Network


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

Hotspot


Dive into the research topics where Anna Otte is active.

Publication


Featured researches published by Anna Otte.


Cell Communication and Signaling | 2012

Mesenchymal stem cells as all-round supporters in a normal and neoplastic microenvironment

Ralf Hass; Anna Otte

Mesenchymal stem cells (MSC) represent a heterogeneous population exhibiting stem cell-like properties which are distributed almost ubiquitously among perivascular niches of various human tissues and organs. Organismal requirements such as tissue damage determine interdisciplinary functions of resident MSC including self-renewal, migration and differentiation, whereby MSC support local tissue repair, angiogenesis and concomitant immunomodulation. However, growth of tumor cells and invasion also causes local tissue damage and injury which subsequently activates repair mechanisms and consequently, attracts MSC. Thereby, MSC exhibit a tissue-specific functional biodiversity which is mediated by direct cell-to-cell communication via adhesion molecule signaling and by a tightly regulated exchange of a multifactorial panel of cytokines, exosomes, and micro RNAs. Such interactions determine either tumor-promoting or tumor-inhibitory support by MSC. Moreover, fusion with necrotic/apoptotic tumor cell bodies contributes to re-program MSC into an aberrant phenotype also suggesting that tumor tissue in general represents different types of neoplastic cell populations including tumor-associated stem cell-like cells. The present work summarizes some functional characteristics and biodiversity of MSC and highlights certain controversial interactions with normal and tumorigenic cell populations, including associated modulations within the MSC microenvironment.


Stem Cells and Development | 2013

Mesenchymal Stem Cells Directly Interact with Breast Cancer Cells and Promote Tumor Cell Growth In Vitro and In Vivo

Katharina Mandel; Yuanyuan Yang; Axel Schambach; Silke Glage; Anna Otte; Ralf Hass

Cellular interactions were investigated between human mesenchymal stem cells (MSC) and human breast cancer cells. Co-culture of the two cell populations was associated with an MSC-mediated growth stimulation of MDA-MB-231 breast cancer cells. A continuous expansion of tumor cell colonies was progressively surrounded by MSC(GFP) displaying elongated cell bodies. Moreover, some MSC(GFP) and MDA-MB-231(cherry) cells spontaneously generated hybrid/chimeric cell populations, demonstrating a dual (green fluorescent protein+cherry) fluorescence. During a co-culture of 5-6 days, MSC also induced expression of the GPI-anchored CD90 molecule in breast cancer cells, which could not be observed in a transwell assay, suggesting the requirement of direct cellular interactions. Indeed, MSC-mediated CD90 induction in the breast cancer cells could be partially blocked by a gap junction inhibitor and by inhibition of the notch signaling pathway, respectively. Similar findings were observed in vivo by which a subcutaneous injection of a co-culture of primary MSC with MDA-MB-231(GFP) cells into NOD/scid mice exhibited an about 10-fold increased tumor size and enhanced metastatic capacity as compared with the MDA-MB-231(GFP) mono-culture. Flow cytometric evaluation of the co-culture tumors revealed more than 90% of breast cancer cells with about 3% of CD90-positive cells, also suggesting an MSC-mediated in vivo induction of CD90 in MDA-MB-231 cells. Furthermore, immunohistochemical analysis demonstrated an elevated neovascularization and viability in the MSC/MDA-MB-231(GFP)-derived tumors. Together, these data suggested an MSC-mediated growth stimulation of breast cancer cells in vitro and in vivo by which the altered MSC morphology and the appearance of hybrid/chimeric cells and breast cancer-expressing CD90(+) cells indicate mutual cellular alterations.


Orphanet Journal of Rare Diseases | 2014

In vitro and in vivo therapeutic approach for a small cell carcinoma of the ovary hypercalcaemic type using a SCCOHT-1 cellular model

Anna Otte; Finn Rauprich; Peter Hillemanns; Tjoung-Won Park-Simon; Juliane von der Ohe; Ralf Hass

BackgroundThe small cell ovarian carcinoma of the hypercalcemic type (SCCOHT) which preferably affects young women during regenerative age represents a rare and aggressive form of ovarian tumors with poor prognosis and lacks an efficient therapy.Methods and resultsIn vitro chemotherapy testing in a fluorescence assay using a recently developed cellular model from a recurrent SCCOHT revealed sensitivity for certain epothilones, methotrexate and topotecan whereas little if any cytotoxicity was observed with other chemotherapeutics including platin-based compounds. In particular, epothilone B demonstrated a high sensitivity in contrast to ixabepilone with only little detectable effects. Western blot and cell cycle analysis revealed that the epothilone B sensitivity was associated with increased Ser15 phosphorylation of p53, a significant G1 and G2 cell cycle accumulation and subsequent cell death in subG1 phase. Moreover, tubulinβ3 expression in SMARCA4/BRG1-defective SCCOHT-1 in contrast to other ovarian cancer cells was also affected during chemotherapy treatment. Increased extracellular Ca2+ levels further enhanced the epothilone B cytotoxicity in SCCOHT-1 cells. These in vitro effects were also confirmed in vivo in NOD/scid mouse xenografts demonstrating an attenuated tumor growth in epothilone B / Ca2+-treated mice. After 4d of subsequent treatment, the tumor sizes were reduced by about 90% as compared to continuously growing control tumors. In parallel, a hypercalcemia in control tumor-carrying mice was reverted to normal serum Ca2+ levels after epothilone B / Ca2+ therapy.ConclusionsTaken together, these data demonstrated anti-tumorigenic effects of epothilone B / Ca2+ in SCCOHT providing a focused therapeutic approach against this rare disease and arising recurrent tumors.


Stem Cell Research & Therapy | 2016

Conditioned umbilical cord tissue provides a natural three-dimensional storage compartment as in vitro stem cell niche for human mesenchymal stroma/stem cells

Yuanyuan Yang; Catharina Melzer; Juliane von der Ohe; Anna Otte; Ralf Hass

BackgroundThe use of large amounts of human multipotent mesenchymal stroma/stem cells (MSC) for cell therapies represents a desirable property in tissue engineering and banking in the field of regenerative medicine.Methods and resultsWhereas cryo-storage of umbilical cord (UC) tissue pieces in liquid nitrogen without ingredients was associated with predominant appearance of apoptotic cells after thawing and re-culture, progressive growth of MSC was observed following use of cryo-medium. Moreover, conditioning of UC tissue pieces by initial explant culture and subsequent cryo-storage with cryo-medium accelerated a further MSC culture after thawing. These findings suggested that conditioning of UC tissue pieces provides an in vitro stem cell niche by maintenance of a 3-dimensional natural microenvironment for continuous MSC outgrowth and expansion. Indeed, culture of GFP-labeled UC tissue pieces was accompanied by increased outgrowth of GFP-labeled cells which was accelerated in conditioned UC tissue after cryo-storage. Moreover, cryopreserved conditioned UC tissue pieces in cryo-medium after thawing and explant culture could be cryopreserved again demonstrating renewed MSC outgrowth after repeated thawing with similar population doublings compared to the initial explant culture. Flow cytometry analysis of outgrowing cells revealed expression of the typical MSC markers CD73, CD90, and CD105. Furthermore, these cells demonstrated little if any senescence and cultures revealed stem cell-like characteristics by differentiation along the adipogenic, chondrogenic and osteogenic lineages.ConclusionsExpression of MSC markers was maintained for at least 10 freeze/thaw/explant culture cycles demonstrating that repeated cryopreservation of conditioned UC tissue pieces provided a reproducible and enriched stem cell source.


Oncotarget | 2015

c-Met inhibitors attenuate tumor growth of small cell hypercalcemic ovarian carcinoma (SCCOHT) populations

Anna Otte; Finn Rauprich; Juliane von der Ohe; Yuanyuan Yang; Friedrich Kommoss; Friedrich Feuerhake; Peter Hillemanns; Ralf Hass

A cellular model (SCCOHT-1) of the aggressive small cell hypercalcemic ovarian carcinoma demonstrated constitutive chemokine and growth factor production including HGF. A simultaneous presence of c-Met in 41% SCCOHT-1 cells suggested an autocrine growth mechanism. Expression of c-Met was also observed at low levels in the corresponding BIN-67 cell line (6.5%) and at high levels in ovarian adenocarcinoma cells (NIH:OVCAR-3 (84.4%) and SK-OV-3 (99.3%)). Immunohistochemistry of c-Met expression in SCCOHT tumors revealed a heterogeneous distribution between undetectable levels and 80%. Further characterization of SCCOHT-1 and BIN-67 cells by cell surface markers including CD90 and EpCAM demonstrated similar patterns with differences to the ovarian adenocarcinoma cells. HGF stimulation of SCCOHT-1 cells was associated with c-Met phosphorylation at Tyr1349 and downstream Thr202/Tyr204 phosphorylation of p44/42 MAP kinase. This HGF-induced signaling cascade was abolished by the c-Met inhibitor foretinib. Cell cycle analysis after foretinib treatment demonstrated enhanced G2 accumulation and increasing apoptosis within 72 h. Moreover, the IC50 of foretinib revealed 12.4 nM in SCCOHT-1 cells compared to 411 nM and 481 nM in NIH:OVCAR-3 and SK-OV-3 cells, respectively, suggesting potential therapeutic effects. Indeed, SCCOHT-1 and BIN-67 tumor xenografts in NODscid mice exhibited an approximately 10-fold and 5-fold reduced tumor size following systemic application of foretinib, respectively. Furthermore, foretinib-treated tumors revealed a significantly reduced vascularization and little if any c-Met-mediated signal transduction. Similar findings of reduced proliferative capacity and declined tumor size were observed after siRNA-mediated c-Met knock-down in SCCOHT-1 cells demonstrating that in vivo inhibition of these pathways contributed to an attenuation of SCCOHT tumor growth.


Cell Communication and Signaling | 2011

Abolished adherence alters signaling pathways in phorbol ester-induced human U937 cells.

Anna Otte; Katharina Mandel; Gesche Reinstrom; Ralf Hass

Phorbol ester (TPA) treatment of human U937 myeloid leukemia cells is associated with increasing adherence and monocyte-like maturation whereby the role of β2 integrin-mediated attachment for subsequent growth properties and the differentiation program remains unclear. Here, stably-transfected U937 cells with a pMTH1 vector containing the β2 integrin gene of CD11b in antisense orientation (asCD11b-U937) demonstrated a significantly reduced proliferative capacity in contrast to control vector transfectants (pMTH1-U937) or wild-type U937 cells. Phorbol ester exposure induced adherence and growth arrest in more than 90% of pMTH1-U937 and wild-type U937 cells after 72 h. In contrast, TPA-treated asCD11b-U937 failed to attach and the proliferation continued in more than 30% of the cells. Moreover, increased apoptosis appeared in asCD11b-U937 after TPA induction in contrast to pMTH1-U937 cells. In addition, non-specific inhibition of adherence on an agarose surface demonstrated internucleosomal DNA fragmentation in both, pMTH1-U937 and asCD11b-U937 after TPA treatment indicating a functional relationship between abolished adherence, regulation of proliferation and induction of apoptosis. Western blot analysis revealed differences in the expression levels and altered phosphorylation patterns of Pyk-2, pp60src and p42/p44 MAP kinases between pMTH1-U937 and asCD11b-U937 following TPA exposure which was also substantiated by Pyk-2 immunoprecipitation. These findings suggested that induced adherence predominantly mediated by a functional CD11b/CD18 integrin in U937 cells is involved in the activation of downstream signaling kinases and contributes to cell cycle regulation and apoptosis during monocytic maturation.


International Journal of Oncology | 2014

Interference of Ca2+ with the proliferation of SCCOHT-1 and ovarian adenocarcinoma cells

Anna Otte; Finn Rauprich; Juliane von der Ohe; Peter Hillemanns; Ralf Hass

A recently established cellular model for the rare small cell carcinoma of the ovary hypercalcemic type (SCCOHT-1) was characterized in comparison to ovarian adenocarcinoma cells (NIH:OVCAR-3 and SK-OV-3). The different cancer populations exhibited a common sensitivity in acidic pH milieu and a continuous proliferation in alkaline medium of pH 8.0-9.0. In the presence of elevated Ca2+ concentrations, the ovarian cancer cells demonstrated a progressively reduced proliferation within 72 h in contrast to other tumor types such as breast cancer cells. This significant growth inhibition was calcium-specific since the proliferation was unaffected after culture of the ovarian cancer cells in the presence of similar concentrations of other cations. The Ca2+ effects on the ovarian cancer cells were associated with marked differences in the activation of intracellular signaling pathways including enhanced phosphorylation of the p42/44 MAP kinase (Thr202/Tyr204). Further analysis of the signaling pathway revealed a significantly enhanced Ca2+-dependent and p42/44 MAP kinase activation-mediated prostaglandin E2 (PGE2) production in SK-OV-3 and SCCOHT-1 and to a lesser extent in NIH:OVCAR-3 cells. Vice versa, exogenous PGE2 did not affect the proliferative capacity of the ovarian cancer cells and inhibition of the Ca2+-mediated MAP kinase activation did not abolish the Ca2+-mediated cytotoxicity. Collectively, these data suggest that multiple pathways are activated by exogenous Ca2+ in the different ovarian cancer cells, including a specific MAP kinase signaling cascade with subsequent PGE2 production and a parallel pathway for the induction of cell death.


Cell Communication and Signaling | 2012

Involvement of CD11b integrin in the alteration of metabolic factors after phorbol ester stimulation of human myeloid leukemia cells

Katharina Mandel; Anna Otte; Ralf Hass

Previous work has demonstrated that phorbol ester (TPA)-induced adherence of human U937 myeloid leukemia cells can be blocked upon down-modulation of the β2-integrin CD11b after stable transfection of U937 cells with a pMTH1 vector-containing the CD11b gene in antisense orientation (asCD11b-U937) [Otte et al., (2011)]. In the present study, alterations in metabolism-associated factors, particularly intra- and extracellular proteases were investigated. A measurement of telomerase activity in the leukemic cells revealed continuously decreasing telomere adducts within 72 h of TPA treatment in pMTH1-U937 cells. In contrast, telomerase activity sustained in asCD11b-U937 upon TPA-induced differentiation. Flow cytometric analysis confirmed unchanged CD11b levels in TPA-induced asCD11b-U937 in contrast to elevated levels in pMTH1-U937 whereby the expression of other β2-integrins including CD11a, CD11c and CD18 was increased in both populations after TPA treatment. Moreover, adherent pMTH1-U937 demonstrated the expression of monocytic differentiation markers including F4-80 and CD14 and an increased MIP-1α production which remained at low or undetectable in TPA-induced asCD11b-U937. These effects indicated an altered response of the different cell populations to the TPA-induced differentiation process. Indeed, Western blot analysis revealed differences in the expression levels of intracellular metabolic factors including MnSOD and p97/VCP and after measurement of 20 S proteasomal proteolytic activity. In addition, increased levels of extracellular metabolic factors including the matrix metalloproteinases MMP-1, MMP-7 and MMP-9 were observed in pMTH1-U937 cells in contrast to unaltered levels in asCD11b-U937 cells.


Tissue Engineering Part C-methods | 2013

Mesenchymal Stem Cells Maintain Long-Term In Vitro Stemness During Explant Culture

Anna Otte; Kerstin Reimers; Ralf Hass


International Journal of Oncology | 2015

Acquisition of new tumor cell properties by MSC-derived exosomes

Yuanyuan Yang; Bucan; Heike Baehre; von der Ohe J; Anna Otte; Ralf Hass

Collaboration


Dive into the Anna Otte's collaboration.

Top Co-Authors

Avatar

Ralf Hass

Hannover Medical School

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge