Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Clin Microbiol Infect ; 24 Suppl 2: S95-S107, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29427804

RESUMO

BACKGROUND: The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) consensus document on the safety of targeted and biological therapies. AIMS: To review, from an infectious diseases perspective, the safety profile of immune checkpoint inhibitors, LFA-3-targeted agents, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors, and to suggest preventive recommendations. SOURCES: Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT: T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death (PD)-1/PD-1 ligand 1 (PD-L1)-targeted agents do not appear to intrinsically increase the risk of infection but can induce immune-related adverse effects requiring additional immunosuppression. Although CD4+ T-cell lymphopenia is associated with alefacept, no opportunistic infections have been observed. Progressive multifocal leukoencephalopathy (PML) may occur during therapy with natalizumab (anti-α4-integrin monoclonal antibody (mAb)) and efalizumab (anti-CD11a mAb), but no cases have been reported to date with vedolizumab (anti-α4ß7 mAb). In patients at high risk for PML (positive anti-JC polyomavirus serology with serum antibody index >1.5 and duration of therapy ≥48 months), the benefit-risk ratio of continuing natalizumab should be carefully considered. Fingolimod induces profound peripheral blood lymphopenia and increases the risk of varicella zoster virus (VZV) infection. Prophylaxis with (val)acyclovir and VZV vaccination should be considered. Proteasome inhibitors also increase the risk of VZV infection, and antiviral prophylaxis with (val)acyclovir is recommended. Anti-Pneumocystis prophylaxis may be considered in myeloma multiple patients with additional risk factors (i.e. high-dose corticosteroids). IMPLICATIONS: Clinicians should be aware of the risk of immune-related adverse effects and PML in patients receiving immune checkpoint and cell adhesion inhibitors respectively.


Assuntos
Terapia Biológica/efeitos adversos , Adesão Celular/efeitos dos fármacos , Doenças Transmissíveis/terapia , Genes cdc/efeitos dos fármacos , Terapia de Alvo Molecular/efeitos adversos , Inibidores de Proteassoma/efeitos adversos , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Terapia Biológica/métodos , Antígeno CTLA-4/antagonistas & inibidores , Ensaios Clínicos como Assunto , Consenso , Humanos , Hospedeiro Imunocomprometido , Leucoencefalopatia Multifocal Progressiva/terapia , Terapia de Alvo Molecular/métodos , Natalizumab/efeitos adversos , Natalizumab/uso terapêutico , Inibidores de Proteassoma/uso terapêutico , Receptores de Lisoesfingolipídeo/efeitos dos fármacos
2.
Mult Scler ; 24(12): 1605-1616, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-28911260

RESUMO

BACKGROUND: Amiselimod, an oral selective sphingosine-1-phosphate 1 receptor modulator, suppressed disease activity dose-dependently without clinically relevant bradyarrhythmia in a 24-week phase 2, placebo-controlled study in relapsing-remitting multiple sclerosis. OBJECTIVE: To assess safety and efficacy of amiselimod over 96 weeks. METHODS: After completing the core study, patients on amiselimod continued at the same dose, whereas those on placebo were randomised 1:1:1 to amiselimod 0.1, 0.2 or 0.4 mg for another 72 weeks. Most patients receiving 0.1 mg were re-randomised to 0.2 or 0.4 mg upon availability of the core study results. RESULTS: Of 415 patients randomised in the core study, 367 (88.4%) entered and 322 (77.6%) completed the extension. One or more adverse events were reported in 303 (82.6%) of 367 patients: 'headache', 'lymphocyte count decreased', 'nasopharyngitis' and 'MS relapse' were most common (14.7%-16.9%). No serious opportunistic infection, macular oedema or malignancy was reported and no bradyarrhythmia of clinical concern was observed by Holter or 12-lead electrocardiogram. The dose-dependent effect of amiselimod on clinical and magnetic resonance imaging-related outcomes from the core study was sustained in those continuing on amiselimod and similarly observed after switching to active drug. CONCLUSION: For up to 2 years of treatment, amiselimod was well tolerated and dose-dependently effective in controlling disease activity.


Assuntos
Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Propanolaminas/administração & dosagem , Propanolaminas/efeitos adversos , Adulto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Tempo , Resultado do Tratamento
3.
Neurosci Lett ; 658: 1-5, 2017 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-28822836

RESUMO

PURPOSE: This study evaluates FTY720/Fingolimod, modulator of sphingosine-1-phosphate (S1P) receptor, as a potential mitigator of radiation-induced neurocognitive dysfunction. METHODS AND MATERIALS: To study radiation-induced neurocognitive deficits, 6 week-old C57/Bl/6J mice received 0 or 7Gy cranial irradiation and were treated with FTY720 or vehicle for seven weeks. Fear conditioning and Morris water maze were then employed to test learning and memory. Immunohistochemical staining for neural progenitor cells (NPCs) and mature neurons was used to assess changes in hippocampal neurogenesis. To test effects on tumor growth, mice harboring brain tumor xenografts were treated with FTY720 or vehicle for six weeks. RESULTS: In irradiated mice, learning deficits were manifested by significantly longer latency times in the Morris Water Maze compared to non-irradiated controls (p=0.001). The deficits were fully restored by FTY720. In irradiated brains, FTY720 maintained the cytoarchitecture of the dentate gyrus granular cell layer and partially restored the pool of NPC. In mice harboring brain tumor stem cell (BTSC) xenografts FTY720 delayed tumor growth and improved survival (p=0.012). CONCLUSIONS: FTY720 mitigates radiation-induced learning dysfunction. A partial restoration of neurogenesis was observed. Furthermore, FTY720 appears to delay tumor growth and improve survival in a xenograft glioma mouse model.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Cloridrato de Fingolimode/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Cognição/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Lesões por Radiação , Radioterapia/efeitos adversos , Receptores de Lisoesfingolipídeo/metabolismo
4.
Pharmacol Ther ; 168: 143-157, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27621206

RESUMO

Sphingosine-1-phosphate (S1P), a simple, bioactive sphingolipid metabolite, plays a key role, both intracellularly and extracellularly, in various cellular processes such as proliferation, survival, migration, inflammation, angiogenesis, and endothelial barrier integrity. The cellular S1P level is low and is tightly regulated by its synthesis and degradation. Sphingosine Kinases (SphKs) 1 and 2, catalyze the ATP-dependent phosphorylation of sphingosine to S1P, while the degradation is mediated by the reversible dephosphorylation catalyzed by the S1P phosphatases and lipid phosphate phosphatases and the irreversible degradation to hexadecenal and ethanolamine phosphate by sphingosine-1-phosphate lyase (S1PL). As a ligand for specific G-protein-coupled receptors, S1P1-5, which are differentially expressed in different cell types, S1P generates downstream signals that play crucial role in developmental and disease related pathologies. In addition to acting extracellularly on receptors located on the plasma membrane, S1P can also act intracellularly, independently of S1P1-5, affecting calcium homeostasis and cell proliferation. The SphKs /S1P /S1PL metabolic pathway is implicated in numerous human pathologies including respiratory disorders, thereby raising the possibility that manipulating intracellular S1P levels could offer therapeutic potential in ameliorating lung diseases. This review focuses on the prospects of targeting S1P signaling and S1P metabolizing enzymes using small molecule inhibitors, receptor agonists, and antagonists in the treatment of lung diseases.


Assuntos
Pneumopatias/tratamento farmacológico , Lisofosfolipídeos/metabolismo , Terapia de Alvo Molecular , Esfingosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Animais , Desenho de Fármacos , Humanos , Ligantes , Pneumopatias/fisiopatologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/metabolismo
5.
Lancet Neurol ; 15(4): 373-81, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26879276

RESUMO

BACKGROUND: Modulation of sphingosine 1-phosphate (S1P) receptors in a non-selective manner decreases disease activity in patients with multiple sclerosis but has potential safety concerns. We assessed the safety and efficacy of the oral selective S1P receptor modulator ozanimod in patients with relapsing multiple sclerosis. METHODS: RADIANCE is a combined phase 2/3 trial. Patients with relapsing multiple sclerosis were recruited from 55 academic and private multiple sclerosis clinics in 13 countries across Europe and the USA. Eligible participants were aged 18-55 years, had an Expanded Disability Status Scale (EDSS) score of 0-5·0, and had either one or more relapses in the previous 12 months, or one or more relapses in the past 24 months and one or more gadolinium-enhancing lesions on MRI in the previous 12 months before screening. Participants were assigned by a computer-generated randomisation sequence in a 1:1:1 ratio to ozanimod (0·5 mg or 1 mg) or matching placebo once daily for 24 weeks by an independent, unmasked, statistical team. Trial participants, study site personnel, MRI assessors, steering committee members, and the study statistician were masked to treatment assignment. To attenuate first-dose cardiac effects, ozanimod was up-titrated from 0·25 mg to 0·5 mg or 1 mg over 8 days. The primary endpoint was the cumulative number of total gadolinium-enhancing MRI lesions measured by an independent MRI analysis centre at weeks 12-24 after treatment initiation. Analysis was by intention to treat. Here, we report results from the 24-week phase 2 trial. This trial is registered with ClinicalTrials.gov, number NCT01628393. The 2-year phase 3 trial is ongoing. FINDINGS: The first patient was randomised on Oct 18, 2012, and the final visit of the last randomised patient was on May 11, 2014. The intention-to-treat and safety population consisted of 258 participants, 88 were assigned placebo, 87 ozanimod 0·5 mg, and 83 ozanimod 1 mg; 252 (98%) patients completed the assigned treatment. The mean cumulative number of gadolinium-enhancing lesions at weeks 12-24 was 11·1 (SD 29·9) with placebo compared with 1·5 (3·7) with ozanimod 0·5 mg (odds ratio 0·16, 95% CI 0·08-0·30; p<0·0001) and 1·5 (3·4) with ozanimod 1 mg (odds ratio 0·11, 95% CI 0·06-0·21; p<0·0001). Three serious adverse events unrelated to treatment were reported in patients assigned ozanimod 0·5 mg: optic neuritis, somatoform autonomic dysfunction, and cervical squamous metaplasia (HPV-related). No serious infectious or cardiac adverse events were reported, and no cases of macular oedema arose. The most common adverse events in the ozanimod 0·5 mg and 1 mg groups compared with placebo were nasopharyngitis (11 and five vs 12), headache (five and three vs eight), and urinary-tract infections (six and two vs two). The maximum reduction in mean heart rate by Holter monitoring during the first 6 h in ozanimod-treated participants was less than 2 beats per min (bpm) compared with baseline, with no patient having a minimum hourly heart rate less than 45 bpm. Electrocardiograms and 24-h Holter monitoring showed no increased incidence of atrioventricular block or sinus pause with ozanimod. INTERPRETATION: Ozanimod significantly reduced MRI lesion activity in participants with relapsing multiple sclerosis, with a favourable safety profile over a period of 24 weeks. These findings warrant phase 3 trials, which are ongoing. FUNDING: Receptos, Inc.


Assuntos
Fatores Imunológicos/farmacologia , Indanos/farmacologia , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Avaliação de Resultados em Cuidados de Saúde , Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Adulto , Feminino , Humanos , Fatores Imunológicos/administração & dosagem , Indanos/administração & dosagem , Masculino , Pessoa de Meia-Idade , Oxidiazóis/administração & dosagem
6.
Chem Biol Interact ; 237: 58-65, 2015 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-25980589

RESUMO

Invasiveness and metastasis are the primary factors indicating poor prognosis in breast cancer patients. To identify a novel lead compound for the development of therapeutics for the treatment of breast cancer through inhibiting invasion, we screened the natural piper amide-like compounds library that we previously constructed. Among the compounds tested, (E)-3-(3,4-dimethoxyphenyl)-N-(4-hydroxyphenethyl)acrylamide (NED-135) showed potent inhibitory effects on matrix metalloproteinase (MMP)-9 and invasiveness of MCF10A human breast epithelial cells treated with an inflammatory lipid, sphingosine-1-phosphate (S1P). The invasive phenotypes of MDA-MB-231 and Hs578T triple-negative breast cancer cells were significantly inhibited by NED-135. NED-135 efficiently inhibited the S1P-induced MMP-9 expression at the transcriptional level with a comparable degree to FTY720, a known antagonist of S1P. We further showed that NED-135 significantly inhibited activation of S1P-induced signaling molecules, Akt, ERKs, and p38 MAPK. Computational similarity analysis led us to postulate that NED-135 and FTY720 may exert anti-invasive effects on breast cells possibly via different mechanisms. Due to its novel structural and functional features, we suggest that NED-135 can be used as a novel lead compound against breast cancer in an inflammatory microenvironment and highly invasive triple-negative breast cancer.


Assuntos
Acrilamidas/farmacologia , Invasividade Neoplásica , Fenetilaminas/farmacologia , Neoplasias de Mama Triplo Negativas/patologia , Acrilamidas/química , Linhagem Celular Tumoral , Feminino , Humanos , Lisofosfolipídeos/metabolismo , Fenetilaminas/química , Proteínas Quinases/metabolismo , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Neoplasias de Mama Triplo Negativas/enzimologia , Neoplasias de Mama Triplo Negativas/metabolismo
7.
Hepatol Int ; 9(1): 149-54, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25788389

RESUMO

PURPOSE: To determine whether FTY720, a sphingosine-1-phosphate (S1P) receptor modulator, would protect sinusoid endothelial cells (SECs) from radiation injury in vitro. MATERIALS AND METHODS: The effect of FTY720 on the viability of irradiated human liver SECs were examined by MTT assay or FACS analysis. The effect of FTY720 on the survival of hepatocellular carcinoma cell line, HepG2 and McA-RH7777, were determined by clonogenic assays. The activation of Akt pathway was tested by western bolt. RESULTS: FTY720 increases the survival of irradiated SECs; in contrast, it does not appear to be radioprotective of tumor cells. Furthermore, the activation of Akt pathway was confirmed in the protective effect of FTY720 on SECs. CONCLUSION: These results suggest that FTY720 will be a potential therapeutic protector for the SEC apoptosis during RILD.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/efeitos da radiação , Cloridrato de Fingolimode/farmacologia , Fígado/efeitos da radiação , Lesões por Radiação/prevenção & controle , Protetores contra Radiação/farmacologia , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Células Endoteliais/patologia , Células Hep G2 , Humanos , Fígado/citologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tolerância a Radiação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
Biochim Biophys Acta ; 1851(5): 519-26, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25601519

RESUMO

Transforming growth factor ß2 (TGF-ß2) is well known to stimulate the expression of pro-fibrotic connective tissue growth factor (CTGF) in several cell types including human mesangial cells. The present study demonstrates that TGF-ß2 enhances sphingosine 1-phosphate receptor 5 (S1P5) mRNA and protein expression in a time and concentration dependent manner. Pharmacological and siRNA approaches reveal that this upregulation is mediated via activation of classical TGF-ß downstream effectors, Smad and mitogen-activated protein kinases. Most notably, inhibition of Gi with pertussis toxin and downregulation of S1P5 by siRNA block TGF-ß2-stimulated upregulation of CTGF, demonstrating that Gi coupled S1P5 is necessary for TGF-ß2-triggered expression of CTGF in human mesangial cells. Overall, these findings indicate that TGF-ß2 dependent upregulation of S1P5 is required for the induction of pro-fibrotic CTGF by TGF-ß. Targeting S1P5 might be an attractive novel approach to treat renal fibrotic diseases.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Células Mesangiais/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Fator de Crescimento Transformador beta2/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Humanos , Células Mesangiais/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fatores de Tempo , Transfecção , Regulação para Cima
9.
Acta Pharmacol Sin ; 35(12): 1537-45, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25399649

RESUMO

AIM: FTY720, a new immunomodulatory drug with low cytotoxicity, is currently used to treat multiple sclerosis. In this study, we investigated the effects of FTY720 on inflammatory cell infiltration in albumin overload-induced nephropathy of rats. METHODS: Male Wistar rats were subjected to right-side nephrectomy and divided into 3 groups. One week after the surgery, albumin overload (AO) group was treated with BSA (5 g·kg(-1)·d(-1), ip) for 9 weeks; AO+FTY720 group was given BSA (5 g·kg(-1)·d(-1), ip) plus FTY720 (0.5 g·kg(-1)·d(-1), ip) for 9 weeks; and control group received daily ip injection of equivalent volume of saline. All rats were killed 9 weeks after nephrectomy. RESULTS: AO rats exhibited gradually increased urinary protein excretion accompanied by elevated urinary N-acetyl-ß-O-glucosaminidase activity, and both reached their peak values at week 7. Furthermore, AO significantly increased lymphocytes and monocytes in circulation and the inflammatory cells recruited to tubulointerstitium, and the expression of inflammatory cytokines MCP-1, TNF-α and IL-6, as well as sphingosine 1-phosphate (S1P) receptors S1pr1 and S1pr3, and S1P-synthesizing enzyme sphingosine kinase 1 (Sphk1) in the kidney. Concomitant administration of FTY720 significantly attenuated all the AO-induced pathological changes. CONCLUSION: FTY720 alleviates tubulointerstitium inflammation in an AO rat model of nephropathy via down-regulation of the Sphk1 pathway.


Assuntos
Albuminúria/tratamento farmacológico , Anti-Inflamatórios/farmacologia , Imunossupressores/farmacologia , Túbulos Renais/efeitos dos fármacos , Nefrite Intersticial/prevenção & controle , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Acetilglucosaminidase/urina , Albuminúria/enzimologia , Albuminúria/patologia , Albuminúria/urina , Animais , Modelos Animais de Doenças , Regulação para Baixo , Cloridrato de Fingolimode , Mediadores da Inflamação/metabolismo , Túbulos Renais/enzimologia , Túbulos Renais/patologia , Linfócitos/efeitos dos fármacos , Linfócitos/enzimologia , Lisofosfolipídeos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Masculino , Nefrite Intersticial/enzimologia , Nefrite Intersticial/patologia , Nefrite Intersticial/urina , Ratos Wistar , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/metabolismo , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Fatores de Tempo
10.
Rev Neurol ; 59(3): 121-8, 2014 Aug 01.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-25030072

RESUMO

INTRODUCTION: Fingolimod, a sphingosine-1-phosphate receptor modulator, was the first oral therapy approved for relapsing-remitting multiple sclerosis, and shows a novel mechanism of action. Upon binding to S1P1 receptors in lymphocytes, the selective retention of naive and central memory T cells in secondary lymphoid tissues is promoted, preventing their egress to the central nervous system (CNS). In addition, fingolimod readily crosses the blood brain barrier, and several reports suggest a direct neuroprotective effect in the CNS. AIM: To review the available data on the central effects of fingolimod. DEVELOPMENT: Imbalances between damage and repair processes are a reflection of chronic demyelination, axonal degeneration and gliosis, and seem to contribute to multiple sclerosis associated disability. Given fingolimod readily crosses the blood brain barrier, it can exert its action directly on S1P receptors present in CNS cells. Fingolimod occupies S1P receptors in oligodendrocytes, oligodendrocyte precursor cells, astrocytes, microglial cells and neurons, promoting remyelination, neuroprotection, and endogenous regeneration processes. Efficacy results from clinical trials are consistent with a mechanism of action that includes direct effects in CNS cells. CONCLUSIONS: Current evidence suggests that the efficacy of fingolimod in the treatment of Multiple Sclerosis is due to its dual action as an immunomodulatory molecule and as a direct modulator of S1PRs in the CNS. In fact, recent reports propose that fingolimod has neuroprotective effects in several models, and open new avenues of potential therapeutic applications, such as Alzheimer's disease, cerebral malaria, neuroblastoma and neuroprotection in cranial irradiation.


TITLE: Efectos del fingolimod en el sistema nervioso central.Introduccion. El fingolimod, un modulador del receptor de la esfingosina-1-fosfato (S1P) dotado de un mecanismo de accion novedoso, fue el primer tratamiento oral aprobado para la esclerosis multiple remitente recurrente. Su union a los receptores S1P1 de los linfocitos promueve la retencion selectiva de los linfocitos T virgenes y de memoria central en los tejidos linfoides secundarios, lo que impide su salida hacia el sistema nervioso central (SNC). Asimismo, el fingolimod atraviesa con facilidad la barrera hematoencefalica, y diversos estudios le atribuyen un efecto neuroprotector directo en el SNC. Objetivo. Revisar la informacion disponible acerca de los efectos centrales del fingolimod. Desarrollo. El desequilibrio entre los procesos lesivos y reparadores constituye un reflejo de la desmielinizacion cronica, la degeneracion axonal y la gliosis, y parece contribuir a la discapacidad que la esclerosis multiple acarrea. La facilidad con la que el fingolimod atraviesa la barrera hematoencefalica le permite actuar directamente sobre los receptores S1P localizados en las celulas del SNC. Una vez en el interior del SNC, ocupa los receptores S1P de los oligodendrocitos y de sus celulas precursoras, de los astrocitos, los microgliocitos y las neuronas, fomentando la remielinizacion, la neuroproteccion y los procesos endogenos de regeneracion. La eficacia evidenciada en los ensayos clinicos concuerda con un mecanismo de accion que incluiria efectos directos sobre las celulas del SNC. Conclusiones. Los datos disponibles indican que la eficacia del fingolimod en el tratamiento de la esclerosis multiple se debe a su ambivalencia como molecula inmunomoduladora y moduladora directa de los receptores S1P del SNC. Tanto es asi que estudios recientes le atribuyen efectos neuroprotectores en varios modelos que suscitan expectativas en torno a su posible aplicacion terapeutica en la enfermedad de Alzheimer, el paludismo cerebral y el neuroblastoma, asi como en la neuroproteccion frente a la radioterapia craneal.


Assuntos
Fármacos Neuroprotetores/farmacologia , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Animais , Barreira Hematoencefálica , Quimiotaxia de Leucócito/efeitos dos fármacos , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Cloridrato de Fingolimode , Humanos , Memória Imunológica , Camundongos , Camundongos Knockout , Camundongos Mutantes Neurológicos , Estudos Multicêntricos como Assunto , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Neurogênese/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacocinética , Propilenoglicóis/farmacocinética , Propilenoglicóis/uso terapêutico , Ratos , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Esfingosina/farmacocinética , Esfingosina/farmacologia , Esfingosina/uso terapêutico , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia
11.
Am J Physiol Cell Physiol ; 307(1): C14-24, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24740542

RESUMO

Understanding vascular growth and maturation in developing tumors has important implications for tumor progression, spread, and ultimately host survival. Modulating the signaling of endothelial G protein-coupled receptors (GPCRs) in blood and lymphatic vessels can enhance or limit tumor progression. Sphingosine 1-phosphate receptor 1 (S1PR1) is a GPCR for circulating lysophospholipid S1P that is highly expressed in blood and lymphatic vessels. Using the S1PR1- enhanced green fluorescent protein (eGFP) mouse model in combination with intravital imaging and pharmacologic modulation of S1PR1 signaling, we show that boundary conditions of high and low S1PR1 signaling retard tumor progression by enhancing or destabilizing neovasculature integrity, respectively. In contrast, midrange S1PR1 signaling, achieved by receptor antagonist titration, promotes abundant growth of small, organized vessels and thereby enhances tumor progression. Furthermore, in vivo S1PR1 antagonism supports lung colonization by circulating tumor cells. Regulation of endothelial S1PR1 dynamically controls vascular integrity and maturation and thus modulates angiogenesis, tumor growth, and hematogenous metastasis.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Permeabilidade Capilar , Células Endoteliais/metabolismo , Neovascularização Patológica , Receptores de Lisoesfingolipídeo/metabolismo , Anilidas/farmacologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Permeabilidade Capilar/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia , Organofosfonatos/farmacologia , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato , Fatores de Tempo , Carga Tumoral
12.
Coron Artery Dis ; 25(4): 311-20, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24448174

RESUMO

OBJECTIVES: Natural killer T (NKT) cells are unique T lymphocytes that recognize glycolipid antigen and produce various cytokines. NKT cells accelerate atherosclerosis in mice. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid and regulates T-lymphocyte trafficking. We aimed to determine the effects of S1P on the production of proinflammatory cytokine, tumor necrosis factor (TNF)-α, in NKT cell hybridomas and mouse NKT cells. MATERIALS AND METHODS: NKT cell hybridomas and sorted mouse NKT cells were stimulated with S1P and α-galactosylceramide (α-GalCer), the major ligand to produce cytokines in NKT cells. TNF-α mRNA expression and protein production were determined by real-time PCR and ELISA, respectively. Cell migration was assayed using chemotaxicell. Plasma S1P was measured using HPLC. RESULTS: Hybridomas expressed S1P receptors, S1P1, S1P2, and S1P4. S1P and α-GalCer increased TNF-α mRNA expression and protein production. S1P enhanced TNF-α induction by α-GalCer. S1P receptor antagonists decreased the TNF-α mRNA expression induced by S1P. FTY720, an immunosuppressive S1P receptor modulator, also decreased the TNF-α mRNA expression. The migration of NKT cell hybridomas was increased by S1P. FTY720 reduced the migration induced by S1P. S1P also increased the TNF-α mRNA expression in mouse NKT cells. Plasma TNF-α levels in patients with high plasma S1P (≥500 nmol/l) were higher than those in patients with low S1P (<500 nmol/l). CONCLUSION: S1P binds to S1P receptors in NKT cells and enhances TNF-α production. TNF-α overproduction may induce atherogenic inflammatory responses. S1P may serve as a novel therapeutic target for amelioration of vascular inflammatory diseases.


Assuntos
Aterosclerose/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Lisofosfolipídeos/metabolismo , Células T Matadoras Naturais/metabolismo , Esfingosina/análogos & derivados , Animais , Aterosclerose/imunologia , Linhagem Celular Tumoral , Quimiotaxia de Leucócito , Técnicas de Cocultura , Feminino , Humanos , Hibridomas , Fatores Imunológicos/farmacologia , Inflamação/imunologia , Ativação Linfocitária , Camundongos Endogâmicos C57BL , Células T Matadoras Naturais/efeitos dos fármacos , Células T Matadoras Naturais/imunologia , Inibidor 1 de Ativador de Plasminogênio/sangue , Ratos , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Esfingosina/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
13.
Eur J Clin Pharmacol ; 70(3): 287-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24362488

RESUMO

PURPOSE: To determine the effects of steady-state concentrations of the selective S1P1 receptor modulator ponesimod on the pharmacokinetics (PK) of a single dose of a combined oral contraceptive, containing 1 mg norethisterone (NET) and 35 µg ethinyl estradiol (EE) and to investigate the effects on heart rate at different ponesimod doses within an up-titration regimen prior to co-administration of the contraceptive. METHODS: Twenty-two healthy women (age: 29-60 years) received twice a single oral dose of the combined oral contraceptive, alone or in combination with multiple doses of 40 mg ponesimod attained by an up-titration regimen. Heart rate (HR) effects were assessed on the first day of each up-titration level. PK parameters of NET and EE were determined by non-compartmental analysis. RESULTS: Geometric mean ratios (ponesimod and contraceptive / contraceptive alone) of Cmax and AUC0-24 of NET were 0.87 (90 % CI: 0.80, 0.94) and 0.84 (90 % CI: 0.76, 0.93), respectively. Geometric mean ratios of Cmax and AUC0-24 of EE were 0.94 (90 % CI: 0.86, 1.03) and 0.95 (90 % CI: 0.89, 1.01), respectively. The maximum mean HR reduction after the first dose of 10 mg ponesimod was 12.4 bpm (SD ± 6.2) at 2.5 h post-dose. On Day 4 (first dose of 20 mg) and Day 7 (first dose of 40 mg) the maximum mean HR reduction was 4.3 bpm (SD ± 5.7) and 1.4 (SD ± 6.4), respectively, at 2.5 h post-dose compared to baseline. CONCLUSION: No clinically relevant PK interactions between ponesimod and the combined oral contraceptive were observed, therefore, efficacy of hormonal contraceptives is not expected to be affected by concomitant administration of ponesimod. The up-titration regimen showed that HR reductions are diminished upon repeated ponesimod administration.


Assuntos
Anticoncepcionais Orais Combinados/farmacocinética , Etinilestradiol/farmacocinética , Noretindrona/farmacocinética , Tiazóis/farmacologia , Adulto , Área Sob a Curva , Anticoncepcionais Orais Combinados/administração & dosagem , Estudos Cross-Over , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Interações Medicamentosas , Etinilestradiol/administração & dosagem , Feminino , Frequência Cardíaca/efeitos dos fármacos , Humanos , Pessoa de Meia-Idade , Noretindrona/administração & dosagem , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/metabolismo , Tiazóis/administração & dosagem , Fatores de Tempo
14.
Handb Exp Pharmacol ; (215): 239-53, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23579459

RESUMO

The recent success of FTY720 (Fingolimod, Gilenya(®)), which has been approved for the treatment of relapsing-remitting multiple sclerosis and is the first-in-class sphingosine-1-phosphate (S1P) receptor modulating drug, has boosted the interest in further drug development in this area. Several selective S1P1 receptor-modulating drugs are being investigated in clinical trials for the treatment of diverse autoimmune disorders. Sphingosine kinase inhibitors are under development for the treatment of cancer, aberrant angiogenesis and inflammatory diseases; an inhibitor of SK2 with relatively low affinity is being analysed in patients with advanced solid tumours. While an indirect S1P lyase inhibitor has just failed the proof of concept in patients with rheumatoid arthritis, S1P lyase is still a promising target for the treatment of inflammatory and autoimmune diseases. Another approach is the development of S1P-scavenging or -clearing agents, including a monoclonal S1P antibody that has successfully passed phase I clinical trials and will be further developed for age-related macular degeneration.


Assuntos
Lisofosfolipídeos/fisiologia , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados , Aldeído Liases/antagonistas & inibidores , Animais , Inibidores Enzimáticos/farmacologia , Cloridrato de Fingolimode , Humanos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Propilenoglicóis/uso terapêutico , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Esfingosina/fisiologia , Esfingosina/uso terapêutico
15.
Med Sci (Paris) ; 28(11): 951-7, 2012 Nov.
Artigo em Francês | MEDLINE | ID: mdl-23171898

RESUMO

Sphingosine 1-phosphate (S1P) mediates critical physiological responses by its binding to G protein-coupled receptor (GPCR) subtypes, known as S1P receptors. Five distinct mammalian S1P receptors, designated S1P1-5 have been identified, each with a different cellular pattern of expression which influences the responses to S1P. In this review, we briefly outline our understanding of the modes of action and the roles of S1P receptors in the regulation of physiological and pathological functions in the cardiovascular, immune and central nervous system.


Assuntos
Lisofosfolipídeos/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Animais , Aterosclerose/fisiopatologia , Sistema Cardiovascular/fisiopatologia , Movimento Celular , Humanos , Linfócitos/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Mamíferos , Camundongos , Camundongos Knockout , Neoplasias/irrigação sanguínea , Neoplasias/imunologia , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurogênese/fisiologia , Receptores de Lisoesfingolipídeo/classificação , Receptores de Lisoesfingolipídeo/deficiência , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Sistemas do Segundo Mensageiro/fisiologia , Esfingosina/fisiologia
16.
Neuro Oncol ; 14(4): 405-15, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22351749

RESUMO

FTY720 is a sphingosine analogue that down regulates expression of sphingosine-1-phosphate receptors and causes apoptosis of multiple tumor cell types, including glioma cells. This study examined the effect of FTY720 on brain tumor stem cells (BTSCs) derived from human glioblastoma (GBM) tissue. FTY720 treatment of BTSCs led to rapid inactivation of ERK MAP kinase, leading to upregulation of the BH3-only protein Bim and apoptosis. In combination with temozolomide (TMZ), the current standard chemotherapeutic agent for GBM, FTY720 synergistically induced BTSC apoptosis. FTY720 also slowed growth of intracranial xenograft tumors in nude mice and augmented the therapeutic effect of TMZ, leading to enhanced survival. Furthermore, the combination of FTY720 and TMZ decreased the invasiveness of BTSCs in mouse brains. FTY720 is known to cross the blood-brain barrier and recently received Food and Drug Administration approval for treatment of relapsing multiple sclerosis. Thus, FTY720 is an excellent potential therapeutic agent for treatment of GBM.


Assuntos
Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Propilenoglicóis/farmacologia , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Esfingosina/análogos & derivados , Animais , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Cloridrato de Fingolimode , Glioblastoma/tratamento farmacológico , Humanos , Quimioterapia de Indução , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Propilenoglicóis/uso terapêutico , Proteínas Proto-Oncogênicas/metabolismo , Esfingosina/farmacologia , Esfingosina/uso terapêutico , Temozolomida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Pathol Res Pract ; 208(2): 82-8, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22244964

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive sophospholipid with various S1P receptor (S1PR) expression profiles in cells of different origin. S1PR1, R3 and - to a lesser extent - R2 were the main receptors expressed in most of endothelial cells (ECs). The balances in the expression and activation of S1PR1, R2 and R3 help to maintain the physiological functions of ECs. Reverse transcription-PCR and Western blotting were used to detect the mRNA transcript level and protein expression of S1PR. Endothelial barrier function was measured by transflux of tracer protein through endothelial monolayer. Human dermal microvascular ECs predominantly expressed S1PR1 and S1PR3. Lipopolysaccharide (LPS) or tumor necrosis factor-α (TNF-α) significantly upregulated S1PR2 mRNA and protein levels. The application of S1PR2 antagonist JTE-013 decreased the endothelial monolayer hyper-permeability response induced by LPS and TNF-α. Inflammatory mediators LPS and TNF-α induce S1PR2 expression in endothelium, suggesting that S1PR2 up-regulation may be involved in LPS and TNF-α elicited endothelial barrier dysfunction.


Assuntos
Derme/irrigação sanguínea , Células Endoteliais/efeitos dos fármacos , Mediadores da Inflamação/farmacologia , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Microvasos/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Western Blotting , Permeabilidade Capilar , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Humanos , Inflamação/genética , Microvasos/metabolismo , Pirazóis/farmacologia , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores de Esfingosina-1-Fosfato , Fatores de Tempo , Regulação para Cima
18.
Neurology ; 76(8 Suppl 3): S15-9, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21339486

RESUMO

Viral infections may have an important role in the precipitation or relapse of multiple sclerosis (MS) and its treatment. This review describes the normal immune response to viral infection, the possible associations between viral infections and MS therapy, and the impact of sphingosine 1-phosphate (S1P) receptor (S1PR) modulation with fingolimod (FTY720) on the immune responses to viral infection. The physiologic immune response to viral infection involves lymphocyte activation and control of the circulation of subsets of lymphocytes with different functions between the lymph nodes, vascular system, and tissues, under the control of the S1P/S1PR signaling mechanism. In MS, it has been postulated that viral infections may play a role in triggering MS relapses, with virus-specific T cells being responsible for the demyelinating lesions within the CNS. Fingolimod-an S1PR modulator approved for the treatment of relapsing MS in some countries-is thought to act by downmodulating lymphatic S1P subtype 1 receptors. This retains naïve T cells and central memory T cells, but not effector memory T cells, within the lymph nodes and prevents their circulation to the CNS. Evidence from infection models supports that the selective effects of fingolimod on T cell subsets allows key immune responses to be preserved during therapy. However, in patients, long-term observation is important as both the risk of cancer and infection is potentially increased by the use of any immunomodulatory agent.


Assuntos
Sistema Imunitário/fisiologia , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Animais , Cloridrato de Fingolimode , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Sistema Imunitário/efeitos dos fármacos , Imunossupressores/farmacologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Lisofosfolipídeos/genética , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Propilenoglicóis/farmacologia , Propilenoglicóis/uso terapêutico , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Esfingosina/genética , Esfingosina/metabolismo , Esfingosina/farmacologia , Esfingosina/uso terapêutico , Viroses/tratamento farmacológico , Viroses/imunologia , Viroses/patologia
19.
J Neuroimmunol ; 230(1-2): 10-6, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20826007

RESUMO

Human microglia, monocyte-derived dendritic cells (DCs) and macrophages ex vivo express relatively higher levels of sphingosine-1-phosphate (S1P) receptor 1 (S1P1) mRNA as compared to other receptor subtypes. The S1P agonist FTY720 decreased ERK phosphorylation and induced myosin light chain (MLC) II phosphorylation only in macrophages and DCs. FTY720 inhibited IL-12p70 production (CD40L induced) by DCs and macrophages but not microglia (poly I:C induced). IL-10 production was increased in DCs and unaffected in other myeloid cells. Despite similar receptor expression patterns, the distinct myeloid cell populations present in the human CNS, under steady-state or inflammatory conditions, exhibit differential responses to FTY720.


Assuntos
Imunossupressores/farmacologia , Microglia/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Western Blotting , Citocinas/biossíntese , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Cloridrato de Fingolimode , Expressão Gênica , Humanos , Lisofosfolipídeos/imunologia , Lisofosfolipídeos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Microglia/imunologia , Microglia/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , RNA Mensageiro/análise , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/imunologia , Receptores de Lisoesfingolipídeo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esfingosina/imunologia , Esfingosina/metabolismo , Esfingosina/farmacologia
20.
Arterioscler Thromb Vasc Biol ; 30(9): 1779-86, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20702813

RESUMO

OBJECTIVE: The goals of this study were to identify the signaling pathway by which sphingosine 1-phosphate (S1P) activates RhoA in smooth muscle cells (SMC) and to evaluate the contribution of this pathway to the regulation of SMC phenotype. METHODS AND RESULTS: Using a combination of receptor-specific agonists and antagonists we identified S1P receptor 2 (S1PR2) as the major S1P receptor subtype that regulates SMC differentiation marker gene expression. Based on the known coupling properties of S1PR2 and our demonstration that overexpression of Galpha(12) or Galpha(13) increased SMC-specific promoter activity, we next tested whether the effects of S1P in SMC were mediated by the regulator of G protein-signaling-Rho guanine exchange factors (RGS-RhoGEFs) (leukemia-associated RhoGEF [LARG], PDZ-RhoGEF [PRG], RhoGEF [p115]). Although each of the RGS-RhoGEFs enhanced actin polymerization, myocardin-related transcription factor-A nuclear localization, and SMC-specific promoter activity when overexpressed in 10T1/2 cells, LARG exhibited the most robust effect and was the only RGS-RhoGEF activated by S1P in SMC. Importantly, siRNA-mediated depletion of LARG significantly inhibited the activation of RhoA and SMC differentiation marker gene expression by S1P. Knockdown of LARG had no effect on SMC proliferation but promoted SMC migration as measured by scratch wound and transwell assays. CONCLUSIONS: These data indicate that S1PR2-dependent activation of RhoA in SMC is mediated by LARG and that this signaling mechanism promotes the differentiated SMC phenotype.


Assuntos
Diferenciação Celular , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Lisofosfolipídeos/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Movimento Celular , Ativação Enzimática , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Camundongos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Fenótipo , Interferência de RNA , RNA Mensageiro/metabolismo , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais/efeitos dos fármacos , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Fatores de Tempo , Transcrição Gênica , Transfecção , Proteína rhoA de Ligação ao GTP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA