Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Nature ; 628(8006): 204-211, 2024 Apr.
Article En | MEDLINE | ID: mdl-38418880

The eye, an anatomical extension of the central nervous system (CNS), exhibits many molecular and cellular parallels to the brain. Emerging research demonstrates that changes in the brain are often reflected in the eye, particularly in the retina1. Still, the possibility of an immunological nexus between the posterior eye and the rest of the CNS tissues remains unexplored. Here, studying immune responses to herpes simplex virus in the brain, we observed that intravitreal immunization protects mice against intracranial viral challenge. This protection extended to bacteria and even tumours, allowing therapeutic immune responses against glioblastoma through intravitreal immunization. We further show that the anterior and posterior compartments of the eye have distinct lymphatic drainage systems, with the latter draining to the deep cervical lymph nodes through lymphatic vasculature in the optic nerve sheath. This posterior lymphatic drainage, like that of meningeal lymphatics, could be modulated by the lymphatic stimulator VEGFC. Conversely, we show that inhibition of lymphatic signalling on the optic nerve could overcome a major limitation in gene therapy by diminishing the immune response to adeno-associated virus and ensuring continued efficacy after multiple doses. These results reveal a shared lymphatic circuit able to mount a unified immune response between the posterior eye and the brain, highlighting an understudied immunological feature of the eye and opening up the potential for new therapeutic strategies in ocular and CNS diseases.


Brain , Eye , Lymphatic System , Animals , Female , Humans , Male , Mice , Rabbits , Bacteria/immunology , Brain/anatomy & histology , Brain/immunology , Dependovirus/immunology , Eye/anatomy & histology , Eye/immunology , Glioblastoma/immunology , Herpesvirus 2, Human/immunology , Intravitreal Injections , Lymphatic System/anatomy & histology , Lymphatic System/immunology , Lymphatic Vessels/anatomy & histology , Lymphatic Vessels/immunology , Macaca mulatta , Meninges/immunology , Optic Nerve/immunology , Swine , Zebrafish , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/pharmacology
2.
Stem Cell Res Ther ; 13(1): 448, 2022 09 05.
Article En | MEDLINE | ID: mdl-36064450

BACKGROUND: Adipose-derived stem cells (ADSCs) have provided promising applications for Crohn's disease (CD). However, the practical efficacy of ADSCs remains controversial, and their mechanism is still unclear. Based on the pathogenesis of dysregulated immune responses and abnormal lymphatic alterations in CD, vascular endothelial growth factor-C (VEGF-C) is thought to be a favourable growth factor to optimize ADSCs. We aimed to investigate the efficacy of VEGF-C-stimulated ADSCs and their dual mechanisms in both inhibiting inflammation "IN" and promoting inflammation "OUT" in the intestine. METHODS: Human stem cells isolated from adipose tissues were identified, pretreated with or without 100 ng/ml VEGF-C and analysed for the secretion of cell culture supernatants in vitro. Lymphatic endothelial cells (LECs) were treated with ADSCs-conditioned medium or co-cultured with ADSCs and VEGF-C stimulated ADSCs. Changes in LECs transmigration, and VEGF-C/VEGFR-3 mRNA levels were assessed by transwell chamber assay and qRT-PCR. ADSCs and VEGF-C-stimulated ADSCs were intraperitoneally injected into mice with TNBS-induced chronic colitis. ADSCs homing and lymphatic vessel density (LVD) were evaluated by immunofluorescence staining. Lymphatic drainage was assessed using Evans blue. Cytokines and growth factors expression was detected respectively by ELISA and qRT-PCR. The protein levels of VEGF-C/VEGFR-3-mediated downstream signals and the NF-κB pathway were assayed by western blot. Faecal microbiota was measured by 16S rRNA sequencing. RESULTS: ADSCs stimulated with VEGF-C released higher levels of growth factors (VEGF-C, TGF-ß1, and FGF-2) and lower expression of cytokines (IFN-γ and IL-6) in cell supernatants than ADSCs in vitro (all P < 0.05). Secretome released by VEGF-C stimulated ADSCs exhibited a stronger LEC migratory capability and led to elevated VEGF-C/VEGFR-3 expression, but these effects were markedly attenuated by VEGFR-3 inhibitor. VEGF-C-stimulated ADSCs homing to the inflamed colon and mesenteric lymph nodes (MLNs) can exert stronger efficacy in improving colitis symptoms, reducing inflammatory cell infiltration, and significantly enhancing lymphatic drainage. The mRNA levels and protein concentrations of anti-inflammatory cytokines and growth factors were markedly increased with decreased proinflammatory cytokines in the mice treated with VEGF-C-stimulated ADSCs. Systemic administration of VEGF-C-stimulated ADSCs upregulated the colonic VEGF-C/VEGFR-3 pathway and activated downstream AKT and ERK phosphorylation signalling, accompanied by decreased NF-κB p65 expression. A higher abundance of faecal p-Bacteroidetes and lower p-Firmicutes were detected in mice treated with VEGF-C-stimulated ADSCs (all P < 0.05). CONCLUSION: VEGF-C-stimulated ADSCs improve chronic intestinal inflammation by promoting lymphatic drainage and enhancing paracrine signalling via activation of VEGF-C/VEGFR-3-mediated signalling and inhibition of the NF-κB pathway. Our study may provide a new insight into optimizing ADSCs treatment and investigating potential mechanisms in CD.


Colitis , Mesenchymal Stem Cells , NF-kappa B , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3 , Angiogenesis Inhibitors , Animals , Colitis/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/therapy , Mesenchymal Stem Cells/immunology , Mice , NF-kappa B/immunology , RNA, Messenger/metabolism , RNA, Ribosomal, 16S/metabolism , Secretome/immunology , Stem Cells/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/pharmacology , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
3.
Toxins (Basel) ; 13(8)2021 08 10.
Article En | MEDLINE | ID: mdl-34437431

Discriminating Polistes dominula and Vespula spp. venom allergy is of growing importance worldwide, as systemic reactions to either species' sting can lead to severe outcomes. Administering the correct allergen-specific immunotherapy is therefore a prerequisite to ensure the safety and health of venom-allergic patients. Component-resolved diagnostics of Hymenoptera venom allergy might be improved by adding additional allergens to the diagnostic allergen panel. Therefore, three potential new allergens from P. dominula venom-immune responsive protein 30 (IRP30), vascular endothelial growth factor C (VEGF C) and phospholipase A2 (PLA2)-were cloned, recombinantly produced and biochemically characterized. Sera sIgE titers of Hymenoptera venom-allergic patients were measured in vitro to assess the allergenicity and potential cross-reactivity of the venom proteins. IRP30 and VEGF C were classified as minor allergens, as sensitization rates lay around 20-40%. About 50% of P. dominula venom-allergic patients had measurable sIgE titers directed against PLA2 from P. dominula venom. Interestingly, PLA2 was unable to activate basophils of allergic patients, questioning its role in the context of clinically relevant sensitization. Although the obtained results hint to a questionable benefit of the characterized P. dominula venom proteins for improved diagnosis of venom-allergic patients, they can contribute to a deeper understanding of the molecular mechanisms of Hymenoptera venoms and to the identification of factors that determine the allergenic potential of proteins.


Allergens , Arthropod Venoms , Hypersensitivity , Insect Proteins , Allergens/genetics , Allergens/immunology , Animals , Arthropod Venoms/chemistry , Arthropod Venoms/immunology , Basophils/immunology , Humans , Hypersensitivity/blood , Hypersensitivity/diagnosis , Hypersensitivity/immunology , Immunoglobulin E/blood , Insect Proteins/genetics , Insect Proteins/immunology , Phospholipases A2/genetics , Phospholipases A2/immunology , Recombinant Proteins/immunology , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/immunology , Wasps
4.
Cells ; 10(5)2021 05 17.
Article En | MEDLINE | ID: mdl-34067671

Despite improvement during the last ten years in the longevity of patients with metastatic clear cell renal cell carcinoma (mccRCC) the disease remains incurable. Hence, new therapeutic strategies are urgently needed. Relapse following anti-angiogenic treatment depends on the over-expression of vascular endothelial growth factor C (VEGFC), one of the main drivers of lymphangiogenesis. Therefore, we developed specific mouse monoclonal antibodies and evaluated their therapeutic efficacy in vitro and in vivo. Immunization of mice with the domain of VEGFC that stimulates the VEGF receptor 3 (VEGFR3) led to the selection of one hybridoma producing specific anti-VEGFC monoclonal antibodies. The selected 1E9 antibodies were sequenced, and the corresponding variable light and heavy chains were subcloned into expression vectors in frame with sequences encoding the human IgG1 constant heavy and light chains. CHO cells were stably transfected and cloned to produce chimeric antibodies. These antibodies inhibited the activation of VEGFR3 signaling, and therefore the proliferation and migration of VEGFC-stimulated endothelial cells. Moreover, they inhibited the proliferation of VEGFC-expressing renal cancer cells through NRP2 signaling. 1E9 antibodies inhibited the growth of experimental RCC, and their therapeutic efficacy was enhanced by the anti-VEGF antibody bevacizumab. Hence, our results suggest that targeting VEGFC could have a relevant therapeutic impact on mccRCC that relapse following anti-angiogenic treatment.


Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Renal Cell/drug therapy , Endothelial Cells/drug effects , Kidney Neoplasms/drug therapy , Neovascularization, Pathologic , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Mice, Nude , Signal Transduction , Tumor Burden/drug effects , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 117(41): 25859-25868, 2020 10 13.
Article En | MEDLINE | ID: mdl-33004630

Endometriosis is a highly prevalent gynecological disease with severe negative impacts on life quality and financial burden. Unfortunately, there is no cure for this disease, which highlights the need for further investigation about the pathophysiology of this disease to provide clues for developing novel therapeutic regimens. Herein, we identified that vascular endothelial growth factor (VEGF)-C, a potent lymphangiogenic factor, is up-regulated in endometriotic cells and contributes to increased lymphangiogenesis. Bioinformatic analysis and molecular biological characterization revealed that VEGF-C is negatively regulated by an orphan nuclear receptor, chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII). Further studies demonstrated that proinflammatory cytokines, via suppression of COUP-TFII level, induce VEGF-C overexpression. More importantly, we show that functional VEGF-C is transported by extracellular vesicles (EVs) to enhance the lymphangiogenic ability of lymphatic endothelial cells. Autotransplanted mouse model of endometriosis showed lenvatinib treatment abrogated the increased lymphatic vessels development in the endometriotic lesion, enlarged retroperitoneal lymph nodes, and immune cells infiltration, indicating that blocking VEGF-C signaling can reduce local chronic inflammation and concomitantly endometriosis development. Evaluation of EV-transmitted VEGF-C from patients' sera demonstrates it is a reliable noninvasive way for clinical diagnosis. Taken together, we identify the vicious cycle of inflammation, COUP-TFII, VEGF-C, and lymphangiogenesis in the endometriotic microenvironment, which opens up new horizons in understanding the pathophysiology of endometriosis. VEGF-C not only can serve as a diagnostic biomarker but also a molecular target for developing therapeutic regimens.


Endometriosis/immunology , Extracellular Vesicles/immunology , Immune System/immunology , Lymphangiogenesis , Vascular Endothelial Growth Factor C/immunology , Animals , COUP Transcription Factor II/genetics , COUP Transcription Factor II/immunology , Cytokines/genetics , Cytokines/immunology , Endometriosis/genetics , Endometriosis/physiopathology , Endothelial Cells/immunology , Extracellular Vesicles/genetics , Female , Humans , Lymphatic Vessels/immunology , Mice , Vascular Endothelial Growth Factor C/genetics
6.
Pharmacol Res Perspect ; 8(2): e00573, 2020 04.
Article En | MEDLINE | ID: mdl-32125783

A phage-derived human monoclonal antibody against VEGF-C was developed as a potential anti-tumor therapeutic and exhibited fast clearance in preclinical species, with notably faster clearance in serum than in plasma. The purpose of this work was to understand the factors contributing to its fast clearance. In vitro incubations in animal and human blood, plasma, and serum were conducted with radiolabeled anti-VEGF-C to determine potential protein and cell-based interactions with the antibody as well as any matrix-dependent recovery dependent upon the matrix. A tissue distribution study was conducted in mice with and without heparin infusion in order to identify a tissue sink and determine whether heparin could affect antibody recovery from serum and/or plasma. Incubation of radiolabeled anti-VEGF-C in human and animal blood, plasma, or serum revealed that the antibody formed a complex with an endogenous protein, likely VEGF-C. This complex was trapped within the blood clot during serum preparation from blood, but not within the blood cell pellet during plasma preparation. Low level heparin infusion in mice slowed down clot formation during serum preparation and allowed for better recovery of the radiolabeled antibody in serum. No tissue sink was found in mice. Thus, during this characterization, we determined that the blood sampling matrix greatly impacted the amount of antibody recovered in the samples, therefore, altering its derived pharmacokinetic parameters. Target biology should be considered when selecting appropriate sampling matrices for PK analysis.


Antibodies, Monoclonal/pharmacokinetics , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Animals , Antibodies, Monoclonal/blood , Artifacts , Blood Coagulation , Female , Humans , Macaca fascicularis , Mice, Nude , Rats, Sprague-Dawley , Tissue Distribution , Vascular Endothelial Growth Factor C/immunology
7.
Int Immunopharmacol ; 78: 106079, 2020 Jan.
Article En | MEDLINE | ID: mdl-31841756

Hereditary angioedema is a disabling, life-threatening condition caused by deficiency (type I) or dysfunction (type II) of the C1 inhibitor protein (C1-INH-HAE) leading to bradykinin accumulation and recurrent episodes of edema attack. Vascular leakage is a complex process sustained by the coordinated production of several permeabilizing factors including vascular endothelial growth factors (VEGFs), angiopoietins (ANGPTs) and phospholipase A2 enzymes (PLA2). We previously reported that patients with C1-INH-HAE in remission have increased plasma levels of VEGFs, ANGPTs and secreted PLA2. In this study, we sought to analyze plasma levels of these mediators in 15 patients with C1-INH-HAE during the acute attack compared to remission. Plasma concentrations of VEGF-A, VEGF-C and VEGF-D were not altered during attack compared to remission. Moreover, VEGF-D concentrations were not altered also in remission phase compared to controls. Concentrations of ANGPT1, a vascular stabilizer, were increased during attacks compared to symptoms-free periods, whereas ANGPT2 levels were not altered. The ANGPT2/ANGPT1 ratio was decreased during angioedema attacks. Platelet activating factor acetylhydrolase activity was increased in patients with C1-INH-HAE in remission compared to controls and was decreased during angioedema attacks. Our results emphasize the complexity by which several vasoactive mediators are involved not only in the pathophysiology of C1-INH-HAE, but also during angioedema attacks and its resolution.


1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , Angiopoietin-1/blood , Complement C1 Inhibitor Protein/metabolism , Hereditary Angioedema Types I and II/immunology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/immunology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Adolescent , Adult , Angiopoietin-1/immunology , Angiopoietin-1/metabolism , Angiopoietin-2/blood , Angiopoietin-2/immunology , Angiopoietin-2/metabolism , Bradykinin/immunology , Bradykinin/metabolism , Capillary Permeability/immunology , Case-Control Studies , Complement C1 Inhibitor Protein/genetics , Female , Healthy Volunteers , Hereditary Angioedema Types I and II/blood , Hereditary Angioedema Types I and II/genetics , Humans , Male , Middle Aged , Symptom Flare Up , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/blood , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/blood , Vascular Endothelial Growth Factor D/immunology , Vascular Endothelial Growth Factor D/metabolism , Young Adult
8.
Nat Commun ; 10(1): 5306, 2019 11 22.
Article En | MEDLINE | ID: mdl-31757960

After stroke, peripheral immune cells are activated and these systemic responses may amplify brain damage, but how the injured brain sends out signals to trigger systemic inflammation remains unclear. Here we show that a brain-to-cervical lymph node (CLN) pathway is involved. In rats subjected to focal cerebral ischemia, lymphatic endothelial cells proliferate and macrophages are rapidly activated in CLNs within 24 h, in part via VEGF-C/VEGFR3 signalling. Microarray analyses of isolated lymphatic endothelium from CLNs of ischemic mice confirm the activation of transmembrane tyrosine kinase pathways. Blockade of VEGFR3 reduces lymphatic endothelial activation, decreases pro-inflammatory macrophages, and reduces brain infarction. In vitro, VEGF-C/VEGFR3 signalling in lymphatic endothelial cells enhances inflammatory responses in co-cultured macrophages. Lastly, surgical removal of CLNs in mice significantly reduces infarction after focal cerebral ischemia. These findings suggest that modulating the brain-to-CLN pathway may offer therapeutic opportunities to ameliorate systemic inflammation and brain injury after stroke.


Brain Infarction/immunology , Brain Ischemia/immunology , Brain/immunology , Endothelium, Lymphatic/immunology , Lymph Nodes/immunology , Macrophages/immunology , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor Receptor-3/immunology , Animals , Brain/metabolism , Brain Infarction/metabolism , Brain Ischemia/metabolism , Cell Proliferation , Endothelial Cells , Endothelium, Lymphatic/metabolism , Inflammation , Lymph Nodes/metabolism , Lymphangiogenesis , Mice , Neck , Rats , Stroke/immunology , Stroke/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/metabolism
9.
Nat Commun ; 10(1): 229, 2019 01 16.
Article En | MEDLINE | ID: mdl-30651548

There are no conventional lymphatic vessels within the CNS parenchyma, although it has been hypothesized that lymphatics near the cribriform plate or dura maintain fluid homeostasis and immune surveillance during steady-state conditions. However, the role of these lymphatic vessels during neuroinflammation is not well understood. We report that lymphatic vessels near the cribriform plate undergo lymphangiogenesis in a VEGFC - VEGFR3 dependent manner during experimental autoimmune encephalomyelitis (EAE) and drain both CSF and cells that were once in the CNS parenchyma. Lymphangiogenesis also contributes to the drainage of CNS derived antigens that leads to antigen specific T cell proliferation in the draining lymph nodes during EAE. In contrast, meningeal lymphatics do not undergo lymphangiogenesis during EAE, suggesting heterogeneity in CNS lymphatics. We conclude that increased lymphangiogenesis near the cribriform plate can contribute to the management of neuroinflammation-induced fluid accumulation and immune surveillance.


Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphangiogenesis/immunology , Lymphatic Vessels/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens/immunology , Antigens/metabolism , Brain/diagnostic imaging , Cell Proliferation , Cerebrospinal Fluid/immunology , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Ethmoid Bone , Evans Blue/administration & dosage , Female , Humans , Immunologic Surveillance/immunology , Lymphatic Vessels/diagnostic imaging , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Pertussis Toxin/administration & dosage , Pertussis Toxin/immunology , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/immunology , Vascular Endothelial Growth Factor Receptor-3/metabolism
10.
JCI Insight ; 3(23)2018 12 06.
Article En | MEDLINE | ID: mdl-30518687

VEGF-C is an important mediator of lymphangiogenesis and has been shown to alleviate chronic inflammation in a variety of disease models. In this study, we investigated whether targeted delivery of VEGF-C to sites of inflammation and site-specific activation of lymphatic vessels would represent a clinically feasible strategy for treating chronic skin inflammation. To this end, we generated a fusion protein consisting of human VEGF-C fused to the F8 antibody (F8-VEGF-C), which is specific for the alternatively spliced, angiogenesis-marking extradomain A (EDA) of fibronectin. In two mouse models of psoriasis-like skin inflammation, mediated by transgenic VEGF-A overexpression or repeated application of imiquimod, intravenous treatment with F8-VEGF-C but not with untargeted VEGF-C significantly reduced ear skin edema and was as effective as the clinically used TNF-α receptor-Fc fusion protein (TNFR-Fc). Treatment with F8-VEGF-C led to a marked expansion of lymphatic vessels in the inflamed skin and significantly improved lymphatic drainage function. At the same time, treatment with F8-VEGF-C significantly reduced leukocyte numbers, including CD4+ and γδ T cells. In sum, our results reveal that targeted delivery of VEGF-C and site-specific induction of lymphatic vessels represent a potentially new and promising approach for the treatment of chronic inflammatory diseases.


Chronic Disease , Dermatitis/immunology , Inflammation/immunology , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , CD4-Positive T-Lymphocytes , Cell Proliferation , Dermatitis/drug therapy , Disease Models, Animal , Etanercept/immunology , Etanercept/metabolism , Etanercept/pharmacology , Female , Fibronectins , Inflammation/drug therapy , Lymphangiogenesis/immunology , Lymphatic Vessels , Mice , Mice, Inbred C57BL , Mice, Transgenic , Psoriasis , Vascular Endothelial Growth Factor C/pharmacology
11.
Cancer Res ; 78(20): 5940-5948, 2018 10 15.
Article En | MEDLINE | ID: mdl-30185550

High expression of VEGFC predicts adverse prognosis in acute myeloid leukemia (AML). We therefore explored VEGFC-targeting efficacy as an AML therapy using a VEGFC mAb. VEGFC antibody therapy enforced myelocytic differentiation of clonal CD34+ AML blasts. Treatment of CD34+ AML blasts with the antibody reduced expansion potential by 30% to 50% and enhanced differentiation via FOXO3A suppression and inhibition of MAPK/ERK proliferative signals. VEGFC antibody therapy also accelerated leukemia cell differentiation in a systemic humanized AML mouse model. Collectively, these results define a regulatory function of VEGFC in CD34+ AML cell fate decisions via FOXO3A and serve as a new potential differentiation therapy for patients with AML.Significance: These findings reveal VEGFC targeting as a promising new differentiation therapy in AML. Cancer Res; 78(20); 5940-8. ©2018 AACR.


Antibodies, Monoclonal/pharmacology , Leukemia, Myeloid, Acute/therapy , Vascular Endothelial Growth Factor C/immunology , Animals , Antigens, CD34/metabolism , Cell Differentiation , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phosphorylation , Prognosis
12.
Int J Oral Maxillofac Surg ; 46(1): 16-23, 2017 Jan.
Article En | MEDLINE | ID: mdl-27745792

The purpose of this study was to evaluate the immunoexpression of vascular endothelial growth factor C (VEGF-C) and VEGF receptor 3 (VEGFR-3) and their correlation with intratumoural lymphatic density (ILD) and peritumoural lymphatic density (PLD) in metastatic and non-metastatic lower lip squamous cell carcinoma (LLSCC). Twenty-five LLSCC with regional nodal metastasis and 25 LLSCC without metastasis were selected. The percentages of VEGF-C and VEGFR-3 staining in each tumour core and at the deep invasive front were assessed. PLD and ILD were determined using anti-podoplanin antibody. Immunohistochemical findings were correlated with nodal metastasis, clinical staging, local recurrence, clinical outcome, and histological grade. Cytoplasmic immunoexpression of VEGFR-3 in the tumour core was associated with metastasis (P=0.009), patient death (P=0.008), and histological grade (P<0.005). PLD, ILD, and VEGF-C expression showed no significant associations with clinicopathological parameters (P>0.05). PLD and ILD were not significantly correlated with the immunoexpression of VEGF-C or VEGFR-3 (P>0.05). There was a significant positive correlation between PLD and ILD (P=0.004), and between cytoplasmic immunoreactivity of VEGF-C and VEGFR-3 (P=0.011). These results suggest an important role for VEGFR-3 in the progression of LLSCC, and highlight the possible influence of its expression on the prognosis of these tumours. ILD and PLD may not be associated with lymph node metastasis in LLSCC.


Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Lip Neoplasms/immunology , Lip Neoplasms/pathology , Lymphatic Metastasis/immunology , Lymphatic Metastasis/pathology , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor Receptor-3/immunology , Female , Humans , Immunoenzyme Techniques , Male , Membrane Glycoproteins/immunology , Neoplasm Recurrence, Local , Neoplasm Staging
13.
Stem Cell Res ; 17(2): 342-351, 2016 09.
Article En | MEDLINE | ID: mdl-27614132

Indirect evidence suggests that adipose tissue-derived stromal cells (ASCs) possess different physiological and biological variations related to the anatomical localization of the adipose depots. Accordingly, to investigate the influence of the tissue origin on the intrinsic properties of ASCs and to assess their response to specific stimuli, we compared the biological, functional and ultrastructural properties of two ASC pools derived from mediastinal and subcutaneous depots (thoracic compartment) by means of supplements such as platelet lysate (PL) and FBS. Subcutaneous ASCs exhibited higher proliferative and clonogenic abilities than mediastinal counterpart, as well as increased secreted levels of IL-6 combined with lower amount of VEGF-C. In contrast, mediastinal ASCs displayed enhanced pro-angiogenic and adipogenic differentiation properties, increased cell diameter and early autophagic processes, highlighted by electron microscopy. Our results further support the hypothesis that the origin of adipose tissue significantly defines the biological properties of ASCs, and that a homogeneric function for all ASCs cannot be assumed.


Stromal Cells/cytology , Subcutaneous Fat/cytology , Adipogenesis , Aged , Antibodies, Neutralizing/immunology , Autophagy , Cell Differentiation , Cell Proliferation , Cell Transdifferentiation , Cells, Cultured , Female , Humans , Interleukin-6/metabolism , Male , Mediastinum , Microscopy, Electron, Transmission , Middle Aged , Stromal Cells/metabolism , Stromal Cells/ultrastructure , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism
14.
J Cell Mol Med ; 20(4): 613-22, 2016 Apr.
Article En | MEDLINE | ID: mdl-26828975

Although lymphatic neovascularization may be a key feature of chronic inflammation, it is almost unexplored in primary Sjögren's syndrome (pSS). A recent study revealed a pro-lymphangiogenic function of interleukin (IL)-17, a leading player in pSS pathogenesis. The aims of the study were to investigate lymphangiogenic mediators and lymphatic vasculature in pSS, as well as their possible association with IL-17. Circulating lymphatic endothelial precursor cells (LEPCs) and Th17 cells were enumerated in pSS patients and healthy donors. VEGF-C and IL-17 levels were assessed in paired serum samples. Lymphatic vasculature, VEGF-C/VEGF receptor (VEGFR)-3 and IL-17 were evaluated in pSS minor salivary glands (MSGs) and compared with normal and non-specific chronic sialadenitis (NSCS) MSGs. Circulating LEPCs were expanded in pSS and correlated with circulating Th17 cells, IL-17 and VEGF-C. In pSS MSGs, a newly formed lymphatic capillary network was found within periductal inflammatory infiltrates and the number of interlobular lymphatic vessels was significantly increased compared with normal and NSCS MSGs. Strong VEGF-C expression was detected in pSS ductal epithelial cells and periductal inflammatory cells. Numerous VEGFR-3(+) infiltrating mononuclear cells were exclusively observed in pSS MSGs. VEGFR-3 expression was strongly increased in lymphatic capillaries of pSS MSGs. IL-17(+) inflammatory cells were preferentially observed around lymphatic vessels in pSS MSGs. This study supports the notion that lymphvasculogenesis and lymphangiogenesis are active in pSS, thereby unmasking a novel aspect of disease pathogenesis. In addition, our results suggest another possible pathogenic role of IL-17 in pSS, further supporting its therapeutic targeting in this disease.


Endothelial Cells/pathology , Interleukin-17/immunology , Neovascularization, Pathologic/pathology , Salivary Glands, Minor/pathology , Sialadenitis/diagnosis , Sjogren's Syndrome/diagnosis , Case-Control Studies , Diagnosis, Differential , Endothelial Cells/immunology , Female , Gene Expression Regulation , Humans , Interleukin-17/genetics , Lymphangiogenesis/genetics , Lymphangiogenesis/immunology , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Male , Middle Aged , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Salivary Glands, Minor/immunology , Sialadenitis/genetics , Sialadenitis/immunology , Sialadenitis/pathology , Signal Transduction , Sjogren's Syndrome/genetics , Sjogren's Syndrome/immunology , Sjogren's Syndrome/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/immunology
15.
Transplantation ; 99(4): 678-86, 2015 Apr.
Article En | MEDLINE | ID: mdl-25606789

BACKGROUND: Graft failure because of immune rejection remains a significant problem in organ transplantation, and lymphatic and blood vessels are important components of the afferent and efferent arms of the host alloimmune response, respectively. We compare the effect of antihemangiogenic and antilymphangiogenic therapies on alloimmunity and graft survival in a murine model of high-risk corneal transplantation. METHODS: Orthotopic corneal transplantation was performed in hemevascularized and lymph-vascularized high-risk host beds, and graft recipients received subconjunctival vascular endothelial growth factor (VEGF)-trap, anti-VEGF-C, sVEGFR-3, or no treatment, beginning at the time of surgery. Fourteen days after transplantation, graft hemeangiogenesis and lymphangiogenesis were evaluated by immunohistochemistry. The frequencies of Th1 cells in regional lymphoid tissue and graft-infiltrating immune cells were evaluated by flow cytometry. Long-term allograft survival was compared using Kaplan-Meier curves. RESULTS: VEGF-trap significantly decreased graft hemangiogenesis as compared to the control group and was most effective in reducing the frequency of graft-infiltrating immune cells. Anti-VEGF-C and sVEGFR3 significantly decreased graft lymphangiogenesis and lymphoid Th1 cell frequencies as compared to control. VEGF-trap (72%), anti-VEGF-C (25%), and sVEGFR-3 (11%) all significantly improved in the 8-week graft survival compared to control (0%), although VEGF-trap was significantly more effective than both anti-VEGF-C (P < 0.05) and sVEGFR-3 (P < 0.05). CONCLUSION: In a clinically relevant model of high-risk corneal transplantation in which blood and lymphatic vessels are present and treatment begins at the time of transplantation, VEGF-trap is significantly more effective in improving long-term graft survival as compared to anti-VEGF-C and sVEGFR-3, but all approaches improve survival when compared to untreated control.


Angiogenesis Inhibitors/pharmacology , Corneal Transplantation/adverse effects , Graft Rejection/prevention & control , Graft Survival/drug effects , Receptors, Vascular Endothelial Growth Factor/pharmacology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Chemotaxis, Leukocyte/drug effects , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/physiopathology , Lymphangiogenesis/drug effects , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Neovascularization, Physiologic/drug effects , Signal Transduction/drug effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Time Factors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/pharmacology
16.
J Histochem Cytochem ; 63(3): 170-80, 2015 03.
Article En | MEDLINE | ID: mdl-25473093

To determine whether vascular endothelial growth factor-C (VEGF-C) and its receptor (VEGFR-3) are involved in the glial reaction elicited by transplanted mesenchymal stem cells (MSCs), we examined the cellular localization of VEGF-C and VEGFR-3 proteins in the striatum of adult normal rats that received bone marrow-derived human MSCs. The MSC grafts were infiltrated with activated microglia/macrophages and astrocytes over a 2-week period post-transplantation, which appeared to parallel the loss of transplanted MSCs. VEGF-C/VEGFR-3 was expressed in activated microglia/macrophages recruited to the graft site, where the induction of VEGF-C protein was rather late compared with that of its receptor. VEGF-C protein was absent or very weak on day 3, whereas VEGFR-3 immunoreactivity was evident within the first three days. Furthermore, within three days, VEGF-C could be detected in the brain macrophages localized immediately adjacent to the needle track. At the same time, almost all the brain macrophages in both regions expressed VEGFR-3. Reactive astrocytes at the graft site expressed VEGFR-3, but not VEGF-C. These data demonstrated the characteristic time- and cell-dependent expression patterns for VEGF-C and VEGFR-3 within the engrafted brain tissue, suggesting that they may contribute to neuroinflammation in MSC transplantation, possibly through the recruitment and/or activation of microglia/macrophages and astrogliosis.


Brain/immunology , Macrophages/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Microglia/immunology , Vascular Endothelial Growth Factor C/analysis , Vascular Endothelial Growth Factor Receptor-3/analysis , Animals , Astrocytes/immunology , Brain/cytology , Cells, Cultured , Humans , Immunohistochemistry , Male , Mesenchymal Stem Cells/cytology , Microglia/cytology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor Receptor-3/immunology
17.
Biochem Biophys Res Commun ; 454(2): 320-7, 2014 Nov 14.
Article En | MEDLINE | ID: mdl-25450396

Vascular endothelial growth factor-C (VEGF-C) is a main inducer of inflammation-associated lymphangiogenesis in various inflammatory disorders including chronic progressive kidney diseases, for which angiotensin II receptor type 1 blockers (ARBs) are widely used as the main treatment. Although proximal renal tubular cells may affect the formation of lymphatic vessels in the interstitial area by producing VEGF-C, the molecular mechanisms of VEGF-C production and its manipulation by ARB have not yet been examined in human proximal renal tubular epithelial cells (HPTECs). In the present study, TNF-α dose-dependently induced the production of VEGF-C in HPTECs. The TNF-α-induced production of VEGF-C was mediated by the phosphorylation of p38MAPK and HSP27, but not by that of ERK or NFkB. Telmisartan, an ARB that can activate the peroxisome proliferator-activated receptor (PPAR), served as a PPAR-δ activator and reduced the TNF-α-stimulated production of VEGF-C. This reduction was partially attributed to a PPAR-δ-dependent decrease in p38MAPK phosphorylation. Our results indicate that TNF-α induced the production of VEGF-C in HPTECs by activating p38MAPK/HSP27, and this was partially inhibited by telmisartan in a PPAR-δ dependent manner. These results provide a novel insight into inflammation-associated lymphangiogenesis.


Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Benzoates/pharmacology , HSP27 Heat-Shock Proteins/antagonists & inhibitors , PPAR delta/agonists , Tumor Necrosis Factor-alpha/immunology , Vascular Endothelial Growth Factor C/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Antihypertensive Agents/pharmacology , Cell Line , Gene Expression Regulation/drug effects , HSP27 Heat-Shock Proteins/immunology , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/metabolism , PPAR delta/immunology , Phosphorylation/drug effects , Signal Transduction/drug effects , Telmisartan , Vascular Endothelial Growth Factor C/genetics , p38 Mitogen-Activated Protein Kinases/immunology
18.
Immunity ; 40(4): 501-14, 2014 Apr 17.
Article En | MEDLINE | ID: mdl-24656836

Toll-like receptors (TLRs) are critical in mediating innate immune responses against infections. However, uncontrolled TLR-triggered inflammation is associated with endotoxin shock. To better understand the homeostatic mechanisms induced by TLR4 signaling, we screened a group of key cytokines, chemokines, growth factors, and their receptors for bacteria- or LPS-induced expression. The surface vascular endothelial growth factor receptor-3 (VEGFR-3) and its ligand VEGF-C were upregulated in macrophages. VEGFR-3 ligation by VEGF-C significantly attenuated proinflammatory cytokine production. Notably, ablation of the ligand-binding domain or tyrosine kinase activity of VEGFR-3 rendered mice more sensitive to septic shock. VEGFR-3 restrained TLR4-NF-κB activation by regulating the PI3-kinase-Akt signaling pathway and SOCS1 expression. Aside from targeting lymphatic vessels, we suggest a key role of VEGFR-3 on macrophages to prevent infections that is complicated with lymphoedema. Thus, VEGFR-3-VEGF-C signaling represents a "self-control" mechanism during antibacterial innate immunity.


Gram-Negative Anaerobic Bacteria/immunology , Macrophages/immunology , Shock, Septic/immunology , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Escherichia coli/immunology , Escherichia coli Infections/immunology , Immunity, Innate/genetics , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Engineering , Protein Structure, Tertiary/genetics , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Toll-Like Receptor 4/metabolism , Vascular Endothelial Growth Factor C/immunology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics
19.
Wound Repair Regen ; 21(4): 604-15, 2013.
Article En | MEDLINE | ID: mdl-23758174

Noninvasive, longitudinal near-infrared fluorescence (NIRF) imaging was used to detect and quantify lymphangiogenesis following a full-dermis thickness incision in the presence and absence of locally administered vascular endothelial growth factor-C (VEGF-C), a well-known regulator of lymphangiogenesis. Peripheral cytokines/chemokines were also measured in treated and sham-injected animals. Lymphangiogenesis was detected via NIRF imaging by day 7-8 and confirmed by intravital microscopy, while angiogenesis was observed by day 2-3 postincision (PI). All lymph vessel parameters quantified were significantly greater on wounded vs. nonwounded sides of mice. Lymph vessel parameters appeared larger on wounded sides of VEGF-C- relative to NaCl-treated mice, although differences were not significant. Interleukin-1α and interleukin-22 were significantly elevated at day 7 PI relative to respective preincision levels in VEGF-C-treated mice, and decreased by day 21 PI to levels nearing those measured preincision. For the majority of cytokines/chemokines measured, mean responses were significantly greater in VEGF-C- vs. NaCl-treated animals. Local VEGF-C administration may stimulate lymphangiogenesis during tissue repair and regeneration via mediating systemic cytokine/chemokine levels. NIRF imaging can be utilized to detect lymphangiogenesis during wound healing, and offers a promising platform to complement current methods for monitoring wound status and studying the effects of growth factors on healing.


Angiogenesis Inducing Agents/pharmacology , Lymphangiogenesis/drug effects , Recombinant Proteins/pharmacology , Skin/drug effects , Vascular Endothelial Growth Factor C/pharmacology , Wound Healing/drug effects , Administration, Cutaneous , Angiogenesis Inducing Agents/immunology , Animals , Chemokines/immunology , Coloring Agents , Cytokines/immunology , Female , Lymphangiogenesis/immunology , Male , Mice , Mice, Inbred C57BL , Optical Imaging , Recombinant Proteins/immunology , Skin/injuries , Spectroscopy, Near-Infrared , Vascular Endothelial Growth Factor C/immunology , Wound Healing/immunology
20.
Cancer Res ; 73(12): 3692-703, 2013 Jun 15.
Article En | MEDLINE | ID: mdl-23576559

Inhibition of VEGF signaling can promote lymph node metastasis in preclinical models, but the mechanism is not fully understood, and successful methods of prevention have not been found. Signaling of hepatocyte growth factor (HGF) and its receptor c-Met can promote the growth of lymphatics and metastasis of some tumors. We sought to explore the contributions of c-Met signaling to lymph node metastasis after inhibition of VEGF signaling. In particular, we examined whether c-Met is upregulated in lymphatics in or near pancreatic neuroendocrine tumors in RIP-Tag2 transgenic mice and whether lymph node metastasis can be reduced by concurrent inhibition of VEGF and c-Met signaling. Inhibition of VEGF signaling by anti-VEGF antibody or sunitinib in mice from the age of 14 to 17 weeks was accompanied by more intratumoral lymphatics, more tumor cells inside lymphatics, and more lymph node metastases. Under these conditions, lymphatic endothelial cells, like tumor cells, had strong immunoreactivity for c-Met and phospho-c-Met. c-Met blockade by the selective inhibitor, PF-04217903, significantly reduced metastasis to local lymph nodes. Together, these results indicate that inhibition of VEGF signaling in RIP-Tag2 mice upregulates c-Met expression in lymphatic endothelial cells, increases the number of intratumoral lymphatics and number of tumor cells within lymphatics, and promotes metastasis to local lymph nodes. Prevention of lymph node metastasis by PF-04217903 in this setting implicates c-Met signaling in tumor cell spread to lymph nodes.


Pancreatic Neoplasms/prevention & control , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazines/pharmacology , Triazoles/pharmacology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Immunohistochemistry , Indoles/pharmacology , Lymphatic Metastasis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/metabolism , Pyrroles/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Sunitinib , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/immunology
...