Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Dermatol Sci ; 74(2): 106-15, 2014 May.
Article in English | MEDLINE | ID: mdl-24507936

ABSTRACT

BACKGROUND: We have previously reported stromal upregulation of the endogenous angiogenesis inhibitor thrombospondin-2 (TSP-2) during multistep carcinogenesis, and we found accelerated and enhanced skin angiogenesis and carcinogenesis in TSP-2 deficient mice. GOALS: To investigate whether enhanced levels of TSP-2 might protect from skin cancer development. METHODS: We established transgenic mice with targeted overexpression of TSP-2 in the skin and subjected hemizygous TSP-2 transgenic mice and their wild-type littermates to a chemical skin carcinogenesis regimen. RESULTS: TSP-2 transgenic mice showed a significantly delayed onset of tumor formation compared to wild-type mice, whereas the ratio of malignant conversion to squamous cell carcinomas was comparable in both genotypes. Computer-assisted morphometric analysis of blood vessels revealed pronounced tumor angiogenesis already in the early stages of carcinogenesis in wild type mice. TSP-2 overexpression significantly reduced tumor blood vessel density in transgenic mice but had no overt effect on LYVE-1 positive lymphatic vessels. The percentage of desmin surrounded, mature tumor-associated blood vessels and the degree of epithelial differentiation remained unaffected. The antiangiogenic effect of transgenic TSP-2 was accompanied by a significantly increased number of apoptotic tumor cells in transgenic mice. CONCLUSION: Our results demonstrate that enhanced levels of TSP-2 in the skin result in reduced susceptibility to chemically-induced skin carcinogenesis and identify TSP-2 as a new target for the prevention of skin cancer.


Subject(s)
Carcinogenesis , Carcinoma, Squamous Cell/etiology , Skin Neoplasms/etiology , Thrombospondins/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Epidermis/metabolism , Epidermis/pathology , Female , Genotype , Mice , Mice, Transgenic , Neovascularization, Pathologic , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Thrombospondins/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
J Perinat Med ; 40(1): 63-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22848904

ABSTRACT

OBJECTIVE: To analyze the relationship of the time interval between two deliveries, done by one obstetric team, on the delivery mode of the subsequent birth; to define the length of this interval; and to evaluate this time interval as a risk factor for increased perinatal mortality in a population-based cohort. METHODS: All singleton deliveries at ≥ 24 weeks' gestation in Lower Saxony, Germany, between 2001 and 2005 (a total of 317,663 deliveries including 402 cases of perinatal mortality) were analyzed. The mode of the previous and the subsequent delivery, the time interval between the two deliveries, the time of birth, the hospital volume, and the existence of an affiliated neonatal ward were investigated. RESULTS: When the first vaginal delivery was <45 min, there was a reduced probability that the subsequent birth would be a cesarean section. In case of a previous cesarean section, the cesarean rate of the following birth was influenced up to 165 min. In a multivariate analysis, vaginal deliveries following an earlier vaginal birth and occurring within <45 min were associated with increased perinatal mortality. Repeated cesarean sections within <165 min were associated with increased perinatal mortality when occurring at night or on weekends. CONCLUSION: A short time interval between two deliveries in an obstetric unit constitutes an independent risk factor for perinatal mortality.


Subject(s)
Delivery, Obstetric/statistics & numerical data , Perinatal Mortality , Female , Humans , Pregnancy , Time Factors
3.
Breast Cancer Res Treat ; 130(3): 783-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21279682

ABSTRACT

Metastasis to bone is a frequent problem of advanced breast cancer. Particularly breast cancers, which do not express estrogen and progesterone receptors and which have no overexpression/amplification of the HER2-neu gene, so called triple-negative breast cancers, are considered as very aggressive and possess a bad prognosis. About 60% of all human breast cancers and about 74% of triple-negative breast cancers express receptors for gonadotropin-releasing hormone (GnRH), which might be used as a therapeutic target. Recently, we could show that bone-directed invasion of human breast cancer cells in vitro is time- and dose-dependently reduced by GnRH analogs. In the present study, we have analyzed whether GnRH analogs are able to reduce metastases of triple-negative breast cancers in vivo. In addition, we have evaluated the effects of GnRH analogs on tumor growth. To quantify formation of metastasis by triple-negative MDA-MB-435 and MDA-MB-231 human breast cancers, we used a real-time PCR method based on detection of human-specific alu sequences measuring accurately the amount of human tumor DNA in athymic mouse organs. To analyze tumor growth, the volumes of breast cancer xenotransplants into nude mice were measured. We could demonstrate that GnRH analogs significantly reduced metastasis formation by triple-negative breast cancer in vivo. In addition, we could show that GnRH analogs significantly inhibited the growth of breast cancer into nude mice. Side effects were not detectable. In conclusion, GnRH analogs seem to be suitable drugs for an efficacious therapy for triple-negative, GnRH receptor-positive human breast cancers to prevent metastasis formation.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Gonadotropin-Releasing Hormone/therapeutic use , Receptors, LHRH/agonists , Receptors, LHRH/antagonists & inhibitors , Alu Elements/genetics , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Breast Neoplasms/classification , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Gonadotropin-Releasing Hormone/analogs & derivatives , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Xenograft Model Antitumor Assays
4.
Breast Cancer Res Treat ; 128(2): 337-46, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20714802

ABSTRACT

Thus far the clinical benefits seen in breast cancer patients treated with drugs targeting the vascular endothelial growth factor (VEGF) pathway are only modest. Consequently, additional antiangiogenic approaches for treatment of breast cancer need to be investigated. Thrombospondin-2 (TSP-2) has been shown to inhibit tumor growth and angiogenesis with a greater potency than the related molecule TSP-1. The systemic effects of TSP-2 on tumor metastasis and the underlying molecular mechanisms of the antiangiogenic activity of TSP-2 have remained poorly understood. We generated a recombinant fusion protein consisting of the N-terminal region of TSP-2 and the IgG-Fc1 fragment (N-TSP2-Fc) and could demonstrate that the antiangiogenic activity of N-TSP2-Fc is dependent on the CD36 receptor. We found that N-TSP2-Fc inhibited VEGF-induced tube formation of human dermal microvascular endothelial cells (HDMEC) on matrigel in vitro and that concurrent incubation of anti-CD36 antibody with N-TSP2-Fc resulted in tube formation that was comparable to untreated control. N-TSP2-Fc potently induced apoptosis of HDMEC in vitro in a CD36-dependent manner. Moreover, we could demonstrate a CD36 receptor-mediated loss of mitochondrial membrane potential and activation of caspase-3 in HDMEC in vitro. Daily intraperitoneal injections of N-TSP2-Fc resulted in a significant inhibition of the growth of human MDA-MB-435 and MDA-MB-231 tumor cells grown in the mammary gland of immunodeficient nude mice and in reduced tumor vascularization. Finally, increased serum concentrations of N-TSP2-Fc significantly inhibited regional metastasis to lymph nodes and distant metastasis to lung as shown by quantitative real-time alu PCR. These results identify N-TSP2-Fc as a potent systemic inhibitor of tumor metastasis and provide strong evidence for an important role of the CD36 receptor in mediating the antiangiogenic activity of TSP-2.


Subject(s)
Apoptosis , Breast Neoplasms/prevention & control , CD36 Antigens/metabolism , Lung Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Recombinant Fusion Proteins/metabolism , Thrombospondins/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Blotting, Western , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , CD36 Antigens/genetics , Caspase 3/metabolism , Cell Adhesion , Cell Movement , Cell Proliferation , Dermis/cytology , Dermis/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Membrane Potential, Mitochondrial , Mice , Mice, Nude , Recombinant Fusion Proteins/genetics , Thrombospondins/genetics , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
5.
Am J Pathol ; 165(2): 541-52, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277228

ABSTRACT

In the present study, the type-1 repeats of thrombospondin-1 (TSP-1) were transfected into A431 cells. Expression of all three type-1 repeats (3TSR) and expression of just the second type-1 repeat containing the transforming growth factor (TGF)-beta activating sequence KRFK (TSR2 + KRFK) significantly inhibited in vivo tumor angiogenesis and growth in nude mice. These tumors expressed increased levels of both active and total TGF-beta. A431 cells expressing the second type-1 repeat without the KRFK sequence (TSR2 - KRFK) produced tumors that were slightly larger than the 3TSR and TSR2 + KRFK tumors. These tumors expressed elevated levels of active TGF-beta but levels of total TGF-beta were not different from control tumors. Injection of the peptide, LSKL, which blocks TSP-1 activation of TGF-beta, reversed the growth inhibition observed with cells expressing TSR2 + KRFK to a level comparable to controls. Various residues in the WSHWSPW region and the VTCG sequence of both TSR2+/- KRFK were mutated. Although mutation of the VTCG sequence had no significant effect on tumor growth, mutation of the WSHWSPW sequence reduced inhibition of tumor growth. These findings suggest that the inhibition of tumor angiogenesis and growth by endogenous TSP-1 involves regulation of both active and total TGF-beta and the sequences KRFK and WSHWSPW in the second type-1 repeat.


Subject(s)
Carcinoma, Squamous Cell/pathology , Gene Expression/physiology , Lung Neoplasms/pathology , Thrombospondin 1/genetics , Transforming Growth Factor beta/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Squamous Cell/blood supply , Humans , Luciferases/metabolism , Lung Neoplasms/blood supply , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Neovascularization, Pathologic/prevention & control , Peptide Fragments/genetics , Repetitive Sequences, Nucleic Acid , Survival Rate , Transfection
6.
Oncogene ; 21(52): 7945-56, 2002 Nov 14.
Article in English | MEDLINE | ID: mdl-12439745

ABSTRACT

The roles played by the endogenous angiogenesis inhibitor thrombospondin-1 (TSP-1) in the early stages of multi-step carcinogenesis and in the control of hematogenous versus lymphatic metastasis are unknown. To investigate these issues we compared tumor development in normal mice and in transgenic mice with targeted overexpression of TSP-1 in the epidermis following a standard two-step chemical skin carcinogenesis regimen. Overexpression of TSP-1 resulted in delayed and reduced development of premalignant epithelial hyperplasias, but did not inhibit the malignant conversion to squamous cell carcinomas. TSP-1 overexpression also suppressed tumor angiogenesis and distant organ metastasis, but failed to inhibit tumor-associated lymphangiogenesis or lymphatic tumor spread to regional lymph nodes. Concomitant with these results, we found that the endothelial TSP-1 receptor CD36 was mostly absent from cutaneous lymphatic vessels. Our findings indicate the potential use of TSP-1 for the prevention of premalignant stages of tumorigenesis and are likely to have implications for the further development of anti-angiogenic cancer therapies.


Subject(s)
Cell Transformation, Neoplastic , Lymphatic Metastasis/prevention & control , Neovascularization, Pathologic/prevention & control , Thrombospondin 1/physiology , Animals , Mice , Mice, Transgenic , Skin Neoplasms/blood supply , Skin Neoplasms/pathology
7.
Cancer Res ; 62(7): 2004-12, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11929817

ABSTRACT

Recent studies indicate that continuous administration improves the antitumoral efficacy of angiogenesis inhibitors, as compared with intermittent dosing, suggesting a potential role of gene therapy in antiangiogenic tumor therapy. We established a tissue-engineered implant system for the continuous in vivo production of thrombospondin-2 (TSP-2), a potent endogenous inhibitor of tumor growth and angiogenesis. Fibroblasts were retrovirally transduced to overexpress TSP-2 and were seeded onto biodegradable polymer scaffolds. After transplantation into the peritoneal cavity of nude mice, bioimplants maintained high levels of TSP-2 secretion over extended time periods, resulting in increased levels of circulating TSP-2. Bioimplant-generated TSP-2 potently inhibited tumor growth and angiogenesis of human squamous cell carcinomas, malignant melanomas, and Lewis lung carcinomas that were implanted at a distant site. These results provide the first proof-of-principle for the feasibility and therapeutic efficiency of systemic, cell-based antiangiogenic gene therapy using biodegradable polymer grafts for the treatment of cancer.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Genetic Therapy/methods , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/therapy , Thrombospondins/genetics , 3T3 Cells/metabolism , 3T3 Cells/transplantation , Animals , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cell Division/genetics , Cell Transplantation/methods , Humans , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Polymers , Prostheses and Implants , Retroviridae/genetics , Thrombospondins/biosynthesis , Thrombospondins/metabolism , Transfection , Tumor Cells, Cultured
8.
Am J Pathol ; 160(4): 1381-92, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11943723

ABSTRACT

The distinct roles of angiopoietin (Ang)-1 and Ang2, counteracting ligands for the endothelium-specific Tie2 receptor, in tumor development and progression have remained poorly understood. We investigated the expression of Ang1 and Ang2 during multistep mouse skin carcinogenesis and in human squamous cell carcinoma (SCC) xenografts. Expression of Ang2, but not of Ang1, was up-regulated in angiogenic tumor vessels already in early stages of skin carcinogenesis and was also strongly increased in SCCs. Stable overexpression of Ang1 in human A431 SCCs resulted in a more than 70% inhibition of tumor growth, associated with enhanced Tie2 phosphorylation levels, as compared with low levels in control transfected tumors. No major changes in the vascular density, vascular endothelial growth factor mRNA and protein expression, and vascular endothelial growth factor receptor-2 phosphorylation levels were observed in Ang1-expressing tumors. However, the fraction of tumor blood vessels with coverage by alpha-smooth muscle actin-positive periendothelial cells was significantly increased, indicative of an increased vascular maturation status. These findings identify an inhibitory role of Ang1/Tie2 receptor-mediated vessel maturation in SCC growth and suggest that up-regulation of its antagonist, Ang2, during early-stage epithelial tumorigenesis contributes to the angiogenic switch by counteracting specific vessel-stabilizing effects of Ang1.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Membrane Glycoproteins/physiology , Neoplasm Proteins/physiology , Neovascularization, Pathologic/physiopathology , Proto-Oncogene Proteins , Angiopoietin-1 , Angiopoietin-2 , Animals , Apoptosis , Blood Vessels/growth & development , Carcinoma, Squamous Cell/physiopathology , Cell Division/physiology , Endothelial Growth Factors/metabolism , Humans , Lymphokines/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Proteins/physiology , Receptor, TIE-2 , Tumor Cells, Cultured , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...