Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 191
Filter
Add more filters

Country/Region as subject
Publication year range
1.
ESMO Open ; 9(8): 103665, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39121813

ABSTRACT

BACKGROUND: Physical activity (PA) reduces the risk of developing breast cancer (BC) and mortality rate in BC patients starting PA after diagnosis. Immunomodulation is considered responsible for these effects. However, limited data exist on the immunomodulation induced by moderate PA (mPA) during neoadjuvant chemotherapy (NACT). We have investigated the longitudinal change of cytokines during NACT alone or combined with mPA. MATERIALS AND METHODS: Twenty-three cytokines were analyzed in BC patients at consecutive timepoints: at baseline (T0), before starting mPA (T1), before surgery (T2), and after surgery (T3). mPA consisted of 3-weekly brisk-walking sessions for 9-10 consecutive weeks. RESULTS: Ninety-two patients were assessed: 21 patients refused mPA (untrained) and 71 agreed (trained). At T1, NACT induced significant up-regulation of interleukin (IL)-5, IL-6, IL-15, chemokine ligand (CCL)-2, interferon-γ, and C-X-C motif ligand (CXCL)-10 and reduction of expression of IL-13 and CCL-22. At T2, NACT and mPA induced up-regulation of IL-21, CCL-2, and tumor necrosis factor-α and reduction of expression of IL-8, IL-15, vascular endothelial growth factor, and soluble interleukin 6 receptor. Only CXCL-10 increased in untrained patients. A cytokine score (CS) was created to analyze, all together, the changes between T1 and T2. At T2 the CS decreased in trained and increased in untrained patients. We clustered the patients using cytokines and predictive factors and identified two clusters. The cluster A, encompassing 90% of trained patients, showed more pathological complete response (pCR) compared to the cluster B: 78% versus 22%, respectively. CONCLUSIONS: mPA interacts with NACT inducing CS reduction in trained patients not observed in untrained patients, suggesting a reduction of inflammation, notwithstanding chemotherapy. This effect may contribute to the higher rate of pCR observed in the cluster A, including most trained patients.


Subject(s)
Breast Neoplasms , Cytokines , Exercise , Inflammation , Neoadjuvant Therapy , Humans , Female , Breast Neoplasms/drug therapy , Neoadjuvant Therapy/methods , Middle Aged , Cytokines/metabolism , Exercise/physiology , Adult , Aged , Prospective Studies
2.
Nat Med ; 2(12): 1371-5, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8946838

ABSTRACT

The HIV-1 Tat protein transactivates HIV, viral and some host cell genes. Tat can be released by infected cells and acts extracellularly in the microenvironment, regulating functions of immunocompetent and mesenchymal cells. One of the most striking effects of Tat is the induction of a functional program in vascular cells related to angiogenesis and inflammation (migration, proliferation and expression of plasminogen activator inhibitor-1 and E selectin). Tat induces growth of Kaposi's sarcoma (KS) spindle cells and is angiogenic in vivo and in transgenic mice10-12. We previously reported that Tat is a direct angiogenic factor and noted the Tat arginine- and lysine-rich sequence is similar to that of other potent angiogenic growth factors, such as vascular endothelial growth factor-A (VEGF-A). It is possible that Tat mimics one of these factors by interacting with its growth factor tyrosine kinase receptor. Here we demonstrate that Tat specifically binds and activates the Flk-1/kinase insert domain receptor (Flk-1/KDR), a VEGF-A tyrosine kinase receptor (for review see ref. 13), and that Tat-induced angiogenesis is blocked by agents blocking the Flk-1/KDR receptor. Endothelial cell stimulation by Tat occurs in the absence of activation of FLT-1, another VEGF-A tyrosine kinase receptor.


Subject(s)
Endothelium, Vascular/metabolism , Gene Products, tat/metabolism , HIV-1/metabolism , Neovascularization, Pathologic , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Animals , Binding Sites , COS Cells , Chemotaxis/drug effects , Collagen , Drug Combinations , Endothelial Growth Factors/metabolism , Endothelial Growth Factors/pharmacology , Enzyme Activation/drug effects , Gene Products, tat/pharmacology , Humans , Laminin , Lymphokines/metabolism , Lymphokines/pharmacology , Phosphorylation , Proteoglycans , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , tat Gene Products, Human Immunodeficiency Virus
3.
Int J Cardiovasc Imaging ; 37(11): 3343-3354, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34114150

ABSTRACT

Right ventricular (RV) involvement is frequently detected in patients presenting with acute left ventricular myocardial infarction. The ischemic right ventricle carries a dismal outcome by predisposing the heart to arrhythmic events and mechanical or hemodynamic complications. A comprehensive RV evaluation by multimodality imaging could guide clinical practice but has always been a conundrum for the imagers. Two-dimensional echocardiography is the best first-line tool due to its availability of bedside capabilities. More advanced imaging techniques provide a more comprehensive evaluation of the complex RV geometry but are mostly reserved for the post-acute setting. Three-dimensional echocardiography has improved the evaluation of RV volumes and function. The recent application of speckle-tracking echocardiography to the right ventricle appears promising, allowing the earlier detection of subtle RV dysfunction. Cardiac magnetic resonance imaging is considered the gold standard for the RV assessment. Cardiac multidetector computed tomography could be a reliable alternative. The aim of this review is to focus on the growing importance of multimodality imaging of the ischemic right ventricle and to propose a diagnostic algorithm, in order to reach a comprehensive assessment of this too frequently neglected chamber.


Subject(s)
Echocardiography, Three-Dimensional , Ventricular Dysfunction, Right , Algorithms , Heart Ventricles/diagnostic imaging , Humans , Predictive Value of Tests , Ventricular Dysfunction, Right/diagnostic imaging , Ventricular Dysfunction, Right/etiology , Ventricular Function, Right
4.
J Cell Biol ; 127(2): 557-65, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7523423

ABSTRACT

The integrin subunit beta 1B, a beta 1 isoform with a unique sequence at the cytoplasmic domain, forms heterodimers with integrin alpha chains and binds fibronectin, but it does not localize to focal adhesion sites (Balzac, F., A. Belkin, V. Koteliansky, Y. Balabanow, F. Altruda, L. Silengo, and G. Tarone. 1993. J. Cell Biol. 121:171-178). Here we analyze the functional properties of human beta 1B by expressing it in hamster CHO cells. When stimulated by specific antibodies, beta 1B does not trigger tyrosine phosphorylation of a 125-kD cytosolic protein, an intracellular signalling pathway that is activated both by the endogenous hamster or the transfected human beta 1A. Moreover, expression of beta 1B results in reduced spreading on fibronectin and laminin, but not on vitronectin. Expression of beta 1B also results in severe reduction of cell motility in the Boyden chamber assay. Reduced cell spreading and motility could not be accounted for by preferential association of beta 1B with a given integrin alpha subunit. These data, together with our previous results, indicate that beta 1B interferes with beta 1A function when expressed in CHO cells resulting in a dominant negative effect on cell adhesion and migration.


Subject(s)
Cell Adhesion , Cell Movement , Integrins/physiology , Animals , CHO Cells , Cell Size , Cricetinae , Fibronectins , Glycoproteins , Integrin alpha3beta1 , Integrin alphaV , Integrin beta1 , Integrins/chemistry , Integrins/genetics , Integrins/metabolism , Laminin , Phosphorylation , Phosphotyrosine , Proteins/metabolism , Receptors, Fibronectin , Transfection , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vitronectin
5.
J Cell Biol ; 136(6): 1375-84, 1997 Mar 24.
Article in English | MEDLINE | ID: mdl-9087450

ABSTRACT

During endochondral bone formation, avascular cartilage differentiates to hypertrophic cartilage that then undergoes erosion and vascularization leading to bone deposition. Resting cartilage produces inhibitors of angiogenesis, shifting to production of angiogenic stimulators in hypertrophic cartilage. A major protein synthesized by hypertrophic cartilage both in vivo and in vitro is transferrin. Here we show that transferrin is a major angiogenic molecule released by hypertrophic cartilage. Endothelial cell migration and invasion is stimulated by transferrins from a number of different sources, including hypertrophic cartilage. Checkerboard analysis demonstrates that transferrin is a chemotactic and chemokinetic molecule. Chondrocyte-conditioned media show similar properties. Polyclonal anti-transferrin antibodies completely block endothelial cell migration and invasion induced by purified transferrin and inhibit the activity produced by hypertrophic chondrocytes by 50-70% as compared with controls. Function-blocking mAbs directed against the transferrin receptor similarly reduce the endothelial migratory response. Chondrocytes differentiating in the presence of serum produce transferrin, whereas those that differentiate in the absence of serum do not. Conditioned media from differentiated chondrocytes not producing transferrin have only 30% of the endothelial cell migratory activity of parallel cultures that synthesize transferrin. The angiogenic activity of transferrins was confirmed by in vivo assays on chicken egg chorioallantoic membrane, showing promotion of neovascularization by transferrins purified from different sources including conditioned culture medium. Based on the above results, we suggest that transferrin is a major angiogenic molecule produced by hypertrophic chondrocytes during endochondral bone formation.


Subject(s)
Cartilage/blood supply , Endothelium, Vascular/drug effects , Neovascularization, Physiologic/physiology , Transferrin/pharmacology , Allantois/blood supply , Allantois/drug effects , Animals , Cartilage/cytology , Cartilage/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemotaxis/drug effects , Chick Embryo , Chorion/blood supply , Chorion/drug effects , Conalbumin/pharmacology , Culture Media, Conditioned/pharmacology , Culture Media, Serum-Free/pharmacology , Endothelium, Vascular/cytology , Fetal Blood/physiology , Growth Plate/cytology , Growth Plate/embryology , Osteogenesis/physiology , Transferrin/biosynthesis
6.
J Cell Biol ; 105(4): 1867-72, 1987 Oct.
Article in English | MEDLINE | ID: mdl-2959668

ABSTRACT

Fibronectin (FN) is a multidomain extracellular matrix protein that induces attachment and chemotactic migration of fibroblastic cells. In this study we analyzed the molecular determinants involved in the FN-induced chemotactic migration of normal and SV40-transformed 3T3 cells. Two different monoclonal antibodies to the cell-binding site of FN blocked chemotaxis to a 140-kD FN fragment (Ca 140) containing the cell-binding domain. A monoclonal antibody to a determinant distant from the cell-binding site did not affect chemotaxis. A synthetic tetrapeptide, RGDS, which represents the major cell-attachment sequence, was able to compete with FN and the Ca 140 fragment in chemotaxis assays, but this peptide itself had no significant chemotactic activity. A larger peptide encompassing this sequence, GRGDSP, was chemotactic, while the peptide GRGESP, where a glutamic acid residue was substituted for aspartic acid, was inactive. Chemotactic migration could be prevented in a dose-dependent manner by a rabbit polyclonal antiserum to a 140-kD cell surface FN receptor. This antibody was more effective on normal than on transformed 3T3 cells. Neither the anti-FN receptor antiserum nor a monoclonal antibody to the cell-binding site of FN blocked migration induced by another potent chemoattractant, platelet-derived growth factor. These data indicate that FN-induced chemotaxis of 3T3 and SV3T3 cells is mediated via the RGDS cell-attachment site of FN and the 140-kD cell surface FN receptor. The interaction is specific and can be altered by transformation.


Subject(s)
Chemotaxis , Fibronectins/physiology , Receptors, Immunologic/physiology , Animals , Binding Sites , Cell Line , Cell Line, Transformed , Chemotaxis/drug effects , Immunologic Techniques , Mice , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Peptide Fragments/physiology , Receptors, Fibronectin
7.
Curr Med Chem ; 13(23): 2737-50, 2006.
Article in English | MEDLINE | ID: mdl-17073625

ABSTRACT

Growth factors and cytokines control and coordinate a broad spectrum of fundamental cellular functions, and are evolutionarily conserved both in vertebrates and invertebrates. In this review, we focus our attention on the functional phylogenetic aspects of growth factors/cytokines like the Transforming Growth Factor-beta (TGF-beta), the Connective Tissue Growth Factor (CTGF), and the Vascular Endothelial Growth Factor (VEGF). We will also delve into the activites of two chemokine families, interleukin (IL)-8 (or CXCL8) and CC chemokine ligand 2/monocyte chemoattractant protein-1 (CCL2). These molecules have been selected for their involvement in immune responses and wound healing processes, where they mediate and finely regulate various regeneration processes like angiogenesis or fibroplasia, not only in vertebrates, but also in invertebrates.


Subject(s)
Chemokines/physiology , Intercellular Signaling Peptides and Proteins/physiology , Invertebrates/physiology , Vertebrates/physiology , Animals , Humans , Invertebrates/growth & development , Neovascularization, Physiologic , Vertebrates/growth & development , Wound Healing/physiology
8.
Curr Pharm Des ; 12(24): 3101-15, 2006.
Article in English | MEDLINE | ID: mdl-16918437

ABSTRACT

The intricate interplay between the endothelium and immune cells has been well recognized in the context of immune responses. However, the fact that this inter-relation extends well beyond immune regulation is becoming increasingly recognized, with particular regards to the influence of the immune system on the essential endothelial process of angiogenesis, where the contribution of cytokines drives the angiogenic process. As angiogenesis is an important component of numerous pathological states, among these chronic inflammatory conditions and cancer, understanding the role of cytokines and chemokines in guiding new vessel formation provides key insight into novel therapeutic modalities. Here we review the actions of principal cytokines and chemokines on the angiogenic process and discuss how both can be considered potential pharmaceutical targets or pharmaceuticals themselves for modulation of angiogenesis in chronic inflammation associated with cancer, rheumatoid arthritis and other inflammatory diseases.


Subject(s)
Chemokines/physiology , Cytokines/physiology , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Chemokines/immunology , Chemokines/pharmacology , Cytokines/immunology , Cytokines/pharmacology , Health Status , Humans , Inflammation/pathology , Inflammation/prevention & control , Neoplasms/pathology , Neoplasms/prevention & control , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/immunology
9.
Int J Immunopathol Pharmacol ; 19(1): 81-96, 2006.
Article in English | MEDLINE | ID: mdl-16569346

ABSTRACT

Kaposi's sarcoma (KS) is a malignancy associated to conditions of immune system impairment such as HIV-1 infection and post-transplantation therapy. Here we report that HIV-1-Tat protein, at concentrations well below those detected in AIDS patients, up-regulates the expression of both CD40 and CD154 on KS cells. This occurred also in the presence of vincristine, that at doses shown to induce apoptosis decreased the expression of both CD40 and CD154 on KS cells. The treatment with a soluble CD40-muIg fusion protein (CD40 fp) that prevents the binding of CD154 with cell surface CD40, as well as the transfection with a vector for soluble CD40 (KS sCD40), decreased the anti-apoptotic effect of Tat. Moreover, Tat-induced motility of KS cells was inhibited by soluble CD40 fp. Tat also enhanced the expression of intracellular proteins known to transduce signals triggered by CD40 engagement, in particular TRAF-3. Tat as well as soluble CD154 (sCD154) prevented vincristine-induced reduction of TRAF-3 in KS cells transfected with a vector for neomycin resistance (KS psv-neo), but not in KS sCD40. Immunoprecipitation studies showed that Tat induced CD40 / TRAF-3 association and that this binding was abrogated upon the incubation with the soluble CD40 fp. These data suggest that Tat activates the CD40-CD154 pathway by enhancing the membrane expression of CD40 and in particular of CD154, and by activating the TRAF-3-dependent signaling pathway of CD40. These findings indicate that the CD40-CD154 pathway mediates the anti-apoptotic and migratory effects of HIV-1- Tat, suggesting the potential therapeutic benefits of blocking CD40 activation in HIV-1-associated KS.


Subject(s)
Apoptosis/drug effects , CD40 Ligand/biosynthesis , Cell Movement/drug effects , Gene Products, tat/pharmacology , Sarcoma, Kaposi/metabolism , Blotting, Western , CD40 Antigens/biosynthesis , CD40 Antigens/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/ultrastructure , Cell Survival/drug effects , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/isolation & purification , HIV-1/metabolism , Humans , Immunoprecipitation , In Situ Nick-End Labeling , Indicators and Reagents , Oligonucleotide Array Sequence Analysis , Transfection , tat Gene Products, Human Immunodeficiency Virus
12.
J Natl Cancer Inst ; 83(11): 775-9, 1991 Jun 05.
Article in English | MEDLINE | ID: mdl-1645772

ABSTRACT

The 72-kd type IV collagenase is a member of the collagenase enzyme family that has been closely linked with the invasive phenotype of cancer cells. Previous studies have shown that both normal cells and highly invasive tumor cells produce the 72-kd type IV procollagenase enzyme in a complexed form consisting of the proenzyme and a novel tissue inhibitor of metalloproteinases, TIMP-2. The balance between activated enzyme and available inhibitor is thought to be a critical determinant of the matrix proteolysis associated with a variety of pathologic processes, including tumor cell invasion. In the present study, we demonstrate that alteration of the metalloproteinase-metalloproteinase-inhibitor balance in favor of excess inhibitor blocks human fibrosarcoma HT-1080 tumor cell invasion of a reconstituted basement membrane. The HT-1080 cell line produces both the 72-kd and the 92-kd type IV collagenases. Alteration of the type IV collagenase-inhibitor balance was achieved by addition of free TIMP-2 or antibodies to 72-kd type IV collagenase. Native, purified TIMP-2 was inhibitory in the range of 1-25 micrograms/mL. Addition of specific antiserum against the 72-kd type IV collagenase, which did not cross-react with the 92-kd type IV collagenase, inhibited HT-1080 cell invasion to the same extent. These results suggest that metalloproteinases, in particular the 72-kd type IV collagenase, are critical for tumor cell invasion of the reconstituted basement membrane. Our findings demonstrate that addition of the endogenous inhibitor TIMP-2 is able to block invasion. Thus, we recommend initiation of in vivo studies of the therapeutic potential of TIMP-2 to block tumor cell invasion and intravasation into the circulation.


Subject(s)
Antineoplastic Agents/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Neoplasm Invasiveness , Neoplasm Proteins/pharmacology , Animals , Humans , Immune Sera/immunology , Mice , Microbial Collagenase/antagonists & inhibitors , Microbial Collagenase/immunology , Microbial Collagenase/physiology , Tissue Inhibitor of Metalloproteinase-2 , Tumor Cells, Cultured
13.
Cancer Res ; 52(8): 2353-6, 1992 Apr 15.
Article in English | MEDLINE | ID: mdl-1313744

ABSTRACT

The metastasis associated 72-kDa type IV collagenase is secreted as a latent proenzyme which is converted to an active 62-kDa form by autoproteolytic removal of an amino terminal profragment. The region immediately upstream from the cleavage site contains a highly conserved peptide sequence, MRKPRCGNPDV, which is present in all known members of the matrix metalloproteinase family. Evidence implicates the cysteine residue of this sequence as critical for maintenance of the latent form through coordination with the catalytic zinc atom of the active site. A synthetic peptide, TMRKPRCGNPDVAN (peptide 74), encompassing this conserved sequence, has been shown to inhibit the activated form of the 72-kDa type IV collagenase in vitro. In the present study we examine the ability of this peptide inhibitor to modulate tumor cell invasiveness. Peptide 74 and the control peptide 78, which contains a single substitution of serine for the "critical" cysteine residue, were added at 30 microM concentrations to the upper compartment of the Boyden chamber in the chemoinvasion assay using HT1080 and A2058 human tumor cells. In this assay a layer of reconstituted basement membrane, Matrigel, is coated onto chemotaxis filters and acts as a barrier to the migration of cells in the Boyden chambers. Only cells with invasive capacity can cross the Matrigel barrier. Peptide 74 containing the cysteine residue inhibited the invasion of both the HT1080 and A2058 cells through the Matrigel barrier; control peptide 78 was not inhibitory. Both peptides were shown to be without cytotoxic action and did not inhibit chemotaxis or affect cell number. This study demonstrates that addition of an excess peptide containing the matrix metalloproteinase prosegment inhibitory sequence can inhibit invasive activity at the cellular level and suggests that this may be a useful strategy to modulate tumor cell invasiveness in vivo.


Subject(s)
Microbial Collagenase/antagonists & inhibitors , Neoplasm Invasiveness , Peptides/pharmacology , Base Sequence , Fibrosarcoma/enzymology , Fibrosarcoma/pathology , Humans , Intercellular Signaling Peptides and Proteins , Matrix Metalloproteinase 9 , Melanoma/enzymology , Melanoma/pathology , Molecular Sequence Data , Peptides/chemistry , Tumor Cells, Cultured
14.
Cancer Res ; 48(23): 6764-8, 1988 Dec 01.
Article in English | MEDLINE | ID: mdl-2846159

ABSTRACT

Estrogen increases the ability of the estrogen-dependent MCF-7 human breast cancer cell line to both proliferate and invade through an artificial basement membrane. In studying the response of MCF-7 cells to various antiestrogens, we found that 4-hydroxytamoxifen and tamoxifen inhibited cell proliferation but increased their invasiveness. In contrast, the structurally unrelated benzothiophene antiestrogens, LY117018 and LY156758, were potent antiproliferative agents which did not stimulate invasiveness. The differential effects of these antiestrogenic agents on invasion correlated with changes in production of collagenase IV, while no significant change was seen in the chemotactic activity of the cells. Invasiveness was increased by 17 beta-estradiol or 4-hydroxytamoxifen after a few hours of treatment and was rapidly lost when 17 beta-estradiol was withdrawn. Stimulation of invasiveness with 17 beta-estradiol was blocked by the antiestrogen, LY117018. Cells from the MDA-MB-231 line which lacks estrogen receptors were not affected by estrogen or antiestrogen in terms of proliferation or invasion. These studies indicate that the invasiveness of MCF-7 cells is regulated by antiestrogens through the estrogen receptor and may be mediated by collagenase IV activity. Antiestrogens which reduce both the proliferation and invasiveness of these cells may be interesting new candidates for clinical application.


Subject(s)
Breast Neoplasms/pathology , Estrogen Antagonists/pharmacology , Neoplasm Invasiveness , Cell Division/drug effects , Chemotaxis , Estradiol/pharmacology , Female , Humans , Microbial Collagenase/biosynthesis , Plasminogen Activators/biosynthesis , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tumor Cells, Cultured
15.
Cancer Res ; 61(14): 5441-6, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454689

ABSTRACT

Gene transfer delivery of endogenous angiogenesis inhibitors such as angiostatin would circumvent problems associated with long-term administration of proteins. Kaposi's sarcoma (KS), a highly vascular neoplasm, is an excellent model for studying tumor angiogenesis and antiangiogenic agent efficacy. We investigated the effects of angiostatin gene transfer in in vitro and in vivo models of KS-induced neovascularization and tumor growth. A eukaryotic expression plasmid and a Moloney leukemia virus-based retroviral vector for expression of murine angiostatin were generated harboring the angiostatin cDNA with cleavable leader signals under the control of either the strong cytomegalovirus promoter/enhancer or the Moloney leukemia virus long terminal repeat. Angiostatin secretion was confirmed by radioimmunoprecipitation and Western blot analysis. Supernatants of angiostatin-transfected cells inhibited endothelial cell migration in vitro. Stable gene transfer of the angiostatin cDNA by retroviral vectors in KS-IMM cells resulted in sustained angiostatin expression and delayed tumor growth in nude mice, which was associated with reduced vascularization. These findings suggest that gene therapy with angiostatin might be useful for treatment of KS and possibly other highly angiogenic tumors.


Subject(s)
Peptide Fragments/physiology , Plasminogen/physiology , Sarcoma, Kaposi/pathology , Angiostatins , Animals , Cell Division , Cell Line , Cell Movement/drug effects , Chemotaxis/drug effects , Culture Media, Conditioned/pharmacology , Genetic Vectors/genetics , Humans , Mice , Peptide Fragments/genetics , Plasminogen/genetics , Sarcoma, Kaposi/genetics , Transfection , Tumor Cells, Cultured
16.
Cancer Res ; 61(22): 8171-8, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719447

ABSTRACT

The thiol N-acetyl-L-cysteine (NAC), an analogue and precursor of reduced glutathione, has cancer chemopreventive properties attributable to its nucleophilicity, antioxidant activity, and a variety of other mechanisms. We demonstrated recently that NAC has anti-invasive, antimetastatic, and antiangiogenic effects in in vitro and in vivo test systems. In the present study, s.c. transplantation of KS-Imm cells in (CD-1)BR nude mice resulted in the local growth of Kaposi's sarcoma, a highly vascularized human tumor. The daily administration of NAC with drinking water, initiated after the tumor mass had become established and detectable, produced a sharp inhibition of tumor growth, with regression of tumors in half of the treated mice along with a markedly prolonged median survival time. The production of vascular endothelial growth factor (VEGF) and certain proliferation markers (proliferating cell nuclear antigen and Ki-67) were significantly lower in Kaposi's sarcomas from NAC-treated mice than from control mice. Treatment of KS-Imm cells with NAC in vitro resulted in a dose-dependent inhibition of chemotaxis and invasion through inhibition of gelatinase-A (matrix metalloproteinase-2, MMP-2) activity without altering MMP-2 or MMP-9 mRNA levels. NAC also significantly inhibited VEGF production but did not affect proliferation markers in vitro. Reverse transcription-PCR analysis indicated that total VEGF mRNAs were reduced by 10 mM NAC. Taken together, these findings provide evidence that NAC, the safety of which even at high doses has been established in almost 40 years of clinical use, in addition to its chemopreventive action, has a strong antiangiogenic potential that could be exploited for preventing cancer progression as well as used in cancer adjuvant therapy.


Subject(s)
Acetylcysteine/pharmacology , Angiogenesis Inhibitors/pharmacology , Neovascularization, Pathologic/drug therapy , Sarcoma, Kaposi/blood supply , Administration, Oral , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Movement/drug effects , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Female , Growth Inhibitors/pharmacology , Humans , Ki-67 Antigen/metabolism , Lymphokines/antagonists & inhibitors , Lymphokines/biosynthesis , Lymphokines/genetics , Male , Mice , Mice, Nude , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sarcoma, Kaposi/pathology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
17.
Cancer Res ; 51(10): 2670-6, 1991 May 15.
Article in English | MEDLINE | ID: mdl-2021945

ABSTRACT

Kaposi's sarcoma (KS) in general, and acquired immunodeficiency syndrome-related KS (AIDS-KS) in particular, is a highly invasive and intensely angiogenic neoplasm of unknown cellular origin. We have recently established AIDS-KS cells in long term culture and reported the development of KS-like lesions in nude mice inoculated with these cells. Here, we have examined the in vitro invasiveness of basement membrane by AIDS-KS cells, as well as the effect(s) of their supernatants on the migration and invasiveness of human vascular endothelial cells. AIDS-KS cells were highly invasive in the Boyden chamber invasion assay and formed invasive, branching colonies in a 3-dimensional gel (Matrigel). Normal endothelial cells form tube-like structures on Matrigel. AIDS-KS cell-conditioned media induced endothelial cells to form invasive clusters in addition to tubes. KS-cell-conditioned media, when placed in the lower compartment of the Boyden chamber, stimulated the migration of human and bovine vascular endothelial cells across filters coated with either small amounts of collagen IV (chemotaxis) or a Matrigel barrier (invasion). Basic fibroblast growth factor could also induce endothelial cell chemotaxis and invasion in these assays. However, when antibodies to basic fibroblast growth factor were used the invasive activity induced by the AIDS-KS-cell-conditioned media was only marginally inhibited, suggesting that the large quantities of basic fibroblast growth factor-like material released by the AIDS-KS cells are not the main mediators of this effect. Specific inhibitors of laminin and collagenase IV action, which represent critical determinants of basement membrane invasion, blocked the invasiveness of the AIDS-KS cell-activated endothelial cells in these assays. These data indicate that KS cells appear to be of smooth muscle origin but secrete a potent inducer of endothelial cell chemotaxis and invasiveness which could be responsible for angiogenesis and the resulting highly vascularized lesions. These assays appear to be a model to study the invasive spread and angiogenic capacity of human AIDS-related KS and should prove useful in the identification of molecular mediators and potential inhibitors of neoplastic neovascularization.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Chemotaxis , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Sarcoma, Kaposi/physiopathology , Acquired Immunodeficiency Syndrome/physiopathology , Animals , Aorta/physiology , Basement Membrane/pathology , Cattle , Cell Communication , Cells, Cultured , Humans , Neoplasm Invasiveness , Sarcoma, Kaposi/etiology , Sarcoma, Kaposi/pathology
18.
Cancer Res ; 42(6): 2277-83, 1982 Jun.
Article in English | MEDLINE | ID: mdl-7074610

ABSTRACT

Aniline of unknown purity has been reported to induce spleen hemangiosarcoma in rats. Aniline has been found to be negative in terms of mutagenicity in both bacteria and yeasts. We have found that both commercial (already rather pure) and repurified aniline are clearly positive to a similar extent in inducing DNA damage in vivo in liver and kidney of rats. Both the commercial and repurified product are also clearly positive in induction of sister chromatid exchanges in vivo in male Swiss mice bone marrow cells. Liver, kidney, and bone marrow DNA damage was absent in male Swiss mice.


Subject(s)
Aniline Compounds/pharmacology , Bone Marrow/physiology , Crossing Over, Genetic/drug effects , DNA Replication/drug effects , Liver/physiology , Sister Chromatid Exchange/drug effects , Spleen/physiology , Aniline Compounds/isolation & purification , Animals , Bone Marrow/drug effects , Cell Survival/drug effects , Chromatography, Gas , Liver/drug effects , Male , Mice , Rats , Rats, Inbred Strains , Spleen/drug effects
19.
Cancer Res ; 47(12): 3239-45, 1987 Jun 15.
Article in English | MEDLINE | ID: mdl-2438036

ABSTRACT

We have reconstituted a matrix of basement membrane onto a filter in a Boyden chamber and assessed the ability of various malignant and nonmalignant cells to penetrate through the coated filter. Cells from all the malignant cell lines tested were able to cross the matrix in 5-6 h, whereas human fibroblasts as well as mouse 3T3 and 10T1/2 cell lines, which are not tumorigenic, were not invasive. In addition, normal primary prostate epithelial cells and benign prostatic hyperplasia cells were not invasive when tested in this assay, whereas malignant prostate carcinoma cells were highly invasive. Parallel experiments with these prostatic cells using the intrasplenic assay for metastasis detection in the nude mouse confirmed the benign behavior of the former cells and the metastatic phenotype of the latter ones. These results suggest that this in vitro test allows the rapid and quantitative assessment of invasiveness and a means to screen for drugs which alter the invasive phenotype of tumor cells.


Subject(s)
Melanoma/pathology , Neoplasm Invasiveness , Animals , Basement Membrane/ultrastructure , Fibroblasts/ultrastructure , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C3H , Microscopy, Phase-Contrast , Phenotype , Prostatic Hyperplasia/pathology
20.
Oncogene ; 9(8): 2253-60, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8036010

ABSTRACT

Oncostatin M (OM) is a polypeptide cytokine that induces autocrine and paracrine effects on AIDS-Kaposi's sarcoma (KS) cells (Nair et al., Science, 255, 1430-1432, 1992; Miles et al., Science, 255, 1432-1434, 1992). The signalling pathways underlying this activation are largely unknown. We have found that OM binding to KS cell lines in vitro identifies a higher affinity binding site (Kd 10-20 pM) with a lower affinity (Kd 1.5 nM), high capacity binding site. The binding of OM to its receptor at the KS cell surface stimulates a rapid tyrosine phosphorylation of multiple proteins, including the p85 regulatory subunit of phosphatidylinositol-3-kinase (PI3K). In addition OM can stimulate the in vivo activation of PI3K and increases the PI3K activity in anti-phosphotyrosine and anti-src kinase family antibody directed immunoprecipitates. Genistein, an inhibitor of tyrosine kinases, inhibits the synthesis of phosphatidylinositol 3,4-biphosphate and the growth of KS cells. Finally, OM enhances tyrosine kinase activity in immune complex kinase assay performed with antibody anti-src kinase family. These data suggest that in KS cells OM can stimulate formation of tyrosine kinase co-ordinate signalling complexes, containing at least src kinase family and PI3K, which can drive the accumulation of the putative second-messengers D3-phosphorylated phosphoinositides.


Subject(s)
Antineoplastic Agents/pharmacology , Cytokines/pharmacology , Peptides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sarcoma, Kaposi/enzymology , Humans , Oncostatin M , Phosphatidylinositol 3-Kinases , Phosphatidylinositols/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL