Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Europace ; 26(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38788213

ABSTRACT

AIMS: Human induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCM) could be a helpful tool to study the physiology and diseases of the human atrium. To fulfil this expectation, the electrophysiology of hiPSC-aCM should closely resemble the situation in the human atrium. Data on the contribution of the slowly activating delayed rectifier currents (IKs) to repolarization are lacking for both human atrium and hiPSC-aCM. METHODS AND RESULTS: Human atrial tissues were obtained from patients with sinus rhythm (SR) or atrial fibrillation (AF). Currents were measured in human atrial cardiomyocytes (aCM) and compared with hiPSC-aCM and used to model IKs contribution to action potential (AP) shape. Action potential was recorded by sharp microelectrodes. HMR-1556 (1 µM) was used to identify IKs and to estimate IKs contribution to repolarization. Less than 50% of hiPSC-aCM and aCM possessed IKs. Frequency of occurrence, current densities, activation/deactivation kinetics, and voltage dependency of IKs did not differ significantly between hiPSC-aCM and aCM, neither in SR nor AF. ß-Adrenoceptor stimulation with isoprenaline did not increase IKs neither in aCM nor in hiPSC-aCM. In tissue from SR, block of IKs with HMR-1556 did not lengthen the action potential duration, even when repolarization reserve was reduced by block of the ultra-rapid repolarizing current with 4-aminopyridine or the rapidly activating delayed rectifier potassium outward current with E-4031. CONCLUSION: I Ks exists in hiPSC-aCM with biophysics not different from aCM. As in adult human atrium (SR and AF), IKs does not appear to relevantly contribute to repolarization in hiPSC-aCM.


Subject(s)
Action Potentials , Atrial Fibrillation , Delayed Rectifier Potassium Channels , Heart Atria , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Myocytes, Cardiac/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Induced Pluripotent Stem Cells/metabolism , Heart Atria/physiopathology , Delayed Rectifier Potassium Channels/metabolism , Atrial Fibrillation/physiopathology , Atrial Fibrillation/metabolism , Female , Cells, Cultured , Male , Middle Aged , Kinetics , Aged , Cell Differentiation , Models, Cardiovascular , Potassium Channel Blockers/pharmacology
2.
Circulation ; 149(11): 860-884, 2024 03 12.
Article in English | MEDLINE | ID: mdl-38152989

ABSTRACT

BACKGROUND: SGLT2 (sodium-glucose cotransporter 2) inhibitors (SGLT2i) can protect the kidneys and heart, but the underlying mechanism remains poorly understood. METHODS: To gain insights on primary effects of SGLT2i that are not confounded by pathophysiologic processes or are secondary to improvement by SGLT2i, we performed an in-depth proteomics, phosphoproteomics, and metabolomics analysis by integrating signatures from multiple metabolic organs and body fluids after 1 week of SGLT2i treatment of nondiabetic as well as diabetic mice with early and uncomplicated hyperglycemia. RESULTS: Kidneys of nondiabetic mice reacted most strongly to SGLT2i in terms of proteomic reconfiguration, including evidence for less early proximal tubule glucotoxicity and a broad downregulation of the apical uptake transport machinery (including sodium, glucose, urate, purine bases, and amino acids), supported by mouse and human SGLT2 interactome studies. SGLT2i affected heart and liver signaling, but more reactive organs included the white adipose tissue, showing more lipolysis, and, particularly, the gut microbiome, with a lower relative abundance of bacteria taxa capable of fermenting phenylalanine and tryptophan to cardiovascular uremic toxins, resulting in lower plasma levels of these compounds (including p-cresol sulfate). SGLT2i was detectable in murine stool samples and its addition to human stool microbiota fermentation recapitulated some murine microbiome findings, suggesting direct inhibition of fermentation of aromatic amino acids and tryptophan. In mice lacking SGLT2 and in patients with decompensated heart failure or diabetes, the SGLT2i likewise reduced circulating p-cresol sulfate, and p-cresol impaired contractility and rhythm in human induced pluripotent stem cell-derived engineered heart tissue. CONCLUSIONS: SGLT2i reduced microbiome formation of uremic toxins such as p-cresol sulfate and thereby their body exposure and need for renal detoxification, which, combined with direct kidney effects of SGLT2i, including less proximal tubule glucotoxicity and a broad downregulation of apical transporters (including sodium, amino acid, and urate uptake), provides a metabolic foundation for kidney and cardiovascular protection.


Subject(s)
Cresols , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Induced Pluripotent Stem Cells , Sodium-Glucose Transporter 2 Inhibitors , Sulfuric Acid Esters , Humans , Mice , Animals , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Uric Acid , Tryptophan , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Proteomics , Uremic Toxins , Induced Pluripotent Stem Cells/metabolism , Glucose , Sodium/metabolism , Diabetes Mellitus, Type 2/complications
3.
Stem Cell Reports ; 18(11): 2123-2137, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37802072

ABSTRACT

Primary carnitine deficiency (PCD) is an autosomal recessive monogenic disorder caused by mutations in SLC22A5. This gene encodes for OCTN2, which transports the essential metabolite carnitine into the cell. PCD patients suffer from muscular weakness and dilated cardiomyopathy. Two OCTN2-defective human induced pluripotent stem cell lines were generated, carrying a full OCTN2 knockout and a homozygous OCTN2 (N32S) loss-of-function mutation. OCTN2-defective genotypes showed lower force development and resting length in engineered heart tissue format compared with isogenic control. Force was sensitive to fatty acid-based media and associated with lipid accumulation, mitochondrial alteration, higher glucose uptake, and metabolic remodeling, replicating findings in animal models. The concordant results of OCTN2 (N32S) and -knockout emphasizes the relevance of OCTN2 for these findings. Importantly, genome-wide analysis and pharmacological inhibitor experiments identified ferroptosis, an iron- and lipid-dependent cell death pathway associated with fibroblast activation as a novel PCD cardiomyopathy disease mechanism.


Subject(s)
Cardiomyopathies , Ferroptosis , Induced Pluripotent Stem Cells , Animals , Humans , Organic Cation Transport Proteins/genetics , Solute Carrier Family 22 Member 5/genetics , Cardiomyopathies/genetics , Lipids
4.
EMBO Mol Med ; 13(6): e13074, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33998164

ABSTRACT

The phospholamban (PLN) p.Arg14del mutation causes dilated cardiomyopathy, with the molecular disease mechanisms incompletely understood. Patient dermal fibroblasts were reprogrammed to hiPSC, isogenic controls were established by CRISPR/Cas9, and cardiomyocytes were differentiated. Mutant cardiomyocytes revealed significantly prolonged Ca2+ transient decay time, Ca2+ -load dependent irregular beating pattern, and lower force. Proteomic analysis revealed less endoplasmic reticulum (ER) and ribosomal and mitochondrial proteins. Electron microscopy showed dilation of the ER and large lipid droplets in close association with mitochondria. Follow-up experiments confirmed impairment of the ER/mitochondria compartment. PLN p.Arg14del end-stage heart failure samples revealed perinuclear aggregates positive for ER marker proteins and oxidative stress in comparison with ischemic heart failure and non-failing donor heart samples. Transduction of PLN p.Arg14del EHTs with the Ca2+ -binding proteins GCaMP6f or parvalbumin improved the disease phenotype. This study identified impairment of the ER/mitochondria compartment without SR dysfunction as a novel disease mechanism underlying PLN p.Arg14del cardiomyopathy. The pathology was improved by Ca2+ -scavenging, suggesting impaired local Ca2+ cycling as an important disease culprit.


Subject(s)
Heart Transplantation , Myocytes, Cardiac , Calcium-Binding Proteins/metabolism , Endoplasmic Reticulum , Humans , Mitochondria , Mutation , Myocytes, Cardiac/metabolism , Proteomics , Tissue Donors
5.
Redox Biol ; 41: 101951, 2021 05.
Article in English | MEDLINE | ID: mdl-33831709

ABSTRACT

Sulforaphane (SFN) is a phytochemical compound extracted from cruciferous plants, like broccoli or cauliflower. Its isothiocyanate group renders SFN reactive, thus allowing post-translational modification of cellular proteins to regulate their function with the potential for biological and therapeutic actions. SFN and stabilized variants recently received regulatory approval for clinical studies in humans for the treatment of neurological disorders and cancer. Potential unwanted side effects of SFN on heart function have not been investigated yet. The present study characterizes the impact of SFN on cardiomyocyte contractile function in cardiac preparations from neonatal rat, adult mouse and human induced-pluripotent stem cell-derived cardiomyocytes. This revealed a SFN-mediated negative inotropic effect, when administered either acutely or chronically, with an impairment of the Frank-Starling response to stretch activation. A direct effect of SFN on myofilament function was excluded in chemically permeabilized mouse trabeculae. However, SFN pretreatment increased lactate formation and enhanced the mitochondrial production of reactive oxygen species accompanied by a significant reduction in the mitochondrial membrane potential. Transmission electron microscopy revealed disturbed sarcomeric organization and inflated mitochondria with whorled membrane shape in response to SFN exposure. Interestingly, administration of the alternative energy source l-glutamine to the medium that bypasses the uptake route of pyruvate into the mitochondrial tricarboxylic acid cycle improved force development in SFN-treated EHTs, suggesting indeed mitochondrial dysfunction as a contributor of SFN-mediated contractile dysfunction. Taken together, the data from the present study suggest that SFN might impact negatively on cardiac contractility in patients with cardiovascular co-morbidities undergoing SFN supplementation therapy. Therefore, cardiac function should be monitored regularly to avoid the onset of cardiotoxic side effects.


Subject(s)
Apoptosis , Isothiocyanates , Animals , Humans , Mice , Mitochondria , Rats , Reactive Oxygen Species , Sulfoxides
6.
Curr Protoc Stem Cell Biol ; 55(1): e127, 2020 12.
Article in English | MEDLINE | ID: mdl-32956561

ABSTRACT

The reproducibility of stem cell research relies on the constant availability of quality-controlled cells. As the quality of human induced pluripotent stem cells (hiPSCs) can deteriorate in the course of a few passages, cell banking is key to achieve consistent results and low batch-to-batch variation. Here, we provide a cost-efficient route to generate master and working cell banks for basic research projects. In addition, we describe minimal protocols for quality assurance including tests for sterility, viability, pluripotency, and genetic integrity. © 2020 The Authors. Basic Protocol 1: Expansion of hiPSCs Basic Protocol 2: Cell banking of hiPSCs Support Protocol 1: Pluripotency assessment by flow cytometry Support Protocol 2: Thawing control: Viability and sterility Support Protocol 3: Potency, viral clearance, and pluripotency: Spontaneous differentiation and qRT-PCR Support Protocol 4: Identity: Short tandem repeat analysis.


Subject(s)
Cryopreservation/methods , Induced Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Cell Line , Humans , Quality Control , Reproducibility of Results
8.
Nat Protoc ; 12(6): 1177-1197, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28492526

ABSTRACT

Since the advent of the generation of human induced pluripotent stem cells (hiPSCs), numerous protocols have been developed to differentiate hiPSCs into cardiomyocytes and then subsequently assess their ability to recapitulate the properties of adult human cardiomyocytes. However, hiPSC-derived cardiomyocytes (hiPSC-CMs) are often assessed in single-cell assays. A shortcoming of these assays is the limited ability to characterize the physiological parameters of cardiomyocytes, such as contractile force, due to random orientations. This protocol describes the differentiation of cardiomyocytes from hiPSCs, which occurs within 14 d. After casting, cardiomyocytes undergo 3D assembly. This produces fibrin-based engineered heart tissues (EHTs)-in a strip format-that generate force under auxotonic stretch conditions. 10-15 d after casting, the EHTs can be used for contractility measurements. This protocol describes parallel expansion of hiPSCs; standardized generation of defined embryoid bodies, growth factor and small-molecule-based cardiac differentiation; and standardized generation of EHTs. To carry out the protocol, experience in advanced cell culture techniques is required.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Tissue Engineering/methods , Humans
9.
J Vis Exp ; (122)2017 04 15.
Article in English | MEDLINE | ID: mdl-28448053

ABSTRACT

Cardiac tissue engineering describes techniques to constitute three dimensional force-generating engineered tissues. For the implementation of these procedures in basic research and preclinical drug development, it is important to develop protocols for automated generation and analysis under standardized conditions. Here, we present a technique to generate engineered heart tissue (EHT) from cardiomyocytes of different species (rat, mouse, human). The technique relies on the assembly of a fibrin-gel containing dissociated cardiomyocytes between elastic polydimethylsiloxane (PDMS) posts in a 24-well format. Three-dimensional, force-generating EHTs constitute within two weeks after casting. This procedure allows for the generation of several hundred EHTs per week and is technically limited only by the availability of cardiomyocytes (0.4-1.0 x 106/EHT). Evaluation of auxotonic muscle contractions is performed in a modified incubation chamber with a mechanical interlock for 24-well plates and a camera placed on top of this chamber. A software controls a camera moved on an XYZ axis system to each EHT. EHT contractions are detected by an automated figure recognition algorithm, and force is calculated based on shortening of the EHT and the elastic propensity and geometry of the PDMS posts. This procedure allows for automated analysis of high numbers of EHT under standardized and sterile conditions. The reliable detection of drug effects on cardiomyocyte contraction is crucial for cardiac drug development and safety pharmacology. We demonstrate, with the example of the hERG channel inhibitor E-4031, that the human EHT system replicates drug responses on contraction kinetics of the human heart, indicating it to be a promising tool for cardiac drug safety screening.


Subject(s)
Drug Evaluation, Preclinical/methods , Tissue Engineering/methods , Animals , Automation , Dimethylpolysiloxanes , Drug Evaluation, Preclinical/instrumentation , ERG1 Potassium Channel/antagonists & inhibitors , Fibrin/pharmacology , Heart/drug effects , Humans , Mice , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Rats
10.
Stem Cell Reports ; 7(1): 29-42, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27211213

ABSTRACT

Analyzing contractile force, the most important and best understood function of cardiomyocytes in vivo is not established in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). This study describes the generation of 3D, strip-format, force-generating engineered heart tissues (EHT) from hiPSC-CM and their physiological and pharmacological properties. CM were differentiated from hiPSC by a growth factor-based three-stage protocol. EHTs were generated and analyzed histologically and functionally. HiPSC-CM in EHTs showed well-developed sarcomeric organization and alignment, and frequent mitochondria. Systematic contractility analysis (26 concentration-response curves) reveals that EHTs replicated canonical response to physiological and pharmacological regulators of inotropy, membrane- and calcium-clock mediators of pacemaking, modulators of ion-channel currents, and proarrhythmic compounds with unprecedented precision. The analysis demonstrates a high degree of similarity between hiPSC-CM in EHT format and native human heart tissue, indicating that human EHTs are useful for preclinical drug testing and disease modeling.


Subject(s)
Heart/growth & development , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Tissue Engineering , Cell Differentiation/genetics , Humans , Mitochondria/metabolism , Myocardial Contraction/genetics , Myocardium/cytology , Myocardium/metabolism , Sarcomeres/metabolism
11.
J Histochem Cytochem ; 61(3): 199-205, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23275449

ABSTRACT

Specialized protein domains bind to posttranslational modifications (PTMs) of proteins, such as phosphorylation or glycosylation. When such PTM-binding protein domains are used as analytical tools, the functional states of cells and tissues can be determined with high precision. Here, we describe the use of recombinant CLEC10A (CD301), a human glycoreceptor of the C-type lectin family, for the detection of ligands in sections from formalin-fixed, paraffin-embedded normal and cancerous mammary tissues. A construct, in which part of the carbohydrate recognition domain (CRD) was deleted, was used as a negative control. In comparison to normal mammary glands, a pronounced staining of tumor tissues was observed. Because the construct with the truncated CRD did not show any tissue staining, the binding of the wild-type glycoreceptor can be attributed to its carbohydrate recognition domain. To distinguish our novel approach from immunohistochemistry, we propose the designation "protein domain histochemistry" (PDH).


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Histocytochemistry/methods , Lectins, C-Type/analysis , Lectins, C-Type/metabolism , Breast Neoplasms/diagnosis , Cloning, Molecular , Female , HEK293 Cells , Humans , Paraffin Embedding/methods , Polysaccharides/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Tissue Fixation/methods
12.
Eur J Cell Biol ; 89(1): 87-94, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20034698

ABSTRACT

Members of the family of carcinoembryonic antigen (CEA)-related cell adhesion molecules (CEACAMs) belonging to the immunoglobulin (Ig) superfamily are expressed in a variety of normal and malignant human tissues. As components of the cell membrane, these glycoproteins can make contact with adjacent cells. CEACAM1 and CEACAM5 (CEA) express Lewis(x) (Le(x)) structures. As shown by mass spectrometry in conjunction with enzymatic digestion, CEACAM1 contains at least seven Le(x) residues. Fucosyltransferase IX is the main fucosyltransferase responsible for attachment of terminal fucose, the key feature of the Le(x) structure, to CEA and CEACAM1. The Le(x) residues of both, CEACAM1 and CEA, interact with the human Le(x)-binding glycan receptors DC-SIGN and SRCL. Since subpopulations of human macrophages express DC-SIGN or SRCL, Le(x)-carrying CEACAMs may modulate the immune response in normal tissues such as the human placenta or in malignant tumours, for example in colorectal, pancreatic or lung carcinomas.


Subject(s)
Antigens, CD/metabolism , Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules/metabolism , Collectins/metabolism , Lectins, C-Type/metabolism , Polysaccharides/metabolism , Receptors, Cell Surface/metabolism , Receptors, Scavenger/metabolism , Recombinant Proteins/metabolism , Cell Line , Colorectal Neoplasms/metabolism , Female , Fucose/metabolism , Fucosyltransferases/metabolism , Humans , Intestinal Mucosa/metabolism , Lewis X Antigen/metabolism , Melanoma/metabolism , Placenta/metabolism , Pregnancy , Protein Binding , Skin Neoplasms/metabolism , Tissue Extracts
13.
Glycobiology ; 17(3): 324-33, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17145745

ABSTRACT

Intercellular adhesion molecule-3 (ICAM-3) binds to the alpha(L)beta(2) integrin and mediates the contact between T cells and antigen-presenting cells. It has been suggested that dendritic cell-specific ICAM-3 grabbing nonintegrin (DC-SIGN), a C-type lectin of macrophages and DCs, is an additional ligand of ICAM-3. So far, the glycan structure mediating the interaction of native ICAM-3 with DC-SIGN is undefined. Here, we demonstrate that native ICAM-3 from human peripheral leukocytes binds recombinant DC-SIGN, is recognized by monoclonal Lewis x antibodies, and specifically interacts with DC-SIGN on immature DCs. The presence of Lewis x residues on ICAM-3 was confirmed by matrix-assisted laser desorption/ionization time-of-flight mass spectroscopy. Investigations on different peripheral blood cell populations revealed that only ICAM-3 from granulocytes bound DC-SIGN. Cotransfection studies demonstrated that fucosyltransferase (FUT) IX and, to a significantly lesser extent, FUT IV, but not FUTs III and VII, mediate the synthesis of Lewis x residues on ICAM-3. These findings indicate that FUT IX is the main FUT mediating the synthesis of Lewis x residues of ICAM-3 in cells of the myeloid lineage, and that these residues bind DC-SIGN. The results suggest that ICAM-3 assists in the interaction of granulocytes with DC-SIGN of DCs.


Subject(s)
Antigens, CD/chemistry , Cell Adhesion Molecules/chemistry , Granulocytes/immunology , Lectins, C-Type/chemistry , Lewis X Antigen/chemistry , Receptors, Cell Surface/chemistry , Antigens, CD/isolation & purification , Cell Adhesion Molecules/isolation & purification , Fucosyltransferases/chemistry , Humans , Leukocytes/immunology , Lewis X Antigen/analysis , Lewis X Antigen/biosynthesis , Recombinant Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
Glycobiology ; 16(3): 197-209, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16282604

ABSTRACT

The CEA-related cell adhesion molecule 1, CEACAM1, is a glycoprotein expressed on the surface of human granulocytes and lymphocytes, endothelia, and many epithelia. CEACAM1 is involved in the regulation of important biological processes, such as tumor growth, angiogenesis, and modulation of the immune response. CEACAM1, a member of the immunoglobulin superfamily carries several Lewis x (Lex) structures as we recently demonstrated by mass spectrometry of native CEACAM1 from human granulocytes. Since Lex residues of pathogens bind to the C-type lectin dendritic cell-specific ICAM-3 grabbing nonintegrin (DC-SIGN) expressed on human DCs, we hypothesized that Lex glycans of CEACAM1 are recognized by DC-SIGN. Here, we demonstrate that CEACAM1, the major carrier of Lex residues in human granulocytes, is specifically recognized by DC-SIGN via Lex residues mediating the internalization of CEACAM1 into immature DCs. Expression studies with CEACAM1 in combination with different fucosyltransferases (FUTs) revealed that FUTIX plays a key role in the synthesis of Lex groups of CEACAM1. As Lex groups on CEACAM1 are selectively attached and specifically interact with DC-SIGN, our findings suggest that CEACAM1 participates in immune regulation in physiological conditions and in pathological conditions, such as inflammation, autoimmune disease, and cancer.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , Fucose/metabolism , Fucosyltransferases/metabolism , Granulocytes/metabolism , Lectins, C-Type/metabolism , Lewis X Antigen/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, CD/chemistry , Cell Adhesion Molecules/chemistry , Cell Differentiation , Cell Line , Cricetinae , Dendritic Cells/cytology , Fucose/chemistry , Gene Expression , Humans , Ligands , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...