Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
PLoS Pathog ; 13(4): e1006309, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28384349

ABSTRACT

Early responses mounted by both tissue-resident and recruited innate immune cells are essential for host defense against bacterial pathogens. In particular, both neutrophils and Ly6Chi monocytes are rapidly recruited to sites of infection. While neutrophils and monocytes produce bactericidal molecules, such as reactive nitrogen and oxygen species, both cell types are also capable of synthesizing overlapping sets of cytokines important for host defense. Whether neutrophils and monocytes perform redundant or non-redundant functions in the generation of anti-microbial cytokine responses remains elusive. Here, we sought to define the contributions of neutrophils and Ly6Chi monocytes to cytokine production and host defense during pulmonary infection with Legionella pneumophila, responsible for the severe pneumonia Legionnaires' disease. We found that both neutrophils and monocytes are critical for host defense against L. pneumophila. Both monocytes and neutrophils contribute to maximal IL-12 and IFNγ responses, and monocytes are also required for TNF production. Moreover, natural killer (NK) cells, NKT cells, and γδ T cells are sources of IFNγ, and monocytes direct IFNγ production by these cell types. Thus, neutrophils and monocytes cooperate in eliciting an optimal cytokine response that promotes effective control of bacterial infection.


Subject(s)
Antigens, Ly/immunology , Cytokines/immunology , Legionella pneumophila/physiology , Legionnaires' Disease/immunology , Lung/microbiology , Monocytes/immunology , Neutrophils/immunology , Animals , Antigens, Ly/genetics , Cytokines/genetics , Humans , Legionnaires' Disease/genetics , Legionnaires' Disease/microbiology , Legionnaires' Disease/prevention & control , Lung/immunology , Mice , Mice, Inbred C57BL
2.
Clin Immunol ; 185: 100-108, 2017 12.
Article in English | MEDLINE | ID: mdl-27570219

ABSTRACT

Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease that can affect multiple end organs. Kidney and brain are two of the organs most commonly involved in SLE. Past studies have suggested the importance of macrophages in the pathogenesis of lupus nephritis (LN). Furthermore, as the immune effectors of the brain, microglia have been implicated in pathways leading to neuropsychiatric SLE (NPSLE). We depleted macrophages and microglia using GW2580, a small colony stimulating factor-1 receptor (CSF-1R) kinase inhibitor, in MRL-lpr/lpr (MRL/lpr) mice, a classic murine lupus model that displays features of both LN and NPSLE. Treatment was initiated before the onset of disease, and mice were followed for the development of LN and neurobehavioral dysfunction throughout the study. Treatment with GW2580 significantly ameliorated kidney disease, as evidenced by decreased proteinuria, BUN, and improved renal histopathology, despite equivalent levels of IgG and C3 deposition in the kidneys of treated and control mice. We were able to confirm macrophage depletion within the kidney via IBA-1 staining. Furthermore, we observed specific improvement in the depression-like behavioral deficit of MRL/lpr mice with GW2580 treatment. Circulating antibody and autoantibody levels were, however, not affected. These results provide additional support for the role of macrophages as a potentially valuable therapeutic target in SLE. Inhibiting CSF-1 receptor signaling would be more targeted than current immunosuppressive therapies, and may hold promise for the treatment of renal and neuropsychiatric end organ disease manifestations.


Subject(s)
Anisoles/therapeutic use , Lupus Nephritis/drug therapy , Lupus Vasculitis, Central Nervous System/drug therapy , Macrophages/drug effects , Pyrimidines/therapeutic use , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Anisoles/pharmacology , Antibodies, Antinuclear/immunology , Brain/drug effects , Brain/immunology , Chromatin/immunology , Complement System Proteins/immunology , Cytokines/immunology , Depression/drug therapy , Depression/immunology , Depression/pathology , Disease Models, Animal , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Locomotion/drug effects , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lupus Nephritis/psychology , Lupus Vasculitis, Central Nervous System/immunology , Lupus Vasculitis, Central Nervous System/pathology , Lupus Vasculitis, Central Nervous System/psychology , Macrophages/immunology , Mice, Inbred MRL lpr , Pyrimidines/pharmacology , Spatial Memory/drug effects
3.
Scand J Gastroenterol ; 52(4): 420-424, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28040992

ABSTRACT

OBJECTIVE: Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) may be involved in the pathogenesis of inflammatory bowel disease. The aim was to investigate if TWEAK may reflect disease activity in inflammatory bowel disease. MATERIALS AND METHODS: In this cohort study, 139 consecutive patients with newly diagnosed and previously untreated inflammatory bowel disease - 95 with ulcerative colitis (UC) and 44 with Crohn's disease (CD) - underwent colonoscopy. Disease activity was assessed by the Mayo score and the Mayo endoscopic score (MES) for UC, or the Simple Endoscopic Score (SES) for CD. Serum C-reactive protein (CRP) and fecal calprotectin were measured in IBD patients, as were plasma TWEAK levels in patients and 85 healthy subjects. Associations between TWEAK levels and disease activity markers were explored. RESULTS: In the total IBD group, the median (interquartile range) TWEAK level was 430 pg/ml (109-6570), in UC 502 pg/ml (109-4547) and in CD patients 352 pg/ml (101-9179), respectively. Healthy subjects had a median (IQR) TWEAK of 307 pg/ml (63-3492). There were no significant differences in TWEAK levels between the total IBD group and healthy control subjects, nor between UC and CD, or between UC/CD and healthy subjects. Furthermore, we found no significant associations between Mayo scores, MES-UC, SES-CD, CRP, and fecal calprotectin with plasma TWEAK levels. CONCLUSIONS: Plasma TWEAK levels do not reflect disease activity or the grade of inflammation in patients with newly diagnosed inflammatory bowel disease. NCT01551563.


Subject(s)
Colitis, Ulcerative/blood , Crohn Disease/blood , Tumor Necrosis Factors/blood , Adolescent , Adult , Aged , Biomarkers/blood , C-Reactive Protein/analysis , Case-Control Studies , Colonoscopy , Cytokine TWEAK , Feces/chemistry , Female , Humans , Leukocyte L1 Antigen Complex/analysis , Linear Models , Male , Middle Aged , Norway , Prospective Studies , Severity of Illness Index , Young Adult
4.
J Neuroinflammation ; 13(1): 73, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27055816

ABSTRACT

BACKGROUND: Neuropsychiatric lupus (NPSLE) can be one of the earliest clinical manifestations in human lupus. However, its mechanisms are not fully understood. In lupus, a compromised blood-brain barrier may allow for the passage of circulating autoantibodies into the brain, where they can induce neuropsychiatric abnormalities including depression-like behavior and cognitive abnormalities. The purpose of this study was to determine the role of B cells and/or autoantibodies in the pathogenesis of murine NPSLE. METHODS: We evaluated neuropsychiatric manifestations, brain pathology, and cytokine expression in constitutively (JhD/MRL/lpr) and conditionally (hCD20-DTA/MRL/lpr, inducible by tamoxifen) B cell-depleted mice as compared to MRL/lpr lupus mice. RESULTS: We found that autoantibody levels were negligible (JhD/MRL/lpr) or significantly reduced (hCD20-DTA/MRL/lpr) in the serum and cerebrospinal fluid, respectively. Nevertheless, both JhD/MRL/lpr and hCD20-DTA/MRL/lpr mice showed profound depression-like behavior, which was no different from MRL/lpr mice. Cognitive deficits were also observed in both JhD/MRL/lpr and hCD20-DTA/MRL/lpr mice, similar to those exhibited by MRL/lpr mice. Furthermore, although some differences were dependent on the timing of depletion, central features of NPSLE in the MRL/lpr strain including increased blood-brain barrier permeability, brain cell apoptosis, and upregulated cytokine expression persisted in B cell-deficient and B cell-depleted mice. CONCLUSIONS: Our study surprisingly found that B cells and/or autoantibodies are not required for key features of neuropsychiatric disease in murine NPSLE.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/immunology , Cognition Disorders/etiology , Cognition Disorders/immunology , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Animals , Behavior, Animal , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , In Situ Nick-End Labeling , Mice , Mice, Inbred MRL lpr , Real-Time Polymerase Chain Reaction
5.
Exp Dermatol ; 25(12): 969-976, 2016 12.
Article in English | MEDLINE | ID: mdl-27305603

ABSTRACT

The cytokine TNF-like weak inducer of apoptosis (TWEAK) and its receptor Fn14 are involved in cell survival and cytokine production. The TWEAK/Fn14 pathway plays a role in the pathogenesis of spontaneous cutaneous lesions in the MRL/lpr lupus strain; however, the role of TWEAK/Fn14 in disease induced by ultraviolet B (UVB) irradiation has not been explored. MRL/lpr Fn14 knockout (KO) was compared to MRL/lpr Fn14 wild-type (WT) mice following exposure to UVB. We found that irradiated MRL/lpr KO mice had significantly attenuated cutaneous disease when compared to their WT counterparts. There were also fewer infiltrating immune cells (CD3+ , IBA-1+ and NGAL+ ) in the UVB-exposed skin of MRL/lpr Fn14KO mice, as compared to Fn14WT. Furthermore, we identified several macrophage-derived proinflammatory chemokines with elevated expression in MRL/lpr mice after UV exposure. Depletion of macrophages, using a CSF-1R inhibitor, was found to be protective against the development of skin lesions after UVB exposure. In combination with the phenotype of the MRL/lpr Fn14KO mice, these findings indicate a critical role for Fn14 and recruited macrophages in UVB-triggered cutaneous lupus. Our data strongly suggest that TWEAK/Fn14 signalling is important in the pathogenesis of UVB-induced cutaneous disease manifestations in the MRL/lpr model of lupus and further support this pathway as a possible target for therapeutic intervention.


Subject(s)
Lupus Erythematosus, Systemic/complications , Photosensitivity Disorders/etiology , Skin/radiation effects , TWEAK Receptor/metabolism , Ultraviolet Rays/adverse effects , Animals , Apoptosis , Chemokines/metabolism , Cytokine TWEAK/metabolism , Female , Lipocalin-2/metabolism , Mice, Knockout , Photosensitivity Disorders/metabolism , Skin/immunology , Skin/metabolism
6.
Brain Behav Immun ; 54: 27-37, 2016 May.
Article in English | MEDLINE | ID: mdl-26721417

ABSTRACT

Fn14, the sole known signaling receptor for the TNF family member TWEAK, is inducibly expressed in the central nervous system (CNS) in endothelial cells, astrocytes, microglia, and neurons. There is increasing recognition of the importance of the TWEAK/Fn14 pathway in autoimmune neurologic conditions, including experimental autoimmune encephalomyelitis and neuropsychiatric lupus. Previously, we had found that Fn14 knockout lupus-prone MRL/lpr mice display significantly attenuated neuropsychiatric manifestations. To investigate whether this improvement in disease is secondary to inhibition of TWEAK/Fn14 signaling within the CNS or the periphery, and determine whether TWEAK-mediated neuropsychiatric effects are strain dependent, we performed intracerebroventricular (ICV) injection of Fc-TWEAK or an isotype matched control protein to C57Bl6/J non-autoimmune mice. We found that Fc-TWEAK injected C57Bl6/J mice developed significant depression-like behavior and cognitive dysfunction. Inflammatory mediators associated with lupus brain disease, including CCL2, C3, and iNOS, were significantly elevated in the brains of Fc-TWEAK treated mice. Furthermore, Fc-TWEAK directly increased blood brain barrier (BBB) permeability, as demonstrated by increased IgG deposition in the brain and reduced aquaporin-4 expression. Finally, Fc-TWEAK increased apoptotic cell death in the cortex and hippocampus. In conclusion, TWEAK can contribute to lupus-associated neurobehavioral deficits including depression and cognitive dysfunction by acting within the CNS to enhance production of inflammatory mediators, promote disruption of the BBB, and induce apoptosis in resident brain cells. Our study provides further support that the TWEAK/Fn14 signaling pathway may be a potential therapeutic target for inflammatory diseases involving the CNS.


Subject(s)
Cognitive Dysfunction/chemically induced , Depression/chemically induced , Tumor Necrosis Factors/administration & dosage , Animals , Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Cytokine TWEAK , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Injections, Intraventricular , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Pregnancy , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Tumor Necrosis Factors/metabolism
7.
Eur J Immunol ; 44(11): 3240-51, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25103845

ABSTRACT

Paraneoplastic neurologic diseases (PND) involving immune responses directed toward intracellular antigens are poorly understood. Here, we examine immunity to the PND antigen Nova2, which is expressed exclusively in central nervous system (CNS) neurons. We hypothesized that ectopic expression of neuronal antigen in the periphery could incite PND. In our C57BL/6 mouse model, CNS antigen expression limits antigen-specific CD4+ and CD8+ T-cell expansion. Chimera experiments demonstrate that this tolerance is mediated by antigen expression in nonhematopoietic cells. CNS antigen expression does not limit tumor rejection by adoptively transferred transgenic T cells but does limit the generation of a memory population that can be expanded upon secondary challenge in vivo. Despite mediating cancer rejection, adoptively transferred transgenic T cells do not lead to paraneoplastic neuronal targeting. Preliminary experiments suggest an additional requirement for humoral activation to induce CNS autoimmunity. This work provides evidence that the requirements for cancer immunity and neuronal autoimmunity are uncoupled. Since humoral immunity was not required for tumor rejection, B-cell targeting therapy, such as rituximab, may be a rational treatment option for PND that does not hamper tumor immunity.


Subject(s)
Autoimmunity , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Paraneoplastic Syndromes, Nervous System/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , B-Lymphocytes/immunology , Central Nervous System/cytology , Immune Tolerance , Immunization , Immunologic Factors/pharmacology , Immunologic Memory , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuro-Oncological Ventral Antigen , Neurons/immunology , RNA-Binding Proteins/immunology , Rituximab , beta-Galactosidase/genetics , beta-Galactosidase/immunology
8.
J Neuroinflammation ; 12: 205, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26546449

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease marked by both B and T cell hyperactivity which commonly affects the joints, skin, kidneys, and brain. Neuropsychiatric disease affects about 40 % of SLE patients, most frequently manifesting as depression, memory deficits, and general cognitive decline. One important and yet unresolved question is whether neuropsychiatric SLE (NPSLE) is a complication of systemic autoimmunity or whether it is primarily driven by brain-intrinsic factors. METHODS: To dissect the relative contributions of the central nervous system from those of the hematopoietic compartment, we generated bone marrow chimeras between healthy control (MRL/+) and lupus-prone MRL/Tnfrsf6 (lpr/lpr) mice (MRL/+ → MRL/lpr), as well as control chimeras. After bone marrow reconstitution, mice underwent extensive behavioral testing, analysis of brain tissue, and histological assessment. RESULTS: Despite transfer of healthy MRL/+ bone marrow and marked attenuation of systemic disease, we found that MRL/+ → MRL/lpr mice had a behavioral phenotype consisting of depressive-like behavior and visuospatial memory deficits, comparable to MRL/lpr → MRL/lpr control transplanted mice and the behavioral profile previously established in MRL/lpr mice. Moreover, MRL/+ → MRL/lpr chimeric mice displayed increased brain RANTES expression, neurodegeneration, and cellular infiltration in the choroid plexus, as well as blood brain barrier disruption, all in the absence of significant systemic autoimmunity. CONCLUSIONS: Chimeric MRL/+ → MRL/lpr mice displayed no attenuation of the behavioral phenotype found in MRL/lpr mice, despite normalized serum autoantibodies and conserved renal function. Therefore, neuropsychiatric disease in the MRL/lpr lupus-prone strain of mice can occur absent any major contributions from systemic autoimmunity.


Subject(s)
Lupus Erythematosus, Systemic/psychology , Mental Disorders/psychology , Animals , Behavior, Animal , Blood-Brain Barrier , Bone Marrow Transplantation , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chimera , Choroid Plexus/pathology , Depression/psychology , Female , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/therapy , Memory Disorders/psychology , Mental Disorders/drug therapy , Mental Disorders/etiology , Mice , Mice, Inbred MRL lpr , Motor Activity , Psychomotor Performance
9.
J Autoimmun ; 60: 40-50, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25911200

ABSTRACT

Neuropsychiatric disease is one of the most common manifestations of human systemic lupus erythematosus, but the mechanisms remain poorly understood. In human brain microvascular endothelial cells in vitro, TNF-like weak inducer of apoptosis (TWEAK) decreases tight junction ZO-1 expression and increases the permeability of monolayer cell cultures. Furthermore, knockout (KO) of the TWEAK receptor, Fn14, in the MRL/lpr lupus mouse strain markedly attenuates neuropsychiatric disease, as demonstrated by significant reductions in depressive-like behavior and improved cognitive function. The purpose of the present study was to determine the mechanisms by which TWEAK signaling is instrumental in the pathogenesis of neuropsychiatric lupus (NPSLE). Evaluating brain sections of MRL/lpr Fn14WT and Fn14KO mice, we found that Fn14KO mice displayed significantly decreased cellular infiltrates in the choroid plexus. To evaluate the integrity of the blood brain barrier (BBB) in MRL/lpr mice, Western blot for fibronectin, qPCR for iNOS, and immunohistochemical staining for VCAM-1/ICAM-1 were performed. We found preserved BBB permeability in MRL/lpr Fn14KO mice, attributable to reduced brain expression of VCAM-1/ICAM-1 and iNOS. Additionally, administration of Fc-TWEAK intravenously directly increased the leakage of a tracer (dextran-FITC) into brain tissue. Furthermore, MRL/lpr Fn14KO mice displayed reduced antibody (IgG) and complement (C3, C6, and C4a) deposition in the brain. Finally, we found that MRL/lpr Fn14KO mice manifested reduced neuron degeneration and hippocampal gliosis. Our studies indicate that TWEAK/Fn14 interactions play an important role in the pathogenesis of NPSLE by increasing the accumulation of inflammatory cells in the choroid plexus, disrupting BBB integrity, and increasing neuronal damage, suggesting a novel target for therapy in this disease.


Subject(s)
Apoptosis/genetics , Blood-Brain Barrier/physiopathology , Neurons/pathology , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factors/immunology , Animals , Apoptosis/immunology , Choroid Plexus/physiopathology , Cognition , Complement C3/immunology , Complement C4a/immunology , Complement C6/immunology , Cytokine TWEAK , Depression/genetics , Disease Models, Animal , Gliosis/genetics , Immunoglobulin G/immunology , Intercellular Adhesion Molecule-1/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Knockout , Nerve Degeneration/genetics , Permeability , Signal Transduction/genetics , Signal Transduction/immunology , TWEAK Receptor , Vascular Cell Adhesion Molecule-1/metabolism , Zonula Occludens-1 Protein/biosynthesis
10.
J Drugs Dermatol ; 14(5): 453-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25942662

ABSTRACT

Sanguinarine has a history of use in both folk medicine and early dermatology for the treatment of cutaneous neoplasms. Applied indiscriminately, bloodroot is an escharotic agent with potential to cause extensive tissue necrosis. However, when used in a controlled fashion, sanguinarine imparts selective cytotoxic/anti-proliferative activity through multiple mechanisms against human/ murine melanoma. To exploit sanguinarine's observed activity against melanoma, a targeted delivery system is required. We present a sol-gel based nanoparticulate platform for encapsulating sanguinarine chloride(sang-np)-a targeted therapeutic capable of steady, reliable delivery of predictable quantities of drug over a sustained time period with minimal undesirable effects. Size and release kinetics of sang-np were characterized using dynamic light scattering and ultraviolet-visible spectroscopy respectively. In vitro efficacy of sang-np was assessed. At both 2 and 24 hours, free sanguinarine killed > 90% of B16 melanoma cells, assessed via MTT assay. At 2 hours, sang-np killed a portion of melanoma cells, increasing to percentages comparable to free sanguinarine by 24 hours. Control(empty) nanoparticles exerted minimal toxicity to melanoma cells at both time points. TUNEL assay revealed that treatment with both sanguinarine and sang-np induces apoptosis in B16 melanoma cells, suggesting that both treatments act via the same mechanism of action. These data confirm controlled release of sanguinarine from sang-np, as well as comparable efficacy and mechanism of action to sanguinarine alone. This suggests that nanoparticle delivery of sanguinarine may be a unique approach to capitalize on this potent agent's inherent anti-tumor activity and overcome many of the limitations with its current formulation.


Subject(s)
Benzophenanthridines/administration & dosage , Drug Delivery Systems , Isoquinolines/administration & dosage , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzophenanthridines/pharmacology , Delayed-Action Preparations , Dynamic Light Scattering , In Situ Nick-End Labeling , Isoquinolines/pharmacology , Melanoma, Experimental/pathology , Mice , Nanocapsules , Particle Size , Skin Neoplasms/pathology , Time Factors
11.
Nanomedicine ; 11(1): 195-206, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25240595

ABSTRACT

Burn wounds are often complicated by bacterial infection, contributing to morbidity and mortality. Agents commonly used to treat burn wound infection are limited by toxicity, incomplete microbial coverage, inadequate penetration, and rising resistance. Curcumin is a naturally derived substance with innate antimicrobial and wound healing properties. Acting by multiple mechanisms, curcumin is less likely than current antibiotics to select for resistant bacteria. Curcumin's poor aqueous solubility and rapid degradation profile hinder usage; nanoparticle encapsulation overcomes this pitfall and enables extended topical delivery of curcumin. In this study, we synthesized and characterized curcumin nanoparticles (curc-np), which inhibited in vitro growth of methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa in dose-dependent fashion, and inhibited MRSA growth and enhanced wound healing in an in vivo murine wound model. Curc-np may represent a novel topical antimicrobial and wound healing adjuvant for infected burn wounds and other cutaneous injuries.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Bacterial Infections/drug therapy , Curcumin/chemistry , Nanoparticles/chemistry , Animals , Burns/therapy , Cell Movement , Dose-Response Relationship, Drug , Drug Delivery Systems , Keratinocytes/cytology , Light , Methicillin-Resistant Staphylococcus aureus , Mice , Mice, Inbred BALB C , Microscopy, Electron, Scanning , Nanomedicine/methods , Scattering, Radiation , Solubility , Stem Cells , Wound Healing , Zebrafish
12.
mBio ; 14(5): e0170723, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37737612

ABSTRACT

IMPORTANCE: Inflammasomes are essential for host defense against intracellular bacterial pathogens like Legionella, as they activate caspases, which promote cytokine release and cell death to control infection. In mice, interferon (IFN) signaling promotes inflammasome responses against bacteria by inducing a family of IFN-inducible GTPases known as guanylate-binding proteins (GBPs). Within murine macrophages, IFN promotes the rupture of the Legionella-containing vacuole (LCV), while GBPs are dispensable for this process. Instead, GBPs facilitate the lysis of cytosol-exposed Legionella. In contrast, the functions of IFN and GBPs in human inflammasome responses to Legionella are poorly understood. We show that IFN-γ enhances inflammasome responses to Legionella in human macrophages. Human GBP1 is required for these IFN-γ-driven inflammasome responses. Furthermore, GBP1 co-localizes with Legionella and/or LCVs in a type IV secretion system (T4SS)-dependent manner and promotes damage to the LCV, which leads to increased exposure of the bacteria to the host cell cytosol. Thus, our findings reveal species- and pathogen-specific differences in how GBPs function to promote inflammasome responses.


Subject(s)
Legionella pneumophila , Legionella , Humans , Animals , Mice , Inflammasomes/metabolism , Legionella/metabolism , Vacuoles/metabolism , Carrier Proteins/metabolism , Signal Transduction , Legionella pneumophila/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism
13.
Arthritis Res Ther ; 20(1): 10, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29370834

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that affects different end organs, including skin and brain. We and others have previously shown the importance of macrophages in the pathogenesis of cutaneous and neuropsychiatric lupus. Additionally, autoantibodies produced by autoreactive B cells are thought to play a role in both the skin and central nervous system pathologies associated with SLE. METHODS: We used a novel inhibitor of Bruton's tyrosine kinase (BTK), BI-BTK-1, to target both macrophage and B cell function in the MRL-lpr/lpr murine model of SLE, and examined the effect of treatment on skin and brain disease. RESULTS: We found that treatment with BI-BTK-1 significantly attenuated the lupus associated cutaneous and neuropsychiatric disease phenotypes in MRL/lpr mice. Specifically, BI-BTK-1 treated mice had fewer macroscopic and microscopic skin lesions, reduced cutaneous cellular infiltration, and diminished inflammatory cytokine expression compared to control mice. BTK inhibition also significantly improved cognitive function, and decreased accumulation of T cells, B cells, and macrophages within the central nervous system, specifically the choroid plexus. CONCLUSIONS: Directed therapies may improve the response rate in lupus-driven target organ involvement, and decrease the dangerous side effects associated with global immunosuppression. Overall, our results suggest that inhibition of BTK may be a promising therapeutic option for cutaneous and neuropsychiatric disease associated with SLE.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Brain Diseases/prevention & control , Enzyme Inhibitors/pharmacology , Lupus Erythematosus, Systemic/complications , Skin Diseases/prevention & control , Agammaglobulinaemia Tyrosine Kinase/immunology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Brain Diseases/etiology , Brain Diseases/immunology , Cognition/drug effects , Cognition/physiology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression/drug effects , Humans , Lupus Erythematosus, Systemic/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred MRL lpr , Skin Diseases/etiology , Skin Diseases/immunology
14.
Sci Rep ; 6: 26164, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27192942

ABSTRACT

Lupus nephritis (LN) is a potentially dangerous end organ pathology that affects upwards of 60% of lupus patients. Bruton's tyrosine kinase (BTK) is important for B cell development, Fc receptor signaling, and macrophage polarization. In this study, we investigated the effects of a novel, highly selective and potent BTK inhibitor, BI-BTK-1, in an inducible model of LN in which mice receive nephrotoxic serum (NTS) containing anti-glomerular antibodies. Mice were treated once daily with vehicle alone or BI-BTK-1, either prophylactically or therapeutically. When compared with control treated mice, NTS-challenged mice treated prophylactically with BI-BTK-1 exhibited significantly attenuated kidney disease, which was dose dependent. BI-BTK-1 treatment resulted in decreased infiltrating IBA-1+ cells, as well as C3 deposition within the kidney. RT-PCR on whole kidney RNA and serum profiling indicated that BTK inhibition significantly decreased levels of LN-relevant inflammatory cytokines and chemokines. Renal RNA expression profiling by RNA-seq revealed that BI-BTK-1 dramatically modulated pathways related to inflammation and glomerular injury. Importantly, when administered therapeutically, BI-BTK-1 reversed established proteinuria and improved renal histopathology. Our results highlight the important role for BTK in the pathogenesis of immune complex-mediated nephritis, and BTK inhibition as a promising therapeutic target for LN.


Subject(s)
Antigen-Antibody Complex/metabolism , Enzyme Inhibitors/administration & dosage , Lupus Nephritis/drug therapy , Lupus Nephritis/prevention & control , Protein-Tyrosine Kinases/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase , Animals , Blood Chemical Analysis , Complement C3/analysis , Cytokines/analysis , Disease Models, Animal , Gene Expression Profiling , Kidney/pathology , Lupus Nephritis/chemically induced , Lupus Nephritis/pathology , Mice , Sequence Analysis, RNA , Treatment Outcome
15.
J Invest Dermatol ; 135(8): 1986-1995, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25826425

ABSTRACT

Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK, TNFSF12) and its sole receptor Fn14, belonging to the TNF ligand and receptor superfamilies respectively, are involved in cell survival and cytokine production. The role of TWEAK/Fn14 interactions in the pathogenesis of cutaneous lupus has not been explored. TWEAK treatment of murine PAM212 keratinocytes stimulated the secretion of RANTES via Fn14 and promoted apoptosis. Parthenolide, but not wortmanin or the MAPK inhibitor PD98059, significantly decreased production of RANTES, indicating that this effect of TWEAK is mediated via NF-κB signaling. UVB irradiation significantly upregulated the expression of Fn14 on keratinocytes in vitro and in vivo and increased RANTES production. MRL/lpr Fn14 knockout (KO) lupus mice were compared with MRL/lpr Fn14 wild-type (WT) mice to evaluate for any possible differences in the severity of cutaneous lesions and the presence of infiltrating immune cells. MRL/lpr Fn14 KO mice had markedly attenuated cutaneous disease as compared with their Fn14 WT littermates, as evidenced by the well-maintained architecture of the skin and significantly decreased skin infiltration of T cells and macrophages. Our data strongly implicate TWEAK/Fn14 signaling in the pathogenesis of the cutaneous manifestations in the MRL/lpr model of spontaneous lupus and suggest a possible target for therapeutic intervention.


Subject(s)
Lupus Erythematosus, Cutaneous/etiology , Lupus Erythematosus, Cutaneous/physiopathology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Tumor Necrosis Factors/physiology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line , Chemokine CCL5/metabolism , Cytokine TWEAK , Disease Models, Animal , Fibroblasts/metabolism , In Vitro Techniques , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , Lupus Erythematosus, Cutaneous/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Mice, Knockout , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/radiation effects , TWEAK Receptor , Tumor Necrosis Factors/pharmacology , Ultraviolet Rays , Up-Regulation/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL