Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 561
Filtrar
1.
Cell Death Dis ; 15(9): 678, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39284838

RESUMEN

SPHK1 (sphingosine kinase type 1) is characterized as a rate-limiting enzyme in sphingolipid metabolism to phosphorylate sphingosine into sphingosine-1-phosphate (S1P) that can bind to S1P receptors (S1PRs) to initiate several signal transductions leading to cell proliferation and survival of normal cell. Many studies have indicated that SPHK1 is involved in several types of cancer development, however, a little is known in bladder cancer. The TCGA database analysis was utilized for analyzing the clinical relevance of SPHK1 in bladder cancer. Through CRISPR/Cas9 knockout (KO) and constitutive activation (CA) strategies on SPHK1 in the bladder cancer cells, we demonstrated the potential downstream target could be programmed cell death 1 ligand 2 (PD-L2). On the other hand, we demonstrated that FDA-approved SPHK1 inhibitor Gilenya® (FTY720) can successfully suppress bladder cancer metastasis by in vitro and in vivo approaches. This finding indicated that SPHK1 as a potent therapeutic target for metastatic bladder cancer by dissecting the mechanism of action, SPHK1/S1P-elicited Akt/ß-catenin activation promoted the induction of PD-L2 that is a downstream effector in facilitating bladder cancer invasion and migration. Notably, PD-L2 interacted with c-Src that further activates FAK. Here, we unveil the clinical relevance of SPHK1 in bladder cancer progression and the driver role in bladder cancer metastasis. Moreover, we demonstrated the inhibitory effect of FDA-approved SPHK1 inhibitor FTY720 on bladder cancer metastasis from both in vitro and in vivo models.


Asunto(s)
Fosfotransferasas (Aceptor de Grupo Alcohol) , Transducción de Señal , Neoplasias de la Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/genética , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/genética , Metástasis de la Neoplasia , Ratones , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Familia-src Quinasas/metabolismo , Movimiento Celular , Ratones Desnudos , Lisofosfolípidos/metabolismo , Proteína Tirosina Quinasa CSK/metabolismo , Clorhidrato de Fingolimod/farmacología , Proliferación Celular
2.
Neuropharmacology ; 261: 110165, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39303855

RESUMEN

Sphingosine-1-phosphate (S1P) receptor (S1PR) agonists, such as fingolimod (FTY720), alleviate nociception in preclinical pain models by either activation (agonism) or inhibition (functional antagonism) of S1PR type-1 (S1PR1). However, the dose-dependence and temporal relationship between reversal of nociception and modulation of S1PR1 signaling has not been systematically investigated. This study examined the relationship between FTY720-induced antinociception and S1PR1 adaptation using a sciatic nerve chronic constriction injury (CCI) model of neuropathic pain in male and female C57Bl/6J mice. Daily injections of FTY720 for 14 days dose-dependently reversed CCI-induced mechanical allodynia without tolerance development, and concomitantly resulted in a dose-dependent reduction of G-protein activation by the S1PR1-selective agonist SEW2871 in the lumbar spinal cord and brain. These findings indicate FTY720-induced desensitization of S1PR1 signaling coincides with its anti-allodynic effects. Consistent with this finding, a single injection of FTY720 reversed mechanical allodynia while concomitantly producing partial desensitization of S1PR1-stimulated G-protein activation in the CNS. However, mechanical allodynia returned 24-hr post injection, despite S1PR1 desensitization at that time, demonstrating a dissociation between these measures. Furthermore, CCI surgery led to elevations of sphingolipid metabolites, including S1P, which were unaffected by daily FTY720 administration, suggesting FTY720 reversed mechanical allodynia by targeting S1PR1 rather than sphingolipid metabolism. Supporting this hypothesis, acute administration of the S1PR1-selective agonist CYM-5442 mimicked the anti-allodynic effect of FTY720. In contrast, the S1PR1-selective antagonist NIBR-0213 prevented the anti-allodynic effect of FTY720, but NIBR-0213 given alone did not affect nociception. These results indicate that FTY720 alleviates CCI-induced allodynia through a mechanism distinct from functional antagonism.


Asunto(s)
Clorhidrato de Fingolimod , Hiperalgesia , Ratones Endogámicos C57BL , Receptores de Esfingosina-1-Fosfato , Animales , Clorhidrato de Fingolimod/farmacología , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Masculino , Femenino , Receptores de Esfingosina-1-Fosfato/agonistas , Receptores de Esfingosina-1-Fosfato/metabolismo , Ratones , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/metabolismo , Modelos Animales de Enfermedad , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Relación Dosis-Respuesta a Droga , Oxadiazoles/farmacología , Neuropatía Ciática/tratamiento farmacológico , Neuropatía Ciática/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Tiofenos
3.
Int J Mol Sci ; 25(17)2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39273153

RESUMEN

BACKGROUND: Schistosomiasis is a parasitic infection that can cause pulmonary hypertension (PH). Th2 CD4 T cells are necessary for experimental Schistosoma-PH. However, if T cells migrate to the lung to initiate, the localized inflammation that drives vascular remodeling and PH is unknown. METHODS: Mice were sensitized to Schistosoma mansoni eggs intraperitoneally and then challenged using tail vein injection. FTY720 was administered, which blocks lymphocyte egress from lymph nodes. T cells were quantified using flow cytometry, PH severity via heart catheterization, and cytokine concentration through ELISA. RESULTS: FTY720 decreased T cells in the peripheral blood, and increased T cells in the mediastinal lymph nodes. However, FTY720 treatment resulted in no change in PH or type 2 inflammation severity in mice sensitized and challenged with S. mansoni eggs, and the number of memory and effector CD4 T cells in the lung parenchyma was also unchanged. Notably, intraperitoneal Schistosoma egg sensitization alone resulted in a significant increase in intravascular lymphocytes and T cells, including memory T cells, although there was no significant change in parenchymal cell density, IL-4 or IL-13 expression, or PH. CONCLUSION: Blocking T cell migration did not suppress PH following Schistosoma egg challenge. Memory CD4 T cells, located in the lung intravascular space following egg sensitization, appear sufficient to cause type 2 inflammation and PH.


Asunto(s)
Hipertensión Pulmonar , Pulmón , Schistosoma mansoni , Animales , Ratones , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/parasitología , Hipertensión Pulmonar/inmunología , Pulmón/parasitología , Pulmón/inmunología , Pulmón/patología , Schistosoma mansoni/inmunología , Clorhidrato de Fingolimod/farmacología , Femenino , Linfocitos T CD4-Positivos/inmunología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/complicaciones , Esquistosomiasis mansoni/patología , Modelos Animales de Enfermedad , Interleucina-4/metabolismo , Citocinas/metabolismo , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Esquistosomiasis/complicaciones , Esquistosomiasis/inmunología , Esquistosomiasis/parasitología
4.
J Integr Neurosci ; 23(8): 142, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39207074

RESUMEN

BACKGROUND: Clinically, ischemic reperfusion injury is the main cause of stroke injury. This study aimed to assess the effectiveness of fingolimod in suppressing inflammation caused by ischemic brain injury and explore its pharmacological mechanisms. METHODS: In total, 75 male Sprague-Dawley rats were randomly and equally assigned to five distinct groups: sham, middle cerebral artery occlusion/reperfusion (MCAO/R) surgery, fingolimod low-dose (F-L), fingolimod medium-dose (F-M), and fingolimod high-dose (F-H). Neurobehavioral tests, 2,3,5-triphenyltetrazolium chloride staining, and the brain tissue drying-wet method were conducted to evaluate neurological impairment, cerebral infarction size, and brain water content. Enzyme-linked immunosorbent assay was employed to quantify pro-inflammatory cytokines interleukin (IL)-1ß, IL-6, and tumor necrosis factor-alpha (TNF-α) protein levels. Western blotting and immunohistochemical staining were performed to assess high mobility group box 1 (HMGB1), toll-like receptor 4 (TLR4), and nuclear factor kappa-B p65 (NF-κBp65) levels. RESULTS: Rats in the F-L, F-M, and F-H groups exhibited lower Longa scores, reduced infarction volumes, and decreased brain edema than those in the MCAO/R group. Additionally, the F-L, F-M, and F-H groups exhibited lower serum levels of IL-1ß, IL-6, and TNF-α than those of the MCAO/R group. Additionally, F-L, F-M, and F-H treatments resulted in decreased HMGB1, TLR4, and NF-κBp65 protein expression levels in the hippocampus of MCAO/R rats. CONCLUSIONS: Fingolimod was found to reduce ischemic brain injury in a dose-dependent manner. Moreover, it was also found to alleviate inflammation following ischemic brain injury via the HMGB1/TLR4/NF­κB signaling pathway.


Asunto(s)
Isquemia Encefálica , Clorhidrato de Fingolimod , Transducción de Señal , Animales , Masculino , Ratas , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/administración & dosificación , Proteína HMGB1/metabolismo , Proteína HMGB1/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/etiología , FN-kappa B/metabolismo , FN-kappa B/efectos de los fármacos , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/efectos de los fármacos
5.
Mult Scler Relat Disord ; 90: 105813, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39154595

RESUMEN

Few data are available regarding vaccine induced SARS-CoV-2 specific T cell responses over time and after booster doses in multiple sclerosis (MS) patients on different disease modifying treatments. We measured SARS-CoV-2 specific CD4+ T cell responses in 72 samples collected from 36 MS patients. The percentage of CD4+ CTVlow CD25+ ICOS+ T cells after stimulation with Spike Recombinant Protein was 29.9 (17.0-43.6) on teriflunomide, 32.4 (11.9-42.5) on ocrelizumab, but much lower (0.6 [0.3-5.9]) on sphingosine-1-phospate receptor modulators (ß = -26.35, p = 0.003). SARS-CoV-2 specific T cells were mainly of Th1 type and stable over time and after booster vaccine doses. mRNA vaccines elicit strong and persistent CD4+ T cell responses against SARS-CoV-2 in MS patients on anti-CD20 and teriflunomide, but not in those on sphingosine-1-phospate receptor modulators.


Asunto(s)
Linfocitos T CD4-Positivos , Vacunas contra la COVID-19 , COVID-19 , Esclerosis Múltiple , Nitrilos , SARS-CoV-2 , Humanos , Masculino , Femenino , Adulto , Vacunas contra la COVID-19/administración & dosificación , Vacunas contra la COVID-19/farmacología , Persona de Mediana Edad , COVID-19/prevención & control , COVID-19/inmunología , SARS-CoV-2/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Linfocitos T CD4-Positivos/inmunología , Nitrilos/farmacología , Toluidinas/farmacología , Toluidinas/uso terapéutico , Crotonatos/farmacología , Crotonatos/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Hidroxibutiratos/farmacología , Vacunas de ARNm , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Factores Inmunológicos/farmacología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
6.
FASEB J ; 38(15): e23872, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39126272

RESUMEN

Paclitaxel is among the most active chemotherapy drugs for the aggressive triple negative breast cancer (TNBC). Unfortunately, it often induces painful peripheral neuropathy (CIPN), a major debilitating side effect. Here we demonstrate that in naive and breast tumor-bearing immunocompetent mice, a clinically relevant dose of FTY720/Fingolimod that targets sphingosine-1-phosphate receptor 1 (S1PR1), alleviated paclitaxel-induced neuropathic pain. FTY720 also significantly attenuated paclitaxel-stimulated glial fibrillary acidic protein (GFAP), a marker for activated astrocytes, and expression of the astrocyte-secreted synaptogenic protein Sparcl1/Hevin, a key regulator of synapse formation. Notably, the formation of excitatory synapses containing VGluT2 in the spinal cord dorsal horn induced by paclitaxel was also inhibited by FTY720 treatment, supporting the involvement of astrocytes and Sparcl1 in CIPN. Furthermore, in this TNBC mouse model that mimics human breast cancer, FTY720 administration also enhanced the anti-tumor effects of paclitaxel, leading to reduced tumor progression and lung metastasis. Taken together, our findings suggest that targeting the S1P/S1PR1 axis with FTY720 is a multipronged approach that holds promise as a therapeutic strategy for alleviating both CIPN and enhancing the efficacy of chemotherapy in TNBC treatment.


Asunto(s)
Clorhidrato de Fingolimod , Neuralgia , Paclitaxel , Animales , Clorhidrato de Fingolimod/farmacología , Paclitaxel/farmacología , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Neuralgia/patología , Ratones , Femenino , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/genética , Línea Celular Tumoral , Receptores de Esfingosina-1-Fosfato/metabolismo , Humanos , Progresión de la Enfermedad , Antineoplásicos Fitogénicos/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteína Ácida Fibrilar de la Glía/genética
7.
Front Immunol ; 15: 1416464, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39076966

RESUMEN

Introduction: Disease-modifying therapies (DMTs) have been shown to improve disease outcomes in multiple sclerosis (MS) patients. They may also impair the immune response to vaccines, including the SARS-CoV-2 vaccine. However, available data on both the intrinsic immune effects of DMTs and their influence on cellular response to the SARS-CoV-2 vaccine are still incomplete. Methods: Here, we evaluated the immune cell effects of 3 DMTs on the response to mRNA SARS-CoV-2 vaccination by comparing MS patients treated with one specific therapy (fingolimod, dimethyl fumarate, or natalizumab) with both healthy controls and untreated patients. We profiled 23 B-cell traits, 57 T-cell traits, and 10 cytokines, both at basal level and after stimulation with a pool of SARS-CoV-2 spike peptides, in 79 MS patients, treated with DMTs or untreated, and 32 healthy controls. Measurements were made before vaccination and at three time points after immunization. Results and Discussion: MS patients treated with fingolimod showed the strongest immune cell dysregulation characterized by a reduction in all measured lymphocyte cell classes; the patients also had increased immune cell activation at baseline, accompanied by reduced specific immune cell response to the SARS-CoV-2 vaccine. Also, anti-spike specific B cells progressively increased over the three time points after vaccination, even when antibodies measured from the same samples instead showed a decline. Our findings demonstrate that repeated booster vaccinations in MS patients are crucial to overcoming the immune cell impairment caused by DMTs and achieving an immune response to the SARS-CoV-2 vaccine comparable to that of healthy controls.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Dimetilfumarato , Clorhidrato de Fingolimod , Esclerosis Múltiple , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/prevención & control , Masculino , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Femenino , SARS-CoV-2/inmunología , Vacunas contra la COVID-19/inmunología , Persona de Mediana Edad , Adulto , Clorhidrato de Fingolimod/uso terapéutico , Clorhidrato de Fingolimod/farmacología , Dimetilfumarato/uso terapéutico , Dimetilfumarato/farmacología , Inmunosupresores/uso terapéutico , Natalizumab/uso terapéutico , Linfocitos B/inmunología , Vacunación , Linfocitos T/inmunología , Citocinas/metabolismo
8.
ACS Appl Mater Interfaces ; 16(30): 38880-38892, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39016239

RESUMEN

Post-translational modification, mitochondrial abruptions, neuroinflammation, and α-synuclein (α-Syn) aggregation are considered as major causes of Parkinson's disease (PD) pathogenesis. The recent literature highlights neuroimmune cross talk and the negative role of immune effector T (Teff) and positive regulation by regulatory T (Treg) cells in PD treatment. Herein, a strategy to endow Treg action paves the path for development of PD treatment. Thus, we explored the neuroprotective efficiency of the immunomodulator and PP2A (protein phosphatase 2) activator, FTY720 nanoparticles in in vivo experimental PD models. Repurposing of FTY720 for PD is known due to its protective effect by reducing PD and its camouflaged role in endowing EZH2-mediated epigenetic regulation of PD. EZH2-FOXP3 interaction is necessary for the neuroprotective Treg cell activity. Therefore, we synthesized FTY720 nanoparticles to improve FTY720 protective efficacy in an in vivo PD model to explore the PP2A mediated signaling. We confirmed the formation of FTY720NPs, and the results of the behavioral and protein expression study showed the significant neuroprotective efficiency of our nanoformulations. In the exploration of neuroprotective mechanism, several lines of evidence confirmed FTY720NPs mediated induction of PP2A/EZH2/FOXP3 signaling in the induction of Treg cells effect in in vivo PD treatment. In summary, our nanoformulations have novel potential to alleviate PD by inducing PP2A-induced epigenetic regulation-mediated neuroimmunomodulation at the clinical setup.


Asunto(s)
Clorhidrato de Fingolimod , Nanopartículas , Fármacos Neuroprotectores , Linfocitos T Reguladores , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Animales , Nanopartículas/química , Ratones , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/química , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/química , Clorhidrato de Fingolimod/uso terapéutico , Ratones Endogámicos C57BL , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Fosfatasa 2/metabolismo , Agentes Inmunomoduladores/farmacología , Agentes Inmunomoduladores/química , Masculino , Factores Inmunológicos/farmacología , Factores Inmunológicos/química , Factores de Transcripción Forkhead/metabolismo , Humanos , Trastornos Parkinsonianos/tratamiento farmacológico
9.
Neurochem Res ; 49(10): 2668-2681, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38918332

RESUMEN

Neuropsychiatric and neurological disorders pose a significant global health burden, highlighting the need for innovative therapeutic approaches. Fingolimod (FTY720), a common drug to treat multiple sclerosis, has shown promising efficacy against various neuropsychiatric and neurological disorders. Fingolimod exerts its neuroprotective effects by targeting multiple cellular and molecular processes, such as apoptosis, oxidative stress, neuroinflammation, and autophagy. By modulating Sphingosine-1-Phosphate Receptor activity, a key regulator of immune cell trafficking and neuronal function, it also affects synaptic activity and strengthens memory formation. In the hippocampus, fingolimod decreases glutamate levels and increases GABA levels, suggesting a potential role in modulating synaptic transmission and neuronal excitability. Taken together, fingolimod has emerged as a promising neuroprotective agent for neuropsychiatric and neurological disorders. Its broad spectrum of cellular and molecular effects, including the modulation of apoptosis, oxidative stress, neuroinflammation, autophagy, and synaptic plasticity, provides a comprehensive therapeutic approach for these debilitating conditions. Further research is warranted to fully elucidate the mechanisms of action of fingolimod and optimize its use in the treatment of neuropsychiatric and neurological disorders.


Asunto(s)
Clorhidrato de Fingolimod , Enfermedades del Sistema Nervioso , Fármacos Neuroprotectores , Clorhidrato de Fingolimod/uso terapéutico , Clorhidrato de Fingolimod/farmacología , Humanos , Animales , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/farmacología , Cognición/efectos de los fármacos , Cognición/fisiología , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/metabolismo , Moduladores de los Receptores de fosfatos y esfingosina 1/uso terapéutico , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Estrés Oxidativo/efectos de los fármacos
10.
Rheumatology (Oxford) ; 63(9): 2578-2589, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38837706

RESUMEN

OBJECTIVES: Increasing studies demonstrated the importance of C5a and anti-neutrophil cytoplasmic antibody (ANCA)-induced neutrophil activation in the pathogenesis of ANCA-associated vasculitis (AAV). Sphingosine-1-phosphate (S1P) acts as a downstream effector molecule of C5a and enhances neutrophil activation induced by C5a and ANCA. The current study investigated the role of a S1P receptor modulator, FTY720, in experimental autoimmune vasculitis (EAV) and explored the immunometabolism-related mechanisms of FTY720 in modulating ANCA-induced neutrophil activation. METHODS: The effects of FTY720 in EAV were evaluated by quantifying haematuria, proteinuria, crescent formation, tubulointerstitial injury and pulmonary haemorrhage. RNA sequencing of renal cortex and gene enrichment analysis were performed. The proteins of key identified pathways were analysed in neutrophils isolated from peripheral blood of patients with active AAV and normal controls. We assessed the effects of FTY720 on ANCA-induced neutrophil respiratory burst and neutrophil extracellular traps formation (NETosis). RESULTS: FTY720 treatment significantly attenuated renal injury and pulmonary haemorrhage in EAV. RNA sequencing analyses of renal cortex demonstrated enhanced fatty acid oxidation (FAO) and peroxisome proliferator-activated receptor (PPAR) signalling in FTY720-treated rats. Compared with normal controls, patients with active AAV showed decreased FAO in neutrophils. FTY720-treated differentiated HL-60 cells showed increased expression of carnitine palmitoyltransferase 1a (CPT1a) and PPARα. Blocking or knockdown of CPT1a or PPARα in isolated human neutrophils and HL-60 cells reversed the inhibitory effects of FTY720 on ANCA-induced neutrophil respiratory burst and NETosis. CONCLUSION: FTY720 attenuated renal injury in EAV through upregulating FAO via the PPARα-CPT1a pathway in neutrophils, offering potential immunometabolic targets in AAV treatment.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos , Ácidos Grasos , Clorhidrato de Fingolimod , Neutrófilos , Oxidación-Reducción , PPAR alfa , Clorhidrato de Fingolimod/farmacología , PPAR alfa/metabolismo , Animales , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/tratamiento farmacológico , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/metabolismo , Neutrófilos/metabolismo , Neutrófilos/efectos de los fármacos , Ratas , Humanos , Ácidos Grasos/metabolismo , Oxidación-Reducción/efectos de los fármacos , Masculino , Peroxidasa/metabolismo , Transducción de Señal/efectos de los fármacos , Modelos Animales de Enfermedad , Activación Neutrófila/efectos de los fármacos , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología
11.
Cancer Res Commun ; 4(7): 1802-1814, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38881188

RESUMEN

Recent progress in single-cell profiling technologies has revealed significant phenotypic and transcriptional heterogeneity in tumor-infiltrating CD8+ T cells. However, the transition between the different states of intratumoral antigen-specific CD8+ T cells remains elusive. Here, we sought to examine the generation, transcriptomic states, and the clinical relevance of melanoma-infiltrating CD8+ T cells expressing a chemokine receptor and T-cell differentiation marker, CX3C chemokine receptor 1 (CX3CR1). Analysis of single-cell datasets revealed distinct human melanoma-infiltrating CD8+ T-cell clusters expressing genes associated with effector T-cell function but with distinguishing expression of CX3CR1 or PDCD1. No obvious impact of CX3CR1 expression in melanoma on the response to immune checkpoint inhibitor therapy was observed while increased pretreatment and on-treatment frequency of a CD8+ T-cell cluster expressing high levels of exhaustion markers was associated with poor response to the treatment. Adoptively transferred antigen-specific CX3CR1- CD8+ T cells differentiated into the CX3CR1+ subset in mice treated with FTY720, which inhibits lymphocyte egress from secondary lymphoid tissues, suggesting the intratumoral generation of CX3CR1+ CD8+ T cells rather than their trafficking from secondary lymphoid organs. Furthermore, analysis of adoptively transferred antigen-specific CD8+ T cells, in which the Cx3cr1 gene was replaced with a marker gene confirmed that CX3CR1+ CD8+ T cells could directly differentiate from the intratumoral CX3CR1- subset. These findings highlight that tumor antigen-specific CX3CR1- CD8+ T cells can fully differentiate outside the secondary lymphoid organs and generate CX3CR1+ CD8+ T cells in the tumor microenvironment, which are distinct from CD8+ T cells that express markers of exhaustion. SIGNIFICANCE: Intratumoral T cells are composed of heterogeneous subpopulations with various phenotypic and transcriptional states. This study illustrates the intratumoral generation of antigen-specific CX3CR1+ CD8+ T cells that exhibit distinct transcriptomic signatures and clinical relevance from CD8+ T cells expressing markers of exhaustion.


Asunto(s)
Linfocitos T CD8-positivos , Receptor 1 de Quimiocinas CX3C , Linfocitos Infiltrantes de Tumor , Melanoma , Transcriptoma , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Melanoma/inmunología , Melanoma/genética , Melanoma/patología , Animales , Humanos , Ratones , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Ratones Endogámicos C57BL , Análisis de la Célula Individual , Relevancia Clínica
12.
Int J Biol Macromol ; 272(Pt 2): 132940, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38848845

RESUMEN

This study reports on the design and synthesis of hypoxia responsive nanoparticles (HRNPs) composed of methoxy polyethylene glycol-4,4 dicarboxylic azolinker-chitosan (mPEG-Azo-chitosan) as ideal drug delivery platform for Fingolimod (FTY720, F) delivery to achieve selective and highly enhanced TNBC therapy in vivo. Herein, HRNPs with an average size of 49.86 nm and a zeta potential of +3.22 mV were synthetized, which after PEG shedding can shift into a more positively-charged NPs (+30.3 mV), possessing self-activation ability under hypoxia situation in vitro, 2D and 3D culture. Treatment with lower doses of HRNPs@F significantly reduced MDA-MB-231 microtumor size to 15 %, induced apoptosis by 88 % within 72 h and reduced highly-proliferative 4 T1 tumor weight by 87.66 % vs. ∼30 % for Fingolimod compared to the untreated controls. To the best of our knowledge, this is the first record for development of hypoxia-responsive chitosan-based NPs with desirable physicochemical properties, and selective self-activation potential to generate highly-charged nanosized tumor-penetrating chitosan NPs. This formulation is capable of localized delivery of Fingolimod to the tumor core, minimizing its side effects while boosting its anti-tumor potential for eradication of TNBC solid tumors.


Asunto(s)
Quitosano , Clorhidrato de Fingolimod , Nanopartículas , Quitosano/química , Quitosano/análogos & derivados , Nanopartículas/química , Humanos , Animales , Línea Celular Tumoral , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/química , Clorhidrato de Fingolimod/administración & dosificación , Ratones , Femenino , Portadores de Fármacos/química , Apoptosis/efectos de los fármacos , Polietilenglicoles/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología
13.
Virology ; 597: 110143, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38917692

RESUMEN

Merkel Cell Carcinoma (MCC) is a rare neuroendocrine skin cancer. In our previous work, we decoded genes specifically deregulated by MCPyV early genes as opposed to other polyomaviruses and established functional importance of NDRG1 in inhibiting cellular proliferation and migration in MCC. In the present work, we found the SET protein, (I2PP2A, intrinsic inhibitor of PP2A) upstream of NDRG1 which was modulated by MCPyV early genes, both in hTERT-HK-MCPyV and MCPyV-positive (+) MCC cell lines. Additionally, MCC dermal tumour nodule tissues showed strong SET expression. Inhibition of the SET-PP2A interaction in hTERT-HK-MCPyV using the small molecule inhibitor, FTY720, increased NDRG1 expression and inhibited cell cycle regulators, cyclinD1 and CDK2. SET inhibition by shRNA and FTY720 also decreased cell proliferation and colony formation in MCPyV(+) MCC cells. Overall, these results pave a path for use of drugs targeting SET protein for the treatment of MCC.


Asunto(s)
Carcinoma de Células de Merkel , Movimiento Celular , Proliferación Celular , Poliomavirus de Células de Merkel , Proteína Fosfatasa 2 , Humanos , Poliomavirus de Células de Merkel/fisiología , Poliomavirus de Células de Merkel/genética , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/genética , Carcinoma de Células de Merkel/virología , Carcinoma de Células de Merkel/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Clorhidrato de Fingolimod/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Línea Celular Tumoral , Chaperonas de Histonas/metabolismo , Chaperonas de Histonas/genética , Infecciones por Polyomavirus/virología , Neoplasias Cutáneas/virología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética
14.
Mult Scler Relat Disord ; 87: 105670, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38772150

RESUMEN

BACKGROUND: The long-term effect of high efficacy disease modifying therapy (DMT) on neurodegeneration in people with multiple sclerosis (pwMS) is largely unknown. The aim of this study was to evaluate the long-term effect of natalizumab (NTZ) or fingolimod (FTY) therapy on the evolution of brain atrophy compared to moderate efficacy DMT in a real-world clinical setting. METHODS: A total of 438 pwMS with 2,439 MRI exams during treatment were analyzed: 252 pwMS treated with moderate efficacy DMT, 130 with NTZ and 56 with FTY. Evolution of brain atrophy was analyzed over an average follow-up of 6.6 years after treatment initiation. Brain segmentation was performed on clinical 3D-FLAIRs using SynthSeg and regional brain volume changes over time were compared between the treatment groups. RESULTS: Total brain, white matter and deep gray matter atrophy rates did not differ between moderate efficacy DMTs, NTZ and FTY. Annualized ventricle growth rates were lower in pwMS treated with NTZ (1.1 %/year) compared with moderate efficacy DMT (2.4 %/year, p < 0.001) and similar to FTY (2.0 %/year, p = 0.051). Cortical atrophy rates were lower in NTZ (-0.08 %/year) compared with moderate efficacy DMT (-0.16 %/year, p = 0.048). CONCLUSION: In a real-world clinical setting, pwMS treated with NTZ had slower ventricular expansion and cortical atrophy compared to those treated with moderate efficacy DMT.


Asunto(s)
Atrofia , Encéfalo , Clorhidrato de Fingolimod , Factores Inmunológicos , Imagen por Resonancia Magnética , Esclerosis Múltiple , Natalizumab , Humanos , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Clorhidrato de Fingolimod/administración & dosificación , Natalizumab/farmacología , Natalizumab/administración & dosificación , Natalizumab/uso terapéutico , Femenino , Masculino , Adulto , Persona de Mediana Edad , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Esclerosis Múltiple/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/patología , Factores Inmunológicos/farmacología , Factores Inmunológicos/administración & dosificación , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/administración & dosificación , Estudios de Seguimiento
15.
J Surg Res ; 300: 25-32, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38795670

RESUMEN

INTRODUCTION: Previous literature suggests that sphingolipids may impact systemic coagulation and platelet aggregation, thus modulating the risks of thrombotic events. The goal of this investigation was to evaluate the role of serum sphingolipids on intrinsic platelet function to assess whether pharmacologic manipulation of sphingolipid metabolites would impact platelet aggregability. METHODS: C57BL/6J mice were injected with either normal saline, 1 mg/kg FTY720 (synthetic sphingosine-1-phosphate [S1P] receptor analog), or 5 mg/kg SLM6031434 (sphingosine kinase two inhibitor). Mice were sacrificed at 6 h and whole blood (WB) was collected for impedance aggregometry assessing platelet responsiveness to arachidonic acid or adenosine diphosphate. Ex vivo studies utilized WB or platelet-rich plasma that was pretreated with S1P, FTY720, amitriptyline, or d-sphingosine then analyzed by aggregability and flow cytometry for platelet and platelet-derived microvesicle characteristics. RESULTS: FTY720 and SLM6031434 pretreated induced similar arachidonic acid and adenosine diphosphate-mediated platelet aggregation as controls. Ex vivo WB and platelet-rich plasma treatment with S1P, FTY720, amitriptyline and d-sphingosine did not impact platelet aggregation. The percentages of CD41+, CD62P+ and CD41+/ceramide+, CD62P+/ceramide + platelets, and platelet-derived microvesicle were not significantly different between amitriptyline-treated and normal saline-treated cohorts. CONCLUSIONS: Sphingolipid modulating agents, such as FTY720, SLM6031434, S1P, amitriptyline, ceramide, and d-sphingosine do not appear to independently impact platelet aggregation in murine models.


Asunto(s)
Plaquetas , Clorhidrato de Fingolimod , Ratones Endogámicos C57BL , Agregación Plaquetaria , Esfingolípidos , Esfingosina , Animales , Agregación Plaquetaria/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Esfingosina/análogos & derivados , Esfingosina/sangre , Ratones , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Esfingolípidos/sangre , Esfingolípidos/metabolismo , Masculino , Lisofosfolípidos/farmacología , Lisofosfolípidos/sangre , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Ácido Araquidónico/farmacología , Amitriptilina/farmacología , Adenosina Difosfato/farmacología
16.
J Neuroimmune Pharmacol ; 19(1): 18, 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38733535

RESUMEN

Suppression of immune functions can be elicited by behavioural conditioning using drugs such as cyclosporin A or rapamycin. Nevertheless, little is known about the underlying mechanisms and generalisability of this phenomenon. Against this background, the present study investigated whether the pharmacological properties of fingolimod (FTY720), an immunosuppressive drug widely applied to treat multiple sclerosis, can be conditioned in rats by means of taste-immune associative learning. For this purpose, a conditioned taste avoidance paradigm was used, pairing the presentation of a novel sweet drinking solution (saccharin or sucrose) as conditioned stimulus (CS) with therapeutically effective doses of FTY720 as unconditioned stimulus (US). Subsequent re-exposure to the CS at a later time point revealed that conditioning with FTY720 induced a mild conditioned taste avoidance only when saccharin was employed as CS. However, on an immunological level, neither re-exposure with saccharin nor sucrose altered blood immune cell subsets or splenic cytokine production. Despite the fact that intraperitonally administered FTY720 could be detected in brain regions known to mediate neuro-immune interactions, the present findings show that the physiological action of FTY720 is not inducible by mere taste-immune associative learning. Whether conditioning generalises across all small-molecule drugs with immunosuppressive properties still needs to be investigated with modified paradigms probably using distinct sensory CS. Moreover, these findings emphasize the need to further investigate the underlying mechanisms of conditioned immunomodulation to assess the generalisability and usability of associative learning protocols as supportive therapies in clinical contexts.


Asunto(s)
Clorhidrato de Fingolimod , Inmunosupresores , Animales , Clorhidrato de Fingolimod/farmacología , Ratas , Inmunosupresores/farmacología , Masculino , Ratas Wistar , Leucocitos/efectos de los fármacos , Reacción de Prevención/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Glicoles de Propileno/farmacología , Gusto/efectos de los fármacos , Sacarina
17.
Int J Nanomedicine ; 19: 4081-4101, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38736654

RESUMEN

Purpose: Spinal cord injury (SCI) is an incurable and disabling event that is accompanied by complex inflammation-related pathological processes, such as the production of excessive reactive oxygen species (ROS) by infiltrating inflammatory immune cells and their release into the extracellular microenvironment, resulting in extensive apoptosis of endogenous neural stem cells. In this study, we noticed the neuroregeneration-promoting effect as well as the ability of the innovative treatment method of FTY720-CDs@GelMA paired with NSCs to increase motor function recovery in a rat spinal cord injury model. Methods: Carbon dots (CDs) and fingolimod (FTY720) were added to a hydrogel created by chemical cross-linking GelMA (FTY720-CDs@GelMA). The basic properties of FTY720-CDs@GelMA hydrogels were investigated using TEM, SEM, XPS, and FTIR. The swelling and degradation rates of FTY720-CDs@GelMA hydrogels were measured, and each group's ability to scavenge reactive oxygen species was investigated. The in vitro biocompatibility of FTY720-CDs@GelMA hydrogels was assessed using neural stem cells. The regeneration of the spinal cord and recovery of motor function in rats were studied following co-treatment of spinal cord injury using FTY720-CDs@GelMA hydrogel in combination with NSCs, utilising rats with spinal cord injuries as a model. Histological and immunofluorescence labelling were used to determine the regeneration of axons and neurons. The recovery of motor function in rats was assessed using the BBB score. Results: The hydrogel boosted neurogenesis and axonal regeneration by eliminating excess ROS and restoring the regenerative environment. The hydrogel efficiently contained brain stem cells and demonstrated strong neuroprotective effects in vivo by lowering endogenous ROS generation and mitigating ROS-mediated oxidative stress. In a follow-up investigation, we discovered that FTY720-CDs@GelMA hydrogel could dramatically boost NSC proliferation while also promoting neuronal regeneration and synaptic formation, hence lowering cavity area. Conclusion: Our findings suggest that the innovative treatment of FTY720-CDs@GelMA paired with NSCs can effectively improve functional recovery in SCI patients, making it a promising therapeutic alternative for SCI.


Asunto(s)
Clorhidrato de Fingolimod , Hidrogeles , Células-Madre Neurales , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal , Animales , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/terapia , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/química , Clorhidrato de Fingolimod/administración & dosificación , Células-Madre Neurales/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Hidrogeles/administración & dosificación , Ratas , Recuperación de la Función/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Puntos Cuánticos/química , Modelos Animales de Enfermedad , Femenino , Médula Espinal/efectos de los fármacos
18.
Bioorg Chem ; 147: 107412, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38696845

RESUMEN

The development of novel topoisomerase I (TOP1) inhibitors is crucial for overcoming the drawbacks and limitations of current TOP1 poisons. Here, we identified two potential TOP1 inhibitors, namely, FTY720 (a sphingosine 1-phosphate antagonist) and COH29 (a ribonucleotide reductase inhibitor), through experimental screening of known active compounds. Biological experiments verified that FTY720 and COH29 were nonintercalative TOP1 catalytic inhibitors that did not induce the formation of DNA-TOP1 covalent complexes. Molecular docking revealed that FTY720 and COH29 interacted favorably with TOP1. Molecular dynamics simulations revealed that FTY720 and COH29 could affect the catalytic domain of TOP1, thus resulting in altered DNA-binding cavity size. The alanine scanning and interaction entropy identified Arg536 as a hotspot residue. In addition, the bioinformatics analysis predicted that FTY720 and COH29 could be effective in treating malignant breast tumors. Biological experiments verified their antitumor activities using MCF-7 breast cancer cells. Their combinatory effects with TOP1 poisons were also investigated. Further, FTY720 and COH29 were found to cause less DNA damage compared with TOP1 poisons. The findings provide reliable lead compounds for the development of novel TOP1 catalytic inhibitors and offer new insights into the potential clinical applications of FTY720 and COH29 in targeting TOP1.


Asunto(s)
Antineoplásicos , ADN-Topoisomerasas de Tipo I , Clorhidrato de Fingolimod , Simulación del Acoplamiento Molecular , Inhibidores de Topoisomerasa I , Humanos , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/química , Clorhidrato de Fingolimod/síntesis química , ADN-Topoisomerasas de Tipo I/metabolismo , ADN-Topoisomerasas de Tipo I/química , Inhibidores de Topoisomerasa I/farmacología , Inhibidores de Topoisomerasa I/química , Inhibidores de Topoisomerasa I/síntesis química , Estructura Molecular , Relación Estructura-Actividad , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Proliferación Celular/efectos de los fármacos , Simulación de Dinámica Molecular , Células MCF-7
19.
Curr Eye Res ; 49(8): 862-871, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38577836

RESUMEN

PURPOSE: FTY720 is an agonist of the Sphingosine-1-phosphate (S1P) receptor 1, 3, 4, and 5 and a functional antagonist of the S1P1 receptor; it can inhibit the activation of mTOR/NF-κB and has therapeutic potential in inflammatory disease. This study was designed to determine the role of the inflammatory process in diabetic retinopathy and investigate the effect of FTY720 on high glucose (HG)-induced rat retinal Müller cells (rMC-1 cells). METHODS: In the present study, the role of FTY720 in inhibiting inflammation and its underlying mechanism were investigated. rMC-1 cells were treated without or with HG, FTY720, CQ, or RAP. Cell viability was examined by CCK-8 assay; cell activation was assessed by western blot analysis and IF staining; and cell migration was evaluated by a scratch wound healing assay. The expression of inflammation-associated proteins and autophagy-related proteins was evaluated by transmission electron microscopy, AO staining, MDC-labeled autophagic vacuoles, western blot analysis and ELISA. RESULTS: Western blot analysis and IF staining showed that the level of the rMC-1 cell marker GFAP was decreased, while GS was increased in FTY720 groups compared to that in the HG group. The healing assay results showed that compared with HG treatment, FTY720 treatment significantly reduced cell migration. Western blot analysis, ELISA and IF staining showed that compared with HG, FTY720 reduced proinflammatory proteins by inhibiting the mechanistic target of the mTOR/NF-κB signaling pathway and regulating autophagy. CONCLUSIONS: This study suggests that in an HG-induced rMC-1 cell model, FTY720 significantly inhibited the production of inflammatory cytokines by inhibiting mTOR/NF-κB signaling and regulating autophagy. These findings were associated with a decrease in rMC-1 cell injury, suggesting that FTY720 or related compounds may be valuable modulators of HG-induced retinal injury.


Asunto(s)
Autofagia , Western Blotting , Movimiento Celular , Supervivencia Celular , Retinopatía Diabética , Células Ependimogliales , Clorhidrato de Fingolimod , FN-kappa B , Transducción de Señal , Serina-Treonina Quinasas TOR , Clorhidrato de Fingolimod/farmacología , Animales , Ratas , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Serina-Treonina Quinasas TOR/metabolismo , FN-kappa B/metabolismo , Autofagia/efectos de los fármacos , Retinopatía Diabética/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Células Cultivadas , Inmunosupresores/farmacología , Microscopía Electrónica de Transmisión , Progresión de la Enfermedad
20.
J Immunol ; 212(11): 1843-1854, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38568091

RESUMEN

Intraepithelial lymphocytes (IELs) are T cells important for the maintenance of barrier integrity in the intestine. Colon IELs are significantly reduced in both MyD88-deficient mice and those lacking an intact microbiota, suggesting that MyD88-mediated detection of bacterial products is important for the recruitment and/or retention of these cells. Here, using conditionally deficient MyD88 mice, we show that myeloid cells are the key mediators of TCRαß+ IEL recruitment to the colon. Upon exposure to luminal bacteria, myeloid cells produce sphingosine-1-phosphate (S1P) in a MyD88-dependent fashion. TCRαß+ IEL recruitment may be blocked using the S1P receptor antagonist FTY720, confirming the importance of S1P in the recruitment of TCRαß+ IELs to the colon epithelium. Finally, using the TNFΔARE/+ model of Crohn's-like bowel inflammation, we show that disruption of colon IEL recruitment through myeloid-specific MyD88 deficiency results in reduced pathology. Our results illustrate one mechanism for recruitment of a subset of IELs to the colon.


Asunto(s)
Colon , Mucosa Intestinal , Linfocitos Intraepiteliales , Lisofosfolípidos , Ratones Noqueados , Células Mieloides , Factor 88 de Diferenciación Mieloide , Receptores de Antígenos de Linfocitos T alfa-beta , Esfingosina , Animales , Lisofosfolípidos/metabolismo , Ratones , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Colon/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Ratones Endogámicos C57BL , Clorhidrato de Fingolimod/farmacología , Enfermedad de Crohn/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA