ANGPTL7 modulates lipid metabolism by regulating lipoprotein lipase (LPL) activity and lipid uptake in adipose tissue and skeletal muscle. It also influences adipogenesis, contributing to obesity-related metabolic complications .
ANGPTL7 stimulates the expansion and repopulation capacity of human hematopoietic stem and progenitor cells (HSPCs). This effect is mediated through activation of Wnt signaling pathways and upregulation of CXCR4 and HOXB4, enhancing HSPC engraftment in xenograft models .
ANGPTL7 is critical for maintaining IOP homeostasis. Genetic studies identified rare coding variants (e.g., Gln175His, Arg177*) in ANGPTL7 associated with reduced IOP and glaucoma protection. These variants likely disrupt ANGPTL7 function, leading to decreased extracellular matrix deposition in the trabecular meshwork .
In murine models of bronchopulmonary dysplasia (BPD), ANGPTL7 restored lung vascular diameter and density, mitigated inflammation, and enhanced VEGF-A levels, suggesting therapeutic potential for preterm lung injuries .
ANGPTL7 inhibition or loss-of-function variants may serve as a therapeutic strategy to lower IOP. In mice, Angptl7 knockout models exhibited reduced IOP, aligning with human genetic associations .
Elevated plasma ANGPTL7 levels correlate with obesity, insulin resistance, and obstructive sleep apnea (OSA). Modulating ANGPTL7 may address metabolic dysregulation linked to these conditions .
Cord blood-derived ANGPTL7 levels are inversely associated with BPD severity. Experimental models show ANGPTL7 administration improves alveolar development and reduces inflammation, positioning it as a candidate for preterm infant therapies .
Variant Type | Effect on IOP | Frequency | Source |
---|---|---|---|
Gln175His (missense) | Reduced IOP (β = –0.21 SD) | 0.7% (MAF) | |
Arg177 (stop-gain)* | Loss of function, lower IOP | 0.03% (MAF) |
In xenograft models, recombinant ANGPTL7 increased human HSPC engraftment efficiency by 2–3 fold compared to controls, driven by Wnt pathway activation .
In OSA patients, circulating ANGPTL7 levels correlate positively with triglycerides (ρ = 0.332) and negatively with HDL cholesterol (ρ = –0.239), highlighting its role in metabolic-inflammatory crosstalk .
angiopoietin like 7, AngX, CDT6, dJ647M16.1, Angiopoietin-like factor; Angiopoietin-like Protein 7, ANGPTL7, angiopoietinrelated protein 7, Cornea-derived transcript 6 protein
HEK293 cells.
QKLSKHKTPA QPQLKAANCC EEVKELKAQV ANLSSLLSEL NKKQERDWVS VVMQVMELES NSKRMESRLT DAESKYSEMN NQIDIMQLQA AQTVTQTSAD AIYDCSSLYQ KNYRISGVYK LPPDDFLGSP ELEVFCDMET SGGGWTIIQR RKSGLVSFYR DWKQYKQGFG SIRGDFWLGN EHIHRLSRQP TRLRVEMEDW EGNLRYAEYS HFVLGNELNS YRLFLGNYTG NVGNDALQYH NNTAFSTKDK DNDNCLDKCA QLRKGGYWYN CCTDSNLNGV YYRLGEHNKH LDGITWYGWH GSTYSLKRVE MKIRPEDFKP LEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK
ANGPTL7 shows tissue-specific expression patterns with notable presence in the trabecular meshwork of the eye, where it plays a role in extracellular matrix formation and organization . The protein is also expressed during hematopoiesis, particularly in cells that support hematopoietic stem and progenitor cell (HSPC) development . In pathological conditions, ANGPTL7 has been found to be over-expressed in several human cancers, particularly under hypoxic conditions . For comprehensive tissue expression analysis, researchers should employ RNA-sequencing methodologies combined with protein detection techniques such as immunohistochemistry or Western blotting across multiple tissue types, with appropriate controls to account for baseline expression levels.
ANGPTL7 has been found to activate several signaling cascades in human cells. In hematopoietic stem and progenitor cells, RNA-sequencing analysis revealed that ANGPTL7 activates the expression of CXCR4, HOXB4, and Wnt downstream targets . The involvement of the Wnt signaling pathway appears particularly significant, as chemical manipulation of this pathway diminished ANGPTL7's effects on human HSPCs in culture . To investigate these pathways, researchers should design experiments incorporating specific pathway inhibitors (e.g., IWP2, CHIR99021 for Wnt signaling) while monitoring downstream gene expression changes through RNA-seq or qPCR. Functional validation using reporter assays for specific pathways would provide more definitive evidence of signaling activation.
Overexpression of ANGPTL7 in primary human trabecular meshwork cells significantly alters the expression of numerous extracellular matrix components, including fibronectin, collagens type I, IV & V, myocilin, versican, and MMP1 . Beyond mere expression changes, ANGPTL7 has been shown to interfere with the fibrillar assembly of fibronectin, suggesting a direct role in matrix organization . Research methodologies should include gene expression analysis of ECM components, immunofluorescence studies of matrix deposition patterns, and functional assays examining matrix stiffness and organization following ANGPTL7 modulation. Proteomics approaches could comprehensively assess the full spectrum of ECM changes induced by ANGPTL7.
While structurally related to other ANGPTL family members, ANGPTL7 has distinct functions. Unlike some ANGPTLs involved primarily in angiogenesis or metabolism, ANGPTL7 demonstrates specialized roles in extracellular matrix formation and hematopoietic stem cell regulation . Furthermore, ANGPTL7 stands out for its significant association with intraocular pressure regulation and glaucoma risk, with rare protein-altering variants conferring protection against glaucoma . Comparative studies examining multiple ANGPTL proteins simultaneously under identical experimental conditions would best elucidate functional differences. This could include parallel overexpression or knockdown experiments across multiple cell types relevant to ANGPTL function.
To investigate ANGPTL7's effects on hematopoietic stem and progenitor cells (HSPCs), researchers should employ a multi-faceted approach:
Ex vivo expansion studies: Culture human CD34+ HSPCs with recombinant ANGPTL7 (typically at 500 ng/mL based on previous studies) in serum-free media supplemented with standard hematopoietic cytokines (SCF, TPO, FLT3L) . Compare cell proliferation, maintenance of stem cell markers, and colony-forming capacity between control and ANGPTL7-treated cultures.
Transplantation assays: Assess HSPC function through xenograft models using immunodeficient mice (e.g., NSI mice) to evaluate repopulation capacity. This approach should include limiting dilution analysis to quantify functional stem cell frequency, with human engraftment defined as ≥1% CD45+ human cells in mouse bone marrow at 8 weeks post-transplant .
Molecular mechanism investigation: Perform RNA-sequencing on ANGPTL7-treated HSPCs to identify activated pathways, with follow-up validation through chemical inhibitors of identified pathways (e.g., Wnt pathway modulators) .
Homing assays: Evaluate HSPC trafficking by comparing the homing efficiency of ANGPTL7-treated cells to bone marrow niches, potentially using CXCR4 blocking antibodies to assess mechanism specificity .
Research into ANGPTL7 variants requires sophisticated genetic and physiological approaches:
Genetic association studies: Large cohort analysis comparing rare protein-altering variants in ANGPTL7 with intraocular pressure measurements and glaucoma diagnoses. UK Biobank and FinnGen data have identified specific variants (rs28991009, p.Gln175His, MAF=0.8% in UK Biobank; rs147660927, p.Arg220Cys, MAF=4.3% in Finland) associated with lower IOP and reduced glaucoma risk .
Functional validation: Express identified variants in trabecular meshwork cell models to assess effects on:
ECM gene expression profiles
Protein secretion and processing
Structural changes to the protein
Downstream signaling pathway activation
Animal models: Generate knock-in mouse models expressing human ANGPTL7 variants to measure IOP and assess for glaucomatous changes in vivo.
Mechanism determination: Compare protein-truncating variants with missense variants to determine if the protective mechanism involves loss of function or altered interaction domains .
ANGPTL7 has been shown to exert pro-angiogenetic effects on human differentiated endothelial cells while not stimulating progenitor endothelial cells . To investigate this differential response:
Parallel assays: Conduct side-by-side experiments with isolated progenitor endothelial cells and mature endothelial cells, assessing:
Receptor profiling: Compare receptor expression profiles between mature and progenitor cells to identify potential differences in ANGPTL7 receptor expression or downstream signaling components.
In vivo validation: Utilize Matrigel sponge assays in mice to assess vascularization responses, with histological examination to differentiate between recruitment of existing vessels versus formation of new vessels from progenitors .
Single-cell analysis: Apply single-cell RNA sequencing to heterogeneous endothelial populations treated with ANGPTL7 to identify cell subtype-specific response patterns.
Given that ANGPTL7 is specifically up-regulated by hypoxia in cancer cells , methodological approaches should include:
Controlled hypoxia systems: Utilize hypoxia chambers with precise O₂ control (typically 1-2% O₂) compared to normoxic conditions (21% O₂), with time-course experiments to determine optimal hypoxic exposure duration.
HIF pathway analysis: Investigate the role of hypoxia-inducible factors (HIFs) in ANGPTL7 regulation through:
ChIP assays to detect HIF binding to the ANGPTL7 promoter
HIF knock-down or inhibition studies to confirm dependency
HIF stabilization under normoxic conditions (using CoCl₂ or HIF-pathway inhibitors) to determine if this mimics hypoxic induction of ANGPTL7
Secretion and exosome studies: Fractionate conditioned media from hypoxic cells to separate exosomal and soluble ANGPTL7, as the protein has been found partially associated with the exosomal fraction . This requires ultracentrifugation protocols and exosome markers for verification.
In vivo hypoxia models: Utilize tumor xenograft models with hypoxic regions (detected via pimonidazole staining) to correlate ANGPTL7 expression with hypoxic zones in tumors.
To investigate ANGPTL7's effects on ECM:
3D matrix assembly assays: Culture cells on fluorescently labeled ECM proteins (particularly fibronectin) to visualize matrix assembly patterns following ANGPTL7 treatment or overexpression .
Atomic force microscopy: Measure changes in ECM stiffness and organization at the nanoscale level following ANGPTL7 modulation.
Quantitative proteomics: Use mass spectrometry-based approaches to comprehensively profile ECM composition changes, including post-translational modifications of matrix proteins.
Time-lapse imaging: Monitor real-time matrix assembly and reorganization in cells expressing fluorescently tagged ECM proteins with or without ANGPTL7 treatment.
Gene expression analysis: Quantify expression changes in key ECM components (fibronectin, collagens, myocilin, versican, and MMP1) through qPCR or RNA-seq following ANGPTL7 treatment .
Since ANGPTL7's effects on HSPCs appear dependent on Wnt signaling , researchers should:
Pathway modulation: Treat ANGPTL7-stimulated HSPCs with:
Reporter assays: Utilize TCF/LEF luciferase reporters to directly measure Wnt pathway activation in HSPCs following ANGPTL7 treatment.
Downstream target analysis: Quantify expression of key Wnt target genes through RNA-seq or targeted qPCR panels, filtering for genes with ≥2-fold expression change and P<0.05 .
β-catenin localization: Track nuclear translocation of β-catenin through immunofluorescence or cellular fractionation studies following ANGPTL7 treatment.
Genetic approaches: Use CRISPR/Cas9 to knock out key Wnt pathway components and test if this abolishes ANGPTL7's effects on HSPCs.
Research into ANGPTL7's oncogenic potential faces several methodological challenges:
Baseline expression variability: ANGPTL7 is marginally expressed under standard growth conditions but significantly upregulated under hypoxia , necessitating careful experimental design with appropriate hypoxic controls.
Dual secretion mechanisms: ANGPTL7 is both freely secreted and associated with exosomes , requiring sophisticated fractionation techniques to distinguish between these pools and their respective functions.
Context-dependent functions: ANGPTL7 may have different roles depending on the cancer type and microenvironment, requiring comparative studies across multiple cancer models.
Angiogenesis quantification: Properly quantifying ANGPTL7's pro-angiogenic effects requires standardized assays including:
Endothelial cell proliferation, migration, and tube formation in vitro
In vivo Matrigel plug assays with quantifiable vascularization metrics
Distinguishing direct effects on endothelial cells from indirect effects via other factors
Therapeutic targeting considerations: Development of ANGPTL7 inhibitors must account for its beneficial effects in other contexts (e.g., glaucoma protection), necessitating tissue-specific targeting approaches.
To study ANGPTL7's role in IOP regulation:
Human genetic correlation: Compare IOP measurements between carriers of different ANGPTL7 variants, as demonstrated in UK Biobank studies where heterozygotes showed -0.53 to -0.67 mmHg lower IOP and homozygotes displayed -3.40 to -2.37 mmHg reductions for the p.Gln175His variant .
Animal models: Develop transgenic mouse models expressing human ANGPTL7 variants or with ANGPTL7 knockout/knockdown to measure:
IOP through tonometry (both Goldman-correlated and corneal-compensated measurements)
Outflow facility through perfusion studies
Trabecular meshwork ultrastructure through electron microscopy
Ex vivo perfusion models: Utilize anterior segment perfusion models to directly measure the effects of recombinant ANGPTL7 or its variants on aqueous humor outflow.
Trabecular meshwork cell models: Study primary human trabecular meshwork cells with ANGPTL7 overexpression or knockdown to assess:
Given ANGPTL7's partial association with exosomes in cancer cells , researchers should:
Exosome isolation: Use differential ultracentrifugation, size exclusion chromatography, or commercial exosome isolation kits to separate exosomes from soluble proteins in conditioned media.
Verification of exosome purity: Confirm exosome preparations using:
Transmission electron microscopy for morphology
Nanoparticle tracking analysis for size distribution
Western blotting for exosomal markers (CD63, CD9, CD81)
ANGPTL7 localization: Determine if ANGPTL7 is on the exosome surface or internal using:
Protease protection assays
Immunogold electron microscopy
Surface biotinylation techniques
Functional studies: Compare the biological activities of:
Purified recombinant ANGPTL7
ANGPTL7-containing exosomes
Exosome-depleted soluble ANGPTL7
In vivo tracking: Use fluorescently labeled exosomes containing ANGPTL7 to track distribution and uptake in target tissues, particularly focusing on endothelial cell interactions.
Research into tissue-specific effects of ANGPTL7 variants requires:
Cross-tissue expression analysis: Compare expression patterns of wild-type and variant ANGPTL7 across relevant tissues (eye trabecular meshwork, hematopoietic niches, tumor microenvironments).
Variant functional characterization: Systematically analyze variants including:
Tissue-specific assays: Develop parallel experimental systems for:
Intraocular pressure regulation (trabecular meshwork cells)
Hematopoietic stem cell expansion (CD34+ culture systems)
Angiogenesis (endothelial cell assays)
to compare variant effects across tissue contexts.
Population genetics approaches: Analyze the distribution and selection pressures on ANGPTL7 variants across populations, noting the >50-fold enrichment of p.Arg220Cys in Finland compared to other populations .
Clinical correlation: Develop registries of individuals with rare ANGPTL7 variants to systematically assess phenotypes across multiple organ systems, potentially revealing previously unrecognized functions.
ANGPTL7 is composed of an N-terminal coiled coil domain and a C-terminal fibrinogen-like domain . This structural composition is crucial for its function. The protein is involved in various biological processes, including the regulation of angiogenesis, which is the formation of new blood vessels from pre-existing ones .
The biological activity of ANGPTL7 has been studied in various experimental settings. For instance, it has been shown to promote the expansion of E16 rat liver mononuclear cells in vitro . This activity is measured in the presence of other recombinant proteins such as mouse SCF/c-kit ligand, mouse thrombopoietin, and mouse Flt-3 ligand .
Recombinant human ANGPTL7 is typically produced using Chinese Hamster Ovary (CHO) cell lines . The recombinant protein is purified to a high degree, with a purity greater than 95% as determined by SDS-PAGE under reducing conditions . The endotoxin level is kept below 1.0 EU per microgram of protein, ensuring its suitability for various experimental applications .
Recombinant ANGPTL7 is used in various research applications, including studies on angiogenesis, glaucoma, and other conditions where its regulatory role in blood vessel formation and maintenance is of interest . The protein is available in different formulations, with or without carrier proteins like Bovine Serum Albumin (BSA), depending on the specific requirements of the experimental setup .