Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Surg Res ; 259: 200-210, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33307511

RESUMO

BACKGROUND: It remains unknown whether epithelial-mesenchymal transition (EMT)-mediated vascular invasion and cancer stemness are associated with sphingosine-1-phosphate receptor-1 (S1PR1) expression in human hepatocellular carcinoma (HCC). The aim of this study was to investigate the correlation between S1PR1 expression and prognosis of patients with primary HCC and to define the potential of S1PR as a therapeutic target. MATERIALS AND METHODS: We investigated 108 patients who underwent primary surgical resection for HCC treatment. Expression of S1PR1 and EMT markers was analyzed to predict prognosis of patients with HCC. Furthermore, three-dimensional organotypic culture, anoikis assay, and cell invasion were performed to validate the association of S1PR1 with EMT and cancer stemness. RESULTS: Among patients with HCC, the high S1PR1 expression group had significantly shorter overall survival than the low expression group. Moreover, high S1PR1 expression was significantly associated with shorter recurrence-free survival, increased risk of portal and hepatic vein invasion, and intrahepatic metastasis. Multivariate analyses revealed that S1PR1 overexpression was an independent prognostic factor in patients with HCC. S1PR1 overexpression positively correlated with vimentin and MMP-9 expression and negatively correlated with E-cadherin. In addition, S1PR1 overexpression induced EMT and enhanced tumor invasion and cancer stemness. CONCLUSIONS: S1PR1 overexpression, via EMT-induced vascular invasion and increased cancer stem cell properties, establishes a metastatic niche, enhances the capacity of hematogenous metastasis, and associates with poor outcomes in patients with HCC. Hence, S1PR1 may serve as a therapeutic target for patients with HCC with vascular invasion.


Assuntos
Carcinoma Hepatocelular/patologia , Transição Epitelial-Mesenquimal , Neoplasias Hepáticas/patologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Idoso , Carcinoma Hepatocelular/mortalidade , Linhagem Celular Tumoral , Feminino , Veias Hepáticas/patologia , Humanos , Neoplasias Hepáticas/mortalidade , Masculino , Metaloproteinase 9 da Matriz/análise , Pessoa de Meia-Idade , Invasividade Neoplásica , Veia Porta/patologia , Vimentina/análise
2.
J Immunol ; 203(9): 2401-2414, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31548329

RESUMO

Ligand-engaged chemoattractant receptors trigger Gαi subunit nucleotide exchange, stimulating the activation of downstream effector molecules. Activated chemoattractant receptors also dock G protein-coupled receptor kinases (GRKs) that help mediate receptor desensitization. In this study, we show that the B cell-specific loss of GRK2 severely disrupts B cell trafficking and immune cell homeostasis. The GRK2 deficiency in developing murine B cells leads to a severe immune phenotype, including a major reduction of bone marrow IgD+ cells, splenomegaly with a loss of white pulp and grossly expanded red pulp, a deficit of Peyer patches, and small lymph nodes with marked reductions in B cell numbers. The major phenotypes in these mice arise from excessive S1PR1 signaling combined with inadequate homeostatic chemokine receptor signaling. CXCL13 signaling is the most severely compromised. In B cells, our data also indicate that S1PR1 signals constitutively, as blocking S1PR1 signaling with an S1PR1 antagonist enhanced CXCL13-triggered wild-type B cell migration. Furthermore, blocking S1PR1 signaling in the GRK2-deficient B cells partially corrected their poor response to chemokines. Treating mice lacking GRK2 expression in their B cells with an S1PR1 antagonist partially normalized B cell trafficking into lymph node and splenic follicles. These findings reveal the critical interdependence of Gαi-linked signaling pathways in controlling B lymphocyte trafficking.


Assuntos
Linfócitos B/fisiologia , Homeostase , Tecido Linfoide/fisiologia , Receptores de Quimiocinas/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Animais , Cálcio/metabolismo , Movimento Celular , Quimiocina CXCL13/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Leucocitose/imunologia , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores CXCR4/fisiologia , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia
3.
Can J Physiol Pharmacol ; 99(12): 1280-1287, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34310896

RESUMO

Skin flap transfer is an important method to repair and reconstruct various tissue defects; however, avascular necrosis largely affects the success of flap transfer. The sphingosine 1-phosphate receptor 1 (S1PR1) agonist SEW2871 has been proven to ameliorate ischemic injury; however, its effect on flap survival has not been reported. In this study, an experimental skin flap model was established in rats to investigate the roles of SEW2871. The results indicated that SEW2871 greatly increased the survival of the skin flap, alleviated pathological injury, promoted the angiogenesis, and inhibited cells apoptosis in skin flap tissues. SEW2871 activated S1PR1 downstream signaling pathways, including heat shock protein 27 (HSP27), extracellular regulated protein kinases (ERK), and protein kinase B (Akt). In addition, SEW2871 promoted the expression of S1PR1. These findings may provide novel insights for skin flap transfer.


Assuntos
Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/genética , Oxidiazóis/farmacologia , Transplante de Pele/métodos , Pele/irrigação sanguínea , Receptores de Esfingosina-1-Fosfato/fisiologia , Retalhos Cirúrgicos/transplante , Tiofenos/farmacologia , Indutores da Angiogênese , Animais , Apoptose/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Pele/patologia , Receptores de Esfingosina-1-Fosfato/genética , Receptores de Esfingosina-1-Fosfato/metabolismo , Retalhos Cirúrgicos/irrigação sanguínea
4.
Int J Neurosci ; 131(1): 85-94, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32148137

RESUMO

Objective: Neuroinflammation is a central part of cerebral ischemia/reperfusion injury. The novel immune suppressant, fingolimod, is a promising candidate to ameliorate stroke-induced damage. Fingolimod is efficacious in experimental ischemic models, but a rigorous meta-analysis is lacking that considers how different experiment variables affect outcomes.Methods: We conducted a systematic literature review of fingolimod in stroke models, with the aim of rigorously evaluating fingolimod's effects on reducing infarct volume improving neurological outcomes. Seventeen variables were evaluated as covariates for the source of heterogeneity, and effect sizes were combined by using normalized mean difference meta-analysis to evaluate efficacy. Study quality was evaluated by the CAMARADES ten-item checklist, and publication bias was evaluated by funnel plots and Egger's tests.Results: About 123 unduplicated articles were identified in the literature research. Of these papers, 118 articles were excluded after reading titles and abstracts. Another 17 articles were selected in this study. Study quality was moderate (median = 6; interquartile range = 4), and publication bias was statistically insignificant. fingolimod reduced infarct volume by 30.4% (95% CI 22.4%-38.3%; n = 24; I2 = 90.0%; p < 0.0001) and consistently enhanced neurobehavioral outcome by 34.2% (95% CI 23.1%-45.2%; n = 14; I2 = 76.5%; p < 0.0001). No single factors accounted for heterogeneity.Conclusions: Our rigorous statistical evaluation confirmed the neuroprotective properties of fingolimod. New data can be used in designing future clinical trials.


Assuntos
Modelos Animais de Doenças , Cloridrato de Fingolimode/uso terapêutico , Moduladores do Receptor de Esfingosina 1 Fosfato/uso terapêutico , Receptores de Esfingosina-1-Fosfato/agonistas , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Cloridrato de Fingolimode/farmacologia , Humanos , Moduladores do Receptor de Esfingosina 1 Fosfato/farmacologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Acidente Vascular Cerebral/patologia , Resultado do Tratamento
5.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34502385

RESUMO

Erythropoietin (Epo) is the critical hormone for erythropoiesis. In adults, Epo is mainly produced by a subset of interstitial fibroblasts in the kidney, with minor amounts being produced in the liver and the brain. In this study, we used the immortalized renal interstitial fibroblast cell line FAIK F3-5 to investigate the ability of the bioactive sphingolipid sphingosine 1-phosphate (S1P) to stimulate Epo production and to reveal the mechanism involved. Stimulation of cells with exogenous S1P under normoxic conditions (21% O2) led to a dose-dependent increase in Epo mRNA and protein levels and subsequent release of Epo into the medium. S1P also enhanced the stabilization of HIF-2α, a key transcription factor for Epo expression. S1P-stimulated Epo mRNA and protein expression was abolished by HIF-2α mRNA knockdown or by the HIF-2 inhibitor compound 2. Furthermore, the approved S1P receptor modulator FTY720, and its active form FTY720-phosphate, both exerted a similar effect on Epo expression as S1P. The effect of S1P on Epo was antagonized by the selective S1P1 and S1P3 antagonists NIBR-0213 and TY-52156, but not by the S1P2 antagonist JTE-013. Moreover, inhibitors of the classical MAPK/ERK, the p38-MAPK, and inhibitors of protein kinase (PK) C and D all blocked the effect of S1P on Epo expression. Finally, the S1P and FTY720 effects were recapitulated in the Epo-producing human neuroblastoma cell line Kelly, suggesting that S1P receptor-dependent Epo synthesis is of general relevance and not species-specific. In summary, these data suggest that, in renal interstitial fibroblasts, which are the primary source of plasma Epo, S1P1 and 3 receptor activation upregulates Epo under normoxic conditions. This may have a therapeutic impact on disease situations such as chronic kidney disease, where Epo production is impaired, causing anemia, but it may also have therapeutic value as Epo can mediate additional tissue-protective effects in various organs.


Assuntos
Eritropoetina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Linhagem Celular , Células Cultivadas , Eritropoese , Eritropoetina/fisiologia , Fibroblastos/metabolismo , Cloridrato de Fingolimode/metabolismo , Humanos , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Rim/metabolismo , Lisofosfolipídeos/metabolismo , Camundongos , Ligação Proteica , Receptores de Lisoesfingolipídeo/metabolismo , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/fisiologia
6.
FASEB J ; 33(6): 7180-7191, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30844311

RESUMO

Epithelial-mesenchymal transition (EMT) is a critical process implicated in the initial stage of cancer metastasis, which is the major cause of tumor recurrence and mortality. Although key transcription factors that regulate EMT, such as snail family transcriptional repressor 2 (SNAI2), are well characterized, the upstream signaling pathways controlling these transcriptional mediators are largely unknown, which limits therapeutic strategies. Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator, generated by sphingosine kinases (SPHK1 and SPHK2), that mainly exerts its effects by binding to the following 5 GPCRs: S1P1 to S1P5. S1P signaling has been reported to regulate different aspects of cancer progression including cell proliferation, apoptosis, and migration; nevertheless, its role in cancer metastasis, specifically via EMT, is not established. Here we show that SPHK1 expression correlates significantly with EMT score in breast cancer cell lines, and with SNAI2 in patient-derived breast tumors. Cell-based assays demonstrate that S1P can rapidly up-regulate the expression of SNAI2 in breast cancer cells via the activation of cognate receptors S1P2 and S1P3. Knockdown studies suggest that S1P2 and S1P3 mediate this effect by activating myocardin-related transcription factor A (MRTF-A) and yes-associated protein (YAP), respectively. Michigan Cancer Foundation 7 cells stably overexpressing S1P2 or S1P3 exhibit a more invasive phenotype, when compared to control cells. Taken together, our findings suggest that S1P produced by SPHK1 induces SNAI2 expression via S1P2-YAP and S1P3-MRTF-A pathways, leading to enhanced cell invasion. Cumulatively, this study reveals a novel mechanism by which S1P activates parallel pathways that regulate the expression of SNAI2, a master regulator of EMT, and provides new insights into druggable therapeutic targets that may limit cancer metastasis. Wang, W., Hind, T., Lam, B. W. S., Herr, D. R. Sphingosine 1-phosphate signaling induces SNAI2 expression to promote cell invasion in breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal/fisiologia , Lisofosfolipídeos/fisiologia , Invasividade Neoplásica/patologia , Proteínas de Neoplasias/biossíntese , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Fatores de Transcrição da Família Snail/biossíntese , Esfingosina/análogos & derivados , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Lisofosfolipídeos/farmacologia , Células MCF-7 , Invasividade Neoplásica/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Interferência de RNA , Estabilidade de RNA , RNA Interferente Pequeno/farmacologia , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/fisiologia , Esfingosina/farmacologia , Esfingosina/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Transativadores/antagonistas & inibidores , Transativadores/genética , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Proteínas de Sinalização YAP
7.
Rev Neurol (Paris) ; 176(1-2): 100-112, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31757428

RESUMO

The ideal treatment for multiple sclerosis (MS) would target both the neuroinflammatory component of the disease (peripheral and central) and its neurodegenerative component, via modulation of a ubiquitous and pleiotropic common target. Sphingosine-1-phosphate (S1P), a product of sphingosine metabolism, regulates many biological functions (including cell proliferation and survival, cell migration, the immune response and cardiovascular function) via five subtypes of receptor. These receptors are expressed in all types of brain cells where they modulate a number of processes involved in neuronal plasticity, including myelination, neurogenesis and neuroprotection. This profile has aroused interest in modulation of S1P function as a therapeutic target in many brain diseases, particularly those in which the immune system plays a role in the development of brain lesions. Fingolimod, a S1P receptor modulator, exerts its beneficial effects in MS through its anti-inflammatory and anti-neurodegenerative effects. This review discusses recent evidence indicating that fingolimod may target both the inflammatory and neurodegenerative components of the disease process in MS.


Assuntos
Imunossupressores/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Receptores de Esfingosina-1-Fosfato/antagonistas & inibidores , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/fisiologia , Imunossupressores/farmacologia , Lisofosfolipídeos/metabolismo , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/fisiologia
8.
J Physiol ; 597(7): 2007-2019, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30793318

RESUMO

KEY POINTS: Sphingosine-1-phosphate (S1P) strongly activates mouse vagal C-fibres in the airways. Airway-specific nodose and jugular C-fibre neurons express mRNA coding for the S1P receptor S1PR3. S1P activation of nodose C-fibres is inhibited by a S1PR3 antagonist. S1P activation of nodose C-fibres does not occur in S1PR3 knockout mice. ABSTRACT: We evaluated the effect of sphingosine-1-phosphate (S1P), a lipid that is elevated during airway inflammatory conditions like asthma, for its ability to stimulate vagal afferent C-fibres in mouse lungs. Single cell RT-PCR on lung-specific vagal afferent neurons revealed that both TRPV1-expressing and TRPV1-non-expressing nodose neurons express mRNA coding for the S1P receptor S1PR3. TRPV1-expressing airway-specific jugular ganglion neurons also express S1PR3 mRNA. S1PR1 and S1PR2 mRNAs were also found to be expressed but only in a limited subset (32% and 22%, respectively) of airway-specific vagal sensory neurons; whereas S1PR4 and S1PR5 were rarely expressed. We used large scale two-photon imaging of the nodose ganglia from our ex vivo preparation isolated from Pirt-Cre;R26-GCaMP6s transgenic mice, which allows for simultaneous monitoring of calcium transients in ∼1000 neuronal cell bodies in the ganglia during tracheal perfusion with S1P (10 µM). We found that S1P in the lungs strongly activated 81.5% of nodose fibres, 70% of which were also activated by capsaicin. Single fibre electrophysiological recordings confirmed that S1P evoked action potential (AP) generation in a concentration-dependent manner (0.1-10 µM). Action potential generation by S1P in nodose C-fibres was effectively inhibited by the S1PR3 antagonist TY 52156 (10 µM). Finally, in S1PR3 knockout mice, S1P was not able to activate any of the airway nodose C-fibres analysed. These results support the hypothesis that S1P may play a role in evoking C-fibre-mediated airway sensations and reflexes that are associated with airway inflammatory diseases.


Assuntos
Lisofosfolipídeos/farmacologia , Células Receptoras Sensoriais/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Esfingosina/análogos & derivados , Nervo Vago/citologia , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato/genética
9.
Neurobiol Dis ; 124: 189-201, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30468865

RESUMO

Multiple sclerosis is an inflammatory disease of the central nervous system (CNS) in which multiple sites of blood-brain barrier (BBB) disruption, focal inflammation, demyelination and tissue destruction are the hallmarks. Here we show that sphingosine-1-phosphate receptor 2 (S1PR2) has a negative role in myelin repair as well as an important role in demyelination by modulating BBB permeability. In lysolecithin-induced demyelination of adult mouse spinal cord, S1PR2 inactivation by either the pharmacological inhibitor JTE-013 or S1PR2 gene knockout led to enhanced myelin repair as determined by higher numbers of differentiated oligodendrocytes and increased numbers of remyelinated axons at the lesion sites. S1PR2 inactivation in lysolecithin-induced demyelination of the optic chiasm, enhanced oligodendrogenesis and improved the behavioral outcome in an optokinetic reflex test. In order to see the effect of S1PR2 inactivation on demyelination, experimental autoimmune encephalitis (EAE) was induced by MOG-peptide. S1PR2 inhibition or knockout decreased the extent of demyelinated areas as well as the clinical disability in this EAE model. Both toxin induced and EAE models showed decreased BBB leakage and reduced numbers of Iba1+ macrophages following S1PR2 inactivation. Our results suggest that S1PR2 activity impairs remyelination and also enhances BBB leakage and demyelination. The former effect could be mediated by Nogo-A, as antagonism of this factor enhances remyelination and S1PR2 can act as a Nogo-A receptor.


Assuntos
Esclerose Múltipla/fisiopatologia , Remielinização , Receptores de Esfingosina-1-Fosfato/fisiologia , Animais , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/fisiologia , Esclerose Múltipla/patologia , Bainha de Mielina/ultraestrutura , Receptores de Esfingosina-1-Fosfato/genética , Medula Espinal/patologia , Medula Espinal/fisiopatologia
10.
Acta Neuropathol ; 138(2): 275-293, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31062076

RESUMO

Glioblastomas strongly invade the brain by infiltrating into the white matter along myelinated nerve fiber tracts even though the myelin protein Nogo-A prevents cell migration by activating inhibitory RhoA signaling. The mechanisms behind this long-known phenomenon remained elusive so far, precluding a targeted therapeutic intervention. This study demonstrates that the prevalent activation of AKT in gliomas increases the ER protein-folding capacity and enables tumor cells to utilize a side effect of RhoA activation: the perturbation of the IRE1α-mediated decay of SPARC mRNA. Once translation is initiated, glioblastoma cells rapidly secrete SPARC to block Nogo-A from inhibiting migration via RhoA. By advanced ultramicroscopy for studying single-cell invasion in whole, undissected mouse brains, we show that gliomas require SPARC for invading into white matter structures. SPARC depletion reduces tumor dissemination that significantly prolongs survival and improves response to cytostatic therapy. Our finding of a novel RhoA-IRE1 axis provides a druggable target for interfering with SPARC production and underscores its therapeutic value.


Assuntos
Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Proteínas de Neoplasias/fisiologia , Proteínas Nogo/biossíntese , Osteonectina/biossíntese , Biossíntese de Proteínas , Substância Branca/patologia , Proteína rhoA de Ligação ao GTP/fisiologia , Animais , Ligação Competitiva , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Invasividade Neoplásica , Proteínas Nogo/genética , Osteonectina/genética , Domínios Proteicos , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato/fisiologia , Células Tumorais Cultivadas , Substância Branca/metabolismo
11.
J Invest Dermatol ; 141(5): 1188-1197.e5, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33197483

RESUMO

The outer layer of the epidermis composes the skin barrier, a sophisticated filter constituted by layers of corneocytes in a lipid matrix. The matrix lipids, especially the ceramide-generated sphingosine 1-phosphate, are the messengers that the skin barrier uses to communicate with the basal layer of the epidermis where replicating keratinocytes are located. Sphingosine 1-phosphate is a bioactive sphingolipid mediator involved in various cellular functions through S1PR1‒5, expressed by keratinocytes. We discovered that the S1pr2 absence is linked to an impairment in the skin barrier function. Although S1pr2-/- mouse skin has no difference in its phenotype and barrier function compared with that of wild-type mouse, after tape stripping, S1pr2-/- mouse showed significantly higher transepidermal water loss and required another 24 hours to normalize their transepidermal water loss levels. Moreover, after epicutaneous Staphylococcus aureus application, impaired S1pr2-/- mouse epidermal barrier function allowed deeper bacterial penetration and denser neutrophil infiltration in the dermis. Microarray and RNA sequence of S1pr2-/- mouse epidermis linked the barrier dysfunction with a decrease in FLG2 and tight junction components. In conclusion, S1pr2-/- mice have compromised skin barrier function and increased bacteria permeability, making them a suitable model for diseases that present similar characteristics, such as atopic dermatitis.


Assuntos
Epiderme/metabolismo , Homeostase/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Animais , Células Cultivadas , Proteínas Filagrinas , Humanos , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Permeabilidade , Proteínas S100/análise , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Estresse Mecânico
12.
Cancer Lett ; 506: 107-119, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33600895

RESUMO

Targeting the metastatic process to prevent disease dissemination in cancer remains challenging. One step in the metastatic cascade involves cancer cells transiting through the vascular endothelium after inflammation has increased the permeability of this cellular layer. Reducing inflammation-mediated gaps in the vascular endothelium could potentially be used to retard metastasis. This study describes the development of a novel ASR396-containing nanoparticle designed to activate the Sphingosine-1-Phosphate Receptor 1 (S1PR1) in order to tighten the junctions between the endothelial cells lining the vascular endothelium thereby inhibiting metastasis. ASR396 was derived from the S1PR1 agonist SEW2871 through chemical modification enabling the new compound to be loaded into a nanoliposome. ASR396 retained S1PR1 binding activity and the nanoliposomal formulation (nanoASR396) made it systemically bioavailable upon intravenous injection. Studies conducted in microvessels demonstrated that nanoASR396 significantly attenuated inflammatory mediator-induced permeability increase through the S1PR1 activation. Similarly, nanoASR396 inhibited gap formation mediated by inflammatory agents on an endothelial cell monolayer by decreasing levels of phosphorylated myosin light chain protein thereby inhibiting cellular contractility. In animal models, nanoASR396 inhibited lung metastasis by up to 80%, indicating its potential for retarding melanoma metastasis. Thus, a novel bioavailable nanoparticle-based S1PR1 agonist has been developed to negate the effects of inflammatory mediators on the vascular endothelium in order to reduce the metastatic dissemination of cancer cells.


Assuntos
Células Endoteliais/metabolismo , Cadeias Leves de Miosina/metabolismo , Metástase Neoplásica/prevenção & controle , Receptores de Esfingosina-1-Fosfato/fisiologia , Animais , Permeabilidade Capilar , Linhagem Celular Tumoral , Humanos , Lipossomos , Camundongos , Nanopartículas , Oxidiazóis/farmacologia , Fosforilação , Transdução de Sinais/fisiologia , Receptores de Esfingosina-1-Fosfato/agonistas , Tiofenos/farmacologia
13.
Neuromolecular Med ; 23(1): 211-223, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32914259

RESUMO

Sphingosine 1-phosphate (S1P) is an important lipid biomolecule that exerts pleiotropic cellular actions as it binds to and activates its five G-protein-coupled receptors, S1P1-5. Through these receptors, S1P can mediate diverse biological activities in both healthy and diseased conditions. S1P is produced by S1P-producing enzymes, sphingosine kinases (SphK1 and SphK2), and is abundantly present in different organs, including the brain. The medically important roles of receptor-mediated S1P signaling are well characterized in multiple sclerosis because FTY720 (Gilenya™, Novartis), a non-selective S1P receptor modulator, is currently used as a treatment for this disease. In cerebral ischemia, its role is also notable because of FTY720's efficacy in both rodent models and human patients with cerebral ischemia. In particular, some of the S1P receptors, including S1P1, S1P2, and S1P3, have been identified as pathogenic players in cerebral ischemia. Other than these receptors, S1P itself and S1P-producing enzymes have been shown to play certain roles in cerebral ischemia. This review aims to compile the current updates and overviews about the roles of S1P signaling, along with a focus on S1P receptors in cerebral ischemia, based on recent studies that used in vivo rodent models of cerebral ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Lisofosfolipídeos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Esfingosina/análogos & derivados , Animais , Dano Encefálico Crônico/etiologia , Dano Encefálico Crônico/metabolismo , Isquemia Encefálica/complicações , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Cloridrato de Fingolimode/uso terapêutico , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Inflamação , AVC Isquêmico/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Ratos , Transdução de Sinais/fisiologia , Esfingosina/fisiologia
14.
Sci Rep ; 11(1): 15308, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321503

RESUMO

Sphingosine 1-phosphate (S1P) is a bioactive signalling sphingolipid that is increased in diseases such as obesity and diabetes. S1P can modulate platelet function, however the direction of effect and S1P receptors (S1PRs) involved are controversial. Here we describe the role of S1P in regulating human platelet function and identify the receptor subtypes responsible for S1P priming. Human platelets were treated with protease-activated receptor 1 (PAR-1)-activating peptide in the presence or absence of S1P, S1PR agonists or antagonists, and sphingosine kinases inhibitors. S1P alone did not induce platelet aggregation but at low concentrations S1P enhanced PAR1-mediated platelet responses, whereas PAR1 responses were inhibited by high concentrations of S1P. This biphasic effect was mimicked by pan-S1PR agonists. Specific agonists revealed that S1PR1 receptor activation has a positive priming effect, S1PR2 and S1PR3 have no effect on platelet function, whereas S1PR4 and S1PR5 receptor activation have an inhibitory effect on PAR-1 mediated platelet function. Although platelets express both sphingosine kinase 1/2, enzymes which phosphorylate sphingosine to produce S1P, only dual and SphK2 inhibition reduced platelet function. These results support a role for SphK2-mediated S1P generation in concentration-dependent positive and negative priming of platelet function, through S1PR1 and S1PR4/5 receptors, respectively.


Assuntos
Lisofosfolipídeos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato/efeitos dos fármacos , Esfingosina/análogos & derivados , Plaquetas/efeitos dos fármacos , Plaquetas/ultraestrutura , Proteínas de Transporte/farmacologia , Forma Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Lisofosfolipídeos/agonistas , Lisofosfolipídeos/antagonistas & inibidores , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Receptor PAR-1/agonistas , Esfingosina/agonistas , Esfingosina/antagonistas & inibidores , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato/fisiologia
15.
J Biomed Mater Res A ; 109(5): 695-712, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32608188

RESUMO

Regeneration of skeletal muscle after volumetric injury is thought to be impaired by a dysregulated immune microenvironment that hinders endogenous repair mechanisms. Such defects result in fatty infiltration, tissue scarring, chronic inflammation, and debilitating functional deficits. Here, we evaluated the key cellular processes driving dysregulation in the injury niche through localized modulation of sphingosine-1-phosphate (S1P) receptor signaling. We employ dimensionality reduction and pseudotime analysis on single cell cytometry data to reveal heterogeneous immune cell subsets infiltrating preclinical muscle defects due to S1P receptor inhibition. We show that global knockout of S1P receptor 3 (S1PR3) is marked by an increase of muscle stem cells within injured tissue, a reduction in classically activated relative to alternatively activated macrophages, and increased bridging of regenerating myofibers across the defect. We found that local S1PR3 antagonism via nanofiber delivery of VPC01091 replicated key features of pseudotime immune cell recruitment dynamics and enhanced regeneration characteristic of global S1PR3 knockout. Our results indicate that local S1P receptor modulation may provide an effective immunotherapy for promoting a proreparative environment leading to improved regeneration following muscle injury.


Assuntos
Ciclopentanos/uso terapêutico , Imunoterapia/métodos , Músculo Esquelético/lesões , Regeneração/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato/fisiologia , Animais , Ciclopentanos/farmacologia , Liberação Controlada de Fármacos , Citometria de Fluxo , Leucopenia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Força Atômica , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Células Mieloides/imunologia , Nanofibras , Tamanho do Órgão , Músculo Quadríceps/imunologia , Músculo Quadríceps/lesões , Músculo Quadríceps/metabolismo , Músculo Quadríceps/patologia , Transdução de Sinais/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato/deficiência , Receptores de Esfingosina-1-Fosfato/genética , Subpopulações de Linfócitos T/imunologia , Alicerces Teciduais
16.
Neuromolecular Med ; 23(1): 47-67, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33180310

RESUMO

Sphingosine 1-phosphates (S1Ps) are bioactive lipids that mediate a diverse range of effects through the activation of cognate receptors, S1P1-S1P5. Scrutiny of S1P-regulated pathways over the past three decades has identified important and occasionally counteracting functions in the brain and cerebrovascular system. For example, while S1P1 and S1P3 mediate proinflammatory effects on glial cells and directly promote endothelial cell barrier integrity, S1P2 is anti-inflammatory but disrupts barrier integrity. Cumulatively, there is significant preclinical evidence implicating critical roles for this pathway in regulating processes that drive cerebrovascular disease and vascular dementia, both being part of the continuum of vascular cognitive impairment (VCI). This is supported by clinical studies that have identified correlations between alterations of S1P and cognitive deficits. We review studies which proposed and evaluated potential mechanisms by which such alterations contribute to pathological S1P signaling that leads to VCI-associated chronic neuroinflammation and neurodegeneration. Notably, S1P receptors have divergent but overlapping expression patterns and demonstrate complex interactions. Therefore, the net effect produced by S1P represents the cumulative contributions of S1P receptors acting additively, synergistically, or antagonistically on the neural, vascular, and immune cells of the brain. Ultimately, an optimized therapeutic strategy that targets S1P signaling will have to consider these complex interactions.


Assuntos
Demência Vascular/fisiopatologia , Lisofosfolipídeos/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Esfingosina/análogos & derivados , Aldeído Liases/antagonistas & inibidores , Aldeído Liases/fisiologia , Doença de Alzheimer/fisiopatologia , Animais , Transtornos Cerebrovasculares/fisiopatologia , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Cloridrato de Fingolimode/uso terapêutico , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/fisiopatologia , Inflamação , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/fisiopatologia , Camundongos , Camundongos Knockout , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/fisiopatologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Transdução de Sinais , Esfingosina/fisiologia , Receptores de Esfingosina-1-Fosfato/efeitos dos fármacos
17.
Brain Res ; 1739: 146831, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32278723

RESUMO

There is thought to be a strong relationship between sphingosine-1-phosphate (S1P) signaling and pathophysiolosy of cerebral ischemia. We examined the change of expression and distribution of S1P receptors (S1PRs) and sphingosine kinases (SphKs) after cerebral ischemia in male C57BL6/J mice using immunohistochemical analysis at 1, 5, 14, and 28 days after 30 min of transient middle cerebral artery occlusion (tMCAO). S1PR1, 3, and 5 were transiently induced in the cells, which were morphologically similar to neurons in the peri-infarct lesion with a peak seen at 1 day after tMCAO (p < 0.01 vs. sham control). S1PR2 appeared in the inner layer of vessels in the ischemic core (p < 0.01 vs. sham control) and the peri-infarct lesion (p < 0.01 vs. sham control) at the acute phase after tMCAO. However, SphK1 was strongly induced at 1 and 5 days after tMCAO (p < 0.01 vs. sham control) in the peri-infarct lesion, whereas SphK2 expression did not change. Western blot analysis at 1 and 5 days after 30 min of tMCAO revealed that the expression of S1PRs were transiently enhanced at the acute phase, which was consistent with the immunohistochemical results. Double immunofluorescent analysis revealed S1PR2/NG2- and S1PR2/CD31-, S1PR3/CD31-, and S1PR5/CD31-double positive cells in the peri-infarct lesion 1 day after tMCAO. The present results suggest that S1PRs and SphK1 may be important therapeutic targets for rescuing the peri-infarct lesion.


Assuntos
Infarto da Artéria Cerebral Média/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/fisiopatologia , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/fisiologia , Ativação Transcricional
18.
Biochem Pharmacol ; 165: 249-262, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30753812

RESUMO

Sphingosine kinase 1 (SphK1) and 2 (SphK2) have been shown contribute to synovial inflammation in animal models of arthritis. However, low levels of intracellular sphingosine-1 phosphate (S1P) were reported in fibroblast-like synoviocytes (FLS) from patients in the end stage of rheumatoid arthritis (RA) compared to normal FLS. Moreover, the S1P receptor-mediated chemokine synthesis was altered in RAFLS in response to chemical hypoxia. Since the mechanisms responsible for low levels of intracellular S1P in RAFLS are not fully identified, we evaluated the contribution of SphKs to the S1P-induced synthesis of chemokines under conditions of chemical hypoxia. Our results show that a chemical hypoxia mimetic cobalt chloride (CoCl2) increased SphK1 expression and activation in normal FLS but not in RAFLS. Using selective inhibitors of SphKs and gene silencing approaches, we provide evidence that both SphK1 and SphK2 are involved in hypoxia-induced chemokine production in normal FLS. In contrast, only SphK2 mediates hypoxia-induced chemokine production in RAFLS. Moreover, CoCl2 increased S1P2 and S1P3 receptor mRNA levels in normal FLS but not in RAFLS. The data suggest that altered expression and/or activation of SphK1 combined with reduced induction of S1P receptor expression by CoCl2 impaired the CoCl2-mediated autocrine S1P receptor signaling loop and chemokine production in RAFLS.


Assuntos
Artrite Reumatoide/enzimologia , Fibroblastos/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Membrana Sinovial/enzimologia , Hipóxia Celular , Células Cultivadas , Quimiocinas/metabolismo , Cobalto/farmacologia , Ativação Enzimática , Humanos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Receptores de Esfingosina-1-Fosfato/fisiologia
19.
Ann Endocrinol (Paris) ; 80(5-6): 263-272, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31455516

RESUMO

Sphingosine-1-phoshate (S1P) is a membrane sphingolipid involved in several physiological processes, including cell proliferation, tissue growth, cell survival and migration, inflammation, vasculogenesis, and angiogenesis. Herein, we review the most critical effects of S1P on ovarian function, including its physiological and pathophysiological effects. Based on the available evidence, S1P plays an important role in ovarian physiology, participating as an essential stimulator of follicular development in both the preantral and antral phases, as well as in ovulation and corpus luteum development. Moreover, S1P may be a good cytoprotective agent against cancer treatment side-effects (chemotherapy with or without radiation therapy). In the future, this compound may be given for fertility preservation to women undergoing cancer treatment. However, further studies are required to confirm its efficacy in ovarian protection and also its safety in terms of cancer prognosis, given the biological action of the compound. Under- or over-production of S1P may be related to ovarian pathologies.


Assuntos
Lisofosfolipídeos/fisiologia , Doenças Ovarianas/fisiopatologia , Ovário/fisiopatologia , Esfingosina/análogos & derivados , Animais , Proliferação de Células , Corpo Lúteo/crescimento & desenvolvimento , Feminino , Preservação da Fertilidade , Humanos , Doenças Ovarianas/patologia , Folículo Ovariano/crescimento & desenvolvimento , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/fisiopatologia , Ovário/patologia , Esfingosina/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia
20.
Thyroid ; 29(5): 700-713, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30963819

RESUMO

Background: There is growing evidence that sphingosine-1-phosphate (S1P), a pleiotropic bioactive sphingolipid metabolite synthesized intracellularly by two closely related sphingosine kinases (SphKs), SphK1 and SphK2, is involved in inflammation. However, the role of SphKs/S1P/S1P receptors (S1PRs) in autoimmune thyroiditis (AIT) has not been studied to date. Methods: This study examined whether SphK1/S1P/S1PR1 signaling is aberrantly altered in thyroid tissues and serum of both AIT patients and a spontaneously autoimmune thyroiditis (SAT) mouse model. Murine CD4+T cells were employed to further investigate the downstream signaling of SphK1/S1P/S1PR1. Furthermore, a total of 102 NOD.H-2h4 mice, randomly divided into different groups, were used to investigate the therapeutic effect of S1PR1 blockade and its potential mechanism. Results: We found that components of the SphK1/S1P/S1PR1 pathway were abnormally expressed in patients with Hashimoto thyroiditis and in a SAT mouse model. In addition, S1P could activate signal transducer and activator of transcription 3 (STAT3) through S1PR1 and its downstream signaling pathways in CD4+T cells of NOD.H-2h4 mice. Furthermore, an in vivo study demonstrated that blocking S1PR1 by FTY720 administration could reduce the incidence and severity of thyroiditis and goiter in SAT mice in a time-dependent manner. The proportions of STAT3-related and inflammation-related cell subtypes, such as T helper 1, T helper 17, and follicular T helper cells, were elevated in the SAT group when compared to the control group, and these cell subtypes decreased after FTY720 administration. Furthermore, the downstream inflammatory cytokines of STAT3 were also downregulated after FTY720 administration. Conclusion: The present study shows that blocking Sphk1/S1P/S1PR1 signaling can ameliorate the severity of AIT, providing evidence of a promising therapeutic target for AIT.


Assuntos
Lisofosfolipídeos/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Esfingosina/análogos & derivados , Tireoidite Autoimune/etiologia , Animais , Citocinas/genética , Modelos Animais de Doenças , Feminino , Cloridrato de Fingolimode/farmacologia , Humanos , Janus Quinase 2/fisiologia , Camundongos , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/fisiologia , Esfingosina/farmacologia , Serina-Treonina Quinases TOR/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA