Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L32-L53, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28860142

RESUMO

The biological and immune-protective properties of surfactant-derived phospholipids and phospholipid subfractions in the context of neonatal inflammatory lung disease are widely unknown. Using a porcine neonatal triple-hit acute respiratory distress syndrome (ARDS) model (repeated airway lavage, overventilation, and LPS instillation into airways), we assessed whether the supplementation of surfactant (S; poractant alfa) with inositol derivatives [inositol 1,2,6-trisphosphate (IP3) or phosphatidylinositol 3,5-bisphosphate (PIP2)] or phosphatidylglycerol subfractions [16:0/18:1-palmitoyloleoyl-phosphatidylglycerol (POPG) or 18:1/18:1-dioleoyl-phosphatidylglycerol (DOPG)] would result in improved clinical parameters and sought to characterize changes in key inflammatory pathways behind these improvements. Within 72 h of mechanical ventilation, the oxygenation index (S+IP3, S+PIP2, and S+POPG), the ventilation efficiency index (S+IP3 and S+POPG), the compliance (S+IP3 and S+POPG) and resistance (S+POPG) of the respiratory system, and the extravascular lung water index (S+IP3 and S+POPG) significantly improved compared with S treatment alone. The inositol derivatives (mainly S+IP3) exerted their actions by suppressing acid sphingomyelinase activity and dependent ceramide production, linked with the suppression of the inflammasome nucleotide-binding domain, leucine-rich repeat-containing protein-3 (NLRP3)-apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-caspase-1 complex, and the profibrotic response represented by the cytokines transforming growth factor-ß1 and IFN-γ, matrix metalloproteinase (MMP)-1/8, and elastin. In addition, IκB kinase activity was significantly reduced. S+POPG and S+DOPG treatment inhibited polymorphonuclear leukocyte activity (MMP-8 and myeloperoxidase) and the production of interleukin-6, maintained alveolar-capillary barrier functions, and reduced alveolar epithelial cell apoptosis, all of which resulted in reduced pulmonary edema. S+DOPG also limited the profibrotic response. We conclude that highly concentrated inositol derivatives and phosphatidylglycerol subfractions in surfactant preparations mitigate key inflammatory pathways in inflammatory lung disease and that their clinical application may be of interest for future treatment of the acute exudative phase of neonatal ARDS.


Assuntos
Modelos Animais de Doenças , Inositol/farmacologia , Fosfatidilgliceróis/farmacologia , Edema Pulmonar/tratamento farmacológico , Surfactantes Pulmonares/farmacologia , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Animais , Animais Recém-Nascidos , Apoptose , Líquido da Lavagem Broncoalveolar , Citocinas/genética , Citocinas/metabolismo , Feminino , Humanos , Masculino , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Edema Pulmonar/metabolismo , Edema Pulmonar/patologia , Troca Gasosa Pulmonar , Distribuição Aleatória , Respiração Artificial , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Suínos , Pesquisa Translacional Biomédica , Complexo Vitamínico B/farmacologia
2.
Am J Physiol Lung Cell Mol Physiol ; 310(8): L720-32, 2016 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-26851257

RESUMO

Transcellular albumin transport occurs via caveolae that are abundant in lung microvascular endothelial cells. Stimulation of albumin transcytosis by proinflammatory mediators may contribute to alveolar protein leak in lung injury, yet the regulation of albumin transport and its underlying molecular mechanisms are so far incompletely understood. Here we tested the hypothesis that thrombin may stimulate transcellular albumin transport across lung microvascular endothelial cells in an acid-sphingomyelinase dependent manner. Thrombin increased the transport of fluorescently labeled albumin across confluent human lung microvascular endothelial cell (HMVEC-L) monolayers to an extent that markedly exceeds the rate of passive diffusion. Thrombin activated acid sphingomyelinase (ASM) and increased ceramide production in HMVEC-L, but not in bovine pulmonary artery cells, which showed little albumin transport in response to thrombin. Thrombin increased total caveolin-1 (cav-1) content in both whole cell lysates and lipid rafts from HMVEC-L, and this effect was blocked by inhibition of ASM or de novo protein biosynthesis. Thrombin-induced uptake of albumin into lung microvascular endothelial cells was confirmed in isolated-perfused lungs by real-time fluorescence imaging and electron microscopy of gold-labeled albumin. Inhibition of ASM attenuated thrombin-induced albumin transport both in confluent HMVEC-L and in intact lungs, whereas HMVEC-L treatment with exogenous ASM increased albumin transport and enriched lipid rafts in cav-1. Our findings indicate that thrombin stimulates transcellular albumin transport in an acid sphingomyelinase-dependent manner by inducing de novo synthesis of cav-1 and its recruitment to membrane lipid rafts.


Assuntos
Albuminas/metabolismo , Células Endoteliais/enzimologia , Esfingomielina Fosfodiesterase/metabolismo , Transcitose , Animais , Bovinos , Caveolina 1/metabolismo , Células Cultivadas , Endotélio Vascular/citologia , Ativação Enzimática , Humanos , Pulmão/irrigação sanguínea , Masculino , Microdomínios da Membrana/metabolismo , Microvasos/citologia , Transporte Proteico , Ratos Sprague-Dawley , Trombina/fisiologia
3.
EMBO J ; 30(2): 379-94, 2011 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-21157428

RESUMO

We previously demonstrated that tumour necrosis factor (TNF)-induced ceramide production by endosomal acid sphingomyelinase (A-SMase) couples to apoptosis signalling via activation of cathepsin D and cleavage of Bid, resulting in caspase-9 and caspase-3 activation. The mechanism of TNF-mediated A-SMase activation within the endolysosomal compartment is poorly defined. Here, we show that TNF-induced A-SMase activation depends on functional caspase-8 and caspase-7 expression. The active forms of all three enzymes, caspase-8, caspase-7 and A-SMase, but not caspase-3, colocalize in internalized TNF receptosomes. While caspase-8 and caspase-3 are unable to induce activation of purified pro-A-SMase, we found that caspase-7 mediates A-SMase activation by direct interaction resulting in proteolytic cleavage of the 72-kDa pro-A-SMase zymogen at the non-canonical cleavage site after aspartate 253, generating an active 57 kDa A-SMase molecule. Caspase-7 down modulation revealed the functional link between caspase-7 and A-SMase, confirming proteolytic cleavage as one further mode of A-SMase activation. Our data suggest a signalling cascade within TNF receptosomes involving sequential activation of caspase-8 and caspase-7 for induction of A-SMase activation by proteolytic cleavage of pro-A-SMase.


Assuntos
Caspase 7/metabolismo , Caspase 8/metabolismo , Endossomos/metabolismo , Ativação Enzimática/fisiologia , Esfingomielina Fosfodiesterase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose , Western Blotting , Linhagem Celular , Ceramidas/metabolismo , Cromatografia em Camada Fina , Clonagem Molecular , Ativação Enzimática/genética , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Células Jurkat , Camundongos , Microscopia Confocal
4.
BMC Cancer ; 14: 74, 2014 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-24507727

RESUMO

BACKGROUND: The cytokine TRAIL represents one of the most promising candidates for the apoptotic elimination of tumor cells, either alone or in combination therapies. However, its efficacy is often limited by intrinsic or acquired resistance of tumor cells to apoptosis. Programmed necrosis is an alternative, molecularly distinct mode of programmed cell death that is elicited by TRAIL under conditions when the classical apoptosis machinery fails or is actively inhibited. The potential of TRAIL-induced programmed necrosis in tumor therapy is, however, almost completely uncharacterized. We therefore investigated its impact on a panel of tumor cell lines of wide-ranging origin. METHODS: Cell death/viability was measured by flow cytometry/determination of intracellular ATP levels/crystal violet staining. Cell surface expression of TRAIL receptors was detected by flow cytometry, expression of proteins by Western blot. Ceramide levels were quantified by high-performance thin layer chromatography and densitometric analysis, clonogenic survival of cells was determined by crystal violet staining or by soft agarose cloning. RESULTS: TRAIL-induced programmed necrosis killed eight out of 14 tumor cell lines. Clonogenic survival was reduced in all sensitive and even one resistant cell lines tested. TRAIL synergized with chemotherapeutics in killing tumor cell lines by programmed necrosis, enhancing their effect in eight out of 10 tested tumor cell lines and in 41 out of 80 chemotherapeutic/TRAIL combinations. Susceptibility/resistance of the investigated tumor cell lines to programmed necrosis seems to primarily depend on expression of the pro-necrotic kinase RIPK3 rather than the related kinase RIPK1 or cell surface expression of TRAIL receptors. Furthermore, interference with production of the lipid ceramide protected all tested tumor cell lines. CONCLUSIONS: Our study provides evidence that TRAIL-induced programmed necrosis represents a feasible approach for the elimination of tumor cells, and that this treatment may represent a promising new option for the future development of combination therapies. Our data also suggest that RIPK3 expression may serve as a potential predictive marker for the sensitivity of tumor cells to programmed necrosis and extend the previously established role of ceramide as a key mediator of death receptor-induced programmed necrosis (and thus as a potential target for future therapies) also to the tumor cell lines examined here.


Assuntos
Apoptose/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/intoxicação , Antineoplásicos/intoxicação , Western Blotting , Morte Celular/genética , Citometria de Fluxo/métodos , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Necrose/patologia , Necrose/prevenção & controle , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Células U937
5.
Pulm Pharmacol Ther ; 28(1): 25-34, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24140177

RESUMO

BACKGROUND: 18:1/18:1-Dioleoyl-phosphatidylgycerol (DOPG) is a surfactant phospholipid that is nearly non-detectable in neonatal surfactant films. When alveolar macrophages are exposed to DOPG in vitro, secretory phospholipase A2 (sPLA2) production is blocked, resulting in suppressed macrophage activity and improved surfactant function. We investigated whether the addition of DOPG to a commercially available surfactant preparation would improve lung function in a neonatal piglet model of acute respiratory distress syndrome. MATERIALS AND METHODS: Respiratory failure was achieved by triple-hit lung injury (repeated broncho-alveolar lavage, injurious ventilation, tracheal lipopolysaccharide instillation, each intervention 24 h apart) in twenty-four domestic piglets aged 2-6 days and subject to mechanical ventilation. Following each lung injury protocol the piglets were treated with surfactant alone or with surfactant + DOPG. RESULTS: Within 72 h of mechanical ventilation, we observed significantly improved gas exchange (oxygenation and ventilation), lung mechanics (compliance and resistance of the respiratory system), and pulmonary oedema (extra-vascular lung water index) in the surfactant + DOPG group. This favourable clinical effect could be attributed to improved surfactant function, reduced sPLA2 secretion, inhibition of macrophage migration, reduced alveolar epithelial apoptosis, and suppression of amphiregulin and TGF-ß1 expression in pulmonary tissues as a prerequisite for fibrous lung repair. CONCLUSIONS: We conclude that surfactant fortified by DOPG preserves lung function, and prevents alveolar epithelial injury and fibrous stimulus by reduction of sPLA2 in a neonatal model of acute respiratory distress syndrome without any relevant discernable side effects. Hence, DOPG supplementation in a neonatal lung exerts important function protecting effects and seems to be justified in cases of overwhelming pulmonary inflammation.


Assuntos
Apoptose/efeitos dos fármacos , Fosfatidilgliceróis/farmacologia , Surfactantes Pulmonares/farmacologia , Síndrome do Desconforto Respiratório do Recém-Nascido/prevenção & controle , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Masculino , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Edema Pulmonar/prevenção & controle , Fibrose Pulmonar/patologia , Fibrose Pulmonar/prevenção & controle , Respiração Artificial , Suínos
6.
Proc Natl Acad Sci U S A ; 107(3): 1112-7, 2010 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-20080539

RESUMO

The phospholipase neutral sphingomyelinase (N-SMase) has been recognized as a major mediator of processes such as inflammation, development and growth, differentiation and death of cells, as well as in diseases such as Alzheimer's, atherosclerosis, heart failure, ischemia/reperfusion damage, or combined pituitary hormone deficiency. Although activation of N-SMase by the proinflammatory cytokine TNF was described almost two decades ago, the underlying signaling pathway is unresolved. Here, we identify the Polycomb group protein EED (embryonic ectodermal development) as an interaction partner of nSMase2. In yeast, the N terminus of EED binds to the catalytic domain of nSMase2 as well as to RACK1, a protein that modulates the activation of nSMase2 by TNF in concert with the TNF receptor 1 (TNF-R1)-associated protein FAN. In mammalian cells, TNF causes endogenous EED to translocate from the nucleus and to colocalize and physically interact with both endogenous nSMase2 and RACK1. As a consequence, EED and nSMase2 are recruited to the TNF-R1.FAN.RACK1-complex in a timeframe concurrent with activation of nSMase2. After knockdown of EED by RNA interference, the TNF-dependent activation of nSMase2 is completely abrogated, identifying EED as a protein that both physically and functionally couples TNF-R1 to nSMase2, and which therefore represents the "missing link" that completes one of the last unresolved signaling pathways of TNF-R1.


Assuntos
Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Repressoras/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Ativação Enzimática , Células HeLa , Humanos , Complexo Repressor Polycomb 2
7.
Am J Respir Cell Mol Biol ; 47(2): 158-69, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22403805

RESUMO

D-myo-inositol-1,2,6-trisphosphate (IP3) is an isomer of the naturally occurring second messenger D-myo-inositol-1,4,5-trisphosphate, and exerts anti-inflammatory and antiedematous effects in the lung. Myo-inositol (Inos) is a component of IP3, and is thought to play an important role in the prevention of neonatal pulmonary diseases such as bronchopulmonary dysplasia and neonatal acute lung injury (nALI). Inflammatory lung diseases are characterized by augmented acid sphingomyelinase (aSMase) activity leading to ceramide production, a pathway that promotes increased vascular permeability, apoptosis, and surfactant alterations. A novel, clinically relevant triple-hit model of nALI was developed, consisting of repeated airway lavage, injurious ventilation, and lipopolysaccharide instillation into the airways, every 24 hours. Thirty-five piglets were randomized to one of four treatment protocols: control (no intervention), surfactant alone, surfactant + Inos, and surfactant + IP3. After 72 hours of mechanical ventilation, lungs were excised from the thorax for subsequent analyses. Clinically, oxygenation and ventilation improved, and extravascular lung water decreased significantly with the S + IP3 intervention. In pulmonary tissue, we observed decreased aSMase activity and ceramide concentrations, decreased caspase-8 concentrations, reduced alveolar epithelial apoptosis, the reduced expression of interleukin-6, transforming growth factor-ß1, and amphiregulin (an epithelial growth factor), reduced migration of blood-borne cells and particularly of CD14(+)/18(+) cells (macrophages) into the airspaces, and lower surfactant surface tensions in S + IP3-treated but not in S + Inos-treated piglets. We conclude that the admixture of IP3 to surfactant, but not of Inos, improves gas exchange and edema in our nALI model by the suppression of the governing enzyme aSMase, and that this treatment deserves clinical evaluation.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Apoptose/efeitos dos fármacos , Fosfatos de Inositol/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Edema Pulmonar/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Anfirregulina , Animais , Animais Recém-Nascidos , Líquido da Lavagem Broncoalveolar , Caspase 8/metabolismo , Ceramidas/metabolismo , Modelos Animais de Doenças , Feminino , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-6/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Linfotoxina-alfa/metabolismo , Masculino , Alvéolos Pulmonares/metabolismo , Edema Pulmonar/metabolismo , Edema Pulmonar/patologia , Troca Gasosa Pulmonar/efeitos dos fármacos , Surfactantes Pulmonares/metabolismo , Surfactantes Pulmonares/farmacologia , Respiração Artificial/métodos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Esfingomielina Fosfodiesterase/metabolismo , Tensão Superficial/efeitos dos fármacos , Suínos
8.
J Cell Mol Med ; 16(11): 2813-26, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22882773

