LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 73

Search options

  1. Article ; Online: An Innovative Toolkit to Investigate the Complex Mechanisms of Cardiac Arrhythmias.

    Clauss, Sebastian

    Journal of visualized experiments : JoVE

    2023  , Issue 192

    Abstract: Articles discussed: Tomsits, P., Schuttler, D., Kaab, S., Clauss, S., Voigt, N. Isolation of high quality murine atrial and ventricular myocytes for simultaneous measurements of Ca2+ transients and L-Type calcium current. Journal of Visualized ... ...

    Abstract Articles discussed: Tomsits, P., Schuttler, D., Kaab, S., Clauss, S., Voigt, N. Isolation of high quality murine atrial and ventricular myocytes for simultaneous measurements of Ca2+ transients and L-Type calcium current. Journal of Visualized Experiments. (165), 10.3791/61964 (2020). Seibertz, F., Reynolds, M., Voigt, N. Single-Cell optical action potential measurement in human induced pluripotent stem cell-derived cardiomyocytes. Journal of Visualized Experiments. (166), 10.3791/61890 (2020). Xia, R. et al. Isolation and culture of resident cardiac macrophages from the murine sinoatrial and atrioventricular node. Journal of Visualized Experiments. (171), 10.3791/62236 (2021). Xia, R. et al. Whole-Mount immunofluorescence staining, confocal imaging and 3D reconstruction of the sinoatrial and atrioventricular node in the mouse. Journal of Visualized Experiments. (166), 10.3791/62058 (2020). Kumar, P., Si, M., Paulhus, K., Glasscock, E. Microelectrode array recording of sinoatrial node firing rate to identify intrinsic cardiac pacemaking defects in mice. Journal of Visualized Experiments. (173), 10.3791/62735 (2021). Scherschel, K. et al. Location, dissection, and analysis of the murine stellate ganglion. Journal of Visualized Experiments. (166), 10.3791/62026 (2020). Shimura, D., Hunter, J., Katsumata, M., Shaw, R. M. Removal of an internal translational start site from mRNA while retaining expression of the full-length protein. Journal of Visualized Experiments. (181), 10.3791/63405 (2022). Rotzer, R. D. et al. Implantation of combined telemetric ECG and blood pressure transmitters to determine spontaneous baroreflex sensitivity in conscious mice. Journal of Visualized Experiments. (168), 10.3791/62101 (2021). Tomsits, P. et al. Analyzing long-term electrocardiography recordings to detect arrhythmias in mice. Journal of Visualized Experiments. (171), 10.3791/62386 (2021). Grab, M. et al. Development and evaluation of 3D-printed cardiovascular phantoms for interventional planning and training. Journal of Visualized Experiments. (167), 10.3791/62063 (2021).
    MeSH term(s) Humans ; Animals ; Mice ; Induced Pluripotent Stem Cells ; Arrhythmias, Cardiac ; Dissection ; Atrioventricular Node ; Baroreflex
    Language English
    Publishing date 2023-02-10
    Publishing country United States
    Document type Editorial ; Research Support, Non-U.S. Gov't
    ZDB-ID 2259946-0
    ISSN 1940-087X ; 1940-087X
    ISSN (online) 1940-087X
    ISSN 1940-087X
    DOI 10.3791/64932
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Book ; Online ; Thesis: Proarrhythmic atrial remodeling mechanisms leading to arrhythmias

    Clauß, Sebastian [Verfasser]

    2021  

    Author's details Sebastian Clauß
    Keywords Medizin, Gesundheit ; Medicine, Health
    Subject code sg610
    Language English
    Publisher Universitätsbibliothek der Ludwig-Maximilians-Universität
    Publishing place München
    Document type Book ; Online ; Thesis
    Database Digital theses on the web

    More links

    Kategorien

  3. Article ; Online: Microdissection and Immunofluorescence Staining of Myocardial Sleeves in Murine Pulmonary Veins.

    Villgrater, Hannes E / Xia, Ruibing / Sharma Chivukula, Aparna / Tomsits, Philipp / Clauss, Sebastian

    Journal of visualized experiments : JoVE

    2023  , Issue 201

    Abstract: Pulmonary veins (PVs) are the major source of ectopic beats in atrial arrhythmias and play a crucial role in the development and progression of atrial fibrillation (AF). PVs contain myocardial sleeves (MS) composed of cardiomyocytes. MS are implicated in ...

    Abstract Pulmonary veins (PVs) are the major source of ectopic beats in atrial arrhythmias and play a crucial role in the development and progression of atrial fibrillation (AF). PVs contain myocardial sleeves (MS) composed of cardiomyocytes. MS are implicated in the initiation and maintenance of AF, as they preserve similarities to the cardiac working myocardium, including the ability to generate ectopic electrical impulses. Rodents are widely used and may represent excellent animal models to study the pulmonary vein myocardium since cardiomyocytes are widely present all over the vessel wall. However, precise microdissection and preparation of murine PVs is challenging due to the small organ size and intricate anatomy. We demonstrate a microscopy-guided microdissection protocol for isolating the murine left atrium (LA) together with the PVs. Immunofluorescence staining using cardiac Troponin-T (cTNT) and connexin 43 (Cx43) antibodies is performed to visualize the LA and PVs in full length. Imaging at 10x and 40x magnification provides a comprehensive view of the PV structure as well as detailed insights into the myocardial architecture, particularly highlighting the presence of connexin 43 within the MS.
    MeSH term(s) Animals ; Mice ; Connexin 43 ; Pulmonary Veins ; Microdissection ; Myocardium ; Atrial Fibrillation/surgery ; Heart Atria ; Fluorescent Antibody Technique ; Catheter Ablation
    Chemical Substances Connexin 43
    Language English
    Publishing date 2023-11-21
    Publishing country United States
    Document type Journal Article ; Video-Audio Media
    ZDB-ID 2259946-0
    ISSN 1940-087X ; 1940-087X
    ISSN (online) 1940-087X
    ISSN 1940-087X
    DOI 10.3791/65836
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Medetomidine/midazolam/fentanyl narcosis alters cardiac autonomic tone leading to conduction disorders and arrhythmias in mice.

    Tomsits, Philipp / Volz, Lina / Xia, Ruibing / Chivukula, Aparna / Schüttler, Dominik / Clauß, Sebastian

    Lab animal

    2023  Volume 52, Issue 4, Page(s) 85–92

    Abstract: Arrhythmias are critical contributors to cardiovascular morbidity and mortality. Therapies are mainly symptomatic and often insufficient, emphasizing the need for basic research to unveil the mechanisms underlying arrhythmias and to enable better and ... ...

    Abstract Arrhythmias are critical contributors to cardiovascular morbidity and mortality. Therapies are mainly symptomatic and often insufficient, emphasizing the need for basic research to unveil the mechanisms underlying arrhythmias and to enable better and ideally causal therapies. In translational approaches, mice are commonly used to study arrhythmia mechanisms in vivo. Experimental electrophysiology studies in mice are performed under anesthesia with medetomidine/midazolam/fentanyl (MMF) and isoflurane/fentanyl (IF) as commonly used regimens. Despite evidence of adverse effects of individual components on cardiac function, few data are available regarding the specific effects of these regimens on cardiac electrophysiology in mice. Here we present a study investigating the effects of MMF and IF narcosis on cardiac electrophysiology in vivo in C57BL/6N wild-type mice. Telemetry transmitters were implanted in a group of mice, which served as controls for baseline parameters without narcosis. In two other groups of mice, electrocardiogram and invasive electrophysiology studies were performed under narcosis (with either MMF or IF). Basic electrocardiogram parameters, heart rate variability parameters, sinus node and atrioventricular node function, and susceptibility to arrhythmias were assessed. Experimental data suggest a remarkable influence of MMF on cardiac electrophysiology compared with IF and awake animals. While IF only moderately reduced heart rate, MMF led to significant bradycardia, spontaneous arrhythmias, heart rate variability alterations as well as sinus and AV node dysfunction, and increased inducibility of ventricular arrhythmias. On the basis of these observed effects, we suggest avoiding MMF in mice, specifically when studying cardiac electrophysiology, but also whenever a regular heartbeat is required for reliable results, such as in heart failure or imaging research.
    MeSH term(s) Mice ; Animals ; Midazolam/adverse effects ; Fentanyl/adverse effects ; Medetomidine/adverse effects ; Stupor/chemically induced ; Mice, Inbred C57BL ; Arrhythmias, Cardiac/chemically induced ; Heart Rate
    Chemical Substances Midazolam (R60L0SM5BC) ; Fentanyl (UF599785JZ) ; Medetomidine (MR15E85MQM)
    Language English
    Publishing date 2023-03-23
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ISSN 1548-4475
    ISSN (online) 1548-4475
    DOI 10.1038/s41684-023-01141-0
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Genetic insight into sick sinus syndrome. Is there a pill for it or how far are we on the translational road to personalized medicine?

    Tomsits, Philipp / Clauss, Sebastian / Kääb, Stefan

    European heart journal

    2021  Volume 42, Issue 20, Page(s) 1972–1975

    Language English
    Publishing date 2021-02-08
    Publishing country England
    Document type Editorial ; Comment
    ZDB-ID 603098-1
    ISSN 1522-9645 ; 0195-668X
    ISSN (online) 1522-9645
    ISSN 0195-668X
    DOI 10.1093/eurheartj/ehab209
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Book ; Online ; Thesis: Structure-property relationships of one-component moisture-curing polyurethane adhesives under thermal load

    Clauss, Sebastian

    2011  

    Author's details by Sebastian Clauss
    Language English
    Size Online-Ressource (1 Band), Ill
    Publisher ETH / IfB, Institut für Baustoffe - Holzphysik
    Publishing place Zürich
    Document type Book ; Online ; Thesis
    Thesis / German Habilitation thesis Diss., Eidgenössische Technische Hochschule ETH Zürich, Nr. 20060--Zürich, 2006
    Database Former special subject collection: coastal and deep sea fishing

    More links

    Kategorien

  7. Book ; Online ; Thesis: Structure-property relationships of one-component moisture-curing polyurethane adhesives under thermal load

    Clauss, Sebastian

    2011  

    Author's details by Sebastian Clauss
    Language English
    Size Online-Ressource (1 Band), Ill
    Publisher ETH / IfB, Institut für Baustoffe - Holzphysik
    Publishing place Zürich
    Document type Book ; Online ; Thesis
    Thesis / German Habilitation thesis Diss., Eidgenössische Technische Hochschule ETH Zürich, Nr. 20060--Zürich, 2006
    Database Library catalogue of the German National Library of Science and Technology (TIB), Hannover

    More links

    Kategorien

  8. Article ; Online: Genetic Burden of Birthweight on Atrial Fibrillation: Translational Challenges in Genetic Atrial Fibrillation Studies.

    Tomsits, Philipp / Clauss, Sebastian / Kääb, Stefan

    Circulation. Genomic and precision medicine

    2020  Volume 13, Issue 3, Page(s) e002987

    MeSH term(s) Atrial Fibrillation/genetics ; Birth Weight/genetics ; Body Mass Index ; Humans ; Mendelian Randomization Analysis ; Risk Factors
    Language English
    Publishing date 2020-06-16
    Publishing country United States
    Document type Editorial ; Comment
    ISSN 2574-8300
    ISSN (online) 2574-8300
    DOI 10.1161/CIRCGEN.120.002987
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article: Chronic Thromboembolic Pulmonary Hypertension - What Have We Learned From Large Animal Models.

    Stam, Kelly / Clauss, Sebastian / Taverne, Yannick J H J / Merkus, Daphne

    Frontiers in cardiovascular medicine

    2021  Volume 8, Page(s) 574360

    Abstract: Chronic thrombo-embolic pulmonary hypertension (CTEPH) develops in a subset of patients after acute pulmonary embolism. In CTEPH, pulmonary vascular resistance, which is initially elevated due to the obstructions in the larger pulmonary arteries, is ... ...

    Abstract Chronic thrombo-embolic pulmonary hypertension (CTEPH) develops in a subset of patients after acute pulmonary embolism. In CTEPH, pulmonary vascular resistance, which is initially elevated due to the obstructions in the larger pulmonary arteries, is further increased by pulmonary microvascular remodeling. The increased afterload of the right ventricle (RV) leads to RV dilation and hypertrophy. This RV remodeling predisposes to arrhythmogenesis and RV failure. Yet, mechanisms involved in pulmonary microvascular remodeling, processes underlying the RV structural and functional adaptability in CTEPH as well as determinants of the susceptibility to arrhythmias such as atrial fibrillation in the context of CTEPH remain incompletely understood. Several large animal models with critical clinical features of human CTEPH and subsequent RV remodeling have relatively recently been developed in swine, sheep, and dogs. In this review we will discuss the current knowledge on the processes underlying development and progression of CTEPH, and on how animal models can help enlarge understanding of these processes.
    Language English
    Publishing date 2021-04-16
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2781496-8
    ISSN 2297-055X
    ISSN 2297-055X
    DOI 10.3389/fcvm.2021.574360
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article: Cardiac Macrophages and Their Effects on Arrhythmogenesis.

    Xia, Ruibing / Tomsits, Philipp / Loy, Simone / Zhang, Zhihao / Pauly, Valerie / Schüttler, Dominik / Clauss, Sebastian

    Frontiers in physiology

    2022  Volume 13, Page(s) 900094

    Abstract: Cardiac electrophysiology is a complex system established by a plethora of inward and outward ion currents in cardiomyocytes generating and conducting electrical signals in the heart. However, not only cardiomyocytes but also other cell types can ... ...

    Abstract Cardiac electrophysiology is a complex system established by a plethora of inward and outward ion currents in cardiomyocytes generating and conducting electrical signals in the heart. However, not only cardiomyocytes but also other cell types can modulate the heart rhythm. Recently, cardiac macrophages were demonstrated as important players in both electrophysiology and arrhythmogenesis. Cardiac macrophages are a heterogeneous group of immune cells including resident macrophages derived from embryonic and fetal precursors and recruited macrophages derived from circulating monocytes from the bone marrow. Recent studies suggest antiarrhythmic as well as proarrhythmic effects of cardiac macrophages. The proposed mechanisms of how cardiac macrophages affect electrophysiology vary and include both direct and indirect interactions with other cardiac cells. In this review, we provide an overview of the different subsets of macrophages in the heart and their possible interactions with cardiomyocytes under both physiologic conditions and heart disease. Furthermore, we elucidate similarities and differences between human, murine and porcine cardiac macrophages, thus providing detailed information for researchers investigating cardiac macrophages in important animal species for electrophysiologic research. Finally, we discuss the pros and cons of mice and pigs to investigate the role of cardiac macrophages in arrhythmogenesis from a translational perspective.
    Language English
    Publishing date 2022-06-22
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2564217-0
    ISSN 1664-042X
    ISSN 1664-042X
    DOI 10.3389/fphys.2022.900094
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top