Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Adv Sci (Weinh) ; 8(14): 2100626, 2021 07.
Article in English | MEDLINE | ID: mdl-34306986

ABSTRACT

Somatic cell reprogramming and tissue repair share relevant factors and molecular programs. Here, Dickkopf-3 (DKK3) is identified as novel factor for organ regeneration using combined transcription-factor-induced reprogramming and RNA-interference techniques. Loss of Dkk3 enhances the generation of induced pluripotent stem cells but does not affect de novo derivation of embryonic stem cells, three-germ-layer differentiation or colony formation capacity of liver and pancreatic organoids. However, DKK3 expression levels in wildtype animals and serum levels in human patients are elevated upon injury. Accordingly, Dkk3-null mice display less liver damage upon acute and chronic failure mediated by increased proliferation in hepatocytes and LGR5+ liver progenitor cell population, respectively. Similarly, recovery from experimental pancreatitis is accelerated. Regeneration onset occurs in the acinar compartment accompanied by virtually abolished canonical-Wnt-signaling in Dkk3-null animals. This results in reduced expression of the Hedgehog repressor Gli3 and increased Hedgehog-signaling activity upon Dkk3 loss. Collectively, these data reveal Dkk3 as a key regulator of organ regeneration via a direct, previously unacknowledged link between DKK3, canonical-Wnt-, and Hedgehog-signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Genomics/methods , Organogenesis/genetics , Organogenesis/physiology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Regeneration/genetics , Regeneration/physiology
2.
Oncogene ; 39(7): 1445-1465, 2020 02.
Article in English | MEDLINE | ID: mdl-31659255

ABSTRACT

Aneuploidy can instigate tumorigenesis. However, mutations in genes that control chromosome segregation are rare in human tumors as these mutations reduce cell fitness. Screening experiments indicate that the knockdown of multiple classes of genes that are not directly involved in chromosome segregation can lead to aneuploidy induction. The possible contribution of these genes to cancer formation remains yet to be defined. Here we identified gene knockdowns that lead to an increase in aneuploidy in checkpoint-deficient human cancer cells. Computational analysis revealed that the identified genes overlap with recurrent mutations in human cancers. The knockdown of the three strongest selected candidate genes (ORP3, GJB3, and RXFP1) enhances the malignant transformation of human fibroblasts in culture. Furthermore, the knockout of Orp3 results in an aberrant expansion of lymphoid progenitor cells and a high penetrance formation of chromosomal instable, pauci-clonal B-cell lymphoma in aging mice. At pre-tumorous stages, lymphoid cells from the animals exhibit deregulated phospholipid metabolism and an aberrant induction of proliferation regulating pathways associating with increased aneuploidy in hematopoietic progenitor cells. Together, these results support the concept that aneuploidy-inducing gene deficiencies contribute to cellular transformation and carcinogenesis involving the deregulation of various molecular processes such as lipid metabolism, proliferation, and cell survival.


Subject(s)
Aneuploidy , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Gene Knockdown Techniques , Lymphoma, B-Cell/genetics , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Fibroblasts/pathology , Humans , Lymphoma, B-Cell/pathology , Mice
3.
Cancers (Basel) ; 11(10)2019 Sep 21.
Article in English | MEDLINE | ID: mdl-31546614

ABSTRACT

BACKGROUND: Most liver tumors arise on the basis of chronic liver diseases that trigger inflammatory responses. Besides inflammation, subsequent defects in the p53-signaling pathway frequently occurs in liver cancer. In this study, we analyzed the consequences of inflammation and p53 loss in liver carcinogenesis. METHODS: We used inducible liver-specific transgenic mouse strains to analyze the consequences of NF-κB/p65 activation mimicking chronic inflammation and subsequent p53 loss. RESULTS: Ikk2ca driven NF-κB/p65 activation in mice results in liver fibrosis, the formation of ectopic lymphoid structures and carcinogenesis independent of p53 expression. Subsequent deletion of Trp53 led to an increased tumor formation, metastasis and a shift in tumor differentiation towards intrahepatic cholangiocarcinoma. In addition, loss of Trp53 in an inflammatory liver resulted in elevated chromosomal instability and indicated a distinct aberration pattern. CONCLUSIONS: In conclusion, activation of NF-κB/p65 mimicking chronic inflammation provokes the formation of liver carcinoma. Collateral disruption of Trp53 supports tumor progression and influences tumor differentiation and heterogeneity.

4.
Int J Mol Sci ; 19(12)2018 Nov 29.
Article in English | MEDLINE | ID: mdl-30501048

ABSTRACT

Liver cholestasis is a chronic liver disease and a major health problem worldwide. Cholestasis is characterised by a decrease in bile flow due to impaired secretion by hepatocytes or by obstruction of bile flow through intra- or extrahepatic bile ducts. Thereby cholestasis can induce ductal proliferation, hepatocyte injury and liver fibrosis. Notch signalling promotes the formation and maturation of bile duct structures. Here we investigated the liver regeneration process in the context of cholestasis induced by disruption of the Notch signalling pathway. Liver-specific deletion of recombination signal binding protein for immunoglobulin kappa j region (Rbpj), which represents a key regulator of Notch signalling, induces severe cholestasis through impaired intra-hepatic bile duct (IHBD) maturation, severe necrosis and increased lethality. Deregulation of the biliary compartment and cholestasis are associated with the change of several signalling pathways including a Kyoto Encyclopedia of Genes and Genomes (KEGG) gene set representing the Hippo pathway, further yes-associated protein (YAP) activation and upregulation of SRY (sex determining region Y)-box 9 (SOX9), which is associated with transdifferentiation of hepatocytes. SOX9 upregulation in cholestatic liver injury in vitro is independent of Notch signalling. We could comprehensively address that in vivo Rbpj depletion is followed by YAP activation, which influences the transdifferentiation of hepatocytes and thereby contributing to liver regeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cholestasis/metabolism , Liver Regeneration/physiology , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bile Ducts/metabolism , Bile Ducts/physiology , Blotting, Western , Cell Cycle Proteins , Cell Transdifferentiation/genetics , Cell Transdifferentiation/physiology , Cells, Cultured , Cholestasis/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Liver Regeneration/genetics , Male , Mice , Phosphoproteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology , YAP-Signaling Proteins
5.
Acta Neuropathol Commun ; 4(1): 87, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27550225

ABSTRACT

Parkinson's disease is one of the most common neurodegenerative disorders of the elderly and ageing hence described to be a major risk factor. Telomere shortening as a result of the inability to fully replicate the ends of linear chromosomes is one of the hallmarks of ageing. The role of telomere dysfunction in neurological diseases and the ageing brain is not clarified and there is an ongoing discussion whether telomere shortening is linked to Parkinson's disease. Here we studied a mouse model of Parkinson's disease (Thy-1 [A30P] α-synuclein transgenic mouse model) in the background of telomere shortening (Terc knockout mouse model). α-synuclein transgenic mice with short telomeres (αSYN(tg/tg) G3Terc(-/-)) developed an accelerated disease with significantly decreased survival. This accelerated phenotype of mice with short telomeres was characterized by a declined motor performance and an increased formation of α-synuclein aggregates. Immunohistochemical analysis and mRNA expression studies revealed that the disease end-stage brain stem microglia showed an impaired response in αSYN(tg/tg) G3Terc(-/-) microglia animals. These results provide the first experimental data that telomere shortening accelerates α-synuclein pathology that is linked to limited microglia function in the brainstem.


Subject(s)
Brain Stem/metabolism , Microglia/metabolism , Parkinsonian Disorders/metabolism , Telomere Shortening/physiology , alpha-Synuclein/metabolism , Animals , Brain Stem/pathology , Disease Progression , Humans , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Motor Activity/physiology , Parkinsonian Disorders/pathology , Phenotype , Postural Balance/physiology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , RNA, Messenger/metabolism , Time Factors
6.
J Cell Sci ; 129(12): 2416-29, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27179075

ABSTRACT

Dynamic regulation of cell-cell adhesion by the coordinated formation and dissolution of E-cadherin-based adherens junctions is crucial for tissue homeostasis. The actin-binding protein cortactin interacts with E-cadherin and enables F-actin accumulation at adherens junctions. Here, we were interested to study the broader functional interactions of cortactin in adhesion complexes. In line with literature, we demonstrate that cortactin binds to E-cadherin, and that a posttranslational modification of cortactin, RhoA-induced phosphorylation by protein kinase D1 (PKD1; also known as PRKD1) at S298, impairs adherens junction assembly and supports their dissolution. Two new S298-phosphorylation-dependent interactions were also identified, namely, that phosphorylation of cortactin decreases its interaction with ß-catenin and the actin-binding protein vinculin. In addition, binding of vinculin to ß-catenin, as well as linkage of vinculin to F-actin, are also significantly compromised upon phosphorylation of cortactin. Accordingly, we found that regulation of cell-cell adhesion by phosphorylation of cortactin downstream of RhoA and PKD1 is vitally dependent on vinculin-mediated protein interactions. Thus, cortactin, unexpectedly, is an important integration node for the dynamic regulation of protein complexes during breakdown and formation of adherens junctions.


Subject(s)
Cadherins/metabolism , Cortactin/metabolism , TRPP Cation Channels/metabolism , Actin Cytoskeleton/metabolism , Adherens Junctions/metabolism , Animals , Antigens, CD , Caco-2 Cells , Cell Adhesion , Colon/metabolism , Epithelium/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phosphorylation , Phosphoserine/metabolism , Vinculin/metabolism
7.
Nat Commun ; 5: 4599, 2014 Aug 13.
Article in English | MEDLINE | ID: mdl-25118183

ABSTRACT

In addition to performing its canonical function, Telomerase Reverse Transcriptase (TERT) has been shown to participate in cellular processes independent of telomerase activity. Furthermore, although TERT mainly localizes to Cajal bodies, it is also present within the nucleolus. Because the nucleolus is the site of rDNA transcription, we investigated the possible role of telomerase in regulating RNA polymerase I (Pol I). Here we show that TERT binds to rDNA and stimulates transcription by Pol I during liver regeneration and Ras-induced hyperproliferation. Moreover, the inhibition of telomerase activity by TERT- or TERC-specific RNA interference, the overexpression of dominant-negative-TERT, and the application of the telomerase inhibitor imetelstat reduce Pol I transcription and the growth of tumour cells. In vitro, telomerase can stimulate the formation of the transcription initiation complex. Our results demonstrate how non-canonical features of telomerase may direct Pol I transcription in oncogenic and regenerative hyperproliferation.


Subject(s)
Cell Proliferation/physiology , DNA, Ribosomal/genetics , DNA, Ribosomal/physiology , RNA Polymerase I/physiology , Telomerase/physiology , Transcription, Genetic/physiology , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Humans , Kidney/cytology , Liver/cytology , Liver Regeneration/genetics , Liver Regeneration/physiology , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/cytology , Protein Binding/physiology , RNA Polymerase I/genetics , Rabbits , Telomerase/genetics , Transcription, Genetic/genetics
8.
Gut ; 63(9): 1501-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24092862

ABSTRACT

BACKGROUND AND AIMS: The cyclin-dependent kinase inhibitor p21 has been implicated as a tumour suppressor. Moreover, recent genetic studies suggest that p21 might be a potential therapeutic target to improve regeneration in chronic diseases. The aim of this study was to delineate the role of p21 in chronic liver injury and to specify its role in hepatocarcinogenesis in a mouse model of chronic cholestatic liver injury. METHODS: The degree of liver injury, regeneration and tumour formation was assessed in Mdr2(-/-) mice and compared with Mdr2/ p21(-/-) mice. Moreover, the role of p21 was evaluated in hepatoma cells in vitro and in human hepatocellular carcinoma (HCC). RESULTS: Mdr2(-/-) mice developed HCCs as a consequence of chronic inflammatory liver injury. In contrast, tumour development was profoundly delayed in Mdr2/ p21(-/-) mice. Delayed tumour development was accompanied by markedly impaired liver regeneration in Mdr2/ p21(-/-) mice. Moreover, the regenerative capacity of the Mdr2/ p21(-/-) livers in response to partial hepatectomy declined with age in these mice. Hepatocyte transplantation experiments revealed that impaired liver regeneration was due to intrinsic factors within the cells and changes in the Mdr2/ p21(-/-) microenvironment. In human HCCs, a subset of tumours expressed p21, which was associated with a significant shorter patient survival. CONCLUSIONS: We provide experimental evidence that p21 is required for sustained liver regeneration and tumour development in chronic liver injury indicating that p21 needs to be tightly regulated in order to balance liver regeneration and cancer risk. Moreover, we identify p21 as a negative prognostic marker in human HCC.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/etiology , Cholestasis, Intrahepatic/complications , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hepatic Insufficiency/physiopathology , Liver Neoplasms/etiology , Liver Regeneration/physiology , Animals , Biomarkers/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Cell Line , Chronic Disease , Disease Progression , Female , Hepatectomy , Hepatic Insufficiency/etiology , Hepatic Insufficiency/metabolism , Hepatic Insufficiency/surgery , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Prognosis
9.
Gastroenterology ; 142(5): 1229-1239.e3, 2012 May.
Article in English | MEDLINE | ID: mdl-22342966

ABSTRACT

BACKGROUND & AIMS: p53 limits the self-renewal of stem cells from various tissues. Loss of p53, in combination with other oncogenic events, results in aberrant self-renewal and transformation of progenitor cells. It is not known whether loss of p53 is sufficient to induce tumor formation in liver. METHODS: We used AlfpCre mice to create mice with liver-specific disruption of Trp53 (AlfpCre(+)Trp53(Δ2-10/Δ2-10) mice). We analyzed colony formation and genomic features and gene expression patterns in liver cells during hepatocarcinogenesis in mice with homozygous, heterozygous, and no disruption of Trp53. RESULTS: Liver-specific disruption of Trp53 consistently induced formation of liver carcinomas that had bilineal differentiation. In nontransformed liver cells and cultured primary liver cells, loss of p53 (but not p21) resulted in chromosomal imbalances and increased clonogenic capacity of liver progenitor cells (LPCs) and hepatocytes. Primary cultures of hepatocytes and LPCs from AlfpCre(+)Trp53(Δ2-10/Δ2-10) mice, but not Cdkn1a(-/-) mice, formed tumors with bilineal differentiation when transplanted into immunocompromised mice. Spontaneous liver tumors that developed in AlfpCre(+)Trp53(Δ2-10/Δ2-10) mice had significant but complex alterations in expression of Rb checkpoint genes compared with chemically induced liver tumors that developed mice with wild-type Trp53. CONCLUSIONS: Deletion of p53 from livers of mice is sufficient to induce tumor formation. The tumors have bilineal differentiation and dysregulation of Rb checkpoint genes.


Subject(s)
Liver Neoplasms, Experimental/etiology , Liver/pathology , Tumor Suppressor Protein p53/physiology , Aging , Animals , Cell Differentiation , Cell Transformation, Neoplastic , Chromosomal Instability , Cyclin-Dependent Kinase Inhibitor p21/physiology , Genes, Retinoblastoma , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL
10.
Gastroenterology ; 142(4): 907-17, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22245845

ABSTRACT

BACKGROUND & AIMS: Ectopic expression of certain transcription factors can reprogram somatic cells to a pluripotent state. Hematopoietic and muscle stem cells can be more efficiently reprogrammed than differentiated blood or muscle cells, yet similar findings have not been shown in other primary organ systems. Moreover, molecular characteristics of the cellular hierarchy of tissues that influence reprogramming capacities need to be delineated. We analyzed the effect of differentiation stage of freshly isolated, mouse liver cells on the reprogramming efficiency. METHODS: Liver progenitor cell (LPC)-enriched cell fractions were isolated from adult (6-8 wk) and fetal (embryonic day 14.5) livers of mice and reprogrammed to become induced pluripotent stem (iPS) cells. Different transcription factors were expressed in liver cells, and markers of pluripotency were examined, along with the ability of iPS cells to differentiate, in vitro and in vivo, into different germ layers. RESULTS: Fetal and adult LPCs had significantly greater reprogramming efficiency after transduction with 3 or 4 reprogramming factors. Transduction efficiency-corrected reprogramming rates of fetal LPCs were 275-fold higher, compared with unsorted fetal liver cells, when 3 reprogramming factors were transduced. The increased reprogramming efficiency of LPCs, compared with differentiated liver cells, occurred independently of proliferation rates, but was associated with endogenous expression of reprogramming factors (Klf4 and c-Myc) and BAF (Brg1/Brm associated factor)-complex members Baf155 and Brg1, which mediate epigenetic changes during reprogramming. Knockdown of BAF complex members negated the increased reprogramming efficiency of LPCs, compared with non-LPCs. CONCLUSIONS: LPCs have intrinsic, cell proliferation-independent characteristics resulting in an increased reprogramming capacity compared to differentiated liver cells.


Subject(s)
Cell Differentiation , DNA Helicases/metabolism , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Liver/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Biomarkers/metabolism , Cell Proliferation , Cells, Cultured , DNA Helicases/genetics , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Genotype , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Liver/embryology , Mice , Nuclear Proteins/genetics , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Time Factors , Transcription Factors/genetics , Transduction, Genetic , Transfection
12.
Hepatology ; 53(5): 1608-17, 2011 May.
Article in English | MEDLINE | ID: mdl-21520174

ABSTRACT

UNLABELLED: Telomere shortening impairs liver regeneration in mice and is associated with cirrhosis formation in humans with chronic liver disease. In humans, telomerase mutations have been associated with familial diseases leading to bone marrow failure or lung fibrosis. It is currently unknown whether telomerase mutations associate with cirrhosis induced by chronic liver disease. The telomerase RNA component (TERC) and the telomerase reverse transcriptase (TERT) were sequenced in 1,121 individuals (521 patients with cirrhosis induced by chronic liver disease and 600 noncirrhosis controls). Telomere length was analyzed in patients carrying telomerase gene mutations. Functional defects of telomerase gene mutations were investigated in primary human fibroblasts and patient-derived lymphocytes. An increased incidence of telomerase mutations was detected in cirrhosis patients (allele frequency 0.017) compared to noncirrhosis controls (0.003, P value 0.0007; relative risk [RR] 1.859; 95% confidence interval [CI] 1.552-2.227). Cirrhosis patients with TERT mutations showed shortened telomeres in white blood cells compared to control patients. Cirrhosis-associated telomerase mutations led to reduced telomerase activity and defects in maintaining telomere length and the replicative potential of primary cells in culture. CONCLUSION: This study provides the first experimental evidence that telomerase gene mutations are present in patients developing cirrhosis as a consequence of chronic liver disease. These data support the concept that telomere shortening can represent a causal factor impairing liver regeneration and accelerating cirrhosis formation in response to chronic liver disease.


Subject(s)
Liver Cirrhosis/genetics , Mutation , Telomerase/genetics , Adult , Aged , Aged, 80 and over , Chronic Disease , Female , Humans , Liver Cirrhosis/etiology , Liver Diseases/complications , Male , Middle Aged
13.
Circulation ; 122(18): 1823-36, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20956206

ABSTRACT

BACKGROUND: Ion channels are key determinants for the function of excitable cells, but little is known about their role and involvement during cardiac development. Earlier work identified Ca(2+)-activated potassium channels of small and intermediate conductance (SKCas) as important regulators of neural stem cell fate. Here we have investigated their impact on the differentiation of pluripotent cells toward the cardiac lineage. METHODS AND RESULTS: We have applied the SKCa activator 1-ethyl-2-benzimidazolinone on embryonic stem cells and identified this particular ion channel family as a new critical target involved in the generation of cardiac pacemaker-like cells: SKCa activation led to rapid remodeling of the actin cytoskeleton, inhibition of proliferation, induction of differentiation, and diminished teratoma formation. Time-restricted SKCa activation induced cardiac mesoderm and commitment to the cardiac lineage as shown by gene regulation, protein, and functional electrophysiological studies. In addition, the differentiation into cardiomyocytes was modulated in a qualitative fashion, resulting in a strong enrichment of pacemaker-like cells. This was accompanied by induction of the sino-atrial gene program and in parallel by a loss of the chamber-specific myocardium. In addition, SKCa activity induced activation of the Ras-Mek-Erk signaling cascade, a signaling pathway involved in the 1-ethyl-2-benzimidazolinone-induced effects. CONCLUSIONS: SKCa activation drives the fate of pluripotent cells toward mesoderm commitment and cardiomyocyte specification, preferentially into nodal-like cardiomyocytes. This provides a novel strategy for the enrichment of cardiomyocytes and in particular, the generation of a specific subtype of cardiomyocytes, pacemaker-like cells, without genetic modification.


Subject(s)
Cell Differentiation/physiology , Heart Conduction System/cytology , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Potassium Channels, Calcium-Activated/physiology , Animals , Benzimidazoles/pharmacology , Calcium Channel Agonists/pharmacology , Cell Line , Cell Proliferation , Cytoskeleton/physiology , Heart Conduction System/physiology , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Mice , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/physiology , Potassium Channels, Calcium-Activated/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...