RESUMO

Hypoxemic respiratory failure of the neonatal organism involves increased acid sphingomyelinase (aSMase) activity and production of ceramide, a second messenger of a pro-inflammatory pathway that promotes increased vascular permeability, surfactant alterations and alveolar epithelial apoptosis. We comparatively assessed the benefits of topical aSMase inhibition by either imipramine (Imi) or phosphatidylinositol-3,5-bisphosphate (PIP2) when administered into the airways together with surfactant (S) for fortification. In this translational study, a triple-hit acute lung injury model was used that entails repeated airway lavage, injurious ventilation and tracheal lipopolysaccharide instillation in newborn piglets subject to mechanical ventilation for 72 hrs. After randomization, we administered an air bolus (control), S, S+Imi, or S+PIP2. Only in the latter two groups we observed significantly improved oxygenation and ventilation, dynamic compliance and pulmonary oedema. S+Imi caused systemic aSMase suppression and ceramide reduction, whereas the S+PIP2 effect remained compartmentalized in the airways because of the molecule's bulky structure. The surfactant surface tensions improved by S+Imi and S+PIP2 interventions, but only to a minor extent by S alone. S+PIP2 inhibited the migration of monocyte-derived macrophages and granulocytes into airways by the reduction of CD14/CD18 expression on cell membranes and the expression of epidermal growth factors (amphiregulin and TGF-ß1) and interleukin-6 as pro-fibrotic factors. Finally we observed reduced alveolar epithelial apoptosis, which was most apparent in S+PIP2 lungs. Exogenous surfactant "fortified" by PIP2, a naturally occurring surfactant component, improves lung function by topical suppression of aSMase, providing a potential treatment concept for neonates with hypoxemic respiratory failure.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Fosfatos de Fosfatidilinositol/administração & dosagem , Lesão Pulmonar Aguda/patologia , Administração Tópica , Anfirregulina , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar/citologia , Antígenos CD18/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Ceramidas/metabolismo , Modelos Animais de Doenças , Feminino , Glicoproteínas/metabolismo , Imipramina/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-6/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Surfactantes Pulmonares , Respiração Artificial , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Suínos , Fator de Crescimento Transformador beta/metabolismo
9.
Pulm Pharmacol Ther ; 25(3): 228-35, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22469869

RESUMO

Airway epithelial NF-κB is a key regulator of host defence in bacterial infections and has recently evolved as a target for therapeutical approaches. Evidence is accumulating that ceramide, generated by acid sphingomyelinase (aSMase), and sphingosine-1-phosphate (S1-P) are important mediators in host defence as well as in pathologic processes of acute lung injury. Little is known about the regulatory mechanisms of pulmonary sphingolipid metabolism in bacterial infections of the lung. The objective of this study was to evaluate the influence of NF-κB on sphingolipid metabolism in Pseudomonas aeruginosa LPS-induced pulmonary inflammation. In a murine acute lung injury model with intranasal Pseudomonas aeruginosa LPS we investigated TNF-α, KC (murine IL-8), IL-6, MCP-1 and neutrophilic infiltration next to aSMase activity and ceramide and S1-P lung tissue concentrations. Airway epithelial NF-κB was inhibited by topically applied IKK NBD, a cell penetrating NEMO binding peptide. This treatment resulted in significantly reduced inflammation and suppression of aSMase activity along with decreased ceramide and S1-P tissue concentrations down to levels observed in healthy animals. In conclusion our results confirm that changes in sphingolipid metabolim due to Pseudomonas aeruginosa LPS inhalation are regulated by NF-κB translocation. This confirms the critical role of airway epithelial NF-κB pathway for the inflammatory response to bacterial pathogens and underlines the impact of sphingolipids in inflammatory host defence mechanisms.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Peptídeos Penetradores de Células/farmacologia , NF-kappa B/metabolismo , Peptídeos/farmacologia , Esfingolipídeos/metabolismo , Lesão Pulmonar Aguda/imunologia , Animais , Ceramidas/metabolismo , Modelos Animais de Doenças , Feminino , Quinase I-kappa B/metabolismo , Inflamação/imunologia , Inflamação/fisiopatologia , Lipopolissacarídeos , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , Pseudomonas aeruginosa/imunologia , Esfingosina/análogos & derivados , Esfingosina/metabolismo
10.
Eur J Immunol ; 40(4): 1162-73, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20104488

RESUMO

Human monocytes respond to a variety of stimuli with a complex spectrum of activities ranging from acute defense mechanisms to cell differentiation or cytokine release. However, the individual intracellular signaling pathways related to these functions are not well understood. CXC chemokine ligand 4 (CXCL4) represents a broad activator of monocytes, which induces acute as well as delayed activities in these cells including cell differentiation, survival, or the release of ROS, and cytokines. Here, we report for the first time that CXCL4-treated monocytes significantly upregulate sphingosine kinase 1 (SphK1) mRNA and that CXCL4 induces SphK1 enzyme activity as well as its translocation to the cell membrane. Furthermore, we could show that pharmacological inhibition of SphK results in reversal of CXCL4-induced monocyte survival, cytokine expression, and release of oxygen radicals, which was confirmed by the use of SphK1-specific siRNA. CXCL4-mediated rescue from apoptosis, which is accompanied by inhibition of caspases, is controlled by SphK1 and its downstream element Erk. Taken together, these data assign SphK1 as a central regulator of acute and delayed monocyte activation and suggest SphK1 as a potential therapeutic target to suppress pro-inflammatory responses induced by CXCL4.


Assuntos
Citocinas/biossíntese , Monócitos/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Fator Plaquetário 4/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Adulto , Apoptose/efeitos dos fármacos , Inibidores de Caspase , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/enzimologia , Citocinas/genética , Citocinas/metabolismo , Indução Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/fisiopatologia , Monócitos/citologia , Monócitos/metabolismo , Toxina Pertussis/farmacologia , Transporte Proteico/efeitos dos fármacos
11.
Nat Med ; 10(2): 155-60, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14704790

RESUMO

Platelet-activating factor (PAF) induces pulmonary edema and has a key role in acute lung injury (ALI). Here we show that PAF induces pulmonary edema through two mechanisms: acid sphingomyelinase (ASM)-dependent production of ceramide, and activation of the cyclooxygenase pathway. Agents that interfere with PAF-induced ceramide synthesis, such as steroids or the xanthogenate D609, attenuate pulmonary edema formation induced by PAF, endotoxin or acid instillation. Our results identify acid sphingomyelinase and ceramide as possible therapeutic targets in acute lung injury.


Assuntos
Ceramidas/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Edema Pulmonar/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Animais , Anticorpos/metabolismo , Anticorpos/uso terapêutico , Hidrocarbonetos Aromáticos com Pontes/metabolismo , Dexametasona/metabolismo , Feminino , Glucocorticoides/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Norbornanos , Inibidores de Fosfodiesterase/metabolismo , Edema Pulmonar/tratamento farmacológico , Ratos , Ratos Wistar , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Tiocarbamatos , Tionas/metabolismo
12.
Exp Cell Res ; 315(17): 3064-75, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19632221

RESUMO

The regulation of mast cell activities and survival is a central issue in inflammatory immune responses. Here, we have investigated the role of mouse interleukin-15, a pro-inflammatory and pleiotropic cytokine, in the control of mast cell survival and homeostasis. We report that aged IL-15-/- mice show a reduced number of peritoneal mast cells compared to WT mice. Furthermore, IL-15 deficiency in bone marrow derived mouse mast cells (BMMCs) results in increased susceptibility to apoptosis mediated by growth factor deprivation and A-SMase-treatment. IL-15-/- BMMCs show a constitutive stronger mRNA and protein expression as well as enzymatic activity of the members of the mitochondrial apoptotic pathways including acidic lysosomal aspartate protease cathepsin D (CTSD), endogenous acid sphingomyelinase (A-SMase), caspase-3 and -7 compared to wild type (WT) BMMCs. Furthermore, IL-15-/- BMMCs constitutively generate more A-SMase-derived ceramide than WT controls and display a decreased expression of pro-survival sphingosin-1-phosphate (SPP) both in cytosol and membrane cell fractions. Furthermore, pre-treatment of mast cells with imipramine or pepstatin A, inhibitors of the intracellular acid sphingomyelinase and cathepsin D pathways respectively, increases survival in IL-15-/- BMMCs. These findings suggest that intracellular IL-15 is a key regulator of pathways controlling primary mouse mast cell homeostasis.


Assuntos
Sobrevivência Celular/fisiologia , Interleucina-15/fisiologia , Mastócitos/citologia , Mastócitos/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Ceramidas/metabolismo , Substâncias de Crescimento/farmacologia , Substâncias de Crescimento/fisiologia , Homeostase/fisiologia , Interleucina-15/deficiência , Interleucina-15/genética , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA Mensageiro/genética
13.
Pulm Pharmacol Ther ; 22(4): 297-304, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19254776

RESUMO

Acute respiratory failure in neonates (e.g. ARDS, meconium aspiration pneumonitis, pneumonia) is characterized by an excessive inflammatory response, governing the migration of polymorpho-nuclear leukocytes (PMNLs) into lung tissue and causing consecutive impairment of gas exchange and lung function. Critical to this inflammatory response is the activation of nuclear factor-kappaB (NF-kappaB) that is required for transcription of the genes for many pro-inflammatory mediators. We asked whether the inhibition of NF-kappaB activity using either a selective inhibitor (IKK-NBD peptide) or dexamethasone would be more effective in decreasing NF-kappaB activity and chemokine expression in pulmonary cells. Changes in lung function were repeatedly assessed for 24h following induction of acute respiratory failure and therapeutic intervention. We conducted a randomized, controlled, prospective animal study with mechanically ventilated newborn piglets which underwent repeated airway lavage (20+/-2 [SEM]) to remove surfactant and to induce lung inflammation. Admixed to 100 mg kg(-1) surfactant, piglets then received either IKK-NBD peptide (S+IKK), a selective inhibitor of NF-kappaB activation, its control peptide without intrinsic activity, dexamethasone (S+Dexa), its solvent aqua, or an air bolus only (all groups n=8). After 24h of mechanical ventilation, the following differences were measured: PaO(2)/FiO(2) (S+IKK 230+/-9 mm Hg vs. S+Dexa 188+/-14, p<0.05); ventilation efficiency index (0.18+/-0.01 [3800/(PIP-PEEP)(*)f(*)PaCO(2)] vs. 0.14+/-0.01, p<0.05); extravascular lung water (24+/-1 ml kg(-1) vs. 29+/-2, p<0.05); PMNL in BAL fluid (112+/-21 cells microl(-1) vs. 208+/-34, p<0.05), IL-8 (351+/-117 pg ml(-1) vs. 491+/-144, p=ns) and leukotriene B(4) (23+/-7 pg ml(-1) vs. 71+/-11, p<0.01) in BAL fluid. NF-kappaB activity in the nucleus of pulmonary cells differed by 32+/-5% vs. 55+/-3, p<0.001. Differences between these two intervention groups were more pronounced in the second half of the observation period (hours 12-24). At 24h of mechanical ventilation, inhibition of NF-kappaB activity by IKK-NBD peptide admixed to surfactant as a carrier caused improved gas exchange, lung function and reduced pulmonary inflammation, as evidenced by reduction in PMNL migration into lung tissue due to reduced nuclear NF-kappaB activity. We conclude that IKK-NBD admixture to surfactant in acute neonatal respiratory failure is superior to dexamethasone administration within the first 24h.


Assuntos
Animais Recém-Nascidos/fisiologia , Anti-Inflamatórios/farmacologia , Dexametasona/farmacologia , Inflamação/complicações , Inflamação/patologia , Pneumopatias/patologia , Pneumopatias/prevenção & controle , NF-kappa B/antagonistas & inibidores , Doenças Respiratórias/complicações , Doenças Respiratórias/patologia , Doença Aguda , Animais , Contagem de Células Sanguíneas , Líquido da Lavagem Broncoalveolar/citologia , Interleucina-8/metabolismo , Leucotrieno B4/metabolismo , Neutrófilos/fisiologia , Tamanho do Órgão , Troca Gasosa Pulmonar , Surfactantes Pulmonares/uso terapêutico , Respiração Artificial/efeitos adversos , Suínos
14.
Am J Respir Crit Care Med ; 177(11): 1233-41, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18310483

RESUMO

RATIONALE: In acute inflammatory lung disease in newborn infants, exogenous surfactant only transiently improves lung function. We hypothesized that the transient nature of this protection is in part explained by elevated acid sphingomyelinase (a-SMase) activity that may inactivate surfactant and promote proinflammatory responses. OBJECTIVES: We investigated the intermediate-term effects (>12 h) of a-SMase inhibition in a neonatal piglet model of repeated airway lavage by the intratracheal use of the a-SMase inhibitor imipramine, together with exogenous surfactant as a carrier substance. METHODS: After surfactant washout and induction of pulmonary inflammation, lung function was monitored over 24 hours of mechanical ventilation and followed by ex vivo analyses. In addition, we studied the effect of lipopolysaccharide inhalation in a-SMase-deficient mice at 48 hours. MEASUREMENTS AND MAIN RESULTS: Surfactant washout increased both pulmonary a-SMase activity and ceramide content; this was attenuated by surfactant and prevented in the surfactant plus imipramine group. Compared with surfactant alone, Pa(O(2)), dynamic compliance, and extravascular lung water were improved in the final 12 hours in the surfactant plus imipramine group. At 24 hours, lavage fluid leukocyte counts and IL-8 concentrations decreased, and physical surfactant film properties improved. In the mouse model at 48 hours, a-SMase-deficient mice showed reduced pulmonary ceramide levels and attenuated leukocyte influx into the alveolar space. CONCLUSIONS: We conclude that stabilization of exogenous surfactant by adding imipramine to create a "fortified surfactant preparation" improves lung function in a clinically relevant piglet model, and that this effect can be attributed to the inhibition of a-SMase as evidenced in the mouse model.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Imipramina/uso terapêutico , Troca Gasosa Pulmonar/fisiologia , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/fisiopatologia , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Lavagem Broncoalveolar , Ceramidas/metabolismo , Modelos Animais de Doenças , Complacência Pulmonar/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Edema Pulmonar/etiologia , Edema Pulmonar/metabolismo , Edema Pulmonar/prevenção & controle , Síndrome do Desconforto Respiratório/etiologia , Suínos , Fatores de Tempo
15.
J Neurochem ; 106(3): 1459-70, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18498440

RESUMO

The physiological function of the cellular prion protein (PrP(c)) is unclear. PrP(c) associates with lipid rafts, highly glycolipid-rich membrane domains containing a large variety of signaling molecules, e.g., sphingolipids (SL). In this study, we investigated possible connections between PrP(c) and sphingolipid-associated signaling pathways. Using PrP(c)-wt and PrP(c)-k.o. hippocampal cell lines and mouse brains we showed higher activity of neutral and acid sphingomyelinase (SMase) in PrP(c)-k.o.-groups, while ceramide and sphingomyelin-levels were unchanged. Furthermore, despite lower basal expression levels of sphingosine kinase (SphK) in PrP(c)-k.o.-groups, the levels of its metabolite sphingosine-1-phosphate were increased, whereas S1P(3)-receptor expression was higher in PrP(c)-wt-groups again. In addition, we detected enhanced activity of phospholipase D1, an enzyme that seems to be suitable to act as a connector between the S1P(3) receptor and continuative signaling. Finally, evidence for an impact on downstream signaling cascades, especially activation of the PI3K/Akt pathway, was found. In summary, our data suggest that PrP(c) is involved in sphingolipid-associated signaling, modulating pathways that exert anti-apoptotic functions, hence indicating that PrP(c) plays a role in neuroprotection.


Assuntos
Proteínas PrPC/fisiologia , Transdução de Sinais/fisiologia , Esfingolipídeos/fisiologia , Animais , Proteínas Reguladoras de Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fármacos Neuroprotetores/metabolismo , Proteínas PrPC/genética
16.
Oncotarget ; 8(12): 20067-20085, 2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-28223543

RESUMO

Acid sphingomyelinase (A-SMase) plays an important role in the initiation of CD95 signaling by forming ceramide-enriched membrane domains that enable clustering and activation of the death receptors. In TNF-R1 and TRAIL-R1/R2 signaling, A-SMase also contributes to the lysosomal apoptosis pathway triggered by receptor internalization. Here, we investigated the molecular mechanism of CD95-mediated A-SMase activation, demonstrating that A-SMase is located in internalized CD95-receptosomes and is activated by the CD95/CD95L complex in a biphasic manner.Since several caspases have been described to be involved in the activation of A-SMase, we evaluated expression levels of caspase-8, caspase-7 and caspase-3 in CD95-receptosomes. The occurrence of cleaved caspase-8 correlated with the first peak of A-SMase activity and translocation of the A-SMase to the cell surface which could be blocked by the caspase-8 inhibitor IETD.Inhibition of CD95-internalization selectively reduced the second phase of A-SMase activity, suggesting a fusion between internalized CD95-receptosomes and an intracellular vesicular pool of A-SMase. Further analysis demonstrated that caspase-7 activity correlates with the second phase of the A-SMase activity, whereas active caspase-3 is present at early and late internalization time points. Blocking caspases-7/ -3 by DEVD reduced the second phase of A-SMase activation in CD95-receptosomes suggesting the potential role of caspase-7 or -3 for late A-SMase activation.In summary, we describe a biphasic A-SMase activation in CD95-receptosomes indicating (I.) a caspase-8 dependent translocation of A-SMase to plasma membrane and (II.) a caspase-7 and/or -3 dependent fusion of internalized CD95-receptosomes with intracellular A-SMase-containing vesicles.


Assuntos
Linfócitos B/patologia , Caspases/metabolismo , Proteína Ligante Fas/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Receptor fas/metabolismo , Apoptose , Linfócitos B/enzimologia , Inibidores de Caspase/farmacologia , Caspases/química , Membrana Celular/metabolismo , Proliferação de Células , Ativação Enzimática , Humanos , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
17.
FASEB J ; 19(14): 1945-56, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16319138

RESUMO

Although numerous studies have implicated the sphingolipid ceramide in the induction of cell death, a causative function of ceramide in caspase-dependent apoptosis remains a highly debated issue. Here, we show that ceramide is a key mediator of a distinct route to programmed cell death (PCD), i.e., caspase-independent PCD. Under conditions where apoptosis is either not initiated or actively inhibited, TNF induces caspase-independent PCD in L929 fibrosarcoma cells, NIH3T3 fibroblasts, human leukemic Jurkat T cells, and lung fibroblasts by increasing intracellular ceramide levels prior to the onset of cell death. Survival is significantly enhanced when ceramide accumulation is prevented, as demonstrated in fibroblasts genetically deficient for acid sphingomyelinase, in L929 cells overexpressing acid ceramidase, by pharmacological intervention, or by RNA interference. Jurkat cells deficient for receptor-interacting protein 1 (RIP1) do not accumulate ceramide and therefore are fully resistant to caspase-independent PCD whereas Jurkat cells overexpressing the mitochondrial protein Bcl-2 are partially protected, implicating RIP1 and mitochondria as components of the ceramide death pathway. Our data point to a role of caspases (but not cathepsins) in suppressing the ceramide death pathway under physiological conditions. Moreover, clonogenic survival of tumor cells is clearly reduced by induction of the ceramide death pathway, promising additional options for the development of novel tumor therapies.


Assuntos
Apoptose , Caspases/metabolismo , Ceramidas/farmacologia , Animais , Benzoquinonas , Linhagem Celular , Ceramidas/metabolismo , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Immunoblotting , Células Jurkat , Lactamas Macrocíclicas , Pulmão/metabolismo , Potenciais da Membrana , Camundongos , Mitocôndrias/metabolismo , Células NIH 3T3 , Proteínas Serina-Treonina Quinases/metabolismo , Quinonas/farmacologia , Interferência de RNA , Espécies Reativas de Oxigênio , Proteína Serina-Treonina Quinases de Interação com Receptores , Esfingomielina Fosfodiesterase/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
19.
J Invest Dermatol ; 122(6): 1423-31, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15175033

RESUMO

A defective permeability barrier leads to the penetration of environmental allergens into the skin and initiates immunological reactions and inflammation crucially involved in the pathogenesis of atopic dermatitis (AD). Decreased stratum corneum ceramide content may cause the defect in permeability barrier function consistently found in AD. Acid and neutral sphingomyelinase (A- and N-SMase) generate ceramides with structural and signal transduction functions in epidermal proliferation and differentiation. We determined epidermal SMase activities, DNA synthesis, involucrin, loricrin, filaggrin, and keratin expression in lesional and non-lesional skin of AD patients. We found decreased epidermal A-SMase activity in lesional and non-lesional skin, correlating with reduced stratum corneum ceramide content and disturbed barrier function. N-SMase activity was reduced in non-lesional skin and more significantly reduced in lesional skin, correlating with impaired expression of cornified envelope proteins and keratins, important for skin barrier function. Changes in involucrin, loricrin, filaggrin, keratin K 5 (basal) and K 16 (proliferation associated) were noticed in non-lesional and lesional skin, whereas changes in K 10 (suprabasal), K 6 (proliferation associated), and K 17 (inflammation associated) were found only in lesional skin. In summary, reduction in SMase-generating ceramides and impaired differentiation are involved in the defective barrier function found in AD.


Assuntos
Dermatite Atópica/metabolismo , Dermatite Atópica/patologia , Epiderme/enzimologia , Epiderme/patologia , Esfingomielina Fosfodiesterase/metabolismo , Biomarcadores , Western Blotting , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Ceramidas/metabolismo , Feminino , Proteínas Filagrinas , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Queratinas/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Precursores de Proteínas/metabolismo , Transdução de Sinais/fisiologia , Especificidade por Substrato , Água/metabolismo
20.
Mol Cell Biol ; 34(17): 3214-28, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24980434

RESUMO

Signaling by tumor necrosis factor (TNF) receptor 1 (TNF-R1), a prototypic member of the death receptor family, mediates pleiotropic biological outcomes ranging from inflammation and cell proliferation to cell death. Although many elements of specific signaling pathways have been identified, the main question of how these selective cell fate decisions are regulated is still unresolved. Here we identified TNF-induced K63 ubiquitination of TNF-R1 mediated by the ubiquitin ligase RNF8 as an early molecular checkpoint in the regulation of the decision between cell death and survival. Downmodulation of RNF8 prevented the ubiquitination of TNF-R1, blocked the internalization of the receptor, prevented the recruitment of the death-inducing signaling complex and the activation of caspase-8 and caspase-3/7, and reduced apoptotic cell death. Conversely, recruitment of the adaptor proteins TRADD, TRAF2, and RIP1 to TNF-R1, as well as activation of NF-κB, was unimpeded and cell growth and proliferation were significantly enhanced in RNF8-deficient cells. Thus, K63 ubiquitination of TNF-R1 can be sensed as a new level of regulation of TNF-R1 signaling at the earliest stage after ligand binding.


Assuntos
Receptores Tipo I de Fatores de Necrose Tumoral/química , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Ubiquitinação , Animais , Apoptose/fisiologia , Caspase 8/metabolismo , Linhagem Celular , Sobrevivência Celular/fisiologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endocitose , Proteína de Domínio de Morte Associada a Fas/metabolismo , Células HeLa , Humanos , Camundongos , Modelos Biológicos , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Células U937 , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa