208 results on '"Andreas Kispert"'
Search Results
2. Combined genomic and proteomic approaches reveal DNA binding sites and interaction partners of TBX2 in the developing lung
- Author
-
Timo H. Lüdtke, Irina Wojahn, Marc-Jens Kleppa, Jasper Schierstaedt, Vincent M. Christoffels, Patrick Künzler, and Andreas Kispert
- Subjects
Tbx2 ,Pulmonary mesenchyme ,Lung development ,NuRD ,HDAC ,CBX3 ,Diseases of the respiratory system ,RC705-779 - Abstract
Abstract Background Tbx2 encodes a transcriptional repressor implicated in the development of numerous organs in mouse. During lung development TBX2 maintains the proliferation of mesenchymal progenitors, and hence, epithelial proliferation and branching morphogenesis. The pro-proliferative function was traced to direct repression of the cell-cycle inhibitor genes Cdkn1a and Cdkn1b, as well as of genes encoding WNT antagonists, Frzb and Shisa3, to increase pro-proliferative WNT signaling. Despite these important molecular insights, we still lack knowledge of the DNA occupancy of TBX2 in the genome, and of the protein interaction partners involved in transcriptional repression of target genes. Methods We used chromatin immunoprecipitation (ChIP)-sequencing and expression analyses to identify genomic DNA-binding sites and transcription units directly regulated by TBX2 in the developing lung. Moreover, we purified TBX2 containing protein complexes from embryonic lung tissue and identified potential interaction partners by subsequent liquid chromatography/mass spectrometry. The interaction with candidate proteins was validated by immunofluorescence, proximity ligation and individual co-immunoprecipitation analyses. Results We identified Il33 and Ccn4 as additional direct target genes of TBX2 in the pulmonary mesenchyme. Analyzing TBX2 occupancy data unveiled the enrichment of five consensus sequences, three of which match T-box binding elements. The remaining two correspond to a high mobility group (HMG)-box and a homeobox consensus sequence motif. We found and validated binding of TBX2 to the HMG-box transcription factor HMGB2 and the homeobox transcription factor PBX1, to the heterochromatin protein CBX3, and to various members of the nucleosome remodeling and deacetylase (NuRD) chromatin remodeling complex including HDAC1, HDAC2 and CHD4. Conclusion Our data suggest that TBX2 interacts with homeobox and HMG-box transcription factors as well as with the NuRD chromatin remodeling complex to repress transcription of anti-proliferative genes in the pulmonary mesenchyme.
- Published
- 2021
- Full Text
- View/download PDF
3. TBX2-positive cells represent a multi-potent mesenchymal progenitor pool in the developing lung
- Author
-
Irina Wojahn, Timo H. Lüdtke, Vincent M. Christoffels, Mark-Oliver Trowe, and Andreas Kispert
- Subjects
Tbx2 ,Lineage tracing ,Pulmonary mesenchyme ,Smooth muscle cells ,Lung development ,Diseases of the respiratory system ,RC705-779 - Abstract
Abstract Background In the embryonic mammalian lung, mesenchymal cells act both as a signaling center for epithelial proliferation, differentiation and morphogenesis as well as a source for a multitude of differentiated cell types that support the structure of the developing and mature organ. Whether the embryonic pulmonary mesenchyme is a homogenous precursor pool and how it diversifies into different cell lineages is poorly understood. We have previously shown that the T-box transcription factor gene Tbx2 is expressed in the pulmonary mesenchyme of the developing murine lung and is required therein to maintain branching morphogenesis. Methods We determined Tbx2/TBX2 expression in the developing murine lung by in situ hybridization and immunofluorescence analyses. We used a genetic lineage tracing approach with a Cre line under the control of endogenous Tbx2 control elements (Tbx2 cre ), and the R26 mTmG reporter line to trace TBX2-positive cells in the murine lung. We determined the fate of the TBX2 lineage by co-immunofluorescence analysis of the GFP reporter and differentiation markers in normal murine lungs and in lungs lacking or overexpressing TBX2 in the pulmonary mesenchyme. Results We show that TBX2 is strongly expressed in mesenchymal progenitors in the developing murine lung. In differentiated smooth muscle cells and in fibroblasts, expression of TBX2 is still widespread but strongly reduced. In mesothelial and endothelial cells expression is more variable and scattered. All fetal smooth muscle cells, endothelial cells and fibroblasts derive from TBX2+ progenitors, whereas half of the mesothelial cells have a different descent. The fate of TBX2-expressing cells is not changed in Tbx2-deficient and in TBX2-constitutively overexpressing mice but the distribution and abundance of endothelial and smooth muscle cells is changed in the overexpression condition. Conclusion The fate of pulmonary mesenchymal progenitors is largely independent of TBX2. Nevertheless, a successive and precisely timed downregulation of TBX2 is necessary to allow proper differentiation and functionality of bronchial smooth muscle cells and to limit endothelial differentiation. Our work suggests expression of TBX2 in an early pulmonary mesenchymal progenitor and supports a role of TBX2 in maintaining the precursor state of these cells.
- Published
- 2019
- Full Text
- View/download PDF
4. Generation of hiPSC-derived low threshold mechanoreceptors containing axonal termini resembling bulbous sensory nerve endings and expressing Piezo1 and Piezo2
- Author
-
Shuyong Zhu, Nancy Stanslowsky, Jorge Fernández-Trillo, Tamrat M. Mamo, Pengfei Yu, Norman Kalmbach, Birgit Ritter, Reto Eggenschwiler, Werner J.D. Ouwendijk, David Mzinza, Likai Tan, Andreas Leffler, Michael Spohn, Richard J.P. Brown, Kai A. Kropp, Volkhard Kaever, Teng-Cheong Ha, Pratibha Narayanan, Adam Grundhoff, Reinhold Förster, Axel Schambach, Georges M.G.M. Verjans, Manuela Schmidt, Andreas Kispert, Tobias Cantz, Ana Gomis, Florian Wegner, and Abel Viejo-Borbolla
- Subjects
Human induced pluripotent stem cells ,Small molecule-derived neural precursor cells ,Low threshold mechanoreceptors ,Bulbous sensory nerve ending ,Piezo1, Piezo2 ,Biology (General) ,QH301-705.5 - Abstract
Somatosensory low threshold mechanoreceptors (LTMRs) sense innocuous mechanical forces, largely through specialized axon termini termed sensory nerve endings, where the mechanotransduction process initiates upon activation of mechanotransducers. In humans, a subset of sensory nerve endings is enlarged, forming bulb-like expansions, termed bulbous nerve endings. There is no in vitro human model to study these neuronal endings. Piezo2 is the main mechanotransducer found in LTMRs. Recent evidence shows that Piezo1, the other mechanotransducer considered absent in dorsal root ganglia (DRG), is expressed at low level in somatosensory neurons. We established a differentiation protocol to generate, from iPSC-derived neuronal precursor cells, human LTMR recapitulating bulbous sensory nerve endings and heterogeneous expression of Piezo1 and Piezo2. The derived neurons express LTMR-specific genes, convert mechanical stimuli into electrical signals and have specialized axon termini that morphologically resemble bulbous nerve endings. Piezo2 is concentrated within these enlarged axon termini. Some derived neurons express low level Piezo1, and a subset co-express both channels. Thus, we generated a unique, iPSCs-derived human model that can be used to investigate the physiology of bulbous sensory nerve endings, and the role of Piezo1 and 2 during mechanosensation.
- Published
- 2021
- Full Text
- View/download PDF
5. A 3D iPSC-differentiation model identifies interleukin-3 as a regulator of early human hematopoietic specification
- Author
-
Mania Ackermann, Kathrin Haake, Henning Kempf, Paul Kaschutnig, Anna-Carina Weiss, Ariane H.H. Nguyen, Markus Abeln, Sylvia Merkert, Mark Phillip Kühnel, Dorothee Hartmann, Danny Jonigk, Thomas Thum, Andreas Kispert, Michael D. Milsom, and Nico Lachmann
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Hematopoietic development is spatiotemporally tightly regulated by defined cell-intrinsic and extrinsic modifiers. The role of cytokines has been intensively studied in adult hematopoiesis; however, their role in embryonic hematopoietic specification remains largely unexplored. Here, we used induced pluripotent stem cell (iPSC) technology and established a 3-dimensional, organoid-like differentiation system (hemanoid) maintaining the structural cellular integrity to evaluate the effect of cytokines on embryonic hematopoietic development. We show, that defined stages of early human hematopoietic development were recapitulated within the generated hemanoids. We identified KDR+/CD34high/CD144+/CD43-/CD45- hemato-endothelial progenitor cells (HEPs) forming organized, vasculature-like structures and giving rise to CD34low/CD144-/CD43+/CD45+ hematopoietic progenitor cells. We demonstrate that the endothelial to hematopoietic transition of HEPs is dependent on the presence of interleukin 3 (IL-3). Inhibition of IL-3 signalling blocked hematopoietic differentiation and arrested the cells in the HEP stage. Thus, our data suggest an important role for IL-3 in early human hematopoiesis by supporting the endothelial to hematopoietic transition of hemato-endothelial progenitor cells and highlight the potential of a hemanoid-based model to study human hematopoietic development.
- Published
- 2020
- Full Text
- View/download PDF
6. The transcription factor GATA4 promotes myocardial regeneration in neonatal mice
- Author
-
Mona Malek Mohammadi, Badder Kattih, Andrea Grund, Natali Froese, Mortimer Korf‐Klingebiel, Anna Gigina, Ulrike Schrameck, Carsten Rudat, Qiangrong Liang, Andreas Kispert, Kai C Wollert, Johann Bauersachs, and Joerg Heineke
- Subjects
cardiac regeneration ,cardiomyocyte proliferation ,GATA4 ,IL‐13 ,neonatal cryoinfarction ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Heart failure is often the consequence of insufficient cardiac regeneration. Neonatal mice retain a certain capability of myocardial regeneration until postnatal day (P)7, although the underlying transcriptional mechanisms remain largely unknown. We demonstrate here that cardiac abundance of the transcription factor GATA4 was high at P1, but became strongly reduced at P7 in parallel with loss of regenerative capacity. Reconstitution of cardiac GATA4 levels by adenoviral gene transfer markedly improved cardiac regeneration after cryoinjury at P7. In contrast, the myocardial scar was larger in cardiomyocyte‐specific Gata4 knockout (CM‐G4‐KO) mice after cryoinjury at P0, indicative of impaired regeneration, which was accompanied by reduced cardiomyocyte proliferation and reduced myocardial angiogenesis in CM‐G4‐KO mice. Cardiomyocyte proliferation was also diminished in cardiac explants from CM‐G4‐KO mice and in isolated cardiomyocytes with reduced GATA4 expression. Mechanistically, decreased GATA4 levels caused the downregulation of several pro‐regenerative genes (among them interleukin‐13, Il13) in the myocardium. Interestingly, systemic administration of IL‐13 rescued defective heart regeneration in CM‐G4‐KO mice and could be evaluated as therapeutic strategy in the future.
- Published
- 2017
- Full Text
- View/download PDF
7. The transcription factor GATA4 promotes myocardial regeneration in neonatal mice
- Author
-
Mona Malek Mohammadi, Badder Kattih, Andrea Grund, Natali Froese, Mortimer Korf‐Klingebiel, Anna Gigina, Ulrike Schrameck, Carsten Rudat, Qiangrong Liang, Andreas Kispert, Kai C Wollert, Johann Bauersachs, and Joerg Heineke
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2019
- Full Text
- View/download PDF
8. Proteomic analysis identifies transcriptional cofactors and homeobox transcription factors as TBX18 binding proteins.
- Author
-
Reginaldo Rivera-Reyes, Marc-Jens Kleppa, and Andreas Kispert
- Subjects
Medicine ,Science - Abstract
The TBX18 transcription factor is a crucial developmental regulator of several organ systems in mice, and loss of its transcriptional repression activity causes dilative nephropathies in humans. The molecular complexes with which TBX18 regulates transcription are poorly understood prompting us to use an unbiased proteomic approach to search for protein interaction partners. Using overexpressed dual tagged TBX18 as bait, we identified by tandem purification and subsequent LC-MS analysis TBX18 binding proteins in 293 cells. Clustering of functional annotations of the identified proteins revealed a highly significant enrichment of transcriptional cofactors and homeobox transcription factors. Using nuclear recruitment assays as well as GST pull-downs, we validated CBFB, GAR1, IKZF2, NCOA5, SBNO2 and CHD7 binding to the T-box of TBX18 in vitro. From these transcriptional cofactors, CBFB, CHD7 and IKZF2 enhanced the transcriptional repression of TBX18, while NCOA5 and SBNO2 dose-dependently relieved it. All tested homeobox transcription factors interacted with the T-box of TBX18 in pull-down assays, with members of the Pbx and Prrx subfamilies showing coexpression with Tbx18 in the developing ureter of the mouse. In summary, we identified and characterized new TBX18 binding partners that may influence the transcriptional activity of TBX18 in vivo.
- Published
- 2018
- Full Text
- View/download PDF
9. A SHH-FOXF1-BMP4 signaling axis regulating growth and differentiation of epithelial and mesenchymal tissues in ureter development.
- Author
-
Tobias Bohnenpoll, Anna B Wittern, Tamrat M Mamo, Anna-Carina Weiss, Carsten Rudat, Marc-Jens Kleppa, Karin Schuster-Gossler, Irina Wojahn, Timo H-W Lüdtke, Mark-Oliver Trowe, and Andreas Kispert
- Subjects
Genetics ,QH426-470 - Abstract
The differentiated cell types of the epithelial and mesenchymal tissue compartments of the mature ureter of the mouse arise in a precise temporal and spatial sequence from uncommitted precursor cells of the distal ureteric bud epithelium and its surrounding mesenchyme. Previous genetic efforts identified a member of the Hedgehog (HH) family of secreted proteins, Sonic hedgehog (SHH) as a crucial epithelial signal for growth and differentiation of the ureteric mesenchyme. Here, we used conditional loss- and gain-of-function experiments of the unique HH signal transducer Smoothened (SMO) to further characterize the cellular functions and unravel the effector genes of HH signaling in ureter development. We showed that HH signaling is not only required for proliferation and SMC differentiation of cells of the inner mesenchymal region but also for survival of cells of the outer mesenchymal region, and for epithelial proliferation and differentiation. We identified the Forkhead transcription factor gene Foxf1 as a target of HH signaling in the ureteric mesenchyme. Expression of a repressor version of FOXF1 in this tissue completely recapitulated the mesenchymal and epithelial proliferation and differentiation defects associated with loss of HH signaling while re-expression of a wildtype version of FOXF1 in the inner mesenchymal layer restored these cellular programs when HH signaling was inhibited. We further showed that expression of Bmp4 in the ureteric mesenchyme depends on HH signaling and Foxf1, and that exogenous BMP4 rescued cell proliferation and epithelial differentiation in ureters with abrogated HH signaling or FOXF1 function. We conclude that SHH uses a FOXF1-BMP4 module to coordinate the cellular programs for ureter elongation and differentiation, and suggest that deregulation of this signaling axis occurs in human congenital anomalies of the kidney and urinary tract (CAKUT).
- Published
- 2017
- Full Text
- View/download PDF
10. Epicardial Lineages
- Author
-
Andreas Kispert and Franziska Greulich
- Subjects
epicardium ,epicardial lineages ,EPDCs ,fibroblasts ,smooth muscle cells ,Biology (General) ,QH301-705.5 - Abstract
The epicardium is the mono-layered epithelium that covers the outer surface of the myocardium from early in cardiac development. Long thought to act merely passively to protect the myocardium from frictional forces in the pericardial cavity during the enduring contraction and expansion cycles of the heart, it is now considered to be a crucial source of cells and signals that direct myocardial growth and formation of the coronary vasculature during development and regeneration. Lineage tracing efforts in the chick, the mouse and the zebrafish unambiguously identified fibroblasts in interstitial and perivascular locations as well as coronary smooth muscle cells as the two major lineages that derive from epithelial-mesenchymal transition and subsequent differentiation from individual epicardial cells. However, controversies exist about an additional endothelial and myocardial fate of epicardial progenitor cells. Here, we review epicardial fate mapping efforts in three vertebrate model systems, describe their conceptual differences and discuss their methodological limitations to reach a consensus of the potential of (pro-)epicardial cells in vitro and in vivo.
- Published
- 2013
- Full Text
- View/download PDF
11. Lack of Genetic Interaction between Tbx18 and Tbx2/Tbx20 in Mouse Epicardial Development.
- Author
-
Franziska Greulich, Carsten Rudat, Henner F Farin, Vincent M Christoffels, and Andreas Kispert
- Subjects
Medicine ,Science - Abstract
The epicardium, the outermost layer of the heart, is an essential source of cells and signals for the formation of the cardiac fibrous skeleton and the coronary vasculature, and for the maturation of the myocardium during embryonic development. The molecular factors that control epicardial mobilization and differentiation, and direct the epicardial-myocardial cross-talk are, however, insufficiently understood. The T-box transcription factor gene Tbx18 is specifically expressed in the epicardium of vertebrate embryos. Loss of Tbx18 is dispensable for epicardial development, but may influence coronary vessel maturation. In contrast, over-expression of an activator version of TBX18 severely impairs epicardial development by premature differentiation of epicardial cells into SMCs indicating a potential redundancy of Tbx18 with other repressors of the T-box gene family. Here, we show that Tbx2 and Tbx20 are co-expressed with Tbx18 at different stages of epicardial development. Using a conditional gene targeting approach we find that neither the epicardial loss of Tbx2 nor the combined loss of Tbx2 and Tbx18 affects epicardial development. Similarly, we observed that the heterozygous loss of Tbx20 with and without additional loss of Tbx18 does not impact on epicardial integrity and mobilization in mouse embryos. Thus, Tbx18 does not function redundantly with Tbx2 or Tbx20 in epicardial development.
- Published
- 2016
- Full Text
- View/download PDF
12. Tbx18 Regulates the Differentiation of Periductal Smooth Muscle Stroma and the Maintenance of Epithelial Integrity in the Prostate.
- Author
-
C Chase Bolt, Soumya Negi, Nuno Guimarães-Camboa, Huimin Zhang, Joseph M Troy, Xiaochen Lu, Andreas Kispert, Sylvia M Evans, and Lisa Stubbs
- Subjects
Medicine ,Science - Abstract
The T-box transcription factor TBX18 is essential to mesenchymal cell differentiation in several tissues and Tbx18 loss-of-function results in dramatic organ malformations and perinatal lethality. Here we demonstrate for the first time that Tbx18 is required for the normal development of periductal smooth muscle stromal cells in prostate, particularly in the anterior lobe, with a clear impact on prostate health in adult mice. Prostate abnormalities are only subtly apparent in Tbx18 mutants at birth; to examine postnatal prostate development we utilized a relatively long-lived hypomorphic mutant and a novel conditional Tbx18 allele. Similar to the ureter, cells that fail to express Tbx18 do not condense normally into smooth muscle cells of the periductal prostatic stroma. However, in contrast to ureter, the periductal stromal cells in mutant prostate assume a hypertrophic, myofibroblastic state and the adjacent epithelium becomes grossly disorganized. To identify molecular events preceding the onset of this pathology, we compared gene expression in the urogenital sinus (UGS), from which the prostate develops, in Tbx18-null and wild type littermates at two embryonic stages. Genes that regulate cell proliferation, smooth muscle differentiation, prostate epithelium development, and inflammatory response were significantly dysregulated in the mutant urogenital sinus around the time that Tbx18 is first expressed in the wild type UGS, suggesting a direct role in regulating those genes. Together, these results argue that Tbx18 is essential to the differentiation and maintenance of the prostate periurethral mesenchyme and that it indirectly regulates epithelial differentiation through control of stromal-epithelial signaling.
- Published
- 2016
- Full Text
- View/download PDF
13. Wnt11 is required for oriented migration of dermogenic progenitor cells from the dorsomedial lip of the avian dermomyotome.
- Author
-
Gabriela Morosan-Puopolo, Ajeesh Balakrishnan-Renuka, Faisal Yusuf, Jingchen Chen, Fangping Dai, Georg Zoidl, Timo H-W Lüdtke, Andreas Kispert, Carsten Theiss, Mohammed Abdelsabour-Khalaf, and Beate Brand-Saberi
- Subjects
Medicine ,Science - Abstract
The embryonic origin of the dermis in vertebrates can be traced back to the dermomyotome of the somites, the lateral plate mesoderm and the neural crest. The dermal precursors directly overlying the neural tube display a unique dense arrangement and are the first to induce skin appendage formation in vertebrate embryos. These dermal precursor cells have been shown to derive from the dorsomedial lip of the dermomyotome (DML). Based on its expression pattern in the DML, Wnt11 is a candidate regulator of dorsal dermis formation. Using EGFP-based cell labelling and time-lapse imaging, we show that the Wnt11 expressing DML is the source of the dense dorsal dermis. Loss-of-function studies in chicken embryos show that Wnt11 is indeed essential for the formation of dense dermis competent to support cutaneous appendage formation. Our findings show that dermogenic progenitors cannot leave the DML to form dense dorsal dermis following Wnt11 silencing. No alterations were noticeable in the patterning or in the epithelial state of the dermomyotome including the DML. Furthermore, we show that Wnt11 expression is regulated in a manner similar to the previously described early dermal marker cDermo-1. The analysis of Wnt11 mutant mice exhibits an underdeveloped dorsal dermis and strongly supports our gene silencing data in chicken embryos. We conclude that Wnt11 is required for dense dermis and subsequent cutaneous appendage formation, by influencing the cell fate decision of the cells in the DML.
- Published
- 2014
- Full Text
- View/download PDF
14. Upk3b is dispensable for development and integrity of urothelium and mesothelium.
- Author
-
Carsten Rudat, Thomas Grieskamp, Christian Röhr, Rannar Airik, Christoph Wrede, Jan Hegermann, Bernhard G Herrmann, Karin Schuster-Gossler, and Andreas Kispert
- Subjects
Medicine ,Science - Abstract
The mesothelium, the lining of the coelomic cavities, and the urothelium, the inner lining of the urinary drainage system, are highly specialized epithelia that protect the underlying tissues from mechanical stress and seal them from the overlying fluid space. The development of these epithelia from simple precursors and the molecular characteristics of the mature tissues are poorly analyzed. Here, we show that uroplakin 3B (Upk3b), which encodes an integral membrane protein of the tetraspanin superfamily, is specifically expressed both in development as well as under homeostatic conditions in adult mice in the mesothelia of the body cavities, i.e., the epicardium and pericardium, the pleura and the peritoneum, and in the urothelium of the urinary tract. To analyze Upk3b function, we generated a creERT2 knock-in allele by homologous recombination in embryonic stem cells. We show that Upk3bcreERT2 represents a null allele despite the lack of creERT2 expression from the mutated locus. Morphological, histological and molecular analyses of Upk3b-deficient mice did not detect changes in differentiation or integrity of the urothelium and the mesothelia that cover internal organs. Upk3b is coexpressed with the closely related Upk3a gene in the urothelium but not in the mesothelium, leaving the possibility of a functional redundancy between the two genes in the urothelium only.
- Published
- 2014
- Full Text
- View/download PDF
15. TSHZ3 and SOX9 regulate the timing of smooth muscle cell differentiation in the ureter by reducing myocardin activity.
- Author
-
Elise Martin, Xavier Caubit, Rannar Airik, Christine Vola, Ahmed Fatmi, Andreas Kispert, and Laurent Fasano
- Subjects
Medicine ,Science - Abstract
Smooth muscle cells are of key importance for the proper functioning of different visceral organs including those of the urogenital system. In the mouse ureter, the two transcriptional regulators TSHZ3 and SOX9 are independently required for initiation of smooth muscle differentiation from uncommitted mesenchymal precursor cells. However, it has remained unclear whether TSHZ3 and SOX9 act independently or as part of a larger regulatory network. Here, we set out to characterize the molecular function of TSHZ3 in the differentiation of the ureteric mesenchyme. Using a yeast-two-hybrid screen, we identified SOX9 as an interacting protein. We show that TSHZ3 also binds to the master regulator of the smooth muscle program, MYOCD, and displaces it from the coregulator SRF, thereby disrupting the activation of smooth muscle specific genes. We found that the initiation of the expression of smooth muscle specific genes in MYOCD-positive ureteric mesenchyme coincides with the down regulation of Sox9 expression, identifying SOX9 as a possible negative regulator of smooth muscle cell differentiation. To test this hypothesis, we prolonged the expression of Sox9 in the ureteric mesenchyme in vivo. We found that Sox9 does not affect Myocd expression but significantly reduces the expression of MYOCD/SRF-dependent smooth muscle genes, suggesting that down-regulation of Sox9 is a prerequisite for MYOCD activity. We propose that the dynamic expression of Sox9 and the interaction between TSHZ3, SOX9 and MYOCD provide a mechanism that regulates the pace of progression of the myogenic program in the ureter.
- Published
- 2013
- Full Text
- View/download PDF
16. Tbx2 terminates shh/fgf signaling in the developing mouse limb bud by direct repression of gremlin1.
- Author
-
Henner F Farin, Timo H-W Lüdtke, Martina K Schmidt, Susann Placzko, Karin Schuster-Gossler, Marianne Petry, Vincent M Christoffels, and Andreas Kispert
- Subjects
Genetics ,QH426-470 - Abstract
Vertebrate limb outgrowth is driven by a positive feedback loop that involves Sonic hedgehog (Shh) and Gremlin1 (Grem1) in the posterior limb bud mesenchyme and Fibroblast growth factors (Fgfs) in the overlying epithelium. Proper spatio-temporal control of these signaling activities is required to avoid limb malformations such as polydactyly. Here we show that, in Tbx2-deficient hindlimbs, Shh/Fgf4 signaling is prolonged, resulting in increased limb bud size and duplication of digit 4. In turn, limb-specific Tbx2 overexpression leads to premature termination of this signaling loop with smaller limbs and reduced digit number as phenotypic manifestation. We show that Tbx2 directly represses Grem1 in distal regions of the posterior limb mesenchyme allowing Bone morphogenetic protein (Bmp) signaling to abrogate Fgf4/9/17 expression in the overlying epithelium. Since Tbx2 itself is a target of Bmp signaling, our data identify a growth-inhibiting positive feedback loop (Bmp/Tbx2/Grem1). We propose that proliferative expansion of Tbx2-expressing cells mediates self-termination of limb bud outgrowth due to their refractoriness to Grem1 induction.
- Published
- 2013
- Full Text
- View/download PDF
17. Phenotypical analysis of atypical PKCs in vivo function display a compensatory system at mouse embryonic day 7.5.
- Author
-
Sebastian Seidl, Ursula Braun, Norbert Roos, Shaohua Li, Timo H-W Lüdtke, Andreas Kispert, and Michael Leitges
- Subjects
Medicine ,Science - Abstract
BACKGROUND: The atypical protein kinases C (PKC) isoforms ι/λ and ζ play crucial roles in many cellular processes including development, cell proliferation, differentiation and cell survival. Possible redundancy between the two isoforms has always been an issue since most biochemical tools do not differentiate between the two proteins. Thus, much effort has been made during the last decades to characterize the functions of aPKCs using gene targeting approaches and depletion studies. However, little is known about the specific roles of each isoform in mouse development. METHODOLOGY/PRINCIPAL FINDINGS: To evaluate the importance of PKCι in mouse development we designed PKCι deletion mutants using the gene targeting approach. We show that the deletion of PKCι, results in a reduced size of the amniotic cavity at E7.5 and impaired growth of the embryo at E8.5 with subsequent absorption of the embryo. Our data also indicate an impaired localization of ZO-1 and disorganized structure of the epithelial tissue in the embryo. Importantly, using electron microscopy, embryoid body formation and immunofluorescence analysis, we found, that in the absence of PKCι, tight junctions and apico-basal polarity were still established. Finally, our study points to a non-redundant PKCι function at E9.5, since expression of PKCζ is able to rescue the E7.5 phenotype, but could not prevent embryonic lethality at a later time-point (E9.5). CONCLUSION: Our data show that PKCι is crucial for mouse embryogenesis but is dispensable for the establishment of polarity and tight junction formation. We present a compensatory function of PKCζ at E7.5, rescuing the phenotype. Furthermore, this study indicates at least one specific, yet unknown, PKCι function that cannot be compensated by the overexpression of PKCζ at E9.5.
- Published
- 2013
- Full Text
- View/download PDF
18. Tbx2 controls lung growth by direct repression of the cell cycle inhibitor genes Cdkn1a and Cdkn1b.
- Author
-
Timo H-W Lüdtke, Henner F Farin, Carsten Rudat, Karin Schuster-Gossler, Marianne Petry, Phil Barnett, Vincent M Christoffels, and Andreas Kispert
- Subjects
Genetics ,QH426-470 - Abstract
Vertebrate organ development relies on the precise spatiotemporal orchestration of proliferation rates and differentiation patterns in adjacent tissue compartments. The underlying integration of patterning and cell cycle control during organogenesis is insufficiently understood. Here, we have investigated the function of the patterning T-box transcription factor gene Tbx2 in lung development. We show that lungs of Tbx2-deficient mice are markedly hypoplastic and exhibit reduced branching morphogenesis. Mesenchymal proliferation was severely decreased, while mesenchymal differentiation into fibrocytes was prematurely induced. In the epithelial compartment, proliferation was reduced and differentiation of alveolar epithelial cells type 1 was compromised. Prior to the observed cellular changes, canonical Wnt signaling was downregulated, and Cdkn1a (p21) and Cdkn1b (p27) (two members of the Cip/Kip family of cell cycle inhibitors) were strongly induced in the Tbx2-deficient lung mesenchyme. Deletion of both Cdkn1a and Cdkn1b rescued, to a large degree, the growth deficits of Tbx2-deficient lungs. Prolongation of Tbx2 expression into adulthood led to hyperproliferation and maintenance of mesenchymal progenitor cells, with branching morphogenesis remaining unaffected. Expression of Cdkn1a and Cdkn1b was ablated from the lung mesenchyme in this gain-of-function setting. We further show by ChIP experiments that Tbx2 directly binds to Cdkn1a and Cdkn1b loci in vivo, defining these two genes as direct targets of Tbx2 repressive activity in the lung mesenchyme. We conclude that Tbx2-mediated regulation of Cdkn1a and Cdkn1b represents a crucial node in the network integrating patterning information and cell cycle regulation that underlies growth, differentiation, and branching morphogenesis of this organ.
- Published
- 2013
- Full Text
- View/download PDF
19. Partial absence of pleuropericardial membranes in Tbx18- and Wt1-deficient mice.
- Author
-
Julia Norden, Thomas Grieskamp, Vincent M Christoffels, Antoon F M Moorman, and Andreas Kispert
- Subjects
Medicine ,Science - Abstract
The pleuropericardial membranes are fibro-serous walls that separate the pericardial and pleural cavities and anchor the heart inside the mediastinum. Partial or complete absence of pleuropericardial membranes is a rare human disease, the etiology of which is poorly understood. As an attempt to better understand these defects, we wished to analyze the cellular and molecular mechanisms directing the separation of pericardial and pleural cavities by pleuropericardial membranes in the mouse. We found by histological analyses that both in Tbx18- and Wt1-deficient mice the pleural and pericardial cavities communicate due to a partial absence of the pleuropericardial membranes in the hilus region. We trace these defects to a persisting embryonic connection between these cavities, the pericardioperitoneal canals. Furthermore, we identify mesenchymal ridges in the sinus venosus region that tether the growing pleuropericardial membranes to the hilus of the lung, and thus, close the pericardioperitoneal canals. In Tbx18-deficient embryos these mesenchymal ridges are not established, whereas in Wt1-deficient embryos the final fusion process between these tissues and the body wall does not occur. We suggest that this fusion is an active rather than a passive process, and discuss the interrelation between closure of the pericardioperitoneal canals, lateral release of the pleuropericardial membranes from the lateral body wall, and sinus horn development.
- Published
- 2012
- Full Text
- View/download PDF
20. Heterozygous variants in the DVL2 interaction region of DACT1 cause CAKUT and features of Townes–Brocks syndrome 2
- Author
-
Anne Christians, Esra Kesdiren, Imke Hennies, Alejandro Hofmann, Mark-Oliver Trowe, Frank Brand, Helge Martens, Ann Christin Gjerstad, Zoran Gucev, Matthias Zirngibl, Robert Geffers, Tomáš Seeman, Heiko Billing, Anna Bjerre, Velibor Tasic, Andreas Kispert, Benno Ure, Dieter Haffner, Jens Dingemann, and Ruthild G. Weber
- Subjects
Genetics ,Genetics (clinical) - Abstract
Most patients with congenital anomalies of the kidney and urinary tract (CAKUT) remain genetically unexplained. In search of novel genes associated with CAKUT in humans, we applied whole-exome sequencing in a patient with kidney, anorectal, spinal, and brain anomalies, and identified a rare heterozygous missense variant in the DACT1 (dishevelled binding antagonist of beta catenin 1) gene encoding a cytoplasmic WNT signaling mediator. Our patient’s features overlapped Townes–Brocks syndrome 2 (TBS2) previously described in a family carrying a DACT1 nonsense variant as well as those of Dact1-deficient mice. Therefore, we assessed the role of DACT1 in CAKUT pathogenesis. Taken together, very rare (minor allele frequency ≤ 0.0005) non-silent DACT1 variants were detected in eight of 209 (3.8%) CAKUT families, significantly more frequently than in controls (1.7%). All seven different DACT1 missense variants, predominantly likely pathogenic and exclusively maternally inherited, were located in the interaction region with DVL2 (dishevelled segment polarity protein 2), and biochemical characterization revealed reduced binding of mutant DACT1 to DVL2. Patients carrying DACT1 variants presented with kidney agenesis, duplex or (multi)cystic (hypo)dysplastic kidneys with hydronephrosis and TBS2 features. During murine development, Dact1 was expressed in organs affected by anomalies in patients with DACT1 variants, including the kidney, anal canal, vertebrae, and brain. In a branching morphogenesis assay, tubule formation was impaired in CRISPR/Cas9-induced Dact1−/− murine inner medullary collecting duct cells. In summary, we provide evidence that heterozygous hypomorphic DACT1 variants cause CAKUT and other features of TBS2, including anomalies of the skeleton, brain, distal digestive and genital tract.
- Published
- 2022
21. Permissive ureter specification by TBX18-mediated repression of metanephric gene expression
- Author
-
Anna-Carina Weiss, Eva Blank, Tobias Bohnenpoll, Marc-Jens Kleppa, Reginaldo Rivera-Reyes, Makoto Mark Taketo, Mark-Oliver Trowe, and Andreas Kispert
- Subjects
Molecular Biology ,Developmental Biology - Abstract
The murine kidney and ureter develop in a regionalized fashion from the ureteric bud and its surrounding mesenchyme. Whereas the factors that establish the metanephric cell lineages have been well characterized, much less is known about the molecular cues that specify the ureter. Here, we have identified a crucial patterning function in this process for Tbx18, a T-box transcription factor gene specifically expressed in the mesenchymal primordium of the ureter. Using misexpression and loss-of-function mice combined with molecular profiling approaches, we show that Tbx18 is required and sufficient to repress metanephric mesenchymal gene programs. We identify Wt1 as a functional target of TBX18. Our work suggests that TBX18 acts as a permissive factor in ureter specification by generating a mesenchymal domain around the distal ureteric bud where SHH and BMP4 signaling can occur.
- Published
- 2023
22. Genetic Variants in ARHGEF6 Cause Congenital Anomalies of the Kidneys and Urinary Tract in Humans, Mice, and Frogs
- Author
-
Verena Klämbt, Florian Buerger, Chunyan Wang, Thomas Naert, Karin Richter, Theresa Nauth, Anna-Carina Weiss, Tobias Sieckmann, Ethan Lai, Dervla M. Connaughton, Steve Seltzsam, Nina Mann, Amar J. Majmundar, Chen-Han W. Wu, Ana C. Onuchic-Whitford, Shirlee Shril, Sophia Schneider, Luca Schierbaum, Rufeng Dai, Mir Reza Bekheirnia, Marieke Joosten, Omer Shlomovitz, Asaf Vivante, Ehud Banne, Shrikant Mane, Richard P. Lifton, Karin M. Kirschner, Andreas Kispert, Georg Rosenberger, Klaus-Dieter Fischer, Soeren S. Lienkamp, Mirjam M.P. Zegers, and Friedhelm Hildebrandt
- Subjects
urinary tract ,ALPHA-PIX ,Women's cancers Radboud Institute for Molecular Life Sciences [Radboudumc 17] ,MUTATIONS ,Biology and Life Sciences ,General Medicine ,monogenic kidney disease ,REGULATES BRANCHING MORPHOGENESIS ,SEQUENCE ,pediatric ,All institutes and research themes of the Radboud University Medical Center ,Nephrology ,NUCLEOTIDE EXCHANGE FACTORS ,EXTRACELLULAR-MATRIX ,INBRED MOUSE ,INTEGRIN-LINKED KINASE ,PROTEIN NEPHRONECTIN ,MDCK CELL-CULTURE ,development ,CAKUT - Abstract
Background: About 40 disease genes have been described to date for isolated congenital anomalies of the kidneys and urinary tract (CAKUT), the most common cause of childhood chronic kidney disease. However, these genes account for only 20% of cases. ARHGEF6, a guanine nucleotide exchange factor that is implicated in such biologic processes as cell migration and focal adhesion, acts downstream of integrin linked kinase (ILK) and parvin proteins. A genetic variant of ILK that causes murine renal agenesis abrogates the interaction of ILK with a murine focal adhesion protein encoded by Parva, leading to CAKUT in mice with this variant. Methods: To identify novel genes that, when mutated, result in CAKUT, we performed exome sequencing in an international cohort of 1265 families with CAKUT. We also assessed the effects in vitro of wild-type and mutant ARHGEF6 proteins, as well as the effects of Arhgef6 deficiency in mouse and frog models. Results: We detected six different hemizygous variants in the gene ARHGEF6 (which is located on the X chromosome in humans) in eight individuals from six families with CAKUT. In kidney cells, overexpression of wild-type ARHGEF6—but not proband-derived mutant ARHGEF6— increased active levels of CDC42/RAC1, induced lamellipodia formation, and stimulated PARVAdependent cell spreading. ARHGEF6 mutant proteins showed loss of interaction with PARVA. Three-dimensional MDCK cell cultures expressing ARHGEF6 mutant proteins exhibited reduced lumen formation and polarity defects. Arhgef6 deficiency in mouse and frog models recapitulated features of human CAKUT. Conclusions: Deleterious variants in ARHGEF6 may cause dysregulation of integrin-parvinRAC1/CDC42 signaling, thereby leading to X-linked CAKUT.
- Published
- 2023
23. GATA6 is a crucial factor for Myocd expression in the visceral smooth muscle cell differentiation program of the murine ureter
- Author
-
Jennifer Kurz, Anna-Carina Weiss, Timo H.-W. Lüdtke, Lena Deuper, Mark-Oliver Trowe, Hauke Thiesler, Herbert Hildebrandt, Joerg Heineke, Stephen A. Duncan, and Andreas Kispert
- Subjects
Mice ,Myocytes, Smooth Muscle ,Animals ,Cell Differentiation ,Muscle, Smooth ,Ureter ,Muscle Development ,Molecular Biology ,Developmental Biology - Abstract
Smooth muscle cells (SMCs) are a crucial component of the mesenchymal wall of the ureter, as they account for the efficient removal of the urine from the renal pelvis to the bladder by means of their contractile activity. Here, we show that the zinc-finger transcription factor gene Gata6 is expressed in mesenchymal precursors of ureteric SMCs under the control of BMP4 signaling. Mice with a conditional loss of Gata6 in these precursors exhibit a delayed onset and reduced level of SMC differentiation and peristaltic activity, as well as dilatation of the ureter and renal pelvis (hydroureternephrosis) at birth and at postnatal stages. Molecular profiling revealed a delayed and reduced expression of the myogenic driver gene Myocd, but the activation of signaling pathways and transcription factors previously implicated in activation of the visceral SMC program in the ureter was unchanged. Additional gain-of-function experiments suggest that GATA6 cooperates with FOXF1 in Myocd activation and SMC differentiation, possibly as pioneer and lineage-determining factors, respectively.
- Published
- 2022
24. S100A1 expression characterizes terminally differentiated superficial cells in the urothelium of the murine bladder and ureter
- Author
-
Fairouz Qasrawi, Max Meuser, Finja Lehnhoff, Marjenna Schulte, and Andreas Kispert
- Subjects
Histology ,Calcium-Binding Proteins ,S100 Proteins ,Urinary Bladder ,Cell Differentiation ,Cell Biology ,Medical Laboratory Technology ,Mice ,Animals ,RNA ,RNA, Messenger ,Ureter ,Urothelium ,Molecular Biology - Abstract
The urothelium is a stratified epithelium that lines the inner surface of the components of the urinary drainage system. It is composed of a layer of basal cells, one or several layers of intermediate cells, and a layer of large luminal superficial or umbrella cells. In the mouse, only a small set of markers is available that allows easy molecular distinction of these urothelial cell types. Here, we analyzed expression of S100A1, a member of the S100 family of calcium-binding proteins, in the urothelium of the two major organs of the murine urinary tract, the ureter and the bladder. Using RNA in situ hybridization analysis, we found exclusive expression of S100a1 mRNA in luminal cells of the ureter from embryonic day (E)17.5 onwards and of the bladder from E15.5 to adulthood. Immunofluorescence analysis showed that expression of S100A1 protein is confined to terminally differentiated superficial cells of both the ureter and bladder where it localized to the nucleus and cytoplasm. We conclude that S100A1 is a suitable marker for mature superficial cells in the urothelial lining of the drainage system of the developing and mature mouse.
- Published
- 2022
25. Mesenchymal FGFR1 and FGFR2 control patterning of the ureteric mesenchyme by balancing SHH and BMP4 signaling
- Author
-
Lena Deuper, Max Meuser, Hauke Thiesler, Ulrich W. H. Jany, Carsten Rudat, Herbert Hildebrandt, Mark-Oliver Trowe, and Andreas Kispert
- Subjects
Mesoderm ,Mice ,Myocytes, Smooth Muscle ,Animals ,Cell Differentiation ,Hedgehog Proteins ,Bone Morphogenetic Protein 4 ,Ureter ,Molecular Biology ,Developmental Biology ,Signal Transduction - Abstract
The coordinated development of the mesenchymal and epithelial progenitors of the murine ureter depends on a complex interplay of diverse signaling activities. We have recently shown that epithelial FGFR2 signaling regulates stratification and differentiation of the epithelial compartment by enhancing epithelial Shh expression, and mesenchymal SHH and BMP4 activity. Here, we show that FGFR1 and FGFR2 expression in the mesenchymal primordium impinges on the SHH/BMP4 signaling axis to regulate mesenchymal patterning and differentiation. Mouse embryos with conditional loss of Fgfr1 and Fgfr2 in the ureteric mesenchyme exhibited reduced mesenchymal proliferation and prematurely activated lamina propria formation at the expense of the smooth muscle cell program. They also manifested hydroureter at birth. Molecular profiling detected increased SHH, WNT and retinoic acid signaling, whereas BMP4 signaling in the mesenchyme was reduced. Pharmacological activation of SHH signaling in combination with inhibition of BMP4 signaling recapitulated the cellular changes in explant cultures of wild-type ureters. Additional experiments suggest that mesenchymal FGFR1 and FGFR2 act as a sink for FGF ligands to dampen activation of Shh and BMP receptor gene expression by epithelial FGFR2 signaling.
- Published
- 2022
26. Expansion of the renal capsular stroma, ureteric bud branching defects and cryptorchidism in mice with<scp>W</scp>ilms tumor 1gene deletion in the stromal compartment of the developing kidney
- Author
-
Christoph Englert, Anna-Carina Weiss, Andreas Kispert, and Reginaldo Rivera-Reyes
- Subjects
Male ,0301 basic medicine ,Stromal cell ,Organogenesis ,Renal cortex ,Retinoic acid ,Biology ,Kidney ,urologic and male genital diseases ,Pathology and Forensic Medicine ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Stroma ,Cryptorchidism ,medicine ,Animals ,WT1 Proteins ,urogenital system ,Mesenchymal stem cell ,Gene Expression Regulation, Developmental ,Wilms' tumor ,medicine.disease ,female genital diseases and pregnancy complications ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Ureteric bud ,Ureter ,Biomarkers ,Gene Deletion - Abstract
Development of the mammalian kidney is orchestrated by reciprocal interactions of stromal and nephrogenic mesenchymal cells with the ureteric bud epithelium. Previous work showed that the transcription factor Wilms tumor 1 (WT1) acts in the nephrogenic lineage to maintain precursor cells, to drive the epithelial transition of aggregating precursors into a renal vesicle and to specify and maintain the podocyte fate. However, WT1 is expressed not only in the nephrogenic lineage but also transiently in stromal progenitors in the renal cortex. Here we report that specific deletion of Wt1 in the stromal lineage using the Foxd1cre driver line results at birth in cryptorchidism and hypoplastic kidneys that harbour fewer and enlarged ureteric bud tips and display an expansion of capsular stroma into the cortical region. In vivo and ex vivo analysis at earlier stages revealed that stromal loss of Wt1 reduces stromal proliferation, and delays and alters branching morphogenesis, resulting in a variant architecture of the collecting duct tree with an increase of single at the expense of bifurcated ureteric bud tips. Molecular analysis identified a transient reduction of Aldh1a2 expression and of retinoic acid signalling activity in stromal progenitors, and of Ret in ureteric bud tips. Administration of retinoic acid partly rescued the branching defects of mutant kidneys in culture. We propose that WT1 maintains retinoic acid signalling in the cortical stroma, which, in turn, assures proper levels and dynamics of Ret expression in the ureteric bud tips, and thus normal ramification of the ureteric tree. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2020
27. Loss ofAnks6leads to YAP deficiency and liver abnormalities
- Author
-
Nathan Herdman, Friedhelm Hildebrandt, Dean Yimlamai, Andreas Kispert, Blake McCourt, Kari Nejak-Bowen, Yijen L. Wu, Markus Schüler, Eugen Widmeier, Anna-Carina Weiss, Merlin Airik, Donna B. Stolz, Timo H. Lüdtke, and Rannar Airik
- Subjects
Muscle Proteins ,Biology ,Ciliopathies ,Cholangiocyte ,Mice ,03 medical and health sciences ,Morphogenesis ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Genetics (clinical) ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Mice, Knockout ,YAP1 ,0303 health sciences ,Hippo signaling pathway ,Bile duct ,Cilium ,030305 genetics & heredity ,TEA Domain Transcription Factors ,Cell Differentiation ,YAP-Signaling Proteins ,General Medicine ,medicine.disease ,Cell biology ,DNA-Binding Proteins ,Ciliopathy ,medicine.anatomical_structure ,Liver ,Hippo signaling ,General Article ,Bile Ducts ,Carrier Proteins ,Signal Transduction ,Transcription Factors - Abstract
ANKS6 is a ciliary protein that localizes to the proximal compartment of the primary cilium, where it regulates signaling. Mutations in the ANKS6 gene cause multiorgan ciliopathies in humans, which include laterality defects of the visceral organs, renal cysts as part of nephronophthisis and congenital hepatic fibrosis (CHF) in the liver. Although CHF together with liver ductal plate malformations are common features of several human ciliopathy syndromes, including nephronophthisis-related ciliopathies, the mechanism by which mutations in ciliary genes lead to bile duct developmental abnormalities is not understood. Here, we generated a knockout mouse model of Anks6 and show that ANKS6 function is required for bile duct morphogenesis and cholangiocyte differentiation. The loss of Anks6 causes ciliary abnormalities, ductal plate remodeling defects and periportal fibrosis in the liver. Our expression studies and biochemical analyses show that biliary abnormalities in Anks6-deficient livers result from the dysregulation of YAP transcriptional activity in the bile duct-lining epithelial cells. Mechanistically, our studies suggest, that ANKS6 antagonizes Hippo signaling in the liver during bile duct development by binding to Hippo pathway effector proteins YAP1, TAZ and TEAD4 and promoting their transcriptional activity. Together, this study reveals a novel function for ANKS6 in regulating Hippo signaling during organogenesis and provides mechanistic insights into the regulatory network controlling bile duct differentiation and morphogenesis during liver development.
- Published
- 2020
28. A 3D iPSC-differentiation model identifies interleukin-3 as a regulator of early human hematopoietic specification
- Author
-
Danny Jonigk, Mania Ackermann, Markus Abeln, Kathrin Haake, Mark Kühnel, Ariane H.H. Nguyen, Anna-Carina Weiss, Michael D. Milsom, Sylvia Merkert, Nico Lachmann, Paul Kaschutnig, Henning Kempf, Thomas Thum, Andreas Kispert, and Dorothee Hartmann
- Subjects
Adult ,Pluripotent Stem Cells ,0301 basic medicine ,Hemogenic endothelium ,CD43 ,Induced Pluripotent Stem Cells ,Regulator ,Cell Differentiation ,Hematology ,Biology ,Embryonic stem cell ,Article ,Hematopoiesis ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,Humans ,Interleukin-3 ,Progenitor cell ,Induced pluripotent stem cell ,030217 neurology & neurosurgery ,Interleukin 3 - Abstract
Hematopoietic development is spatiotemporally tightly regulated by defined cell-intrinsic and extrinsic modifiers. The role of cytokines has been intensively studied in adult hematopoiesis; however, their role in embryonic hematopoietic specification remains largely unexplored. Here, we used induced pluripotent stem cell (iPSC) technology and established a 3-dimensional, organoid-like differentiation system (hemanoid) maintaining the structural cellular integrity to evaluate the effect of cytokines on embryonic hematopoietic development. We show, that defined stages of early human hematopoietic development were recapitulated within the generated hemanoids. We identified KDR+/CD34high/CD144+/CD43-/CD45- hemato-endothelial progenitor cells (HEPs) forming organized, vasculature-like structures and giving rise to CD34low/CD144-/CD43+/CD45+ hematopoietic progenitor cells. We demonstrate that the endothelial to hematopoietic transition of HEPs is dependent on the presence of interleukin 3 (IL-3). Inhibition of IL-3 signalling blocked hematopoietic differentiation and arrested the cells in the HEP stage. Thus, our data suggest an important role for IL-3 in early human hematopoiesis by supporting the endothelial to hematopoietic transition of hemato-endothelial progenitor cells and highlight the potential of a hemanoid-based model to study human hematopoietic development.
- Published
- 2020
29. Inflammation-like changes in the urothelium of Lifr-deficient mice and LIFR-haploinsufficient humans with urinary tract anomalies
- Author
-
Ruthild G. Weber, Imke Hennies, Andreas Kispert, Dieter Haffner, Anne Christians, Maximilian Georg Klopf, Anna-Carina Weiss, and Helge Martens
- Subjects
Heterozygote ,Leukemia Inhibitory Factor Receptor alpha Subunit ,Urinary system ,030232 urology & nephrology ,Leukemia inhibitory factor receptor ,Context (language use) ,Haploinsufficiency ,Biology ,Kidney ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Exome Sequencing ,Genetics ,medicine ,Animals ,Humans ,Exome ,Urothelium ,Progenitor cell ,Urinary Tract ,Molecular Biology ,Genetics (clinical) ,030304 developmental biology ,Inflammation ,0303 health sciences ,General Medicine ,Pedigree ,medicine.anatomical_structure ,Urogenital Abnormalities ,Mutation ,Cancer research - Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of end-stage kidney disease in children. While the genetic aberrations underlying CAKUT pathogenesis are increasingly being elucidated, their consequences on a cellular and molecular level commonly remain unclear. Recently, we reported rare heterozygous deleterious LIFR variants in 3.3% of CAKUT patients, including a novel de novo frameshift variant, identified by whole-exome sequencing, in a patient with severe bilateral CAKUT. We also demonstrated CAKUT phenotypes in Lifr−/− and Lifr+/− mice, including a narrowed ureteric lumen due to muscular hypertrophy and a thickened urothelium. Here, we show that both in the ureter and bladder of Lifr−/− and Lifr+/− embryos, differentiation of the three urothelial cell types (basal, intermediate and superficial cells) occurs normally but that the turnover of superficial cells is elevated due to increased proliferation, enhanced differentiation from their progenitor cells (intermediate cells) and, importantly, shedding into the ureteric lumen. Microarray-based analysis of genome-wide transcriptional changes in Lifr−/− versus Lifr+/+ ureters identified gene networks associated with an antimicrobial inflammatory response. Finally, in a reverse phenotyping effort, significantly more superficial cells were detected in the urine of CAKUT patients with versus without LIFR variants indicating conserved LIFR-dependent urinary tract changes in the murine and human context. Our data suggest that LIFR signaling is required in the epithelium of the urinary tract to suppress an antimicrobial response under homeostatic conditions and that genetically induced inflammation-like changes underlie CAKUT pathogenesis in Lifr deficiency and LIFR haploinsufficiency.
- Published
- 2020
30. FGFR2 signaling enhances the SHH-BMP4 signaling axis in early ureter development
- Author
-
Max Meuser, Lena Deuper, Carsten Rudat, Nurullah Aydoğdu, Hauke Thiesler, Patricia Zarnovican, Herbert Hildebrandt, Mark-Oliver Trowe, and Andreas Kispert
- Subjects
animal structures ,Organogenesis ,Bone Morphogenetic Protein 4 ,Mesoderm ,Mice ,embryonic structures ,Animals ,Hedgehog Proteins ,Receptor, Fibroblast Growth Factor, Type 2 ,Ureter ,Urothelium ,Molecular Biology ,Signal Transduction ,Developmental Biology - Abstract
The patterned array of basal, intermediate and superficial cells in the urothelium of the mature ureter arises from uncommitted epithelial progenitors of the distal ureteric bud. Urothelial development requires signaling input from surrounding mesenchymal cells, which, in turn, depend on cues from the epithelial primordium to form a layered fibro-muscular wall. Here, we have identified FGFR2 as a crucial component in this reciprocal signaling crosstalk in the murine ureter. Loss of Fgfr2 in the ureteric epithelium led to reduced proliferation, stratification, intermediate and basal cell differentiation in this tissue, and affected cell survival and smooth muscle cell differentiation in the surrounding mesenchyme. Loss of Fgfr2 impacted negatively on epithelial expression of Shh and its mesenchymal effector gene Bmp4. Activation of SHH or BMP4 signaling largely rescued the cellular defects of mutant ureters in explant cultures. Conversely, inhibition of SHH or BMP signaling in wild-type ureters recapitulated the mutant phenotype in a dose-dependent manner. Our study suggests that FGF signals from the mesenchyme enhance, via epithelial FGFR2, the SHH-BMP4 signaling axis to drive urothelial and mesenchymal development in the early ureter.
- Published
- 2022
31. TBX2-positive cells represent a multi-potent mesenchymal progenitor pool in the developing lung
- Author
-
Vincent M. Christoffels, Timo H. Lüdtke, Mark-Oliver Trowe, Irina Wojahn, Andreas Kispert, Medical Biology, ACS - Heart failure & arrhythmias, and Amsterdam Reproduction & Development (AR&D)
- Subjects
0301 basic medicine ,Mesenchyme ,Cellular differentiation ,Cell ,Morphogenesis ,Mice, Transgenic ,Biology ,Lineage tracing ,03 medical and health sciences ,Mice ,Pregnancy ,medicine ,Animals ,Cell Lineage ,Progenitor cell ,Lung ,Cells, Cultured ,Progenitor ,lcsh:RC705-779 ,030102 biochemistry & molecular biology ,Research ,Mesenchymal stem cell ,Tbx2 ,Mesenchymal Stem Cells ,lcsh:Diseases of the respiratory system ,Embryonic stem cell ,Cell biology ,Pulmonary mesenchyme ,030104 developmental biology ,medicine.anatomical_structure ,Smooth muscle cells ,Lung development ,Female ,T-Box Domain Proteins - Abstract
BackgroundIn the embryonic mammalian lung, mesenchymal cells act both as a signaling center for epithelial proliferation, differentiation and morphogenesis as well as a source for a multitude of differentiated cell types that support the structure of the developing and mature organ. Whether the embryonic pulmonary mesenchyme is a homogenous precursor pool and how it diversifies into different cell lineages is poorly understood. We have previously shown that the T-box transcription factor geneTbx2is expressed in the pulmonary mesenchyme of the developing murine lung and is required therein to maintain branching morphogenesis.MethodsWe determined Tbx2/TBX2 expression in the developing murine lung by in situ hybridization and immunofluorescence analyses. We used a genetic lineage tracing approach with aCreline under the control of endogenousTbx2control elements (Tbx2cre), and theR26mTmGreporter line to trace TBX2-positive cells in the murine lung. We determined the fate of the TBX2 lineage by co-immunofluorescence analysis of the GFP reporter and differentiation markers in normal murine lungs and in lungs lacking or overexpressing TBX2 in the pulmonary mesenchyme.ResultsWe show that TBX2 is strongly expressed in mesenchymal progenitors in the developing murine lung. In differentiated smooth muscle cells and in fibroblasts, expression of TBX2 is still widespread but strongly reduced. In mesothelial and endothelial cells expression is more variable and scattered. All fetal smooth muscle cells, endothelial cells and fibroblasts derive from TBX2+progenitors, whereas half of the mesothelial cells have a different descent. The fate of TBX2-expressing cells is not changed inTbx2-deficient and inTBX2-constitutively overexpressing mice but the distribution and abundance of endothelial and smooth muscle cells is changed in the overexpression condition.ConclusionThe fate of pulmonary mesenchymal progenitors is largely independent of TBX2. Nevertheless, a successive and precisely timed downregulation of TBX2 is necessary to allow proper differentiation and functionality of bronchial smooth muscle cells and to limit endothelial differentiation. Our work suggests expression of TBX2 in an early pulmonary mesenchymal progenitor and supports a role of TBX2 in maintaining the precursor state of these cells.
- Published
- 2019
32. Combined genomic and proteomic approaches reveal DNA binding sites and interaction partners of TBX2 in the developing lung
- Author
-
Marc-Jens Kleppa, Patrick Künzler, Timo H. Lüdtke, Irina Wojahn, Jasper Schierstaedt, Andreas Kispert, Vincent M. Christoffels, Medical Biology, ACS - Heart failure & arrhythmias, and Amsterdam Reproduction & Development (AR&D)
- Subjects
0301 basic medicine ,Proteomics ,Chromosomal Proteins, Non-Histone ,Fluorescent Antibody Technique ,Polymerase Chain Reaction ,0302 clinical medicine ,Tandem Mass Spectrometry ,HDAC ,Lung ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,HMGB2 ,PBX1 ,Pre-B-Cell Leukemia Transcription Factor 1 ,Wnt signaling pathway ,Gene Expression Regulation, Developmental ,Tbx2 ,Genomics ,Cell biology ,030220 oncology & carcinogenesis ,Chromatin Immunoprecipitation Sequencing ,Lung development ,Mi-2 Nucleosome Remodeling and Deacetylase Complex ,Spectrometry, Mass, Electrospray Ionization ,Biology ,Chromatin remodeling ,CCN Intercellular Signaling Proteins ,03 medical and health sciences ,Proto-Oncogene Proteins ,NuRD ,Nucleosome ,Animals ,HMGB2 Protein ,Humans ,Transcription factor ,Cell Proliferation ,lcsh:RC705-779 ,Binding Sites ,Gene Expression Profiling ,Research ,lcsh:Diseases of the respiratory system ,Interleukin-33 ,DNA binding site ,Pulmonary mesenchyme ,030104 developmental biology ,HEK293 Cells ,CBX3 ,Frzb ,Homeobox ,T-Box Domain Proteins ,Chromatin immunoprecipitation ,Chromatography, Liquid - Abstract
Background Tbx2 encodes a transcriptional repressor implicated in the development of numerous organs in mouse. During lung development TBX2 maintains the proliferation of mesenchymal progenitors, and hence, epithelial proliferation and branching morphogenesis. The pro-proliferative function was traced to direct repression of the cell-cycle inhibitor genes Cdkn1a and Cdkn1b, as well as of genes encoding WNT antagonists, Frzb and Shisa3, to increase pro-proliferative WNT signaling. Despite these important molecular insights, we still lack knowledge of the DNA occupancy of TBX2 in the genome, and of the protein interaction partners involved in transcriptional repression of target genes. Methods We used chromatin immunoprecipitation (ChIP)-sequencing and expression analyses to identify genomic DNA-binding sites and transcription units directly regulated by TBX2 in the developing lung. Moreover, we purified TBX2 containing protein complexes from embryonic lung tissue and identified potential interaction partners by subsequent liquid chromatography/mass spectrometry. The interaction with candidate proteins was validated by immunofluorescence, proximity ligation and individual co-immunoprecipitation analyses. Results We identified Il33 and Ccn4 as additional direct target genes of TBX2 in the pulmonary mesenchyme. Analyzing TBX2 occupancy data unveiled the enrichment of five consensus sequences, three of which match T-box binding elements. The remaining two correspond to a high mobility group (HMG)-box and a homeobox consensus sequence motif. We found and validated binding of TBX2 to the HMG-box transcription factor HMGB2 and the homeobox transcription factor PBX1, to the heterochromatin protein CBX3, and to various members of the nucleosome remodeling and deacetylase (NuRD) chromatin remodeling complex including HDAC1, HDAC2 and CHD4. Conclusion Our data suggest that TBX2 interacts with homeobox and HMG-box transcription factors as well as with the NuRD chromatin remodeling complex to repress transcription of anti-proliferative genes in the pulmonary mesenchyme.
- Published
- 2021
33. Proteomic analysis identifies ZMYM2 as endogenous binding partner of TBX18 protein in 293 and A549 cells
- Author
-
Timo H.-W. Lüdtke, Marc-Jens Kleppa, Reginaldo Rivera-Reyes, Fairouz Qasrawi, Dervla M. Connaughton, Shirlee Shril, Friedhelm Hildebrandt, and Andreas Kispert
- Subjects
Cell Nucleus ,Proteomics ,Vesico-Ureteral Reflux ,Embryonic Development ,Gene Expression Regulation, Developmental ,Cell Biology ,Transfection ,Biochemistry ,DNA-Binding Proteins ,Mice ,A549 Cells ,Pregnancy ,Urogenital Abnormalities ,Mutation ,Animals ,Humans ,Female ,Gene Knock-In Techniques ,Ureter ,T-Box Domain Proteins ,Molecular Biology ,Protein Binding ,Signal Transduction ,Transcription Factors - Abstract
The TBX18 transcription factor regulates patterning and differentiation programs in the primordia of many organs yet the molecular complexes in which TBX18 resides to exert its crucial transcriptional function in these embryonic contexts have remained elusive. Here, we used 293 and A549 cells as an accessible cell source to search for endogenous protein interaction partners of TBX18 by an unbiased proteomic approach. We tagged endogenous TBX18 by CRISPR/Cas9 targeted genome editing with a triple FLAG peptide, and identified by anti-FLAG affinity purification and subsequent LC–MS analysis the ZMYM2 protein to be statistically enriched together with TBX18 in both 293 and A549 nuclear extracts. Using a variety of assays, we confirmed the binding of TBX18 to ZMYM2, a component of the CoREST transcriptional corepressor complex. Tbx18 is coexpressed with Zmym2 in the mesenchymal compartment of the developing ureter of the mouse, and mutations in TBX18 and in ZMYM2 were recently linked to congenital anomalies in the kidney and urinary tract (CAKUT) in line with a possible in vivo relevance of TBX18–ZMYM2 protein interaction in ureter development.
- Published
- 2021
34. Growth differentiation factor 11 attenuates liver fibrosis via expansion of liver progenitor cells
- Author
-
Hildegard Büning, Xizhong Shen, Amar Deep Sharma, Michael Ott, Taihua Yang, Elmar Jaeckel, Guangqi Song, Tobias Cantz, Anna-Carina Weiss, Qinggong Yuan, Xuemei Jiang, Asha Balakrishnan, Michael Manns, Selina Möbus, Zhen Dai, Martin Bentler, Heike Bantel, Arndt Vogel, Andreas Kispert, Anna Saborowski, and Ji-Min Zhu
- Subjects
Gene Flow ,Liver Cirrhosis ,Male ,Cirrhosis ,Fluorescent Antibody Technique ,Chronic liver disease ,Mice ,Fibrosis ,Animals ,Humans ,Medicine ,Progenitor cell ,In Situ Hybridization ,Mice, Inbred BALB C ,business.industry ,Stem Cells ,Liver cell ,Gastroenterology ,Growth differentiation factor ,medicine.disease ,Up-Regulation ,Growth Differentiation Factors ,Disease Models, Animal ,Liver ,Bone Morphogenetic Proteins ,Cancer research ,Hepatic stellate cell ,Stem cell ,business - Abstract
ObjectiveLiver fibrosis and cirrhosis resulting from chronic liver injury represent a major healthcare burden worldwide. Growth differentiation factor (GDF) 11 has been recently investigated for its role in rejuvenation of ageing organs, but its role in chronic liver diseases has remained unknown. Here, we investigated the expression and function of GDF11 in liver fibrosis, a common feature of most chronic liver diseases.DesignWe analysed the expression of GDF11 in patients with liver fibrosis, in a mouse model of liver fibrosis and in hepatic stellate cells (HSCs) as well as in other liver cell types. The functional relevance of GDF11 in toxin-induced and cholestasis-induced mouse models of liver fibrosis was examined by in vivo modulation of Gdf11 expression using adeno-associated virus (AAV) vectors. The effect of GDF11 on leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5)+ liver progenitor cells was studied in mouse and human liver organoid culture. Furthermore, in vivo depletion of LGR5+ cells was induced by injecting AAV vectors expressing diptheria toxin A under the transcriptional control of Lgr5 promoter.ResultsWe showed that the expression of GDF11 is upregulated in patients with liver fibrosis and in experimentally induced murine liver fibrosis models. Furthermore, we found that therapeutic application of GDF11 mounts a protective response against fibrosis by increasing the number of LGR5+ progenitor cells in the liver.ConclusionCollectively, our findings uncover a protective role of GDF11 during liver fibrosis and suggest a potential application of GDF11 for the treatment of chronic liver disease.
- Published
- 2019
35. Dexamethasone improves therapeutic outcomes in a preclinical bacterial epididymitis mouse model
- Author
-
Carsten Rudat, Kate L Loveland, Sudhanshu Bhushan, Rukmali Wijayarathna, Mark P. Hedger, Adrian Pilatz, Ralf Middendorff, Swapnila Pant, Julia Kautz, Britta Klein, Andreas Kispert, and Andreas Meinhardt
- Subjects
Male ,Epithelial-Mesenchymal Transition ,Anti-Inflammatory Agents ,Drug Evaluation, Preclinical ,Physiology ,Levofloxacin ,Adaptive Immunity ,Dexamethasone ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Animals ,Humans ,Medicine ,Infertility, Male ,030304 developmental biology ,Subclinical infection ,Epididymis ,Epididymitis ,0303 health sciences ,030219 obstetrics & reproductive medicine ,biology ,business.industry ,Rehabilitation ,Obstetrics and Gynecology ,medicine.disease ,Fibrosis ,Bacterial Load ,Anti-Bacterial Agents ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Immunoglobulin M ,Reproductive Medicine ,Immunoglobulin G ,biology.protein ,Cytokines ,Drug Therapy, Combination ,business ,medicine.drug - Abstract
STUDY QUESTION Can dexamethasone improve infertility-related cauda epididymidal tissue damage caused by bacterial epididymitis? SUMMARY ANSWER Dexamethasone in addition to anti-microbial treatment effectively reduces long-term deleterious epididymal tissue damage by dampening the host’s adaptive immune response. WHAT IS KNOWN ALREADY Despite effective anti-microbial treatment, ~40% of patients with epididymitis experience subsequent sub- or infertility. An epididymitis mouse model has shown that the host immune response is mainly responsible for the magnitude of epididymal tissue damage that is fundamentally causative of the subsequent fertility issues. STUDY DESIGN, SIZE, DURATION Bacterial epididymitis was induced in male mice by using uropathogenic Escherichia coli (UPEC). From Day 3 after infection onwards, mice were treated with daily doses of levofloxacin (20 mg/kg, total n = 12 mice), dexamethasone (0.5 mg/kg, total n = 9) or both in combination (total n = 11) for seven consecutive days. Control animals were left untreated, i.e. given no interventional treatment following UPEC infection (total n = 11). Half of the animals from each group were killed either at 10 or 31 days post-infection. PARTICIPANTS/MATERIALS, SETTING, METHODS A mouse model of induced bacterial epididymitis was applied to adult male C57BL/6J mice. At the respective endpoints (10 or 31 days post-infection), epididymides were collected. Effectiveness of antibiotic treatment was assessed by plating of epididymal homogenates onto lysogeny broth agar plates. Overall tissue morphology and the degree and nature of tissue damage were assessed histologically. Quantitative RT-PCR was used to assess local cytokine transcript levels. Blood was drawn and serum analysed for systemic IgG and IgM levels by ELISA. In addition, correlation analyses of clinical data and serum-analyses of IgG and IgM levels in patients with epididymitis were performed. MAIN RESULTS AND THE ROLE OF CHANCE The addition of dexamethasone to the standard anti-microbial treatment did not further worsen epididymal tissue integrity. In fact, an obviously dampened immune response and reduced tissue reaction/damage was observed at both 10 and 31 days post-infection following combined treatment. More specifically, epididymal duct continuity was preserved, enabling sperm transit. In contrast, in untreated or antibiotic-treated animals, damage of the epididymal duct and duct constrictions were observed, associated with a lack of cauda spermatozoa. In line with the bacteriostatic/bactericidal effect of levofloxacin (alone as well as in combination), local cytokine transcript levels were significantly and similarly reduced in animals treated with levofloxacin alone (P LIMITATIONS, REASONS FOR CAUTION Breeding studies to address the fertility-protecting effect of the combined treatment were not possible in the experimental animals because the vas deferens was ligated (model specific). WIDER IMPLICATIONS OF THE FINDINGS Whereas innate immunity is necessary and involved in acute bacterial clearance, adaptive immunity seems to be responsible for long-term, subclinical immunological activities that may negatively affect the pathogenesis of bacterial epididymitis even after effective bacterial eradication. These effects can be reduced in mice by the additional treatment with dexamethasone. This immunological characteristic of bacterial epididymitis shows similarities to the Jarisch–Herxheimer reaction known from other types of bacterial infection. STUDY FUNDING/COMPETING INTEREST(S) The study was supported by grants from the Deutsche Forschungsgemeinschaft, Monash University and the Medical Faculty of Justus-Liebig University to the International Research Training Group on ‘Molecular pathogenesis of male reproductive disorders’ (GRK 1871). R.W., K.L.L. and M.P.H. were supported by grants from the National Health and Medical Research Council of Australia (ID1079646, ID1081987, ID1020269 and ID1063843) and by the Victorian Government’s Operational Infrastructure Support Program. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER No clinical trial involved.
- Published
- 2019
36. Corrigendum to: ‘Hepatocyte-specific suppression of microRNA-221-3p mitigates liver fibrosis’ [J Hepatol (2019) 722-734]
- Author
-
Hsin-Chieh Tsay, Qinggong Yuan, Asha Balakrishnan, Marina Kaiser, Selina Möbus, Emilia Kozdrowska, Marwa Farid, Pia-Katharina Tegtmeyer, Katharina Borst, Florian W.R. Vondran, Ulrich Kalinke, Andreas Kispert, Michael P. Manns, Michael Ott, and Amar Deep Sharma
- Subjects
Hepatology - Published
- 2022
37. Uridine diphosphate-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase deletion in mice leads to lethal intracerebral hemorrhage during embryonic development
- Author
-
Markus Abeln, Anja K. Münster-Kühnel, Anna-Carina Weiss, Henri Wedekind, Andreas Kispert, Harald Neumann, Christine Klaus, Rüdiger Horstkorte, Elina Kats, Hauke Thiesler, and Birgit Weinhold
- Subjects
Mice, Knockout ,Fetus ,Angiogenesis ,Embryogenesis ,Embryonic Development ,Embryo ,Biology ,Biochemistry ,Embryonic stem cell ,N-Acetylneuraminic Acid ,Sialic acid ,Cell biology ,Neuroepithelial cell ,chemistry.chemical_compound ,Mice ,medicine.anatomical_structure ,chemistry ,Multienzyme Complexes ,Placenta ,embryonic structures ,medicine ,Biocatalysis ,Animals ,Cerebral Hemorrhage - Abstract
Among the enzymes of the biosynthesis of sialoglycoconjugates, uridine diphosphate-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase (GNE), catalyzing the first essential step of the sialic acid (Sia) de novo biosynthesis, and cytidine monophosphate (CMP)-Sia synthase (CMAS), activating Sia to CMP-Sia, are particularly important. The knockout of either of these enzymes in mice is embryonically lethal. While the lethality of Cmas−/− mice has been attributed to a maternal complement attack against asialo fetal placental cells, the cause of lethality in Gne-deficient embryos has remained elusive. Here, we advanced the significance of sialylation for embryonic development through detailed histological analyses of Gne−/− embryos and placentae. We found that Gne−/− embryonic and extraembryonic tissues are hyposialylated rather than being completely deficient of sialoglycans, which holds true for Cmas−/− embryos. Residual sialylation of Gne−/− cells can be explained by scavenging free Sia from sialylated maternal serum glycoconjugates via the lysosomal salvage pathway. The placental architecture of Gne−/− mice was unaffected, but severe hemorrhages in the neuroepithelium with extensive bleeding into the cephalic ventricles were present at E12.5 in the mutants. At E13.5, the vast majority of Gne−/− embryos were asystolic. This phenotype persisted when Gne−/− mice were backcrossed to a complement component 3-deficient background, confirming distinct pathomechanisms of Cmas−/− and Gne−/− mice. We conclude that the low level of sialylation observed in Gne−/− mice is sufficient both for immune homeostasis at the fetal–maternal interface and for embryonic development until E12.5. However, formation of the neural microvasculature is the first critical process, depending on a higher degree of sialylation during development of the embryo proper.
- Published
- 2021
38. Notch signaling is a novel regulator of visceral smooth muscle cell differentiation in the murine ureter
- Author
-
Jennifer Kurz, Anna-Carina Weiss, Hauke Thiesler, Fairouz Qasrawi, Lena Deuper, Jaskiran Kaur, Carsten Rudat, Timo H. Lüdtke, Irina Wojahn, Herbert Hildebrandt, Mark-Oliver Trowe, and Andreas Kispert
- Subjects
Male ,Mice, Knockout ,Receptors, Notch ,Myocytes, Smooth Muscle ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Cell Differentiation ,Diamines ,Actins ,Mice ,Thiazoles ,Viscera ,Immunoglobulin J Recombination Signal Sequence-Binding Protein ,cardiovascular system ,Trans-Activators ,Animals ,Female ,Ureter ,Molecular Biology ,Jagged-1 Protein ,Developmental Biology ,Signal Transduction - Abstract
The contractile phenotype of smooth muscle cells (SMCs) is transcriptionally controlled by a complex of the DNA-binding protein SRF and the transcriptional co-activator MYOCD. The pathways that activate expression of Myocd and of SMC structural genes in mesenchymal progenitors are diverse, reflecting different intrinsic and extrinsic signaling inputs. Taking the ureter as a model, we analyzed whether Notch signaling, a pathway previously implicated in vascular SMC development, also affects visceral SMC differentiation. We show that mice with a conditional deletion of the unique Notch mediator RBPJ in the undifferentiated ureteric mesenchyme exhibit altered ureter peristalsis with a delayed onset, and decreased contraction frequency and intensity at fetal stages. They also develop hydroureter 2 weeks after birth. Notch signaling is required for precise temporal activation of Myocd expression and, independently, for expression of a group of late SMC structural genes. Based on additional expression analyses, we suggest that a mesenchymal JAG1-NOTCH2/NOTCH3 module regulates visceral SMC differentiation in the ureter in a biphasic and bimodal manner, and that its molecular function differs from that in the vascular system.
- Published
- 2021
39. Rare heterozygous GDF6 variants in patients with renal anomalies
- Author
-
Anne Christians, Soeren S. Lienkamp, Maike Getwan, Ruthild G. Weber, Robert Geffers, Imke Hennies, Zoran Gucev, Arne Christians, Frank Brand, Andreas Kispert, Anna-Carina Weiss, Ann Christin Gjerstad, Helge Martens, Dieter Haffner, Velibor Tasic, Tomáš Seeman, Anna Bjerre, HZI,Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr. 7,38124 Braunschweig, Germany., University of Zurich, and Weber, Ruthild G
- Subjects
0301 basic medicine ,Adult ,Male ,Pathology ,medicine.medical_specialty ,2716 Genetics (clinical) ,Heterozygote ,10017 Institute of Anatomy ,Adolescent ,Xenopus ,030232 urology & nephrology ,610 Medicine & health ,In situ hybridization ,Biology ,Growth Differentiation Factor 6 ,Development ,Microphthalmia ,Article ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,1311 Genetics ,Genetics research ,Genetics ,medicine ,Animals ,Humans ,Child ,Genetics (clinical) ,Vesico-Ureteral Reflux ,Coloboma ,Kidney ,Renal ectopia ,Medical genetics ,Infant ,medicine.disease ,Phenotype ,Pronephros ,030104 developmental biology ,medicine.anatomical_structure ,Kidney Tubules ,GDF6 ,10076 Center for Integrative Human Physiology ,Child, Preschool ,Urogenital Abnormalities ,Mutation ,570 Life sciences ,biology ,Female - Abstract
Although over 50 genes are known to cause renal malformation if mutated, the underlying genetic basis, most easily identified in syndromic cases, remains unsolved in most patients. In search of novel causative genes, whole-exome sequencing in a patient with renal, i.e., crossed fused renal ectopia, and extrarenal, i.e., skeletal, eye, and ear, malformations yielded a rare heterozygous variant in the GDF6 gene encoding growth differentiation factor 6, a member of the BMP family of ligands. Previously, GDF6 variants were reported to cause pleiotropic defects including skeletal, e.g., vertebral, carpal, tarsal fusions, and ocular, e.g., microphthalmia and coloboma, phenotypes. To assess the role of GDF6 in the pathogenesis of renal malformation, we performed targeted sequencing in 193 further patients identifying rare GDF6 variants in two cases with kidney hypodysplasia and extrarenal manifestations. During development, gdf6 was expressed in the pronephric tubule of Xenopus laevis, and Gdf6 expression was observed in the ureteric tree of the murine kidney by RNA in situ hybridization. CRISPR/Cas9-derived knockout of Gdf6 attenuated migration of murine IMCD3 cells, an effect rescued by expression of wild-type but not mutant GDF6, indicating affected variant function regarding a fundamental developmental process. Knockdown of gdf6 in Xenopus laevis resulted in impaired pronephros development. Altogether, we identified rare heterozygous GDF6 variants in 1.6% of all renal anomaly patients and 5.4% of renal anomaly patients additionally manifesting skeletal, ocular, or auricular abnormalities, adding renal hypodysplasia and fusion to the phenotype spectrum of GDF6 variant carriers and suggesting an involvement of GDF6 in nephrogenesis.
- Published
- 2020
40. WNT6/ACC2-induced storage of triacylglycerols in macrophages is exploited by Mycobacterium tuberculosis
- Author
-
Lara Linnemann, Andreas Kispert, Michael Leitges, Martina Hein, Tim Vierbuchen, Annette S. Gross, Alexandra Hölscher, Christoph Lange, Franziska Waldow, Dominik Schwudke, Julius Brandenburg, Christoph Hölscher, Ulrich E. Schaible, Holger Heine, Jan Rupp, Barbara Kalsdorf, Thomas Scholzen, Simone C. Tazoll, Norbert Reiling, Jochen Behrends, Torsten Goldmann, Stefan Niemann, Maja Reimann, Svenja Goldenbaum, and Sebastian Marwitz
- Subjects
Tuberculosis ,medicine.medical_treatment ,Antitubercular Agents ,Biology ,WNT6 ,Mycobacterium tuberculosis ,Mice ,Proto-Oncogene Proteins ,medicine ,Isoniazid ,Animals ,Humans ,Enzyme Inhibitors ,Lung ,Tuberculosis, Pulmonary ,Triglycerides ,Foam cell ,Mice, Knockout ,Host Microbial Interactions ,Macrophages ,Wnt signaling pathway ,General Medicine ,medicine.disease ,biology.organism_classification ,Mice, Inbred C57BL ,Wnt Proteins ,Cytokine ,Adjunctive treatment ,Cancer research ,Intracellular ,Acetyl-CoA Carboxylase ,Foam Cells ,Signal Transduction ,Research Article - Abstract
In view of emerging drug-resistant tuberculosis (TB), host-directed adjunct therapies are urgently needed to improve treatment outcomes with currently available anti-TB therapies. One approach is to interfere with the formation of lipid-laden "foamy" macrophages in the host, as they provide a nutrient-rich host cell environment for Mycobacterium tuberculosis (Mtb). Here, we provide evidence that Wnt family member 6 (WNT6), a ligand of the evolutionarily conserved Wingless/Integrase 1 (WNT) signaling pathway, promotes foam cell formation by regulating key lipid metabolic genes including acetyl-CoA carboxylase 2 (ACC2) during pulmonary TB. Using genetic and pharmacological approaches, we demonstrated that lack of functional WNT6 or ACC2 significantly reduced intracellular triacylglycerol (TAG) levels and Mtb survival in macrophages. Moreover, treatment of Mtb-infected mice with a combination of a pharmacological ACC2 inhibitor and the anti-TB drug isoniazid (INH) reduced lung TAG and cytokine levels, as well as lung weights, compared with treatment with INH alone. This combination also reduced Mtb bacterial numbers and the size of mononuclear cell infiltrates in livers of infected mice. In summary, our findings demonstrate that Mtb exploits WNT6/ACC2-induced storage of TAGs in macrophages to facilitate its intracellular survival, a finding that opens new perspectives for host-directed adjunctive treatment of pulmonary TB.
- Published
- 2020
41. WNT6-ACC2-induced accumulation of triacylglycerol rich lipid droplets is exploited by M. tuberculosis
- Author
-
Christoph Hoelscher, Franziska Waldow, Martina Hein, Jan Rupp, Julius Brandenburg, Andreas Kispert, Barbara Kalsdorf, Sebastian Marwitz, Michael Leitges, Christoph Lange, Alexandra Hoelscher, Annette S. Gross, Tim Vierbuchen, Dominik Schwudke, Simone C. Tazoll, Jochen Behrends, Norbert Reiling, Stefan Niemann, Ulrich E. Schaible, Svenja Goldenbaum, Thomas Scholzen, Holger Heine, Lara Linnemann, Torsten Goldmann, and Maja Reimann
- Subjects
Mycobacterium tuberculosis ,WNT6 ,Tuberculosis ,biology ,Lipid droplet ,Wnt signaling pathway ,medicine ,Signal transduction ,biology.organism_classification ,medicine.disease ,Intracellular ,Cell biology ,Foam cell - Abstract
In view of emerging drug-resistant tuberculosis, host directed therapies are urgently needed to improve treatment outcomes with currently available anti-tuberculosis therapies. One option is to interfere with the formation of lipid-laden “foamy” macrophages in the infected host. Here, we provide evidence that WNT6, a member of the evolutionary conserved WNT signaling pathway, promotes foam cell formation by regulating key lipid metabolic genes including acetyl-CoA carboxylase-2 (ACC2) during pulmonary TB. In addition, we demonstrate that Mycobacterium tuberculosis (Mtb) facilitates its intracellular growth and dissemination in the host by exploiting the WNT6-ACC2 pathway. Using genetic and pharmacological approaches, we show that lack of functional WNT6 or ACC2 significantly reduces intracellular TAG levels, Mtb growth and necrotic cell death of macrophages. In combination with the anti-TB drug isoniazid, pharmacological inhibition of ACC2 improved anti-mycobacterial treatment in vitro and in vivo. Therefore, we propose the WNT6-ACC2 signaling pathway as a promising target for a host-directed therapy to reduce intracellular replication of Mtb by modulating neutral lipid metabolism.
- Published
- 2020
42. Growth differentiation factor 11 mitigates liver fibrosis via expansion of liver progenitor cells
- Author
-
Guangqi Song, Tobias Cantz, Heike Bantel, Hildegard Büning, Elmar Jaeckel, Martin Bentler, Zhen Dai, Amar Deep Sharma, Taihua Yang, Asha Balakrishnan, Andreas Kispert, Selina Möbus, Anna-Carina Weiss, Michael P. Manns, Michael Ott, Qinggong Yuan, Arndt Vogel, and Anna Saborowski
- Subjects
Liver fibrosis ,GDF11 ,Cancer research ,Biology ,Progenitor cell - Published
- 2020
43. Generation of hiPSC-derived low threshold mechanoreceptors containing axonal termini resembling bulbous sensory nerve endings and expressing Piezo1 and Piezo2
- Author
-
Andreas Kispert, Norman Kalmbach, Adam Grundhoff, Florian Wegner, Reto Eggenschwiler, Teng-Cheong Ha, Ana Gomis, Tobias Cantz, Tamrat M. Mamo, Georges M. G. M. Verjans, Andreas Leffler, Kai A. Kropp, Likai Tan, Shuyong Zhu, Axel Schambach, Richard J. C. Brown, Volkhard Kaever, Manuela Schmidt, Pengfei Yu, David Twapokera Mzinza, Birgit Ritter, Jorge Fernández-Trillo, Abel Viejo-Borbolla, Werner J. D. Ouwendijk, Michael Spohn, Nancy Stanslowsky, Pratibha Narayanan, Reinhold Förster, Virology, European Commission, German Research Foundation, Ministry for Science and Culture of Lower Saxony, Agencia Estatal de Investigación (España), and Ministerio de Ciencia, Innovación y Universidades (España)
- Subjects
Small molecule-derived neural precursor cells ,Sensory Receptor Cells ,QH301-705.5 ,Low threshold mechanoreceptors ,Induced Pluripotent Stem Cells ,Piezo1, Piezo2 ,Biology ,Somatosensory system ,Mechanotransduction, Cellular ,Ion Channels ,Bulbous sensory nerve ending ,medicine ,Humans ,Biology (General) ,Axon ,Mechanotransduction ,Process (anatomy) ,Nerve Endings ,Mechanosensation ,PIEZO1 ,Piezo2 ,Human induced pluripotent stem cells ,Cell Biology ,General Medicine ,Piezo1 ,Sensory Nerve Endings ,medicine.anatomical_structure ,nervous system ,Stammzelle ,Mechanoreceptors ,Neuroscience ,Free nerve ending ,Developmental Biology - Abstract
Somatosensory low threshold mechanoreceptors (LTMRs) sense innocuous mechanical forces, largely through specialized axon termini termed sensory nerve endings, where the mechanotransduction process initiates upon activation of mechanotransducers. In humans, a subset of sensory nerve endings is enlarged, forming bulb-like expansions, termed bulbous nerve endings. There is no in vitro human model to study these neuronal endings. Piezo2 is the main mechanotransducer found in LTMRs. Recent evidence shows that Piezo1, the other mechanotransducer considered absent in dorsal root ganglia (DRG), is expressed at low level in somatosensory neurons. We established a differentiation protocol to generate, from iPSC-derived neuronal precursor cells, human LTMR recapitulating bulbous sensory nerve endings and heterogeneous expression of Piezo1 and Piezo2. The derived neurons express LTMR-specific genes, convert mechanical stimuli into electrical signals and have specialized axon termini that morphologically resemble bulbous nerve endings. Piezo2 is concentrated within these enlarged axon termini. Some derived neurons express low level Piezo1, and a subset co-express both channels. Thus, we generated a unique, iPSCs-derived human model that can be used to investigate the physiology of bulbous sensory nerve endings, and the role of Piezo1 and 2 during mechanosensation., This work was supported by N-RENNT of the Ministry of Science and Culture of Lower Saxony to A.V.B., by a Marie Curie Career Integration Grant to A.V.B. (FP7-PEOPLE-2013-CIG, project number 631792, acronym INMA), by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany’s Excellence Strategy – EXC 2155 “RESIST” – Project ID 39087428, by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) - SFB-900 – 158989968 to R.F. (B1) and A.V.B. (B9), and by a Spanish Government project to A.G. (PID2019-108194RB-I00).
- Published
- 2021
44. Retinoic acid signaling maintains epithelial and mesenchymal progenitors in the developing mouse ureter
- Author
-
Mark-Oliver Trowe, Anna-Carina Weiss, Tobias Bohnenpoll, Timo H. Lüdtke, Maurice Labuhn, and Andreas Kispert
- Subjects
0301 basic medicine ,Cellular differentiation ,Mesenchyme ,Myocytes, Smooth Muscle ,Retinoic acid ,lcsh:Medicine ,Mice, Transgenic ,Tretinoin ,Biology ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Fibrocyte ,medicine ,Animals ,Progenitor cell ,lcsh:Science ,Lamina propria ,Multidisciplinary ,Mesenchymal stem cell ,lcsh:R ,Gene Expression Regulation, Developmental ,Epithelial Cells ,Mesenchymal Stem Cells ,Anatomy ,Embryo, Mammalian ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Ureteric bud ,lcsh:Q ,Ureter ,Signal Transduction - Abstract
The differentiated cell types of the mature ureter arise from the distal ureteric bud epithelium and its surrounding mesenchyme. Uncommitted epithelial cells first become intermediate cells from which both basal and superficial cells develop. Mesenchymal progenitors give rise to separated layers of adventitial fibrocytes, smooth muscle cells and lamina propria fibrocytes. How progenitor expansion and differentiation are balanced is poorly understood. Here, we addressed the role of retinoic acid (RA) signaling in these programs. Using expression analysis of components and target genes, we show that pathway activity is restricted to the mesenchymal and epithelial progenitor pools. Inhibition of RA signaling in ureter explant cultures resulted in tissue hypoplasia with a relative expansion of smooth muscle cells at the expense of lamina propria fibroblasts in the mesenchyme, and of superficial cells at the expense of intermediate cells in the ureteric epithelium. Administration of RA led to a slight reduction of smooth muscle cells, and almost completely prevented differentiation of intermediate cells into basal and superficial cells. We identified cellular programs and transcriptional targets of RA signaling that may account for this activity. We conclude that RA signaling is required and sufficient to maintain mesenchymal and epithelial progenitors in early ureter development.
- Published
- 2017
45. A Dominant Mutation in Nuclear Receptor Interacting Protein 1 Causes Urinary Tract Malformations via Dysregulation of Retinoic Acid Signaling
- Author
-
Soeren S. Lienkamp, Richard P. Lifton, Benjamin Dekel, Anna Carina Weiss, Friedhelm Hildebrandt, Vincent Cavaillès, Michael M. Kaminski, Amelie T. van der Ven, Tobias Bohnenpoll, Johanna Magdalena Schmidt, Rachel Shukrun, Hadas Ityel, Ali G. Gharavi, Eugen Widmeier, Weining Lu, Hagith Yonath, Jing Chen, Yair Anikster, Andreas Kispert, Nina Mann, Stuart B. Bauer, Daniella Magen, Asaf Vivante, Robert Kleta, Velibor Tasic, Shirlee Shril, Maike Getwan, Catherine Teyssier, Horia Stanescu, Simone Sanna-Cherchi, Sheba Medical Center, Medizinische Hochschule Hannover (MHH), Department of Neurology, Children's Hospital [Boston], Boston Children's Hospital, Columbia University, New York, NY, United States, Rambam Health Care Campus, Tel Aviv University [Tel Aviv], Yale University School of Medicine, Department of Pediatric Nephrology, University Children’s Hospital, Centre for Nephrology [London, UK], University College of London [London] (UCL), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Centre for Biological Signaling Studies [Freiburg] (BIOSS), University of Freiburg [Freiburg], Pediatrics, University of Michigan [Ann Arbor], and University of Michigan System-University of Michigan System
- Subjects
0301 basic medicine ,Retinoic acid ,Tretinoin ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Retinoic acid receptor beta ,030105 genetics & heredity ,Biology ,Retinoic acid-inducible orphan G protein-coupled receptor ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,retinoic acid ,Animals ,Urinary Tract ,ComputingMilieux_MISCELLANEOUS ,CAKUT ,Adaptor Proteins, Signal Transducing ,NRIP1 ,Nuclear Proteins ,General Medicine ,Retinoic acid receptor gamma ,Retinoid X receptor gamma ,Molecular biology ,Nuclear Receptor Interacting Protein 1 ,3. Good health ,Retinoic acid receptor ,Basic Research ,030104 developmental biology ,Nuclear receptor ,chemistry ,Nephrology ,Retinoic acid receptor alpha ,Mutation ,Signal Transduction - Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of CKD in the first three decades of life. However, for most patients with CAKUT, the causative mutation remains unknown. We identified a kindred with an autosomal dominant form of CAKUT. By whole-exome sequencing, we identified a heterozygous truncating mutation (c.279delG, p.Trp93fs*) of the nuclear receptor interacting protein 1 gene (NRIP1) in all seven affected members. NRIP1 encodes a nuclear receptor transcriptional cofactor that directly interacts with the retinoic acid receptors (RARs) to modulate retinoic acid transcriptional activity. Unlike wild-type NRIP1, the altered NRIP1 protein did not translocate to the nucleus, did not interact with RARα, and failed to inhibit retinoic acid–dependent transcriptional activity upon expression in HEK293 cells. Notably, we also showed that treatment with retinoic acid enhanced NRIP1 binding to RARα. RNA in situ hybridization confirmed Nrip1 expression in the developing urogenital system of the mouse. In explant cultures of embryonic kidney rudiments, retinoic acid stimulated Nrip1 expression, whereas a pan-RAR antagonist strongly reduced it. Furthermore, mice heterozygous for a null allele of Nrip1 showed a CAKUT-spectrum phenotype. Finally, expression and knockdown experiments in Xenopus laevis confirmed an evolutionarily conserved role for NRIP1 in renal development. These data indicate that dominant NRIP1 mutations can cause CAKUT by interference with retinoic acid transcriptional signaling, shedding light on the well documented association between abnormal vitamin A levels and renal malformations in humans, and suggest a possible gene-environment pathomechanism in this disease.
- Published
- 2017
46. Mutations in the leukemia inhibitory factor receptor (LIFR) gene and Lifr deficiency cause urinary tract malformations
- Author
-
Lars Pape, Frank Brand, Stephanie Schubert, Dieter Haffner, Ruthild G. Weber, Kerstin Amann, Lars Krogvold, Imke Hennies, Jan Hinrich Bräsen, Michaela Goerk, Christoph Daniel, Anna Bjerre, Andreas Kispert, Anne-Kathrin Schäfer, Anne Kosfeld, Vera Riehmer, Helge Martens, Anna-Carina Weiss, and Martin Kreuzer
- Subjects
Adult ,Male ,0301 basic medicine ,Heterozygote ,Adolescent ,Leukemia Inhibitory Factor Receptor alpha Subunit ,Receptors, OSM-LIF ,DNA Mutational Analysis ,Leukemia inhibitory factor receptor ,Biology ,Kidney ,Leukemia Inhibitory Factor ,Frameshift mutation ,Mice ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Humans ,Missense mutation ,Exome ,Child ,Urinary Tract ,Molecular Biology ,Genetics (clinical) ,Exome sequencing ,Mice, Knockout ,Infant ,Heterozygote advantage ,Sequence Analysis, DNA ,General Medicine ,030104 developmental biology ,medicine.anatomical_structure ,Child, Preschool ,Urogenital Abnormalities ,Mutation ,Knockout mouse ,Cancer research ,Female ,Ureter ,Leukemia inhibitory factor - Abstract
Congenital anomalies of the kidneys and urinary tract (CAKUT) are the most common cause of chronic kidney disease in children. As CAKUT is a genetically heterogeneous disorder and most cases are genetically unexplained, we aimed to identify new CAKUT causing genes. Using whole-exome sequencing and trio-based de novo analysis, we identified a novel heterozygous de novo frameshift variant in the leukemia inhibitory factor receptor (LIFR) gene causing instability of the mRNA in a patient presenting with bilateral CAKUT and requiring kidney transplantation at one year of age. LIFR encodes a transmembrane receptor utilized by IL-6 family cytokines, mainly by the leukemia inhibitory factor (LIF). Mutational analysis of 121 further patients with severe CAKUT yielded two rare heterozygous LIFR missense variants predicted to be pathogenic in three unrelated patients. LIFR mutants showed decreased half-life and cell membrane localization resulting in reduced LIF-stimulated STAT3 phosphorylation. LIFR showed high expression in human fetal kidney and the human ureter, and was also expressed in the developing murine urogenital system. Lifr knockout mice displayed urinary tract malformations including hydronephrosis, hydroureter, ureter ectopia, and, consistently, reduced ureteral lumen and muscular hypertrophy, similar to the phenotypes observed in patients carrying LIFR variants. Additionally, a form of cryptorchidism was detected in all Lifr-/- mice and the patient carrying the LIFR frameshift mutation. Altogether, we demonstrate heterozygous novel or rare LIFR mutations in 3.3% of CAKUT patients, and provide evidence that Lifr deficiency and deactivating LIFR mutations cause highly similar anomalies of the urogenital tract in mice and humans.
- Published
- 2017
47. Diversification of Cell Lineages in Ureter Development
- Author
-
Tobias Bohnenpoll, Sarah Feraric, Max Meuser, Anna-Carina Weiss, Carsten Rudat, Andreas Kispert, Marvin Nattkemper, and Mark-Oliver Trowe
- Subjects
0301 basic medicine ,Cell type ,Cellular differentiation ,Biology ,Mesoderm ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Fibrocyte ,medicine ,Animals ,Cell Lineage ,Progenitor cell ,Lamina propria ,Cell growth ,Mesenchymal stem cell ,Cell Differentiation ,Epithelial Cells ,Muscle, Smooth ,General Medicine ,Embryonic stem cell ,Cell biology ,Basic Research ,030104 developmental biology ,medicine.anatomical_structure ,Nephrology ,030220 oncology & carcinogenesis ,Ureter - Abstract
The mammalian ureter consists of a mesenchymal wall composed of smooth muscle cells and surrounding fibrocytes of the tunica adventitia and the lamina propria and an inner epithelial lining composed of layers of basal, intermediate, and superficial cells. How these cell types arise from multipotent progenitors is poorly understood. Here, we performed marker analysis, cell proliferation assays, and genetic lineage tracing to define the lineage relations and restrictions of the mesenchymal and epithelial cell types in the developing and mature mouse ureter. At embryonic day (E) 12.5, the mesenchymal precursor pool began to subdivide into an inner and outer compartment that began to express markers of smooth muscle precursors and adventitial fibrocytes, respectively, by E13.5. Smooth muscle precursors further diversified into lamina propria cells directly adjacent to the ureteric epithelium and differentiated smooth muscle cells from E16.5 onwards. Uncommitted epithelial progenitors of the ureter differentiated into intermediate cells at E14.5. After stratification into two layers at E15.5 and three cell layers at E18.5, intermediate cells differentiated into basal cells and superficial cells. In homeostasis, proliferation of all epithelial and mesenchymal cell types remained low but intermediate cells still gave rise to basal cells, whereas basal cells divided only into basal cells. These studies provide a framework to further determine the molecular mechanisms of cell differentiation in the tissues of the developing ureter.
- Published
- 2016
48. Heparan Sulfate-Editing Extracellular Sulfatases Enhance VEGF Bioavailability for Ischemic Heart Repair
- Author
-
Mortimer Korf-Klingebiel, Marc R. Reboll, Karsten Grote, Hauke Schleiner, Yong Wang, Xuekun Wu, Stefanie Klede, Yuliya Mikhed, André Nobre, Johann Bauersachs, Michael Klintschar, Carsten Rudat, Andreas Kispert, Hans W. Niessen, Torben Lübke, Thomas Dierks, Kai C. Wollert, Cardio-thoracic surgery, Pathology, and ACS - Heart failure & arrhythmias
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,Physiology ,Myocardial Ischemia ,Biological Availability ,Inflammation ,030204 cardiovascular system & hematology ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,medicine ,Extracellular ,Animals ,Humans ,Myocardial infarction ,Mice, Knockout ,business.industry ,Sulfatase ,Heparan sulfate ,medicine.disease ,Bioavailability ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Heart failure ,medicine.symptom ,Sulfatases ,Cardiology and Cardiovascular Medicine ,Ischemic heart ,business ,Extracellular Space - Abstract
Rationale: Mechanistic insight into the inflammatory response after acute myocardial infarction may inform new molecularly targeted treatment strategies to prevent chronic heart failure. Objective: We identified the sulfatase SULF2 in an in silico secretome analysis in bone marrow cells from patients with acute myocardial infarction and detected increased sulfatase activity in myocardial autopsy samples. SULF2 (Sulf2 in mice) and its isoform SULF1 (Sulf1) act as endosulfatases removing 6- O -sulfate groups from heparan sulfate (HS) in the extracellular space, thus eliminating docking sites for HS-binding proteins. We hypothesized that the Sulfs have a role in tissue repair after myocardial infarction. Methods and Results: Both Sulfs were dynamically upregulated after coronary artery ligation in mice, attaining peak expression and activity levels during the first week after injury. Sulf2 was expressed by monocytes and macrophages, Sulf1 by endothelial cells and fibroblasts. Infarct border zone capillarization was impaired, scar size increased, and cardiac dysfunction more pronounced in mice with a genetic deletion of either Sulf1 or Sulf2. Studies in bone marrow-chimeric Sulf-deficient mice and Sulf-deficient cardiac endothelial cells established that inflammatory cell-derived Sulf2 and endothelial cell-autonomous Sulf1 promote angiogenesis. Mechanistically, both Sulfs reduced HS sulfation in the infarcted myocardium, thereby diminishing Vegfa (vascular endothelial growth factor A) interaction with HS. Along this line, both Sulfs rendered infarcted mouse heart explants responsive to the angiogenic effects of HS-binding Vegfa 164 but did not modulate the angiogenic effects of non-HS-binding Vegfa 120 . Treating wild-type mice systemically with the small molecule HS-antagonist surfen (bis-2-methyl-4-amino-quinolyl-6-carbamide, 1 mg/kg/day) for 7 days after myocardial infarction released Vegfa from HS, enhanced infarct border-zone capillarization, and exerted sustained beneficial effects on cardiac function and survival. Conclusions: These findings establish HS-editing Sulfs as critical inducers of postinfarction angiogenesis and identify HS sulfation as a therapeutic target for ischemic tissue repair.
- Published
- 2019
49. The transcription factor <scp>GATA</scp> 4 promotes myocardial regeneration in neonatal mice
- Author
-
Carsten Rudat, Johann Bauersachs, Anna Gigina, Joerg Heineke, Badder Kattih, Qiangrong Liang, Natali Froese, Mortimer Korf-Klingebiel, Andrea Grund, Ulrike Schrameck, Kai C. Wollert, Mona Malek Mohammadi, and Andreas Kispert
- Subjects
0301 basic medicine ,Medicine (General) ,Transcription, Genetic ,Angiogenesis ,ComputingMethodologies_IMAGEPROCESSINGANDCOMPUTERVISION ,Gene Expression ,Biology ,QH426-470 ,IL‐13 ,Regenerative Medicine ,Cardiovascular System ,neonatal cryoinfarction ,03 medical and health sciences ,GATA4 ,Mice ,R5-920 ,Downregulation and upregulation ,Transduction, Genetic ,medicine ,Genetics ,Animals ,Regeneration ,Transcription factor ,Research Articles ,ComputingMilieux_MISCELLANEOUS ,Mice, Knockout ,Regeneration (biology) ,Stem Cells ,cardiac regeneration ,Heart ,medicine.disease ,Cell biology ,GATA4 Transcription Factor ,030104 developmental biology ,Animals, Newborn ,Gene Expression Regulation ,Heart Injuries ,Heart failure ,cardiomyocyte proliferation ,Interleukin 13 ,Immunology ,Systemic administration ,cardiovascular system ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Molecular Medicine ,Corrigendum ,Gene Deletion ,Research Article - Abstract
Heart failure is often the consequence of insufficient cardiac regeneration. Neonatal mice retain a certain capability of myocardial regeneration until postnatal day (P)7, although the underlying transcriptional mechanisms remain largely unknown. We demonstrate here that cardiac abundance of the transcription factor GATA4 was high at P1, but became strongly reduced at P7 in parallel with loss of regenerative capacity. Reconstitution of cardiac GATA4 levels by adenoviral gene transfer markedly improved cardiac regeneration after cryoinjury at P7. In contrast, the myocardial scar was larger in cardiomyocyte‐specific Gata4 knockout (CM‐G4‐KO) mice after cryoinjury at P0, indicative of impaired regeneration, which was accompanied by reduced cardiomyocyte proliferation and reduced myocardial angiogenesis in CM‐G4‐KO mice. Cardiomyocyte proliferation was also diminished in cardiac explants from CM‐G4‐KO mice and in isolated cardiomyocytes with reduced GATA4 expression. Mechanistically, decreased GATA4 levels caused the downregulation of several pro‐regenerative genes (among them interleukin‐13, Il13) in the myocardium. Interestingly, systemic administration of IL‐13 rescued defective heart regeneration in CM‐G4‐KO mice and could be evaluated as therapeutic strategy in the future.
- Published
- 2019
50. Mesothelial mobilization in the developing lung and heart differs in timing, quantity, and pathway dependency
- Author
-
Andreas Kispert, Franziska Greulich, Nurullah Aydoğdu, Mark-Oliver Trowe, Marc-Jens Kleppa, Tamrat M. Mamo, Regine Häfner, Irina Wojahn, Carsten Rudat, Tobias Bohnenpoll, Jennifer Kurz, Timo H. Lüdtke, and Makoto Mark Taketo
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Male ,Pathology ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Physiology ,Gestational Age ,Mice, Transgenic ,030204 cardiovascular system & hematology ,Epithelium ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Movement ,Pregnancy ,Physiology (medical) ,medicine ,Animals ,WT1 Proteins ,Lung ,Wnt Signaling Pathway ,Heart development ,Thoracic cavity ,business.industry ,Myocardium ,Gestational age ,Heart ,Cell Biology ,medicine.disease ,Embryonic stem cell ,Immunohistochemistry ,Mice, Mutant Strains ,Mesothelium ,030104 developmental biology ,medicine.anatomical_structure ,Female ,business ,Signal Transduction - Abstract
The mesothelial lining of the lung, the visceral pleura, and of the heart, the epicardium, derive from a common multipotent precursor tissue, the mesothelium of the embryonic thoracic cavity that also contributes to organ-specific mesenchymal cell types. Insight into mesothelial mobilization and differentiation has prevailedin the developing heart while the mesenchymal transition and fate of the visceral pleura are poorly understood. Here, we use the fact that the early mesothelium of both the lung and the heart expresses the transcription factor gene Wt1, to comparatively analyze mesothelial mobilization in the two organs by a genetic cre-loxP-based conditional approach. We show that epicardial cells are mobilized in a large number between E12.5 and E14.5, whereas pleural mobilization occurs only sporadically and variably in few regions of the lung in a temporally highly confined manner shortly after E12.5. Mesothelium-specific inactivation of unique pathway components using a Wt1creERT2line excluded a requirement for canonical WNT, NOTCH, HH, TGFB, PDGFRA, and FGFR1/FGFR2 signaling in the mesenchymal transition of the visceral pleura but indicated a deleterious effect of activated WNT, NOTCH, and HH signaling on lung development. Epicardial mobilization was negatively impacted on by loss of HH, PDGFRA, FGFR1/2 signaling. Epicardial overactivation of WNT, NOTCH, and HH disturbed epicardial and myocardial integrity. We conclude that mesothelial mobilization in the developing lung and heart differs in timing, quantity and pathway dependency, indicating the organ specificity of the program.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.