Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cardiovasc Med ; 10: 1182752, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38094126

RESUMEN

Introduction: Despite increasing use of transcatheter approaches, endoscopic mitral valve surgery (MVS) remains an established option for treatment of mitral regurgitation (MR). Nevertheless, as perioperative risk increases with age, outcome of endoscopic MVS in elderly patients is uncertain. Methods: We retrospectively analyzed 756 consecutive patients with MR ≥2, who underwent minimally-invasive MVS at our institution between 2016 and 2022. Patients were stratified by age ≥75 (elderly-group; n = 91) or <75 years (control-group; n = 665). All patients received endoscopic MVS via right anterolateral minithoracotomy with non-rib spreading soft tissue retraction and 3D-camera visualization. Results: Overall surgical risk was increased in the elderly-group (median age of 77 (76-80) years vs. 58 (51-67) years, p < 0.001) with STS-PROM Scores of 1.9% vs. 0.4% (p < 0.001) and increased prevalence of hypertension, diabetes, coronary artery disease and atrial fibrillation (AFib). Elderly patients were also more symptomatic (NYHA class III 45.7% vs. 29.8%; p = 0.002). Axillo-femoral perfusion was more frequently used in the elderly-group (27.5% vs. 4.2%; p < 0.001). Cross-clamp and cardiopulmonary bypass times were similar. Rate of MV repair was 85.7% vs. 93.8% (p = 0.005). Closure of the left atrial appendage was more frequently performed in the elderly-group (45.1% vs. 23.9%; p < 0.001), whereas rate of concomitant tricuspid valve repair was similar (11.0% vs. 8.9%; p = 0.511). Postoperative complications including perioperative hemodialysis (3.3% vs. 2.9%; p = 0.739), low cardiac output (5.3% vs. 3.8%; p = 0.393), perioperative stroke (1.1% vs. 0.15%; p = 0.224) and myocardial infarction (0% vs. 0.15%) were favorably low in both groups. Acute mortality at 30 days was 2.2% vs. 0.4% (p = 0.112). Conclusion: Despite increased prevalence of outcome-relevant comorbidities and surgical risk, perioperative outcome of patients aged ≥75 years undergoing endoscopic MVS is favorable. Therefore, endoscopic MVS is a valuable therapeutic option for selected elderly patients and should be taken in consideration during routine heart-team discussion.

2.
Artículo en Inglés | MEDLINE | ID: mdl-37726023

RESUMEN

BACKGROUND: Alterations in left ventricular (LV) diastolic function following native tissue-preserving aortic valve (AV) procedures have not been systematically investigated. Furthermore, no comparisons have been made between these changes and those observed after prosthetic AV replacement. METHODS: From October 2017 to August 2020, 74 patients aged <65 years were referred to our institution for elective AV surgery. Preoperative and postoperative (i.e., discharge, 3-month and 1-year follow-up) transthoracic echocardiography was analyzed. RESULTS: Native tissue-preserving surgery was performed in 55 patients (AV repair: n = 42, Ross procedure: n = 13). The remaining 19 patients underwent prosthetic AV replacement. Preoperatively and at discharge, transvalvular hemodynamics and LV diastolic function were comparable in both groups. At 1-year follow-up, native valve (NV) patients showed significantly lower mean transvalvular gradient (7 ± 5 vs. 9 ± 3 mmHg, p = 0.046) and peak velocity (1.74 ± 0.51 vs. 2.26 ± 0.96 m/s, p = 0.004), and significantly better septal e' (9.1 ± 2.7 vs. 7.7 ± 2.5 cm/s, p = 0.043) and lateral e' (14.7 ± 3.1 vs. 11.7 ± 3.7 cm/s, p = 0.001). From preoperatively to 1-year postoperatively, septal and lateral e' and E/e' improved markedly after NV preservation (septal e': +0.7 cm/s, p = 0.075; lateral e': +2.3 cm/s, p < 0.001; E/e': -1.5, p = 0.001) but not after AV replacement (septal e': +0.2 cm/s, p = 0.809; lateral e': +0.8 cm/s, p = 0.574; E/e': -1.2, p = 0.347). Significant negative linear correlations between postoperative transvalvular gradients and absolute changes in lateral e' and E/e' were detected during follow-up. CONCLUSION: Preservation of native tissue in AV surgery results in superior transvalvular hemodynamics compared with prosthetic AV replacement. This may induce faster LV reverse remodeling and may explain more pronounced improvement in LV diastolic function.

3.
Innovations (Phila) ; 17(4): 317-323, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35983699

RESUMEN

Objective: Subannular mitral valve (MV) repair techniques have been developed to address increased rates of recurrent mitral regurgitation (MR) in patients with secondary MR (SMR) type IIIb. Endoscopic papillary muscle relocation (PMR) is feasible via minithoracotomy. Nevertheless, the periprocedural outcome of patients with severe left ventricular (LV) dysfunction remains unknown. Methods: A total of 98 consecutive patients with SMR type IIIb underwent PMR at our institution. Due to concomitant coronary artery bypass grafting, 62 patients underwent sternotomy and were excluded from the current analysis, whereas 36 patients were treated by a minimally invasive technique using 3-dimensional endoscopy. Of these, 18 patients had severely depressed LV ejection fraction (LVEF) ≤35% (study group) and were compared to the remaining 18 patients with LVEF >35% (control group). Periprocedural outcome was retrospectively analyzed. Results: Although LVEF was significantly worse in the study group (30% ± 4% vs 43% ± 6%, P < 0.001), the severity of SMR and the degree of MV leaflet tethering were similar. The prevalence of concomitant procedures and the duration of surgery, cardiopulmonary bypass, and aortic cross-clamp were comparable. Periprocedural low cardiac output syndrome was favorably low in both groups (16.7% vs 5.6%, P = 0.29). Postoperative ventilation time (5.7 h [4.2 to 8.7 h] vs 6.0 h [4.6 to 9.8 h], P = 0.43) and duration of intensive care unit stay (2 days [1 to 3 days] vs 2 days [1 to 3 days], P = 0.22) were similar. There was no 30-day mortality in either group. Conclusions: Standardized endoscopic PMR resulted in favorable periprocedural outcomes in patients with severe LV dysfunction, suggesting that minimally invasive surgery can safely be extended to this patient population.


Asunto(s)
Anuloplastia de la Válvula Mitral , Insuficiencia de la Válvula Mitral , Disfunción Ventricular Izquierda , Endoscopía , Humanos , Anuloplastia de la Válvula Mitral/métodos , Músculos Papilares/cirugía , Estudios Retrospectivos , Resultado del Tratamiento , Disfunción Ventricular Izquierda/complicaciones , Disfunción Ventricular Izquierda/cirugía
4.
Eur J Cardiothorac Surg ; 62(1)2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35511127

RESUMEN

OBJECTIVES: The major drawback of isolated annuloplasty for treatment of secondary mitral regurgitation (SMR) with restricted leaflet motion during systole (type IIIb) is the recurrence of SMR, leading to adverse clinical outcome. Additional papillary muscle relocation (PMR) specifically addresses leaflet tethering to restore mitral valve geometry. We aimed to compare the 2-year outcome of annuloplasty with additional PMR vs isolated annuloplasty. METHODS: A total of 105 consecutive type IIIb SMR patients with preoperative left ventricular (LV) ejection faction <45%, left ventricular end-diastolic diameter >55 mm and a tenting height >10 mm reached 2-year postoperative follow-up after mitral valve repair and were included in the current analysis. A total of 51 patients underwent annuloplasty and additional PMR (study group). A total of 54 patients underwent isolated annuloplasty (control group). Primary composite study end point comprised death or recurrence of mitral regurgitation ≥2 at 2 years postoperatively. RESULTS: Echocardiographic baseline variables indicating the severity of LV dysfunction and mitral leaflet tethering were similar. Procedural and periprocedural outcome was comparable in both groups. The primary composite end point was significantly improved in the study group (19.6% [10/51]) in comparison to the control group (44.4% [24/54]; P = 0.009). Two-year all-cause mortality was 7.8% [4/51] in the study group vs 18.5% [10/54] in the control group (P = 0.098). After 2 years, a significant improvement in New York Heart Association functional class as compared to the baseline values was observed in the study group. CONCLUSIONS: Additional PMR to treat SMR type IIIb resulted in an improved 2-year outcome in comparison to isolated annuloplasty. PMR specifically addressing mitral leaflet tethering represents a valid therapeutic option for heart failure patients with SMR type IIIb.


Asunto(s)
Anuloplastia de la Válvula Mitral , Insuficiencia de la Válvula Mitral , Ecocardiografía , Humanos , Válvula Mitral/diagnóstico por imagen , Válvula Mitral/cirugía , Anuloplastia de la Válvula Mitral/métodos , Músculos Papilares/cirugía , Resultado del Tratamiento
5.
Transpl Int ; 30(11): 1181-1189, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28672061

RESUMEN

Cardiac allograft vasculopathy (CAV) affects approximately 30% of cardiac transplant patients at 5 years post-transplantation. To date, there are few CAV treatment or prevention options, none of which are highly effective. The aim of the study was to investigate the effect of thalidomide on the development of CAV. The effect of thalidomide treatment on chronic rejection was assessed in rat orthotopic aortic transplants in allogeneic F344 or syngeneic Lew rats (n = 6 per group). Animals were left untreated or received thalidomide for 30 days post-transplant, and evidence of graft CAV was determined by histology (trichrome and immunohistochemistry) and intragraft cytokine measurements. Animals that received thalidomide treatment post-transplant showed markedly reduced luminal obliteration, with concomitant rescue of smooth muscle cells (SMCs) in the aortic media of grafts. Thalidomide counteracted neointimal hyperplasia by preventing dedifferentiation of vascular SMCs. Measurement of intragraft cytokine levels after thalidomide treatment revealed downregulation of matrix metalloproteinase 8 and monocyte chemotactic protein 1, cytokines involved in tissue remodelling and inflammation, respectively. Importantly, no negative side effects of thalidomide were observed. Thalidomide treatment prevents CAV development in a rodent model and is therefore potentially useful in clinical applications to prevent post-transplant heart rejection.


Asunto(s)
Aorta Torácica/trasplante , Enfermedad de la Arteria Coronaria/prevención & control , Rechazo de Injerto/prevención & control , Inmunosupresores/uso terapéutico , Talidomida/uso terapéutico , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Enfermedad Crónica , Enfermedad de la Arteria Coronaria/etiología , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Rechazo de Injerto/complicaciones , Inmunosupresores/farmacología , Linfocitos/efectos de los fármacos , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Talidomida/farmacología , Túnica Media/efectos de los fármacos
6.
Transpl Int ; 29(12): 1337-1348, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27614085

RESUMEN

Bronchiolitis obliterans syndrome (BOS) is a main cause of allograft dysfunction and mortality after lung transplantation (LTx). A better understanding of BOS pathogenesis is needed to overcome this treatment-refractory complication. Orthotopic tracheal transplantation using human bronchus was performed in Brown Norway (BN) and nude (RNU) rats. Allografts were recovered in both strains at Day 7 (BN7 , n = 6; RNU7 , n = 7) or Day 28 (BN28 , n = 6; RNU28 , n = 6). Immune response of the host against the bronchial graft was assessed. Human samples from BOS patients were used to compare with the histological features of the animal model. Obstruction of the allograft lumen associated with significant infiltration of CD3+ and CD68+ cells was observed in the BN group on Day 28. Immune response from type 1 T-helper cells against the tracheal xenograft was higher in BN animals compared to nude animals on Days 7 and 28 (P < 0.001 and P = 0.035). Xenoreactive antibodies were significantly higher at Day 7 (IgM) and Day 28 (IgG) in the BN group compared to RNU (respectively, 37.6 ± 6.5 vs. 5.8 ± 0.7 mean fluorescence, P = 0.039; and 22.4 ± 3.8 vs. 6.9 ± 1.6 mean fluorescence, P = 0.011). Immunocompetent animals showed a higher infiltration of S100A4+ cells inside the bronchial wall after 28 days, associated with cartilage damage ranging from invasion to complete destruction. In vitro expression of S100A4 by human fibroblasts was higher when stimulated by mononuclear cells (MNCs) from BN rats than from RNU (2.9 ± 0.1 vs. 2.4 ± 0.1 mean fluorescence intensity, P = 0.005). Similarly, S100A4 was highly expressed in response to human MNCs compared to stimulation by T-cell-depleted human MNCs (4.3 ± 0.2 vs. 2.7 ± 0.1 mean fluorescence intensity, P < 0.001). Obliterative bronchiolitis has been induced in a new xenotransplant model in which chronic airway obstruction was associated with immune activation against the xenograft. Cartilage infiltration by S100A4+ cells might be stimulated by T cells.


Asunto(s)
Bronquios/trasplante , Bronquiolitis Obliterante/etiología , Tráquea/trasplante , Trasplante Heterólogo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Complejo CD3/metabolismo , Modelos Animales de Enfermedad , Ensayo de Immunospot Ligado a Enzimas , Rechazo de Injerto/patología , Supervivencia de Injerto , Humanos , Sistema Inmunológico , Trasplante de Hígado , Periodo Posoperatorio , Distribución Aleatoria , Ratas , Ratas Endogámicas BN , Proteína de Unión al Calcio S100A4/metabolismo , Resultado del Tratamiento
7.
Transplantation ; 100(5): 1022-31, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26910327

RESUMEN

BACKGROUND: The efficacy of selective Janus kinase 1/3 inhibitor R507 to prevent obliterative airway disease was analyzed in preclinical airway transplantation models. METHODS: Orthotopic trachea transplantations were performed between Lewis donors and Brown Norway rat recipients. Oral everolimus (4 mg/kg once per day) or oral respective inhaled R507 (60 mg/kg twice per day, each) was used for immunosuppression. Grafts were retrieved after 6 or 60 days. Toxicity and anti-inflammatory effects of R507 were analyzed on human airway epithelial cells. RESULTS: In 6-day animals, oral and inhaled R507 more potently diminished mononuclear graft infiltration than everolimus and preserved ciliated pseudostratified columnar respiratory epithelium. Everolimus and R507 similarly suppressed systemic cellular and humoral immune activation. In untreated rats, marked obliterative airway disease developed over 60 days. Oral and inhaled R507 was significantly more effective in reducing airway obliteration and preserved the morphology of the airway epithelium. Luciferase-positive donors revealed that a substantial amount of smooth muscle cells within the obliterative tissue was of donor origin. Only everolimus but not R507, adversely altered kidney function and lipid profiles. The R507 aerosol did not show airway toxicity in vitro but effectively suppressed activation of inflammatory signaling pathways induced by IL-1ß. CONCLUSIONS: The Janus kinase 1/3 inhibitor R507 is a very well-tolerated immunosuppressant that similarly diminished obliterative airway disease with systemic or inhaled administration.


Asunto(s)
Bronquiolitis Obliterante/prevención & control , Inmunosupresores/administración & dosificación , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/administración & dosificación , Tráquea/trasplante , Administración por Inhalación , Administración Oral , Aerosoles/química , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Everolimus/administración & dosificación , Humanos , L-Lactato Deshidrogenasa/metabolismo , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Endogámicas Lew , Transducción de Señal , Resultado del Tratamiento
8.
Stem Cells Transl Med ; 4(6): 625-31, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25947338

RESUMEN

UNLABELLED: Different tissue-engineering approaches have been developed to induce and promote cardiac regeneration; however, the impact of the immune system and its responses to the various scaffold components of the engineered grafts remains unclear. Fibrin-based engineered heart tissue (EHT) was generated from neonatal Lewis (Lew) rat heart cells and transplanted onto the left ventricular surface of three different rat strains: syngeneic Lew, allogeneic Brown Norway, and immunodeficient Rowett Nude rats. Interferon spot frequency assay results showed similar degrees of systemic immune activation in the syngeneic and allogeneic groups, whereas no systemic immune response was detectable in the immunodeficient group (p < .001 vs. syngeneic and allogeneic). Histological analysis revealed much higher local infiltration of CD3- and CD68-positive cells in syngeneic and allogeneic rats than in immunodeficient animals. Enzyme-linked immunospot and immunofluorescence experiments revealed matrix-directed TH1-based rejection in syngeneic recipients without collateral impairment of heart cell survival. Bioluminescence imaging was used for in vivo longitudinal monitoring of transplanted luciferase-positive EHT constructs. Survival was documented in syngeneic and immunodeficient recipients for a period of up to 110 days after transplant, whereas in the allogeneic setting, graft survival was limited to only 14 ± 1 days. EHT strategies using autologous cells are promising approaches for cardiac repair applications. Although fibrin-based scaffold components elicited an immune response in our studies, syngeneic cells carried in the EHT were relatively unaffected. SIGNIFICANCE: An initial insight into immunological consequences after transplantation of engineered heart tissue was gained through this study. Most important, this study was able to demonstrate cell survival despite rejection of matrix components. Generation of syngeneic human engineered heart tissue, possibly using human induced pluripotent stem cell technology with subsequent directed rejection of matrix components, may be a potential future approach to replace diseased myocardium.


Asunto(s)
Fibrina/química , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Miocardio/inmunología , Células TH1/inmunología , Ingeniería de Tejidos , Aloinjertos , Animales , Fibrina/inmunología , Rechazo de Injerto/patología , Ratas , Andamios del Tejido , Trasplante Isogénico
9.
Cell Stem Cell ; 16(1): 33-8, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25465116

RESUMEN

The generation of pluripotent stem cells by somatic cell nuclear transfer (SCNT) has recently been achieved in human cells and sparked new interest in this technology. The authors reporting this methodical breakthrough speculated that SCNT would allow the creation of patient-matched embryonic stem cells, even in patients with hereditary mitochondrial diseases. However, herein we show that mismatched mitochondria in nuclear-transfer-derived embryonic stem cells (NT-ESCs) possess alloantigenicity and are subject to immune rejection. In a murine transplantation setup, we demonstrate that allogeneic mitochondria in NT-ESCs, which are nucleus-identical to the recipient, may trigger an adaptive alloimmune response that impairs the survival of NT-ESC grafts. The immune response is adaptive, directed against mitochondrial content, and amenable for tolerance induction. Mitochondrial alloantigenicity should therefore be considered when developing therapeutic SCNT-based strategies.


Asunto(s)
Células Madre Embrionarias/citología , Inmunidad , Mitocondrias/metabolismo , Técnicas de Transferencia Nuclear , Animales , Antígenos/inmunología , Células Madre Embrionarias/metabolismo , Humanos , Ratones Endogámicos BALB C , Trasplante Homólogo
10.
Am J Med Sci ; 349(1): 67-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25275340

RESUMEN

Cardiac c-kit+ cells isolated from cardiac explant-derived cells modestly improve cardiac functions after myocardial infarction; however, their full potential has not yet been realized. The present study was undertaken to determine the isolation and culture of c-kit+ cardiac stem cells (CSCs), and the roles of myocardial injection of CSCs on the survival of rat cardiac allograft. Recipient Sprague-Dawley rats were transplanted with hearts from Wistar rats. In the in vitro experiment, c-kit+ cells were isolated from mouse heart fragment culture by magnetic cell sorting. CSCs expressed of cardiomyocyte specific protein cardiac troponin I, α smooth muscle actin and von Willebrand factor in conditioned culture. CSC injection increased graft survival of cardiac allograft rats. The effects of CSCs on increase in graft survival of cardiac allograft rats were blocked by stromal-derived factor-1 (SDF-1) knockdown. The expression of SDF-1 was increased after CSC injection into the cardiac of cardiac allograft rats. These results indicate that CSC injection into the cardiac prolongs graft survival of cardiac allograft rats. SDF-1 plays an important role in the effects of CSCs on the graft survival of cardiac allograft rats.


Asunto(s)
Trasplante de Corazón , Trasplante de Células Madre , Animales , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Supervivencia de Injerto , Masculino , Miocardio/citología , Proteínas Proto-Oncogénicas c-kit , Ratas Sprague-Dawley , Ratas Wistar , Células Madre , Trasplante Homólogo
11.
J Vis Exp ; (87)2014 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-24893977

RESUMEN

Various in vivo laboratory rodent models for the induction of artery stenosis have been established to mimic diseases that include arterial plaque formation and stenosis, as observed for example in ischemic heart disease. Two highly reproducible mouse models - both resulting in artery stenosis but each underlying a different pathway of development - are introduced here. The models represent the two most common causes of artery stenosis; namely one mouse model for each myointimal hyperplasia, and atherosclerosis are shown. To induce myointimal hyperplasia, a balloon catheter injury of the abdominal aorta is performed. For the development of atherosclerotic plaque, the ApoE -/- mouse model in combination with western fatty diet is used. Different model-adapted options for the measurement and evaluation of the results are named and described in this manuscript. The introduction and comparison of these two models provides information for scientists to choose the appropriate artery stenosis model in accordance to the scientific question asked.


Asunto(s)
Aterosclerosis/patología , Modelos Animales de Enfermedad , Túnica Íntima/patología , Animales , Apolipoproteínas E/deficiencia , Femenino , Hiperplasia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Placa Aterosclerótica/patología , Reproducibilidad de los Resultados
12.
Nature ; 509(7502): 641-4, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24747400

RESUMEN

Despite the introduction of antiproliferative drug-eluting stents, coronary heart disease remains the leading cause of death in the United States. In-stent restenosis and bypass graft failure are characterized by excessive smooth muscle cell (SMC) proliferation and concomitant myointima formation with luminal obliteration. Here we show that during the development of myointimal hyperplasia in human arteries, SMCs show hyperpolarization of their mitochondrial membrane potential (ΔΨm) and acquire a temporary state with a high proliferative rate and resistance to apoptosis. Pyruvate dehydrogenase kinase isoform 2 (PDK2) was identified as a key regulatory protein, and its activation proved necessary for relevant myointima formation. Pharmacologic PDK2 blockade with dichloroacetate or lentiviral PDK2 knockdown prevented ΔΨm hyperpolarization, facilitated apoptosis and reduced myointima formation in injured human mammary and coronary arteries, rat aortas, rabbit iliac arteries and swine (pig) coronary arteries. In contrast to several commonly used antiproliferative drugs, dichloroacetate did not prevent vessel re-endothelialization. Targeting myointimal ΔΨm and alleviating apoptosis resistance is a novel strategy for the prevention of proliferative vascular diseases.


Asunto(s)
Aorta/lesiones , Arterias/lesiones , Constricción Patológica/prevención & control , Ácido Dicloroacético/farmacología , Ácido Dicloroacético/uso terapéutico , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Angioplastia de Balón/efectos adversos , Animales , Aorta/efectos de los fármacos , Aorta/patología , Apoptosis/efectos de los fármacos , Arterias/efectos de los fármacos , Arterias/patología , Proliferación Celular/efectos de los fármacos , Constricción Patológica/patología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/lesiones , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Hiperplasia/tratamiento farmacológico , Hiperplasia/patología , Arteria Ilíaca/efectos de los fármacos , Arteria Ilíaca/lesiones , Arteria Ilíaca/patología , Arterias Mamarias/efectos de los fármacos , Arterias Mamarias/lesiones , Arterias Mamarias/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Conejos , Ratas , Prevención Secundaria , Stents/efectos adversos , Porcinos , Túnica Íntima/lesiones
13.
Stem Cells Dev ; 22(19): 2619-29, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23711207

RESUMEN

Mesenchymal stromal cells (MSCs) have been isolated from many tissues, including gestational tissue. To date, a study comparing the properties and suitability of these cells in cell-based therapies is lacking. In this study, we compared the phenotype, proliferation rate, migration, immunogenicity, and immunomodulatory capabilities of human MSCs derived from umbilical cord lining (CL-MSCs), umbilical cord blood (CB-MSCs), placenta (P-MSCs), and Wharton's jelly (WJ-MSCs). Differences were noted in differentiation, proliferation, and migration, with CL-MSCs showing the highest proliferation and migration rates resulting in prolonged survival in immunodeficient mice. Moreover, CL-MSCs showed a prolongation in survival in xenogeneic BALB/c mice, which was attributed to their ability to dampen TH1 and TH2 responses. Weaker human cellular immune responses were detected against CL-MSCs and P-MSCs, which were correlated with their lower HLA I expression. Furthermore, HLA II was upregulated less substantially by CL-MSCs and CB-MSCs after IFN-γ stimulation. MSC types did not differ in indolamine 2,3-dioxygenase (IDO) expression after IFN-γ stimulation. Despite their lower IDO, HLA-G, and TGF-ß1 expression, only CL-MSCs were able to reduce the release of IFN-γ by lymphocytes in a mixed lymphocyte reaction. In summary, CL-MSCs showed the best characteristics for cell-based strategies, as they are hypo-immunogenic and show high proliferation and migration rates. In addition, these studies show for the first time that although immunomodulatory molecules HLA-G, HLA-E, and TGF-ß play an important role in MSC immune evasion, basal and induced HLA expression seems to be decisive in determining the immunogenicity of MSCs.


Asunto(s)
Sangre Fetal/citología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Placenta/citología , Cordón Umbilical/citología , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Antígenos HLA-G/biosíntesis , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase II/biosíntesis , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Interferón gamma/metabolismo , Prueba de Cultivo Mixto de Linfocitos , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos BALB C , Embarazo , Linfocitos T Colaboradores-Inductores/inmunología , Factor de Crecimiento Transformador beta1/biosíntesis , Trasplante Heterólogo , Regulación hacia Arriba , Antígenos HLA-E
14.
Transplantation ; 95(2): 285-92, 2013 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-23325003

RESUMEN

BACKGROUND: The calcium-activated potassium channel KCa3.1 is critically involved in T-cell activation as well as in the proliferation of smooth muscle cells and fibroblasts. We sought to investigate whether KCa3.1 contributes to the pathogenesis of obliterative airway disease (OAD) and whether knockout or pharmacologic blockade would prevent the development of OAD. METHODS: Tracheas from CBA donors were heterotopically transplanted into the omentum of C57Bl/6J wild-type or KCa3.1 mice. C57Bl/6J recipients were either left untreated or received the KCa3.1 blocker TRAM-34 (120 mg/kg/day). Histopathology and immunologic assays were performed on postoperative day 5 or 28. RESULTS: Subepithelial T-cell and macrophage infiltration on postoperative day 5, as seen in untreated allografts, was significantly reduced in the KCa3.1 and TRAM-34 groups. Also, systemic Th1 activation was significantly and Th2 mildly reduced by KCa3.1 knockout or blockade. After 28 days, luminal obliteration of tracheal allografts was reduced from 89%±21% in untreated recipients to 53%±26% (P=0.010) and 59%±33% (P=0.032) in KCa3.1 and TRAM-34-treated animals, respectively. The airway epithelium was mostly preserved in syngeneic grafts, mostly destroyed in the KCa3.1 and TRAM-34 groups, and absent in untreated allografts. Allografts triggered an antibody response in untreated recipients, which was significantly reduced in KCa3.1 animals. KCa3.1 was detected in T cells, airway epithelial cells, and myofibroblasts. TRAM-34 dose-dependently suppressed proliferation of wild-type C57B/6J splenocytes but did not show any effect on KCa3.1 splenocytes. CONCLUSIONS: Our findings suggest that KCa3.1 channels are involved in the pathogenesis of OAD and that KCa3.1 blockade holds promise to reduce OAD development.


Asunto(s)
Bronquiolitis Obliterante/prevención & control , Terapia Genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Pirazoles/farmacología , Tráquea/efectos de los fármacos , Animales , Bronquiolitis Obliterante/genética , Bronquiolitis Obliterante/inmunología , Bronquiolitis Obliterante/metabolismo , Bronquiolitis Obliterante/patología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Ensayo de Immunospot Ligado a Enzimas , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/deficiencia , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Isoanticuerpos/sangre , Activación de Linfocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/efectos de los fármacos , Células Th2/inmunología , Células Th2/metabolismo , Factores de Tiempo , Tráquea/inmunología , Tráquea/metabolismo , Tráquea/patología , Tráquea/trasplante
15.
Transplantation ; 94(7): 695-702, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22971540

RESUMEN

BACKGROUND: Selective inhibition of lymphocyte activation through abrogation of signal 3-cytokine transduction emerges as a new strategy for immunosuppression. This is the first report on the novel Janus kinase (JAK)1/3 inhibitors R507 and R545 for prevention of acute allograft rejection. METHODS: Pharmacokinetic and in vitro enzyme inhibition assays were performed to characterize the drugs. Heterotopic Brown Norway-Lewis heart transplantations were performed to study acute cardiac allograft rejection, graft survival, suppression of cellular host responsiveness, and antibody production. Therapeutic and subtherapeutic doses of R507 (60 and 15 mg/kg 2 times per day) and R545 (20 and 5 mg/kg 2 times per day) were compared with those of tacrolimus (Tac; 4 and 1 mg/kg once per day). RESULTS: Plasma levels of R507 and R545 were sustained high for several hours. Cell-based enzyme assays showed selective inhibition of JAK1/3-dependent pathways with 20-fold or greater selectivity over JAK2 and Tyrosine kinase 2 kinases. After heart transplantation, both JAK1/3 inhibitors reduced early mononuclear graft infiltration, even significantly more potent than Tac. Intragraft interferon-γ release was significantly reduced by R507 and R545, and for interleukin-10 suppression, they were even significantly more potent than Tac. Both JAK1/3 inhibitors and Tac were similarly effective in reducing the host Th1 and Th2, but not Th17, responsiveness and similarly prevented donor-specific immunoglobulin M antibody production. Subtherapeutic and therapeutic R507 and R545 doses prolonged the mean graft survival and were similarly effective as 1 and 4 mg/kg Tac, respectively. In combination regimens, however, only R507 showed highly beneficial synergistic drug interactions with Tac. CONCLUSIONS: Both R507 and R545 are potent novel immunosuppressants with favorable pharmacokinetics and high JAK1/3 selectivity, but only R507 synergistically interacts with Tac.


Asunto(s)
Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón/inmunología , Inmunosupresores/farmacología , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 3/antagonistas & inhibidores , Miocardio/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Células Cultivadas , Técnicas de Cocultivo , Esquema de Medicación , Sinergismo Farmacológico , Quimioterapia Combinada , Ensayo de Inmunoadsorción Enzimática , Rechazo de Injerto/enzimología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Trasplante de Corazón/efectos adversos , Inmunoglobulina M/sangre , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacocinética , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Janus Quinasa 1/metabolismo , Janus Quinasa 3/metabolismo , Activación de Linfocitos/efectos de los fármacos , Prueba de Cultivo Mixto de Linfocitos , Masculino , Miocardio/inmunología , Miocardio/patología , Fosforilación , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacocinética , Ratas , Ratas Endogámicas BN , Ratas Endogámicas Lew , Factor de Transcripción STAT3/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Tacrolimus/farmacología
16.
J Vis Exp ; (63): e3663, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22617624

RESUMEN

Preclinical in vivo research models to investigate pathobiological and pathophysiological processes in the development of intimal hyperplasia after vessel stenting are crucial for translational approaches (1,2). The commonly used animal models include mice, rats, rabbits, and pigs (3-5). However, the translation of these models into clinical settings remains difficult, since those biological processes are already studied in animal vessels but never performed before in human research models (6,7). In this video we demonstrate a new humanized model to overcome this translational gap. The shown procedure is reproducible, easy, and fast to perform and is suitable to study the development of intimal hyperplasia and the applicability of diverse stents. This video shows how to perform the stent technique in human vessels followed by transplantation into immunodeficient rats, and identifies the origin of proliferating cells as human.


Asunto(s)
Oclusión de Injerto Vascular/etiología , Arterias Mamarias/trasplante , Stents , Injerto Vascular/métodos , Animales , Procesos de Crecimiento Celular/fisiología , Oclusión de Injerto Vascular/patología , Humanos , Arterias Mamarias/citología , Ratas , Ratas Desnudas , Trasplante Heterólogo
17.
Circ Res ; 110(10): 1303-10, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22427341

RESUMEN

RATIONALE: The transcription factor Islet-1 is a marker of cardiovascular progenitors during embryogenesis. The isolation of Islet-1-positive (Islet-1(+)) cells from early postnatal hearts suggested that Islet-1 also marks cardiac progenitors in adult life. OBJECTIVE: We investigated the distribution and identity of Islet-1(+) cells in adult murine heart and evaluated whether their number or distribution change with age or after myocardial infarction. METHODS AND RESULTS: Distribution of Islet-1(+) cells in adult heart was investigated using gene targeted mice with nuclear ß-galactosidase inserted into the Islet-1 locus. nLacZ-positive cells were only present in 3 regions of the adult heart: clusters in the interatrial septum and around the pulmonary veins, scattered within the wall of the great vessels, and a strictly delimited cluster between the right atrium and superior vena cava. Islet-1(+) cells in the first type of clusters coexpressed markers for parasympathetic neurons. Positive cells in the great arteries coexpressed smooth muscle actin and myosin heavy chain, indicating a smooth muscle cell identity. Very few Islet-1(+) cells within the outflow tract expressed the cardiomyocyte marker α-actinin. Islet-1(+) cells in the right atrium coexpressed the sinoatrial node pacemaker cell marker HCN4. Cell number and localization remained unchanged between 1 to 18 months of age. Consistently Islet-1 mRNA was detected in human sinoatrial node. Islet-1(+) cells could not be detected in the infarct zone 2 to 28 days after myocardial infarction, aside from 10 questionable cells in 1/13 hearts. CONCLUSIONS: Our results identify Islet-1 as a novel marker of the adult sinoatrial node and do not provide evidence for Islet-1(+) cells to serve as cardiac progenitors.


Asunto(s)
Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Nodo Sinoatrial/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Aorta/citología , Aorta/metabolismo , Biomarcadores/metabolismo , Compuestos Cromogénicos , Galactósidos , Indoles , Operón Lac , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/patología , Miocardio/citología , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , ARN Mensajero/metabolismo , Nodo Sinoatrial/citología
18.
J Cell Sci ; 124(Pt 17): 3029-37, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21878509

RESUMEN

Human embryonic stem cells (hESCs) can serve as a universal cell source for emerging cell or tissue replacement strategies, but immune rejection of hESC derivatives remains an unsolved problem. Here, we sought to describe the mechanisms of rejection for naïve hESCs and upon HLA class I (HLA I) knockdown (hESC(KD)). hESCs were HLA I-positive but negative for HLA II and co-stimulatory molecules. Transplantation of naïve hESC into immunocompetent Balb/c mice induced substantial T helper cell 1 and 2 (Th1 and Th2) responses with rapid cell death, but hESCs survived in immunodeficient SCID-beige recipients. Histology revealed mainly macrophages and T cells, but only scattered natural killer (NK) cells. A surge of hESC-specific antibodies against hESC class I, but not class II antigens, was observed. Using HLA I RNA interference and intrabody technology, HLA I surface expression of hESC(KD) was 88%-99% reduced. T cell activation after hESC(KD) transplantation into Balb/c was significantly diminished, antibody production was substantially alleviated, the levels of graft-infiltrating immune cells were reduced and the survival of hESC(KD) was prolonged. Because of their very low expression of stimulatory NK ligands, NK-susceptibility of naïve hESCs and hESC(KD) was negligible. Thus, HLA I recognition by T cells seems to be the primary mechanism of hESC recognition, and T cells, macrophages and hESC-specific antibodies participate in hESC killing.


Asunto(s)
Células Madre Embrionarias/inmunología , Células Madre Embrionarias/trasplante , Rechazo de Injerto/inmunología , Antígenos HLA/genética , Antígenos HLA/inmunología , Animales , Células Madre Embrionarias/citología , Técnicas de Silenciamiento del Gen/métodos , Rechazo de Injerto/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Trasplante Heterólogo
19.
Circulation ; 124(11 Suppl): S3-9, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21911816

RESUMEN

BACKGROUND: Although human embryonic stem cells (hESC) have enormous potential for cell replacement therapy of heart failure, immune rejection of hESC derivatives inevitably would occur after transplantation. We therefore aimed to generate a hypoantigeneic hESC line with improved survival characteristics. METHODS AND RESULTS: Using various in vivo, nonischemic, hindlimb xenotransplant models (immunocompetent and defined immunodefective mouse strains) as well as human in vitro T-cell and natural killer (NK)-cell assays, we revealed a central role for T cells in mediating hESC rejection. The NK-cell susceptibility of hESC in vivo was found to be low, and the NK response to hESC challenge in vitro was negligible. To reduce the antigenicity of hESC, we successfully generated human leukocyte antigen (HLA) I knockdown cells (hESC(siRNA+IB)) using both HLA I RNA interference (siRNA) and intrabody (IB) technology. HLA I expression was ≈99% reduced after 7 days and remained low for weeks. Cellular immune recognition of these hESC(siRNA+IB) was strongly reduced in both xenogeneic and allogeneic settings. Immune rejection was profoundly mitigated after hESC(siRNA+IB) transplantation into immunocompetent mice, and even long-term graft survival was achieved in one third of the animals without any immunosuppression. The survival benefit of hESC(siRNA+IB) was further confirmed under ischemic conditions in a left anterior descending coronary artery ligation model. CONCLUSIONS: HLA I knockdown hESC(siRNA+IB) provoke T-cell ignorance and experience largely mitigated xenogeneic rejection. By generating hypoantigeneic hESC lines, the generation of acceptable hESC derivatives may become a practical concept and push cell replacement strategies forward.


Asunto(s)
Células Madre Embrionarias/inmunología , Técnicas de Silenciamiento del Gen , Supervivencia de Injerto/inmunología , Antígenos HLA/genética , Tolerancia Inmunológica/inmunología , Trasplante de Células Madre , Trasplante Heterólogo/inmunología , Animales , Supervivencia Celular/inmunología , Células Cultivadas , Células Madre Embrionarias/citología , Humanos , Terapia de Inmunosupresión , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Modelos Animales , Linfocitos T/inmunología , Factores de Tiempo
20.
Circulation ; 122(11 Suppl): S170-8, 2010 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-20837910

RESUMEN

BACKGROUND: ε protein kinase C (εPKC) is involved in vascular smooth muscle cell (VSMC) activation, but little is known about its function in vascular pathology. We aimed at assessing the role of εPKC in the development of restenosis. METHODS AND RESULTS: Rat models of aortic balloon injury with or without subsequent stenting were used. Rats were treated with the selective ψεPKC activator ε receptor for activated protein kinase C (ψεRACK), the selective εPKC inhibitor εV1-2, or saline. Both down-stream cascades of the platelet-derived growth factor receptor via extracellular signal-regulated kinase and Akt, respectively, were evaluated in vivo and in VSMC cultures. Intimal hyperplasia with luminal obliteration developed in saline-treated balloon-injured rat aortas (20.3±8.0%), and ψεRACK significantly promoted neointima development (32.4±4.9%, P=0.033), whereas εV1-2 significantly inhibited luminal narrowing (9.2±4.3%, P=0.039). εPKC inhibition led to significantly reduced VSMC extracellular signal-regulated kinase phosphorylation in vivo, whereas Akt phosphorylation was not markedly affected. Neointimal proliferation in vivo and platelet-derived growth factor-induced VSMC proliferation/migration in vitro were significantly inhibited by εV1-2. The inhibition of the platelet-derived growth factor pathway was mediated by inhibiting down-stream extracellular signal-regulated kinase and Akt phosphorylation. In vitro, εV1-2 showed inhibitory properties on endothelial cell proliferation, but that did not prevent reendothelialization in vivo. εV1-2 showed proapoptotic effects on VSMC in vitro. After stent implantation, luminal restenosis (quantified by optical coherence tomography imaging) was significantly reduced with εV1-2 (8.0±2.0%) compared with saline (20.2±9.8%, P=0.028). CONCLUSIONS: εPKC seems to be centrally involved in the development of neointimal hyperplasia. We suggest that εPKC inhibition may be mediated via inhibition of extracellular signal-regulated kinase and Akt activation. εPKC modulation may become a new therapeutic target against vascular restenosis.


Asunto(s)
Aorta , Células Endoteliales/enzimología , Oclusión de Injerto Vascular , Miocitos del Músculo Liso/enzimología , Proteína Quinasa C-epsilon , Inhibidores de Proteínas Quinasas/farmacología , Animales , Aorta/enzimología , Aorta/lesiones , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Oclusión de Injerto Vascular/enzimología , Humanos , Masculino , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...