Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 214
Filtrar
1.
BMC Complement Med Ther ; 24(1): 214, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38840248

RESUMEN

BACKGROUND: Traditional Chinese medicine (TCM) has been found widespread application in neoplasm treatment, yielding promising therapeutic candidates. Previous studies have revealed the anti-cancer properties of Brevilin A, a naturally occurring sesquiterpene lactone derived from Centipeda minima (L.) A.Br. (C. minima), a TCM herb, specifically against lung cancer. However, the underlying mechanisms of its effects remain elusive. This study employs network pharmacology and experimental analyses to unravel the molecular mechanisms of Brevilin A in lung cancer. METHODS: The Batman-TCM, Swiss Target Prediction, Pharmmapper, SuperPred, and BindingDB databases were screened to identify Brevilin A targets. Lung cancer-related targets were sourced from GEO, Genecards, OMIM, TTD, and Drugbank databases. Utilizing Cytoscape software, a protein-protein interaction (PPI) network was established. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene set enrichment analysis (GSEA), and gene-pathway correlation analysis were conducted using R software. To validate network pharmacology results, molecular docking, molecular dynamics simulations, and in vitro experiments were performed. RESULTS: We identified 599 Brevilin A-associated targets and 3864 lung cancer-related targets, with 155 overlapping genes considered as candidate targets for Brevilin A against lung cancer. The PPI network highlighted STAT3, TNF, HIF1A, PTEN, ESR1, and MTOR as potential therapeutic targets. GO and KEGG analyses revealed 2893 enriched GO terms and 157 enriched KEGG pathways, including the PI3K-Akt signaling pathway, FoxO signaling pathway, and HIF-1 signaling pathway. GSEA demonstrated a close association between hub genes and lung cancer. Gene-pathway correlation analysis indicated significant associations between hub genes and the cellular response to hypoxia pathway. Molecular docking and dynamics simulations confirmed Brevilin A's interaction with PTEN and HIF1A, respectively. In vitro experiments demonstrated Brevilin A-induced dose- and time-dependent cell death in A549 cells. Notably, Brevilin A treatment significantly reduced HIF-1α mRNA expression while increasing PTEN mRNA levels. CONCLUSIONS: This study demonstrates that Brevilin A exerts anti-cancer effects in treating lung cancer through a multi-target and multi-pathway manner, with the HIF pathway potentially being involved. These results lay a theoretical foundation for the prospective clinical application of Brevilin A.


Asunto(s)
Neoplasias Pulmonares , Simulación del Acoplamiento Molecular , Sesquiterpenos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Sesquiterpenos/farmacología , Sesquiterpenos/química , Lactonas/farmacología , Lactonas/química , Células A549 , Mapas de Interacción de Proteínas , Farmacología en Red , Crotonatos
2.
Sci Rep ; 14(1): 10295, 2024 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704415

RESUMEN

Lysine crotonylation (Kcr) is a recently discovered histone acylation modification that is closely associated with gene expression, cell proliferation, and the maintenance of stem cell pluripotency and indicates the transcriptional activity of genes and the regulation of various biological processes. During cell culture, the introduction of exogenous croconic acid disodium salt (Nacr) has been shown to modulate intracellular Kcr levels. Although research on Kcr has increased, its role in cell growth and proliferation and its potential regulatory mechanisms remain unclear compared to those of histone methylation and acetylation. Our investigation demonstrated that the addition of 5 mM Nacr to cultured bovine fibroblasts increased the expression of genes associated with Kcr modification, ultimately promoting cell growth and stimulating cell proliferation. Somatic cell nuclear transfer of donor cells cultured in 5 mM Nacr resulted in 38.1% blastocyst development, which was significantly greater than that in the control group (25.2%). This research is important for elucidating the crotonylation modification mechanism in fibroblast proliferation to promote the efficacy of somatic cell nuclear transfer.


Asunto(s)
Proliferación Celular , Fibroblastos , Histonas , Técnicas de Transferencia Nuclear , Animales , Bovinos , Fibroblastos/metabolismo , Fibroblastos/citología , Proliferación Celular/efectos de los fármacos , Histonas/metabolismo , Desarrollo Embrionario , Blastocisto/metabolismo , Blastocisto/citología , Lisina/metabolismo , Crotonatos/metabolismo , Células Cultivadas , Procesamiento Proteico-Postraduccional , Femenino
4.
Cell Cycle ; 23(1): 70-82, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38273425

RESUMEN

Our previous findings confirmed the high enrichment of Bacteroides fragilis (BF) in fecal samples from patients with colorectal cancer (CRC). The intestinal mucosal barrier is the first defense of the organism against commensal flora and intestinal pathogens and is closely associated with the occurrence and development of CRC. Therefore, this study aimed to investigate the molecular mechanisms through which BF mediates intestinal barrier injury and CRC progression. SW480 cells and a Caco2 intestinal barrier model were treated with entero-toxigenic BF (ETBF), its enterotoxin (B. fragilis toxin, BFT), and non-toxigenic BF (NTBF). Cell counting kit-8, flow cytometry, wound healing and transwell assays were performed to analyze the proliferation, apoptosis, migration, and invasion of SW480 cells. Transmission electron microscopy, FITC-dextran, and transepithelial electrical resistance (TEER) were used to analyze damage in the Caco2 intestinal barrier model. The Azoxymethane/Dextran Sulfate Sodium (AOM/DSS) animal model was established to evaluate the effect of ETBF on intestinal barrier injury and CRC progression in vivo. ETBF and BFT enhanced the viability, wound healing ratio, invasion, and EMT of SW480 cells. In addition, ETBF and BFT disrupted the tight junctions and villus structure in the intestinal barrier model, resulting in increased permeability and reduced TEER. Similarly, the expression of intestinal barrier-related proteins (MUC2, Occludin and Zo-1) was restricted by ETBF and BFT. Interestingly, the STAT3/ZEB2 axis was activated by ETBF and BFT, and treatment with Brevilin A (a STAT3 inhibitor) or knockdown of ZEB2 limited the promotional effect of ETBF and BFT on the SW480 malignant phenotype. In vivo experiments also confirmed that ETBF colonization accelerated tumor load, carcinogenesis, and intestinal mucosal barrier damage in the colorectum of the AOM/DSS animal model, and that treatment with Brevilin A alleviated these processes. ETBF-secreted BFT accelerated intestinal barrier damage and CRC by activating the STAT3/ZEB2 axis. Our findings provide new insights and perspectives for the application of ETBF in CRC treatment.


Asunto(s)
Toxinas Bacterianas , Bacteroides fragilis , Neoplasias Colorrectales , Factor de Transcripción STAT3 , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Animales , Humanos , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Bacteroides fragilis/genética , Bacteroides fragilis/metabolismo , Infecciones por Bacteroides/patología , Células CACO-2 , Neoplasias Colorrectales/patología , Crotonatos , Sesquiterpenos , Factor de Transcripción STAT3/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo
5.
Mult Scler Relat Disord ; 70: 104472, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36566698

RESUMEN

BACKGROUND: The prevalence of multiple sclerosis (MS) in older people is increasing due to population aging and availability of effective disease-modifying therapies (DMTs). Treating older people with MS is complicated by age-related and MS-related comorbidities, immunologic effects of prior DMTs, and immunosenescence. Teriflunomide is a once-daily oral immunomodulator that has demonstrated efficacy and acceptable safety in clinical trials of adults with relapsing forms of MS (RMS). However, there are limited clinical trial and real-world data regarding teriflunomide use in people with MS aged >55 years. We analyzed real-world data to assess the effectiveness and safety of teriflunomide in older people with RMS who had switched to this agent from other DMTs. METHODS: People with RMS (relapsing remitting and active secondary progressive MS) aged ≥55 years who had switched from other DMTs to teriflunomide (7 mg or 14 mg) for ≥1 year were identified retrospectively by chart review at four sites in the United States. Data were extracted from medical records from 1 year pre-index to 2 years post-index (index defined as the teriflunomide start date). Assessments of effectiveness included annualized relapse rate (ARR), Expanded Disability Status Scale (EDSS) score, and magnetic resonance imaging (MRI) outcomes. Assessments of safety included lymphocyte counts, infections, and malignancies. We examined the effectiveness outcomes and lymphocyte counts within sub-groups defined by age (55-64, ≥65 years), sex, MS type, and prior route of DMT administration (oral, injectable, infusible). RESULTS: In total, 182 patients with RMS aged ≥55 years who switched from other DMTs to teriflunomide were identified (mean [SD] age: 62.5 [5.4] years). Mean ARR decreased from the start of teriflunomide treatment (mean [SD]: 0.43 [0.61]) to year 1 post-index (0.13 [0.65]) and year 2 post-index (0.05 [0.28]). Mean EDSS score remained unchanged from index (mean [SD]: 4.5 [1.8]) to 1 year post-treatment (4.5 [1.8]) and increased slightly at 2 years post-treatment (4.7 [1.7]). MRI scans from index and years 1 and 2 post-index compared with scans from the previous year indicated that most patients had stable or improved MRI outcomes at index (87.7%) and remained stable or improved at years 1 (96.0%) and 2 (93.6%). Lymphopenia decreased at years 1 (21.4%) and 2 post-index (14.8%, compared to index (23.5%). By 1 year post-index, fewer patients had grade 3 or 4 lymphopenia, and at 2 years post-index, there were no patients with grade 3 or 4 lymphopenia. Infection incidence was low (n = 40, 22.0%) and none were related to teriflunomide. The decreases in lymphopenia were driven by decreases among people who switched from a prior oral DMT; there were no notable differences in lymphopenia across the other sub-groups examined. ARR, EDSS score, and MRI outcomes across all sub-groups were similar to the results of the overall population. CONCLUSION: Our multicenter, longitudinal, retrospective study demonstrated that patients with RMS aged 55 or older switching to teriflunomide from other DMTs had significantly improved ARR, stable disability, and stable or improved MRI over up to 2 years' follow up. Safety results were acceptable with fewer patients exhibiting lymphopenia at years 1 and 2 post-index.


Asunto(s)
Leucopenia , Linfopenia , Esclerosis Múltiple Recurrente-Remitente , Esclerosis Múltiple , Adulto , Humanos , Anciano , Persona de Mediana Edad , Esclerosis Múltiple/tratamiento farmacológico , Estudios Retrospectivos , Crotonatos/uso terapéutico , Toluidinas/uso terapéutico , Recurrencia , Linfopenia/inducido químicamente , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico
6.
Cell Mol Neurobiol ; 43(5): 2071-2087, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36219379

RESUMEN

Myasthenia gravis (MG) is an autoantibody-mediated autoimmune disease characterized by skeletal muscle weakness exacerbated with exercise. There is a need for novel drugs effective in refractory MG. We aimed to test the potential of teriflunomide, an immunomodulatory drug currently used in rheumatoid arthritis and multiple sclerosis treatment, in a murine experimental autoimmune myasthenia gravis (EAMG) model. EAMG was induced by immunizations with recombinant acetylcholine receptor (AChR). Teriflunomide treatment (10 mg/kg/day, intraperitoneal) was initiated to one group of mice (n = 21) following the third immunization and continued for 5 weeks. The disease control group (n = 19) did not receive medication. Naïve mice (n = 10) received only mock immunization. In addition to the clinical scorings, the numbers of B cells and T cells, and cytokine profiles of T cells were examined by flow cytometry. Anti-AChR-specific antibodies in the peripheral blood serum were quantified by ELISA. Teriflunomide significantly reduced clinical disease scores and the absolute numbers of CD4+ T cells and some of their cytokine-producing subgroups (IFN-γ, IL 2, IL22, IL-17A, GM-CSF) in the spleen and the lymph nodes. The thymic CD4+ T cells were also significantly reduced. Teriflunomide mostly spared CD8+ T cells' numbers and cytokine production, while reducing CD138+CD19+lambda+ plasma B cells' absolute numbers and CD138 mean fluorescent intensities, probably decreasing the number of IgG secreting more mature plasma cells. It also led to some selective changes in the measurements of anti-AChR-specific antibodies in the serum. Our results showed that teriflunomide may be beneficial in the treatment of MG in humans.


Asunto(s)
Miastenia Gravis , Humanos , Animales , Ratones , Crotonatos/farmacología , Crotonatos/uso terapéutico , Hidroxibutiratos , Nitrilos
7.
Mult Scler Relat Disord ; 68: 104377, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36544305

RESUMEN

BACKGROUND: Given its potential antiviral activity, we investigated the effect of teriflunomide on EBV in patients with relapsing-remitting MS (RRMS). METHODS: Saliva samples were collected at home and analysed for EBV DNA presence in patients with RRMS treated with teriflunomide for ≥3 months. RESULTS: The proportion of patients with detectable EBV in the teriflunomide cohort was lower than in the reference cohorts. The proportion of samples with EBV DNA or shedding from teriflunomide-treated patients was reduced relative to each reference cohort (P<0.0001; >5.8 virus copies/µL cut-off). CONCLUSION: This pilot study demonstrated the feasibility of at-home saliva sample collection and revealed a possible effect of teriflunomide on EBV shedding.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Esclerosis Múltiple Recurrente-Remitente , Esclerosis Múltiple , Humanos , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Herpesvirus Humano 4 , Proyectos Piloto , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Crotonatos/farmacología , Crotonatos/uso terapéutico , Estudios de Cohortes
8.
Eur J Med Chem ; 238: 114489, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35640328

RESUMEN

Human dihydroorotate dehydrogenase (hDHODH) is a key enzyme in the de novo synthesis pathway of pyrimidine nucleotide in cells. The moderate efficiency of teriflunomide, an approved hDHODH inhibitor for the treatment of multiple sclerosis, limited its therapeutic application of malignancy. Herein, thirty-seven novel teriflunomide derivatives with a biphenyl scaffold were designed, synthesized and evaluated. As a result, the optimal compound A37 exhibited a potent hDHODH inhibitory activity with an IC50 value of 10.2 nM, which was about 40-fold stronger than that of teriflunomide (IC50 = 407.8 nM), and showed favorable antiproliferative activities against HCT116 cells with an IC50 value of 0.3 µM. Meanwhile, A37 displayed an acceptable safety profile at an oral dosage of 400 mg/kg in the acute toxicity assay, and exhibited a promising antitumor effect with tumor growth inhibition rate of 54.4% at an oral dosage of 30 mg/kg in HCT116 xenograft model. These results indicate that A37 is an efficacious hDHODH inhibitor with potential in the treatment of colorectal carcinoma.


Asunto(s)
Neoplasias Colorrectales , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Neoplasias Colorrectales/tratamiento farmacológico , Crotonatos , Dihidroorotato Deshidrogenasa , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Hidroxibutiratos , Nitrilos , Relación Estructura-Actividad , Toluidinas
9.
Sci Rep ; 12(1): 3049, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35197552

RESUMEN

Astrocytes utilize both glycolytic and mitochondrial pathways to power cellular processes that are vital to maintaining normal CNS functions. These cells also mount inflammatory and acute phase reactive programs in response to diverse stimuli. While the metabolic functions of astrocytes under homeostatic conditions are well-studied, the role of cellular bioenergetics in astrocyte reactivity is poorly understood. Teriflunomide exerts immunomodulatory effects in diseases such as multiple sclerosis by metabolically reprogramming lymphocytes and myeloid cells. We hypothesized that teriflunomide would constrain astrocytic inflammatory responses. Purified murine astrocytes were grown under serum-free conditions to prevent acquisition of a spontaneous reactive state. Stimulation with TNFα activated NFκB and increased secretion of Lcn2. TNFα stimulation increased basal respiration, maximal respiration, and ATP production in astrocytes, as assessed by oxygen consumption rate. TNFα also increased glycolytic reserve and glycolytic capacity of astrocytes but did not change the basal glycolytic rate, as assessed by measuring the extracellular acidification rate. TNFα specifically increased mitochondrial ATP production and secretion of Lcn2 required ATP generated by oxidative phosphorylation. Inhibition of dihydroorotate dehydrogenase via teriflunomide transiently increased both oxidative phosphorylation and glycolysis in quiescent astrocytes, but only the increased glycolytic ATP production was sustained over time, resulting in a bias away from mitochondrial ATP production even at doses down to 1 µM. Preconditioning with teriflunomide prevented the TNFα-induced skew toward oxidative phosphorylation, reduced mitochondrial ATP production, and reduced astrocytic inflammatory responses, suggesting that this drug may limit neuroinflammation by acting as a metabolomodulator.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Astrocitos/metabolismo , Crotonatos/farmacología , Hidroxibutiratos/farmacología , Inflamación/metabolismo , Nitrilos/farmacología , Toluidinas/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Células Cultivadas , Quimiocinas/metabolismo , Metabolismo Energético/efectos de los fármacos , Glucólisis/efectos de los fármacos , Lipocalina 2/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
10.
Mult Scler Relat Disord ; 58: 103506, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35066272

RESUMEN

BACKGROUND: Diarrhea is generally a benign and self-limited adverse effect of teriflunomide. Small intestinal pathology has yet to be described with teriflunomide associated diarrhea. OBJECTIVE: To report small intestinal pathology in teriflunomide associated diarrhea. METHODS: Small intestinal and colonic biopsies were obtained from a patient with teriflunomide associated diarrhea. RESULTS: Small intestinal biopsy demonstrated blunting of villi, increased intraepithelial lymphocytes and expansion of lamina propria. Gliadin and t-transglutaminase antibodies were negative. Diarrhea resolved following elimination of teriflunomide with cholestyramine. CONCLUSION: This is the first reported case of small intestinal inflammation similar to celiac disease in teriflunomide associated diarrhea.


Asunto(s)
Crotonatos , Toluidinas , Crotonatos/efectos adversos , Diarrea/inducido químicamente , Humanos , Hidroxibutiratos , Nitrilos , Toluidinas/efectos adversos
11.
CNS Drugs ; 36(1): 45-59, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34940954

RESUMEN

Pediatric-onset multiple sclerosis (POMS) is a rare neuroinflammatory and neurodegenerative disease that has a significant impact on long-term physical and cognitive patient outcomes. A small percentage of multiple sclerosis (MS) diagnoses occur before the age of 18 years. Before treatment initiation, a careful differential diagnosis and exclusion of other similar acquired demyelinating syndromes such as anti-aquaporin-4-associated neuromyelitis optica spectrum disorder (AQP4-NMOSD) and myelin oligodendrocyte glycoprotein antibody spectrum disorder (MOGSD) is warranted. The recent 2017 changes to the McDonald criteria can successfully predict up to 71% of MS diagnoses and have good specificity of 95% and sensitivity of 71%. Additional measures such as the presence of T1-weighted hypointense lesions and/or contrast-enhancing lesions significantly increase the accuracy of diagnosis. In adults, early use of disease-modifying therapies (DMTs) is instrumental to a better long-term prognosis, including lower rates of relapse and disability worsening, and numerous FDA-approved therapies for adult-onset MS are available. However, unlike their adult counterparts, the development, testing, and regulatory approval of POMS treatments have been significantly slower and hindered by logistic and/or ethical considerations. Currently, only two MS DMTs (fingolimod and teriflunomide) have been tested in large phase III trials and approved by regulatory agencies for use in POMS. First-line therapies not approved by the FDA for use in children (interferon-ß and glatiramer acetate) are also commonly used and result in a significant reduction in inflammatory activity when compared with non-treated POMS patients. An increasing number of POMS patients are now treated with moderate efficacy therapies such as dimethyl fumarate and high-efficacy therapies such as natalizumab, anti-CD20 monoclonal antibodies, anti-CD52 monoclonal antibodies, and/or autologous hematopoietic stem cell transplantation. These high-efficacy DMTs generally provide additional reduction in inflammatory activity when compared with the first-line medications (up to 62% of relapse-rate reduction). Therefore, a number of phase II and III trials are currently investigating their efficacy and safety in POMS patients. In this review, we discuss potential changes in the regulatory approval process for POMS patients that are recommended for DMTs already approved for the adult MS population, including smaller sample size for pharmacokinetic/pharmacodynamic studies, MRI-centered primary outcomes, and/or inclusion of teenagers in the adult trials.


Asunto(s)
Esclerosis Múltiple/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Niño , Crotonatos/uso terapéutico , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Hidroxibutiratos/uso terapéutico , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/diagnóstico , Nitrilos/uso terapéutico , Pronóstico , Moduladores de los Receptores de fosfatos y esfingosina 1/uso terapéutico , Toluidinas/uso terapéutico
12.
J Med Chem ; 64(24): 18175-18192, 2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-34905371

RESUMEN

Human dihydroorotate dehydrogenase (hDHODH), as the fourth and rate-limiting enzyme of the de novo pyrimidine synthesis pathway, is regarded as an attractive target for malignancy therapy. In the present study, a novel series of teriflunomide derivatives were designed, synthesized, and evaluated as hDHODH inhibitors. 13t was the optimal compound with promising enzymatic activity (IC50 = 16.0 nM), potent antiproliferative activity against human lymphoma Raji cells (IC50 = 7.7 nM), and excellent aqueous solubility (20.1 mg/mL). Mechanistically, 13t directly inhibited hDHODH and induced cell cycle S-phase arrest in Raji cells. The acute toxicity assay indicated a favorable safety profile of 13t. Notably, 13t displayed significant tumor growth inhibition activity with a tumor growth inhibition (TGI) rate of 81.4% at 30 mg/kg in a Raji xenograft model. Together, 13t is a promising inhibitor of hDHODH and a preclinical candidate for antitumor therapy, especially for lymphoma.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Crotonatos/química , Crotonatos/farmacología , Dihidroorotato Deshidrogenasa/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hidroxibutiratos/química , Hidroxibutiratos/farmacología , Neoplasias/tratamiento farmacológico , Nitrilos/química , Nitrilos/farmacología , Toluidinas/química , Toluidinas/farmacología , Antineoplásicos/síntesis química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Crotonatos/síntesis química , Inhibidores Enzimáticos/síntesis química , Humanos , Hidroxibutiratos/síntesis química , Neoplasias/patología , Nitrilos/síntesis química , Relación Estructura-Actividad , Toluidinas/síntesis química
13.
Front Immunol ; 12: 730342, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721394

RESUMEN

Background and Objectives: Inhibition of de novo pyrimidine synthesis in proliferating T and B lymphocytes by teriflunomide, a pharmacological inhibitor of dihydroorotate dehydrogenase (DHODH), has been shown to be an effective therapy to treat patients with MS in placebo-controlled phase 3 trials. Nevertheless, the underlying mechanism contributing to the efficacy of DHODH inhibition has been only partially elucidated. Here, we aimed to determine the impact of teriflunomide on the immune compartment in a longitudinal high-dimensional follow-up of patients with relapse-remitting MS (RRMS) treated with teriflunomide. Methods: High-dimensional spectral flow cytometry was used to analyze the phenotype and the function of innate and adaptive immune system of patients with RRMS before and 12 months after teriflunomide treatment. In addition, we assessed the impact of teriflunomide on the migration of memory CD8 T cells in patients with RRMS, and we defined patient immune metabolic profiles. Results: We found that 12 months of treatment with teriflunomide in patients with RRMS does not affect the B cell or CD4 T cell compartments, including regulatory TREG follicular helper TFH cell and helper TH cell subsets. In contrast, we observed a specific impact of teriflunomide on the CD8 T cell compartment, which was characterized by decreased homeostatic proliferation and reduced production of TNFα and IFNγ. Furthermore, we showed that DHODH inhibition also had a negative impact on the migratory velocity of memory CD8 T cells in patients with RRMS. Finally, we showed that the susceptibility of memory CD8 T cells to DHODH inhibition was not related to impaired metabolism. Discussion: Overall, these findings demonstrate that the clinical efficacy of teriflunomide results partially in the specific susceptibility of memory CD8 T cells to DHODH inhibition in patients with RRMS and strengthens active roles for these T cells in the pathophysiological process of MS.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Crotonatos/uso terapéutico , Dihidroorotato Deshidrogenasa/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Hidroxibutiratos/uso terapéutico , Memoria Inmunológica/efectos de los fármacos , Inmunosupresores/uso terapéutico , Células T de Memoria/efectos de los fármacos , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Nitrilos/uso terapéutico , Toluidinas/uso terapéutico , Adulto , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Crotonatos/efectos adversos , Dihidroorotato Deshidrogenasa/metabolismo , Inhibidores Enzimáticos/efectos adversos , Femenino , Humanos , Hidroxibutiratos/efectos adversos , Inmunosupresores/efectos adversos , Interferón gamma/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Células T de Memoria/enzimología , Células T de Memoria/inmunología , Esclerosis Múltiple Recurrente-Remitente/diagnóstico , Esclerosis Múltiple Recurrente-Remitente/enzimología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Nitrilos/efectos adversos , Fenotipo , Factores de Tiempo , Toluidinas/efectos adversos , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/metabolismo
14.
Int J Mol Sci ; 22(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34575975

RESUMEN

Several classes of immunomodulators are used for treating relapsing-remitting multiple sclerosis (RRMS). Most of these disease-modifying therapies, except teriflunomide, carry the risk of progressive multifocal leukoencephalopathy (PML), a severely debilitating, often fatal virus-induced demyelinating disease. Because teriflunomide has been shown to have antiviral activity against DNA viruses, we investigated whether treatment of cells with teriflunomide inhibits infection and spread of JC polyomavirus (JCPyV), the causative agent of PML. Treatment of choroid plexus epithelial cells and astrocytes with teriflunomide reduced JCPyV infection and spread. We also used droplet digital PCR to quantify JCPyV DNA associated with extracellular vesicles isolated from RRMS patients. We detected JCPyV DNA in all patients with confirmed PML diagnosis (n = 2), and in six natalizumab-treated (n = 12), two teriflunomide-treated (n = 7), and two nonimmunomodulated (n = 2) patients. Of the 21 patients, 12 (57%) had detectable JCPyV in either plasma or serum. CSF was uniformly negative for JCPyV. Isolation of extracellular vesicles did not increase the level of detection of JCPyV DNA versus bulk unprocessed biofluid. Overall, our study demonstrated an effect of teriflunomide inhibiting JCPyV infection and spread in glial and choroid plexus epithelial cells. Larger studies using patient samples are needed to correlate these in vitro findings with patient data.


Asunto(s)
Crotonatos/farmacología , Virus ADN/efectos de los fármacos , Hidroxibutiratos/farmacología , Leucoencefalopatía Multifocal Progresiva/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Neuroglía/efectos de los fármacos , Nitrilos/farmacología , Toluidinas/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/virología , Línea Celular , Plexo Coroideo/efectos de los fármacos , Plexo Coroideo/virología , Virus ADN/patogenicidad , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/virología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/virología , Humanos , Factores Inmunológicos/efectos adversos , Factores Inmunológicos/uso terapéutico , Virus JC/efectos de los fármacos , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva/inducido químicamente , Leucoencefalopatía Multifocal Progresiva/patología , Leucoencefalopatía Multifocal Progresiva/virología , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/patología , Esclerosis Múltiple Recurrente-Remitente/virología , Neuroglía/virología , Virosis/tratamiento farmacológico , Virosis/genética , Virosis/virología
15.
Molecules ; 26(18)2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34577124

RESUMEN

Leflunomide, an anti-inflammatory agent, has been shown to be effective in multiple myeloma (MM) treatment; however, the mechanism of this phenomenon has not been fully elucidated. The aim of the study was to assess the role of mitochondria and dihydroorotate dehydrogenase (DHODH) inhibition in the cytotoxicity of leflunomide in relation to the MM cell line RPMI 8226. The cytotoxic effect of teriflunomide-an active metabolite of leflunomide-was determined using MTT assay, apoptosis detection, and cell cycle analysis. To evaluate DHODH-dependent toxicity, the cultures treated with teriflunomide were supplemented with uridine. Additionally, the level of cellular thiols as oxidative stress symptom was measured as well as mitochondrial membrane potential and protein tyrosine kinases (PTK) activity. The localization of the compound in cell compartments was examined using HPLC method. Teriflunomide cytotoxicity was not abolished in uridine presence. Observed apoptosis occurred in a mitochondria-independent manner, there was also no decrease in cellular thiols level. Teriflunomide arrested cell cycle in the G2/M phase which is not typical for DHODH deficiency. PTK activity was decreased only at the highest drug concentration. Interestingly, teriflunomide was not detected in the mitochondria. The aforementioned results indicate DHODH- and mitochondria-independent mechanism of leflunomide toxicity against RPMI 8226 cell line.


Asunto(s)
Leflunamida , Mieloma Múltiple , Antineoplásicos , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Crotonatos , Inhibidores Enzimáticos/farmacología , Humanos , Hidroxibutiratos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Nitrilos , Toluidinas
16.
Sci Rep ; 11(1): 12533, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131191

RESUMEN

Neoplasm development in Multiple Sclerosis (MS) patients treated with disease-modifying therapies (DMTs) has been widely discussed. The aim of this work is to determine neoplasm frequency, relationship with the prescription pattern of DMTs, and influence of the patients' baseline characteristics. Data from 250 MS outpatients were collected during the period 1981-2019 from the medical records of the Neurology Service of the HUPM (Hospital Universitario Puerta del Mar)-in Southern Spain-and analysed using Cox models. Neoplasm prevalence was 24%, mainly located on the skin, with cancer prevalence as expected for MS (6.8%). Latency period from MS onset to neoplasm diagnosis was 10.4 ± 6.9 years (median 9.30 [0.9-30.5]). During the observation period ß-IFN (70.4% of patients), glatiramer acetate (30.4%), natalizumab (16.8%), fingolimod (24.8%), dimethyl fumarate (24.0%), alemtuzumab (6.0%), and teriflunomide (4.8%) were administered as monotherapy. Change of pattern in step therapy was significantly different in cancer patients vs unaffected individuals (p = 0.011) (29.4% did not receive DMTs [p = 0.000]). Extended Cox model: Smoking (HR = 3.938, CI 95% 1.392-11.140, p = 0.010), being female (HR = 2.006, 1.070-3.760, p = 0.030), and age at MS diagnosis (AGE-DG) (HR = 1.036, 1.012-1.061, p = 0.004) were risk factors for neoplasm development. Secondary progressive MS (SPMS) phenotype (HR = 0.179, 0.042-0.764, p = 0.020) and treatment-time with IFN (HR = 0.923, 0.873-0.977, p = 0.006) or DMF (HR = 0.725, 0.507-1.036, p = 0.077) were protective factors. Tobacco and IFN lost their negative/positive influence as survival time increased. Cox PH model: Tobacco/AGE-DG interaction was a risk factor for cancer (HR = 1.099, 1.001-1.208, p = 0.049), followed by FLM treatment-time (HR = 1.219, 0.979-1.517). In conclusion, smoking, female sex, and AGE-DG were risk factors, and SPMS and IFN treatment-time were protective factors for neoplasm development; smoking/AGE-DG interaction was the main cancer risk factor.


Asunto(s)
Inmunosupresores/efectos adversos , Esclerosis Múltiple Crónica Progresiva/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico , Neoplasias/epidemiología , Adulto , Anciano , Alemtuzumab/efectos adversos , Alemtuzumab/uso terapéutico , Crotonatos/efectos adversos , Crotonatos/uso terapéutico , Dimetilfumarato/efectos adversos , Dimetilfumarato/uso terapéutico , Femenino , Clorhidrato de Fingolimod/efectos adversos , Clorhidrato de Fingolimod/uso terapéutico , Acetato de Glatiramer/efectos adversos , Acetato de Glatiramer/uso terapéutico , Humanos , Hidroxibutiratos/efectos adversos , Hidroxibutiratos/uso terapéutico , Inmunosupresores/uso terapéutico , Interferón beta/efectos adversos , Interferón beta/uso terapéutico , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/epidemiología , Esclerosis Múltiple/patología , Esclerosis Múltiple Crónica Progresiva/complicaciones , Esclerosis Múltiple Crónica Progresiva/epidemiología , Esclerosis Múltiple Crónica Progresiva/patología , Esclerosis Múltiple Recurrente-Remitente/complicaciones , Esclerosis Múltiple Recurrente-Remitente/epidemiología , Esclerosis Múltiple Recurrente-Remitente/patología , Natalizumab/efectos adversos , Natalizumab/uso terapéutico , Neoplasias/inducido químicamente , Neoplasias/patología , Nitrilos/efectos adversos , Nitrilos/uso terapéutico , Pacientes Ambulatorios , Modelos de Riesgos Proporcionales , Factores de Riesgo , Fumar/efectos adversos , España , Toluidinas/efectos adversos , Toluidinas/uso terapéutico
17.
Presse Med ; 50(2): 104068, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34033862

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system. In recent years, many disease-modifying therapies (DMT) have been approved for MS treatment. For this reason, a profound knowledge of the characteristics and indications of the available compounds is required to tailor the therapeutic strategy to the individual patient characteristics. This should include the mechanism of action and pharmacokinetic of the drug, the safety and efficacy profile provided by clinical trials, as well as the understanding of possible side effects. Moreover, the evolving knowledge of the disease is paving the way to new and innovative therapeutic approaches, as well as the development of new biomarkers to monitor the therapeutic response and to guide the clinician's therapeutic choices. In this review we provide a comprehensive overview on currently approved therapies in MS and the emerging evidence-based strategies to adopt for initiating, monitoring, and eventually adapting a therapeutic regimen with DMT.


Asunto(s)
Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/prevención & control , Algoritmos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Cladribina/uso terapéutico , Crotonatos/uso terapéutico , Dimetilfumarato/uso terapéutico , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Humanos , Hidroxibutiratos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Indanos/uso terapéutico , Interferón beta/uso terapéutico , Masculino , Mitoxantrona/uso terapéutico , Natalizumab/uso terapéutico , Nitrilos/uso terapéutico , Oxadiazoles/uso terapéutico , Embarazo , Moduladores de los Receptores de fosfatos y esfingosina 1/uso terapéutico , Receptores de Esfingosina-1-Fosfato/uso terapéutico , Toluidinas/uso terapéutico
18.
Clin Immunol ; 227: 108750, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33945870

RESUMEN

The co-existence of an autoinflammatory syndrome with a demyelinating disorder is a very rare occurrence raising the question whether there is a pathophysiological connection between them. We describe the case of a man with symptoms of cryopyrin-associated periodic syndrome (CAPS) since infancy who later developed multiple sclerosis (MS). As CAPS was genetically confirmed, the inhibition of interleukin-1 (IL-1) with anakinra led to a swift resolution of the CAPS symptoms and also, in combination with teriflunomide, to a clinical and imaging improvement of MS. In vitro studies showed that, upon a CAPS flare, the patient's peripheral neutrophils released neutrophil extracellular traps (NETs) decorated with IL-1ß, while NET release was markedly decreased following anakinra-induced remission of CAPS. Taking into account the growing evidence on the involvement of IL-1ß in experimental models of MS, this rare patient case suggests that the role of neutrophils/NETs and IL-1ß in MS should be further studied.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina/inmunología , Trampas Extracelulares/inmunología , Interleucina-1beta/inmunología , Esclerosis Múltiple/inmunología , Neutrófilos/inmunología , Adulto , Antirreumáticos/uso terapéutico , Encéfalo/diagnóstico por imagen , Crotonatos/uso terapéutico , Síndromes Periódicos Asociados a Criopirina/complicaciones , Síndromes Periódicos Asociados a Criopirina/tratamiento farmacológico , Síndromes Periódicos Asociados a Criopirina/fisiopatología , Humanos , Hidroxibutiratos/uso terapéutico , Inmunosupresores/uso terapéutico , Técnicas In Vitro , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Imagen por Resonancia Magnética , Masculino , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/tratamiento farmacológico , Nitrilos/uso terapéutico , Toluidinas/uso terapéutico
19.
Artículo en Inglés | MEDLINE | ID: mdl-33837058

RESUMEN

OBJECTIVE: To test the hypothesis that teriflunomide can reduce ex vivo spontaneous proliferation of peripheral blood mononuclear cells (PBMCs) from patients with human T-cell lymphotropic virus type 1 (HTLV-1)-associated myelopathy/tropical spastic paraparesis (HAM/TSP). METHODS: PBMCs from patients with HAM/TSP were cultured in the presence and absence of teriflunomide and assessed for cell viability, lymphocyte proliferation, activation markers, HTLV-1 tax and HTLV-1 hbz messenger ribonucleic acid (mRNA) expression, and HTLV-1 Tax protein expression. RESULTS: In culture, teriflunomide did not affect cell viability. A concentration-dependent reduction in spontaneous proliferation of PBMCs was observed with 25 µM (38.3% inhibition), 50 µM (65.8% inhibition), and 100 µM (90.7% inhibition) teriflunomide. The inhibitory effects of teriflunomide were detected in both CD8+ and CD4+ T-cell subsets, which are involved in the immune response to HTLV-1 infection and the pathogenesis of HAM/TSP. There was no significant change in HTLV-1 proviral load (PVL) or tax mRNA/Tax protein expression in these short-term cultures, but there was a significant reduction of HTLV-1 PVL due to inhibition of proliferation of CD4+ T cells obtained from a subset of patients with HAM/TSP. CONCLUSIONS: These results suggest that teriflunomide inhibits abnormal T-cell proliferation associated with HTLV-1 infection and may have potential as a therapeutic option in patients with HAM/TSP.


Asunto(s)
Crotonatos/farmacología , Hidroxibutiratos/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Nitrilos/farmacología , Paraparesia Espástica Tropical/tratamiento farmacológico , Toluidinas/farmacología , Adulto , Anciano , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Productos del Gen tax/metabolismo , Infecciones por HTLV-I/complicaciones , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Masculino , Persona de Mediana Edad , Cultivo Primario de Células , ARN Mensajero/metabolismo , Subgrupos de Linfocitos T/efectos de los fármacos
20.
Mol Immunol ; 135: 116-126, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33892379

RESUMEN

Brevilin A (BA), is a natural biologically active ingredient derived from Centipeda minima with several reports of anti-cancer, while its anti-inflammatory activity is rarely reported. Current studies have found the dysregulated activation of NLRP3 inflammasome cause a variety of inflammatory diseases. Targeting the NLRP3 inflammasome contributes to the treatment of NLRP3-induced diseases. Here, we found that BA significantly attenuates the activation of caspase-1 and the subsequent secretion of the interleukin-1ß (IL-1ß) in mouse macrophages and human THP-1 cells, showing the inhibitory effect of BA on the NLRP3 inflammasome activation. Moreover, BA specifically inhibits NLRs inflammasomes activation triggered by multi-stimuli, but it has no effect on the AIM2 inflammasome activation, indicating that BA is a specific inhibitor of the NLRs inflammasomes. Research on the mechanism found BA inhibits NLRP3 inflammasome activation by blocking the upstream of ASC oligomerization. Importantly, in vivo experiments showed that BA markedly reduces the secretion of IL-1ß to suppress NLRP3 inflammasome in the LPS-induced inflammation and MSU-challenged peritonitis model. In conclusion, our experiments show that BA is an effective NLRP3 inflammasome inhibitor and can be regarded as a drug candidate for NLRP3 inflammasome-driven diseases.


Asunto(s)
Inhibidores de Caspasas/farmacología , Crotonatos/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Sesquiterpenos/farmacología , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células THP-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA