Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where André Lechel is active.

Publication


Featured researches published by André Lechel.


Nature Genetics | 2007

Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation

Aaheli Roy Choudhury; Zhenyu Ju; Meta Wulandari Djojosubroto; Andrea Schienke; André Lechel; Sonja Schaetzlein; Hong Jiang; Anna Stepczynska; Chunfang Wang; Jan Buer; Han-Woong Lee; Thomas von Zglinicki; Arnold Ganser; Peter Schirmacher; Hiromitsu Nakauchi; K. Lenhard Rudolph

Telomere shortening limits the proliferative lifespan of human cells by activation of DNA damage pathways, including upregulation of the cell cycle inhibitor p21 (encoded by Cdkn1a, also known as Cip1 and Waf1)) (refs. 1–5). Telomere shortening in response to mutation of the gene encoding telomerase is associated with impaired organ maintenance and shortened lifespan in humans and in mice. The in vivo function of p21 in the context of telomere dysfunction is unknown. Here we show that deletion of p21 prolongs the lifespan of telomerase-deficient mice with dysfunctional telomeres. p21 deletion improved hematolymphopoiesis and the maintenance of intestinal epithelia without rescuing telomere function. Moreover, deletion of p21 rescued proliferation of intestinal progenitor cells and improved the repopulation capacity and self-renewal of hematopoietic stem cells from mice with dysfunctional telomeres. In these mice, apoptotic responses remained intact, and p21 deletion did not accelerate chromosomal instability or cancer formation. This study provides experimental evidence that telomere dysfunction induces p21-dependent checkpoints in vivo that can limit longevity at the organismal level.


Cell | 2012

A Differentiation Checkpoint Limits Hematopoietic Stem Cell Self-Renewal in Response to DNA Damage

Jianwei Wang; Qian Sun; Yohei Morita; Hong Jiang; Alexander Groß; André Lechel; Kai Hildner; Luis Miguel Guachalla; Anne Gompf; Daniel Hartmann; Axel Schambach; Torsten Wuestefeld; Daniel Dauch; Hubert Schrezenmeier; Wolf-Karsten Hofmann; Hiromitsu Nakauchi; Zhenyu Ju; Hans A. Kestler; Lars Zender; K. Lenhard Rudolph

Checkpoints that limit stem cell self-renewal in response to DNA damage can contribute to cancer protection but may also promote tissue aging. Molecular components that control stem cell responses to DNA damage remain to be delineated. Using in vivo RNAi screens, we identified basic leucine zipper transcription factor, ATF-like (BATF) as a major component limiting self-renewal of hematopoietic stem cells (HSCs) in response to telomere dysfunction and γ-irradiation. DNA damage induces BATF in a G-CSF/STAT3-dependent manner resulting in lymphoid differentiation of HSCs. BATF deletion improves HSC self-renewal and function in response to γ-irradiation or telomere shortening but results in accumulation of DNA damage in HSCs. Analysis of bone marrow from patients with myelodysplastic syndrome supports the conclusion that DNA damage-dependent induction of BATF is conserved in human HSCs. Together, these results provide experimental evidence that a BATF-dependent differentiation checkpoint limits self-renewal of HSCs in response to DNA damage.


Cell | 2007

Exonuclease-1 Deletion Impairs DNA Damage Signaling and Prolongs Lifespan of Telomere-Dysfunctional Mice

Sonja Schaetzlein; N.R. Kodandaramireddy; Zhenyu Ju; André Lechel; Anna Stepczynska; Dana R. Lilli; Alan B. Clark; Cornelia Rudolph; Florian Kühnel; Kaichun Wei; Brigitte Schlegelberger; Peter Schirmacher; Thomas A. Kunkel; Roger A. Greenberg; Winfried Edelmann; K. Lenhard Rudolph

Exonuclease-1 (EXO1) mediates checkpoint induction in response to telomere dysfunction in yeast, but it is unknown whether EXO1 has similar functions in mammalian cells. Here we show that deletion of the nuclease domain of Exo1 reduces accumulation of DNA damage and DNA damage signal induction in telomere-dysfunctional mice. Exo1 deletion improved organ maintenance and lifespan of telomere-dysfunctional mice but did not increase chromosomal instability or cancer formation. Deletion of Exo1 also ameliorated the induction of DNA damage checkpoints in response to gamma-irradiation and conferred cellular resistance to 6-thioguanine-induced DNA damage. Exo1 deletion impaired upstream induction of DNA damage responses by reducing ssDNA formation and the recruitment of Replication Protein A (RPA) and ATR at DNA breaks. Together, these studies provide evidence that EXO1 contributes to DNA damage signal induction in mammalian cells, and deletion of Exo1 can prolong survival in the context of telomere dysfunction.


Cancer Cell | 2013

Loss of p53 in Enterocytes Generates an Inflammatory Microenvironment Enabling Invasion and Lymph Node Metastasis of Carcinogen-Induced Colorectal Tumors

Sarah Schwitalla; Paul K. Ziegler; David Horst; Valentin Becker; Irina Kerle; Yvonne Begus-Nahrmann; André Lechel; K. Lenhard Rudolph; Rupert Langer; Julia Slotta-Huspenina; Franz G. Bader; Olivia Prazeres da Costa; Markus F. Neurath; Alexander Meining; Thomas Kirchner; Florian R. Greten

Loss of p53 is considered to allow progression of colorectal tumors from the adenoma to the carcinoma stage. Using mice with an intestinal epithelial cell (IEC)-specific p53 deletion, we demonstrate that loss of p53 alone is insufficient to initiate intestinal tumorigenesis but markedly enhances carcinogen-induced tumor incidence and leads to invasive cancer and lymph node metastasis. Whereas p53 controls DNA damage and IEC survival during the initiation stage, loss of p53 during tumor progression is associated with increased intestinal permeability, causing formation of an NF-κB-dependent inflammatory microenvironment and the induction of epithelial-mesenchymal transition. Thus, we propose a p53-controlled tumor-suppressive function that is independent of its well-established role in cell-cycle regulation, apoptosis, and senescence.


Hepatology | 2007

Telomere shortening and inactivation of cell cycle checkpoints characterize human hepatocarcinogenesis

Ruben R. Plentz; Young Nyun Park; André Lechel; Haeryoung Kim; Friederike Nellessen; Britta Heike Langkopf; Ludwig Wilkens; Annarita Destro; Barbara Fiamengo; Michael P. Manns; Massimo Roncalli; Karl Lenhard Rudolph

Telomere shortening and inactivation of cell cycle checkpoints characterize carcinogenesis. Whether these molecular features coincide at specific stages of human hepatocarcinogenesis is unknown. The preneoplasia–carcinoma sequence of human HCC is not well defined. Small cell changes (SCC) and large cell changes (LCC) are potential precursor lesions. We analyzed hepatocellular telomere length, the prevalence of DNA damage, and the expression of p21 and p16 in biopsy specimens of patients with chronic liver disease (n = 27) that showed different precursor lesions and/or HCC: liver cirrhosis (n = 25), LCC (n = 26), SCC (n = 13), and HCC (n = 13). The study shows a decrease in telomere length in nondysplastic cirrhotic liver compared with normal liver and a further significant shortening of telomeres in LCC, SCC, and HCC. HCC had the shortest telomeres, followed by SCC and LCC. Hepatocytes showed an increased p21 labeling index (p21‐LI) at the cirrhosis stage, which remained elevated in most LCC. In contrast, most SCC and HCC showed a strongly reduced p21‐LI. Similarly, p16 was strongly expressed in LCC but reduced in SCC and not detectable in HCC. γH2AX‐DNA‐damage‐foci were not detected in LCC but were present in SCC and more frequently in HCC. These data indicate that LCC and SCC represent clonal expansions of hepatocytes with shortened telomeres. Conclusion: The inactivation of cell cycle checkpoints coincides with further telomere shortening and an accumulation of DNA damage in SCC and HCC, suggesting that SCC represent more advanced precursor lesions compared with LCC. (HEPATOLOGY 2007;45:968–976.)


Hepatology | 2011

Telomerase gene mutations are associated with cirrhosis formation

Daniel Hartmann; Ujala Srivastava; Michaela Thaler; Karin N. Kleinhans; G. Nkontchou; Annika Scheffold; Kerstin Bauer; Ramona F. Kratzer; Natalia Kloos; Sarah-Fee Katz; Zhangfa Song; Yvonne Begus-Nahrmann; Alexander Kleger; Guido von Figura; Pavel Strnad; André Lechel; Cagatay Günes; Andrej Potthoff; Katja Deterding; Heiner Wedemeyer; Zhenyu Ju; Ge Song; Feng Xiao; Sonja Gillen; Hubert Schrezenmeier; Thomas Mertens; Marianne Ziol; Helmut Friess; Michael Jarek; Michael P. Manns

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. (HEPATOLOGY 2011;)


Nature Genetics | 2009

p53 deletion impairs clearance of chromosomal-instable stem cells in aging telomere-dysfunctional mice

Yvonne Begus-Nahrmann; André Lechel; Anna C. Obenauf; Kodandaramireddy Nalapareddy; Elvira Peit; Eva Maria Hoffmann; Falk Schlaudraff; Birgit Liss; Peter Schirmacher; Hans A. Kestler; Esther Danenberg; Nick Barker; Hans Clevers; Michael R. Speicher; K. Lenhard Rudolph

Telomere dysfunction limits the proliferative capacity of human cells and induces organismal aging by activation of p53 and p21 (refs. 1, 2, 3, 4, 5, 6). Although deletion of p21 elongates the lifespan of telomere-dysfunctional mice, a direct analysis of p53 in telomere-related aging has been hampered by early tumor formation in p53 knockout mice. Here we analyzed the functional consequences of conditional p53 deletion. Intestinal deletion of p53 shortened the lifespan of telomere-dysfunctional mice without inducing tumor formation. In contrast to p21 deletion, the deletion of p53 impaired the depletion of chromosomal-instable intestinal stem cells in aging telomere-dysfunctional mice. These instable stem cells contributed to epithelial regeneration leading to an accumulation of chromosomal instability, increased apoptosis, altered epithelial cell differentiation and premature intestinal failure. Together, these results provide the first experimental evidence for an organ system in which p53-dependent mechanisms prevent tissue destruction in response to telomere dysfunction by depleting genetically instable stem cells.


EMBO Reports | 2005

The cellular level of telomere dysfunction determines induction of senescence or apoptosis in vivo

André Lechel; Ande Satyanarayana; Zhenyu Ju; Ruben R. Plentz; Sonja Schaetzlein; Cornelia Rudolph; Ludwig Wilkens; Stephanie U Wiemann; Gabriele Saretzki; Nisar P. Malek; Michael P. Manns; Jan Buer; K. Lenhard Rudolph

Telomere dysfunction induces two types of cellular response: cellular senescence and apoptosis. We analysed the extent to which the cellular level of telomere dysfunction and p53 gene status affect these cellular responses in mouse liver using the experimental system of TRF2 inhibition by a dominant‐negative version of the protein (TRF2ΔBΔM). We show that the level of telomere dysfunction correlates with the level of TRF2ΔBΔM protein expression resulting in chromosomal fusions, aberrant mitotic figures and aneuploidy of liver cells. These alterations provoked p53‐independent apoptosis, but a strictly p53‐dependent senescence response in distinct populations of mouse liver cells depending on the cellular level of TRF2ΔBΔM expression. Apoptosis was associated with higher expression of TRF2ΔBΔM, whereas cellular senescence was associated with low levels of TRF2ΔBΔM expression. Our data provide experimental evidence that induction of senescence or apoptosis in vivo depends on the cellular level of telomere dysfunction and differentially on p53 gene function.


Mechanisms of Ageing and Development | 2009

Telomeres shorten while Tert expression increases during ageing of the short-lived fish Nothobranchius furzeri

Nils Hartmann; Kathrin Reichwald; André Lechel; Michael Graf; Jeanette Kirschner; Alexander Dorn; Eva Terzibasi; Juliane Wellner; Matthias Platzer; Karl Lenhard Rudolph; Alessandro Cellerino; Christoph Englert

Age research in vertebrates is often limited by the longevity of available models. The teleost fish Nothobranchius furzeri has an exceptionally short lifespan with 3.5 months for the laboratory strain GRZ and about 6 months for the wild-derived strain MZM-0403. Here we have investigated telomere length in muscle and skin tissue of young and old fish of both strains using different methods. We found age-dependent telomere shortening in the MZM-0403 strain with the longer lifespan, whereas the short-lived GRZ strain showed no significant telomere shortening with advanced age. Sequencing of the two main telomerase genes Tert and Terc revealed that both genes are highly conserved between the N. furzeri strains while there is little conservation to other fish species and humans. Both genes are ubiquitously expressed in N. furzeri and expression levels of Tert and Terc correlate with telomerase activity in a tissue-specific manner. Unexpectedly, the expression level of Tert is increased in aged muscle and skin tissue of MZM-0403 suggesting that telomeres shorten upon ageing despite increased Tert expression and hence high telomerase activity. We further conclude that the extremely short lifespan of the GRZ strain is not caused by diminished telomerase activity or accelerated telomere shortening.


Nature Cell Biology | 2012

Puma and p21 represent cooperating checkpoints limiting self-renewal and chromosomal instability of somatic stem cells in response to telomere dysfunction

Tobias Sperka; Zhangfa Song; Yohei Morita; Kodandaramireddy Nalapareddy; Luis Miguel Guachalla; André Lechel; Yvonne Begus-Nahrmann; Martin D. Burkhalter; Monika Mach; Falk Schlaudraff; Birgit Liss; Zhenyu Ju; Michael R. Speicher; K. Lenhard Rudolph

The tumour suppressor p53 activates Puma-dependent apoptosis and p21-dependent cell-cycle arrest in response to DNA damage. Deletion of p21 improved stem-cell function and organ maintenance in progeroid mice with dysfunctional telomeres, but the function of Puma has not been investigated in this context. Here we show that deletion of Puma improves stem- and progenitor-cell function, organ maintenance and lifespan of telomere-dysfunctional mice. Puma deletion impairs the clearance of stem and progenitor cells that have accumulated DNA damage as a consequence of critically short telomeres. However, further accumulation of DNA damage in these rescued progenitor cells leads to increasing activation of p21. RNA interference experiments show that upregulation of p21 limits proliferation and evolution of chromosomal imbalances of Puma-deficient stem and progenitor cells with dysfunctional telomeres. These results provide experimental evidence that p53-dependent apoptosis and cell-cycle arrest act in cooperating checkpoints limiting tissue maintenance and evolution of chromosomal instability at stem- and progenitor-cell levels in response to telomere dysfunction. Selective inhibition of Puma-dependent apoptosis can result in temporary improvements in maintenance of telomere-dysfunctional organs.

Collaboration


Dive into the André Lechel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Schirmacher

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qiong Lin

RWTH Aachen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge