Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Funct Biomater ; 15(5)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38786639

RESUMEN

Probiotic microorganisms are used in a variety of food supplements and medical formulations to promote human health. In periodontal therapy, probiotics are mainly used in the form of gels, tablets or rinses that often tend to leak from the periodontal pocket, resulting in a strongly reduced therapeutic effect. In this pilot in vitro study, we present biodegradable alginate-based particles as an alternative, highly efficient system for a periodontal delivery of probiotic bacteria to the inflammation site. For this purpose, Lactococcus (L.) lactis was encapsulated using a standardized pump-controlled extrusion-dripping method. Time-dependent bacterial release in artificial saliva was investigated over 9 days. The effect of freeze drying was explored to ensure long-term storage of L. lactis-loaded particles. Additionally, the particles were bound to dentin surface using approved bioadhesives and subjected to shear stress in a hydrodynamic flow chamber that mimics the oral cavity in vitro. Thus, round particles within the range of 0.80-1.75 mm in radius could be produced, whereby the diameter of the dripping tip had the most significant impact on the size. Although both small and large particles demonstrated a similar release trend of L. lactis, the release rate was significantly higher in the former. Following lyophilization, particles could restore their original shape within 4 h in artificial saliva; thereby, the bacterial viability was not affected. The attachment strength to dentin intensified by an adhesive could resist forces between 10 and 25 N/m2. Full degradation of the particles was observed after 20 days in artificial saliva. Therefore, alginate particles display a valuable probiotic carrier for periodontal applications that have several crucial advantages over existing preparations: a highly stable form, prolonged continuous release of therapeutic bacteria, precise manufacturing according to required dimensions at the application site, strong attachment to the tooth with low risk of dislocation, high biocompatibility and biodegradability.

2.
J Transl Med ; 22(1): 187, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38378655

RESUMEN

BACKGROUND: The initial idea of functional tissue replacement has shifted to the concept that injected cells positively modulate myocardial healing by a non-specific immune response of the transplanted cells within the target tissue. This alleged local modification of the scar requires assessment of regional properties of the left ventricular wall in addition to commonly applied measures of global morphological and functional parameters. Hence, we aimed at investigating the effect of cardiac cell therapy with cardiovascular progenitor cells, so-called cardiac induced cells, on both global and regional properties of the left ventricle by a multimodal imaging approach in a mouse model. METHODS: Myocardial infarction was induced in mice by ligation of the left anterior descending artery, the therapy group received an intramyocardial injection of 1 × 106 cardiac induced cells suspended in matrigel, the control group received matrigel only. [18F]FDG positron emission tomography imaging was performed after 17 days, to assess regional glucose metabolism. Three weeks after myocardial infarction, cardiac magnetic resonance imaging was performed for morphological and functional assessment of the left ventricle. Following these measurements, hearts were excised for histological examinations. RESULTS: Cell therapy had no significant effect on global morphological parameters. Similarly, there was no difference in scar size and capillary density between therapy and control group. However, there was a significant improvement in contractile function of the left ventricle - left ventricular ejection fraction, stroke volume and cardiac output. Regional analysis of the left ventricle identified changes of wall properties in the scar area as the putative mechanism. Cell therapy reduced the thinning of the scar and significantly improved its radial contractility. Furthermore, the metabolic defect, assessed by [18F]FDG, was significantly reduced by the cell therapy. CONCLUSION: Our data support the relevance of extending the assessment of global left ventricular parameters by a structured regional wall analysis for the evaluation of therapies targeting at modulation of healing myocardium. This approach will enable a deeper understanding of mechanisms underlying the effect of experimental regenerative therapies, thus paving the way for a successful translation into clinical application.


Asunto(s)
Fluorodesoxiglucosa F18 , Infarto del Miocardio , Animales , Ratones , Volumen Sistólico , Fluorodesoxiglucosa F18/metabolismo , Cicatriz/patología , Función Ventricular Izquierda , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/terapia , Infarto del Miocardio/patología , Miocardio/patología
3.
Genome Med ; 15(1): 61, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37563727

RESUMEN

BACKGROUND: The immune response is a crucial factor for mediating the benefit of cardiac cell therapies. Our previous research showed that cardiomyocyte transplantation alters the cardiac immune response and, when combined with short-term pharmacological CCR2 inhibition, resulted in diminished functional benefit. However, the specific role of innate immune cells, especially CCR2 macrophages on the outcome of cardiomyocyte transplantation, is unclear. METHODS: We compared the cellular, molecular, and functional outcome following cardiomyocyte transplantation in wildtype and T cell- and B cell-deficient Rag2del mice. The cardiac inflammatory response was assessed using flow cytometry. Gene expression profile was assessed using single-cell and bulk RNA sequencing. Cardiac function and morphology were determined using magnetic resonance tomography and immunohistochemistry respectively. RESULTS: Compared to wildtype mice, Rag2del mice show an increased innate immune response at steady state and disparate macrophage response after MI. Subsequent single-cell analyses after MI showed differences in macrophage development and a lower prevalence of CCR2 expressing macrophages. Cardiomyocyte transplantation increased NK cells and monocytes, while reducing CCR2-MHC-IIlo macrophages. Consequently, it led to increased mRNA levels of genes involved in extracellular remodelling, poor graft survival, and no functional improvement. Using machine learning-based feature selection, Mfge8 and Ccl7 were identified as the primary targets underlying these effects in the heart. CONCLUSIONS: Our results demonstrate that the improved functional outcome following cardiomyocyte transplantation is dependent on a specific CCR2 macrophage response. This work highlights the need to study the role of the immune response for cardiomyocyte cell therapy for successful clinical translation.


Asunto(s)
Infarto del Miocardio , Miocitos Cardíacos , Ratones , Animales , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Macrófagos/metabolismo , Monocitos/metabolismo , Ratones Endogámicos C57BL
4.
Cell Death Dis ; 14(8): 523, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37582912

RESUMEN

Long-living individuals (LLIs) escape age-related cardiovascular complications until the very last stage of life. Previous studies have shown that a Longevity-Associated Variant (LAV) of the BPI Fold Containing Family B Member 4 (BPIFB4) gene correlates with an extraordinarily prolonged life span. Moreover, delivery of the LAV-BPIFB4 gene exerted therapeutic action in murine models of atherosclerosis, limb ischemia, diabetic cardiomyopathy, and aging. We hypothesize that downregulation of BPIFB4 expression marks the severity of coronary artery disease (CAD) in human subjects, and supplementation of the LAV-BPIFB4 protects the heart from ischemia. In an elderly cohort with acute myocardial infarction (MI), patients with three-vessel CAD were characterized by lower levels of the natural logarithm (Ln) of peripheral blood BPIFB4 (p = 0.0077). The inverse association between Ln BPIFB4 and three-vessel CAD was confirmed by logistic regression adjusting for confounders (Odds Ratio = 0.81, p = 0.0054). Moreover, in infarcted mice, a single administration of LAV-BPIFB4 rescued cardiac function and vascularization. In vitro studies showed that LAV-BPIFB4 protein supplementation exerted chronotropic and inotropic actions on induced pluripotent stem cell (iPSC)-derived cardiomyocytes. In addition, LAV-BPIFB4 inhibited the pro-fibrotic phenotype in human cardiac fibroblasts. These findings provide a strong rationale and proof of concept evidence for treating CAD with the longevity BPIFB4 gene/protein.


Asunto(s)
Enfermedad de la Arteria Coronaria , Péptidos y Proteínas de Señalización Intercelular , Longevidad , Anciano , Animales , Humanos , Ratones , Envejecimiento/genética , Haplotipos/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Isquemia , Longevidad/genética
5.
Int J Mol Sci ; 24(10)2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37240361

RESUMEN

Cardiovascular diseases are the leading cause of death in industrialized nations. Due to the high number of patients and expensive treatments, according to the Federal Statistical Office (2017) in Germany, cardiovascular diseases account for around 15% of total health costs. Advanced coronary artery disease is mainly the result of chronic disorders such as high blood pressure, diabetes, and dyslipidemia. In the modern obesogenic environment, many people are at greater risk of being overweight or obese. The hemodynamic load on the heart is influenced by extreme obesity, which often leads to myocardial infarction (MI), cardiac arrhythmias, and heart failure. In addition, obesity leads to a chronic inflammatory state and negatively affects the wound-healing process. It has been known for many years that lifestyle interventions such as exercise, healthy nutrition, and smoking cessation drastically reduce cardiovascular risk and have a preventive effect against disorders in the healing process. However, little is known about the underlying mechanisms, and there is significantly less high-quality evidence compared to pharmacological intervention studies. Due to the immense potential of prevention in heart research, the cardiologic societies are calling for research work to be intensified, from basic understanding to clinical application. The topicality and high relevance of this research area are also evident from the fact that in March 2018, a one-week conference on this topic with contributions from top international scientists took place as part of the renowned "Keystone Symposia" ("New Insights into the Biology of Exercise"). Consistent with the link between obesity, exercise, and cardiovascular disease, this review attempts to draw lessons from stem-cell transplantation and preventive exercise. The application of state-of-the-art techniques for transcriptome analysis has opened new avenues for tailoring targeted interventions to very individual risk factors.


Asunto(s)
Cardiomioplastia , Infarto del Miocardio , Humanos , Obesidad/terapia , Sobrepeso , Estilo de Vida , Infarto del Miocardio/etiología , Infarto del Miocardio/prevención & control
6.
Cytotherapy ; 25(6): 640-652, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36890093

RESUMEN

Backgound Aims: This meta-analysis aims at summarizing the whole body of research on cell therapies for acute myocardial infarction (MI) in the mouse model to bring forward ongoing research in this field of regenerative medicine. Despite rather modest effects in clinical trials, pre-clinical studies continue to report beneficial effects of cardiac cell therapies for cardiac repair following acute ischemic injury. Results: The authors' meta-analysis of data from 166 mouse studies comprising 257 experimental groups demonstrated a significant improvement in left ventricular ejection fraction of 10.21% after cell therapy compared with control animals. Subgroup analysis indicated that second-generation cell therapies such as cardiac progenitor cells and pluripotent stem cell derivatives had the highest therapeutic potential for minimizing myocardial damage post-MI. Conclusions: Whereas the vision of functional tissue replacement has been replaced by the concept of regional scar modulation in most of the investigated studies, rather basic methods for assessing cardiac function were most frequently used. Hence, future studies will highly benefit from integrating methods for assessment of regional wall properties to evolve a deeper understanding of how to modulate cardiac healing after acute MI.


Asunto(s)
Infarto del Miocardio , Función Ventricular Izquierda , Animales , Ratones , Volumen Sistólico , Corazón , Infarto del Miocardio/terapia , Trasplante de Células Madre/métodos
7.
Int J Mol Sci ; 23(16)2022 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-36012110

RESUMEN

Ventricular arrhythmias associated with myocardial infarction (MI) have a significant impact on mortality in patients following heart attack. Therefore, targeted reduction of arrhythmia represents a therapeutic approach for the prevention and treatment of severe events after infarction. Recent research transplanting mesenchymal stem cells (MSC) showed their potential in MI therapy. Our study aimed to investigate the effects of MSC injection on post-infarction arrhythmia. We used our murine double infarction model, which we previously established, to more closely mimic the clinical situation and intramyocardially injected hypoxic pre-conditioned murine MSC to the infarction border. Thereafter, various types of arrhythmias were recorded and analyzed. We observed a homogenous distribution of all types of arrhythmias after the first infarction, without any significant differences between the groups. Yet, MSC therapy after double infarction led to a highly significant reduction in simple and complex arrhythmias. Moreover, RNA-sequencing of samples from stem cell treated mice after re-infarction demonstrated a significant decline in most arrhythmias with reduced inflammatory pathways. Additionally, following stem-cell therapy we found numerous highly expressed genes to be either linked to lowering the risk of heart failure, cardiomyopathy or sudden cardiac death. Moreover, genes known to be associated with arrhythmogenesis and key mutations underlying arrhythmias were downregulated. In summary, our stem-cell therapy led to a reduction in cardiac arrhythmias after MI and showed a downregulation of already established inflammatory pathways. Furthermore, our study reveals gene regulation pathways that have a potentially direct influence on arrhythmogenesis after myocardial infarction.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Infarto del Miocardio , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/terapia , Modelos Animales de Enfermedad , Células Madre Mesenquimatosas/metabolismo , Ratones , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia
8.
Clin Implant Dent Relat Res ; 24(5): 664-675, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35709098

RESUMEN

BACKGROUND: Inflammation triggered by bacterial biofilms in the surrounding tissue is a major etiological factor for peri-implantitis and subsequent implant failure. However, little is known about the direct effects of bacterial corrosion and recolonization on implant failure PURPOSE: To investigate the influence of oral commensals on bacterial corrosion and recolonization of titanium surfaces. MATERIALS AND METHODS: Streptococcus sanguinis (S. sanguinis) and Porphyromonas gingivalis (P. gingivalis), which are key bacteria in oral biofilm formation, were cultured on commercially pure titanium and titanium-aluminum-vanadium (Ti6Al4V) plates in artificial saliva/brain heart infusion medium under aerobic or anaerobic conditions. Biofilm formation was examined after 7 and 21 days by crystal violet and live/dead staining. Titanium ions released into culture supernatants were analyzed over a period of 21 days by atomic absorption spectrometry. Visual changes in surface morphology were investigated using scanning electron microscopy. Biofilm formation on sterilized, biocorroded, and recolonized implant surfaces was determined by crystal violet staining. RESULTS: S. sanguinis and P. gingivalis formed stable biofilms on the titanium samples. Bacterial corrosion led to a significant increase in titanium ion release from these titanium plates (p < 0.01), which was significantly higher under aerobic conditions on pure titanium (p ≤ 0.001). No obvious morphological surface changes, such as pitting and discoloration, were detected in the titanium samples. During early biofilm formation, the addition of titanium ions significantly decreased the number of live cells. In contrast, a significant effect on biofilm mass was only detected with P. gingivalis. Bacterial corrosion had no influence on bacterial recolonization following sterilization of titanium and Ti6Al4V surfaces. CONCLUSION: Bacterial corrosion differs between oral commensal bacteria and leads to increased titanium ion release from titanium plates. The titanium ion release did not influence biofilm formation or bacterial recolonization under in vitro conditions.


Asunto(s)
Implantes Dentales , Titanio , Aleaciones , Aluminio , Biopelículas , Corrosión , Implantes Dentales/microbiología , Violeta de Genciana , Porphyromonas gingivalis , Saliva Artificial , Propiedades de Superficie , Titanio/química , Vanadio
9.
Cell Mol Life Sci ; 79(3): 149, 2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35199227

RESUMEN

The in vitro generation of human cardiomyocytes derived from induced pluripotent stem cells (iPSC) is of great importance for cardiac disease modeling, drug-testing applications and for regenerative medicine. Despite the development of various cultivation strategies, a sufficiently high degree of maturation is still a decisive limiting factor for the successful application of these cardiac cells. The maturation process includes, among others, the proper formation of sarcomere structures, mediating the contraction of cardiomyocytes. To precisely monitor the maturation of the contractile machinery, we have established an imaging-based strategy that allows quantitative evaluation of important parameters, defining the quality of the sarcomere network. iPSC-derived cardiomyocytes were subjected to different culture conditions to improve sarcomere formation, including prolonged cultivation time and micro patterned surfaces. Fluorescent images of α-actinin were acquired using super-resolution microscopy. Subsequently, we determined cell morphology, sarcomere density, filament alignment, z-Disc thickness and sarcomere length of iPSC-derived cardiomyocytes. Cells from adult and neonatal heart tissue served as control. Our image analysis revealed a profound effect on sarcomere content and filament orientation when iPSC-derived cardiomyocytes were cultured on structured, line-shaped surfaces. Similarly, prolonged cultivation time had a beneficial effect on the structural maturation, leading to a more adult-like phenotype. Automatic evaluation of the sarcomere filaments by machine learning validated our data. Moreover, we successfully transferred this approach to skeletal muscle cells, showing an improved sarcomere formation cells over different differentiation periods. Overall, our image-based workflow can be used as a straight-forward tool to quantitatively estimate the structural maturation of contractile cells. As such, it can support the establishment of novel differentiation protocols to enhance sarcomere formation and maturity.


Asunto(s)
Señalización del Calcio/fisiología , Diferenciación Celular/fisiología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Sarcómeros/metabolismo , Actinina/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Humanos , Aprendizaje Automático , Ratones , Microscopía Fluorescente/métodos , Músculo Esquelético/citología , Miocardio/citología , Fenotipo , ARN/genética , ARN/aislamiento & purificación
10.
Biomed Pharmacother ; 145: 112429, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34801854

RESUMEN

The human contact system consists of plasma proteins, which - after contact to foreign surfaces - are bound to them, thereby activating the zymogens of the system into enzymes. This activation mechanism gave the system its name - contact system. It is considered as a procoagulant and proinflammatory response mechanism, as activation finally leads to the generation of fibrin and bradykinin. To date, no physiological processes have been described that are mediated by contact activation. However, contact system factors play a pathophysiological role in numerous diseases, such as cardiovascular diseases, arthritis, colitis, sepsis, and cancer. Contact system factors are therefore an interesting target for new therapeutic options in different clinical conditions.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Inflamación/fisiopatología , Neoplasias/patología , Sepsis/fisiopatología , Animales , Proteínas Sanguíneas/metabolismo , Bradiquinina/metabolismo , Fibrina/metabolismo , Humanos
11.
Sci Rep ; 11(1): 11625, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34079005

RESUMEN

Novel therapeutic strategies aiming at improving the healing process after an acute myocardial infarction are currently under intense investigation. The mouse model plays a central role for deciphering the underlying mechanisms on a molecular and cellular level. Therefore, we intended to assess in-vivo post-infarct remodeling as comprehensively as possible using an expedient native magnetic resonance imaging (MRI) in the two most prominent infarct models, permanent ligation (PL) of the left anterior descending artery (LAD) versus ischemia reperfusion (I/R). Mice were subjected to either permanent or transient (45 min) occlusion of the LAD. After 3 weeks, examinations were performed with a 7-Tesla small animal MRI system. Data analysis was performed with the freely available software Segment. PL resulted in a massive dilation of the left ventricle, accompanied by hypertrophy of the non-infarcted myocardium and a decline of contractile function. These effects were less pronounced following I/R compared to healthy animals. Single plane assessments were not sufficient to capture the specific differences of left ventricular (LV) properties between the two infarct models. Bulls-eye plots were found to be an ideal tool for qualitative LV wall assessment, whereas a multi-slice sector-based analysis of wall regions is ideal to determine differences in hypertrophy, lateral wall thinning and wall thickening on a quantitative level. We combine the use of polar map-based analysis of LV wall properties with volumetric measurements using simple CINE CMR imaging. Our strategy represents a versatile and easily available tool for serial assessment of the LV during the remodeling process. Our study contributes to a better understanding of the effects of novel therapies targeting the healing of damaged myocardium.


Asunto(s)
Trastornos Cerebrovasculares/diagnóstico por imagen , Ventrículos Cardíacos/diagnóstico por imagen , Corazón/diagnóstico por imagen , Infarto de la Arteria Cerebral Anterior/diagnóstico por imagen , Infarto del Miocardio/diagnóstico por imagen , Daño por Reperfusión/diagnóstico por imagen , Animales , Trastornos Cerebrovasculares/fisiopatología , Modelos Animales de Enfermedad , Corazón/fisiopatología , Ventrículos Cardíacos/fisiopatología , Infarto de la Arteria Cerebral Anterior/fisiopatología , Ligadura/métodos , Imagen por Resonancia Cinemagnética/métodos , Ratones , Infarto del Miocardio/fisiopatología , Daño por Reperfusión/fisiopatología , Factores de Tiempo , Remodelación Ventricular
12.
Cells ; 9(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32756334

RESUMEN

We investigated the influence of syngeneic cardiomyocyte transplantation after myocardial infarction (MI) on the immune response and cardiac function. Methods and Results: We show for the first time that the immune response is altered as a result of syngeneic neonatal cardiomyocyte transplantation after MI leading to improved cardiac pump function as observed by magnetic resonance imaging in C57BL/6J mice. Interestingly, there was no improvement in the capillary density as well as infarct area as observed by CD31 and Sirius Red staining, respectively. Flow cytometric analysis revealed a significantly different response of monocyte-derived macrophages and regulatory T cells after cell transplantation. Interestingly, the inhibition of monocyte infiltration accompanied by cardiomyocyte transplantation diminished the positive effect of cell transplantation alone. The number of CD68+ macrophages in the remote area of the heart observed after four weeks was also different between the groups. Transcriptome analysis showed several changes in the gene expression involving circadian regulation, mitochondrial metabolism and immune responses after cardiomyocyte transplantation. Conclusion: Our work shows that cardiomyocyte transplantation alters the immune response after myocardial infarction with the recruited monocytes playing a role in the beneficial effect of cell transplantation. It also paves the way for further optimization of the efficacy of cardiomyocyte transplantation and their successful translation in the clinic.


Asunto(s)
Infarto del Miocardio/terapia , Miocardio/inmunología , Miocitos Cardíacos/trasplante , Animales , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Corazón/fisiología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/inmunología , Receptores CCR2/inmunología , Linfocitos T Reguladores/inmunología
13.
Cells ; 9(6)2020 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-32486211

RESUMEN

Angiogenesis plays a central role in the healing process following acute myocardial infarction. The PET tracer [68Ga]-NODAGA-RGD, which is a ligand for the αvß3 integrin, has been investigated for imaging angiogenesis in the process of healing myocardium in both animal and clinical studies. It´s value as a prognostic marker of functional outcome remains unclear. Therefore, the aim of this work was to establish [68Ga]-NODAGA-RGD for imaging angiogenesis in the murine infarct model and evaluate the tracer as a predictor for cardiac remodeling in the context of cardiac stem cell therapy. [68Ga]-NODAGA-RGD PET performed seven days after left anterior descending coronary artery (LAD) occlusion in 129S6 mice showed intense tracer accumulation within the infarct region. The specificity was shown in a sub-group of animals by application of the competitive inhibitor cilengitide prior to tracer injection in a subgroup of animals. Myocardial infarction (MI) significantly reduced cardiac function and resulted in pronounced left ventricular remodeling after three weeks, as measured by cardiac MRI in a separate group. Cardiac induced cells (CiC) that were derived from mESC injected intramyocardially in the therapy group significantly improved left ventricular ejection fraction (LVEF). Surprisingly, CiC transplantation resulted in significantly lower tracer accumulation seven days after MI induction. Accordingly, we successfully established the PET tracer [68Ga]-NODAGA-RGD for the assessment of αvß3 integrin expression in the healing process after MI in the mouse model. Yet, our results indicate that the mere extent of angiogenesis following MI does not serve as a sufficient prognostic marker for functional outcome.


Asunto(s)
Acetatos/química , Radioisótopos de Galio/química , Compuestos Heterocíclicos con 1 Anillo/química , Infarto del Miocardio/diagnóstico por imagen , Neovascularización Fisiológica , Oligopéptidos/química , Tomografía de Emisión de Positrones , Trasplante de Células Madre , Remodelación Ventricular , Animales , Integrina alfaVbeta3/metabolismo , Imagen por Resonancia Magnética , Ratones , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia
14.
Int J Mol Sci ; 21(9)2020 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-32397287

RESUMEN

Cellular inflammation is an integral part of the healing process following acute myocardial infarction and has been under intense investigation for both therapeutic and prognostic approaches. Monocytes and macrophages are metabolically highly active and show increased uptake rates of glucose and its analog, 18F-FDG. Yet, the specific allocation of the radioactivity to the inflammatory cells via positron emission tomography (PET) imaging requires the suppression of glucose metabolism in viable myocardium. In mice, the most important model organism in basic research, this can be achieved by the application of ketamine/xylazine (KX) for anesthesia instead of isoflurane. Yet, while the consensus exists that glucose metabolism is effectively suppressed, a strategy for reproducible image analysis is grossly lacking and causes uncertainty concerning data interpretation. We introduce a simple strategy for systematic image analysis, which is a prerequisite to evaluate therapies targeting myocardial inflammation. Mice underwent permanent occlusion of the left anterior descending artery (LAD), inducing an acute myocardial infarction (MI). Five days after MI induction, 10MBq 18F-FDG was injected intravenously and a static PET/CT scan under ketamine/xylazine anesthesia was performed. For image reconstruction, we used an algorithm based on three-dimensional ordered subsets expectation maximization (3D-OSEM) followed by three-dimensional ordinary Poisson maximum a priori (MAP) reconstruction. Using this approach, high focal tracer uptake was typically located in the border zone of the infarct by visual inspection. To precisely demarcate the border zone for reproducible volume of interest (VOI) positioning, our protocol relies on positioning VOIs around the whole left ventricle, the inferobasal wall and the anterolateral wall guided by anatomical landmarks. This strategy enables comparable data in mouse studies, which is an important prerequisite for using a PET-based assessment of myocardial inflammation as a prognostic tool in therapeutic applications.


Asunto(s)
Fluorodesoxiglucosa F18/metabolismo , Inflamación/metabolismo , Infarto del Miocardio/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Anestesia/métodos , Animales , Modelos Animales de Enfermedad , Glucosa/metabolismo , Procesamiento de Imagen Asistido por Computador , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Radiofármacos/metabolismo
15.
J Adv Res ; 21: 35-47, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31641536

RESUMEN

Prospective severity assessment is legally required in many countries to ensure high-quality research along with high welfare standards for laboratory animals. Mice and rats, the most common laboratory species, are prey animals that usually suppress signs of pain and suffering. Therefore, highly sensitive readout parameters are necessary to adequately quantify distress. The present study compared the performance of different non-invasive methods in determining animal distress, such as measuring body weight, distress score, faecal corticosterone metabolites, burrowing, and nesting behaviour, with continuous monitoring of heart rate, body temperature and activity by telemetry. The distress caused by two surgical interventions was compared and the burden caused by tumour growth was described. Transmitter implantation caused higher distress than laparotomy plus carcinoma cell injection into the pancreas. Surprisingly, no significant increase in distress was observed during tumour growth. The receiver operating characteristic curve analysis revealed that some non-invasive distress-parameters, i.e., distress-score and burrowing activity, exhibited slightly better performance to quantify distress than the most suitable parameters measured by telemetry. Due to the high burden caused by the implantation of the telemetric device, the use of non-invasive methods to assess distress in laboratory animals after surgical interventions should be favoured in future studies.

16.
Cells ; 8(12)2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31835854

RESUMEN

Cellular inflammation following acute myocardial infarction has gained increasing importance as a target mechanism for therapeutic approaches. We sought to investigate the effect of syngeneic cardiac induced cells (CiC) on myocardial inflammation using 18F-FDG PET (Positron emission tomography)-based imaging and the resulting effect on cardiac pump function using cardiac magnetic resonance (CMR) imaging in a mouse model of myocardial infarction. Mice underwent permanent left anterior descending coronary artery (LAD) ligation inducing an acute inflammatory response. The therapy group received an intramyocardial injection of 106 CiC into the border zone of the infarction. Five days after myocardial infarction, 18F-FDG PET was performed under anaesthesia with ketamine and xylazine (KX) to image the inflammatory response in the heart. Flow cytometry of the mononuclear cells in the heart was performed to analyze the inflammatory response. The effect of CiC therapy on cardiac function was determined after three weeks by CMR. The 18F-FDG PET imaging of the heart five days after myocardial infarction (MI) revealed high focal tracer accumulation in the border zone of the infarcted myocardium, whereas no difference was observed in the tracer uptake between infarct and remote myocardium. The CiC transplantation induced a shift in 18F-FDG uptake pattern, leading to significantly higher 18F-FDG uptake in the whole heart, as well as the remote area of the heart. Correspondingly, high numbers of CD11+ cells could be measured by flow cytometry in this region. The CiC transplantation significantly improved the left ventricular ejection function (LVEF) three weeks after myocardial infarction. The CiC transplantation after myocardial infarction leads to an improvement in pump function through modulation of the cellular inflammatory response five days after myocardial infarction. By combining CiC transplantation and the cardiac glucose uptake suppression protocol with KX in a mouse model, we show for the first time, that imaging of cellular inflammation after myocardial infarction using 18F-FDG PET can be used as an early prognostic tool for assessing the efficacy of cardiac stem cell therapies.


Asunto(s)
Antígenos CD11/metabolismo , Fluorodesoxiglucosa F18/administración & dosificación , Corazón/diagnóstico por imagen , Células Madre Embrionarias de Ratones/trasplante , Infarto del Miocardio/terapia , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Corazón/fisiopatología , Humanos , Imagen por Resonancia Cinemagnética , Ratones , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/inmunología , Infarto del Miocardio/fisiopatología , Tomografía de Emisión de Positrones , Resultado del Tratamiento , Función Ventricular Izquierda
17.
Cells ; 8(12)2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31757119

RESUMEN

BACKGROUND: Ventricular arrhythmias (VA) are a common cause of sudden death after myocardial infarction (MI). Therefore, developing new therapeutic methods for the prevention and treatment of VA is of prime importance. METHODS: Human bone marrow derived CD271+ mesenchymal stem cells (MSC) were tested for their antiarrhythmic effect. This was done through the development of a novel mouse model using an immunocompromised Rag2-/- γc-/- mouse strain subjected to myocardial "infarction-reinfarction". The mice underwent a first ischemia-reperfusion through the left anterior descending (LAD) artery closure for 45 minutes with a subsequent second permanent LAD ligation after seven days from the first infarct. RESULTS: This mouse model induced various types of VA detected with continuous electrocardiogram (ECG) monitoring via implanted telemetry device. The immediate intramyocardial delivery of CD271+ MSC after the first MI significantly reduced VA induced after the second MI. CONCLUSIONS: In addition to the clinical relevance, more closely reflecting patients who suffer from severe ischemic heart disease and related arrhythmias, our new mouse model bearing reinfarction warrants the time required for stem cell engraftment and for the first time enables us to analyze and verify significant antiarrhythmic effects of human CD271+ stem cells in vivo.


Asunto(s)
Adapaleno/inmunología , Antiarrítmicos/uso terapéutico , Modelos Animales de Enfermedad , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/terapia , Adapaleno/análisis , Animales , Femenino , Humanos , Inmunofenotipificación , Ratones , Ratones Noqueados
18.
Cells Tissues Organs ; 206(1-2): 35-45, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30630170

RESUMEN

In-depth knowledge of the mechanisms induced by early postischemic cardiac endogenous mesenchymal stem cells (MSCs) in the acutely ischemic heart could advance our understanding of cardiac regeneration. Herein, we aimed to identify, isolate, and initially characterize the origin, kinetics and fate of cardiac MSCs. This was facilitated by in vivo genetic cell fate mapping through green fluorescent protein (GFP) expression under the control of vimentin induction after acute myocardial infarction (MI). Following permanent ligation of the left anterior descending coronary artery in CreER+ mTom/mGFP+ mice, vimentin/GFP+ cells revealed ischemia-responsive activation, survival, and local enrichment inside the peri-infarction border zone. Fluorescence-activated cell sorting (FACS)-isolated vimentin/GFP+ cells could be strongly expanded in vitro with clonogenic precursor formation and revealed MSC-typical cell morphology. Flow-cytometric analyses demonstrated an increase in cardiac vimentin/GFP+ cells in the ischemic heart, from a 0.6% cardiac mononuclear cell (MNC) fraction at 24 h to 1.6% at 72 h following MI. Sca-1+CD45- cells within the vimentin/GFP+ subtype of this MNC fraction increased from 35.2% at 24 h to 74.6% at 72 h after MI. The cardiac postischemic vimentin/GFP+ MNC subtype showed multipotent adipogenic, chondrogenic, and osteogenic differentiation potential, which is distinctive for MSCs. In conclusion, we demonstrated a seemingly proliferative first response of vimentin- induced cardiac endogenous MSCs in the acutely ischemic heart. Genetically, GFP-targeted in vivo cell tracking, isolation, and in vitro expansion of this cardiac MSC subtype could help to clarify their reparative status in inflammation, fibrogenesis, cell turnover, tissue homeostasis, and myocardial regeneration.


Asunto(s)
Células Madre Mesenquimatosas/citología , Infarto del Miocardio/patología , Miocardio/patología , Vimentina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Separación Celular , Supervivencia Celular , Células Cultivadas , Femenino , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/metabolismo , Antígenos Comunes de Leucocito/análisis , Antígenos Comunes de Leucocito/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Vimentina/análisis
19.
Adv Drug Deliv Rev ; 120: 2-24, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29054357

RESUMEN

After two decades of intensive research and attempts of clinical translation, stem cell based therapies for cardiac diseases are not getting closer to clinical success. This review tries to unravel the obstacles and focuses on underlying mechanisms as the target for regenerative therapies. At present, the principal outcome in clinical therapy does not reflect experimental evidence. It seems that the scientific obstacle is a lack of integration of knowledge from tissue repair and disease mechanisms. Recent insights from clinical trials delineate mechanisms of stem cell dysfunction and gene defects in repair mechanisms as cause of atherosclerosis and heart disease. These findings require a redirection of current practice of stem cell therapy and a reset using more detailed analysis of stem cell function interfering with disease mechanisms. To accelerate scientific development the authors suggest intensifying unified computational data analysis and shared data knowledge by using open-access data platforms.


Asunto(s)
Cardiopatías/terapia , Trasplante de Células Madre , Animales , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Trasplante de Células Madre/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...