Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
J Biomed Inform ; 148: 104546, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37984546

ABSTRACT

OBJECTIVE: Computational models are at the forefront of the pursuit of personalized medicine thanks to their descriptive and predictive abilities. In the presence of complex and heterogeneous data, patient stratification is a prerequisite for effective precision medicine, since disease development is often driven by individual variability and unpredictable environmental events. Herein, we present GreatNectorworkflow as a valuable tool for (i) the analysis and clustering of patient-derived longitudinal data, and (ii) the simulation of the resulting model of patient-specific disease dynamics. METHODS: GreatNectoris designed by combining an analytic strategy composed of CONNECTOR, a data-driven framework for the inspection of longitudinal data, and an unsupervised methodology to stratify the subjects with GreatMod, a quantitative modeling framework based on the Petri Net formalism and its generalizations. RESULTS: To illustrate GreatNectorcapabilities, we exploited longitudinal data of four immune cell populations collected from Multiple Sclerosis patients. Our main results report that the T-cell dynamics after alemtuzumab treatment separate non-responders versus responders patients, and the patients in the non-responders group are characterized by an increase of the Th17 concentration around 36 months. CONCLUSION: GreatNectoranalysis was able to stratify individual patients into three model meta-patients whose dynamics suggested insight into patient-tailored interventions.


Subject(s)
Precision Medicine , Humans , Workflow , Computer Simulation , Precision Medicine/methods , Cluster Analysis
2.
Crit Rev Immunol ; 40(3): 249-253, 2020.
Article in English | MEDLINE | ID: mdl-33389888

ABSTRACT

The contribution of Eli E. Sercarz to immunology and immunopathology has been remarkable and achieved many milestones in the understanding of the processes of the mechanisms fine-tuning immune responses. A part of his work was dedicated to the study of the deep complexity of the lymphocyte T cell repertoire and its importance during the physiologic development and disease, such as clonal heterogeneity of T cell responses. Starting from these studies, under his mentoring, we had the opportunity to implement the spectratyping method and apply it to human and experimental autoimmune diseases, obtaining intriguing results. The open question of this brief review is the possible role of this fine and complex technique, the immunoscope analysis, in the era of the big data and omics.


Subject(s)
Genes, T-Cell Receptor/genetics , Immunophenotyping/methods , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Allergy and Immunology/history , Allergy and Immunology/trends , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , High-Throughput Nucleotide Sequencing , History, 20th Century , Humans , Immunophenotyping/history , Immunophenotyping/trends , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/immunology , Polymorphism, Genetic , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , V(D)J Recombination
3.
BMC Bioinformatics ; 21(Suppl 17): 550, 2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33308135

ABSTRACT

BACKGROUND: Multiple Sclerosis (MS) represents nowadays in Europe the leading cause of non-traumatic disabilities in young adults, with more than 700,000 EU cases. Although huge strides have been made over the years, MS etiology remains partially unknown. Furthermore, the presence of various endogenous and exogenous factors can greatly influence the immune response of different individuals, making it difficult to study and understand the disease. This becomes more evident in a personalized-fashion when medical doctors have to choose the best therapy for patient well-being. In this optics, the use of stochastic models, capable of taking into consideration all the fluctuations due to unknown factors and individual variability, is highly advisable. RESULTS: We propose a new model to study the immune response in relapsing remitting MS (RRMS), the most common form of MS that is characterized by alternate episodes of symptom exacerbation (relapses) with periods of disease stability (remission). In this new model, both the peripheral lymph node/blood vessel and the central nervous system are explicitly represented. The model was created and analysed using Epimod, our recently developed general framework for modeling complex biological systems. Then the effectiveness of our model was shown by modeling the complex immunological mechanisms characterizing RRMS during its course and under the DAC administration. CONCLUSIONS: Simulation results have proven the ability of the model to reproduce in silico the immune T cell balance characterizing RRMS course and the DAC effects. Furthermore, they confirmed the importance of a timely intervention on the disease course.


Subject(s)
Immune System/physiology , Models, Biological , Multiple Sclerosis, Relapsing-Remitting/immunology , User-Computer Interface , Algorithms , Daclizumab/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/pathology , Stochastic Processes
4.
Int J Mol Sci ; 18(5)2017 Apr 29.
Article in English | MEDLINE | ID: mdl-28468254

ABSTRACT

Natalizumab is a monoclonal antibody highly effective in the treatment of relapsing remitting multiple sclerosis (RRMS) patients. Despite its effectiveness, there are growing concerns regarding the risk of progressive multifocal leukoencephalopathy (PML), a brain infection caused by John Cunningham virus (JCV), particularly after 24 doses and in patients who previously received immunosuppressive drugs. Long-term natalizumab treated, immunosuppressive-pretreated, and JCV antibody-positive patients are asked to rediscuss natalizumab continuation or withdrawal after 24 doses. Until now, there has not been a clear strategy that should be followed to avoid PML risk and in parallel reduce clinical and radiological rebound activity. In this review, we analyzed the results of clinical trials and case reports in relation to the following situations: natalizumab continuation, natalizumab discontinuation followed by full therapeutic suspension or switch to other first or second line MS treatments. Quitting all MS treatment after natalizumab increases MS activity occurrence. The results regarding the therapeutic switch are not homogeneous, so at the moment there are no established guidelines regarding natalizumab treatment after 24 administrations; the choice is currently based on the professional experience of the neurologist, and on patients' clinical features and preferences.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunologic Factors/therapeutic use , Multiple Sclerosis/drug therapy , Natalizumab/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , JC Virus/isolation & purification , Leukoencephalopathy, Progressive Multifocal/diagnosis , Leukoencephalopathy, Progressive Multifocal/etiology , Natalizumab/administration & dosage , Natalizumab/adverse effects
5.
Clin Sci (Lond) ; 130(3): 193-203, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26558403

ABSTRACT

The mechanisms responsible for the evolution of steatosis towards NASH (non-alcoholic steatohepatitis) and fibrosis are not completely defined. In the present study we evaluated the role of CD4(+) T-helper (Th) cells in this process. We analysed the infiltration of different subsets of CD4(+) Th cells in C57BL/6 mice fed on a MCD (methionine choline-deficient) diet, which is a model reproducing all phases of human NASH progression. There was an increase in Th17 cells at the beginning of NASH development and at the NASH-fibrosis transition, whereas levels of Th22 cells peaked between the first and the second expansion of Th17 cells. An increase in the production of IL (interleukin)-6, TNFα (tumour necrosis factor α), TGFß (transforming growth factor ß) and CCL20 (CC chemokine ligand 20) accompanied the changes in Th17/Th22 cells. Livers of IL-17(-/-) mice were protected from NASH development and characterized by an extensive infiltration of Th22 cells. In vitro, IL-17 exacerbated the JNK (c-Jun N-terminal kinase)-dependent mouse hepatocyte lipotoxicity induced by palmitate. IL-22 prevented lipotoxicity through PI3K (phosphoinositide 3-kinase)-mediated inhibition of JNK, but did not play a protective role in the presence of IL-17, which up-regulated the PI3K/Akt inhibitor PTEN (phosphatase and tensin homologue deleted on chromosome 10). Consistently, livers of IL-17(-/-) mice fed on the MCD diet displayed decreased activation of JNK, reduced expression of PTEN and increased phosphorylation of Akt compared with livers of wild-type mice. Hepatic infiltration of Th17 cells is critical for NASH initiation and development of fibrosis in mice, and reflects an infiltration of Th22 cells. Th22 cells are protective in NASH, but only in the absence of IL-17. These data strongly support the potentiality of clinical applications of IL-17 inhibitors that can prevent NASH by both abolishing the lipotoxic action of IL-17 and allowing IL-22-mediated protection.


Subject(s)
Non-alcoholic Fatty Liver Disease/immunology , T-Lymphocytes, Helper-Inducer/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Disease Models, Animal , Interleukin-17/deficiency , Interleukins/metabolism , Liver/immunology , Male , Mice, Inbred C57BL , Interleukin-22
6.
Clin Sci (Lond) ; 130(4): 247-58, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26590104

ABSTRACT

PDAC (pancreatic ductal adenocarcinoma) is the fifth leading cause of cancer-related death. The causes of this cancer remain unknown, but increasing evidence indicates a key role of the host immune response and cytokines in human carcinogenesis. Intra-tumoral IL (interleukin)-22 levels have been shown to be elevated in PDAC patients. However, little is known regarding the expression and clinical relevance of Th22 cells in human PDAC and, furthermore, which TILs (tumour-infiltrating lymphocytes) are the main producers of IL-22 is unknown. In the present study, we characterized the functional proprieties of the different subsets of IL-22-producing TILs and analysed their relationship with the TNM staging system and patient survival. We have demonstrated for the first time that, in PDAC patients, the T-cells co-producing IFN-γ (interferon γ) and exerting perforin-mediated cytotoxicity are the major intra-tumoral source of IL-22. In addition, isolated Th22 cells were able to induce apoptosis, which was antagonized by IL-22. Finally, we observed that the IL-22-producing T-cells were significantly increased in tumour tissue and that this increase was positively correlated with TNM staging of PDAC and poorer patient survival. These novel findings support the dual role of the anti-tumour immune system and that IL-22-producing cells may participate in PDAC pathogenesis. Therefore monitoring Th22 levels could be a good diagnostic parameter, and blocking IL-22 signalling may represent a viable method for anti-PDAC therapies.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Interferon-gamma/metabolism , Interleukins/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Pancreatic Neoplasms/metabolism , Paracrine Communication , T-Lymphocytes, Helper-Inducer/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , Female , Granzymes/metabolism , Humans , Interferon-gamma/immunology , Interleukins/immunology , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Perforin/metabolism , Phenotype , Signal Transduction , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Interleukin-22
7.
J Immunol ; 191(11): 5451-9, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24154625

ABSTRACT

Hemopexin (Hx) is an acute-phase protein synthesized by hepatocytes in response to the proinflammatory cytokines IL-6, IL-1ß, and TNF-α. Hx is the plasma protein with the highest binding affinity to heme and controls heme-iron availability in tissues and also in T lymphocytes, where it modulates their responsiveness to IFN-γ. Recent data have questioned regarding an anti-inflammatory role of Hx, a role that may be both heme-binding dependent and independent. The aim of this study was to investigate the role of Hx in the development of a T cell-mediated inflammatory autoimmune response. During experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis, Hx content in serum increased and remained high. When EAE was induced in Hx knockout (Hx(-/-)) mice, they developed a clinically earlier and exacerbated EAE compared with wild-type mice, associated to a higher amount of CD4(+)-infiltrating T cells. The severe EAE developed by Hx(-/-) mice could be ascribed to an enhanced expansion of Th17 cells accounting for both a higher disposition of naive T cells to differentiate toward the Th17 lineage and a higher production of Th17 differentiating cytokines IL-6 and IL-23 by APCs. When purified human Hx was injected in Hx(-/-) mice before EAE induction, Th17 expansion, as well as disease severity, were comparable with those of wild-type mice. Taken together, these data indicate that Hx has a negative regulatory role in Th17-mediated inflammation and prospect its pharmacological use to limit the expansion of this cell subset in inflammatory and autoimmune disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Hemopexin/metabolism , Hepatocytes/immunology , Multiple Sclerosis/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Hemopexin/genetics , Hemopexin/immunology , Humans , Immunity, Cellular/genetics , Immunosuppression Therapy , Inflammation Mediators/immunology , Male , Mice , Mice, 129 Strain , Mice, Knockout , Multiple Sclerosis/genetics
8.
Gastroenterology ; 144(5): 1098-106, 2013 May.
Article in English | MEDLINE | ID: mdl-23333712

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDA) is an aggressive tumor, and patients typically present with late-stage disease; rates of 5-year survival after pancreaticoduodenectomy are low. Antibodies against α-enolase (ENO1), a glycolytic enzyme, are detected in more than 60% of patients with PDA, and ENO1-specific T cells inhibit the growth of human pancreatic xenograft tumors in mice. We investigated whether an ENO1 DNA vaccine elicits antitumor immune responses and prolongs survival of mice that spontaneously develop autochthonous, lethal pancreatic carcinomas. METHODS: We injected and electroporated a plasmid encoding ENO1 (or a control plasmid) into Kras(G12D)/Cre (KC) mice and Kras(G12D)/Trp53(R172H)/Cre (KPC) mice at 4 weeks of age (when pancreatic intraepithelial lesions are histologically evident). Antitumor humoral and cellular responses were analyzed by histology, immunohistochemistry, enzyme-linked immunosorbent assays, flow cytometry, and enzyme-linked immunosorbent spot and cytotoxicity assays. Survival was analyzed by Kaplan-Meier analysis. RESULTS: The ENO1 vaccine induced antibody and a cellular response and increased survival times by a median of 138 days in KC mice and 42 days in KPC mice compared with mice given the control vector. On histologic analysis, the vaccine appeared to slow tumor progression. The vaccinated mice had increased serum levels of anti-ENO1 immunoglobulin G, which bound the surface of carcinoma cells and induced complement-dependent cytotoxicity. ENO1 vaccination reduced numbers of myeloid-derived suppressor cells and T-regulatory cells and increased T-helper 1 and 17 responses. CONCLUSIONS: In a genetic model of pancreatic carcinoma, vaccination with ENO1 DNA elicits humoral and cellular immune responses against tumors, delays tumor progression, and significantly extends survival. This vaccination strategy might be developed as a neoadjuvant therapy for patients with PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Immunity, Cellular/immunology , Neoplasms, Experimental/drug therapy , Pancreatic Neoplasms/drug therapy , Phosphopyruvate Hydratase/immunology , Vaccination/methods , Vaccines, DNA/pharmacology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Mutant Strains , Neoplasms, Experimental/genetics , Neoplasms, Experimental/mortality , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Survival Rate
9.
J Immunol ; 188(3): 1011-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22219326

ABSTRACT

IFN-ß inhibits the expansion of Th17 cells in active multiple sclerosis (AMS), and this might contribute to improve the clinical symptoms. The effectiveness of this inhibition, however, requires intact IFN-γ signaling in T cells. In this study, we report that both mRNA and cell surface expression of the signaling chain of the IFN-γ receptor (IFN-γR2) and its cognate tyrosine kinase JAK2 are enhanced in peripheral blood Th17 cells and clones from patients with AMS compared with those with inactive multiple sclerosis (IMS) or healthy subjects (HS). IFN-γ decreased the frequency of Th17 peripheral cells and proliferation of Th17 clones from AMS patients. Stimulation of PBMCs from HS in Th17-polarizing conditions resulted in the enhancement of JAK2 expression and accumulation of cell surface IFN-γR2. The role of JAK2 in the modulation of IFN-γR2 was demonstrated as its transduction prevented rapid internalization and degradation of IFN-γR2 in JAK2-deficient γ2A cells. In conclusion, these data identify JAK2 as a critical factor that stabilizes IFN-γR2 surface expression in Th17 cells from AMS patients, making them sensitive to IFN-γ. These data may have clinical implications for a better use of IFNs in multiple sclerosis and possibly other inflammatory diseases.


Subject(s)
Janus Kinase 2/metabolism , Multiple Sclerosis/immunology , Receptors, Interferon/metabolism , Th17 Cells/metabolism , Case-Control Studies , Cell Proliferation , Humans , Interferons , Multiple Sclerosis/pathology , RNA, Messenger/analysis , Receptors, Interferon/analysis , Th17 Cells/immunology , Interferon gamma Receptor
10.
Nutrients ; 16(6)2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38542790

ABSTRACT

The Mediterranean Diet (MD) is described in the literature as a beneficial dietary pattern for neurodegenerative diseases such as Multiple Sclerosis (MS). The objective of this study was to evaluate the dietary habits in people with MS (pwMS) and to test whether adherence to the MD could have an impact on the severity of the disease measured as the MS severity score (MSSS). Adherence to the MD was assessed in 31 PwMS using the Mediterranean Diet Adherence Screener (MEDAS), the Pyramid-based Mediterranean Diet Score (PyrMDS) index, and the Italian Mediterranean Index (IMI), and their eating habits were recorded in a food diary for a one-year follow-up. When data obtained from dietary analysis were compared to the MSSS, results showed that pwMS with lower MSSS adhere more to the MD than the other pwMS groups according to the MEDAS index. Furthermore, a high consumption of fiber in the MS mild severity class was observed. Further studies are needed to clarify which of the nutritional components of the MD may impact the course of MS and if the sensitization of pwMS to MD adherence can be a strategy for mitigating the disease.


Subject(s)
Diet, Mediterranean , Multiple Sclerosis , Humans , Feeding Behavior , Italy , Dietary Patterns
11.
Front Immunol ; 14: 1176016, 2023.
Article in English | MEDLINE | ID: mdl-37304278

ABSTRACT

Multiple sclerosis (MS) is a multifactorial neurological disease characterized by chronic inflammation and immune-driven demyelination of the central nervous system (CNS). The rising number of MS cases in the last decade could be partially attributed to environmental changes, among which the alteration of the gut microbiome driven by novel dietary habits is now of particular interest. The intent of this review is to describe how diet can impact the development and course of MS by feeding the gut microbiome. We discuss the role of nutrition and the gut microbiota in MS disease, describing preclinical studies on experimental autoimmune encephalomyelitis (EAE) and clinical studies on dietary interventions in MS, with particular attention to gut metabolites-immune system interactions. Possible tools that target the gut microbiome in MS, such as the use of probiotics, prebiotics and postbiotics, are analyzed as well. Finally, we discuss the open questions and the prospects of these microbiome-targeted therapies for people with MS and for future research.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Gastrointestinal Microbiome , Microbiota , Multiple Sclerosis , Humans , Animals , Multiple Sclerosis/therapy , Central Nervous System
12.
Front Immunol ; 14: 1205879, 2023.
Article in English | MEDLINE | ID: mdl-37409134

ABSTRACT

Background and objectives: Long lasting immune response to anti-SARS-CoV-2 vaccination in people with Multiple Sclerosis (pwMS) is still largely unexplored. Our study aimed at evaluating the persistence of the elicited amount of neutralizing antibodies (Ab), their activity and T cell response after three doses of anti-SARS-CoV-2 vaccine in pwMS. Methods: We performed a prospective observational study in pwMS undergoing SARS-CoV-2 mRNA vaccinations. Anti-Region Binding Domain (anti-RBD) of the spike (S) protein immunoglobulin G (IgG) titers were measured by ELISA. The neutralization efficacy of collected sera was measured by SARS-CoV-2 pseudovirion-based neutralization assay. The frequency of Spike-specific IFNγ-producing CD4+ and CD8+ T cells was measured by stimulating Peripheral Blood Mononuclear Cells (PBMCs) with a pool of peptides covering the complete protein coding sequence of the SARS-CoV-2 S. Results: Blood samples from 70 pwMS (11 untreated pwMS, 11 under dimethyl fumarate, 9 under interferon-γ, 6 under alemtuzumab, 8 under cladribine, 12 under fingolimod and 13 under ocrelizumab) and 24 healthy donors were collected before and up to six months after three vaccine doses. Overall, anti-SARS-CoV-2 mRNA vaccine elicited comparable levels of anti-RBD IgGs, neutralizing activity and anti-S T cell response both in untreated, treated pwMS and HD that last six months after vaccination. An exception was represented by ocrelizumab-treated pwMS that showed reduced levels of IgGs (p<0.0001) and a neutralizing activity under the limit of detection (p<0.001) compared to untreated pwMS. Considering the occurrence of a SARS-CoV-2 infection after vaccination, the Ab neutralizing efficacy (p=0.04), as well as CD4+ (p=0.016) and CD8+ (p=0.04) S-specific T cells, increased in treated COVID+ pwMS compared to uninfected treated pwMS at 6 months after vaccination. Discussion: Our follow-up provides a detailed evaluation of Ab, especially in terms of neutralizing activity, and T cell responses after anti-SARS-CoV-2 vaccination in MS context, over time, considering a wide number of therapies, and eventually breakthrough infection. Altogether, our observations highlight the vaccine response data to current protocols in pwMS and underline the necessity to carefully follow-up anti-CD20- treated patients for higher risk of breakthrough infections. Our study may provide useful information to refine future vaccination strategies in pwMS.


Subject(s)
COVID-19 , Multiple Sclerosis , Humans , Antibodies, Neutralizing , Multiple Sclerosis/drug therapy , COVID-19/prevention & control , Leukocytes, Mononuclear , SARS-CoV-2 , Breakthrough Infections
13.
J Immunol ; 184(11): 6124-32, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20435927

ABSTRACT

Rat (r)Erbb2 transgenic BALB-neuT mice genetically predestined to develop multiple invasive carcinomas allow an assessment of the potential of a vaccine against the stages of cancer progression. Because of rErbb2 expression in the thymus and its overexpression in the mammary gland, CD8(+) T cell clones reacting at high avidity with dominant rErbb2 epitopes are deleted in these mice. In BALB-neuT mice with diffuse and invasive in situ lesions and almost palpable carcinomas, a temporary regulatory T cells depletion combined with anti-rErbb2 vaccine markedly enhanced the anti-rErbb2 Ab response and allowed the expansion of latent pools of low-avidity CD8(+) T cells bearing TCRs repertoire reacting with the rErbb2 dominant peptide. This combination of a higher Ab response and activation of a low-avidity cytotoxic response persistently blocked tumor progression at stages in which the vaccine alone was ineffective. However, when diffuse and invasive microscopic cancers become almost palpable, this combination was no longer able to secure a significant extension of mice survival.


Subject(s)
Cancer Vaccines/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Mammary Neoplasms, Experimental/immunology , Receptor, ErbB-2/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Vaccines, DNA/immunology , Animals , Antibodies/immunology , Cancer Vaccines/genetics , Cancer Vaccines/pharmacology , Cell Separation , Electroporation , Female , Flow Cytometry , Genes, erbB-2/genetics , Genes, erbB-2/immunology , Immunohistochemistry , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, SCID , Mice, Transgenic , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/drug effects , Vaccines, DNA/pharmacology
14.
Mult Scler Relat Disord ; 61: 103776, 2022 May.
Article in English | MEDLINE | ID: mdl-35364386

ABSTRACT

Anti-SARS-CoV2 mRNA vaccines showed a blunted antibody (Ab) response in people with MS (pwMS) on high efficacy therapies, suggesting the need for a booster dose. We evaluated the kinetics of the production of anti-receptor binding domain (RBD) Immunoglobulins G (IgG) after the vaccination cycle and the booster in pwMS receiving ocrelizumab, fingolimod and cladribine. A significant increase of anti-RBD IgG seroconversion was observed after booster respect to the vaccination cycle. Results obtained from this study will be useful for the management of pwMS in relation to their disease modifying therapy (DMT) and for any future vaccination campaign.


Subject(s)
COVID-19 , Multiple Sclerosis , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Immunoglobulin G/therapeutic use , Multiple Sclerosis/drug therapy , SARS-CoV-2
15.
Front Immunol ; 13: 818325, 2022.
Article in English | MEDLINE | ID: mdl-35296069

ABSTRACT

Introduction: Alemtuzumab is highly effective in the treatment of patients with relapsing multiple sclerosis (PwRMS) and selectively targets the CD52 antigen, with a consequent profound lymphopenia, particularly of CD4+ T lymphocytes. However, the immunological basis of its long-term efficacy has not been clearly elucidated. Methods: We followed up 29 alemtuzumab-treated RMS patients over a period of 72 months and studied the immunological reconstitution of their CD4+ T cell subsets by means of phenotypic and functional analysis and through mRNA-related molecule expression, comparing them to healthy subject (HS) values (rate 2:1). Results: In patients receiving only two-course alemtuzumab, the percentage of CD4+ lymphocytes decreased and returned to basal levels only at month 48. Immune reconstitution of the CD4+ subsets was characterized by a significant increase (p < 0.001) in Treg cell percentage at month 24, when compared to baseline, and was accompanied by restoration of the Treg suppressor function that increased within a range from 2- to 6.5-fold compared to baseline and that persisted through to the end of the follow-up. Furthermore, a significant decrease in self-reactive myelin basic protein-specific Th17 (p < 0.0001) and Th1 (p < 0.05) cells reaching HS values was observed starting from month 12. There was a change in mRNA of cytokines, chemokines, and transcriptional factors related to Th17, Th1, and Treg cell subset changes, consequently suggesting a shift toward immunoregulation and a reduction of T cell recruitment to the central nervous system. Conclusions: These data provide further insight into the mechanism that could contribute to the long-term 6-year persistence of the clinical effect of alemtuzumab on RMS disease activity.


Subject(s)
Multiple Sclerosis , Alemtuzumab/pharmacology , Alemtuzumab/therapeutic use , CD4-Positive T-Lymphocytes , Follow-Up Studies , Humans , Multiple Sclerosis/drug therapy , RNA, Messenger/therapeutic use
16.
Genes (Basel) ; 12(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34440354

ABSTRACT

As a complex disease, Multiple Sclerosis (MS)'s etiology is determined by both genetic and environmental factors. In the last decade, the gut microbiome has emerged as an important environmental factor, but its interaction with host genetics is still unknown. In this review, we focus on these dual aspects of MS pathogenesis: we describe the current knowledge on genetic factors related to MS, based on genome-wide association studies, and then illustrate the interactions between the immune system, gut microbiome and central nervous system in MS, summarizing the evidence available from Experimental Autoimmune Encephalomyelitis mouse models and studies in patients. Finally, as the understanding of influence of host genetics on the gut microbiome composition in MS is in its infancy, we explore this issue based on the evidence currently available from other autoimmune diseases that share with MS the interplay of genetic with environmental factors (Inflammatory Bowel Disease, Rheumatoid Arthritis and Systemic Lupus Erythematosus), and discuss avenues for future research.


Subject(s)
Gastrointestinal Microbiome , Multiple Sclerosis/genetics , Multiple Sclerosis/microbiology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Genome-Wide Association Study , Humans , Mice
17.
Front Immunol ; 12: 637974, 2021.
Article in English | MEDLINE | ID: mdl-33767708

ABSTRACT

Tobacco smoking is known to impact circulating levels of major immune cells populations, but its effect on specific immune cell subsets remains poorly understood. Here, using high-resolution data from 223 healthy women (25 current and 198 never smokers), we investigated the association between smoking status and 35,651 immune traits capturing immune cell subset frequencies. Our results confirmed that active tobacco smoking is associated with increased frequencies of circulating CD8+ T cells expressing the CD25 activation marker. Moreover, we identified novel associations between smoking status and relative abundances of CD8+ CD25+ memory T cells, CD8+ memory T cells expressing the CCR4 chemokine receptor, and CD4+CD8+ (double-positive) CD25+ T cells. We also observed, in current smokers, a decrease in the relative frequencies of CD4+ T cells expressing the CD38 activation marker and an increase in class-switched memory B cell isotypes IgA, IgG, and IgE. Finally, using data from 135 former female smokers, we showed that the relative frequencies of immune traits associated with active smoking are usually completely restored after smoking cessation, with the exception of subsets of CD8+ and CD8+ memory T cells, which persist partially altered. Our results are consistent with previous findings and provide further evidence on how tobacco smoking shapes leukocyte cell subsets proportion toward chronic inflammation.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Subsets/immunology , Monocytes/immunology , Smoking/adverse effects , Adult , Aged , CD4 Lymphocyte Count , Female , Humans , Immunologic Memory/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Middle Aged , Smoking/immunology , Smoking Cessation
18.
Cells ; 9(6)2020 06 03.
Article in English | MEDLINE | ID: mdl-32503344

ABSTRACT

Alemtuzumab is a monoclonal antibody that binds to CD52, a protein present on the surface of mature lymphocytes, but not on the stem cells from which these lymphocytes are derived. It is currently used as an immune reconstitution therapy in patients with relapsing-remitting multiple sclerosis. Alemtuzumab treatment is an intermittent infusion that induces long-term remission of Multiple Sclerosis also in the treatment-free period. After the robust T and B cell depletion induced by alemtuzumab, the immune system undergoes radical changes during its reconstitution. In this review, we will discuss the current knowledge on the reconstitution of the lymphocyte repertoire after alemtuzumab treatment and how it could affect the development of side effects, which led to its temporary suspension by the European Medical Agency.


Subject(s)
Alemtuzumab/therapeutic use , Immune Reconstitution , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Animals , Clinical Trials as Topic , Disease Models, Animal , Humans , Translational Research, Biomedical
19.
Cells ; 8(10)2019 10 19.
Article in English | MEDLINE | ID: mdl-31635066

ABSTRACT

Multiple sclerosis (MS) is a chronic central nervous system inflammatory disease that leads to demyelination and neurodegeneration. The third trimester of pregnancy, which is characterized by high levels of estrogens, has been shown to be associated with reduced relapse rates compared with the rates before pregnancy. These effects could be related to the anti-inflammatory properties of estrogens, which orchestrate the reshuffling of the immune system toward immunotolerance to allow for fetal growth. The action of these hormones is mediated by the transcriptional regulation activity of estrogen receptors (ERs). Estrogen levels and ER expression define a specific balance of immune cell types. In this review, we explore the role of estradiol (E2) and ERs in the adaptive immune system, with a focus on estrogen-mediated cellular, molecular, and epigenetic mechanisms related to immune tolerance and neuroprotection in MS. The epigenome dynamics of immune systems are described as key molecular mechanisms that act on the regulation of immune cell identity. This is a completely unexplored field, suggesting a future path for more extensive research on estrogen-induced coregulatory complexes and molecular circuitry as targets for therapeutics in MS.


Subject(s)
Estrogens/metabolism , Multiple Sclerosis/metabolism , Receptors, Estrogen/metabolism , Adaptive Immunity/genetics , Adaptive Immunity/physiology , Animals , Epigenome/genetics , Epigenome/physiology , Estradiol/metabolism , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Multiple Sclerosis/genetics , Pregnancy , Receptors, Estrogen/genetics
20.
Hum Gene Ther ; 19(3): 229-40, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18269312

ABSTRACT

We have shown that electroporation of plasmid carrying extracellular and transmembrane domains (EC-TM plasmid) encoded by the rat neu oncogene triggers a protective immune response toward rat p185(neu)-positive tumors in both wild-type BALB/c mice and cancer-prone rat neu-transgenic BALB-neuT mice. To identify the critical fragments that confer this protective immunity, mice were electroporated with plasmids encoding the TM domain associated with decreasing fragments of the EC domain and the antitumor protection afforded, the titer of antibody, and cytotoxic T lymphocyte (CTL) activity elicited to Neu protein were evaluated. Plasmids encoding EC fragments shortened by 70 (EC1-TM plasmid), 150 (EC2-TM), 230 (EC3-TM), 310 (EC4-TM), and 390 (EC5-TM) NH(2)-terminal residues afforded effective protection. Plasmids encoding shorter truncated proteins were ineffective. When the immunogenic protein was retained in the cytoplasm (EC1-TM, EC2-TM, and EC5-TM), only a CTL response was elicited, whereas when it was also expressed on the membrane (EC4-TM) both CTLs and antibodies were induced. EC4-TM encoding a truncated protein with an EC portion of only 344 amino acids conferred protection on both BALB/c and BALB-neuT mice comparable to that of EC-TM.


Subject(s)
Genes, erbB-2 , Mammary Neoplasms, Animal/immunology , Receptor, ErbB-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies/immunology , Cell Line , Electroporation , Female , Fibroblasts , Genetic Therapy , Mammary Neoplasms, Animal/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Plasmids/genetics , Rats , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL