Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Neuroinflammation ; 21(1): 8, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38178143

ABSTRACT

Progressive brain diseases create a huge social and economic burden on modern societies as a major cause of disability and death. Incidence of brain diseases has a significantly increasing trend and merits new therapeutic strategies. At the base of many progressive brain malfunctions is a process of unresolved, chronic inflammation. Macrophage migration inhibitory factor, MIF, is an inflammatory mediator that recently gained interest of neuro-researchers due to its varied effects on the CNS such as participation of nervous system development, neuroendocrine functions, and modulation of neuroinflammation. MIF appears to be a candidate as a new biomarker and target of novel therapeutics against numerous neurologic diseases ranging from cancer, autoimmune diseases, vascular diseases, neurodegenerative pathology to psychiatric disorders. In this review, we will focus on MIF's crucial role in neurological diseases such as multiple sclerosis (MS), Alzheimer's disease (AD) and glioblastoma (GBM).


Subject(s)
Brain Diseases , Macrophage Migration-Inhibitory Factors , Multiple Sclerosis , Nervous System Diseases , Humans , Macrophage Migration-Inhibitory Factors/genetics , Inflammation , Calgranulin A , Calgranulin B , Intramolecular Oxidoreductases
2.
Eur J Clin Invest ; 54(4): e14142, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38071404

ABSTRACT

BACKGROUND AND AIMS: We found a higher incidence of myocarditis in young males who had received at least two Pfizer-BioNTech BNT162b2 vaccinations. The human leukocyte antigens (HLA) are known to play an important role in infectious and autoinflammatory diseases. We hypothesized that certain HLA alleles might be associated with vaccination-induced myocarditis. METHODS: HLA typing was performed using next-generation sequencing technology with the Illumina Iseq100 platform. HLA class I and II loci were genotyped in 29 patients with post-vaccination myocarditis and compared with HLA data from 300 healthy controls. RESULTS: We demonstrate that the DRB1*14:01, DRB1*15:03 alleles and the motifs in HLA-A - Leu62 and Gln63, which are part of binding pocket B and HLA-DR Tyr47, His60, Arg70 and Glu74, which are part of binding pockets P4, P7 and P9, were significantly associated with disease susceptibility. CONCLUSIONS: Our findings suggest that immunogenetic fingerprints in HLA peptide-binding grooves may affect the presentation of peptides derived from the Pfizer-BioNTech BNT162b2 vaccination to T cells and induce an inflammatory process that results in myocarditis.


Subject(s)
BNT162 Vaccine , Myocarditis , Male , Humans , Myocarditis/chemically induced , Myocarditis/genetics , HLA-DRB1 Chains/metabolism , HLA Antigens , Disease Susceptibility , Peptides
3.
Cell Immunol ; 378: 104561, 2022 08.
Article in English | MEDLINE | ID: mdl-35738135

ABSTRACT

Our previous studies demonstrated increased serum levels of macrophage migration inhibitory factor (MIF-1) and its homologue, MIF-2, in males during MS progression; and that genetically high-MIF-expressing male subjects with relapsing multiple sclerosis (MS) had a significantly greater risk of conversion to progressive MS than lower-MIF-expressing males and females. However, female MS subjects with severe disease expressed higher levels of CD74, the common MIF-1/MIF-2 receptor, on blood cells. In the murine model of MS, experimental autoimmune encephalomyelitis (EAE), both male and female mice lacking MIF-1 and/or MIF-2 were clinically improved during development of moderately severe disease, thus implicating both homologs as co-pathogenic contributors. The current study using MIF-deficient mice with severe acute EAE revealed a highly significant reduction of EAE scores in MIF-1-deficient females, in contrast to only minor and delayed reduction of clinical signs in MIF-1-deficient males. However, clinical EAE scores and factor expression were strongly suppressed in males and further reduced in females after treatment of WT and MIF-1-, MIF-2- and MIF-1/2-DUAL-deficient female and male mice with a MHCII DRα1-MOG-35-55 molecular construct that competitively inhibits MIF-1 & MIF-2 signaling through CD74 as well as T cell activation. These results suggest sex-dependent differences in which the absence of the MIF-1 and/or MIF-2 genotypes may permit stronger compensatory CD74-dependent EAE-inducing responses in males than in females. However, EAE severity in both sexes could still be reduced nearly to background (a "near cure") with DRα1-MOG-35-55 blockade of compensatory MIF and CD74-dependent factors known to attract peripheral inflammatory cells into the spinal cord tissue.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , MSH Release-Inhibiting Hormone , Macrophage Migration-Inhibitory Factors , Multiple Sclerosis , Animals , Female , Histocompatibility Antigens Class II/immunology , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , MSH Release-Inhibiting Hormone/metabolism , MSH Release-Inhibiting Hormone/therapeutic use , Macrophage Migration-Inhibitory Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Spinal Cord
4.
Cell Immunol ; 370: 104439, 2021 12.
Article in English | MEDLINE | ID: mdl-34607646

ABSTRACT

Macrophage migration inhibitory factor (MIF-1) and its homologue d-dopachrome tautomerase (MIF-2) share the common CD74 receptor and function innately to enhance severity of multiple sclerosis (MS) as well as the experimental autoimmune encephalomyelitis (EAE) model for MS. We previously demonstrated that genetically high-MIF-expressing male subjects with relapsing MS had a significantly greater risk of conversion to progressive MS (PMS) than lower-MIF-expressing males. To expand on this observation, we utilized MIF-1, MIF-2, and MIF-1/2-DUAL-deficient male mice to discern if there would be a greater contribution of these inflammatory factors in EAE mice with severe vs. moderate clinical disease signs. As shown previously, mice deficient in either MIF-1 or MIF-2 each had a ∼25% reduction of moderate EAE compared to WT mice, with significant differences in disease onset and trajectory. However, EAE induction in mice deficient in both MIF-1 and MIF-2 genes did not result in a further reduction in EAE severity. This result suggests that the two MIF homologues were likely affecting the same pathogenic pathways such that each could partially compensate for the other but not in an additive or synergistic manner. However, MIF-1-KO, MIF-2-KO, and MIF-1/2-DUAL-KO mice with severe EAE did not exhibit a significant reduction in cumulative EAE scores compared with WT mice, but the MIF-1-KO and, to a lesser extent, MIF-1/2-DUAL-KO mice did show a significant reduction in daily EAE scores over the last 3 days of observation, and MIF-2-KO mice showed a more modest but still consistent reduction over the same span. Furthermore, deletion of MIF-1 resulted in a massive reduction in the expression of EAE- and Complete Freund's Adjuvant-associated inflammatory factors, suggesting delayed involvement of the MIF/CD74 axis in promoting disease expression. To further explore modulation of MIF-1 and MIF-2 effects on EAE, we treated WT mice with moderate EAE using DRα1-mMOG-35-55, an inhibitor of CD74 that blocks both MIF-1 and MIF-2 action. This treatment reduced ongoing moderate EAE severity in excess of 25%, suggesting efficient blockade of the MIF/CD74 axis in disease-enhancing pathways. Moreover, DRα1-mMOG-35-55 treatment of mice with severe EAE strongly reversed EAE- and CFA-associated expression of inflammatory cytokines and chemokines including Tnf, Ccr7, Ccr6, Ccl8, Cxcr3, and Ccl19 in MIF-deficient mouse genotypes, and also exceeded innate MIF-1 and MIF-2 EAE enhancing effects, especially in MIF-1-KO mice. These results illustrate the therapeutic potential of targeting the disease-enhancing MIF/CD74 pathway in male mice with moderate and severe EAE, with implications for treatment of high-MIF-expressing RRMS human males at risk of conversion to progressive MS as well as those that have already transitioned to PMS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Neuroprotective Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
J Clin Invest ; 131(20)2021 10 15.
Article in English | MEDLINE | ID: mdl-34651586

ABSTRACT

Neoantigens are now recognized drivers of the antitumor immune response. Recurrent neoantigens, shared among groups of patients, have thus become increasingly coveted therapeutic targets. Here, we report on the data-driven identification of a robustly presented, immunogenic neoantigen that is derived from the combination of HLA-A*01:01 and RAS.Q61K. Analysis of large patient cohorts indicated that this combination applies to 3% of patients with melanoma. Using HLA peptidomics, we were able to demonstrate robust endogenous presentation of the neoantigen in 10 tumor samples. We detected specific reactivity to the mutated peptide within tumor-infiltrating lymphocytes (TILs) from 2 unrelated patients, thus confirming its natural immunogenicity. We further investigated the neoantigen-specific clones and their T cell receptors (TCRs) via a combination of TCR sequencing, TCR overexpression, functional assays, and single-cell transcriptomics. Our analysis revealed a diverse repertoire of neoantigen-specific clones with both intra- and interpatient TCR similarities. Moreover, 1 dominant clone proved to cross-react with the highly prevalent RAS.Q61R variant. Transcriptome analysis revealed a high association of TCR clones with specific T cell phenotypes in response to cognate melanoma, with neoantigen-specific cells showing an activated and dysfunctional phenotype. Identification of recurrent neoantigens and their reactive TCRs can promote "off-the-shelf" precision immunotherapies, alleviating limitations of personalized treatments.


Subject(s)
Antigens, Neoplasm/immunology , Melanoma/immunology , ras Proteins/immunology , Cell Line, Tumor , HLA-A Antigens/immunology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, Antigen, T-Cell/immunology , ras Proteins/genetics
6.
PLoS Negl Trop Dis ; 15(10): e0009821, 2021 10.
Article in English | MEDLINE | ID: mdl-34662363

ABSTRACT

Nodding syndrome (NS) is a catastrophic and enigmatic childhood epilepsy, accompanied by multiple neurological impairments and neuroinflammation. Of all the infectious, environmental and psychological factors associated with NS, the major culprit is Onchocerca Volvulus (Ov)-a parasitic worm transmitted to human by blackflies. NS seems to be an 'Autoimmune Epilepsy' in light of the recent findings of deleterious autoimmune antibodies to Glutamate receptors and to Leiomodin-I in NS patients. Moreover, we recently found immunogenetic fingerprints in HLA peptide-binding grooves associate with protection or susceptibility to NS. Macrophage migration inhibitory factor (MIF) is an immune-regulatory cytokine playing a central role in modulating innate and adaptive immunity. MIF is also involved in various pathologies: infectious, autoimmune and neurodegenerative diseases, epilepsy and others. Herein, two functional polymorphisms in the MIF gene, a -794 CATT5-8 microsatellite repeat and a -173 G/C single-nucleotide polymorphism, were assessed in 49 NS patients and 51 healthy controls from South Sudan. We also measured MIF plasma levels in established NS patients and healthy controls. We discovered that the frequency of the high-expression MIF -173C containing genotype was significantly lower in NS patients compared to healthy controls. Interestingly however, MIF plasma levels were significantly elevated in NS patients than in healthy controls. We further demonstrated that the HLA protective and susceptibility associations are dominant over the MIF association with NS. Our findings suggest that MIF might have a dual role in NS. Genetically controlled high-expression MIF genotype is associated with disease protection. However, elevated MIF in the plasma may contribute to the detrimental autoimmunity, neuroinflammation and epilepsy.


Subject(s)
Macrophage Migration-Inhibitory Factors/genetics , Nodding Syndrome/genetics , Adolescent , Adult , Animals , Child , Child, Preschool , Female , Genotype , Humans , Macrophage Migration-Inhibitory Factors/blood , Male , Microsatellite Repeats , Nodding Syndrome/blood , Nodding Syndrome/parasitology , Onchocerca volvulus/physiology , Polymorphism, Single Nucleotide , Young Adult
7.
Cell Rep ; 35(13): 109305, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34166618

ABSTRACT

The human leukocyte antigen (HLA)-bound viral antigens serve as an immunological signature that can be selectively recognized by T cells. As viruses evolve by acquiring mutations, it is essential to identify a range of presented viral antigens. Using HLA peptidomics, we are able to identify severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-derived peptides presented by highly prevalent HLA class I (HLA-I) molecules by using infected cells as well as overexpression of SARS-CoV-2 genes. We find 26 HLA-I peptides and 36 HLA class II (HLA-II) peptides. Among the identified peptides, some are shared between different cells and some are derived from out-of-frame open reading frames (ORFs). Seven of these peptides were previously shown to be immunogenic, and we identify two additional immunoreactive peptides by using HLA multimer staining. These results may aid the development of the next generation of SARS-CoV-2 vaccines based on presented viral-specific antigens that span several of the viral genes.


Subject(s)
Antigens, Viral/immunology , COVID-19/immunology , COVID-19/virology , Peptides/immunology , SARS-CoV-2/immunology , Antigen Presentation , Antigens, Viral/metabolism , COVID-19 Vaccines , Cell Line , Epitopes, T-Lymphocyte/immunology , HEK293 Cells , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Peptidomimetics , SARS-CoV-2/genetics , T-Lymphocytes
8.
Nature ; 592(7852): 138-143, 2021 04.
Article in English | MEDLINE | ID: mdl-33731925

ABSTRACT

A variety of species of bacteria are known to colonize human tumours1-11, proliferate within them and modulate immune function, which ultimately affects the survival of patients with cancer and their responses to treatment12-14. However, it is not known whether antigens derived from intracellular bacteria are presented by the human leukocyte antigen class I and II (HLA-I and HLA-II, respectively) molecules of tumour cells, or whether such antigens elicit a tumour-infiltrating T cell immune response. Here we used 16S rRNA gene sequencing and HLA peptidomics to identify a peptide repertoire derived from intracellular bacteria that was presented on HLA-I and HLA-II molecules in melanoma tumours. Our analysis of 17 melanoma metastases (derived from 9 patients) revealed 248 and 35 unique HLA-I and HLA-II peptides, respectively, that were derived from 41 species of bacteria. We identified recurrent bacterial peptides in tumours from different patients, as well as in different tumours from the same patient. Our study reveals that peptides derived from intracellular bacteria can be presented by tumour cells and elicit immune reactivity, and thus provides insight into a mechanism by which bacteria influence activation of the immune system and responses to therapy.


Subject(s)
Antigens, Bacterial/analysis , Antigens, Bacterial/immunology , Bacteria/immunology , HLA Antigens/immunology , Melanoma/immunology , Melanoma/microbiology , Peptides/analysis , Peptides/immunology , Antigen Presentation , Bacteria/classification , Bacteria/genetics , Cell Line, Tumor , Coculture Techniques , HLA Antigens/analysis , Humans , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/pathology , Neoplasm Metastasis/immunology , Phylogeny , RNA, Ribosomal, 16S/genetics
9.
PLoS One ; 16(3): e0249149, 2021.
Article in English | MEDLINE | ID: mdl-33784369

ABSTRACT

Conducting numerous, rapid, and reliable PCR tests for SARS-CoV-2 is essential for our ability to monitor and control the current COVID-19 pandemic. Here, we tested the sensitivity and efficiency of SARS-CoV-2 detection in clinical samples collected directly into a mix of lysis buffer and RNA preservative, thus inactivating the virus immediately after sampling. We tested 79 COVID-19 patients and 20 healthy controls. We collected two samples (nasopharyngeal swabs) from each participant: one swab was inserted into a test tube with Viral Transport Medium (VTM), following the standard guideline used as the recommended method for sample collection; the other swab was inserted into a lysis buffer supplemented with nucleic acid stabilization mix (coined NSLB). We found that RT-qPCR tests of patients were significantly more sensitive with NSLB sampling, reaching detection threshold 2.1±0.6 (Mean±SE) PCR cycles earlier then VTM samples from the same patient. We show that this improvement is most likely since NSLB samples are not diluted in lysis buffer before RNA extraction. Re-extracting RNA from NSLB samples after 72 hours at room temperature did not affect the sensitivity of detection, demonstrating that NSLB allows for long periods of sample preservation without special cooling equipment. We also show that swirling the swab in NSLB and discarding it did not reduce sensitivity compared to retaining the swab in the tube, thus allowing improved automation of COVID-19 tests. Overall, we show that using NSLB instead of VTM can improve the sensitivity, safety, and rapidity of COVID-19 tests at a time most needed.


Subject(s)
Limit of Detection , SARS-CoV-2/isolation & purification , Safety , Specimen Handling/methods , Adult , Buffers , Female , Humans , Male , Pandemics , Polymerase Chain Reaction , SARS-CoV-2/genetics , Time Factors
10.
Sci Transl Med ; 13(589)2021 04 14.
Article in English | MEDLINE | ID: mdl-33619081

ABSTRACT

Pooling multiple swab samples before RNA extraction and real-time reverse transcription polymerase chain reaction (RT-PCR) analysis has been proposed as a strategy to reduce costs and increase throughput of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) tests. However, reports on practical large-scale group testing for SARS-CoV-2 have been scant. Key open questions concern reduced sensitivity due to sample dilution, the rate of false positives, the actual efficiency (number of tests saved by pooling), and the impact of infection rate in the population on assay performance. Here, we report an analysis of 133,816 samples collected between April and September 2020 and tested by Dorfman pooling for the presence of SARS-CoV-2. We spared 76% of RNA extraction and RT-PCR tests, despite the frequently changing prevalence (0.5 to 6%). We observed pooling efficiency and sensitivity that exceeded theoretical predictions, which resulted from the nonrandom distribution of positive samples in pools. Overall, our findings support the use of pooling for efficient large-scale SARS-CoV-2 testing.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19 Testing , Humans , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Specimen Handling
11.
PLoS Negl Trop Dis ; 14(7): e0008436, 2020 07.
Article in English | MEDLINE | ID: mdl-32639997

ABSTRACT

Nodding syndrome (NS) is a devastating and enigmatic childhood epilepsy. NS is accompanied by multiple neurological impairments and neuroinflammation, and associated with the parasite Onchocerca volvulus (Ov) and other environmental factors. Moreover, NS seems to be an 'Autoimmune Epilepsy' since: 1. ~50% of NS patients have neurotoxic cross-reactive Ov/Leimodin-I autoimmune antibodies. 2. Our recently published findings: Most (~86%) of NS patients have glutamate-receptor AMPA-GluR3B peptide autoimmune antibodies that bind, induce Reactive Oxygen Species, and kill both neural cells and T cells. Furthermore, NS patient's IgG induce seizures, brain multiple damage alike occurring in brains of NS patients, and elevation of T cells and activated microglia and astrocytes, in brains of normal mice. Human Leukocyte antigen (HLA) class I and II molecules are critical for initiating effective beneficial immunity against foreign microorganisms and contributing to proper brain function, but also predispose to detrimental autoimmunity against self-peptides. We analyzed seven HLA loci, either by next-generation-sequencing or Sequence-Specific-Oligonucleotide-Probe, in 48 NS patients and 51 healthy controls from South Sudan. We discovered that NS associates significantly with both protective HLA haplotype: HLA-B*42:01, C*17:01, DRB1*03:02, DQB1*04:02 and DQA1*04:01, and susceptible motif: Ala24, Glu63 and Phe67, in the HLA-B peptide-binding groove. These amino acids create a hydrophobic and sterically closed peptide-binding HLA pocket, favoring proline residue. Our findings suggest that immunogenetic fingerprints in HLA peptide-binding grooves tentatively associate with protection or susceptibility to NS. Accordingly, different HLA molecules may explain why under similar environmental factors, only some children, within the same families, tribes and districts, develop NS, while others do not.


Subject(s)
HLA Antigens/chemistry , HLA Antigens/immunology , Nodding Syndrome/immunology , Adolescent , Adult , Amino Acid Motifs , Autoantibodies/immunology , Case-Control Studies , Child , Child, Preschool , Disease Susceptibility , Female , HLA Antigens/genetics , Humans , Male , Nodding Syndrome/genetics , Nodding Syndrome/prevention & control , Receptors, AMPA/genetics , Receptors, AMPA/immunology , South Sudan , Young Adult
12.
J Neuroimmunol ; 339: 577120, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31790982

ABSTRACT

Our knowledge about genetic factors that drive the worsening of neuromyelitis optica (NMO) is limited. Herein, we analyzed the macrophage migration inhibitory factor (MIF) -173G/C functional polymorphism in NMO patients and controls. Our data reveal that the frequency of the high-expression MIF genotypes (CC/GC) did not differ between the two groups. However, frequency of this genotypes was elevated in patients diagnosed with both optic neuritis and myelitis compared with patients that were diagnosed with only one symptom. Furthermore, patients carrying the CC/CG genotypes had significantly higher disability score. We conclude that MIF is associated with NMO severity rather than susceptibility.


Subject(s)
Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Neuromyelitis Optica/diagnosis , Neuromyelitis Optica/genetics , Polymorphism, Single Nucleotide/genetics , Severity of Illness Index , Adolescent , Adult , Aged , Cohort Studies , Female , Humans , Male , Middle Aged , Young Adult
13.
HLA ; 94(4): 380-381, 2019 10.
Article in English | MEDLINE | ID: mdl-31271260

ABSTRACT

HLA-B*15:539 differs from HLA-B*15:151 by two nucleotide substitutions.


Subject(s)
Alleles , HLA-B15 Antigen/genetics , Exons , High-Throughput Nucleotide Sequencing , Humans , Polymorphism, Genetic , South Sudan
14.
J Neuroinflammation ; 16(1): 14, 2019 Jan 23.
Article in English | MEDLINE | ID: mdl-30683115

ABSTRACT

Neurovascular, autoimmune, and traumatic injuries of the central nervous system (CNS) all have in common an initial acute inflammatory response mediated by influx across the blood-brain barrier of activated mononuclear cells followed by chronic and often progressive disability. Although some anti-inflammatory therapies can reduce cellular infiltration into the initial lesions, there are essentially no effective treatments for the progressive phase. We here review the successful treatment of animal models for four separate neuroinflammatory and neurodegenerative CNS conditions using a single partial MHC class II construct called DRa1-hMOG-35-55 or its newest iteration, DRa1(L50Q)-hMOG-35-55 (DRhQ) that can be administered without a need for class II tissue type matching due to the conserved DRα1 moiety of the drug. These constructs antagonize the cognate TCR and bind with high affinity to their cell-bound CD74 receptor on macrophages and dendritic cells, thereby competitively inhibiting downstream signaling and pro-inflammatory effects of macrophage migration inhibitory factor (MIF) and its homolog, D-dopachrome tautomerase (D-DT=MIF-2) that bind to identical residues of CD74 leading to progressive disease. These effects suggest the existence of a common pathogenic mechanism involving a chemokine-driven influx of activated monocytes into the CNS tissue that can be reversed by parenteral injection of the DRa1-MOG-35-55 constructs that also induce anti-inflammatory macrophages and microglia within the CNS. Due to their ability to block this common pathway, these novel drugs appear to be prime candidates for therapy of a wide range of neuroinflammatory and neurodegenerative CNS conditions.


Subject(s)
Amphetamine-Related Disorders/therapy , Anti-Inflammatory Agents/therapeutic use , Brain Injuries, Traumatic/therapy , Brain Ischemia/therapy , Multiple Sclerosis/therapy , Animals , Chloride-Bicarbonate Antiporters , Humans , Myelin-Oligodendrocyte Glycoprotein/therapeutic use , Peptide Fragments/therapeutic use , Sulfate Transporters
15.
Metab Brain Dis ; 34(1): 153-164, 2019 02.
Article in English | MEDLINE | ID: mdl-30353480

ABSTRACT

Multiple sclerosis (MS) is a demyelinating and degenerative disease of the central nervous system (CNS) with a strong inflammatory component that affects more than 2 million people worldwide (and at least 400,000 in the United States). In MS, macrophage migration inhibitory factor (MIF) and D-dopachrome tautomerase (D-DT) enhance the inflammatory event as a result of their interaction with their cognate receptor CD74. Therefore, the search for new agents aimed at blocking this interaction is critical for therapeutic purposes and will be of paramount importance for the treatment of MS. DRα1-MOG-35-55 constructs have been demonstrated to be effective in the treatment of experimental autoimmune encephalomyelitis (EAE) a mouse model for MS. This effect is directly correlated with the binding to its cell surface receptor, CD74, apparently preventing or blocking the binding of two inflammatory factors, MIF and D-DT. Here we report that a single amino acid substitution (L50Q) in the DRα1 domain of the human and mouse DRα1-MOG-35-55 constructs (notated as DRhQ and DRmQ, respectively) possessed increased affinity for CD74, a greater capacity to block MIF binding, the ability to inhibit pERK1/2 signaling and increased therapeutic activity in mice with EAE. These data suggest that binding affinity for CD74 could serve as an in vitro indicator of biological potency of DRhQ and thus support its possible clinical utility as an effective therapy for MS and perhaps other diseases in which there is an inflammatory reaction driven by MIF and D-DT.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Histocompatibility Antigens Class II/metabolism , Neuroprotective Agents/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/drug effects , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Male , Mice , Neuroprotective Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Signal Transduction/physiology , Treatment Outcome
16.
Metab Brain Dis ; 33(5): 1599-1607, 2018 10.
Article in English | MEDLINE | ID: mdl-29916184

ABSTRACT

A seven day pretreatment course of an oral antibiotic cocktail (Ampicillin, Metronidazole, Neomycin Sulfate, and Vancomycin) was shown to induce changes in peripheral immune regulation and protect mice from signs of experimental autoimmune encephalomyelitis (EAE). To determine if a shorter course of antibiotic pretreatment could also protect the mice from EAE and induce regulatory immune cells, studies were conducted using the same oral antibiotic cocktail for three days. In addition, the CNS was examined to determine the effects of antibiotic pretreatment on EAE disease course and immune modulation within the affected tissue. The shorter three day pretreatment course was also significantly protective against severe EAE in C57BL/6 mice. Moreover, our study found increased frequencies of regulatory cells and a decrease in the frequency of anti-inflammatory macrophages in the spleen of EAE protected mice. Additionally, a chemokine and chemokine receptor array run on mRNA from spinal cords revealed that genes associated with regulatory T cells and macrophage recruitment were strongly upregulated in the antibiotic pretreated mice. Additional RT-PCR data showed genes associated with anti-inflammatory microglia/macrophages were upregulated and pro-inflammatory genes were downregulated. This suggests the macrophages recruited to the spinal cord by chemokines are subsequently polarized toward an anti-inflammatory phenotype. These results lend strong support to the conclusion that a three day course of antibiotic treatment given prior to the induction of severe EAE profoundly protected the mice by inducing regulatory lymphocytes in the periphery and an anti-inflammatory milieu in the affected spinal cord tissue.


Subject(s)
Anti-Bacterial Agents/pharmacology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immunomodulation , Macrophages/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Chemokines/genetics , Down-Regulation/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Lymph Nodes/cytology , Lymph Nodes/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Receptors, Chemokine/genetics , Spinal Cord/drug effects , Spinal Cord/physiopathology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Up-Regulation/genetics
17.
Proc Natl Acad Sci U S A ; 114(40): E8421-E8429, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28923927

ABSTRACT

Little is known about mechanisms that drive the development of progressive multiple sclerosis (MS), although inflammatory factors, such as macrophage migration inhibitory factor (MIF), its homolog D-dopachrome tautomerase (D-DT), and their common receptor CD74 may contribute to disease worsening. Our findings demonstrate elevated MIF and D-DT levels in males with progressive disease compared with relapsing-remitting males (RRMS) and female MS subjects, with increased levels of CD74 in females vs. males with high MS disease severity. Furthermore, increased MIF and D-DT levels in males with progressive disease were significantly correlated with the presence of two high-expression promoter polymorphisms located in the MIF gene, a -794CATT5-8 microsatellite repeat and a -173 G/C SNP. Conversely, mice lacking MIF or D-DT developed less-severe signs of experimental autoimmune encephalomyelitis, a murine model of MS, thus implicating both homologs as copathogenic contributors. These findings indicate that genetically controlled high MIF expression (and D-DT) promotes MS progression in males, suggesting that these two factors are sex-specific disease modifiers and raising the possibility that aggressive anti-MIF treatment of clinically isolated syndrome or RRMS males with a high-expresser genotype might slow or prevent the onset of progressive MS. Additionally, selective targeting of MIF:CD74 signaling might provide an effective, trackable therapeutic approach for MS subjects of both sexes.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Histocompatibility Antigens Class II/metabolism , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/physiology , Multiple Sclerosis/pathology , Severity of Illness Index , Adult , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Female , Histocompatibility Antigens Class II/genetics , Humans , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Knockout , Middle Aged , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Polymorphism, Genetic
18.
J Neuroimmunol ; 310: 51-59, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28778445

ABSTRACT

Sex hormones promote immunoregulatory effects on multiple sclerosis. In the current study we evaluated the composition of the gut microbiota and the mucosal-associated regulatory cells in estrogen or sham treated female mice before and after autoimmune encephalomyelitis (EAE) induction. Treatment with pregnancy levels of estrogen induces changes in the composition and diversity of gut microbiota. Additionally, estrogen prevents EAE-associated changes in the gut microbiota and might promote the enrichment of bacteria that are associated with immune regulation. Our results point to a possible cross-talk between the sex hormones and the gut microbiota, which could promote neuroprotection.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Estrogens/therapeutic use , Intestines/microbiology , Microbiota/drug effects , Mucous Membrane/drug effects , Mucous Membrane/pathology , Animals , Antigens, CD/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Feces/microbiology , Female , Interleukin-10/genetics , Interleukin-10/metabolism , Intestines/drug effects , Leukocytes/drug effects , Lymph Nodes/drug effects , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Spinal Cord/pathology , Time Factors
19.
Metab Brain Dis ; 32(5): 1747-1754, 2017 10.
Article in English | MEDLINE | ID: mdl-28689297

ABSTRACT

Autoimmune diseases including multiple sclerosis predominantly affect females. Although high levels of sex hormones, particularly estrogen (E2), can reduce proinflammatory immune responses, it remains unclear if a lack of endogenous sex hormones might affect treatment with exogenous sex hormones. Pretreatment with E2 almost completely prevents intact female and male mice from developing clinical and histological signs of experimental autoimmune encephalomyelitis (EAE) by promoting various regulatory immune cell phenotypes. To evaluate the effects of exogenous estrogen in the absence of endogenous sex hormones, the current study compared EAE severity and the emergence of different immunoregulatory cell populations after E2 pretreatment of ovariectomized (OVX) female versus male mice. We found that E2 equally protected both OVX females and males from EAE over a 21 day observation period concomitant with reduced total cell numbers in spleen and spinal cord (males only), but enhanced percentages of CD19+CD5+CD1dhi, CD19+CD138+CD44hi and CD19+Tim-1+ Breg cells, CD8+CD122+ Treg cells and CD11b+CD 206+ARG-1+ anti-inflammatory M2-like monocytes/macrophages in both groups. In contrast, E2 decreased the percentage of CD4+CD25+FoxP3+ Treg cells in OVX females but increased these Treg cells in males and intact female mice. These data suggest that with the exception of CD4+CD25+FoxP3+ Treg cells, E2 protection against EAE promotes highly overlapping immunoregulatory subsets in OVX females and males.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Estradiol/therapeutic use , Animals , Antigens, CD/metabolism , B-Lymphocytes, Regulatory/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Estradiol/metabolism , Female , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Ovariectomy , Spinal Cord/pathology , Spleen/pathology , T-Lymphocytes, Regulatory
20.
Cell Immunol ; 318: 49-54, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28606360

ABSTRACT

Stroke is the leading cause of disability in the United States. Sex differences, including smaller infarcts in females and greater involvement of immune-mediated inflammation in males may affect the efficacy of immune-modulating interventions. To address these differences, we sought to identify distinct stroke-modifying mechanisms in female vs. male mice. The current study demonstrated smaller infarcts and increased levels of regulatory CD19+CD5+CD1dhi B10 cells as well as anti-inflammatory CD11b+CD206+ microglia/macrophages in the ipsilateral vs. contralateral hemisphere of female but not male mice undergoing 60min middle cerebral artery occlusion followed by 96h of reperfusion. Moreover, female mice with MCAO had increased total spleen cell numbers but lower B10 levels in spleens. These results elucidate differing sex-dependent regulatory mechanisms that account for diminished stroke severity in females and underscore the need to test immune-modulating therapies for stroke in both males and females.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Brain/immunology , Macrophages/immunology , Sex , Stroke/immunology , Animals , Brain/pathology , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Cerebral Artery/surgery , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...