Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Ther Nucleic Acids ; 27: 797-809, 2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35116191

RESUMEN

Since Paul Ehrlich's introduction of the "magic bullet" concept in 1908, drug developers have been seeking new ways to target drug activity to diseased cells while limiting effects on normal tissues. In recent years, it has been proposed that coupling riboswitches capable of detecting RNA biomarkers to small interfering RNAs (siRNAs) to create siRNA pro-drugs could selectively activate RNA interference (RNAi) activity in specific cells. However, this concept has not been achieved previously. We report here that we have accomplished this goal, validating a simple and programmable new design that functions reliably in mammalian cells. We show that these conditionally activated siRNAs (Cond-siRNAs) can switch RNAi activity against different targets between clearly distinguished OFF and ON states in response to different cellular RNA biomarkers. Notably, in a rat cardiomyocyte cell line (H9C2), one version of our construct demonstrated biologically meaningful inhibition of a heart-disease-related target gene protein phosphatase 3 catalytic subunit alpha (PPP3CA) in response to increased expression of the pathological marker atrial natriuretic peptide (NPPA) messenger RNA (mRNA). Our results demonstrate the ability of synthetic riboswitches to regulate gene expression in mammalian cells, opening a new path for development of programmable siRNA pro-drugs.

2.
Life Sci Alliance ; 4(12)2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34663679

RESUMEN

Extracellular vesicles (EVs) mediate intercellular signaling by transferring their cargo to recipient cells, but the functional consequences of signaling are not fully appreciated. RBC-derived EVs are abundant in circulation and have been implicated in regulating immune responses. Here, we use a transgenic mouse model for fluorescence-based mapping of RBC-EV recipient cells to assess the role of this intercellular signaling mechanism in heart disease. Using fluorescent-based mapping, we detected an increase in RBC-EV-targeted cardiomyocytes in a murine model of ischemic heart failure. Single cell nuclear RNA sequencing of the heart revealed a complex landscape of cardiac cells targeted by RBC-EVs, with enrichment of genes implicated in cell proliferation and stress signaling pathways compared with non-targeted cells. Correspondingly, cardiomyocytes targeted by RBC-EVs more frequently express cellular markers of DNA synthesis, suggesting the functional significance of EV-mediated signaling. In conclusion, our mouse model for mapping of EV-recipient cells reveals a complex cellular network of RBC-EV-mediated intercellular communication in ischemic heart failure and suggests a functional role for this mode of intercellular signaling.


Asunto(s)
Eritrocitos/metabolismo , Vesículas Extracelulares/metabolismo , Insuficiencia Cardíaca/sangre , Infarto del Miocardio/sangre , Miocardio/metabolismo , ARN Nuclear/genética , RNA-Seq/métodos , Transducción de Señal/genética , Análisis de la Célula Individual/métodos , Animales , Comunicación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Voluntarios Sanos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/metabolismo
3.
Circ Heart Fail ; 14(2): e007279, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33517669

RESUMEN

BACKGROUND: Mineralocorticoid receptor (MR) antagonists decrease heart failure (HF) hospitalization and mortality, but the mechanisms are unknown. Preclinical studies reveal that the benefits on cardiac remodeling and dysfunction are not completely explained by inhibition of MR in cardiomyocytes, fibroblasts, or endothelial cells. The role of MR in smooth muscle cells (SMCs) in HF has never been explored. METHODS: Male mice with inducible deletion of MR from SMCs (SMC-MR-knockout) and their MR-intact littermates were exposed to HF induced by 27-gauge transverse aortic constriction versus sham surgery. HF phenotypes and mechanisms were measured 4 weeks later using cardiac ultrasound, intracardiac pressure measurements, exercise testing, histology, cardiac gene expression, and leukocyte flow cytometry. RESULTS: Deletion of MR from SMC attenuated transverse aortic constriction-induced HF with statistically significant improvements in ejection fraction, cardiac stiffness, chamber dimensions, intracardiac pressure, pulmonary edema, and exercise capacity. Mechanistically, SMC-MR-knockout protected from adverse cardiac remodeling as evidenced by decreased cardiomyocyte hypertrophy and fetal gene expression, interstitial and perivascular fibrosis, and inflammatory and fibrotic gene expression. Exposure to pressure overload resulted in a statistically significant decline in cardiac capillary density and coronary flow reserve in MR-intact mice. These vascular parameters were improved in SMC-MR-knockout mice compared with MR-intact littermates exposed to transverse aortic constriction. CONCLUSIONS: These results provide a novel paradigm by which MR inhibition may be beneficial in HF by blocking MR in SMC, thereby improving cardiac blood supply in the setting of pressure overload-induced hypertrophy, which in turn mitigates the adverse cardiac remodeling that contributes to HF progression and symptoms.


Asunto(s)
Insuficiencia Cardíaca/genética , Miocitos del Músculo Liso/metabolismo , Receptores de Mineralocorticoides/genética , Remodelación Ventricular/genética , Animales , Aorta/cirugía , Presión Arterial , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Constricción Patológica , Modelos Animales de Enfermedad , Ecocardiografía , Técnicas de Inactivación de Genes , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Ratones , Músculo Liso Vascular/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología
4.
Circulation ; 143(12): 1242-1255, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33463362

RESUMEN

BACKGROUND: Despite the well-established association between T-cell-mediated inflammation and nonischemic heart failure, the specific mechanisms triggering T-cell activation during the progression of heart failure and the antigens involved are poorly understood. We hypothesized that myocardial oxidative stress induces the formation of isolevuglandin (IsoLG)-modified proteins that function as cardiac neoantigens to elicit CD4+ T-cell receptor (TCR) activation and promote heart failure. METHODS: We used transverse aortic constriction in mice to trigger myocardial oxidative stress and T-cell infiltration. We profiled the TCR repertoire by mRNA sequencing of intramyocardial activated CD4+ T cells in Nur77GFP reporter mice, which transiently express GFP on TCR engagement. We assessed the role of antigen presentation and TCR specificity in the development of cardiac dysfunction using antigen presentation-deficient MhcII-/- mice and TCR transgenic OTII mice that lack specificity for endogenous antigens. We detected IsoLG protein adducts in failing human hearts. We also evaluated the role of reactive oxygen species and IsoLGs in eliciting T-cell immune responses in vivo by treating mice with the antioxidant TEMPOL and the IsoLG scavenger 2-hydroxybenzylamine during transverse aortic constriction, and ex vivo in mechanistic studies of CD4+ T-cell proliferation in response to IsoLG-modified cardiac proteins. RESULTS: We discovered that TCR antigen recognition increases in the left ventricle as cardiac dysfunction progresses and identified a limited repertoire of activated CD4+ T-cell clonotypes in the left ventricle. Antigen presentation of endogenous antigens was required to develop cardiac dysfunction because MhcII-/- mice reconstituted with CD4+ T cells and OTII mice immunized with their cognate antigen were protected from transverse aortic constriction-induced cardiac dysfunction despite the presence of left ventricle-infiltrated CD4+ T cells. Scavenging IsoLGs with 2-hydroxybenzylamine reduced TCR activation and prevented cardiac dysfunction. Mechanistically, cardiac pressure overload resulted in reactive oxygen species-dependent dendritic cell accumulation of IsoLG protein adducts, which induced robust CD4+ T-cell proliferation. CONCLUSIONS: Our study demonstrates an important role of reactive oxygen species-induced formation of IsoLG-modified cardiac neoantigens that lead to TCR-dependent CD4+ T-cell activation within the heart.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Cardiopatías/complicaciones , Lípidos/efectos adversos , Animales , Humanos , Lípidos/farmacología , Ratones
5.
Circ Res ; 128(1): e1-e23, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33092465

RESUMEN

RATIONALE: Previous translational studies implicate plasma extracellular microRNA-30d (miR-30d) as a biomarker in left ventricular remodeling and clinical outcome in heart failure (HF) patients, although precise mechanisms remain obscure. OBJECTIVE: To investigate the mechanism of miR-30d-mediated cardioprotection in HF. METHODS AND RESULTS: In rat and mouse models of ischemic HF, we show that miR-30d gain of function (genetic, lentivirus, or agomiR-mediated) improves cardiac function, decreases myocardial fibrosis, and attenuates cardiomyocyte (CM) apoptosis. Genetic or locked nucleic acid-based knock-down of miR-30d expression potentiates pathological left ventricular remodeling, with increased dysfunction, fibrosis, and cardiomyocyte death. RNA sequencing of in vitro miR-30d gain and loss of function, together with bioinformatic prediction and experimental validation in cardiac myocytes and fibroblasts, were used to identify and validate direct targets of miR-30d. miR-30d expression is selectively enriched in cardiomyocytes, induced by hypoxic stress and is acutely protective, targeting MAP4K4 (mitogen-associate protein kinase 4) to ameliorate apoptosis. Moreover, miR-30d is secreted primarily in extracellular vesicles by cardiomyocytes and inhibits fibroblast proliferation and activation by directly targeting integrin α5 in the acute phase via paracrine signaling to cardiac fibroblasts. In the chronic phase of ischemic remodeling, lower expression of miR-30d in the heart and plasma extracellular vesicles is associated with adverse remodeling in rodent models and human subjects and is linked to whole-blood expression of genes implicated in fibrosis and inflammation, consistent with observations in model systems. CONCLUSIONS: These findings provide the mechanistic underpinning for the cardioprotective association of miR-30d in human HF. More broadly, our findings support an emerging paradigm involving intercellular communication of extracellular vesicle-contained miRNAs (microRNAs) to transregulate distinct signaling pathways across cell types. Functionally validated RNA biomarkers and their signaling networks may warrant further investigation as novel therapeutic targets in HF.


Asunto(s)
MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Comunicación Paracrina , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Apoptosis , Células Cultivadas , Modelos Animales de Enfermedad , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas Sprague-Dawley , Ratas Transgénicas , Transducción de Señal , Quinasa de Factor Nuclear kappa B
6.
Curr Atheroscler Rep ; 22(10): 57, 2020 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-32772195

RESUMEN

PURPOSE OF REVIEW: Atherosclerosis (AS) is a chronic inflammatory disease that contributes to the development of coronary artery disease, which has become a leading health burden worldwide. Though several strategies such as pharmacological treatment, exercise intervention, and surgery have been used in clinical practice, there is still no effective strategy to cure AS. Exosomes are extensively studied both as diagnostic markers as well as for therapeutic purposes due to their role in pathological processes related to AS. To elucidate the role of exosomes in AS and thus provide a new insight into AS therapy, we review recent advances concerning exosome targets and their function in mediating intercellular communication in AS, and expect to provide a reference for novel effective strategies to cure AS. RECENT FINDINGS: Exosomes exert important roles in the diagnosis, development, and potential therapy of AS. For AS development, (1) activation of CD-137 in endothelial cells represses exosomal-TET2 production, causing a phenotypic switch of vascular smooth muscle cells (VSMC) and promoting plaque formation; (2) exosomal-MALTA1 derived from endothelial cells causes neutrophil extracellular traps (NETs) and M2 macrophage polarization, which aggravates AS; and (3) exosomal-miR-21-3p derived from macrophages inhibits PTEN expression and further promotes VSMC migration/proliferation, leading to AS development. For AS diagnosis, plasma exosomal-miR30e and miR-92a are considered to be potential diagnostic markers. For AS therapy, adipose mesenchymal stem cell-derived exosomes protect endothelial cells from AS aggravation, via inhibiting miR-342-5p. Exosome-mediated cross-talk between different cells within the vasculature exerts crucial roles in regulating endothelial function, proliferation and differentiation of vascular smooth muscle cells, and platelet activation as well as macrophage activation, collectively leading to the development and progression of AS. Exosomes can potentially be used as diagnostic biomarkers and constitute as a new therapeutic strategy for AS.


Asunto(s)
Aterosclerosis/metabolismo , Exosomas/metabolismo , Animales , Biomarcadores/metabolismo , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Humanos , Macrófagos/metabolismo , MicroARNs/metabolismo , Miocitos del Músculo Liso/metabolismo , Fosfohidrolasa PTEN/metabolismo
7.
Circ Res ; 127(5): 631-646, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32418505

RESUMEN

RATIONALE: Cardiac CITED4 (CBP/p300-interacting transactivators with E [glutamic acid]/D [aspartic acid]-rich-carboxylterminal domain4) is induced by exercise and is sufficient to cause physiological hypertrophy and mitigate adverse ventricular remodeling after ischemic injury. However, the role of endogenous CITED4 in response to physiological or pathological stress is unknown. OBJECTIVE: To investigate the role of CITED4 in murine models of exercise and pressure overload. METHODS AND RESULTS: We generated cardiomyocyte-specific CITED4 knockout mice (C4KO) and subjected them to an intensive swim exercise protocol as well as transverse aortic constriction (TAC). Echocardiography, Western blotting, qPCR, immunohistochemistry, immunofluorescence, and transcriptional profiling for mRNA and miRNA (microRNA) expression were performed. Cellular crosstalk was investigated in vitro. CITED4 deletion in cardiomyocytes did not affect baseline cardiac size or function in young adult mice. C4KO mice developed modest cardiac dysfunction and dilation in response to exercise. After TAC, C4KOs developed severe heart failure with left ventricular dilation, impaired cardiomyocyte growth accompanied by reduced mTOR (mammalian target of rapamycin) activity and maladaptive cardiac remodeling with increased apoptosis, autophagy, and impaired mitochondrial signaling. Interstitial fibrosis was markedly increased in C4KO hearts after TAC. RNAseq revealed induction of a profibrotic miRNA network. miR30d was decreased in C4KO hearts after TAC and mediated crosstalk between cardiomyocytes and fibroblasts to modulate fibrosis. miR30d inhibition was sufficient to increase cardiac dysfunction and fibrosis after TAC. CONCLUSIONS: CITED4 protects against pathological cardiac remodeling by regulating mTOR activity and a network of miRNAs mediating cardiomyocyte to fibroblast crosstalk. Our findings highlight the importance of CITED4 in response to both physiological and pathological stimuli.


Asunto(s)
Cardiomegalia Inducida por el Ejercicio , Hipertrofia Ventricular Izquierda/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Comunicación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/patología , Ratas , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transcriptoma
8.
JACC Clin Electrophysiol ; 6(1): 70-79, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31971908

RESUMEN

OBJECTIVES: This study evaluated whether plasma miRNAs were specifically associated with sudden cardiac and/or arrhythmic death (SCD) in a cohort of patients with coronary heart disease (CHD), most of whom were without primary prevention implantable cardioverter-defibrillators. BACKGROUND: Novel biomarkers for sudden death risk stratification are needed in patients with CHD to more precisely target preventive therapies, such as implantable cardioverter-defibrillators. miRNAs have been implicated in regulating inflammation and cardiac fibrosis in cells, and plasma miRNAs have been shown to predict cardiovascular death in patients with CHD. METHODS: We performed a nested case control study within a multicenter cohort of 5,956 patients with CHD followed prospectively for SCD. Plasma levels of 18 candidate miRNAs previously associated with cardiac remodeling were measured in 129 SCD cases and 258 control subjects matched on age, sex, race, and left ventricular ejection fraction. RESULTS: miR-150-5p, miR-29a-3p, and miR-30a-5p were associated with increased SCD risk (odds ratios and 95% confidence intervals: 2.03 [1.12 to 3.67]; p = 0.02; 1.93 [1.07 to 3.50]; p = 0.02; 0.55 [0.31 to 0.97]; p = 0.04, respectively, for third vs. first tertile miRNA level). Unfavorable levels of all 3 miRNAs was associated with a 4.8-fold increased SCD risk (1.59 to 14.51; p = 0.006). A bioinformatics-based approach predicted miR-150-5p, miR-29a-3p, and miR-30a-5p to be involved in apoptosis, fibrosis, and inflammation. CONCLUSIONS: These findings suggest that plasma miRNAs may regulate pathways important for remodeling and may be useful in identifying patients with CHD at increased risk of SCD.


Asunto(s)
Enfermedad Coronaria , Muerte Súbita Cardíaca/epidemiología , MicroARNs , Anciano , Biomarcadores/sangre , Biomarcadores/metabolismo , Estudios de Casos y Controles , Enfermedad Coronaria/genética , Enfermedad Coronaria/metabolismo , Citocinas/sangre , Femenino , Humanos , Masculino , MicroARNs/sangre , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Medición de Riesgo
9.
JCI Insight ; 4(7)2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30779709

RESUMEN

Heart failure (HF) is associated in humans and mice with increased circulating levels of CXCL9 and CXCL10, chemokine ligands of the CXCR3 receptor, predominantly expressed on CD4+ Th1 cells. Chemokine engagement of receptors is required for T cell integrin activation and recruitment to sites of inflammation. Th1 cells drive adverse cardiac remodeling in pressure overload-induced cardiac dysfunction, and mice lacking the integrin ligand ICAM-1 show defective T cell recruitment to the heart. Here, we show that CXCR3+ T cells infiltrate the heart in humans and mice with pressure overload-induced cardiac dysfunction. Genetic deletion of CXCR3 disrupts CD4+ T cell heart infiltration and prevents adverse cardiac remodeling. We demonstrate that cardiac fibroblasts and cardiac myeloid cells that include resident and infiltrated macrophages are the source of CXCL9 and CXCL10, which mechanistically promote Th1 cell adhesion to ICAM-1 under shear conditions in a CXCR3-dependent manner. To our knowledge, our findings identify a previously unrecognized role for CXCR3 in Th1 cell recruitment into the heart in pressure overload-induced cardiac dysfunction.


Asunto(s)
Insuficiencia Cardíaca/inmunología , Miocardio/inmunología , Receptores CXCR3/metabolismo , Células TH1/inmunología , Animales , Presión Sanguínea , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/inmunología , Quimiocina CXCL9/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Fibroblastos , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/patología , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Macrófagos , Masculino , Ratones , Miocardio/citología , Miocardio/patología , Miofibroblastos , Receptores CXCR3/inmunología , Células TH1/metabolismo
10.
Immunology ; 157(1): 52-69, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30690734

RESUMEN

T helper type 17 lymphocytes (Th17 cells) infiltrate the central nervous system (CNS), induce inflammation and demyelination and play a pivotal role in the pathogenesis of multiple sclerosis. Sialomucin CD43 is highly expressed in Th17 cells and mediates adhesion to endothelial selectin (E-selectin), an initiating step in Th17 cell recruitment to sites of inflammation. CD43-/- mice have impaired Th17 cell recruitment to the CNS and are protected from experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis. However, E-selectin is dispensable for the development of EAE, in contrast to intercellular and vascular cell adhesion molecules (ICAM-1 and VCAM-1). We report that CD43-/- mice have decreased demyelination and T-cell infiltration, but similar up-regulation of ICAM-1 and VCAM-1 in the spinal cord, compared with wild-type (WT) mice, at the initiation of EAE. CD43-/- Th17 cells have impaired adhesion to ICAM-1 under flow conditions in vitro, despite having similar expression of LFA-1, the main T-cell ligand for ICAM-1, as WT Th17 cells. Regardless of the route of integrin activation, CD43-/- Th17 cell firm arrest on ICAM-1 was comparable to that of WT Th17 cells, but CD43-/- Th17 cells failed to optimally apically migrate on immobilized ICAM-1-coated coverslips and endothelial cells, and to transmigrate under shear flow conditions in an ICAM-1-dependent manner. Collectively, these findings unveil novel roles for CD43, facilitating adhesion of Th17 cells to ICAM-1 and modulating apical and transendothelial migration, as mechanisms potentially responsible for Th17 cell recruitment to sites of inflammation such as the CNS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Leucosialina/metabolismo , Esclerosis Múltiple/inmunología , Células Th17/inmunología , Animales , Adhesión Celular , Movimiento Celular , Modelos Animales de Enfermedad , Humanos , Molécula 1 de Adhesión Intercelular/genética , Leucosialina/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Migración Transendotelial y Transepitelial , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
Circulation ; 138(12): 1236-1252, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-29653926

RESUMEN

BACKGROUND: Fibronectin (FN) polymerization is necessary for collagen matrix deposition and is a key contributor to increased abundance of cardiac myofibroblasts (MFs) after cardiac injury. We hypothesized that interfering with FN polymerization or its genetic ablation in fibroblasts would attenuate MF and fibrosis and improve cardiac function after ischemia/reperfusion (I/R) injury. METHODS: Mouse and human MFs were used to assess the impact of the FN polymerization inhibitor (pUR4) in attenuating pathological cellular features such as proliferation, migration, extracellular matrix deposition, and associated mechanisms. To evaluate the therapeutic potential of inhibiting FN polymerization in vivo, wild-type mice received daily intraperitoneal injections of either pUR4 or control peptide (III-11C) immediately after cardiac surgery for 7 consecutive days. Mice were analyzed 7 days after I/R to assess MF markers and inflammatory cell infiltration or 4 weeks after I/R to evaluate long-term effects of FN inhibition on cardiac function and fibrosis. Furthermore, inducible, fibroblast-restricted, FN gene-ablated (Tcf21MerCreMer; Fnflox) mice were used to evaluate cell specificity of FN expression and polymerization in the heart. RESULTS: pUR4 administration on activated MFs reduced FN and collagen deposition into the extracellular matrix and attenuated cell proliferation, likely mediated through decreased c-myc signaling. pUR4 also ameliorated fibroblast migration accompanied by increased ß1 integrin internalization and reduced levels of phosphorylated focal adhesion kinase protein. In vivo, daily administration of pUR4 for 7 days after I/R significantly reduced MF markers and neutrophil infiltration. This treatment regimen also significantly attenuated myocardial dysfunction, pathological cardiac remodeling, and fibrosis up to 4 weeks after I/R. Last, inducible ablation of FN in fibroblasts after I/R resulted in significant functional cardioprotection with reduced hypertrophy and fibrosis. The addition of pUR4 to the FN-ablated mice did not confer further cardioprotection, suggesting that the salutary effects of inhibiting FN polymerization may be mediated largely through effects on FN secreted from the cardiac fibroblast lineage. CONCLUSIONS: Inhibiting FN polymerization or cardiac fibroblast gene expression attenuates pathological properties of MFs in vitro and ameliorates adverse cardiac remodeling and fibrosis in an in vivo model of heart failure. Interfering with FN polymerization may be a new therapeutic strategy for treating cardiac fibrosis and heart failure.


Asunto(s)
Fibronectinas/antagonistas & inhibidores , Insuficiencia Cardíaca/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miofibroblastos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis , Quinasa 1 de Adhesión Focal/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Integrina beta1/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Infiltración Neutrófila/efectos de los fármacos , Fosforilación , Polimerizacion , Transducción de Señal/efectos de los fármacos
12.
J Exp Med ; 214(11): 3311-3329, 2017 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-28970239

RESUMEN

Despite emerging data indicating a role for T cells in profibrotic cardiac repair and healing after ischemia, little is known about whether T cells directly impact cardiac fibroblasts (CFBs) to promote cardiac fibrosis (CF) in nonischemic heart failure (HF). Recently, we reported increased T cell infiltration in the fibrotic myocardium of nonischemic HF patients, as well as the protection from CF and HF in TCR-α-/- mice. Here, we report that T cells activated in such a context are mainly IFN-γ+, adhere to CFB, and induce their transition into myofibroblasts. Th1 effector cells selectively drive CF both in vitro and in vivo, whereas adoptive transfer of Th1 cells, opposite to activated IFN-γ-/- Th cells, partially reconstituted CF and HF in TCR-α-/- recipient mice. Mechanistically, Th1 cells use integrin α4 to adhere to and induce TGF-ß in CFB in an IFN-γ-dependent manner. Our findings identify a previously unrecognized role for Th1 cells as integrators of perivascular CF and cardiac dysfunction in nonischemic HF.


Asunto(s)
Fibroblastos/metabolismo , Insuficiencia Cardíaca/patología , Miocardio/patología , Células TH1/patología , Animales , Adhesión Celular , Fibrosis , Citometría de Flujo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Integrina alfa4/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Miocardio/metabolismo , Miofibroblastos/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Células TH1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
13.
Physiol Rep ; 5(12)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28637706

RESUMEN

Heart Failure (HF) is associated with increased circulating levels of aldosterone and systemic inflammation. Mineralocorticoid receptor (MR) antagonists block aldosterone action and decrease mortality in patients with congestive HF However, the molecular mechanisms underlying the therapeutic benefits of MR antagonists remain unclear. MR is expressed in all cell types in the heart, including the endothelial cells (EC), in which aldosterone induces the expression of intercellular adhesion molecule 1 (ICAM-1). Recently, we reported that ICAM-1 regulates cardiac inflammation and cardiac function in mice subjected to transverse aortic constriction (TAC). Whether MR specifically in endothelial cells (EC) contributes to the several mechanisms of pathological cardiac remodeling and cardiac dysfunction remains unclear. Basal cardiac function and LV dimensions were comparable in mice with MR selectively deleted from ECs (EC-MR-/-) and wild-type littermate controls (EC-MR+/+). MR was specifically deleted in heart EC, and in EC-containing tissues, but not in leukocytes of TAC EC-MR-/- mice. While EC-MR-/- TAC mice showed preserved systolic function and some alterations in the expression of fetal genes, the proinflammatory cytokine TNFα and the endothelin receptors in the LV as compared to EC-MR+/+ TAC mice, no difference was observed between both TAC groups in overall cardiac hypertrophy, ICAM-1 LV expression and leukocyte infiltration, cardiac fibrosis or capillary rarefaction, all hallmarks of pathological cardiac remodeling. Our data indicate that EC-MR contributes to the transition of cardiac hypertrophy to systolic dysfunction independently of other maladaptive changes induced by LV pressure overload.


Asunto(s)
Cardiomegalia/metabolismo , Endotelio Vascular/metabolismo , Receptores de Mineralocorticoides/genética , Sístole , Disfunción Ventricular , Animales , Cardiomegalia/fisiopatología , Células Cultivadas , Endotelio Vascular/patología , Fibrosis , Inflamación , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Mineralocorticoides/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Remodelación Ventricular
14.
J Am Heart Assoc ; 5(3): e003126, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-27068635

RESUMEN

BACKGROUND: Left ventricular dysfunction and heart failure are strongly associated in humans with increased circulating levels of proinflammatory cytokines, T cells, and soluble intercellular cell adhesion molecule 1 (ICAM1). In mice, infiltration of T cells into the left ventricle contributes to pathological cardiac remodeling, but the mechanisms regulating their recruitment to the heart are unclear. We hypothesized that ICAM1 regulates cardiac inflammation and pathological cardiac remodeling by mediating left ventricular T-cell recruitment and thus contributing to cardiac dysfunction and heart failure. METHODS AND RESULTS: In a mouse model of pressure overload-induced heart failure, intramyocardial endothelial ICAM1 increased within 48 hours in response to thoracic aortic constriction and remained upregulated as heart failure progressed. ICAM1-deficient mice had decreased T-cell and proinflammatory monocyte infiltration in the left ventricle in response to thoracic aortic constriction, despite having numbers of circulating T cells and activated T cells in the heart-draining lymph nodes that were similar to those of wild-type mice. ICAM1-deficient mice did not develop cardiac fibrosis or systolic and diastolic dysfunction in response to thoracic aortic constriction. Exploration of the mechanisms regulating ICAM1 expression revealed that endothelial ICAM1 upregulation and T-cell infiltration were not mediated by endothelial mineralocorticoid receptor signaling, as demonstrated in thoracic aortic constriction studies in mice with endothelial mineralocorticoid receptor deficiency, but rather were induced by the cardiac cytokines interleukin 1ß and 6. CONCLUSIONS: ICAM1 regulates pathological cardiac remodeling by mediating proinflammatory leukocyte infiltration in the left ventricle and cardiac fibrosis and dysfunction and thus represents a novel target for treatment of heart failure.


Asunto(s)
Aorta Torácica/fisiopatología , Presión Arterial , Quimiotaxis de Leucocito , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Linfocitos T/metabolismo , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Aorta Torácica/cirugía , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Cultivadas , Constricción , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/prevención & control , Molécula 1 de Adhesión Intercelular/genética , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Activación de Linfocitos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Miofibroblastos/patología , Transducción de Señal , Linfocitos T/patología , Factores de Tiempo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
15.
J Immunol ; 196(3): 1305-1316, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26700769

RESUMEN

Endothelial E- and P-selectins mediate lymphocyte trafficking in inflammatory processes by interacting with lymphocyte selectin ligands. These are differentially expressed among different T cell subsets and function alone or in cooperation to mediate T cell adhesion. In this study, we characterize the expression and functionality of E-selectin ligands in Th type 17 lymphocytes (Th17 cells) and report that CD43 functions as a Th17 cell E-selectin ligand in vitro that mediates Th17 cell rolling on the vascular endothelium and recruitment in vivo. We demonstrate Th17 cells express CD44, P-selectin glycoprotein ligand (PSGL)-1, and CD43. Few PSGL-1(-/-)CD43(-/-) Th17 cells accumulated on E-selectin under shear flow conditions compared with wild-type cells. CD43(-/-) Th17 cell accumulation on E-selectin was impaired as compared with wild-type and PSGL-1(-/-), and similar to that observed for PSGL-1(-/-)CD43(-/-) Th17 cells, indicating that CD43 alone is a dominant ligand for E-selectin. Notably, this finding is Th17 cell subset specific because CD43 requires cooperation with PSGL-1 in Th1 cells for binding to E-selectin. In vivo, Th17 cell recruitment into the air pouch was reduced in CD43(-/-) mice in response to CCL20 or TNF-α, and intravital microscopy studies demonstrated that CD43(-/-) Th17 cells had impaired rolling on TNF-α-treated microvessels. Furthermore, CD43(-/-) mice were protected from experimental autoimmune encephalomyelitis and had impaired recruitment of Th17 cells in the spinal cord. Our findings demonstrate that CD43 is a major E-selectin ligand in Th17 cells that functions independent of PSGL-1, and they suggest that CD43 may hold promise as a therapeutic target to modulate Th17 cell recruitment.


Asunto(s)
Selectina E/inmunología , Inflamación/inmunología , Rodamiento de Leucocito/inmunología , Leucosialina/inmunología , Células Th17/inmunología , Animales , Western Blotting , Separación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Endotelio Vascular/inmunología , Citometría de Flujo , Inmunoprecipitación , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Circ Heart Fail ; 8(4): 776-87, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26022677

RESUMEN

BACKGROUND: Despite the emerging association between heart failure (HF) and inflammation, the role of T cells, major players in chronic inflammation, has only recently begun to be explored. Whether T-cell recruitment to the left ventricle (LV) participates in the development of HF requires further investigation to identify novel mechanisms that may serve for the design of alternative therapeutic interventions. METHODS AND RESULTS: Real-time videomicroscopy of T cells from nonischemic HF patients or from mice with HF induced by transverse aortic constriction revealed enhanced adhesion to activated vascular endothelial cells under flow conditions in vitro compared with T cells from healthy subjects or sham mice. T cells in the mediastinal lymph nodes and the intramyocardial endothelium were both activated in response to transverse aortic constriction and the kinetics of LV T-cell infiltration was directly associated with the development of systolic dysfunction. In response to transverse aortic constriction, T cell-deficient mice (T-cell receptor, TCRα(-/-)) had preserved LV systolic and diastolic function, reduced LV fibrosis, hypertrophy and inflammation, and improved survival compared with wild-type mice. Furthermore, T-cell depletion in wild-type mice after transverse aortic constriction prevented HF. CONCLUSIONS: T cells are major contributors to nonischemic HF. Their activation combined with the activation of the LV endothelium results in LV T-cell infiltration negatively contributing to HF progression through mechanisms involving cytokine release and induction of cardiac fibrosis and hypertrophy. Reduction of T-cell infiltration is thus identified as a novel translational target in HF.


Asunto(s)
Quimiotaxis de Leucocito , Insuficiencia Cardíaca/inmunología , Ventrículos Cardíacos/inmunología , Activación de Linfocitos , Linfocitos T/inmunología , Adulto , Animales , Estudios de Casos y Controles , Adhesión Celular , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/inmunología , Fibrosis , Genes Codificadores de la Cadena alfa de los Receptores de Linfocito T , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Hipertrofia Ventricular Izquierda/inmunología , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Cinética , Masculino , Ratones Noqueados , Microscopía por Video , Persona de Mediana Edad , Contracción Miocárdica , Disfunción Ventricular Izquierda/inmunología , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda , Remodelación Ventricular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...