Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Growth Factors ; 42(2): 49-61, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38299881

RESUMO

Breast cancer represents a collection of pathologies with different molecular subtypes, histopathology, risk factors, clinical behavior, and responses to treatment. "Basal-like" breast cancers predominantly lack the receptors for estrogen and progesterone (ER/PR), lack amplification of human epidermal growth factor receptor 2 (HER2) but account for 10-15% of all breast cancers, are largely insensitive to targeted treatment and represent a disproportionate number of metastatic cases and deaths. Analysis of interleukin (IL)-3 and the IL-3 receptor subunits (IL-3RA + CSF2RB) reveals elevated expression in predominantly the basal-like group. Further analysis suggests that IL-3 itself, but not the IL-3 receptor subunits, associates with poor patient outcome. Histology on patient-derived xenografts supports the notion that breast cancer cells are a significant source of IL-3 that may promote disease progression. Taken together, these observations suggest that IL-3 may be a useful marker in solid tumors, particularly triple negative breast cancer, and warrants further investigation into its contribution to disease pathogenesis.


Assuntos
Neoplasias da Mama , Interleucina-3 , Humanos , Feminino , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Interleucina-3/metabolismo , Animais , Prognóstico , Camundongos , Linhagem Celular Tumoral
2.
BMC Cancer ; 21(1): 765, 2021 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-34215227

RESUMO

BACKGROUND: The formation of blood vessels within solid tumors directly contributes to cancer growth and metastasis. Until recently, tumor vasculature was thought to occur exclusively via endothelial cell (EC) lined structures (i.e. angiogenesis), but a second source of tumor vasculature arises from the cancer cells themselves, a process known as vasculogenic mimicry (VM). While it is generally understood that the function of VM vessels is the same as that of EC-lined vessels (i.e. to supply oxygen and nutrients to the proliferating cancer cells), the molecular mechanisms underpinning VM are yet to be fully elucidated. METHODS: Human VM-competent melanoma cell lines were examined for their VM potential using the in vitro angiogenesis assays (Matrigel), together with inhibition studies using small interfering RNA and blocking monoclonal antibodies. Invasion assays and adhesion assays were used to examine cancer cell function. RESULTS: Herein we demonstrate that CD36, a cell surface glycoprotein known to promote angiogenesis by ECs, also supports VM formation by human melanoma cancer cells. In silico analysis of CD36 expression within the melanoma cohort of The Cancer Genome Atlas suggests that melanoma patients with high expression of CD36 have a poorer clinical outcome. Using in vitro 'angiogenesis' assays and CD36-knockdown approaches, we reveal that CD36 supports VM formation by human melanoma cells as well as adhesion to, and invasion through, a cancer derived extracellular matrix substrate. Interestingly, thrombospondin-1 (TSP-1), a ligand for CD36 on ECs that inhibits angiogenesis, has no effect on VM formation. Further investigation revealed a role for laminin, but not collagen or fibronectin, as ligands for CD36 expressing melanoma cells. CONCLUSIONS: Taken together, this study suggests that CD36 is a novel regulator of VM by melanoma cancer cells that is facilitated, at least in part, via integrin-α3 and laminin. Unlike angiogenesis, VM is not perturbed by the presence of TSP-1, thus providing new information on differences between these two processes of tumor vascularization which may be exploited to combat cancer progression.


Assuntos
Antígenos CD36/metabolismo , Matriz Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Melanoma , Microambiente Tumoral
3.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-34948000

RESUMO

Cutaneous squamous cell carcinoma (cSCC) accounts for 25% of cutaneous malignancies diagnosed in Caucasian populations. Surgical removal in combination with radiation and chemotherapy are effective treatments for cSCC. Nevertheless, the aggressive metastatic forms of cSCC still have a relatively poor patient outcome. Studies have linked actin cytoskeletal dynamics and the Wnt/ß-catenin signaling pathway as important modulators of cSCC pathogenesis. Previous studies have also shown that the actin-remodeling protein Flightless (Flii) is a negative regulator of cSCC. The aim of this study was to investigate if the functional effects of Flii on cSCC involve the Wnt/ß-catenin signaling pathway. Flii knockdown was performed using siRNA in a human late stage aggressive metastatic cSCC cell line (MET-1) alongside analysis of Flii genetic murine models of 3-methylcholanthrene induced cSCC. Flii was increased in a MET-1 cSCC cell line and reducing Flii expression led to fewer PCNA positive cells and a concomitant reduction in cellular proliferation and symmetrical division. Knockdown of Flii led to decreased ß-catenin and a decrease in the expression of the downstream effector of ß-catenin signaling protein SOX9. 3-Methylcholanthrene (MCA)-induced cSCC in Flii overexpressing mice showed increased markers of cancer metastasis including talin and keratin-14 and a significant increase in SOX9 alongside a reduction in Flii associated protein (Flap-1). Taken together, this study demonstrates a role for Flii in regulating proteins involved in cSCC proliferation and tumor progression and suggests a potential role for Flii in aggressive metastatic cSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Proteínas dos Microfilamentos/genética , Neoplasias Cutâneas/genética , Transativadores/genética , Regulação para Cima , Via de Sinalização Wnt , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Metilcolantreno/efeitos adversos , Camundongos , Neoplasias Cutâneas/induzido quimicamente
4.
J Virol ; 92(14)2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29743365

RESUMO

Severe dengue virus (DENV) infection is associated with overactivity of the complement alternative pathway (AP) in patient studies. Here, the molecular changes in components of the AP during DENV infection in vitro were investigated. mRNA for factor H (FH), a major negative regulator of the AP, was significantly increased in DENV-infected endothelial cells (EC) and macrophages, but, in contrast, production of extracellular FH protein was not. This discord was not seen for the AP activator factor B (FB), with DENV induction of both FB mRNA and protein, nor was it seen with Toll-like receptor 3 or 4 stimulation of EC and macrophages, which induces both FH and FB mRNA and protein. Surface-bound and intracellular FH protein was, however, induced by DENV, but only in DENV antigen-positive cells, while in two other DENV-susceptible immortalized cell lines (ARPE-19 and human retinal endothelial cells), FH protein was induced both intracellularly and extracellularly by DENV infection. Regardless of the cell type, there was an imbalance in AP components and an increase in markers of complement AP activity associated with DENV-infected cells, with lower FH relative to FB protein, an increased ability to promote AP-mediated lytic activity, and increased deposition of complement component C3b on the surface of DENV-infected cells. For EC in particular, these changes are predicted to result in higher complement activity in the local cellular microenvironment, with the potential to induce functional changes that may result in increased vascular permeability, a hallmark of dengue disease.IMPORTANCE Dengue virus (DENV) is a significant human viral pathogen with a global medical and economic impact. DENV may cause serious and life-threatening disease, with increased vascular permeability and plasma leakage. The pathogenic mechanisms underlying these features remain unclear; however, overactivity of the complement alternative pathway has been suggested to play a role. In this study, we investigate the molecular events that may be responsible for this observed alternative pathway overactivity and provide novel findings of changes in the complement system in response to DENV infection in primary cell types that are a major target for DENV infection (macrophages) and pathogenesis (endothelial cells) in vivo Our results suggest a new dimension of cellular events that may influence endothelial cell barrier function during DENV infection that could expand strategies for developing therapeutics to prevent or control DENV-mediated vascular disease.


Assuntos
Fator B do Complemento/imunologia , Fator H do Complemento/imunologia , Via Alternativa do Complemento , Proteínas do Sistema Complemento/imunologia , Vírus da Dengue/imunologia , Dengue/imunologia , Células Cultivadas , Fator B do Complemento/metabolismo , Fator H do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Dengue/metabolismo , Dengue/virologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Endotélio Vascular/virologia , Humanos , Retina/imunologia , Retina/patologia , Retina/virologia
5.
Respirology ; 24(11): 1095-1103, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30977250

RESUMO

BACKGROUND AND OBJECTIVE: Pulmonary arterial hypertension (PAH) is characterized by increased resistance in the distal pulmonary arteries, ultimately leading to right heart failure and, despite the available therapeutics, survival remains poor. Reduced expression of bone morphogenetic protein receptor type 2 (BMPR2) is strongly associated with PAH. Cell therapies are of interest in PAH, but whether this approach can upregulate BMPR2 is not known. Our objective was to evaluate a preclinical cell therapy approach based on upregulation of BMPR2. METHODS: We assessed the therapeutic effect of intravenously injected BMPR2-augmented rat bone marrow-derived endothelial-like progenitor cells (BMPR2-BM-ELPC) on PAH in the rat monocrotaline (MCT) model. RESULTS: The cells accumulate in the lungs with negligible systemic distribution, but the vast majority are lost from the lungs by 24 h. Lungs from rats treated with BMPR2-BM-ELPC exhibited an immediate increase in BMPR2 and related intracellular signalling proteins. Treatment with BMPR2-BM-ELPC attenuated PAH as demonstrated by a reduction in right ventricular hypertrophy as well as right ventricular systolic and mean pulmonary arterial pressures. In addition, this treatment reversed PAH-induced vascular remodelling with a significant reduction in vessel thickness and muscularization. In view of the short retention time of injected cells in the lungs, the mechanism for the effects seen may be intracellular communication via exosomes. In support of this hypothesis, we demonstrate that BMPR2-transduced outgrowth endothelial progenitor cells (OECs) release BMPR2-expressing exosomes. CONCLUSION: BMPR2-augmented ELPC demonstrate therapeutic benefits in the rat model and may have clinical translation potential.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Células Progenitoras Endoteliais , Hipertensão Arterial Pulmonar , Resistência Vascular , Animais , Medula Óssea/metabolismo , Terapia Baseada em Transplante de Células e Tecidos/métodos , Modelos Animais de Doenças , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/transplante , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/fisiopatologia , Hipertensão Arterial Pulmonar/terapia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Ratos , Resultado do Tratamento , Regulação para Cima , Remodelação Vascular
6.
Lung ; 197(5): 671-680, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31300872

RESUMO

PURPOSE: Mechanical ventilation is a well-established therapy for patients with acute respiratory failure. However, up to 35% of mortality in acute respiratory distress syndrome may be attributed to ventilation-induced lung injury (VILI). We previously demonstrated the efficacy of the synthetic tripeptide feG for preventing and ameliorating acute pancreatitis-associated lung injury. However, as the mechanisms of induction of injury during mechanical ventilation may differ, we aimed to investigate the effect of feG in a rodent model of VILI, with or without secondary challenge, as a preventative treatment when administered before injury (prophylactic), or as a therapeutic treatment administered following initiation of injury (therapeutic). METHODS: Lung injury was assessed following prophylactic or therapeutic intratracheal feG administration in a rodent model of ventilation-induced lung injury, with or without secondary intratracheal lipopolysaccharide challenge. RESULTS: Prophylactic feG administration resulted in significant improvements in arterial blood oxygenation and respiratory mechanics, and decreased lung oedema, bronchoalveolar lavage protein concentration, histological tissue injury scores, blood vessel activation, bronchoalveolar lavage cell infiltration and lung myeloperoxidase activity in VILI, both with and without lipopolysaccharide. Therapeutic feG administration similarly ameliorated the severity of tissue damage and encouraged the resolution of injury. feG associated decreases in endothelial adhesion molecules may indicate a mechanism for these effects. CONCLUSIONS: This study supports the potential for feG as a pharmacological agent in the prevention or treatment of lung injury associated with mechanical ventilation.


Assuntos
Pulmão/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Administração por Inalação , Animais , Modelos Animais de Doenças , Lipopolissacarídeos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Peroxidase/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Edema Pulmonar/metabolismo , Edema Pulmonar/patologia , Edema Pulmonar/prevenção & controle , Ratos Sprague-Dawley , Respiração Artificial , Mecânica Respiratória/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/etiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
7.
J Immunol ; 196(9): 3854-64, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27001955

RESUMO

The prevalence of allergies, including rhinitis, eczema, and anaphylaxis, is rising dramatically worldwide. This increase is especially problematic in children who bear the greatest burden of this rising trend. Increasing evidence identifies neutrophils as primary perpetrators of the more severe and difficult to manage forms of inflammation. A newly recognized mechanism by which neutrophils are recruited during the early phase of histamine-induced inflammation involves the sphingosine kinase (SK)/sphingosine-1-phosphate axis. This study examines whether topical application of fingolimod, an established SK/sphingosine-1-phosphate antagonist already in clinical use to treat multiple sclerosis, may be repurposed to treat cutaneous inflammation. Using two mouse models of ear skin inflammation (histamine- and IgE-mediated passive cutaneous anaphylaxis) we topically applied fingolimod prophylactically, as well as after establishment of the inflammatory response, and examined ear swelling, SK activity, vascular permeability, leukocyte recruitment, and production of proinflammatory mediators. The present study reveals that when applied topically, fingolimod attenuates both immediate and late-phase responses to histamine with reduced extravasation of fluid, SK-1 activity, proinflammatory cytokine and chemokine production, and neutrophil influx and prevents ear swelling. Intravital microscopy demonstrates that histamine-induced neutrophil rolling and adhesion to the postcapillary venules in the mouse ears is significantly attenuated even after 24 h. More importantly, these effects are achievable even once inflammation is established. Translation into humans was also accomplished with epicutaneous application of fingolimod resolving histamine-induced and allergen-induced inflammatory reactions in forearm skin. Overall, this study demonstrates, to our knowledge for the first time, that fingolimod may be repurposed to treat cutaneous inflammation.


Assuntos
Dermatite/tratamento farmacológico , Cloridrato de Fingolimode/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Neutrófilos/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Pele/efeitos dos fármacos , Administração Tópica , Animais , Permeabilidade Capilar/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Cloridrato de Fingolimode/farmacologia , Histamina/metabolismo , Humanos , Imunoglobulina E/sangue , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Pele/imunologia
8.
Nephrology (Carlton) ; 23(11): 1055-1062, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29265637

RESUMO

AIMS: Cardiovascular disease (CVD) is the leading cause of mortality in patients with end-stage renal disease (ESRD) receiving maintenance haemodialysis treatment. This study investigated the effect of a 12-week intradialytic progressive resistance training (PRT) intervention on pulse wave velocity (PWV) and associated haemodynamic, anthropometric, and hematologic outcomes in patients with ESRD. METHODS: Twenty-two patients with ESRD (59% men, 71.3 ± 11.0 years) were recruited. Supervised PRT (three sets of 11 exercises) was prescribed three times per week during routine dialysis. The primary outcome was brachial-ankle PWV via applanation tonometry. Secondary outcomes included augmentation index, brachial and aortic blood pressures, endothelial progenitor cells, C-reactive protein, blood lipids and anthropometrics. RESULTS: The intradialytic PRT regimen resulted in no significant change in PWV between control and intervention periods [mean difference = 0 (95% CI = -0.1 to 0.1); P = 0.58]. Similarly, no significant change was noted in any secondary outcome measures between the control and intervention periods. Post-hoc analyses limited to high adherers (≥75% attendance; n = 11) did not differ from the primary analysis, indicating no dose-response effect of our intervention. CONCLUSION: Our 12-week PRT intervention did not change PWV or any secondary outcomes. Future studies should determine if higher dosages of intradialytic PRT (i.e. longer duration and/or higher intensity) can be applied as a method to improve arterial stiffness to potentially reduce cardiovascular disease and associated mortality this cohort.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Falência Renal Crônica/complicações , Análise de Onda de Pulso , Diálise Renal/efeitos adversos , Treinamento Resistido , Idoso , Idoso de 80 Anos ou mais , Biomarcadores , Estudos Cross-Over , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Avaliação de Resultados em Cuidados de Saúde
9.
Pflugers Arch ; 469(9): 1121-1134, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28456852

RESUMO

Administration of bolus intravenous fluid is associated with respiratory dysfunction and increased mortality, findings with no clear mechanistic explanation. The objective of this study was to examine whether bolus intravenous (i.v.) fluid administration results in acute lung injury in a rat model and further, to examine whether this injury is associated with transient receptor potential vallinoid (TRPV)4 channel function and endothelial inflammatory response. Healthy male Sprague-Dawley rats were administered 60 ml/kg 0.9% saline i.v. over 30 min. Manifestation of acute lung injury was assessed by lung physiology, morphology, and markers of inflammation. The role of TRPV4 channels in fluid-induced lung injury was subsequently examined by the administration of ruthenium red (RR) in this established rat model and again in TRPV4 KO mice. In endothelial cell culture, permeability and P-selectin expression were measured following TRPV4 agonist with and without antagonist; 0.9% saline resulted in an increase in lung water, lavage protein and phospholipase A2, and plasma angiopoietin-2, with worsening in arterial blood oxygen (PaO2), lung elastance, surfactant activity, and lung histological injury score. These effects were ameliorated following i.v. fluid in rats receiving RR. TRPV4 KO mice did not develop lung edema. Expression of P-selectin increased in endothelial cells following administration of a TRPV4 agonist, which was ameliorated by simultaneous addition of RR. Bolus i.v. 0.9% saline resulted in permeability pulmonary edema. Data from ruthenium red, TRPV4 KO mice, and endothelial cell culture suggest activation of TRPV4 and release of angiopoietin 2 and P-selectin as the central mechanism.


Assuntos
Lesão Pulmonar/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Endotélio/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Permeabilidade , Edema Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley , Rutênio Vermelho/metabolismo
10.
Exp Dermatol ; 26(7): 607-614, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27992081

RESUMO

Skin cancer is associated with abnormal cellular metabolism which if identified early introduces the possibility of intervention to prevent its progress to a deadly metastatic stage. This study combines multiphoton microscopy with fluorescence lifetime imaging (FLIM) using a syngeneic melanoma mouse model, to detect changes in metabolic state of single epidermal cells as a metabolic marker to monitor the progress of tumor growth. This method utilizes imaging of the ratio of the amounts of the free and protein-bound forms of the intracellular autofluorescent metabolic co-enzyme nicotinamide adenine dinucleotide (NADH). Here, we investigate the impact of the primary tumor lesion on the epidermal layers at three different growth stages of melanoma lesion compared to normal skin as a control. We showed a significant increase in the free-to-bound NADH ratio with the growth of the solid melanoma tumor, while concurrently the short and the long lifetime components of NADH remained constant. These results demonstrate the ability of FLIM for rapid, non-invasive and sensitive assessment of melanoma progression revealing its potential as a diagnostic tool for melanoma detection and as an aid for melanoma staging.


Assuntos
Melanoma/diagnóstico por imagem , Microscopia de Fluorescência por Excitação Multifotônica , Neoplasias Cutâneas/diagnóstico por imagem , Animais , Linhagem Celular Tumoral , Progressão da Doença , Epiderme/metabolismo , Feminino , Queratinócitos/citologia , Melanoma/fisiopatologia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , NAD/química , Estadiamento de Neoplasias , Neoplasias Cutâneas/fisiopatologia
11.
Biomacromolecules ; 18(6): 1697-1704, 2017 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-28437084

RESUMO

The propensity of glycosaminoglycans to mediate cell-cell and cell-matrix interactions opens the door to capture cells, including circulating blood cells, onto biomaterial substrates. Chondroitin sulfate (CS)-B is of particular interest, since it interacts with the receptor (EGF)-like module-containing mucin-like hormone receptor-like 2 precursor (EMR2) displayed on the surface of leukocytes and endothelial progenitor cells. Herein, CS-B and its isomer CS-A were covalently immobilized onto heptylamine plasma polymer films via three different binding chemistries to develop platform technology for the capture of EMR2 expressing cells onto solid carriers. Surface characterization verified the successful immobilization of both glycosaminoglycans. The EMR2 expressing human myeloid cell line U937 preferentially bound onto CS-B-modified substrates, and U937 cells preincubated with CS-B in solution exhibited reduced affinity for the substrate. The direct capture of hematopoietic and blood-circulating endothelial cell types via a glycosaminoglycan-binding surface receptor opens an unexplored route for the development of biomaterials targeted at these cell types.


Assuntos
Separação Celular/métodos , Materiais Revestidos Biocompatíveis/química , Dermatan Sulfato/química , Receptores Acoplados a Proteínas G/metabolismo , Aminas/química , Adesão Celular , Sulfatos de Condroitina/química , Materiais Revestidos Biocompatíveis/metabolismo , Dermatan Sulfato/metabolismo , Expressão Gênica , Humanos , Gases em Plasma , Ligação Proteica , Receptores Acoplados a Proteínas G/genética , Propriedades de Superfície , Células U937
12.
Angiogenesis ; 19(4): 463-86, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27338829

RESUMO

Desmogleins (DSG) are a family of cadherin adhesion proteins that were first identified in desmosomes and provide cardiomyocytes and epithelial cells with the junctional stability to tolerate mechanical stress. However, one member of this family, DSG2, is emerging as a protein with additional biological functions on a broader range of cells. Here we reveal that DSG2 is expressed by non-desmosome-forming human endothelial progenitor cells as well as their mature counterparts [endothelial cells (ECs)] in human tissue from healthy individuals and cancer patients. Analysis of normal blood and bone marrow showed that DSG2 is also expressed by CD34(+)CD45(dim) hematopoietic progenitor cells. An inability to detect other desmosomal components, i.e., DSG1, DSG3 and desmocollin (DSC)2/3, on these cells supports a solitary role for DSG2 outside of desmosomes. Functionally, we show that CD34(+)CD45(dim)DSG2(+) progenitor cells are multi-potent and pro-angiogenic in vitro. Using a 'knockout-first' approach, we generated a Dsg2 loss-of-function strain of mice (Dsg2 (lo/lo)) and observed that, in response to reduced levels of Dsg2: (i) CD31(+) ECs in the pancreas are hypertrophic and exhibit altered morphology, (ii) bone marrow-derived endothelial colony formation is impaired, (iii) ex vivo vascular sprouting from aortic rings is reduced, and (iv) vessel formation in vitro and in vivo is attenuated. Finally, knockdown of DSG2 in a human bone marrow EC line reveals a reduction in an in vitro angiogenesis assay as well as relocalisation of actin and VE-cadherin away from the cell junctions, reduced cell-cell adhesion and increased invasive properties by these cells. In summary, we have identified DSG2 expression in distinct progenitor cell subpopulations and show that, independent from its classical function as a component of desmosomes, this cadherin also plays a critical role in the vasculature.


Assuntos
Desmogleína 2/metabolismo , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Animais , Diferenciação Celular , Células Cultivadas , Desmogleína 2/deficiência , Desmogleína 2/genética , Células Endoteliais/citologia , Feminino , Técnicas de Silenciamento de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Neovascularização Fisiológica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética
13.
Microcirculation ; 23(3): 248-65, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26822263

RESUMO

OBJECTIVE: A key mediator of vascular EC barrier integrity, S1P, is derived from phosphorylation of sphingosine by the SK-1 and SK-2. While previous work indicates that SK-1 can regulate EC barrier integrity, whether SK-2 has a similar role remains to be determined. METHODS: A cell impedance assay was used to assess human umbilical vein EC and bone marrow EC barrier integrity in vitro, with application of the SK inhibitors ABC294640, PF543, SKi, and MP-A08. In vivo studies were conducted using intravital microscopy to assess EC barrier integrity in SK-1 (Sphk1(-/-)) and SK-2 (Sphk2(-/-)) knock-out mice. RESULTS: Only ABC294640 and MP-A08, which can both inhibit SK-2, caused a decrease in EC barrier integrity in vitro in both cell types. Intravital microscopy revealed that Sphk1(-/-) mice had reduced EC barrier integrity compared to WT mice, whereas no change was evident in Sphk2(-/-) mice. CONCLUSIONS: Our data suggest that in vitro inhibition of SK-2, can compromise the integrity of the EC monolayer, while SK-1 exerts a more dominant control in vivo. These data may have clinical implications and could aid in the development of new treatments for disorders of vascular barrier function.


Assuntos
Adamantano/análogos & derivados , Permeabilidade Capilar/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Piridinas/farmacocinética , Adamantano/farmacocinética , Animais , Permeabilidade Capilar/genética , Humanos , Camundongos , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/genética
14.
FASEB J ; 29(9): 3638-53, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25985799

RESUMO

Sphingosine 1-phosphate (S1P) is a bioactive lipid that can function both extracellularly and intracellularly to mediate a variety of cellular processes. Using lipid affinity matrices and a radiolabeled lipid binding assay, we reveal that S1P directly interacts with the transcription factor peroxisome proliferator-activated receptor (PPAR)γ. Herein, we show that S1P treatment of human endothelial cells (ECs) activated a luciferase-tagged PPARγ-specific gene reporter by ∼12-fold, independent of the S1P receptors. More specifically, in silico docking, gene reporter, and binding assays revealed that His323 of the PPARγ ligand binding domain is important for binding to S1P. PPARγ functions when associated with coregulatory proteins, and herein we identify that peroxisome proliferator-activated receptor-γ coactivator 1 (PGC1)ß binds to PPARγ in ECs and their progenitors (nonadherent endothelial forming cells) and that the formation of this PPARγ:PGC1ß complex is increased in response to S1P. ECs treated with S1P selectively regulated known PPARγ target genes with PGC1ß and plasminogen-activated inhibitor-1 being increased, no change to adipocyte fatty acid binding protein 2 and suppression of CD36. S1P-induced in vitro tube formation was significantly attenuated in the presence of the PPARγ antagonist GW9662, and in vivo application of GW9662 also reduced vascular development in Matrigel plugs. Interestingly, activation of PPARγ by the synthetic ligand troglitazone also reduced tube formation in vitro and in vivo. To support this, Sphk1(-/-)Sphk2(+/-) mice, with low circulating S1P levels, demonstrated a similar reduction in vascular development. Taken together, our data reveal that the transcription factor, PPARγ, is a bona fide intracellular target for S1P and thus suggest that the S1P:PPARγ:PGC1ß complex may be a useful target to manipulate neovascularization.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Lisofosfolipídeos/metabolismo , Neovascularização Fisiológica/fisiologia , PPAR gama/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Lisofosfolipídeos/genética , Camundongos , Camundongos Knockout , PPAR gama/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Proteínas de Ligação a RNA , Receptores de Lisoesfingolipídeo/genética , Serpina E2/genética , Serpina E2/metabolismo , Esfingosina/genética , Esfingosina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células U937
15.
Biomacromolecules ; 17(11): 3724-3731, 2016 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-27744681

RESUMO

Porous silicon (pSi) substrates are a promising platform for cell expansion, since pore size and chemistry can be tuned to control cell behavior. In addition, a variety of bioactives can be loaded into the pores and subsequently released to act on cells adherent to the substrate. Here, we construct a cell microarray on a plasma polymer coated pSi substrate that enables the simultaneous culture of human endothelial cells on printed immobilized protein factors, while a second soluble growth factor is released from the same substrate. This allows three elements of candidate pSi scaffold materials-topography, surface functionalization, and controlled factor release-to be assessed simultaneously in high throughput. We show that protein conjugation within printed microarray spots is more uniform on the pSi substrate than on flat glass or silicon surfaces. Active growth factors are released from the pSi surface over a period of several days. Using an endothelial progenitor cell line, we investigate changes in cell behavior in response to the microenvironment. This platform facilitates the design of advanced functional biomaterials, including scaffolds, and carriers for regenerative medicine and cell therapy.


Assuntos
Materiais Biocompatíveis/química , Sistemas de Liberação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Polímeros/química , Materiais Biocompatíveis/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/química , Humanos , Polímeros/farmacologia , Porosidade , Silício/química , Análise Serial de Tecidos
16.
Respirology ; 21(4): 727-33, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26809239

RESUMO

BACKGROUND AND OBJECTIVE: Pulmonary arterial hypertension (PAH) continues to be a fatal disease and is associated with downregulation of bone morphogenetic protein receptor type-2 (BMPR2). Our approach is to upregulate BMPR2 in the pulmonary vasculature allowing us to examine the changes in endothelial cell signalling and better understand what pathways are altered when disease is attenuated using this treatment approach. METHODS: We used gene delivery of BMPR2 to human pulmonary endothelial cells to investigate downstream signalling, then assessed the impact of this approach on downstream signalling in vivo in rats with PAH using the monocrotaline (MCT) model. RESULTS: Gene delivery of BMPR2 leads to an increase in BMPR2 protein expression, and this is associated with increased Smad1/5/8 and reduced Smad2/3 signalling. Additionally, we have found that BMPR2 modulation has effects on non-Smad signalling with increases found in phosphoinositide-3 kinase (PI3K) and a decrease in phosphorylated-p38-mitogen activated protein kinase (p38-MAPK) in vivo. These findings are associated with amelioration of PAH (reduced right ventricular, mean pulmonary artery pressures and Fulton Index). CONCLUSION: These results indicate that the therapeutic effect of BMPR2 gene delivery on PAH is associated with a switch between TGF-ß-Smad2/3 signalling to BMPR2-Smad1/5/8 signalling. This supports the further development of this treatment approach.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Terapia Genética , Hipertensão Pulmonar/terapia , Artéria Pulmonar/fisiopatologia , Transdução de Sinais , Proteínas Smad Reguladas por Receptor/metabolismo , Animais , Pressão Arterial , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/uso terapêutico , Linhagem Celular , Células Endoteliais/metabolismo , Humanos , Hipertensão Pulmonar/fisiopatologia , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Immunol Rev ; 250(1): 277-302, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23046136

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3), and IL-5 are members of a discrete family of cytokines that regulates the growth, differentiation, migration and effector function activities of many hematopoietic cells and immunocytes. These cytokines are involved in normal responses to infectious agents, bridging innate and adaptive immunity. However, in certain cases, the overexpression of these cytokines or their receptors can lead to excessive or aberrant initiation of signaling resulting in pathological conditions, with chronic inflammatory diseases and myeloid leukemias the most notable examples. Recent crystal structures of the GM-CSF receptor ternary complex and the IL-5 binary complex have revealed new paradigms of cytokine receptor activation. Together with a wealth of associated structure-function studies, they have significantly enhanced our understanding of how these receptors recognize cytokines and initiate signals across cell membranes. Importantly, these structures provide opportunities for structure-based approaches for the discovery of novel and disease-specific therapeutics. In addition, recent biochemical evidence has suggested that the GM-CSF/IL-3/IL-5 receptor family is capable of interacting productively with other membrane proteins at the cell surface. Such interactions may afford additional or unique biological activities and might be harnessed for selective modulation of the function of these receptors in disease.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/química , Interleucina-3/química , Interleucina-5/química , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/química , Receptores de Interleucina-3/química , Receptores de Interleucina-5/química , Cristalografia por Raios X , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-3/imunologia , Interleucina-3/metabolismo , Interleucina-5/imunologia , Interleucina-5/metabolismo , Leucemia Mieloide/imunologia , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Linfócitos/imunologia , Linfócitos/metabolismo , Linfócitos/patologia , Modelos Moleculares , Ligação Proteica , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/imunologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Receptores de Interleucina-3/imunologia , Receptores de Interleucina-3/metabolismo , Receptores de Interleucina-5/imunologia , Receptores de Interleucina-5/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
18.
Mediators Inflamm ; 2015: 509306, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26609198

RESUMO

One of the main pathogenic effects of severe dengue virus (DENV) infection is a vascular leak syndrome. There are no available antivirals or specific DENV treatments and without hospital support severe DENV infection can be life-threatening. The cause of the vascular leakage is permeability changes in the endothelial cells lining the vasculature that are brought about by elevated vasoactive cytokine and chemokines induced following DENV infection. The source of these altered cytokine and chemokines is traditionally believed to be from DENV-infected cells such as monocyte/macrophages and dendritic cells. Herein we discuss the evidence for the endothelium as an additional contributor to inflammatory and innate responses during DENV infection which may affect endothelial cell function, in particular the ability to maintain vascular integrity. Furthermore, we hypothesise roles for two factors, sphingosine kinase-1 and microRNAs (miRNAs), with a focus on several candidate miRNAs, which are known to control normal vascular function and inflammatory responses. Both of these factors may be potential therapeutic targets to regulate inflammation of the endothelium during DENV infection.


Assuntos
Dengue/fisiopatologia , Endotélio Vascular/patologia , Inflamação/etiologia , MicroRNAs/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Permeabilidade Capilar , Humanos
19.
Diabetologia ; 57(2): 402-12, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24292564

RESUMO

AIMS/HYPOTHESIS: Skin lesions and ulcerations are severe complications of diabetes that often result in leg amputations. In this study we investigated the function of the cytoskeletal protein flightless I (FLII) in diabetic wound healing. We hypothesised that overexpression of FLII would have a negative effect on diabetic wound closure and modulation of this protein using specific FLII-neutralising antibodies (FnAb) would enhance cellular proliferation, migration and angiogenesis within the diabetic wound. METHODS: Using a streptozotocin-induced model of diabetes we investigated the effect of altered FLII levels through Flii genetic knockdown, overexpression or treatment with FnAb on wound healing. Diabetic wounds were assessed using histology, immunohistochemistry and biochemical analysis. In vitro and in vivo assays of angiogenesis were used to assess the angiogenic response. RESULTS: FLII levels were elevated in the wounds of both diabetic mice and humans. Reduction in the level of FLII improved healing of murine diabetic wounds and promoted a robust pro-angiogenic response with significantly elevated von Willebrand factor (vWF) and vascular endothelial growth factor (VEGF)-positive endothelial cell infiltration. Diabetic mouse wounds treated intradermally with FnAb showed improved healing and a significantly increased rate of re-epithelialisation. FnAb improved the angiogenic response through enhanced formation of capillary tubes and functional neovasculature. Reducing the level of FLII led to increased numbers of mature blood vessels, increased recruitment of smooth muscle actin-α-positive cells and improved tight junction formation. CONCLUSIONS/INTERPRETATION: Reducing the level of FLII in a wound may be a potential therapeutic approach for the treatment of diabetic foot ulcers.


Assuntos
Proteínas do Citoesqueleto/farmacologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Angiopatias Diabéticas/patologia , Pele/patologia , Cicatrização/imunologia , Indutores da Angiogênese , Animais , Anticorpos Neutralizantes/metabolismo , Proteínas de Transporte , Proliferação de Células , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/imunologia , Angiopatias Diabéticas/imunologia , Feminino , Humanos , Imuno-Histoquímica , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos , Pele/lesões , Transativadores , Úlcera/patologia
20.
Rev Med Virol ; 23(2): 73-84, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22639116

RESUMO

Sphingosine kinase 1 (SphK1) is an enzyme that phosphorylates the lipid sphingosine to generate sphingosine-1-phosphate (S1P). S1P can act intracellularly as a signaling molecule and extracellularly as a receptor ligand. The SphK1/S1P axis has well-described roles in cell signaling, the cell death/survival decision, the production of a pro-inflammatory response, immunomodulation, and control of vascular integrity. Agents targeting the SphK1/S1P axis are being actively developed as therapeutics for cancer and immunological and inflammatory disorders. Control of cell death/survival and pro-inflammatory immune responses is central to the pathology of infectious disease, and we can capitalize on the knowledge provided by investigations of SphK1/S1P in cancer and immunology to assess its application to selected human infections. We have herein reviewed the growing literature relating viral infections to changes in SphK1 and S1P. SphK1 activity is reportedly increased following human cytomegalovirus and respiratory syncytial virus infections, and elevated SphK1 enhances influenza virus infection. In contrast, SphK1 activity is reduced in bovine viral diarrhea virus and dengue virus infections. Sphingosine analogs that modulate S1P receptors have proven useful in animal models in alleviating influenza virus infection but have shown no benefit in simian human immunodeficiency virus and lymphocytic choriomeningitis virus infections. We have rationalized a role for SphK1/S1P in dengue virus, chikungunya virus, and Ross River virus infections, on the basis of the biology and the pathology of these diseases. The increasing number of effective SphK1 and S1P modulating agents currently in development makes it timely to investigate these roles with the potential for developing modulators of SphK1 and S1P for novel anti-viral therapies.


Assuntos
Lisofosfolipídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Viroses/imunologia , Animais , Modelos Animais de Doenças , Humanos , Fatores Imunológicos/metabolismo , Esfingosina/metabolismo , Viroses/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA