44 research outputs found

    Gentherapie : Etablierung und Optimierung retroviraler Vektoren für den T-Zell-spezifischen Gentransfer

    Get PDF
    Das Ziel von Gentherapie ist die Behandlung bzw. Heilung einer Erkrankung durch das Einbringen eines oder mehrerer Gene. Dazu werden Gentransfervektoren benötigt, die effizient therapeutische Gene in die zu behandelnden Zellen einbringen. Für eine systemische Applikation müssen Gentransfervektoren die Eigenschaft besitzen, ausschließlich die erkrankten Zellen zu transduzieren. Der Tropismus retroviraler Vektoren wird durch das Envelopeprotein (Env) festgelegt. Maus Leukämie Virus (MLV) basierende Kapsidpartikel können mit dem Envelopeprotein des humanen Immundefizienzvirus Typ-1 (HIV-1) pseudotypisiert werden. Diese MLV/HIV-1 Pseudotypvektoren besitzen einen Tropismus für humane CD4 positive T-Helferzellen. Diese Vektoren sind geeigneten Kandidaten für die gen-therapeutische Behandlung der HIV-lnfektion und von kutanen T-Zell Lymphomen, einer lymphproliferativen Erkrankung von CD4 Zellen. Im ersten Teil dieser Arbeit wurden MLV/HIV Pseudotypvektoren exprimierende Verpackungszelllinien mit Hüllproteinen verschiedener Subtypen etabliert und charakterisiert. Die verwendeten Subtypen waren das T-trophe HIV-1 Isolat BH10, das T-trophe HIV-2 Isolat ISY und das dualotrophe SHIV Isolat 89.6P. Dabei zeigte sich eine Abhängigkeit der Vektortiter vom Subtyp. Die höchsten Titer wurden mit der MLV/HIV-1 Pseudotypvektoren exprimierenden Verpackungszelllinie FLY-HIV-87 erhalten und lagen in Abhängigkeit vom retroviralen Vektor zwischen 1 x 10 hoch 5 und 1 x 10 hoch 6 IU/ml. Die Transduktionsspezifität entsprach dem Korezeptorgebrauch des HIV-Subtyps. Da für die geplanten in vivo Experimente höhere Titer notwendig waren, wurden verschiedene Methoden zur Anreicherung MLV/HIV-1 pseudotypisierter Vektoren zunächst getestet, sowie die Beste dieser Methoden für die Konzentrierung der Partikel optimiert. Im zweiten Teil dieser Arbeit wurde an zwei Mausmodellen die in vivo Applikation MLV/HIV-1 pseudotypisierter Vektoren untersucht. An transgenen hCD4 Mäusen wurde der Gentransfer nach systemischer Applikation von MLV/HIV-1 LacZ Pseudotypvektoren untersucht. In den transduzierten Mäusen konnte Gentransfer in Lymphknoten und Thymus beobachtet werden. Durch subkutane Implantation der humanen kutanen T-Zell Lymphomzellen MyLa in Nacktmäuse wurde ein Tiermodell Modell für humane kutane T-Zell Lymphome etabliert. Die intratumorale Applikation von MLV/HIV-1 Partikeln, deren Vektorgenom für das grüne Fluoreszenzprotein (EGFP) kodiert ergab, daß es zum effektiven und spezifischen Gentransfer in die CD4 positiven MyLa Zellen kam. In dem anschließend durchgeführte Therapieversuch mit Herpes Simplex Virus Thymidinkinase kodierenden Vektoren und darauf folgender systemischer Ganciclovir Behandlung konnte eine Verlangsamung des Tumorwachstums erzielt werden Die Ergebnisse der vorliegenden Arbeit haben gezeigt, daß MLV/HIV-1 Pseudotypvektoren für den spezifischen und effizienten Transfer von Genen in primäre humane CD4 T-Helferzellen geeignet sind und daß sowohl die systemische als auch die intratumorale Applikation dieser Vektoren möglich ist

    RASSF1A-Mediated Suppression of Estrogen Receptor Alpha (ERα)-Driven Breast Cancer Cell Growth Depends on the Hippo-Kinases LATS1 and 2

    Get PDF
    Around 70% of breast cancers express the estrogen receptor alpha (ERα). This receptor is of central importance for breast cancer development and estrogen-dependent tumor growth. However, the molecular mechanisms that are responsible for the control of ERα expression and function in the context of breast carcinogenesis are complex and not fully understood. In previous work, we have demonstrated that the tumor suppressor RASSF1A suppresses estrogen-dependent growth of breast cancer cells through a complex network that keeps ERα expression and function under control. We observed that RASSF1A mediates the suppression of ERα expression through modulation of the Hippo effector Yes-associated protein 1 (YAP1) activity. Here we report that RASSF1A-mediated alteration of YAP1 depends on the Hippo-kinases LATS1 and LATS2. Based on these results, we conclude that inactivation of RASSF1A causes changes in the function of the Hippo signaling pathway and altered activation of YAP1, and as a consequence, increased expression and function of ERα. Thus, the inactivation of RASSF1A might constitute a fundamental event that supports the initiation of ERα-dependent breast cancer. Furthermore, our results support the notion that the Hippo pathway is important for the suppression of luminal breast cancers, and that the tumor-suppressor function of RASSF1A depends on LATS1 and LATS2

    Assessment of HIV-1 entry inhibitors by MLV/HIV-1 pseudotyped vectors

    Get PDF
    BACKGROUND: Murine leukemia virus (MLV) vector particles can be pseudotyped with a truncated variant of the human immunodeficiency virus type 1 (HIV-1) envelope protein (Env) and selectively target gene transfer to human cells expressing both CD4 and an appropriate co-receptor. Vector transduction mimics the HIV-1 entry process and is therefore a safe tool to study HIV-1 entry. RESULTS: Using FLY cells, which express the MLV gag and pol genes, we generated stable producer cell lines that express the HIV-1 envelope gene and a retroviral vector genome encoding the green fluorescent protein (GFP). The BH10 or 89.6 P HIV-1 Env was expressed from a bicistronic vector which allowed the rapid selection of stable cell lines. A codon-usage-optimized synthetic env gene permitted high, Rev-independent Env expression. Vectors generated by these producer cells displayed different sensitivity to entry inhibitors. CONCLUSION: These data illustrate that MLV/HIV-1 vectors are a valuable screening system for entry inhibitors or neutralizing antisera generated by vaccines

    RASSF1A Suppresses Estrogen-Dependent Breast Cancer Cell Growth through Inhibition of the Yes-Associated Protein 1 (YAP1), Inhibition of the Forkhead Box Protein M1 (FOXM1), and Activation of Forkhead Box Transcription Factor 3A (FOXO3A)

    Get PDF
    The estrogen receptor alpha (ERα) is expressed by the majority of breast cancers and plays an important role in breast cancer development and tumor outgrowth. Although ERα is well known to be a specific and efficient therapeutic target, the molecular mechanisms that are responsible for the control of ERα expression and function in the context of breast cancer initiation and progression are complex and not completely elucidated. In previous work, we have demonstrated that the tumor suppressor RASSF1A inhibits ERα expression and function in ERα-positive breast cancer cells through an AKT-dependent mechanism. Transcriptional activators such as forkhead box protein M1 (FOXM1) and forkhead transcription factor 3A (FOXO3A) and signaling pathways such as the Hippo pathway are also known to modulate ERα expression and activity. Here we report that RASSF1A acts as an inhibitor of ERα-driven breast cancer cell growth through a complex, hierarchically organized network that initially involves suppression of the Hippo effector Yes-associated protein 1 (YAP1), which is followed by inhibition of AKT1 activity, increased FOXO3A activity as well as a blockade of FOXM1 and ERα expression. Together our findings provide important new mechanistic insights into how the loss of RASSF1A contributes to ERα+ breast cancer initiation and progression

    Detection of Cellular Senescence Reveals the Existence of Senescent Tumor Cells within Invasive Breast Carcinomas and Related Metastases

    Get PDF
    Oncogene-induced senescence is thought to constitute a barrier to carcinogenesis by arresting cells at risk of malignant transformation. However, numerous findings suggest that senescent cells may conversely promote tumor growth and metastatic progression, for example, through the senescence-associated secretory phenotype (SASP) they produce. Here, we investigated the degree to which senescent tumor cells exist within untreated human primary breast carcinomas and whether the presence of senescent cancer cells in primary tumors is recapitulated in their matched lymph node metastases. For the detection of senescence, we used SA-β-galactosidase (SA-β-gal) staining and other senescence markers such as Ki67, p21, p53, and p16. In patients with invasive luminal A and B breast carcinomas, we found broad similarities in the appearance of cancer cells between primary tumors and their corresponding metastases. Analysis of lymph nodes from patients with other breast cancer subtypes also revealed senescent tumor cells within metastatic lesions. Collectively, our findings show that senescent tumor cells exist within primary breast carcinomas and metastatic lesions. These results suggest a potential role for senescent breast tumor cells during metastatic progression and raise the question as to whether the targeting of senescent tumor cells with anti-senescent drugs might represent a novel avenue for improved treatment of breast and other cancers

    Functional characterization of circulating tumor cells (CTCs) from metastatic ER+/HER2− breast cancer reveals dependence on HER2 and FOXM1 for endocrine therapy resistance and tumor cell survival: Implications for treatment of ER+/HER2− breast cancer

    Get PDF
    Mechanisms of acquired endocrine resistance and late recurrence in patients with ER+/HER2− breast cancer are complex and not fully understood. Here, we evaluated mechanisms of acquired resistance in circulating tumor cells (CTCs) from an ER+/HER2− breast cancer patient who initially responded but later progressed under endocrine treatment. We found a switch from ERα-dependent to HER2-dependent and ERα-independent expression of FOXM1, which may enable disseminated ER+/HER2− cells to re-initiate tumor cell growth and metastasis formation in the presence of endocrine treatment. Our results also suggest a role for HER2 in resistance, even in ER+ breast cancer cells that have neither HER2 amplification nor activating HER2 mutations. We found that NFkB signaling sustains HER2 and FOXM1 expression in CTCs in the presence of ERα inhibitors. Inhibition of NFkB signaling blocked expression of HER2 and FOXM1 in the CTCs, and induced apoptosis. Thus, targeting of NFkB and FOXM1 might be an efficient therapeutic approach to prevent late recurrence and to treat endocrine resistance. Collectively our data show that CTCs from patients with endocrine resistance allow mechanisms of acquired endocrine resistance to be delineated, and can be used to test potential drug regimens for combatting resistance

    Proteasome inhibitors prevent bi-directional HER2/estrogen-receptor cross-talk leading to cell death in endocrine and lapatinib-resistant HER2+/ER+ breast cancer cells

    Get PDF
    Amplification and/or overexpression of the human epidermal growth factor 2 (HER2) oncogene occurs in about 13–15% of invasive breast cancer and triggers breast cancer cell proliferation, survival and metastatic progression. Around half of all breast cancers with HER2 overexpression co-express hormone receptors (HR) such as those for estrogen and progesterone. Aberrant signaling through HER2 and other members of the HER-family mediates endocrine-resistance in estrogen receptor alpha (ERα) positive breast cancer. On the other hand, ERα co-expression has been shown to attenuate the efficiency of anti-HER2 therapies. These findings indicate that HER2 and ERα synergize to escape from both anti-ERα and anti-HER2-targeted therapies. Rationally designed clinical trials that combine endocrine therapy with anti-HER2 agents to interfere with HER2/ERα cross-talk have been conducted. However, the outcome of these trials suggests that novel therapeutic approaches are needed to further improve inhibition of HER2 and other HER-family members in conjunction with a more efficient ERα blockade. Here, we demonstrate that carfilzomib and bortezomib stabilize the HER2-specific protein tyrosine phosphatase BDP1 leading to decreased HER2 autophosphorylation, reduced HER2 activity and subsequently attenuated activation of the PI3K/Akt-pathway, together with blockade of ERα expression. We further observed that proteasome inhibitors (PIs) reverse autophosphorylation and thereby inhibit the activity of constitutively active mutant HER2. We also demonstrate that PIs cause cell death in lapatinib and endocrine-resistant HER2+/ER+ breast cancer cells. These findings suggest that PIs might have the potential to improve the management of HER2+/ER+ breast cancer patients by efficiently disrupting the bi-directional HER2/ERα cross-talk

    \u27Normalizing\u27 the malignant phenotype of luminal breast cancer cells via alpha(v)beta(3)-integrin

    Get PDF
    Reestablishing tissue organization of breast cancer cells into acini was previously shown to override their malignant phenotype. In our study, we demonstrate that alpha(v)beta(3) integrin (Int-αvβ3), previously shown to play a role in cancer progression, promoted differentiation and growth arrest of organoids derived from luminal A breast cancer cells grown in their relevant three-dimensional microenvironment. These organoids differentiated into normal-like acini resembling a benign stage of breast tissue. Likewise, we demonstrate that Int-αvβ3 is selectively expressed in the epithelium of the benign stage of breast tissues, and is lost during the early stages of luminal A breast cancer progression. Notably, the organoids’ reversion into normal-like acini was mediated by cancer luminal progenitor-like cells expressing both EpCAMhigh^{high}CD49flow^{low}CD24+^{+} and Int-αvβ3. Furthermore, downregulation of Notch4 expression and downstream signaling was shown to mediate Int-αvβ3-induced reversion. Intriguingly, when luminal A breast cancer cells expressing Int-αvβ3 were injected into a humanized mouse model, differentiated tumors developed when compared with that generated by control cells. Hence, our data suggest that promoting differentiation of luminal A breast cancer cells by signaling emanating from Int-αvβ3 can potentially promote ‘normalization’ of their malignant phenotype and may prevent the malignant cells from progressing
    corecore