Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Allergy ; 78(10): 2712-2723, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37548395

RESUMEN

BACKGROUND: Dupilumab, a monoclonal anti-IL-4Rα antibody, is approved for several type 2 mediated inflammatory diseases like asthma, atopic dermatitis, and diffuse type 2 chronic rhinosinusitis (CRS). Clinical studies had reported a transient increase in blood eosinophils during dupilumab therapy. This study aimed to assess the impact of elevated blood eosinophils on clinical outcome and to investigate the cause of high blood eosinophil levels under dupilumab therapy. METHODS: Patients suffering from diffuse type 2 CRS treated with dupilumab were examined on days 0, 28, 90, and 180 after therapy start. Sino-Nasal-Outcome-Test Score (SNOT-22), Total Nasal Polyp Score (TNPS), and blood samples were collected. Cytokine measurements and proteomics analysis were conducted. Flow cytometry analysis measured receptor expression on eosinophils. RESULTS: Sixty-eighty patients were included. Baseline eosinophilia ≥0.3G/L was observed in 63.2% of patients, and in 30.9% of patients, eosinophils increased by ≥0.5G/L under dupilumab. Subjects with eosinophilia ≥0.3G/L at baseline had the best SNOT-22 mean change compared to no eosinophilia. Eosinophil elevation during dupilumab therapy had no impact on clinical scores. The eosinophil adhesion molecule VCAM-1 decreased significantly during therapy in all patients. The chemokine receptor CXCR4 was significantly down- and IL-4 upregulated in subjects with eosinophil increase. CONCLUSION: Our findings suggest that increased eosinophils in type 2 CRS are associated with a good clinical response to dupilumab. Patients with elevated IL-4 at baseline developed dupilumab-induced transient eosinophilia. We identified the downregulation of VCAM-1 and surface markers CD49d and CXCR4 on eosinophils as possible explanations of dupilumab-induced eosinophilia.


Asunto(s)
Eosinofilia , Pólipos Nasales , Rinitis , Sinusitis , Humanos , Molécula 1 de Adhesión Celular Vascular/metabolismo , Rinitis/complicaciones , Interleucina-4/metabolismo , Eosinofilia/metabolismo , Sinusitis/complicaciones , Eosinófilos , Enfermedad Crónica , Anticuerpos Monoclonales/uso terapéutico , Pólipos Nasales/complicaciones
2.
Br J Dermatol ; 189(5): 603-611, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37409661

RESUMEN

BACKGROUND: Extracorporeal photopheresis (ECP) has emerged as a systemic first-line immunomodulatory therapy in leukaemic cutaneous T-cell lymphoma (L-CTCL) and is now beginning to be utilized in other T-cell-mediated diseases. Although ECP has been used for nearly 30 years, its mechanisms of action are not sufficiently understood, and biomarkers for response are scarce. OBJECTIVES: We aimed to investigate the immunomodulatory effects of ECP on cytokine secretion patterns in patients with L-CTCL, to help elucidate its mechanism of action. METHODS: A total of 25 patients with L-CTCL and 15 healthy donors (HDs) were enrolled in this retrospective cohort study. Concentrations of 22 cytokines were simultaneously quantified by using multiplex bead-based immunoassays. Neoplastic cells in patients' blood were evaluated by flow cytometry. RESULTS: Firstly, we observed a distinct cytokine profile pattern difference between L-CTCLs and HDs. There was a significant loss of tumour necrosis factor (TNF)-α, and significant increase of interleukins (IL)-9, IL-12 and IL-13 in the sera of patients with L-CTCL compared with HDs. Secondly, patients with L-CTCL who received ECP were classified as treatment responders and nonresponders according to the quantitative reduction of malignant burden in their blood. We evaluated cytokine levels in culture supernatants from patients' peripheral blood mononuclear cells (PBMCs) at baseline and 27 weeks after ECP initiation. Strikingly, PBMCs purified from ECP responders released statistically higher concentrations of innate immune cytokines IL-1α, IL-1ß, granulocyte-macrophage colony-stimulating factor (GM-CSF) and TNF-α in comparison with ECP nonresponders. In parallel, responders showed clearance of erythema, reduction of malignant clonal T cells in the blood, and a potent boost of relevant innate immune cytokines in individual patients with L-CTCL. CONCLUSIONS: Taken together, our results demonstrate that ECP stimulates the innate immune network, and facilitates redirection of the tumour-biased immunosuppressive microenvironment towards proactive antitumour immune responses. The alterations of IL-1α, IL-1ß, GM-CSF and TNF-α can be used as biomarkers of response to ECP in patients with L-CTCL.


Asunto(s)
Linfoma Cutáneo de Células T , Fotoféresis , Neoplasias Cutáneas , Humanos , Citocinas , Fotoféresis/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Factor de Necrosis Tumoral alfa , Estudios Retrospectivos , Leucocitos Mononucleares , Linfoma Cutáneo de Células T/patología , Inmunidad Innata , Neoplasias Cutáneas/terapia , Biomarcadores , Microambiente Tumoral
3.
Allergy ; 77(4): 1274-1284, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34467524

RESUMEN

BACKGROUND: Systemic sclerosis (SSc) is a multiorgan autoimmune disease characterized by inflammation, vascular modification, and progressive fibrosis of the skin and several visceral organs. Innate and adaptive immune cells, including myeloid, B and T cells, are believed to be central to the pathogenesis of SSc. However, the role and functional state of neutrophil granulocytes (neutrophils) are ill-defined in SSc. METHODS: We performed a prospective study of neutrophils freshly isolated from SSc patients and healthy donors (HD) by measuring in these neutrophils (i) functional cell surface markers, including CD16, CD62L, CD66b, CD66c, CXCR1, CXCR2, and CXCR4; (ii) cytokine-activated intracellular signal transducer and activator of transcription (STAT) pathways, such as phosphorylated STAT3 (pSTAT3), pSTAT5, and pSTAT6; (iii) production of neutrophil extracellular traps (NET) and intracellular myeloperoxidase (MPO); and (iv) phagocytosis of bacteria by the neutrophils. RESULTS: Neutrophils of SSc patients expressed lower CD16 and CD62L and higher pSTAT3 and pSTAT6 compared to HD. Moreover, neutrophils of SSc patients lacked CXCR1 and CXCR2, the receptors responding to the potent neutrophil chemoattractant CXCL8. Neutrophils of SSc patients were also deficient in MPO levels, NET formation, and phagocytosis of bacteria. CONCLUSIONS: Neutrophils of patients with SSc display several functional defects affecting cell migration, NET formation, and phagocytosis of bacteria.


Asunto(s)
Trampas Extracelulares , Esclerodermia Sistémica , Humanos , Neutrófilos , Fagocitosis , Estudios Prospectivos , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología
4.
J Allergy Clin Immunol ; 147(2): 545-557.e9, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33221383

RESUMEN

BACKGROUND: Whereas severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific antibody tests are increasingly being used to estimate the prevalence of SARS-CoV-2 infection, the determinants of these antibody responses remain unclear. OBJECTIVES: Our aim was to evaluate systemic and mucosal antibody responses toward SARS-CoV-2 in mild versus severe coronavirus disease 2019 (COVID-19) cases. METHODS: Using immunoassays specific for SARS-CoV-2 spike proteins, we determined SARS-CoV-2-specific IgA and IgG in sera and mucosal fluids of 2 cohorts, including SARS-CoV-2 PCR-positive patients (n = 64) and PCR-positive and PCR-negtive health care workers (n = 109). RESULTS: SARS-CoV-2-specific serum IgA titers in patients with mild COVID-19 were often transiently positive, whereas serum IgG titers remained negative or became positive 12 to 14 days after symptom onset. Conversely, patients with severe COVID-19 showed a highly significant increase of SARS-CoV-2-specific serum IgA and IgG titers after symptom onset. Very high titers of SARS-CoV-2-specific serum IgA were correlated with severe acute respiratory distress syndrome. Interestingly, some health care workers with negative SARS-CoV-2-specific serum antibody titers showed SARS-CoV-2-specific IgA in mucosal fluids with virus-neutralizing capacity in some cases. SARS-CoV-2-specific IgA titers in nasal fluids were inversely correlated with age. CONCLUSIONS: Systemic antibody production against SARS-CoV-2 develops mainly in patients with severe COVID-19, with very high IgA titers seen in patients with severe acute respiratory distress syndrome, whereas mild disease may be associated with transient production of SARS-CoV-2-specific antibodies but may stimulate mucosal SARS-CoV-2-specific IgA secretion.


Asunto(s)
Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Membrana Mucosa/inmunología , SARS-CoV-2/inmunología , Adulto , Anciano , Anticuerpos Antivirales/sangre , COVID-19/sangre , Femenino , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Saliva/inmunología , Índice de Severidad de la Enfermedad , Lágrimas/inmunología
5.
Circulation ; 142(23): 2240-2258, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33070627

RESUMEN

BACKGROUND: Cardiac hypertrophy is a key biological response to injurious stresses such as pressure overload and, when excessive, can lead to heart failure. Innate immune activation by danger signals, through intracellular pattern recognition receptors such as nucleotide-binding oligomerization domain 1 (Nod1) and its adaptor receptor-interacting protein 2 (RIP2), might play a major role in cardiac remodeling and progression to heart failure. We hypothesize that Nod1/RIP2 are major contributors to cardiac hypertrophy, but may not be sufficient to fully express the phenotype alone. METHODS: To elucidate the contribution of Nod1/RIP2 signaling to cardiac hypertrophy, we randomized Nod1-/-, RIP2-/-, or wild-type mice to transverse aortic constriction or sham operations. Cardiac hypertrophy, fibrosis, and cardiac function were examined in these mice. RESULTS: Nod1 and RIP2 proteins were upregulated in the heart after transverse aortic constriction, and this was paralleled by increased expression of mitochondrial proteins, including mitochondrial antiviral signaling protein (MAVS). Nod1-/- and RIP2-/- mice subjected to transverse aortic constriction exhibited better survival, improved cardiac function, and decreased cardiac hypertrophy. Downstream signal transduction pathways that regulate inflammation and fibrosis, including NF (nuclear factor) κB and MAPK (mitogen-activated protein kinase)-GATA4/p300, were reduced in both Nod1-/- and RIP2-/- mice after transverse aortic constriction compared with wild-type mice. Coimmunoprecipitation of extracted cardiac proteins and confocal immunofluorescence microscopy showed that Nod1/RIP2 interaction was robust and that this complex also included MAVS as an essential component. Suppression of MAVS expression attenuated the complex formation, NF κB signaling, and myocyte hypertrophy. Interrogation of mitochondrial function compared in the presence or ablation of MAVS revealed that MAVS serves to suppress mitochondrial energy output and mediate fission/fusion related dynamic changes. The latter is possibly linked to mitophagy during cardiomyocytes stress, which may provide an intriguing link between innate immune activation and mitochondrial energy balance under stress or injury conditions. CONCLUSIONS: We have identified that innate immune Nod1/RIP2 signaling is a major contributor to cardiac remodeling after stress. This process is critically joined by and regulated through the mitochondrial danger signal adapter MAVS. This novel complex coordinates remodeling, inflammatory response, and mitochondrial energy metabolism in stressed cardiomyocytes. Thus, Nod1/RIP2/MAVS signaling complex may represent an attractive new therapeutic approach toward heart failure.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Cardiomegalia/inmunología , Metabolismo Energético/fisiología , Inmunidad Innata/fisiología , Proteína Adaptadora de Señalización NOD1/inmunología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Recién Nacidos , Cardiomegalia/metabolismo , Cardiomegalia/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Transducción de Señal/fisiología
6.
Cancer Immunol Immunother ; 70(7): 1867-1876, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33394095

RESUMEN

INTRODUCTION: Lung cancer is the leading cause of death by cancer. In recent years, immunotherapy with checkpoint inhibitors (ICI) emerged as a promising new therapeutic approach. However, a deeper understanding of the immunologic responses adjacent to the tumor known as tumor microenvironment (TME) is needed. Our study investigated TME of lung cancer by analyzing cytokines in bronchoalveolar lavage fluid (BALF). MATERIALS AND METHODS: Between January 2018 and June 2019, 119 patients were prospectively enrolled in this study. For each cancer patient, levels of 16 cytokines (fractalkine, granulocyte-macrophage colony-stimulating factor (GM-CSF), interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukins (IL): IL-1b, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12p70, IL-13, IL-17A, and IL-23) were measured in BALF and serum and compared to healthy individuals and patients with other lung diseases. RESULTS: There were several significant differences of cytokine levels of patients with lung cancer compared to healthy individuals. However, none of them remained in the multivariate analysis compared to other lung diseases in either BALF or serum. Furthermore, there were no significant differences between the groups in cell differentiation of either BALF or serum. Cytokine levels in BALF were generally near the lower detection limit and showed almost no correlation with their respective levels measured in serum of the same individual. CONCLUSIONS: Cytokines in BALF and serum of lung cancer patients may indicate unspecific inflammation. BAL is not recommendable as a tool to investigate TME of lung cancer. Therefore, cytokines measured in BALF are probably not appropriate as predictors in patients treated with ICIs.


Asunto(s)
Biomarcadores de Tumor/análisis , Líquido del Lavado Bronquioalveolar/química , Citocinas/metabolismo , Neoplasias Pulmonares/patología , Microambiente Tumoral/inmunología , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos
7.
Mol Cell Biochem ; 476(9): 3241-3252, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33881711

RESUMEN

Heart inflammation is one of the major causes of heart damage that leads to dilated cardiomyopathy and often progresses to end-stage heart failure. In the present study, we aimed to assess whether human cardiac cells could release immune mediators upon stimulation of Toll-like receptors (TLRs) and Retinoic acid-inducible gene (RIG)-I-like receptors (RLRs).Commercially available human cardiac fibroblasts and an immortalized human cardiomyocyte cell line were stimulated in vitro with TLR2, TLR3, and TLR4 agonists. In addition, cytosolic RLRs were activated in cardiac cells after transfection of polyinosinic-polycytidylic acid (PolyIC). Upon stimulation of TLR3, TLR4, MDA5, and RIG-I, but not upon stimulation of TLR2, human cardiac fibroblasts produced high amounts of the pro-inflammatory cytokines IL-6 and IL-8. On the contrary, the immortalized human cardiomyocyte cell line was unresponsive to the tested TLRs agonists. Upon RLRs stimulation, cardiac fibroblasts, and to a lesser extent the cardiomyocyte cell line, induced anti-viral IFN-ß expression.These data demonstrate that human cardiac fibroblasts and an immortalized human cardiomyocyte cell line differently respond to various TLRs and RLRs ligands. In particular, human cardiac fibroblasts were able to induce pro-inflammatory and anti-viral cytokines on their own. These aspects will contribute to better understand the immunological function of the different cell populations that make up the cardiac tissue.


Asunto(s)
Citocinas/metabolismo , Inmunidad Innata/inmunología , Mediadores de Inflamación/metabolismo , Miofibroblastos/inmunología , Miofibroblastos/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Receptores Toll-Like/metabolismo , Células Cultivadas , Humanos
8.
Cytokine ; 123: 154768, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31276936

RESUMEN

Measurement of cytokines in peripheral blood and bronchoalveolar lavage fluid (BALF) is a useful method to assess human immune responses in a large range of pulmonary diseases. One of the major pre-analytical challenges of cytokine analysis is the quality and stability of cytokines in the timeframe between sample collection and the separation of supernatant from cells. To evaluate if the method of storage may affect cytokine quantification, whole blood and BALF were collected, aliquoted, and left at room temperature (RT) to be processed at different time points. In addition, sera and BALF were left either at RT or at 4 °C for 24 h after cell separation to test cytokine variations in the absence of cells. Samples were analysed by a multiple array containing ten cytokines. Most of the cytokines analysed (interleukin (IL)-4, IL-5, IL-6, IL-12p70, IL-13, IL-17A, IL-23, interferon (IFN)-γ, and tumour necrosis factor (TNF)-α) did not show significant variations in whole blood and BALF. Levels of IL-8 however, increased after storage of whole blood and BALF for 24 h at RT. Ex vivo IL-8 production seems to correlate with higher numbers of macrophages in collected BALF. These data demonstrate that many cytokines are stable for a brief time after sample collection. For IL-8, freshly collected whole blood and BALF should be quickly processed and frozen to avoid false positive results.


Asunto(s)
Líquido del Lavado Bronquioalveolar , Lavado Broncoalveolar , Citocinas/sangre , Enfermedades Pulmonares/sangre , Preservación Biológica , Femenino , Humanos , Masculino , Factores de Tiempo
9.
Circulation ; 128(14): 1542-54, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24030499

RESUMEN

BACKGROUND: Viral myocarditis follows a fatal course in ≈30% of patients. Interleukin-1 receptor-associated kinase 4 (IRAK4), a major nodal signal transducer in innate immunity, can play a pivotal role in host inflammatory response. We sought to determine how IRAK4 modulates inflammation and outcome in a mouse model of viral myocarditis. METHODS AND RESULTS: Myocarditis was induced after intraperitoneal inoculation of coxsackievirus B3 into C57Bl/6 IRAK4-deficient mice and their littermate controls. Mortality and viral proliferation were markedly reduced in IRAK4(-/-) mice compared with their IRAK4(+/+) littermates. Disease resistance of IRAK4(-/-) mice paralleled increased amounts of protective heart-infiltrating CCR5(+) monocytes/macrophages and enhanced interferon-α and interferon-γ production 2 days after infection. Competitive bone marrow chimera demonstrated that intact IRAK4 function inhibited heart-specific migration of bone marrow-derived CCR5(+) cells. Mechanistically, lack of IRAK4 resulted in interferon regulatory factor 5 homodimerization via reduced melanoma differentiation-associated protein 5 degradation and enhanced Stat1 and Stat5 phosphorylation. Consequently, antiviral interferon-α and interferon-γ production, as well as CCR5(+) cell recruitment, increased, whereas the overall proinflammatory response was drastically reduced in the absence of IRAK4. CONCLUSIONS: Innate immunity signal transducer IRAK4 exacerbates viral myocarditis through inhibition of interferon production and reduced mobilization of protective CCR5(+) monocytes/macrophages to the heart. The combination of IRAK4 inhibitors and antiviral adjuvants may become an attractive therapeutic approach against viral myocarditis in the future.


Asunto(s)
Antígeno CD11b/análisis , Infecciones por Coxsackievirus/inmunología , Interferones/biosíntesis , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Monocitos/fisiología , Miocarditis/inmunología , Receptores CCR5/análisis , Traslado Adoptivo , Animales , Movimiento Celular/fisiología , Quimiocina CCL5/deficiencia , Quimiocina CCL5/fisiología , Infecciones por Coxsackievirus/fisiopatología , Infecciones por Coxsackievirus/virología , ARN Helicasas DEAD-box/metabolismo , Dimerización , Resistencia a la Enfermedad , Enterovirus Humano B/fisiología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/fisiopatología , Factores Reguladores del Interferón/química , Factores Reguladores del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Quinasas Asociadas a Receptores de Interleucina-1/deficiencia , Quinasas Asociadas a Receptores de Interleucina-1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/fisiopatología , Miocarditis/virología , Enfermedades de Inmunodeficiencia Primaria , Procesamiento Proteico-Postraduccional , Quimera por Radiación , Receptores CCR5/deficiencia , Receptores CCR5/fisiología , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT5/metabolismo , Replicación Viral
10.
Basic Res Cardiol ; 109(3): 411, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24763933

RESUMEN

c-Cbl-associated protein (CAP), also called Sorbs1 or ponsin, has been described as an essential adapter protein in the insulin-signalling pathway. Here, we describe for the first time a unique protective role for CAP in viral myocarditis. Mortality and heart failure development were increased in CAP(-/-) mice compared to CAP(+/+) littermates after Coxsackievirus (CVB3) infection. Mechanistically, CAP protected from tissue apoptosis because of reduced CD8(+) T and natural killer cell cytotoxicity. Despite reduced cytotoxic elimination of CVB3-infected cells in CAP(+/+) hearts, however, CAP enhanced interferon regulatory factor 3 (IRF3)-dependent antiviral type I interferon production and decreased viral proliferation in vitro by binding to the cytoplasmic RIG-I-like receptor melanoma differentiation-associated protein 5 (MDA5). Taken together, these findings reveal a novel modulatory role for CAP in the heart as a key protein stabilizing antiviral type I interferon production, while protecting from excessive cytotoxic responses. Our study will help to define future strategies to develop treatments to limit detrimental responses during viral heart inflammation.


Asunto(s)
Apoptosis , Infecciones por Coxsackievirus/prevención & control , Enterovirus Humano B/inmunología , Interferón Tipo I/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocarditis/prevención & control , Miocardio/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Células Cultivadas , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/virología , ARN Helicasas DEAD-box/metabolismo , Modelos Animales de Enfermedad , Enterovirus Humano B/genética , Enterovirus Humano B/crecimiento & desarrollo , Enterovirus Humano B/patogenicidad , Factor 3 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/virología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Miocarditis/genética , Miocarditis/inmunología , Miocarditis/metabolismo , Miocarditis/patología , Miocarditis/virología , Miocardio/inmunología , Miocardio/patología , Factores de Tiempo , Replicación Viral
11.
J Cancer Res Clin Oncol ; 148(7): 1711-1720, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34347128

RESUMEN

PURPOSE: Immune-checkpoint inhibitors (ICI) present a new treatment for malignancies by boosting the immune system. This has led to a variety of immune-related adverse events, including ICI-associated pneumonitis (ICIaP). Diagnosis thereof is often challenging, and its pathogenesis has not yet been fully understood. The aim of this cross-sectional case-control study was to investigate cytokines in serum and bronchoalveolar lavage fluid (BALF) expressed in patients with ICIaP compared to controls consisting of healthy individuals, patients with lung cancer and patients with interstitial lung diseases (ILD) other than ICIaP. METHODS: From January 2018 until June 2019, 401 adult patients with various lung diseases were prospectively enrolled in a BALF- and serum biobank, called BALOTHEK. Of these, 12 patients were diagnosed with ICIaP (Pembrolizumab, Ipilimumab, or both, and Durvalumab) serving as case group. Subjects with one of three diagnosis groups from BALOTHEK, including lung cancer, ILD other than ICIaP, and healthy individuals, served as matched controls. The following 11 cytokines were simultaneously analyzed in BALF and serum of each study participant: interferon gamma, tumor necrosis factor alpha, interleukin (IL) 1b, IL-2, IL-4, IL-5, IL-6, IL-8, IL-12p70, IL-13 and IL-17A. This study was approved by the local ethic review committee (BASEC-ID 2017-02,307 and 2018-01,724). RESULTS: Absolute number and percentage of lymphocytes in BALF of patients with ICIaP were significantly higher compared to control groups. For the investigated cytokines in BALF, a significant increase of IL-6 level was shown for patients with ICIaP compared to control groups (p = 0.031, adjusted for multiple comparisons). CONCLUSION: Cytokine profile assessed in BALF shows promising potential for facilitating diagnosis and understanding of pathophysiology of ICIaP. IL-6 may not only contribute to better understanding of pathophysiology but also herald therapeutic implications for Tocilizumab.


Asunto(s)
Enfermedades Pulmonares Intersticiales , Neoplasias Pulmonares , Neumonía , Adulto , Líquido del Lavado Bronquioalveolar , Estudios de Casos y Controles , Estudios Transversales , Citocinas , Humanos , Inhibidores de Puntos de Control Inmunológico , Interleucina-6 , Enfermedades Pulmonares Intersticiales/inducido químicamente , Neoplasias Pulmonares/tratamiento farmacológico
12.
Circ Res ; 105(5): 462-70, 2009 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-19628793

RESUMEN

RATIONALE: Myocardial fibrosis is a hallmark of inflammation-triggered end-stage heart disease, a common cause of heart failure in young patients. OBJECTIVE: We used CD4(+) T-cell-mediated experimental autoimmune myocarditis model to determine the parameters regulating cardiac fibrosis in inflammatory heart disease. METHODS AND RESULTS: alpha-Myosin heavy chain peptide/complete Freund's adjuvant immunization was used to induce experimental autoimmune myocarditis in BALB/c mice. Chimeric mice, reconstituted with enhanced green fluorescence protein (EGFP)(+) bone marrow, were used to track the fate of inflammatory cells. Prominin-1(+) cells were isolated from the inflamed hearts, cultured in vitro and injected intracardially at different stages of experimental autoimmune myocarditis. Transforming growth factor (TGF)-beta-mediated fibrosis was addressed using anti-TGF-beta antibody treatment. Myocarditis peaked 21 days after immunization and numbers of cardiac fibroblasts progressively increased on follow-up. In chimeric mice, >60% of cardiac fibroblasts were EGFP(+) 46 days after immunization. At day 21, cardiac infiltrates contained approximately 30% of prominin-1(+) progenitors. In vitro and in vivo experiments confirmed that prominin-1(+) but not prominin-1(-) cells isolated from acutely inflamed hearts represented the cellular source of cardiac fibroblasts at late stages of disease, characterized by increased TGF-beta levels within the myocardium. Mechanistically, the in vitro differentiation of heart-infiltrating prominin-1(+) cells into fibroblasts depended on TGF-beta-mediated phosphorylation of Smad proteins. Accordingly, anti-TGF-beta antibody treatment prevented myocardial fibrosis in immunized mice. CONCLUSIONS: Taken together, heart-infiltrating prominin-1(+) progenitors are the major source of subsequent TGF-beta-triggered cardiac fibrosis in experimental autoimmune myocarditis. Recognizing the critical, cytokine-dependent role of bone marrow-derived progenitors in cardiac remodeling might result in novel treatment concepts against inflammatory heart failure.


Asunto(s)
Antígenos CD/metabolismo , Enfermedades Autoinmunes/metabolismo , Glicoproteínas/metabolismo , Miocarditis/metabolismo , Miocardio/metabolismo , Péptidos/metabolismo , Transducción de Señal , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Antígeno AC133 , Animales , Anticuerpos/administración & dosificación , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/prevención & control , Linfocitos T CD4-Positivos/inmunología , Linaje de la Célula , Transdiferenciación Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis , Adyuvante de Freund , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Antígenos Comunes de Leucocito/análisis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocarditis/inmunología , Miocarditis/patología , Miocarditis/prevención & control , Miocardio/inmunología , Miocardio/patología , Cadenas Pesadas de Miosina , Fosforilación , Proteínas Smad/metabolismo , Trasplante de Células Madre , Células Madre/inmunología , Células Madre/patología , Factores de Tiempo , Factor de Crecimiento Transformador beta/inmunología
13.
Circ Res ; 105(9): 912-20, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19762681

RESUMEN

RATIONALE: The myeloid differentiation factor (MyD)88/interleukin (IL)-1 axis activates self-antigen-presenting cells and promotes autoreactive CD4(+) T-cell expansion in experimental autoimmune myocarditis, a mouse model of inflammatory heart disease. OBJECTIVE: The aim of this study was to determine the role of MyD88 and IL-1 in the progression of acute myocarditis to an end-stage heart failure. METHODS AND RESULTS: Using alpha-myosin heavy chain peptide (MyHC-alpha)-loaded, activated dendritic cells, we induced myocarditis in wild-type and MyD88(-/-) mice with similar distributions of heart-infiltrating cell subsets and comparable CD4(+) T-cell responses. Injection of complete Freund's adjuvant (CFA) or MyHC-alpha/CFA into diseased mice promoted cardiac fibrosis, induced ventricular dilation, and impaired heart function in wild-type but not in MyD88(-/-) mice. Experiments with chimeric mice confirmed the bone marrow origin of the fibroblasts replacing inflammatory infiltrates and showed that MyD88 and IL-1 receptor type I signaling on bone marrow-derived cells was critical for development of cardiac fibrosis during progression to heart failure. CONCLUSIONS: Our findings indicate a critical role of MyD88/IL-1 signaling in the bone marrow compartment in postinflammatory cardiac fibrosis and heart failure and point to novel therapeutic strategies against inflammatory cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/inmunología , Insuficiencia Cardíaca/inmunología , Interleucina-1beta/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Miocarditis/inmunología , Miocardio/inmunología , Transducción de Señal , Animales , Autoinmunidad , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibroblastos/inmunología , Fibrosis , Adyuvante de Freund , Proteínas Fluorescentes Verdes/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Inmunidad Innata , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Miocarditis/complicaciones , Miocarditis/patología , Miocarditis/fisiopatología , Miocardio/patología , Cadenas Pesadas de Miosina/inmunología , Fenotipo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Quimera por Trasplante
14.
Sci Rep ; 11(1): 20961, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702968

RESUMEN

Cardioprotective effect of prostaglandin-E2 receptor-4 (EP4) stimulation on the ischemic heart has been demonstrated. Its effect on the heart affected by myocarditis, however, remains uncertain. In this study, we investigated therapeutic effect of EP4 stimulant using a mouse model of autoimmune myocarditis (EAM) that progresses to dilated cardiomyopathy (DCM). EP4 was present in the hearts of EAM mice. Treatment with EP4 agonist (ONO-0260164: 20 mg/kg/day) improved an impaired left ventricular (LV) contractility and reduction of blood pressure on day 21, a peak myocardial inflammation. Alternatively, DCM phenotype, characterized by LV dilation, LV systolic dysfunction, and collagen deposition, was observed on day 56, along with activation of matrix metalloproteinase (MMP)-2 critical for myocardial extracellular matrix disruption, indicating an important molecular mechanism underlying adverse ventricular remodeling after myocarditis. Continued treatment with ONO-0260164 alleviated the DCM phenotype, but this effect was counteracted by its combination with a EP4 antagonist. Moreover, ONO-0260164 inhibited in vivo proteolytic activity of MMP-2 in association with up-regulation of tissue inhibitor of metalloproteinase (TIMP)-3. EP4 stimulant may be a promising and novel therapeutic agent that rescues cardiac malfunction during myocarditis and prevents adverse ventricular remodeling after myocarditis by promoting the TIMP-3/MMP-2 axis.


Asunto(s)
Miocarditis/tratamiento farmacológico , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiomiopatía Dilatada/tratamiento farmacológico , Modelos Animales de Enfermedad , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Miocarditis/inmunología , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
15.
Am J Respir Crit Care Med ; 179(10): 939-49, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19234103

RESUMEN

RATIONALE: The mouse model of bleomycin-induced lung injury offers an approach to study idiopathic pulmonary fibrosis, a progressive interstitial lung disease with poor prognosis. Progenitor cell-based treatment strategies might combine antiinflammatory effects and the capacity for tissue repair. OBJECTIVES: To expand progenitor cells with reparative and regenerative capacities and to evaluate their protective effects on pulmonary fibrosis in vivo. METHODS: Prominin-1/CD133(+) epithelial progenitor cells (PEPs) were expanded from adult mouse lungs after digestion and culture of distal airways. Lung fibrosis was induced in C57Bl/6 mice by instillation of bleomycin. Two hours later, animals were transplanted with PEPs. Inflammation and fibrosis were assessed by immunohistochemistry, bronchoalveolar lavage fluid differentials, and real-time polymerase chain reaction. MEASUREMENTS AND MAIN RESULTS: PEPs expanded from mouse lungs were of bone marrow origin, coexpressed stem and hematopoietic cell markers, and differentiated in vitro into alveolar type II surfactant protein-C(+) epithelial cells. In bleomycin-challenged mice, intratracheally injected PEPs engrafted into the lungs and differentiated into type II pneumocytes. Furthermore, PEPs suppressed proinflammatory and profibrotic gene expression, prevented the recruitment of inflammatory cells, and protected bleomycin-challenged mice from pulmonary fibrosis. Mechanistically, the protective effect depended on upregulation of inducible nitric oxide synthase in PEPs and nitric oxide-mediated suppression of alveolar macrophage proliferation. Accordingly, PEPs from iNOS(-/-) but not iNOS(+/+) mice failed to protect from bleomycin-induced lung injury. CONCLUSIONS: The combined antiinflammatory and regenerative capacity of bone marrow-derived pulmonary epithelial progenitors offers a promising approach for development of cell-based therapeutic strategies against pulmonary fibrosis.


Asunto(s)
Antígenos CD/biosíntesis , Glicoproteínas/biosíntesis , Fibrosis Pulmonar Idiopática/terapia , Trasplante de Células Madre/métodos , Antígeno AC133 , Animales , Bleomicina/administración & dosificación , Líquido del Lavado Bronquioalveolar/química , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/inmunología , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/inmunología , Fibrosis Pulmonar Idiopática/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/biosíntesis , Óxido Nítrico/inmunología , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/metabolismo , Péptidos , Alveolos Pulmonares/citología , Alveolos Pulmonares/inmunología , Regeneración , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Células Madre/inmunología
16.
J Pharm Biomed Anal ; 179: 113010, 2020 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-31816469

RESUMEN

Measurement of Tumour Necrosis Factor alpha (TNF-α) in peripheral blood is a useful tool to assess inflammatory responses in a large range of diseases. One of the major challenges for cytokine analysis is the availability of a proper analytical tool with high specificity, accuracy, linearity, precision, stability, and analytical sensitivity. Although available immunoassays are usually robust and reproducible, it is also true that they are not interchangeable. Two ELISA, four flow cytometric bead array (CBA) and four Luminex immunoassays were compared. Correlation between different techniques was almost absent, while some immunoassays based on the same technique showed significant correlation. Among the ten different assays evaluated, just few of them complied with the pre-established acceptance validation criteria. Interestingly, sera and plasma collected from the same healthy donor had significant different reference values. Samples stability was maintained in serum up to one week at four degrees, while plasma was stable only when it was frozen. Since several anti-inflammatory treatments are based on biologics targeting TNF-α (anti-TNF-α antibodies), potential interference with the immunoassays was tested and resulted relevant. This study shows that although each immunoassay presents benefits and drawbacks, just few assays are suitable for the measurement of TNF-α in clinical laboratories, demonstrating that, so far, the measurement of TNF-α in human blood is still not yet harmonised. In addition, we found that false negative results caused by anti-TNF-α treatments should be carefully considered for results interpretation.


Asunto(s)
Inflamación/diagnóstico , Factor de Necrosis Tumoral alfa/sangre , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Células HEK293 , Humanos , Inmunoensayo/métodos , Inflamación/sangre , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
17.
Int Immunopharmacol ; 87: 106822, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32738595

RESUMEN

C-Cbl-associated protein (CAP), also known as Sorbin and SH3 domain-containing protein 1 (Sorbs1) or ponsin, an adaptor protein of the insulin-signalling pathway, mediates anti-viral and anti-cytotoxic protection in acute viral heart disease. In the present study we describe a novel protective immuno-modulatory function of CAP in inflammation. Among the three members of the Sorbs family of adapter molecules, which include CAP (Sorbs1), ArgBP2 (Sorbs2), and Vinexin (Sorbs3), CAP consistently down-regulated the expression of pro-inflammatory cytokines in mouse fibroblasts, cardiomyocytes, and myeloid-derived leukocytes, after Toll-like receptor (TLR) stimulation. Upon the same TLR stimulation, ArgBP2 partially down-regulated pro-inflammatory cytokine production in mouse fibroblasts and cardiomyocytes, while Vinexin rather promoted their production. Mechanistically, CAP limited pro-inflammatory cytokine expression by suppressing the phosphorylation of Inhibitor of kappa B (IκB) kinase (Iκκ)-α and Iκκ-ß and their downstream NF-κB-dependent signalling pathway. Molecular affinity between CAP and Iκκ-α/ Iκκ-ß was necessary to block the NF-κB pathway. The CAP-dependent inhibitory mechanism - in vivo exclusively IL-6 inhibition - was confirmed after collecting blood from mice with systemic inflammation induced by lipopolysaccharide (LPS) and in the heart tissue collected from mice infected with the cardiotropic Coxsackievirus B3 (CVB3). Taken together, CAP down-regulates pro-inflammatory cytokines by interfering with the normal function of the NF-κB pathway. The promotion of CAP production could support the development of new strategies aiming to limit excessive and detrimental activation of the immune system.


Asunto(s)
Citocinas/inmunología , FN-kappa B/inmunología , Proteínas Proto-Oncogénicas c-cbl/inmunología , Animales , Línea Celular , Infecciones por Coxsackievirus/inmunología , Enterovirus Humano B , Fibroblastos , Humanos , Leucocitos , Ratones Noqueados , Miocitos Cardíacos , Proteínas Proto-Oncogénicas c-cbl/genética , Transducción de Señal
18.
Cardiovasc Res ; 80(2): 236-45, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18621802

RESUMEN

AIMS: Experimental autoimmune myocarditis (EAM) is a CD4(+) T cell-mediated mouse model of inflammatory heart disease. Tissue-resident bone marrow-derived cells adopt different cellular phenotypes depending on the local milieu. We expanded a specific population of bone marrow-derived prominin-1-expressing progenitor cells (PPC) from healthy heart tissue, analysed their plasticity, and evaluated their capacity to protect mice from EAM and heart failure. METHODS AND RESULTS: PPC were expanded from healthy mouse hearts. Analysis of CD45.1/CD45.2 chimera mice confirmed bone marrow origin of PPC. Depending on in vitro culture conditions, PPC differentiated into macrophages, dendritic cells, or cardiomyocyte-like cells. In vivo, PPC acquired a cardiac phenotype after direct injection into healthy hearts. Intravenous injection of PPC into myosin alpha heavy chain/complete Freund's adjuvant (MyHC-alpha/CFA)-immunized BALB/c mice resulted in heart-specific homing and differentiation into the macrophage phenotype. Histology revealed reduced severity scores for PPC-treated mice compared with control animals [treated with phosphate-buffered saline (PBS) or crude bone marrow at day 21 after MyHC-alpha/CFA immunization]. Echocardiography showed preserved fractional shortening and velocity of circumferential shortening in PPC but not PBS-treated MyHC-alpha/CFA-immunized mice. In vitro and in vivo data suggested that interferon-gamma signalling on PPC was critical for nitric oxide-mediated suppression of heart-specific CD4(+) T cells. Accordingly, PPC from interferon-gamma receptor-deficient mice failed to protect MyHC-alpha/CFA-immunized mice from EAM. CONCLUSION: Prominin-1-expressing, heart-resident, bone marrow-derived cells combine high plasticity, T cell-suppressing capacity, and anti-inflammatory in vivo effects.


Asunto(s)
Antígenos CD/metabolismo , Enfermedades Autoinmunes/prevención & control , Glicoproteínas/metabolismo , Miocarditis/prevención & control , Miocardio/inmunología , Péptidos/metabolismo , Trasplante de Células Madre , Células Madre/inmunología , Antígeno AC133 , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Adyuvante de Freund , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Contracción Miocárdica , Miocarditis/inmunología , Miocarditis/patología , Miocarditis/fisiopatología , Miocardio/patología , Cadenas Pesadas de Miosina , Fenotipo , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptor de Interferón gamma
19.
Curr Pharm Des ; 22(4): 440-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26696258

RESUMEN

Myocarditis, which is commonly known as heart inflammation, is a multifaceted disease that includes at least three phases. The host's immune system is mostly active during the first viral and the second autoimmune phase, when several inflammatory pathways are activated. One of the pivotal transcription factors that regulate immune responses is the nuclear factor kappa B (NF-κB). If, on one side, the acute response to heart injury activates the production of inflammatory cytokines to protect and limit host damage, on the other side sustained and long-term inflammation is one of the leading causes of cardiac hypertrophy and chronic heart failure. An update on the current knowledge of inhibitors and treatments that limit excessive inflammation in experimental and viral autoimmune myocarditis, and therapeutic approaches to cure patients with myocarditis, are described and discussed in this review.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Miocarditis/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Virosis/tratamiento farmacológico , Adiponectina/administración & dosificación , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Ácidos Grasos Monoinsaturados/administración & dosificación , Fluvastatina , Humanos , Indoles/administración & dosificación , Miocarditis/inmunología , Miocarditis/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Resultado del Tratamiento , Virosis/inmunología , Virosis/metabolismo
20.
Mol Immunol ; 74: 47-58, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27148820

RESUMEN

Interferon (IFN)-γ is highly upregulated during heart inflammation and enhances the production of pro-inflammatory cytokines. Absent in Melanoma 2 (AIM2) is an IFN-inducible protein implicated as a component of the inflammasome. Here we seek to determine the role of AIM2 during inflammation in cardiac cells. We found that the presence of AIM2, but not of the other inflammasome components Nod-like receptor (NLR) NLRP3 or NLRC4, specifically limited the transcription of the pro-inflammatory cytokines interleukin (IL)-6, IP-10, and tumor necrosis factor (TNF)-α in HL-1 mouse cardiomyocytes stimulated with IFN-γ and lipopolysaccharides (LPS). Similarly, AIM2 reduced pro-inflammatory cytokine transcription in primary mouse neonatal cardiomyocytes (MNC), but not in primary mouse neonatal cardiac fibroblasts (MNF). Interestingly, AIM2-dependent reduction of pro-inflammatory cytokines in cardiomyocytes was independent of Caspase-1. Mechanistically, AIM2 reduced pro-inflammatory cytokine transcription in cardiomyocytes by interacting with and inhibiting the phosphorylation of STAT1. In AIM2-depleted cardiomyocytes, increased STAT1 phosphorylation enhanced the NF-κB pathway by promoting NF-κB p65 phosphorylation and acetylation. These results show for the first time that AIM2 plays an important anti-inflammatory, yet inflammasome-independent function in cardiomyocytes. Our findings will help to further understand how the various heart cell types differently react to inflammatory stimuli.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Citocinas/biosíntesis , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Inmunoprecipitación , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Fosforilación , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA