Network


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

Hotspot


Dive into the research topics where Mathias Dahlmann is active.

Publication


Featured researches published by Mathias Dahlmann.


Journal of Molecular Medicine | 2010

MACC1 — more than metastasis? Facts and predictions about a novel gene

Ulrike Stein; Mathias Dahlmann; Wolfgang Walther

We recently identified the metastasis-associated in colon cancer 1 (MACC1) gene by a genome-wide search for differentially expressed genes in human colon cancer tissues, metastases, and normal tissues. Based on MACC1 expression in primary colon cancers, which did not present with metastases, our negative and positive prediction for metachronous metastasis was correct in 80% and 74% of cases, respectively. The 5-year-survival was 80% for MACC1 low expressors, but 15% for individuals who showed high MACC1 expression in their primary tumors. MACC1 induces migration, invasion and proliferation in cell culture, and liver and lung metastases in xenograft models. Here, we describe features of MACC1 beyond its utility as an indicator of metastasis. We elucidate its genomic localization and organization, its predicted splice variants, and single nucleotide polymorphisms. We discuss the MACC1 protein domain structure, posttranslational modifications, its conservation through evolution, and some family ties to SH3BP4. Furthermore, we summarize the predicted expressions of MACC1 in normal and malignant human tissues. We also evaluate the MACC1 levels in the context of one of its transcriptional targets, the receptor tyrosine kinase Met that activates the hepatocyte growth factor/Met signaling pathway, leading to enhanced cell motility, invasion, and metastasis.


Cancers | 2016

S100A4 in Cancer Metastasis: Wnt Signaling-Driven Interventions for Metastasis Restriction.

Mathias Dahlmann; Dennis Kobelt; Wolfgang Walther; Giridhar Mudduluru; Ulrike Stein

The aberrant activity of Wnt signaling is an early step in the transformation of normal intestinal cells to malignant tissue, leading to more aggressive tumors, and eventually metastases. In colorectal cancer (CRC), metastasis accounts for about 90% of patient deaths, representing the most lethal event during the course of the disease and is directly linked to patient survival, critically limiting successful therapy. This review focuses on our studies of the metastasis-inducing gene S100A4, which we identified as transcriptional target of β-catenin. S100A4 increased migration and invasion in vitro and metastasis in mice. In patient CRC samples, high S100A4 levels predict metastasis and reduced patient survival. Our results link pathways important for tumor progression and metastasis: the Wnt signaling pathway and S100A4, which regulates motility and invasiveness. S100A4 suppression by interdicting Wnt signaling has potential for therapeutic intervention. As proof of principle, we applied S100A4 shRNA systemically and prevented metastasis in mice. Furthermore, we identified small molecule inhibitors from high-throughput screens of pharmacologically active compounds employing an S100A4 promoter-driven reporter. Best hits act, as least in part, via intervening in the Wnt pathway and restricted metastasis in mouse models. We currently translate our findings on restricting S100A4-driven metastasis into clinical practice. The repositioned FDA-approved drug niclosamide, targeting Wnt signaling, is being tested in a prospective phase II clinical trial for treatment of CRC patients. Our assay for circulating S100A4 transcripts in patient blood is used to monitor treatment success.


Oncogene | 2016

SPON2, a newly identified target gene of MACC1, drives colorectal cancer metastasis in mice and is prognostic for colorectal cancer patient survival.

Felicitas Schmid; Q Wang; Matthew R. Huska; Miguel A. Andrade-Navarro; Margit Lemm; Iduna Fichtner; Mathias Dahlmann; Dennis Kobelt; Wolfgang Walther; Janice Smith; Peter M. Schlag; U. Stein

MACC1 (metastasis associated in colon cancer 1) is a prognostic biomarker for tumor progression, metastasis and survival of a variety of solid cancers including colorectal cancer (CRC). Here we aimed to identify the MACC1-induced transcriptome and key players mediating the MACC1-induced effects in CRC. We performed microarray analyses using CRC cells ectopically overexpressing MACC1. We identified more than 1300 genes at least twofold differentially expressed, including the gene SPON2 (Spondin 2) as 90-fold upregulated transcriptional target of MACC1. MACC1-dependent SPON2 expression regulation was validated on mRNA and protein levels in MACC1 high (endogenously or ectopically) and low (endogenously or by knockdown) expressing cells. Chromatin immunoprecipitation analysis demonstrated the binding of MACC1 to the gene promoter of SPON2. In cell culture, ectopic SPON2 overexpression induced cell viability, migration, invasion and colony formation in endogenously MACC1 and SPON2 low expressing cells, whereas SPON2 knockdown reduced proliferative, migratory and invasive abilities in CRC cells with high endogenous MACC1 and SPON2 expression. In intrasplenically transplanted NOD/SCID mice, metastasis induction was analyzed with control or SPON2-overexpressing CRC cells. Tumors with SPON2 overexpression induced liver metastasis (vs control animals without any metastases, P=0.0036). In CRC patients, SPON2 expression was determined in primary tumors (stages I–III), and survival time was analyzed by Kaplan–Meier method. CRC patients with high SPON2 expressing primary tumors demonstrated 8 months shorter metastasis-free survival (MFS) compared with patients with low SPON2 levels (P=0.053). Combining high levels of SPON2 and MACC1 improved the identification of high-risk patients with a 20-month shorter MFS vs patients with low biomarker expression. In summary, SPON2 is a transcriptional target of the metastasis gene MACC1. SPON2 induces cell motility in vitro and CRC metastasis in mice. In patients, SPON2 serves as prognostic indicator for CRC metastasis and survival, and might represent a promising target for therapeutic approaches.


Thyroid | 2015

RAGE Mediates the Pro-Migratory Response of Extracellular S100A4 in Human Thyroid Cancer Cells

Manoj Reddy Medapati; Mathias Dahlmann; Saeid Ghavami; Kumar Alok Pathak; Lydia Lucman; Thomas Klonisch; Cuong Hoang-Vu; Ulrike Stein; Sabine Hombach-Klonisch

BACKGROUND Expression of the small calcium-binding protein S100A4 is associated with poor prognosis in patients with thyroid cancer (TC). The authors have previously shown that S100A4 is a target for relaxin and insulin-like peptide 3 signaling in TC cells and that S100A4 is secreted from human TC cells. Although the pro-migratory role of intracellular S100A4 in binding to non-muscle myosin is well known, this study investigated here whether extracellular S100A4 contributes to TC migration. METHODS Human cell lines of follicular, papillary, and undifferentiated thyroid cancer, primary patient TC cells, and TC tissues were utilized to discover the presence of the receptor of advanced glycation end products (RAGE) in TC cells and TC tissues. Fluorescence imaging, protein pull-down assays, Western blot, siRNA protein silencing, small GTPase inhibitors, cell proliferation, and cell migration assays were used to investigate the interaction of extracellular S100A4 with RAGE in promoting a TC migratory response. RESULTS It was demonstrated that RAGE served as receptor for extracellular S100A4 mediating cell migration in TC cells. The RAGE-mediated increase in cell migration was dependent on the intracellular RAGE signaling partner diaphanous-1 (Dia-1) and involved the activation of the small GTPases Cdc42 and RhoA. Although extracellular S100A4 consistently activated ERK signaling in TC cells, it was shown that ERK signaling was not mediated by RAGE and not essential for the migratory response in TC cells. CONCLUSION The data have identified the RAGE/Dia-1 signaling system as a mediator for the pro-migratory response of extracellular S100A4 in human TC. Thus, therapeutic targeting of the RAGE/Dia-1/small GTPases signaling may successfully reduce local invasion and metastasis in TC.


Drug Resistance Updates | 2016

Repositioning of drugs for intervention in tumor progression and metastasis: Old drugs for new targets

Giridhar Mudduluru; Wolfgang Walther; Dennis Kobelt; Mathias Dahlmann; Christoph Treese; Yehuda G. Assaraf; Ulrike Stein

The increasing unraveling of the molecular basis of cancer offers manifold novel options for intervention strategies. However, the discovery and development of new drugs for potential clinical applications is a tremendously time-consuming and costly process. Translating a novel lead candidate compound into an approved clinical drug takes often more than a decade, and the success rate is very low due to versatile efforts including defining its pharmacokinetics, pharmacodynamics, side effects as well as lack of sufficient efficacy. Thus, strategies are needed to minimize time and costs, while maximizing success rates. A very attractive strategy for novel cancer therapeutic options is the repositioning of already approved drugs. These medicines, approved for the treatment of non-malignant disorders, have already passed some early costs and time, have been tested in humans and are ready for clinical trials as anti-cancer drugs. Here we discuss the repositioning of nonsteroidal anti-inflammatory drugs (NSAID), statins, anti-psychotic drugs, anti-helminthic drugs and vitamin D as anti-tumor agents. We focus on their novel actions and potential for inhibition of cancer growth and metastasis by interfering with target molecules and pathways, which drive these malignant processes. Furthermore, important pre-clinical and clinical data are reviewed herein, which elucidate their therapeutic mechanisms which enable their repositioning for cancer therapy and disruption of metastasis.


PLOS Biology | 2017

Statin and rottlerin small-molecule inhibitors restrict colon cancer progression and metastasis via MACC1

Manisha Juneja; Dennis Kobelt; Wolfgang Walther; Cynthia Voss; Janice Smith; Edgar Specker; Martin Neuenschwander; Bjoern-Oliver Gohlke; Mathias Dahlmann; Silke Radetzki; Robert Preissner; Jens Peter von Kries; Peter M. Schlag; Ulrike Stein

MACC1 (Metastasis Associated in Colon Cancer 1) is a key driver and prognostic biomarker for cancer progression and metastasis in a large variety of solid tumor types, particularly colorectal cancer (CRC). However, no MACC1 inhibitors have been identified yet. Therefore, we aimed to target MACC1 expression using a luciferase reporter-based high-throughput screening with the ChemBioNet library of more than 30,000 compounds. The small molecules lovastatin and rottlerin emerged as the most potent MACC1 transcriptional inhibitors. They remarkably inhibited MACC1 promoter activity and expression, resulting in reduced cell motility. Lovastatin impaired the binding of the transcription factors c-Jun and Sp1 to the MACC1 promoter, thereby inhibiting MACC1 transcription. Most importantly, in CRC-xenografted mice, lovastatin and rottlerin restricted MACC1 expression and liver metastasis. This is—to the best of our knowledge—the first identification of inhibitors restricting cancer progression and metastasis via the novel target MACC1. This drug repositioning might be of therapeutic value for CRC patients.


Archive | 2017

MACC1, a Novel Player in Solid Cancer Carcinogenesis

Giridhar Mudduluru; Katharina Ilm; Mathias Dahlmann; Ulrike Stein

Cancer develops due to uncontrolled proliferation of cells initiated by genetic instability, mutations, and environmental stress. Cells acquire these fundamental abnormalities in a multistep process due to changes in complex multilayer molecular network signaling axes equipping them with increased capacity of proliferation, survival, extracellular matrix (ECM) degradation, migration, invasion, and metastasis. Recently, metastasis-associated in colon cancer 1 (MACC1) was identified with differential display RT-PCR by analyzing the normal mucosa, primary, and metastasis specimens of colon cancer. We discovered that MACC1 is an important transcriptional regulator of hepatocyte growth factor (HGF) receptor c-Met and showed that MACC1 plays an important role in tumorigenesis, migration, invasion, and distant metastasis. In this book chapter, we discuss in-depth the structure and function of MACC1 in different aspects of carcinogenesis like gene regulation, signaling, cell proliferation, apoptosis, migration, invasion, metastasis, angiogenesis, epithelial mesenchymal transition (EMT), its role in cell metabolism, and also the impact of MACC1 as predictive and prognostic marker. In addition, we describe MACC1 as druggable target molecule by different approaches to reduce tumorigenesis and metastasis.


Methods of Molecular Biology | 2015

RNA Interference for Antimetastatic Therapy

Mathias Dahlmann; Ulrike Stein

The suppression of genes involved in tumor progression, metastasis formation, or therapy resistance by RNA interference is a promising tool to treat cancer disease. Efficient delivery of interfering molecules and their sustained presence in tumor cells are required for therapeutic success. This chapter describes a method of systemic application of shRNA expression plasmid via tail vein injection in xenograft mice, causing the sustained reduction of target gene expression in the primary tumor. By choosing S100A4 as a metastasis driving target gene, this therapeutic approach restricted the formation of distant colorectal cancer metastases after intrasplenic transplantation. In vivo imaging of bioluminescent cancer cells allows the monitoring of tumor growth and metastasis formation over time. End point analysis of the trial included scoring of the metastatic burden and the quantification of target gene expression in the tumor. Average S100A4 expression in tumor tissues was reduced by 30 %, causing a 70 % decrease of liver metastases.


Oncotarget | 2014

RAGE mediates S100A4-induced cell motility via MAPK/ERK and hypoxia signaling and is a prognostic biomarker for human colorectal cancer metastasis

Mathias Dahlmann; Anna Okhrimenko; Patrick Marcinkowski; Marc Osterland; Pia Herrmann; Janice Smith; Claus W. Heizmann; Peter M. Schlag; Ulrike Stein


Oncotarget | 2012

Systemic shRNA mediated knock-down of S100A4 in colorectal cancer xenografted mice reduces metastasis formation

Mathias Dahlmann; Ulrike Sack; Pia Herrmann; Margit Lemm; Iduna Fichtner; Peter M. Schlag; Ulrike Stein

Collaboration


Dive into the Mathias Dahlmann's collaboration.

Top Co-Authors

Avatar

Ulrike Stein

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Dennis Kobelt

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Wolfgang Walther

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Pia Herrmann

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Giridhar Mudduluru

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Janice Smith

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Iduna Fichtner

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Margit Lemm

Max Delbrück Center for Molecular Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge