Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 219
Filtrar
1.
Pediatr Dev Pathol ; 27(3): 228-234, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38512910

RESUMEN

INTRODUCTION: Blue rubber bleb nevus syndrome (BRBNS) is an uncommon vascular anomaly characterized by multifocal cutaneous, visceral, and other soft tissue or solid organ venous malformations. We observed that BRBNS lesions express immunohistochemical markers of lymphatic differentiation. METHODS: BRBNS histopathologic specimens assessed at our institution during the past 27 years were reviewed. Slides from 19 BRBNS lesions were selected from 14 patients (9 cutaneous, 9 gastrointestinal, and 1 hepatic). We recorded the involved anatomical compartments and presence/absence of thrombi or vascular smooth muscle. Immunohistochemical endothelial expression of PROX1 (nuclear) and D2-40 (membranous/cytoplasmic) was evaluated semi-quantitatively. RESULTS: Endothelial PROX1 immunopositivity was noted in all specimens; the majority (89.5%) demonstrated staining in more than 10% of cells. D2-40 immunopositivity was present in one-third (33%) of cutaneous lesions and only 1 gastrointestinal lesion. CONCLUSION: Endothelial cells in BRBNS almost always express 1 or more immunohistochemical markers of lymphatic differentiation.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Gastrointestinales , Inmunohistoquímica , Nevo Azul , Neoplasias Cutáneas , Humanos , Nevo Azul/metabolismo , Nevo Azul/patología , Nevo Azul/diagnóstico , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/diagnóstico , Masculino , Niño , Femenino , Preescolar , Adolescente , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/diagnóstico , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/análisis , Lactante , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/análisis , Proteínas de Homeodominio/metabolismo , Endotelio Linfático/metabolismo , Endotelio Linfático/patología , Anticuerpos Monoclonales de Origen Murino/metabolismo
2.
Int J Exp Pathol ; 102(6): 268-278, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34791715

RESUMEN

Lymphangiogenesis makes an important contribution to the tumour microenvironment (TME), but little is known about this in oral squamous cell carcinoma (OSCC). Archival formalin-fixed paraffin-embedded specimens (28 OSCC, 10 inflamed and 6 normal oral mucosa controls) were processed using immunohistochemistry (IHC) with antibodies against lymphatic markers D2-40 (podoplanin), LYVE-1, VEGFR3 and Prox1. After the endothelial cells had been highlighted by the various markers for lymphatic endothelium, the positive stained cells and vessels were identified and counted in a systematic manner to determine microvessel density. Double-labelling immunofluorescence (DLIF) was used to investigate the specificity of D2-40 and LYVE-1 to lymphatic endothelial cells (LECs) as opposed to blood ECs. There was higher D2-40 and Prox1 lymphatic vessel density (P = .001) in the OSCC group when compared with both control groups. Some malignant keratinocytes expressed lymphatic markers, as did a much smaller number of epithelial cells in the control groups. DLIF showed that no vessels co-expressed D2-40/CD34 or LYVE/CD34. Some D2/40+ LVs were LYVE- . D2-40 was the most specific LEC marker in OSCC tissues. These results establish that the OSCC TME contains significantly more lymphatic vessels expressing D2-40 and Prox1 than the control groups, which may play a role in facilitating lymphatic invasion and metastases.


Asunto(s)
Células Endoteliales/metabolismo , Linfangiogénesis/fisiología , Neoplasias de la Boca/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Antígenos CD34/metabolismo , Biomarcadores de Tumor/metabolismo , Células Endoteliales/patología , Endotelio Linfático/metabolismo , Endotelio Linfático/patología , Técnica del Anticuerpo Fluorescente , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Vasos Linfáticos/metabolismo , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Proteínas Supresoras de Tumor/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Transporte Vesicular/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 41(12): 2943-2960, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34670407

RESUMEN

OBJECTIVE: Cerebral cavernous malformations (CCMs) can happen anywhere in the body, although they most commonly produce symptoms in the brain. The role of CCM genes in other vascular beds outside the brain and retina is not well-examined, although the 3 CCM-associated genes (CCM1, CCM2, and CCM3) are ubiquitously expressed in all tissues. We aimed to determine the role of CCM gene in lymphatics. Approach and Results: Mice with an inducible pan-endothelial cell (EC) or lymphatic EC deletion of Ccm3 (Pdcd10ECKO or Pdcd10LECKO) exhibit dilated lymphatic capillaries and collecting vessels with abnormal valve structure. Morphological alterations were correlated with lymphatic dysfunction in Pdcd10LECKO mice as determined by Evans blue dye and fluorescein isothiocyanate(FITC)-dextran transport assays. Pdcd10LECKO lymphatics had increased VEGFR3 (vascular endothelial growth factor receptor-3)-ERK1/2 (extracellular signal-regulated kinase 1/2) signaling with lymphatic hyperplasia. Mechanistic studies suggested that VEGFR3 is primarily regulated at a transcriptional level in Ccm3-deficient lymphatic ECs, in an NF-κB (nuclear factor κB)-dependent manner. CCM3 binds to importin alpha 2/KPNA2 (karyopherin subunit alpha 2), and a CCM3 deletion releases KPNA2 to activate NF-κB P65 by facilitating its nuclear translocation and P65-dependent VEGFR3 transcription. Moreover, increased VEGFR3 in lymphatic EC preferentially activates ERK1/2 signaling, which is critical for lymphatic EC proliferation. Importantly, inhibition of VEGFR3 or ERK1/2 rescued the lymphatic defects in structure and function. CONCLUSIONS: Our data demonstrate that CCM3 deletion augments the VEGFR3-ERK1/2 signaling in lymphatic EC that drives lymphatic hyperplasia and malformation and warrant further investigation on the potential clinical relevance of lymphatic dysfunction in patients with CCM.


Asunto(s)
Endotelio Linfático/fisiopatología , Hemangioma Cavernoso del Sistema Nervioso Central/fisiopatología , Sistema de Señalización de MAP Quinasas/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Células Cultivadas , Células Endoteliales/fisiología , Endotelio Linfático/patología , Femenino , Eliminación de Gen , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Hiperplasia , Masculino , Ratones Endogámicos , Modelos Animales , FN-kappa B/genética , Translocación Genética
4.
Sci Adv ; 7(29)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34272244

RESUMEN

The mechanisms maintaining adult lymphatic vascular specialization throughout life and their role in coordinating inter-organ communication to sustain homeostasis remain elusive. We report that inactivation of the mechanosensitive transcription factor Foxc2 in adult lymphatic endothelium leads to a stepwise intestine-to-lung systemic failure. Foxc2 loss compromised the gut epithelial barrier, promoted dysbiosis and bacterial translocation to peripheral lymph nodes, and increased circulating levels of purine metabolites and angiopoietin-2. Commensal microbiota depletion dampened systemic pro-inflammatory cytokine levels, corrected intestinal lymphatic dysfunction, and improved survival. Foxc2 loss skewed the specialization of lymphatic endothelial subsets, leading to populations with mixed, pro-fibrotic identities and to emergence of lymph node-like endothelial cells. Our study uncovers a cross-talk between lymphatic vascular function and commensal microbiota, provides single-cell atlas of lymphatic endothelial subtypes, and reveals organ-specific and systemic effects of dysfunctional lymphatics. These effects potentially contribute to the pathogenesis of diseases, such as inflammatory bowel disease, cancer, or lymphedema.


Asunto(s)
Vasos Linfáticos , Linfedema , Células Endoteliales/metabolismo , Endotelio Linfático/metabolismo , Endotelio Linfático/patología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Vasos Linfáticos/metabolismo , Linfedema/metabolismo , Linfedema/patología
5.
Pediatr Hematol Oncol ; 38(4): 367-377, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33641614

RESUMEN

Multifocal lymphangioendotheliomatosis with thrombocytopenia (MLT) is a recently recognized disorder characterized by vascular lesions marked by distinct endothelial proliferation. Lesions affect multiple tissues, and MLT can be associated with refractory thrombocytopenia resulting in life-threatening bleeding. Diagnosing MLT may be challenging given its rarity and phenotypic variability. There is no consensus on the optimal management or treatment duration. We report a 4-month-old male who presented with multiple vascular malformations involving the gastrointestinal tract, lung, bones, choroid plexus, and spleen, with minimal cutaneous involvement and no thrombocytopenia. Wedge resection of a pulmonary nodule was strongly positive for lymphatic vessel endothelial hyaluronan receptor 1 favoring MLT despite the lack of thrombocytopenia. The patient's clinical symptoms and vascular lesions improved on sirolimus therapy. We review the literature to highlight the clinical variability of MLT and discuss the diagnostic and therapeutic options for MLT.


Asunto(s)
Angiomatosis/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Vasos Linfáticos/patología , Sirolimus/uso terapéutico , Trombocitopenia/tratamiento farmacológico , Angiomatosis/complicaciones , Angiomatosis/patología , Endotelio Linfático/efectos de los fármacos , Endotelio Linfático/patología , Humanos , Lactante , Vasos Linfáticos/efectos de los fármacos , Masculino , Trombocitopenia/complicaciones , Trombocitopenia/patología
6.
Trends Cancer ; 7(3): 214-225, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33132107

RESUMEN

Secondary lymphoid organs (SLOs) are important initiators and regulators of immunity. To carry out this function, the blood vasculature must deliver oxygen and nutrients and recruit circulating lymphocytes into the SLO parenchyma, where they encounter cognate antigen. High endothelial venules (HEVs) are specialised postcapillary venules that specifically serve this function and are found in all SLOs except spleen. It is becoming clear that alterations to HEV network density and/or morphology can result in immune activation or, as recently implicated, in providing an exit route for tumour cell dissemination and metastases. In this review, the structural plasticity of HEVs, the regulatory pathways underpinning this plasticity, and the relevance of these pathways to cancer progression will be discussed.


Asunto(s)
Endotelio Linfático/patología , Neoplasias/inmunología , Neoplasias/patología , Ganglio Linfático Centinela/patología , Movimiento Celular/inmunología , Plasticidad de la Célula/inmunología , Progresión de la Enfermedad , Endotelio Linfático/inmunología , Humanos , Ganglio Linfático Centinela/inmunología
7.
Int J Oncol ; 56(4): 1034-1044, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32319559

RESUMEN

Metastatic cancer cells cross endothelial barriers and travel through the blood or lymphatic fluid to pre­metastatic niches, leading to their colonisation. 'S' stereoisomer 12S­hydroxy­5Z,8Z,10E,14Z­eicosatetraenoic acid [12(S)­HETE] is secreted by a variety of cancer cell types and has been indicated to open up these barriers. In the present study, another aspect of the endothelial unlocking mechanism was elucidated. This was achieved by investigating 12(S)­HETE­treated lymph endothelial cells (LECs) with regard to their expression and mutual interaction with v­rel avian reticuloendotheliosis viral oncogene homolog A (RELA), intercellular adhesion molecule 1, SRY­box transcription factor 18 (SOX18), prospero homeobox 1 (PROX1) and focal adhesion kinase (FAK). These key players of LEC retraction, which is a prerequisite for cancer cell transit into vasculature, were analysed using western blot analysis, reverse transcription­quantitative PCR and transfection with small interfering (si)RNA. The silencing of a combination of these signalling and executing molecules using siRNA, or pharmacological inhibition with defactinib and Bay11­7082, extended the mono­culture experiments to co­culture settings using HCT116 colon cancer cell spheroids that were placed on top of LEC monolayers to measure their retraction using the validated 'circular chemorepellent­induced defect' assay. 12(S)­HETE was indicated to induce the upregulation of the RELA/SOX18 feedback loop causing the subsequent phosphorylation of FAK, which fed back to RELA/SOX18. Therefore, 12(S)­HETE was demonstrated to be associated with circuits involving RELA, SOX18 and FAK, which transduced signals causing the retraction of LECs. The FAK­inhibitor defactinib and the NF­κB inhibitor Bay11­7082 attenuated LEC retraction additively, which was similar to the suppression of FAK and PROX1 (the target of SOX18) by the transfection of respective siRNAs. FAK is an effector molecule at the distal end of a pro­metastatic signalling cascade. Therefore, targeting the endothelial­specific activity of FAK through the pathway demonstrated herein may provide a potential therapeutic method to combat cancer dissemination via vascular routes.


Asunto(s)
Movimiento Celular , Endotelio Linfático/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Ácidos Hidroxieicosatetraenoicos/farmacología , Neoplasias/patología , Factores de Transcripción SOXF/metabolismo , Factor de Transcripción ReIA/metabolismo , Línea Celular Tumoral , Endotelio Linfático/efectos de los fármacos , Endotelio Linfático/patología , Retroalimentación Fisiológica , Quinasa 1 de Adhesión Focal/genética , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Factores de Transcripción SOXF/genética , Transducción de Señal , Factor de Transcripción ReIA/genética
8.
Cells ; 8(8)2019 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-31426584

RESUMEN

Crohn's disease (CD) is a chronic inflammatory condition that can affect different portions of the gastrointestinal tract. Lymphatic drainage was demonstrated to be dysfunctional in CD pathogenesis, ultimately causing the failure of the resolution of intestinal inflammation. To investigate the molecular mechanisms underlying these dysfunctions, we isolated human intestinal lymphatic endothelial cells (HILECs) from surgical specimens of patients undergoing resection for complicated CD (CD HILEC) and from a disease-free margin of surgical specimens of patients undergoing resection for cancer (healthy HILEC). Both cell types underwent transcriptomic profiling, and their barrier functionality was tested using a transwell-based co-culture system between HILEC and lamina propria mononuclear cells (LPMCs). Results showed CD HILEC displayed a peculiar transcriptomic signature that highlighted mTOR signaling as an orchestrator of leukocyte trafficking through the lymphatic barrier of CD patients. Moreover, we demonstrated that LPMC transmigration through the lymphatic endothelium of patients with CD depends on the capability of mTOR to trigger interleukin 20 receptor subunit α (IL20RA)-mediated intracellular signaling. Conclusively, our study suggests that leukocyte trafficking through the intestinal lymphatic microvasculature can be controlled by modulating IL20RA, thus leading to the resolution of chronic inflammation in patients with CD.


Asunto(s)
Enfermedad de Crohn/inmunología , Células Endoteliales/inmunología , Endotelio Linfático/inmunología , Intestinos/inmunología , Serina-Treonina Quinasas TOR/fisiología , Anciano , Movimiento Celular/inmunología , Células Endoteliales/patología , Endotelio Linfático/patología , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Intestinos/patología , Masculino , Persona de Mediana Edad , Receptores de Interleucina/inmunología
9.
Am J Pathol ; 189(11): 2269-2292, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31421071

RESUMEN

Lymphatic metastasis is a high-impact prognostic factor for mortality of breast cancer (BC) patients, and it directly depends on tumor-associated lymphatic vessels. We previously reported that lipopolysaccharide-induced inflammatory lymphangiogenesis is strongly promoted by myeloid-derived lymphatic endothelial cell progenitors (M-LECPs) derived from the bone marrow (BM). As BC recruits massive numbers of provascular myeloid cells, we hypothesized that M-LECPs, within this recruited population, are specifically programmed to promote tumor lymphatics that increase lymph node metastasis. In support of this hypothesis, high levels of M-LECPs were found in peripheral blood and tumor tissues of BC patients. Moreover, the density of M-LECPs and lymphatic vessels positive for myeloid marker proteins strongly correlated with patient node status. It was also established that tumor M-LECPs coexpress lymphatic-specific, stem/progenitor and M2-type macrophage markers that indicate their BM hematopoietic-myeloid origin and distinguish them from mature lymphatic endothelial cells, tumor-infiltrating lymphoid cells, and tissue-resident macrophages. Using four orthotopic BC models, we show that mouse M-LECPs are similarly recruited to tumors and integrate into preexisting lymphatics. Finally, we demonstrate that adoptive transfer of in vitro differentiated M-LECPs, but not naïve or nondifferentiated BM cells, significantly increased metastatic burden in ipsilateral lymph nodes. These data support a causative role of BC-induced lymphatic progenitors in tumor lymphangiogenesis and suggest molecular targets for their inhibition.


Asunto(s)
Neoplasias de la Mama/patología , Células Progenitoras Endoteliales/fisiología , Endotelio Linfático/patología , Células Mieloides/fisiología , Animales , Células de la Médula Ósea/fisiología , Línea Celular Tumoral , Femenino , Humanos , Linfangiogénesis/fisiología , Metástasis Linfática , Vasos Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID
10.
J Clin Invest ; 129(11): 4912-4921, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31415243

RESUMEN

Molecular heterogeneity of endothelial cells underlies their highly specialized functions during changing physiological conditions within diverse vascular beds. For example, placental spiral arteries (SAs) undergo remarkable remodeling to meet the ever-growing demands of the fetus - a process which is deficient in preeclampsia. The extent to which maternal endothelial cells coordinate with immune cells and pregnancy hormones to promote SA remodeling remains largely unknown. Here we found that remodeled SAs expressed the lymphatic markers PROX1, LYVE1, and VEGFR3, mimicking lymphatic identity. Uterine natural killer (uNK) cells, which are required for SA remodeling and secrete VEGFC, were both sufficient and necessary for VEGFR3 activation in vitro and in mice lacking uNK cells, respectively. Using Flt4Chy/+ mice with kinase inactive VEGFR3 and Vegfcfl/fl Vav1-Cre mice, we demonstrated that SA remodeling required VEGFR3 signaling, and that disrupted maternal VEGFR3 signaling contributed to late-gestation fetal growth restriction. Collectively, we identified a novel instance of lymphatic mimicry by which maternal endothelial cells promote SA remodeling, furthering our understanding of the vascular heterogeneity employed for the mitigation of pregnancy complications such as fetal growth restriction and preeclampsia.


Asunto(s)
Arterias/inmunología , Retardo del Crecimiento Fetal/inmunología , Imitación Molecular , Placenta/inmunología , Preeclampsia/inmunología , Útero/inmunología , Remodelación Vascular/inmunología , Animales , Antígenos de Diferenciación , Arterias/patología , Endotelio Linfático/inmunología , Endotelio Linfático/patología , Femenino , Retardo del Crecimiento Fetal/patología , Humanos , Ratones , Placenta/irrigación sanguínea , Placenta/patología , Preeclampsia/patología , Embarazo , Útero/irrigación sanguínea , Útero/patología
11.
EBioMedicine ; 38: 79-88, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30497977

RESUMEN

BACKGROUND: Nanomedicine offers an excellent opportunity to tackle treatment-refractory malignancies by enhancing the delivery of therapeutics to the tumor site. High endothelial venules (HEVs) are found primarily in lymph nodes or formed de novo in peripheral tissues during inflammatory responses. They express peripheral node addressin (PNAd), which is recognized by the monoclonal antibody MECA79. METHODS: Here, we demonstrated that HEVs form de novo in human pancreatic ductal adenocarcinoma (PDAC). We engineered MECA79 coated nanoparticles (MECA79-NPs) that recognize these ectopic HEVs in PDAC. FINDINGS: The trafficking of MECA79-NPs following intravenous delivery to human PDAC implanted in a humanized mouse model was more robust than non-conjugated NPs. Treatment with MECA79-Taxol-NPs augmented the delivery of Paclitaxel (Taxol) to the tumor site and significantly reduced the tumor size. This effect was associated with a higher apoptosis rate of PDAC cells and reduced vascularization within the tumor. INTERPRETATION: Targeting the HEVs of PDAC using MECA79-NPs could lay the ground for the localized delivery of a wide variety of drugs including chemotherapeutic agents. FUND: National Institutes of Health (NIH) grants: T32-EB016652 (B·B.), NIH Cancer Core Grant CA034196 (L.D.S.), National Institute of Allergy and Infectious Diseases grants R01-AI126596 and R01-HL141815 (R.A.).


Asunto(s)
Carcinoma Ductal Pancreático/patología , Endotelio Linfático/patología , Ganglios Linfáticos/patología , Neovascularización Patológica , Neoplasias Pancreáticas/patología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Biomarcadores , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Endotelio Linfático/efectos de los fármacos , Endotelio Linfático/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Terapia Molecular Dirigida , Nanopartículas/química , Nanopartículas/ultraestructura , Neovascularización Patológica/tratamiento farmacológico , Paclitaxel/administración & dosificación , Paclitaxel/química , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Nanomedicina Teranóstica , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
12.
Arterioscler Thromb Vasc Biol ; 38(10): 2410-2422, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354217

RESUMEN

Objective- Maintenance of lymphatic permeability is essential for normal lymphatic function during adulthood, but the precise signaling pathways that control lymphatic junctions during development are not fully elucidated. The Gs-coupled AM (adrenomedullin) signaling pathway is required for embryonic lymphangiogenesis and the maintenance of lymphatic junctions during adulthood. Thus, we sought to elucidate the downstream effectors mediating junctional stabilization in lymphatic endothelial cells. Approach and Results- We knocked-down both Rap1A and Rap1B isoforms in human neonatal dermal lymphatic cells (human lymphatic endothelial cells) and genetically deleted the mRap1 gene in lymphatic endothelial cells by producing 2 independent, conditional Rap1a/b knockout mouse lines. Rap1A/B knockdown caused disrupted junctional formation with hyperpermeability and impaired AM-induced lymphatic junctional tightening, as well as rescue of histamine-induced junctional disruption. Less than 60% of lymphatic- Rap1a/b knockout embryos survived to E13.5 exhibiting interstitial edema, blood-filled lymphatics, disrupted lymphovenous valves, and defective lymphangiogenesis. Consistently, inducible lymphatic- Rap1a/b deletion in adult animals prevented AM-rescue of histamine-induced lymphatic leakage and dilation. Conclusions- Rap1 (Ras-related protein) serves as the dominant effector downstream of AM to stabilize lymphatic junctions. Rap1 is required for maintaining lymphatic permeability and driving normal lymphatic development.


Asunto(s)
Adrenomedulina/farmacología , Células Endoteliales/efectos de los fármacos , Endotelio Linfático/efectos de los fármacos , Uniones Intercelulares/efectos de los fármacos , Linfangiogénesis/efectos de los fármacos , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/patología , Endotelio Linfático/enzimología , Endotelio Linfático/patología , Histamina/farmacología , Humanos , Uniones Intercelulares/enzimología , Uniones Intercelulares/patología , Ratones , Ratones Noqueados , Permeabilidad , Transducción de Señal , Proteínas de Unión al GTP rap/genética , Proteínas de Unión al GTP rap1/genética
13.
J Immunol ; 201(6): 1735-1747, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30045970

RESUMEN

Lymph node (LN) expansion during an immune response is a complex process that involves the relaxation of the fibroblastic network, germinal center formation, and lymphatic vessel growth. These processes require the stromal cell network of the LN to act deliberately to accommodate the influx of immune cells to the LN. The molecular drivers of these processes are not well understood. Therefore, we asked whether the immediate cytokines type 1 IFN produced during viral infection influence the lymphatic network of the LN in mice. We found that following an IFN-inducing stimulus such as viral infection or polyI:C, programmed cell death ligand 1 (PD-L1) expression is dynamically upregulated on lymphatic endothelial cells (LECs). We found that reception of type 1 IFN by LECs is important for the upregulation of PD-L1 of mouse and human LECs and the inhibition of LEC expansion in the LN. Expression of PD-L1 by LECs is also important for the regulation of LN expansion and contraction after an IFN-inducing stimulus. We demonstrate a direct role for both type 1 IFN and PD-L1 in inhibiting LEC division and in promoting LEC survival. Together, these data reveal a novel mechanism for the coordination of type 1 IFN and PD-L1 in manipulating LEC expansion and survival during an inflammatory immune response.


Asunto(s)
Antígeno B7-H1/inmunología , Proliferación Celular , Células Endoteliales/inmunología , Endotelio Linfático/inmunología , Interferón Tipo I/inmunología , Animales , Antígeno B7-H1/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Endoteliales/patología , Endotelio Linfático/patología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interferón Tipo I/genética , Ratones , Ratones Noqueados , Poli I-C/farmacología
14.
Microcirculation ; 25(7): e12492, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30025187

RESUMEN

OBJECTIVE: Lymphatic vessel dysfunction and increased lymph leakage have been directly associated with several metabolic diseases. However, the underlying cellular mechanisms causing lymphatic dysfunction have not been determined. Aberrant insulin signaling affects the metabolic function of cells and consequently impairs tissue function. We hypothesized that insulin resistance in LECs decreases eNOS activity, disrupts barrier integrity increases permeability, and activates mitochondrial dysfunction and pro-inflammatory signaling pathways. METHODS: LECs were treated with insulin and/or glucose to determine the mechanisms leading to insulin resistance. RESULTS: Acute insulin treatment increased eNOS phosphorylation and NO production in LECs via activation of the PI3K/Akt signaling pathway. Prolonged hyperglycemia and hyperinsulinemia induced insulin resistance in LECs. Insulin-resistant LECs produced less NO due to a decrease in eNOS phosphorylation and showed a significant decrease in impedance across an LEC monolayer that was associated with disruption in the adherence junctional proteins. Additionally, insulin resistance in LECs impaired mitochondrial function by decreasing basal-, maximal-, and ATP-linked OCRs and activated NF-κB nuclear translocation coupled with increased pro-inflammatory signaling. CONCLUSION: Our data provide the first evidence that insulin resistance disrupts endothelial barrier integrity, decreases eNOS phosphorylation and mitochondrial function, and activates inflammation in LECs.


Asunto(s)
Endotelio Linfático/metabolismo , Resistencia a la Insulina , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Endotelio Linfático/patología , Glucosa/farmacología , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Insulina/farmacología , Uniones Intercelulares/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos
15.
Biomed Pharmacother ; 106: 1-7, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29940537

RESUMEN

BACKGROUND: Lymphangiogenesis is critical for metastasis of a variety of cancers, including breast cancer. CPT1A (carnitine palmitoyltransferase 1a) has been reported to play a critical role in breast cancer progress. However, the molecular mechanism remains elusive. METHODS: In order to investigate the role of CPT1A in HDLEC cells, short hairpin RNA approach was utilized to knock down the CPT1A gene expression. We employed transwell and lymphatic vessel formation assay to examine invasion and lymphangiogenesis of HDLEC (Human dermal lymphatic endothelial cells). RT-qPCR and westernblot analyses were used to determine genes expression in HDLEC and breast cancer cells. Finally, we determined the relative rate of acetyl-CoA/CoA in shNC and shCPT1A HDLEC cells by LC-MS approach. RESULTS: Knockdown of CPT1A in breast cancer cells (MCF-7 and MDA-MB-231) abolished invasion and lymphangiogenesis of HDLEC cells. Mechanistically, CPT1A depletion suppressed the expression of VEGF-C and VEGF-D in MCF-7 and MDA-MB-231 cells. Interestingly, CPT1A knockdown in HDLEC cells exhibited attenuated expression of lymphangiogenic markers (podoplanin, VEGFR-3, VEGF-C, VEGF-D and PROX-1). Consistently, CPT1A -null HDLEC cells displayed compromised invasion and lymphangiogenesis compared with negative control. Further investigation revealed that CPT1A regulated VEGFR3 via acetyl-CoA mediated H3K9ac, which could be abrogated by supplement of acetate. CONCLUSIONS: In present study, we revealed the mechanism by which CPT1A regulates breast cancer-associated invasion and lymphangiogenesis. Our findings provide insights into CPT1A -promoted breast tumor metastasis and provide rationale for understanding breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/enzimología , Carnitina O-Palmitoiltransferasa/metabolismo , Células Endoteliales/enzimología , Endotelio Linfático/enzimología , Linfangiogénesis , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Acetilcoenzima A/metabolismo , Acetilación , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carnitina O-Palmitoiltransferasa/genética , Comunicación Celular , Movimiento Celular , Técnicas de Cocultivo , Células Endoteliales/patología , Endotelio Linfático/patología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Linfangiogénesis/genética , Células MCF-7 , Metástasis de la Neoplasia , Factor A de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor D de Crecimiento Endotelial Vascular/genética , Factor D de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Elife ; 72018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29712618

RESUMEN

Lymphatic invasion and lymph node metastasis correlate with poor clinical outcome in melanoma. However, the mechanisms of lymphatic dissemination in distant metastasis remain incompletely understood. We show here that exposure of expansively growing human WM852 melanoma cells, but not singly invasive Bowes cells, to lymphatic endothelial cells (LEC) in 3D co-culture facilitates melanoma distant organ metastasis in mice. To dissect the underlying molecular mechanisms, we established LEC co-cultures with different melanoma cells originating from primary tumors or metastases. Notably, the expansively growing metastatic melanoma cells adopted an invasively sprouting phenotype in 3D matrix that was dependent on MMP14, Notch3 and ß1-integrin. Unexpectedly, MMP14 was necessary for LEC-induced Notch3 induction and coincident ß1-integrin activation. Moreover, MMP14 and Notch3 were required for LEC-mediated metastasis of zebrafish xenografts. This study uncovers a unique mechanism whereby LEC contact promotes melanoma metastasis by inducing a reversible switch from 3D growth to invasively sprouting cell phenotype.


Asunto(s)
Neoplasias de la Mama/patología , Endotelio Linfático/patología , Integrina beta1/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Metaloproteinasa 14 de la Matriz/metabolismo , Receptor Notch3/metabolismo , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Endotelio Linfático/metabolismo , Femenino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Metástasis Linfática , Ratones , Ratones SCID , Invasividad Neoplásica , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
17.
Elife ; 72018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29620526

RESUMEN

Patients with Gorham-Stout disease (GSD) have lymphatic vessels in their bones and their bones gradually disappear. Here, we report that mice that overexpress VEGF-C in bone exhibit a phenotype that resembles GSD. To drive VEGF-C expression in bone, we generated Osx-tTA;TetO-Vegfc double-transgenic mice. In contrast to Osx-tTA mice, Osx-tTA;TetO-Vegfc mice developed lymphatics in their bones. We found that inhibition of VEGFR3, but not VEGFR2, prevented the formation of bone lymphatics in Osx-tTA;TetO-Vegfc mice. Radiological and histological analysis revealed that bones from Osx-tTA;TetO-Vegfc mice were more porous and had more osteoclasts than bones from Osx-tTA mice. Importantly, we found that bone loss in Osx-tTA;TetO-Vegfc mice could be attenuated by an osteoclast inhibitor. We also discovered that the mutant phenotype of Osx-tTA;TetO-Vegfc mice could be reversed by inhibiting the expression of VEGF-C. Taken together, our results indicate that expression of VEGF-C in bone is sufficient to induce the pathologic hallmarks of GSD in mice.


Asunto(s)
Resorción Ósea/patología , Huesos/patología , Endotelio Linfático/patología , Vasos Linfáticos/patología , Osteoclastos/patología , Factor C de Crecimiento Endotelial Vascular/fisiología , Animales , Resorción Ósea/metabolismo , Huesos/metabolismo , Células Cultivadas , Endotelio Linfático/metabolismo , Humanos , Vasos Linfáticos/metabolismo , Ratones , Ratones Transgénicos , Osteoclastos/metabolismo , Fenotipo , Transducción de Señal , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Lymphat Res Biol ; 16(1): 56-64, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29359999

RESUMEN

It is theorized that toxic agents are transported from the hyperpermeable gut of burn victims through the lymph, to the systemic circulation, causing global injury. We believe that immune cells respond to leakage of "toxic lymph" following trauma causing the attraction of these cells to the perilymphatic space. To test this, we utilized a model of burn on rats to examine changes in a single immune cell population associated with mesenteric lymphatic dysfunction. We examined the ability of serum from these animals to increase permeability in lymphatic endothelial monolayers and disrupt cellular junctions. We also treated burn animals with doxycycline, an inhibitor of microvascular permeability, and observed the effects on immune cell populations, morphometry, and lymphatic endothelial permeability. Burn injury increased the number of MHCII+ immune cells along the vessel (>50%). The size and shape of these cells also changed significantly following burn injury. Serum from burn animals increased lymphatic endothelial permeability (∼1.5-fold) and induced breaks in VE-cadherin staining. Doxycycline treatment blocked the accumulation of immune cells along the vessel, whereas serum from doxycycline-treated animals failed to increase lymphatic endothelial permeability. The size of cells along the vessel in doxycycline-treated burn animals was not affected, suggesting that the cells already present on the lymphatic vessels still respond to substances in the lymph. These findings suggest that factors produced during burn can induce lymphatic endothelial barrier disruption and lymph produced during traumatic injury can influence the attraction and morphology of immune cell populations along the vessel.


Asunto(s)
Células Presentadoras de Antígenos/efectos de los fármacos , Quemaduras/tratamiento farmacológico , Doxiciclina/farmacología , Células Endoteliales/efectos de los fármacos , Antígenos de Histocompatibilidad Clase II/inmunología , Vasos Linfáticos/efectos de los fármacos , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/patología , Antígenos CD/genética , Antígenos CD/inmunología , Biomarcadores/metabolismo , Quemaduras/genética , Quemaduras/inmunología , Quemaduras/patología , Cadherinas/genética , Cadherinas/inmunología , Permeabilidad Capilar , Movimiento Celular/efectos de los fármacos , Tamaño de la Célula , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/patología , Endotelio Linfático/efectos de los fármacos , Endotelio Linfático/inmunología , Endotelio Linfático/patología , Expresión Génica , Antígenos de Histocompatibilidad Clase II/genética , Linfa/citología , Linfa/efectos de los fármacos , Linfa/inmunología , Vasos Linfáticos/inmunología , Vasos Linfáticos/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Mesenterio/efectos de los fármacos , Mesenterio/inmunología , Mesenterio/patología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/patología , Ratas , Ratas Sprague-Dawley
19.
World J Surg Oncol ; 15(1): 204, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162097

RESUMEN

BACKGROUND: The purpose of this study is to examine the expression levels of lymphatic endothelial markers in colorectal cancer and to explore the correlation between the expression levels of markers and lymph node status. METHODS: Forty-seven paired fresh tumor tissues and para-cancerous tissues were collected from colorectal cancer patients who received surgical treatment between August 2015 and March 2016 in Cancer Hospital, Chinese Academy of Medical Sciences. Real-time quantitative PCR (RTQ-PCR) was used to check the expression levels of LYVE-1, VEGFR-3, Podoplanin, and Prox-1 in tumor and para-cancerous tissues. RESULTS: The positive expression rates of LYVE-1, VEGFR-3, Podoplanin, and Prox-1 in tumor tissues were 100, 93.6, 100, and 91.4%, but 100, 100, 100, and 87.2% in para-cancerous tissues. Comparing with para-cancerous tissues, tumor tissues had significantly lower expression levels of LYVE-1 (P < 0.001) and VEGFR-3 (P = 0.013) and higher levels of Podoplanin (P = 0.016) and Prox-1 (P = 0.078). There was no correlation between lymph node status and the expression level of LYVE-1 in tumor tissues (P = 0.354) or par-cancerous tissues (P = 0.617); similar results were found for VEGFR-3 (P = 0.631, 0.738), Podoplanin (P = 0.490, 0.625), and Prox-1 (P = 0.503, 0.174). Meanwhile, there was no correlation between N-staging and the expression level of LYVE-1 in tumor tissues (P = 0.914) or para-cancerous tissues (P = 0.784); similar results were found for VEGFR-3 (P = 0.493, 0.955), Podoplanin (P = 0.199, 0.370), and Prox-1 (P = 0.780, 0.234). CONCLUSIONS: There was no correlation between expression levels of lymphatic endothelial markers and lymph node status; LYVE-1, VEGFR-3, Podoplanin, and Prox-1 could not be used for predicting the lymph node status or N-staging of colorectal cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Endotelio Linfático/patología , Ganglios Linfáticos/patología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Metástasis Linfática , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Proteínas Supresoras de Tumor/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Transporte Vesicular/metabolismo
20.
Arterioscler Thromb Vasc Biol ; 37(9): 1732-1735, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28705793

RESUMEN

OBJECTIVE: The purpose of this study was to investigate the role of Fat4 and Dachsous1 signaling in the lymphatic vasculature. APPROACH AND RESULTS: Phenotypic analysis of the lymphatic vasculature was performed in mice lacking functional Fat4 or Dachsous1. The overall architecture of lymphatic vasculature is unaltered, yet both genes are specifically required for lymphatic valve morphogenesis. Valve endothelial cells (Prox1high [prospero homeobox protein 1] cells) are disoriented and failed to form proper valve leaflets. Using Lifeact-GFP (green fluorescent protein) mice, we revealed that valve endothelial cells display prominent actin polymerization. Finally, we showed the polarized recruitment of Dachsous1 to membrane protrusions and cellular junctions of valve endothelial cells in vivo and in vitro. CONCLUSIONS: Our data demonstrate that Fat4 and Dachsous1 are critical regulators of valve morphogenesis. This study highlights that valve defects may contribute to lymphedema in Hennekam syndrome caused by Fat4 mutations.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Células Endoteliales/metabolismo , Endotelio Linfático/metabolismo , Linfangiogénesis , Vasos Linfáticos/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Cadherinas/deficiencia , Cadherinas/genética , Células Cultivadas , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Anomalías Craneofaciales/patología , Células Endoteliales/patología , Endotelio Linfático/patología , Técnica del Anticuerpo Fluorescente , Predisposición Genética a la Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Humanos , Linfangiectasia Intestinal/genética , Linfangiectasia Intestinal/metabolismo , Linfangiectasia Intestinal/patología , Vasos Linfáticos/patología , Linfedema/genética , Linfedema/metabolismo , Linfedema/patología , Ratones Noqueados , Mutación , Fenotipo , Multimerización de Proteína , Transducción de Señal , Transfección , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...