Network


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

Hotspot


Dive into the research topics where Jessica Roos is active.

Publication


Featured researches published by Jessica Roos.


Frontiers in Pharmacology | 2010

5-Lipoxygenase: Underappreciated Role of a Pro-Inflammatory Enzyme in Tumorigenesis

Dieter Steinhilber; As Fischer; Julia Metzner; Svenja Dorothea Steinbrink; Jessica Roos; Martin Ruthardt; Thorsten J. Maier

Leukotrienes constitute a group of bioactive lipids generated by the 5-lipoxygenase (5-LO) pathway. An increasing body of evidence supports an acute role for 5-LO products already during the earliest stages of pancreatic, prostate, and colorectal carcinogenesis. Several pieces of experimental data form the basis for this hypothesis and suggest a correlation between 5-LO expression and tumor cell viability. First, several independent studies documented an overexpression of 5-LO in primary tumor cells as well as in established cancer cell lines. Second, addition of 5-LO products to cultured tumor cells also led to increased cell proliferation and activation of anti-apoptotic signaling pathways. 5-LO antisense technology approaches demonstrated impaired tumor cell growth due to reduction of 5-LO expression. Lastly, pharmacological inhibition of 5-LO potently suppressed tumor cell growth by inducing cell cycle arrest and triggering cell death via the intrinsic apoptotic pathway. However, the documented strong cytotoxic off-target effects of 5-LO inhibitors, in combination with the relatively high concentrations of 5-LO products needed to achieve mitogenic effects in cell culture assays, raise concern over the assignment of the cause, and question the relationship between 5-LO products and tumorigenesis.


Cancer Research | 2014

5-Lipoxygenase Is a Candidate Target for Therapeutic Management of Stem Cell–like Cells in Acute Myeloid Leukemia

Jessica Roos; Claudia Oancea; Maria Heinssmann; Dilawar Khan; Hannelore Held; Astrid S. Kahnt; Ricardo Capelo; Estel.la Buscató; Ewgenij Proschak; Elena Puccetti; Dieter Steinhilber; Ingrid Fleming; Thorsten J. Maier; Martin Ruthardt

Nonsteroidal anti-inflammatory drugs such as sulindac inhibit Wnt signaling, which is critical to maintain cancer stem cell-like cells (CSC), but they also suppress the activity of 5-lipoxygenase (5-LO) at clinically feasible concentrations. Recently, 5-LO was shown to be critical to maintain CSC in a model of chronic myeloid leukemia. For these reasons, we hypothesized that 5-LO may offer a therapeutic target to improve the management of acute myeloid leukemia (AML), an aggressive disease driven by CSCs. Pharmacologic and genetic approaches were used to evaluate the effects of 5-LO blockade in a PML/RARα-positive model of AML. As CSC models, we used Sca-1(+)/lin(-) murine hematopoietic stem and progenitor cells (HSPC), which were retrovirally transduced with PML/RARα. We found that pharmacologic inhibition of 5-LO interfered strongly with the aberrant stem cell capacity of PML/RARα-expressing HSPCs. Through small-molecule inhibitor studies and genetic disruption of 5-LO, we also found that Wnt and CSC inhibition is mediated by the enzymatically inactive form of 5-LO, which hinders nuclear translocation of β-catenin. Overall, our findings revealed that 5-LO inhibitors also inhibit Wnt signaling, not due to the interruption of 5-LO-mediated lipid signaling but rather due to the generation of a catalytically inactive form of 5-LO, which assumes a new function. Given the evidence that CSCs mediate AML relapse after remission, eradication of CSCs in this setting by 5-LO inhibition may offer a new clinical approach for immediate evaluation in patients with AML. Cancer Res; 74(18); 5244-55. ©2014 AACR.


Cell Cycle | 2012

Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells.

Annette Romanski; Kerstin Schwarz; Maren Keller; Sarah Wietbrauk; Anja Vogel; Jessica Roos; Claudia Oancea; Boris Brill; Oliver H. Krämer; Hubert Serve; Martin Ruthardt; Gesine Bug

Acute myeloid leukemia (AML) is a highly malignant disease that is not curable in the majority of patients. Numerous non-random genetic abnormalities are known, among which several translocations such as PLZF/RARα or AML1/ETO are known to aberrantly recruit histone deacetylases. Deacetylase inhibitors (DACi) are promising drugs leading to growth inhibition, cell cycle arrest, premature senescence and apoptosis in malignant cells. It is believed that DACi may have clinical efficacy by eradicating the most primitive population of leukemic stem and progenitor cells, possibly by interfering with self-renewal. The aim of the study was to investigate the effects of DACi on leukemic stem and progenitor cells using murine transduction-transplantation models of hematopoietic cells harboring the leukemia-associated fusion proteins (LAFP) PLZF/RARα or a truncated AML1/ETO protein (AML1/ETO exon 9). We show that the self-renewal and short-term repopulation capacity of AML1/ETO- or PLZF/RARα-expressing Sca1+/lin- stem and progenitor cells are profoundly inhibited by clinically applicable concentrations of the DACi dacinostat and vorinostat. To further investigate the mechanisms underlying these effects, we examined the impact of DACi on the transcription factor c-MYC and the Polycomb group protein BMI1, which are induced by LAFP and involved in leukemic transformation. In AML1/ETO or PLZF/RARα-positive 32D cells, DACi-mediated antiproliferative effects were associated with downregulation of BMI1 and c-MYC protein levels. Similar effects were demonstrated in primary samples of cytogenetically defined high-risk AML patients. In conclusion, DACi may be effective as maintenance therapy by negatively interfering with signaling pathways that control survival and proliferation of leukemic stem and progenitor cells.


Antioxidants & Redox Signaling | 2014

Electrophilic Fatty Acid Species Inhibit 5-Lipoxygenase and Attenuate Sepsis-Induced Pulmonary Inflammation

Khader Awwad; Svenja Dorothea Steinbrink; Timo Frömel; Nicole Lill; Johann Isaak; Ann-Kathrin Häfner; Jessica Roos; Bettina Hofmann; Heinrich Heide; Gerd Geisslinger; Dieter Steinhilber; Bruce A. Freeman; Thorsten J. Maier; Ingrid Fleming

AIMS The reaction of nitric oxide and nitrite-derived species with polyunsaturated fatty acids yields electrophilic fatty acid nitroalkene derivatives (NO2-FA), which display anti-inflammatory properties. Given that the 5-lipoxygenase (5-LO, ALOX5) possesses critical nucleophilic amino acids, which are potentially sensitive to electrophilic modifications, we determined the consequences of NO2-FA on 5-LO activity in vitro and on 5-LO-mediated inflammation in vivo. RESULTS Stimulation of human polymorphonuclear leukocytes (PMNL) with nitro-oleic (NO2-OA) or nitro-linoleic acid (NO2-LA) (but not the parent lipids) resulted in the concentration-dependent and irreversible inhibition of 5-LO activity. Similar effects were observed in cell lysates and using the recombinant human protein, indicating a direct reaction with 5-LO. NO2-FAs did not affect the activity of the platelet-type 12-LO (ALOX12) or 15-LO-1 (ALOX15) in intact cells or the recombinant protein. The NO2-FA-induced inhibition of 5-LO was attributed to the alkylation of Cys418, and the exchange of Cys418 to serine rendered 5-LO insensitive to NO2-FA. In vivo, the systemic administration of NO2-OA to mice decreased neutrophil and monocyte mobilization in response to lipopolysaccharide (LPS), attenuated the formation of the 5-LO product 5-hydroxyeicosatetraenoic acid (5-HETE), and inhibited lung injury. The administration of NO2-OA to 5-LO knockout mice had no effect on LPS-induced neutrophil or monocyte mobilization as well as on lung injury. INNOVATION Prophylactic administration of NO2-OA to septic mice inhibits inflammation and promotes its resolution by interfering in 5-LO-mediated inflammatory processes. CONCLUSION NO2-FAs directly and irreversibly inhibit 5-LO and attenuate downstream acute inflammatory responses.


Pharmacology & Therapeutics | 2016

Regulation of tumorigenic Wnt signaling by cyclooxygenase-2, 5-lipoxygenase and their pharmacological inhibitors: A basis for novel drugs targeting cancer cells?

Jessica Roos; Sabine Grösch; Oliver Werz; Peter Schröder; Slava Ziegler; Simone Fulda; Patrick Paulus; Anja Urbschat; Benjamin Kühn; Isabelle V. Maucher; Jasmin Fettel; Thomas Vorup-Jensen; Matthias Piesche; Carmela Matrone; Dieter Steinhilber; Michael J. Parnham; Thorsten J. Maier

Canonical Wnt signaling is a highly conserved pathway with a prominent role in embryogenic development, adult tissue homeostasis, cell polarization, stem cell biology, cell differentiation, and proliferation. Furthermore, canonical Wnt signaling is of pivotal importance in the pathogenesis of a number of cancer types and crucially affects tumor initiation, cancer cell proliferation, cancer cell apoptosis, and metastasis. Reports over the last decade have provided strong evidence for a pathophysiological role of Wnt signaling in non-malignant classical inflammatory and neurodegenerative diseases. Although, several agents suppressing the Wnt pathway at different levels have been identified, the development of clinically relevant Wnt-inhibiting agents remains challenging due to selectivity and toxicity issues. Several studies have shown that long-term administration of non-steroidal anti-inflammatory drugs protects against colon cancer and potentially other tumor types by interfering both with the COX and the Wnt pathway. Our own studies have shown that non-steroidal anti-inflammatory drugs suppress Wnt signaling by targeting the pro-inflammatory enzyme 5-lipoxygenase which is the key enzyme pathophysiologically involved in the synthesis of leukotrienes. Furthermore, we found a direct link between the 5-lipoxygenase and Wnt signaling pathways, which is essential for the maintenance of leukemic stem cells. Accordingly, genetic and pharmacological inhibition of 5-lipoxygenase led to an impairment of Wnt-dependent acute and chronic myeloid leukemic stem cells. We believe that 5-lipoxygenase inhibitors might represent a novel type of Wnt inhibitor activating a potentially naturally occurring novel mechanism of suppression of Wnt signaling that is non-toxic, at least in mice, and is potentially well tolerated in patients.


PLOS ONE | 2011

Sulindac sulfide reverses aberrant self-renewal of progenitor cells induced by the AML-associated fusion proteins PML/RARα and PLZF/RARα.

Gunnar Steinert; Claudia Oancea; Jessica Roos; Heike Hagemeyer; Thorsten J. Maier; Martin Ruthardt; Elena Puccetti

Chromosomal translocations can lead to the formation of chimeric genes encoding fusion proteins such as PML/RARα, PLZF/RARα, and AML-1/ETO, which are able to induce and maintain acute myeloid leukemia (AML). One key mechanism in leukemogenesis is increased self renewal of leukemic stem cells via aberrant activation of the Wnt signaling pathway. Either X-RAR, PML/RARα and PLZF/RARα or AML-1/ETO activate Wnt signaling by upregulating γ-catenin and β-catenin. In a prospective study, a lower risk of leukemia was observed with aspirin use, which is consistent with numerous studies reporting an inverse association of aspirin with other cancers. Furthermore, a reduction in leukemia risk was associated with use of non-steroidal anti-inflammatory drug (NSAID), where the effects on AML risk was FAB subtype-specific. To better investigate whether NSAID treatment is effective, we used Sulindac Sulfide in X-RARα-positive progenitor cell models. Sulindac Sulfide (SSi) is a derivative of Sulindac, a NSAID known to inactivate Wnt signaling. We found that SSi downregulated both β-catenin and γ-catenin in X-RARα-expressing cells and reversed the leukemic phenotype by reducing stem cell capacity and increasing differentiation potential in X-RARα-positive HSCs. The data presented herein show that SSi inhibits the leukemic cell growth as well as hematopoietic progenitors cells (HPCs) expressing PML/RARα, and it indicates that Sulindac is a valid molecular therapeutic approach that should be further validated using in vivo leukemia models and in clinical settings.


Biochimica et Biophysica Acta | 2015

5-Lipoxygenase is a direct p53 target gene in humans.

Bianca Gilbert; Khalil Ahmad; Jessica Roos; Christoph Lehmann; Tomohiro Chiba; Sandra Ulrich-Rückert; Leonie Smeenk; Simon J. van Heeringen; Thorsten J. Maier; Bernd Groner; Dieter Steinhilber

The p53 tumor suppressor plays a critical role in cancer, and more than 50% of human tumors contain mutations or deletions of the TP53 gene. p53 can transactivate or repress target genes in response to diverse stress signals, such as transient growth arrest, DNA repair, cellular differentiation, senescence and apoptosis. Through an unbiased genome-wide ChIP-seq analysis, we have found that 5-lipoxygenase (ALOX5, 5-LO) which is a key enzyme of leukotriene (LT) biosynthesis, is a direct target gene of p53 and its expression is induced by genotoxic stress via actinomycin D (Act.D) or etoposide (Eto) treatment. 5-LO and LTs play a role in immunological diseases as well as in tumorigenesis and tumor growth. p53 binds to a specific binding site consisting of a complete p53 consensus-binding motif in ALOX5 intron G which is located about 64kbp downstream of the transcriptional start site. We confirmed the strong binding of p53 to the 5-LO target site in ChIP-qPCR experiments. Expression analyses by qRT-PCR and immunoblot further revealed that genotoxic stress induces the ALOX5 mRNA and protein expression in a p53-dependent manner. Knockdown of p53 in U2OS cells leads to a downregulation of 5-LO mRNA and protein expression. In addition, immunofluorescence and immunoprecipitation assays indicate the direct binding of 5-LO to p53 protein. Furthermore, we found that 5-LO can inhibit the transcriptional activity of p53 suggesting that 5-LO acts in a negative feedback loop to limit induction of p53 target genes.


Biochemical Pharmacology | 2017

Characterization of the molecular mechanism of 5-lipoxygenase inhibition by 2-aminothiazoles

Simon B.M. Kretschmer; Stefano Woltersdorf; Dominik Vogt; Felix F. Lillich; Michael Rühl; Michael Karas; Isabelle V. Maucher; Jessica Roos; Ann-Kathrin Häfner; Astrid Kaiser; Mario Wurglics; Manfred Schubert-Zsilavecz; Carlo Angioni; Gerd Geisslinger; Holger Stark; Dieter Steinhilber; Bettina Hofmann

Graphical abstract Figure. No Caption available. Abstract 5‐Lipoxygenase (5‐LO, EC1.13.11.34) has been implicated in the pathogenesis of inflammatory and immune diseases. Recently, aminothiazole comprising inhibitors have been discovered for this valuable target. Yet, the molecular mode of action of this class of substances is only poorly understood. Here, we present the detailed molecular mechanism of action of the compound class and the in vitro pharmacological profile of two lead compounds ST‐1853 and ST‐1906. Mechanistic studies with recombinant proteins as well as intact cell assays enabled us to define this class as a novel type of 5‐LO inhibitors with unique characteristics. The parent compounds herein presented a certain reactivity concerning oxidation and thiol binding: Unsubstituted aminophenols bound covalently to C159 and C418 of human 5‐LO. Yet, dimethyl substitution of the aminophenol prevented this reactivity and slowed down phase II metabolism. Both ST‐1853 and ST‐1906 confirmed their lead likeness by retaining their high potency in physiologically relevant 5‐LO activity assays, high metabolic stability, high specificity and non‐cytotoxicity.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Nitro-fatty acids are formed in response to virus infection and are potent inhibitors of STING palmitoylation and signaling.

Anne Louise Hansen; Gregory J. Buchan; Michael Rühl; Kojiro Mukai; Sonia R. Salvatore; Emari Ogawa; Sidsel D. Andersen; Marie B. Iversen; Anne L. Thielke; Camilla Gunderstofte; Mona Motwani; Charlotte T. Møller; Andreas S. Jakobsen; Katherine A. Fitzgerald; Jessica Roos; Rongtuan Lin; Thorsten J. Maier; Raphaela Goldbach-Mansky; Cathrine Miner; Wei Qian; Jonathan J. Miner; Rachel E. Rigby; Jan Rehwinkel; Martin R. Jakobsen; Hiroyuki Arai; Tomohiko Taguchi; Francisco J. Schopfer; David Olagnier; Christian K. Holm

Significance Several chronic inflammatory conditions have recently been shown to depend on abnormally high activity of the signaling protein stimulator of IFN genes (STING). These conditions include examples from systemic lupus erythematosus, Aicardi–Goutiéres syndrome, and STING-associated vasculopathy with onset in infancy. The involvement of STING in these diseases points to an unmet demand to identify inhibitors of STING signaling, which could form the basis of anti-STING therapeutics. With this report, we identify distinct endogenously formed lipid species as potent inhibitors of STING signaling—and propose that these lipids could have pharmaceutical potential for treatment of STING-dependent inflammatory diseases. The adaptor molecule stimulator of IFN genes (STING) is central to production of type I IFNs in response to infection with DNA viruses and to presence of host DNA in the cytosol. Excessive release of type I IFNs through STING-dependent mechanisms has emerged as a central driver of several interferonopathies, including systemic lupus erythematosus (SLE), Aicardi–Goutières syndrome (AGS), and stimulator of IFN genes-associated vasculopathy with onset in infancy (SAVI). The involvement of STING in these diseases points to an unmet need for the development of agents that inhibit STING signaling. Here, we report that endogenously formed nitro-fatty acids can covalently modify STING by nitro-alkylation. These nitro-alkylations inhibit STING palmitoylation, STING signaling, and subsequently, the release of type I IFN in both human and murine cells. Furthermore, treatment with nitro-fatty acids was sufficient to inhibit production of type I IFN in fibroblasts derived from SAVI patients with a gain-of-function mutation in STING. In conclusion, we have identified nitro-fatty acids as endogenously formed inhibitors of STING signaling and propose for these lipids to be considered in the treatment of STING-dependent inflammatory diseases.


Biochemical Pharmacology | 2017

Michael acceptor containing drugs are a novel class of 5-lipoxygenase inhibitor targeting the surface cysteines C416 and C418

Isabelle V. Maucher; Michael Rühl; Simon B.M. Kretschmer; Bettina Hofmann; Benjamin Kühn; Jasmin Fettel; Anja Vogel; Karsten T. Flügel; Georg Manolikakes; Nadine Hellmuth; Ann-Kathrin Häfner; Vahid Golghalyani; Ann-Katrin Ball; Matthias Piesche; Carmela Matrone; Gerd Geisslinger; Michael J. Parnham; Michael Karas; Dieter Steinhilber; Jessica Roos; Thorsten J. Maier

Graphical abstract Figure. No Caption available. ABSTRACT Recently, we published that nitro‐fatty acids (NFA) are potent electrophilic molecules which inhibit 5‐lipoxygenase (5‐LO) by interacting catalytically with cysteine residues next to a substrate entry channel. The electrophilicity is derived from an intramolecular Michael acceptor moiety consisting of an electron‐withdrawing group in close proximity to a double bond. The potential of the Michael acceptor moiety to interact with functionally relevant cysteines of proteins potentially renders them effective and sustained enzyme activity modulators. We screened a large library of naturally derived and synthetic electrophilic compounds to investigate whether other types of Michael acceptor containing drugs suppress 5‐LO enzyme activity. The activity was measured by assessing the effect on the 5‐LO product formation of intact human polymorphonuclear leukocytes. We demonstrated that a number of structurally different compounds were suppressive in the activity assays and showed that Michael acceptors of the quinone and nitro‐alkene group produced the strongest inhibition of 5‐LO product formation. Reactivity with the catalytically relevant cysteines 416 and 418 was confirmed using mutated recombinant 5‐LO and mass spectrometric analysis (MALDI‐MS). In the present study, we show for the first time that a number of well‐recognized naturally occurring or synthetic anti‐inflammatory compounds carrying a Michael acceptor, such as thymoquinone (TQ), the paracetamol metabolite NAPQI, the 5‐LO inhibitor AA‐861, and bardoxolone methyl (also known as RTA 402 or CDDO‐methyl ester) are direct covalent 5‐LO enzyme inhibitors that target the catalytically relevant cysteines 416 and 418.

Collaboration


Dive into the Jessica Roos's collaboration.

Top Co-Authors

Avatar

Dieter Steinhilber

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Thorsten J. Maier

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Martin Ruthardt

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Benjamin Kühn

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Claudia Oancea

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jasmin Fettel

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Hubert Serve

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Michael Rühl

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge