Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mult Scler Relat Disord ; 87: 105604, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38718750

RESUMO

BACKGROUND: Multiple Sclerosis (MS) may cluster in families, an entity known as familial MS (FMS), possibly due to aggregation of genetic and environmental factors. Though previous studies have characterized FMS in different populations, no study to the best of our knowledge has yet characterized FMS in the unique Israeli population, which is comprised of relatively endogamous ethnicities. Our goal in this study was to compare demographic and clinical characteristics between FMS and sporadic MS (SMS), and to search for intra-familial patterns. METHODS: In a retrospective study of 101 FMS patients and 508 SMS patients, ethnicity and sex distribution was assessed. Clinical aspects were compared between 172 paired FMS and SMS patients, matched for sex, age and ethnicity, and between generations of the FMS cohort. RESULTS: Females comprised 75.3 % of FMS and 67.5 % of SMS patients (p = 0.1). Ethnic distribution was significantly different between FMS and SMS (p = 0.014), with the former comprising a higher proportion of Christian-Arabs (15.4% vs. 5.1 %, p = 0.004) and lower proportion of Jews (60% vs. 74.2 %, p = 0.016). Age at disease onset or diagnosis, frequency of positive Oligoclonal bands and comorbidity of other autoimmune/inflammatory disease or chronic diseases was comparable between FMS and SMS, yet motor symptoms at onset were more prevalent in FMS (34% vs. 20 %, p = 0.02). Annualized relapse rates throughout 10 years from onset were comparable. Among FMS, mean Expanded-Disability-Status-Scale (EDSS) and slope of deterioration in EDSS over 20 years from diagnosis were higher (p = 0.0004 and p = 0.023, respectively), time to EDSS ≥ 3 was shorter (7.1 vs. 12.1 years, HR 1.6, p = 0.036) and MS-Severity-Score (MSSS) was higher (3.84 vs. 2.95, p = 0.04), compared to SMS. Following adjustment for smoking, which tended to be higher among FMS patients (P = 0.06), mean EDSS and slope of deterioration in EDSS over 20 years remained significantly higher (p = 0.0006 and p = 0.025, respectively) in FMS, time to EDSS ≥ 3 tended to be higher (HR 1.5, p = 0.06), while MSSS was comparable. An inter-generational analysis of the total FMS cohort, as well as an intra-familial analysis, both adjusted for year of diagnosis, revealed significantly earlier age of onset (p < 0.0001 and p < 0.0001) and diagnosis (p = 0.001 and p < 0.0001) in the younger compared to the older generations, respectively. CONCLUSION: In this Israeli cohort, the proportions of specific ethnicities differ between FMS and SMS, indicating that FMS has a population-specific prevalence pattern, and that further investigation for susceptibility genes is warranted. Disease progression is faster in FMS patients and anticipation is observed in families with multiple cases of MS. Closer surveillance and application of a pro-active induction or early highly-effective therapeutic strategy for FMS patients should be considered, to reduce high disease activity and fast disability progression.


Assuntos
Progressão da Doença , Esclerose Múltipla , Humanos , Feminino , Masculino , Israel/epidemiologia , Israel/etnologia , Adulto , Estudos Retrospectivos , Esclerose Múltipla/etnologia , Esclerose Múltipla/genética , Esclerose Múltipla/epidemiologia , Pessoa de Meia-Idade , Idade de Início , Adulto Jovem
2.
Int J Mol Sci ; 24(19)2023 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-37834472

RESUMO

Microbiome dysbiosis is increasingly being recognized as implicated in immune-mediated disorders including multiple sclerosis (MS). The microbiome is modulated by genetic and environmental factors including lifestyle, diet, and drug intake. This study aimed to characterize the MS-associated gut microbiome in the Israeli populations and to identify associations with demographic, dietary, and clinical features. The microbiota from 57 treatment-naive patients with MS (PwMS) and 43 age- and gender-matched healthy controls (HCs) was sequenced and abundance compared. Associations between differential microbes with demographic or clinical characteristics, as well as diet and nutrient intake, were assessed. While there was no difference in α- or ß-diversity of the microbiome, we identified 40 microbes from different taxonomic levels that differ in abundance between PwMS and HCs, including Barnesiella, Collinsella, Egerthella, Mitsuokella, Olsenella Romboutsia, and Succinivibrio, all enhanced in PwMS, while several members of Lacnospira were reduced. Additional MS-differential microbes specific to ethnicity were identified. Several MS-specific microbial patterns were associated with gender, vitamin D level, Mediterranean diet, nutrient intake, or disability status. Thus, PwMS have altered microbiota composition, with distinctive patterns related to geographic locations and population. Microbiome dysbiosis seem to be implicated in disease progression, gender-related differences, and vitamin D-mediated immunological effects recognized in MS. Dietary interventions may be beneficial in restoring a "healthy microbiota" as part of applying comprehensive personalized therapeutic strategies for PwMS.


Assuntos
Dieta Mediterrânea , Microbioma Gastrointestinal , Esclerose Múltipla , Humanos , Vitamina D , Etnicidade , Disbiose/complicações , Israel , Dieta , Vitaminas
3.
Front Immunol ; 13: 868915, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35432335

RESUMO

Background: Immunomodulatory/immunosuppressive activity of multiple sclerosis (MS) disease modifying therapies (DMTs) might affect immune responses to SARS-CoV-2 exposure or vaccination in patients with MS (PwMS). We evaluated the effect of DMTs on humoral and cell-mediated immune responses to 2 and 3 vaccinations and the longevity of SARS-Cov-2 IgG levels in PwMS. Methods: 522 PwMS and 68 healthy controls vaccinated with BNT162b2-Pfizer mRNA vaccine against SARS-CoV-2, or recovering from COVID-19, were recruited in a nation-wide multi-center study. Blood was collected at 3 time-points: 2-16 weeks and ~6 months post 2nd vaccination and 1-16 weeks following 3rd vaccination. Serological responses were measured by quantifying IgG levels against the spike-receptor-binding-domain of SARS-CoV-2, and cellular responses (in a subgroup analysis) by quantifying IFNγ secretion in blood incubated with COVID-19 spike-antigen. Results: 75% PwMS were seropositive post 2nd or 3rd vaccination. IgG levels decreased by 82% within 6 months from vaccination (p<0.0001), but were boosted 10.3 fold by the 3rd vaccination (p<0.0001), and 1.8 fold compared to ≤3m post 2nd vaccination (p=0.025). Patients treated with most DMTs were seropositive post 2nd and 3rd vaccinations, however only 38% and 44% of ocrelizumab-treated patients and 54% and 46% of fingolimod-treated patients, respectively, were seropositive. Similarly, in COVID-19-recovered patients only 54% of ocrelizumab-treated, 75% of fingolimod-treated and 67% of cladribine-treated patients were seropositive. A time interval of ≥5 months between ocrelizumab infusion and vaccination was associated with higher IgG levels (p=0.039 post-2nd vaccination; p=0.036 post-3rd vaccination), and with higher proportions of seropositive patients. Most fingolimod- and ocrelizumab-treated patients responded similarly to 2nd and 3rd vaccination. IFNγ-T-cell responses were detected in 89% and 63% of PwMS post 2nd and 3rd vaccination, however in only 25% and 0% of fingolimod-treated patients, while in 100% and 86% of ocrelizumab-treated patients, respectively. Conclusion: PwMS treated with most DMTs developed humoral and T-cell responses following 2 and 3 mRNA SARS-CoV-2 vaccinations. Fingolimod- or ocrelizumab-treated patients had diminished humoral responses, and fingolimod compromised the cellular responses, with no improvement after a 3rd booster. Vaccination following >5 months since ocrelizumab infusion was associated with better sero-positivity. These findings may contribute to the development of treatment-stratified vaccination guidelines for PwMS.


Assuntos
COVID-19 , Esclerose Múltipla , Vacina BNT162 , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Cloridrato de Fingolimode/uso terapêutico , Humanos , Imunidade Celular , Imunoglobulina G/uso terapêutico , Israel , Esclerose Múltipla/tratamento farmacológico , RNA Mensageiro/uso terapêutico , SARS-CoV-2 , Vacinação , Vacinas Sintéticas , Vacinas de mRNA
4.
J Neuroimmunol ; 343: 577230, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32247228

RESUMO

Dimethyl Fumarate (DMF), known for its mechanism of action targeting Nrf2 and related redox homeostasis, is an approved immunotherapy for patients with Multiple Sclerosis (PwMS) in the relapsing form. We assessed how DMF modulates immune cell functions, namely the cytokine profile of co-cultured B and T cells, and the chemokine-mediated migration of immune cells. Following DMF therapy, LTα+, TNFα+ and IFNγ+ B cells were reduced while TGFß and IL10 expression elevated. B cells from DMF-treated patients increased TGFß and LTα expression on T cells, while DMF directly reduced TNFα+ and IFNγ+ T cells. CXCL12/CXCL13-mediated migration of B cells, Monocytes, CD4 and CD8 T cells was reduced, with altered CXCR5 and CXCR4 expression. Induction of regulatory B and T cells and reduced migration of immune cells may be part of the beneficial mechanism of DMF in PwMS.


Assuntos
Linfócitos B/efeitos dos fármacos , Fumarato de Dimetilo/uso terapêutico , Imunossupressores/uso terapêutico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Linfócitos B/imunologia , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Humanos , Esclerose Múltipla Recidivante-Remitente/imunologia , Linfócitos T/imunologia
5.
Mult Scler Relat Disord ; 34: 29-37, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31228713

RESUMO

BACKGROUND: Fingolimod, an oral therapy for patients with relapsing Multiple Sclerosis (MS), traps CC chemokine receptor type 7 (CCR7)-expresssing lymphocytes within lymphoid tissues in the periphery, thereby supposedly reducing the infiltration of pathogenic cells into the central nervous system. Additional immunomodulatory effects of Fingolimod, involving cell function, B and T cells interactions and cross-regulation, have scarcely been studied. The objective of this study was to assess how Fingolimod therapy affects B cells functions, namely cell migration, immunoglobullin production and T cell stimulation. METHODS: B cells from 36 patients with relapsing MS were obtained before and after 3 months Fingolimod therapy, while CD4 T cells were collected pre-treatment. Clinical follow-up was performed for 1 year. For in-vitro validation, Lymphoblastoid cell-lines from 16 patients were cultured with Fingolimod. B cell migration towards C-X-C Motif Chemokine Ligand 12 (CXCL12) was assessed using a transwell system. C-X-C chemokine receptor 4 (CXCR4) expression was assessed by flow cytometry and western blot. Plasma immunoglobullins and Brain-derived Neurotrophic Factor (BDNF) were assessed by ELISA or RT-PCR. Drug effect on interacting co-cultured B and T cells on cytokine profiles and T cell proliferation was explored by flow cytometry. RESULTS: Lymphocyte count reduction did not predict clinical response of patients. Fingolimod therapy reduced CXCR4 expression and B cell migration towards CXCL12. No effect was found on immunoglobulins and BDNF. B cells from Fingolimod-treated patients induced a reduction in pro-inflammatory cytokines in T cells, while increased transforming growth factor beta (TGFß)+ B and T cells, and downregulated IL2-secretion from proliferative T cells. CONCLUSIONS: Fingolimod promotes anti-inflammatory cytokine profiles of B and T cells, through induction of regulatory B cells. Reduced B cell migration capacity in Fingolimod-treated patients leading to decreased cerebral inflammatory infiltration, may be part of the mechanism by which Fingolimod reduces disease activity in MS.


Assuntos
Linfócitos B/imunologia , Movimento Celular/imunologia , Cloridrato de Fingolimode/uso terapêutico , Imunossupressores/uso terapêutico , Esclerose Múltipla Recidivante-Remitente/terapia , Receptores CXCR4/metabolismo , Adulto , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proliferação de Células , Quimiocina CXCL12/metabolismo , Feminino , Seguimentos , Humanos , Inflamação/imunologia , Inflamação/terapia , Interleucina-2/metabolismo , Masculino , Esclerose Múltipla Recidivante-Remitente/imunologia , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/metabolismo , Resultado do Tratamento
6.
Neurol Neuroimmunol Neuroinflamm ; 5(6): e508, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30345334

RESUMO

OBJECTIVE: To elucidate the immunomodulatory effects of dimethyl fumarate (DMF) on B cells in patients with relapsing MS receiving DMF as a "1st-line" vs "2nd-line" therapy. METHODS: B cells were isolated from 43 patients with MS at baseline and after 15-week DMF therapy. Phenotype and functional markers and cytokine profile were assessed by flow cytometry. Analysis included clinical and MRI parameters recorded during a 1-year follow-up. RESULTS: 1st-line and 2nd-line patients presented several differences in their baseline immune profile, which corresponded with differences in their immunologic response to DMF treatment. DMF reduced the proportions of B cells and CD8 T cells whereas increased monocytes. DMF reduced memory B cells, including plasma cells in 2nd-line patients only, whereas strongly increased transitional B cells. Several IL10+ B-cell subsets and TGFß+ B cells were increased. Proinflammatory LTα+ and TNFα+ B cells were reduced, while IL4+ B cells elevated, whereas IFNγ+ B cells showed opposite effects in 1st-line and 2nd-line patients. HLA and ICAM-1 expression was increased, but % CD86+ B cells reduced. The expression of B-cell activating factor receptor and the proportion of activated CD69 B cells were increased. CONCLUSIONS: DMF is associated with increased transitional and IL10+ and TGFß+ regulatory B cells and a shift toward a more anti-inflammatory immune profile. Cell activation with reduced costimulatory capacity may induce immune hyporesponsiveness. Carryover effects of preceding therapies in 2nd-line patients and the stage of disease influence the immune profile of the patients and the immunomodulatory effects of DMF.

7.
Clin Immunol ; 184: 11-25, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28461106

RESUMO

The role of B cells in the pathogenesis of Multiple Sclerosis (MS), an autoimmune neurodegenerative disease, is becoming eminent in recent years, but the specific contribution of the distinct B cell subsets remains to be elucidated. Several B cell subsets have shown regulatory, anti-inflammatory capacities in response to stimuli in vitro, as well as in the animal model of MS: Experimental Autoimmune Encephalomyelitis (EAE). However, the functional role of the B regulatory cells (Bregs) in vivo and specifically in the human disease is yet to be clarified. In the present review, we have summarized the updated information on the roles of effector and regulatory B cells in MS and the immune-modulatory effects of MS therapeutic agents on their phenotype and function.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B Reguladores/imunologia , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Alemtuzumab/uso terapêutico , Animais , Crotonatos/uso terapêutico , Fumarato de Dimetilo/uso terapêutico , Cloridrato de Fingolimode/uso terapêutico , Acetato de Glatiramer/uso terapêutico , Humanos , Hidroxibutiratos , Fatores Imunológicos/uso terapêutico , Imunossupressores/uso terapêutico , Interferon beta/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Natalizumab/uso terapêutico , Nitrilas , Rituximab/uso terapêutico , Toluidinas/uso terapêutico
8.
J Autoimmun ; 82: 62-73, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28549776

RESUMO

OBJECTIVE: To identify novel genetic and epigenetic factors associated with Myasthenia gravis (MG) using an identical twins experimental study design. METHODS: The transcriptome and methylome of peripheral monocytes were compared between monozygotic (MZ) twins discordant and concordant for MG, as well as with MG singletons and healthy controls, all females. Sets of differentially expressed genes and differentially methylated CpGs were validated using RT-PCR for expression and target bisulfite sequencing for methylation on additional samples. RESULTS: >100 differentially expressed genes and ∼1800 differentially methylated CpGs were detected in peripheral monocytes between MG patients and controls. Several transcripts associated with immune homeostasis and inflammation resolution were reduced in MG patients. Only a relatively few genes differed between the discordant healthy and MG co-twins, and both their expression and methylation profiles demonstrated very high similarity. INTERPRETATION: This is the first study to characterize the DNA methylation profile in MG, and the expression profile of immune cells in MZ twins with MG. Results suggest that numerous small changes in gene expression or methylation might together contribute to disease. Impaired monocyte function in MG and decreased expression of genes associated with inflammation resolution could contribute to the chronicity of the disease. Findings may serve as potential new predictive biomarkers for disease and disease activity, as well as potential future targets for therapy development. The high similarity between the healthy and the MG discordant twins, suggests that a molecular signature might precede a clinical phenotype, and that genetic predisposition may have a stronger contribution to disease than previously assumed.


Assuntos
Metilação de DNA , Miastenia Gravis/genética , Transcriptoma , Gêmeos Monozigóticos , Adulto , Idoso , Estudos de Casos e Controles , Ilhas de CpG , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Pessoa de Meia-Idade , Monócitos/imunologia , Monócitos/metabolismo , Miastenia Gravis/metabolismo , Transdução de Sinais , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo , Adulto Jovem
9.
Curr Opin Neurol ; 29(3): 354-61, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27070218

RESUMO

PURPOSE OF REVIEW: In recent years we notice paradigm shifts in the understanding of multiple sclerosis (MS), leading to important transition in the patients' management. This review discusses some of the recent findings and developments underlying the conceptual changes being translated from 'treating the disease' to 'treating the patient' with MS (PwMS). RECENT FINDINGS: Applying advanced technologies combined with cross-disciplinary efforts in the fields of neuropathology, neuroimmunology, neurobiology, and neuroimaging, together with clinical neurology provided support for the notion that MS is not a single disease but rather a spectrum. Predictive markers of disease subtypes, disease activity and response to therapy are being developed; some already applied to practice, allowing informed management. In parallel, population-specific issues, some genetic-driven, others caused by environmental (sun-exposure, life-style, etc.), gender-related (hormones) and epigenetic factors, are being elucidated. Additionally, patient empowerment-based approaches, including integration of patient-reported outcome measures (PRO) as well as tools to enhance patients' adherence to medications, are being developed, some already provided as part of emerging mobile-health technologies. SUMMARY: Developments in the MS field, elucidating disease subtypes and interpopulation diversities, together with integration of patient-centered approaches, allow transition toward precision medicine in MS clinical trials and patient care.


Assuntos
Fatores Imunológicos/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Gerenciamento Clínico , Humanos , Esclerose Múltipla/etiologia , Medicina de Precisão
10.
J Autoimmun ; 70: 40-51, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27055778

RESUMO

Fingolimod, an oral therapeutic agent approved for patients with relapsing-remitting Multiple Sclerosis (MS), has been shown to prevent lymphocyte egress from secondary lymphoid tissues; however the specific drug effect on B cells in fingolimod-treated patients remains to be fully elucidated. We present here a comprehensive analysis on the proportions of B cell subsets in the periphery, and the levels of activation, functional surface markers and cytokine profile of B cells in MS patients, following initiation of fingolimod therapy, using flow cytometry and cytokine bead array. Fingolimod therapy increased the ratio of naïve to memory cells, elevated the percentage of plasma cells and highly increased the proportion of transitional B cells as well as additional regulatory subsets, including: IL10(+), CD25(+) and CD5(+) B cells. The percentage of activated CD69(+) cells was highly elevated in the remaining circulating B cells, which produced increased levels of IL10, TGFß, IL6, IL4, LTα, TNFα and IFNγ cytokines, with an overall increased ratio of TGFß to pro-inflammatory cytokines. Furthermore, fingolimod therapy reduced ICAM-1(+) cells, suggesting a possible reduction in antigen-presenting capacity. Phosphorylated-fingolimod was shown in vitro to reduce S1PR1 RNA and protein, to slightly increase viability and to activate anti-apoptotic Bcl2 in transformed B cells of patients with MS. In conclusion, fingolimod therapy modulates significantly the composition of circulating B cells, promoting regulatory subsets and an anti-inflammatory cytokine repertoire.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Interleucina-10/biossíntese , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Adulto , Apresentação de Antígeno/imunologia , Subpopulações de Linfócitos B/citologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Citocinas/metabolismo , Feminino , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Expressão Gênica , Humanos , Memória Imunológica , Imunofenotipagem , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Mediadores da Inflamação/metabolismo , Contagem de Leucócitos , Ativação Linfocitária , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/tratamento farmacológico , Plasmócitos/imunologia , Plasmócitos/metabolismo , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Receptores de Esfingosina-1-Fosfato , Adulto Jovem
11.
Epigenetics ; 10(10): 943-57, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26291385

RESUMO

The relationship between DNA methylation and gene expression is complex and elusive. To further elucidate these relations, we performed an integrative analysis of the methylome and transcriptome of 4 circulating immune cell subsets (B cells, monocytes, CD4(+), and CD8(+) T cells) from healthy females. Additionally, in light of the known sex bias in the prevalence of several immune-mediated diseases, the female datasets were compared with similar public available male data sets. Immune cell-specific differentially methylated regions (DMRs) were found to be highly similar between sexes, with an average correlation coefficient of 0.82; however, numerous sex-specific DMRs, shared by the cell subsets, were identified, mainly on autosomal chromosomes. This provides a list of highly interesting candidate genes to be studied in disorders with sexual dimorphism, such as autoimmune diseases. Immune cell-specific DMRs were mainly located in the gene body and intergenic region, distant from CpG islands but overlapping with enhancer elements, indicating that distal regulatory elements are important in immune cell specificity. In contrast, sex-specific DMRs were overrepresented in CpG islands, suggesting that the epigenetic regulatory mechanisms of sex and immune cell specificity may differ. Both positive and, more frequently, negative correlations between subset-specific expression and methylation were observed, and cell-specific DMRs of both interactions were associated with similar biological pathways, while sex-specific DMRs were linked to networks of early development or estrogen receptor and immune-related molecules. Our findings of immune cell- and sex-specific methylome and transcriptome profiles provide novel insight on their complex regulatory interactions and may particularly contribute to research of immune-mediated diseases.


Assuntos
Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Metilação de DNA/genética , Monócitos/metabolismo , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Ilhas de CpG/genética , Ilhas de CpG/imunologia , Metilação de DNA/imunologia , Epigênese Genética , Feminino , Genoma Humano , Humanos , Masculino , Monócitos/imunologia , Caracteres Sexuais , Transcriptoma/genética , Transcriptoma/imunologia
12.
BMC Neurol ; 13: 60, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23767916

RESUMO

BACKGROUND: Flu-like symptoms (FLS) are common side effects of interferon beta (IFN-ß) treatment in patients with Multiple Sclerosis (PwMS) and are associated with post-injection cytokine surge. We hypothesized that vitamin D3 supplementation would ameliorate FLS by decreasing related serum cytokines' levels. METHODS: In a randomized, double blind study of 45 IFNß-treated PwMS, 21 patients were assigned to 800 IU of vitamin D3 per day (low dose), while 24 patients received 4,370 IU per day (high dose) for one year. FLS were assessed monthly by telephonic interviews. Serum levels of 25-hydroxy-D (25-OH-D), calcium, PTH, IL-17, IL-10 and IFN-γ were measured periodically. EDSS, relapses, adverse events and quality of life (QoL) were documented. RESULTS: 25-OH-D levels increased to a significantly higher levels and PTH levels decreased in the high dose group. There was no significant change in FLS. IL-17 levels were significantly increased in the low dose group, while patients receiving high dose vitamin D had a heterogeneous IL-17 response. No significant differences in relapse rate, EDSS, QoL, serum IL-10 and IFNγ were found. Hypercalcemia or other potential major adverse events were not observed. CONCLUSION: Vitamin D supplementation to IFN-ß treated PwMS, at the doses used, seems safe and associated with dose-dependent changes in IL-17 serum levels, while not affecting IFN-ß related FLS. TRIAL REGISTRATION: ClinicalTrials.gov ID: NCT01005095.


Assuntos
Colecalciferol/farmacologia , Citocinas/sangue , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Interferon beta/efeitos adversos , Esclerose Múltipla Recidivante-Remitente , Adulto , Idoso , Colecalciferol/administração & dosagem , Colecalciferol/efeitos adversos , Método Duplo-Cego , Feminino , Humanos , Interleucina-17/sangue , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/sangue , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/fisiopatologia , Resultado do Tratamento
13.
Brain Behav Immun ; 32: 180-5, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23665342

RESUMO

BACKGROUND: Multiple sclerosis (MS) incidence is higher in geographic regions with less sunlight exposure. Both vitamin D and melatonin are essential mediators of the effect of sunlight in health, and as such are candidates to play a key role in MS. We hypothesized that vitamin D and melatonin may have related influences in patients with MS. METHODS: In a randomized, double blind study of 40 IFN-ß treated MS patients, 21 patients were assigned to 800 IU of vitamin D3 per day (low dose), while 19 patients received 4,370 IU vitamin D3 per day (high dose) for one year. Serum 25-hydroxy-vitamin-D (25-OH-D) and nighttime urine melatonin metabolite, 6-sulphatoxy-melatonin (6-SMT), were measured at baseline, 3 months and 1 year from enrolment. RESULTS: After 3 months supplementation, 25-OH-D levels increased and nighttime melatonin secretion decreased significantly in the high dose group, but not in the low dose group. After 1 year, a decrease in 25-OH-D levels, accompanied by an increase of urine nighttime 6-SMT were observed in the high dose group. Percent change in serum 25-OH-D was significantly and negatively correlated with percent change in urine 6-SMT after 3 months and between 3 months to 1 year. 25-OH-D levels by the end of the study were significantly and negatively correlated to BMI. CONCLUSIONS: Melatonin secretion is negatively correlated with alterations in serum 25-OH-D in IFN-ß treated patients with MS. The finding suggests that melatonin should be considered as a potential mediator of vitamin D neuro-immunomodulatory effects in patients with MS.


Assuntos
Melatonina/metabolismo , Esclerose Múltipla Recidivante-Remitente/metabolismo , Vitamina D/uso terapêutico , Vitaminas/uso terapêutico , Adulto , Colecalciferol/sangue , Interpretação Estatística de Dados , Depressão/psicologia , Suplementos Nutricionais , Feminino , Humanos , Hidroxicolecalciferóis/sangue , Masculino , Melatonina/análogos & derivados , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/psicologia , Vitamina D/administração & dosagem , Vitaminas/administração & dosagem
14.
PLoS One ; 8(4): e62366, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23626809

RESUMO

BACKGROUND: Monocytes, which are key players in innate immunity, are outnumbered by neutrophils and lymphocytes among peripheral white blood cells. The cytokine interferon-ß (IFN-ß) is widely used as an immunomodulatory drug for multiple sclerosis and its functional pathways in peripheral blood mononuclear cells (PBMCs) have been previously described. The aim of the present study was to identify novel, cell-specific IFN-ß functions and pathways in tumor necrosis factor (TNF)-α-activated monocytes that may have been missed in studies using PBMCs. METHODOLOGY/PRINCIPAL FINDINGS: Whole genome gene expression profiles of human monocytes and T cells were compared following in vitro priming to TNF-α and overnight exposure to IFN-ß. Statistical analyses of the gene expression data revealed a cell-type-specific change of 699 transcripts, 667 monocyte-specific transcripts, 21 T cell-specific transcripts and 11 transcripts with either a difference in the response direction or a difference in the magnitude of response. RT-PCR revealed a set of differentially expressed genes (DEGs), exhibiting responses to IFN-ß that are modulated by TNF-α in monocytes, such as RIPK2 and CD83, but not in T cells or PBMCs. Known IFN-ß promoter response elements, such as ISRE, were enriched in T cell DEGs but not in monocyte DEGs. The overall directionality of the gene expression regulation by IFN-ß was different in T cells and monocytes, with up-regulation more prevalent in T cells, and a similar extent of up and down-regulation recorded in monocytes. CONCLUSIONS: By focusing on the response of distinct cell types and by evaluating the combined effects of two cytokines with pro and anti-inflammatory activities, we were able to present two new findings First, new IFN-ß response pathways and genes, some of which were monocytes specific; second, a cell-specific modulation of the IFN-ß response transcriptome by TNF-α.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon beta/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , ADP-Ribosil Ciclase 1/genética , ADP-Ribosil Ciclase 1/metabolismo , Análise por Conglomerados , Redes Reguladoras de Genes , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Especificidade de Órgãos , Reprodutibilidade dos Testes , Transdução de Sinais , Transcrição Gênica , Fator de Necrose Tumoral alfa/farmacologia
15.
J Neuroimmunol ; 251(1-2): 45-54, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22846497

RESUMO

Laquinimod is an orally administered drug under development for the treatment of Multiple Sclerosis (MS), lacking a fully elucidated mode of action. We assessed the immunomodulatory effects of laquinimod in vitro on human B cells from healthy or MS patients, cultured alone or with CD4(+) T cells. Laquinimod modulated B cell markers, mainly by increasing the regulatory ones CD25, IL10 and CD86, and decreased IL4, while increasing IL10 and TGFß in both B and T cells, in a B cell-mediated manner. These findings shed additional light on the mechanisms underlying the effects of laquinimod in MS and potentially other immune-mediated diseases.


Assuntos
Linfócitos B/efeitos dos fármacos , Fatores Imunológicos/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Quinolonas/uso terapêutico , Adulto , Antígeno B7-2/biossíntese , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Interleucina-10/biossíntese , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Interleucina-4/biossíntese , Masculino , Pessoa de Meia-Idade , Linfócitos T/efeitos dos fármacos , Fator de Crescimento Transformador beta/biossíntese , Adulto Jovem
16.
J Neuroimmunol ; 232(1-2): 200-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21112096

RESUMO

We investigated the modulation of Cathepsin S, Cathepsin B and Cystatin C expression in immune cells by interferon (IFN)-ß, and their role in cell migration. Cathepsin levels were increased in monocytic and T line cells upon activation. IFN-ß abolished this increase of Cathepsin B in monocytes and of Cathepsin S in T cells, while increased Cystatin C. Neutralizing Cystatin C increased cell migration, while migration of monocytes and T cells was inhibited by IFN-ß, by Cystatin C, and T cell migration was suppressed by Cathepsin S and B inhibitors. These findings support further studies in the importance of Cathepsins and Cystatins in immunomodulation.


Assuntos
Catepsina B/metabolismo , Catepsinas/metabolismo , Movimento Celular , Quimiotaxia de Leucócito/fisiologia , Cistatina C/metabolismo , Interferon beta/metabolismo , Western Blotting , Catepsina B/imunologia , Catepsinas/imunologia , Cistatina C/imunologia , Humanos , Interferon beta/imunologia , Células Jurkat , Monócitos/citologia , Monócitos/imunologia , Monócitos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células U937
17.
Reprod Biol Endocrinol ; 7: 135, 2009 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19939245

RESUMO

BACKGROUND: We have previously shown that Matrix metalloproteinase (MMP) -2 is a key-enzyme in early trophoblast invasion and that Protein Kinase A (PKA) increases MMP-2 expression and trophoblast invasion. The aim of this study was to examine MMP -2 regulation by PKA in invasive trophoblasts: JAR choriocarcinoma cell-line and 6-8 w first trimester trophoblasts. METHODS: The effect of Forskolin (PKA) on MMP-2 expression was assessed by Northern Blot and RT-PCR. Possible transcription factors binding to consensus MMP-2 promoter sequences in response to Forskolin, were detected by EMSA binding assay and their expression assessed by western blot analysis. Antisense transfection of relevant transcription factors was performed and the inhibitory effect assessed on MMP-2 expression (RT-PCR), secretion (zymography) and trophoblast invasiveness (transwell migration assay). RESULTS: We found that Forskolin increased MMP-2 mRNA in JAR cells within 24 hours, and induced binding to p53, Ets, C/EBP and AP-2. Transcription factors Ets-2, phospho- p53, C/EBP epsilon, C/EBP lambda and AP-2 alpha bound to their respective binding sequences in response to Forskolin and the expressions of these transcription factors were all elevated in Forskolin- treated cells. Inhibition of Ets-2 and p53 reduced MMP-2 expression, secretion and invasiveness of Forskolin treated cells. CONCLUSION: MMP-2 is regulated by PKA through several binding sites and transcription factors including Ets-2, p53, C/EBP, C/EBP lambda and AP-2 alpha. Ets-2 and p53 mediate cAMP- induced trophoblast invasiveness, through regulation of MMP-2.


Assuntos
AMP Cíclico/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Proteína Proto-Oncogênica c-ets-2/fisiologia , Trofoblastos/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia , Sítios de Ligação/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Colforsina/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteína Proto-Oncogênica c-ets-2/metabolismo , Trofoblastos/metabolismo , Trofoblastos/fisiologia , Proteína Supressora de Tumor p53/metabolismo
18.
Reprod Biol Endocrinol ; 3: 56, 2005 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16236179

RESUMO

The implantation process involves complex and synchronized molecular and cellular events between the uterus and the implanting embryo. These events are regulated by paracrine and autocrine factors. Trophoblast invasion and migration through the uterine wall is mediated by molecular and cellular interactions, controlled by the trophoblast and the maternal microenvironment. This review is focused on the molecular constituents of the human trophoblast, their actions and interactions, including interrelations with the uterine endometrium.


Assuntos
Implantação do Embrião/fisiologia , Trofoblastos/fisiologia , Animais , Moléculas de Adesão Celular/fisiologia , Gonadotropina Coriônica/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Fator de Crescimento Epidérmico/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Feminino , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Integrinas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-1/fisiologia , Interleucina-6/fisiologia , Fator Inibidor de Leucemia , Metaloproteinases da Matriz/fisiologia , Mucina-1/fisiologia , Progesterona/fisiologia , Prostaglandinas/fisiologia , Serina Endopeptidases/fisiologia , Inibidores Teciduais de Metaloproteinases/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
19.
Reprod Biol Endocrinol ; 2: 59, 2004 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-15294019

RESUMO

BACKGROUND: The aim of this study was to examine the invasiveness of first trimester trophoblasts according to the secretion profile of MMP-2 and -9 at different gestational stages, and to test the similarity between primary trophoblast cell-culture and the JAR choriocarcinoma cell-line. METHODS: First trimester trophoblasts were divided into two groups: 6-8 weeks (early) and 9-12 w (late) of gestation. The two trophoblast groups and JAR cells were cultured in medium, with various concentrations of forskolin and Epidermal Growth Factor (EGF). Proteolytic activity was detected by zymography and invasiveness was assessed by Matrigel invasion assay. Student's T-test was used for statistical analysis. RESULTS: In 6-8 w trophoblast, proMMP-2 was only slightly dominant over proMMP-9 (53.2% vs. 46.8% respectively), whereas in 9-12 w, proMMP-9 was clearly dominant over proMMP-2 (61.7% vs.38.3% respectively). In JAR cells proMMP-2 was strongly dominant (90.2% vs.9.8% respectively). In JAR cells forskolin significantly increased proMMP-2 and -9 secretion (128.5% +/- 12 and 183.2% +/- 27.9 of control, respectively). EGF had a dual effect on JAR cells: at 8 ng/ml both proMMP-2 and -9 were increased (133.5% +/-15 and 223.9% +/- 32.4 of control, respectively) while at 80 ng/ml both proMMP-2 and -9 were decreased (65.1% +/- 18.3 and 66.6% +/- 37 of control, respectively). Forskolin significantly increased both proMMP-2 and -9 secretion in 6-8 w and 9-12 w trophoblasts (125.9% +/- 6.3,128.4% +/- 6.4; 169.7% +/- 20.3, 120.3% +/- 4.5 of control, respectively). EGF also significantly increased both proMMP-2 and -9 secretion in 6-8 w and 9-12 w trophoblasts (141.22% +/- 14.8, 138.8% +/- 10.3; 168.3% +/- 18.2, 117.3 +/- 3.8 of control, respectively). Both forskolin and EGF increased trophoblast cells invasiveness in all groups. The invasive ability of trophoblast cells, induced by forskolin, was reduced by MMP-2 antibody in: JAR cells, 6-8 w and 9-12 w trophoblasts. Likewise trophoblast invasion induced by EGF was reduced by MMP-2 antibody in all groups. However the invasive ability induced by forskolin or EGF was inhibited by MMP-9 antibody only in trophoblasts from 9-12 w. CONCLUSIONS: First trimester trophoblasts express differential gelatinase secretion profile according to the gestational week. In JAR and early trophoblasts (6-8 w) MMP-2 is the main gelatinase and the key enzyme in trophoblast invasion. Thereafter in late first trimester trophoblasts (9-12 w), both MMP-2 and -9 participate in trophoblast invasion.


Assuntos
Regulação Enzimológica da Expressão Gênica/genética , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Trofoblastos/enzimologia , Trofoblastos/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Coriocarcinoma/enzimologia , Coriocarcinoma/genética , Coriocarcinoma/metabolismo , Coriocarcinoma/patologia , Colforsina/administração & dosagem , Relação Dose-Resposta a Droga , Fator de Crescimento Epidérmico/administração & dosagem , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Placenta/enzimologia , Placenta/fisiologia , Gravidez , Primeiro Trimestre da Gravidez/genética , Primeiro Trimestre da Gravidez/fisiologia , Trofoblastos/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA