Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 28(1): 172-189.e7, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31269438

RESUMEN

Two types of monocytes, Ly6Chi and Ly6Clo, infiltrate the heart in murine experimental autoimmune myocarditis (EAM). We discovered a role for cardiac fibroblasts in facilitating monocyte-to-macrophage differentiation of both Ly6Chi and Ly6Clo cells, allowing these macrophages to perform divergent functions in myocarditis progression. During the acute phase of EAM, IL-17A is highly abundant. It signals through cardiac fibroblasts to attenuate efferocytosis of Ly6Chi monocyte-derived macrophages (MDMs) and simultaneously prevents Ly6Clo monocyte-to-macrophage differentiation. We demonstrated an inverse clinical correlation between heart IL-17A levels and efferocytic receptor expressions in humans with heart failure (HF). In the absence of IL-17A signaling, Ly6Chi MDMs act as robust phagocytes and are less pro-inflammatory, whereas Ly6Clo monocytes resume their differentiation into MHCII+ macrophages. We propose that MHCII+Ly6Clo MDMs are associated with the reduction of cardiac fibrosis and prevention of the myocarditis sequalae.


Asunto(s)
Antígenos Ly/metabolismo , Diferenciación Celular/efectos de los fármacos , Fibroblastos/metabolismo , Interleucina-17/farmacología , Macrófagos/metabolismo , Monocitos/metabolismo , Miocarditis/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/ultraestructura , Humanos , Inflamación/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Macrófagos/citología , Macrófagos/ultraestructura , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Microscopía Electrónica de Transmisión , Monocitos/citología , Miocarditis/inducido químicamente , Miocarditis/patología , Miocardio/citología , Miocardio/patología , Parabiosis , Transducción de Señal , Transcriptoma/genética , Tirosina Quinasa c-Mer/metabolismo
2.
Exp Mol Pathol ; 103(2): 141-152, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28822770

RESUMEN

The extensive, diverse communities that constitute the microbiome are increasingly appreciated as important regulators of human health and disease through inflammatory, immune, and metabolic pathways. We sought to elucidate pathways by which microbiota contribute to inflammatory, autoimmune cardiac disease. We employed an animal model of experimental autoimmune myocarditis (EAM), which results in inflammatory and autoimmune pathophysiology and subsequent maladaptive cardiac remodeling and heart failure. Antibiotic dysbiosis protected mice from EAM and fibrotic cardiac dysfunction. Additionally, mice derived from different sources with different microbiome colonization profiles demonstrated variable susceptibility to disease. Unexpectedly, it did not track with segmented filamentous bacteria (SFB)-driven Th17 programming of CD4+ T cells in the steady-state gut. Instead, we found disease susceptibility to track with presence of type 3 innate lymphoid cells (ILC3s). Ablating ILCs by antibody depletion or genetic tools in adoptive transfer variants of the EAM model demonstrated that ILCs and microbiome profiles contributed to the induction of CCL20/CCR6-mediated inflammatory chemotaxis to the diseased heart. From these data, we conclude that sensing of the microbiome by ILCs is an important checkpoint in the development of inflammatory cardiac disease processes through their ability to elicit cardiotropic chemotaxis.


Asunto(s)
Antibacterianos/farmacología , Enfermedades Autoinmunes/inmunología , Corazón/fisiopatología , Linfocitos/inmunología , Microbiota , Miocarditis/inmunología , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/metabolismo , Modelos Animales de Enfermedad , Disbiosis/prevención & control , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Miocarditis/tratamiento farmacológico , Miocarditis/metabolismo
3.
Immun Inflamm Dis ; 5(2): 163-176, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28474508

RESUMEN

INTRODUCTION: Complete Freund's Adjuvant (CFA) emulsified with an antigen is a widely used method to induce autoimmune disease in animal models, yet the contribution of CFA to the immune response is not well understood. We compared the effectiveness of CFA with Incomplete Freund's Adjuvant (IFA) or TiterMax Gold Adjuvant (TMax) in experimental autoimmune myocarditis (EAM) in male mice. METHODS: EAM was induced in A/J, BALB/c, and IL6KO BALB/c male mice by injection of the myocarditogenic peptide in CFA, IFA, or TMax on days 0 and 7. EAM severity was analyzed by histology on day 21. In addition, specific flow cytometry outcomes were evaluated on day 21. RESULTS: Only mice immunized with CFA and myocarditogenic peptide on both days 0 and 7 developed substantial myocarditis as measured by histology. We observed a significantly increased level of IL6 in the spleen 3 days after CFA immunization. In the spleen and heart on day 21, there was an expansion of myeloid cells in CFA-immunized mice, as compared to IFA or TMax-immunized animals. Recombinant IL-6 at the time of IFA immunization partially restored susceptibility of the mice to EAM. We also treated EAM-resistant IL-6 knockout mice with recombinant IL-6 around the time of the first immunization, on days -1 to 2, completely restoring disease susceptibility, showing that the requirement for IL-6 coincides with primary immunization. Examining APC populations in the lymph node draining the immunization site evidenced the contribution of IL-6 to the CFA-dependence of EAM was through controlling local dendritic cell (DC) trafficking. CONCLUSIONS: CFA used with myocarditogenic peptide twice is required to induce EAM in both A/J and Balb/c mice. Although IFA and TiterMax induce antibody responses, only CFA preferentially induced autoantigen-specific responses. CFA expands monocytes in the heart and in the spleen. IL-6 signaling is required during short window around primary immunization to induce EAM. In addition, IL-6 deficient mice resistance to EAM could be reversed by injecting IL-6 around first immunization. IL-6 expands dendritic cell and monocytic populations and ultimately leads to a robust T-cell driven immune response in CFA immunized mice.


Asunto(s)
Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/inmunología , Adyuvante de Freund/efectos adversos , Interleucina-6/inmunología , Miocarditis/inducido químicamente , Miocarditis/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Adyuvante de Freund/farmacología , Interleucina-6/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Miocarditis/genética , Miocarditis/patología
4.
J Exp Med ; 214(4): 943-957, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28302646

RESUMEN

Inflammatory dilated cardiomyopathy (DCMi) is a major cause of heart failure in children and young adults. DCMi develops in up to 30% of myocarditis patients, but the mechanisms involved in disease progression are poorly understood. Patients with eosinophilia frequently develop cardiomyopathies. In this study, we used the experimental autoimmune myocarditis (EAM) model to determine the role of eosinophils in myocarditis and DCMi. Eosinophils were dispensable for myocarditis induction but were required for progression to DCMi. Eosinophil-deficient ΔdblGATA1 mice, in contrast to WT mice, showed no signs of heart failure by echocardiography. Induction of EAM in hypereosinophilic IL-5Tg mice resulted in eosinophilic myocarditis with severe ventricular and atrial inflammation, which progressed to severe DCMi. This was not a direct effect of IL-5, as IL-5TgΔdblGATA1 mice were protected from DCMi, whereas IL-5-/- mice exhibited DCMi comparable with WT mice. Eosinophils drove progression to DCMi through their production of IL-4. Our experiments showed eosinophils were the major IL-4-expressing cell type in the heart during EAM, IL-4-/- mice were protected from DCMi like ΔdblGATA1 mice, and eosinophil-specific IL-4 deletion resulted in improved heart function. In conclusion, eosinophils drive progression of myocarditis to DCMi, cause severe DCMi when present in large numbers, and mediate this process through IL-4.


Asunto(s)
Cardiomiopatía Dilatada/etiología , Eosinófilos/fisiología , Interleucina-4/fisiología , Miocarditis/complicaciones , Animales , Enfermedades Autoinmunes/complicaciones , Progresión de la Enfermedad , Fibrosis , Humanos , Interferón gamma/fisiología , Interleucina-13/fisiología , Interleucina-17/fisiología , Interleucina-5/fisiología , Ratones , Ratones Endogámicos BALB C
5.
Eur J Immunol ; 46(12): 2749-2760, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27621211

RESUMEN

Cardiac manifestations are a major cause of morbidity and mortality in patients with eosinophil-associated diseases. Eosinophils are thought to play a pathogenic role in myocarditis. We investigated the pathways that recruit eosinophils to the heart using a model of eosinophilic myocarditis, in which experimental autoimmune myocarditis (EAM) is induced in IFNγ-/- IL-17A-/- mice. Two conditions are necessary for efficient eosinophil trafficking to the heart: high eotaxin (CCL11, CCL24) expression in the heart and expression of the eotaxin receptor CCR3 by eosinophils. We identified cardiac fibroblasts as the source of CCL11 in the heart interstitium. CCL24 is produced by F4/80+ macrophages localized at inflammatory foci in the heart. Expression of CCL11 and CCL24 is controlled by Th2 cytokines, IL-4 and IL-13. To determine the relevance of this pathway in humans, we analyzed endomyocardial biopsy samples from myocarditis patients. Expression of CCL11 and CCL26 was significantly increased in eosinophilic myocarditis compared to chronic lymphocytic myocarditis and positively correlated with the number of eosinophils. Thus, eosinophil trafficking to the heart is dependent on the eotaxin-CCR3 pathway in a mouse model of EAM and associated with cardiac eotaxin expression in patients with eosinophilic myocarditis. Blocking this pathway may prevent eosinophil-mediated cardiac damage.


Asunto(s)
Quimiocina CCL11/metabolismo , Quimiocina CCL24/metabolismo , Eosinófilos/inmunología , Fibroblastos/inmunología , Macrófagos/inmunología , Miocarditis/inmunología , Miocardio/inmunología , Enfermedad Autoinmune Experimental del Sistema Nervioso/inmunología , Adulto , Anciano , Animales , Miosinas Cardíacas/inmunología , Movimiento Celular , Células Cultivadas , Femenino , Humanos , Interferón gamma/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Miocardio/patología , Receptores CCR3/genética , Balance Th1 - Th2
6.
J Autoimmun ; 75: 20-29, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27545842

RESUMEN

In the broad field of autoimmunity and clinical immunology, experimental evidence over the past few years have demonstrated several connections between the immune system and the nervous system, both central and peripheral, leading to the definition of neuroimmunology and of an immune-brain axis. Indeed, the central nervous system as an immune-privileged site, thanks to the blood-brain barrier, is no longer a dogma as the barrier may be altered during chronic inflammation with disruptive changes of endothelial cells and tight junctions, largely mediated by adenosine receptors and the expression of CD39/CD73. The diseases that encompass the neuroimmunology field vary from primary nervous diseases such as multiple sclerosis to systemic conditions with neuropsychiatric complications, such as systemic lupus erythematosus or vasculitidies. Despite potentially similar clinical manifestations, the pathogenesis of each condition is different, but the interaction between the ultra-specialized structure that is the nervous system and inflammation mediators are crucial. Two examples come from anti-dsDNA cross-reacting with anti-N-Methyl-d-Aspartate receptor (NMDAR) antibodies in neuropsychiatric lupus or the new family of antibody-associated neuronal autoimmune diseases including classic paraneoplastic syndromes with antibodies directed to intracellular antigens (Hu, Yo, Ri) and autoimmune encephalitis. In the case of multiple sclerosis, the T cell paradigm is now complicated by the growing evidence of a B cell involvement, particularly via aquaporin antibodies, and their influence on Th1 and Th17 lineages. Inspired by a productive AARDA-sponsored colloquium among experts we provide a critical review of the literature on the pathogenesis of different immune-mediated diseases with neurologic manifestations and we discuss the basic immunology of the central nervous system and the interaction between immune cells and the peripheral nervous system.


Asunto(s)
Autoinmunidad/inmunología , Encéfalo/inmunología , Sistema Nervioso Central/inmunología , Sistema Inmunológico/inmunología , Sistema Nervioso Periférico/inmunología , Autoanticuerpos/inmunología , Encefalitis/inmunología , Enfermedad de Hashimoto/inmunología , Humanos , Lupus Eritematoso Sistémico/inmunología , Esclerosis Múltiple/inmunología
7.
Am J Pathol ; 186(9): 2337-52, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27470712

RESUMEN

Infections with Staphylococcus aureus are a continuing and growing problem in community and hospital settings. Preclinical animal modeling of S. aureus relies on experimental infection, which carries some limitations. We describe here a novel, spontaneous model of oral staphylococcal infection in double knockout mice, deficient in the receptors for IL-17 (IL-17RA) and interferon (IFN)-γ (IFNγRI), beginning at 6 to 8 weeks of age. IFNγRI(-/-)IL17RA(-/-) (GRAKO) mice developed progressive oral abscesses. Cytometric methods revealed extensive neutrophilic infiltration of oral tissues in GRAKO mice; further investigation evidenced that IL-17 predominated neutrophil defects in these mice. To investigate the contribution of IFN-γ signaling to this native host defense to S. aureus, we observed perturbations of monocyte recruitment and macrophage differentiation in the oral tissues of GRAKO mice, and CXCL9/chemokine ligand receptor (CXCR)3-driven recruitment of T-cell oral tissues and draining lymph nodes. To address the former finding, we depleted macrophages and monocytes in vivo from IL17RA(-/-) mice using liposomes loaded with clodronate. This treatment elicited oral abscesses, recapitulating the phenotype of GRAKO mice. From these findings, we propose novel collaborative functions of IL-17 and IFN-γ, acting through neutrophils and macrophages, respectively, in native mucocutaneous host defenses to S. aureus.


Asunto(s)
Interferón gamma/inmunología , Interleucina-17/inmunología , Mucosa Bucal/inmunología , Mucosa Bucal/microbiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal/inmunología
8.
Eur J Immunol ; 46(3): 582-92, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26660726

RESUMEN

Using a mouse model of experimental autoimmune myocarditis (EAM), we showed for the first time that IL-23 stimulation of CD4(+) T cells is required only briefly at the initiation of GM-CFS-dependent cardiac autoimmunity. IL-23 signal, acting as a switch, turns on pathogenicity of CD4(+) T cells, and becomes dispensable once autoreactivity is established. Il23a(-/-) mice failed to mount an efficient Th17 response to immunization, and were protected from myocarditis. However, remarkably, transient IL-23 stimulation ex vivo fully restored pathogenicity in otherwise nonpathogenic CD4(+) T cells raised from Il23a(-/-) donors. Thus, IL-23 may no longer be necessary to uphold inflammation in established autoimmune diseases. In addition, we demonstrated that IL-23-induced GM-CSF mediates the pathogenicity of CD4(+) T cells in EAM. The neutralization of GM-CSF abrogated cardiac inflammation. However, sustained IL-23 signaling is required to maintain IL-17A production in CD4(+) T cells. Despite inducing inflammation in Il23a(-/-) recipients comparable to wild-type (WT), autoreactive CD4(+) T cells downregulated IL-17A production without persistent IL-23 signaling. This divergence on the controls of GM-CSF-dependent pathogenicity on one side and IL-17A production on the other side may contribute to the discrepant efficacies of anti-IL-23 therapy in different autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Interleucina-23/metabolismo , Miocarditis/inmunología , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Femenino , Interleucina-17/biosíntesis , Interleucina-17/genética , Interleucina-23/deficiencia , Interleucina-23/genética , Interleucina-23/farmacología , Ratones , Miocarditis/fisiopatología , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Células Th17/inmunología
9.
Clin Immunol ; 159(2): 183-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26003843

RESUMEN

Although the mechanisms by which the human microbiome influences the onset and progression of autoimmune diseases remain to be determined, established animal models of autoimmune diseases indicate that local and systemic bidirectional interactions with the microbiome play a signaling or promoting role through the immune system. Whether alterations in the microbiome are a pathogenic cause or simply an effect of inflammation and autoimmune disease remains an essential question to be addressed in disease-specific research, as well as whether particular conditions of the microbiome promote health or promote disease. Future research in this area needs to account for sex differences in microbiome composition because autoimmune diseases disproportionately affect women. Probiotic and other treatments that manipulate assemblage of the microbiome may offer methods of preventing or mitigating the effects of autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Dieta , Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa/inmunología , Animales , Enfermedades Autoinmunes/microbiología , Epigénesis Genética , Femenino , Microbioma Gastrointestinal/genética , Humanos , Masculino , Ratones , Microbiota/genética , Microbiota/inmunología , Factores Sexuales
10.
Am J Pathol ; 185(3): 847-61, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25622543

RESUMEN

Myocarditis is a leading cause of sudden cardiac failure in young adults. Natural killer (NK) cells, a subset of the innate lymphoid cell compartment, are protective in viral myocarditis. Herein, we demonstrated that these protective qualities extend to suppressing autoimmune inflammation. Experimental autoimmune myocarditis (EAM) was initiated in BALB/c mice by immunization with myocarditogenic peptide. During EAM, activated cardiac NK cells secreted interferon γ, perforin, and granzyme B, and expressed CD69, tumor necrosis factor-related apoptosis-inducing ligand treatment, and CD27 on their cell surfaces. The depletion of NK cells during EAM with anti-asialo GM1 antibody significantly increased myocarditis severity, and was accompanied by elevated fibrosis and a 10-fold increase in the percentage of cardiac-infiltrating eosinophils. The resultant influx of eosinophils to the heart was directly responsible for the increased disease severity in the absence of NK cells, because treatment with polyclonal antibody asialogangloside GM-1 did not augment myocarditis severity in eosinophil-deficient ΔdoubleGATA1 mice. We demonstrate that NK cells limit eosinophilic infiltration both indirectly, through altering eosinophil-related chemokine production by cardiac fibroblasts, and directly, by inducing eosinophil apoptosis in vitro. Altogether, we define a new pathway of eosinophilic regulation through interactions with NK cells.


Asunto(s)
Eosinófilos/inmunología , Células Asesinas Naturales/inmunología , Miocarditis/inmunología , Miocardio/inmunología , Animales , Apoptosis/inmunología , Eosinófilos/patología , Fibrosis/inmunología , Fibrosis/patología , Inflamación/patología , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos BALB C , Miocarditis/patología , Miocardio/patología
11.
J Exp Med ; 211(7): 1449-64, 2014 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-24935258

RESUMEN

Inflammatory dilated cardiomyopathy (DCMi) is a major cause of heart failure in individuals below the age of 40. We recently reported that IL-17A is required for the development of DCMi. We show a novel pathway connecting IL-17A, cardiac fibroblasts (CFs), GM-CSF, and heart-infiltrating myeloid cells with the pathogenesis of DCMi. Il17ra(-/-) mice were protected from DCMi, and this was associated with significantly diminished neutrophil and Ly6Chi monocyte/macrophage (MO/MΦ) cardiac infiltrates. Depletion of Ly6Chi MO/MΦ also protected mice from DCMi. Mechanistically, IL-17A stimulated CFs to produce key chemokines and cytokines that are critical downstream effectors in the recruitment and differentiation of myeloid cells. Moreover, IL-17A directs Ly6Chi MO/MΦ in trans toward a more proinflammatory phenotype via CF-derived GM-CSF. Collectively, this IL-17A-fibroblast-GM-CSF-MO/MΦ axis could provide a novel target for the treatment of DCMi and related inflammatory cardiac diseases.


Asunto(s)
Cardiomiopatía Dilatada/inmunología , Fibroblastos/inmunología , Interleucina-17/inmunología , Miocardio/inmunología , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Fibroblastos/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Interleucina-17/genética , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Miocardio/patología , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/patología
12.
J Immunol ; 191(8): 4038-47, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24048893

RESUMEN

CD4(+) T cells play a central role in inflammatory heart disease, implicating a cytokine product associated with Th cell effector function as a necessary mediator of this pathophysiology. IFN-γ-deficient mice developed severe experimental autoimmune myocarditis (EAM), in which mice are immunized with cardiac myosin peptide, whereas IL-17A-deficient mice were protected from progression to dilated cardiomyopathy. We generated IFN-γ(-/-)IL-17A(-/-) mice to assess whether IL-17 signaling was responsible for the severe EAM of IFN-γ(-/-) mice. Surprisingly, IFN-γ(-/-)IL-17A(-/-) mice developed a rapidly fatal EAM. Eosinophils constituted a third of infiltrating leukocytes, qualifying this disease as eosinophilic myocarditis. We found increased cardiac production of CCL11/eotaxin, as well as Th2 deviation, among heart-infiltrating CD4(+) cells. Ablation of eosinophil development improved survival of IFN-γ(-/-)IL-17A(-/-) mice, demonstrating the necessity of eosinophils in fatal heart failure. The severe and rapidly fatal autoimmune inflammation that developed in the combined absence of IFN-γ and IL-17A constitutes a novel model of eosinophilic heart disease in humans. This is also, to our knowledge, the first demonstration that eosinophils have the capacity to act as necessary mediators of morbidity in an autoimmune process.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Eosinófilos/inmunología , Interferón gamma/deficiencia , Interleucina-17/deficiencia , Miocarditis/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/prevención & control , Biomarcadores , Miosinas Cardíacas/inmunología , Cardiomiopatías/inmunología , Quimiocina CCL11/biosíntesis , Inflamación , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Miocarditis/genética , Miocarditis/prevención & control , Miocardio/inmunología , Miositis
13.
Ann N Y Acad Sci ; 1285: 80-96, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23692566

RESUMEN

This review focuses on autoimmune myocarditis and its sequela, inflammatory dilated cardiomyopathy (DCMI), and the inflammatory and immune mechanisms underlying the pathogenesis of these diseases. Several mouse models of myocarditis and DCMI have improved our knowledge of the pathogenesis of these diseases, informing more general problems of cardiac remodeling and heart failure. CD4(+) T cells are critical in driving the pathogenesis of myocarditis. We discuss in detail the role of T helper cell subtypes in the pathogenesis of myocarditis, the biology of T cell-derived effector cytokines, and the participation of other leukocytic effectors in mediating disease pathophysiology. We discuss interactions between these subsets in both suppressive and collaborative fashions. These findings indicate that cardiac inflammatory disease, and autoimmunity in general, may be more diverse in divergent effector mechanisms than has previously been appreciated.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Miocarditis/inmunología , Miocarditis/patología , Animales , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/patología , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/patología , Ratones
14.
Circ Heart Fail ; 5(3): 366-75, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22454393

RESUMEN

BACKGROUND: IL-33 through its receptor ST2 protects the heart from myocardial infarct and hypertrophy in animal models but, paradoxically, increases autoimmune disease. In this study, we examined the effect of IL-33 or ST2 administration on autoimmune heart disease. METHODS AND RESULTS: We used pressure-volume relationships and isoproterenol challenge to assess the effect of recombinant (r) IL-33 or rST2 (eg, soluble ST2) administration on the development of autoimmune coxsackievirus B3 myocarditis and dilated cardiomyopathy in male BALB/c mice. The rIL-33 treatment significantly increased acute perimyocarditis (P=0.006) and eosinophilia (P=1.3×10(-5)), impaired cardiac function (maximum ventricular power, P=0.0002), and increased ventricular dilation (end-diastolic volume, P=0.01). The rST2 treatment prevented eosinophilia and improved heart function compared with rIL-33 treatment (ejection fraction, P=0.009). Neither treatment altered viral replication. The rIL-33 treatment increased IL-4, IL-33, IL-1ß, and IL-6 levels in the heart during acute myocarditis. To determine whether IL-33 altered cardiac function on its own, we administered rIL-33 to undiseased mice and found that rIL-33 induced eosinophilic pericarditis and adversely affected heart function. We used cytokine knockout mice to determine that this effect was due to IL-33-mediated signaling but not to IL-1ß or IL-6. CONCLUSIONS: We show for the first time to our knowledge that IL-33 induces eosinophilic pericarditis, whereas soluble ST2 prevents eosinophilia and improves systolic function, and that IL-33 independently adversely affects heart function through the IL-33 receptor.


Asunto(s)
Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/prevención & control , Eosinofilia/etiología , Corazón/fisiopatología , Interleucinas/efectos adversos , Pericarditis/etiología , Receptores de Interleucina/uso terapéutico , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/prevención & control , Enfermedades Autoinmunes/virología , Cardiomiopatía Dilatada/metabolismo , Infecciones por Coxsackievirus/complicaciones , Modelos Animales de Enfermedad , Eosinofilia/prevención & control , Eosinofilia/virología , Corazón/efectos de los fármacos , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-33 , Interleucina-6/deficiencia , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucinas/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Pericarditis/prevención & control , Pericarditis/virología , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico
15.
Immunobiology ; 217(5): 468-75, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21820754

RESUMEN

Cardiac inflammatory disease represents a significant public health burden, and interesting questions of immunopathologic science and clinical inquiry. Novel insights into the diverse programming and functions within the macrophage lineages in recent years have yielded a view of these cells as dynamic effectors and regulators of immunity, host defense, and inflammatory disease. In this review, we examine and discuss recent investigations into the complex participation of mononuclear phagocytic cells in the pathology of animal models of myocarditis.


Asunto(s)
Macrófagos/inmunología , Miocarditis/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Humanos , Monocitos/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología
16.
Eur J Immunol ; 42(3): 726-36, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22161142

RESUMEN

The involvement of macrophages (MΦs) in Th17-cell responses is still poorly understood. While neutrophils are thought to be the predominant effector of Th17-cell responses, IL-17 is also known to induce myelotropic chemokines and growth factors. Other T-cell-derived cytokines induce non-classical functions, suggesting that IL-17 sigxnaling may similarly elicit unique MΦ functions. Here, we characterized the expression of subunits of the IL-17 receptor on primary murine MΦs from different anatomical compartments. The greatest expression of IL-17 receptors was observed on mucosal Ly6C(hi) "inflammatory" MΦs. We further observed upregulation of IL-17 receptors in vitro on bone marrow-derived macrophages (BMMΦs) in response to peptidoglycan or CpG oligonucleotide stimuli, and in vivo, upon CFA administration. Macrophages expressing IL-17 receptors were observed infiltrating the hearts of mice with myocarditis, and genetic ablation of IL-17RA altered MΦ recruitment. Treating primary MΦs from a wide variety of different anatomic sources (as well as cell lines) with IL-17A induced the production of unique profiles of cytokines and chemokines, including GM-CSF, IL-3, IL-9, CCL4/MIP-1ß and CCL5/RANTES. IL-17A also induced production of IL-12p70; IL-17-signaling-deficient MΦs elicited diminished IFN-γ production by responding DO11.10 CD4(+) T cells when used as APCs. These data indicate that MΦs from different anatomic locations direct IL-17-mediated responses.


Asunto(s)
Interleucina-17/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Miocarditis/inmunología , Animales , Quimiocinas/inmunología , Femenino , Citometría de Flujo , Inflamación/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores de Interleucina-17/inmunología , Organismos Libres de Patógenos Específicos , Regulación hacia Arriba/inmunología
17.
Exp Mol Pathol ; 89(2): 83-91, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20599938

RESUMEN

A protective effect of interferon-gamma (IFNγ) has been described in a number of models of autoimmune disease, including experimental autoimmune myocarditis (EAM). Some reports have suggested that regulation of apoptosis in autoreactive lymphocytes mediate these protective functions. We examined the potential of IFNγ to regulate apoptotic mechanisms in detail, both in vitro and in vivo in EAM. We observed multiple apoptotic defects in caspase activity, and the expression of TNF superfamily members on CD4(+) T cells. In addition, we observed selective defects in CD4(+) T cell activation in response to antigenic stimulation. These activation and apoptotic defects were CD4(+) cell autonomous, independent of the genotype of APCs. Inhibition of nitric oxide production in vivo did not reproduce the severe form of EAM of IFNγ-deficient mice, indicating that this pathway does not mediate the protective effect of IFNγ. Crosswise adoptive transfer of wild type, IFNγ(-/-), and IFNγR(-/-)EAM demonstrated that IFNγ signaling was critical in CD4(+) cells, but that non-CD4(+) sources of IFNγ production were also involved in the control of disease. Together, these data indicate multiple mechanisms of autonomous and non-autonomous CD4(+) T cell regulation mediated by IFNγ in the control of autoimmune heart disease.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/fisiología , Interferón gamma/fisiología , Miocarditis/genética , Miocarditis/inmunología , Animales , Apoptosis , Enfermedades Autoinmunes/genética , Femenino , Interferón gamma/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Recombinantes , Organismos Libres de Patógenos Específicos
18.
Circ Res ; 106(10): 1646-55, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20378858

RESUMEN

RATIONALE: One-third of myocarditis cases progresses to dilated cardiomyopathy (DCM), but the mechanisms controlling this process are largely unknown. CD4(+) T helper (Th)17 cells have been implicated in the pathogenesis of autoimmune diseases, but the role of Th17-produced cytokines during inflammation-induced cardiac remodeling has not been previously studied. OBJECTIVE: We examined the importance of interleukin (IL)-17A in the progression of myocarditis to DCM using a mouse model. METHODS AND RESULTS: Immunization of mice with myocarditogenic peptide in complete Freund's adjuvant induced the infiltration of IL-17A-producing Th17 cells into the inflamed heart. Unexpectedly, IL-17A-deficient mice developed myocarditis with similar incidence and severity compared to wild-type mice. Additionally, IL-17A deficiency did not ameliorate the severe myocarditis of interferon (IFN)gamma-deficient mice, suggesting that IL-17A plays a minimal role during acute myocarditis. In contrast, IL-17A-deficient mice were protected from postmyocarditis remodeling and did not develop DCM. Flow cytometric and cytokine analysis revealed an important role for IL-17A in heart-specific upregulation of IL-6, TNFalpha, and IL-1beta and the recruitment of CD11b(+) monocyte and Gr1(+) granulocyte populations into the heart. Furthermore, IL-17A-deficient mice had reduced interstitial myocardial fibrosis, downregulated expression of matrix metalloproteinase-2 and -9 and decreased gelatinase activity. Treatment of BALB/c mice with anti-IL-17A monoclonal antibody administered after the onset of myocarditis abrogated myocarditis-induced cardiac fibrosis and preserved ventricular function. CONCLUSIONS: Our findings reveal a critical role for IL-17A in postmyocarditis cardiac remodeling and the progression to DCM. Targeting IL-17A may be an attractive therapy for patients with inflammatory dilated cardiomyopathy.


Asunto(s)
Cardiomiopatías/fisiopatología , Interleucina-17/fisiología , Miocarditis/fisiopatología , Animales , Antígenos CD/inmunología , Linfocitos T CD4-Positivos/inmunología , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Citometría de Flujo , Inmunización , Inflamación/fisiopatología , Interleucina-17/deficiencia , Interleucina-17/genética , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Función Ventricular Izquierda/fisiología , Remodelación Ventricular/fisiología
19.
Ann N Y Acad Sci ; 1173: 378-83, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19758176

RESUMEN

Sex differences in a NOD.H2(h4) murine model of Sjögren's syndrome were analyzed. Compared to males, female NOD.H2(h4) mice have increased severity of sialoadenitis and have a significantly increased percentage of CD4(+) T cells in salivary gland infiltrates. CD4(+) T cells in female infiltrates produce more Th2 and Th17 cytokines than in males, while males have greater Th1 responses. Females also have enhanced B cell responses, with higher levels of SSA and SSB serum antibodies, and B cell activation factor F (BAFF). Thus, sex has a strong impact on the severity of murine Sjögren's syndrome by affecting the immune mechanisms driving the autoimmune inflammation.


Asunto(s)
Citocinas/metabolismo , Síndrome de Sjögren/metabolismo , Síndrome de Sjögren/patología , Animales , Antígeno CD11b/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Interleucina-9/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos NOD , Glándulas Salivales/metabolismo , Glándulas Salivales/patología , Factores Sexuales , Sialadenitis/metabolismo , Sialadenitis/patología , Factor de Necrosis Tumoral alfa/metabolismo
20.
Int Immunopharmacol ; 8(5): 624-33, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18387504

RESUMEN

We evaluated the efficacy of the Ligand Epitope Antigen Presentation System (L.E.A.P.S.trade mark) in preventing or treating experimental autoimmune myocarditis (EAM) in A/J mice. L.E.A.P.S. (here, J-My-1) is a conjugate of the myocarditogenic peptide of cardiac myosin MyHCalpha(334-352) (My-1) and J peptide, derived from the sequence of human beta-2 microglobulin. Remarkably, early prophylactic (J-My-1 injected on days -14 and -7 before EAM induction), late prophylactic (J-My-1 injected on days 0, 7, 14, and 21), and therapeutic (J-My-1 injected on days 7, 14, and 21 or 10, 17 and 24) administration of J-My-1 significantly decreased the incidence and severity of EAM. However, extended therapeutic treatment was associated with anaphylaxis and death, corresponding with global immune activation associated with J-My-1 treatment. In J-My1-treated animals, we observed expanded numbers of activated CD69+ and CD44+ CD4+ and CD8+ T cells in the spleens. J-My-1 treatment also increased the proportion of CD11c+ dendritic cells in spleens and induced strong production of anti-J-My-1 specific antibodies. J-My-1 injections resulted in decreased levels of chemokines MIP-1alpha and IP-10 in hearts. We propose that J-My-1 treatment interferes with trafficking of autoaggressive immune cells to the heart.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Enfermedades Autoinmunes/tratamiento farmacológico , Epítopos/efectos de los fármacos , Cadenas J de Inmunoglobulina/farmacología , Miocarditis/tratamiento farmacológico , Miocardio/patología , Animales , Enfermedades Autoinmunes/patología , Proliferación Celular/efectos de los fármacos , Quimiocina CCL3/biosíntesis , Quimiocina CXCL10/biosíntesis , Quimiocinas/biosíntesis , Anergia Clonal/efectos de los fármacos , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Liberación de Histamina/efectos de los fármacos , Ligandos , Ratones , Ratones Endogámicos A , Miocarditis/patología , Miocardio/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Células TH1/efectos de los fármacos , Células Th2/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...