Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.184
Filter
1.
Anticancer Res ; 44(10): 4435-4448, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39348958

ABSTRACT

BACKGROUND/AIM: Predicting lymph node metastasis (LNM) in early gastric cancer (EGC) is crucial for making treatment decisions. This study aimed to confirm risk factors for LNM and identify novel auxiliary biomarkers for predicting LNM in EGC. PATIENTS AND METHODS: We established a training set, comprising 63 patients with LNM-EGC and 274 patients with non-LNM EGC, and a test set, comprising 19 patients with LNM-EGC and 146 non-LNM EGC. Immunohistochemistry for lymphangiogenic and related pathway components (VEGF-C, TGF-ß1, SMAD2/3, VEGF-D, pSTAT3, E-cadherin, CD44, c-MET, YAP, and HER2), in situ hybridization for Epstein-Barr virus-encoded small RNAs, and multiplex PCR for microsatellite instability were conducted. RESULTS: In the training set, Lauren's diffuse/mixed classification, stromal desmoplasia, submucosal invasion ≥500 µm, lymphatic invasion, and high VEGF-C and SMAD2/3 expression were independent risk factors for LNM (p<0.05). A large tumor size, mixed histology, submucosal invasion, perineural invasion, and ulceration were determined as risk factors using univariate analysis (p<0.05). The tumor cutoff size for predicting LNM was 2.65 cm, based on a ROC analysis. The test set study verified that stromal desmoplasia, submucosal invasion, and high VEGF-C expression were independent risk factors for LNM (p<0.05). Moreover, mixed histology, lymphatic invasion, ulceration, and high SMAD 2/3 expression were identified as additional risk factors using univariate analysis (p<0.05). CONCLUSION: Stromal desmoplasia, submucosal invasion, and high VEGF-C expression are potential biomarkers for LNM in EGC. VEGF-C expression might serve as an adjunct biomarker for predicting LNM on forceps-biopsy tissue at initial diagnosis.


Subject(s)
Biomarkers, Tumor , Herpesvirus 4, Human , Lymphatic Metastasis , Microsatellite Instability , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/virology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Male , Female , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Middle Aged , Herpesvirus 4, Human/genetics , Aged , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/pathology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Adult , Smad2 Protein/metabolism , Smad2 Protein/genetics , Risk Factors
2.
Int J Mol Sci ; 25(17)2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39273646

ABSTRACT

Corneal transparency and avascularity are essential for vision. The avascular cornea transitions into the vascularized conjunctiva at the limbus. Here, we explore a limbal stromal cell sub-population that expresses ABCB5 and has mesenchymal stem cell characteristics. Human primary corneal stromal cells were enriched for ABCB5 by using FACS sorting. ABCB5+ cells expressed the MSC markers CD90, CD73, and CD105. ABCB5+ but not ABCB5- cells from the same donor displayed evidence of pluripotency with a significantly higher colony-forming efficiency and the ability of trilineage differentiation (osteogenic, adipogenic, and chondrogenic). The ABCB5+ cell secretome demonstrated lower levels of the pro-inflammatory protein MIF (macrophage migration inhibitory factor) as well as of the pro-(lymph)angiogenic growth factors VEGFA and VEGFC, which correlated with reduced proliferation of Jurkat cells co-cultured with ABCB5+ cells and decreased proliferation of blood and lymphatic endothelial cells cultured in ABCB5+ cell-conditioned media. These data support the hypothesis that ABCB5+ limbal stromal cells are a putative MSC population with potential anti-inflammatory and anti-(lymph)angiogenic effects. The therapeutic modulation of ABCB5+ limbal stromal cells may prevent cornea neovascularization and inflammation and, if transplanted to other sites in the body, provide similar protective properties to other tissues.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Mesenchymal Stem Cells , Vascular Endothelial Growth Factor A , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Vascular Endothelial Growth Factor A/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Cell Differentiation , Vascular Endothelial Growth Factor C/metabolism , Cell Proliferation , Limbus Corneae/metabolism , Limbus Corneae/cytology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Jurkat Cells , Cells, Cultured , Stromal Cells/metabolism , Coculture Techniques , Endothelial Cells/metabolism
3.
Immunity ; 57(10): 2328-2343.e8, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39217987

ABSTRACT

The precise neurophysiological changes prompted by meningeal lymphatic dysfunction remain unclear. Here, we showed that inducing meningeal lymphatic vessel ablation in adult mice led to gene expression changes in glial cells, followed by reductions in mature oligodendrocyte numbers and specific lipid species in the brain. These phenomena were accompanied by altered meningeal adaptive immunity and brain myeloid cell activation. During brain remyelination, meningeal lymphatic dysfunction provoked a state of immunosuppression that contributed to delayed spontaneous oligodendrocyte replenishment and axonal loss. The deficiencies in mature oligodendrocytes and neuroinflammation due to impaired meningeal lymphatic function were solely recapitulated in immunocompetent mice. Patients diagnosed with multiple sclerosis presented reduced vascular endothelial growth factor C in the cerebrospinal fluid, particularly shortly after clinical relapses, possibly indicative of poor meningeal lymphatic function. These data demonstrate that meningeal lymphatics regulate oligodendrocyte function and brain myelination, which might have implications for human demyelinating diseases.


Subject(s)
Brain , Lymphatic Vessels , Meninges , Multiple Sclerosis , Myelin Sheath , Oligodendroglia , Animals , Oligodendroglia/metabolism , Mice , Meninges/immunology , Brain/metabolism , Brain/immunology , Humans , Myelin Sheath/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Vascular Endothelial Growth Factor C/metabolism , Mice, Inbred C57BL , Cell Survival , Remyelination , Female , Male , Adaptive Immunity
4.
Commun Biol ; 7(1): 962, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122919

ABSTRACT

With increases in life expectancy, the number of patients requiring joint replacement therapy and experiencing periprosthetic osteolysis, the most common complication leading to implant failure, is growing or underestimated. In this study, we found that osteolysis progression and osteoclast differentiation in the surface of the skull bone of adult mice were accompanied by significant expansion of lymphatic vessels within bones. Using recombinant VEGF-C protein to activate VEGFR3 and promote proliferation of lymphatic vessels in bone, we counteracted excessive differentiation of osteoclasts and osteolysis caused by titanium alloy particles or inflammatory cytokines LPS/TNF-α. However, this effect was not observed in aged mice because adipogenically differentiated mesenchymal stem cells (MSCs) inhibited the response of lymphatic endothelial cells to agonist proteins. The addition of the JAK inhibitor ruxolitinib restored the response of lymphatic vessels to external stimuli in aged mice to protect against osteolysis progression. These findings suggest that inhibiting SASP secretion by adipogenically differentiated MSCs while activating lymphatic vessels in bone offers a new method to prevent periprosthetic osteolysis during joint replacement follow-up.


Subject(s)
Lymphatic Vessels , Mesenchymal Stem Cells , Osteolysis , Animals , Osteolysis/prevention & control , Mice , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Aging , Mice, Inbred C57BL , Osteoclasts/metabolism , Osteoclasts/drug effects , Cell Differentiation/drug effects , Male , Phenotype , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Skull/pathology , Skull/drug effects , Vascular Endothelial Growth Factor Receptor-3/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Titanium
5.
FASEB J ; 38(16): e23879, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39162663

ABSTRACT

Both lymphatic vessels and macrophages are key factors influencing the inflammatory response. During the inflammatory response, lymphatic vessels undergo dilation and growth, playing a beneficial role in alleviating inflammation by facilitating the drainage of exudate, inflammatory mediators, and leukocytes. Consequently, the promotion of lymphangiogenesis has emerged as a novel therapeutic approach to treating inflammation. Macrophages play a crucial role in promoting lymphangiogenesis by secreting several pro-lymphatic growth factors, including vascular endothelial growth factor (VEGF)-C, and undergoing transdifferentiation into lymphatic endothelial cell progenitors (LECP), which integrate into newly formed lymphatic vessels. Macrophages exhibit heterogeneity and perform diverse functions based on their phenotypes. The regulation of macrophage polarization is crucial in inflammatory responses. Notably, macrophages promote lymphangiogenesis, while lymphatic vessels, in turn, serve as a conduit for macrophages to drain out inflamed tissue and also affect macrophage polarization. Thus, there is an interactive relationship between them. In this review, we discuss current work on the effects of macrophages on lymphangiogenesis as well as lymphatic vessel recruitment of macrophages and regulation of macrophage polarization. Furthermore, we explore the roles of lymphatic vessels and macrophages in various inflammation-related diseases, emphasizing potential therapeutic targets within the context of lymphatic-macrophage interactions.


Subject(s)
Inflammation , Lymphangiogenesis , Lymphatic Vessels , Macrophages , Macrophages/immunology , Macrophages/metabolism , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Animals , Lymphangiogenesis/physiology , Vascular Endothelial Growth Factor C/metabolism
6.
Cell Oncol (Dordr) ; 47(5): 1927-1942, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39141315

ABSTRACT

PURPOSE: Dysfunctional lymphangiogenesis is pivotal for various pathological processes including tumor lymph node metastasis which is a crucial cause of therapeutic failure for ESCC. In this study, we aim to elucidate the molecular mechanisms and clinical relevance of Zinc-finger protein ZNF468 in lymphangiogenesis and lymphatic metastasis in ESCC. METHODS: Immunohistochemistry, Western blot, Kaplan-Meier plotter analysis and Gene Set Enrichment Analysis were preformed to detect the association of ZNF468 with lymphangiogenesis and poor prognosis in ESCC patients. Foot-pads lymph node metastasis model, tube formation assay, 3D-culture assay and invasion assay were preformed to verify the effect of ZNF468 on lymphangiogenesis and lymph node metastasis. CUT&Tag analysis, immunoprecipitation and mass spectrometry analysis and ChIP-PCR assay were preformed to study the molecular mechanisms of ZNF468 in lymphangiogenesis. RESULTS: We found that ectopic expression of ZNF468 was correlated with higher microlymphatic vessel density in ESCC tissues, leading to poorer prognosis of ESCC patients. ZNF468 enhanced the capacity of lymphangiogenesis and promoted lymphatic metastasis in ESCC both in vitro and in vivo. However, silencing ZNF468 reversed these phenotypes in ESCC. Mechanically, we demonstrated that ZNF468 recruits the histone modification factors (PRMT1/HAT1) to increase the levels of H4R2me2a and H3K9ac, which then leads to the recruitment of the transcription initiation complex on the VEGF-C promoter, ultimately promoting the upregulation of VEGF-C transcription. Strikingly, the promoting effect of lymphatic metastasis induced by ZNF468 in ESCC was abrogated by targeting PRMT1 using Arginine methyltransferase inhibitor-1 or silencing VEGF-C. Furthermore, we found that the activation of PI3K/AKT and ERK1/2 signaling is required for ZNF468-medicated lymphatic metastasis in ESCC. Importantly, the clinical relevance between ZNF468 and VEGF-C were confirmed not only in ESCC samples and but also in multiple cancer types. CONCLUSION: Our results identified a precise mechanism underlying ZNF468-induced epigenetic upregulation of VEGF-C in facilitating lymphangiogenesis and lymph node metastasis of ESCC, which might provide a novel prognostic biomarker and potential therapeutic for ESCC patients.


Subject(s)
Epigenesis, Genetic , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic , Lymphangiogenesis , Lymphatic Metastasis , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Up-Regulation , Vascular Endothelial Growth Factor C , Lymphangiogenesis/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Humans , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation/genetics , Cell Line, Tumor , Phosphatidylinositol 3-Kinases/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Animals , MAP Kinase Signaling System/genetics , Female , Male , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Mice, Nude , Mice , Middle Aged , Mice, Inbred BALB C , Prognosis , Transcription Factors/metabolism , Transcription Factors/genetics , Signal Transduction/genetics
7.
J Clin Invest ; 134(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949026

ABSTRACT

Ubiquitination plays an essential role in protein stability, subcellular localization, and interactions. Crosstalk between different types of ubiquitination results in distinct biological outcomes for proteins. However, the role of ubiquitination-related crosstalk in lymph node (LN) metastasis and the key regulatory factors controlling this process have not been determined. Using high-throughput sequencing, we found that ubiquitin-conjugating enzyme E2 C (UBE2C) was overexpressed in bladder cancer (BCa) and was strongly associated with an unfavorable prognosis. Overexpression of UBE2C increased BCa lymphangiogenesis and promoted LN metastasis both in vitro and in vivo. Mechanistically, UBE2C mediated sodium-coupled neutral amino acid transporter 2 (SNAT2) monoubiquitination at lysine 59 to inhibit K63-linked polyubiquitination at lysine 33 of SNAT2. Crosstalk between monoubiquitination and K63-linked polyubiquitination increased SNAT2 membrane protein levels by suppressing epsin 1-mediated (EPN1-mediated) endocytosis. SNAT2 facilitated glutamine uptake and metabolism to promote VEGFC secretion, ultimately leading to lymphangiogenesis and LN metastasis in patients with BCa. Importantly, inhibition of UBE2C significantly attenuated BCa lymphangiogenesis in a patient-derived xenograft model. Our results reveal the mechanism by which UBE2C mediates crosstalk between the monoubiquitination and K63-linked polyubiquitination of SNAT2 to promote BCa metastasis and identify UBE2C as a promising target for treating LN-metastatic BCa.


Subject(s)
Lymphatic Metastasis , Ubiquitin-Conjugating Enzymes , Ubiquitination , Urinary Bladder Neoplasms , Animals , Female , Humans , Male , Mice , Amino Acid Transport System ASC , Cell Line, Tumor , Lymphangiogenesis/genetics , Minor Histocompatibility Antigens , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics
8.
Int J Mol Sci ; 25(14)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39063073

ABSTRACT

Secondary lymphedema is caused by damage to the lymphatic system from surgery, cancer treatment, infection, trauma, or obesity. This damage induces stresses such as oxidative stress and hypoxia in lymphatic tissue, impairing the lymphatic system. In response to damage, vascular endothelial growth factor C (VEGF-C) levels increase to induce lymphangiogenesis. Unfortunately, VEGF-C often fails to repair the lymphatic damage in lymphedema. The underlying mechanism contributing to lymphedema is not well understood. In this study, we found that surgery-induced tail lymphedema in a mouse model increased oxidative damage and cell death over 16 days. This corresponded with increased VEGF-C levels in mouse tail lymphedema tissue associated with macrophage infiltration. Similarly, in the plasma of patients with secondary lymphedema, we found a positive correlation between VEGF-C levels and redox imbalance. To determine the effect of oxidative stress in the presence or absence of VEGF-C, we found that hydrogen peroxide (H2O2) induced cell death in human dermal lymphatic endothelial cells (HDLECs), which was potentiated by VEGF-C. The cell death induced by VEGF-C and H2O2 in HDLECs was accompanied by increased reactive oxygen species (ROS) levels and a loss of mitochondrial membrane potential. Antioxidant pre-treatment rescued HDLECs from VEGF-C-induced cell death and decreased ROS under oxidative stress. As expected, VEGF-C increased the number of viable and proliferating HDLECs. However, upon H2O2 treatment, VEGF-C failed to increase either viable or proliferating cells. Since oxidative stress leads to DNA damage, we also determined whether VEGF-C treatment induces DNA damage in HDLECs undergoing oxidative stress. Indeed, DNA damage, detected in the form of gamma H2AX (γH2AX), was increased by VEGF-C under oxidative stress. The potentiation of oxidative stress damage induced by VEFG-C in HDLECs was associated with p53 activation. Finally, the inhibition of vascular endothelial growth factor receptor-3 (VEGFR-3) activation blocked VEGF-C-induced cell death following H2O2 treatment. These results indicate that VEGF-C further sensitizes lymphatic endothelial cells to oxidative stress by increasing ROS and DNA damage, potentially compromising lymphangiogenesis.


Subject(s)
Apoptosis , DNA Damage , Endothelial Cells , Hydrogen Peroxide , Lymphedema , Mitochondria , Oxidative Stress , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor C/metabolism , Oxidative Stress/drug effects , Animals , Humans , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Lymphedema/metabolism , Lymphedema/pathology , Lymphedema/etiology , Mitochondria/metabolism , Mitochondria/drug effects , Mice , Apoptosis/drug effects , Hydrogen Peroxide/pharmacology , Reactive Oxygen Species/metabolism , Lymphangiogenesis/drug effects , Female
9.
Eur J Oral Sci ; 132(4): e13006, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38989803

ABSTRACT

Lymphatics are involved in the resolution of inflammation and wound healing, but their role in the oral wound healing process after tooth extraction has never been investigated. We therefore sought to evaluate the healing process following the extraction of maxillary molars in two transgenic mouse models: K14-VEGFR3-Ig mice, which lack initial mucosal lymphatic vessels, and K14-VEGFC mice, which have hyperplastic mucosal lymphatics. Maxillary molars were extracted from both transgenic mouse types and their corresponding wild-type (WT) controls. Mucosal and alveolar bone healing were evaluated. A delayed epithelialization and bone regeneration were observed in K14-VEGFR3-Ig mice compared with their WT littermates. The hampered wound closure was accompanied by decreased levels of epidermal growth factor (EGF) and persistent inflammation, characterized by infiltrates of immune cells and elevated levels of pro-inflammatory markers in the wounds. Hyperplastic mucosal lymphatics did not enhance the healing process after tooth extraction in K14-VEGFC mice. The findings indicate that initial mucosal lymphatics play a major role in the initial phase of the oral wound healing process.


Subject(s)
Lymphatic Vessels , Mice, Transgenic , Tooth Extraction , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3 , Wound Healing , Animals , Wound Healing/physiology , Mice , Vascular Endothelial Growth Factor C/metabolism , Lymphatic Vessels/pathology , Vascular Endothelial Growth Factor Receptor-3/metabolism , Molar , Mouth Mucosa/pathology , Bone Regeneration/physiology , Epidermal Growth Factor/analysis , Epidermal Growth Factor/metabolism , Re-Epithelialization
10.
Aesthet Surg J ; 44(10): NP737-NP748, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-38870037

ABSTRACT

BACKGROUND: Persistent macrophage infiltration may lead to adverse consequences, such as calcifications and nodules in fat grafts. Lymphatic vessels, which transport inflammatory cells, are involved in regulating inflammatory responses. Less is known, however, about lymphatic vessels after fat grafting. OBJECTIVES: The aim of this study was to explore the regulation of fat graft survival by lymphatic vessels. METHODS: A common adipose graft model was constructed to assess the processes responsible for changes in the number of lymphatic vessels in grafts. Adipose tissue samples from C57/BL6 mice and green fluorescent protein-expressing mice were cross-grafted to determine the source of lymphatic vessels. The number of lymphatic vessels in the grafts was increased by treatment with vascular endothelial growth factor C, and the effects of this increase on fat grafting were evaluated. RESULTS: The number of lymphatic vessels was greater in postgrafted fat than in inguinal fat before transplantation, with lymphatic vessels in these grafts gradually transitioning from donor to recipient sources. Lymphatic vessels grew more slowly than blood vessels during early stages of grafting; during later stages, however, the number of blood vessels declined markedly, with more lymphatic vessels than blood vessels being observed 60 days after grafting. Vascular endothelial growth factor C treatment increased graft lymphatics and distant volume retention, while reducing fibrosis and oil sacs. Lymphatic vessels acted as drainage channels for macrophages, with the degree of sustained macrophage infiltration decreasing with increases in the number of lymphatic vessels. CONCLUSIONS: Increasing the number of lymphatic vessels is beneficial for fat graft survival, which may be related to a reduction in prolonged macrophage infiltration.


Subject(s)
Adipose Tissue , Graft Survival , Lymphatic Vessels , Macrophages , Mice, Inbred C57BL , Animals , Macrophages/immunology , Macrophages/metabolism , Adipose Tissue/transplantation , Mice , Vascular Endothelial Growth Factor C/metabolism , Models, Animal , Mice, Transgenic , Male , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lymphangiogenesis/physiology
11.
J Cell Mol Med ; 28(11): e18462, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38847478

ABSTRACT

Osteosarcoma (OS) is the most common primary malignant bone tumour in children and young adults. Account for 80% of all OS cases, conventional OS are characterized by the presence of osteoblastic, chondroblastic and fibroblastic cell types. Despite this heterogeneity, therapeutic treatment and prognosis of OS are essentially the same for all OS subtypes. Here, we report that DEC2, a transcriptional repressor, is expressed at higher levels in chondroblastic OS compared with osteoblastic OS. This difference suggests that DEC2 is disproportionately involved in the progression of chondroblastic OS, and thus, DEC2 may represent a possible molecular target for treating this type of OS. In the human chondroblastic-like OS cell line MNNG/HOS, we found that overexpression of DEC2 affects the proliferation of the cells by activating the VEGFC/VEGFR2 signalling pathway. Enhanced expression of DEC2 increased VEGFR2 expression, as well as increased the phosphorylation levels at sites Y951 and Y1175 of VEGFR2. On the one hand, activation of VEGFR2Y1175 enhanced cell proliferation through VEGFR2Y1175-PLCγ1-PKC-SPHK-MEK-ERK signalling. On the other hand, activation of VEGFR2Y951 decreased mitochondria-dependent apoptosis rate through VEGFR2Y951-VARP-PI3K-AKT signalling. Activation of these two signalling pathways resulted in enhanced progression of chondroblastic OS. In conclusion, DEC2 plays a pivotal role in cell proliferation and apoptosis-resistance in chondroblastic OS via the VEGFC/VEGFR2 signalling pathway. These findings lay the groundwork for developing focused treatments that target specific types of OS.


Subject(s)
Bone Neoplasms , Cell Proliferation , Gene Expression Regulation, Neoplastic , Osteosarcoma , Signal Transduction , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-2 , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Cell Line, Tumor , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Animals , Apoptosis/genetics , Phosphorylation
12.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38742432

ABSTRACT

Development of the vascular system is regulated by multiple signaling pathways mediated by receptor tyrosine kinases. Among them, angiopoietin (Ang)/Tie signaling regulates lymphatic and blood vessel development in mammals. Of the two Tie receptors, Tie2 is well known as a key mediator of Ang/Tie signaling, but, unexpectedly, recent studies have revealed that the Tie2 locus has been lost in many vertebrate species, whereas the Tie1 gene is more commonly present. However, Tie1-driven signaling pathways, including ligands and cellular functions, are not well understood. Here, we performed comprehensive mutant analyses of angiopoietins and Tie receptors in zebrafish and found that only angpt1 and tie1 mutants show defects in trunk lymphatic vessel development. Among zebrafish angiopoietins, only Angpt1 binds to Tie1 as a ligand. We indirectly monitored Ang1/Tie1 signaling and detected Tie1 activation in sprouting endothelial cells, where Tie1 inhibits nuclear import of EGFP-Foxo1a. Angpt1/Tie1 signaling functions in endothelial cell migration and proliferation, and in lymphatic specification during early lymphangiogenesis, at least in part by modulating Vegfc/Vegfr3 signaling. Thus, we show that Angpt1/Tie1 signaling constitutes an essential signaling pathway for lymphatic development in zebrafish.


Subject(s)
Angiopoietin-1 , Lymphangiogenesis , Receptor, TIE-1 , Signal Transduction , Zebrafish Proteins , Zebrafish , Animals , Angiopoietin-1/metabolism , Angiopoietin-1/genetics , Cell Movement , Cell Proliferation , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Lymphangiogenesis/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/embryology , Mutation/genetics , Protein Binding , Receptor, TIE-1/metabolism , Receptor, TIE-1/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
13.
Invest Ophthalmol Vis Sci ; 65(5): 32, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38771570

ABSTRACT

Purpose: To evaluate VEGF-C-induced lymphoproliferation in conjunction with 5-fluorouracil (5-FU) antimetabolite treatment in a rabbit glaucoma filtration surgery (GFS) model. Methods: Thirty-two rabbits underwent GFS and were assigned to four groups (n = 8 each) defined by subconjunctival drug treatment: (a) VEGF-C combined with 5-FU, (b) 5-FU, (c) VEGF-C, (d) and control. Bleb survival, bleb measurements, and IOP were evaluated over 30 days. At the end, histology and anterior segment OCT were performed on some eyes. mRNA was isolated from the remaining eyes for RT-PCR evaluation of vessel-specific markers (lymphatics, podoplanin and LYVE-1; and blood vessels, CD31). Results: Qualitatively and quantitatively, VEGF-C combined with 5-FU resulted in blebs which were posteriorly longer and wider than the other conditions: vs. 5-FU (P = 0.043 for longer, P = 0.046 for wider), vs. VEGF-C (P < 0.001, P < 0.001) and vs. control (P < 0.001, P < 0.001). After 30 days, the VEGF-C combined with 5-FU condition resulted in longer bleb survival compared with 5-FU (P = 0.025), VEGF-C (P < 0.001), and control (P < 0.001). Only the VEGF-C combined with 5-FU condition showed a negative correlation between IOP and time that was statistically significant (r = -0.533; P = 0.034). Anterior segment OCT and histology demonstrated larger blebs for the VEGF-C combined with 5-FU condition. Only conditions including VEGF-C led to increased expression of lymphatic markers (LYVE-1, P < 0.001-0.008 and podoplanin, P = 0.002-0.011). Expression of CD31 was not different between the groups (P = 0.978). Conclusions: Adding VEGF-C lymphoproliferation to standard antimetabolite treatment improved rabbit GFS success and may suggest a future strategy to improve human GFSs.


Subject(s)
Disease Models, Animal , Fluorouracil , Glaucoma , Intraocular Pressure , Trabeculectomy , Vascular Endothelial Growth Factor C , Animals , Rabbits , Fluorouracil/therapeutic use , Fluorouracil/pharmacology , Glaucoma/surgery , Glaucoma/physiopathology , Glaucoma/drug therapy , Vascular Endothelial Growth Factor C/metabolism , Trabeculectomy/methods , Intraocular Pressure/physiology , Antimetabolites/pharmacology , Antimetabolites/therapeutic use , Tomography, Optical Coherence , Conjunctiva , RNA, Messenger/genetics
14.
Sci Rep ; 14(1): 9976, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38693148

ABSTRACT

Inflammation and fibrosis often occur in the kidney after acute injury, resulting in chronic kidney disease and consequent renal failure. Recent studies have indicated that lymphangiogenesis can drive renal inflammation and fibrosis in injured kidneys. However, whether and how this pathogenesis affects the contralateral kidney remain largely unknown. In our study, we uncovered a mechanism by which the contralateral kidney responded to injury. We found that the activation of mineralocorticoid receptors and the increase in vascular endothelial growth factor C in the contralateral kidney after unilateral ureteral obstruction could promote lymphangiogenesis. Furthermore, mineralocorticoid receptor activation in lymphatic endothelial cells resulted in the secretion of myofibroblast markers, thereby contributing to renal fibrosis. We observed that this process could be attenuated by administering the mineralocorticoid receptor blocker eplerenone, which, prevented the development of fibrotic injury in the contralateral kidneys of rats with unilateral ureteral obstruction. These findings offer valuable insights into the intricate mechanisms underlying kidney injury and may have implications for the development of therapeutic strategies to mitigate renal fibrosis in the context of kidney disease.


Subject(s)
Eplerenone , Fibrosis , Kidney , Lymphangiogenesis , Mineralocorticoid Receptor Antagonists , Ureteral Obstruction , Animals , Eplerenone/pharmacology , Lymphangiogenesis/drug effects , Rats , Fibrosis/drug therapy , Kidney/metabolism , Kidney/drug effects , Kidney/pathology , Ureteral Obstruction/drug therapy , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Ureteral Obstruction/complications , Mineralocorticoid Receptor Antagonists/pharmacology , Male , Receptors, Mineralocorticoid/metabolism , Spironolactone/analogs & derivatives , Spironolactone/pharmacology , Vascular Endothelial Growth Factor C/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Rats, Sprague-Dawley , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Myofibroblasts/pathology
15.
Ecotoxicol Environ Saf ; 278: 116444, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38728943

ABSTRACT

Silicosis is a disease characterized by lung inflammation and fibrosis caused by long-term inhalation of free silicon dioxide (SiO2). Recent studies have found that a large number of lymphatic hyperplasia occurs during the occurrence and development of silicosis. miRNAs play an important role in lymphangiogenesis. However, the regulation and mechanism of miRNAs on lymphangiogenesis in silicosis remain unclear. In this study, lymphangiogenesis was observed in silicosis rats, and VEGF-C-targeted miRNAs were screened, and the effect of miRNAs on the formation of human lymphatic endothelial cells (HLECs) tubular structure was investigated in vitro. The results showed that SiO2 promoted the expressions of Collagen Ι and α-SMA, TNF-α, IL-6 and VEGF-C increased first and then decreased, and promoted the formation of lymphatic vessels. Bioinformatics methods screened miR-455-3p for targeted binding to VEGF-C, and dual luciferase reporter genes confirmed VEGF-C as the target gene of miR-455-3p, and miR-455-3p was down-regulated in the lung tissue of silicosis rats. Transfection of miR-455-3p Inhibitors down-regulated the expression level of miR-455-3p and up-regulated the expression levels of VEGF-C and VEGFR-3 in HLECs, enhanced migration ability and increased tube formation. Transfection of miR-455-3p Mimics showed an opposite trend. These results suggest that miR-455-3p further regulates the tubular structure formation of HLECs by regulating VEGF-C/VEGFR3. Therefore, targeting miR-455-3p may provide a new therapeutic strategy for SiO2-induced silicosis injury.


Subject(s)
Lymphangiogenesis , MicroRNAs , Silicosis , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3 , Animals , Humans , Male , Rats , Endothelial Cells/drug effects , Lymphangiogenesis/drug effects , MicroRNAs/genetics , Rats, Sprague-Dawley , Silicon Dioxide/toxicity , Silicosis/pathology , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
16.
PLoS One ; 19(4): e0301416, 2024.
Article in English | MEDLINE | ID: mdl-38603681

ABSTRACT

INTRODUCTION: Portal hypertension (PH) drives the progression of liver cirrhosis to decompensation and death. Hepatic venous pressure gradient (HVPG) measurement is the standard of PH quantification, and HVPG≥10 mmHg defines clinically significant PH (CSPH). We performed proteomics-based serum profiling to search for a proteomic signature of CSPH in patients with compensated advanced chronic liver disease (cACLD). MATERIALS AND METHODS: Consecutive patients with histologically confirmed cACLD and results of HVPG measurements were prospectively included. Serum samples were pooled according to the presence/absence of CSPH and analysed by liquid chromatography-mass spectrometry. Gene set enrichment analysis was performed, followed by comprehensive literature review for proteins identified with the most striking difference between the groups. RESULTS: We included 48 patients (30 with, and 18 without CSPH). Protein CD44, involved in the inflammatory response, vascular endothelial growth factor C (VEGF-C) and lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), both involved in lymphangiogenesis were found solely in the CSPH group. Although identified in both groups, proteins involved in neutrophil extracellular traps (NET) formation, as well as tenascin C, autotaxin and nephronectin which mediate vascular contractility and lymphangiogenesis were more abundant in CSPH. DISCUSSION AND CONCLUSION: We propose that altered inflammatory response, including NET formation, vascular contractility and formation of new lymph vessels are key steps in PH development. Proteins such as CD44, VEGF-C, LYVE-1, tenascin C, Plasminogen activator inhibitor 1, Nephronectin, Bactericidal permeability-increasing protein, Autotaxin, Myeloperoxidase and a disintegrin and metalloproteinase with thrombospondin motifs-like protein 4 might be considered for further validation as potential therapeutic targets and candidate biomarkers of CSPH in cACLD.


Subject(s)
Elasticity Imaging Techniques , Hypertension, Portal , Humans , Vascular Endothelial Growth Factor C , Tenascin , Proteomics , Liver , Liver Cirrhosis , Portal Pressure
17.
J Ovarian Res ; 17(1): 77, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594780

ABSTRACT

PURPOSE: Our explorative study assessed a panel of molecules for their association with epithelial ovarian carcinomas and their prognostic implications. The panel included tissue expression of VEGF-C, COX-2, Ki-67 and eNOS alongside plasma levels of VEGF-C and nitric oxide. METHODS: 130 cases were enrolled in the study. Plasma levels were quantified by ELISA and tissue expressions were scored by immunohistochemistry. The Chi square and Fischer's exact test were applied to examine the impact of markers on clinicopathological factors. Non-parametric Spearman's rank correlation test was applied to define the association among test factors. RESULTS: Plasma VEGF-C levels and COX-2 tissue expression strongly predicted recurrence and poor prognosis (< 0.001). Tissue Ki-67 was strongly indicative of late-stage disease (< 0.001). The aforementioned markers significantly associated with clinicopathological factors. Nuclear staining of VEGF-C was intriguing and was observed to correlate with high grade-stage malignancies, highly elevated plasma VEGF-C, and with recurrence. eNOS tissue expression showed no significant impact while nitric oxide associated positively with ascites levels. Tissue expression of VEGF-C did not associate significantly with poor prognosis although the expression was highly upregulated in most of the cases. CONCLUSION: Plasma VEGF-C holds immense promise as a prognostic marker and the nuclear staining of VEGF-C seems to have some significant implication in molecular carcinogenesis and is a novel finding that commands further robust scrutiny. We present a first such study that assesses a set of biomarkers for prognostic implications in clinical management of epithelial ovarian carcinomas in a pan-Indian (Asian) population.


Subject(s)
Ovarian Neoplasms , Humans , Female , Carcinoma, Ovarian Epithelial/pathology , Prognosis , Ovarian Neoplasms/pathology , Cyclooxygenase 2/metabolism , Vascular Endothelial Growth Factor C , Ki-67 Antigen , Nitric Oxide , Neoplasm Staging , Biomarkers, Tumor/metabolism
18.
Exp Eye Res ; 243: 109891, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615832

ABSTRACT

The aim of this study is to investigate the relationship between age-related macular degeneration (AMD) and lymphangiogenesis biomarkers, namely LYVE-1, Podoplanin, VEGF-C, VEGFR-2 and VEGFR-3. This prospective and interventional study includes 30 patients with AMD which may be dry or wet type and 30 controls for whom vitrectomy and phacoemulsification was indicated due to additional pathologies (epiretinal membrane, macular hole, retinal detachment, and cataract). 0.1-0,2 ml of aqueous humor and 0.5-1 ml of vitreous sample was taken during the operations. Before the operations 1 tube serum was also taken. All the lymphangiogenesis biomarkers in the study are examined by ELISA method. LYVE-1 (p = 0.001) and Podoplanin (p = 0.004) levels in the vitreous for the patient group are found to be significantly lower than the control group. Serum (p = 0.019), vitreous (p = 0.001), aqueous (p < 0.001) levels of VEGF-C for the patient group are significantly higher than the control group. VEGF-C/VEGFR-2 (p < 0.001), VEGF-C/VEGFR-3 (p < 0.001) ratios in the vitreous for the patient group are found to be significantly higher than the control group. Especially in wet AMD patients, LYVE-1 level is significantly lower in the vitreous (p = 0.002) and aqueous (p = 0.002) than the control group. In addition, Podoplanin level is observed as significantly lower in the vitreous (p = 0.014) and serum (p = 0.002) in comparison to control group. In the wet AMD group, VEGF-C level in the vitreous (p < 0.001), aqueous (p < 0.001) and serum (p = 0.001) is higher than the control group. The result of this study indicates a valid relationship between the weakening of lymphangiogenesis and the pathophysiology of AMD, especially for the wet type. It is observed that the levels of receptors that bind VEGF-C (VEGFR-2 and VEGFR-3) do not increase at the same rate as VEGF-C to compensate for the increase in VEGF-C. The absence of an increase in VEGFR-3, which is especially necessary for lymphangiogenesis, also suggests that lymphangiogenesis is weakened or decreased in AMD. In the future interventional studies with larger series, examination of lymphangiogenic biomarkers in inflammatory retinal diseases and glaucoma may reveal unexplored details.


Subject(s)
Aqueous Humor , Biomarkers , Enzyme-Linked Immunosorbent Assay , Lymphangiogenesis , Membrane Glycoproteins , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3 , Vesicular Transport Proteins , Vitreous Body , Humans , Male , Female , Biomarkers/metabolism , Biomarkers/blood , Prospective Studies , Aged , Vascular Endothelial Growth Factor Receptor-3/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/blood , Aqueous Humor/metabolism , Vitreous Body/metabolism , Vitreous Body/pathology , Membrane Glycoproteins/metabolism , Vesicular Transport Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Middle Aged , Aged, 80 and over , Macular Degeneration/metabolism , Macular Degeneration/diagnosis , Wet Macular Degeneration/metabolism , Wet Macular Degeneration/diagnosis
19.
Cell Rep ; 43(4): 114049, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38573853

ABSTRACT

Heterotopic ossification (HO) is a challenging condition that occurs after musculoskeletal injury and is characterized by the formation of bone in non-skeletal tissues. While the effect of HO on blood vessels is well established, little is known about its impact on lymphatic vessels. Here, we use a mouse model of traumatic HO to investigate the relationship between HO and lymphatic vessels. We show that injury triggers lymphangiogenesis at the injury site, which is associated with elevated vascular endothelial growth factor C (VEGF-C) levels. Through single-cell transcriptomic analyses, we identify mesenchymal progenitor cells and tenocytes as sources of Vegfc. We demonstrate by lineage tracing that Vegfc-expressing cells undergo osteochondral differentiation and contribute to the formation of HO. Last, we show that Vegfc haploinsufficiency results in a nearly 50% reduction in lymphangiogenesis and HO formation. These findings shed light on the complex mechanisms underlying HO formation and its impact on lymphatic vessels.


Subject(s)
Lymphangiogenesis , Mesenchymal Stem Cells , Ossification, Heterotopic , Vascular Endothelial Growth Factor C , Animals , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Ossification, Heterotopic/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Mice , Mesenchymal Stem Cells/metabolism , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Cell Differentiation , Tenocytes/metabolism , Osteogenesis , Haploinsufficiency , Mice, Inbred C57BL , Disease Models, Animal , Male
20.
J Exp Med ; 221(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38442272

ABSTRACT

Meningeal lymphatic vessels (MLVs) promote tissue clearance and immune surveillance in the central nervous system (CNS). Vascular endothelial growth factor-C (VEGF-C) regulates MLV development and maintenance and has therapeutic potential for treating neurological disorders. Herein, we investigated the effects of VEGF-C overexpression on brain fluid drainage and ischemic stroke outcomes in mice. Intracerebrospinal administration of an adeno-associated virus expressing mouse full-length VEGF-C (AAV-mVEGF-C) increased CSF drainage to the deep cervical lymph nodes (dCLNs) by enhancing lymphatic growth and upregulated neuroprotective signaling pathways identified by single nuclei RNA sequencing of brain cells. In a mouse model of ischemic stroke, AAV-mVEGF-C pretreatment reduced stroke injury and ameliorated motor performances in the subacute stage, associated with mitigated microglia-mediated inflammation and increased BDNF signaling in brain cells. Neuroprotective effects of VEGF-C were lost upon cauterization of the dCLN afferent lymphatics and not mimicked by acute post-stroke VEGF-C injection. We conclude that VEGF-C prophylaxis promotes multiple vascular, immune, and neural responses that culminate in a protection against neurological damage in acute ischemic stroke.


Subject(s)
Ischemic Stroke , Stroke , Animals , Mice , Vascular Endothelial Growth Factor C , Neuroinflammatory Diseases , Drainage
SELECTION OF CITATIONS
SEARCH DETAIL