Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 6890, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767269

ABSTRACT

Clinical use of human mesenchymal stem cells (hMSCs) is limited due to their rapid clearance, reducing their therapeutic efficacy. The inflammatory cytokine IL-1ß activates hMSCs and is known to enhance their engraftment. Consequently, understanding the molecular mechanism of this inflammation-triggered adhesion is of great clinical interest to improving hMSC retention at sites of tissue damage. Integrins are cell-matrix adhesion receptors, and clustering of integrins at the nanoscale underlies cell adhesion. Here, we found that IL-1ß enhances adhesion of hMSCs via increased focal adhesion contacts in an α5ß1 integrin-specific manner. Further, through quantitative super-resolution imaging we elucidated that IL-1ß specifically increases nanoscale integrin α5ß1 availability and clustering at the plasma membrane, whilst conserving cluster area. Taken together, these results demonstrate that hMSC adhesion via IL-1ß stimulation is partly regulated through integrin α5ß1 spatial organization at the cell surface. These results provide new insight into integrin clustering in inflammation and provide a rational basis for design of therapies directed at improving hMSC engraftment.


Subject(s)
Bone Marrow Cells/physiology , Cell Adhesion , Extracellular Matrix/metabolism , Integrin alpha5beta1/metabolism , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/physiology , Bone Marrow Cells/cytology , Cell Membrane/metabolism , Cell Movement , Fibronectins/metabolism , Humans , Integrin alpha5beta1/genetics , Mesenchymal Stem Cells/cytology
2.
Nanoscale ; 12(38): 19844-19854, 2020 Oct 14.
Article in English | MEDLINE | ID: mdl-32969445

ABSTRACT

Extracellular vesicles (EVs) represent a promising cell-free alternative for treatment of cardiovascular diseases. Nevertheless, the lack of standardised and reproducible isolation methods capable of recovering pure, intact EVs presents a significant obstacle. Additionally, there is significant interest in investigating the interactions of EVs with different cardiac cell types. Here we established a robust technique for the production and isolation of EVs harvested from an enriched (>97% purity) population of human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) with size exclusion chromatography. Utilizing an advanced fluorescence labelling strategy, we then investigated the interplay of the CM-EVs with the three major cellular components of the myocardium (fibroblasts, cardiomyocytes and endothelial cells) and identified that cardiac endothelial cells show preferential uptake of these EVs. Overall, our findings provide a great opportunity to overcome the translational hurdles associated with the isolation of intact, non-aggregated human iPSC-CM EVs at high purity. Furthermore, understanding in detail the interaction of the secreted EVs with their surrounding cells in the heart may open promising new avenues in the field of EV engineering for targeted delivery in cardiac regeneration.


Subject(s)
Extracellular Vesicles , Induced Pluripotent Stem Cells , Biological Transport , Endothelial Cells , Extracellular Vesicles/metabolism , Humans , Myocytes, Cardiac
3.
Adv Mater ; 32(6): e1904598, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31833108

ABSTRACT

The ability to manipulate cellular organization within soft materials has important potential in biomedicine and regenerative medicine; however, it often requires complex fabrication procedures. Here, a simple, cost-effective, and one-step approach that enables the control of cell orientation within 3D collagen hydrogels is developed to dynamically create various tailored microstructures of cardiac tissues. This is achieved by incorporating iron oxide nanoparticles into human cardiomyocytes and applying a short-term external magnetic field to orient the cells along the applied field to impart different shapes without any mechanical support. The patterned constructs are viable and functional, can be detected by T2 *-weighted magnetic resonance imaging, and induce no alteration to normal cardiac function after grafting onto rat hearts. This strategy paves the way to creating customized, macroscale, 3D tissue constructs with various cell-types for therapeutic and bioengineering applications, as well as providing powerful models for investigating tissue behavior.


Subject(s)
Collagen/chemistry , Magnetite Nanoparticles/chemistry , Myocytes, Cardiac/cytology , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Cell Line , Equipment Design , Humans , Hydrogels/chemistry , Magnetic Fields , Tissue Engineering/methods
4.
Adv Mater ; 29(26)2017 Jul.
Article in English | MEDLINE | ID: mdl-28474844

ABSTRACT

Inspired by biological systems, many biomimetic methods suggest fabrication of functional materials with unique physicochemical properties. Such methods frequently generate organic-inorganic composites that feature highly ordered hierarchical structures with intriguing properties, distinct from their individual components. A striking example is that of DNA-inorganic hybrid micro/nanostructures, fabricated by the rolling circle technique. Here, a novel concept for the encapsulation of bioactive proteins in DNA flowers (DNF) while maintaining the activity of protein payloads is reported. A wide range of proteins, including enzymes, can be simultaneously associated with the growing DNA strands and Mg2 PPi crystals during the rolling circle process, ultimately leading to the direct immobilization of proteins into DNF. The unique porous structure of this construct, along with the abundance of Mg ions and DNA molecules present, provides many interaction sites for proteins, enabling high loading efficiency and enhanced stability. Further, as a proof of concept, it is demonstrated that the DNF can deliver payloads of cytotoxic protein (i.e., RNase A) to the cells without a loss in its biological function and structural integrity, resulting in highly increased cell death compared to the free protein.


Subject(s)
DNA/chemistry , Flowers , Nanostructures , Proteins
5.
Chem Mater ; 28(17): 6080-6088, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27656042

ABSTRACT

Poly(ethylene dioxythiophene) with functional pendant groups bearing double bonds is synthesized and employed for the fabrication of electroactive hydrogels with advantageous characteristics: covalently cross-linked porous 3D scaffolds with notable swelling ratio, appropriate mechanical properties, electroactivity in physiological conditions, and suitability for proliferation and differentiation of C2C12 cells. This is a new approach for the fabrication of conductive engineered constructs.

6.
Eur Heart J ; 34(21): 1575-86, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22621821

ABSTRACT

AIMS: Myocardial cell replacement therapies are hampered by a paucity of sources for human cardiomyocytes and by the expected immune rejection of allogeneic cell grafts. The ability to derive patient-specific human-induced pluripotent stem cells (hiPSCs) may provide a solution to these challenges. We aimed to derive hiPSCs from heart failure (HF) patients, to induce their cardiomyocyte differentiation, to characterize the generated hiPSC-derived cardiomyocytes (hiPSC-CMs), and to evaluate their ability to integrate with pre-existing cardiac tissue. METHODS AND RESULTS: Dermal fibroblasts from two HF patients were reprogrammed by retroviral delivery of Oct4, Sox2, and Klf4 or by using an excisable polycistronic lentiviral vector. The resulting HF-hiPSCs displayed adequate reprogramming properties and could be induced to differentiate into cardiomyocytes with the same efficiency as control hiPSCs (derived from human foreskin fibroblasts). Gene expression and immunostaining studies confirmed the cardiomyocyte phenotype of the differentiating HF-hiPSC-CMs. Multi-electrode array recordings revealed the development of a functional cardiac syncytium and adequate chronotropic responses to adrenergic and cholinergic stimulation. Next, functional integration and synchronized electrical activities were demonstrated between hiPSC-CMs and neonatal rat cardiomyocytes in co-culture studies. Finally, in vivo transplantation studies in the rat heart revealed the ability of the HF-hiPSC-CMs to engraft, survive, and structurally integrate with host cardiomyocytes. CONCLUSIONS: Human-induced pluripotent stem cells can be established from patients with advanced heart failure and coaxed to differentiate into cardiomyocytes, which can integrate with host cardiac tissue. This novel source for patient-specific heart cells may bring a unique value to the emerging field of cardiac regenerative medicine.


Subject(s)
Heart Failure/pathology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Cell Differentiation , Cell Survival , Cellular Reprogramming/drug effects , Female , Genetic Vectors , Heart Failure/therapy , Humans , Induced Pluripotent Stem Cells/transplantation , Karyotype , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/pharmacology , Octamer Transcription Factor-3/pharmacology , Rats , Rats, Sprague-Dawley , SOXB1 Transcription Factors/pharmacology , Transgenes , Transplantation, Heterologous
7.
Cell Mol Life Sci ; 69(19): 3285-99, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22960788

ABSTRACT

Myocardial stem cell therapies are emerging as novel therapeutic paradigms for myocardial repair, but are hampered by the lack of sources for autologous human cardiomyocytes. An exciting development in the field of cardiovascular regenerative medicine is the ability to reprogram adult somatic cells into pluripotent stem cell lines (induced pluripotent stem cells, iPSCs) and to coax their differentiation into functional cardiomyocytes. This technology holds great promise for the emerging disciplines of personalized and regenerative medicine, because of the ability to derive patient-specific iPSCs that could potentially elude the immune system. The current review describes the latest techniques of generating iPSCs as well as the methods used to direct their differentiation towards the cardiac lineage. We then detail the unique potential as well as the possible hurdles on the road to clinical utilizing of the iPSCs derived cardiomyocytes in the emerging field of cardiovascular regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Myocardium/cytology , Myocytes, Cardiac/cytology , Regenerative Medicine/methods , Animals , Cell Differentiation , Fibroblasts/cytology , Heart Diseases/therapy , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/physiology , Stem Cell Transplantation/methods , Tissue Engineering/methods
8.
PLoS One ; 6(4): e18037, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21483779

ABSTRACT

BACKGROUND: The ability to establish human induced pluripotent stem cells (hiPSCs) by reprogramming of adult fibroblasts and to coax their differentiation into cardiomyocytes opens unique opportunities for cardiovascular regenerative and personalized medicine. In the current study, we investigated the Ca(2+)-handling properties of hiPSCs derived-cardiomyocytes (hiPSC-CMs). METHODOLOGY/PRINCIPAL FINDINGS: RT-PCR and immunocytochemistry experiments identified the expression of key Ca(2+)-handling proteins. Detailed laser confocal Ca(2+) imaging demonstrated spontaneous whole-cell [Ca(2+)](i) transients. These transients required Ca(2+) influx via L-type Ca(2+) channels, as demonstrated by their elimination in the absence of extracellular Ca(2+) or by administration of the L-type Ca(2+) channel blocker nifedipine. The presence of a functional ryanodine receptor (RyR)-mediated sarcoplasmic reticulum (SR) Ca(2+) store, contributing to [Ca(2+)](i) transients, was established by application of caffeine (triggering a rapid increase in cytosolic Ca(2+)) and ryanodine (decreasing [Ca(2+)](i)). Similarly, the importance of Ca(2+) reuptake into the SR via the SR Ca(2+) ATPase (SERCA) pump was demonstrated by the inhibiting effect of its blocker (thapsigargin), which led to [Ca(2+)](i) transients elimination. Finally, the presence of an IP3-releasable Ca(2+) pool in hiPSC-CMs and its contribution to whole-cell [Ca(2+)](i) transients was demonstrated by the inhibitory effects induced by the IP3-receptor blocker 2-Aminoethoxydiphenyl borate (2-APB) and the phospholipase C inhibitor U73122. CONCLUSIONS/SIGNIFICANCE: Our study establishes the presence of a functional, SERCA-sequestering, RyR-mediated SR Ca(2+) store in hiPSC-CMs. Furthermore, it demonstrates the dependency of whole-cell [Ca(2+)](i) transients in hiPSC-CMs on both sarcolemmal Ca(2+) entry via L-type Ca(2+) channels and intracellular store Ca(2+) release.


Subject(s)
Calcium/metabolism , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Animals , Biological Transport , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Line , Gene Expression Regulation , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Intracellular Space/metabolism , Mice , Myocytes, Cardiac/enzymology , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcolemma/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
9.
Nature ; 471(7337): 225-9, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21240260

ABSTRACT

The ability to generate patient-specific human induced pluripotent stem cells (iPSCs) offers a new paradigm for modelling human disease and for individualizing drug testing. Congenital long QT syndrome (LQTS) is a familial arrhythmogenic syndrome characterized by abnormal ion channel function and sudden cardiac death. Here we report the development of a patient/disease-specific human iPSC line from a patient with type-2 LQTS (which is due to the A614V missense mutation in the KCNH2 gene). The generated iPSCs were coaxed to differentiate into the cardiac lineage. Detailed whole-cell patch-clamp and extracellular multielectrode recordings revealed significant prolongation of the action-potential duration in LQTS human iPSC-derived cardiomyocytes (the characteristic LQTS phenotype) when compared to healthy control cells. Voltage-clamp studies confirmed that this action-potential-duration prolongation stems from a significant reduction of the cardiac potassium current I(Kr). Importantly, LQTS-derived cells also showed marked arrhythmogenicity, characterized by early-after depolarizations and triggered arrhythmias. We then used the LQTS human iPSC-derived cardiac-tissue model to evaluate the potency of existing and novel pharmacological agents that may either aggravate (potassium-channel blockers) or ameliorate (calcium-channel blockers, K(ATP)-channel openers and late sodium-channel blockers) the disease phenotype. Our study illustrates the ability of human iPSC technology to model the abnormal functional phenotype of an inherited cardiac disorder and to identify potential new therapeutic agents. As such, it represents a promising paradigm to study disease mechanisms, optimize patient care (personalized medicine), and aid in the development of new therapies.


Subject(s)
Drug Evaluation, Preclinical/methods , Induced Pluripotent Stem Cells/pathology , Long QT Syndrome/pathology , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Adult , Cell Transdifferentiation , Cells, Cultured , Cellular Reprogramming/genetics , ERG1 Potassium Channel , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Female , Fibroblasts/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Long QT Syndrome/classification , Long QT Syndrome/drug therapy , Long QT Syndrome/genetics , Mutation, Missense/genetics , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Phenotype , Precision Medicine/methods
10.
Tissue Eng Part A ; 17(7-8): 1027-37, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21087204

ABSTRACT

Cardiomyocytes derived from induced pluripotent stem (iPS) cells hold great promise for basic and translational cardiovascular research. For the successful implementation of this unique technology, however, it is essential to establish efficient, reproducible, and safe strategies to produce cardiomyocytes in a scalable manner. The aim of the current study was to establish scalable bioprocess that allows direct embryoid bodies formation for the differentiation of murine iPS cells (generated without the oncogene c-Myc) into cardiomyocytes. The cardiomyocytes' structural, molecular, and functional properties were then compared to ones derived by the well-established static culture system. Similar gene expression patterns were observed in both differentiation systems with the sequential expression of mesoderm markers, cardiac transcription factors, and cardiomyocyte structural genes. Cells in the contracting embryoid bodies were stained positively for cardiac troponin-I, sarcomeric α-actinin, cardiac troponin-T, and connexin-43. Electrophysiological measurements using multielectrode array recordings demonstrated that the bioreactor-derived cardiomyocytes were functionally similar to static derived cardiomyocytes and responded appropriately to different drugs, including adrenergic and muscarinic agonists (isoproterenol and carbamylcholine, respectively) and the gap junction uncoupler heptanol. Our study describes, for the first time, a strategy for scalable differentiation of c-Myc-free iPS cells into cardiomyocytes with the appropriate molecular, structural, and functional properties. The result of this study should have important implications for several cardiovascular research areas and specifically for the emerging field of regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Differentiation/physiology , Cell Line , Flow Cytometry , Induced Pluripotent Stem Cells/metabolism , Mice , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-myc/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...