Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Circ Res ; 132(5): 565-582, 2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-36744467

RESUMEN

BACKGROUND: In the past years, several studies investigated how distinct immune cell subsets affects post-myocardial infarction repair. However, whether and how the tissue environment controls these local immune responses has remained poorly understood. We sought to investigate how antigen-specific T-helper cells differentiate under myocardial milieu's influence. METHODS: We used a transgenic T cell receptor (TCR-M) model and major histocompatibility complex-II tetramers, both myosin-specific, combined with single-cell transcriptomics (single-cell RNA sequencing [scRNA-seq]) and functional phenotyping to elucidate how the antigen-specific CD4+ T cells differentiate in the murine infarcted myocardium and influence tissue repair. Additionally, we transferred proinflammatory versus regulatory predifferentiated TCR-M-cells to dissect how they specially contribute to post-myocardial infarction inflammation. RESULTS: Flow cytometry and scRNA-/TCR-seq analyses revealed that transferred TCR-M cells acquired an induced regulatory phenotype (induced regulatory T cell) in the infarcted myocardium and blunted local inflammation. Myocardial TCR-M cells differentiated into 2 main lineages enriched with either cell activation and profibrotic transcripts (eg, Tgfb1) or suppressor immune checkpoints (eg, Pdcd1), which we also found in human myocardial tissue. These cells produced high levels of LAP (latency-associated peptide) and inhibited IL-17 (interleukin-17) responses. Endogenous myosin-specific T-helper cells, identified using genetically barcoded tetramers, also accumulated in infarcted hearts and exhibited a regulatory phenotype. Notably, TCR-M cells that were predifferentiated toward a regulatory phenotype in vitro maintained stable in vivo FOXP3 (Forkhead box P3) expression and anti-inflammatory activity whereas TH17 partially converted toward a regulatory phenotype in the injured myocardium. Overall, the myosin-specific Tregs dampened post-myocardial infarction inflammation, suppressed neighboring T cells, and were associated with improved cardiac function. CONCLUSIONS: These findings provide novel evidence that the heart and its draining lymph nodes actively shape local immune responses by promoting the differentiation of antigen-specific Tregs poised with suppressive function.


Asunto(s)
Infarto del Miocardio , Linfocitos T Reguladores , Ratones , Animales , Humanos , Miocardio/metabolismo , Infarto del Miocardio/metabolismo , Antígenos/metabolismo , Diferenciación Celular , Miosinas/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Inflamación/metabolismo , Factores de Transcripción Forkhead/genética
2.
Basic Res Cardiol ; 119(2): 261-275, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38436707

RESUMEN

Myocardial infarction (MI) induces the generation of proinflammatory Ly6Chigh monocytes in the spleen and the recruitment of these cells to the myocardium. CD4+ Foxp3+ CD25+ T-cells (Tregs) promote the healing process after myocardial infarction by engendering a pro-healing differentiation state in myocardial monocyte-derived macrophages. We aimed to study the effects of CD4+ T-cells on splenic myelopoiesis and monocyte differentiation. We instigated MI in mice and found that MI-induced splenic myelopoiesis is abrogated in CD4+ T-cell deficient animals. Conventional CD4+ T-cells promoted myelopoiesis in vitro by cell-cell-contact and paracrine mechanisms, including interferon-gamma (IFN-γ) signalling. Depletion of regulatory T-cells enhanced myelopoiesis in vivo, as evidenced by increases in progenitor cell numbers and proliferative activity in the spleen 5 days after MI. The frequency of CD4+ T-cells-producing factors that promote myelopoiesis increased within the spleen of Treg-depleted mice. Moreover, depletion of Tregs caused a proinflammatory bias in splenic Ly6Chigh monocytes, which showed predominantly upregulated expression of IFN-γ responsive genes after MI. Our results indicate that conventional CD4+ T-cells promote and Tregs attenuate splenic myelopoiesis and proinflammatory differentiation of monocytes.


Asunto(s)
Monocitos , Infarto del Miocardio , Ratones , Animales , Monocitos/metabolismo , Mielopoyesis , Bazo/metabolismo , Infarto del Miocardio/metabolismo , Linfocitos T Reguladores/metabolismo , Interferón gamma/farmacología , Ratones Endogámicos C57BL
3.
Basic Res Cardiol ; 119(3): 453-479, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38491291

RESUMEN

Though myocardial infarction (MI) in pigs is a well-established translational large animal model, it has not yet been widely used for immunotherapy studies, and a comprehensive description of the immune response to MI in this species is lacking. We induced MI in Landrace pigs by balloon occlusion of the left anterior descending artery over 90 min. Within 14 days, the necrotic myocardium was progressively replaced by scar tissue with involvement of myofibroblasts. We characterized the immune response in the heart ex vivo by (immuno)histology, flow cytometry, and RNA sequencing of myocardial tissue on days 3, 7, and 14 after MI. Besides a clear predominance of myeloid cells among heart-infiltrating leukocytes, we detected activated T cells and an increasing proportion of CD4+ Foxp3+ regulatory T cells (Treg), especially in the infarct core-findings that closely mirror what has been observed in mice and humans after MI. Transcriptome data indicated inflammatory activity that was persistent but markedly changing in character over time and linked to extracellular matrix biology. Analysis of lymphocytes in heart-draining lymph nodes revealed significantly higher proliferation rates of T helper cell subsets, including Treg on day 7 after MI, compared to sham controls. Elevated frequencies of myeloid progenitors in the spleen suggest that it might be a site of emergency myelopoiesis after MI in pigs, as previously shown in mice. We thus provide a first description of the immune response to MI in pigs, and our results can aid future research using the species for preclinical immunotherapy studies.


Asunto(s)
Modelos Animales de Enfermedad , Infarto del Miocardio , Miocardio , Linfocitos T Reguladores , Animales , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Linfocitos T Reguladores/inmunología , Miocardio/patología , Miocardio/inmunología , Sus scrofa , Porcinos , Activación de Linfocitos , Masculino , Transcriptoma , Femenino , Factores de Tiempo
4.
J Immunol ; 207(10): 2473-2488, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34625520

RESUMEN

Because of its size, anatomical similarities, and now also accessibility to genetic manipulations, pigs are used as animal models for human diseases and immune system development. However, expression and function of CD28, the most important costimulatory receptor expressed by T cells, so far is poorly understood in this species. Using a newly generated mAb (mAb 3D11) with specificity for pig CD28, we detected CD28 on CD8+ and CD4+ αß T cells. Among γδ T cells, CD28 expression was restricted to a small CD2+ subpopulation of phenotypically naive cells. Functionally, CD28 ligation with mAb 3D11-costimulated porcine T cells, enhanced proliferation and cytokine secretion in vitro. We used a second, likewise newly generated but superagonistic, anti-CD28 mAb (CD28-SA; mAb 4D12) to test the function of CD28 on porcine T cells in a pilot study in vivo. Injection of the CD28-SA into pigs in vivo showed a very similar dose-response relationship as in humans (i.e., 100 µg/kg body weight [BW]) of CD28-SA induced a cytokine release syndrome that was avoided at a dose of 10 µg/kg BW and below. The data further suggest that low-dose (10 µg/kg BW) CD28-SA infusion was sufficient to increase the proportion of Foxp3+ regulatory T cells among CD4+ T cells in vivo. The pig is thus a suitable animal model for testing novel immunotherapeutics. Moreover, data from our pilot study in pigs further suggest that low-dose CD28-SA infusion might allow for selective expansion of CD4+ regulatory T cells in humans.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos CD28/inmunología , Modelos Animales , Porcinos/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Humanos , Activación de Linfocitos/inmunología
5.
MAGMA ; 36(2): 279-293, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37027119

RESUMEN

INTRODUCTION: MRI of excised hearts at ultra-high field strengths ([Formula: see text]≥7 T) can provide high-resolution, high-fidelity ground truth data for biomedical studies, imaging science, and artificial intelligence. In this study, we demonstrate the capabilities of a custom-built, multiple-element transceiver array customized for high-resolution imaging of excised hearts. METHOD: A dedicated 16-element transceiver loop array was implemented for operation in parallel transmit (pTx) mode (8Tx/16Rx) of a clinical whole-body 7 T MRI system. The initial adjustment of the array was performed using full-wave 3D-electromagnetic simulation with subsequent final fine-tuning on the bench. RESULTS: We report the results of testing the implemented array in tissue-mimicking liquid phantoms and excised porcine hearts. The array demonstrated high efficiency of parallel transmits characteristics enabling efficient pTX-based B1+-shimming. CONCLUSION: The receive sensitivity and parallel imaging capability of the dedicated coil were superior to that of a commercial 1Tx/32Rx head coil in both SNR and T2*-mapping. The array was successfully tested to acquire ultra-high-resolution (0.1 × 0.1 × 0.8 mm voxel) images of post-infarction scar tissue. High-resolution (isotropic 1.6 mm3 voxel) diffusion tensor imaging-based tractography provided high-resolution information about normal myocardial fiber orientation.


Asunto(s)
Inteligencia Artificial , Imagen de Difusión Tensora , Porcinos , Animales , Relación Señal-Ruido , Diseño de Equipo , Imagen por Resonancia Magnética/métodos , Fantasmas de Imagen
6.
Eur Heart J ; 43(28): 2698-2709, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35417553

RESUMEN

AIMS: Newborn mice and humans display transient cardiac regenerative potential that rapidly declines postnatally. Patients who survive a myocardial infarction (MI) often develop chronic heart failure due to the heart's poor regeneration capacity. We hypothesized that the cardiac 'regenerative-to-scarring' transition might be driven by the perinatal shifts observed in the circulating T-cell compartment. METHODS AND RESULTS: Post-MI immune responses were characterized in 1- (P1) vs. 7-day-old (P7) mice subjected to left anterior descending artery ligation. Myocardial infarction induced robust early inflammatory responses (36 h post-MI) in both age groups, but neonatal hearts exhibited rapid resolution of inflammation and full functional recovery. The perinatal loss of myocardial regenerative capacity was paralleled by a baseline increase in αß-T cell (CD4+ and CD8+) numbers. Strikingly, P1-infarcted mice reconstituted with adult T-cells shifted to an adult-like healing phenotype, marked by irreversible cardiac functional impairment and increased fibrosis. Infarcted neonatal mice harbouring adult T-cells also had more monocyte-derived macrophage recruitment, as typically seen in adults. At the transcriptome level, infarcted P1 hearts that received isolated adult T-cells showed enriched gene sets linked to fibrosis, inflammation, and interferon-gamma (IFN-γ) signalling. In contrast, newborn mice that received isolated Ifng-/- adult T-cells prior to MI displayed a regenerative phenotype that resembled that of its age-matched untreated controls. CONCLUSION: Physiological T-cell development or adoptive transfer of adult IFN-γ-producing T-cells into neonates contributed to impaired cardiac regeneration and promoted irreversible structural and functional cardiac damage. These findings reveal a trade-off between myocardial regenerative potential and the development of T-cell competence.


Asunto(s)
Infarto del Miocardio , Miocitos Cardíacos , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Fibrosis , Humanos , Inflamación/patología , Interferón gamma , Ratones , Miocardio/patología , Miocitos Cardíacos/fisiología , Embarazo , Regeneración/fisiología
7.
J Mol Cell Cardiol ; 173: 25-29, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36122767

RESUMEN

T-cells contribute to pathophysiological processes in myocardial diseases, including myocardial infarction (MI) and heart failure (HF). Antigen-specificity is a hallmark of T-cell responses but the cardiac antigens that trigger heart-directed T-cell responses in patients have not yet been uncovered, thus posing a roadblock to translation. In the present exploratory study, we identified a peptide fragment of the beta-1 adrenergic receptor (ADRB1) that elicits CD4+ T-cell responses after myocardial infarction in patients with a defined HLA haplotype. Our observations may advance the development of tools to monitor other antigen-specific immune responses in patients.


Asunto(s)
Linfocitos T CD4-Positivos , Infarto del Miocardio , Humanos , Epítopos , Corazón
8.
Mol Cell Biochem ; 477(6): 1789-1801, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35312907

RESUMEN

Calsequestrin (CSQ2) is the main Ca2+-binding protein in the sarcoplasmic reticulum of the mammalian heart. In order to understand the function of calsequestrin better, we compared two age groups (young: 4-5 months of age versus adult: 18 months of age) of CSQ2 knock-out mice (CSQ2(-/-)) and littermate wild-type mice (CSQ2(+/+)). Using echocardiography, in adult mice, the basal left ventricular ejection fraction and the spontaneous beating rate were lower in CSQ2(-/-) compared to CSQ2(+/+). The increase in ejection fraction by ß-adrenergic stimulation (intraperitoneal injection of isoproterenol) was lower in adult CSQ2(-/-) versus adult CSQ2(+/+). After hypoxia in vitro (isolated atrial preparations) by gassing the organ bath buffer with 95% N2, force of contraction in electrically driven left atria increased to lower values in young CSQ2(-/-) than in young CSQ2(+/+). In addition, after global ischemia and reperfusion (buffer-perfused hearts according to Langendorff; 20-min ischemia and 15-min reperfusion), the rate of tension development was higher in young CSQ2(-/-) compared to young CSQ2(+/+). Finally, we evaluated signs of inflammation (immune cells, autoantibodies, and fibrosis). However, whereas no immunological alterations were found between all investigated groups, pronounced fibrosis was found in the ventricles of adult CSQ2(-/-) compared to all other groups. We suggest that in young mice, CSQ2 is important for cardiac performance especially in isolated cardiac preparations under conditions of impaired oxygen supply, but with differences between atrium and ventricle. Lack of CSQ2 leads age dependently to fibrosis and depressed cardiac performance in echocardiographic studies.


Asunto(s)
Calcio , Calsecuestrina , Animales , Calcio/metabolismo , Calsecuestrina/genética , Calsecuestrina/metabolismo , Fibrosis , Atrios Cardíacos/metabolismo , Hipoxia/metabolismo , Isquemia/metabolismo , Mamíferos/metabolismo , Ratones , Ratones Noqueados , Contracción Miocárdica , Retículo Sarcoplasmático/metabolismo , Volumen Sistólico , Función Ventricular Izquierda
9.
Int J Mol Sci ; 23(18)2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36142218

RESUMEN

Acute ischemic cardiac injury predisposes one to cognitive impairment, dementia, and depression. Pathophysiologically, recent positron emission tomography data suggest astroglial activation after experimental myocardial infarction (MI). We analyzed peripheral surrogate markers of glial (and neuronal) damage serially within 12 months after the first ST-elevation MI (STEMI). Serum levels of glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL) were quantified using ultra-sensitive molecular immunoassays. Sufficient biomaterial was available from 45 STEMI patients (aged 28 to 78 years, median 56 years, 11% female). The median (quartiles) of GFAP was 63.8 (47.0, 89.9) pg/mL and of NfL 10.6 (7.2, 14.8) pg/mL at study entry 0-4 days after STEMI. GFAP after STEMI increased in the first 3 months, with a median change of +7.8 (0.4, 19.4) pg/mL (p = 0.007). It remained elevated without further relevant increases after 6 months (+11.7 (0.6, 23.5) pg/mL; p = 0.015), and 12 months (+10.3 (1.5, 22.7) pg/mL; p = 0.010) compared to the baseline. Larger relative infarction size was associated with a higher increase in GFAP (ρ = 0.41; p = 0.009). In contrast, NfL remained unaltered in the course of one year. Our findings support the idea of central nervous system involvement after MI, with GFAP as a potential peripheral biomarker of chronic glial damage as one pathophysiologic pathway.


Asunto(s)
Infarto del Miocardio , Infarto del Miocardio con Elevación del ST , Materiales Biocompatibles , Biomarcadores , Femenino , Proteína Ácida Fibrilar de la Glía , Humanos , Filamentos Intermedios , Masculino , Infarto del Miocardio/diagnóstico por imagen , Proteínas de Neurofilamentos
10.
Arterioscler Thromb Vasc Biol ; 40(3): 682-696, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31893950

RESUMEN

OBJECTIVE: Activated perivascular mast cells (MCs) participate in different cardiovascular diseases. Many factors provoking MC degranulation have been described, while physiological counterregulators are barely known. Endothelial CNP (C-type natriuretic peptide) participates in the maintenance of vascular barrier integrity, but the target cells and mechanisms are unclear. Here, we studied whether MCs are regulated by CNP. Approach and Results: In cultured human and murine MCs, CNP activated its specific GC (guanylyl cyclase)-B receptor and cyclic GMP signaling. This enhanced cyclic GMP-dependent phosphorylation of the cytoskeleton-associated VASP (vasodilator-stimulated phosphoprotein) and inhibited ATP-evoked degranulation. To elucidate the relevance in vivo, mice with a floxed GC-B (Npr2) gene were interbred with a Mcpt5-CreTG line to generate mice lacking GC-B in connective tissue MCs (MC GC-B knockout). In anesthetized mice, acute ischemia-reperfusion of the cremaster muscle microcirculation provoked extensive MC degranulation and macromolecule extravasation. Superfusion of CNP markedly prevented MC activation and endothelial barrier disruption in control but not in MC GC-B knockout mice. Notably, already under resting conditions, such knockout mice had increased numbers of degranulated MCs in different tissues, together with elevated plasma chymase levels. After transient coronary occlusion, their myocardial areas at risk and with infarction were enlarged. Moreover, MC GC-B knockout mice showed augmented perivascular neutrophil infiltration and deep vein thrombosis in a model of inferior vena cava ligation. CONCLUSIONS: CNP, via GC-B/cyclic GMP signaling, stabilizes resident perivascular MCs at baseline and prevents their excessive activation under pathological conditions. Thereby CNP contributes to the maintenance of vascular integrity in physiology and disease.


Asunto(s)
Degranulación de la Célula , Células Endoteliales/metabolismo , Mastocitos/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Péptido Natriurético Tipo-C/metabolismo , Comunicación Paracrina , Receptores del Factor Natriurético Atrial/metabolismo , Trombosis/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Permeabilidad Capilar , Moléculas de Adhesión Celular/metabolismo , Degranulación de la Célula/efectos de los fármacos , Línea Celular , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Mastocitos/efectos de los fármacos , Mastocitos/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Péptido Natriurético Tipo-C/farmacología , Infiltración Neutrófila , Fosfoproteínas/metabolismo , Fosforilación , Receptores del Factor Natriurético Atrial/agonistas , Receptores del Factor Natriurético Atrial/genética , Transducción de Señal , Trombosis/genética , Trombosis/patología
11.
Sensors (Basel) ; 21(16)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34451104

RESUMEN

This work investigates elimination methods for cardiogenic artifacts in respiratory surface electromyographic (sEMG) signals and compares their performance with respect to subsequent fatigue detection with different fatigue algorithms. The analysis is based on artificially constructed test signals featuring a clearly defined expected fatigue level. Test signals are additively constructed with different proportions from sEMG and electrocardiographic (ECG) signals. Cardiogenic artifacts are eliminated by high-pass filtering (HP), template subtraction (TS), a newly introduced two-step approach (TSWD) consisting of template subtraction and a wavelet-based damping step and a pure wavelet-based damping (DSO). Each method is additionally combined with the exclusion of QRS segments (gating). Fatigue is subsequently quantified with mean frequency (MNF), spectral moments ratio of order five (SMR5) and fuzzy approximate entropy (fApEn). Different combinations of artifact elimination methods and fatigue detection algorithms are tested with respect to their ability to deliver invariant results despite increasing ECG contamination. Both DSO and TSWD artifact elimination methods displayed promising results regarding the intermediate, "cleaned" EMG signal. However, only the TSWD method enabled superior results in the subsequent fatigue detection across different levels of artifact contamination and evaluation criteria. SMR5 could be determined as the best fatigue detection algorithm. This study proposes a signal processing chain to determine neuromuscular fatigue despite the presence of cardiogenic artifacts. The results furthermore underline the importance of selecting a combination of algorithms that play well together to remove cardiogenic artifacts and to detect fatigue. This investigation provides guidance for clinical studies to select optimal signal processing to detect fatigue from respiratory sEMG signals.


Asunto(s)
Artefactos , Fatiga Muscular , Algoritmos , Electrocardiografía , Electromiografía , Contracción Muscular , Músculos Respiratorios , Procesamiento de Señales Asistido por Computador
12.
Nanomedicine ; 23: 102096, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31669855

RESUMEN

Phosphatidylserine (PS) and phosphatidylglycerol (PG) are endogenous phospholipids with putative anti-inflammatory potential. However, studies comparing PS and PG are rare and were mainly conducted with phospholipid-dispersions of large size and broad distributions. Thus, we prepared small-sized PS- and PG-loaded liposomes exhibiting narrow distribution, and additionally studied the impact of liposome-pegylation on the reduction of the TNFα-production caused by the PS- and PG-liposomes. These PS- and PG-containing nanodispersions had a small size around 100nm and a narrow distribution (PDI<0.1). The liposome-dispersions showed no toxicity in NHDF- and 3T3-cells and virtually no hemolytic activity. They decreased the TNFα-production of LPS-(lipopolysaccharide)-stimulated mouse peritoneal macrophages in vitro. PG-liposomes always decreased the TNFα-levels more potently than PS-liposomes. Pegylation of PS- and PG-liposomes caused different Zeta potentials, but did not change biological activity. The results of the current study indicate a high potential of the tested formulations for phospholipid-based anti-inflammatory therapies.


Asunto(s)
Macrófagos Peritoneales/metabolismo , Nanopartículas , Fosfatidilgliceroles , Fosfatidilserinas , Células 3T3 , Animales , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Liposomas , Macrófagos Peritoneales/patología , Ratones , Nanopartículas/química , Nanopartículas/uso terapéutico , Fosfatidilgliceroles/química , Fosfatidilgliceroles/farmacología , Fosfatidilserinas/química , Fosfatidilserinas/farmacología
13.
Proc Natl Acad Sci U S A ; 114(12): E2420-E2429, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28255084

RESUMEN

In recent years, the myocardium has been rediscovered under the lenses of immunology, and lymphocytes have been implicated in the pathogenesis of cardiomyopathies with different etiologies. Aging is an important risk factor for heart diseases, and it also has impact on the immune system. Thus, we sought to determine whether immunological activity would influence myocardial structure and function in elderly mice. Morphological, functional, and molecular analyses revealed that the age-related myocardial impairment occurs in parallel with shifts in the composition of tissue-resident leukocytes and with an accumulation of activated CD4+ Foxp3- (forkhead box P3) IFN-γ+ T cells in the heart-draining lymph nodes. A comprehensive characterization of different aged immune-deficient mouse strains revealed that T cells significantly contribute to age-related myocardial inflammation and functional decline. Upon adoptive cell transfer, the T cells isolated from the mediastinal lymph node (med-LN) of aged animals exhibited increased cardiotropism, compared with cells purified from young donors or from other irrelevant sites. Nevertheless, these cells caused rather mild effects on cardiac functionality, indicating that myocardial aging might stem from a combination of intrinsic and extrinsic (immunological) factors. Taken together, the data herein presented indicate that heart-directed immune responses may spontaneously arise in the elderly, even in the absence of a clear tissue damage or concomitant infection. These observations might shed new light on the emerging role of T cells in myocardial diseases, which primarily affect the elderly population.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD4-Positivos/inmunología , Miocardio/inmunología , Traslado Adoptivo , Animales , Corazón/crecimiento & desarrollo , Humanos , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL
15.
Opt Express ; 27(6): 9097-9098, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-31052719

RESUMEN

Erroneous absolute luminance values of the pixelated phosphor structure B25 (pixel size of 25 microns by 25 microns) have been corrected. The updated figure shows clearly the potential of the investigated structure as light-converter for high-resolution lighting systems.

16.
Am J Physiol Heart Circ Physiol ; 315(5): H1358-H1367, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30095974

RESUMEN

Heart-specific antibodies have been widely associated with myocardial infarction (MI). However, it remains unclear whether autoantibodies mediate disease progression or are a byproduct of cardiac injury. To disambiguate the role of immunoglobulins in MI, we characterized the development of ischemic heart failure in agammaglobulinemic mice (AID-/-µS-/-). Although these animals can produce functional B cells, they cannot synthesize secretory IgM (µS-/-) or perform Ig class switching (AID-/-), leading to complete antibody deficiency. Agammaglobulinemia did not affect overall post-MI survival but resulted in a significant reduction in infarct size. Echocardiographic analyses showed that, compared with wild-type infarcted control mice, AID-/-µS-/- mice exhibited improved cardiac function and reduced remodeling on day 56 post-MI. These differences remained significant even after animals with matched infarct sizes were compared. Infarcted AID-/-µS-/- mice also showed reduced myocardial expression levels of transcripts known to promote adverse remodeling, such as matrix metalloproteinase-9, collagen type I a1, collagen type III a1, and IL-6. An unbiased screening of the heart reactivity potential in the plasma of wild-type MI animals revealed the presence of antibodies that target the myocardial scar and collagenase-sensitive epitopes. Moreover, we found that IgG accumulated within the scar tissues of infarcted mice and remained in close proximity with cells expressing Fcγ receptors (CD16/32), suggesting the existence of an in situ IgG-Fcγ receptor axis. Collectively, our study results confirm that antibodies contribute to ischemic heart failure progression and provide novel insights into the mechanisms underlying this phenomenon. NEW & NOTEWORTHY Our study sheds some light on the long-standing debate over the relevance of autoantibodies in heart failure and might stimulate future research in the field. The observation of extracellular matrix-specific antibodies and the detection of Fcγ receptor-expressing cells within the scar provide novel insights into the mechanisms by which antibodies may contribute to adverse remodeling.


Asunto(s)
Agammaglobulinemia/inmunología , Autoanticuerpos/inmunología , Insuficiencia Cardíaca/prevención & control , Cambio de Clase de Inmunoglobulina , Inmunoglobulina M/inmunología , Infarto del Miocardio/inmunología , Miocarditis/prevención & control , Miocardio/inmunología , Agammaglobulinemia/complicaciones , Agammaglobulinemia/genética , Agammaglobulinemia/metabolismo , Animales , Autoanticuerpos/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosis , Insuficiencia Cardíaca/inmunología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Cambio de Clase de Inmunoglobulina/genética , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocarditis/inmunología , Miocarditis/metabolismo , Miocarditis/patología , Miocardio/metabolismo , Miocardio/patología , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular
17.
Ann Neurol ; 82(5): 729-743, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29023958

RESUMEN

OBJECTIVE: Cardiac diseases are established risk factors for ischemic stroke incidence and severity. Conversely, there is increasing evidence that brain ischemia can cause cardiac dysfunction. The mechanisms underlying this neurogenic heart disease are incompletely understood. Although it is established that ischemic stroke is associated with cardiac arrhythmias, myocardial damage, elevated cardiac enzymes, and plasma catecholamines in the acute phase, nothing is known about the delayed consequences of ischemic stroke on cardiovascular function. METHODS: To determine the long-term cardiac consequences of a focal cerebral ischemia, we subjected young and aged mice to a 30-minute transient middle cerebral artery occlusion and analyzed cardiac function by serial transthoracic echocardiography and hemodynamic measurements up to week 8 after surgery. Finally, animals were treated with metoprolol to evaluate a pharmacologic treatment option to prevent the development of heart failure. RESULTS: Focal cerebral ischemia induced a long-term cardiac dysfunction with a reduction in left ventricular ejection fraction and an increase in left ventricular volumes; this development was associated with higher peripheral sympathetic activity. Metoprolol treatment prevented the development of chronic cardiac dysfunction by decelerating extracellular cardiac remodeling and inhibiting sympathetic signaling relevant to chronic autonomic dysfunction. INTERPRETATION: Focal cerebral ischemia in mice leads to the development of chronic systolic dysfunction driven by increased sympathetic activity. If these results can be confirmed in a clinical setting, treating physicians should be attentive to clinical signs of heart failure in every patient after ischemic stroke. Therapeutically, the successful ß-blockade with metoprolol in mice could also have future clinical implications. Ann Neurol 2017;82:729-743.


Asunto(s)
Isquemia Encefálica/fisiopatología , Hemodinámica/fisiología , Accidente Cerebrovascular/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Antagonistas de Receptores Adrenérgicos beta 1/uso terapéutico , Animales , Isquemia Encefálica/sangre , Isquemia Encefálica/complicaciones , Ecocardiografía , Epinefrina/sangre , Insuficiencia Cardíaca/prevención & control , Hemodinámica/efectos de los fármacos , Hidrocortisona/sangre , Infarto de la Arteria Cerebral Media , Masculino , Metoprolol/uso terapéutico , Ratones , Péptido Natriurético Encefálico/sangre , Norepinefrina/sangre , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/complicaciones , Sistema Nervioso Simpático/efectos de los fármacos
18.
Opt Express ; 26(20): 26134-26144, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30469704

RESUMEN

Porous phosphor microstructures are studied for their potential as light converter in laser-based, high-resolution lighting systems. Phosphor particles are filled into pre-patterned silicon molds and coated by an atomic layer deposition with a thin layer of Al2O3 for mechanical stability. Pixel sizes of 2 mm by 2 mm down to 25 µm by 25 µm are fabricated. The structures show a significant drop in luminance between the illuminated and the non-illuminated, adjacent pixel. The high thermal conductivity of the silicon allows an efficient cooling of the structures. Having removed the backside silicon, an active air flow cooling of the porous phosphor structure is possible.

19.
Circ Res ; 119(2): 237-48, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27142162

RESUMEN

RATIONALE: In patients after acute myocardial infarction (AMI), the initial extent of necrosis and inflammation determine clinical outcome. One early event in AMI is the increased cardiac expression of atrial natriuretic peptide (NP) and B-type NP, with their plasma levels correlating with severity of ischemia. It was shown that NPs, via their cGMP-forming guanylyl cyclase-A (GC-A) receptor and cGMP-dependent kinase I (cGKI), strengthen systemic endothelial barrier properties in acute inflammation. OBJECTIVE: We studied whether endothelial actions of local NPs modulate myocardial injury and early inflammation after AMI. METHODS AND RESULTS: Necrosis and inflammation after experimental AMI were compared between control mice and littermates with endothelial-restricted inactivation of GC-A (knockout mice with endothelial GC-A deletion) or cGKI (knockout mice with endothelial cGKI deletion). Unexpectedly, myocardial infarct size and neutrophil infiltration/activity 2 days after AMI were attenuated in knockout mice with endothelial GC-A deletion and unaltered in knockout mice with endothelial cGKI deletion. Molecular studies revealed that hypoxia and tumor necrosis factor-α, conditions accompanying AMI, reduce the endothelial expression of cGKI and enhance cGMP-stimulated phosphodiesterase 2A (PDE2A) levels. Real-time cAMP measurements in endothelial microdomains using a novel fluorescence resonance energy transfer biosensor revealed that PDE2 mediates NP/cGMP-driven decreases of submembrane cAMP levels. Finally, intravital microscopy studies of the mouse cremaster microcirculation showed that tumor necrosis factor-α-induced endothelial NP/GC-A/cGMP/PDE2 signaling impairs endothelial barrier functions. CONCLUSIONS: Hypoxia and cytokines, such as tumor necrosis factor-α, modify the endothelial postreceptor signaling pathways of NPs, with downregulation of cGKI, induction of PDE2A, and altered cGMP/cAMP cross talk. Increased expression of PDE2 can mediate hyperpermeability effects of paracrine endothelial NP/GC-A/cGMP signaling and facilitate neutrophil extravasation during the early phase after MI.


Asunto(s)
Factor Natriurético Atrial/farmacología , Endotelio Vascular/metabolismo , Mediadores de Inflamación/metabolismo , Infarto del Miocardio/metabolismo , Animales , Factor Natriurético Atrial/biosíntesis , Endotelio Vascular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Factor de Necrosis Tumoral alfa/farmacología
20.
Circ Res ; 116(2): 354-67, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25593279

RESUMEN

A large body of evidence produced during decades of research indicates that myocardial injury activates innate immunity. On the one hand, innate immunity both aggravates ischemic injury and impedes remodeling after myocardial infarction (MI). On the other hand, innate immunity activation contributes to myocardial healing, as exemplified by monocytes' central role in the formation of a stable scar and protection against intraventricular thrombi after acute infarction. Although innate leukocytes can recognize a wide array of self-antigens via pattern recognition receptors, adaptive immunity activation requires highly specific cooperation between antigen-presenting cells and distinct antigen-specific receptors on lymphocytes. We have only recently begun to examine lymphocyte activation's relationship to adaptive immunity and significance in the context of ischemic myocardial injury. There is some experimental evidence that CD4(+) T-cells contribute to ischemia-reperfusion injury. Several studies have shown that CD4(+) T-cells, especially CD4(+) T-regulatory cells, improve wound healing after MI, whereas depleting B-cells is beneficial post MI. That T-cell activation after MI is induced by T-cell receptor signaling implicates autoantigens that have not yet been identified in this context. Also, the significance of lymphocytes in humans post MI remains unclear, primarily as a result of methodology. This review summarizes current experimental evidence of lymphocytes' activation, functional role, and crosstalk with innate leukocytes in myocardial ischemia-reperfusion injury, wound healing, and remodeling after myocardial infarction.


Asunto(s)
Linfocitos/fisiología , Infarto del Miocardio/inmunología , Daño por Reperfusión Miocárdica/inmunología , Remodelación Ventricular/inmunología , Cicatrización de Heridas/inmunología , Animales , Humanos , Inmunidad Innata/inmunología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda