Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
PLoS Biol ; 21(4): e3002084, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37098006

RESUMEN

Systemic autoimmune diseases are characteristically associated with aberrant autoreactive innate and adaptive immune responses that lead to tissue damage and increased morbidity and mortality. Autoimmunity has been linked to alterations in the metabolic functions of immune cells (immunometabolism) and, more specifically, to mitochondrial dysfunction. Much has been written about immunometabolism in autoimmunity in general, so this Essay focuses on recent research into the role of mitochondrial dysfunction in the dysregulation of innate and adaptive immunity that is characteristic of systemic autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Enhancing the understanding of mitochondrial dysregulation in autoimmunity will hopefully contribute to accelerating the development of immunomodulatory treatments for these challenging diseases.


Asunto(s)
Artritis Reumatoide , Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Humanos , Autoinmunidad , Sistema Inmunológico
2.
J Immunol ; 213(4): 419-434, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38949522

RESUMEN

The Krebs cycle enzyme aconitate decarboxylase 1 (ACOD1) mediates itaconate synthesis in monocytes and macrophages. Previously, we reported that administration of 4-octyl itaconate to lupus-prone mice abrogated immune dysregulation and clinical features. In this study, we explore the role of the endogenous ACOD1/itaconate pathway in the development of TLR7-induced lupus (imiquimod [IMQ] model). We found that, in vitro, ACOD1 was induced in mouse bone marrow-derived macrophages and human monocyte-derived macrophages following TLR7 stimulation. This induction was partially dependent on type I IFN receptor signaling and on specific intracellular pathways. In the IMQ-induced mouse model of lupus, ACOD1 knockout (Acod1-/-) displayed disruptions of the splenic architecture, increased serum levels of anti-dsDNA and proinflammatory cytokines, and enhanced kidney immune complex deposition and proteinuria, when compared with the IMQ-treated wild-type mice. Consistent with these results, Acod1-/- bone marrow-derived macrophages treated in vitro with IMQ showed higher proinflammatory features. Furthermore, itaconate serum levels in systemic lupus erythematosus patients were decreased compared with healthy individuals, in association with disease activity and specific perturbed cardiometabolic parameters. These findings suggest that the ACOD1/itaconate pathway plays important immunomodulatory and vasculoprotective roles in systemic lupus erythematosus, supporting the potential therapeutic role of itaconate analogs in autoimmune diseases.


Asunto(s)
Carboxiliasas , Lupus Eritematoso Sistémico , Macrófagos , Ratones Noqueados , Succinatos , Animales , Lupus Eritematoso Sistémico/inmunología , Ratones , Humanos , Femenino , Macrófagos/inmunología , Succinatos/farmacología , Enfermedades Cardiovasculares/inmunología , Biomarcadores , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Adulto , Masculino , Modelos Animales de Enfermedad , Persona de Mediana Edad , Citocinas/metabolismo , Receptor Toll-Like 7/metabolismo , Hidroliasas
3.
Proc Natl Acad Sci U S A ; 117(10): 5409-5419, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32094169

RESUMEN

Type III IFN lambdas (IFN-λ) have recently been described as important mediators of immune responses at barrier surfaces. However, their role in autoimmune diseases such as systemic lupus erythematosus (SLE), a condition characterized by aberrant type I IFN signaling, has not been determined. Here, we identify a nonredundant role for IFN-λ in immune dysregulation and tissue inflammation in a model of TLR7-induced lupus. IFN-λ protein is increased in murine lupus and IFN-λ receptor (Ifnlr1) deficiency significantly reduces immune cell activation and associated organ damage in the skin and kidneys without effects on autoantibody production. Single-cell RNA sequencing in mouse spleen and human peripheral blood revealed that only mouse neutrophils and human B cells are directly responsive to this cytokine. Rather, IFN-λ activates keratinocytes and mesangial cells to produce chemokines that induce immune cell recruitment and promote tissue inflammation. These data provide insights into the immunobiology of SLE and identify type III IFNs as important factors for tissue-specific pathology in this disease.


Asunto(s)
Interferones/fisiología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Animales , Linfocitos B/inmunología , Línea Celular , Eliminación de Gen , Humanos , Imiquimod/farmacología , Inflamación/inmunología , Inflamación/patología , Inductores de Interferón/farmacología , Interferón Tipo I/fisiología , Interferones/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/patología , Células Mesangiales/efectos de los fármacos , Células Mesangiales/inmunología , Células Mesangiales/patología , Ratones Endogámicos C57BL , Ratones Mutantes , Receptores de Interferón/genética , Transducción de Señal , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/fisiología , Interferón lambda
4.
Proc Natl Acad Sci U S A ; 117(28): 16481-16491, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32601182

RESUMEN

Differences between female and male immunity may contribute to variations in response to infections and predisposition to autoimmunity. We previously reported that neutrophils from reproductive-age males are more immature and less activated than their female counterparts. To further characterize the mechanisms that drive differential neutrophil phenotypes, we performed RNA sequencing on circulating neutrophils from healthy adult females and males. Female neutrophils displayed significant up-regulation of type I IFN (IFN)-stimulated genes (ISGs). Single-cell RNA-sequencing analysis indicated that these differences are neutrophil specific, driven by a distinct neutrophil subset and related to maturation status. Neutrophil hyperresponsiveness to type I IFNs promoted enhanced responses to Toll-like receptor agonists. Neutrophils from young adult males had significantly increased mitochondrial metabolism compared to those from females and this was modulated by estradiol. Assessment of ISGs and neutrophil maturation genes in Klinefelter syndrome (47, XXY) males and in prepubescent children supported that differences in neutrophil phenotype between adult male and female neutrophils are hormonally driven and not explained by X chromosome gene dosage. Our results indicate that there are distinct sex differences in neutrophil biology related to responses to type I IFNs, immunometabolism, and maturation status that may have prominent functional and pathogenic implications.


Asunto(s)
Interferón Tipo I/inmunología , Neutrófilos/inmunología , Adulto , Femenino , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Síndrome de Klinefelter/genética , Síndrome de Klinefelter/inmunología , Síndrome de Klinefelter/metabolismo , Masculino , Factores Sexuales , Adulto Joven
5.
Curr Opin Rheumatol ; 29(5): 442-449, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28639951

RESUMEN

PURPOSE OF REVIEW: Upon antigen exposure, immune cells rely on cell-specific metabolic pathways to mount an efficient immune response. In autoimmunity, failure in critical metabolic checkpoints may lead to immune cell hyperactivation and tissue damage. Oxidative stress in autoimmune patients can also contribute to immune dysregulation and injury to the host. Recent insights into the immune cell metabolism signatures, specifically associated with systemic lupus erythematosus (SLE) and the consequences of heightened oxidative stress in patients, are discussed herein. RECENT FINDINGS: Glucose metabolism inhibitors, mechanistic target of rapamycin pathway modulators, and peroxisome proliferator-activated receptor gamma-activating compounds demonstrate therapeutic benefit in experimental models of lupus. Mitochondrial-derived reactive oxygen species (ROS) and molecular modifications induced by oxidative stress appear to be detrimental in lupus. Effective therapies tailored toward the reconfiguration of metabolic imbalances in lupus immune cells and the reduction of mitochondrial ROS production/availability are currently being tested. SUMMARY: A paucity of knowledge exists regarding the metabolic needs of a number of immune cells involved in the pathogenesis of SLE, including myeloid cells and B cells. Nonetheless, SLE-specific metabolic signatures have been identified and their specific targeting, along with mitochondrial ROS inhibitors/scavengers, could show therapeutic advantage in lupus patients.


Asunto(s)
Autoinmunidad , Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Humanos , Neutrófilos/patología
6.
J Immunol ; 192(6): 2722-33, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24532579

RESUMEN

Nasal administration of an oil-in-water nanoemulsion (NE) adjuvant W805EC produces potent systemic and mucosal, Th-1- and Th-17-balanced cellular responses. However, its molecular mechanism of action has not been fully characterized and is of particular interest because NE does not contain specific ligands for innate immune receptors. In these studies, we demonstrate that W805EC NE adjuvant activates innate immunity, induces specific gene transcription, and modulates NF-κB activity via TLR2 and TLR4 by a mechanism that appears to be distinct from typical TLR agonists. Nasal immunization with NE-based vaccine showed that the TLR2, TLR4, and MyD88 pathways and IL-12 and IL-12Rß1 expression are not required for an Ab response, but they are essential for the induction of balanced Th-1 polarization and Th-17 cellular immunity. NE adjuvant induces MHC class II, CD80, and CD86 costimulatory molecule expression and dendritic cell maturation. Further, upon immunization with NE, adjuvant mice deficient in the CD86 receptor had normal Ab responses but significantly reduced Th-1 cellular responses, whereas animals deficient in both CD80 and CD86 or lacking CD40 failed to produce either humoral or cellular immunity. Overall, our data show that intranasal administration of Ag with NE induces TLR2 and TLR4 activation along with a MyD88-independent Ab response and a MyD88-dependent Th-1 and Th-17 cell-mediated immune response. These findings suggest that the unique properties of NE adjuvant may offer novel opportunities for understanding previously unrecognized mechanisms of immune activation important for generating effective mucosal and systemic immune responses.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Emulsiones/farmacología , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Línea Celular , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Emulsiones/administración & dosificación , Femenino , Células HEK293 , Humanos , Inmunidad Celular/genética , Inmunidad Celular/inmunología , Inmunidad Humoral/genética , Inmunidad Humoral/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-12/metabolismo , Sudunidad beta 1 del Receptor de Interleucina-12/genética , Sudunidad beta 1 del Receptor de Interleucina-12/inmunología , Sudunidad beta 1 del Receptor de Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Transcriptoma/inmunología
7.
Proc Natl Acad Sci U S A ; 110(7): 2629-34, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23359678

RESUMEN

Bacterial biofilm formation is a complex developmental process involving cellular differentiation and the formation of intricate 3D structures. Here we demonstrate that exposure to ferric chloride triggers rugose biofilm formation by the uropathogenic Escherichia coli strain UTI89 and by enteric bacteria Citrobacter koseri and Salmonella enterica serovar typhimurium. Two unique and separable cellular populations emerge in iron-triggered, rugose biofilms. Bacteria at the air-biofilm interface express high levels of the biofilm regulator csgD, the cellulose activator adrA, and the curli subunit operon csgBAC. Bacteria in the interior of rugose biofilms express low levels of csgD and undetectable levels of matrix components curli and cellulose. Iron activation of rugose biofilms is linked to oxidative stress. Superoxide generation, either through addition of phenazine methosulfate or by deletion of sodA and sodB, stimulates rugose biofilm formation in the absence of high iron. Additionally, overexpression of Mn-superoxide dismutase, which can mitigate iron-derived reactive oxygen stress, decreases biofilm formation in a WT strain upon iron exposure. Not only does reactive oxygen stress promote rugose biofilm formation, but bacteria in the rugose biofilms display increased resistance to H(2)O(2) toxicity. Altogether, we demonstrate that iron and superoxide stress trigger rugose biofilm formation in UTI89. Rugose biofilm development involves the elaboration of two distinct bacterial populations and increased resistance to oxidative stress.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Cloruros/farmacología , Citrobacter koseri/crecimiento & desarrollo , Escherichia coli Enteropatógena/crecimiento & desarrollo , Compuestos Férricos/farmacología , Salmonella typhimurium/crecimiento & desarrollo , Biopelículas/efectos de los fármacos , Western Blotting , Citrobacter koseri/efectos de los fármacos , Escherichia coli Enteropatógena/efectos de los fármacos , Proteínas de Escherichia coli/metabolismo , Peróxido de Hidrógeno/metabolismo , Microscopía Confocal , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Salmonella typhimurium/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Transactivadores/metabolismo , beta-Galactosidasa/metabolismo
8.
J Virol ; 87(23): 12685-93, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24049163

RESUMEN

Noroviruses (NoVs) are the causative agent of the vast majority of nonbacterial gastroenteritis worldwide. Due to the inability to culture human NoVs and the inability to orally infect a small animal model, little is known about the initial steps of viral entry. One particular step that is not understood is how NoVs breach the intestinal epithelial barrier. Murine NoV (MNV) is the only NoV that can be propagated in vitro by infecting murine macrophages and dendritic cells, making this virus an attractive model for studies of different aspects of NoV biology. Polarized murine intestinal epithelial mICcl2 cells were used to investigate how MNV interacts with and crosses the intestinal epithelium. In this in vitro model of the follicle-associated epithelium (FAE), MNV is transported across the polarized cell monolayer in the absence of viral replication or disruption of tight junctions by a distinct epithelial cell with microfold (M) cell properties. In addition to transporting MNV, these M-like cells also transcytose microbeads and express an IgA receptor. Interestingly, B myeloma cells cultured in the basolateral compartment underlying the epithelial monolayer did not alter the number of M-like cells but increased their transcytotic activity. Our data demonstrate that MNV can cross an intact intestinal epithelial monolayer in vitro by hijacking the M-like cells' intrinsic transcytotic pathway and suggest a potential mechanism for MNV entry into the host.


Asunto(s)
Infecciones por Caliciviridae/virología , Células Epiteliales/virología , Intestinos/citología , Norovirus/fisiología , Transcitosis , Animales , Línea Celular , Polaridad Celular , Células Epiteliales/citología , Humanos , Intestinos/virología , Ratones , Internalización del Virus
9.
medRxiv ; 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38605883

RESUMEN

Objective: The Krebs cycle enzyme Aconitate Decarboxylase 1 (ACOD1) mediates itaconate synthesis in myeloid cells.. Previously, we reported that administration of 4-octyl itaconate abrogated lupus phenotype in mice. Here, we explore the role of the endogenous ACOD1/itaconate pathway in the development of murine lupus as well as their relevance in premature cardiovascular damage in SLE. Methods: We characterized Acod1 protein expression in bone marrow-derived macrophages and human monocyte-derived macrophages, following a TLR7 agonist (imiquimod, IMQ). Wild type and Acod1-/- mice were exposed to topical IMQ for 5 weeks to induce an SLE phenotype and immune dysregulation was quantified. Itaconate serum levels were quantified in SLE patients and associated to cardiometabolic parameters and disease activity. Results: ACOD1 was induced in mouse bone marrow-derived macrophages (BMDM) and human monocyte-derived macrophages following in vitro TLR7 stimulation. This induction was partially dependent on type I Interferon receptor signaling and specific intracellular pathways. In the IMQ-induced mouse model of lupus, ACOD1 knockout (Acod1-/-) displayed disruptions of the splenic architecture, increased serum anti-dsDNA and proinflammatory cytokine levels, enhanced kidney immune complex deposition and proteinuria, when compared to the IMQ-treated WT mice. Consistent with these results, Acod1-/- BMDM exposed to IMQ showed higher proinflammatory features in vitro. Itaconate levels were decreased in SLE serum compared to healthy control sera, in association with specific perturbed cardiometabolic parameters and subclinical vascular disease. Conclusion: These findings suggest that the ACOD1/itaconate pathway plays important immunomodulatory and vasculoprotective roles in SLE, supporting the potential therapeutic role of itaconate analogs in autoimmune diseases.

10.
Eur J Immunol ; 42(8): 2073-86, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22653620

RESUMEN

While the nasal mucosa is a potentially useful site for human immunization, toxin-based nasal adjuvants are generally unsafe and less effective in humans. Safe mucosal adjuvants that activate protective immunity via mucosal administration are highly dependent on barrier antigen sampling by epithelial and DCs. Here, we demonstrate that protein antigens formulated in unique oil-in-water nanoemulsions (NEs) result in distinctive transcellular antigen uptake in ciliated nasal epithelial cells, leading to delivery into nasal associated lymphoid tissue. NE formulation also enhances MHC class II expression in epithelial cells and DC activation/trafficking to regional lymphoid tissues in mice. These materials appear to induce local epithelial cell apoptosis and heterogeneous cytokine production by mucosal epithelial cells and mixed nasal tissues, including G-CSF, GM-CSF, IL-1a, IL-1b, IL-5, IL-6, IL-12, IP-10, KC, MIP-1a, TGF-ß, and TSLP. This is the first observation of a nasal adjuvant that activates calreticulin-associated apoptosis of ciliated nasal epithelial cells to generate broad cytokine/chemokine responses in mucosal tissue.


Asunto(s)
Adyuvantes Inmunológicos , Apoptosis , Citocinas/biosíntesis , Células Dendríticas/inmunología , Mucosa Nasal/inmunología , Animales , Transporte Biológico/inmunología , Calreticulina , Movimiento Celular , Células Cultivadas , Células Dendríticas/metabolismo , Emulsiones , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Genes MHC Clase II , Interleucina-6/inmunología , Interleucina-6/metabolismo , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Mucosa Nasal/metabolismo
11.
Arthritis Rheumatol ; 74(12): 1971-1983, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35791960

RESUMEN

OBJECTIVE: Itaconic acid, a Krebs cycle-derived immunometabolite, is synthesized by myeloid cells in response to danger signals to control inflammasome activation, type I interferon (IFN) responses, and oxidative stress. As these pathways are dysregulated in systemic lupus erythematosus (SLE), we investigated the role of an itaconic acid derivative in the treatment of established murine lupus. METHODS: Female (NZW × NZB)F1 lupus-prone mice were administered 4-octyl itaconate (4-OI) or vehicle starting after clinical onset of disease (30 weeks of age) for 4 weeks (n = 10 mice /group). At 34 weeks of age (peak disease activity), animals were euthanized, organs and serum were collected, and clinical, metabolic, and immunologic parameters were evaluated. RESULTS: Proteinuria, kidney immune complex deposition, renal scores of severity and inflammation, and anti-RNP autoantibodies were significantly reduced in the 4-OI treatment group compared to the vehicle group. Splenomegaly decreased in the 4-OI group compared to vehicle, with decreases in activation markers in innate and adaptive immune cells, increases in CD8+ T cell numbers, and inhibition of JAK1 activation. Gene expression analysis in splenocytes showed significant decreases in type I IFN and proinflammatory cytokine genes and increased Treg cell-associated markers in the 4-OI group compared to the vehicle group. In human control and lupus myeloid cells, 4-OI in vitro treatment decreased proinflammatory responses and B cell responses. CONCLUSIONS: These results support targeting immunometabolism as a potentially viable approach in autoimmune disease treatment, with 4-OI displaying beneficial roles attenuating immune dysregulation and organ damage in lupus.


Asunto(s)
Lupus Eritematoso Sistémico , Ratones , Femenino , Humanos , Animales , Recién Nacido , Ratones Endogámicos NZB , Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/tratamiento farmacológico , Anticuerpos Antinucleares
12.
J Clin Invest ; 132(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35025762

RESUMEN

BACKGROUNDFasting and NAD+-boosting compounds, including NAD+ precursor nicotinamide riboside (NR), confer antiinflammatory effects. However, the underlying mechanisms and therapeutic potential are incompletely defined.METHODSWe explored the underlying biology in myeloid cells from healthy volunteers following in vivo placebo or NR administration and subsequently tested the findings in vitro in monocytes extracted from patients with systemic lupus erythematosus (SLE).RESULTSRNA-Seq of unstimulated and LPS-activated monocytes implicated NR in the regulation of autophagy and type I IFN signaling. In primary monocytes, NR blunted LPS-induced IFN-ß production, and genetic or pharmacological disruption of autophagy phenocopied this effect. Given that NAD+ is a coenzyme in oxidoreductive reactions, metabolomics was performed and identified that NR increased the inosine level. Inosine supplementation similarly blunted autophagy and IFN-ß release. Finally, because SLE exhibits type I IFN dysregulation, we assessed the NR effect on monocytes from patients with SLE and found that NR reduced autophagy and IFN-ß release.CONCLUSIONWe conclude that NR, in an NAD+-dependent manner and in part via inosine signaling, mediated suppression of autophagy and attenuated type I IFN in myeloid cells, and we identified NR as a potential adjunct for SLE management.TRIAL REGISTRATIONClinicalTrials.gov registration numbers NCT02812238, NCT00001846, and NCT00001372.FUNDINGThis work was supported by the NHLBI and NIAMS Intramural Research divisions.


Asunto(s)
Lupus Eritematoso Sistémico , NAD , Estudios Clínicos como Asunto , Humanos , Inosina , Interferón beta , Lipopolisacáridos , Monocitos , Niacinamida , Receptor Toll-Like 4
13.
Crit Rev Immunol ; 30(2): 189-99, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20370629

RESUMEN

T17 (T-helper-17) cytokine responses have been recently recognized as an important component for the protective immunity produced by vaccination. However, the mechanism by which immune adjuvants induce T17 immunity has not been defined. We have developed a novel mucosal nanoemulsion (NE) adjuvant that produces a robust humoral and T1 cellular immunity. Herein, we demonstrate that immunization with NE adjuvant induces a T17 response to diverse antigens in both outbred and inbred mice. CD86 deficiency had a limited effect on the induction of IL-17, however, double CD80/CD86, CD40, and IL-6 (interleukin 6) mutant mice failed to produce T17 immunity in response to NE adjuvant. Mice deficient in TLR2 and TLR4 (Toll-like receptors 2 and 4) had a diminished IL-17 response. Our data indicate that nasal mucosal immunization with NE adjuvant produces T1 and T17 immunity; that this process requires IL-6, CD40, and at least one of the CD80/CD86 molecules; and that the induction of TH17 is enhanced by the presence of TLR2 and TLR4 receptors. This unique approach to vaccination may have a significant role in protection against mucosal and intracellular pathogens.


Asunto(s)
Adyuvantes Inmunológicos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Interleucina-17/inmunología , Nanoestructuras , Animales , Emulsiones , Vacunación
14.
Arthritis Rheumatol ; 73(12): 2282-2292, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33983685

RESUMEN

OBJECTIVE: Neutrophil extracellular traps (NETs) are extracellular lattices composed of nucleic material bound to neutrophil granule proteins. NETs may play pathogenic roles in the development and severity of autoimmune diseases such as systemic lupus erythematosus (SLE), at least in part, through induction of type I interferon (IFN) responses via externalization of oxidized immunostimulatory DNA. A distinct subset of SLE proinflammatory neutrophils (low-density granulocytes [LDGs]) displays enhanced ability to form proinflammatory NETs that damage the vasculature. We undertook this study to assess whether NET-bound RNA can contribute to inflammatory responses in endothelial cells (ECs) and the pathways that mediate this effect. METHODS: Expression of newly synthesized and total RNA was quantified in NETs from healthy controls and lupus patients. The ability of ECs to take up NET-bound RNA and downstream induction of type I IFN responses were quantified. RNAs present in NETs were sequenced and specific small RNAs were tested for induction of endothelial type I IFN pathways. RESULTS: NETs extruded RNA that was internalized by ECs, and this was enhanced when NET-bound nucleic acids were oxidized, particularly in lupus LDG-derived NETs. Internalization of NET-bound RNA by ECs was dependent on endosomal Toll-like receptors (TLRs) and the actin cytoskeleton and induced type I IFN-stimulated genes (ISGs). This ISG induction was dependent on NET-associated microRNA let-7b, a small RNA expressed at higher levels in LDG-derived NETs, which acted as a TLR-7 agonist. CONCLUSION: These findings highlight underappreciated roles for small RNAs externalized in NETs in the induction of proinflammatory responses in vascular cells, with implications for lupus vasculopathy.


Asunto(s)
Células Endoteliales/metabolismo , Inflamación/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Aorta/metabolismo , Línea Celular , Trampas Extracelulares , Humanos , MicroARNs/metabolismo , Neutrófilos/metabolismo
15.
Arthritis Rheumatol ; 72(12): 2118-2129, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32692482

RESUMEN

OBJECTIVE: Gasdermin D (GSDMD) is the key executioner of an inflammatory cell death mechanism known as pyroptosis. Recent reports have also implicated GSDMD in other mechanisms of cell death, including apoptosis, necroptosis, and NETosis. Given the role of dysregulated cell death in autoimmune syndromes such as systemic lupus erythematosus (SLE), this study was undertaken in a murine lupus model to investigate whether GSDMD plays a pathogenic role in systemic autoimmunity by promoting inflammatory cell death, leading to increased generation of nuclear autoantigens and autoantibodies. METHODS: An imiquimod-induced model of SLE was tested in GSDMD-/- mice (n = 30), with wild-type (WT) mice as controls (n = 34), on a C57BL/6 background. At the time of euthanasia, the mice were examined for serum autoantibodies, immune complex deposition, organ inflammation, immune dysregulation, and type I interferon responses. A model of pristane-induced lung injury in GSDMD-/- mice (n = 7), with WT mice as controls (n = 10), was used to confirm the pulmonary phenotype. Regulation of various mechanisms of cell death by GSDMD was investigated in the mice. RESULTS: Unexpectedly, GSDMD-/- mice developed enhanced mortality, more severe renal and pulmonary inflammation, and exacerbated autoantibody production in response to imiquimod. Pulmonary involvement was also more severe in the absence of GSDMD in mice with pristane-induced lung injury. Compared to WT mice, lack of GSDMD was associated with increased levels of circulating nuclear autoantigens (P < 0.01), anti-double-stranded DNA autoantibodies (P < 0.01), tissue immune complex deposition (P < 0.05), expansion of myeloid cell subsets (P < 0.05), and enhanced B cell activation and plasma cell differentiation (P = 0.001). Moreover, in the absence of GSDMD, enhanced autoantigen generation was associated with increased local induction of cell death in vivo. CONCLUSION: GSDMD negatively regulates autoantigen generation and immune dysregulation in response to tissue injury and may play previously unappreciated protective roles in systemic autoimmunity.


Asunto(s)
Muerte Celular/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Animales , Autoanticuerpos/sangre , Autoinmunidad , Diferenciación Celular/fisiología , ADN/inmunología , Modelos Animales de Enfermedad , Imiquimod , Inflamación/genética , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/genética , Ratones , Ratones Noqueados , Proteínas de Unión a Fosfato/genética
16.
Arthritis Rheumatol ; 72(3): 454-464, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31566908

RESUMEN

OBJECTIVE: A role for mitochondrial dysfunction has been proposed in the immune dysregulation and organ damage characteristic of systemic lupus erythematosus (SLE). Idebenone is a coenzyme Q10 synthetic quinone analog and an antioxidant that has been used in humans to treat diverse diseases in which mitochondrial function is impaired. This study was undertaken to assess whether idebenone ameliorates lupus in murine models. METHODS: Idebenone was administered orally to MRL/lpr mice at 2 different doses (1 gm/kg or 1.5 gm/kg idebenone-containing diet) for 8 weeks. At peak disease activity, clinical, immunologic, and metabolic parameters were analyzed and compared to those in untreated mice (n = 10 per treatment group). Results were confirmed in the lupus-prone NZM2328 mouse model. RESULTS: In MRL/lpr mice, idebenone-treated mice showed a significant reduction in mortality incidence (P < 0.01 versus untreated mice), and the treatment attenuated several disease features, including glomerular inflammation and fibrosis (each P < 0.05 versus untreated mice), and improved renal function in association with decreased renal expression of interleukin-17A (IL-17A) and mature IL-18. Levels of splenic proinflammatory cytokines and inflammasome-related genes were significantly decreased (at least P < 0.05 and some with higher significance) in mice treated with idebenone, while no obvious drug toxicity was observed. Idebenone inhibited neutrophil extracellular trap formation in neutrophils from lupus-prone mice (P < 0.05) and human patients with SLE. Idebenone also improved mitochondrial metabolism (30% increase in basal respiration and ATP production), reduced the extent of heart lipid peroxidation (by one-half that of untreated mice), and significantly improved endothelium-dependent vasorelaxation (P < 0.001). NZM2328 mice exposed to idebenone also displayed improvements in renal and systemic inflammation, reducing the kidney pathology score (P < 0.05), IgG/C3 deposition (P < 0.05), and the gene expression of interferon, proinflammatory, and inflammasome-related genes (at least P < 0.05 and some with higher significance). CONCLUSION: Idebenone ameliorates murine lupus disease activity and the severity of organ damage, supporting the hypothesis that agents that modulate mitochondrial biologic processes may have a therapeutic role in human SLE.


Asunto(s)
Antioxidantes/administración & dosificación , Lupus Eritematoso Sistémico/terapia , Ubiquinona/análogos & derivados , Animales , Modelos Animales de Enfermedad , Trampas Extracelulares/efectos de los fármacos , Inflamación , Interleucina-17/metabolismo , Interleucina-18/metabolismo , Riñón/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Mitocondrias/metabolismo , Ubiquinona/administración & dosificación
17.
Lupus Sci Med ; 7(1)2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32343673

RESUMEN

OBJECTIVES: Recent investigations in humans and mouse models with lupus have revealed evidence of mitochondrial dysfunction and production of mitochondrial reactive oxygen species (mROS) in T cells and neutrophils. This can provoke numerous cellular changes including oxidation of nucleic acids, proteins, lipids and even induction of cell death. We have previously observed that in T cells from patients with lupus, the increased mROS is capable of provoking oligomerisation of mitochondrial antiviral stimulator (MAVS) and production of type I interferon (IFN-I). mROS in SLE neutrophils also promotes the formation of neutrophil extracellular traps (NETs), which are increased in lupus and implicated in renal damage. As a result, in addition to traditional immunosuppression, more comprehensive treatments for lupus may also include non-immune therapy, such as antioxidants. METHODS: Lupus-prone MRL-lpr mice were treated from weaning for 11 weeks with the mitochondria-targeted antioxidant, MitoQ (200 µM) in drinking water. Mice were then assessed for ROS production in neutrophils, NET formation, MAVS oligomerisation, serum IFN-I, autoantibody production and renal function. RESULTS: MitoQ-treated mice manifested reduced neutrophil ROS and NET formation, decreased MAVS oligomerisation and serum IFN-I, and reduced immune complex formation in kidneys, despite no change in serum autoantibody . CONCLUSIONS: These findings reveal the potential utility of targeting mROS in addition to traditional immunosuppressive therapy for lupus.


Asunto(s)
Trampas Extracelulares/inmunología , Enfermedades Renales/metabolismo , Lupus Eritematoso Sistémico/inmunología , Mitocondrias/metabolismo , Compuestos Organofosforados/farmacología , Ubiquinona/análogos & derivados , Animales , Autoanticuerpos/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Interferón Tipo I/inmunología , Riñón/metabolismo , Riñón/fisiopatología , Enfermedades Renales/fisiopatología , Lupus Eritematoso Sistémico/fisiopatología , Masculino , Ratones , Ratones Endogámicos MRL lpr , Neutrófilos/inmunología , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/inmunología , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/inmunología , Ubiquinona/farmacología
18.
Front Immunol ; 11: 554725, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33072095

RESUMEN

Systemic Lupus Erythematosus (SLE) is a chronic inflammatory autoimmune disease in which type I interferons (IFN) play a key role. The IFN response can be triggered when oxidized DNA engages the cytosolic DNA sensing platform cGAS-STING, but the repair mechanisms that modulate this process and govern disease progression are unclear. To gain insight into this biology, we interrogated the role of oxyguanine glycosylase 1 (OGG1), which repairs oxidized guanine 8-Oxo-2'-deoxyguanosine (8-OH-dG), in the pristane-induced mouse model of SLE. Ogg1-/- mice showed increased influx of Ly6Chi monocytes into the peritoneal cavity and enhanced IFN-driven gene expression in response to short-term exposure to pristane. Loss of Ogg1 was associated with increased auto-antibodies (anti-dsDNA and anti-RNP), higher total IgG, and expression of interferon stimulated genes (ISG) to longer exposure to pristane, accompanied by aggravated skin pathology such as hair loss, thicker epidermis, and increased deposition of IgG in skin lesions. Supporting a role for type I IFNs in this model, skin lesions of Ogg1-/- mice had significantly higher expression of type I IFN genes (Isg15, Irf9, and Ifnb). In keeping with loss of Ogg1 resulting in dysregulated IFN responses, enhanced basal and cGAMP-dependent Ifnb expression was observed in BMDMs from Ogg1-/- mice. Use of the STING inhibitor, H151, reduced both basal and cGAMP-driven increases, indicating that OGG1 regulates Ifnb expression through the cGAS-STING pathway. Finally, in support for a role for OGG1 in the pathology of cutaneous disease, reduced OGG1 expression in monocytes associated with skin involvement in SLE patients and the expression of OGG1 was significantly lower in lesional skin compared with non-lesional skin in patients with Discoid Lupus. Taken together, these data support an important role for OGG1 in protecting against IFN production and SLE skin disease.


Asunto(s)
Daño del ADN/inmunología , Lupus Eritematoso Cutáneo/inmunología , Lupus Eritematoso Sistémico/inmunología , Piel/inmunología , Terpenos/efectos adversos , Animales , ADN Glicosilasas/deficiencia , ADN Glicosilasas/inmunología , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Lupus Eritematoso Cutáneo/inducido químicamente , Lupus Eritematoso Cutáneo/genética , Lupus Eritematoso Cutáneo/patología , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Oxidación-Reducción/efectos de los fármacos , Piel/patología , Terpenos/farmacología
19.
Science ; 366(6472): 1531-1536, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31857488

RESUMEN

Mitochondrial stress releases mitochondrial DNA (mtDNA) into the cytosol, thereby triggering the type Ι interferon (IFN) response. Mitochondrial outer membrane permeabilization, which is required for mtDNA release, has been extensively studied in apoptotic cells, but little is known about its role in live cells. We found that oxidatively stressed mitochondria release short mtDNA fragments via pores formed by the voltage-dependent anion channel (VDAC) oligomers in the mitochondrial outer membrane. Furthermore, the positively charged residues in the N-terminal domain of VDAC1 interact with mtDNA, promoting VDAC1 oligomerization. The VDAC oligomerization inhibitor VBIT-4 decreases mtDNA release, IFN signaling, neutrophil extracellular traps, and disease severity in a mouse model of systemic lupus erythematosus. Thus, inhibiting VDAC oligomerization is a potential therapeutic approach for diseases associated with mtDNA release.


Asunto(s)
ADN Mitocondrial/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Membranas Mitocondriales/metabolismo , Multimerización de Proteína , Canales Aniónicos Dependientes del Voltaje/metabolismo , Animales , Modelos Animales de Enfermedad , Endodesoxirribonucleasas/genética , Humanos , Interferones/metabolismo , Lupus Eritematoso Sistémico/tratamiento farmacológico , Ratones , Estrés Oxidativo , Dominios Proteicos , Multimerización de Proteína/efectos de los fármacos , Ratas , Canales Aniónicos Dependientes del Voltaje/antagonistas & inhibidores , Canales Aniónicos Dependientes del Voltaje/genética
20.
Arthritis Rheumatol ; 75(1): 143-144, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35969518
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA