standardize housing, handling, and experimental procedures

    • Document and report all relevant environmental conditions

  • Reproducibility considerations:

    • "While your goal as a biomedical researcher may be to test your hypothesis in a living organism, a mouse's goal is simple: be a mouse"

    • Replicate critical findings independently

    • Avoid the "just one more replicate" trap that can lead to irreproducible results

What are the potential clinical applications of LXN research findings?

The findings from LXN mouse models suggest several potential clinical applications:

  • HSC expansion for transplantation:

    • Understanding how Lxn regulates HSC expansion could inform approaches for ex vivo expansion

    • This might lead to "development of safe and effective approaches to manipulate HSCs for clinical benefit"

  • Stress resistance applications:

    • The enhanced recovery of Lxn−/− HSCs from 5-FU-induced stress suggests potential protective strategies for patients undergoing chemotherapy

    • Temporary inhibition of Lxn function might protect HSCs during cytotoxic treatments

  • Biomarker development:

    • Altered Lxn expression in various cancers suggests potential utility as a diagnostic or prognostic biomarker

    • Monitoring Lxn and downstream targets like Thbs1 could provide valuable clinical information

How can LXN mouse models inform our understanding of leukemogenesis?

While Lxn−/− mice do not spontaneously develop leukemia, they provide valuable insights into hematopoietic regulation relevant to leukemogenesis:

  • Expanded but regulated HSC pool:

    • Lxn−/− mice maintain an expanded but non-malignant HSC population

    • This allows study of the boundary between physiological expansion and pathological transformation

  • Aging effects:

    • Aged Lxn−/− mice (28 months) maintain nearly 2-fold more LT-HSCs without developing malignancy

    • This suggests additional genetic or environmental factors are required for leukemic transformation

  • Gene expression alterations:

    • The altered gene expression profile in Lxn−/− HSCs may reveal pathways that, when further dysregulated, contribute to leukemogenesis

    • The relationship between Lxn and Thbs1 provides a specific pathway for investigation

Understanding the molecular mechanisms that prevent malignant transformation in Lxn−/− mice despite HSC expansion could provide critical insights into leukemia development and potential therapeutic targets.

What are the most promising areas for future research using LXN mouse models?

Several high-priority research directions emerge from current LXN findings:

  • Molecular mechanism studies:

    • Further characterization of the relationship between Lxn and Thbs1

    • Identification of additional downstream targets and interacting partners

    • Detailed mapping of signaling pathways regulated by Lxn

  • Stress response investigations:

    • Systematic analysis of how Lxn modulates responses to different types of hematopoietic stress

    • Investigation of potential protective effects of Lxn inhibition during chemotherapy or radiation exposure

  • Conditional knockout models:

    • Development of tissue-specific and inducible Lxn knockout models to precisely control timing and location of Lxn inactivation

    • This would help distinguish developmental from adult homeostatic functions

  • Translational research:

    • Testing whether temporary pharmacological inhibition of Lxn function could enhance HSC expansion or stress resistance

    • Exploration of Lxn as a potential target in leukemia treatment based on its downregulation in various cancers

What experimental design improvements could enhance reproducibility in LXN mouse research?

To enhance reproducibility in LXN mouse research, consider implementing these experimental design improvements:

  • Standardized reporting:

    • Detailed reporting of mouse strain, age, sex, housing conditions, and experimental procedures

    • Documentation of genetic background and breeding strategy

    • Transparency about sample sizes and statistical approaches

  • Multi-laboratory validation:

    • Collaborative studies across multiple laboratories to confirm key findings

    • Standardized protocols for phenotypic characterization

    • Pre-registration of study designs and analysis plans

  • Comprehensive phenotyping:

    • Integration of multiple complementary assays (in vitro, in vivo, molecular)

    • Assessment of both basal and stress conditions

    • Longitudinal studies across different age points

  • Genetic validation:

    • Genetic rescue experiments (e.g., Thbs1 restoration)

    • Use of alternative genetic approaches to validate key findings

    • Exploration of strain-dependent effects

Product Science Overview

Discovery and Function

Latexin was initially identified as a molecular marker for the regional specification of the neocortex in the developing brain of rats . It is highly expressed in various tissues, including the heart, prostate, ovary, kidney, pancreas, and colon . The protein plays a crucial role in inhibiting carboxypeptidase A1, an enzyme involved in protein degradation .

Structure and Expression

The recombinant mouse latexin protein is typically expressed in E. coli and consists of 228 amino acids with a calculated molecular mass of approximately 26.3 kDa . When analyzed under reducing conditions using SDS-PAGE, the apparent molecular mass is around 32 kDa . The protein is often tagged with a polyhistidine tag at the N-terminus to facilitate purification .

Biochemical Properties

Latexin’s activity is measured by its ability to inhibit carboxypeptidase A1 cleavage of the colorimetric peptide substrate Ac-Phe-Thiaphe-OH in the presence of 5,5’-dithiobis (2-nitrobenzoic acid) (DTNB) . The IC50 value, which indicates the concentration required to inhibit 50% of the enzyme activity, is less than 2.0 nM .

Applications and Storage

Recombinant mouse latexin is used in various biochemical and physiological studies to understand its role in enzyme inhibition and its potential therapeutic applications . The protein is typically lyophilized and stored under sterile conditions at -20°C to -80°C to maintain stability . It is recommended to avoid repeated freeze-thaw cycles to preserve its activity .

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.