Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Cell ; 19(1): 31-44, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21215706

ABSTRACT

Polarization of tumor-associated macrophages (TAMs) to a proangiogenic/immune-suppressive (M2-like) phenotype and abnormal, hypoperfused vessels are hallmarks of malignancy, but their molecular basis and interrelationship remains enigmatic. We report that the host-produced histidine-rich glycoprotein (HRG) inhibits tumor growth and metastasis, while improving chemotherapy. By skewing TAM polarization away from the M2- to a tumor-inhibiting M1-like phenotype, HRG promotes antitumor immune responses and vessel normalization, effects known to decrease tumor growth and metastasis and to enhance chemotherapy. Skewing of TAM polarization by HRG relies substantially on downregulation of placental growth factor (PlGF). Besides unveiling an important role for TAM polarization in tumor vessel abnormalization, and its regulation by HRG/PlGF, these findings offer therapeutic opportunities for anticancer and antiangiogenic treatment.


Subject(s)
Down-Regulation/genetics , Macrophages/immunology , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic/immunology , Pregnancy Proteins/metabolism , Proteins/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotactic Factors/metabolism , Clodronic Acid/pharmacology , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Humans , Hypoxia/genetics , Hypoxia/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microvessels/drug effects , Microvessels/pathology , Microvessels/ultrastructure , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Placenta Growth Factor , Pregnancy Proteins/genetics , Pregnancy Proteins/immunology , Proteins/genetics , Proteins/pharmacology
2.
Arterioscler Thromb Vasc Biol ; 31(4): 741-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21205984

ABSTRACT

OBJECTIVE: The role of semaphorins in tumor progression is still poorly understood. In this study, we aimed at elucidating the regulatory role of semaphorin 3A (SEMA3A) in primary tumor growth and metastatic dissemination. METHODS AND RESULTS: We used 3 different experimental approaches in mouse tumor models: (1) overexpression of SEMA3A in tumor cells, (2) systemic expression of SEMA3A following liver gene transfer in mice, and (3) tumor-targeted release of SEMA3A using gene modified Tie2-expressing monocytes as delivery vehicles. In each of these experimental settings, SEMA3A efficiently inhibited tumor growth by inhibiting vessel function and increasing tumor hypoxia and necrosis, without promoting metastasis. We further show that the expression of the receptor neuropilin-1 in tumor cells is required for SEMA3A-dependent inhibition of tumor cell migration in vitro and metastatic spreading in vivo. CONCLUSIONS: In sum, both systemic and tumor-targeted delivery of SEMA3A inhibits tumor angiogenesis and tumor growth in multiple mouse models; moreover, SEMA3A inhibits the metastatic spreading from primary tumors. These data support the rationale for further investigation of SEMA3A as an anticancer molecule.


Subject(s)
Breast Neoplasms/therapy , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Lung Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Semaphorin-3A/metabolism , Stem Cells/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Necrosis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neuropilin-1/metabolism , Paracrine Communication , RNA Interference , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Recombinant Fusion Proteins/metabolism , Semaphorin-3A/genetics , Signal Transduction , Stromal Cells/metabolism , Time Factors , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
3.
Cancer Res ; 66(4): 2089-97, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16489009

ABSTRACT

Histidine-rich glycoprotein (HRGP) is an abundant heparin-binding plasma protein that efficiently arrests growth and vascularization of mouse tumor models. We have shown that the antiangiogenic effect of HRGP is dependent on its histidine/proline-rich domain, which needs to be released from the mother protein to exert its effects. Here we identify a 35-amino-acid peptide, HRGP330, derived from the histidine/proline-rich domain as endowed with antiangiogenic properties in vitro and in vivo. The mechanism of action of HRGP330 involves subversion of focal adhesion function by disruption of integrin-linked kinase (ILK) and focal adhesion kinase (FAK) functions, inhibition of vascular endothelial growth factor (VEGF)-induced tyrosine phosphorylation of the FAK substrate alpha-actinin, and, as a consequence, an arrest in endothelial cell motility. The disturbed focal adhesion function is reflected in the ability of HRGP as well as of HRGP330 to prevent endothelial cell adhesion to vitronectin in a manner involving alpha(v)beta3 integrin. In conclusion, HRGP330, which we define as the minimal antiangiogenic domain of HRGP, exerts its effects through signal transduction targeting focal adhesions, thereby interrupting VEGF-induced endothelial cell motility.


Subject(s)
Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Proteins/pharmacology , Actinin/metabolism , Amino Acid Sequence , Animals , Cattle , Cell Movement/drug effects , Cell Movement/physiology , Endothelial Cells/cytology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alphaVbeta3/metabolism , Molecular Sequence Data , Paxillin/antagonists & inhibitors , Paxillin/biosynthesis , Peptide Fragments/chemistry , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proteins/chemistry , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
J Virol Methods ; 133(1): 48-52, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16297456

ABSTRACT

Recovery of infectious Equine arteritis virus (EAV) from the semen of persistently infected Swedish stallions was attempted by classical cell culture isolation and by transfection of extracted total RNA. Whereas virus from semen samples stored for several months at -20 degrees C or from extended semen could only be recovered by transfection of extracted RNA, isolation in cell culture was achieved readily with fresh, unextended semen stored at -70 degrees C or directly used after sampling. In parallel, the viruses were examined in the variable region of the large glycoprotein GP5 by nested RT-PCR and direct nucleotide sequencing. The resulting sequences were placed into a large phylogenetic tree from this region, demonstrating that Swedish strains belonged to very diverse phylogenetic groups. This represents the first report of recovery of infectious EAV from archived semen samples by RNA transfection.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/isolation & purification , Horse Diseases/virology , RNA, Viral/analysis , Semen/virology , Animals , Arterivirus Infections/virology , Cell Line , Cricetinae , Horses , Male , Phylogeny , RNA, Viral/genetics , Rabbits , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Analysis, RNA , Sweden/epidemiology , Transfection
5.
Cancer Res ; 64(24): 9012-7, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15604266

ABSTRACT

Endostatin constitutes the COOH-terminal 20,000 Da proteolytic fragment of collagen XVIII and has been shown to possess antiangiogenic and antitumorigenic properties. In the present study, we have investigated the role of the heparin-binding sites in the in vivo mechanism of action of endostatin. The majority of the heparin binding is mediated by arginines 155/158/184/270 in endostatin, but there is also a minor site constituted by arginines 193/194. Using endostatin mutants lacking either of these two sites, we show that inhibition of fibroblast growth factor-2-induced angiogenesis in the chicken chorioallantoic membrane requires both heparin-binding sites. In contrast, inhibition of vascular endothelial growth factor-A-induced chorioallantoic membrane angiogenesis by endostatin was only dependent on the minor heparin-binding site (R193/194). These arginines were also required for endostatin to inhibit fibroblast growth factor-2- and vascular endothelial growth factor-A-induced chemotaxis of primary endothelial cells. Moreover, we show that a synthetic peptide corresponding to amino acids 180-199 of human endostatin (which covers the minor heparin-binding site) inhibits endothelial cell chemotaxis and reduces tumor vascularization in vivo. Substitution of arginine residues 193/194 for alanine attenuates the antiangiogenic effects of the peptide. These data show an essential role for heparin binding in the antiangiogenic action of endostatin.


Subject(s)
Endostatins/pharmacology , Fibrosarcoma/blood supply , Heparin/metabolism , Pancreatic Neoplasms/blood supply , Amino Acid Sequence , Animals , Binding Sites , Cattle , Chemotaxis/drug effects , Chick Embryo , Chorioallantoic Membrane/blood supply , Endostatins/genetics , Endostatins/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/pharmacology
6.
Cancer Res ; 64(2): 599-605, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14744774

ABSTRACT

In this study, we show that recombinant human histidine-rich glycoprotein (HRGP) has potent antiangiogenic properties as judged from effects on a syngeneic tumor model in C57/bl6 mice. Growth of fibrosarcoma, a very aggressive tumor, was reduced by >60% by HRGP treatment, and tumor angiogenesis was dramatically decreased. Treatment with HRGP led to increased apoptosis and reduced proliferation in the tumors. In contrast, HRGP did not affect apoptosis or DNA synthesis in endothelial cells or tumor cells in vitro. The mechanism of action of HRGP involves rearrangement of focal adhesions and decreased attachment of endothelial cells to vitronectin and, as a consequence, reduced endothelial cell migration. By using truncated versions of HRGP, we demonstrate that the isolated 150 amino acid-residue His/Pro-rich domain, which is also released by spontaneous proteolysis from purified HRGP, mediates the inhibitory effect on chemotaxis. Moreover, the His/Pro-rich domain must be released from HRGP to exert its effect. This study shows for the first time inhibitory effects of HRGP on tumor vascularization in vivo, thus providing proof of concept that HRGP is an angiogenesis inhibitor.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bone Neoplasms/blood supply , Endothelium, Vascular/physiology , Fibrosarcoma/blood supply , Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Peptide Fragments/pharmacology , Proteins/pharmacology , Animals , Aorta , Bone Neoplasms/drug therapy , Cell Adhesion/drug effects , Disease Models, Animal , Endothelium, Vascular/drug effects , Fibrosarcoma/drug therapy , Humans , Mice , Mice, Inbred C57BL , Proteins/chemistry , Recombinant Proteins/pharmacology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...