Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
bioRxiv ; 2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37461569

ABSTRACT

While TGF-ß signaling is essential for microglial function, the cellular source of TGF-ß ligand and its spatial regulation remains unclear in the adult CNS. Our data support that microglia, not astrocytes or neurons, are the primary producers of TGF-ß1 ligands needed for microglial homeostasis. Microglia (MG)-Tgfb1 inducible knockout (iKO) leads to the activation of microglia featuring a dyshomeostatic transcriptomic profile that resembles disease-associated microglia (DAMs), injury-associated microglia, and aged microglia, suggesting that microglial self-produced TGF-ß1 ligands are important in the adult CNS. Interestingly, astrocytes in MG-Tgfb1 iKO mice show a transcriptome profile that closely aligns with A1-like astrocytes. Additionally, using sparse mosaic single-cell microglia iKO of TGF-ß1 ligand, we established an autocrine mechanism for TGF-ß signaling. Importantly MG-Tgfb1 iKO mice show cognitive deficits, supporting that precise spatial regulation of TGF-ß1 ligand derived from microglia is critical for the maintenance of brain homeostasis and normal cognitive function in the adult brain.

2.
Front Immunol ; 13: 887649, 2022.
Article in English | MEDLINE | ID: mdl-36059473

ABSTRACT

Cancer treatment utilizing infusion therapies to enhance the patient's own immune response against the tumor have shown significant functionality in a small subpopulation of patients. Additionally, advances have been made in the utilization of nanotechnology for the treatment of disease. We have previously reported the potent effects of 3-4 daily intravenous infusions of immune modifying poly(lactic-co-glycolic acid) (PLGA) nanoparticles (IMPs; named ONP-302) for the amelioration of acute inflammatory diseases by targeting myeloid cells. The present studies describe a novel use for ONP-302, employing an altered dosing scheme to reprogram myeloid cells resulting in significant enhancement of tumor immunity. ONP-302 infusion decreased tumor growth via the activation of the cGAS/STING pathway within myeloid cells, and subsequently increased NK cell activation via an IL-15-dependent mechanism. Additionally, ONP-302 treatment increased PD-1/PD-L1 expression in the tumor microenvironment, thereby allowing for functionality of anti-PD-1 for treatment in the B16.F10 melanoma tumor model which is normally unresponsive to monotherapy with anti-PD-1. These findings indicate that ONP-302 allows for tumor control via reprogramming myeloid cells via activation of the STING/IL-15/NK cell mechanism, as well as increasing anti-PD-1 response rates.


Subject(s)
Melanoma, Experimental , Nanoparticles , Animals , Humans , Immunotherapy/methods , Interleukin-15 , Melanoma, Experimental/therapy , Membrane Proteins/metabolism , Myeloid Cells/metabolism , Nucleotidyltransferases/metabolism , Tumor Microenvironment
3.
JCI Insight ; 7(5)2022 03 08.
Article in English | MEDLINE | ID: mdl-35104243

ABSTRACT

Systemic sclerosis (SSc) is a chronic, multisystem orphan disease with a highly variable clinical course, high mortality rate, and a poorly understood complex pathogenesis. We have identified an important role for a subpopulation of monocytes and macrophages characterized by surface expression of the scavenger receptor macrophage receptor with collagenous structure (MARCO) in chronic inflammation and fibrosis in SSc and in preclinical disease models. We show that MARCO+ monocytes and macrophages accumulate in lesional skin and lung in topographic proximity to activated myofibroblasts in patients with SSc and in the bleomycin-induced mouse model of SSc. Short-term treatment of mice with a potentially novel nanoparticle, poly(lactic-co-glycolic) acid (PLG), which is composed of a carboxylated, FDA-approved, biodegradable polymer and modulates activation and trafficking of MARCO+ inflammatory monocytes, markedly attenuated bleomycin-induced skin and lung inflammation and fibrosis. Mechanistically, in isolated cells in culture, PLG nanoparticles inhibited TGF-dependent fibrotic responses in vitro. Thus, MARCO+ monocytes are potent effector cells of skin and lung fibrosis and can be therapeutically targeted in SSc using PLG nanoparticles.


Subject(s)
Nanoparticles , Scleroderma, Systemic , Animals , Bleomycin/toxicity , Fibroblasts/metabolism , Fibrosis , Humans , Mice , Monocytes/metabolism , Receptors, Immunologic/metabolism , Scleroderma, Systemic/drug therapy
4.
Cells ; 10(12)2021 12 07.
Article in English | MEDLINE | ID: mdl-34943952

ABSTRACT

We have shown that PLG nanoparticles loaded with peptide antigen can reduce disease in animal models of autoimmunity and in a phase 1/2a clinical trial in celiac patients. Clarifying the mechanisms by which antigen-loaded nanoparticles establish tolerance is key to further adapting them to clinical use. The mechanisms underlying tolerance induction include the expansion of antigen-specific CD4+ regulatory T cells and sequestration of autoreactive cells in the spleen. In this study, we employed nanoparticles loaded with two model peptides, GP33-41 (a CD8 T cell epitope derived from lymphocytic choriomeningitis virus) and OVA323-339 (a CD4 T cell epitope derived from ovalbumin), to modulate the CD8+ and CD4+ T cells from two transgenic mouse strains, P14 and DO11.10, respectively. Firstly, it was found that the injection of P14 mice with particles bearing the MHC I-restricted GP33-41 peptide resulted in the expansion of CD8+ T cells with a regulatory cell phenotype. This correlated with reduced CD4+ T cell viability in ex vivo co-cultures. Secondly, both nanoparticle types were able to sequester transgenic T cells in secondary lymphoid tissue. Flow cytometric analyses showed a reduction in the surface expression of chemokine receptors. Such an effect was more prominently observed in the CD4+ cells rather than the CD8+ cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Celiac Disease/therapy , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Antigens/pharmacology , Antigens, Viral/immunology , Antigens, Viral/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Celiac Disease/genetics , Celiac Disease/immunology , Cell Lineage/drug effects , Cell Lineage/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/pharmacology , Glycoproteins/immunology , Glycoproteins/pharmacology , Humans , Immune Tolerance/drug effects , Mice , Mice, Transgenic , Nanoparticles/chemistry , Ovalbumin/immunology , Ovalbumin/pharmacology , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Peptides/immunology , Peptides/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , T-Lymphocytes, Regulatory/drug effects , Viral Proteins/immunology , Viral Proteins/pharmacology
5.
Cell Rep ; 36(8): 109602, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433042

ABSTRACT

Inappropriate CD4+ T helper (Th) differentiation can compromise host immunity or promote autoimmune disease. To identify disease-relevant regulators of T cell fate, we examined mutations that modify risk for multiple sclerosis (MS), a canonical organ-specific autoimmune disease. This analysis identified a role for Zinc finger E-box-binding homeobox (ZEB1). Deletion of ZEB1 protects against experimental autoimmune encephalitis (EAE), a mouse model of multiple sclerosis (MS). Mechanistically, ZEB1 in CD4+ T cells is required for pathogenic Th1 and Th17 differentiation. Genomic analyses of paired human and mouse expression data elucidated an unexpected role for ZEB1 in JAK-STAT signaling. ZEB1 inhibits miR-101-3p that represses JAK2 expression, STAT3/STAT4 phosphorylation, and subsequent expression of interleukin-17 (IL-17) and interferon gamma (IFN-γ). Underscoring its clinical relevance, ZEB1 and JAK2 downregulation decreases pathogenic cytokines expression in T cells from MS patients. Moreover, a Food and Drug Administration (FDA)-approved JAK2 inhibitor is effective in EAE. Collectively, these findings identify a conserved, potentially targetable mechanism regulating disease-relevant inflammation.


Subject(s)
Cell Differentiation/physiology , Interleukin-17/metabolism , Multiple Sclerosis/pathology , Th17 Cells/immunology , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Cell Differentiation/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Interleukin-17/immunology , Mice , Multiple Sclerosis/immunology , Th1 Cells/immunology , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/immunology
6.
Front Immunol ; 11: 571897, 2020.
Article in English | MEDLINE | ID: mdl-33123148

ABSTRACT

Multiple Sclerosis (MS) is characterized by immune cell infiltration to the central nervous system (CNS) as well as loss of myelin. Characterization of the cells in lesions of MS patients revealed an important accumulation of myeloid cells such as macrophages and dendritic cells (DCs). Data from the experimental autoimmune encephalomyelitis (EAE) model of MS supports the importance of peripheral myeloid cells in the disease pathology. However, the majority of MS therapies focus on lymphocytes. As we will discuss in this review, multiple strategies are now in place to target myeloid cells in clinical trials. These strategies have emerged from data in both human and mouse studies. We discuss strategies targeting myeloid cell migration, growth factors and cytokines, biological functions (with a focus on miRNAs), and immunological activities (with a focus on nanoparticles).


Subject(s)
Central Nervous System/physiology , Encephalomyelitis, Autoimmune, Experimental/therapy , Multiple Sclerosis/therapy , Myeloid Cells/physiology , Neurogenic Inflammation/therapy , Animals , Cell Movement , Cytokines/metabolism , Humans , Mice , MicroRNAs/genetics , Nanoparticles
7.
Nature ; 582(7812): 416-420, 2020 06.
Article in English | MEDLINE | ID: mdl-32499641

ABSTRACT

Regulatory T (Treg) cells are required to control immune responses and maintain homeostasis, but are a significant barrier to antitumour immunity1. Conversely, Treg instability, characterized by loss of the master transcription factor Foxp3 and acquisition of proinflammatory properties2, can promote autoimmunity and/or facilitate more effective tumour immunity3,4. A comprehensive understanding of the pathways that regulate Foxp3 could lead to more effective Treg therapies for autoimmune disease and cancer. The availability of new functional genetic tools has enabled the possibility of systematic dissection of the gene regulatory programs that modulate Foxp3 expression. Here we developed a CRISPR-based pooled screening platform for phenotypes in primary mouse Treg cells and applied this technology to perform a targeted loss-of-function screen of around 500 nuclear factors to identify gene regulatory programs that promote or disrupt Foxp3 expression. We identified several modulators of Foxp3 expression, including ubiquitin-specific peptidase 22 (Usp22) and ring finger protein 20 (Rnf20). Usp22, a member of the deubiquitination module of the SAGA chromatin-modifying complex, was revealed to be a positive regulator that stabilized Foxp3 expression; whereas the screen suggested that Rnf20, an E3 ubiquitin ligase, can serve as a negative regulator of Foxp3. Treg-specific ablation of Usp22 in mice reduced Foxp3 protein levels and caused defects in their suppressive function that led to spontaneous autoimmunity but protected against tumour growth in multiple cancer models. Foxp3 destabilization in Usp22-deficient Treg cells could be rescued by ablation of Rnf20, revealing a reciprocal ubiquitin switch in Treg cells. These results reveal previously unknown modulators of Foxp3 and demonstrate a screening method that can be broadly applied to discover new targets for Treg immunotherapies for cancer and autoimmune disease.


Subject(s)
CRISPR-Cas Systems , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmunity/immunology , Cells, Cultured , Forkhead Transcription Factors/biosynthesis , Gene Editing , Gene Expression Regulation , Humans , Immunotherapy , Male , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/prevention & control , Protein Stability , Reproducibility of Results , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
8.
mBio ; 11(3)2020 05 12.
Article in English | MEDLINE | ID: mdl-32398314

ABSTRACT

Ocular herpes simplex virus 1 (HSV-1) infection leads to an immunopathogenic disease called herpes stromal keratitis (HSK), in which CD4+ T cell-driven inflammation contributes to irreversible damage to the cornea. Herpesvirus entry mediator (HVEM) is an immune modulator that activates stimulatory and inhibitory cosignals by interacting with its binding partners, LIGHT (TNFSF14), BTLA (B and T lymphocyte attenuator), and CD160. We have previously shown that HVEM exacerbates HSK pathogenesis, but the involvement of its binding partners and its connection to the pathogenic T cell response have not been elucidated. In this study, we investigated the role of HVEM and its binding partners in mediating the T cell response using a murine model of ocular HSV-1 infection. By infecting mice lacking the binding partners, we demonstrated that multiple HVEM binding partners were required for HSK pathogenesis. Surprisingly, while LIGHT-/-, BTLA-/-, and CD160-/- mice did not show differences in disease compared to wild-type mice, BTLA-/- LIGHT-/- and CD160-/- LIGHT-/- double knockout mice showed attenuated disease characterized by decreased clinical symptoms, increased retention of corneal sensitivity, and decreased infiltrating leukocytes in the cornea. We determined that the attenuation of disease in HVEM-/-, BTLA-/- LIGHT-/-, and CD160-/- LIGHT-/- mice correlated with a decrease in gamma interferon (IFN-γ)-producing CD4+ T cells. Together, these results suggest that HVEM cosignaling through multiple binding partners induces a pathogenic Th1 response to promote HSK. This report provides new insight into the mechanism of HVEM in HSK pathogenesis and highlights the complexity of HVEM signaling in modulating the immune response following ocular HSV-1 infection.IMPORTANCE Herpes simplex virus 1 (HSV-1), a ubiquitous human pathogen, is capable of causing a progressive inflammatory ocular disease called herpes stromal keratitis (HSK). HSV-1 ocular infection leads to persistent inflammation in the cornea resulting in outcomes ranging from significant visual impairment to complete blindness. Our previous work showed that herpesvirus entry mediator (HVEM) promotes the symptoms of HSK independently of viral entry and that HVEM expression on CD45+ cells correlates with increased infiltration of leukocytes into the cornea during the chronic inflammatory phase of the disease. Here, we elucidated the role of HVEM in the pathogenic Th1 response following ocular HSV-1 infection and the contribution of HVEM binding partners in HSK pathogenesis. Investigating the molecular mechanisms of HVEM in promoting corneal inflammation following HSV-1 infection improves our understanding of potential therapeutic targets for HSK.


Subject(s)
Herpesvirus 1, Human/physiology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/pathology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Virus Internalization , Animals , Cornea/immunology , Cornea/pathology , Cornea/virology , Disease Models, Animal , Female , Herpesvirus 1, Human/immunology , Host Microbial Interactions/immunology , Inflammation , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 14/immunology , Signal Transduction , T-Lymphocytes/immunology
9.
Ann Neurol ; 87(3): 442-455, 2020 03.
Article in English | MEDLINE | ID: mdl-31925846

ABSTRACT

OBJECTIVE: There are currently no definitive disease-modifying therapies for traumatic brain injury (TBI). In this study, we present a strong therapeutic candidate for TBI, immunomodulatory nanoparticles (IMPs), which ablate a specific subset of hematogenous monocytes (hMos). We hypothesized that prevention of infiltration of these cells into brain acutely after TBI would attenuate secondary damage and preserve anatomic and neurologic function. METHODS: IMPs, composed of US Food and Drug Administration-approved 500nm carboxylated-poly(lactic-co-glycolic) acid, were infused intravenously into wild-type C57BL/6 mice following 2 different models of experimental TBI, controlled cortical impact (CCI), and closed head injury (CHI). RESULTS: IMP administration resulted in remarkable preservation of both tissue and neurological function in both CCI and CHI TBI models in mice. After acute treatment, there was a reduction in the number of immune cells infiltrating into the brain, mitigation of the inflammatory status of the infiltrating cells, improved electrophysiologic visual function, improved long-term motor behavior, reduced edema formation as assessed by magnetic resonance imaging, and reduced lesion volumes on anatomic examination. INTERPRETATION: Our findings suggest that IMPs are a clinically translatable acute intervention for TBI with a well-defined mechanism of action and beneficial anatomic and physiologic preservation and recovery. Ann Neurol 2020;87:442-455.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Immunologic Factors/therapeutic use , Administration, Intravenous , Animals , Brain/immunology , Brain/pathology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Cell Movement/drug effects , Edema/complications , Edema/drug therapy , Immunologic Factors/administration & dosage , Immunologic Factors/chemistry , Magnetic Resonance Imaging , Male , Mice , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neuroimaging , Recovery of Function/drug effects
10.
Bio Protoc ; 10(11): e3644, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-33659314

ABSTRACT

Methods to test both the functionality and mechanism of action for human recombinant proteins and antibodies in vitro have been limited by multiple factors. To test the functionality of a recombinant protein or antibody, the receptor, the receptor-associated ligand, or both must be expressed by the cells present within the in vitro culture. While the use of transfected cell lines can circumvent this gap, the use of transfected cell lines does not allow for studying the native signaling pathway(s) modulated by the specific recombinant protein or antibody in primary cells. The present protocol utilizes sort purified CD14+ monocytes and T cells, both CD4+ T cells and CD8+ T cells, from healthy donors in a co-culture system. This methodology is particularly relevant for testing recombinant proteins or antibodies that are putative therapeutics for the treatment of autoimmune disease and cancer. While the current protocol focuses on co-cultures containing B7-H4 expressing monocytes plus either autologous CD4+ T cells or CD8+ T cells, the protocol can be modified for the user's specific needs.

11.
Am J Physiol Heart Circ Physiol ; 318(1): H116-H123, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31809213

ABSTRACT

In humans, loss of central tolerance for the cardiac self-antigen α-myosin heavy chain (α-MHC) leads to circulation of cardiac autoreactive T cells and renders the heart susceptible to autoimmune attack after acute myocardial infarction (MI). MI triggers profound tissue damage, releasing danger signals and self-antigen by necrotic cardiomyocytes, which lead to recruitment of inflammatory monocytes. We hypothesized that excessive inflammation by monocytes contributes to the initiation of adaptive immune responses to cardiac self-antigen. Using an experimental model of MI in α-MHC-mCherry reporter mice, which specifically express mCherry in cardiomyocytes, we detected α-MHC antigen in myeloid cells in the heart-draining mediastinal lymph node (MLN) 7 days after MI. To test whether monocytes were required for cardiac self-antigen trafficking to the MLN, we blocked monocyte recruitment with a C-C motif chemokine receptor type 2 (CCR2) antagonist or immune modifying nanoparticles (IMP). Blockade of monocyte recruitment reduced α-MHC antigen detection in the MLN after MI. Intramyocardial injection of the model antigen ovalbumin into OT-II transgenic mice demonstrated the requirement for monocytes in antigen trafficking and T-cell activation in the MLN. Finally, in nonobese diabetic mice, which are prone to postinfarction autoimmunity, blockade of monocyte recruitment reduced α-MHC-specific responses leading to improved tissue repair and ventricular function 28 days after MI. Taken together, these data support a role for monocytes in the onset of pathological cardiac autoimmunity following MI and suggest that therapeutic targeting of monocytes may mitigate postinfarction autoimmunity in humans.NEW & NOTEWORTHY Our study newly identifies a role for inflammatory monocytes in priming an autoimmune T-cell response after myocardial infarction. Select inhibition of monocyte recruitment to the infarct prevents trafficking of cardiac self-antigen and activation of cardiac myosin reactive T cells in the heart-draining lymph node. Therapeutic targeting of inflammatory monocytes may limit autoimmune responses to improve cardiac remodeling and preserve left ventricular function after myocardial infarction.


Subject(s)
Adaptive Immunity , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Cell Communication , Lymphocyte Activation , Monocytes/immunology , Myocardial Infarction/immunology , Myocardium/immunology , Animals , Antigens, Ly/immunology , Antigens, Ly/metabolism , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Monocytes/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myosin Heavy Chains/immunology , Myosin Heavy Chains/metabolism , Signal Transduction , Ventricular Function, Left , Ventricular Remodeling
12.
J Immunol ; 201(3): 897-907, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29898965

ABSTRACT

The potent immune regulatory function of an agonistic B7-H4-Ig fusion protein (B7-H4Ig) has been demonstrated in multiple experimental autoimmune models; however, the identity of a functional B7-H4 receptor remained unknown. The biological activity of B7-H4 is associated with decreased inflammatory CD4+ T cell responses as supported by a correlation between B7-H4-expressing tumor-associated macrophages and Foxp3+ T cells within the tumor microenvironment. Recent data indicate that members of the semaphorin (Sema)/plexin/neuropilin (Nrp) family of proteins both positively and negatively modulate immune cell function. In this study, we show that B7-H4 binds the soluble Sema family member Sema3a. Additionally, B7-H4Ig-induced inhibition of inflammatory CD4+ T cell responses is lost in both Sema3a functional mutant mice and mice lacking Nrp-1 expression in Foxp3+ T cells. These findings indicate that B7-H4Ig binds to Sema3a, which acts as a functional bridge to stimulate an Nrp-1/Plexin A4 heterodimer to form a functional immunoregulatory receptor complex resulting in increased levels of phosphorylated PTEN and enhanced regulatory CD4+ T cell number and function.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Neuropilin-1/metabolism , Receptors, Cell Surface/metabolism , Semaphorin-3A/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Female , Forkhead Transcription Factors/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/metabolism , Tumor Microenvironment/immunology
13.
J Exp Med ; 215(4): 1169-1186, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29487082

ABSTRACT

The pathophysiology of drug-resistant pediatric epilepsy is unknown. Flow cytometric analysis of inflammatory leukocytes in resected brain tissues from 29 pediatric patients with genetic (focal cortical dysplasia) or acquired (encephalomalacia) epilepsy demonstrated significant brain infiltration of blood-borne inflammatory myeloid cells and memory CD4+ and CD8+ T cells. Significantly, proinflammatory (IL-17- and GM-CSF-producing) γδ T cells were concentrated in epileptogenic lesions, and their numbers positively correlated with disease severity. Conversely, numbers of regulatory T (T reg) cells inversely correlated with disease severity. Correspondingly, using the kainic acid model of status epilepticus, we show ameliorated seizure activity in both γδ T cell- and IL-17RA-deficient mice and in recipients of T reg cells, whereas T reg cell depletion heightened seizure severity. Moreover, both IL-17 and GM-CSF induced neuronal hyperexcitability in brain slice cultures. These studies support a major pathological role for peripherally derived innate and adaptive proinflammatory immune responses in the pathogenesis of intractable epilepsy and suggest testing of immunomodulatory therapies.


Subject(s)
Drug Resistant Epilepsy/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity/drug effects , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Brain/pathology , Cell Survival/drug effects , Child , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Drug Resistant Epilepsy/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunity, Innate/drug effects , Inflammation/pathology , Interleukin-17/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neurons/drug effects , Neurons/pathology , T-Lymphocytes, Regulatory/drug effects
14.
Nat Commun ; 8(1): 1319, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29105655

ABSTRACT

Metastatic cancers produce exosomes that condition pre-metastatic niches in remote microenvironments to favor metastasis. In contrast, here we show that exosomes from poorly metastatic melanoma cells can potently inhibit metastasis to the lung. These "non-metastatic" exosomes stimulate an innate immune response through the expansion of Ly6Clow patrolling monocytes (PMo) in the bone marrow, which then cause cancer cell clearance at the pre-metastatic niche, via the recruitment of NK cells and TRAIL-dependent killing of melanoma cells by macrophages. These events require the induction of the Nr4a1 transcription factor and are dependent on pigment epithelium-derived factor (PEDF) on the outer surface of exosomes. Importantly, exosomes isolated from patients with non-metastatic primary melanomas have a similar ability to suppress lung metastasis. This study thus demonstrates that pre-metastatic tumors produce exosomes, which elicit a broad range of PMo-reliant innate immune responses via trigger(s) of immune surveillance, causing cancer cell clearance at the pre-metastatic niche.


Subject(s)
Exosomes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Monocytes/immunology , Animals , Cell Differentiation/immunology , Eye Proteins/immunology , Female , Humans , Immunity, Innate , Immunologic Surveillance , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Macrophages/immunology , Macrophages/pathology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Monocytes/pathology , Nerve Growth Factors/immunology , Phagocytosis/immunology , Serpins/immunology , Tumor Microenvironment/immunology
15.
Neurobiol Dis ; 108: 73-82, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28823935

ABSTRACT

Intravenously infused synthetic 500nm nanoparticles composed of poly(lactide-co-glycolide) are taken up by blood-borne inflammatory monocytes via a macrophage scavenger receptor (macrophage receptor with collagenous structure), and the monocytes no longer traffic to sites of inflammation. Intravenous administration of the nanoparticles after experimental spinal cord injury in mice safely and selectively limited infiltration of hematogenous monocytes into the injury site. The nanoparticles did not bind to resident microglia, and did not change the number of microglia in the injured spinal cord. Nanoparticle administration reduced M1 macrophage polarization and microglia activation, reduced levels of inflammatory cytokines, and markedly reduced fibrotic scar formation without altering glial scarring. These findings thus implicate early-infiltrating hematogenous monocytes as highly selective contributors to fibrosis that do not play an indispensable role in gliosis after SCI. Further, the nanoparticle treatment reduced accumulation of chondroitin sulfate proteoglycans, increased axon density inside and caudal to the lesion site, and significantly improved functional recovery after both moderate and severe injuries to the spinal cord. These data provide further evidence that hematogenous monocytes contribute to inflammatory damage and fibrotic scar formation after spinal cord injury in mice. Further, since the nanoparticles are simple to administer intravenously, immunologically inert, stable at room temperature, composed of an FDA-approved material, and have no known toxicity, these findings suggest that the nanoparticles potentially offer a practical treatment for human spinal cord injury.


Subject(s)
Immunologic Factors/administration & dosage , Nanoparticles/administration & dosage , Polyglactin 910/administration & dosage , Spinal Cord Injuries/drug therapy , Administration, Intravenous , Animals , Axons/drug effects , Axons/immunology , Axons/pathology , Cell Size , Chondroitin Sulfate Proteoglycans/metabolism , Cicatrix/drug therapy , Cicatrix/immunology , Cicatrix/pathology , Disease Models, Animal , Female , Fibrosis/drug therapy , Fibrosis/immunology , Fibrosis/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , Motor Activity/drug effects , Recovery of Function/drug effects , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
16.
J Autoimmun ; 84: 1-11, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28641926

ABSTRACT

In multiple sclerosis (MS), there is a growing interest in inhibiting the pro-inflammatory effects of granulocyte-macrophage colony-stimulating factor (GM-CSF). We sought to evaluate the therapeutic potential and underlying mechanisms of GM-CSF receptor alpha (Rα) blockade in animal models of MS. We show that GM-CSF signaling inhibition at peak of chronic experimental autoimmune encephalomyelitis (EAE) results in amelioration of disease progression. Similarly, GM-CSF Rα blockade in relapsing-remitting (RR)-EAE model prevented disease relapses and inhibited T cell responses specific for both the inducing and spread myelin peptides, while reducing activation of mDCs and inflammatory monocytes. In situ immunostaining of lesions from human secondary progressive MS (SPMS), but not primary progressive MS patients shows extensive recruitment of GM-CSF Rα+ myeloid cells. Collectively, this study reveals a pivotal role of GM-CSF in disease relapses and the benefit of GM-CSF Rα blockade as a potential novel therapeutic approach for treatment of RRMS and SPMS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Central Nervous System/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Myeloid Cells/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Animals , Autoimmunity , Cell Differentiation , Cell Movement , Cells, Cultured , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Humans , Immunosuppression Therapy , Male , Mice , Mice, Inbred C57BL , Middle Aged , Molecular Targeted Therapy , Multiple Sclerosis/therapy , Myelin Sheath/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Signal Transduction
17.
Invest Ophthalmol Vis Sci ; 58(1): 282-291, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28114589

ABSTRACT

Purpose: To determine cellular and temporal expression patterns of herpes virus entry mediator (HVEM, Tnfrsf14) in the murine cornea during the course of herpes simplex virus 1 (HSV-1) infection, the impact of this expression on pathogenesis, and whether alterations in HVEM or downstream HVEM-mediated effects ameliorate corneal disease. Methods: Corneal HVEM levels were assessed in C57BL/6 mice after infection with HSV-1(17). Leukocytic infiltrates and corneal sensitivity loss were measured in the presence, global absence (HVEM knockout [KO] mice; Tnfrsf14-/-), or partial absence of HVEM (HVEM conditional KO). Effects of immune-modifying nanoparticles (IMPs) on viral replication, corneal sensitivity, and corneal infiltrates were measured. Results: Corneal HVEM+ populations, particularly monocytes/macrophages during acute infection (3 days post infection [dpi]) and polymorphonuclear neutrophils (PMN) during the chronic inflammatory phase (14 dpi), increased after HSV-1 infection. Herpes virus entry mediator increased leukocytes in the cornea and corneal sensitivity loss. Ablation of HVEM from CD45+ cells, or intravenous IMP therapy, reduced infiltrates in the chronic phase and maintained corneal sensitivity. Conclusions: Herpes virus entry mediator was expressed on two key populations: corneal monocytes/macrophages and PMNs. Herpes virus entry mediator promoted the recruitment of myeloid cells to the cornea in the chronic phase. Herpes virus entry mediator-associated corneal sensitivity loss preceded leukocytic infiltration, suggesting it may play an active role in recruitment. We propose that HVEM on resident corneal macrophages increases nerve damage and immune cell invasion, and we showed that prevention of late-phase infiltration of PMN and CD4+ T cells by IMP therapy improved clinical symptoms and mortality and reduced corneal sensitivity loss caused by HSV-1.


Subject(s)
Eye Infections, Viral/therapy , Herpesviridae Infections/therapy , Herpesvirus 1, Human/pathogenicity , Immunotherapy/methods , Keratitis, Herpetic/therapy , Lactic Acid/administration & dosage , Polyglycolic Acid/administration & dosage , Receptors, Tumor Necrosis Factor, Member 14/administration & dosage , Animals , Biocompatible Materials/administration & dosage , Cornea/metabolism , Cornea/pathology , Cornea/virology , Disease Models, Animal , Eye Infections, Viral/diagnosis , Eye Infections, Viral/immunology , Flow Cytometry , Herpesviridae Infections/diagnosis , Herpesviridae Infections/immunology , Immunity, Cellular , Immunohistochemistry , Injections, Intravenous , Keratitis, Herpetic/diagnosis , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer
18.
Proc Natl Acad Sci U S A ; 113(18): 5059-64, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27091976

ABSTRACT

Specific immunotherapy (SIT) is the most widely used treatment for allergic diseases that directly targets the T helper 2 (Th2) bias underlying allergy. However, the most widespread clinical applications of SIT require a long period of dose escalation with soluble antigen (Ag) and carry a significant risk of adverse reactions, particularly in highly sensitized patients who stand to benefit most from a curative treatment. Thus, the development of safer, more efficient methods to induce Ag-specific immune tolerance is critical to advancing allergy treatment. We hypothesized that antigen-associated nanoparticles (Ag-NPs), which we have used to prevent and treat Th1/Th17-mediated autoimmune disease, would also be effective for the induction of tolerance in a murine model of Th2-mediated ovalbumin/alum-induced allergic airway inflammation. We demonstrate here that antigen-conjugated polystyrene (Ag-PS) NPs, although effective for the prophylactic induction of tolerance, induce anaphylaxis in presensitized mice. Antigen-conjugated NPs made of biodegradable poly(lactide-co-glycolide) (Ag-PLG) are similarly effective prophylactically, are well tolerated by sensitized animals, but only partially inhibit Th2 responses when administered therapeutically. PLG NPs containing encapsulated antigen [PLG(Ag)], however, were well tolerated and effectively inhibited Th2 responses and airway inflammation both prophylactically and therapeutically. Thus, we illustrate progression toward PLG(Ag) as a biodegradable Ag carrier platform for the safe and effective inhibition of allergic airway inflammation without the need for nonspecific immunosuppression in animals with established Th2 sensitization.


Subject(s)
Antigens/administration & dosage , Antigens/immunology , Asthma/immunology , Asthma/therapy , Drug Implants/administration & dosage , Nanocapsules/administration & dosage , Th2 Cells/immunology , Absorbable Implants , Animals , Asthma/diagnosis , Female , Immunization/methods , Injections, Intravenous , Mice , Mice, Inbred BALB C , Particle Size , Polyglactin 910/administration & dosage , Polyglactin 910/chemistry , Th2 Cells/drug effects , Treatment Outcome
19.
J Immunol ; 196(4): 1455-1459, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26783338

ABSTRACT

Myeloid cells play a crucial role in the induction and sustained inflammation in neuroinflammatory disorders, such as multiple sclerosis. miR-223, a myeloid cell-specific microRNA, is one of the most upregulated microRNAs in multiple sclerosis patients. We demonstrate that miR-223-knockout mice display significantly reduced active and adoptive-transfer experimental autoimmune encephalomyelitis that is characterized by reduced numbers of myeloid dendritic cells (mDCs) and Th17 cells in the CNS. Knockout mDCs have increased PD-L1 and decreased IL-1ß, IL-6, and IL-23 expression, as well as a reduced capacity to drive Th17, but not Th1, cell differentiation. Thus, miR-223 controls mDC-induced activation of pathologic Th17 responses during autoimmune inflammation.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , MicroRNAs/metabolism , Th17 Cells/immunology , Animals , Cell Differentiation , Cytokines/biosynthesis , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Th1 Cells/immunology
20.
Methods Mol Biol ; 1304: 145-60, 2016.
Article in English | MEDLINE | ID: mdl-25005074

ABSTRACT

Experimental autoimmune encephalitis (EAE), the animal model of multiple sclerosis (MS), has provided significant insight into the mechanisms that initiate and drive autoimmunity. Several central nervous system proteins and peptides have been used to induce disease, in a number of different mouse strains, to model the diverse clinical presentations of MS. In this chapter, we detail the materials and methods used to induce active and adoptive EAE. We focus on disease induction in the SJL/J, C57BL/6, and BALB/c mouse strains, using peptides derived from proteolipid protein, myelin basic protein, and myelin oligodendrocyte glycoprotein. We also include a protocol for the isolation of leukocytes from the spinal cord and brain for flow cytometric analysis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Leukocytes/immunology , Leukocytes/pathology , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...