Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(3): e2213317120, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36634143

RESUMO

There is an urgent need to develop novel drugs to reduce the mortality from severe infectious diseases with the emergence of new pathogens, including Coronavirus disease 2019 (COVID-19). Although current drugs effectively suppress the proliferation of pathogens, immune cell activation, and inflammatory cytokine functions, they cannot completely reduce mortality from severe infections and sepsis. In this study, we focused on the endothelial cell-specific protein, Roundabout 4 (Robo4), which suppresses vascular permeability by stabilizing endothelial cells, and investigated whether enhanced Robo4 expression could be a novel therapeutic strategy against severe infectious diseases. Endothelial-specific overexpression of Robo4 suppresses vascular permeability and reduces mortality in lipopolysaccharide (LPS)-treated mice. Screening of small molecules that regulate Robo4 expression and subsequent analysis revealed that two competitive small mothers against decapentaplegic (SMAD) signaling pathways, activin receptor-like kinase 5 (ALK5)-SMAD2/3 and ALK1-SMAD1/5, positively and negatively regulate Robo4 expression, respectively. An ALK1 inhibitor was found to increase Robo4 expression in mouse lungs, suppress vascular permeability, prevent extravasation of melanoma cells, and decrease mortality in LPS-treated mice. The inhibitor suppressed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced endothelial barrier disruption and decreased mortality in mice infected with SARS-CoV-2. These results indicate that enhancing Robo4 expression is an efficient strategy to suppress vascular permeability and mortality in severe infectious diseases, including COVID-19, and that small molecules that upregulate Robo4 can be potential therapeutic agents against these diseases.


Assuntos
COVID-19 , Endotoxemia , Animais , Camundongos , Receptores de Superfície Celular/metabolismo , Permeabilidade Capilar , Células Endoteliais/metabolismo , Transdução de Sinais , Regulação para Cima , Endotoxemia/metabolismo , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , COVID-19/metabolismo , SARS-CoV-2/metabolismo
2.
Angiogenesis ; 26(4): 567-580, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37563497

RESUMO

Tissue-resident vascular endothelial stem cells (VESCs), marked by expression of CD157, possess long-term repopulating potential and contribute to vascular regeneration and homeostasis in mice. Stem cell exhaustion is regarded as one of the hallmarks of aging and is being extensively studied in several types of tissue-resident stem cells; however, how aging affects VESCs has not been clarified yet. In the present study, we isolated VESCs from young and aged mice to compare their potential to differentiate into endothelial cells in vitro and in vivo. Here, we report that the number of liver endothelial cells (ECs) including VESCs was lower in aged (27-28 month-old) than young (2-3 month-old) mice. In vitro culture of primary VESCs revealed that the potential to generate ECs is impaired in aged VESCs isolated from liver and lung relative to young VESCs. Orthotopic transplantation of VESCs showed that aged VESCs and their progeny expand less efficiently than their young counterparts when transplanted into aged mice, but they are equally functional in young recipients. Gene expression analysis indicated that inflammatory signaling was more activated in aged ECs including VESCs. Using single-cell RNA sequencing data from the Tabula Muris Consortium, we show that T cells and monocyte/macrophage lineage cells including Kupffer cells are enriched in the aged liver. These immune cells produce IL-1ß and several chemokines, suggesting the possible involvement of age-associated inflammation in the functional decline of VESCs with age.


Assuntos
Células Progenitoras Endoteliais , Camundongos , Animais , Células-Tronco/metabolismo , Fígado , Envelhecimento
3.
Rinsho Ketsueki ; 64(7): 661-664, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-37544727

RESUMO

By carrying a systemic circulation, hematopoietic and vascular systems coordinately govern the functional organ connections in the body. Blood vessels play an important role in the development, regeneration, and maintenance of organs by acting as conduits for environmental factors in the blood to tissues and secreting organ-specific cytokines as angiocrine signals. Recently, it has become clear that vascular endothelial cells, which are the main constituent cells of the blood vessels and play a role in homeostasis, are diverse. It has also been established that the cells of stem cell fraction exist in endothelial cells. The vascular endothelial cells in various organs are functionally different. For example, it has been discovered that sinusoidal blood vessels in the liver produce coagulation factor VIII as an organ-specific vascular function. Determining how such tissue-/organ-specific function of the endothelial cells is induced is a topic of interest in the vascular field of study.


Assuntos
Capilares , Células Endoteliais , Hemofilia A , Fígado , Humanos , Fígado/irrigação sanguínea , Fígado/fisiologia , Vasos Sanguíneos , Capilares/fisiologia
4.
Angiogenesis ; 25(2): 151-154, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34617195

RESUMO

Microvascular dysfunction accompanied by a dramatic alteration of stable capillary structure is a major hallmark of numerous age-related diseases. In skin, although the role of angiogenesis during dermal reconstitution is well documented, the functional relevance of the extracellular matrix (ECM) stiffness to vascular remodeling and its molecular mechanisms was poorly understood. Here, we developed an ex vivo 3-dimensional angiogenic model using human fat, revealing that "appropriate" stiffness induces vascular maturation associated with upregulated APJ expression, whereas the overexpression of APJ promotes the formation of large vessels even in the absence of the "appropriate" stiffness. Taken together, APJ could be a novel mechanotransducer that accelerates the maturation of cutaneous blood vessels, leading to the prevention of human skin aging.


Assuntos
Matriz Extracelular , Pele , Vasos Sanguíneos , Capilares , Matriz Extracelular/metabolismo , Humanos , Neovascularização Patológica/metabolismo , Pele/irrigação sanguínea
5.
Cancer Sci ; 112(4): 1348-1356, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33587826

RESUMO

It has been widely accepted that the regulation of the tumor microenvironment is an important strategy in cancer treatment. Particularly, control of the tumor vasculature has been suggested to be critical for antitumor immunotherapy. Effectiveness of cancer immunotherapy depends on the quality and quantity of immune cells infiltrating into tumor tissues, which may be affected by the status of the tumor vasculature. Under physiological conditions, immune cells migrate from the intravascular lumen into the parenchyma especially by passing through the vascular wall of venulae. Extravasation of immune cells is induced from venulae where endothelial cells (ECs) are fully covered with pericytes from the basal side. Interaction of pericytes with ECs contributes to immune cell extravasation by several steps, ie, adhesion of immune cells to intraluminal ECs, transmigration, and chemotaxis of immune cells. Blood vessels are structurally immature and non-functional in tumors, and therefore, induction of maturation in the tumor vasculature is a promising strategy for effective cancer therapies and is relevant not only for immune cell migration but also drug delivery.


Assuntos
Endotélio Vascular/patologia , Neoplasias/patologia , Animais , Movimento Celular/fisiologia , Células Endoteliais/patologia , Humanos , Imunoterapia/métodos , Pericitos/patologia , Microambiente Tumoral/fisiologia
6.
Int Immunol ; 32(5): 295-305, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31996897

RESUMO

The vast blood-vessel network of the circulatory system is crucial for maintaining bodily homeostasis, delivering essential molecules and blood cells, and removing waste products. Blood-vessel dysfunction and dysregulation of new blood-vessel formation are related to the onset and progression of many diseases including cancer, ischemic disease, inflammation and immune disorders. Endothelial cells (ECs) are fundamental components of blood vessels and their proliferation is essential for new vessel formation, making them good therapeutic targets for regulating the latter. New blood-vessel formation occurs by vasculogenesis and angiogenesis during development. Induction of ECs termed tip, stalk and phalanx cells by interactions between vascular endothelial growth factor A (VEGF-A) and its receptors (VEGFR1-3) and between Notch and Delta-like Notch ligands (DLLs) is crucial for regulation of angiogenesis. Although the importance of angiogenesis is unequivocal in the adult, vasculogenesis effected by endothelial progenitor cells (EPCs) may also contribute to post-natal vessel formation. However, the definition of these cells is ambiguous and they include several distinct cell types under the simple classification of 'EPC'. Furthermore, recent evidence indicates that ECs within the intima show clonal expansion in some situations and that they may harbor vascular-resident endothelial stem cells. In this article, we summarize recent knowledge on vascular development and new blood-vessel formation in the adult. We also introduce concepts of EC heterogeneity and EC clonal expansion, referring to our own recent findings.


Assuntos
Vasos Sanguíneos/citologia , Proliferação de Células , Células Endoteliais/citologia , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Células Endoteliais/metabolismo , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Immunity ; 37(6): 1024-36, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23200825

RESUMO

Jdp2 is an AP-1 family transcription factor that regulates the epigenetic status of histones. Previous in vitro studies revealed that Jdp2 is involved in osteoclastogenesis. However, the roles of Jdp2 in vivo and its pleiotropic functions are largely unknown. Here we generated Jdp2(-/-) mice and discovered its crucial roles not only in bone metabolism but also in differentiation of neutrophils. Jdp2(-/-) mice exhibited osteopetrosis resulting from impaired osteoclastogenesis. Jdp2(-/-) neutrophils were morphologically normal but had impaired surface expression of Ly6G, bactericidal function, and apoptosis. We also found that ATF3 was an inhibitor of neutrophil differentiation and that Jdp2 directly suppresses its expression via inhibition of histone acetylation. Strikingly, Jdp2(-/-) mice were highly susceptible to Staphylococcus aureus and Candida albicans infection. Thus, Jdp2 plays pivotal roles in in vivo bone homeostasis and host defense by regulating osteoclast and neutrophil differentiation.


Assuntos
Osso e Ossos/metabolismo , Neutrófilos/imunologia , Osteoclastos/citologia , Proteínas Repressoras/genética , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Animais , Antígenos Ly/genética , Antígenos Ly/metabolismo , Apoptose/genética , Apoptose/imunologia , Osso e Ossos/imunologia , Candidíase/genética , Diferenciação Celular/genética , Regulação da Expressão Gênica , Predisposição Genética para Doença , Homeostase , Camundongos , Camundongos Knockout , Neutrófilos/citologia , Neutrófilos/metabolismo , Osteoclastos/metabolismo , Osteopetrose/genética , Osteopetrose/imunologia , Proteínas Repressoras/metabolismo , Infecções Estafilocócicas/genética
8.
Arterioscler Thromb Vasc Biol ; 40(10): 2425-2439, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32787520

RESUMO

OBJECTIVE: The calcineurin-NFAT (nuclear factor for activated T cells)-DSCR (Down syndrome critical region)-1 pathway plays a crucial role as the downstream effector of VEGF (vascular endothelial growth factor)-mediated tumor angiogenesis in endothelial cells. A role for DSCR-1 in different organ microenvironment such as the cornea and its role in ocular diseases is not well understood. Corneal changes can be indicators of various disease states and are easily detected through ocular examinations. Approach and Results: The presentation of a corneal arcus or a corneal opacity due to lipid deposition in the cornea often indicates hyperlipidemia and in most cases, hypercholesterolemia. Although the loss of Apo (apolipoprotein) E has been well characterized and is known to lead to elevated serum cholesterol levels, there are few corneal changes observed in ApoE-/- mice. In this study, we show that the combined loss of ApoE and DSCR-1 leads to a dramatic increase in serum cholesterol levels and severe corneal opacity with complete penetrance. The cornea is normally maintained in an avascular state; however, loss of Dscr-1 is sufficient to induce hyper-inflammatory and -oxidative condition, increased corneal neovascularization, and lymphangiogenesis. Furthermore, immunohistological analysis and genome-wide screening revealed that loss of Dscr-1 in mice triggers increased immune cell infiltration and upregulation of SDF (stromal derived factor)-1 and its receptor, CXCR4 (C-X-C motif chemokine ligand receptor-4), potentiating this signaling axis in the cornea, thereby contributing to pathological corneal angiogenesis and opacity. CONCLUSIONS: This study is the first demonstration of the critical role for the endogenous inhibitor of calcineurin, DSCR-1, and pathological corneal angiogenesis in hypercholesterolemia induced corneal opacity.


Assuntos
Proteínas de Ligação ao Cálcio/deficiência , Neovascularização da Córnea/etiologia , Opacidade da Córnea/etiologia , Células Endoteliais/metabolismo , Endotélio Corneano/metabolismo , Hipercolesterolemia/complicações , Proteínas Musculares/deficiência , Animais , Proteínas de Ligação ao Cálcio/genética , Quimiocina CXCL12/metabolismo , Quimiotaxia de Leucócito , Neovascularização da Córnea/genética , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Opacidade da Córnea/genética , Opacidade da Córnea/metabolismo , Opacidade da Córnea/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/patologia , Endotélio Corneano/patologia , Infecções Oculares Fúngicas/metabolismo , Infecções Oculares Fúngicas/patologia , Células HEK293 , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Linfangiogênese , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Estresse Oxidativo , Receptores CXCR4/metabolismo , Transdução de Sinais , Síndrome de Stevens-Johnson/metabolismo , Síndrome de Stevens-Johnson/patologia , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Circulation ; 139(22): 2570-2584, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30922063

RESUMO

BACKGROUND: Heart failure, which is a major global health problem, is often preceded by pathological cardiac hypertrophy. The expansion of the cardiac vasculature, to maintain adequate supply of oxygen and nutrients, is a key determinant of whether the heart grows in a physiological compensated manner or a pathological decompensated manner. Bidirectional endothelial cell (EC)-cardiomyocyte (CMC) cross talk via cardiokine and angiocrine signaling plays an essential role in the regulation of cardiac growth and homeostasis. Currently, the mechanisms involved in the EC-CMC interaction are not fully understood, and very little is known about the EC-derived signals involved. Understanding how an excess of angiogenesis induces cardiac hypertrophy and how ECs regulate CMC homeostasis could provide novel therapeutic targets for heart failure. METHODS: Genetic mouse models were used to delete vascular endothelial growth factor (VEGF) receptors, adeno-associated viral vectors to transduce the myocardium, and pharmacological inhibitors to block VEGF and ErbB signaling in vivo. Cell culture experiments were used for mechanistic studies, and quantitative polymerase chain reaction, microarrays, ELISA, and immunohistochemistry were used to analyze the cardiac phenotypes. RESULTS: Both EC deletion of VEGF receptor (VEGFR)-1 and adeno-associated viral vector-mediated delivery of the VEGFR1-specific ligands VEGF-B or placental growth factor into the myocardium increased the coronary vasculature and induced CMC hypertrophy in adult mice. The resulting cardiac hypertrophy was physiological, as indicated by preserved cardiac function and exercise capacity and lack of pathological gene activation. These changes were mediated by increased VEGF signaling via endothelial VEGFR2, because the effects of VEGF-B and placental growth factor on both angiogenesis and CMC growth were fully inhibited by treatment with antibodies blocking VEGFR2 or by endothelial deletion of VEGFR2. To identify activated pathways downstream of VEGFR2, whole-genome transcriptomics and secretome analyses were performed, and the Notch and ErbB pathways were shown to be involved in transducing signals for EC-CMC cross talk in response to angiogenesis. Pharmacological or genetic blocking of ErbB signaling also inhibited part of the VEGF-B-induced effects in the heart. CONCLUSIONS: This study reveals that cross talk between the EC VEGFR2 and CMC ErbB signaling pathways coordinates CMC hypertrophy with angiogenesis, contributing to physiological cardiac growth.


Assuntos
Cardiomegalia/metabolismo , Células Endoteliais/metabolismo , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica , Comunicação Parácrina , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Receptores ErbB/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/patologia , Receptor Cross-Talk , Receptores Notch/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
10.
Cancer Sci ; 111(7): 2400-2412, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32391593

RESUMO

Escape of cancer cells from chemotherapy is a problem in the management of cancer patients. Research on chemotherapy resistance has mainly focused on the heterogeneity of cancer cells, multiple gene mutations, and quiescence of malignant cancer cells. However, some studies have indicated that interactions between cancer cells and vascular cells promote resistance to chemotherapy. Here, we established mouse leukemia models using the cell lines THP-1 or MEG-1. These were derived from acute and chronic myeloid leukemias, respectively, and highly expressed DNA replication factor PSF1, a member of the GINS complex. We found that, after anti-cancer drug administration, surviving GFP-positive leukemia cells in the bone marrow were located adjacent to blood vessels, as previously reported in a subcutaneous solid tumor transplantation model. Treating THP-1 and MEG-1 cells with anti-cancer drugs in vitro revealed that those most strongly expressing PSF1 were most chemoresistant, suggesting that PSF1 induces not only cell cycle progression but also facilitates cell survival. Indeed, when PSF1 expression was suppressed by shRNA, the growth rate was reduced and cell death was enhanced in both cell lines. Furthermore, PSF1 knockdown in leukemia cells led to a change in their location at a distance from the blood vessels in a bone marrow transplantation model. These findings potentially reflect a mechanism of escape of leukemic cells from chemotherapy and suggest that PSF1 may be a possible therapeutic target to enhance the effect of chemotherapy.


Assuntos
Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Ciclo Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Expressão Gênica , Leucemia/genética , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Antineoplásicos/farmacologia , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Development ; 144(13): 2392-2401, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28576770

RESUMO

The placental labyrinth is the interface for gas and nutrient exchange between the embryo and the mother; hence its proper development is essential for embryogenesis. However, the molecular mechanism underlying development of the placental labyrinth, particularly in terms of its endothelial organization, is not well understood. Here, we determined that fibronectin leucine-rich transmembrane protein 2 (FLRT2), a repulsive ligand of the UNC5 receptor family for neurons, is unexpectedly expressed in endothelial cells specifically in the placental labyrinth. Mice lacking FLRT2 in endothelial cells exhibited embryonic lethality at mid-gestation, with systemic congestion and hypoxia. Although they lacked apparent deformities in the embryonic vasculature and heart, the placental labyrinths of these embryos exhibited aberrant alignment of endothelial cells, which disturbed the feto-maternal circulation. Interestingly, this vascular deformity was related to endothelial repulsion through binding to the UNC5B receptor. Our results suggest that the proper organization of the placental labyrinth depends on coordinated inter-endothelial repulsion, which prevents uncontrolled layering of the endothelium.


Assuntos
Glicoproteínas de Membrana/metabolismo , Organogênese , Placenta/embriologia , Placenta/metabolismo , Transdução de Sinais , Animais , Sobrevivência Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células Endoteliais/metabolismo , Feminino , Deleção de Genes , Hipóxia/patologia , Glicoproteínas de Membrana/deficiência , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Receptores de Netrina , Placenta/irrigação sanguínea , Placenta/citologia , Gravidez , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/metabolismo
12.
Am J Pathol ; 189(4): 900-910, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30653955

RESUMO

Galectin-3 (Gal-3; gene LGALS3) is a member of the ß-galactose-binding lectin family. Previous studies showed that Gal-3 is expressed in several tissues across species and functions as a regulator of cell proliferation, apoptosis, adhesion, and migration, thus affecting many aspects of events, such as angiogenesis and tumorigenesis. Although several reports have suggested that the level of Gal-3 expression correlates positively with tumor progression, herein we show that highly metastatic mouse melanoma B16/BL6 cells express less Gal-3 than B16 cells with a lower metastatic potential. It was found that overexpression of Gal-3 in melanoma cells in fact suppresses metastasis. In contrast, knocking out Gal-3 expression in cancer cells promoted cell aggregation mediated through interactions with platelets and fibrinogen in vitro and increased the number of metastatic foci in vivo. Thus, reduced Gal-3 expression results in the up-regulation of ß3 integrin expression, and this contributes to metastatic potential. These findings indicate that changes of Gal-3 expression in cancer cells during tumor progression influence the characteristics of metastatic cells.


Assuntos
Galectina 3/fisiologia , Regulação Neoplásica da Expressão Gênica , Integrina beta3/metabolismo , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/prevenção & controle , Neovascularização Patológica/prevenção & controle , Animais , Apoptose , Adesão Celular , Proliferação de Células , Humanos , Integrina beta3/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus
13.
Pediatr Int ; 62(3): 257-304, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32202048

RESUMO

The objective was to prepare guidelines to perform the current optimum treatment by organizing effective and efficient treatments of hemangiomas and vascular malformations, confirming the safety, and systematizing treatment, employing evidence-based medicine (EBM) techniques and aimed at improvement of the outcomes. Clinical questions (CQs) were decided based on the important clinical issues. For document retrieval, key words for literature searches were set for each CQ and literature published from 1980 to the end of September 2014 was searched in Pubmed, Cochrane Library, and Japana Centra Revuo Medicina (JCRM). The strengths of evidence and recommendations acquired by systematic reviews were determined following the Medical Information Network Distribution System (MINDS) technique. A total of 33 CQs were used to compile recommendations and the subjects included efficacy of resection, sclerotherapy/embolization, drug therapy, laser therapy, radiotherapy, and other conservative treatment, differences in appropriate treatment due to the location of lesions and among symptoms, appropriate timing of treatment and tests, and pathological diagnosis deciding the diagnosis. Thus, the Japanese Clinical Practice Guidelines for Vascular Anomalies 2017 have been prepared as the evidence-based guidelines for the management of vascular anomalies.


Assuntos
Hemangioma/terapia , Malformações Vasculares/terapia , Malformações Arteriovenosas/terapia , Embolização Terapêutica/métodos , Medicina Baseada em Evidências , Humanos , Terapia a Laser/métodos , Escleroterapia/métodos , Resultado do Tratamento
14.
Rinsho Ketsueki ; 61(9): 1440-1445, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-33162547

RESUMO

Angiogenesis is the process of new vascular formation from preexisting blood vessels, and the contribution of endothelial stem cell population to this process has been elucidated. Vascular endothelial growth factor (VEGF) promotes not only the proliferation of endothelial cells (ECs) but also the vascular permeability by inducing cell-to-cell dissociation between ECs. Chronic hyper vascular leakage induces intra-tumoral interstitial hypertension resulting in disturbed vascular perfusion. Hypoxia in the nonfunctional vasculatures causes chromosomal instability of cancer cells resulting in the development of malignant cancer cells, such as cancer stem cells. Hypoxia also induces exhaustion of immune cells. Moreover, the blood vessel structures in tumors are abnormal and nonfunctional, and the infiltration of leukocytes are suppressed. Moreover, drug delivery, including anti-cancer drugs and immune checkpoint inhibitors, is limited. A drug inducing normalization or maturation of abnormal blood vessels in tumor is warranted for the improvement of malignant tumor microenvironment.


Assuntos
Antineoplásicos , Imunoterapia , Neoplasias , Microambiente Tumoral , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Células Endoteliais , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neovascularização Patológica/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular
15.
Am J Pathol ; 188(5): 1300-1314, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29650228

RESUMO

Vascular endothelial cells (ECs) isolated from tumors characteristically express certain genes. It has recently been suggested that tumor vessel normalization facilitates effective drug delivery into tumors; however, how tumor vessel normalization can be recognized on the basis of the molecules expressed by tumor ECs is not clearly defined. The degree of cell proliferation is an important indicator to characterize the condition of the ECs. Herein, we generated transgenic mice expressing enhanced green fluorescent protein (EGFP) under the transcriptional control of the DNA replication factor partner of Sld5-1 (PSF1; official name GINS1) promoter to assess whether active ECs can be distinguished from dormant ECs. Predictably, ECs in the adult skin exhibited no EGFP signals. However, after s.c. injection of tumor cells, some ECs shifted to EGFP positivity, enabling distinction of EGFP-positive from EGFP-negative cells. We found that only a fraction of the EGFP-negative ECs strongly expressed the glycosylphosphatidylinositol-anchor protein CD109 associated with the phosphatidylinositol 3-kinase pathway. Taken together, these data indicate that areas of vascular normalization in tumors can be detected by CD109 expression, and this provides a window of opportunity for timing chemotherapy.


Assuntos
Proteínas de Transporte/metabolismo , Células Endoteliais/metabolismo , Regiões Promotoras Genéticas , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Células Endoteliais/patologia , Camundongos , Microscopia de Fluorescência , Transplante de Neoplasias
16.
Int Immunol ; 30(2): 69-78, 2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29528433

RESUMO

Schlafen-8 (Slfn8) is a member of the Schlafen family of proteins, which harbor helicase domains and are induced by LPS and interferons. It has been reported that the Schlafen family are involved in various cellular functions, including proliferation, differentiation and regulation of virus replication. Slfn8 has been implicated in T-cell differentiation in the thymus. However, the roles of Slfn8 in the immune system remains unclear. In this study, we generated Slfn8 knockout mice (Slfn8-/-) and investigated the immunological role of Slfn8 using the T-cell-mediated autoimmune model experimental autoimmune encephalomyelitis (EAE). We found that the clinical score was reduced in Slfn8-/- mice. IL-6 and IL-17A cytokine production, which are associated with EAE onset and progression, were decreased in the lymph nodes of Slfn8-/- mice. Immune cell populations in Slfn8-/- mice, including macrophages, neutrophils, T cells and B cells, did not reveal significant differences compared with wild-type mice. In vitro activation of Slfn8-/- T cells in response to TCR stimulation also did not reveal significant differences. To confirm the involvement of non-hematopoietic cells, we isolated CD45- CD31+ endothelial cells and CD45-CD31- gp38+ fibroblastic reticular cells by FACS sorting. We showed that the levels of IL-6 and Slfn8 mRNA in CD45- CD31+ endothelial cells were increased after EAE induction. In contrast, the level of IL-6 mRNA after EAE induction was markedly decreased in CD31+ endothelial cells from Slfn8-/- mice. These results indicate that Slfn8 may play a role in EAE by regulating inflammation in endothelial cells.


Assuntos
Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Células Endoteliais/metabolismo , Animais , Biomarcadores , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Imunidade Inata , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/efeitos adversos , Fragmentos de Peptídeos/efeitos adversos , Índice de Gravidade de Doença , Linfócitos T/imunologia , Linfócitos T/metabolismo
17.
Nature ; 495(7442): 524-8, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23515163

RESUMO

Macrophages consist of at least two subgroups, M1 and M2 (refs 1-3). Whereas M1 macrophages are proinflammatory and have a central role in host defence against bacterial and viral infections, M2 macrophages are associated with responses to anti-inflammatory reactions, helminth infection, tissue remodelling, fibrosis and tumour progression. Trib1 is an adaptor protein involved in protein degradation by interacting with COP1 ubiquitin ligase. Genome-wide association studies in humans have implicated TRIB1 in lipid metabolism. Here we show that Trib1 is critical for the differentiation of F4/80(+)MR(+) tissue-resident macrophages--that share characteristics with M2 macrophages (which we term M2-like macrophages)--and eosinophils but not for the differentiation of M1 myeloid cells. Trib1 deficiency results in a severe reduction of M2-like macrophages in various organs, including bone marrow, spleen, lung and adipose tissues. Aberrant expression of C/EBPα in Trib1-deficient bone marrow cells is responsible for the defects in macrophage differentiation. Unexpectedly, mice lacking Trib1 in haematopoietic cells show diminished adipose tissue mass accompanied by evidence of increased lipolysis, even when fed a normal diet. Supplementation of M2-like macrophages rescues the pathophysiology, indicating that a lack of these macrophages is the cause of lipolysis. In response to a high-fat diet, mice lacking Trib1 in haematopoietic cells develop hypertriglyceridaemia and insulin resistance, together with increased proinflammatory cytokine gene induction. Collectively, these results demonstrate that Trib1 is critical for adipose tissue maintenance and suppression of metabolic disorders by controlling the differentiation of tissue-resident M2-like macrophages.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Contagem de Células , Proteínas de Ciclo Celular/deficiência , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Citocinas/genética , Dieta Hiperlipídica/efeitos adversos , Eosinófilos/citologia , Eosinófilos/metabolismo , Feminino , Hipertrigliceridemia/induzido quimicamente , Hipertrigliceridemia/genética , Mediadores da Inflamação/metabolismo , Resistência à Insulina/genética , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipodistrofia/induzido quimicamente , Lipodistrofia/metabolismo , Lipodistrofia/patologia , Lipólise , Pulmão/citologia , Macrófagos/classificação , Masculino , Camundongos , Neutrófilos/citologia , Neutrófilos/metabolismo , Especificidade de Órgãos , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Baço/citologia , Ubiquitina/metabolismo
18.
Neurobiol Dis ; 120: 51-62, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30176352

RESUMO

Krabbe disease (KD), or globoid cell leukodystrophy, is an inherited lysosomal storage disease with leukodystrophy caused by a mutation in the galactosylceramidase (GALC) gene. The majority of patients show the early onset form of KD dominated by cerebral demyelination with apoptotic oligodendrocyte (OL) death. However, the initial pathophysiological changes in developing OLs remain poorly understood. Here, we show that OLs of twitcher mice, an authentic mouse model of KD, exhibited developmental defects and impaired myelin formation in vivo and in vitro. In twitcher mouse brain, abnormal myelination and reduced expression of myelin genes during the period of most active OL differentiation and myelination preceded subsequent progressive OL death and demyelination. Importantly, twitcher mouse OL precursor cells proliferated normally, but their differentiation and survival were intrinsically defective. These defects were associated with aberrant accumulation of endogenous psychosine (galactosylsphingosine) and reduced activation of the Erk1/2 and Akt/mTOR pathways before apoptotic cell death. Collectively, our results demonstrate that GALC deficiency in developing KD OLs profoundly affects their differentiation and maturation, indicating the critical contribution of OL dysfunction to KD pathogenesis.


Assuntos
Modelos Animais de Doenças , Leucodistrofia de Células Globoides/metabolismo , Oligodendroglia/metabolismo , Psicosina/metabolismo , Animais , Proliferação de Células/fisiologia , Células Cultivadas , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligodendroglia/patologia , Psicosina/genética
19.
Circ J ; 83(1): 12-17, 2018 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-30487375

RESUMO

The roles that blood vessels play in the maintenance of organs and tissues in addition to the delivery of oxygen and nutrients are being gradually clarified. The maintenance of tissue-specific organ stem cells, such as hematopoietic and neuronal stem cells, is supported by endothelial cells (ECs), which represent an important component of the stem cell niche. The maintenance of organogenesis, for example, osteogenesis and liver generation/regeneration, is supported by molecules referred to as "angiocrine signals" secreted by EC. The mechanisms responsible for the well-known functions of blood vessels, such as thermoregulation and metabolism, especially removal of local metabolites, have now been determined at the molecular level. Following the development of single-cell genetic analysis, blood cell heterogeneity, especially of mural cell populations, has been established and tissue-specific blood vessel formation and function are now also understood at the molecular level. Among the heterogeneous populations of ECs, it seems that a stem cell population with the ability to maintain the production of ECs long-term is present in pre-existing blood vessels. Neovascularization by therapeutic angiogenesis yields benefits in many diseases, not only ischemic disease but also metabolic disease and other vascular diseases. Therefore, vascular endothelial stem cells should be considered to use in vascular regeneration therapy.


Assuntos
Células Progenitoras Endoteliais/metabolismo , Isquemia/metabolismo , Doenças Metabólicas/metabolismo , Neovascularização Fisiológica , Regeneração , Doenças Vasculares/metabolismo , Animais , Células Progenitoras Endoteliais/patologia , Humanos , Isquemia/patologia , Isquemia/terapia , Doenças Metabólicas/patologia , Doenças Metabólicas/terapia , Doenças Vasculares/patologia , Doenças Vasculares/terapia
20.
World J Surg ; 42(9): 2879-2886, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29511870

RESUMO

BACKGROUND: PDGFR-ß is used as a stromal biomarker and is functional in mesenchymal cells of the tumor microenvironment. The significance of stromal PDGFR-ß expression in non-small cell lung cancer (NSCLC) in patients undergoing preoperative chemo- or chemoradiotherapy had not been determined. METHODS: Patients with NSCLC undergoing preoperative chemo- or chemoradiotherapy between 1996 and 2014 were assessed for expression of stromal PDGFR-ß by immunohistochemistry using resected specimens. Relationships between stromal PDGFR-ß expression and survival after operation were analyzed. Forty-three patients who underwent surgery without preoperative treatment in 2005 were also analyzed as a chemo-naïve control group. RESULTS: The mean age of the 92 patients was 60.2 years. Seventy-eight (85%) were male, and 14 (15%) were female. Fifty-four patients (59%) underwent preoperative chemoradiotherapy, and 38 patients (41%) underwent preoperative chemotherapy. Regimens for preoperative chemotherapy were cisplatin (CDDP) based in 48 patients (52%) and carboplatin (CBDCA) based in 43 (42%). While stromal cells expressed PDGFR-ß in 21 chemo-naïve patients (49%), stromal cells expressed PDGFR-ß in 65 patients who underwent preoperative therapy (p = 0.02). The 5-year disease-free survival rate (DFS) of the PDGFR-ß-positive group was significantly worse than that of the negative group (27 vs. 48%, p = 0.04). The 5-year disease-specific survival rate (DSS) in the stromal PDGFR-ß-positive group was also significantly worse than in the negative group (43 vs. 70%, p = 0.01). On the other hand, stromal PDGFR-ß expression did not influence survival in chemo-naïve patients. CONCLUSIONS: Stromal PDGFR-ß expression is negatively associated with DFS and DSS in patients with NSCLC undergoing preoperative chemo- or chemoradiotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Quimiorradioterapia , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Adulto , Idoso , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/terapia , Cisplatino , Feminino , Humanos , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/terapia , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante , Período Pós-Operatório , Taxa de Sobrevida , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa