Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mol Divers ; 28(1): 183-195, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37566199

ABSTRACT

Ten hexahydropyrimido[4,5-d]pyrimidine derivatives have been synthesized by using a green and time-efficient microwave method. The synthesized motifs were evaluated for their anticancer activity, antimicrobial activity, molecular docking, drug likeliness and ADMET studies. Comparatively, the hetero-aromatic pyrazole substituted compound 4a exhibited the highest anticancer activity [Mean growth percent: 35.57], while EDG [-N(CH3)2] substituted compound 4i indicated very good activity [Mean growth percent: 60.92] against various cell lines. From the computational studies, Compound 4a passed the drug-likeness and ADME properties, fewer toxic properties, and potent inhibitory potential against the RIPK2 with significant binding affinity. In-silico molecular docking revealed that the compound 4a has significant binding energy (- 9.8 kcal/mol) and dissociation constant (0.54 µM) properties. Additionally, synthesized motifs were evaluated for antimicrobial activity by MIC referencing the standards. According to the SAR evaluations, the compounds 4f (4-NO2), 4g (3-NO2), and 4h (2-Cl) that include EWGs substituted aldehydes performed well as antimicrobials against selected bacterial and fungal strains. Thus, the synthesized pyrimido[4,5-d]pyrimidine with the heterocyclic and EWGs substituents could act as a potential candidate after further structural optimization for anticancer and antimicrobial drug discovery, respectively.


Subject(s)
Anti-Infective Agents , Antineoplastic Agents , Molecular Docking Simulation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nitrogen Dioxide , Anti-Infective Agents/pharmacology , Pyrimidines/pharmacology , Pyrimidines/chemistry , Molecular Structure , Structure-Activity Relationship
2.
PLoS One ; 17(8): e0264800, 2022.
Article in English | MEDLINE | ID: mdl-35984843

ABSTRACT

BACKGROUND: Upfront docetaxel or novel hormonal agents (NHA) such as abiraterone and enzalutamide have become the standard of care for metastatic hormone sensitive prostate cancer (mHSPC). We evaluated real-world management of patients treated with these agents at a single center. PATIENTS AND METHODS: Patients with de novo mHSPC treated with upfront docetaxel or an NHA between January 2014 and April 2019 at Mount Sinai Health System were included. We evaluated time to next treatment (TTNT), PSA progression free survival (PFS) and overall survival (OS) after initial treatment with these drugs. Kaplan Meier method and multivariable Cox proportional hazards models were used for analysis. We additionally assessed the prognostic value of post-treatment PSA. RESULTS: We identified 94 de novo mHSPC patients; 52 and 42 treated with upfront docetaxel and NHAs, respectively. NHAs were associated with a median TTNT of 20.7 months compared to 10.1 months with docetaxel (log-rank p = 0.023). We also observed median PSA PFS of 19 months for NHAs and 13.2 months for docetaxel (p = 0.069). However, OS between the two treatment groups was unchanged. Among docetaxel treated patients, TTNT was shorter among those with high metastasis burden (9.63 vs 25.5 months, p = 0.026) which was not observed among NHA treated patients (25.1 vs 20.7 months, p = 0.79). Regardless of treatment, lower post-treatment PSA levels were associated with improved TTNT (58.95 vs. 11.57 vs. 9.4 months for PSA ≤0.2, 0.2-0.4, >0.4ng/ml, respectively; p<0.001). CONCLUSION: Real world data demonstrated a shorter duration of treatment with docetaxel than NHAs, reflecting the time-limited nature of docetaxel regimens compared to the treat-till-progression approach of NHAs. While TTNT was generally longer for NHAs than docetaxel, some docetaxel-treated patients achieved significant periods of time off treatment. In addition, the depth of PSA response following combination treatment may hold prognostic value for mHSPC outcomes.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms, Castration-Resistant , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Docetaxel/therapeutic use , Hormones/therapeutic use , Humans , Male , Prostate-Specific Antigen/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Retrospective Studies , Treatment Outcome
3.
Scand J Clin Lab Invest ; 80(6): 508-514, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32706999

ABSTRACT

Familial hypercholesterolemia (FH) is a monogenic disease characterized by a lifelong exposure to high LDL-C levels that can lead to early onset coronary heart disease (CHD). The main causes of FH identified to date include loss-of-function mutations in LDLR or APOB, or gain-of-function mutations in PCSK9. Early diagnosis and genetic testing of FH suspects is critical for improved prognosis of affected individuals as lipid lowering treatments are effective in preventing CHD related morbidity and mortality. In the present study, we carried out a comprehensive screening, using a next-generation sequencing (NGS) panel, for FH culprit mutations in two Icelandic studies representative of either FH families or the general population. We confirmed all previously known mutations in the FH families, and identified two subjects that had been misdiagnosed clinically at young age. We identified six new mutations in the Icelandic FH families and detected three pathogenic mutations in the general population-based study. The application of the NGS panel revealed substantial diagnostic yields in identifying pathogenic mutations, or 68.2% of those with definite clinical diagnosis of FH in the family material and 5.6-fold enrichment in the population-based genetic testing.


Subject(s)
Genetic Testing/methods , Hyperlipoproteinemia Type II/diagnosis , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Hyperlipoproteinemia Type II/genetics , Iceland , Loss of Function Mutation , Mutation , Prospective Studies
4.
J Assist Reprod Genet ; 35(8): 1489-1496, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29779145

ABSTRACT

PURPOSE: To develop a comprehensive genetic test for female and male infertility in support of medical decisions during assisted reproductive technology (ART) protocols. METHODS: We developed a next-generation sequencing (NGS) gene panel consisting of 87 genes including promoters, 5' and 3' untranslated regions, exons, and selected introns. In addition, sex chromosome aneuploidies and Y chromosome microdeletions were analyzed concomitantly using the same panel. RESULTS: The NGS panel was analytically validated by retrospective analysis of 118 genomic DNA samples with known variants in loci representative of female and male infertility. Our results showed analytical accuracy of > 99%, with > 98% sensitivity for single-nucleotide variants (SNVs) and > 91% sensitivity for insertions/deletions (indels). Clinical sensitivity was assessed with samples containing variants representative of male and female infertility, and it was 100% for SNVs/indels, CFTR IVS8-5T variants, sex chromosome aneuploidies, and copy number variants (CNVs) and > 93% for Y chromosome microdeletions. Cost analysis shows potential savings when comparing this single NGS assay with the standard approach, which includes multiple assays. CONCLUSIONS: A single, comprehensive, NGS panel can simplify the ordering process for healthcare providers, reduce turnaround time, and lower the overall cost of testing for genetic assessment of infertility in females and males, while maintaining accuracy.


Subject(s)
Genetic Testing , High-Throughput Nucleotide Sequencing , Infertility, Female/genetics , Infertility, Male/genetics , DNA Copy Number Variations/genetics , Exons , Female , Humans , INDEL Mutation/genetics , Infertility, Female/diagnosis , Infertility, Female/pathology , Infertility, Male/diagnosis , Infertility, Male/pathology , Male , Polymorphism, Single Nucleotide/genetics
5.
Nat Commun ; 8(1): 1600, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29150604

ABSTRACT

We have previously reported the molecular signature of murine pathogenic TH17 cells that induce experimental autoimmune encephalomyelitis (EAE) in animals. Here we show that human peripheral blood IFN-γ+IL-17+ (TH1/17) and IFN-γ-IL-17+ (TH17) CD4+ T cells display distinct transcriptional profiles in high-throughput transcription analyses. Compared to TH17 cells, TH1/17 cells have gene signatures with marked similarity to mouse pathogenic TH17 cells. Assessing 15 representative signature genes in patients with multiple sclerosis, we find that TH1/17 cells have elevated expression of CXCR3 and reduced expression of IFNG, CCL3, CLL4, GZMB, and IL10 compared to healthy controls. Moreover, higher expression of IL10 in TH17 cells is found in clinically stable vs. active patients. Our results define the molecular signature of human pro-inflammatory TH17 cells, which can be used to both identify pathogenic TH17 cells and to measure the effect of treatment on TH17 cells in human autoimmune diseases.


Subject(s)
Gene Expression Profiling , Interleukin-10/genetics , Multiple Sclerosis/genetics , Th17 Cells/metabolism , Adult , Animals , Cells, Cultured , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/metabolism , Male , Mice , Middle Aged , Multiple Sclerosis/metabolism , Th1 Cells/metabolism
6.
Cell Rep ; 17(5): 1318-1329, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27783946

ABSTRACT

Existing therapies for inflammatory bowel disease that are based on broad suppression of inflammation result in variable clinical benefit and unwanted side effects. A potential therapeutic approach for promoting immune tolerance is the in vivo induction of regulatory T cells (Tregs). Here we report that activation of the aryl hydrocarbon receptor using the non-toxic agonist 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) induces human Tregs in vitro that suppress effector T cells through a mechanism mediated by CD39 and Granzyme B. We then developed a humanized murine system whereby human CD4+ T cells drive colitis upon exposure to 2,4,6-trinitrobenzenesulfonic acid and assessed ITE as a potential therapeutic. ITE administration ameliorated colitis in humanized mice with increased CD39, Granzyme B, and IL10-secreting human Tregs. These results develop an experimental model to investigate human CD4+ T responses in vivo and identify the non-toxic AHR agonist ITE as a potential therapy for promoting immune tolerance in the intestine.


Subject(s)
Colitis/immunology , Colitis/metabolism , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Mice , Thiazoles , Transforming Growth Factor beta1/pharmacology , Trinitrobenzenesulfonic Acid
7.
Nat Commun ; 7: 12015, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27352007

ABSTRACT

The gut microbiome plays an important role in immune function and has been implicated in several autoimmune disorders. Here we use 16S rRNA sequencing to investigate the gut microbiome in subjects with multiple sclerosis (MS, n=60) and healthy controls (n=43). Microbiome alterations in MS include increases in Methanobrevibacter and Akkermansia and decreases in Butyricimonas, and correlate with variations in the expression of genes involved in dendritic cell maturation, interferon signalling and NF-kB signalling pathways in circulating T cells and monocytes. Patients on disease-modifying treatment show increased abundances of Prevotella and Sutterella, and decreased Sarcina, compared with untreated patients. MS patients of a second cohort show elevated breath methane compared with controls, consistent with our observation of increased gut Methanobrevibacter in MS in the first cohort. Further study is required to assess whether the observed alterations in the gut microbiome play a role in, or are a consequence of, MS pathogenesis.


Subject(s)
Gastrointestinal Microbiome , Multiple Sclerosis, Relapsing-Remitting/microbiology , RNA, Ribosomal, 16S/genetics , Adult , Breath Tests , Case-Control Studies , Female , Genes, Bacterial , Humans , Immunomodulation , Male , Methane/analysis , Middle Aged , Monocytes/metabolism , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/therapy , Phylogeny , T-Lymphocytes/metabolism
8.
Sci Signal ; 9(433): ra61, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27330188

ABSTRACT

Type 1 diabetes (T1D) is a T cell-dependent autoimmune disease that is characterized by the destruction of insulin-producing ß cells in the pancreas. The administration to patients of ex vivo-differentiated FoxP3(+) regulatory T (Treg) cells or tolerogenic dendritic cells (DCs) that promote Treg cell differentiation is considered a potential therapy for T1D; however, cell-based therapies cannot be easily translated into clinical practice. We engineered nanoparticles (NPs) to deliver both a tolerogenic molecule, the aryl hydrocarbon receptor (AhR) ligand 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), and the ß cell antigen proinsulin (NPITE+Ins) to induce a tolerogenic phenotype in DCs and promote Treg cell generation in vivo. NPITE+Ins administration to 8-week-old nonobese diabetic mice suppressed autoimmune diabetes. NPITE+Ins induced a tolerogenic phenotype in DCs, which was characterized by a decreased ability to activate inflammatory effector T cells and was concomitant with the increased differentiation of FoxP3(+) Treg cells. The induction of a tolerogenic phenotype in DCs by NPs was mediated by the AhR-dependent induction of Socs2, which resulted in inhibition of nuclear factor κB activation and proinflammatory cytokine production (properties of tolerogenic DCs). Together, these data suggest that NPs constitute a potential tool to reestablish tolerance in T1D and potentially other autoimmune disorders.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Humans , Indoles/chemistry , Indoles/pharmacology , Insulin-Secreting Cells/pathology , Mice, Inbred NOD , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes, Regulatory/pathology , Thiazoles/chemistry , Thiazoles/pharmacology
9.
Nat Med ; 22(6): 586-97, 2016 06.
Article in English | MEDLINE | ID: mdl-27158906

ABSTRACT

Astrocytes have important roles in the central nervous system (CNS) during health and disease. Through genome-wide analyses we detected a transcriptional response to type I interferons (IFN-Is) in astrocytes during experimental CNS autoimmunity and also in CNS lesions from patients with multiple sclerosis (MS). IFN-I signaling in astrocytes reduces inflammation and experimental autoimmune encephalomyelitis (EAE) disease scores via the ligand-activated transcription factor aryl hydrocarbon receptor (AHR) and the suppressor of cytokine signaling 2 (SOCS2). The anti-inflammatory effects of nasally administered interferon (IFN)-ß are partly mediated by AHR. Dietary tryptophan is metabolized by the gut microbiota into AHR agonists that have an effect on astrocytes to limit CNS inflammation. EAE scores were increased following ampicillin treatment during the recovery phase, and CNS inflammation was reduced in antibiotic-treated mice by supplementation with the tryptophan metabolites indole, indoxyl-3-sulfate, indole-3-propionic acid and indole-3-aldehyde, or the bacterial enzyme tryptophanase. In individuals with MS, the circulating levels of AHR agonists were decreased. These findings suggest that IFN-Is produced in the CNS function in combination with metabolites derived from dietary tryptophan by the gut flora to activate AHR signaling in astrocytes and suppress CNS inflammation.


Subject(s)
Astrocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Gastrointestinal Microbiome , Interferon Type I/immunology , Multiple Sclerosis/immunology , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes/immunology , Tryptophan/metabolism , Animals , Case-Control Studies , Cell Proliferation , Central Nervous System/immunology , Central Nervous System/metabolism , Chemokine CCL2/metabolism , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Knockdown Techniques , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunoblotting , Indican/urine , Indoles/metabolism , Inflammation , Interferon-beta/pharmacology , Limosilactobacillus reuteri , Mice , Mice, Knockout , Multiple Sclerosis/metabolism , Myxovirus Resistance Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Optical Imaging , Polymerase Chain Reaction , Receptor, Interferon alpha-beta/genetics , Receptors, Aryl Hydrocarbon/metabolism , STAT1 Transcription Factor/metabolism , Serotonin , Suppressor of Cytokine Signaling Proteins , Tryptophanase/metabolism
10.
Neurol Neuroimmunol Neuroinflamm ; 3(2): e200, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26894204

ABSTRACT

OBJECTIVE: To determine whether peripheral immune responses as measured by serum antigen arrays are linked to cerebral MRI measures of disease severity in multiple sclerosis (MS). METHODS: In this cross-sectional study, serum samples were obtained from patients with relapsing-remitting MS (n = 21) and assayed using antigen arrays that contained 420 antigens including CNS-related autoantigens, lipids, and heat shock proteins. Normalized compartment-specific global brain volumes were obtained from 3-tesla MRI as surrogates of atrophy, including gray matter fraction (GMF), white matter fraction (WMF), and total brain parenchymal fraction (BPF). Total brain T2 hyperintense lesion volume (T2LV) was quantified from fluid-attenuated inversion recovery images. RESULTS: We found serum antibody patterns uniquely correlated with BPF, GMF, WMF, and T2LV. Furthermore, we identified immune signatures linked to MRI markers of neurodegeneration (BPF, GMF, WMF) that differentiated those linked to T2LV. Each MRI measure was correlated with a specific set of antibodies. Strikingly, immunoglobulin G (IgG) antibodies to lipids were linked to brain MRI measures. Based on the association between IgG antibody reactivity and each unique MRI measure, we developed a lipid index. This comprised the reactivity directed against all of the lipids associated with each specific MRI measure. We validated these findings in an additional independent set of patients with MS (n = 14) and detected a similar trend for the correlations between BPF, GMF, and T2LV vs their respective lipid indexes. CONCLUSIONS: We propose serum antibody repertoires that are associated with MRI measures of cerebral MS involvement. Such antibodies may serve as biomarkers for monitoring disease pathology and progression.

11.
J Neuroinflammation ; 12: 245, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26714756

ABSTRACT

BACKGROUND: Fingolimod (FTY720), the first oral treatment for multiple sclerosis (MS), blocks immune cell trafficking and prevents disease relapses by downregulation of sphingosine-1-phosphate receptor. We determined the effect of FTY720 on human T cell activation and effector function. METHODS: T cells from MS patients and healthy controls were isolated to measure gene expression profiles in the presence or absence of FTY720 using nanostring and quantitative real-time polymerase chain reaction (qPCR). Cytokine protein expression was measured using luminex assay and flow cytometry analysis. Lentivirus vector carrying short hairpin RNA (shRNA) was used to knock down the expression of specific genes in CD4+ T cells. Chromatin immunoprecipitation was performed to assess T cell factor 1 (TCF-1) binding to promoter regions. Luciferase assays were performed to test the direct regulation of interferon gamma (IFN-γ) and granzyme B (GZMB) by TCF-1. Western blot analysis was used to assess the phosphorylation status of Akt and GSK3ß. RESULTS: We showed that FTY720 treatment not only affects T cell trafficking but also T cell activation. Patients treated with FTY720 showed a significant reduction in circulating CD4 T cells. Activation of T cells in presence of FTY720 showed a less inflammatory phenotype with reduced production of IFN-γ and GZMB. This decreased effector phenotype of FTY720-treated T cells was dependent on the upregulation of TCF-1. FTY720-induced TCF-1 downregulated the pathogenic cytokines IFN-γ and GZMB by binding to their promoter/enhancer regions and mediating epigenetic modifications. Furthermore, we observed that TCF-1 expression was lower in T cells from multiple sclerosis patients than in those from healthy individuals, and FTY720 treatment increased TCF-1 expression in multiple sclerosis patients. CONCLUSIONS: These results reveal a previously unknown mechanism of the effect of FTY720 on human CD4+ T cell modulation in multiple sclerosis and demonstrate the role of TCF-1 in human T cell activation and effector function.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/pharmacology , Multiple Sclerosis/metabolism , T Cell Transcription Factor 1/biosynthesis , Up-Regulation/physiology , CD4-Positive T-Lymphocytes/drug effects , Female , Fingolimod Hydrochloride/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Male , Multiple Sclerosis/drug therapy , Up-Regulation/drug effects
12.
Cell ; 162(6): 1338-52, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26359987

ABSTRACT

Seasonal changes in disease activity have been observed in multiple sclerosis, an autoimmune disorder that affects the CNS. These epidemiological observations suggest that environmental factors influence the disease course. Here, we report that melatonin levels, whose production is modulated by seasonal variations in night length, negatively correlate with multiple sclerosis activity in humans. Treatment with melatonin ameliorates disease in an experimental model of multiple sclerosis and directly interferes with the differentiation of human and mouse T cells. Melatonin induces the expression of the repressor transcription factor Nfil3, blocking the differentiation of pathogenic Th17 cells and boosts the generation of protective Tr1 cells via Erk1/2 and the transactivation of the IL-10 promoter by ROR-α. These results suggest that melatonin is another example of how environmental-driven cues can impact T cell differentiation and have implications for autoimmune disorders such as multiple sclerosis.


Subject(s)
Melatonin/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Light , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Recurrence , Seasons , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
13.
Nat Med ; 21(6): 638-46, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26005855

ABSTRACT

Our understanding of the pathways that regulate lymphocyte metabolism, as well as the effects of metabolism and its products on the immune response, is still limited. We report that a metabolic program controlled by the transcription factors hypoxia inducible factor-1α (HIF1-α) and aryl hydrocarbon receptor (AHR) supports the differentiation of type 1 regulatory T cell (Tr1) cells. HIF1-α controls the early metabolic reprograming of Tr1 cells. At later time points, AHR promotes HIF1-α degradation and takes control of Tr1 cell metabolism. Extracellular ATP (eATP) and hypoxia, linked to inflammation, trigger AHR inactivation by HIF1-α and inhibit Tr1 cell differentiation. Conversely, CD39 promotes Tr1 cell differentiation by depleting eATP. CD39 also contributes to Tr1 suppressive activity by generating adenosine in cooperation with CD73 expressed by responder T cells and antigen-presenting cells. These results suggest that HIF1-α and AHR integrate immunological, metabolic and environmental signals to regulate the immune response.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Cellular , Inflammation/immunology , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Regulatory/metabolism , Adult , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Apyrase/immunology , Apyrase/metabolism , Cell Differentiation/immunology , Female , Hematopoiesis/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Inflammation/metabolism , Inflammation/pathology , Lymphocyte Activation/immunology , Mice , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes, Regulatory/immunology
14.
Blood ; 125(25): 3886-95, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-25833964

ABSTRACT

Mice reconstituted with a human immune system provide a tractable in vivo model to assess human immune cell function. To date, reconstitution of murine strains with human hematopoietic stem cells (HSCs) from patients with monogenic immune disorders have not been reported. One obstacle precluding the development of immune-disease specific "humanized" mice is that optimal adaptive immune responses in current strains have required implantation of autologous human thymic tissue. To address this issue, we developed a mouse strain that lacks murine major histocompatibility complex class II (MHC II) and instead expresses human leukocyte antigen DR1 (HLA-DR1). These mice displayed improved adaptive immune responses when reconstituted with human HSCs including enhanced T-cell reconstitution, delayed-type hypersensitivity responses, and class-switch recombination. Following immune reconstitution of this novel strain with HSCs from a patient with immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, associated with aberrant FOXP3 function, mice developed a lethal inflammatory disorder with multiorgan involvement and autoantibody production mimicking the pathology seen in affected humans. This humanized mouse model permits in vivo evaluation of immune responses associated with genetically altered HSCs, including primary immunodeficiencies, and should facilitate the study of human immune pathobiology and the development of targeted therapeutics.


Subject(s)
Autoimmunity/immunology , Disease Models, Animal , Forkhead Transcription Factors/immunology , Immunologic Deficiency Syndromes/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Hematopoietic Stem Cell Transplantation , Humans , Immunohistochemistry , Immunophenotyping , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
15.
Neurology ; 83(24): 2219-26, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25381299

ABSTRACT

OBJECTIVES: For most adults with initial clinical presentation of multiple sclerosis (MS), biological disease was likely initiated many years prior. Pediatric-onset MS provides an opportunity to study early disease processes. METHODS: Using antigen microarrays, including CNS-related proteins, lipids, and other autoantigens, we studied early immunologic events involved in clinical onset of pediatric MS. Serum samples were collected at the time of incident acquired CNS demyelinating syndromes (ADS) in children who, in subsequent prospective follow-up, were ascertained to have either pediatric MS (ADS-MS) or a monophasic illness (ADS-mono). Samples were obtained both at the time of ADS presentation and 3 months into follow-up. We used an initial training set of samples to implicate antibody signatures associated with each group, and then a test set. An additional set of follow-up samples (stability set) was used as a form of internal validation. RESULTS: Children with ADS-MS tended to have distinguishable serum antibody patterns both at the time of ADS presentation and 3 months into follow-up. At the time of ADS, serum samples from patients with ADS-MS or ADS-mono reacted against similar numbers of CNS antigens, although CNS antigens implicated in adult MS were more often targeted in children with ADS-MS. The follow-up ADS-MS samples reacted against a broader panel of CNS antigens, while corresponding ADS-mono samples exhibited a contraction of the initial antibody response. CONCLUSIONS: Our findings in this prospective cohort of pediatric-onset CNS demyelinating diseases point to an active process of epitope spreading during early stages of MS, not seen in monophasic CNS inflammatory conditions.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Central Nervous System Diseases/immunology , Demyelinating Diseases/immunology , Epitopes/immunology , Multiple Sclerosis/immunology , Adolescent , Autoantibodies/immunology , Brain/pathology , Central Nervous System Diseases/diagnosis , Central Nervous System Diseases/pathology , Child , Demyelinating Diseases/diagnosis , Demyelinating Diseases/pathology , Disease Progression , Female , Follow-Up Studies , Humans , Immunoglobulin G/blood , Male , Microarray Analysis , Multiple Sclerosis/diagnosis , Multiple Sclerosis/pathology , Prospective Studies , ROC Curve
16.
Nat Med ; 20(10): 1147-56, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25216636

ABSTRACT

Astrocytes have complex roles in health and disease, thus it is important to study the pathways that regulate their function. Here we report that lactosylceramide (LacCer) synthesized by ß-1,4-galactosyltransferase 6 (B4GALT6) is upregulated in the central nervous system (CNS) of mice during chronic experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). LacCer acts in an autocrine manner to control astrocyte transcriptional programs that promote neurodegeneration. In addition, LacCer in astrocytes controls the recruitment and activation of microglia and CNS-infiltrating monocytes in a non-cell autonomous manner by regulating production of the chemokine CCL2 and granulocyte-macrophage colony-stimulating factor (GM-CSF), respectively. We also detected high B4GALT6 gene expression and LacCer concentrations in CNS MS lesions. Inhibition of LacCer synthesis in mice suppressed local CNS innate immunity and neurodegeneration in EAE and interfered with the activation of human astrocytes in vitro. Thus, B4GALT6 regulates astrocyte activation and is a potential therapeutic target for MS and other neuroinflammatory disorders.


Subject(s)
Astrocytes/immunology , Astrocytes/metabolism , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Galactosyltransferases/metabolism , Glycolipids/metabolism , Animals , Antigens, CD/metabolism , Central Nervous System/pathology , Chemokine CCL2/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Galactosyltransferases/genetics , Gene Knockdown Techniques , Glial Fibrillary Acidic Protein , Humans , Immunity, Innate , Lactosylceramides/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Up-Regulation
17.
Nat Commun ; 5: 3753, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24796415

ABSTRACT

Interleukin (IL)-22 produced by innate lymphoid cells (ILCs) and CD4+ T cells plays an important role in host defence and mucosal homeostasis, thus it is important to investigate the mechanisms that regulate IL-22 production. We investigated the regulation IL-22 production by CD4+ T cells. Here we show that IL-21 triggers IL-22, but not IL-17 production by CD4+ T cells. STAT3, activated by IL-21, controls the epigenetic status of the il22 promoter and its interaction with the aryl hydrocarbon receptor (AhR). Moreover, IL-21 and AhR signalling in T cells control IL-22 production and the development of dextran sodium sulphate-induced colitis in ILC-deficient mice. Thus, we have identified IL-21 as an inducer of IL-22 production in CD4+ T cells in vitro and in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Interleukins/biosynthesis , Interleukins/physiology , Gene Expression Profiling , Humans , Interleukins/genetics , Interleukins/metabolism , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/metabolism , STAT3 Transcription Factor/physiology , Transcription, Genetic , Interleukin-22
18.
Immunity ; 40(4): 569-81, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24745333

ABSTRACT

Foxp3(+) T regulatory (Treg) cells regulate immune responses and maintain self-tolerance. Recent work shows that Treg cells are comprised of many subpopulations with specialized regulatory functions. Here we identified Foxp3(+) T cells expressing the coinhibitory molecule TIGIT as a distinct Treg cell subset that specifically suppresses proinflammatory T helper 1 (Th1) and Th17 cell, but not Th2 cell responses. Transcriptional profiling characterized TIGIT(+) Treg cells as an activated Treg cell subset with high expression of Treg signature genes. Ligation of TIGIT on Treg cells induced expression of the effector molecule fibrinogen-like protein 2 (Fgl2), which promoted Treg-cell-mediated suppression of T effector cell proliferation. In addition, Fgl2 was necessary to prevent suppression of Th2 cytokine production in a model of allergic airway inflammation. TIGIT expression therefore identifies a Treg cell subset that demonstrates selectivity for suppression of Th1 and Th17 cell but not Th2 cell responses.


Subject(s)
Fibrinogen/metabolism , Receptors, Immunologic/metabolism , Respiratory Hypersensitivity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Eosinophils/immunology , Fibrinogen/genetics , Fibrinogen/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation , Immunosuppression Therapy , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Th1-Th2 Balance
19.
Nat Immunol ; 14(10): 1054-63, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995234

ABSTRACT

Dendritic cells (DCs) control the balance between effector T cells and regulatory T cells in vivo. Hence, the study of DCs might identify mechanisms of disease pathogenesis and guide new therapeutic approaches for disorders mediated by the immune system. We found that interleukin 27 (IL-27) signaling in mouse DCs limited the generation of effector cells of the TH1 and TH17 subsets of helper T cells and the development of experimental autoimmune encephalomyelitis (EAE). The effects of IL-27 were mediated at least in part through induction of the immunoregulatory molecule CD39 in DCs. IL-27-induced CD39 decreased the extracellular concentration of ATP and downregulated nucleotide-dependent activation of the NLRP3 inflammasome. Finally, therapeutic vaccination with IL-27-conditioned DCs suppressed established relapsing-remitting EAE. Thus, IL-27 signaling in DCs limited pathogenic T cell responses and the development of autoimmunity.


Subject(s)
Antigens, CD/genetics , Apyrase/genetics , Autoimmunity , Dendritic Cells/drug effects , Dendritic Cells/immunology , Interleukin-17/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Animals , Antibodies/immunology , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigens, CD/metabolism , Apyrase/metabolism , Autoantibodies/immunology , Autoimmunity/drug effects , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression , Gene Expression Regulation/drug effects , Immune Tolerance/immunology , Mice , Mice, Knockout , Myelin Sheath/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Interleukin , Signal Transduction , T-Lymphocyte Subsets/cytology , Transcription, Genetic/drug effects
20.
Ann Neurol ; 73(6): 729-40, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23494648

ABSTRACT

OBJECTIVE: MicroRNAs (miRNAs) are single-stranded, small noncoding RNAs that regulate gene expression. Because they are stable in serum, they are being developed as biomarkers for cancer and other diseases. In multiple sclerosis (MS), miRNAs have been studied in cell populations but not in the circulation. In MS, a major challenge is to develop immune biomarkers to monitor disease. We asked whether circulating miRNAs could be identified in MS and whether they are linked to disease stage and/or disability. METHODS: A total of 368 miRNAs were measured in ethylenediaminetetraacetic acid plasma in 10 relapsing-remitting MS (RRMS) patients, 9 secondary progressive MS (SPMS) patients, and 9 healthy controls (HCs) using miRCURY LNA™ Universal RT microRNA polymerase chain reaction panels. Nineteen miRNAs from this discovery set were validated using qPCR on an independent set of 50 RRMS patients, 51 SPMS patients, and 32 HCs. RESULTS: We found that circulating miRNAs are differentially expressed in RRMS and SPMS versus HCs and in RRMS versus SPMS. We also found miRNAs to be linked to Expanded Disability Status Scale (EDSS). hsa-miR-92a-1* was identified in the largest number of comparisons. It was different in RRMS versus SPMS, and RRMS versus HCs, and showed an association with EDSS and disease duration. miR-92 has target genes involved in cell cycle regulation and cell signaling. The let-7 family of miRNAs differentiated SPMS from HCs and RRMS from SPMS. let-7 miRNAs regulate stem cell differentiation and T cell activation, activate Toll-like receptor 7, and are linked to neurodegeneration. hsa-miR-454 differentiated RRMS from SPMS, and hsa-miR-145 differentiated RRMS from HCs and RRMS from SPMS. Interestingly, the same circulating miRNAs (let-7 and miR-92) that were differentially expressed in RRMS versus SPMS also differentiated amyotrophic lateral sclerosis (ALS) from RRMS subjects, but were not different between SPMS and ALS, suggesting that similar processes may occur in SPMS and ALS. INTERPRETATION: Our results establish circulating miRNAs as a readily accessible blood biomarker to monitor disease in MS.


Subject(s)
Disease Progression , MicroRNAs/blood , Multiple Sclerosis/blood , Multiple Sclerosis/diagnosis , Adult , Aged , Biomarkers/blood , Cohort Studies , Female , Humans , Longitudinal Studies , Male , Middle Aged , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...