Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Sci Rep ; 13(1): 19322, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37935729

RESUMEN

The immune response in patients with Coronavirus Disease 2019 (COVID-19) is highly variable and is linked to disease severity and mortality. However, antibody and cytokine responses in the early disease stage and their association with disease course and outcome are still not completely understood. In this large, multi-centre cohort study, blood samples of 434 Belgian COVID-19 hospitalized patients with different disease severities (ranging from asymptomatic/mild to critically ill) from the first wave of the COVID-19 pandemic were obtained. Baseline antibody and cytokine responses were characterized and associations with several clinical outcome parameters were determined. Anti-spike immunoglobulin (Ig)G and IgM levels were elevated in patients with a more severe disease course. This increased baseline antibody response however was associated with decreased odds for hospital mortality. Levels of the pro-inflammatory cytokines IL-6, IP-10 and IL-8, the anti-inflammatory cytokine IL-10 and the antiviral cytokines IFN-α, IFN-ß and IFN-λ1 were increased with disease severity. Remarkably, we found significantly lower levels of IFN-λ2,3 in critically ill patients compared to patients of the moderate and severe disease category. Finally, levels of IL-8, IL-6, IP-10, IL-10, IFN-α, IFN-ß, IFN-γ and IFN-λ1 at baseline were positively associated with mortality, whereas higher IFN-λ2,3 levels were negatively associated with mortality.


Asunto(s)
COVID-19 , Humanos , Interleucina-10 , Interleucina-6 , Quimiocina CXCL10 , Interleucina-8 , Pandemias , Enfermedad Crítica , Bélgica/epidemiología , Estudios de Cohortes , Citocinas , Interferón-alfa , Inmunoglobulina G
2.
Cancers (Basel) ; 14(4)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35205782

RESUMEN

Multiple myeloma (MM), or Kahler's disease, is an incurable plasma cell (PC) cancer in the bone marrow (BM). This malignancy is preceded by one or more asymptomatic precursor conditions, monoclonal gammopathy of undetermined significance (MGUS) and/or smoldering multiple myeloma (SMM). The molecular mechanisms and exact cause of this progression are still not completely understood. In this study, the mutational profile underlying the progression from low-intermediate risk myeloma precursor conditions to MM was studied in serial BM smears. A custom capture-based sequencing platform was developed, including 81 myeloma-related genes. The clonal evolution of single nucleotide variants and short insertions and deletions was studied in serial BM smears from 21 progressed precursor patients with a median time of progression of six years. From the 21 patients, four patients had no variation in one of the 81 studied genes. Interestingly, in 16 of the 17 other patients, at least one variant present in MM was also detected in its precursor BM, even years before progression. Here, the variants were present in the pre-stage at a median of 62 months before progression to MM. Studying these paired BM samples contributes to the knowledge of the evolutionary genetic landscape and provides additional insight into the mutational behavior of mutant clones over time throughout progression.

3.
Int J Mol Sci ; 22(17)2021 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-34502175

RESUMEN

Myocardial infarction (MI) occurs when the coronary blood supply is interrupted. As a consequence, cardiomyocytes are irreversibly damaged and lost. Unfortunately, current therapies for MI are unable to prevent progression towards heart failure. As the renewal rate of cardiomyocytes is minimal, the optimal treatment should achieve effective cardiac regeneration, possibly with stem cells transplantation. In that context, our research group identified the cardiac atrial appendage stem cells (CASCs) as a new cellular therapy. However, CASCs are transplanted into a hostile environment, with elevated levels of advanced glycation end products (AGEs), which may affect their regenerative potential. In this study, we hypothesize that pyridoxamine (PM), a vitamin B6 derivative, could further enhance the regenerative capacities of CASCs transplanted after MI by reducing AGEs' formation. Methods and Results: MI was induced in rats by ligation of the left anterior descending artery. Animals were assigned to either no therapy (MI), CASCs transplantation (MI + CASCs), or CASCs transplantation supplemented with PM treatment (MI + CASCs + PM). Four weeks post-surgery, global cardiac function and infarct size were improved upon CASCs transplantation. Interstitial collagen deposition, evaluated on cryosections, was decreased in the MI animals transplanted with CASCs. Contractile properties of resident left ventricular cardiomyocytes were assessed by unloaded cell shortening. CASCs transplantation prevented cardiomyocyte shortening deterioration. Even if PM significantly reduced cardiac levels of AGEs, cardiac outcome was not further improved. Conclusion: Limiting AGEs' formation with PM during an ischemic injury in vivo did not further enhance the improved cardiac phenotype obtained with CASCs transplantation. Whether AGEs play an important deleterious role in the setting of stem cell therapy after MI warrants further examination.


Asunto(s)
Apéndice Atrial/citología , Infarto del Miocardio/terapia , Piridoxamina/uso terapéutico , Trasplante de Células Madre , Animales , Terapia Combinada , Femenino , Infarto del Miocardio/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley
4.
J Clin Med ; 10(13)2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34279448

RESUMEN

BACKGROUND: During myocardial infarction (MI), billions of cardiomyocytes are lost. The optimal therapy should effectively replace damaged cardiomyocytes, possibly with stem cells able to engraft and differentiate into adult functional cardiomyocytes. As such, cardiac atrial appendage stem cells (CASCs) are suitable candidates. However, the presence of elevated levels of advanced glycation end products (AGEs) in cardiac regions where CASCs are transplanted may affect their regenerative potential. In this study, we examine whether and how AGEs alter CASCs properties in vitro. METHODS AND RESULTS: CASCs in culture were exposed to ranging AGEs concentrations (50 µg/mL to 400 µg/mL). CASCs survival, proliferation, and migration capacity were significantly decreased after 72 h of AGEs exposure. Apoptosis significantly increased with rising AGEs concentration. The harmful effects of these AGEs were partially blunted by pre-incubation with a receptor for AGEs (RAGE) inhibitor (25 µM FPS-ZM1), indicating the involvement of RAGE in the observed negative effects. CONCLUSION: AGEs have a time- and concentration-dependent negative effect on CASCs survival, proliferation, migration, and apoptosis in vitro, partially mediated through RAGE activation. Whether anti-AGEs therapies are an effective treatment in the setting of stem cell therapy after MI warrants further examination.

5.
Arch Toxicol ; 95(7): 2235-2253, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34027561

RESUMEN

Metabolic-associated fatty liver disease (MAFLD) is a chronic liver disease that affects about a quarter of the world population. MAFLD encompasses different disease stadia ranging from isolated liver steatosis to non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis and hepatocellular carcinoma. Although MAFLD is considered as the hepatic manifestation of the metabolic syndrome, multiple concomitant disease-potentiating factors can accelerate disease progression. Among these risk factors are diet, lifestyle, genetic traits, intake of steatogenic drugs, male gender and particular infections. Although infections often outweigh the development of fatty liver disease, pre-existing MAFLD could be triggered to progress towards more severe disease stadia. These combined disease cases might be underreported because of the high prevalence of both MAFLD and infectious diseases that can promote or exacerbate fatty liver disease development. In this review, we portray the molecular and cellular mechanisms by which the most relevant viral, bacterial and parasitic infections influence the progression of fatty liver disease and steatohepatitis. We focus in particular on how infectious diseases, including coronavirus disease-19, hepatitis C, acquired immunodeficiency syndrome, peptic ulcer and periodontitis, exacerbate MAFLD. We specifically underscore the synergistic effects of these infections with other MAFLD-promoting factors.


Asunto(s)
Infecciones Bacterianas/complicaciones , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedades Parasitarias/complicaciones , Brote de los Síntomas , Virosis/complicaciones , Síndrome de Inmunodeficiencia Adquirida/complicaciones , Infecciones Bacterianas/microbiología , COVID-19/complicaciones , Hepatitis Viral Humana/complicaciones , Humanos , Hígado/fisiopatología , Síndrome Metabólico , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/parasitología , Enfermedad del Hígado Graso no Alcohólico/virología , Enfermedades Parasitarias/parasitología , Úlcera Péptica , Periodontitis , Factores de Riesgo , Virosis/virología
6.
Nat Commun ; 12(1): 1861, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33767199

RESUMEN

Multiple myeloma (MM) is consistently preceded by precursor conditions recognized clinically as monoclonal gammopathy of undetermined significance (MGUS) or smoldering myeloma (SMM). We interrogate the whole genome sequence (WGS) profile of 18 MGUS and compare them with those from 14 SMMs and 80 MMs. We show that cases with a non-progressing, clinically stable myeloma precursor condition (n = 15) are characterized by later initiation in the patient's life and by the absence of myeloma defining genomic events including: chromothripsis, templated insertions, mutations in driver genes, aneuploidy, and canonical APOBEC mutational activity. This data provides evidence that WGS can be used to recognize two biologically and clinically distinct myeloma precursor entities that are either progressive or stable.


Asunto(s)
Genoma Humano/genética , Gammopatía Monoclonal de Relevancia Indeterminada/genética , Mieloma Múltiple/genética , Mieloma Múltiple Quiescente/genética , Variaciones en el Número de Copia de ADN/genética , Progresión de la Enfermedad , Humanos , Gammopatía Monoclonal de Relevancia Indeterminada/patología , Mieloma Múltiple/patología , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Mieloma Múltiple Quiescente/patología , Secuenciación Completa del Genoma
7.
Int J Mol Sci ; 21(11)2020 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-32486259

RESUMEN

Human cardiac stem cells isolated from atrial appendages based on aldehyde dehydrogenase activity (CASCs) can be expanded in vitro and differentiate into mature cardiomyocytes. In this study, we assess whether Wnt activation stimulates human CASC proliferation, whereas Wnt inhibition induces cardiac maturation. CASCs were cultured as described before. Conventional PCR confirmed the presence of the Frizzled receptors. Small-molecule inhibitors (IWP2, C59, XAV939, and IWR1-endo) and activator (CHIR99021) of the Wnt/ß -catenin signaling pathway were applied, and the effect on ß-catenin and target genes for proliferation and differentiation was assessed by Western blot and RT-qPCR. CASCs express multiple early cardiac differentiation markers and are committed toward myocardial differentiation. They express several Frizzled receptors, suggesting a role for Wnt signaling in clonogenicity, proliferation, and differentiation. Wnt activation increases total and active ß-catenin levels. However, this does not affect CASC proliferation or clonogenicity. Wnt inhibition upregulated early cardiac markers but could not induce mature myocardial differentiation. When CASCs are committed toward myocardial differentiation, the Wnt pathway is active and can be modulated. However, despite its role in cardiogenesis and myocardial differentiation of pluripotent stem-cell populations, our data indicate that Wnt signaling has limited effects on CASC clonogenicity, proliferation, and differentiation.


Asunto(s)
Apéndice Atrial/citología , Diferenciación Celular , Regulación de la Expresión Génica , Miocitos Cardíacos/citología , Células Madre/citología , Vía de Señalización Wnt , Anciano , Anciano de 80 o más Años , Animales , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Femenino , Corazón/fisiología , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Porcinos
8.
Acta Cardiol ; 75(5): 406-410, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31057075

RESUMEN

Background: Coronary artery bypass graft (CABG) surgery is known to induce significant muscle wasting. It remains to be investigated whether muscle wasting after CABG surgery relates to a worse clinical status at entry of rehabilitation and exercise-based rehabilitation remediates such muscle wasting.Design: Prospective observational study.Methods: In 21 males, changes in lean tissue mass (LTM) after CABG surgery were assessed and during a 12-week endurance exercise-based rehabilitation intervention. Changes in blood parameters and cardiopulmonary exercise capacity were assessed, and relations with changes in LTM were analysed.Results: LTM decreased by -1.9 ± 2.5 kg (p < .05) within 3 weeks after CABG surgery: greater LTM loss related to a lower ventilatory threshold at entry of rehabilitation (r = 0.58-0.61, p < .05). LTM was fully restored (+2.1 ± 2.4 kg, p < .05) during rehabilitation.Conclusion: In males, CABG-induced LTM reduction was associated with a worse aerobic exercise tolerance at entry of rehabilitation, but this LTM reduction was fully remediated by endurance exercise-based rehabilitation.


Asunto(s)
Puente de Arteria Coronaria/rehabilitación , Enfermedad de la Arteria Coronaria/cirugía , Entrenamiento Aeróbico/métodos , Atrofia Muscular , Complicaciones Posoperatorias , Absorciometría de Fotón/métodos , Ejercicio Físico/fisiología , Prueba de Esfuerzo/métodos , Tolerancia al Ejercicio , Humanos , Masculino , Persona de Mediana Edad , Atrofia Muscular/diagnóstico , Atrofia Muscular/etiología , Atrofia Muscular/fisiopatología , Atrofia Muscular/terapia , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/fisiopatología , Complicaciones Posoperatorias/terapia , Resultado del Tratamiento
9.
Front Med (Lausanne) ; 6: 224, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31750305

RESUMEN

Irreproducibility of research results is one of the major contributing factors to the failure of translating basic research results into tangible bedside progress. To address this, the University Biobank Limburg (UBiLim) was founded by a collaboration between Hasselt University, the Hospital East-Limburg, and the Jessa Hospital. This paper describes the evolution of this process and the barriers encountered on the way. UBiLim evolved from an archival collection over a single-site biobank into a federated structure, supporting translational research at the founding institutions. Currently, UBiLim is a federated biobank, with an established organizational structure and processing, and storage facilities at each of the three sites. All activities are integrated in an ISO15189-accredited Quality Management System and based on (inter)national biobank guidelines. Common methods for processing and storage of a plethora of sample types, suitable for state-of-the-art applications, were validated and implemented. Because the biobank is embedded in two hospitals, the request of researchers to include certain sample types or enroll specific patient groups can quickly be met. Funding has been a major challenge in each step of its evolution and remains the biggest issue for long-term biobank sustainability. To a lesser extent, the Belgian legislation and the operational cost of information management system are also concerns for smooth biobank operations. Nonetheless, UBiLim serves as a facilitator and accelerator for translational research in the Limburg area of Belgium that, given the fields of research, may have an impact on international patient care.

10.
Biopreserv Biobank ; 17(4): 274-281, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30412415

RESUMEN

Biobanking is increasingly important in studying complex heterogeneous diseases. Therefore, it is essential to ensure the sample quality after long-term storage for reliable downstream analyses. The Clinical Biobank of the Jessa Hospital and the University Biobank Limburg (UBiLim) hold a continuously growing collection of hematological samples, including May-Grünwald-Giemsa (MGG)- and Perls' Prussian Blue (PPB)-stained bone marrow (BM) smears, stored at room temperature (RT) for up to 20 years. In this study, we investigated the effect of short- and long-term storage on the quality of DNA and RNA extracted from these BM smears to assess their fitness-for-purpose in downstream molecular applications, including agarose gel electrophoresis, bio-analyzer analysis, quantitative polymerase chain reaction (qPCR), and targeted next-generation sequencing (NGS). The RNA quality was very low for all samples, independent of storage time or staining method. The DNA from PPB-stained BM smears was already degraded after 1 year of storage and correspondingly could not be used for reliable downstream molecular analysis. In contrast, DNA extracted from MGG-stained BM smears stored for up to 10 years was able to generate high-quality data in qPCR and targeted NGS analyses. Longer storage periods (>15 years) of these samples revealed a high degree of degradation and a significant amount of DNA transitions and transversions. In conclusion, the DNA extracted from archival MGG-stained BM smears with a storage time up to at least 10 years was qualitatively good and fit for downstream analysis, including targeted NGS. This indicates that these samples are an eligible source for molecular DNA research and for studying complex diseases.


Asunto(s)
Bancos de Muestras Biológicas , Médula Ósea/metabolismo , Eosina Amarillenta-(YS)/metabolismo , Azul de Metileno/metabolismo , ADN/metabolismo , Humanos , Control de Calidad , ARN/metabolismo
11.
Clin Lymphoma Myeloma Leuk ; 18(4): 235-248, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29506935

RESUMEN

Multiple myeloma (MM), characterized by malignant plasma cells in the bone marrow, is consistently preceded by asymptomatic premalignant stage monoclonal gammopathy of undetermined significance (MGUS). These MGUS patients have an annual risk of 1% to progress to MM. Clinical, imaging, and genomic (genetic and epigenetic) factors were identified, whose presence increased the risk of progression from MGUS to MM. In this systematic review we summarize the currently identified clinical, imaging, and genomic biomarkers suggested to increase the progression risk or shown to be differentially expressed/present between both cohorts of patients. Despite the wide range of proposed markers, there are still no reliable biomarkers to individually predict which MGUS patient will progress to MM and which will not. Research on biomarkers in the progression from MGUS to MM will give more insight in the unknown pathogenesis of this hematological malignancy. This would improve research by elucidating new pathways and potential therapeutic targets as well as clinical management by closer follow-up and earlier treatment of high-risk MGUS patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Gammopatía Monoclonal de Relevancia Indeterminada/diagnóstico , Gammopatía Monoclonal de Relevancia Indeterminada/patología , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/patología , Progresión de la Enfermedad , Humanos , Pronóstico
12.
J Mol Cell Cardiol ; 97: 235-44, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27291064

RESUMEN

Cardiac atrial appendage stem cells (CASCs) show extraordinary myocardial differentiation properties, making them ideal candidates for myocardial regeneration. However, since the myocardium is a highly vascularized tissue, revascularization of the ischemic infarct area is essential for functional repair. Therefore, this study assessed if CASCs contribute to cardiac angiogenesis via paracrine mechanisms. First, it was demonstrated that CASCs produce and secrete high levels of numerous angiogenic growth factors, including vascular endothelial growth factor (VEGF), endothelin-1 (ET-1) and insulin-like growth factor binding protein 3 (IGFBP-3). Functional in vitro assays with a human microvascular endothelial cell line (HMEC-1) and CASC CM showed that CASCs promote endothelial cell proliferation, migration and tube formation, the most important steps of the angiogenesis process. Addition of inhibitory antibodies against identified growth factors could significantly reduce these effects, indicating their importance in CASC-induced neovascularization. The angiogenic potential of CASCs and CASC CM was also confirmed in a chorioallantoic membrane assay, demonstrating that CASCs promote blood vessel formation in vivo. In conclusion, this study shows that CASCs not only induce myocardial repair by cardiomyogenic differentiation, but also stimulate blood vessel formation by paracrine mechanisms. The angiogenic properties of CASCs further strengthen their therapeutic potential and make them an optimal stem cell source for the treatment of ischemic heart disease.


Asunto(s)
Apéndice Atrial/citología , Neovascularización Fisiológica , Células Madre/metabolismo , Inductores de la Angiogénesis/metabolismo , Animales , Biomarcadores , Células Cultivadas , Embrión de Pollo , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelina-1/metabolismo , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteómica/métodos , Análisis de Matrices Tisulares , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Curr Med Chem ; 23(23): 2421-38, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27222265

RESUMEN

Traditionally the heart is considered a terminally differentiated organ. However, at the beginning of this century increased mitotic activity was reported in ischemic and idiopathic dilated cardiomyopathy hearts, compared to healthy controls, underscoring the potential of regeneration after injury. Due to the presence of adult stem cells in bone marrow and their purported ability to differentiate into other cell lineages, this cell population was soon estimated to be the most suited candidate for cardiac regeneration. Clinical trials with autologous bone marrow-derived mononuclear cells, using either an intracoronary or direct intramyocardial injection approach consistently showed only minor improvement in global left ventricular ejection fraction. This was explained by their limited cardiomyogenic differentiation potential. To obtain more convincing improvement in cardiac function, based on true myocardial regeneration, the focus of research has shifted towards resident cardiac progenitor cells. Several isolation procedures have been described: the c-kit surface marker was the first to be used, however experimental research has clearly shown that c-kit+ cells only marginally contribute to regeneration post myocardial infarction. Sphere formation was used to isolate the so-called cardiosphere derived cells (CDC), and also in this cell population cardiomyogenic differentiation is a rare event. Recently a new type of stem cells derived from atrial tissue (cardiac atrial stem cells - CASCs) was identified, based on the presence of the enzyme aldehyde dehydrogenase (ALDH). Those cells significantly improve both regional and global LV ejection fraction, based on substantial engraftment and consistent differentiation into mature cardiomyocytes (98%).


Asunto(s)
Apéndice Atrial/citología , Células de la Médula Ósea/citología , Células Madre Mesenquimatosas/citología , Aldehído Deshidrogenasa/metabolismo , Diferenciación Celular , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Regeneración , Función Ventricular/fisiología
14.
PLoS One ; 11(4): e0154038, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27101000

RESUMEN

The inevitable switch from standard molecular methods to next-generation sequencing for the molecular profiling of tumors is challenging for most diagnostic laboratories. However, fixed validation criteria for diagnostic accreditation are not in place because of the great variability in methods and aims. Here, we describe the validation of a custom panel of hotspots in 24 genes for the detection of somatic mutations in non-small cell lung carcinoma, colorectal carcinoma and malignant melanoma starting from FFPE sections, using 14, 36 and 5 cases, respectively. The targeted hotspots were selected for their present or future clinical relevance in solid tumor types. The target regions were enriched with the TruSeq approach starting from limited amounts of DNA. Cost effective sequencing of 12 pooled libraries was done using a micro flow cell on the MiSeq and subsequent data analysis with MiSeqReporter and VariantStudio. The entire workflow was diagnostically validated showing a robust performance with maximal sensitivity and specificity using as thresholds a variant allele frequency >5% and a minimal amplicon coverage of 300. We implemented this method through the analysis of 150 routine diagnostic samples and identified clinically relevant mutations in 16 genes including KRAS (32%), TP53 (32%), BRAF (12%), APC (11%), EGFR (8%) and NRAS (5%). Importantly, the highest success rate was obtained when using also the low quality DNA samples. In conclusion, we provide a workflow for the validation of targeted NGS by a custom-designed pan-solid tumor panel in a molecular diagnostic lab and demonstrate its robustness in a clinical setting.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Mutación , Neoplasias/diagnóstico , Humanos , Límite de Detección , Neoplasias/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
16.
Int J Cardiol ; 201: 10-9, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26285182

RESUMEN

BACKGROUND: This study assessed whether autologous transplantation of cardiac atrial appendage stem cells (CASCs) preserves cardiac function after myocardial infarction (MI) in a minipig model. METHODS AND RESULTS: CASCs were isolated from right atrial appendages of Göttingen minipigs based on high aldehyde dehydrogenase activity and expanded. MI was induced by a 2h snare ligation of the left anterior descending coronary artery. Upon reperfusion, CASCs were intramyocardially injected under NOGA guidance (MI-CASC, n=10). Non-transplanted pigs (MI, n=8) received sham treatment. 3D electromechanical mapping (EMM) and cardiac MRI were performed to assess left ventricular (LV) function. MI pigs developed LV dilatation at 2 months (2M), while in the MI-CASC group volumes remained stable. Global LV ejection fraction decreased by 16 ± 8% in MI animals vs 3 ± 10% in MI-CASC animals and regional wall thickening in border areas was better preserved in the MI-CASC group. EMM showed decreased viability and wall motion in the LV for both groups POST-MI, whereas at 2M these parameters only improved in the MI-CASC. Substantial cell retention was accompanied by cardiomyogenic differentiation in 98±1% of the transplanted CASCs, which functionally integrated. Second harmonic generation microscopy confirmed the formation of mature sarcomeres in transplanted CASCs. Absence of cardiac arrhythmias indicated the safety of CASC transplantation. CONCLUSION: CASCs preserve cardiac function by extensive engraftment and cardiomyogenic differentiation. Our data indicate the enormous potential of CASCs in myocardial repair.


Asunto(s)
Apéndice Atrial/fisiología , Apéndice Atrial/trasplante , Infarto del Miocardio/terapia , Miocitos Cardíacos/fisiología , Trasplante de Células Madre/métodos , Animales , Apéndice Atrial/citología , Femenino , Infarto del Miocardio/patología , Células Madre/fisiología , Porcinos , Porcinos Enanos , Trasplante Autólogo
17.
Exp Physiol ; 100(7): 818-28, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25963928

RESUMEN

NEW FINDINGS: What is the central question of this study? It remains uncertain whether significant fat-free mass wasting occurs early after coronary artery bypass graft surgery, and the aetiology of this wasting in these particular conditions is unexplored. What is the main finding and its importance? Significant fat-free mass wasting is present after coronary artery bypass graft surgery, and this wasting effect is greater in younger patients and in patients with greater increments in blood cortisol-to-testosterone ratios after surgery. The magnitude and aetiology of muscle wasting early after coronary artery bypass graft (CABG) surgery remains unknown. In the present study, we assessed changes in fat-free mass early after CABG surgery and explored the possible aetiology (relationships with postsurgical changes in blood hormones, insulin resistance, subject characteristics and inflammation) for these changes. Fat-free mass was assessed before and 23 (range: 25) days after CABG surgery in 25 subjects. Blood testosterone, cortisol, insulin-like growth factor-1, growth hormone, sex hormone-binding globulin, glucose, insulin, C-peptide and C-reactive protein concentrations were determined, and free androgen index, cortisol-to-testosterone ratio and HOMA-IR index were all calculated before surgery, during the first 3 days after surgery and at reassessment of body composition. Relationships between changes in fat-free mass and changes in blood parameters after surgery or subject characteristics were studied. After surgery, free androgen index and blood sex hormone-binding globulin, testosterone and insulin-like growth factor-1 concentrations decreased significantly, while HOMA-IR index, cortisol-to-testosterone ratio, blood growth hormone, insulin and C-reactive protein concentrations increased significantly (P < 0.0025, observed α > 0.80). Whole-body fat-free mass decreased significantly [by -1.9 (range: 9.1) kg, P < 0.0025, observed α = 0.99] after surgery. According to regression analysis, greater absolute loss of fat-free mass was observed after CABG surgery in subjects who were younger, who experienced a greater increase in blood cortisol-to-testosterone ratio after surgery and/or who underwent earlier reassessment of body composition (P < 0.05). Significant decrements in fat-free mass were observed early after CABG surgery, especially in younger subjects and/or subjects with elevated blood cortisol-to-testosterone ratios after surgery. Interventions to preserve fat-free mass soon after CABG surgery are thus warranted.


Asunto(s)
Puente de Arteria Coronaria , Vasos Coronarios/cirugía , Tejido Adiposo/metabolismo , Adulto , Anciano , Composición Corporal/fisiología , Femenino , Humanos , Insulina/sangre , Resistencia a la Insulina/fisiología , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Globulina de Unión a Hormona Sexual/metabolismo , Testosterona/sangre , Injerto Vascular
18.
J Mol Cell Cardiol ; 66: 177-88, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24326234

RESUMEN

Mesenchymal stem cells (MSCs) modulate cardiac healing after myocardial injury through the release of paracrine factors, but the exact mechanisms are still unknown. One possible mechanism is through mobilization of endogenous cardiac stem cells (CSCs). This study aimed to test the pro-migratory effect of MSC conditioned medium (MSC-CM) on endogenous CSCs from human cardiac tissue. By using a three-dimensional collagen assay, we found that MSC-CM improved migration of cells from human cardiac tissue. Cell counts, perimeter and area measurements were utilized to quantify migration effects. To examine whether resident stem cells were among the migrating cells, specific stem cell properties were investigated. The migrating cells displayed strong similarities with resident Cardiac Atrial appendage Stem Cells (CASCs), including a clonogenic potential of ~21.5% and expression of pluripotency associated genes like Oct-4, Nanog, c-Myc and Klf-4. Similar to CASCs, migrating cells demonstrated high aldehyde dehydrogenase activity and were able to differentiate towards cardiomyocytes. Receptor tyrosine kinase analysis and collagen assays performed with recombinant platelet derived growth factor (PDGF)-AA and Imatinib Mesylate, a PDGF receptor inhibitor, suggested a role for the PDGF-AA/PDGF receptor α axis in enhancing the migration process of CASCs. In conclusion, our findings demonstrate that factors present in MSC-CM improve migration of resident stem cells from human cardiac tissue. These data open doors towards future therapies in which MSC secreted factors, like PDGF-AA, can be utilized to enhance the recruitment of CASCs towards the site of myocardial injury.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Biomarcadores/metabolismo , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Expresión Génica , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Mesenquimatosas/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas
19.
Gut ; 62(4): 550-60, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22535374

RESUMEN

OBJECTIVE: Bone marrow-derived mesenchymal stem cells (BM-MSC) migrate to primary tumours and drive tumour progression. This study aimed to identify the molecular mechanisms associated with these heterotypic cellular interactions and analyse their relevance in colorectal cancer (CRC). DESIGN: Paracrine interactions of BM-MSC with CRC cells were studied using collagen invasion assays, cell counts, flow cytometric cell-cycle analysis and tumour xenograft models. The role of neuregulin 1 (NRG1) and the human epidermal growth factor receptor (HER) family pathways were investigated using tyrosine kinase assays, mass spectrometry, pharmacological inhibition, antibody-mediated neutralisation and RNA interference. Transmembrane neuregulin 1 (tNRG1), HER2 and HER3 expression was analysed in primary CRC (n=54), adjacent normal colorectal tissues (n=4), liver metastases (n=3) and adjacent normal liver tissues (n=3) by immunohistochemistry. RESULTS: BM-MSC stimulate invasion, survival and tumorigenesis of CRC through the release of soluble NRG1, activating the HER2/HER3-dependent PI3K/AKT signalling cascade in CRC cells. Similarly, tumour-associated mesenchymal cells (T-MC) in CRC demonstrate high tNRG1 expression, which is significantly associated with advanced Union for International Cancer Control stage (p=0.005) and invasion depth (p=0.04) and decreased 5-year progression-free survival (p=0.01). HER2 and HER3 show membrane localisation in cancer cells of CRC tissue. CONCLUSION: Paracrine NRG1/HER3 signals initiated by BM-MSC and T-MC promote CRC cell progression, and high tNRG1 expression is associated with poor prognosis in CRC.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Madre Mesenquimatosas/patología , Neurregulina-1/metabolismo , Receptor ErbB-3/metabolismo , Análisis de Varianza , Western Blotting , Recuento de Células , Línea Celular Tumoral , Movimiento Celular/fisiología , Distribución de Chi-Cuadrado , Cromatografía Liquida , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Espectrometría de Masas , Comunicación Paracrina , Fosforilación , Interferencia de ARN , Receptor ErbB-2/metabolismo , Transducción de Señal , Estadísticas no Paramétricas
20.
Cardiovasc Res ; 97(3): 413-23, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23257022

RESUMEN

AIMS: Considerable shortcomings in the treatment of myocardial infarction (MI) still exist and therefore mortality remains high. Cardiac stem cell (CSC) therapy is a promising approach for myocardial repair. However, identification and isolation of candidate CSCs is mainly based on the presence or absence of certain cell surface markers, which suffers from some drawbacks. In order to find a more specific and reliable identification and isolation method, we investigated whether CSCs can be isolated based on the high expression of aldehyde dehydrogenase (ALDH). METHODS AND RESULTS: An ALDH(+) stem cell population, the cardiac atrial appendage stem cells (CASCs), was isolated from human atrial appendages. CASCs possess a unique phenotype that is clearly different from c-kit(+) CSCs but that seems more related to the recently described cardiac colony-forming-unit fibroblasts. Based on immunophenotype and in vitro differentiation studies, we suggest that CASCs are an intrinsic stem cell population and are not mobilized from bone marrow or peripheral blood. Indeed, they possess a clonogenicity of 16% and express pluripotency-associated genes. Furthermore, compared with cardiosphere-derived cells, CASCs possess an enhanced cardiac differentiation capacity. Indeed, differentiated cells express the most important cardiac-specific genes, produce troponin T proteins, and have an electrophysiological behaviour similar to that of adult cardiomyocytes (CMs). Transplanting CASCs in the minipig MI model resulted in extensive cardiomyogenic differentiation without teratoma formation. CONCLUSION: We have identified a new human CSC population able to differentiate into functional CMs. This opens interesting perspectives for cell therapy in patients with ischaemic heart disease.


Asunto(s)
Apéndice Atrial/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Infarto del Miocardio/terapia , Isquemia Miocárdica/terapia , Trasplante de Células Madre/métodos , Células Madre/citología , Aldehído Deshidrogenasa/metabolismo , Animales , Apéndice Atrial/metabolismo , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Fenotipo , Células Madre/metabolismo , Porcinos , Porcinos Enanos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA