Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Karolina Åkesson is active.

Publication


Featured researches published by Karolina Åkesson.


Stem Cells | 2008

Molecular Signature of Cardiomyocyte Clusters Derived from Human Embryonic Stem Cells

Jane Synnergren; Karolina Åkesson; Kerstin Dahlenborg; Hilmar Vidarsson; Caroline Améen; Daniella Steel; Anders Lindahl; Björn Olsson; Peter Sartipy

Human embryonic stem cells (hESCs) can differentiate in vitro into spontaneously contracting cardiomyocytes (CMs). These cells may prove extremely useful for various applications in basic research, drug discovery, and regenerative medicine. To fully use the potential of the cells, they need to be extensively characterized, and the regulatory mechanisms that control hESC differentiation toward the cardiac lineage need to be better defined. In this study, we used microarrays to analyze, for the first time, the global gene expression profile of isolated hESC‐derived CM clusters. By comparing the clusters with undifferentiated hESCs and using stringent selection criteria, we identified 530 upregulated and 40 downregulated genes in the contracting clusters. To further characterize the family of upregulated genes in the hESC‐derived CM clusters, the genes were classified according to their Gene Ontology annotation. The results indicate that the hESC‐derived CM clusters display high similarities, on a molecular level, to human heart tissue. Moreover, using the family of upregulated genes, we created protein interaction maps that revealed topological characteristics. We also searched for cellular pathways among the upregulated genes in the hESC‐derived CM clusters and identified eight significantly upregulated pathways. Real‐time quantitative polymerase chain reaction and immunohistochemical analysis confirmed the expression of a subset of the genes identified by the microarrays. Taken together, the results presented here provide a molecular signature of hESC‐derived CM clusters and further our understanding of the biological processes that are active in these cells.


Experimental Biology and Medicine | 2006

Molecular and Pharmacological Properties of Human Embryonic Stem Cell–Derived Cardiomyocytes

Anders Norström; Karolina Åkesson; Thorir Hardarson; L. Hamberger; Petter Björquist; Peter Sartipy

Human embryonic stem cells (hESCs) can be coaxed to differentiate Into specific cell types, including cardiomyocyte-like cells. These cells express cardiac-specific markers and display functional similarities to their adult counterparts. Based on these properties, hESC-derived cardiomyocytes have the potential to be extremely useful in various in vitro applications and to provide the opportunity for cardiac cell replacement therapies. However, before this can become a reality, the molecular and functional characteristics of these cells need to be Investigated in more detail. In the present study we differentiate hESCs into cardiomyocyte-like cells via embryoid bodies (EBs). The fraction of spontaneously beating clusters obtained from the EBs averaged approximately 30% of the total number of EBs used. These cell clusters were isolated, dissociated into single-cell suspensions, and frozen for long-term storage. The cryopreserved cells could be successfully thawed and subcultured. Using electron microscopy, we observed Z discs and tight Junctions in the hESC-derived cardiomyocytes, and by Immunohistochemical analysis we detected expression of cardiac-specific markers (cTnl and cMHC). Notably, using BrdU labeling we also could demonstrate that some of the hESC-derived cardiomyocytes retain a proliferative capacity. Furthermore, pharmacological stimulation of the cells resulted in responses Indicative of functional adrenergic and muscarinic receptor coupling systems. Taken together, these results lend support to the notion that hESCs can be used as a source for the procurement of cardiomyocytes for in vitro and in vivo applications.


Toxicology | 2015

Identification of novel biomarkers for doxorubicin-induced toxicity in human cardiomyocytes derived from pluripotent stem cells

Gustav Holmgren; Jane Synnergren; Yalda Bogestål; Caroline Améen; Karolina Åkesson; Sandra Holmgren; Anders Lindahl; Peter Sartipy

Doxorubicin is a chemotherapeutic agent indicated for the treatment of a variety of cancer types, including leukaemia, lymphomas, and many solid tumours. The use of doxorubicin is, however, associated with severe cardiotoxicity, often resulting in early discontinuation of the treatment. Importantly, the toxic symptoms can occur several years after the termination of the doxorubicin administration. In this study, the toxic effects of doxorubicin exposure have been investigated in cardiomyocytes derived from human embryonic stem cells (hESC). The cells were exposed to different concentrations of doxorubicin for up to 2 days, followed by a 12 day recovery period. Notably, the cell morphology was altered during drug treatment and the cells showed a reduced contractile ability, most prominent at the highest concentration of doxorubicin at the later time points. A general cytotoxic response measured as Lactate dehydrogenase leakage was observed after 2 days’ exposure compared to the vehicle control, but this response was absent during the recovery period. A similar dose-dependant pattern was observed for the release of cardiac specific troponin T (cTnT) after 1 day and 2 days of treatment with doxorubicin. Global transcriptional profiles in the cells revealed clusters of genes that were differentially expressed during doxorubicin exposure, a pattern that in some cases was sustained even throughout the recovery period, suggesting that these genes could be used as sensitive biomarkers for doxorubicin-induced toxicity in human cardiomyocytes. The results from this study show that cTnT release can be used as a measurement of acute cardiotoxicity due to doxorubicin. However, for the late onset of doxorubicin-induced cardiomyopathy, cTnT release might not be the most optimal biomarker. As an alternative, some of the genes that we identified as differentially expressed after doxorubicin exposure could serve as more relevant biomarkers, and may also help to explain the cellular mechanisms behind the late onset apoptosis associated with doxorubicin-induced cardiomyopathy.


PLOS ONE | 2017

A data analysis framework for biomedical big data: Application on mesoderm differentiation of human pluripotent stem cells

Benjamin Ulfenborg; Alexander Karlsson; Maria Riveiro; Caroline Améen; Karolina Åkesson; Christian X. Andersson; Peter Sartipy; Jane Synnergren

The development of high-throughput biomolecular technologies has resulted in generation of vast omics data at an unprecedented rate. This is transforming biomedical research into a big data discipline, where the main challenges relate to the analysis and interpretation of data into new biological knowledge. The aim of this study was to develop a framework for biomedical big data analytics, and apply it for analyzing transcriptomics time series data from early differentiation of human pluripotent stem cells towards the mesoderm and cardiac lineages. To this end, transcriptome profiling by microarray was performed on differentiating human pluripotent stem cells sampled at eleven consecutive days. The gene expression data was analyzed using the five-stage analysis framework proposed in this study, including data preparation, exploratory data analysis, confirmatory analysis, biological knowledge discovery, and visualization of the results. Clustering analysis revealed several distinct expression profiles during differentiation. Genes with an early transient response were strongly related to embryonic- and mesendoderm development, for example CER1 and NODAL. Pluripotency genes, such as NANOG and SOX2, exhibited substantial downregulation shortly after onset of differentiation. Rapid induction of genes related to metal ion response, cardiac tissue development, and muscle contraction were observed around day five and six. Several transcription factors were identified as potential regulators of these processes, e.g. POU1F1, TCF4 and TBP for muscle contraction genes. Pathway analysis revealed temporal activity of several signaling pathways, for example the inhibition of WNT signaling on day 2 and its reactivation on day 4. This study provides a comprehensive characterization of biological events and key regulators of the early differentiation of human pluripotent stem cells towards the mesoderm and cardiac lineages. The proposed analysis framework can be used to structure data analysis in future research, both in stem cell differentiation, and more generally, in biomedical big data analytics.


Archive | 2007

Novel population of multipotent cardiac precursor cells derived from human blastocysts derived stem cells

Peter Sartipy; Karolina Åkesson; Caroline Améen


Archive | 2008

Cardiomyocyte-like cell clusters derived from hbs cells

Peter Sartipy; Karolina Åkesson; Caroline Améen; Jane Synnergren; Kerstin Dahlenborg; Daniella Steel


Journal of Pharmacological and Toxicological Methods | 2018

Evaluation of human iPS cell-derived cardiomyocytes in high-throughput toxicity screening applications

Caroline Améen; Kerstin Dahlenborg; Karolina Åkesson; Maria Flood; Anders Aspegren; Krisztina Juhasz


Journal of Pharmacological and Toxicological Methods | 2017

Physiological and Reproducible Cardiotoxic Response in Cardiomyocytes Analyzed with MEA Technology

Yuki Yamamoto; Kerstin Dahlenborg; Karolina Åkesson; Maria Flood; Anders Aspegren; Tatsuji Enoki; Caroline Améen


Journal of Pharmacological and Toxicological Methods | 2015

Doxorubicin-induced toxicity in cardiomyocytes derived from human pluripotent stem cells

Gustav Holmgren; Jane Synnergren; Yalda Bogestål; Caroline Améen; Karolina Åkesson; Sandra Holmgren; Anders Lindahl; Peter Sartipy


Journal of Pharmacological and Toxicological Methods | 2008

Global gene expression profiling of human embryonic stem cell derived cardiomyocytes

Daniella Steel; Jane Synnergren; Karolina Åkesson; Kerstin Dahlenborg; Caroline Améen; Peter Sartipy

Collaboration


Dive into the Karolina Åkesson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anders Lindahl

Sahlgrenska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anders Norström

Sahlgrenska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge