Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Clin Invest ; 131(24)2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34710063

RESUMO

Acute COVID-19, caused by SARS-CoV-2, is characterized by diverse clinical presentations, ranging from asymptomatic infection to fatal respiratory failure, and often associated with varied longer-term sequelae. Over the past 18 months, it has become apparent that inappropriate immune responses contribute to the pathogenesis of severe COVID-19. Researchers working at the intersection of COVID-19 and autoimmunity recently gathered at an American Autoimmune Related Diseases Association Noel R. Rose Colloquium to address the current state of knowledge regarding two important questions: Does established autoimmunity predispose to severe COVID-19? And, at the same time, can SARS-CoV-2 infection trigger de novo autoimmunity? Indeed, work to date has demonstrated that 10% to 15% of patients with critical COVID-19 pneumonia exhibit autoantibodies against type I interferons, suggesting that preexisting autoimmunity underlies severe disease in some patients. Other studies have identified functional autoantibodies following infection with SARS-CoV-2, such as those that promote thrombosis or antagonize cytokine signaling. These autoantibodies may arise from a predominantly extrafollicular B cell response that is more prone to generating autoantibody-secreting B cells. This Review highlights the current understanding, evolving concepts, and unanswered questions provided by this unique opportunity to determine mechanisms by which a viral infection can be exacerbated by, and even trigger, autoimmunity. The potential role of autoimmunity in post-acute sequelae of COVID-19 is also discussed.


Assuntos
Autoanticorpos/química , Autoimunidade/imunologia , COVID-19/imunologia , COVID-19/fisiopatologia , Transdução de Sinais , Animais , Doenças Autoimunes , Linfócitos B/citologia , Citocinas/metabolismo , Progressão da Doença , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Inflamação , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Ativação de Macrófagos , Masculino , Camundongos , Fosfolipídeos/metabolismo , SARS-CoV-2
2.
J Vasc Surg Venous Lymphat Disord ; 9(3): 585-591.e2, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32979557

RESUMO

BACKGROUND: Infection with the novel severe acute respiratory syndrome coronavirus 2 has been associated with a hypercoagulable state. Emerging data from China and Europe have consistently shown an increased incidence of venous thromboembolism (VTE). We aimed to identify the VTE incidence and early predictors of VTE at our high-volume tertiary care center. METHODS: We performed a retrospective cohort study of 147 patients who had been admitted to Temple University Hospital with coronavirus disease 2019 (COVID-19) from April 1, 2020 to April 27, 2020. We first identified the VTE (pulmonary embolism [PE] and deep vein thrombosis [DVT]) incidence in our cohort. The VTE and no-VTE groups were compared by univariable analysis for demographics, comorbidities, laboratory data, and treatment outcomes. Subsequently, multivariable logistic regression analysis was performed to identify the early predictors of VTE. RESULTS: The 147 patients (20.9% of all admissions) admitted to a designated COVID-19 unit at Temple University Hospital with a high clinical suspicion of acute VTE had undergone testing for VTE using computed tomography pulmonary angiography and/or extremity venous duplex ultrasonography. The overall incidence of VTE was 17% (25 of 147). Of the 25 patients, 16 had had acute PE, 14 had had acute DVT, and 5 had had both PE and DVT. The need for invasive mechanical ventilation (adjusted odds ratio, 3.19; 95% confidence interval, 1.07-9.55) and the admission D-dimer level ≥1500 ng/mL (adjusted odds ratio, 3.55; 95% confidence interval, 1.29-9.78) were independent markers associated with VTE. The all-cause mortality in the VTE group was greater than that in the non-VTE group (48% vs 22%; P = .007). CONCLUSIONS: Our study represents one of the earliest reported from the United States on the incidence rate of VTE in patients with COVID-19. Patients with a high clinical suspicion and the identified risk factors (invasive mechanical ventilation, admission D-dimer level ≥1500 ng/mL) should be considered for early VTE testing. We did not screen all patients admitted for VTE; therefore, the true incidence of VTE could have been underestimated. Our findings require confirmation in future prospective studies.


Assuntos
COVID-19 , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Embolia Pulmonar , Respiração Artificial/métodos , Trombose Venosa , COVID-19/sangue , COVID-19/complicações , COVID-19/epidemiologia , Angiografia por Tomografia Computadorizada/métodos , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Philadelphia/epidemiologia , Prognóstico , Embolia Pulmonar/diagnóstico , Embolia Pulmonar/epidemiologia , Embolia Pulmonar/etiologia , Estudos Retrospectivos , Fatores de Risco , SARS-CoV-2 , Trombofilia/sangue , Trombofilia/diagnóstico , Trombofilia/etiologia , Ultrassonografia Doppler Dupla/métodos , Trombose Venosa/diagnóstico , Trombose Venosa/epidemiologia , Trombose Venosa/etiologia
3.
Cureus ; 12(5): e8130, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32550050

RESUMO

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening condition caused by overproduction of inflammatory cytokines and overactivation of macrophages that can progress to multiorgan dysfunction and failure. Although there are guidelines that attempt to recognize the condition in its early stage, diagnosis can be very challenging due to heterogeneous presentations of HLH. Symptoms and clinical findings include fever, neurologic complaints, respiratory issues, liver dysfunction, cytopenias, amongst others most of which are not specific to HLH. In addition, response to treatment can be highly variable, necessitating an individualized treatment plan based on the presentation. We present a case of a 21-year-old female with a history of biopsy-proven inflammatory myositis on azathioprine and prednisone who presented with fever, hypotension, and pancytopenia. Additional imaging studies showed multiorgan involvement, including pneumonia, pyelonephritis, and splenomegaly. A bone marrow biopsy of her iliac crest showed hemophagocytosis and the infectious workup confirmed cytomegalovirus (CMV) infection, which led to the diagnosis of CMV-induced HLH. She was treated initially with anakinra for macrophage activation syndrome (MAS) in addition to dexamethasone and ganciclovir. Unfortunately, she did not respond to anakinra and was subsequently switched to etoposide with dexamethasone and valganciclovir, which subsequently helped our patient to recover clinically. Our case highlights the challenging nature of HLH and the importance of early detection and a personalized treatment plan in achieving optimal outcomes in patients with HLH.

4.
J Leukoc Biol ; 106(4): 967-975, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31216373

RESUMO

IL-10 is elevated in the autoimmune disease systemic lupus erythematosus (SLE). Here, we show that conventional dendritic cells (cDCs) from predisease lupus-prone B6.NZM Sle1/Sle2/Sle3 triple congenic (TCSle) mice produce more IL-10 than wild-type congenic cDCs upon TLR stimulation, and this overproduction is prevented by blocking the type I IFN receptor (IFNAR) with specific Abs. Priming wild-type cDCs with type I IFN mimics the IL-10 overproduction of TCSle cDCs. The MAPK ERK is more phosphorylated in lupus cDCs, partially contributing to IL-10 overproduction. Moreover, we found that TCSle cDCs express higher levels of IL-27 upon TLR7/TLR9 stimulation, and IFNAR blockade reduced IL-27 levels in TCSle cDCs. These results suggest that dysregulated type I IFNs in cDCs contribute to the increased IL-10 and IL-27 in SLE. Since IL-27 neutralization did not inhibit TLR-induced IL-10 production, we propose that type I IFNs enhanced IL-10 in TCSle cDCs independently from IL-27. Moreover, RNA sequencing analysis of a cohort of SLE patients reveals higher gene expression of these cytokines in SLE patients expressing a high IFN signature. Since IL-27 and IL-10 have both pro- and anti-inflammatory effects, our results also suggest that these cytokines can be modulated by the therapeutic IFN blockade in trials in SLE patients and have complex effects on the autoimmune response.


Assuntos
Interferon Tipo I/metabolismo , Interleucina-10/metabolismo , Interleucina-27/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Animais , Antígenos CD40/metabolismo , Quimiocina CXCL10/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Imidazóis/farmacologia , Ligantes , Lipopolissacarídeos/farmacologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/farmacologia , Receptor de Interferon alfa e beta/metabolismo , Receptores Toll-Like/metabolismo
5.
Front Immunol ; 10: 30, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30761126

RESUMO

Attenuating the innate immunity activation could ameliorate inflammation and disease in settings such as transplant rejection or autoimmunity. Recently, a pivotal role for metabolic re-programming in TLR-induced dendritic cell (DC) activation has emerged. Ethyl pyruvate (EP), a pyruvate derivative, possesses anti-inflammatory properties in vitro and in animal models of disease. However, its effects on DCs remain elusive. We found that EP attenuated LPS-induced activation of murine GM-CSF bone marrow-derived dendritic cells (DCs) in vitro, reducing pro-inflammatory cytokine and IL-10 production, costimulatory molecule and MHC expression, the type I Interferon (IFN-I) response, the LPS-induced cell death, and the ability of DCs to stimulate allogeneic T cells. DC activation induced by TLR7 and TLR9 ligands was also suppressed by EP in vitro. Finally, EP decreased TLR-induced activation stimulated in vivo in conventional DCs and inflammatory monocytes. Investigating EP mechanisms, we found that EP decreased glycolysis and mitochondrial respiration, upon and in absence of TLR stimulation, by reducing ERK, AKT, and nitric oxide (NO) activation. These results indicate that EP inhibits most of the DC biological responses to TLR triggering, altering the metabolic reprogramming necessary for DC activation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Metabolismo Energético , Imunomodulação , Piruvatos/metabolismo , Animais , Sobrevivência Celular/imunologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imunidade Inata , Imunomodulação/efeitos dos fármacos , Lipopolissacarídeos/imunologia , Linfonodos/imunologia , Linfonodos/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Óxido Nítrico/biossíntese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piruvatos/farmacologia , Baço/imunologia , Baço/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
J Immunol Res ; 2018: 1601079, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29850618

RESUMO

Type I interferons (IFN) are pathogenic in systemic lupus erythematosus (SLE) and were proposed to control the immunometabolism of dendritic cells (DCs). We previously reported that DCs from female lupus-prone mice constitutively overexpress IFN-responsive genes resembling the IFN signature found in SLE patients. As SLE has higher incidence in women than men, more so in women of reproductive age, estrogens are suggested to affect lupus pathogenesis. We investigated the effects of sex and estrogens on the IFN signature in conventional GM-CSF-bone marrow-derived DCs (cDCs), from male and female Triple Congenic B6.NZM.Sle1/Sle2/Sle3 (TCSle) lupus-prone mice or from wild-type C57BL/6 mice, generated with titrations of 17-beta-estradiol (E2). We found that cDCs from prediseased TCSle male mice express the IFN signature as female TCSle cDCs do. Estrogens are necessary but not sufficient to express this IFN signature, but high doses of E2 can compensate for other steroidal components. E2 stimulation, regardless of sex, modulates type I IFN-dependent and type I IFN-independent activation of cDCs in response to TLR stimulation. Finally, we found that TCSle cDCs from both sexes have elevated markers of immunometabolism and estrogens enhance the metabolic pathways in cDCs, suggesting a mechanistic link between estrogens, immunometabolism, and the IFN signature in lupus.


Assuntos
Células Dendríticas/imunologia , Estradiol/metabolismo , Interferon Tipo I/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Nefrite Lúpica/metabolismo , Animais , Autoanticorpos/metabolismo , Células da Medula Óssea/citologia , Modelos Animais de Doenças , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Interferon Tipo I/genética , Lúpus Eritematoso Sistêmico/imunologia , Nefrite Lúpica/genética , Nefrite Lúpica/imunologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transcriptoma
7.
J Immunol ; 200(2): 512-522, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29237779

RESUMO

Glomerulonephritis is one of the most serious manifestations of systemic lupus erythematous (SLE). Because SLE is ≥10 times more common in women, a role for estrogens in disease pathogenesis has long been suspected. Estrogen receptor α (ERα) is highly expressed in renal tissue. We asked whether ERα expression contributes to the development of immune-mediated nephropathies like in lupus nephritis. We tested the overall effects of estrogen receptors on the immune response by immunization with OVA and induction of chronic graft-versus-host disease in female ERα-knockout mice. We used nephrotoxic serum nephritis as a model of immune-mediated nephropathy. We investigated the influence of ERα on molecular pathways during nephritis by microarray analysis of glomerular extract gene expression. We performed RNA sequencing of lupus patient whole blood to determine common pathways in murine and human nephritis. Absence of ERα protects female mice from developing nephritis, despite the presence of immune complexes and the production of proinflammatory cytokines in the kidneys and normal humoral responses to immunization. Time-course microarray analysis of glomeruli during nephrotoxic serum nephritis revealed significant upregulation of genes related to PPAR-mediated lipid metabolism and downregulation of genes in the retinol metabolism in wild-type females compared with ERα-knockout females. Similarly, RNA sequencing of lupus patient blood revealed similar expression patterns of these same pathways. During nephritis, the altered activity of metabolic pathways, such as retinol metabolism, occurs downstream of ERα activation and is essential for the progression to end-stage renal failure.


Assuntos
Metabolismo Energético , Receptor alfa de Estrogênio/metabolismo , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Transdução de Sinais , Animais , Complexo Antígeno-Anticorpo/imunologia , Complexo Antígeno-Anticorpo/metabolismo , Autoanticorpos/imunologia , Biologia Computacional , Modelos Animais de Doenças , Progressão da Doença , Suscetibilidade a Doenças , Receptor alfa de Estrogênio/genética , Expressão Gênica , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Glomerulonefrite/patologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Humanos , Metabolismo dos Lipídeos , Lúpus Eritematoso Sistêmico/complicações , Nefrite Lúpica/imunologia , Nefrite Lúpica/metabolismo , Nefrite Lúpica/patologia , Camundongos , Camundongos Knockout , Fatores Sexuais
8.
J Immunol ; 196(3): 1070-80, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26712946

RESUMO

Notch signaling regulates multiple helper CD4(+) T cell programs. We have recently demonstrated that dendritic cells (DCs) expressing the Notch ligand DLL4 are critical for eliciting alloreactive T cell responses and induction of graft-versus-host disease in mice. However, the human counterpart of murine DLL4(+) DCs has yet to be examined. We report the identification of human DLL4(+) DCs and their critical role in regulating Th1 and Th17 differentiation. CD1c(+) DCs and plasmacytoid DCs (pDCs) from the peripheral blood (PB) of healthy donors did not express DLL4. In contrast, patients undergoing allogeneic hematopoietic stem cell transplantation had a 16-fold more DLL4(+)CD1c(+) DCs than healthy donors. Upon activation of TLR signaling, healthy donor-derived CD1c(+) DCs dramatically upregulated DLL4, as did pDCs to a lesser extent. Activated DLL4(+) DCs were better able to promote Th1 and Th17 differentiation than unstimulated PB DCs. Blocking DLL4 using a neutralizing Ab decreased Notch signaling in T cells stimulated with DLL4(+) DCs, and it reduced the generation of Th1 and Th17 cells. Both NF-κB and STAT3 were crucial for inducing DLL4 in human DCs. Interestingly, STAT3 directly activated DLL4 transcription and inhibiting STAT3 alone was sufficient to reduce DLL4 in activated PB DCs. Thus, DLL4 is a unique functional molecule of human circulating DCs critical for directing Th1 and Th17 differentiation. These findings identify a pathway for therapeutic intervention for inflammatory disorders in humans, such as graft-versus-host disease after allogeneic hematopoietic stem cell transplantation, autoimmunity, and tumor immunity.


Assuntos
Diferenciação Celular , Células Dendríticas/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Ativação Linfocitária/imunologia , Células Th1/imunologia , Células Th17/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Aloenxertos/imunologia , Western Blotting , Proteínas de Ligação ao Cálcio , Diferenciação Celular/imunologia , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Humanos , Teste de Cultura Mista de Linfócitos , Reação em Cadeia da Polimerase em Tempo Real , Células Th1/citologia , Células Th17/citologia
9.
Immunity ; 42(6): 1171-84, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26084027

RESUMO

Research on the human microbiome has established that commensal and pathogenic bacteria can influence obesity, cancer, and autoimmunity through mechanisms mostly unknown. We found that a component of bacterial biofilms, the amyloid protein curli, irreversibly formed fibers with bacterial DNA during biofilm formation. This interaction accelerated amyloid polymerization and created potent immunogenic complexes that activated immune cells, including dendritic cells, to produce cytokines such as type I interferons, which are pathogenic in systemic lupus erythematosus (SLE). When given systemically, curli-DNA composites triggered immune activation and production of autoantibodies in lupus-prone and wild-type mice. We also found that the infection of lupus-prone mice with curli-producing bacteria triggered higher autoantibody titers compared to curli-deficient bacteria. These data provide a mechanism by which the microbiome and biofilm-producing enteric infections may contribute to the progression of SLE and point to a potential molecular target for treatment of autoimmunity.


Assuntos
Amiloide/metabolismo , Proteínas de Bactérias/metabolismo , DNA Bacteriano/metabolismo , Células Dendríticas/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Amiloide/imunologia , Animais , Autoanticorpos/biossíntese , Proteínas de Bactérias/imunologia , Biofilmes/crescimento & desenvolvimento , Células Cultivadas , DNA Bacteriano/imunologia , Humanos , Interferon Tipo I/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Polimerização
10.
Clin Immunol ; 159(1): 13-22, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25926428

RESUMO

We showed previously that 17ß estradiol (E2) led to improved survival in nephrotoxic serum induced nephritis (NTN) in male mice. In this study we determined whether E2 regulates vascular cell adhesion molecule (VCAM)-1, an adhesion molecule that is upregulated in kidney during autoimmune nephritis, in mesangial cells (MC). We show that E2 inhibited VCAM-1 up-regulation in kidneys in vivo during NTN, and in MCs upon TNFα stimulation. VCAM-1 up-regulation in MCs was controlled by the transcription factor NFκB. E2 inhibited RNA polymerase II recruitment to the VCAM-1 promoter, but not p65 recruitment. Interestingly E2 inhibited TNFα stimulated interaction between poly (ADP-ribose) polymerase-1 (PARP-1) and p65. As PARP-1 is required for VCAM-1 upregulation in MCs, our data suggest that E2 may inhibit pre-initiation complex formation at VCAM-1 promoter by inhibiting PARP-1 recruitment to p65. We propose that E2 plays an important role in regulating renal inflammation locally.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Glomerulonefrite , Células Mesangiais/efeitos dos fármacos , RNA Mensageiro/metabolismo , Molécula 1 de Adesão de Célula Vascular/efeitos dos fármacos , Animais , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/metabolismo , Células Mesangiais/metabolismo , Camundongos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Polimerase II/efeitos dos fármacos , RNA Polimerase II/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição RelA/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/genética
11.
Sci Signal ; 8(366): ra23, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25737585

RESUMO

Cytosolic Ca2+ signals, generated through the coordinated translocation of Ca2+ across the plasma membrane (PM) and endoplasmic reticulum (ER) membrane, mediate diverse cellular responses. Mitochondrial Ca2+ is important for mitochondrial function, and when cytosolic Ca2+ concentration becomes too high, mitochondria function as cellular Ca2+ sinks. By measuring mitochondrial Ca2+ currents, we found that mitochondrial Ca2+ uptake was reduced in chicken DT40 B lymphocytes lacking either the ER-localized inositol trisphosphate receptor (IP3R), which releases Ca2+ from the ER, or Orai1 or STIM1, components of the PM-localized Ca2+ -permeable channel complex that mediates store-operated calcium entry (SOCE) in response to depletion of ER Ca2+ stores. The abundance of MCU, the pore-forming subunit of the mitochondrial Ca2+ uniporter, was reduced in cells deficient in IP3R, STIM1, or Orai1. Chromatin immunoprecipitation and promoter reporter analyses revealed that the Ca2+ -regulated transcription factor CREB (cyclic adenosine monophosphate response element-binding protein) directly bound the MCU promoter and stimulated expression. Lymphocytes deficient in IP3R, STIM1, or Orai1 exhibited altered mitochondrial metabolism, indicating that Ca2+ released from the ER and SOCE-mediated signals modulates mitochondrial function. Thus, our results showed that a transcriptional regulatory circuit involving Ca2+ -dependent activation of CREB controls the Ca2+ uptake capability of mitochondria and hence regulates mitochondrial metabolism.


Assuntos
Proteínas Aviárias/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Mitocondriais/metabolismo , Animais , Proteínas Aviárias/genética , Canais de Cálcio/genética , Linhagem Celular , Galinhas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Retículo Endoplasmático , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Proteína ORAI1 , Molécula 1 de Interação Estromal
12.
EMBO Mol Med ; 3(10): 605-15, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21882343

RESUMO

MicroRNAs (miRNAs) have been implicated in B cell lineage commitment, regulation of T cell differentiation, TCR signalling, regulation of IFN signalling, and numerous other immunological processes. However, their function in autoimmunity, and specifically in systemic lupus erythematosus (SLE), remains poorly understood. B6.Sle123 is a spontaneous genetic mouse model of SLE characterized by autoantibody production, lymphosplenomegaly, and glomerulonephritis. We identified several differentially regulated miRNAs in B and T lymphocytes of B6.Sle123 mice. We found that miR-21 expression in lupus B and T cells is up-regulated and that in vivo silencing of miR-21 using a tiny seed-targeting LNA reversed splenomegaly, one of the cardinal manifestations of autoimmunity in B6.Sle123 mice, and de-repressed PDCD4 expression in vivo and in vitro. In addition, treatment with anti-miR-21 altered CD4/CD8 T cell ratios and reduced Fas receptor-expressing lymphocyte populations. Our study shows that tiny LNAs can be used to efficiently antagonize endogenous miRNAs in peripheral lymphocytes in vivo and in primary lymphocytes cultured ex vivo and can alter the course of a spontaneous genetic disease in mice.


Assuntos
Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Inativação Gênica , Lúpus Eritematoso Sistêmico/genética , MicroRNAs/genética , Esplenomegalia/genética , Esplenomegalia/imunologia , Envelhecimento/efeitos dos fármacos , Envelhecimento/patologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Relação CD4-CD8 , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/imunologia , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Oligonucleotídeos/farmacologia , Proteínas de Ligação a RNA/metabolismo , Esplenomegalia/complicações , Esplenomegalia/patologia , Transcrição Gênica/efeitos dos fármacos , Receptor fas/metabolismo
13.
J Immunol ; 185(12): 7151-5, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21076068

RESUMO

A hallmark of autoimmune lymphoproliferative syndrome (ALPS), caused by mutation of the Fas death receptor, is massive lymphadenopathy from aberrant expansion of CD4(-)CD8(-) (double-negative [DN]) T cells. Eomesodermin (Eomes) is a member of the T-box family of transcription factors and plays critical roles in effector cell function and memory cell fitness of CD8(+) T lymphocytes. We provide evidence in this study that DN T cells exhibit dysregulated expression of Eomes in humans and mice with ALPS. We also find that T cell-specific deletion of Eomes prevents lymphoid hypertrophy and accumulation of DN T cells in Fas-mutant mice. Although Eomes has critical physiological roles in the function and homeostasis of CD8(+) T cells, overexpression of Eomes appears to enable pathological induction or expansion of unusual CD8-related T cell subsets. Thus, antagonism of Eomes emerges as a therapeutic target for DN T cell ablation in ALPS.


Assuntos
Síndrome Linfoproliferativa Autoimune/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas com Domínio T/imunologia , Subpopulações de Linfócitos T/imunologia , Receptor fas , Animais , Síndrome Linfoproliferativa Autoimune/genética , Síndrome Linfoproliferativa Autoimune/patologia , Síndrome Linfoproliferativa Autoimune/terapia , Linfócitos T CD8-Positivos/patologia , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Memória Imunológica/genética , Memória Imunológica/imunologia , Masculino , Camundongos , Camundongos Knockout , Proteínas com Domínio T/genética , Subpopulações de Linfócitos T/patologia
14.
J Immunol ; 182(11): 7297-306, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19454727

RESUMO

Necrotic lesions and necrotic cell death characterize severe autoimmune nephritides, and contribute to local inflammation and to progression of the disease. Poly(ADP-ribose) polymerase-1 (PARP-1), a DNA repair enzyme, is involved in the induction of necrosis and is a key player in the acute and chronic inflammation. Therefore, we hypothesized that PARP-1 controls the severity of nephritis by mediating the induction of necrosis in the kidney. We used lupus and anti-glomerular basement membrane models of nephritis to determine the effects of PARP-1 on the inflammatory response in the kidney. We show in this study that PARP-1 is indeed activated during the course of glomerulonephritis. We also show that the absence of PARP-1 or its pharmacological inhibition results in milder nephritis, with lower blood urea nitrogen levels, reduced necrotic lesions, and higher survival rates. The relevance of PARP-1 showed a strong male sex specificity, and treatment of male mice with 17beta-estradiol prolonged their survival during the course of nephritis. PARP-1 also regulated TNF-alpha expression and up-regulation of adhesion molecules, further supporting a role of PARP-1 in the inflammatory process within the kidney. Our results demonstrate that PARP-1 activation and consequent necrotic cell death play an important role in the pathogenesis of male nephritis, and suggest that PARP-1 can be a novel therapeutic target in glomerulonephritis.


Assuntos
Glomerulonefrite/patologia , Inflamação/patologia , Necrose/etiologia , Poli(ADP-Ribose) Polimerases/fisiologia , Animais , Doenças Autoimunes/patologia , Progressão da Doença , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Rim/patologia , Masculino , Camundongos , Poli(ADP-Ribose) Polimerases/deficiência , Fatores Sexuais , Taxa de Sobrevida
15.
Arthritis Res Ther ; 8(2): R49, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16507174

RESUMO

We analyzed the activation and function of dendritic cells (DCs) in the spleens of diseased, lupus-prone NZM2410 and NZB-W/F1 mice and age-matched BALB/c and C57BL/6 control mice. Lupus DCs showed an altered ex vivo costimulatory profile, with a significant increase in the expression of CD40, decreased expression of CD80 and CD54, and normal expression of CD86. DCs from young lupus-prone NZM2410 mice, before the development of the disease, expressed normal levels of CD80 and CD86 but already overexpressed CD40. The increase in CD40-positive cells was specific for DCs and involved the subset of myeloid and CD8alpha+ DCs before disease onset, with a small involvement of plasmacytoid DCs in diseased mice. In vitro data from bone marrow-derived DCs and splenic myeloid DCs suggest that the overexpression of CD40 is not due to a primary alteration of CD40 regulation in DCs but rather to an extrinsic stimulus. Our analyses suggest that the defect of CD80 in NZM2410 and NZB-W/F1 mice, which closely resembles the costimulatory defect found in DCs from humans with systemic lupus erythematosus, is linked to the autoimmune disease. The increase in CD40 may instead participate in disease pathogenesis, being present months before any sign of autoimmunity, and its downregulation should be explored as an alternative to treatment with anti-CD40 ligand in lupus.


Assuntos
Células Dendríticas , Lúpus Vulgar/patologia , Lúpus Vulgar/fisiopatologia , Fenótipo , Baço/patologia , Envelhecimento/imunologia , Animais , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Antígenos CD40/metabolismo , Células Dendríticas/imunologia , Suscetibilidade a Doenças , Lúpus Vulgar/imunologia , Camundongos , Camundongos Endogâmicos , Índice de Gravidade de Doença , Baço/imunologia
16.
J Immunol ; 176(4): 2095-104, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16455965

RESUMO

Increased monocyte/macrophage (Mphi) apoptosis occurs in patients with systemic lupus erythematosus (SLE) and is mediated, at least in part, by an autoreactive CD4(+) T cell subset. Furthermore, autoreactive murine CD4(+) T cells that kill syngeneic Mphi in vitro induce a lupus-like disease in vivo. However, it is unclear whether increased Mphi apoptosis in SLE per se is sufficient to accelerate/promote autoimmunity. We have investigated whether increased Mphi apoptosis in vivo, induced by the administration of clodronate liposomes, can exacerbate the autoimmune phenotype in NZB x SWR (SNF(1)) lupus-prone mice, and induce autoantibody production in haplotype-matched BALB/c x DBA1 (DBF(1)) non-lupus-prone mice. Lupus-prone mice SNF(1) mice that were treated with clodronate liposomes, but not mice treated with vehicle, developed significant increases in autoantibodies to dsDNA, nucleosomes, and the idiotypically related family of nephritic Abs Id(LN)F(1), when compared with untreated SNF(1) mice. Furthermore, clodronate treatment hastened the onset of proteinuria and worsened SNF(1) lupus nephritis. When compared with vehicle-treated controls, clodronate-treated non-lupus-prone DBF(1) mice developed significantly higher levels of anti-nucleosome and Id(LN)F(1) Abs but did not develop lupus nephritis. We propose that Mphi apoptosis contributes to the pathogenesis of autoantibody formation and organ damage through both an increase in the apoptotic load and impairment in the clearance of apoptotic material. This study suggests that mechanisms that induce scavenger cell apoptosis, such as death induced by autoreactive cytotoxic T cells observed in SLE, could play a pathogenic role and contribute to the severity of the disease.


Assuntos
Formação de Anticorpos/imunologia , Apoptose , Autoanticorpos/biossíntese , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Macrófagos/citologia , Animais , Autoanticorpos/imunologia , Feminino , Rim/patologia , Lipossomos , Macrófagos/imunologia , Camundongos , Baço/patologia
17.
J Immunol ; 175(7): 4777-82, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16177126

RESUMO

Intraperitoneal injection of the hydrocarbon oil pristane into normal mice leads to a lupus-like autoimmune syndrome. Although advances in defining the roles of cellular and humoral mediators involved in this syndrome have been made, the mechanisms that initiate a break in tolerance leading to autoimmunity remain unknown. We describe in this study that pristane induces apoptosis both in vivo and in vitro. Pristane arrests cell growth and induces cell death by apoptosis via the mitochondrial pathway of caspase activation in a dose-dependent manner. Nuclear autoantigens created by pristane-induced apoptosis of lymphoid cells within the peritoneal cavity in the setting of a profoundly altered cytokine milieu may be the initiating event in the development of autoimmunity in this syndrome. These findings suggest that apoptosis may be a critical initial event in the pathogenesis of pristane-induced lupus and are of potential relevance for human systemic lupus erythematosus.


Assuntos
Apoptose/efeitos dos fármacos , Lúpus Eritematoso Sistêmico/induzido quimicamente , Terpenos/farmacologia , Animais , Líquido Ascítico/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Injeções Intraperitoneais , Células Jurkat , Lúpus Eritematoso Sistêmico/patologia , Linfócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Terpenos/administração & dosagem
18.
J Immunol ; 175(4): 2692-701, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16081846

RESUMO

Autoreactivity in lupus requires the delivery of autoantigens to APCs in a proinflammatory context. It has been proposed that apoptotic cells are a source of lupus autoantigens and targets for autoantibodies. Using a histone H2B-GFP fusion protein as traceable Ag, we show here that lupus autoantibodies, directed against nuclear autoantigens, can opsonize apoptotic cells, enhance their uptake through induction of proinflammatory Fc gammaR-mediated phagocytosis, and augment Ag-specific T cell proliferation by increasing Ag loading. Apoptotic blebs and bodies seemed to be a preferred target of DC phagocytosis, via both "eat-me signals" and Fc gammaR-mediated mechanisms; furthermore, inhibition of nuclear Ag redistribution, by blockade of chromatin fragmentation, could stop binding and opsonization of apoptotic cells by autoantibodies, and inhibited Fc gamma-R-mediated enhancement of phagocytosis. Our results suggest that DC uptake of opsonized histones and other nuclear Ags from apoptotic cells is a novel pathway for the presentation of nuclear Ags in a highly inflammatory context. Blockade of chromatin fragmentation in lupus is a potential therapeutic approach, which could theoretically limit DC access to autoantigens delivered in proinflammatory context, while leaving available for tolerization those delivered in a noninflammatory context.


Assuntos
Anticorpos Antinucleares/metabolismo , Apresentação de Antígeno/imunologia , Autoantígenos/metabolismo , Células Dendríticas/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Proteínas Nucleares/metabolismo , Proteínas Opsonizantes/metabolismo , Fagocitose/imunologia , Transporte Ativo do Núcleo Celular/imunologia , Animais , Anticorpos Antinucleares/fisiologia , Apoptose/imunologia , Apoptose/efeitos da radiação , Autoantígenos/imunologia , Sítios de Ligação de Anticorpos , Linhagem Celular Tumoral , Cromatina/metabolismo , Fragmentação do DNA/imunologia , Células Dendríticas/metabolismo , Proteínas de Fluorescência Verde/genética , Histonas/genética , Histonas/imunologia , Histonas/metabolismo , Humanos , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/imunologia , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo
19.
J Immunol ; 171(11): 5778-86, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14634086

RESUMO

The nuclear self-Ags targeted in systemic lupus erythematosus translocate to the cell membrane of UV-irradiated apoptotic keratinocytes and may represent an important source of self-immunization. It is hard to understand how the noninflammatory milieu accompanying most apoptosis might provoke an immunogenic response leading to autoantibodies. We have found that the precise amount of keratinocyte UV exposure is crucial in determining the rate of apoptosis, the amount of inflammatory cytokine production, and the degree of autoantigen translocation. Low doses of UVB (

Assuntos
Apoptose/imunologia , Apoptose/efeitos da radiação , Autoantígenos/metabolismo , Autoantígenos/efeitos da radiação , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Raios Ultravioleta , Antígenos Nucleares/metabolismo , Morte Celular/imunologia , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Fragmentação do DNA/efeitos da radiação , Relação Dose-Resposta Imunológica , Relação Dose-Resposta à Radiação , Ativação Enzimática/efeitos da radiação , Humanos , Hidrólise/efeitos da radiação , Inflamação/enzimologia , Inflamação/imunologia , Interleucina-1/biossíntese , Interleucina-1/efeitos da radiação , Proteínas Quinases JNK Ativadas por Mitógeno , Queratinócitos/citologia , Queratinócitos/enzimologia , Queratinócitos/imunologia , Queratinócitos/efeitos da radiação , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos da radiação , Poli(ADP-Ribose) Polimerases/metabolismo , Poli(ADP-Ribose) Polimerases/efeitos da radiação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/efeitos da radiação
20.
J Exp Med ; 196(1): 135-40, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12093878

RESUMO

Mice lacking the membrane tyrosine kinase c-mer have been shown to have altered macro-phage cytokine production and defective phagocytosis of apoptotic cells despite normal phagocytosis of other particles. We show here that c-mer-deficient mice have impaired clearance of infused apoptotic cells and that they develop progressive lupus-like autoimmunity, with antibodies to chromatin, DNA, and IgG. The autoimmunity appears to be driven by endogenous antigens, with little polyclonal B cell activation. These mice should be an excellent model for studying the role of apoptotic debris as an immunogenic stimulus for systemic autoimmunity.


Assuntos
Apoptose/imunologia , Autoimunidade/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Proteínas Tirosina Quinases/deficiência , Proteínas Proto-Oncogênicas/deficiência , Receptores Proteína Tirosina Quinases , Animais , Autoanticorpos/sangue , Linfócitos B/imunologia , Cardiolipinas/imunologia , Cromatina/imunologia , DNA/imunologia , Modelos Animais de Doenças , Feminino , Corantes Fluorescentes , Mesângio Glomerular/patologia , Imunoglobulina G/imunologia , Lúpus Eritematoso Sistêmico/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fagocitose/imunologia , Proteínas Tirosina Quinases/genética , Proteinúria/complicações , Proteinúria/patologia , Proteínas Proto-Oncogênicas/genética , Fator Reumatoide/sangue , Rodaminas , c-Mer Tirosina Quinase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA