Endoglin, also known as ENG or CD105, is a 180 kDa homodimeric transmembrane glycoprotein that functions as a co-receptor for ligands of the transforming growth factor-β (TGF-β) superfamily, particularly bone morphogenetic proteins 9 and 10 (BMP9/10), thereby modulating cellular signaling pathways essential for vascular development and angiogenesis.[1]Encoded by the ENG gene located on chromosome 9q34.11, endoglin consists of an extracellular domain featuring a zona pellucida (ZP) module with an RGD integrin-binding motif, a short transmembrane region, and a cytoplasmic tail that exists in two main isoforms: long endoglin (L-endoglin) with 47 cytoplasmic amino acids and short endoglin (S-endoglin) with 14 amino acids.[2][1] These isoforms differentially influence TGF-β signaling, with L-endoglin promoting pro-angiogenic ALK1/Smad1/5/8 pathways and S-endoglin favoring anti-proliferative ALK5/Smad2/3 routes.[3] Beyond TGF-β modulation, endoglin facilitates integrin-mediated cell adhesion, supporting interactions between endothelial cells, leukocytes, platelets, and mural cells during inflammation and vascular repair.[4][2]Primarily expressed on endothelial cells of small and large vessels, endoglin is also found on vascular smooth muscle cells, activated monocytes/macrophages, fibroblasts, syncytiotrophoblasts, and progenitor cells such as endothelial colony-forming cells, with expression upregulated during angiogenesis, wound healing, and pathological conditions like inflammation or hypoxia.[1][4] A soluble form of endoglin (sEng), generated by proteolytic cleavage via matrix metalloproteinases (e.g., MMP12/14), circulates in plasma and acts as a decoy receptor, antagonizing membrane-bound endoglin functions and contributing to vascular dysfunction.[2][1]In clinical contexts, heterozygous mutations in ENG cause hereditary hemorrhagic telangiectasia type 1 (HHT1), an autosomal dominant disorder affecting approximately 1 in 5,000–8,000 individuals, characterized by arteriovenous malformations, recurrent epistaxis, and gastrointestinal bleeding due to impaired vascular integrity.[4] Elevated endoglin expression on proliferating endothelial cells makes it a biomarker for tumor angiogenesis in various cancers, including breast, colorectal, and prostate, where it correlates with poor prognosis and metastasis.[5][3] Additionally, increased sEng levels are implicated in preeclampsia, where concentrations can exceed 400 ng/mL in severe cases like HELLP syndrome, exacerbating endothelial damage and hypertension, as well as in metabolic disorders such as diabetes, obesity, and atherosclerosis.[2][1] Therapeutic strategies targeting endoglin, including monoclonal antibodies like TRC105, are under investigation for inhibiting pathological angiogenesis in cancer and HHT.[5]
Genetics and Expression
Gene Structure and Location
The ENG gene, encoding the endoglin protein, is located on the long arm of human chromosome 9 at cytogenetic band 9q34.11, spanning genomic coordinates 127,815,016–127,854,658 on reference sequence NC_000009.12.[6] This positioning places it within a region associated with vascular regulatory functions, though the gene itself measures approximately 40 kb in length.[6] Orthologs of ENG are conserved in other mammals, including the Eng gene on mouse chromosome 2 at position 22.09 cM.[7]The gene comprises 16 exons, with the first exon being entirely non-coding and contributing to the 5' untranslated region (UTR).[6][8] The coding sequence begins in exon 2 and extends through the remaining exons, producing multiple transcript isoforms via alternative splicing, such as the predominant long (L-ENG) and short (S-ENG) forms differing by a 141-nucleotide insertion in the cytoplasmic tail region that encodes 47 additional amino acids.[9] Introns interrupt the exons variably, with larger introns flanking the extracellular domain-coding regions, reflecting a modular genomic organization typical of transmembrane receptor genes.[6]The proximal promoter of ENG lacks canonical TATA and CAAT boxes but features GC-rich regions that bind Sp1 transcription factors, along with a functional hypoxia-inducible factor-1α (HIF-1α) binding site at nucleotides +338 to +344 relative to the transcription start site.[10] This HIF-1α site enables transcriptional upregulation in response to hypoxia, a key regulatory mechanism influencing endoglin expression in vascular contexts.[10]Evolutionarily, the ENG gene is highly conserved across vertebrates, descending from an ancestral form present in early chordates, as evidenced by phylogenetic analyses showing sequence similarity greater than 70% in the coding regions among mammals, birds, and fish.[11] Exons encoding the transmembrane domain, particularly exons 13 and 14, display the highest conservation, with nucleotide identity exceeding 90% between human and murine sequences, highlighting their critical role in maintaining protein membraneintegration and function.[11] This conservation underscores ENG's fundamental involvement in vasculogenesis from zebrafish to humans.[12]
Tissue-Specific Expression and Regulation
Endoglin exhibits high expression in endothelial cells of both developing and adult vasculature, as well as in vascular smooth muscle cells, syncytiotrophoblasts, and activated monocytes.[13] In the vascular system, it is particularly prominent in microvascular endothelium and proliferating endothelial cells during tissue remodeling.[14] Syncytiotrophoblasts in the placenta show constitutive high levels, supporting trophoblast differentiation and placental vascular development.[13] Activated monocytes upregulate endoglin during differentiation into macrophages, aiding in immune responses.[13]Endoglin expression is upregulated during angiogenesis, inflammation, and wound healing, reflecting its role in dynamic vascular and tissue repair processes.[13] In angiogenic contexts, such as tumor or regenerating tissues, it increases in endothelial cells to facilitate vessel formation.[14] During inflammation and wound healing, expression rises in endothelial and immune cells, including monocytes, to modulate leukocyte recruitment and tissue repair.[3] Basal expression occurs in pre-B cells, particularly in leukemic lines, and in erythroid precursors like pro-erythroblasts and CFU-E, where it supports hematopoietic differentiation.[13]Several regulatory factors control endoglin expression, including hypoxia mediated by HIF-1α, inflammatory cytokines such as TNF-α and IL-1β, and shear stress in endothelial cells. Hypoxia induces endoglin transcription through a hypoxia-responsive element in the promoter, where HIF-1α binds in cooperation with Sp1 and Smad3 to form a multiprotein complex.[15] Inflammatory cytokines like TNF-α and IL-1β upregulate endoglin in monocytes and endothelial cells during inflammatory responses, enhancing macrophage polarization and vascular permeability.[13]Shear stress in endothelium activates endoglin via the ALK1-Endoglin-SMAD1/5 pathway, promoting gene expression changes that maintain vascular tone and integrity.[16]During development, endoglin expression peaks in embryogenesis, particularly in the heart and yolk sac vasculature, before declining postnatally except in proliferating endothelium. Expression is first detected at embryonic day 6.5 in mice within the amniotic fold and allantois, expanding by days 7.5 to 8.5 to mesodermal derivatives, yolk sac endothelium, and developing heart vessels.[17] It reaches high levels around embryonic day 10.5 during active angiogenesis in these sites, supporting hemangioblast specification and early vascular patterning.[3] Postnatally, levels decrease in quiescent tissues but remain elevated in areas of ongoing endothelial proliferation, such as during adult angiogenesis.[14]
Protein Structure and Biochemistry
Primary Structure and Domains
Endoglin is a type I transmembrane glycoprotein that exists primarily as a disulfide-linked homodimer with an apparent molecular weight of approximately 180 kDa due to extensive glycosylation. The human endoglin precursor protein consists of 658 amino acids, with a 25-amino-acid signal peptide cleaved to yield the mature polypeptide of 633 amino acids. This mature form features a large extracellular domain spanning 561 amino acids (residues 26–586), a short hydrophobic transmembrane helix of about 25 amino acids (residues 587–611), and a cytoplasmic tail of 47 amino acids (residues 612–658).[18][19][20]The extracellular domain is divided into two main structural modules: an N-terminal orphan domain (residues 26–359) and a C-terminal zona pellucida (ZP) module (residues 360–586). The orphan domain, which lacks significant homology to other known proteins, adopts a β-propeller-like structure. The ZP module, characteristic of the zona pellucida superfamily, consists of approximately 260 amino acids, forms a rigid, β-sheet-rich domain that contributes to the overall stability of the extracellular region, and contains a conserved RGD integrin-binding motif at residues Arg374-Gly375-Asp376.[21][20][18]The cytoplasmic tail is rich in serine and threonine residues, harboring multiple potential phosphorylation sites that may regulate protein interactions and trafficking, though endoglin possesses no intrinsic kinase activity. In comparison to other members of the transforming growth factor-β (TGF-β) receptor family, endoglin functions as an accessory receptor without specific ligand-binding specificity in its extracellular domain.[20][22][23]
Post-Translational Modifications and Oligomerization
Endoglin, a transmembrane glycoprotein, undergoes extensive post-translational modifications that are essential for its proper folding, stability, and cell surface expression. The extracellular domain features N-linked glycosylation at eight consensus sites (Asn-X-Ser/Thr), primarily within the orphan and zona pellucida (ZP) domains, which accounts for a substantial portion of the mature monomer's apparent molecular weight of approximately 90 kDa. These glycosylations involve the attachment of complex N-glycans, including high-mannose and hybrid forms processed in the endoplasmic reticulum and Golgi apparatus. Additionally, O-linked glycosylation occurs on serine and threonine residues, particularly in a mucin-like region rich in these amino acids proximal to the transmembrane domain, while sialylation of both N- and O-glycans adds negative charge and influences protein conformation and interactions.[24][20][19]Disulfide bonds play a critical role in stabilizing endoglin's structure, with 9-10 intramolecular bonds per monomer forming within the orphandomain (e.g., Cys30-Cys207 and Cys53-Cys182) and ZP domain, including additional conserved cysteines that maintain the beta-sandwich folds characteristic of these modules. Intermolecular disulfide bonds, specifically two per dimer involving Cys516 and Cys582 in the ZP domain, covalently link the two monomers to form the predominant homodimeric configuration observed on the cell surface. These covalent linkages, combined with non-covalent interactions, ensure the protein's quaternary stability against proteolytic degradation and mechanical stress in the vascular environment. The primary sequence of the extracellular domain, featuring cysteine-rich ZP modules, provides the framework for these disulfide pairings.[25][26][27]Proteolytic processing generates soluble forms of endoglin (sEng) through ectodomain shedding, primarily mediated by matrix metalloproteinase-14 (MMP-14) at sites within the extracellular domain, releasing anti-angiogenic fragments into circulation. Recent studies have identified thrombin as a key protease cleaving near the transmembrane domain (e.g., at Arg-Val or similar motifs), particularly under coagulopathic conditions, yielding heterogeneous sEng isoforms of 60-100 kDa. These cleavage events are regulated by the protein's glycosylated and disulfide-stabilized structure, which protects core regions but exposes juxtamembrane sites to proteases.[28][29][30]Endoglin primarily exists as disulfide-linked homodimers on the plasma membrane, with each monomer spanning approximately 658 amino acids, but evidence suggests potential for higher-order oligomerization within specialized membrane microdomains. In lipid rafts enriched with caveolin-1, endoglin dimers may associate into transient multimers, facilitated by glycosylphosphatidylinositol-anchored proteins and cholesterol, enhancing local concentration for ligand presentation without altering the core dimeric architecture.[31][32]
Molecular Interactions and Signaling
Binding Partners
Endoglin functions as a co-receptor for multiple ligands within the transforming growth factor-β (TGF-β) superfamily, serving as an auxiliary component for TGF-β1 and TGF-β3 in association with type II receptors (TGFBR2), while directly binding bone morphogenetic proteins 9 and 10 (BMP9/10) with high affinity through its extracellular domain, thereby facilitating their presentation to signaling receptors.[33][34] It also interacts with BMP-2 and BMP-7, as well as activin-A, via associations with activin type II receptors (ActRII and ActRIIB), thereby modulating these ligands' access to their canonical receptor complexes.[33] These bindings occur predominantly in the zona pellucida (ZP) domain and orphan regions of endoglin's extracellular portion, enabling auxiliary roles in ligand-receptor assembly on endothelial cells.In terms of receptor partnerships, endoglin forms heterotetrameric complexes with TGFBR1 (also known as ALK5), ACVRL1 (ALK1), and TGFBR2, enhancing TGF-β-dependent signaling in endothelial contexts.[35] Similarly, it associates with BMP type I receptors (BMPR1A and BMPR1B) and the type II receptor BMPR2, particularly for BMP-9 and BMP-10 ligands, stabilizing these complexes to promote Smad1/5/8 pathway activation. These interactions are mediated by endoglin's transmembrane and extracellular domains, allowing cooperative ligand binding without altering the core kinase activities of the associated receptors.[36]Beyond TGF-β family components, endoglin engages integrin α5β1 via an RGD motif in its extracellular domain, supporting endothelial-leukocyte adhesion and fibronectin-mediated crosstalk during inflammatory responses.[37] The cytoplasmic tail of endoglin further binds β-arrestin-2 and GIPC-1 (GAIP-interacting protein C-terminus-1), facilitating receptor endocytosis and intracellular trafficking independent of canonical Smad signaling.Soluble endoglin, generated by proteolytic shedding of the membrane-bound form, acts as an antagonist by binding BMP9 and BMP10, thereby trapping them and preventing their interaction with cell-surface endoglin or signaling receptors. This soluble variant competes with membrane endoglin for BMP9/10 availability, particularly in pathological conditions like preeclampsia where elevated levels disrupt vascular homeostasis. It exhibits low-affinity binding to TGF-β1 and TGF-β3 but does not significantly sequester these ligands on its own.[38][34]
Modulation of TGF-β Signaling Pathways
Endoglin serves as a co-receptor in the TGF-β superfamily signaling, exerting a dual role in endothelial cells by promoting the ALK1/Smad1/5/8 pathway, which drives proliferative and pro-angiogenic responses, while inhibiting the ALK5/Smad2/3 pathway associated with anti-proliferative effects.[39] This pathway selection is critical for maintaining endothelial function, as endoglin facilitates the balance between cell proliferation and quiescence.[40]The mechanistic basis involves endoglin's recruitment of ALK1 and TGFBR2 into signaling complexes on the cell surface, which enhances phosphorylation of Smad1/5/8 in response to TGF-β ligands, thereby amplifying downstream transcriptional activity via elements like the BMP-responsive element (BRE).[39] Conversely, this enhancement indirectly suppresses ALK5/Smad2/3 signaling by competing for ligand availability and receptor interactions, reducing Smad2 phosphorylation and responses measured by CAGA-luciferase reporters.[39] Soluble endoglin primarily antagonizes BMP9/10-induced ALK1/Smad1/5/8 signaling by sequestering these ligands; its effect on TGF-β pathways is limited alone but can be synergistic with other soluble TGF-β receptors to inhibit both ALK1/Smad1/5/8 and ALK5/Smad2/3 pathways.[41]Endoglin integrates cross-talk between TGF-β and BMP signaling through shared Smad1/5/8 effectors, as it binds BMP9/10 to promote similar pro-angiogenic outputs via ALK1 complexes. Additionally, the cytoplasmic tail of endoglin undergoes basal phosphorylation by ALK5 and TGFBR2, primarily at serines 646 and 649, which regulates the magnitude of Smad1/5/8 activation and endothelial migration; disruption of these sites impairs pathway inhibition and BMP9 responsiveness.[42] Endoglin expression levels further dictate pathway bias, with high levels favoring ALK1/Smad1/5/8 signaling and low levels shifting toward ALK5 dominance, as observed in heterozygous endothelial cells where reduced endoglin correlates with decreased ALK5 expression and altered Smad responses.[39]
Physiological Functions
Role in Vascular Development and Angiogenesis
Endoglin plays a critical role in embryonic vascular development, particularly in vasculogenesis and angiogenesis. Endoglin-null mice exhibit embryonic lethality between E10.5 and E11.5, characterized by severe impairments in yolk sac vascularization and cardiac development. Specifically, these embryos display disorganized endothelial cells in the yolk sac with multiple pockets of red blood cells and no organized vascular structures, alongside failure of the cardiac tube to complete rotation, accompanied by serosanguinous pericardial effusion. These defects highlight endoglin's necessity for proper vessel assembly and heart outflow tract formation during early embryogenesis.At the molecular level, endoglin facilitates endothelial cell behaviors essential for angiogenesis, including migration, proliferation, and tube formation, primarily through modulation of TGF-β signaling via the ALK1/Smad1/5/8 pathway. In endothelial cells, endoglin enhances ALK1-mediated Smad1/5/8 phosphorylation in response to TGF-β, promoting proliferative and migratory responses required for vessel sprouting and network formation. Additionally, endoglin indirectly interacts with VEGF signaling by associating with VEGFR2, thereby augmenting VEGF-induced endothelial activation and angiogenic responses without directly binding VEGF ligands. This dual regulation ensures coordinated endothelial dynamics during vascular patterning.[43][44][45]While heterozygous loss-of-function mutations in endoglin cause hereditary hemorrhagic telangiectasia type 1 with postnatal vascular malformations, complete loss of endoglin leads to embryonic lethality, as observed in endoglin-null mice with impaired yolk sac vascularization and heart outflow tract defects; similar lethality occurs in rare reported cases of homozygous mutations in humans.[46] These findings underscore endoglin's conserved role across species in embryonic angiogenesis. Postnatally, endoglin is required for physiological vessel sprouting in contexts such as retinal vascularization and wound healing. Conditional endothelial-specific endoglin knockout in mice results in abnormal retinal vascular plexuses with delayed remodeling and increased vascular fragility during neonatal angiogenesis. Similarly, endoglin heterozygosity impairs wound-induced angiogenesis by reducing endothelial proliferation and nitric oxide-mediated vessel formation. This postnatal function involves interplay with hypoxia-inducible factor (HIF), where HIF-1α upregulates endoglin expression to support angiogenic adaptation in low-oxygen environments.[47][48][49][50]
Functions in Adult Vascular Homeostasis
In adult vasculature, endoglin maintains endothelial barrier integrity by regulating adherens junctions and cytoskeletal dynamics, thereby preventing vascular leakage. Endoglin deficiency in endothelial cells increases permeability through dysregulated RhoA activation and enhanced stress fiber formation, leading to disrupted VE-cadherin organization at cell-cell contacts. Conditional knockout of endoglin in adult mice results in hemorrhage and vascular fragility, underscoring its essential role in stabilizing mature vessel architecture and averting arteriovenous malformations. This function is mediated via modulation of TGF-β signaling, which supports endothelial quiescence and junctional stability under physiological conditions.Following vascular injury, endoglin expression is upregulated in endothelial cells to facilitate re-endothelialization and repair processes. It promotes endothelial regeneration after arterial damage by enhancing cell proliferation and migration, as evidenced by reduced re-endothelialization in endoglin-deficient models post-injury. Additionally, endoglin modulates shear stress-induced signaling in endothelium, activating ALK1-Smad1/5 pathways in response to fluid flow to maintain vascular tone and prevent excessive remodeling. This shear-responsive mechanism helps endothelium adapt to hemodynamic changes, ensuring homeostasis during repair without aberrant angiogenesis.Endoglin expressed in bone marrow stromal endothelial cells supports hematopoietic homeostasis by preserving vascular niche integrity and aiding stem cell functions. It maintains the bone marrow vasculature's barrier properties, which is critical for long-term hematopoietic stem cell (HSC) retention and mobilization. Endoglin-positive endothelial subsets in the bone marrow niche facilitate HSC homing through regulated adhesion and TGF-β signaling, while also contributing to erythropoiesis by supporting erythroid progenitor differentiation in the stromal environment.During pregnancy, endoglin plays a vital role in placental vascular homeostasis by regulating trophoblastinvasion and spiral artery remodeling. Expressed on extravillous trophoblasts, it promotes their migratory and invasive differentiation via TGF-β and ERK/AKT pathways, enabling proper endometrial penetration and arterial wall modification. This ensures adequate uteroplacental blood flow; disruptions in endoglin function impair trophoblast endovascular invasion, compromising spiral arterytransformation and maternal-fetal nutrient exchange.
Pathological Implications
Mutations in Hereditary Hemorrhagic Telangiectasia
Hereditary hemorrhagic telangiectasia type 1 (HHT1) is an autosomal dominant disorder caused by heterozygous mutations in the ENG gene, accounting for approximately 50% of all HHT cases. These mutations typically result in haploinsufficiency or production of a dysfunctional endoglin protein, as the majority (~80%) are loss-of-function variants such as nonsense, frameshift, or splice-site alterations that lead to truncated or absent protein. Missense mutations, which may disrupt protein folding or interactions, constitute a smaller proportion.[51][52][53]Over 500 distinct pathogenic ENG variants have been identified and cataloged in specialized databases, with mutations distributed across nearly all exons of the 14-exon gene, though no clear hotspots predominate. Common examples include frameshift deletions and nonsense mutations that prematurely terminate translation, often in the extracellular domain. These variants are documented in comprehensive repositories like the Leiden Open Variation Database (LOVD) for ENG, facilitating clinical genetic testing and counseling.[53][54]At the molecular level, reduced endoglin expression impairs TGF-β signaling through the ALK1 receptor complex, disrupting endothelial cell function and leading to defective vascular remodeling during development and adulthood. This manifests as fragile telangiectasias and arteriovenous malformations (AVMs) primarily in the nasal mucosa, gastrointestinal tract, lungs, liver, and brain, due to abnormal arteriovenous shunting and vessel wall instability.[55][56]Clinically, HHT1 presents with recurrent epistaxis typically onset in the teenage years (average age 12), mucocutaneous telangiectasias, and visceral involvement, including pulmonary AVMs in 30-50% of cases—higher than in HHT2—and cerebral AVMs in about 10-20%. Genotype-phenotype correlations indicate that certain truncating ENG mutations, such as those resulting in severe haploinsufficiency, are associated with more aggressive pulmonary AVMs requiring early intervention, while milder missense variants may correlate with later-onset or less severe manifestations.[54][57][58]
Upregulation in Cancer and Tumor Angiogenesis
Endoglin is overexpressed in the endothelium of tumor blood vessels across various malignancies, where it serves as a marker of proliferating endothelial cells and promotes neovascularization essential for tumor growth. This upregulation is particularly evident in solid tumors such as breast, prostate, and colorectal cancers, where endoglin expression correlates with increased microvessel density (MVD) and is associated with aggressive disease features including metastasis and reduced patient survival.[59] In addition to endothelial cells, endoglin is upregulated in certain cancer cells, contributing to their invasive potential, though its role in epithelial compartments can vary by tumor type.[5] High endoglin levels in tumor tissues have been linked to poor overall survival (OS), disease-free survival (DFS), and cancer-specific survival (CSS) in meta-analyses of multiple cancer cohorts.[60]The mechanisms underlying endoglin's role in tumor angiogenesis involve enhancement of pro-angiogenic signaling pathways. Endoglin acts as a co-receptor for transforming growth factor-β (TGF-β), preferentially directing signals through the activin receptor-like kinase 1 (ALK1)/Smad1/5/8 pathway in endothelial cells, which promotes cell proliferation, migration, and tube formation.[61] It synergizes with vascular endothelial growth factor (VEGF) signaling by interacting with VEGFR2, stabilizing the receptor on the cell surface and amplifying VEGF-induced tip cell formation during sprouting angiogenesis.[44] Soluble endoglin, generated by proteolytic shedding, paradoxically supports metastasis by acting as a decoy receptor that sequesters TGF-β ligands, thereby disrupting the cytokine's tumor-suppressive effects on epithelial cells and facilitating epithelial-to-mesenchymal transition (EMT).[62] Elevated circulating soluble endoglin levels are observed in patients with advanced cancers and correlate with metastatic progression.[63]In specific cancers, endoglin upregulation is prominent in androgen-independent prostate cancer, where its expression in the tumor microenvironment promotes androgen receptor variant 7 (AR-V7) signaling, driving castration-resistant progression and metastasis.[64] Similarly, in head and neck squamous cell carcinoma (HNSCC), endoglin is highly expressed in neo-vessels and cancer-associated fibroblasts, correlating with increased tumor invasiveness and lymph nodemetastasis.[65] Recent studies from 2022-2023 have linked elevated endoglin-positive MVD to enhanced glioma invasion and poorer progression-free survival in recurrent glioblastoma, highlighting its role in peritumoral vascular remodeling that supports tumor cell dissemination.[66]Experimental models demonstrate that inhibiting endoglin reduces tumor angiogenesis and growth. In breast cancer xenografts, anti-endoglin antibodies decrease vessel density and impair metastatic spread by disrupting ALK1 signaling.[67]Prostate cancer xenograft studies show that endoglin blockade suppresses tumor vascularization and limits progression to androgen-independent states.[68] These findings underscore endoglin's pro-angiogenic function in cancer, distinct from its regulated role in physiological vascular development.[5]
Involvement in Fibrosis and Inflammatory Diseases
Endoglin contributes to the pathogenesis of liver fibrosis by promoting the activation of hepatic stellate cells (HSCs) through enhancement of the TGF-β/ALK5/Smad3 signaling pathway, leading to increased expression of profibrotic genes such as α-smooth muscle actin and collagen I.[69] In patients with cirrhosis, endoglin expression is markedly elevated in fibrotic liver tissue and serum levels of soluble endoglin are significantly higher compared to healthy individuals, correlating with disease severity.[70] Experimental models demonstrate that targeting endoglin with monoclonal antibodies reduces HSC activation, decreases collagen deposition, and attenuates fibrosis progression in choline-deficient L-amino acid-defined high-fat diet (CDAA-HFD)-induced metabolic dysfunction-associated steatohepatitis (MASH).[71] A 2025 study further confirmed that anti-endoglin monoclonal antibodies prevent progression of liver sinusoidal endothelial inflammation and fibrosis in this MASH model.[72]Beyond the liver, endoglin is upregulated in fibrotic lesions of systemic sclerosis (SSc), where it drives fibroblast activation and excessive extracellular matrix production in both skin and lung tissues. In SSc skin biopsies, endoglin mRNA and protein levels are significantly higher in lesional fibroblasts from diffuse cutaneous SSc patients compared to healthy controls, promoting TGF-β-mediated collagen synthesis.[73] Similarly, in SSc-associated interstitial lung disease, elevated endoglin expression in lung fibroblasts correlates with reduced diffusing capacity for carbon monoxide and increased fibrotic gene expression.[73] In kidney fibrosis models, such as unilateral ureteral obstruction, overexpression of the short isoform of endoglin (S-endoglin) exerts anti-fibrotic effects by suppressing TGF-β/Smad signaling, reducing myofibroblast differentiation, and limiting inflammation and extracellular matrix accumulation.[74]In inflammatory processes, endoglin modulates monocyte activation by facilitating their adhesion and transmigration across endothelial barriers, thereby amplifying vascular inflammation. Endoglin expression on activated monocytes enhances integrin-mediated interactions with endothelial cells, promoting inflammatory infiltration in response to chemokines like CXCL12. Additionally, endoglin influences Th17 cell responses by regulating TGF-β signaling in CD4+ T cells, where it can shift the balance toward proinflammatory Th17 differentiation and IL-17 production during chronic inflammation. In preeclampsia, elevated circulating levels of soluble endoglin (sEng) induce endothelial dysfunction by antagonizing TGF-β and BMP signaling, leading to increased vascular permeability, hypertension, and a preeclampsia-like syndrome in animal models. High sEng concentrations, often exceeding 40 ng/mL near term, correlate with severe maternal hypertension and proteinuria in affected pregnancies.In COVID-19, soluble endoglin levels predict disease severity and clinical outcomes, with higher concentrations in severe cases linked to endothelial damage and vascular inflammation, as shown in a 2024 cohort study of hospitalized patients.[75] These findings underscore endoglin's role in disrupting vascular homeostasis during acute inflammatory insults.
Endoglin functions as a valuable biomarker in both membrane-bound and soluble forms, enabling the diagnosis, prognosis, and monitoring of vascular-related diseases such as cancer, preeclampsia, and hereditary hemorrhagic telangiectasia (HHT). Membrane-bound endoglin, also known as CD105, is particularly useful in assessing tumor angiogenesis through immunohistochemical staining, which highlights activated endothelial cells in tumor vasculature. In breast cancer biopsies, for example, CD105-based quantification of microvessel density provides insights into neovascularization and correlates with tumor aggressiveness and metastatic potential.[76][60]Soluble endoglin (sEng), a circulating fragment generated by proteolytic cleavage, serves as a serum biomarker for conditions involving dysregulated angiogenesis. In preeclampsia, sEng levels rise more than twofold in affected pregnancies compared to normotensive ones, with elevations detectable weeks before clinical onset and serving as a predictor of disease severity and associated complications like eclampsia or fetal growth restriction.[77][78] Similarly, in HHT, altered sEng levels—often reduced due to underlying mutations—aid in screening for arteriovenous malformations (AVMs), helping to identify patients at risk for complications such as hemorrhage or organ dysfunction.[79][80]The prognostic utility of endoglin extends to oncology and fibrotic disorders. High tissue expression of endoglin in prostate and ovarian cancers is associated with increased metastatic risk and poorer disease-free survival, reflecting its role in promoting tumor invasion and vascular remodeling. As of 2025, elevated plasma endoglin levels have also been associated with better prognosis and increased sensitivity to pemetrexed in non-small cell lung cancer patients.[81][82][83] In fibrosis, elevated sEng levels correlate with disease progression, as seen in conditions like systemic sclerosis or liver fibrogenesis, where it indicates ongoing endothelial activation and extracellular matrix deposition.[84][85]Clinical measurement of endoglin relies on established assays, with enzyme-linked immunosorbent assay (ELISA) being the standard for quantifying sEng in serum or plasma; normal levels typically range below 10 ng/mL in healthy individuals, though values can vary slightly by population and assay kit.[86][87] Emerging approaches include liquid biopsies that detect circulating endothelial cells expressing endoglin, offering a non-invasive method to monitor endothelial dysfunction and tumor burden in real-time.[88] These tools enhance precision in disease stratification and therapeutic response evaluation.
Strategies for Therapeutic Targeting
One prominent strategy for therapeutic targeting of endoglin involves monoclonal antibodies that bind to the protein on endothelial cells to disrupt angiogenesis. TRC105 (carotuximab), a chimeric IgG1 anti-endoglin antibody, inhibits endothelial cell proliferation and induces antibody-dependent cellular cytotoxicity, thereby suppressing tumor vascularization in preclinical models.[89] In clinical trials, TRC105 has advanced to phase II and III evaluations for solid tumors, demonstrating tolerability and preliminary efficacy when combined with pazopanib in advanced angiosarcoma, where it enhanced progression-free survival compared to pazopanib alone.[90] Additionally, in combination with bevacizumab for bevacizumab-refractory advanced solid tumors, including platinum-resistant ovarian cancer, TRC105 led to radiographic tumor reductions in a subset of patients and was well-tolerated, supporting its role in overcoming VEGF pathway resistance.[91][92]Soluble endoglin (sEng) decoys represent another approach to modulate endoglin function by sequestering TGF-β ligands and inhibiting downstream signaling. Engineered recombinant sEng acts as a competitive antagonist, binding TGF-β and preventing its interaction with membrane-bound receptors.[93] In preclinical models of cardiac fibrosis, administration of sEng reduced TGF-β1-induced activation of cardiac fibroblasts, decreased collagen deposition, and attenuated fibrotic remodeling, highlighting its potential to counteract pro-fibrotic TGF-β effects.[94] Similar decoy strategies have been explored in preeclampsia models, where modulating sEng levels influences TGF-β-mediated vascular dysfunction, though therapeutic application remains investigational.[95]Gene therapy offers promise for restoring endoglin function in hereditary hemorrhagic telangiectasia (HHT), particularly for loss-of-function mutations. Nonviral gene delivery systems, such as lipid-complexed plasmids expressing human endoglin cDNA under endothelial-specific promoters, have been tested in Eng heterozygous HHT mouse models, resulting in restored endoglin expression on lung endothelium, normalization of vascular architecture, and reduced lesion formation that correlates with decreased bleeding propensity.[96]CRISPR/Cas9-based editing has also enabled precise correction of ENG mutations in patient-derived induced pluripotent stem cells, yielding endothelial cells with normalized endoglin levels and improved angiogenic responses in vitro, paving the way for autologous cell therapies.[97]Emerging strategies include small molecules disrupting endoglin-integrin interactions and radioimmunoconjugates leveraging anti-endoglin antibodies. Preclinical data indicate that endoglin's RGD motif facilitates binding to integrins like α5β1 and αIIbβ3, promoting leukocyte adhesion and thrombus stability; targeting these interactions with small molecules could mitigate pathological vascular remodeling, with 2023 studies reporting inhibition of endothelial migration in vitro.[2] In hepatocellular carcinoma models, radioimmunotherapy using 177Lu-labeled anti-CD105 monoclonal antibodies selectively targets endoglin-expressing tumor vasculature, achieving significant tumor regression and prolonged survival in mice with minimal off-target toxicity.[98] As of 2025, preclinical development of endoglin-directed chimeric antigen receptor (CAR) T cells has shown comprehensive targeting of tumors in xenograft models of bone and soft-tissue sarcomas. Additionally, endothelium-specific endoglin has been implicated in triggering astrocyte reactivity in Alzheimer's disease models, suggesting potential therapeutic strategies targeting ENG to mitigate neurovascular dysfunction.[99][100] Patient selection for these therapies may incorporate endoglin as a biomarker to identify high-expression cases responsive to targeting.[89]