53 results on '"Lesche M"'
Search Results
2. Controlling distinct signaling states in cultured cancer cells provides a new platform for drug discovery
- Author
-
Poser, S.W. Otto, O. Arps-Forker, C. Ge, Y. Herbig, M. Andree, C. Gruetzmann, K. Adasme, M.F. Stodolak, S. Nikolakopoulou, P. Park, D.M. Mcintyre, A. Lesche, M. Dahl, A. Lennig, P. Bornstein, S.R. Schroeck, E. Klink, B. Leker, R.R. Bickle, M. Chrousos, G.P. Schroeder, M. Cannistraci, C.V. Guck, J. Androutsellis-Theotokis, A.
- Abstract
Cancer cells can switch between signaling pathways to regulate growth under different conditions. In the tumor microenvironment, this likely helps them evade therapies that target specific pathways. We must identify all possible states and utilize them in drug screening programs. One such state is characterized by expression of the transcription factor Hairy and Enhancer of Split 3 (HES3) and sensitivity to HES3 knockdown, and it can be modeled in vitro. Here, we cultured 3 primary human brain cancer cell lines under 3 different culture conditions that maintain low, medium, and high HES3 expression and characterized gene regulation and mechanical phenotype in these states. We assessed gene expression regulation following HES3 knockdown in the HES3-high conditions. We then employed a commonly used human brain tumor cell line to screen Food and Drug Administration (FDA)-approved compounds that specifically target the HES3-high state. We report that cells from multiple patients behave similarly when placed under distinct culture conditions. We identified 37 FDA-approved compounds that specifically kill cancer cells in the high-HES3–expression conditions. Our work reveals a novel signaling state in cancer, biomarkers, a strategy to identify treatments against it, and a set of putative drugs for potential repurposing.—Poser, S. W., Otto, O., Arps-Forker, C., Ge, Y., Herbig, M., Andree, C., Gruetzmann, K., Adasme, M. F., Stodolak, S., Nikolakopoulou, P., Park, D. M., Mcintyre, A., Lesche, M., Dahl, A., Lennig, P., Bornstein, S. R., Schroeck, E., Klink, B., Leker, R. R., Bickle, M., Chrousos, G. P., Schroeder, M., Cannistraci, C. V., Guck, J., Androutsellis-Theotokis, A. Controlling distinct signaling states in cultured cancer cells provides a new platform for drug discovery. FASEB J. 33, 9235–9249 (2019). www.fasebj.org. © FASEB
- Published
- 2019
3. The RNA binding protein HuR is a master regulator of hepatic-lipid homeostasis
- Author
-
Subramanian, P, additional, Grzybek, M, additional, Palladini, A, additional, Sinha, A, additional, Gargani, S, additional, Mitroulis, I, additional, Lesche, M, additional, Burkhardt, R, additional, Brosch, M, additional, Techritz, N, additional, Witt, A, additional, Chung, KJ, additional, Nati, M, additional, Dahl, A, additional, Mirtschink, P, additional, Zamboni, N, additional, Coskun, Ü, additional, Hampe, J, additional, Kontoyiannis, D, additional, and Chavakis, T, additional
- Published
- 2019
- Full Text
- View/download PDF
4. Systems biology of the IMIDIA biobank from organ donors and pancreatectomised patients defines a novel transcriptomic signature of islets from individuals with type 2 diabetes
- Author
-
Solimena, Michele, Schulte, A., Marselli, L., Ehehalt, F., Richter, D., Kleeberg, M., Mziaut, H., Knoch, K., Parnis, J., Bugliani, M., Siddiq, A., Jörns, A., Burdet, F., Liechti, R., Suleiman, M., Margerie, D., Syed, F., Distler, M., Grützmann, R., Petretto, E., Moreno-Moral, A., Wegbrod, C., Sönmez, A., Pfriem, K., Friedrich, A., Meinel, J., Wollheim, C., Baretton, G., Scharfmann, R., Nogoceke, E., Bonifacio, E., Sturm, D., Meyer-Puttlitz, B., Boggi, U., Saeger, H., Filipponi, F., Lesche, M., Meda, P., Dahl, A., Wigger, L., Xenarios, I., Falchi, M., Thorens, B., Weitz, J., Bokvist, K., Lenzen, S., Rutter, G., Froguel, P., von Bülow, M., Ibberson, M., Marchetti, P., Solimena, Michele, Schulte, A., Marselli, L., Ehehalt, F., Richter, D., Kleeberg, M., Mziaut, H., Knoch, K., Parnis, J., Bugliani, M., Siddiq, A., Jörns, A., Burdet, F., Liechti, R., Suleiman, M., Margerie, D., Syed, F., Distler, M., Grützmann, R., Petretto, E., Moreno-Moral, A., Wegbrod, C., Sönmez, A., Pfriem, K., Friedrich, A., Meinel, J., Wollheim, C., Baretton, G., Scharfmann, R., Nogoceke, E., Bonifacio, E., Sturm, D., Meyer-Puttlitz, B., Boggi, U., Saeger, H., Filipponi, F., Lesche, M., Meda, P., Dahl, A., Wigger, L., Xenarios, I., Falchi, M., Thorens, B., Weitz, J., Bokvist, K., Lenzen, S., Rutter, G., Froguel, P., von Bülow, M., Ibberson, M., and Marchetti, P.
- Abstract
© 2017, The Author(s). Aims/hypothesis: Pancreatic islet beta cell failure causes type 2 diabetes in humans. To identify transcriptomic changes in type 2 diabetic islets, the Innovative Medicines Initiative for Diabetes: Improving beta-cell function and identification of diagnostic biomarkers for treatment monitoring in Diabetes (IMIDIA) consortium (www.imidia.org) established a comprehensive, unique multicentre biobank of human islets and pancreas tissues from organ donors and metabolically phenotyped pancreatectomised patients (PPP). Methods: Affymetrix microarrays were used to assess the islet transcriptome of islets isolated either by enzymatic digestion from 103 organ donors (OD), including 84 non-diabetic and 19 type 2 diabetic individuals, or by laser capture microdissection (LCM) from surgical specimens of 103 PPP, including 32 non-diabetic, 36 with type 2 diabetes, 15 with impaired glucose tolerance (IGT) and 20 with recent-onset diabetes (<1 year), conceivably secondary to the pancreatic disorder leading to surgery (type 3c diabetes). Bioinformatics tools were used to (1) compare the islet transcriptome of type 2 diabetic vs non-diabetic OD and PPP as well as vs IGT and type 3c diabetes within the PPP group; and (2) identify transcription factors driving gene co-expression modules correlated with insulin secretion ex vivo and glucose tolerance in vivo. Selected genes of interest were validated for their expression and function in beta cells. Results: Comparative transcriptomic analysis identified 19 genes differentially expressed (false discovery rate =0.05, fold change =1.5) in type 2 diabetic vs non-diabetic islets from OD and PPP. Nine out of these 19 dysregulated genes were not previously reported to be dysregulated in type 2 diabetic islets. Signature genes included TMEM37, which inhibited Ca2+-influx and insulin secretion in beta cells, and ARG2 and PPP1R1A, which promoted insulin secretion. Systems biology approaches identified HNF1A, PDX1 and RE
- Published
- 2018
5. Analyses of clonality of BcrAbl-induced leukemia by Genetic Barcodes
- Author
-
Cornils, K, additional, Thielecke, L, additional, Winkelmann, D, additional, Lesche, M, additional, Aranyossy, T, additional, Dahl, A, additional, Roeder, I, additional, Fehse, B, additional, and Glauche, I, additional
- Published
- 2017
- Full Text
- View/download PDF
6. Detection of low frequency variants of the NLRP3 gene in “mutation- negative” CAPS patients using massive parallel sequencing
- Author
-
Thiem, J, primary, Lesche, M, additional, Dahl, A, additional, Kränkel, A, additional, Roesler, J, additional, and Rösen-Wolff, A, additional
- Published
- 2015
- Full Text
- View/download PDF
7. Nachweis von low frequency Varianten des NLRP3 Gens bei 'mutations-negativen' CAPS Patienten mit massive parallel sequencing
- Author
-
Lesche, M, Dahl, A, Kränkel, A, Roesler, J, Thiem, J, Rösen-Wolff, A, Lesche, M, Dahl, A, Kränkel, A, Roesler, J, Thiem, J, and Rösen-Wolff, A
- Published
- 2014
8. PW02-026 - Low frequency variants of NLRP3 in CAPS patients
- Author
-
Lesche, M, primary, Dahl, A, additional, Kränkel, A, additional, Roesler, J, additional, and Rösen-Wolff, A, additional
- Published
- 2013
- Full Text
- View/download PDF
9. The clinical evaluation of various prostheses.
- Author
-
Lesche, M.
- Subjects
DENTURES ,DENTAL implants ,PROSTHODONTICS ,DENTAL metallurgy ,DENTAL caries ,DENTAL abutments ,DENTAL care ,TOOTH care & hygiene ,DENTAL research ,MAINTENANCE - Abstract
The article offers information on a study that examines the 613 metal-framework prostheses and 133 cast metal splinting prostheses which had been placed in the mouth for five to eight years. It explores the impact of the metal framework on the development of dental caries. It indicates the increase in caries development in teeth bearing prosthesis clasps and the rate of extraction after eight years. It cites the need for a yearly check-up appointments to maintain abutment teeth for a long time and increase the serviceable period of the prosthesis.
- Published
- 1979
10. Lifelong persistence of nuclear RNAs in the mouse brain.
- Author
-
Zocher S, McCloskey A, Karasinsky A, Schulte R, Friedrich U, Lesche M, Rund N, Gage FH, Hetzer MW, and Toda T
- Subjects
- Animals, Mice, Gene Expression Regulation, Heterochromatin genetics, Brain metabolism, Chromatin, RNA, Nuclear genetics
- Abstract
Genomic DNA that resides in the nuclei of mammalian neurons can be as old as the organism itself. The life span of nuclear RNAs, which are critical for proper chromatin architecture and transcription regulation, has not been determined in adult tissues. In this work, we identified and characterized nuclear RNAs that do not turn over for at least 2 years in a subset of postnatally born cells in the mouse brain. These long-lived RNAs were stably retained in nuclei in a neural cell type-specific manner and were required for the maintenance of heterochromatin. Thus, the life span of neural cells may depend on both the molecular longevity of DNA for the storage of genetic information and also the extreme stability of RNA for the functional organization of chromatin.
- Published
- 2024
- Full Text
- View/download PDF
11. BRAF and MEK inhibitor combinations induce potent molecular and immunological effects in NRAS-mutant melanoma cells: Insights into mode of action and resistance mechanisms.
- Author
-
Dinter L, Karitzky PC, Schulz A, Wurm AA, Mehnert MC, Sergon M, Tunger A, Lesche M, Wehner R, Müller A, Käubler T, Niessner H, Dahl A, Beissert S, Schmitz M, Meier F, Seliger B, and Westphal D
- Subjects
- Humans, Proto-Oncogene Proteins B-raf, Vemurafenib, Protein Kinase Inhibitors adverse effects, Mitogen-Activated Protein Kinase Kinases, Mutation, Drug Resistance, Neoplasm genetics, Membrane Proteins genetics, GTP Phosphohydrolases genetics, Melanoma drug therapy, Melanoma genetics, Melanoma metabolism
- Abstract
About 25% of melanoma harbor activating NRAS mutations, which are associated with aggressive disease therefore requiring a rapid antitumor intervention. However, no efficient targeted therapy options are currently available for patients with NRAS-mutant melanoma. MEK inhibitors (MEKi) appear to display a moderate antitumor activity and also immunological effects in NRAS-mutant melanoma, providing an ideal backbone for combination treatments. In our study, the MEKi binimetinib, cobimetinib and trametinib combined with the BRAF inhibitors (BRAFi) encorafenib, vemurafenib and dabrafenib were investigated for their ability to inhibit proliferation, induce apoptosis and alter the expression of immune modulatory molecules in sensitive NRAS-mutant melanoma cells using two- and three-dimensional cell culture models as well as RNA sequencing analyses. Furthermore, NRAS-mutant melanoma cells resistant to the three BRAFi/MEKi combinations were established to characterize the mechanisms contributing to their resistance. All BRAFi induced a stress response in the sensitive NRAS-mutant melanoma cells thereby significantly enhancing the antiproliferative and proapoptotic activity of the MEKi analyzed. Furthermore, BRAFi/MEKi combinations upregulated immune relevant molecules, such as ICOS-L, components of antigen-presenting machinery and the "don't eat me signal" molecule CD47 in the melanoma cells. The BRAFi/MEKi-resistant, NRAS-mutant melanoma cells counteracted the molecular and immunological effects of BRAFi/MEKi by upregulating downstream mitogen-activated protein kinase pathway molecules, inhibiting apoptosis and promoting immune escape mechanisms. Together, our study reveals potent molecular and immunological effects of BRAFi/MEKi in sensitive NRAS-mutant melanoma cells that may be exploited in new combinational treatment strategies for patients with NRAS-mutant melanoma., (© 2023 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)
- Published
- 2024
- Full Text
- View/download PDF
12. Compartmentalization and synergy of osteoblasts drive bone formation in the regenerating fin.
- Author
-
Cudak N, López-Delgado AC, Rost F, Kurth T, Lesche M, Reinhardt S, Dahl A, Rulands S, and Knopf F
- Abstract
Zebrafish regenerate their fins which involves a component of cell plasticity. It is currently unclear how regenerate cells divide labor to allow for appropriate growth and patterning. Here, we studied lineage relationships of fluorescence-activated cell sorting-enriched epidermal, bone-forming (osteoblast), and (non-osteoblast) blastemal fin regenerate cells by single-cell RNA sequencing, lineage tracing, targeted osteoblast ablation, and electron microscopy. Most osteoblasts in the outgrowing regenerate derive from osterix + osteoblasts, while mmp9 + cells reside at segment joints. Distal blastema cells contribute to distal osteoblast progenitors, suggesting compartmentalization of the regenerating appendage. Ablation of osterix + osteoblasts impairs segment joint and bone matrix formation and decreases regenerate length which is partially compensated for by distal regenerate cells. Our study characterizes expression patterns and lineage relationships of rare fin regenerate cell populations, indicates inherent detection and compensation of impaired regeneration, suggests variable dependence on growth factor signaling, and demonstrates zonation of the elongating fin regenerate., Competing Interests: The authors declare no competing interests., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Regulation of multiple signaling pathways promotes the consistent expansion of human pancreatic progenitors in defined conditions.
- Author
-
Jarc L, Bandral M, Zanfrini E, Lesche M, Kufrin V, Sendra R, Pezzolla D, Giannios I, Khattak S, Neumann K, Ludwig B, and Gavalas A
- Subjects
- Humans, Pancreas, Wnt Signaling Pathway, Biological Assay, Pluripotent Stem Cells, Diabetes Mellitus
- Abstract
The unlimited expansion of human progenitor cells in vitro could unlock many prospects for regenerative medicine. However, it remains an important challenge as it requires the decoupling of the mechanisms supporting progenitor self-renewal and expansion from those mechanisms promoting their differentiation. This study focuses on the expansion of human pluripotent stem (hPS) cell-derived pancreatic progenitors (PP) to advance novel therapies for diabetes. We obtained mechanistic insights into PP expansion requirements and identified conditions for the robust and unlimited expansion of hPS cell-derived PP cells under GMP-compliant conditions through a hypothesis-driven iterative approach. We show that the combined stimulation of specific mitogenic pathways, suppression of retinoic acid signaling, and inhibition of selected branches of the TGFβ and Wnt signaling pathways are necessary for the effective decoupling of PP proliferation from differentiation. This enabled the reproducible, 2000-fold, over 10 passages and 40-45 d, expansion of PDX1
+ /SOX9+ /NKX6-1+ PP cells. Transcriptome analyses confirmed the stabilization of PP identity and the effective suppression of differentiation. Using these conditions, PDX1+ /SOX9+ /NKX6-1+ PP cells, derived from different, both XY and XX, hPS cell lines, were enriched to nearly 90% homogeneity and expanded with very similar kinetics and efficiency. Furthermore, non-expanded and expanded PP cells, from different hPS cell lines, were differentiated in microwells into homogeneous islet-like clusters (SC-islets) with very similar efficiency. These clusters contained abundant β-cells of comparable functionality as assessed by glucose-stimulated insulin secretion assays. These findings established the signaling requirements to decouple PP proliferation from differentiation and allowed the consistent expansion of hPS cell-derived PP cells. They will enable the establishment of large banks of GMP-produced PP cells derived from diverse hPS cell lines. This approach will streamline SC-islet production for further development of the differentiation process, diabetes research, personalized medicine, and cell therapies., Competing Interests: LJ, MB, EZ, ML, VK, RS, DP, IG, SK, KN, BL, AG No competing interests declared, (© 2023, Jarc, Bandral et al.)- Published
- 2024
- Full Text
- View/download PDF
14. Activation of the cGAS/STING Axis in Genome-Damaged Hematopoietic Cells Does Not Impact Blood Cell Formation or Leukemogenesis.
- Author
-
Dressel N, Natusch L, Munz CM, Costas Ramon S, Morcos MNF, Loff A, Hiller B, Haase C, Schulze L, Müller P, Lesche M, Dahl A, Luksch H, Rösen-Wolff A, Roers A, Behrendt R, and Gerbaulet A
- Subjects
- Animals, Mice, Hematopoiesis genetics, Nucleotidyltransferases genetics, Nucleotidyltransferases metabolism, Signal Transduction, Interferon Type I metabolism, Leukemia genetics
- Abstract
Genome damage is a main driver of malignant transformation, but it also induces aberrant inflammation via the cGAS/STING DNA-sensing pathway. Activation of cGAS/STING can trigger cell death and senescence, thereby potentially eliminating genome-damaged cells and preventing against malignant transformation. Here, we report that defective ribonucleotide excision repair (RER) in the hematopoietic system caused genome instability with concomitant activation of the cGAS/STING axis and compromised hematopoietic stem cell function, ultimately resulting in leukemogenesis. Additional inactivation of cGAS, STING, or type I IFN signaling, however, had no detectable effect on blood cell generation and leukemia development in RER-deficient hematopoietic cells. In wild-type mice, hematopoiesis under steady-state conditions and in response to genome damage was not affected by loss of cGAS. Together, these data challenge a role of the cGAS/STING pathway in protecting the hematopoietic system against DNA damage and leukemic transformation., Significance: Loss of cGAS/STING signaling does not impact DNA damage-driven leukemogenesis or alter steady-state, perturbed or malignant hematopoiesis, indicating that the cGAS/STING axis is not a crucial antioncogenic mechanism in the hematopoietic system. See related commentary by Zierhut, p. 2807., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
15. Transcriptome analysis reveals an Atoh1b-dependent gene set downstream of Dlx3b/4b during early inner ear development in zebrafish.
- Author
-
Ezhkova D, Schwarzer S, Spieß S, Geffarth M, Machate A, Zöller D, Stucke J, Alexopoulou D, Lesche M, Dahl A, and Hans S
- Subjects
- Animals, Humans, Gene Expression Profiling, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Transcription Factors genetics, Transcription Factors metabolism, Zebrafish Proteins genetics, Ear, Inner metabolism, Zebrafish genetics, Zebrafish metabolism
- Abstract
The vertebrate inner ear is the sensory organ mediating hearing and balance. The entire organ develops from the otic placode, which itself originates from the otic-epibranchial progenitor domain (OEPD). Multiple studies in various species have shown the importance of the forkhead-box and distal-less homeodomain transcription factor families for OEPD and subsequent otic placode formation. However, the transcriptional networks downstream of these factors are only beginning to be understood. Using transcriptome analysis, we here reveal numerous genes regulated by the distal-less homeodomain transcription factors Dlx3b and Dlx4b (Dlx3b/4b). We identify known and novel transcripts displaying widespread OEPD expression in a Dlx3b/4b-dependent manner. Some genes, with a known OEPD expression in other vertebrate species, might be members of a presumptive vertebrate core module required for proper otic development. Moreover, we identify genes controlling early-born sensory hair cell formation as well as regulating biomineral tissue development, both consistent with defective sensory hair cell and otolith formation observed in dlx3b/4b mutants. Finally, we show that ectopic Atoh1b expression can rescue early sensorigenesis even in the absence of Dlx3b/4b. Taken together, our data will help to unravel the gene regulatory network underlying early inner ear development and provide insights into the molecular control of vertebrate inner ear formation to restore hearing loss in humans ultimately., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
16. Cohesin SMC1β promotes closed chromatin and controls TERRA expression at spermatocyte telomeres.
- Author
-
Biswas U, Deb Mallik T, Pschirer J, Lesche M, Sameith K, and Jessberger R
- Subjects
- Male, Spermatocytes, Telomere genetics, Animals, Cohesins, Chromatin genetics, RNA, Long Noncoding genetics, Cell Cycle Proteins
- Abstract
Previous data showed that meiotic cohesin SMC1β protects spermatocyte telomeres from damage. The underlying reason, however, remained unknown as the expressions of telomerase and shelterin components were normal in Smc1 β
-/- spermatocytes. Here. we report that SMC1β restricts expression of the long noncoding RNA TERRA (telomeric repeat containing RNA) in spermatocytes. In somatic cell lines increased TERRA was reported to cause telomere damage through altering telomere chromatin structure. In Smc1 β-/- spermatocytes, we observed strongly increased levels of TERRA which accumulate on damaged chromosomal ends, where enhanced R-loop formation was found. This suggested a more open chromatin configuration near telomeres in Smc1 β-/- spermatocytes, which was confirmed by ATAC-seq. Telomere-distal regions were not affected by the absence of SMC1β but RNA-seq revealed increased transcriptional activity in telomere-proximal regions. Thus, SMC1β promotes closed chromatin specifically near telomeres and limits TERRA expression in spermatocytes., (© 2023 Biswas et al.)- Published
- 2023
- Full Text
- View/download PDF
17. Targeting cardiomyocyte ADAM10 ectodomain shedding promotes survival early after myocardial infarction.
- Author
-
Klapproth E, Witt A, Klose P, Wiedemann J, Vavilthota N, Künzel SR, Kämmerer S, Günscht M, Sprott D, Lesche M, Rost F, Dahl A, Rauch E, Kattner L, Weber S, Mirtschink P, Kopaliani I, Guan K, Lorenz K, Saftig P, Wagner M, and El-Armouche A
- Subjects
- Animals, Mice, Amyloid Precursor Protein Secretases genetics, Leukocytes, Membrane Proteins genetics, Humans, ADAM10 Protein genetics, Myocardial Infarction genetics
- Abstract
After myocardial infarction the innate immune response is pivotal in clearing of tissue debris as well as scar formation, but exaggerated cytokine and chemokine secretion with subsequent leukocyte infiltration also leads to further tissue damage. Here, we address the value of targeting a previously unknown a disintegrin and metalloprotease 10 (ADAM10)/CX3CL1 axis in the regulation of neutrophil recruitment early after MI. We show that myocardial ADAM10 is distinctly upregulated in myocardial biopsies from patients with ischemia-driven cardiomyopathy. Intriguingly, upon MI in mice, pharmacological ADAM10 inhibition as well as genetic cardiomycyte-specific ADAM10 deletion improves survival with markedly enhanced heart function and reduced scar size. Mechanistically, abolished ADAM10-mediated CX3CL1 ectodomain shedding leads to diminished IL-1β-dependent inflammation, reduced neutrophil bone marrow egress as well as myocardial tissue infiltration. Thus, our data shows a conceptual insight into how acute MI induces chemotactic signaling via ectodomain shedding in cardiomyocytes., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. A gene expression map of host immune response in human brucellosis.
- Author
-
Mitroulis I, Chrysanthopoulou A, Divolis G, Ioannidis C, Ntinopoulou M, Tasis A, Konstantinidis T, Antoniadou C, Soteriou N, Lallas G, Mitka S, Lesche M, Dahl A, Gembardt S, Panopoulou M, Sideras P, Wielockx B, Coskun Ü, Ritis K, and Skendros P
- Subjects
- Gene Expression, Humans, Immunity, Innate, Leukocytes, Mononuclear metabolism, Brucella abortus, Brucellosis
- Abstract
Brucellosis is a common zoonotic disease caused by intracellular pathogens of the genus Brucella . Brucella infects macrophages and evades clearance mechanisms, thus resulting in chronic parasitism. Herein, we studied the molecular changes that take place in human brucellosis both in vitro and ex vivo. RNA sequencing was performed in primary human macrophages (Mφ) and polymorphonuclear neutrophils (PMNs) infected with a clinical strain of Brucella spp. We observed a downregulation in the expression of genes involved in host response, such as TNF signaling, IL-1β production, and phagosome formation in Mφ, and phosphatidylinositol signaling and TNF signaling in PMNs, being in line with the ability of the pathogen to survive within phagocytes. Further transcriptomic analysis of isolated peripheral blood mononuclear cells (PBMCs) and PMNs from patients with acute brucellosis before treatment initiation and after successful treatment revealed a positive correlation of the molecular signature of active disease with pathways associated with response to interferons (IFN). We identified 24 common genes that were significantly altered in both PMNs and PBMCs, including genes involved in IFN signaling that were downregulated after treatment in both cell populations, and IL1R1 that was upregulated. The concentration of several inflammatory mediators was measured in the serum of these patients, and levels of IFN-γ, IL-1β and IL-6 were found significantly increased before the treatment of acute brucellosis. An independent cohort of patients with chronic brucellosis also revealed increased levels of IFN-γ during relapse compared to remissions. Taken together, this study provides for the first time an in-depth analysis of the transcriptomic alterations that take place in human phagocytes upon infection, and in peripheral blood immune populations during active disease., Competing Interests: Authors NS and GL were employed by the company P. Zafiropoulos S.A. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Mitroulis, Chrysanthopoulou, Divolis, Ioannidis, Ntinopoulou, Tasis, Konstantinidis, Antoniadou, Soteriou, Lallas, Mitka, Lesche, Dahl, Gembardt, Panopoulou, Sideras, Wielockx, Coskun, Ritis and Skendros.)
- Published
- 2022
- Full Text
- View/download PDF
19. Distinguishing activated T regulatory cell and T conventional cells by single-cell technologies.
- Author
-
Reinhardt J, Sharma V, Stavridou A, Lindner A, Reinhardt S, Petzold A, Lesche M, Rost F, Bonifacio E, and Eugster A
- Subjects
- Biomarkers metabolism, Flow Cytometry, Forkhead Transcription Factors metabolism, Lymphocyte Count, Lymphocyte Activation, T-Lymphocytes, Regulatory
- Abstract
Resting conventional T cells (Tconv) can be distinguished from T regulatory cells (Treg) by the canonical markers FOXP3, CD25 and CD127. However, the expression of these proteins alters after T-cell activation leading to overlap between Tconv and Treg. The objective of this study was to distinguish resting and antigen-responsive T effector (Tconv) and Treg using single-cell technologies. CD4
+ Treg and Tconv cells were stimulated with antigen and responsive and non-responsive populations processed for targeted and non-targeted single-cell RNAseq. Machine learning was used to generate a limited set of genes that could distinguish responding and non-responding Treg and Tconv cells and which was used for single-cell multiplex qPCR and to design a flow cytometry panel. Targeted scRNAseq clearly distinguished the four-cell populations. A minimal set of 27 genes was identified by machine learning algorithms to provide discrimination of the four populations at >95% accuracy. In all, 15 of the genes were validated to be differentially expressed by single-cell multiplex qPCR. Discrimination of responding Treg from responding Tconv could be achieved by a flow cytometry strategy that included staining for CD25, CD127, FOXP3, IKZF2, ITGA4, and the novel marker TRIM which was strongly expressed in Tconv and weakly expressed in both responding and non-responding Treg. A minimal set of genes was identified that discriminates responding and non-responding CD4+ Treg and Tconv cells and, which have identified TRIM as a marker to distinguish Treg by flow cytometry., (© 2022 The Authors. Immunology published by John Wiley & Sons Ltd.)- Published
- 2022
- Full Text
- View/download PDF
20. The RNA binding protein human antigen R is a gatekeeper of liver homeostasis.
- Author
-
Subramanian P, Gargani S, Palladini A, Chatzimike M, Grzybek M, Peitzsch M, Papanastasiou AD, Pyrina I, Ntafis V, Gercken B, Lesche M, Petzold A, Sinha A, Nati M, Thangapandi VR, Kourtzelis I, Andreadou M, Witt A, Dahl A, Burkhardt R, Haase R, Domingues AMJ, Henry I, Zamboni N, Mirtschink P, Chung KJ, Hampe J, Coskun Ü, Kontoyiannis DL, and Chavakis T
- Subjects
- Animals, Homeostasis, Inflammation metabolism, Liver pathology, Liver Cirrhosis metabolism, Mice, Mice, Inbred C57BL, RNA, Triglycerides metabolism, Carcinoma, Hepatocellular pathology, ELAV-Like Protein 1 metabolism, Liver Neoplasms pathology, Non-alcoholic Fatty Liver Disease pathology
- Abstract
Background and Aims: NAFLD is initiated by steatosis and can progress through fibrosis and cirrhosis to HCC. The RNA binding protein human antigen R (HuR) controls RNAs at the posttranscriptional level; hepatocyte HuR has been implicated in the regulation of diet-induced hepatic steatosis. The present study aimed to understand the role of hepatocyte HuR in NAFLD development and progression to fibrosis and HCC., Approach and Results: Hepatocyte-specific, HuR-deficient mice and control HuR-sufficient mice were fed either a normal diet or an NAFLD-inducing diet. Hepatic lipid accumulation, inflammation, fibrosis, and HCC development were studied by histology, flow cytometry, quantitative PCR, and RNA sequencing. The liver lipidome was characterized by lipidomics analysis, and the HuR-RNA interactions in the liver were mapped by RNA immunoprecipitation sequencing. Hepatocyte-specific, HuR-deficient mice displayed spontaneous hepatic steatosis and fibrosis predisposition compared to control HuR-sufficient mice. On an NAFLD-inducing diet, hepatocyte-specific HuR deficiency resulted in exacerbated inflammation, fibrosis, and HCC-like tumor development. A multi-omic approach, including lipidomics, transcriptomics, and RNA immunoprecipitation sequencing revealed that HuR orchestrates a protective network of hepatic-metabolic and lipid homeostasis-maintaining pathways. Consistently, HuR-deficient livers accumulated, already at steady state, a triglyceride signature resembling that of NAFLD livers. Moreover, up-regulation of secreted phosphoprotein 1 expression mediated, at least partially, fibrosis development in hepatocyte-specific HuR deficiency on an NAFLD-inducing diet, as shown by experiments using antibody blockade of osteopontin., Conclusions: HuR is a gatekeeper of liver homeostasis, preventing NAFLD-related fibrosis and HCC, suggesting that the HuR-dependent network could be exploited therapeutically., (© 2021 The Authors. Hepatology published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.)
- Published
- 2022
- Full Text
- View/download PDF
21. Hoxb1 Regulates Distinct Signaling Pathways in Neuromesodermal and Hindbrain Progenitors to Promote Cell Survival and Specification.
- Author
-
Pazur K, Giannios I, Lesche M, Rodriguez-Aznar E, and Gavalas A
- Subjects
- Animals, Cell Differentiation genetics, Cell Survival, Fibroblast Growth Factors metabolism, Gene Expression Regulation, Developmental, Homeodomain Proteins genetics, Mice, Signal Transduction, Transcription Factors metabolism, Homeodomain Proteins metabolism, Rhombencephalon metabolism
- Abstract
Hox genes play key roles in the anterior-posterior (AP) specification of all 3 germ layers during different developmental stages. It is only partially understood how they function in widely different developmental contexts, particularly with regards to extracellular signaling, and to what extent their function can be harnessed to guide cell specification in vitro. Here, we addressed the role of Hoxb1 in 2 distinct developmental contexts; in mouse embryonic stem cells (mES)-derived neuromesodermal progenitors (NMPs) and hindbrain neural progenitors. We found that Hoxb1 promotes NMP survival through the upregulation of Fgf8, Fgf17, and other components of Fgf signaling as well as the repression of components of the apoptotic pathway. Additionally, it upregulates other anterior Hox genes suggesting that it plays an active role in the early steps of AP specification. In neural progenitors, Hoxb1 synergizes with shh to repress anterior and dorsal neural markers, promote the expression of ventral neural markers and direct the specification of facial branchiomotorneuron (FBM)-like progenitors. Hoxb1 and shh synergize in regulating the expression of diverse signals and signaling molecules, including the Ret tyrosine kinase receptor. Finally, Hoxb1 synergizes with exogenous Glial cell line-derived neurotrophic factor (GDNF) to strengthen Ret expression and further promote the generation of FBM-like progenitors. Facial branchiomotorneuron-like progenitors survived for at least 6 months and differentiated into postmitotic neurons after orthotopic transplantation near the facial nucleus of adult mice. These results suggested that the patterning activity of Hox genes in combination with downstream signaling molecules can be harnessed for the generation of defined neural populations and transplantations with implications for neurodegenerative diseases., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
22. Multi-omics profiling of living human pancreatic islet donors reveals heterogeneous beta cell trajectories towards type 2 diabetes.
- Author
-
Wigger L, Barovic M, Brunner AD, Marzetta F, Schöniger E, Mehl F, Kipke N, Friedland D, Burdet F, Kessler C, Lesche M, Thorens B, Bonifacio E, Legido-Quigley C, Barbier Saint Hilaire P, Delerive P, Dahl A, Klose C, Gerl MJ, Simons K, Aust D, Weitz J, Distler M, Schulte AM, Mann M, Ibberson M, and Solimena M
- Subjects
- Biomarkers, Blood Glucose, Disease Susceptibility, Energy Metabolism, Gene Expression Profiling, Gene Expression Regulation, Humans, Insulin metabolism, Living Donors, Metabolomics, Proteomics, Diabetes Mellitus, Type 2 etiology, Diabetes Mellitus, Type 2 metabolism, Insulin-Secreting Cells metabolism, Islets of Langerhans metabolism
- Abstract
Most research on human pancreatic islets is conducted on samples obtained from normoglycaemic or diseased brain-dead donors and thus cannot accurately describe the molecular changes of pancreatic islet beta cells as they progress towards a state of deficient insulin secretion in type 2 diabetes (T2D). Here, we conduct a comprehensive multi-omics analysis of pancreatic islets obtained from metabolically profiled pancreatectomized living human donors stratified along the glycemic continuum, from normoglycemia to T2D. We find that islet pools isolated from surgical samples by laser-capture microdissection display remarkably more heterogeneous transcriptomic and proteomic profiles in patients with diabetes than in non-diabetic controls. The differential regulation of islet gene expression is already observed in prediabetic individuals with impaired glucose tolerance. Our findings demonstrate a progressive, but disharmonic, remodelling of mature beta cells, challenging current hypotheses of linear trajectories toward precursor or transdifferentiation stages in T2D. Furthermore, through integration of islet transcriptomics with preoperative blood plasma lipidomics, we define the relative importance of gene coexpression modules and lipids that are positively or negatively associated with HbA1c levels, pointing to potential prognostic markers., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
23. Environmental enrichment preserves a young DNA methylation landscape in the aged mouse hippocampus.
- Author
-
Zocher S, Overall RW, Lesche M, Dahl A, and Kempermann G
- Subjects
- Age Factors, Animals, Dentate Gyrus metabolism, Epigenomics methods, Female, Hippocampus cytology, Humans, Mice, Inbred C57BL, Neurogenesis genetics, Neuronal Plasticity genetics, Neurons metabolism, Mice, Aging, CpG Islands genetics, DNA Methylation, Environment, Hippocampus metabolism
- Abstract
The decline of brain function during aging is associated with epigenetic changes, including DNA methylation. Lifestyle interventions can improve brain function during aging, but their influence on age-related epigenetic changes is unknown. Using genome-wide DNA methylation sequencing, we here show that experiencing a stimulus-rich environment counteracts age-related DNA methylation changes in the hippocampal dentate gyrus of mice. Specifically, environmental enrichment prevented the aging-induced CpG hypomethylation at target sites of the methyl-CpG-binding protein Mecp2, which is critical to neuronal function. The genes at which environmental enrichment counteracted aging effects have described roles in neuronal plasticity, neuronal cell communication and adult hippocampal neurogenesis and are dysregulated with age-related cognitive decline in the human brain. Our results highlight the stimulating effects of environmental enrichment on hippocampal plasticity at the level of DNA methylation and give molecular insights into the specific aspects of brain aging that can be counteracted by lifestyle interventions.
- Published
- 2021
- Full Text
- View/download PDF
24. SETDB1 is required for intestinal epithelial differentiation and the prevention of intestinal inflammation.
- Author
-
Južnić L, Peuker K, Strigli A, Brosch M, Herrmann A, Häsler R, Koch M, Matthiesen L, Zeissig Y, Löscher BS, Nuber A, Schotta G, Neumeister V, Chavakis T, Kurth T, Lesche M, Dahl A, von Mässenhausen A, Linkermann A, Schreiber S, Aden K, Rosenstiel PC, Franke A, Hampe J, and Zeissig S
- Subjects
- Animals, Cell Differentiation, Epithelial Cells metabolism, Female, Gene Silencing, Homeostasis genetics, Humans, Loss of Function Mutation, Male, Mice, Histone-Lysine N-Methyltransferase genetics, Inflammatory Bowel Diseases genetics, Intestinal Mucosa metabolism
- Abstract
Objective: The intestinal epithelium is a rapidly renewing tissue which plays central roles in nutrient uptake, barrier function and the prevention of intestinal inflammation. Control of epithelial differentiation is essential to these processes and is dependent on cell type-specific activity of transcription factors which bind to accessible chromatin. Here, we studied the role of SET Domain Bifurcated Histone Lysine Methyltransferase 1, also known as ESET (SETDB1), a histone H3K9 methyltransferase, in intestinal epithelial homeostasis and IBD., Design: We investigated mice with constitutive and inducible intestinal epithelial deletion of Setdb1 , studied the expression of SETDB1 in patients with IBD and mouse models of IBD, and investigated the abundance of SETDB1 variants in healthy individuals and patients with IBD., Results: Deletion of intestinal epithelial Setdb1 in mice was associated with defects in intestinal epithelial differentiation, barrier disruption, inflammation and mortality. Mechanistic studies showed that loss of SETDB1 leads to de-silencing of endogenous retroviruses, DNA damage and intestinal epithelial cell death. Predicted loss-of-function variants in human SETDB1 were considerably less frequently observed than expected, consistent with a critical role of SETDB1 in human biology. While the vast majority of patients with IBD showed unimpaired mucosal SETDB1 expression, comparison of IBD and non-IBD exomes revealed over-representation of individual rare missense variants in SETDB1 in IBD, some of which are predicted to be associated with loss of function and may contribute to the pathogenesis of intestinal inflammation., Conclusion: SETDB1 plays an essential role in intestinal epithelial homeostasis. Future work is required to investigate whether rare variants in SETDB1 contribute to the pathogenesis of IBD., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
25. A switch in pdgfrb + cell-derived ECM composition prevents inhibitory scarring and promotes axon regeneration in the zebrafish spinal cord.
- Author
-
Tsata V, Möllmert S, Schweitzer C, Kolb J, Möckel C, Böhm B, Rosso G, Lange C, Lesche M, Hammer J, Kesavan G, Beis D, Guck J, Brand M, and Wehner D
- Subjects
- Animals, Cicatrix physiopathology, Models, Biological, Recovery of Function, Signal Transduction, Spinal Cord physiopathology, Spinal Cord Injuries pathology, Spinal Cord Injuries physiopathology, Zebrafish Proteins metabolism, Axons metabolism, Cicatrix pathology, Extracellular Matrix metabolism, Nerve Regeneration, Receptor, Platelet-Derived Growth Factor beta metabolism, Spinal Cord pathology, Zebrafish physiology
- Abstract
In mammals, perivascular cell-derived scarring after spinal cord injury impedes axonal regrowth. In contrast, the extracellular matrix (ECM) in the spinal lesion site of zebrafish is permissive and required for axon regeneration. However, the cellular mechanisms underlying this interspecies difference have not been investigated. Here, we show that an injury to the zebrafish spinal cord triggers recruitment of pdgfrb
+ myoseptal and perivascular cells in a PDGFR signaling-dependent manner. Interference with pdgfrb+ cell recruitment or depletion of pdgfrb+ cells inhibits axonal regrowth and recovery of locomotor function. Transcriptional profiling and functional experiments reveal that pdgfrb+ cells upregulate expression of axon growth-promoting ECM genes (cthrc1a and col12a1a/b) and concomitantly reduce synthesis of matrix molecules that are detrimental to regeneration (lum and mfap2). Our data demonstrate that a switch in ECM composition is critical for axon regeneration after spinal cord injury and identify the cellular source and components of the growth-promoting lesion ECM., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
26. Innate Immune Training of Granulopoiesis Promotes Anti-tumor Activity.
- Author
-
Kalafati L, Kourtzelis I, Schulte-Schrepping J, Li X, Hatzioannou A, Grinenko T, Hagag E, Sinha A, Has C, Dietz S, de Jesus Domingues AM, Nati M, Sormendi S, Neuwirth A, Chatzigeorgiou A, Ziogas A, Lesche M, Dahl A, Henry I, Subramanian P, Wielockx B, Murray P, Mirtschink P, Chung KJ, Schultze JL, Netea MG, Hajishengallis G, Verginis P, Mitroulis I, and Chavakis T
- Subjects
- Adaptive Immunity, Adoptive Transfer, Animals, Epigenesis, Genetic, Interferon Type I metabolism, Mice, Inbred C57BL, Monocytes metabolism, Neoplasms pathology, Neutrophils metabolism, Phenotype, Receptor, Interferon alpha-beta deficiency, Receptor, Interferon alpha-beta metabolism, Transcription, Genetic, Transcriptome genetics, beta-Glucans metabolism, Granulocytes immunology, Immunity, Innate, Neoplasms immunology
- Abstract
Trained innate immunity, induced via modulation of mature myeloid cells or their bone marrow progenitors, mediates sustained increased responsiveness to secondary challenges. Here, we investigated whether anti-tumor immunity can be enhanced through induction of trained immunity. Pre-treatment of mice with β-glucan, a fungal-derived prototypical agonist of trained immunity, resulted in diminished tumor growth. The anti-tumor effect of β-glucan-induced trained immunity was associated with transcriptomic and epigenetic rewiring of granulopoiesis and neutrophil reprogramming toward an anti-tumor phenotype; this process required type I interferon signaling irrespective of adaptive immunity in the host. Adoptive transfer of neutrophils from β-glucan-trained mice to naive recipients suppressed tumor growth in the latter in a ROS-dependent manner. Moreover, the anti-tumor effect of β-glucan-induced trained granulopoiesis was transmissible by bone marrow transplantation to recipient naive mice. Our findings identify a novel and therapeutically relevant anti-tumor facet of trained immunity involving appropriate rewiring of granulopoiesis., Competing Interests: Declaration of interests M.G.N. is scientific founder of Trained Therapeutics and Discovery (TTxD)., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
27. A smart polymer for sequence-selective binding, pulldown, and release of DNA targets.
- Author
-
Krieg E, Gupta K, Dahl A, Lesche M, Boye S, Lederer A, and Shih WM
- Subjects
- Base Sequence genetics, DNA genetics, DNA metabolism, DNA, Complementary genetics, DNA, Single-Stranded genetics, Electrophoresis, Polyacrylamide Gel, Fractionation, Field Flow, Glucagon genetics, High-Throughput Nucleotide Sequencing methods, Humans, Insulin genetics, Methanol, Pancreas metabolism, Prealbumin genetics, Transcriptome, DNA isolation & purification, Stimuli Responsive Polymers
- Abstract
Selective isolation of DNA is crucial for applications in biology, bionanotechnology, clinical diagnostics and forensics. We herein report a smart methanol-responsive polymer (MeRPy) that can be programmed to bind and separate single- as well as double-stranded DNA targets. Captured targets are quickly isolated and released back into solution by denaturation (sequence-agnostic) or toehold-mediated strand displacement (sequence-selective). The latter mode allows 99.8% efficient removal of unwanted sequences and 79% recovery of highly pure target sequences. We applied MeRPy for the depletion of insulin, glucagon, and transthyretin cDNA from clinical next-generation sequencing (NGS) libraries. This step improved the data quality for low-abundance transcripts in expression profiles of pancreatic tissues. Its low cost, scalability, high stability and ease of use make MeRPy suitable for diverse applications in research and clinical laboratories, including enhancement of NGS libraries, extraction of DNA from biological samples, preparative-scale DNA isolations, and sorting of DNA-labeled non-nucleic acid targets.
- Published
- 2020
- Full Text
- View/download PDF
28. MicroRNA profiling of mouse cortical progenitors and neurons reveals miR-486-5p as a regulator of neurogenesis.
- Author
-
Dori M, Cavalli D, Lesche M, Massalini S, Alieh LHA, de Toledo BC, Khudayberdiev S, Schratt G, Dahl A, and Calegari F
- Subjects
- Animals, Blotting, Northern, Computational Biology methods, Electroporation, Female, Gene Expression Profiling methods, High-Throughput Nucleotide Sequencing methods, Immunohistochemistry, In Situ Hybridization, Mice, Mice, Inbred C57BL, MicroRNAs genetics, Neurogenesis genetics, Neurogenesis physiology, MicroRNAs metabolism, Neural Stem Cells cytology, Neural Stem Cells metabolism, Neurons cytology, Neurons metabolism
- Abstract
MicroRNAs (miRNAs) are short (∼22 nt) single-stranded non-coding RNAs that regulate gene expression at the post-transcriptional level. Over recent years, many studies have extensively characterized the involvement of miRNA-mediated regulation in neurogenesis and brain development. However, a comprehensive catalog of cortical miRNAs expressed in a cell-specific manner in progenitor types of the developing mammalian cortex is still missing. Overcoming this limitation, here we exploited a double reporter mouse line previously validated by our group to allow the identification of the transcriptional signature of neurogenic commitment and provide the field with the complete atlas of miRNA expression in proliferating neural stem cells, neurogenic progenitors and newborn neurons during corticogenesis. By extending the currently known list of miRNAs expressed in the mouse brain by over twofold, our study highlights the power of cell type-specific analyses for the detection of transcripts that would otherwise be diluted out when studying bulk tissues. We further exploited our data by predicting putative miRNAs and validated the power of our approach by providing evidence for the involvement of miR-486 in brain development., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2020. Published by The Company of Biologists Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
29. Single cell sequencing of radial glia progeny reveals the diversity of newborn neurons in the adult zebrafish brain.
- Author
-
Lange C, Rost F, Machate A, Reinhardt S, Lesche M, Weber A, Kuscha V, Dahl A, Rulands S, and Brand M
- Subjects
- Animals, Animals, Genetically Modified, Animals, Newborn anatomy & histology, Diencephalon cytology, Gene Expression Profiling, Mice, Sequence Analysis, RNA, Single-Cell Analysis, Telencephalon cytology, Zebrafish growth & development, Brain cytology, Cell Lineage, Ependymoglial Cells cytology, Neurogenesis, Neurons cytology, Zebrafish anatomy & histology
- Abstract
Zebrafish display widespread and pronounced adult neurogenesis, which is fundamental for their regeneration capability after central nervous system injury. However, the cellular identity and the biological properties of adult newborn neurons are elusive for most brain areas. Here, we have used short-term lineage tracing of radial glia progeny to prospectively isolate newborn neurons from the her4.1
+ radial glia lineage in the homeostatic adult forebrain. Transcriptome analysis of radial glia, newborn neurons and mature neurons using single cell sequencing identified distinct transcriptional profiles, including novel markers for each population. Specifically, we detected two separate newborn neuron types, which showed diversity of cell fate commitment and location. Further analyses showed that these cell types are homologous to neurogenic cells in the mammalian brain, identified neurogenic commitment in proliferating radial glia and indicated that glutamatergic projection neurons are generated in the adult zebrafish telencephalon. Thus, we prospectively isolated adult newborn neurons from the adult zebrafish forebrain, identified markers for newborn and mature neurons in the adult brain, and revealed intrinsic heterogeneity among adult newborn neurons and their homology with mammalian adult neurogenic cell types., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2020. Published by The Company of Biologists Ltd.)- Published
- 2020
- Full Text
- View/download PDF
30. Aldh1b1 expression defines progenitor cells in the adult pancreas and is required for Kras-induced pancreatic cancer.
- Author
-
Mameishvili E, Serafimidis I, Iwaszkiewicz S, Lesche M, Reinhardt S, Bölicke N, Büttner M, Stellas D, Papadimitropoulou A, Szabolcs M, Anastassiadis K, Dahl A, Theis F, Efstratiadis A, and Gavalas A
- Subjects
- Aldehyde Dehydrogenase 1 Family genetics, Aldehyde Dehydrogenase, Mitochondrial genetics, Animals, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Cell Differentiation, Cell Transformation, Neoplastic metabolism, Disease Models, Animal, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Mice, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Signal Transduction, Single-Cell Analysis, Stem Cells metabolism, Aldehyde Dehydrogenase 1 Family metabolism, Aldehyde Dehydrogenase, Mitochondrial metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Transformation, Neoplastic pathology, Mutation, Pancreatic Neoplasms pathology, Proto-Oncogene Proteins p21(ras) genetics, Stem Cells pathology
- Abstract
The presence of progenitor or stem cells in the adult pancreas and their potential involvement in homeostasis and cancer development remain unresolved issues. Here, we show that mouse centroacinar cells can be identified and isolated by virtue of the mitochondrial enzyme Aldh1b1 that they uniquely express. These cells are necessary and sufficient for the formation of self-renewing adult pancreatic organoids in an Aldh1b1-dependent manner. Aldh1b1-expressing centroacinar cells are largely quiescent, self-renew, and, as shown by genetic lineage tracing, contribute to all 3 pancreatic lineages in the adult organ under homeostatic conditions. Single-cell RNA sequencing analysis of these cells identified a progenitor cell population, established its molecular signature, and determined distinct differentiation pathways to early progenitors. A distinct feature of these progenitor cells is the preferential expression of small GTPases, including Kras, suggesting that they might be susceptible to Kras-driven oncogenic transformation. This finding and the overexpression of Aldh1b1 in human and mouse pancreatic cancers, driven by activated Kras, prompted us to examine the involvement of Aldh1b1 in oncogenesis. We demonstrated genetically that ablation of Aldh1b1 completely abrogates tumor development in a mouse model of Kras
G12D -induced pancreatic cancer., Competing Interests: The authors declare no conflict of interest.- Published
- 2019
- Full Text
- View/download PDF
31. Renin cells with defective Gsα/cAMP signaling contribute to renal endothelial damage.
- Author
-
Steglich A, Kessel F, Hickmann L, Gerlach M, Lachmann P, Gembardt F, Lesche M, Dahl A, Federlein A, Schweda F, Hugo CPM, and Todorov VT
- Subjects
- Animals, Biomarkers metabolism, Juxtaglomerular Apparatus, Mice, Mice, Transgenic, Phenotype, Cyclic AMP metabolism, Endothelial Cells metabolism, GTP-Binding Protein alpha Subunits, Gs metabolism, Kidney metabolism, Renin metabolism, Signal Transduction physiology
- Abstract
Synthesis of renin in renal renin-producing cells (RPCs) is controlled via the intracellular messenger cAMP. Interference with cAMP-mediated signaling by inducible knockout of Gs-alpha (Gsα) in RPCs of adult mice resulted in a complex adverse kidney phenotype. Therein, glomerular endothelial damage was most striking. In this study, we investigated whether Gsα knockout leads to a loss of RPCs, which itself may contribute to the endothelial injury. We compared the kidney phenotype of three RPC-specific conditional mouse lines during continuous induction of recombination. Mice expressing red fluorescent reporter protein tdTomato (tdT) in RPCs served as controls. tdT was also expressed in RPCs of the other two strains used, namely with RPC-specific Gsα knockout (Gsα mice) or with RPC-specific diphtheria toxin A expression (DTA mice, in which the RPCs should be diminished). Using immunohistological analysis, we found that RPCs decreased by 82% in the kidneys of Gsα mice as compared with controls. However, the number of tdT-positive cells was similar in the two strains, demonstrating that after Gsα knockout, the RPCs persist as renin-negative descendants. In contrast, both renin-positive and tdT-labeled cells decreased by 80% in DTA mice suggesting effective RPC ablation. Only Gsα mice displayed dysregulated endothelial cell marker expression indicating glomerular endothelial damage. In addition, a robust induction of genes involved in tissue remodelling with microvascular damage was identified in tdT-labeled RPCs isolated from Gsα mice. We concluded that Gsα/renin double-negative RPC progeny essentially contributes for the development of glomerular endothelial damage in our Gsα-deficient mice.
- Published
- 2019
- Full Text
- View/download PDF
32. Controlling distinct signaling states in cultured cancer cells provides a new platform for drug discovery.
- Author
-
Poser SW, Otto O, Arps-Forker C, Ge Y, Herbig M, Andree C, Gruetzmann K, Adasme MF, Stodolak S, Nikolakopoulou P, Park DM, Mcintyre A, Lesche M, Dahl A, Lennig P, Bornstein SR, Schroeck E, Klink B, Leker RR, Bickle M, Chrousos GP, Schroeder M, Cannistraci CV, Guck J, and Androutsellis-Theotokis A
- Subjects
- Cell Line, Tumor, Drug Discovery, Gene Expression Profiling, Gene Expression Regulation genetics, Gene Expression Regulation physiology, Glioblastoma genetics, Humans, RNA Interference, Repressor Proteins genetics, Signal Transduction genetics, Signal Transduction physiology, Glioblastoma metabolism, Repressor Proteins metabolism
- Abstract
Cancer cells can switch between signaling pathways to regulate growth under different conditions. In the tumor microenvironment, this likely helps them evade therapies that target specific pathways. We must identify all possible states and utilize them in drug screening programs. One such state is characterized by expression of the transcription factor Hairy and Enhancer of Split 3 ( HES3 ) and sensitivity to HES3 knockdown, and it can be modeled in vitro . Here, we cultured 3 primary human brain cancer cell lines under 3 different culture conditions that maintain low, medium, and high HES3 expression and characterized gene regulation and mechanical phenotype in these states. We assessed gene expression regulation following HES3 knockdown in the HES3 -high conditions. We then employed a commonly used human brain tumor cell line to screen Food and Drug Administration (FDA)-approved compounds that specifically target the HES3 -high state. We report that cells from multiple patients behave similarly when placed under distinct culture conditions. We identified 37 FDA-approved compounds that specifically kill cancer cells in the high- HES3 -expression conditions. Our work reveals a novel signaling state in cancer, biomarkers, a strategy to identify treatments against it, and a set of putative drugs for potential repurposing.-Poser, S. W., Otto, O., Arps-Forker, C., Ge, Y., Herbig, M., Andree, C., Gruetzmann, K., Adasme, M. F., Stodolak, S., Nikolakopoulou, P., Park, D. M., Mcintyre, A., Lesche, M., Dahl, A., Lennig, P., Bornstein, S. R., Schroeck, E., Klink, B., Leker, R. R., Bickle, M., Chrousos, G. P., Schroeder, M., Cannistraci, C. V., Guck, J., Androutsellis-Theotokis, A. Controlling distinct signaling states in cultured cancer cells provides a new platform for drug discovery.
- Published
- 2019
- Full Text
- View/download PDF
33. Protein Methyltransferase Inhibition Decreases Endocrine Specification Through the Upregulation of Aldh1b1 Expression.
- Author
-
Giannios I, Serafimidis I, Anastasiou V, Pezzolla D, Lesche M, Andree C, Bickle M, and Gavalas A
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Benzoates pharmacology, Gene Expression Regulation, Developmental drug effects, Humans, Insulin-Secreting Cells metabolism, Mice, Mouse Embryonic Stem Cells drug effects, Mouse Embryonic Stem Cells enzymology, Nerve Tissue Proteins genetics, Pancreas drug effects, Pancreas growth & development, Protein Methyltransferases antagonists & inhibitors, Xanthenes pharmacology, Aldehyde Dehydrogenase 1 Family genetics, Aldehyde Dehydrogenase, Mitochondrial genetics, Cell Differentiation genetics, Diabetes Mellitus therapy, Pluripotent Stem Cells transplantation, Protein Methyltransferases genetics
- Abstract
Understanding the mechanisms that promote the specification of pancreas progenitors and regulate their self-renewal and differentiation will help to maintain and expand pancreas progenitor cells derived from human pluripotent stem (hPS) cells. This will improve the efficiency of current differentiation protocols of hPS cells into β-cells and bring such cells closer to clinical applications for the therapy of diabetes. Aldehyde dehydrogenase 1b1 (Aldh1b1) is a mitochondrial enzyme expressed specifically in progenitor cells during mouse pancreas development, and we have shown that its functional inactivation leads to accelerated differentiation and deficient β-cells. In this report, we aimed to identify small molecule inducers of Aldh1b1 expression taking advantage of a mouse embryonic stem (mES) cell Aldh1b1 lacZ reporter line and a pancreas differentiation protocol directing mES cells into pancreatic progenitors. We identified AMI-5, a protein methyltransferase inhibitor, as an Aldh1b1 inducer and showed that it can maintain Aldh1b1 expression in embryonic pancreas explants. This led to a selective reduction in endocrine specification. This effect was due to a downregulation of Ngn3, and it was mediated through Aldh1b1 since the effect was abolished in Aldh1b1 null pancreata. The findings implicated methyltransferase activity in the regulation of endocrine differentiation and showed that methyltransferases can act through specific regulators during pancreas differentiation. Stem Cells 2019;37:640-651., (© 2019 The Authors. Stem Cells published by Wiley Periodicals, Inc. on behalf of AlphaMed Press 2019.)
- Published
- 2019
- Full Text
- View/download PDF
34. Sequence and expression levels of circular RNAs in progenitor cell types during mouse corticogenesis.
- Author
-
Dori M, Haj Abdullah Alieh L, Cavalli D, Massalini S, Lesche M, Dahl A, and Calegari F
- Subjects
- Alternative Splicing genetics, Animals, Binding Sites, Cerebellar Cortex cytology, Exons genetics, Female, Mice, Mice, Transgenic, MicroRNAs genetics, MicroRNAs metabolism, Pregnancy, Protein Biosynthesis genetics, RNA, Circular metabolism, RNA, Messenger genetics, Base Sequence genetics, Neurogenesis genetics, RNA, Circular genetics, Stem Cells physiology, Transcriptome genetics
- Abstract
Circular (circ) RNAs have recently emerged as a novel class of transcripts whose identification and function remain elusive. Among many tissues and species, the mammalian brain is the organ in which circRNAs are more abundant and first evidence of their functional significance started to emerge. Yet, even within this well-studied organ, annotation of circRNAs remains fragmentary, their sequence is unknown, and their expression in specific cell types was never investigated. Overcoming these limitations, here we provide the first comprehensive identification of circRNAs and assessment of their expression patterns in proliferating neural stem cells, neurogenic progenitors, and newborn neurons of the developing mouse cortex. Extending the current knowledge about the diversity of this class of transcripts by the identification of nearly 4,000 new circRNAs, our study is the first to provide the full sequence information and expression patterns of circRNAs in cell types representing the lineage of neurogenic commitment. We further exploited our data by evaluating the coding potential, evolutionary conservation, and biogenesis of circRNAs that we found to arise from a specific subclass of linear mRNAs. Our study provides the arising field of circRNA biology with a powerful new resource to address the complexity and potential biological significance of this new class of transcripts., (© 2019 Dori et al.)
- Published
- 2019
- Full Text
- View/download PDF
35. Hematopoietic Stem Cells but Not Multipotent Progenitors Drive Erythropoiesis during Chronic Erythroid Stress in EPO Transgenic Mice.
- Author
-
Singh RP, Grinenko T, Ramasz B, Franke K, Lesche M, Dahl A, Gassmann M, Chavakis T, Henry I, and Wielockx B
- Subjects
- Animals, Computational Biology methods, Erythroid Precursor Cells cytology, Erythroid Precursor Cells metabolism, Gene Expression Profiling, Mice, Mice, Transgenic, Erythropoiesis, Erythropoietin genetics, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Stress, Physiological
- Abstract
The hematopoietic stem cell (HSC) compartment consists of a small pool of cells capable of replenishing all blood cells. Although it is established that the hematopoietic system is assembled as a hierarchical organization under steady-state conditions, emerging evidence suggests that distinct differentiation pathways may exist in response to acute stress. However, it remains unclear how different hematopoietic stem and progenitor cell subpopulations behave under sustained chronic stress. Here, by using adult transgenic mice overexpressing erythropoietin (EPO; Tg6) and a combination of in vivo, in vitro, and deep-sequencing approaches, we found that HSCs respond differentially to chronic erythroid stress compared with their closely related multipotent progenitors (MPPs). Specifically, HSCs exhibit a vastly committed erythroid progenitor profile with enhanced cell division, while MPPs display erythroid and myeloid cell signatures and an accumulation of uncommitted cells. Thus, our results identify HSCs as master regulators of chronic stress erythropoiesis, potentially circumventing the hierarchical differentiation-detour., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
36. SETD1A protects HSCs from activation-induced functional decline in vivo.
- Author
-
Arndt K, Kranz A, Fohgrub J, Jolly A, Bledau AS, Di Virgilio M, Lesche M, Dahl A, Höfer T, Stewart AF, and Waskow C
- Subjects
- Animals, Histone-Lysine N-Methyltransferase genetics, Mice, Mice, Knockout, Myeloid-Lymphoid Leukemia Protein genetics, Myeloid-Lymphoid Leukemia Protein metabolism, Cell Proliferation, DNA Damage, DNA Repair, Hematopoietic Stem Cells enzymology, Histone-Lysine N-Methyltransferase metabolism
- Abstract
The regenerative capacity of hematopoietic stem cells (HSCs) is limited by the accumulation of DNA damage. Conditional mutagenesis of the histone 3 lysine 4 (H3K4) methyltransferase, Setd1a , revealed that it is required for the expression of DNA damage recognition and repair pathways in HSCs. Specific deletion of Setd1a in adult long-term (LT) HSCs is compatible with adult life and has little effect on the maintenance of phenotypic LT-HSCs in the bone marrow. However, SETD1A-deficient LT-HSCs lose their transcriptional cellular identity, accompanied by loss of their proliferative capacity and stem cell function under replicative stress in situ and after transplantation. In response to inflammatory stimulation, SETD1A protects HSCs and progenitors from activation-induced attrition in vivo. The comprehensive regulation of DNA damage responses by SETD1A in HSCs is clearly distinct from the key roles played by other epigenetic regulators, including the major leukemogenic H3K4 methyltransferase MLL1, or MLL5, indicating that HSC identity and function is supported by cooperative specificities within an epigenetic framework., (© 2018 by The American Society of Hematology.)
- Published
- 2018
- Full Text
- View/download PDF
37. Systems biology of the IMIDIA biobank from organ donors and pancreatectomised patients defines a novel transcriptomic signature of islets from individuals with type 2 diabetes.
- Author
-
Solimena M, Schulte AM, Marselli L, Ehehalt F, Richter D, Kleeberg M, Mziaut H, Knoch KP, Parnis J, Bugliani M, Siddiq A, Jörns A, Burdet F, Liechti R, Suleiman M, Margerie D, Syed F, Distler M, Grützmann R, Petretto E, Moreno-Moral A, Wegbrod C, Sönmez A, Pfriem K, Friedrich A, Meinel J, Wollheim CB, Baretton GB, Scharfmann R, Nogoceke E, Bonifacio E, Sturm D, Meyer-Puttlitz B, Boggi U, Saeger HD, Filipponi F, Lesche M, Meda P, Dahl A, Wigger L, Xenarios I, Falchi M, Thorens B, Weitz J, Bokvist K, Lenzen S, Rutter GA, Froguel P, von Bülow M, Ibberson M, and Marchetti P
- Subjects
- Aged, Aged, 80 and over, Computational Biology, Female, Humans, Male, Pancreatectomy, Biological Specimen Banks, Diabetes Mellitus, Type 2 metabolism, Systems Biology methods, Tissue Donors, Transcriptome genetics
- Abstract
Aims/hypothesis: Pancreatic islet beta cell failure causes type 2 diabetes in humans. To identify transcriptomic changes in type 2 diabetic islets, the Innovative Medicines Initiative for Diabetes: Improving beta-cell function and identification of diagnostic biomarkers for treatment monitoring in Diabetes (IMIDIA) consortium ( www.imidia.org ) established a comprehensive, unique multicentre biobank of human islets and pancreas tissues from organ donors and metabolically phenotyped pancreatectomised patients (PPP)., Methods: Affymetrix microarrays were used to assess the islet transcriptome of islets isolated either by enzymatic digestion from 103 organ donors (OD), including 84 non-diabetic and 19 type 2 diabetic individuals, or by laser capture microdissection (LCM) from surgical specimens of 103 PPP, including 32 non-diabetic, 36 with type 2 diabetes, 15 with impaired glucose tolerance (IGT) and 20 with recent-onset diabetes (<1 year), conceivably secondary to the pancreatic disorder leading to surgery (type 3c diabetes). Bioinformatics tools were used to (1) compare the islet transcriptome of type 2 diabetic vs non-diabetic OD and PPP as well as vs IGT and type 3c diabetes within the PPP group; and (2) identify transcription factors driving gene co-expression modules correlated with insulin secretion ex vivo and glucose tolerance in vivo. Selected genes of interest were validated for their expression and function in beta cells., Results: Comparative transcriptomic analysis identified 19 genes differentially expressed (false discovery rate ≤0.05, fold change ≥1.5) in type 2 diabetic vs non-diabetic islets from OD and PPP. Nine out of these 19 dysregulated genes were not previously reported to be dysregulated in type 2 diabetic islets. Signature genes included TMEM37, which inhibited Ca
2+ -influx and insulin secretion in beta cells, and ARG2 and PPP1R1A, which promoted insulin secretion. Systems biology approaches identified HNF1A, PDX1 and REST as drivers of gene co-expression modules correlated with impaired insulin secretion or glucose tolerance, and 14 out of 19 differentially expressed type 2 diabetic islet signature genes were enriched in these modules. None of these signature genes was significantly dysregulated in islets of PPP with impaired glucose tolerance or type 3c diabetes., Conclusions/interpretation: These studies enabled the stringent definition of a novel transcriptomic signature of type 2 diabetic islets, regardless of islet source and isolation procedure. Lack of this signature in islets from PPP with IGT or type 3c diabetes indicates differences possibly due to peculiarities of these hyperglycaemic conditions and/or a role for duration and severity of hyperglycaemia. Alternatively, these transcriptomic changes capture, but may not precede, beta cell failure.- Published
- 2018
- Full Text
- View/download PDF
38. Modulation of Myelopoiesis Progenitors Is an Integral Component of Trained Immunity.
- Author
-
Mitroulis I, Ruppova K, Wang B, Chen LS, Grzybek M, Grinenko T, Eugster A, Troullinaki M, Palladini A, Kourtzelis I, Chatzigeorgiou A, Schlitzer A, Beyer M, Joosten LAB, Isermann B, Lesche M, Petzold A, Simons K, Henry I, Dahl A, Schultze JL, Wielockx B, Zamboni N, Mirtschink P, Coskun Ü, Hajishengallis G, Netea MG, and Chavakis T
- Subjects
- Animals, Cells, Cultured, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Interleukin-1beta metabolism, Male, Mice, Mice, Inbred C57BL, Myeloid Progenitor Cells drug effects, Myelopoiesis immunology, beta-Glucans pharmacology, Immunity, Innate, Immunologic Memory, Myeloid Progenitor Cells immunology
- Abstract
Trained innate immunity fosters a sustained favorable response of myeloid cells to a secondary challenge, despite their short lifespan in circulation. We thus hypothesized that trained immunity acts via modulation of hematopoietic stem and progenitor cells (HSPCs). Administration of β-glucan (prototypical trained-immunity-inducing agonist) to mice induced expansion of progenitors of the myeloid lineage, which was associated with elevated signaling by innate immune mediators, such as IL-1β and granulocyte-macrophage colony-stimulating factor (GM-CSF), and with adaptations in glucose metabolism and cholesterol biosynthesis. The trained-immunity-related increase in myelopoiesis resulted in a beneficial response to secondary LPS challenge and protection from chemotherapy-induced myelosuppression in mice. Therefore, modulation of myeloid progenitors in the bone marrow is an integral component of trained immunity, which to date, was considered to involve functional changes of mature myeloid cells in the periphery., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
39. Although Abundant in Tumor Tissue, Mast Cells Have No Effect on Immunological Micro-milieu or Growth of HPV-Induced or Transplanted Tumors.
- Author
-
Ghouse SM, Polikarpova A, Muhandes L, Dudeck J, Tantcheva-Poór I, Hartmann K, Lesche M, Dahl A, Eming S, Müller W, Behrendt R, and Roers A
- Subjects
- Animals, Cell Line, Epithelial Cells immunology, Epithelial Cells pathology, Epithelial Cells virology, Mice, Mice, Transgenic, Neoplasm Transplantation, Cell Proliferation, Cell Transformation, Viral immunology, Mast Cells immunology, Mast Cells pathology, Neoplasms, Experimental blood supply, Neoplasms, Experimental immunology, Neoplasms, Experimental pathology, Neoplasms, Experimental virology, Neovascularization, Pathologic immunology, Neovascularization, Pathologic pathology, Neovascularization, Pathologic virology, Papillomaviridae immunology
- Abstract
High numbers of mast cells populate the stroma of many types of neoplasms, including human papilloma virus-induced benign and malignant tumors in man and mouse. Equipped with numerous pattern recognition receptors and capable of executing important pro-inflammatory responses, mast cells are considered innate sentinels that significantly impact tumor biology. Mast cells were reported to promote human papilloma virus (HPV)-induced epithelial hyperproliferation and neo-angiogenesis in an HPV-driven mouse model of skin cancer. We analyzed HPV-induced epithelial hyperplasia and squamous cell carcinoma formation, as well as growth of tumors inoculated into the dermis, in mice lacking skin mast cells. Unexpectedly, the absence of mast cells had no effect on HPV-induced epithelial growth or angiogenesis, on growth kinetics of inoculated tumors, or on the immunological tumor micro-milieu. Thus, the conspicuous recruitment of mast cells into tumor tissues cannot necessarily be equated with important mast cell functions in tumor growth., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
40. Correction: TDRD6 mediates early steps of spliceosome maturation in primary spermatocytes.
- Author
-
Akpınar M, Lesche M, Fanourgakis G, Fu J, Anastassiadis K, Dahl A, and Jessberger R
- Abstract
[This corrects the article DOI: 10.1371/journal.pgen.1006660.].
- Published
- 2017
- Full Text
- View/download PDF
41. Clonal competition in BcrAbl-driven leukemia: how transplantations can accelerate clonal conversion.
- Author
-
Cornils K, Thielecke L, Winkelmann D, Aranyossy T, Lesche M, Dahl A, Roeder I, Fehse B, and Glauche I
- Subjects
- Animals, Base Sequence, Carcinogenesis pathology, Clone Cells, Computer Simulation, Gene Expression Regulation, Leukemic, Genetic Vectors metabolism, Interleukin-3 metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Mice, Inbred BALB C, Models, Biological, RNA, Messenger genetics, RNA, Messenger metabolism, Transcriptome genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Neoplasm Transplantation
- Abstract
Background: Clonal competition in cancer describes the process in which the progeny of a cell clone supersedes or succumbs to other competing clones due to differences in their functional characteristics, mostly based on subsequently acquired mutations. Even though the patterns of those mutations are well explored in many tumors, the dynamical process of clonal selection is underexposed., Methods: We studied the dynamics of clonal competition in a BcrAbl-induced leukemia using a γ-retroviral vector library encoding the oncogene in conjunction with genetic barcodes. To this end, we studied the growth dynamics of transduced cells on the clonal level both in vitro and in vivo in transplanted mice., Results: While we detected moderate changes in clonal abundancies in vitro, we observed monoclonal leukemias in 6/30 mice after transplantation, which intriguingly were caused by only two different BcrAbl clones. To analyze the success of these clones, we applied a mathematical model of hematopoietic tissue maintenance, which indicated that a differential engraftment capacity of these two dominant clones provides a possible explanation of our observations. These findings were further supported by additional transplantation experiments and increased BcrAbl transcript levels in both clones., Conclusion: Our findings show that clonal competition is not an absolute process based on mutations, but highly dependent on selection mechanisms in a given environmental context.
- Published
- 2017
- Full Text
- View/download PDF
42. TDRD6 mediates early steps of spliceosome maturation in primary spermatocytes.
- Author
-
Akpınar M, Lesche M, Fanourgakis G, Fu J, Anastassiadis K, Dahl A, and Jessberger R
- Subjects
- Animals, Arginine chemistry, Chromatids chemistry, Coiled Bodies metabolism, DNA Methylation, Deoxyadenosines chemistry, Exons, Fatty Acids, Unsaturated chemistry, Introns, Male, Methylation, Mice, Mice, Transgenic, Microscopy, Fluorescence, Protein Domains, RNA Splicing, RNA, Messenger metabolism, Spermatocytes cytology, Thionucleosides chemistry, Transcriptome, Protein-Arginine N-Methyltransferases metabolism, Ribonucleoprotein, U5 Small Nuclear metabolism, Ribonucleoproteins genetics, Ribonucleoproteins physiology, Spermatocytes metabolism, Spliceosomes metabolism
- Abstract
Tudor containing protein 6 (TDRD6) is a male germ line-specific protein essential for chromatoid body (ChB) structure, elongated spermatid development and male fertility. Here we show that in meiotic prophase I spermatocytes TDRD6 interacts with the key protein arginine methyl transferase PRMT5, which supports splicing. TDRD6 also associates with spliceosomal core protein SmB in the absence of RNA and in an arginine methylation dependent manner. In Tdrd6-/- diplotene spermatocytes PRMT5 association with SmB and arginine dimethylation of SmB are much reduced. TDRD6 deficiency impairs the assembly of spliceosomes, which feature 3.5-fold increased levels of U5 snRNPs. In the nucleus, these deficiencies in spliceosome maturation correlate with decreased numbers of SMN-positive bodies and Cajal bodies involved in nuclear snRNP maturation. Transcriptome analysis of TDRD6-deficient diplotene spermatocytes revealed high numbers of splicing defects such as aberrant usage of intron and exons as well as aberrant representation of splice junctions. Together, this study demonstrates a novel function of TDRD6 in spliceosome maturation and mRNA splicing in prophase I spermatocytes.
- Published
- 2017
- Full Text
- View/download PDF
43. Pancreas lineage allocation and specification are regulated by sphingosine-1-phosphate signalling.
- Author
-
Serafimidis I, Rodriguez-Aznar E, Lesche M, Yoshioka K, Takuwa Y, Dahl A, Pan D, and Gavalas A
- Subjects
- Acinar Cells cytology, Adaptor Proteins, Signal Transducing metabolism, Animals, Body Patterning, Cell Cycle Proteins, Cell Differentiation, Cell Survival, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Mice, Models, Biological, Phosphoproteins metabolism, Phosphotransferases (Alcohol Group Acceptor) metabolism, Protein Subunits metabolism, Receptors, Lysosphingolipid metabolism, Receptors, Notch metabolism, Sphingosine metabolism, Stem Cells cytology, YAP-Signaling Proteins, Cell Lineage, Lysophospholipids metabolism, Pancreas cytology, Signal Transduction, Sphingosine analogs & derivatives
- Abstract
During development, progenitor expansion, lineage allocation, and implementation of differentiation programs need to be tightly coordinated so that different cell types are generated in the correct numbers for appropriate tissue size and function. Pancreatic dysfunction results in some of the most debilitating and fatal diseases, including pancreatic cancer and diabetes. Several transcription factors regulating pancreas lineage specification have been identified, and Notch signalling has been implicated in lineage allocation, but it remains unclear how these processes are coordinated. Using a combination of genetic approaches, organotypic cultures of embryonic pancreata, and genomics, we found that sphingosine-1-phosphate (S1p), signalling through the G protein coupled receptor (GPCR) S1pr2, plays a key role in pancreas development linking lineage allocation and specification. S1pr2 signalling promotes progenitor survival as well as acinar and endocrine specification. S1pr2-mediated stabilisation of the yes-associated protein (YAP) is essential for endocrine specification, thus linking a regulator of progenitor growth with specification. YAP stabilisation and endocrine cell specification rely on Gαi subunits, revealing an unexpected specificity of selected GPCR intracellular signalling components. Finally, we found that S1pr2 signalling posttranscriptionally attenuates Notch signalling levels, thus regulating lineage allocation. Both S1pr2-mediated YAP stabilisation and Notch attenuation are necessary for the specification of the endocrine lineage. These findings identify S1p signalling as a novel key pathway coordinating cell survival, lineage allocation, and specification and linking these processes by regulating YAP levels and Notch signalling. Understanding lineage allocation and specification in the pancreas will shed light in the origins of pancreatic diseases and may suggest novel therapeutic approaches.
- Published
- 2017
- Full Text
- View/download PDF
44. Loss of Trex1 in Dendritic Cells Is Sufficient To Trigger Systemic Autoimmunity.
- Author
-
Peschke K, Achleitner M, Frenzel K, Gerbaulet A, Ada SR, Zeller N, Lienenklaus S, Lesche M, Poulet C, Naumann R, Dahl A, Ravens U, Günther C, Müller W, Knobeloch KP, Prinz M, Roers A, and Behrendt R
- Subjects
- Animals, Antigens, CD19 physiology, B-Lymphocytes physiology, Brain immunology, Exodeoxyribonucleases deficiency, Interferon Type I biosynthesis, Mice, Mice, Inbred C57BL, Mice, Knockout, Phosphoproteins deficiency, Autoimmunity, Dendritic Cells physiology, Exodeoxyribonucleases physiology, Phosphoproteins physiology
- Abstract
Defects of the intracellular enzyme 3' repair exonuclease 1 (Trex1) cause the rare autoimmune condition Aicardi-Goutières syndrome and are associated with systemic lupus erythematosus. Trex1(-/-) mice develop type I IFN-driven autoimmunity, resulting from activation of the cytoplasmic DNA sensor cyclic GMP-AMP synthase by a nucleic acid substrate of Trex1 that remains unknown. To identify cell types responsible for initiation of autoimmunity, we generated conditional Trex1 knockout mice. Loss of Trex1 in dendritic cells was sufficient to cause IFN release and autoimmunity, whereas Trex1-deficient keratinocytes and microglia produced IFN but did not induce inflammation. In contrast, B cells, cardiomyocytes, neurons, and astrocytes did not show any detectable response to the inactivation of Trex1. Thus, individual cell types differentially respond to the loss of Trex1, and Trex1 expression in dendritic cells is essential to prevent breakdown of self-tolerance ensuing from aberrant detection of endogenous DNA., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
45. Chromatoid Body Protein TDRD6 Supports Long 3' UTR Triggered Nonsense Mediated mRNA Decay.
- Author
-
Fanourgakis G, Lesche M, Akpinar M, Dahl A, and Jessberger R
- Subjects
- 3' Untranslated Regions genetics, Animals, Germ Cells growth & development, Germ Cells metabolism, Glutathione genetics, Male, Mice, Mice, Knockout, Protein Domains genetics, RNA, Messenger genetics, RNA-Binding Proteins genetics, Ribonucleoproteins metabolism, Spermatids growth & development, Spermatids metabolism, Carrier Proteins genetics, Glutathione analogs & derivatives, RNA Stability genetics, Ribonucleoproteins genetics, Spermatogenesis genetics
- Abstract
Chromatoid bodies (CBs) are spermiogenesis-specific organelles of largely unknown function. CBs harbor various RNA species, RNA-associated proteins and proteins of the tudor domain family like TDRD6, which is required for a proper CB architecture. Proteome analysis of purified CBs revealed components of the nonsense-mediated mRNA decay (NMD) machinery including UPF1. TDRD6 is essential for UPF1 localization to CBs, for UPF1-UPF2 and UPF1-MVH interactions. Upon removal of TDRD6, the association of several mRNAs with UPF1 and UPF2 is disturbed, and the long 3' UTR-stimulated but not the downstream exon-exon junction triggered pathway of NMD is impaired. Reduced association of the long 3' UTR mRNAs with UPF1 and UPF2 correlates with increased stability and enhanced translational activity. Thus, we identified TDRD6 within CBs as required for mRNA degradation, specifically the extended 3' UTR-triggered NMD pathway, and provide evidence for the requirement of NMD in spermiogenesis. This function depends on TDRD6-promoted assembly of mRNA and decay enzymes in CBs.
- Published
- 2016
- Full Text
- View/download PDF
46. CCND1-CDK4-mediated cell cycle progression provides a competitive advantage for human hematopoietic stem cells in vivo.
- Author
-
Mende N, Kuchen EE, Lesche M, Grinenko T, Kokkaliaris KD, Hanenberg H, Lindemann D, Dahl A, Platz A, Höfer T, Calegari F, and Waskow C
- Subjects
- Animals, Blotting, Western, Cell Proliferation, DNA Primers genetics, Flow Cytometry, Gene Transfer Techniques, High-Throughput Nucleotide Sequencing, Human Umbilical Vein Endothelial Cells, Humans, Mice, Mice, Inbred C57BL, Polymerase Chain Reaction, Cell Cycle physiology, Cyclin D1 metabolism, Cyclin-Dependent Kinase 4 metabolism, Hematopoietic Stem Cells physiology, Models, Biological
- Abstract
Maintenance of stem cell properties is associated with reduced proliferation. However, in mouse hematopoietic stem cells (HSCs), loss of quiescence results in a wide range of phenotypes, ranging from functional failure to extensive self-renewal. It remains unknown whether the function of human HSCs is controlled by the kinetics of cell cycle progression. Using human HSCs and human progenitor cells (HSPCs), we report here that elevated levels of CCND1-CDK4 complexes promoted the transit from G0 to G1 and shortened the G1 cell cycle phase, resulting in protection from differentiation-inducing signals in vitro and increasing human leukocyte engraftment in vivo. Further, CCND1-CDK4 overexpression conferred a competitive advantage without impacting HSPC numbers. In contrast, accelerated cell cycle progression mediated by elevated levels of CCNE1-CDK2 led to the loss of functional HSPCs in vivo. Collectively, these data suggest that the transition kinetics through the early cell cycle phases are key regulators of human HSPC function and important for lifelong hematopoiesis., (© 2015 Mende et al.)
- Published
- 2015
- Full Text
- View/download PDF
47. Identification and expression patterns of novel long non-coding RNAs in neural progenitors of the developing mammalian cortex.
- Author
-
Aprea J, Lesche M, Massalini S, Prenninger S, Alexopoulou D, Dahl A, Hiller M, and Calegari F
- Abstract
Long non-coding (lnc)RNAs play key roles in many biological processes. Elucidating the function of lncRNAs in cell type specification during organ development requires knowledge about their expression in individual progenitor types rather than in whole tissues. To achieve this during cortical development, we used a dual-reporter mouse line to isolate coexisting proliferating neural stem cells, differentiating neurogenic progenitors and newborn neurons and assessed the expression of lncRNAs by paired-end, high-throughput sequencing. We identified 379 genomic loci encoding novel lncRNAs and performed a comprehensive assessment of cell-specific expression patterns for all, annotated and novel, lncRNAs described to date. Our study provides a powerful new resource for studying these elusive transcripts during stem cell commitment and neurogenesis.
- Published
- 2015
- Full Text
- View/download PDF
48. Transcriptome sequencing during mouse brain development identifies long non-coding RNAs functionally involved in neurogenic commitment.
- Author
-
Aprea J, Prenninger S, Dori M, Ghosh T, Monasor LS, Wessendorf E, Zocher S, Massalini S, Alexopoulou D, Lesche M, Dahl A, Groszer M, Hiller M, and Calegari F
- Subjects
- Alternative Splicing, Animals, Brain cytology, Cerebral Cortex cytology, Cerebral Cortex embryology, Gene Expression Regulation, Developmental, Green Fluorescent Proteins genetics, Mice, Mice, Transgenic, Neurogenesis, Neurons, Phenotype, Proteins genetics, Proto-Oncogene Proteins genetics, Wnt Proteins genetics, Brain embryology, Gene Expression Profiling methods, Neural Stem Cells physiology, RNA, Long Noncoding genetics
- Abstract
Transcriptome analysis of somatic stem cells and their progeny is fundamental to identify new factors controlling proliferation versus differentiation during tissue formation. Here, we generated a combinatorial, fluorescent reporter mouse line to isolate proliferating neural stem cells, differentiating progenitors and newborn neurons that coexist as intermingled cell populations during brain development. Transcriptome sequencing revealed numerous novel long non-coding (lnc)RNAs and uncharacterized protein-coding transcripts identifying the signature of neurogenic commitment. Importantly, most lncRNAs overlapped neurogenic genes and shared with them a nearly identical expression pattern suggesting that lncRNAs control corticogenesis by tuning the expression of nearby cell fate determinants. We assessed the power of our approach by manipulating lncRNAs and protein-coding transcripts with no function in corticogenesis reported to date. This led to several evident phenotypes in neurogenic commitment and neuronal survival, indicating that our study provides a remarkably high number of uncharacterized transcripts with hitherto unsuspected roles in brain development. Finally, we focussed on one lncRNA, Miat, whose manipulation was found to trigger pleiotropic effects on brain development and aberrant splicing of Wnt7b. Hence, our study suggests that lncRNA-mediated alternative splicing of cell fate determinants controls stem-cell commitment during neurogenesis.
- Published
- 2013
- Full Text
- View/download PDF
49. [Clinical use and technology of metalloceramics].
- Author
-
Weiskopf J, Lesche M, and Gehre G
- Subjects
- Crowns, Dental Caries prevention & control, Denture Design, Denture Retention, Humans, Patient Care Planning, Periodontal Diseases prevention & control, Technology, Dental, Dental Alloys, Dental Porcelain, Denture, Partial, Fixed
- Published
- 1984
50. [Follow-up studies and clinical evaluation of model cast dentures with periodontal and periodonto-gingival support].
- Author
-
Ebersbach W and Lesche M
- Subjects
- Dental Abutments, Denture Design, Denture Rebasing, Denture Repair, Humans, Tooth Extraction, Dental Caries etiology, Denture Retention, Dentures adverse effects
- Abstract
The authors examined 746 cast denture constructions with an average wearing time of 6 years to study the influence of cast denture constructions on caries increment. The mode of wear of the cast denture constructions had no influence on caries incidence, whereas effects exerted by the duration of wear and the presence of soft deposits could be detected. The evaluation of the functional performance of cast denture constructions showed that the clinical serviceability amounts to more than 8 years.
- Published
- 1977
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.