Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
HNO ; 70(5): 380-388, 2022 May.
Artigo em Alemão | MEDLINE | ID: mdl-35420312

RESUMO

BACKGROUND: A multitude of vascular anomalies exist and can lead to severe complications. Treatment can be complex. OBJECTIVE: This overview aims to provide important information for the management of vascular anomalies. MATERIALS AND METHODS: In addition to current literature, experiences from the interdisciplinary Vascular Anomalies Center in Marburg were included in this review. RESULTS: Hemangiomas at critical sites, arteriovenous malformations, and vascular anomalies of uncertain etiology require particular attention. CONCLUSION: Self-help and support groups, specialized interdisciplinary centers, scientific medical societies, and networks can provide help for the treatment of vascular anomalies.


Assuntos
Malformações Arteriovenosas , Hemangioma , Malformações Vasculares , Malformações Arteriovenosas/diagnóstico , Malformações Arteriovenosas/terapia , Hemangioma/diagnóstico , Hemangioma/terapia , Humanos , Malformações Vasculares/diagnóstico , Malformações Vasculares/terapia
2.
Cell Mol Life Sci ; 79(2): 88, 2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35067832

RESUMO

Junctional adhesion molecule (JAM)-A is a cell adhesion receptor localized at epithelial cell-cell contacts with enrichment at the tight junctions. Its role during cell-cell contact formation and epithelial barrier formation has intensively been studied. In contrast, its role during collective cell migration is largely unexplored. Here, we show that JAM-A regulates collective cell migration of polarized epithelial cells. Depletion of JAM-A in MDCK cells enhances the motility of singly migrating cells but reduces cell motility of cells embedded in a collective by impairing the dynamics of cryptic lamellipodia formation. This activity of JAM-A is observed in cells grown on laminin and collagen-I but not on fibronectin or vitronectin. Accordingly, we find that JAM-A exists in a complex with the laminin- and collagen-I-binding α3ß1 integrin. We also find that JAM-A interacts with tetraspanins CD151 and CD9, which both interact with α3ß1 integrin and regulate α3ß1 integrin activity in different contexts. Mapping experiments indicate that JAM-A associates with α3ß1 integrin and tetraspanins CD151 and CD9 through its extracellular domain. Similar to depletion of JAM-A, depletion of either α3ß1 integrin or tetraspanins CD151 and CD9 in MDCK cells slows down collective cell migration. Our findings suggest that JAM-A exists with α3ß1 integrin and tetraspanins CD151 and CD9 in a functional complex to regulate collective cell migration of polarized epithelial cells.


Assuntos
Moléculas de Adesão Celular/metabolismo , Integrina alfa3beta1/metabolismo , Tetraspanina 24/metabolismo , Tetraspanina 29/metabolismo , Animais , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Cães , Doxorrubicina/farmacologia , Humanos , Molécula A de Adesão Juncional/antagonistas & inibidores , Molécula A de Adesão Juncional/genética , Células Madin Darby de Rim Canino , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo
3.
Nat Commun ; 12(1): 3624, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131132

RESUMO

The LIM and SH3 domain protein 1 (Lasp1) was originally cloned from metastatic breast cancer and characterised as an adaptor molecule associated with tumourigenesis and cancer cell invasion. However, the regulation of Lasp1 and its function in the aggressive transformation of cells is unclear. Here we use integrative epigenomic profiling of invasive fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA) and from mouse models of the disease, to identify Lasp1 as an epigenomically co-modified region in chronic inflammatory arthritis and a functionally important binding partner of the Cadherin-11/ß-Catenin complex in zipper-like cell-to-cell contacts. In vitro, loss or blocking of Lasp1 alters pathological tissue formation, migratory behaviour and platelet-derived growth factor response of arthritic FLS. In arthritic human TNF transgenic mice, deletion of Lasp1 reduces arthritic joint destruction. Therefore, we show a function of Lasp1 in cellular junction formation and inflammatory tissue remodelling and identify Lasp1 as a potential target for treating inflammatory joint disorders associated with aggressive cellular transformation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Junções Aderentes/metabolismo , Artrite/metabolismo , Transformação Celular Neoplásica/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Artrite/patologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Caderinas/metabolismo , Proteínas do Citoesqueleto/genética , Feminino , Proteínas de Homeodomínio , Proteínas com Domínio LIM/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos , beta Catenina/metabolismo
4.
J Biol Chem ; 296: 100136, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33268376

RESUMO

To migrate, cells assume a polarized morphology, extending forward with a leading edge with their trailing edge retracting back toward the cell body. Both cell extension and retraction critically depend on the organization and dynamics of the actin cytoskeleton, and the small, monomeric GTPases Rac and Rho are important regulators of actin. Activation of Rac induces actin polymerization and cell extension, whereas activation of Rho enhances acto-myosin II contractility and cell retraction. To coordinate migration, these processes must be carefully regulated. The myosin Myo9b, a Rho GTPase-activating protein (GAP), negatively regulates Rho activity and deletion of Myo9b in leukocytes impairs cell migration through increased Rho activity. However, it is not known whether cell motility is regulated by global or local inhibition of Rho activity by Myo9b. Here, we addressed this question by using Myo9b-deficient macrophage-like cells that expressed different recombinant Myo9b constructs. We found that Myo9b accumulates in lamellipodial extensions generated by Rac-induced actin polymerization as a function of its motor activity. Deletion of Myo9b in HL-60-derived macrophages altered cell morphology and impaired cell migration. Reintroduction of Myo9b or Myo9b motor and GAP mutants revealed that local GAP activity rescues cell morphology and migration. In summary, Rac activation leads to actin polymerization and recruitment of Myo9b, which locally inhibits Rho activity to enhance directional cell migration.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Miosinas/metabolismo , Pseudópodes/metabolismo , Movimento Celular/fisiologia , Células Cultivadas , Proteínas Ativadoras de GTPase/genética , Humanos , Miosinas/genética
5.
Cell Rep ; 29(4): 1010-1026.e6, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31644899

RESUMO

Actin-binding proteins are essential for linear and branched actin filament dynamics that control shape change, cell migration, and cell junction remodeling in vascular endothelium (endothelial cells [ECs]). The epithelial protein lost in neoplasm (EPLIN) is an actin-binding protein, expressed as EPLIN-α and EPLIN-ß by alternative promoters; however, the isoform-specific functions are not yet understood. Aortic compared to cava vein ECs and shear stress-exposed cultured ECs express increased EPLIN-ß levels that stabilize stress fibers. In contrast, EPLIN-α expression is increased in growing and migrating ECs, is targeted to membrane protrusions, and terminates their growth via interaction with the Arp2/3 complex. The data indicate that EPLIN-α controls protrusion dynamics while EPLIN-ß has an actin filament stabilizing role, which is consistent with FRAP analyses demonstrating a lower EPLIN-ß turnover rate compared to EPLIN-α. Together, EPLIN isoforms differentially control actin dynamics in ECs, essential in shear stress responses, cell migration, and barrier function.


Assuntos
Actinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Endotélio Vascular/metabolismo , Animais , Aorta/citologia , Aorta/metabolismo , Proliferação de Células , Proteínas do Citoesqueleto/genética , Endotélio Vascular/citologia , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fibras de Estresse/metabolismo
6.
Biophys J ; 116(8): 1547-1559, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30878197

RESUMO

Blood vessels are covered with endothelial cells on their inner surfaces, forming a selective and semipermeable barrier between the blood and the underlying tissue. Many pathological processes, such as inflammation or cancer metastasis, are accompanied by an increased vascular permeability. Progress in live cell imaging techniques has recently revealed that the structure of endothelial cell contacts is constantly reorganized and that endothelial junctions display high heterogeneities at a subcellular level even within one cell. Although it is assumed that this dynamic remodeling is associated with a local change in endothelial barrier function, a direct proof is missing mainly because of a lack of appropriate experimental techniques. Here, we describe a new assay to dynamically measure local endothelial barrier function with a lateral resolution of ∼15 µm and a temporal resolution of 1 min. In this setup, fluorescence-labeled molecules are added to the apical compartment of an endothelial monolayer, and the penetration of molecules from the apical to the basal compartment is recorded by total internal reflection fluorescence microscopy utilizing the generated evanescent field. With this technique, we found a remarkable heterogeneity in the local permeability for albumin within confluent endothelial cell layers. In regions with low permeability, stimulation with the proinflammatory agent histamine results in a transient increase in paracellular permeability. The effect showed a high variability along the contact of one individual cell, indicating a local regulation of endothelial barrier function. In regions with high basal permeability, histamine had no obvious effect. In contrast, the barrier-enhancing drug forskolin reduces the permeability for albumin and dextran uniformly along the cell junctions. Because this new approach can be readily combined with other live cell imaging techniques, it will contribute to a better understanding of the mechanisms underlying subcellular junctional reorganization during wound healing, inflammation, and angiogenesis.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Microscopia/métodos , Sobrevivência Celular/efeitos dos fármacos , Colforsina/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Processamento de Imagem Assistida por Computador , Cinética , Permeabilidade/efeitos dos fármacos
7.
Ann Anat ; 221: 179-185, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30393181

RESUMO

The practice of human and veterinary medicine is based on the science of anatomy and dissection courses are still irreplaceable in the teaching of anatomy. Embalming is required to preserve body donors, for which process formaldehyde (FA) is the most frequently used and well characterized biocidal substance. Since January 2016, a new occupational exposure limit (OEL) for FA of 0.37mg/m3 issued by the European Committee on Hazardous Substances is obligatory since FA has been classified as a human 1B carcinogen. The anatomical institutes in the German-speaking region are called upon to consolidate efforts to reduce use of FA in anatomical curricula and body donations. As a result, the Anatomische Gesellschaft (AG) has formed a "Working Group for Reduction of Formaldehyde Exposure in Dissection Courses" tasked with discussion and recommendation of measures to reduce FA. Based on the assessment of the Working Group, the AG has issued an official opinion to the effect that, at this point in time, embalming of body donors without FA completely is not feasible. Therefore, a combination of approaches are to be used to reduce FA exposure, including technical and structural (architectural) adaptations, modification of protocols for fixation and preservation as well as organizational measures. One structural measure considered unavoidable is the integration of air supply and exhaust of individual dissecting tables into the ventilation system of the anatomy building. To embalm human body donors, intra-arterial perfusion fixation with up to 4% FA and a total fluid volume of 150mL/kg body weight will suffice. For animals where body weights and biology of bodies vary widely (i.e. special needs of fixation for ruminants, large animals as horses) perfusion fixation with up to 4% FA and a quantity of fixative solution of 10-15% of the body weight may be required. Preservation of body donors in storage (immersion) can be done with 40% ethanol or in a full bath preservation containing up to 2% FA. Corpse humidification in the dissecting room is possible with 2% phenoxyethanol, in each case without FA. In veterinary anatomy, microbiological burden is often higher and therefore might lead to a need of FA in long-time storage. Compliance with the current OEL in all institutes would appear to be feasible in combination with various organizational measures.


Assuntos
Anatomia/educação , Formaldeído/efeitos adversos , Exposição Ocupacional/prevenção & controle , Hipersensibilidade Respiratória/prevenção & controle , Humanos , Guias de Prática Clínica como Assunto
8.
Histochem Cell Biol ; 149(1): 15-30, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29143117

RESUMO

The cadherin switch has profound consequences on cancer invasion and metastasis. The endothelial-specific vascular endothelial cadherin (VE-cadherin) has been demonstrated in diverse cancer types including breast cancer and is supposed to modulate tumor progression and metastasis, but underlying mechanisms need to be better understood. First, we evaluated VE-cadherin expression by tissue microarray in 392 cases of breast cancer tumors and found a diverse expression and distribution of VE-cadherin. Experimental expression of fluorescence-tagged VE-cadherin (VE-EGFP) in undifferentiated, fibroblastoid and E-cadherin-negative MDA-231 (MDA-VE-EGFP) as well as in differentiated E-cadherin-positive MCF-7 human breast cancer cell lines (MCF-VE-EGFP), respectively, displayed differentiation-dependent functional differences. VE-EGFP expression reversed the fibroblastoid MDA-231 cells to an epithelial-like phenotype accompanied by increased ß-catenin expression, actin and vimentin remodeling, increased cell spreading and barrier function and a reduced migration ability due to formation of VE-cadherin-mediated cell junctions. The effects were largely absent in both MDA-VE-EGFP and in control MCF-EGFP cell lines. However, MCF-7 cells displayed a VE-cadherin-independent planar cell polarity and directed cell migration that both developed in MDA-231 only after VE-EGFP expression. Furthermore, VE-cadherin expression had no effect on tumor cell proliferation in monocultures while co-culturing with endothelial cells enhanced tumor cell proliferation due to integration of the tumor cells into monolayer where they form VE-cadherin-mediated cell contacts with the endothelium. We propose an interactive VE-cadherin-based crosstalk that might activate proliferation-promoting signals. Together, our study shows a VE-cadherin-mediated cell dynamics and an endothelial-dependent proliferation in a differentiation-dependent manner.


Assuntos
Antígenos CD/biossíntese , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/biossíntese , Diferenciação Celular , Células Endoteliais/metabolismo , Antígenos CD/metabolismo , Caderinas/metabolismo , Proliferação de Células , Células Endoteliais/citologia , Feminino , Humanos , Células MCF-7 , Células Tumorais Cultivadas
9.
Nat Commun ; 8(1): 2210, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29263363

RESUMO

VEGFR-2/Notch signalling regulates angiogenesis in part by driving the remodelling of endothelial cell junctions and by inducing cell migration. Here, we show that VEGF-induced polarized cell elongation increases cell perimeter and decreases the relative VE-cadherin concentration at junctions, triggering polarized formation of actin-driven junction-associated intermittent lamellipodia (JAIL) under control of the WASP/WAVE/ARP2/3 complex. JAIL allow formation of new VE-cadherin adhesion sites that are critical for cell migration and monolayer integrity. Whereas at the leading edge of the cell, large JAIL drive cell migration with supportive contraction, lateral junctions show small JAIL that allow relative cell movement. VEGFR-2 activation initiates cell elongation through dephosphorylation of junctional myosin light chain II, which leads to a local loss of tension to induce JAIL-mediated junctional remodelling. These events require both microtubules and polarized Rac activity. Together, we propose a model where polarized JAIL formation drives directed cell migration and junctional remodelling during sprouting angiogenesis.


Assuntos
Actinas/metabolismo , Antígenos CD/metabolismo , Caderinas/metabolismo , Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Células Endoteliais/metabolismo , Junções Intercelulares/metabolismo , Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína 2 Relacionada a Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Proteína 3 Relacionada a Actina/metabolismo , Actinas/efeitos dos fármacos , Antígenos CD/efeitos dos fármacos , Caderinas/efeitos dos fármacos , Miosinas Cardíacas/metabolismo , Adesão Celular , Movimento Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Endotélio Vascular , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Modelos Cardiovasculares , Cadeias Leves de Miosina/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Pseudópodes/fisiologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Remodelação Vascular , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo
10.
Pflugers Arch ; 469(10): 1401-1412, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28550472

RESUMO

High dietary salt intake may lead to vascular stiffness, which predicts cardiovascular diseases such as heart failure, and myocardial and cerebral infarctions as well as renal impairment. The vascular endothelium is a primary target for deleterious salt effects leading to dysfunction and endothelial stiffness. We hypothesize that the Ca2+- and bicarbonate-activated soluble adenylyl cyclase (sAC) contributes to Na+/K+-ATPase expression regulation in vascular endothelial cells and is an important regulator of endothelial stiffness. In vitro stimulation of vascular endothelial cells with high sodium (150 mM Na+)-induced Na+/K+-ATPase-α and Na+/K+-ATPase-ß protein expression determined by western blot. Promoter analyses revealed increased cAMP response element (CRE)-mediated Na+/K+-ATPase-α transcriptional activity under high sodium concentrations. Inhibition of sAC by the specific inhibitor KH7 or siRNA reduced the sodium effects. Flame photometry revealed increased intracellular sodium concentrations in response to high sodium stimulations, which were paralleled by elevated ATP levels. Using atomic force microscopy, a nano-technique that measures cellular stiffness and deformability, we detected significant endothelial stiffening under increased sodium concentrations, which was prevented by inhibition of sAC using KH7 and Na+/K+-ATPase using ouabain. Furthermore, analysis of primary aortic endothelial cells in an in vitro aging model revealed an impaired Na+/K+-ATPase-α sodium response and elevated intracellular sodium levels with cellular aging. We conclude that sAC mediates sodium-induced Na+/K+-ATPase expression in vascular endothelium and is an important regulator of endothelial stiffness. The reactivity of Na+/K+-ATPase-α expression regulation in response to high sodium seems to be impaired in aging endothelial cells and might be a component of endothelial dysfunction.


Assuntos
Adenilil Ciclases/metabolismo , Células Endoteliais/metabolismo , Cloreto de Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Sódio/metabolismo , Animais , Endotélio Vascular/metabolismo , Ouabaína/farmacologia , Cloreto de Sódio na Dieta/metabolismo
11.
Nat Commun ; 8: 14832, 2017 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-28327544

RESUMO

Migration frequently involves Rac-mediated protrusion of lamellipodia, formed by Arp2/3 complex-dependent branching thought to be crucial for force generation and stability of these networks. The formins FMNL2 and FMNL3 are Cdc42 effectors targeting to the lamellipodium tip and shown here to nucleate and elongate actin filaments with complementary activities in vitro. In migrating B16-F1 melanoma cells, both formins contribute to the velocity of lamellipodium protrusion. Loss of FMNL2/3 function in melanoma cells and fibroblasts reduces lamellipodial width, actin filament density and -bundling, without changing patterns of Arp2/3 complex incorporation. Strikingly, in melanoma cells, FMNL2/3 gene inactivation almost completely abolishes protrusion forces exerted by lamellipodia and modifies their ultrastructural organization. Consistently, CRISPR/Cas-mediated depletion of FMNL2/3 in fibroblasts reduces both migration and capability of cells to move against viscous media. Together, we conclude that force generation in lamellipodia strongly depends on FMNL formin activity, operating in addition to Arp2/3 complex-dependent filament branching.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas/metabolismo , Pseudópodes/metabolismo , Animais , Fenômenos Biomecânicos , Sistemas CRISPR-Cas/genética , Movimento Celular , Fibroblastos/metabolismo , Forminas , Técnicas de Silenciamento de Genes , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Células NIH 3T3 , Fenótipo , Polimerização , Pseudópodes/ultraestrutura , Interferência de RNA
12.
Oncotarget ; 8(68): 112268-112282, 2017 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-29348824

RESUMO

Early metastasis leads to poor prognosis of lung cancer patients, whose 5-year survival rate is only 15%. We could recently show that the Ca2+ sensitive K+ channel KCa3.1 promotes aggressive behavior of non-small cell lung cancer (NSCLC) cells and that it can serve as a prognostic marker in NSCLC. Since NSCLC patients die of metastases, we investigated whether KCa3.1 channels contribute to poor patient prognosis by regulating distinct steps of the metastatic cascade. We investigated the extravasation of NSCLC cells and focused on their adhesion to endothelial cells and on transendothelial migration. We quantified the adhesion forces between NSCLC cells and endothelial cells by applying single cell force spectroscopy, and we monitored transendothelial migration using live-cell imaging. Inhibition of KCa3.1 channels with senicapoc or KCa3.1 silencing increases the adhesion force of A549 lung cancer cells to human microvascular endothelial cells (HMEC-1). Western blotting, immunofluorescence staining and biotinylation assays indicate that the elevated adhesion force is due to increased expression of ICAM-1 in both cell lines when KCa3.1 channels are downregulated. Consistent with this interpretation, an anti-ICAM-1 blocking antibody abolishes the KCa3.1-dependent increase in adhesion. Senicapoc inhibits transendothelial migration of A549 cells by 50%. Selectively silencing KCa3.1 channels in either NSCLC or endothelial cells reveals that transendothelial migration depends predominantly on endothelial KCa3.1 channels. In conclusion, our findings disclose a novel function of KCa3.1 channels in cancer. KCa3.1 channels regulate ICAM-1 dependent cell-cell adhesion between endothelial and cancer cells that affects the transmigration step of the metastatic cascade.

13.
Exp Dermatol ; 26(1): 73-81, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27576129

RESUMO

Although there is increasing evidence that oxidative stress is involved in collagen synthesis and myofibroblast activation, the NADPH oxidase (Nox) system is incompletely investigated in the context of human dermal fibroblasts (HDFs) and skin fibrosis. Using the pan-Nox inhibitor diphenyleneiodonium (DPI) as an initial tool, we show that gene expression of collagen type I, α-smooth muscle actin (α-SMA) and fibronectin 1 is suppressed in HDFs. Detailed expression analysis of all Nox isoforms and adaptors revealed expression of RNA and protein expression of Nox4, p22phox and Poldip2 but neither Nox1 nor Nox2. Nox4 could be immunolocalized to the endoplasmic reticulum. Importantly, TGF-ß1 had a dose- and time-dependent upregulating effect on NADH activity and Nox4 gene expression in HDFs. Genetic silencing of Nox4 as demonstrated by siRNA in HDFs as well as in murine fibroblasts established from Nox4 knockout mice confirmed that TGF-ß1 -mediated collagen type I gene, α-SMA and fibronectin 1 gene expressions were Nox4-dependent. This TGF-ß1 effect was mediated by Smad3 as shown by in silico promoter analysis, pharmacological inhibition and gene silencing of Smad3. The relevance of these findings is highlighted in the bleomycin-induced scleroderma mouse model. DPI treatment attenuated skin fibrosis and myofibroblast activation. Moreover, Nox4 knockdown by siRNA reduced skin collagen synthesis, α-SMA and fibronectin 1 expression in vivo. Finally, analyses of HDFs from patients with systemic sclerosis confirmed the expression of Nox4 and its adaptors, whereas Nox1 and Nox2 were not detectable. Our findings indicate that Nox4 targeting is a promising future treatment for fibrotic skin diseases.


Assuntos
Fibroblastos/enzimologia , NADPH Oxidase 4/genética , Escleroderma Sistêmico/enzimologia , Pele/enzimologia , Pele/patologia , Actinas/genética , Adulto , Animais , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Simulação por Computador , Citocinas/genética , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Fibronectinas , Fibrose , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Inativação Gênica , Humanos , Recém-Nascido , Isoenzimas/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Complexos Multienzimáticos/metabolismo , Miofibroblastos , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidase 1/genética , NADPH Oxidase 2/genética , NADPH Oxidase 4/metabolismo , Oniocompostos/farmacologia , Cultura Primária de Células , RNA Mensageiro/metabolismo , Escleroderma Sistêmico/induzido quimicamente , Escleroderma Sistêmico/patologia , Fator de Crescimento Transformador beta/farmacologia , Adulto Jovem
14.
Basic Res Cardiol ; 108(4): 362, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23744056

RESUMO

Arteries and veins show marked differences in their anatomy, physiology and genetic expression pattern. In this study, we analyzed impact of overexpression or downregulation of arterial marker gene Hey2 and venous marker gene COUP-TFII in human venous and arterial endothelial cells on genes involved in arteriovenous differentiation. Lentiviral overexpression of venous marker gene COUP-TFII in arterial endothelial cells led to downregulation of NICD4, arterial marker gene Hey2 and EphrinB2. Downregulation of Hey2 could be mediated by direct binding of COUP-TFII to Hey2 promoter as shown by ChIP, EMSA and promoter analysis. Downregulation of Hey2 by shRNA causes downregulation of EphrinB2 expression. Overexpression of arterial marker Hey2 in venous endothelial cells did not change expression pattern of COUP-TFII. Downregulation of venous marker gene COUP-TFII in venous endothelial cells resulted in upregulation of VEGF-A, Dll4 and EphrinB2 expression. Our data support an important role of Hey2 and COUP-TFII in arteriovenous differentiation of human endothelial cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fator II de Transcrição COUP/metabolismo , Diferenciação Celular/fisiologia , Endotélio Vascular/metabolismo , Proteínas Repressoras/metabolismo , Artérias Umbilicais/metabolismo , Veias Umbilicais/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Regulação para Baixo/fisiologia , Endotélio Vascular/citologia , Efrina-B2/metabolismo , Humanos , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Artérias Umbilicais/citologia , Veias Umbilicais/citologia , Regulação para Cima/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Mol Imaging Biol ; 15(6): 693-702, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23632953

RESUMO

PURPOSE: Profound changes of the vasculature in tumors critically impact drug delivery and therapy response. We aimed at developing a procedure to monitor morphological and functional parameters of the vasculature in subcutaneous xenograft models commonly applied for therapy testing by using probe-based confocal laser endomicroscopy. PROCEDURES: By monitoring various normal and diseased tissues, we established an experimental and analytical set-up to systematically analyze tracer extravasation from the microvasculature. Application of the approach in two xenograft models (HCT-116 and SW620) was realized consecutively throughout tumor growth. RESULTS: The incidence of dilated vessels increased with xenograft size in both models while macromolecule extravasation and tracer accumulation in the tumor tissue, respectively, was significantly reduced throughout growth. The development of dilated/ultradilated vessels correlated with tracer extravasation only in the HCT-116 but not the SW620 model. The underlying mechanisms are still ambiguous and discussed. CONCLUSIONS: Our findings clearly indicate that both xenograft type and size matter for drug delivery and therapy testing.


Assuntos
Permeabilidade Capilar , Extravasamento de Materiais Terapêuticos e Diagnósticos/patologia , Microscopia Confocal/métodos , Neovascularização Patológica/patologia , Animais , Feminino , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Músculos/irrigação sanguínea , Músculos/patologia , Neoplasias Experimentais , Língua/irrigação sanguínea , Língua/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Breast Cancer Res ; 14(6): R154, 2012 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-23216791

RESUMO

INTRODUCTION: Deregulation of cadherin expression, in particular the loss of epithelial (E)-cadherin and gain of neural (N)-cadherin, has been implicated in carcinoma progression. We previously showed that endothelial cell-specific vascular endothelial (VE)-cadherin can be expressed aberrantly on tumor cells both in human breast cancer and in experimental mouse mammary carcinoma. Functional analyses revealed that VE-cadherin promotes tumor cell proliferation and invasion by stimulating transforming growth factor (TGF)-ß signaling. Here, we investigate the functional interplay between N-cadherin and VE-cadherin in breast cancer. METHODS: The expression of N-cadherin and VE-cadherin was evaluated by immunohistochemistry in a tissue microarray with 84 invasive human breast carcinomas. VE-cadherin and N-cadherin expression in mouse mammary carcinoma cells was manipulated by RNA interference or overexpression, and cells were then analyzed by immunofluorescence, reverse transcriptase-polymerase chain reaction, and western blot. Experimental tumors were generated by transplantation of the modified mouse mammary carcinoma cells into immunocompetent mice. Tumor growth was monitored, and tumor tissue was subjected to histological analysis. RESULTS: VE-cadherin and N-cadherin were largely co-expressed in invasive human breast cancers. Silencing of N-cadherin in mouse mammary carcinoma cells led to decreased VE-cadherin expression and induced changes indicative of mesenchymal-epithelial transition, as indicated by re-induction of E-cadherin, localization of ß-catenin at the cell membrane, decreased expression of vimentin and SIP1, and gain of epithelial morphology. Suppression of N-cadherin expression also inhibited tumor growth in vivo, even when VE-cadherin expression was forced. CONCLUSIONS: Our results highlight the critical role of N-cadherin in breast cancer progression and show that N-cadherin is involved in maintaining the malignant tumor cell phenotype. The presence of N-cadherin prevents the re-expression of E-cadherin and localization of ß-catenin at the plasma membrane of mesenchymal mammary carcinoma cells. N-cadherin is also required to maintain the expression of VE-cadherin in malignant tumor cells but not vice versa. Thus, N-cadherin acts in concert with VE-cadherin to promote tumor growth.


Assuntos
Antígenos CD/biossíntese , Neoplasias da Mama/patologia , Caderinas/biossíntese , Endotélio Vascular/metabolismo , beta Catenina/metabolismo , Animais , Antígenos CD/genética , Mama/patologia , Caderinas/genética , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Progressão da Doença , Transição Epitelial-Mesenquimal , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Proteínas do Tecido Nervoso/biossíntese , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA/biossíntese , Vimentina/biossíntese
17.
J Infect Dis ; 204 Suppl 3: S947-52, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21987774

RESUMO

Ebola virus infection is associated with the release of a soluble glycoprotein (sGP) from infected cells. The sGP has been proposed to modulate Ebola virus pathogenesis in primates but little is known about the role of this protein during infection and disease manifestation. So far sGP has been shown to revert the effect of tumor necrosis factor α (TNF-α) on endothelial permeability, indicating that the function of sGP might be antiinflammatory. Since bystander apoptosis of lymphocytes has been demonstrated in Ebola virus infections, we aimed to investigate the ability of sGP to modulate lymphocyte apoptosis and adhesion of lymphocytes to activated endothelium. Recombinant sGP alone or together with TNF-α and the death receptors TRAIL and FAS neither increased nor decreased apoptosis of Jurkat cells, a well-established human lymphocytic cell line. In addition, Jurkat cell adhesion to native or activated human umbilical vein endothelial cells was also found to be not altered by sGP.


Assuntos
Apoptose/efeitos dos fármacos , Ebolavirus/metabolismo , Endotélio/fisiologia , Glicoproteínas/metabolismo , Linfócitos/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Glicoproteínas/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Células Jurkat , Linfócitos/citologia , Linfócitos/fisiologia , Receptores de Morte Celular/metabolismo , Internalização do Vírus
18.
J Infect Dis ; 204 Suppl 3: S957-67, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21987776

RESUMO

Virus entry into host cells is the first step of infection and a crucial determinant of pathogenicity. Here we show that Ebola virus-like particles (EBOV-VLPs) composed of the glycoprotein GP(1,2) and the matrix protein VP40 use macropinocytosis and clathrin-mediated endocytosis to enter cells. EBOV-VLPs applied to host cells induced actin-driven ruffling and enhanced FITC-dextran uptake, which indicated macropinocytosis as the main entry mechanism. This was further supported by inhibition of entry through inhibitors of actin polymerization (latrunculin A), Na(+)/H(+)-exchanger (EIPA), and PI3-kinase (wortmannin). A fraction of EBOV-VLPs, however, colocalized with clathrin heavy chain (CHC), and VLP uptake was reduced by CHC small interfering RNA transfection and expression of the dominant negative dynamin II-K44A mutant. In contrast, we found no evidence that EBOV-VLPs enter cells via caveolae. This work identifies macropinocytosis as the major, and clathrin-dependent endocytosis as an alternative, entry route for EBOV particles. Therefore, EBOV seems to utilize different entry pathways depending on both cell type and virus particle size.


Assuntos
Clatrina/fisiologia , Ebolavirus/fisiologia , Endocitose/fisiologia , Células Epiteliais/virologia , Internalização do Vírus , Actinas/fisiologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Androstadienos/farmacologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Caveolina 1/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Inativação Gênica , Interações Hospedeiro-Patógeno , Humanos , RNA Interferente Pequeno , Transdução de Sinais , Tiazolidinas/farmacologia , Replicação Viral , Wortmanina
19.
Thromb Haemost ; 102(6): 1135-43, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19967143

RESUMO

The need for uncovering molecular mechanisms in endothelial cell biology has tremendously increased in the last decades as it became more and more clear that the endothelium is an important target in nearly all diseases and treatments (drug delivery) and plays a central role in regeneration processes. One of the critical methods generally applied in cell biology research to uncover structural and functional aspects is the modulation of protein expression by over-expression, expression of mutant variants or gene silencing. This strategy, however, requires genetic manipulation of the respective cells. The classical gene transfer by chemical transfection techniques works pretty well in a large variety of cultured cells but fails for most endothelial cell types. Insufficient transfection rates and gene expression levels as well as the sensitivity of the endothelium against chemical transfection reagents limits utilisation of this technique for endothelial cell biology research. This holds true not only for primary endothelial cell cultures and endothelial cells in vivo but also for endothelial cell lines, e.g. endothelioma cells. The development of viral vectors originally designed for gene therapy approaches has significantly improved the methodological spectrum in endothelial cell research. Two viral vector systems, based on retroviruses and adenoviruses, deliver transgenic information highly efficient into both cultured endothelial cells and in endothelial cells in vivo, respectively. This review aims to give a comprehensive overview of these two vector systems that appear to be reliable and efficient tools for gene delivery into endothelial cell types.


Assuntos
Células Endoteliais/metabolismo , Células Endoteliais/virologia , Técnicas de Transferência de Genes , Vetores Genéticos , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Células Cultivadas , Humanos , Modelos Genéticos , Retroviridae/genética , Retroviridae/fisiologia , Replicação Viral
20.
Cancer Res ; 68(5): 1388-97, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18316602

RESUMO

Epithelial-to-mesenchymal transition (EMT) is an important event during carcinoma progression and leads to increased tumor cell malignancy. Here, we show that vascular endothelial (VE)-cadherin is induced during EMT in mammary tumor cells and is aberrantly expressed in invasive human breast carcinomas. VE-cadherin enhanced the capacity of fibroblastoid tumor cells to proliferate, form cord-like invasive structures, and adhere to endothelial cells, characteristics that are key contributors to their increased malignancy and metastatic potential. Consistently, VE-cadherin expression in malignant fibroblastoid tumor cells promoted the growth of experimental mammary carcinomas in vivo. Analysis of the signaling mechanisms involved revealed that VE-cadherin expression influences the levels of Smad2 phosphorylation and expression of target genes of transforming growth factor-beta (TGF-beta), a major mediator of advanced tumor progression and malignant tumor cell proliferation. VE-cadherin might thus promote tumor progression not only by contributing to tumor angiogenesis but also by enhancing tumor cell proliferation via the TGF-beta signaling pathway. This article provides evidence for a novel function of VE-cadherin in tumor progression and reveals a previously unknown molecular link between VE-cadherin expression and TGF-beta signaling. Our findings may have important implications for the clinical application of anti-VE-cadherin strategies.


Assuntos
Antígenos CD/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Perfilação da Expressão Gênica , Humanos , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias , Neovascularização Patológica , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA