Xinlai Cheng
Heidelberg University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Xinlai Cheng.
Molecular Cancer | 2014
Xinlai Cheng; Palvo Holenya; Suzan Can; Hamed Alborzinia; Riccardo Rubbiani; Ingo Ott; Stefan Wölfl
BackgroundCancer cells in the advanced stage show aberrant antioxidant capacity to detoxify excessive ROS resulting in the compensation for intrinsic oxidative stress and therapeutic resistance. PDAC is one of the most lethal cancers and often associated with a high accumulation of ROS. Recent studies identified gold(I) NHC complexes as potent TrxR inhibitors suppressing cell growth in a wide spectrum of human malignant cell lines at the low micromolar concentration. However, the mechanism of action is not completely elucidated yet.MethodsTo understand the biological function of gold(I) NHC complexes in PDAC, we used a recently published gold(I) NHC complex, MC3, and evaluated its anti-proliferative effect in four PDAC cell lines, determined by MTT and SRB assays. In further detailed analysis, we analyzed cellular ROS levels using the ROS indicator DHE and mitochondrial membrane potential indicated by the dye JC-1 in Panc1. We also analyzed cell cycle arrest and apoptosis by FACS. To elucidate the role of specific cell signaling pathways in MC3-induced cell death, co-incubation with ROS scavengers, a p38-MAPK inhibitor and siRNA mediated depletion of ASK1 were performed, and results were analyzed by immunoblotting, ELISA-microarrays, qRT-PCR and immunoprecipitation.ResultsOur data demonstrate that MC3 efficiently suppressed cell growth, and induced cell cycle arrest and apoptosis in pancreatic cancer cells, in particular in the gemcitabine-resistant cancer cells Panc1 and ASPC1. Treatment with MC3 resulted in a substantial alteration of the cellular redox homeostasis leading to increased ROS levels and a decrease in the mitochondrial membrane potential. ROS scavengers suppressed ROS formation and rescued cells from damage. On the molecular level, MC3 blocked the interaction of Trx with ASK1 and subsequently activated p38-associated signaling. Furthermore, inhibition of this pathway by using ASK1 siRNA or a p38 inhibitor clearly attenuated the effect of MC3 on cell proliferation in Panc1 and ASPC1.ConclusionsOur results confirm that MC3 is a TrxR inhibitor and show MC3 induced apoptosis in gemcitabine-resistant PDACs. MC3 mediated cell death could be blocked by using anti-oxidants, ASK1 siRNA or p38 inhibitor suggesting that the Trx-ASK1-p38 signal cascade played an important role in gold(I) NHC complexes-mediated cellular damage.
Molecular Oncology | 2012
Sangkil Nam; Anna Scuto; Fan Yang; WenYong Chen; Sungman Park; Hwa Seung Yoo; Heiko Konig; Ravi Bhatia; Xinlai Cheng; Karl Heinz Merz; Gerhard Eisenbrand; Richard Jove
Indirubin is the major active anti‐tumor component of a traditional Chinese herbal medicine used for treatment of chronic myelogenous leukemia (CML). While previous studies indicate that indirubin is a promising therapeutic agent for CML, the molecular mechanism of action of indirubin is not fully understood. We report here that indirubin derivatives (IRDs) potently inhibit Signal Transducer and Activator of Transcription 5 (Stat5) protein in CML cells. Compound E804, which is the most potent in this series of IRDs, blocked Stat5 signaling in human K562 CML cells, imatinib‐resistant human KCL‐22 CML cells expressing the T315I mutant Bcr‐Abl (KCL‐22M), and CD34‐positive primary CML cells from patients. Autophosphorylation of Src family kinases (SFKs) was strongly inhibited in K562 and KCL‐22M cells at 5 μM E804, and in primary CML cells at 10 μM E804, although higher concentrations partially inhibited autophosphorylation of Bcr‐Abl. Previous studies indicate that SFKs cooperate with Bcr‐Abl to activate downstream Stat5 signaling. Activation of Stat5 was strongly blocked by E804 in CML cells. E804 down‐regulated expression of Stat5 target proteins Bcl‐xL and Mcl‐1, associated with induction of apoptosis. In sum, our findings identify IRDs as potent inhibitors of the SFK/Stat5 signaling pathway downstream of Bcr‐Abl, leading to apoptosis of K562, KCL‐22M and primary CML cells. IRDs represent a promising structural class for development of new therapeutics for wild type or T315I mutant Bcr‐Abl‐positive CML patients.
Molecular Oncology | 2013
Sangkil Nam; Wei Wen; Anne Schroeder; Andreas Herrmann; Hua Yu; Xinlai Cheng; Karl-Heinz Merz; Gerhard Eisenbrand; Hongzhi Li; Yate-Ching Yuan; Richard Jove
Constitutively‐activated JAK/Stat3 or Src/Stat3 signaling plays a crucial role in tumor cell survival, proliferation, angiogenesis and immune suppression. Activated JAK/Stat3 or Src/Stat3 has been validated as a promising molecular target for cancer therapy. However, prolonged inhibition of Src family kinases (SFKs) leads to reactivation of signal transducer and activator of transcript 3 (Stat3) and tumor cell survival through altered JAK/Stat3 interaction. This compensatory feedback suggests that dual inhibition of Janus kinases (JAKs) and SFKs might be a promising strategy for targeting downstream Stat3 signaling in the clinic. In this study, we identify that the natural product derivative E738 is a novel dual inhibitor of JAKs and SFKs. The IC50 values of E738 against recombinant JAKs and SFKs in vitro are in the ranges of 0.7–74.1 nM and 10.7–263.9 nM, respectively. We observed that phosphorylation of both Jak2 and Src was substantially inhibited in the submicromolar range by E738 in cultured human pancreatic tumor cells, followed by blockade of downstream Stat3 activation. E738 down‐regulated expression of the Stat3 target proteins Mcl‐1 and survivin, associated with induction of apoptosis. Computational models and molecular dynamics simulations of E738/Tyk2 or E738/Src in silico suggest that E738 inhibits both tyrosine kinase 2 (Tyk2) and Src as an ATP‐competitive ligand. Moreover, the planar E738 molecule demonstrates a strong binding affinity in the compact ATP‐binding site of Tyk2. In sum, E738 is the first dual inhibitor of JAKs and SFKs, followed by inhibition of Stat3 signaling. Thus, according to in vitro experiments, E738 is a promising new therapeutic agent for human pancreatic cancer treatment by blocking both oncogenic pathways simultaneously.
Bioorganic & Medicinal Chemistry | 2010
Xinlai Cheng; Paul Rasqué; Sandra Vatter; Karl-Heinz Merz; Gerhard Eisenbrand
Indirubins have been reported to act as potent inhibitors of protein kinases relevant to tumorigenesis and of tumor cell growth, but their development to antitumor drugs suffer from their poor water solubility. We synthesized a novel class of indirubin derivatives, indirubin-5-carboxamides, carrying amide substituents with basic centers. Quaternization or protonation of these alkylamino substituents provided indirubins with significantly improved solubility without loss of bioactivity.
Journal of Cell Science | 2013
Hamed Alborzinia; Hannah Schmidt-Glenewinkel; Iryna Ilkavets; Katja Breitkopf-Heinlein; Xinlai Cheng; Peter Hortschansky; Steven Dooley; Stefan Wölfl
Summary Bone morphogenetic proteins (BMPs) are members of the TGF&bgr; family of signaling proteins and play an important role during development and in tissue formation. BMP signaling is a well-studied process, which is initiated through binding of cognate receptors and processed through activation of Smad downstream mediators. A hallmark of BMP signaling is its modulation at the extracellular level through specific antagonists. Although it had been shown that BMP and TGF&bgr; receptors are internalized following activation, little is known about the fate of BMP ligands. We prepared biologically active fluorescently labeled BMP2 and quantitatively analyzed its binding and uptake in cells using flow cytometry and confocal microscopy. Exogenous BMP2 was rapidly bound to the cell surface and subsequently internalized in a time-dependent manner and accumulated in the cell center. Although binding to the cell surface was limited by binding sites at the beginning, internalization continously increased with time, after a short delay. Using different inhibitors we found that internalization of BMP2 through endosomal particles occurred in a clathrin-dependent pathway. Furthermore, uptake of BMP2 was modulated in strikingly different ways by BMP2 antagonists. Although Noggin and Gremlin increased BMP2 uptake, Chordin blocked BMP2 uptake, which was concentration dependent in both cases. In conclusion, our findings present interesting mechanisms for the modulation of BMP signaling by concentration gradients of BMP ligands and antagonists in a dose- and time-dependent manner, which can provide an explanation of some properties of the BMP regulatory network.
Chemistry & Biology | 2012
Xinlai Cheng; Hamed Alborzinia; Karl-Heinz Merz; Herbert Steinbeisser; Ralf Mrowka; Catharina Scholl; Igor Kitanovic; Gerhard Eisenbrand; Stefan Wölfl
Regulatory Smads (R-Smads), Smad1/5/8 and Smad2/3, are the central mediators of TGFβ and BMP signaling pathways. Here, we screened indirubin derivatives, known kinase inhibitors, and observed strong interference with BMP signaling. We found that indirubin derivative E738 inhibited both TGFβ and BMP pathways through ubiquitin-proteasome-mediated depletion of total R-Smad pools, although phospho-R-Smad levels were initially stabilized by GSK3β and cyclin-dependent kinase inhibition. E738 also enhanced p38 and JNK phosphorylation, involved in Smad-independent TGFβ/BMP signaling. Additionally, using a small siRNA screen, we showed that depletion of ubiquitin proteases USP9x and USP34 significantly reduced total R-Smad levels, mimicking E738 treatment. In fact, both USP9x and USP34 levels were significantly reduced in E738-treated cells. Our findings not only describe the complex activity profile of the indirubin derivative E738, but also reveal a mechanism for controlling TGFβ/BMP signaling, the control of R-Smad protein levels through deubiquitination.
Molecular Oncology | 2016
Xinlai Cheng; Jee Young Kim; Shahrouz Ghafoory; Tijen Duvaci; Roya Rafiee; Jannick Theobald; Hamed Alborzinia; Pavlo Holenya; Johannes Fredebohm; Karl Heinz Merz; Arianeb Mehrabi; Mohammadreza Hafezi; Arash Saffari; Gerhard Eisenbrand; Jörg D. Hoheisel; Stefan Wölfl
Pancreatic ductal adenocarcinoma (PDAC) clinically has a very poor prognosis. No small molecule is available to reliably achieve cures. Meisoindigo is chemically related to the natural product indirubin and showed substantial efficiency in clinical chemotherapy for CML in China. However, its effect on PDAC is still unknown. Our results showed strong anti‐proliferation effect of meisoindigo on gemcitabine‐resistant PDACs. Using a recently established primary PDAC cell line, called Jopaca‐1 with a larger CSCs population as model, we observed a reduction of CD133+ and ESA+/CD44+/CD24+ populations upon treatment and concomitantly a decreased expression of CSC‐associated genes, and reduced cellular mobility and sphere formation. Investigating basic cellular metabolic responses, we detected lower oxygen consumption and glucose uptake, while intracellular ROS levels increased. This was effectively neutralized by the addition of antioxidants, indicating an essential role of the cellular redox balance. Further analysis on energy metabolism related signaling revealed that meisoindigo inhibited LKB1, but activated AMPK. Both of them were involved in cellular apoptosis. Additional in situ hybridization in tissue sections of PDAC patients reproducibly demonstrated co‐expression and ‐localization of LKB1 and CD133 in malignant areas. Finally, we detected that CD133+/CD44+ were more vulnerable to meisoindigo, which could be mimicked by LKB1 siRNAs. Our results provide the first evidence, to our knowledge, that LKB1 sustains the CSC population in PDACs and demonstrate a clear benefit of meisoindigo in treatment of gemcitabine‐resistant cells. This novel mechanism may provide a promising new treatment option for PDAC.
Phytomedicine | 2015
Shan Su; Xinlai Cheng; Michael Wink
Arctium lappa is a well-known traditional medicinal plant in China (TCM) and Europe that has been used for thousands of years to treat arthritis, baldness or cancer. The plant produces lignans as secondary metabolites which have a wide range of bioactivities. Yet, their ability to reverse multidrug resistance (MDR) in cancer cells has not been explored. In this study, we isolated six lignans from A. lappa seeds, namely arctigenin, matairesinol, arctiin, (iso)lappaol A, lappaol C, and lappaol F. The MDR reversal potential of the isolated lignans and the underlying mechanism of action were studied using two MDR cancer cell lines, CaCo2 and CEM/ADR 5000 which overexpress P-gp and other ABC transporters. In two-drug combinations of lignans with the cytotoxic doxorubicin, all lignans exhibited synergistic effects in CaCo2 cells and matairesinol, arctiin, lappaol C and lappaol F display synergistic activity in CEM/ADR 5000 cells. Additionally, in three-drug combinations of lignans with the saponin digitonin and doxorubicin MDR reversal activity was even stronger enhanced. The lignans can increase the retention of the P-gp substrate rhodamine 123 in CEM/ADR 5000 cells, indicating that lignans can inhibit the activity of P-gp. Our study provides a first insight into the potential chemosensitizing activity of a series of natural lignans, which might be candidates for developing novel adjuvant anticancer agents.
Journal of Medicinal Chemistry | 2015
Xinlai Cheng; Eleni Dimou; Hamed Alborzinia; Frank Wenke; Axel Göhring; Stefanie Reuter; Nancy Mah; Heiko Fuchs; Miguel A. Andrade-Navarro; James Adjaye; Sheraz Gul; Christoph Harms; Jochen Utikal; Edda Klipp; Ralf Mrowka; Stefan Wölfl
Reprogramming somatic cells into induced-pluripotent cells (iPSCs) provides new access to all somatic cell types for clinical application without any ethical controversy arising from the use of embryonic stem cells (ESCs). Established protocols for iPSCs generation based on viral transduction with defined factors are limited by low efficiency and the risk of genetic abnormality. Several small molecules have been reported as replacements for defined transcriptional factors, but a chemical able to replace Oct3/4 allowing the generation of human iPSCs is still unavailable. Using a cell-based High Throughput Screening (HTS) campaign, we identified that 2-[4-[(4-methoxyphenyl)methoxy]phenyl]acetonitrile (1), termed O4I1, enhanced Oct3/4 expression. Structural verification and modification by chemical synthesis showed that O4I1 and its derivatives not only promoted expression and stabilization of Oct3/4 but also enhanced its transcriptional activity in diverse human somatic cells, implying the possible benefit from using this class of compounds in regenerative medicine.
Journal of Pharmacy and Pharmacology | 2014
Nasim Heshmati; Xinlai Cheng; Else Dapat; Philip Sassene; Gerhard Eisenbrand; Gert Fricker; Anette Müllertz
Anticancer indirubins are poorly soluble in water. Here, digestion of four self‐emulsifying drug delivery systems (SEDDS) containing E804 (indirubin‐3′‐oxime 2,3‐dihydroxypropyl ether) was compared by dynamic lipolysis and bioavailability studies. Used lipids were either medium‐chain or long‐chain glycerides.