Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Exp Dermatol ; 32(10): 1848-1855, 2023 10.
Article in English | MEDLINE | ID: mdl-37587642

ABSTRACT

The mechanotransduction of light-touch sensory stimuli is considered to be the main physiological function of epidermal Merkel cells (MCs). Recently, however, MCs have been demonstrated to be also thermo-sensitive, suggesting that their role in skin physiologically extends well beyond mechanosensation. Here, we demonstrate that in healthy human skin epidermal MCs express functional olfactory receptors, namely OR2AT4, just like neighbouring keratinocytes. Selective stimulation of OR2AT4 by topical application of the synthetic odorant, Sandalore®, significantly increased Piccolo protein expression in MCs, as assessed by quantitative immunohistomorphometry, indicating increased vesicle trafficking and recycling, and significantly reduced nerve growth factor (NGF) immunoreactivity within MCs, possibly indicating increased neurotrophin release upon OR2AT4 activation. Live-cell imaging showed that Sandalore® rapidly induces a loss of FFN206-dependent fluorescence in MCs, suggesting OR2AT4-dependent MC depolarization and subsequent vesicle secretion. Yet, in contrast to keratinocytes, OR2AT4 stimulation by Sandalore® altered neither the number nor the proliferation status of MCs. These preliminary ex vivo findings demonstrate that epidermal MCs also exert OR-dependent chemosensory functions in human skin, and invite one to explore whether these newly identified properties are dysregulated in selected skin disorders, for example, in pruritic dermatoses, and if these novel MC functions can be therapeutically targeted to maintain/promote skin health.


Subject(s)
Merkel Cells , Humans , Butanols/metabolism , Epidermis/metabolism , Mechanoreceptors/physiology , Mechanotransduction, Cellular/physiology , Merkel Cells/metabolism , Merkel Cells/physiology , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Skin/metabolism
2.
Cells ; 12(4)2023 02 08.
Article in English | MEDLINE | ID: mdl-36831217

ABSTRACT

Viral myocarditis is pathologically associated with RNA viruses such as coxsackievirus B3 (CVB3), or more recently, with SARS-CoV-2, but despite intensive research, clinically proven treatment is limited. Here, by use of a transgenic mouse strain (TG) containing a CVB3ΔVP0 genome we unravel virus-mediated cardiac pathophysiological processes in vivo and in vitro. Cardiac function, pathologic ECG alterations, calcium homeostasis, intracellular organization and gene expression were significantly altered in transgenic mice. A marked alteration of mitochondrial structure and gene expression indicates mitochondrial impairment potentially contributing to cardiac contractile dysfunction. An extended picture on viral myocarditis emerges that may help to develop new treatment strategies and to counter cardiac failure.


Subject(s)
COVID-19 , Coxsackievirus Infections , Myocarditis , Virus Diseases , Mice , Animals , Mice, Transgenic , Enterovirus B, Human , SARS-CoV-2
3.
Nutrients ; 14(16)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36014862

ABSTRACT

Female pattern hair loss (FPHL) is a non-scarring alopecia resulting from the progressive conversion of the terminal (t) scalp hair follicles (HFs) into intermediate/miniaturized (i/m) HFs. Although data supporting nutrient deficiency in FPHL HFs are lacking, therapeutic strategies are often associated with nutritional supplementation. Here, we show by metabolic analysis that selected nutrients important for hair growth such as essential amino acids and vitamins are indeed decreased in affected iHFs compared to tHFs in FPHL scalp skin, confirming nutrient insufficiency. iHFs also displayed a more quiescent metabolic phenotype, as indicated by altered metabolite abundance in freshly collected HFs and release/consumption during organ culture of products/substrates of TCA cycle, aerobic glycolysis, and glutaminolysis. Yet, as assessed by exogenous nutrient supplementation ex vivo, nutrient uptake mechanisms are not impaired in affected FPHL iHFs. Moreover, blood vessel density is not diminished in iHFs versus tHFs, despite differences in tHFs from different FPHL scalp locations or versus healthy scalp or changes in the expression of angiogenesis-associated growth factors. Thus, our data reveal that affected iHFs in FPHL display a relative nutrient insufficiency and dormant metabolism, but are still capable of absorbing nutrients, supporting the potential of nutritional supplementation as an adjunct therapy for FPHL.


Subject(s)
Alopecia , Hair Follicle , Alopecia/drug therapy , Female , Humans , Nutrients , Phenotype , Scalp
5.
Sci Rep ; 10(1): 16804, 2020 10 08.
Article in English | MEDLINE | ID: mdl-33033381

ABSTRACT

A detailed description of pathophysiological effects that viruses exert on their host is still challenging. For the first time, we report a highly controllable viral expression model based on an iPS-cell line from a healthy human donor. The established viral model system enables a dose-dependent and highly localized RNA-virus expression in a fully controllable environment, giving rise for new applications for the scientific community.


Subject(s)
Induced Pluripotent Stem Cells/virology , RNA Virus Infections/virology , RNA Viruses/physiology , Cell Line , Doxycycline/pharmacology , Humans , Models, Biological , Myocytes, Cardiac/virology , Virus Activation/drug effects
6.
Methods Mol Biol ; 2154: 143-152, 2020.
Article in English | MEDLINE | ID: mdl-32314214

ABSTRACT

Primary cicatricial alopecia is characterized by a permanent "scarring" alopecia. This condition is characterized by the irreversible loss of hair follicles (HF) as a result of apoptosis and epithelial-mesenchymal transition (EMT) of epithelial stem cells localized in the HF bulge.We here report the procedure for experimentally induced EMT in healthy human epidermal stem cells (eSCs) using full-length HF organ culture ex vivo. The present model can be used to recapitulate the complex processes observed in scarring alopecia patient tissues, to further investigate the mechanisms involved in EMT transformation of HFeSCs, and to test substances that could prevent and/or rescue HFeSCs from EMT for the management of scarring alopecias.


Subject(s)
Alopecia/etiology , Alopecia/metabolism , Epithelial-Mesenchymal Transition , Hair Follicle/metabolism , Hair Follicle/pathology , Stem Cells/metabolism , Stem Cells/pathology , Biomarkers , Cell Culture Techniques , Cell Separation/methods , Cells, Cultured , Disease Susceptibility , Fluorescent Antibody Technique , Humans , Microdissection/methods , Tissue Culture Techniques
7.
Methods Mol Biol ; 2154: 249-254, 2020.
Article in English | MEDLINE | ID: mdl-32314223

ABSTRACT

Wound healing is a complex, multifactorial process that is divided in sequential and overlapping phases in order to restore the skin barrier. For the study of wound healing, different in vivo, in vitro, and ex vivo models have been used in the past. Here we describe in detail the methodology of the human skin punch-in-a-punch ex vivo wound healing model.


Subject(s)
Biomarkers , Wound Healing , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Skin/metabolism , Skin/pathology , Wound Healing/genetics
8.
Nat Commun ; 9(1): 440, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382828

ABSTRACT

Master cell fate determinants are thought to induce specific cell lineages in gastrulation by orchestrating entire gene programs. The T-box transcription factor EOMES (eomesodermin) is crucially required for the development of the heart-yet it is equally important for endoderm specification suggesting that it may act in a context-dependent manner. Here, we define an unrecognized interplay between EOMES and the WNT signaling pathway in controlling cardiac induction by using loss and gain-of-function approaches in human embryonic stem cells. Dose-dependent EOMES induction alone can fully replace a cocktail of signaling molecules otherwise essential for the specification of cardiogenic mesoderm. Highly efficient cardiomyocyte programming by EOMES mechanistically involves autocrine activation of canonical WNT signaling via the WNT3 ligand, which necessitates a shutdown of this axis at a subsequent stage. Our findings provide insights into human germ layer induction and bear biotechnological potential for the robust production of cardiomyocytes from engineered stem cells.


Subject(s)
Cellular Reprogramming Techniques/methods , Pluripotent Stem Cells/cytology , T-Box Domain Proteins/genetics , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Doxycycline/administration & dosage , Doxycycline/pharmacology , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Human Embryonic Stem Cells/cytology , Humans , Mesoderm , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/drug effects , T-Box Domain Proteins/metabolism , Wnt Signaling Pathway , Wnt3 Protein/metabolism
9.
Elife ; 72018 01 16.
Article in English | MEDLINE | ID: mdl-29337667

ABSTRACT

The transcription factor ISL1 is thought to be key for conveying the multipotent and proliferative properties of cardiac precursor cells. Here, we investigate its function upon cardiac induction of human embryonic stem cells. We find that ISL1 does not stabilize the transient cardiac precursor cell state but rather serves to accelerate cardiomyocyte differentiation. Conversely, ISL1 depletion delays cardiac differentiation and respecifies nascent cardiomyocytes from a ventricular to an atrial identity. Mechanistic analyses integrate this unrecognized anti-atrial function of ISL1 with known and newly identified atrial inducers. In this revised view, ISL1 is antagonized by retinoic acid signaling via a novel player, MEIS2. Conversely, ISL1 competes with the retinoic acid pathway for prospective cardiomyocyte fate, which converges on the atrial specifier NR2F1. This study reveals a core regulatory network putatively controlling human heart chamber formation and also bears implications for the subtype-specific production of human cardiomyocytes with enhanced functional properties.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Homeodomain Proteins/metabolism , Human Embryonic Stem Cells/physiology , LIM-Homeodomain Proteins/metabolism , Myocytes, Cardiac/physiology , Transcription Factors/metabolism , COUP Transcription Factor I/metabolism , Humans
10.
Front Physiol ; 8: 705, 2017.
Article in English | MEDLINE | ID: mdl-28959214

ABSTRACT

The fight-or-flight response (FFR), a physiological acute stress reaction, involves positive chronotropic and inotropic effects on heart muscle cells mediated through ß-adrenoceptor activation. Increased systolic calcium is required to enable stronger heart contractions whereas elevated potassium currents are to limit the duration of the action potentials and prevent arrhythmia. The latter effect is accomplished by an increased functional activity of the Kv7.1 channel encoded by KCNQ1. Current knowledge, however, does not sufficiently explain the full extent of rapid Kv7.1 activation and may hence be incomplete. Using inducible genetic KCNQ1 complementation in KCNQ1-deficient human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), we here reinvestigate the functional role of Kv7.1 in adapting human CMs to adrenergic stress. Under baseline conditions, Kv7.1 was barely detectable at the plasma membrane of hiPSC-CMs, yet it fully protected these from adrenergic stress-induced beat-to-beat variability of repolarization and torsade des pointes-like arrhythmia. Furthermore, isoprenaline treatment increased field potential durations specifically in KCNQ1-deficient CMs to cause these adverse macroscopic effects. Mechanistically, we find that the protective action by Kv7.1 resides in a rapid translocation of channel proteins from intracellular stores to the plasma membrane, induced by adrenergic signaling. Gene silencing experiments targeting RAB GTPases, mediators of intracellular vesicle trafficking, showed that fast Kv7.1 recycling under acute stress conditions is RAB4A-dependent.Our data reveal a key mechanism underlying the rapid adaptation of human cardiomyocytes to adrenergic stress. These findings moreover aid to the understanding of disease pathology in long QT syndrome and bear important implications for safety pharmacological screening.

11.
Stem Cell Res ; 21: 26-28, 2017 05.
Article in English | MEDLINE | ID: mdl-28677534

ABSTRACT

Loss-of-function mutations in the PITX2 transcription factor gene have been shown to cause familial atrial fibrillation (AF). To potentially model aspects of AF and unravel PITX2-regulated downstream genes for drug target discovery, we here report the generation of integration-free PITX2-deficient hiPS cell lines. We also show that both PITX2 knockout hiPS cells and isogenic wild-type controls can selectively be differentiated into human atrial cardiomyocytes, to potentially uncover differentially expressed gene sets between these groups.


Subject(s)
Atrial Fibrillation/metabolism , Cell Differentiation , Gene Knockdown Techniques , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Transcription Factors/deficiency , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Cell Line , Homeodomain Proteins , Humans , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/pathology , Homeobox Protein PITX2
12.
Front Physiol ; 8: 469, 2017.
Article in English | MEDLINE | ID: mdl-28729840

ABSTRACT

The ultrarapid delayed rectifier K+ current (IKur), mediated by Kv1.5 channels, constitutes a key component of the atrial action potential. Functional mutations in the underlying KCNA5 gene have been shown to cause hereditary forms of atrial fibrillation (AF). Here, we combine targeted genetic engineering with cardiac subtype-specific differentiation of human induced pluripotent stem cells (hiPSCs) to explore the role of Kv1.5 in atrial hiPSC-cardiomyocytes. CRISPR/Cas9-mediated mutagenesis of integration-free hiPSCs was employed to generate a functional KCNA5 knockout. This model as well as isogenic wild-type control hiPSCs could selectively be differentiated into ventricular or atrial cardiomyocytes at high efficiency, based on the specific manipulation of retinoic acid signaling. Investigation of electrophysiological properties in Kv1.5-deficient cardiomyocytes compared to isogenic controls revealed a strictly atrial-specific disease phentoype, characterized by cardiac subtype-specific field and action potential prolongation and loss of 4-aminopyridine sensitivity. Atrial Kv1.5-deficient cardiomyocytes did not show signs of arrhythmia under adrenergic stress conditions or upon inhibiting additional types of K+ current. Exposure of bulk cultures to carbachol lowered beating frequencies and promoted chaotic spontaneous beating in a stochastic manner. Low-frequency, electrical stimulation in single cells caused atrial and mutant-specific early afterdepolarizations, linking the loss of KCNA5 function to a putative trigger mechanism in familial AF. These results clarify for the first time the role of Kv1.5 in atrial hiPSC-cardiomyocytes and demonstrate the feasibility of cardiac subtype-specific disease modeling using engineered hiPSCs.

13.
Pflugers Arch ; 469(10): 1233-1243, 2017 10.
Article in English | MEDLINE | ID: mdl-28573409

ABSTRACT

The possibility to generate induced pluripotent stem cells (iPSC) opens the way to generate virtually all cell types of our human body. In combination with modern gene editing techniques like CRISPR/CAS, a new set of powerful tools becomes available for life science. Scientific fields like genotype and cell type-specific pharmacology, disease modeling, stem cell biology, and developmental biology have been dramatically fostered and their faces have been changed. However, as golden as the age of iPSC-derived cells and their manipulation has started, the shine begins to tarnish. Researchers face more and more practical problems intrinsic to the system. These problems are related to the specific culturing conditions which are not yet sufficient to mimic the natural environment of native stem cells differentiating towards adult cells. However, researchers work hard to uncover these factors. Here, we review a common standard approach to generate iPSCs and transduce these to iPSC cardiomyocytes. Further, we review recent achievements and discuss their current limitations and future perspectives. We are on track, but the road is still under construction.


Subject(s)
Cell Differentiation/genetics , Gene Editing , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Animals , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Humans
14.
Cell Physiol Biochem ; 40(6): 1549-1558, 2016.
Article in English | MEDLINE | ID: mdl-27997884

ABSTRACT

BACKGROUND/AIMS: Acquired as well as inherited channelopathies are disorders that are caused by altered ion channel function. A family of channels whose malfunction is associated with different channelopathies is the Kv7 K+ channel family; and restoration of normal Kv7 channel function by small molecule modulators is a promising approach for treatment of these often fatal diseases. METHODS: Here, we show the modulation of Kv7 channels by the natural compound Rottlerin heterologously expressed in Xenopus laevis oocytes and on iPSC cardiomyocytes overexpressing Kv7.1 channels. RESULTS: We show that currents carried by Kv7.1 (EC50 = 1.48 µM), Kv7.1/KCNE1 (EC50 = 4.9 µM), and Kv7.4 (EC50 = 0.148 µM) are strongly enhanced by the compound, whereas Kv7.2, Kv7.2/Kv7.3, and Kv7.5 are not sensitive to Rottlerin. Studies on Kv7.1/KCNE1 mutants and in silico modelling indicate that Rottlerin binds to the R-L3-activator site. Rottlerin mediated activation of Kv7.1/KCNE1 channels might be a promising approach in long QT syndrome. As a proof of concept, we show that Rottlerin shortens cardiac repolarisation in iPSC-derived cardiomyocytes expressing Kv7.1. CONCLUSION: Rottlerin or an optimized derivative holds a potential as QT interval correcting drug.


Subject(s)
Acetophenones/pharmacology , Benzopyrans/pharmacology , Biological Products/pharmacology , Ion Channel Gating/drug effects , KCNQ1 Potassium Channel/metabolism , Acetophenones/chemistry , Animals , Benzopyrans/chemistry , Biological Products/chemistry , Computer Simulation , Humans , Induced Pluripotent Stem Cells/cytology , KCNQ1 Potassium Channel/chemistry , Membrane Potentials/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Domains , Protein Multimerization/drug effects , Xenopus laevis
15.
Genom Data ; 10: 71-74, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27722090

ABSTRACT

Cardiac induction of human embryonic stem cells (hESCs) is a process bearing increasing medical relevance, yet it is poorly understood from a developmental biology perspective. Anticipated technological progress in deriving stably expandable cardiac precursor cells or in advancing cardiac subtype specification protocols will likely require deeper insights into this fascinating system. Recent improvements in controlling hESC differentiation now enable a near-homogeneous induction of the cardiac lineage. This is based on an optimized initial stimulation of mesoderm-inducing signaling pathways such as Activin and/or FGF, BMP, and WNT, followed by WNT inhibition as a secondary requirement. Here, we describe a comprehensive data set based on varying hESC differentiation conditions in a systematic manner and recording high-resolution differentiation time-courses analyzed by genome-wide expression profiling (GEO accession number GSE67154). As a baseline, hESCs were differentiated into cardiomyocytes under optimal conditions. Moreover, in additional time-series, individual signaling factors were withdrawn from the initial stimulation cocktail to reveal their specific roles via comparison to the standard condition. Hence, this data set presents a rich resource for hypothesis generation in studying human cardiac induction, as we reveal numbers of known as well as uncharacterized genes prominently marking distinct intermediate stages in the process. These data will also be useful for identifying putative cardiac master regulators in the human system as well as for characterizing expandable cardiac stem cells.

17.
Mol Biol Evol ; 33(5): 1231-44, 2016 05.
Article in English | MEDLINE | ID: mdl-26814189

ABSTRACT

A substantial fraction of phenotypic differences between closely related species are likely caused by differences in gene regulation. While this has already been postulated over 30 years ago, only few examples of evolutionary changes in gene regulation have been verified. Here, we identified and investigated binding sites of the transcription factor GA-binding protein alpha (GABPa) aiming to discover cis-regulatory adaptations on the human lineage. By performing chromatin immunoprecipitation-sequencing experiments in a human cell line, we found 11,619 putative GABPa binding sites. Through sequence comparisons of the human GABPa binding regions with orthologous sequences from 34 mammals, we identified substitutions that have resulted in 224 putative human-specific GABPa binding sites. To experimentally assess the transcriptional impact of those substitutions, we selected four promoters for promoter-reporter gene assays using human and African green monkey cells. We compared the activities of wild-type promoters to mutated forms, where we have introduced one or more substitutions to mimic the ancestral state devoid of the GABPa consensus binding sequence. Similarly, we introduced the human-specific substitutions into chimpanzee and macaque promoter backgrounds. Our results demonstrate that the identified substitutions are functional, both in human and nonhuman promoters. In addition, we performed GABPa knock-down experiments and found 1,215 genes as strong candidates for primary targets. Further analyses of our data sets link GABPa to cognitive disorders, diabetes, KRAB zinc finger (KRAB-ZNF), and human-specific genes. Thus, we propose that differences in GABPa binding sites played important roles in the evolution of human-specific phenotypes.


Subject(s)
GA-Binding Protein Transcription Factor/genetics , GA-Binding Protein Transcription Factor/metabolism , Gene Expression Regulation , Animals , Binding Sites , Biological Evolution , COS Cells , Chlorocebus aethiops , Chromatin Immunoprecipitation , Chromosome Mapping , Evolution, Molecular , Genetic Speciation , HEK293 Cells , Humans , Promoter Regions, Genetic , Protein Binding , Sequence Alignment , Zinc Fingers/genetics
18.
Cell Stem Cell ; 18(3): 341-53, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26748419

ABSTRACT

Cardiac induction requires stepwise integration of BMP and WNT pathway activity. Human embryonic stem cells (hESCs) are developmentally and clinically relevant for studying the poorly understood molecular mechanisms downstream of these cascades. We show that BMP and WNT signaling drive cardiac specification by removing sequential roadblocks that otherwise redirect hESC differentiation toward competing fates, rather than activating a cardiac program per se. First, BMP and WNT signals pattern mesendoderm through cooperative repression of SOX2, a potent mesoderm antagonist. BMP signaling promotes miRNA-877 maturation to induce SOX2 mRNA degradation, while WNT-driven EOMES induction transcriptionally represses SOX2. Following mesoderm formation, cardiac differentiation requires inhibition of WNT activity. We found that WNT inhibition serves to restrict expression of anti-cardiac regulators MSX1 and CDX2/1. Conversely, their simultaneous disruption partially abrogates the requirement for WNT inactivation. These results suggest that human cardiac induction depends on multi-stage repression of alternate lineages, with implications for deriving expandable cardiac stem cells.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Wnt Signaling Pathway , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , Cell Line , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Human Embryonic Stem Cells/cytology , Humans , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/cytology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
20.
Genom Data ; 4: 69-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26484180

ABSTRACT

Cardiomyocyte-like cells (CMs) derived from human pluripotent stem cells (hPSCs) present a valuable model for human disease modeling, studying early human development and, potentially, developing cell therapeutic approaches. However, the specification of early hPSC-derived CMs into defined cardiac subtypes such as atrial and ventricular cells is not well understood and, thus, poorly controlled. Moreover, the maturation status of hPSC-CMs is not well defined, yet it is known that these cells undergo at least some degree of maturation upon longer term in vitro culture. To gain insight into this process, and to assess their developmental status, we have recently generated a data set of hPSC-CMs monitoring global changes in gene expression upon long term maintenance in vitro, in comparison to human atrial and ventricular heart samples (GEO accession number GEO: GSE64189). These data present a rich resource for evaluating the maturation status of hPSC-CMs, for identifying suitable markers for subtype-specific gene expression, as well as for the generation of functional hypotheses. Here, we provide additional details and quality checks of this data set, and exemplify how it can be used to identify maturation-associated as well as cardiac subtype-specific markers.

SELECTION OF CITATIONS
SEARCH DETAIL
...