Latexin is a 222-amino-acid protein with a unique α/β-hydrolase fold, organized into two cystatin-like subdomains:
N-terminal subdomain (NTS): Met1–Glu92, featuring an α-helix wrapped by a curved β-sheet.
C-terminal subdomain (CTS): Lys114–Lys217, structurally homologous to NTS.
These subdomains bind to the active site of carboxypeptidases (e.g., CPA4, CPA1, CPB) through a large interface, achieving nanomolar inhibition constants (Table 1) .
MCP Target | Kᵢ (nM) |
---|---|
hCPA1 | 1.6 |
hCPA4 | 3.0 |
Thrombin-activatable fibrinolysis inhibitor | 1.8 |
hCPB | 1.1 |
Latexin does not inhibit N/E subfamily MCPs or invertebrate carboxypeptidases due to divergent loop structures near the active site .
Latexin is downregulated in hematological malignancies (e.g., leukemia, lymphoma) and solid tumors (e.g., gastric, pancreatic, prostate cancers). Key findings include:
Promoter hypermethylation silences LXN in leukemia/lymphoma cells, and 5-aza-2′-deoxycytidine treatment restores its expression .
Overexpression in A20 lymphoma cells reduces tumor growth by 60% in vivo via apoptosis induction (↓Bcl-2, ↓Pim-2) .
In pancreatic ductal adenocarcinoma, low latexin correlates with larger tumor size (p=0.002) and poor differentiation (p<0.001) .
Feature | Latexin-Low (%) | Latexin-High (%) | p-value |
---|---|---|---|
Tumor size >3 cm | 18 | 6 | 0.002 |
High histologic grade | 17 | 2 | <0.001 |
Atherosclerosis: LXN knockout in ApoE−/− mice reduces plaque formation by 40% via JAK1/STAT3 pathway activation, enhancing cholesterol efflux and reducing foam cell formation .
Neointimal hyperplasia: Smooth muscle cell (SMC)-specific LXN deletion inhibits PDGFR-driven proliferation, while myeloid-specific deletion attenuates macrophage migration (↓ERK phosphorylation) .
Latexin is upregulated in pro-inflammatory M1 macrophages and regulates polarization. Its deficiency promotes anti-inflammatory cytokine secretion (e.g., IL-10) and reduces oxidative stress .
Epigenetic control: Hypermethylation of the LXN promoter suppresses transcription in cancer cells .
Genetic variation: The SNP rs31528793 (G/C) in the LXN promoter modulates expression, with the G allele linked to higher levels and reduced hematopoietic stem cell numbers .
Transcriptional suppression: HMGB2 binds the LXN promoter, repressing its expression in hematopoietic stem cells .
Recombinant human latexin (27.16 kDa) is produced in E. coli with >95% purity. Key applications include:
Therapeutic development: Adeno-associated virus (AAV)-delivered LXN shRNA reduces atherosclerosis in mice .
Cancer: Restoring LXN expression via demethylating agents or gene therapy could suppress tumor growth.
Cardiovascular disease: Targeting LXN in SMCs or macrophages may prevent restenosis or atherosclerosis.
Human Latexin (LXN), also known as tissue carboxypeptidase inhibitor, is the only mammalian carboxypeptidase inhibitor identified to date. This 222 amino acid protein (spanning from Glu2 to Glu222) functions primarily in inflammation and innate immunity pathways . Latexin was initially identified as a marker of neurons in the lateral neocortex of the developing brain (hence the name "latexin") . Since its discovery, research has revealed that Latexin is highly expressed in mast cells and macrophages, where it plays critical regulatory roles in inflammatory processes . Its ability to inhibit carboxypeptidases from the pancreas (CPA1 and CPA2) and mast cells (CPA3) further highlights its importance in modulating proteolytic activities during inflammation .
Unlike carboxypeptidase inhibitors derived from plants and parasites, Human Latexin possesses distinct structural features that suggest a unique mechanism of action. At 222 amino acids, Human Latexin is significantly larger than its non-mammalian counterparts . A key distinguishing characteristic is its lack of conserved C-terminal residues that typically interact with target carboxypeptidases in a substrate-like manner in other inhibitors . This structural difference indicates that Human Latexin employs an alternative mechanism for carboxypeptidase inhibition, potentially involving different binding domains or allosteric regulation. Understanding these structural peculiarities is essential for researchers investigating the molecular interactions of Latexin with its target enzymes.
Human Latexin exhibits a distinct tissue distribution pattern that provides insights into its physiological roles. While initially identified in neurons of the lateral neocortex, subsequent research has demonstrated that Latexin is highly expressed in immune cells, particularly mast cells and macrophages . Recent studies have also revealed enriched Latexin expression in human and murine atherosclerotic lesions, predominantly localized to macrophages within these pathological sites . Additionally, Latexin expression is induced during acute pancreatitis and lung inflammatory disease, suggesting a responsive role during tissue inflammation . Researchers should note this varied distribution pattern when designing tissue-specific studies and interpreting expression data across different physiological and pathological conditions.
Researchers investigating Human Latexin expression should consider multiple complementary techniques to ensure robust data. Immunofluorescence and immunohistochemistry have been successfully employed to examine LXN expression in human and mouse atherosclerotic lesions . For protein detection, commercially available antibodies such as the Human Latexin Antibody (AF3246) can be utilized in various applications . When analyzing expression at the genetic level, standard molecular biology techniques including RT-PCR and qPCR are appropriate. For visualization of cellular localization, immunofluorescence with appropriate co-staining (e.g., with macrophage markers) can provide valuable insights into the cell types expressing Latexin in various tissues and pathological specimens .
The generation of Latexin knockout models has proven valuable for investigating LXN's functional roles. Researchers have successfully created both LXN knockout and LXN/ApoE double-knockout mice to evaluate Latexin functions in atherosclerosis . When developing such models, researchers should verify knockout efficiency at both genomic (PCR-based genotyping) and protein levels (western blotting, immunohistochemistry). Bone marrow transplantation (BMT) experiments represent an effective approach for investigating cell-specific effects, as demonstrated by studies showing that deletion of LXN in bone marrow protects ApoE mice against atherosclerosis . For therapeutic proof-of-concept studies, adeno-associated virus harboring LXN-depleting shRNA has been successfully employed to attenuate disease phenotypes in atherosclerotic mice .
Several commercial antibodies and reagents are available for Human Latexin research. The Human Latexin Antibody (AF3246) is a polyclonal antibody derived from E. coli-expressed recombinant human Latexin (Glu2-Glu222) . This antibody can be stored for 12 months at -20 to -70°C as supplied, 1 month at 2 to 8°C under sterile conditions after reconstitution, or 6 months at -20 to -70°C under sterile conditions after reconstitution . For proteome-wide studies, researchers can utilize the Proteome Profiler Human Protease/Protease Inhibitor Array or the Proteome Profiler Human Protease Inhibitor Array Kit, which contain membranes spotted with antibodies against multiple protease inhibitors, including Latexin . When selecting reagents, researchers should validate antibody specificity using appropriate positive and negative controls, particularly when studying tissues with potential cross-reactivity.
Human Latexin plays a significant role in atherosclerosis development through its effects on macrophage function and foam cell formation. Research has demonstrated that LXN is enriched in human and murine atherosclerotic lesions, primarily localized to macrophages . Mechanistically, LXN targets and inhibits JAK1 in macrophages, affecting downstream signaling pathways critical for lipid metabolism and inflammatory responses . In atherosclerosis models, LXN deficiency markedly improves disease outcomes by inhibiting foam cell formation . This improvement occurs through enhanced activity of the JAK1/STAT3/ABC transporter pathway, which promotes an anti-inflammatory and anti-oxidant phenotype while facilitating cholesterol efflux from macrophages . These findings suggest that Latexin functions as a pro-atherogenic factor by inhibiting protective cellular mechanisms that would otherwise limit foam cell formation and inflammation in the arterial wall.
Human Latexin exhibits important relationships with multiple inflammatory conditions beyond atherosclerosis. The protein is induced during acute pancreatitis and lung inflammatory disease, suggesting responsive expression during tissue inflammation . As the only identified mammalian carboxypeptidase inhibitor, Latexin regulates specific pancreatic and mast cell carboxypeptidases that contribute to inflammatory processes . Latexin's high expression in macrophages and mast cells—key mediators of innate immunity—further supports its role in inflammatory regulation . Research indicates that Latexin deficiency enhances an anti-inflammatory phenotype through JAK1/STAT3 pathway stimulation . This growing body of evidence positions Latexin as an important regulatory protein in inflammatory diseases, with potential relevance to conditions characterized by chronic inflammation and macrophage dysfunction.
Studies have documented significant changes in Latexin expression during disease progression, particularly in inflammatory and metabolic disorders. In atherosclerosis, Latexin is enriched in both human and murine lesions as the disease advances . The protein shows dynamic expression patterns during acute inflammatory conditions, with induction observed during acute pancreatitis and lung inflammatory disease . Researchers investigating expression changes should employ temporal analysis methods to capture these dynamics. Importantly, comparative studies between healthy and diseased tissues have demonstrated that expression alterations are often cell-type specific, with macrophages showing particularly notable changes in Latexin levels during pathological states . These expression patterns provide valuable biomarkers for disease progression and potential therapeutic intervention points.
Human Latexin influences several critical signaling pathways, most notably the JAK1/STAT3 pathway in macrophages. Research has established that LXN targets and inhibits JAK1, directly impacting downstream STAT3 signaling and ABC transporter expression . This mechanism has significant implications for cellular function, as the JAK1/STAT3/ABC transporter pathway regulates inflammatory responses, antioxidant status, and cholesterol efflux . To study these pathways effectively, researchers should employ phosphorylation-specific antibodies to detect active JAK1 and STAT3, use pharmacological inhibitors to confirm pathway involvement, and measure downstream gene expression through quantitative PCR or RNA sequencing. Protein-protein interaction studies using co-immunoprecipitation can help elucidate the direct binding between Latexin and JAK1. Additionally, chromatin immunoprecipitation (ChIP) assays can reveal STAT3 binding to target gene promoters, providing insights into the transcriptional consequences of Latexin-mediated pathway modulation.
Latexin deficiency significantly reduces foam cell formation through multiple molecular mechanisms. At the pathway level, LXN deficiency stimulates the JAK1/STAT3/ABC transporter pathway, enhancing the expression and activity of cholesterol efflux transporters . This increased cholesterol efflux capacity prevents excessive lipid accumulation within macrophages that would otherwise lead to foam cell formation . Concurrently, Latexin deficiency promotes an anti-inflammatory and anti-oxidant phenotype in macrophages, reducing inflammatory responses that exacerbate lipid uptake and retention . These molecular changes collectively minimize foam cell formation and subsequent atherosclerotic plaque development . For comprehensive analysis of these effects, researchers should employ cholesterol efflux assays, quantify ABC transporter expression and localization, measure oxidative stress markers, and assess inflammatory cytokine production in both wild-type and Latexin-deficient macrophages under lipid-loading conditions.
Targeting Human Latexin offers promising therapeutic applications, particularly for atherosclerosis and inflammatory conditions. Gene therapy approaches using adeno-associated virus harboring LXN-depleting shRNA have demonstrated efficacy in attenuating atherosclerosis in mouse models . These findings position Latexin as a potential new anti-atherosclerosis therapeutic target . Beyond cardiovascular applications, Latexin's role in inflammation suggests potential therapeutic relevance for inflammatory diseases such as pancreatitis and lung inflammatory disorders . Therapeutic strategies could include small molecule inhibitors of Latexin-JAK1 interaction, neutralizing antibodies, or RNA interference approaches to modulate Latexin expression or function. For drug development purposes, researchers should establish high-throughput screening platforms to identify compounds that interfere with Latexin-carboxypeptidase interactions or Latexin-JAK1 binding. Efficacy of Latexin-targeting therapies should be evaluated across multiple disease models to fully appreciate their potential clinical applications.
Resolving contradictory findings in Human Latexin research requires systematic approaches to identify sources of discrepancy. First, researchers should examine methodological differences, as variations in experimental models (cell lines, animal strains, knockout strategies), detection methods (antibody specificities, detection thresholds), and experimental conditions (timing, disease models) can yield apparently contradictory results . A comprehensive metadata analysis that accounts for these variables can help reconcile disparate findings. When analyzing contradictions in multi-omics datasets, researchers should apply embedding-based paragraph clustering methodologies similar to those used for detecting inconsistencies in complex reports . This approach groups related information to identify contextual factors that might explain apparent contradictions. Additionally, collaboration across research groups using standardized protocols can help validate findings and resolve contradictions, particularly for studies investigating Latexin's tissue-specific functions or its varied roles in different disease contexts.
When analyzing Human Latexin expression data, researchers should select statistical approaches that address the specific characteristics of their experimental design and data distribution. For comparative studies of Latexin expression between different tissues or disease states, parametric tests (t-tests, ANOVA) are appropriate when data follow normal distributions, while non-parametric alternatives (Mann-Whitney U test, Kruskal-Wallis test) should be employed for non-normally distributed data. For correlation analyses between Latexin expression and clinical parameters, Pearson's correlation is suitable for linear relationships with normally distributed variables, while Spearman's rank correlation accommodates non-linear relationships. Time-course studies of Latexin expression during disease progression benefit from repeated measures ANOVA or mixed-effects models. When analyzing complex datasets with multiple variables, multivariate analyses such as principal component analysis or hierarchical clustering can reveal patterns in Latexin expression across different experimental conditions. Statistical significance should be adjusted for multiple comparisons using methods such as Bonferroni correction or false discovery rate control to minimize Type I errors.
Emerging technologies offer exciting opportunities for advancing Human Latexin research. CRISPR-Cas9 gene editing allows for precise modification of the LXN gene, enabling the creation of specific mutations to study structure-function relationships. Single-cell RNA sequencing can reveal cell-type-specific expression patterns of Latexin across tissues and disease states with unprecedented resolution. For protein interaction studies, proximity labeling methods such as BioID or APEX provide in situ identification of Latexin's protein interactome. Advanced imaging techniques, including super-resolution microscopy and intravital imaging, allow visualization of Latexin's subcellular localization and dynamics in living systems. Proteomics approaches combined with mass spectrometry can identify post-translational modifications of Latexin that may regulate its function. For therapeutic development, high-throughput screening platforms using FRET-based assays can identify small molecules that modulate Latexin-JAK1 interaction. These technologies, implemented individually or in combination, promise to significantly advance our understanding of Latexin's complex roles in normal physiology and disease pathogenesis.
Multi-omics approaches offer powerful frameworks for comprehensive investigation of Human Latexin biology across multiple levels of biological organization. Integration of genomics, transcriptomics, proteomics, and metabolomics data can provide holistic views of how Latexin functions within cellular networks. Genomic analyses can identify regulatory elements controlling Latexin expression and genetic variants associated with altered function. Transcriptomic profiling before and after Latexin manipulation reveals downstream gene expression changes that define its functional impact. Proteomics approaches identify Latexin's interacting partners and post-translational modifications that regulate its activity. Metabolomics can detect alterations in cellular metabolism resulting from Latexin's effects on signaling pathways. When combined with computational biology approaches, these multi-omics datasets can be integrated to construct predictive models of Latexin's role in health and disease. Researchers should employ standardized data collection protocols and robust computational pipelines for data integration to maximize the value of multi-omics investigations in Latexin research.
Translational research approaches for Human Latexin-targeted therapies are gaining momentum based on recent discoveries about its role in disease. Preclinical development of small molecule inhibitors targeting the Latexin-JAK1 interaction represents a promising approach for atherosclerosis treatment . RNA interference strategies using siRNA or shRNA have demonstrated efficacy in animal models, with adeno-associated virus delivery systems showing particular promise for in vivo applications . For clinical translation, biomarker development is essential—measuring Latexin levels in plasma or within specific immune cell populations may help identify patients most likely to benefit from Latexin-targeted therapies. Patient stratification approaches based on Latexin expression or genetic variants could enhance clinical trial design. Additionally, repurposing existing JAK inhibitors might provide indirect means of counteracting Latexin's effects on downstream pathways. As translational efforts advance, researchers should establish reliable pharmacodynamic markers to monitor therapeutic responses and develop combination strategies that target multiple aspects of Latexin-mediated pathology for enhanced efficacy in complex diseases like atherosclerosis.
When working with recombinant Human Latexin, researchers should follow specific guidelines to maintain protein integrity and activity. Recombinant Human Latexin derived from E. coli expression systems (Glu2-Glu222) has been successfully used in previous studies . For storage, use a manual defrost freezer and avoid repeated freeze-thaw cycles to preserve protein structure and function . Unopened products can be stored for 12 months from date of receipt at -20 to -70°C . After reconstitution, the protein can be stored for 1 month at 2 to 8°C under sterile conditions or for 6 months at -20 to -70°C under sterile conditions . When preparing working solutions, use appropriate buffers (typically phosphate-buffered saline) with protease inhibitors to prevent degradation. For quantification, standard protein assays such as Bradford or BCA are suitable. Researchers should validate protein activity using functional assays that measure carboxypeptidase inhibition before proceeding with experiments to ensure the prepared Latexin retains its biological activity.
Effective documentation and reporting of Human Latexin research findings are essential for reproducibility and advancement of the field. Researchers should employ electronic laboratory notebooks (ELNs) for comprehensive documentation of experimental procedures, results, and analyses . When describing experiments in publications, provide detailed methodological information including exact antibody catalog numbers, dilutions, incubation times, and validation procedures . For reporting expression data, clearly specify the detection method, quantification approach, and normalization strategy. When documenting contradictory findings, explicitly address potential sources of discrepancy and include negative results that may provide valuable context . Statistical analyses should be thoroughly described with appropriate measures of central tendency, dispersion, sample sizes, and exact p-values. For complex datasets, follow standardized reporting guidelines for various experimental approaches (e.g., ARRIVE for animal studies). Use LaTeX or similar document preparation systems for preparing manuscripts to ensure proper formatting of complex scientific content, particularly for mathematical expressions and statistical analyses .
Recombinant human latexin is typically expressed in E. coli with an N-terminal His tag. The protein consists of amino acids Glu2-Glu222 . The molecular weight of latexin is predicted to be around 27 kDa, but it migrates to 30-35 kDa based on Bis-Tris PAGE results .
Latexin is expressed in small nociceptive neurons in sensory ganglia and in a subset of neurons in the telencephalon . It was initially identified as a marker of neurons in the lateral neocortex of the developing brain .
Recent studies have identified the latexin (LXN) gene as a potential tumor suppressor in several types of solid tumors and lymphoma. The expression of LXN is often absent or downregulated in leukemic cells . This suggests that latexin could play a significant role in cancer biology and might be a target for therapeutic interventions.
Recombinant human latexin is typically lyophilized from a 0.22 µm filtered solution in PBS (pH 7.4) with 8% trehalose added as a protectant before lyophilization . The reconstituted protein is stable at -80°C for up to 12 months and at 4°C for up to one week. It is important to use a manual defrost freezer and avoid repeated freeze-thaw cycles to maintain the protein’s stability .