Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Infect Immun ; 89(8): e0012421, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34031127

RESUMEN

Vascular remodeling is a phenomenon seen in the cutaneous lesions formed during infection with Leishmania parasites. Within the lesion, Leishmania major infection leads to the infiltration of inflammatory cells, including macrophages, and is associated with hypoxic conditions and lymphangiogenesis in the local site. This low-oxygen environment is concomitant with the expression of hypoxic inducible factors (HIFs), which initiate the expression of vascular endothelial growth factor-A (VEGF-A) in macrophages during the infection. Here, we found that macrophage hypoxia is elevated in the skin, and the HIF target Vegfa is preferentially expressed at the site of infection. Further, transcripts indicative of both HIF-1α and HIF-2α activation were increased at the site of infection. Given that HIF mediates VEGF-A and that VEGF-A/VEGFR-2 signaling induces lymphangiogenesis, we wanted to investigate the link between myeloid HIF activation and lymphangiogenesis during L. major infection. We show that myeloid aryl hydrocarbon receptor nuclear translocator (ARNT)/HIF/VEGF-A signaling promotes lymphangiogenesis (the generation of newly formed vessels within the local lymphatic network), which helps resolve the lesion by draining away inflammatory cells and fluid. Concomitant with impaired lymphangiogenesis, we find the deletion of myeloid ARNT/HIF signaling leads to an exacerbated inflammatory response associated with a heightened CD4+ Th1 immune response following L. major infection. Altogether, our data suggest that VEGF-A-mediated lymphangiogenesis occurs through myeloid ARNT/HIF activation following Leishmania major infection and this process is critical in limiting immunopathology.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Leishmania major/fisiología , Leishmaniasis Cutánea/etiología , Leishmaniasis Cutánea/metabolismo , Linfangiogénesis/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Transducción de Señal , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Biomarcadores , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Susceptibilidad a Enfermedades/inmunología , Interacciones Huésped-Patógeno/inmunología , Leishmaniasis Cutánea/patología
2.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33434186

RESUMEN

Lymphatic filariasis is the major global cause of nonhereditary lymphedema. We demonstrate that the filarial nematode Brugia malayi induced lymphatic remodeling and impaired lymphatic drainage following parasitism of limb lymphatics in a mouse model. Lymphatic insufficiency was associated with elevated circulating lymphangiogenic mediators, including vascular endothelial growth factor C. Lymphatic insufficiency was dependent on type 2 adaptive immunity, the interleukin-4 receptor, and recruitment of C-C chemokine receptor-2-positive monocytes and alternatively activated macrophages with a prolymphangiogenic phenotype. Oral treatments with second-generation tetracyclines improved lymphatic function, while other classes of antibiotic had no significant effect. Second-generation tetracyclines directly targeted lymphatic endothelial cell proliferation and modified type 2 prolymphangiogenic macrophage development. Doxycycline treatment impeded monocyte recruitment, inhibited polarization of alternatively activated macrophages, and suppressed T cell adaptive immune responses following infection. Our results determine a mechanism of action for the antimorbidity effects of doxycycline in filariasis and support clinical evaluation of second-generation tetracyclines as affordable, safe therapeutics for lymphedemas of chronic inflammatory origin.


Asunto(s)
Brugia Malayi/inmunología , Filariasis Linfática/tratamiento farmacológico , Linfangiogénesis/inmunología , Receptores de Interleucina-4/inmunología , Tetraciclinas/farmacología , Inmunidad Adaptativa , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Filariasis Linfática/genética , Filariasis Linfática/inmunología , Filariasis Linfática/patología , Linfangiogénesis/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Receptores de Interleucina-4/genética , Linfocitos T/inmunología , Linfocitos T/patología
3.
Methods Mol Biol ; 2193: 85-96, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32808261

RESUMEN

Lymphangiogenesis, the formation of lymphatic vessels from preexisting ones, is an important process in wound-healing physiology. Deregulation of lymphangiogenesis and lymphatic vascular remodeling have been implicated in a range of inflammatory conditions, such as lymphedema, lymphadenopathy, tumor growth, and cancer metastasis. Any attempt in understanding various parameters of the lymphangiogenic process and developing desirable therapeutic targets requires recapitulating these conditions in in vivo models. One pitfall with some experimental models is the absence of immune response, an important regulatory factor for lymphangiogenesis. We overcome this issue by using immune competent mice. In this chapter, by using Angiopoietin-2 (Ang2), a protein that belongs to the Ang/Tie signaling pathway, we describe the ear sponge assay with important adaptations, highlighting a reproducible and quantitative tool for assessment of in vivo lymphangiogenesis.


Asunto(s)
Bioensayo/métodos , Oído/fisiopatología , Linfangiogénesis/fisiología , Vasos Linfáticos/fisiología , Angiopoyetina 2/genética , Animales , Oído/cirugía , Humanos , Inmunidad/inmunología , Inmunidad/fisiología , Linfangiogénesis/genética , Linfangiogénesis/inmunología , Vasos Linfáticos/inmunología , Ratones , Transducción de Señal/genética , Remodelación Vascular/genética , Remodelación Vascular/inmunología , Remodelación Vascular/fisiología , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
4.
Gastroenterology ; 160(4): 1315-1329.e13, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33227282

RESUMEN

BACKGROUND & AIMS: Hepatic encephalopathy (HE) is a serious neurologic complication in patients with liver cirrhosis. Very little is known about the role of the meningeal lymphatic system in HE. We tested our hypothesis that enhancement of meningeal lymphatic drainage could decrease neuroinflammation and ameliorate HE. METHODS: A 4-week bile duct ligation model was used to develop cirrhosis with HE in rats. Brain inflammation in patients with HE was evaluated by using archived GSE41919. The motor function of rats was assessed by the rotarod test. Adeno-associated virus 8-vascular endothelial growth factor C (AAV8-VEGF-C) was injected into the cisterna magna of HE rats 1 day after surgery to induce meningeal lymphangiogenesis. RESULTS: Cirrhotic rats with HE showed significantly increased microglia activation in the middle region of the cortex (P < .001) as well as increased neuroinflammation, as indicated by significant increases in interleukin 1ß, interferon γ, tumor necrosis factor α, and ionized calcium binding adaptor molecule 1 (Iba1) expression levels in at least 1 of the 3 regions of the cortex. Motor function was also impaired in rats with HE (P < .05). Human brains of patients with cirrhosis with HE also exhibited up-regulation of proinflammatory genes (NFKB1, IbA1, TNF-α, and IL1ß) (n = 6). AAV8-VEGF-C injection significantly increased meningeal lymphangiogenesis (P = .035) and tracer dye uptake in the anterior and middle regions of the cortex (P = .006 and .003, respectively), their corresponding meninges (P = .086 and .006, respectively), and the draining lymph nodes (P = .02). Furthermore, AAV8-VEGF-C decreased microglia activation (P < .001) and neuroinflammation and ameliorated motor dysfunction (P = .024). CONCLUSIONS: Promoting meningeal lymphatic drainage and enhancing waste clearance improves HE. Manipulation of meningeal lymphangiogenesis could be a new therapeutic strategy for the treatment of HE.


Asunto(s)
Sistema Glinfático/patología , Encefalopatía Hepática/inmunología , Cirrosis Hepática/complicaciones , Trastornos Motores/inmunología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular , Corteza Cerebral/inmunología , Corteza Cerebral/patología , Cisterna Magna/inmunología , Cisterna Magna/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Sistema Glinfático/inmunología , Encefalopatía Hepática/patología , Humanos , Cirrosis Hepática/inmunología , Linfangiogénesis/inmunología , Masculino , Microglía/inmunología , Microglía/patología , Trastornos Motores/patología , Ratas , Factor C de Crecimiento Endotelial Vascular/genética
5.
Int J Mol Sci ; 21(11)2020 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-32485955

RESUMEN

Currently, there is no definitive treatment for lymphatic disorders. Adipose-derived stem cells (ADSCs) have been reported to promote lymphatic regeneration in lymphedema models, but the mechanisms underlying the therapeutic effects remain unclear. Here, we tested the therapeutic effects of ADSC transplantation on lymphedema using a secondary lymphedema mouse model. The model was established in C57BL/6J mice by x-irradiation and surgical removal of the lymphatic system in situ. The number of lymphatic vessels with anti-lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) immunoreactivity increased significantly in mice subjected to transplantation of 7.5 × 105 ADSCs. X-irradiation suppressed lymphatic vessel dilation, which ADSC transplantation could mitigate. Proliferative cell nuclear antigen staining showed increased lymphatic endothelial cell (LEC) and extracellular matrix proliferation. Picrosirius red staining revealed normal collagen fiber orientation in the dermal tissue after ADSC transplantation. These therapeutic effects were not related to vascular endothelial growth factor (VEGF)-C expression. Scanning electron microscopy revealed structures similar to the intraluminal pillar during intussusceptive angiogenesis on the inside of dilated lymphatic vessels. We predicted that intussusceptive lymphangiogenesis occurred in lymphedema. Our findings indicate that ADSC transplantation contributes to lymphedema reduction by promoting LEC proliferation, improving fibrosis and dilation capacity of lymphatic vessels, and increasing the number of lymphatic vessels via intussusceptive lymphangiogenesis.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/metabolismo , Linfangiogénesis/inmunología , Piel/inmunología , Células Madre/citología , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Fibrosis/inmunología , Perfilación de la Expresión Génica , Intususcepción/inmunología , Intususcepción/patología , Vasos Linfáticos/patología , Masculino , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Regeneración , Piel/patología , Piel/efectos de la radiación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Rayos X
6.
Kidney Int ; 97(1): 89-94, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31718844

RESUMEN

Renal allograft rejection can be prevented by immunological tolerance, which may be associated with de novo formed lymphatic vessels in the donor kidney after transplantation in man. A suitable mouse model of renal allograft rejection in which lymphangiogenesis can be deliberately induced in the graft is critical for elucidating the mechanisms responsible for the association between attenuated transplant rejection and abundance of lymphatic vessels. Here we describe the development of a novel mouse model of rapid renal transplant rejection in which transgenic induction of lymphangiogenesis in the immune-incompatible graft greatly extends its survival time. Thus, our novel approach may facilitate exploitation of lymphangiogenesis in the grafted organ.


Asunto(s)
Rechazo de Injerto/prevención & control , Supervivencia de Injerto/inmunología , Enfermedades Renales/cirugía , Trasplante de Riñón/efectos adversos , Linfangiogénesis/inmunología , Aloinjertos/inmunología , Aloinjertos/patología , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Humanos , Riñón/inmunología , Riñón/patología , Longevidad/inmunología , Vasos Linfáticos/inmunología , Vasos Linfáticos/patología , Masculino , Ratones , Ratones Transgénicos , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo
7.
Int J Mol Sci ; 20(9)2019 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-31035644

RESUMEN

Gastric cancer is diagnosed in nearly one million new patients each year and it remains the second leading cause of cancer-related deaths worldwide. Although gastric cancer represents a heterogeneous group of diseases, chronic inflammation has been shown to play a role in tumorigenesis. Cancer development is a multistep process characterized by genetic and epigenetic alterations during tumour initiation and progression. The stromal microenvironment is important in maintaining normal tissue homeostasis or promoting tumour development. A plethora of immune cells (i.e., lymphocytes, macrophages, mast cells, monocytes, myeloid-derived suppressor cells, Treg cells, dendritic cells, neutrophils, eosinophils, natural killer (NK) and natural killer T (NKT) cells) are components of gastric cancer microenvironment. Mast cell density is increased in gastric cancer and there is a correlation with angiogenesis, the number of metastatic lymph nodes and the survival of these patients. Mast cells exert a protumorigenic role in gastric cancer through the release of angiogenic (VEGF-A, CXCL8, MMP-9) and lymphangiogenic factors (VEGF-C and VEGF-F). Gastric mast cells express the programmed death ligands (PD-L1 and PD-L2) which are relevant as immune checkpoints in cancer. Several clinical undergoing trials targeting immune checkpoints could be an innovative therapeutic strategy in gastric cancer. Elucidation of the role of subsets of mast cells in different human gastric cancers will demand studies of increasing complexity beyond those assessing merely mast cell density and microlocalization.


Asunto(s)
Linfangiogénesis , Mastocitos/inmunología , Neovascularización Patológica , Neoplasias Gástricas/etiología , Neoplasias Gástricas/patología , Animales , Biomarcadores , Regulación Neoplásica de la Expresión Génica , Humanos , Linfangiogénesis/genética , Linfangiogénesis/inmunología , Mastocitos/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
8.
Front Immunol ; 10: 557, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30972059

RESUMEN

Background: Inflammatory bowel disease (IBD) is characterized by both acute and chronic phase inflammation of the gastro-intestinal (GI) tract that affect a large and growing number of people worldwide with little to no effective treatments. This is in part due to the lack of understanding of the disease pathogenesis and also the currently poorly described involvement of other systems such as the lymphatics. During DSS induced colitis, mice also develop a severe inflammation of terminal ileum with many features similar to IBD. As well as inflammation within the ileum we have previously demonstrated lymphatic remodeling within the mesentery and mesenteric lymph nodes of DSS-treated mice. The lymphatic remodeling includes lymphangiogenesis, lymphatic vessel dilation and leakiness, as well as cellular infiltration into the surrounding tissue and peripheral draining lymph nodes. Methods: Intestinal inflammation was induced in C57BL/6 mice by administration of 2.5% DSS in drinking water for 7 days. Mice were treated with TLR4 blocker C34 or Polymyxin-B (PMXB) daily from days 3 to 7 of DSS treatment via I.P. injection, and their therapeutic effects on disease activity and lymphatic function were examined. TLR activity and subsequent effect on lymphangiogenesis, lymphadenopathy, and mesenteric lymph node cellular composition were assessed. Results: DSS Mice treated with TLR4 inhibitor, C34, had a significantly improved disease phenotype characterized by reduced ileal and colonic insult. The change correlated with significant reduction in colonic and mesenteric inflammation, resolved mesenteric lymphangiectasia, and CD103+ DC migration similar to that of healthy control. PMXB treatment however did not resolve inflammation within the colon or associated mesenteric lymphatic dysfunction but did however prevent lymphadenopathy within the MLN through alteration of CCL21 gradients and CD103+ DC migration. Conclusions: TLR4 appears to mediate several changes within the mesenteric lymphatics, more specifically it is shown to have different outcomes whether stimulation occurs through pathogen derived factors such as LPS or tissue derived DAMPs, a novel phenomenon.


Asunto(s)
Colitis/inmunología , Ileítis/inmunología , Ganglios Linfáticos/inmunología , Vasos Linfáticos/inmunología , Mesenterio/inmunología , Receptores Toll-Like/inmunología , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Ileítis/inducido químicamente , Linfangiogénesis/inmunología , Ratones Endogámicos C57BL , Receptores Toll-Like/genética
9.
Front Immunol ; 10: 720, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024552

RESUMEN

The lymphatic system comprises a network of lymphoid tissues and vessels that drains the extracellular compartment of most tissues. During tumor development, lymphatic endothelial cells (LECs) substantially expand in response to VEGFR-3 engagement by VEGF-C produced in the tumor microenvironment, a process known as tumor-associated lymphangiogenesis. Lymphatic drainage from the tumor to the draining lymph nodes consequently increases, powering interstitial flow in the tumor stroma. The ability of a tumor to induce and activate lymphatic growth has been positively correlated with metastasis. Much effort has been made to identify genes responsible for tumor-associated lymphangiogenesis. Inhibition of lymphangiogenesis with soluble VEGFR-3 or with specific monoclonal antibodies decreases tumor spread to LNs in rodent models. Importantly, tumor-associated lymphatics do not only operate as tumor cell transporters but also play critical roles in anti-tumor immunity. Therefore, metastatic as well as primary tumor progression can be affected by manipulating tumor-associated lymphatic remodeling or function. Here, we review and discuss our current knowledge on the contribution of LECs immersed in the tumor microenvironment as immunoregulators, as well as a possible functional remodeling of LECs subsets depending on the organ microenvironment.


Asunto(s)
Inmunomodulación/inmunología , Vasos Linfáticos/inmunología , Animales , Células Endoteliales/inmunología , Humanos , Linfangiogénesis/inmunología , Microambiente Tumoral/inmunología
10.
Med Sci (Paris) ; 35(2): 132-137, 2019 Feb.
Artículo en Francés | MEDLINE | ID: mdl-30774080

RESUMEN

The lymphatic system is made up of vessels that drain interstitial fluids throughout the body. The circulation of the lymph (liquid in the lymphatic system) in the lymphatic vessels is unidirectional: tissues to the lymph nodes and then to the veins. Ganglia are mechanical filters but also immune barriers that can block the progression of certain pathogens as well as cancer cells. However, most studies on the lymphatic system and cancer highlight the role of the lymphatic network in metastatic dissemination as tumor cells use this network to reach other organs. However, recent studies describe a beneficial role of the lymphatic system and of the vascular endothelial growth factor C (VEGF-C) which is one of the main factors responsible for the development of lymphatic vessels in cancer. In this review, we will illustrate this ambivalent and emerging role of VEGF-C and the lymphatic system in cancer aggressiveness.


Asunto(s)
Carcinogénesis , Linfangiogénesis , Metástasis de la Neoplasia , Factor C de Crecimiento Endotelial Vascular/fisiología , Animales , Carcinogénesis/genética , Carcinogénesis/inmunología , Humanos , Linfangiogénesis/genética , Linfangiogénesis/inmunología , Vasos Linfáticos/fisiología , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/patología , Escape del Tumor/fisiología
11.
Front Immunol ; 10: 76, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30761143

RESUMEN

The debilitating condition known as secondary lymphedema frequently occurs after lymphadenectomy and/or radiotherapy for the treatment of cancer. These therapies can damage lymphatic vessels leading to edema, fibrosis, inflammation and dysregulated adipogenesis, which result in profound swelling of an affected limb. Importantly, lymphedema patients often exhibit impaired immune function which predisposes them to a variety of infections. It is known that lymphadenectomy can compromise the acquisition of adaptive immune responses and antibody production; however the cellular mechanisms involved are poorly understood. Here we discuss recent progress in revealing the cellular and molecular mechanisms underlying poor immune function in secondary lymphedema, which has indicated a key role for regulatory T cells in immunosuppression in this disease. Furthermore, the interaction of CD4+ T cells and macrophages has been shown to play a role in driving proliferation of lymphatic endothelial cells and aberrant lymphangiogenesis, which contribute to interstitial fluid accumulation in lymphedema. These new insights into the interplay between lymphatic vessels and the immune system in lymphedema will likely provide opportunities for novel therapeutic approaches designed to improve clinical outcomes in this problematic disease.


Asunto(s)
Linfangiogénesis/inmunología , Vasos Linfáticos/inmunología , Linfedema/inmunología , Animales , Comunicación Celular , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Inmunidad Humoral , Inflamación/inmunología , Ganglios Linfáticos/inmunología , Macrófagos/inmunología , Ratones , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología
12.
Nat Commun ; 10(1): 229, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30651548

RESUMEN

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.


Asunto(s)
Encéfalo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Linfangiogénesis/inmunología , Vasos Linfáticos/inmunología , Linfocitos T/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Antígenos/inmunología , Antígenos/metabolismo , Encéfalo/diagnóstico por imagen , Proliferación Celular , Líquido Cefalorraquídeo/inmunología , Encefalomielitis Autoinmune Experimental/diagnóstico por imagen , Hueso Etmoides , Azul de Evans/administración & dosificación , Femenino , Humanos , Vigilancia Inmunológica/inmunología , Vasos Linfáticos/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Glicoproteína Mielina-Oligodendrócito/inmunología , Toxina del Pertussis/administración & dosificación , Toxina del Pertussis/inmunología , Factor C de Crecimiento Endotelial Vascular/inmunología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/inmunología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Int Immunopharmacol ; 66: 224-235, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30476824

RESUMEN

Inflammation-induced lymphangiogenesis is a widely accepted concept. However, most of the inflammatory factors and their related mechanisms have not been clarified. It has been reported that sphingosine-1-phosphate (S1P) is not only closely related to the chronic inflammatory process but also affects angiogenesis. Therefore, we investigated the inflammatory effects of S1P on human lymphatic endothelial cells (HLECs). Our results showed that S1P promotes tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) secretion in HLECs. We also confirmed that S1P-stimulated TNF-α and IL-1ß secretion is mediated through S1P receptor 1 (S1PR1). Using TNF-α siRNA and IL-1ß siRNA, we found that TNF-α and IL-1ß play essential roles in S1P-induced HLEC proliferation, migration, and tube formation. S1P induces phosphorylation of NF-κB p65 and activation of NF-κB nuclear translocation. A S1PR1 antagonist (W146) and NF-κB inhibitor (BAY11-7082) inhibited S1P-induced TNF-α and IL-1ß secretion and prevented NF-κB nuclear translocation. Taken together, the results demonstrated for the first time that S1P promotes the secretion of TNF-α and IL-1ß in HLECs via S1PR1-mediated NF-κB signaling pathways, thus affecting lymphangiogenesis. The study provides a new strategy for finding treatments for lymphangiogenesis-related diseases.


Asunto(s)
Células Endoteliales/fisiología , Inflamación/inmunología , Linfangiogénesis/inmunología , Organogénesis/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lisofosfolípidos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/inmunología , Receptores de Esfingosina-1-Fosfato , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
14.
JCI Insight ; 3(23)2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30518687

RESUMEN

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.


Asunto(s)
Enfermedad Crónica , Dermatitis/inmunología , Inflamación/inmunología , Factor C de Crecimiento Endotelial Vascular/inmunología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Linfocitos T CD4-Positivos , Proliferación Celular , Dermatitis/tratamiento farmacológico , Modelos Animales de Enfermedad , Etanercept/inmunología , Etanercept/metabolismo , Etanercept/farmacología , Femenino , Fibronectinas , Inflamación/tratamiento farmacológico , Linfangiogénesis/inmunología , Vasos Linfáticos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Psoriasis , Factor C de Crecimiento Endotelial Vascular/farmacología
15.
Lymphat Res Biol ; 16(6): 498-506, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30272526

RESUMEN

Many basic and clinical studies have demonstrated that atherosclerosis is a chronic inflammatory disease. Although there are many factors affecting atherosclerosis, the role of lymphatic vessels in this disease has been neglected. Traditionally, lymphatic vessels have been considered to be passages for transporting interstitial fluid to the blood circulation. However, as early as the last century, researchers found that there are numerous lymphatic vessels surrounding sites of atherosclerosis; however, the relationship between lymphatic vessels and atherosclerosis is not clear. With further research, lymphatic vessels were determined to be involved in the induction and resolution of arterial inflammation and also to play a positive role in plaque cholesterol transport. There are abundant immune cells around atherosclerosis, and these immune cells not only have a significant impact on plaque formation but also affect local lymphangiogenesis (IAL). This promotion of IAL seems to relieve the progression of atherosclerosis. Therefore, research into the relationship between lymphatic vessels and atherosclerosis is of great importance for improving atherosclerosis treatment. This review highlights what is known about the relationship between lymphatic vessels and atherosclerosis, including the effect of immune cells on IAL, and reverse cholesterol transport. In addition, we present some of our views on the improvement of atherosclerosis treatment, which have significant clinical value in research.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Colesterol/metabolismo , Hipolipemiantes/uso terapéutico , Vasos Linfáticos/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Vasodilatadores/uso terapéutico , Animales , Aterosclerosis/inmunología , Aterosclerosis/patología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Transporte Biológico/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/patología , Progresión de la Enfermedad , Humanos , Linfangiogénesis/efectos de los fármacos , Linfangiogénesis/inmunología , Vasos Linfáticos/inmunología , Vasos Linfáticos/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
16.
Medicine (Baltimore) ; 97(39): e12458, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30278527

RESUMEN

Glial tumors constitute the majority of primary intracranial brain tumors. The expression of specific markers of lymphangiogenesis in gliomas still remains unclear.A total of 40 surgical specimens from 20 patients with recurrent gliomas were included in the study. The expression of D2-40, vascular endothelial growth factor (VEGF)-C, VEGF-D, and VEGF receptor-3 (VEGR-3) was detected by immunohistochemistry (IHC). The clinicopathologic data (p53 and Ki67) were also collected and analyzed.At relapse malignant transformation rate was 65% (13/20 cases). D2-40, VEGF-C, VEGF-D, and VEGFR-3 were expressed in 20%, 30%, 60%, and 20% of primary and 45%, 30%, 75%, and 35% of recurrent glioma tumors (P < .01, P = 1.00, P = .03, P = .03). In 13 cases with increased malignancy grade, the expression of Ki67 and p53 were higher at relapse compared with the primary tumors (P = .001, P = .045). Multivariate survival analysis showed VEGF-D was an independent prognostic factor for malignant transformation (HR = 0.376, P = .045).Glioma is easy to relapse with tumor progression. VEGF-D was an independent prognostic factor for malignant transformation.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Linfangiogénesis/inmunología , Recurrencia Local de Neoplasia/metabolismo , Adulto , Anticuerpos Monoclonales de Origen Murino/metabolismo , Biomarcadores/metabolismo , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Progresión de la Enfermedad , Femenino , Glioma/diagnóstico por imagen , Glioma/patología , Glioma/cirugía , Humanos , Antígeno Ki-67/metabolismo , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/diagnóstico por imagen , Recurrencia Local de Neoplasia/patología , Pronóstico , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor D de Crecimiento Endotelial Vascular/metabolismo
17.
Microbiol Immunol ; 62(11): 732-736, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30239037

RESUMEN

IL-4 plays an important role in the pathogenesis of atopic dermatitis (AD) by dysregulating many key factors at the transcriptional level. In this study, a microRNA array technique and IL-4 transgenic mice were used to demonstrate that IL-4 dysregulates microRNAs involved in inflammation, angiogenesis, lymphangiogenesis and apoptosis. Of the 372 common microRNAs examined, 26 and one microRNAs were found to be up- and down-regulated, respectively. MicroRNA-101-5p, -122-5p, -142-3p, -204-5p, -335-3p, -376a-3p, -378a-5p, -639 and -9-5p are among the most significantly up-regulated microRNAs. MicroRNA-147a, the only one that was down- regulated in the present study, attenuates TLR-induced inflammatory responses. These dysregulated microRNAs may provide post-transcriptional regulation of key genes in AD.


Asunto(s)
Inductores de la Angiogénesis/inmunología , Inflamación/inmunología , Interleucina-4/inmunología , Queratinocitos/inmunología , MicroARNs/inmunología , Animales , Apoptosis/inmunología , Dermatitis Atópica/genética , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Regulación hacia Abajo , Humanos , Inflamación/genética , Interleucina-4/genética , Queratinocitos/citología , Linfangiogénesis/genética , Linfangiogénesis/inmunología , Ratones , Ratones Transgénicos , MicroARNs/genética , Regulación hacia Arriba
18.
Curr Opin Immunol ; 53: 180-186, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29879585

RESUMEN

Lymph node (LN) expansion during inflammation is essential to establish immune responses and relies on the development of blood and lymph vessels. Human dendritic cells (DCs), subdivided into two main subsets, namely conventional DCs (cDCs) and plasmacytoid DCs (pDCs), are professional antigen presenting cells endowed with the capability to produce soluble mediators regulating inflammation and tissue repair. cDCs support angiogenesis in secondary LNs both directly and indirectly through the secretion of vascular endothelial growth factor-A (VEGF)-A and VEGF-C and the production of several other mediators endowed with angiogenic properties. Finally, cDCs can affect neovascular formation via a transdifferentiation process. At variance with cDCs, the angiogenic properties of pDCs still remain poorly explored.


Asunto(s)
Células Dendríticas/inmunología , Linfangiogénesis/inmunología , Neovascularización Patológica/inmunología , Animales , Transdiferenciación Celular , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo
19.
Am J Reprod Immunol ; 80(1): e12970, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29756666

RESUMEN

PROBLEM: The lymphatic vasculature controls leukocytes trafficking and limits the adaptive immune response. In previous models of preeclampsia (PE), defective immune function caused by disruption of lymphangiogenesis was shown to be involved in the disease pathophysiology. Especially, the dysfunction of regulatory T cells (Treg) at the maternal-fetal interface may be one of the causes of severe PE. In particular, activation of Tregs to obtain immune tolerance requires adequate antigen presentation through the lymphatic system. We hypothesized that impaired lymphangiogenesis and imbalanced Tregs at the maternal-fetal interface are associated with the pathophysiology of severe PE. However, the current research addressing this hypothesis is limited. Therefore, to compare differences in lymphangiogenesis in severe PE and normal conditions, we aimed to examine the location of lymphatics at the maternal-fetal interface and to investigate the association between lymphangiogenesis and Tregs in severe PE. METHOD OF STUDY: We obtained entire uterus from normal pregnant mice. Placental and fetal membranes, including decidua, were obtained from 10 pregnant women with severe PE and 10 gestational age-matched controls. Immunohistochemistry for LYVE1 was used to localize the distribution of lymphatic vessels and CD4, CD25, and FOXP3 for Treg. RESULTS: LYVE1-positive vessels were present in the uterine wall of mice. LYVE1-positive lymphatic vessels were localized on the human decidua. Tubular lymphatics were abundant in the control decidua, but significantly reduced in severe PE. Furthermore, lymphatic vessel density correlated with the number of decidual Tregs. CONCLUSION: Abnormal decidual lymphangiogenesis is associated with reduced numbers of decidual Tregs in severe PE.


Asunto(s)
Vasos Linfáticos/inmunología , Preeclampsia/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD4/inmunología , Membranas Extraembrionarias/inmunología , Femenino , Factores de Transcripción Forkhead/inmunología , Edad Gestacional , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Linfangiogénesis/inmunología , Ratones , Ratones Endogámicos C57BL , Placenta/inmunología , Embarazo , Útero/inmunología , Proteínas de Transporte Vesicular/inmunología
20.
Nat Commun ; 9(1): 1970, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773802

RESUMEN

T cell-mediated responses have been implicated in the development of fibrosis, impaired lymphangiogenesis, and lymphatic dysfunction in secondary lymphedema. Here we show that CD4+ T cells are necessary for lymphedema pathogenesis by utilizing adoptive transfer techniques in CD4 knockout mice that have undergone tail skin and lymphatic excision or popliteal lymph node dissection. We also demonstrate that T cell activation following lymphatic injury occurs in regional skin-draining lymph nodes after interaction with antigen-presenting cells such as dendritic cells. CD4+ T cell activation is associated with differentiation into a mixed T helper type 1 and 2 phenotype, as well as upregulation of adhesion molecules and chemokines that promote migration to the skin. Most importantly, we find that blocking T cell release from lymph nodes using a sphingosine-1-phosphate receptor modulator prevents lymphedema, suggesting that this approach may have clinical utility.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inmunosupresores/uso terapéutico , Linfedema/inmunología , Activación de Linfocitos/inmunología , Traslado Adoptivo , Animales , Antígenos CD4/genética , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Inmunosupresores/farmacología , Ganglios Linfáticos/citología , Ganglios Linfáticos/patología , Linfangiogénesis/inmunología , Vasos Linfáticos/citología , Vasos Linfáticos/inmunología , Vasos Linfáticos/patología , Linfedema/tratamiento farmacológico , Linfedema/patología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Lisoesfingolípidos/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Piel/citología , Piel/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...