Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Physiol ; 13: 1023563, 2022.
Article in English | MEDLINE | ID: mdl-36439258

ABSTRACT

Cardiac contractility modulation (CCM) is a medical device therapy whereby non-excitatory electrical stimulations are delivered to the myocardium during the absolute refractory period to enhance cardiac function. We previously evaluated the effects of the standard CCM pulse parameters in isolated rabbit ventricular cardiomyocytes and 2D human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) monolayers, on flexible substrate. In the present study, we sought to extend these results to human 3D microphysiological systems to develop a robust model to evaluate various clinical CCM pulse parameters in vitro. HiPSC-CMs were studied in conventional 2D monolayer format, on stiff substrate (i.e., glass), and as 3D human engineered cardiac tissues (ECTs). Cardiac contractile properties were evaluated by video (i.e., pixel) and force-based analysis. CCM pulses were assessed at varying electrical 'doses' using a commercial pulse generator. A robust CCM contractile response was observed for 3D ECTs. Under comparable conditions, conventional 2D monolayer hiPSC-CMs, on stiff substrate, displayed no contractile response. 3D ECTs displayed enhanced contractile properties including increased contraction amplitude (i.e., force), and accelerated contraction and relaxation slopes under standard acute CCM stimulation. Moreover, 3D ECTs displayed enhanced contractility in a CCM pulse parameter-dependent manner by adjustment of CCM pulse delay, duration, amplitude, and number relative to baseline. The observed acute effects subsided when the CCM stimulation was stopped and gradually returned to baseline. These data represent the first study of CCM in 3D hiPSC-CM models and provide a nonclinical tool to assess various CCM device signals in 3D human cardiac tissues prior to in vivo animal studies. Moreover, this work provides a foundation to evaluate the effects of additional cardiac medical devices in 3D ECTs.

2.
J Pharmacol Toxicol Methods ; 105: 106886, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32629159

ABSTRACT

To develop therapeutics for cardiovascular disease, especially heart failure, translational models for assessing cardiac contractility are necessary for preclinical target validation and lead optimization. The availability of stem cell-derived cardiomyocytes (SC-CM) has generated a great opportunity in developing new in-vitro models for assessing cardiac contractility. However, the immature phenotype of SC-CM is a well-recognized limitation in inotropic evaluation, especially regarding the lack of or diminished positive inotropic response to ß-adrenergic agonists. Recent development of 3D engineered cardiac tissues (ECTs) using human induced pluripotent stem cell derived-cardiomyocytes (hiPSC-CM) in the BiowireTM II platform has shown improved maturation. To evaluate their suitability to detect drug-induced changes in cardiac contractility, positive inotropes with diverse mechanisms, including ß-adrenergic agonists, PDE3 inhibitors, Ca2+-sensitizers, myosin and troponin activators, and an apelin receptor agonist, were tested blindly. A total of 8 compounds were evaluated, including dobutamine, milrinone, pimobendan, levosimendan, omecamtiv mecarbil, AMG1, AMG2, and pyr-apelin-13. Contractility was evaluated by analyzing the amplitude, velocity and duration of contraction and relaxation. All tested agents, except pyr-apelin-13, increased contractility by increasing the amplitude of contraction and velocity. In addition, myosin and troponin activators increase contraction duration. These results indicate that ECTs generated in the BiowireTM II platform can identify inotropes with different mechanisms and provides a human-based in-vitro model for evaluating potential therapeutics.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Regeneration/physiology , Tissue Scaffolds/chemistry , Cells, Cultured , Humans , Myocardial Contraction/physiology
3.
Toxicol Sci ; 172(1): 89-97, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31385592

ABSTRACT

Recent advances in techniques to differentiate human induced pluripotent stem cells (hiPSCs) hold the promise of an unlimited supply of human derived cardiac cells from both healthy and disease populations. That promise has been tempered by the observation that hiPSC-derived cardiomyocytes (hiPSC-CMs) typically retain a fetal-like phenotype, raising concern about the translatability of the in vitro data obtained to drug safety, discovery, and development studies. The Biowire II platform was used to generate 3D engineered cardiac tissues (ECTs) from hiPSC-CMs and cardiac fibroblasts. Long term electrical stimulation was employed to obtain ECTs that possess a phenotype like that of adult human myocardium including a lack of spontaneous beating, the presence of a positive force-frequency response from 1 to 4 Hz and prominent postrest potentiation. Pharmacology studies were performed in the ECTs to confirm the presence and functionality of pathways that modulate cardiac contractility in humans. Canonical responses were observed for compounds that act via the ß-adrenergic/cAMP-mediated pathway, eg, isoproterenol and milrinone; the L-type calcium channel, eg, FPL64176 and nifedipine; and indirectly effect intracellular Ca2+ concentrations, eg, digoxin. Expected positive inotropic responses were observed for compounds that modulate proteins of the cardiac sarcomere, eg, omecamtiv mecarbil and levosimendan. ECTs generated in the Biowire II platform display adult-like properties and have canonical responses to cardiotherapeutic and cardiotoxic agents that affect contractility in humans via a variety of mechanisms. These data demonstrate that this human-based model can be used to assess the effects of novel compounds on contractility early in the drug discovery and development process.

4.
J Tissue Eng Regen Med ; 12(2): 349-359, 2018 02.
Article in English | MEDLINE | ID: mdl-28482139

ABSTRACT

One of the main efforts in myocardial tissue engineering is towards designing cardiac tissues able to rescue the reduction in heart function once implanted at the site of myocardial infarction. To date, the efficiency of this approach in preclinical applications is limited in part by our incomplete understanding of the inflammatory environment known to be present at the site of myocardial infarct and by poor vascularization. It was recently reported that polarized macrophages known to be present at the site of myocardial infarction secrete bone morphogenetic proteins (BMPs)-2 and -4 causing changes in the expression of cardiac proteins in a 2D in vitro model. Here, these findings were extended towards cardiac tissues composed of human embryonic stem cell derived cardiomyocytes embedded in collagen gel. By preconditioning cardiac tissues with BMPs, constructs were obtained with enhanced expression of cardiac markers. Additionally, after BMP preconditioning, the resulting cardiac-tissues were able to sustain diffusion of the BMPs with the added benefit of supporting human umbilical vein endothelial cell tube formation. Here, a model is proposed of cardiac tissues preconditioned with BMPs that results in stimulation of cardiomyocyte function and diffusion of BMPs able to support angiogenesis. This platform represents a step towards the validation of more complex bioengineered constructs for in vivo applications.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Imaging, Three-Dimensional , Models, Biological , Myocardium/pathology , Neovascularization, Physiologic/drug effects , Wound Healing/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Gene Expression Regulation/drug effects , Heart, Artificial , Human Umbilical Vein Endothelial Cells/metabolism , Humans
5.
J Tissue Eng Regen Med ; 11(5): 1466-1478, 2017 05.
Article in English | MEDLINE | ID: mdl-26103914

ABSTRACT

Following cardiac injury, the ischaemic heart tissue is characterized by the invasion of pro-inflammatory (M1) and pro-healing (M2) macrophages. Any engineered cardiac tissue will inevitably interact with the inflammatory environment found at the site of myocardial infarction at the time of implantation. However, the interactions between the inflammatory and the cardiac repair cells remain poorly understood. Here we recapitulated in vitro some of the important cellular events found at the site of myocardial injury, such as macrophage recruitment and their effect on cardiac differentiation and maturation, by taking into account the involvement of paracrine-mediated signalling. By using a 3D inverted invasion assay, we found that cardiomyocyte (CM) conditioned medium can trigger the recruitment of pro-inflammatory (M1) macrophages, through a mechanism that involves, in part, CM-derived BMP4. Pro-inflammatory (M1) macrophages were also found to affect CM proliferation and differentiation potential, in part due to BMP molecules secreted by macrophages. These effects involved the activation of the canonical outside-in signalling pathways, such as SMAD1,5,8, which are known to be activated during myocardial injury in vivo. In the present study we propose a new role for CM- and macrophage-derived BMP proteins during the recruitment of macrophage subtypes and the maturation of repair cells, representing an important step towards creating a functional cardiac patch with superior therapeutic properties. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Cell Communication , Macrophages/metabolism , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Macrophages/pathology , Myocytes, Cardiac/pathology , Pluripotent Stem Cells/pathology , Smad Proteins/metabolism
6.
Exp Cell Res ; 347(1): 1-13, 2016 09 10.
Article in English | MEDLINE | ID: mdl-26500109

ABSTRACT

The mechanisms by which macrophages control the inflammatory response, wound healing, biomaterial-interactions, and tissue regeneration appear to be related to their activation/differentiation states. Studies of macrophage behavior in vitro can be useful for elucidating their mechanisms of action, but it is not clear to what extent the source of macrophages affects their apparent behavior, potentially affecting interpretation of results. Although comparative studies of macrophage behavior with respect to cell source have been conducted, there has been no direct comparison of the three most commonly used cell sources: murine bone marrow, human monocytes from peripheral blood (PB), and the human leukemic monocytic cell line THP-1, across multiple macrophage phenotypes. In this study, we used multivariate discriminant analysis to compare the in vitro expression of genes commonly chosen to assess macrophage phenotype across all three sources of macrophages, as well as those derived from induced pluripotent stem cells (iPSCs), that were polarized towards four distinct phenotypes using the same differentiation protocols: M(LPS,IFN) (aka M1), M(IL4,IL13) (aka M2a), M(IL10) (aka M2c), and M(-) (aka M0) used as control. Several differences in gene expression trends were found among the sources of macrophages, especially between murine bone marrow-derived and human blood-derived M(LPS,IFN) and M(IL4,IL13) macrophages with respect to commonly used phenotype markers like CCR7 and genes associated with angiogenesis and tissue regeneration like FGF2 and MMP9. We found that the genes with the most similar patterns of expression among all sources were CXCL-10 and CXCL-11 for M(LPS,IFN) and CCL17 and CCL22 for M(IL4,IL13). Human PB-derived macrophages and human iPSC-derived macrophages showed similar gene expression patterns among the groups and genes studied here, suggesting that iPSC-derived monocytes have the potential to be used as a reliable cell source of human macrophages for in vitro studies. These findings could help select appropriate markers when testing macrophage behavior in vitro and highlight those markers that may confuse interpretation of results from experiments employing macrophages from different sources.


Subject(s)
Cell Polarity/genetics , Gene Expression Profiling , Macrophages/cytology , Macrophages/metabolism , Animals , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Polarity/drug effects , Discriminant Analysis , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/pharmacology , Least-Squares Analysis , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice, Inbred BALB C , Phenotype
7.
Biomaterials ; 35(11): 3678-87, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24480538

ABSTRACT

Platelet gel, a fibrin network containing activated platelets, is widely used in regenerative medicine due the capacity of platelet-derived growth factors to accelerate and direct healing processes. However, limitations to this approach include poor mechanical properties, relatively rapid degradation, and the lack of control of release of growth factors at the site of injection. These issues compromise the ability of platelet gels for sustained function in regenerative medicine. In the present study, a combination of platelet gels with silk fibroin gel was studied to address the above limitations. Mixing sonicated silk gels with platelet gels extended the release of growth factors without inhibiting gel-forming ability. The released growth factors were biologically active and their delivery was modified further by manipulation of the charge of the silk protein. Moreover, the silk gel augmented both the rheological properties and compressive stiffness of the platelet gel, tuned by the silk concentration and/or silk/platelet gel ratio. Silk-platelet gel injections in nude rats supported enhanced cell infiltration and blood vessel formation representing a step towards new platelet gel formulations with enhanced therapeutic impact.


Subject(s)
Blood Platelets/chemistry , Gels/pharmacology , Silk/pharmacology , Animals , Cell Proliferation/drug effects , Compressive Strength/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , MAP Kinase Signaling System/drug effects , Platelet-Derived Growth Factor/metabolism , Rats , Rats, Nude , Rheology/drug effects , Time Factors , Vascular Endothelial Growth Factor A/metabolism
8.
Haematologica ; 98(4): 514-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23403314

ABSTRACT

Megakaryocytes release platelets into the bloodstream by elongating proplatelets. In this study, we showed that human megakaryocytes constitutively release Transforming Growth Factor ß1 and express its receptors. Importantly, Transforming Growth Factor ß1 downstream signaling, through SMAD2/3 phosphorylation, was shown to be active in megakaryocytes extending proplatelets, indicating a type of autocrine stimulation on megakaryocyte development. Furthermore, inactivation of Transforming Growth Factor ß1 signaling, by the receptor inhibitors SB431542 and Stemolecule ALK5 inhibitor, determined a significant decrease in proplatelet formation. Recent studies indicated a crucial role of Transforming Growth Factor ß1 in the pathogenesis of primary myelofibrosis. We demonstrated that primary myelofibrosis-derived megakaryocytes expressed increased levels of bioactive Transforming Growth Factor ß1; however, higher levels of released Transforming Growth Factor ß1 did not lead to enhanced activation of downstream pathways. Overall, these data propose Transforming Growth Factor ß1 as a new element in the autocrine regulation of proplatelet formation in vitro. Despite the increase in Transforming Growth Factor ß1 this mechanism seems to be preserved in primary myelofibrosis.


Subject(s)
Autocrine Communication , Megakaryocytes/metabolism , Primary Myelofibrosis/genetics , Transforming Growth Factor beta1/genetics , Benzamides/pharmacology , Blood Platelets/cytology , Blood Platelets/metabolism , Blotting, Western , Cells, Cultured , Dioxoles/pharmacology , Gene Expression , Humans , Megakaryocytes/cytology , Phosphorylation , Primary Myelofibrosis/blood , Primary Myelofibrosis/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
9.
Haematologica ; 97(11): 1657-65, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22689668

ABSTRACT

BACKGROUND: The interaction of adenosine diphosphate with its P2Y(1) and P2Y(12) receptors on platelets is important for platelet function. However, nothing is known about adenosine diphosphate and its function in human megakaryocytes. DESIGN AND METHODS: We studied the role of adenosine diphosphate and P2Y receptors on proplatelet formation by human megakaryocytes in culture. RESULTS: Megakaryocytes expressed all the known eight subtypes of P2Y receptors, and constitutively released adenosine diphosphate. Proplatelet formation was inhibited by the adenosine diphosphate scavengers apyrase and CP/CPK by 60-70% and by the P2Y(12) inhibitors cangrelor and 2-MeSAMP by 50-60%, but was not inhibited by the P2Y(1) inhibitor MRS 2179. However, the active metabolites of the anti-P2Y(12) drugs, clopidogrel and prasugrel, did not inhibit proplatelet formation. Since cangrelor and 2-MeSAMP also interact with P2Y(13), we hypothesized that P2Y(13), rather than P2Y(12) is involved in adenosine diphosphate-regulated proplatelet formation. The specific P2Y(13) inhibitor MRS 2211 inhibited proplatelet formation in a concentration-dependent manner. Megakaryocytes from a patient with severe congenital P2Y(12) deficiency showed normal proplatelet formation, which was inhibited by apyrase, cangrelor or MRS 2211 by 50-60%. The platelet count of patients with congenital delta-storage pool deficiency, who lack secretable adenosine diphosphate, was significantly lower than that of patients with other platelet function disorders, confirming the important role of secretable adenosine diphosphate in platelet formation. CONCLUSIONS: This is the first demonstration that adenosine diphosphate released by megakaryocytes regulates their function by interacting with P2Y(13). The clinical relevance of this not previously described physiological role of adenosine diphosphate and P2Y(13) requires further exploration.


Subject(s)
Adenosine Diphosphate/metabolism , Blood Platelets/metabolism , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/metabolism , Receptors, Purinergic P2Y12/metabolism , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2/metabolism , Apyrase/pharmacology , Blood Platelets/cytology , Cells, Cultured , Female , Fetal Blood , Humans , Male , Megakaryocyte Progenitor Cells/cytology , Megakaryocytes/cytology , Purinergic P2Y Receptor Antagonists/pharmacology
10.
Tissue Eng Part C Methods ; 17(12): 1223-32, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21895494

ABSTRACT

Platelets are specialized cells produced by megakaryocytes in the bone marrow that represent the first defense against hemorrhage, yet they also play a pathological role in thrombosis, inflammation, and cancer. Millions of platelet transfusions are conducted each year, and the supply of this blood component is limited. There are many diseases where platelet production or function is impaired with severe consequences for patients. With such clinical need, new insight into the formation of platelets would have a major impact on patients and healthcare. We developed an innovative 3D system to study platelet production that represents the first spatial reconstruction of the bone marrow environment. In this system human megakaryocytes were able to migrate toward the vascular niche, extend proplatelets, and release functional platelets into vascular tubes. The combination of different bone marrow components and the compliance of silk-based vascular tubes makes this model a unique tool for the study of platelet formation and production for use in healthcare needs.


Subject(s)
Blood Platelets/cytology , Blood Vessels/drug effects , Cell Culture Techniques/methods , Megakaryocytes/cytology , Silk/pharmacology , Tissue Scaffolds/chemistry , Animals , Bioreactors , Blood Platelets/drug effects , Cell Adhesion/drug effects , Cell Movement/drug effects , Humans , Megakaryocytes/drug effects
11.
Blood ; 118(16): 4449-53, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21828129

ABSTRACT

Cell interactions with matrices via specific receptors control many functions, with chemistry, physics, and membrane elasticity as fundamental elements of the processes involved. Little is known about how biochemical and biophysical processes integrate to generate force and, ultimately, to regulate hemopoiesis into the bone marrow-matrix environment. To address this hypothesis, in this work we focus on the regulation of MK development by type I collagen. By atomic force microscopy analysis, we demonstrate that the tensile strength of fibrils in type I collagen structure is a fundamental requirement to regulate cytoskeleton contractility of human MKs through the activation of integrin-α2ß1-dependent Rho-ROCK pathway and MLC-2 phosphorylation. Most importantly, this mechanism seemed to mediate MK migration, fibronectin assembly, and platelet formation. On the contrary, a decrease in mechanical tension caused by N-acetylation of lysine side chains in type I collagen completely reverted these processes by preventing fibrillogenesis.


Subject(s)
Collagen Type I/metabolism , Collagen Type I/ultrastructure , Extracellular Matrix/metabolism , Megakaryocytes/cytology , Cells, Cultured , Collagen Type I/chemistry , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Extracellular Matrix/chemistry , Humans , Integrin alpha2beta1/metabolism , Megakaryocytes/metabolism , Megakaryocytes/ultrastructure , Microscopy, Atomic Force , Tensile Strength , Thrombopoiesis
12.
PLoS One ; 4(12): e8359, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-20027303

ABSTRACT

BACKGROUND: The mechanism by which megakaryocytes (Mks) proliferate, differentiate, and release platelets into circulation are not well understood. Growing evidence indicates that a complex regulatory mechanism, involving cellular interactions, composition of the extracellular matrix and physical parameters such as oxygen tension, may contribute to the quiescent or permissive microenvironment related to Mk differentiation and maturation within the bone marrow. METHODOLOGY/PRINCIPAL FINDINGS: Differentiating human mesenchymal stem cells (hMSCs) into osteoblasts (hOSTs), we established an in vitro model for the osteoblastic niche. We demonstrated for the first time that the combination of HSCs, Mks and hypoxia sustain and promote bone formation by increasing type I collagen release from hOSTs and enhancing its fibrillar organization, as revealed by second harmonic generation microscopy. Through co-culture, we demonstrated that direct cell-cell contact modulates Mk maturation and differentiation. In particular we showed that low oxygen tension and direct interaction of hematopoietic stem cells (HSCs) with hOSTs inhibits Mk maturation and proplatelet formation (PPF). This regulatory mechanism was dependent on the fibrillar structure of type I collagen released by hOSTs and on the resulting engagement of the alpha2beta1 integrin. In contrast, normoxic conditions and the direct interaction of HSCs with undifferentiated hMSCs promoted Mk maturation and PPF, through a mechanism involving the VCAM-1 pathway. CONCLUSIONS/SIGNIFICANCE: By combining cellular, physical and biochemical parameters, we mimicked an in vitro model of the osteoblastic niche that provides a physiological quiescent microenvironment where Mk differentiation and PPF are prevented. These findings serve as an important step in developing suitable in vitro systems to use for the study and manipulation of Mk differentiation and maturation in both normal and diseased states.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation , Megakaryocytes/cytology , Megakaryocytes/physiology , Models, Biological , Osteoblasts/cytology , Blood Platelets/cytology , Blood Platelets/metabolism , Bone Marrow Cells/metabolism , Cell Hypoxia , Cell Shape , Collagen Type I/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Integrin alpha2beta1/metabolism , Integrin alpha4beta1/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...