38 research outputs found

    Aging-associated deficit in CCR7 is linked to worsened glymphatic function, cognition, neuroinflammation, and β-amyloid pathology

    Get PDF
    Aging leads to a progressive deterioration of meningeal lymphatics and peripheral immunity, which may accelerate cognitive decline. We hypothesized that an age-related reduction in C-C chemokine receptor type 7 (CCR7)-dependent egress of immune cells through the lymphatic vasculature mediates some aspects of brain aging and potentially exacerbates cognitive decline and Alzheimer\u27s disease-like brain β-amyloid (Aβ) pathology. We report a reduction in CCR7 expression by meningeal T cells in old mice that is linked to increased effector and regulatory T cells. Hematopoietic CCR7 deficiency mimicked the aging-associated changes in meningeal T cells and led to reduced glymphatic influx and cognitive impairment. Deletion of CCR7 in 5xFAD transgenic mice resulted in deleterious neurovascular and microglial activation, along with increased Aβ deposition in the brain. Treating old mice with anti-CD25 antibodies alleviated the exacerbated meningeal regulatory T cell response and improved cognitive function, highlighting the therapeutic potential of modulating meningeal immunity to fine-tune brain function in aging and in neurodegenerative diseases

    Smoking-induced aggravation of experimental arthritis is dependent of aryl hydrocarbon receptor activation in Th17 cells.

    Get PDF
    Background: Epidemiologic studies have highlighted the association of environmental factors with the development and progression of autoimmune and chronic inflammatory diseases. Among the environmental factors, smoking has been associated with increased susceptibility and poor prognosis in rheumatoid arthritis (RA). However, the immune and molecular mechanism of smoking-induced arthritis aggravation remains unclear. The transcription factor aryl hydrocarbon receptor (AHR) regulates the generation of Th17 cells, CD4 T cells linked the development of autoimmune diseases. AHR is activated by organic compounds including polycyclic aromatic hydrocarbons (PAHs), which are environmental pollutants that are also present in cigarette smoke. In this study, we investigated the role of AHR activation in the aggravation of experiment arthritis induced by exposure to cigarette smoke. Methods: Mice were exposed to cigarette smoke during the developmental phase of antigen-induced arthritis and collagen-induced arthritis to evaluate the effects of smoking on disease development. Aggravation of articular inflammation was assessed by measuring neutrophil migration to the joints, increase in articular hyperalgesia and changes in the frequencies of Th17 cells. In vitro studies were performed to evaluate the direct effects of cigarette smoke and PAH on Th17 differentiation. We also used mice genetically deficient for AHR (Ahr KO) and IL-17Ra (Il17ra KO) to determine the in vivo mechanism of smoking-induced arthritis aggravation. Results: We found that smoking induces arthritis aggravation and increase in the frequencies of Th17 cells. The absence of IL-17 signaling (Il17ra KO) conferred protection to smoking-induced arthritis aggravation. Moreover, in vitro experiments showed that cigarette smoke can directly increase Th17 differentiation of T cells by inducing AHR activation. Indeed, Ahr KO mice were protected from cigarette smoke-induced arthritis aggravation and did not display increase in TH17 frequencies, suggesting that AHR activation is an important mechanism for cigarette smoke effects on arthritis. Finally, we demonstrate that PAHs are also able to induce arthritis aggravation. Conclusions: Our data demonstrate that the disease-exacerbating effects of cigarette smoking are AHR dependent and environmental pollutants with AHR agonist activity can induce arthritis aggravation by directly enhancing Th17 cell development

    Cigarette smoke induces miR-132 in Th17 cells that enhance osteoclastogenesis in inflammatory arthritis

    Get PDF
    Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by joint destruction and severe morbidity. Cigarette smoking (CS) can exacerbate the incidence and severity of RA. Although Th17 cells and the Aryl hydrocarbon receptor (AhR) have been implicated, the mechanism by which CS induces RA development remains unclear. Here, using transcriptomic analysis, we show that microRNA-132 is specifically induced in Th17 cells in the presence of either AhR agonist or CS-enriched medium. miRNA-132 thus induced is packaged into extracellular vesicles produced by Th17 and acts as a proinflammatory mediator increasing osteoclastogenesis through the down-regulation of COX2. In vivo, articular knockdown of miR-132 in murine arthritis models reduces the number of osteoclasts in the joints. Clinically, RA patients express higher levels of miR-132 than do healthy individuals. This increase is further elevated by cigarette smoking. Together, these results reveal a hitherto unrecognized mechanism by which CS could exacerbate RA and further advance understanding of the impact of environmental factors on the pathogenesis of chronic inflammatory diseases

    Erratum to: 36th International Symposium on Intensive Care and Emergency Medicine

    Get PDF
    [This corrects the article DOI: 10.1186/s13054-016-1208-6.]

    Effects of alirocumab on types of myocardial infarction: insights from the ODYSSEY OUTCOMES trial

    Get PDF
    Aims  The third Universal Definition of Myocardial Infarction (MI) Task Force classified MIs into five types: Type 1, spontaneous; Type 2, related to oxygen supply/demand imbalance; Type 3, fatal without ascertainment of cardiac biomarkers; Type 4, related to percutaneous coronary intervention; and Type 5, related to coronary artery bypass surgery. Low-density lipoprotein cholesterol (LDL-C) reduction with statins and proprotein convertase subtilisin–kexin Type 9 (PCSK9) inhibitors reduces risk of MI, but less is known about effects on types of MI. ODYSSEY OUTCOMES compared the PCSK9 inhibitor alirocumab with placebo in 18 924 patients with recent acute coronary syndrome (ACS) and elevated LDL-C (≥1.8 mmol/L) despite intensive statin therapy. In a pre-specified analysis, we assessed the effects of alirocumab on types of MI. Methods and results  Median follow-up was 2.8 years. Myocardial infarction types were prospectively adjudicated and classified. Of 1860 total MIs, 1223 (65.8%) were adjudicated as Type 1, 386 (20.8%) as Type 2, and 244 (13.1%) as Type 4. Few events were Type 3 (n = 2) or Type 5 (n = 5). Alirocumab reduced first MIs [hazard ratio (HR) 0.85, 95% confidence interval (CI) 0.77–0.95; P = 0.003], with reductions in both Type 1 (HR 0.87, 95% CI 0.77–0.99; P = 0.032) and Type 2 (0.77, 0.61–0.97; P = 0.025), but not Type 4 MI. Conclusion  After ACS, alirocumab added to intensive statin therapy favourably impacted on Type 1 and 2 MIs. The data indicate for the first time that a lipid-lowering therapy can attenuate the risk of Type 2 MI. Low-density lipoprotein cholesterol reduction below levels achievable with statins is an effective preventive strategy for both MI types.For complete list of authors see http://dx.doi.org/10.1093/eurheartj/ehz299</p

    Effect of alirocumab on mortality after acute coronary syndromes. An analysis of the ODYSSEY OUTCOMES randomized clinical trial

    Get PDF
    Background: Previous trials of PCSK9 (proprotein convertase subtilisin-kexin type 9) inhibitors demonstrated reductions in major adverse cardiovascular events, but not death. We assessed the effects of alirocumab on death after index acute coronary syndrome. Methods: ODYSSEY OUTCOMES (Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab) was a double-blind, randomized comparison of alirocumab or placebo in 18 924 patients who had an ACS 1 to 12 months previously and elevated atherogenic lipoproteins despite intensive statin therapy. Alirocumab dose was blindly titrated to target achieved low-density lipoprotein cholesterol (LDL-C) between 25 and 50 mg/dL. We examined the effects of treatment on all-cause death and its components, cardiovascular and noncardiovascular death, with log-rank testing. Joint semiparametric models tested associations between nonfatal cardiovascular events and cardiovascular or noncardiovascular death. Results: Median follow-up was 2.8 years. Death occurred in 334 (3.5%) and 392 (4.1%) patients, respectively, in the alirocumab and placebo groups (hazard ratio [HR], 0.85; 95% CI, 0.73 to 0.98; P=0.03, nominal P value). This resulted from nonsignificantly fewer cardiovascular (240 [2.5%] vs 271 [2.9%]; HR, 0.88; 95% CI, 0.74 to 1.05; P=0.15) and noncardiovascular (94 [1.0%] vs 121 [1.3%]; HR, 0.77; 95% CI, 0.59 to 1.01; P=0.06) deaths with alirocumab. In a prespecified analysis of 8242 patients eligible for ≥3 years follow-up, alirocumab reduced death (HR, 0.78; 95% CI, 0.65 to 0.94; P=0.01). Patients with nonfatal cardiovascular events were at increased risk for cardiovascular and noncardiovascular deaths (P<0.0001 for the associations). Alirocumab reduced total nonfatal cardiovascular events (P<0.001) and thereby may have attenuated the number of cardiovascular and noncardiovascular deaths. A post hoc analysis found that, compared to patients with lower LDL-C, patients with baseline LDL-C ≥100 mg/dL (2.59 mmol/L) had a greater absolute risk of death and a larger mortality benefit from alirocumab (HR, 0.71; 95% CI, 0.56 to 0.90; Pinteraction=0.007). In the alirocumab group, all-cause death declined wit h achieved LDL-C at 4 months of treatment, to a level of approximately 30 mg/dL (adjusted P=0.017 for linear trend). Conclusions: Alirocumab added to intensive statin therapy has the potential to reduce death after acute coronary syndrome, particularly if treatment is maintained for ≥3 years, if baseline LDL-C is ≥100 mg/dL, or if achieved LDL-C is low. Clinical Trial Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT01663402

    Global gene expression analysis of mesenchymal stromal cells and hematopoietic stem cells isolated from bone marrow of type 1 diabetes patients

    No full text
    O diabetes mellitus do tipo 1 (T1D) é uma doença autoimune mediada por células T e caracterizada pela destruição seletiva das células ? pancreáticas produtoras de insulina. Células estromais mesenquimais (MSCs) e células tronco hematopoéticas (HSCs) são os principais componentes do nicho hematopoético na medula óssea. Estas células vêm sendo utilizadas nos últimos anos em transplantes autólogos para tratamento do T1D. O objetivo geral do presente trabalho foi avaliar o perfil de expressão gênica global de MSCs e HSCs de pacientes com T1D e compará-lo com células isoladas de indivíduos saudáveis através da técnica de microarray e programas específicos de bioinformática. As MSCs e HSCs foram isoladas da medula óssea de pacientes com T1D antes e após o tratamento com imunossupressão em altas doses seguida pelo transplante autólogo de células tronco hematopoéticas (AHSCT). As MSCs apresentaram valor elevado de expressão absoluta de diversas moléculas potencialmente relacionadas com suas funções de suporte à hematopoese. MSCs de pacientes diabéticos apresentaram perfil de expressão gênica global distinto das isoladas de indivíduos saudáveis, com hiper-regulação da sinalização via proteína G e hiporregulação da atividade transcricional. O receptor ?3 adrenérgico, assim como a sinalização simpática, foram hiper-expressos nas células dos pacientes. Genes que codificam moléculas que suportam a hematopoese e regulados pelo sistema nervoso simpático, VCAM1 e CXCL12, foram hiporregulados em nossa análise. Após o AHSCT, houve atenuação do perfil de expressão diferencial das MSCs dos pacientes, entretanto elas permaneceram com hiperatividade da sinalização via proteína G e déficit da atividade transcricional. As HSCs apresentaram altos níveis de expressão absoluta de diversas integrinas e receptores de citocinas e fatores de crescimento, potencialmente relacionados com funções na hematopoese. HSCs de pacientes com T1D apresentaram perfil de expressão gênica global distinto das de indivíduos saudáveis, com hiper-regulação de genes associados com a atividade transcricional. Os fatores de transcrição TCFL2 e p53, que têm papel fundamental na regulação do ciclo celular das HSCs, foram diferencialmente expressos entre as HSCs de pacientes diabéticos e controles. Assim, nossos resultados de expressão gênica global apontaram alterações intrínsecas nas HSCs e MSCs de pacientes diabéticos que podem estar relacionadas com a falha terapêutica dos transplantes autólogos. A implicação dessas alterações no desenvolvimento e patogênese do T1D permanece desconhecida e a realização de ensaios funcionais poderá esclarecer o significado biológico das mesmas.Type 1 diabetes mellitus (T1D) is a T cell-mediated autoimmune disease, characterized by selective destruction of insulin-producing pancreatic ? cells. Mesenchymal stromal cells (MSCs) and hematopoietic stem cells (HSCs) are the main components of hematopoietic niches. In the last years, these cells are being used in autologous transplantation settings for T1D treatment. The main goal of this study was to evaluate the global gene expression profile of MSCs and HSCs from T1D patients, by using microarrays and bioinformatics specific programs. MSCs and HSCs were isolated from bone marrow of T1D patients before and after treatment with high dose immunossupression followed by hematopoietic stem cell transplantation. MSCs showed high absolute expression values of several molecules potentially related to their function of hematopoiesis support. MSCs from T1D patients exhibited distinct gene expression profile from control MSCs and presented up-regulation of the G protein-coupled receptor signaling pathway and down-regulation of transcriptional activity. The ?3 adrenergic receptor, as well the sympathetic nervous system signaling were up-regulated on patient´s cells. Genes that codify molecules which support hematopoeisis and are regulated by the symphatic nervous system, VCAM1 and CXCL12, were downregulated on our analysis. After AHSCT, there was an attenuation of the differential expression profile of MSCs from T1D patients, however they remained with G proteincoupled receptor signaling pathway hyperactivity and transcriptional activity deficit. HSCs exhibited high absolute expression values of integrins, cytokine receptors and growth factors, molecules potencially related to hematopoietic functions. HSCs from T1D patients showed distinct expression profile from control HSCs and demonstrated up-regulation of genes related to transcriptional activity. The transcription factors TCFL2 and p53, which have important role in regulating HSC cycle, were differentially expressed between HSCs from T1D patients and controls. Thus, our global gene expression analysis has revealed intrinsic alterations on MSCs and HSCs from T1D patients that could be related to the autologous transplant therapeutic failures. The implications of these alterations on the development and pathogenesis of T1D remain unknown and functional assays could unravel their biological meaning

    Global gene expression analysis of mesenchymal stromal cells and hematopoietic stem cells isolated from bone marrow of type 1 diabetes patients

    No full text
    O diabetes mellitus do tipo 1 (T1D) é uma doença autoimune mediada por células T e caracterizada pela destruição seletiva das células ? pancreáticas produtoras de insulina. Células estromais mesenquimais (MSCs) e células tronco hematopoéticas (HSCs) são os principais componentes do nicho hematopoético na medula óssea. Estas células vêm sendo utilizadas nos últimos anos em transplantes autólogos para tratamento do T1D. O objetivo geral do presente trabalho foi avaliar o perfil de expressão gênica global de MSCs e HSCs de pacientes com T1D e compará-lo com células isoladas de indivíduos saudáveis através da técnica de microarray e programas específicos de bioinformática. As MSCs e HSCs foram isoladas da medula óssea de pacientes com T1D antes e após o tratamento com imunossupressão em altas doses seguida pelo transplante autólogo de células tronco hematopoéticas (AHSCT). As MSCs apresentaram valor elevado de expressão absoluta de diversas moléculas potencialmente relacionadas com suas funções de suporte à hematopoese. MSCs de pacientes diabéticos apresentaram perfil de expressão gênica global distinto das isoladas de indivíduos saudáveis, com hiper-regulação da sinalização via proteína G e hiporregulação da atividade transcricional. O receptor ?3 adrenérgico, assim como a sinalização simpática, foram hiper-expressos nas células dos pacientes. Genes que codificam moléculas que suportam a hematopoese e regulados pelo sistema nervoso simpático, VCAM1 e CXCL12, foram hiporregulados em nossa análise. Após o AHSCT, houve atenuação do perfil de expressão diferencial das MSCs dos pacientes, entretanto elas permaneceram com hiperatividade da sinalização via proteína G e déficit da atividade transcricional. As HSCs apresentaram altos níveis de expressão absoluta de diversas integrinas e receptores de citocinas e fatores de crescimento, potencialmente relacionados com funções na hematopoese. HSCs de pacientes com T1D apresentaram perfil de expressão gênica global distinto das de indivíduos saudáveis, com hiper-regulação de genes associados com a atividade transcricional. Os fatores de transcrição TCFL2 e p53, que têm papel fundamental na regulação do ciclo celular das HSCs, foram diferencialmente expressos entre as HSCs de pacientes diabéticos e controles. Assim, nossos resultados de expressão gênica global apontaram alterações intrínsecas nas HSCs e MSCs de pacientes diabéticos que podem estar relacionadas com a falha terapêutica dos transplantes autólogos. A implicação dessas alterações no desenvolvimento e patogênese do T1D permanece desconhecida e a realização de ensaios funcionais poderá esclarecer o significado biológico das mesmas.Type 1 diabetes mellitus (T1D) is a T cell-mediated autoimmune disease, characterized by selective destruction of insulin-producing pancreatic ? cells. Mesenchymal stromal cells (MSCs) and hematopoietic stem cells (HSCs) are the main components of hematopoietic niches. In the last years, these cells are being used in autologous transplantation settings for T1D treatment. The main goal of this study was to evaluate the global gene expression profile of MSCs and HSCs from T1D patients, by using microarrays and bioinformatics specific programs. MSCs and HSCs were isolated from bone marrow of T1D patients before and after treatment with high dose immunossupression followed by hematopoietic stem cell transplantation. MSCs showed high absolute expression values of several molecules potentially related to their function of hematopoiesis support. MSCs from T1D patients exhibited distinct gene expression profile from control MSCs and presented up-regulation of the G protein-coupled receptor signaling pathway and down-regulation of transcriptional activity. The ?3 adrenergic receptor, as well the sympathetic nervous system signaling were up-regulated on patient´s cells. Genes that codify molecules which support hematopoeisis and are regulated by the symphatic nervous system, VCAM1 and CXCL12, were downregulated on our analysis. After AHSCT, there was an attenuation of the differential expression profile of MSCs from T1D patients, however they remained with G proteincoupled receptor signaling pathway hyperactivity and transcriptional activity deficit. HSCs exhibited high absolute expression values of integrins, cytokine receptors and growth factors, molecules potencially related to hematopoietic functions. HSCs from T1D patients showed distinct expression profile from control HSCs and demonstrated up-regulation of genes related to transcriptional activity. The transcription factors TCFL2 and p53, which have important role in regulating HSC cycle, were differentially expressed between HSCs from T1D patients and controls. Thus, our global gene expression analysis has revealed intrinsic alterations on MSCs and HSCs from T1D patients that could be related to the autologous transplant therapeutic failures. The implications of these alterations on the development and pathogenesis of T1D remain unknown and functional assays could unravel their biological meaning

    Citrullinated human fibrinogen triggers arthritis through an inflammatory response mediated by IL-23/IL-17 immune axis

    No full text
    Rheumatoid arthritis (RA) is an autoimmune disease that causes joint destruction. Although its etiology remains unknown, citrullinated proteins have been considered as an auto-antigen able to trigger an inflammatory response in RA. Herein, we modified the classical antigen-induced arthritis (AIA) model by using citrullinated human plasma fibrinogen (hFIB) as an immunogen to investigate the mechanism of inflammation-driven joint damage by citrullinated hFIB in C57BL/6 mice. We found that hFIB-immunized mice showed high serum levels of anti-citrullinated peptides antibodies (ACPAs). Moreover, hFIB immunized mice showed increased mechanical hyperalgesia, massive leukocyte infiltration, high levels of inflammatory mediators, and progressive joint damage after the intra-articular challenge with citrullinated hFIB. Interestingly, hFIB-induced arthritis was dependent on IL-23/IL-17 immune axis-mediated inflammatory responses since leukocyte infiltration and mechanical hyperalgesia were abrogated in Il17ra−/− and Il23a−/− mice. Thus, we have characterized a novel model of experimental arthritis suitable to investigate the contribution of ACPAs and Th17 cell-mediated immune response in the pathogenesis of RA
    corecore