Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
J Endocrinol Invest ; 43(4): 413-429, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31584143

ABSTRACT

The main role of vitamin D is to control mineral homeostasis. However, recent studies suggested the existence of a number of extraskeletal effects. Among the latter, preclinical studies provided consistent data on the involvement of vitamin D in innate and adaptive immunity and autoimmunity. Molecular biology studies showed that both vitamin D receptor and vitamin D enzymatic complexes are expressed in a large number of cells and tissues unrelated to mineral homeostasis. In contrast, only a few randomized clinical trials in humans investigated the possible role of vitamin D in the prevention or treatment of immunological disorders. In this regard, low serum vitamin D levels have been reported in observational trials in human autoimmune disorders. The aim of the present paper was to review the potential implications of vitamin D in immune modulation, with special focus on thyroid autoimmune disorders.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/drug effects , Thyroid Diseases/immunology , Vitamin D/therapeutic use , Autoimmune Diseases/blood , Autoimmune Diseases/drug therapy , Autoimmune Diseases/prevention & control , Humans , Thyroid Diseases/blood , Thyroid Diseases/drug therapy , Thyroid Diseases/prevention & control , Thyroid Gland/drug effects , Vitamin D/administration & dosage , Vitamin D/blood
2.
Health Educ Res ; 33(1): 81-88, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29309599

ABSTRACT

Text-based interventions are effective for smoking cessation, but have not been tested in rural older adults. The purpose of this study was to compare the feasibility, acceptability and preliminary efficacy of a text-based Scheduled Gradual Reduction (SGR) program to a non-SGR text messaging support condition among rural older adults. Adults over 60 years were randomized to either: (i) the SGR program (n = 20), a text-based program to reduce smoking over 4-weeks plus text-based support messages; or (ii) control (n = 20), receipt of text-based support messages only. Participants completed surveys at baseline and end of program to assess feasibility and acceptability of the intervention, and biochemically validated 7-day point prevalence cessation was assessed at end of treatment. Most participants (81%) reported reading all the messages they received. Participants found both interventions useful in quitting smoking (SGR = 57%, Control = 63%) and would recommend it to a friend (SGR = 72%, Control = 79%). Although not statically significant, the SGR group had a higher rate of biochemically validated cessation (SGR = 15%, Control = 5%, Cohen d = 0.67). Among those still smoking, the median percent reduction in cigarettes was 33.3% for both groups. Text-based cessation interventions are feasible, acceptable and can be easily disseminated to rural older adult tobacco users.


Subject(s)
Rural Population , Smoking Cessation/methods , Text Messaging , Aged , Female , Humans , Male , Middle Aged , Socioeconomic Factors , Time Factors
3.
Leukemia ; 30(12): 2351-2363, 2016 12.
Article in English | MEDLINE | ID: mdl-27311934

ABSTRACT

Galectin-1 (Gal-1) is involved in tumoral angiogenesis, hypoxia and metastases. Actually the Gal-1 expression profile in multiple myeloma (MM) patients and its pathophysiological role in MM-induced angiogenesis and tumoral growth are unknown. In this study, we found that Gal-1 expression by MM cells was upregulated in hypoxic conditions and that stable knockdown of hypoxia inducible factor-1α significantly downregulated its expression. Therefore, we performed Gal-1 inhibition using lentivirus transfection of shRNA anti-Gal-1 in human myeloma cell lines (HMCLs), and showed that its suppression modified transcriptional profiles in both hypoxic and normoxic conditions. Interestingly, Gal-1 inhibition in MM cells downregulated proangiogenic genes, including MMP9 and CCL2, and upregulated the antiangiogenic ones SEMA3A and CXCL10. Consistently, Gal-1 suppression in MM cells significantly decreased their proangiogenic properties in vitro. This was confirmed in vivo, in two different mouse models injected with HMCLs transfected with anti-Gal-1 shRNA or the control vector. Gal-1 suppression in both models significantly reduced tumor burden and microvascular density as compared with the control mice. Moreover, Gal-1 suppression induced smaller lytic lesions on X-ray in the intratibial model. Overall, our data indicate that Gal-1 is a new potential therapeutic target in MM blocking angiogenesis.


Subject(s)
Galectin 1/metabolism , Multiple Myeloma/pathology , Neovascularization, Pathologic/drug therapy , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Galectin 1/antagonists & inhibitors , Humans , Mice , Multiple Myeloma/blood supply , RNA, Small Interfering/pharmacology , Transfection , Tumor Burden/drug effects
4.
Clin Exp Immunol ; 183(3): 469-79, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26462606

ABSTRACT

Activated human neutrophils produce a fibrillar DNA network [neutrophil extracellular traps (NETs)] for entrapping and killing bacteria, fungi, protozoa and viruses. Our results suggest that the neutrophil extracellular traps show a resistant amyloidogenic backbone utilized for addressing reputed proteins and DNA against the non-self. The formation of amyloid fibrils in neutrophils is regulated by the imbalance of reactive oxygen species (ROS) in the cytoplasm. The intensity and source of the ROS signal is determinant for promoting stress-associated responses such as amyloidogenesis and closely related events: autophagy, exosome release, activation of the adrenocorticotrophin hormone/α-melanocyte-stimulating hormone (ACTH/α-MSH) loop and synthesis of specific cytokines. These interconnected responses in human activated neutrophils, that have been evaluated from a morphofunctional and quantitative viewpoint, represent primitive, but potent, innate defence mechanisms. In invertebrates, circulating phagocytic immune cells, when activated, show responses similar to those described previously for activated human neutrophils. Invertebrate cells within endoplasmic reticulum cisternae produce a fibrillar material which is then assembled into an amyloidogenic scaffold utilized to convey melanin close to the invader. These findings, in consideration to the critical role played by NET in the development of several pathologies, could explain the structural resistance of these scaffolds and could provide the basis for developing new diagnostic and therapeutic approaches in immunomediated diseases in which the innate branch of the immune system has a pivotal role.


Subject(s)
Amyloid/metabolism , Extracellular Traps/immunology , Extracellular Traps/physiology , Neutrophil Activation , Neutrophils/immunology , Adrenocorticotropic Hormone/physiology , Animals , Autophagy , Exosomes/physiology , Humans , Immunity, Innate , Neutrophils/ultrastructure , Reactive Oxygen Species , alpha-MSH/metabolism
5.
Curr Med Chem ; 21(5): 630-40, 2014.
Article in English | MEDLINE | ID: mdl-23862621

ABSTRACT

Several substances widely dispersed in the environment including hormones, industrial by-products and pollutants exert hormone like activity affecting steroid-responsive physiological systems. These compounds, named endocrine disruptors, are suspected to affect the mammalian reproductive system. However it is still unclear whether these substances are able to elicit estrogen like activity at the low concentrations encountered in the environment. Here we compare the effects of the endocrine disruptor nonylphenol with the effects elicited by 17-ß-estradiol on gene transcription in the human breast cancer cell line MCF7. The correlation of the nonylphenol induced gene expression alterations with a reference profile of estradiol treated cells shows that nonylphenol at a concentration of 100 nM exerts a significant effect on estrogen responsive gene transcription in MCF7 cells. Most of the genes regulated by 17-ß-estradiol respond to the nonylphenol in the same direction though to a much lesser extent. Molecular modeling of the potential interaction of nonylphenol with the estrogen receptor α shows that nonylphenol is likely to bind to the estrogen receptor α.


Subject(s)
Endocrine Disruptors/pharmacology , Phenols/pharmacology , Receptors, Estrogen/metabolism , Transcription, Genetic/drug effects , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Endocrine Disruptors/chemistry , Estradiol/pharmacology , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Molecular Docking Simulation , Phenols/chemistry , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Estrogen/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
6.
Curr Med Chem ; 21(9): 1129-45, 2014.
Article in English | MEDLINE | ID: mdl-24304271

ABSTRACT

Observations on the role of ovarian hormones in breast cancer growth, as well as interest in contraception, stimulated research into the biology of estrogens. The identification of the classical receptors ERα and ERß and the transmembrane receptor GPER and the resolution of the structure of the ligand bound to its receptor established the principal molecular mechanisms of estrogen action. The presence of estrogen-like compounds in many plants used in traditional medicine or ingested as food ingredients, phytoestrogens, as well as the estrogenic activities of many industrial pollutants and pesticides, xenoestrogens, have prompted investigations into their role in human health. Phyto- and xenoestrogens bind to the estrogen receptors with a lower affinity than the endogenous estrogens and can compete or substitute the hormone. Xenoestrogens, which accumulate in the body throughout life, are believed to increase breast cancer risk, especially in cases of prenatal and prepuberal exposure whereas the role of phytoestrogens is still a matter of debate. At present, the application of phytoestrogens appears to be limited to the treatment of post-menopausal symptoms in women where the production of endogenous estrogens has ceased. In this review we discuss chemistry, structure and classification, estrogen signaling and the consequences of the interactions of estrogens, phytoestrogens and xenoestrogens with their receptors, the complex interactions of endogenous and exogenous ligands, the evaluation of the health risks related to xenoestrogens, and the perspectives toward the synthesis of potent third generation selective estrogen receptor modulators (SERMs).


Subject(s)
Breast Neoplasms/metabolism , Endocrine Disruptors/metabolism , Phytoestrogens/metabolism , Animals , Humans , Receptors, Estrogen/metabolism , Risk Factors , Signal Transduction
7.
Br J Pharmacol ; 167(6): 1218-31, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22624859

ABSTRACT

BACKGROUND AND PURPOSE: Several studies have demonstrated anti-proliferative and pro-apoptotic actions of cannabinoids on various tumours, together with their anti-angiogenic properties. The non-psychoactive cannabinoid cannabidiol (CBD) effectively inhibits the growth of different types of tumours in vitro and in vivo and down-regulates some pro-angiogenic signals produced by glioma cells. As its anti-angiogenic properties have not been thoroughly investigated to date, and given its very favourable pharmacological and toxicological profile, here, we evaluated the ability of CBD to modulate tumour angiogenesis. EXPERIMENTAL APPROACH: Firstly, we evaluated the effect of CBD on human umbilical vein endothelial cell (HUVEC) proliferation and viability - through [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay and FACS analysis - and in vitro motility - both in a classical Boyden chamber test and in a wound-healing assay. We next investigated CBD effects on different angiogenesis-related proteins released by HUVECs, using an angiogenesis array kit and an ELISA directed at MMP2. Then we evaluated its effects on in vitro angiogenesis in treated HUVECs invading a Matrigel layer and in HUVEC spheroids embedded into collagen gels, and further characterized its effects in vivo using a Matrigel sponge model of angiogenesis in C57/BL6 mice. KEY RESULTS: CBD induced HUVEC cytostasis without inducing apoptosis, inhibited HUVEC migration, invasion and sprouting in vitro, and angiogenesis in vivo in Matrigel sponges. These effects were associated with the down-modulation of several angiogenesis-related molecules. CONCLUSIONS AND IMPLICATIONS: This study reveals that CBD inhibits angiogenesis by multiple mechanisms. Its dual effect on both tumour and endothelial cells supports the hypothesis that CBD has potential as an effective agent in cancer therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Cannabidiol/therapeutic use , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Cannabidiol/pharmacology , Cell Movement/drug effects , Heparin , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/chemically induced , Tumor Necrosis Factor-alpha , Vascular Endothelial Growth Factor A
8.
Curr Drug Targets ; 12(13): 1909-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21158708

ABSTRACT

Cardiovascular diseases and cancer are the leading causes of death in most countries. These diseases share many common risk factors as well as pathogenetic determinants, and their incidence is related to age in an exponential manner. Furthermore, it has become apparent that several treatments used in therapy or even in prevention of cancer can impair the structural and functional integrity of the cardiovascular system, giving rise to an interdisciplinary field: cardio-oncology. However, tumors and cardiovascular diseases also share common protective factors: they can be prevented either by avoiding exposure to recognized risk factors, and/or by favoring the intake of protective compounds and by modulating the host defense machinery. These latter approaches are generally known as chemoprevention. A great variety of dietary and pharmacological agents have been shown to be potentially capable of preventing cancer in preclinical models, most of which are of plant origin. Phytochemicals, in particular diet-derived compounds, have therefore been proposed and applied in clinical trials as cancer chemopreventive agents. There is now increasing evidence that some phytochemicals can be also protective for the heart, having the potential to reduce cancer, cardiovascular disease and even anticancer drug-induced cardiotoxicity. We introduce the concept that these compounds induce pre-conditioning, a low level cellular stress that induces strong protective mechanisms conferring resistance to toxins such as cancer chemotherapeutics. Cancer cells and cardiomyocytes have fundamental differences in their metabolism and sensitivity to preconditioning, autophagy and apoptosis, so that dosage of the prevention compounds is important. Here we discuss the mechanisms responsible for the cardiotoxicity of anticancer drugs, the possibility to prevent them and provide examples of diet-derived phytochemicals and other biological substances that could be exploited for protecting the cardiovascular system according to a joint cardio-oncological preventative approach.


Subject(s)
Cardiovascular Diseases/prevention & control , Chemoprevention/trends , Diet , Neoplasms/prevention & control , Animals , Brassicaceae/chemistry , Cardiovascular Diseases/diet therapy , Cardiovascular Diseases/mortality , Chemoprevention/methods , Garlic/chemistry , Heart/drug effects , Heart/physiology , Humans , Neoplasms/diet therapy , Neoplasms/mortality , Plants/chemistry , Polyphenols/chemistry , Polyphenols/therapeutic use
9.
Curr Pharm Biotechnol ; 12(2): 171-81, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21044010

ABSTRACT

The idea of cancer stem cell (CSC) has recently moved to the forefront of cancer research. There is still a lack of a widespread consensus on their description and definition. ,The increasing literature on CSCs has compelled researchers worldwide to rewrite the natural history of cancer including those cells as principal players as well as to revise their views on tumor formation and progression. CSCs are tumor cell components that can initiate a new tumor after an apparent therapeutic eradication. A functional definition of cancer stem cell or cancer initiating cell is that of a cell which, when transplanted in a mouse model, can give rise to a tumor recapitulating the original one or even a phenotypically diverse tumor related to the tumor of origin. Since the characteristic asymmetric division of stem cells is somewhat anomalous in cancer, it might be advisable to refer to them as "stemloids". Stemness in cancer is not as much as an identity but rather a status. There is increasing evidence of the importance of the tumor and the host micro-environment in conditioning the stem cell status itself. The cancer stem cell micro-environment may be the key in the development of therapeutic strategies. We must think in terms of targeting "standard" tumor cells, cancer stem cells, and also their niche and tumor micro-environment. Here we discuss some features of cancer stem cells, and the role of the micro-environment, envisaging a choral view of cancer stem cell development and-or latency, towards development of specific therapeutic approaches. Here we propose models of replication and quiescence and the modulation by cells, genes and miRNAs. We also summarize in a table surface makers useful for the identification and isolation of CSCs.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment , Animals , Humans , Mice , Molecular Targeted Therapy , Neoplastic Stem Cells/cytology , Stem Cell Niche/physiology
10.
Q J Nucl Med Mol Imaging ; 54(3): 244-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20639811

ABSTRACT

The tumor microenvironment is a "complex society" of many cell types and their extracellular matrix. All these cell types and the matrix take part to the generation of a tumor "tissue". It is well established that preneoplastic proliferating cells cannot give origin to a tumor without an appropriate blood supply. In fact, angiogenesis could be considered the rate limiting step of tumor growth. In this context microenvironment components play a pivotal role in the regulation of the angiogenic switch and in cancer progression. For these reasons the comprehension of biological and molecular mechanisms involved in the relationship between tumor cells and the microenvironment could unveil new therapeutic and preventive approaches to cancer. In this complex scenario molecular imaging of the microenvironment is crucial to dissect cellular and stromal dynamic contributions.


Subject(s)
Tumor Microenvironment , Animals , Cell Proliferation , Extracellular Matrix/physiology , Humans , Molecular Imaging , Neoplasms/blood supply , Neoplasms/pathology , Neoplasms/physiopathology , Neovascularization, Pathologic , Signal Transduction
11.
Oncogene ; 29(38): 5286-98, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20603611

ABSTRACT

Hepatocyte growth factor (HGF) and its receptor Met are responsible for a wide variety of cellular responses, both physiologically during embryo development and tissue homeostasis, and pathologically, particularly during tumor growth and dissemination. In cancer, Met can act as an oncogene on tumor cells, as well as a pro-angiogenic factor activating endothelial cells and inducing new vessel formation. Molecules interfering with Met activity could be valuable therapeutic agents. Here we have investigated the antiangiogenic properties of a synthetic peptide mimicking the docking site of the Met carboxyl-terminal tail, which was delivered into the cells by fusion with the internalization sequences from Antennapedia or HIV-Tat. We showed that these peptides inhibit ligand-dependent endothelial cell proliferation, motility, invasiveness and morphogenesis in vitro to an even greater extent and with much less toxicity than the Met inhibitor PHA-665752, which correlated with interference of HGF-dependent downstream signaling. In vivo, the peptides inhibited HGF-induced angiogenesis in the matrigel sponge assay and impaired xenograft tumor growth and vascularization in Kaposi's sarcoma. These data show that interference with the Met receptor intracellular sequence impairs HGF-induced angiogenesis, suggesting the use of antidocking site compounds as a therapeutic strategy to counteract angiogenesis in cancer as well as in other diseases.


Subject(s)
Neoplasms/pathology , Neovascularization, Pathologic , Proto-Oncogene Proteins c-met/metabolism , Receptors, Growth Factor/metabolism , Amino Acid Sequence , Animals , Apoptosis , Blotting, Western , Cell Line , Humans , Mice , Molecular Sequence Data , Neoplasms/blood supply , Proto-Oncogene Proteins c-met/chemistry , Receptors, Growth Factor/chemistry , Transplantation, Heterologous
12.
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
13.
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
14.
Microvasc Res ; 72(3): 128-35, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16920158

ABSTRACT

The human Kaposi sarcoma represents one of the most common skin lesions associated with AIDS. Its clinical presentation and anatomopathological structure seem to demonstrate a particularly rich vascularity. The latest therapies aim to limit its intrinsic angiogenic activity in an attempt to reduce vascular density and the formation of new vessels. For these reasons, we decided to study the microvascular architecture of Kaposi sarcoma in three dimensions. We used a corrosion casting technique applied to nude mice previously transplanted subcutaneously with human modified neoplastic Kaposi sarcoma cells. The cooption of host vessels made by the tumor was demonstrated by three-dimensional scanning electron microscopy (SEM) images. At high magnification several angiogenic patterns were observed in the form of potato-shaped vessels, sprouts, intussusceptions and mouse tailed end tipped capillaries along with some ultrastructural features such as intercellular extravasations and endothelial cell modifications. Our investigation allowed us to build a detailed map of tumor vasculature in human Kaposi sarcoma. Furthermore, this study want to shed light on the sharp morphological three-dimensional conformation of angiogenic sprouts so to be able to better understand their modifications occurred during time and after antiangiogenic experimental therapies, by now observed only by immunohistochemical or immunofluorescent assays.


Subject(s)
Neovascularization, Pathologic , Sarcoma, Kaposi/blood supply , Sarcoma, Kaposi/ultrastructure , Animals , Arteries/pathology , Arteries/ultrastructure , Arterioles/pathology , Arterioles/ultrastructure , Capillaries/pathology , Capillaries/ultrastructure , Cell Line, Tumor , Humans , Image Processing, Computer-Assisted , Male , Mice , Mice, Nude , Microcirculation/pathology , Microcirculation/ultrastructure , Microscopy, Electron, Scanning , Neoplasm Transplantation , Sarcoma, Kaposi/pathology , Transplantation, Heterologous , Veins/pathology , Veins/ultrastructure , Venules/pathology , Venules/ultrastructure
15.
Br J Cancer ; 94(10): 1428-35, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16622457

ABSTRACT

Tumour growth is tightly related to new blood vessel formation, tissue remodelling and invasiveness capacity. A number of tissular factors fuel the growth of glioblastoma multiforme, the most aggressive brain neoplasm. In fact, gene array analyses demonstrated that the proapoptotic cytokine tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) inhibited mRNA expression of VEGF, along with those of matrix metalloproteinase-2 (MMP-2), its inhibitor tissue inhibitor of matrix metalloproteinases-2 (TIMP-2), as well as the tumour invasiveness-related gene secreted protein acid rich in cysteine (SPARC) in different human glioblastoma cell lines. Particularly, VEGF mRNA and protein expression and release from glioblastoma cells were also inhibited by TRAIL. The latter also exerted antimitogenic effects on human umbilical vein endothelial cells (HUVECs). With the same cells, TRAIL inhibited new vessel formation in the in vitro matrigel model, as well as it exerted powerful inhibition of blood vessel formation induced by an angiogenic cocktail administered in subcutaneous pellets in vivo in the C57 mouse. Moreover, the expression of MMP-2, its inhibitor TIMP-2 and the tumour invasiveness-related protein SPARC were effectively inhibited by TRAIL in glioblastoma cell lines. In conclusion, our data indicate that TRAIL inhibits the orchestra of factors contributing to glioblastoma biological aggressiveness. Thus, the TRAIL system could be regarded as a molecular target to exploit for innovative therapy of this type of tumour.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/therapeutic use , Brain Neoplasms/blood supply , Glioblastoma/blood supply , Membrane Glycoproteins/therapeutic use , Neovascularization, Pathologic/drug therapy , Tumor Necrosis Factor-alpha/therapeutic use , Vascular Endothelial Growth Factor A/genetics , Animals , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Survival , Endothelium, Vascular/drug effects , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Osteonectin/genetics , Osteonectin/metabolism , RNA, Messenger/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
16.
Gene Ther ; 11(3): 284-91, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14737088

ABSTRACT

IL-12 is thought to induce a cytokine cascade with antiangiogenic effects mediated by IFN-gamma and angiostatic CXCR3 chemokine ligands. Naked DNA intramuscular injection of an expression vector plasmid producing IL-12 resulted in significant, well-tolerated elevation of serum IL-12 levels. Injection of the IL-12 plasmid at least 2 days, and up to 20 days, before subcutaneous injection of matrigel with angiogenic factors resulted in strong prevention of angiogenesis in both C57/bl and nude mice. A single injection of the IL-12 plasmid contemporarily with the matrigel or 2 days after resulted in partial, statistically not significant, inhibition. Control plasmid injection did not affect either angiogenesis or angiogenesis inhibition by IL-12 protein in vivo. Angiogenesis inhibition was observed in NK cell-depleted C57/bl and nude mice as well as in IFN-gamma(-/-) and CXCR3(-/-) knockout mice, indicating that NK- and/or T-cell-initiated IFN-gamma-chemokine cascades were not involved in the angiogenesis inhibition observed in vivo. Finally, IL-12 plasmid DNA gene transfer significantly prevented the growth and vascularization of highly angiogenic KS-Imm Kaposi's sarcoma and TS/A murine mammary carcinoma tumors in nude and/or syngeneic mice. These data suggest that a preventive gene therapy approach using antiangiogenic cytokines can effectively inhibit tumor angiogenesis and KS, representing an example of angioimmunoprevention.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Interleukin-12/genetics , Neovascularization, Pathologic/prevention & control , Sarcoma, Kaposi/blood supply , Animals , Genetic Vectors , Interferon-gamma/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/immunology , Plasmids/genetics , Receptors, CXCR3 , Receptors, Chemokine/immunology , Sarcoma, Kaposi/pathology , Sarcoma, Kaposi/therapy , T-Lymphocytes/immunology
17.
Int J Biol Markers ; 18(1): 70-4, 2003.
Article in English | MEDLINE | ID: mdl-12699068

ABSTRACT

Tumors growing within the host form dynamic aberrant tissue that consists of host components, including the stroma, an expanding vasculature and often chronic inflammation, in addition to the tumor cells themselves. These host components can contribute to, rather than limit, tumor expansion, whereas deprivation of vessel formation has the potential to confine tumors in small, clinically silent foci. Therapeutic inhibition of vessel formation could be best suited to preventive strategies aimed at the suppression of angiogenesis in primary tumors in subjects at risk, or of micrometastases after surgical removal of a primary tumor. Our analysis of potential cancer chemopreventive molecules including N-acetylcysteine, green tea flavonoids and 4-hydroxyphenyl-retinamide has identified antiangiogenic activities that could account--at least in part--for the tumor prevention effects observed with these compounds. These drugs appear to target common mechanisms of tumor angiogenesis that may permit identification of critical targets for antiangiogenic therapy and antiangiogenic chemoprevention.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Anticarcinogenic Agents/pharmacology , Catechin/analogs & derivatives , Fenretinide/pharmacology , Flavonoids/pharmacology , Neoplasms/prevention & control , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Catechin/pharmacology , Chemotaxis , Endothelium, Vascular/metabolism , Humans , Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/prevention & control , Tea , Vascular Endothelial Growth Factor A/metabolism
18.
Endocrinology ; 144(4): 1574-84, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12639942

ABSTRACT

Somatostatin was reported to inhibit Kaposi's sarcoma (KS) cell (KS-Imm) xenografts through an antiangiogenic activity. Here, we show that somatostatin blocks growth of established KS-Imm tumors with the same efficacy as adriamycin, a clinically effective cytotoxic drug. Whereas KS-Imm cells do not express somatostatin receptors (SSTRs), endothelial cells express several SSTRs, in particular SSTR3. We investigated the molecular mechanisms and receptor specificity of somatostatin inhibition of angiogenesis. Somatostatin significantly inhibited angiogenesis in vivo in the matrigel sponge assay; this inhibition was mimicked by the SSTR3 agonist L-796778 and reversed by the SSTR3 antagonist BN81658, demonstrating involvement of SSTR3. In vitro experiments showed that somatostatin directly affected different endothelial cell line proliferation through a block of growth-factor-stimulated MAPK and endothelial nitric oxide (NO) synthase (eNOS) activities. BN81658 reversed somatostatin inhibition of cell proliferation, NO production, and MAPK activity, indicating that SSTR3 activation is required for the effects of somatostatin in vitro. Finally in vivo angiogenesis assays demonstrated that eNOS inhibition was a prerequisite for the antiangiogenic effects of somatostatin, because high concentrations of sodium nitroprusside, an NO donor, abolished the somatostatin effects. In conclusion, we demonstrate that somatostatin is a powerful antitumor agent in vivo that inhibits tumor angiogenesis through SSTR3-mediated inhibition of both eNOS and MAPK activities.


Subject(s)
Hormones/pharmacology , Neovascularization, Pathologic/drug therapy , Nitric Oxide Synthase/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/pharmacology , Amides/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme Activation/drug effects , Humans , In Vitro Techniques , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitrobenzenes/pharmacology , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Umbilical Veins/cytology
19.
Gene Ther ; 9(13): 867-78, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12080381

ABSTRACT

The administration of different angiogenesis inhibitors by gene transfer has been shown to result in inhibition of tumor growth in animal tumor models, but the potency of these genes has been only partially evaluated in comparative studies to date. To identify the most effective anti-angiogenic molecule for delivery by retroviral vectors, we investigated the effects of angiostatin, endostatin and interferon(IFN)-alpha(1) gene transfer in in vivo models of breast cancer induced neovascularization and tumor growth. Moloney leukemia virus-based retroviral vectors for expression of murine angiostatin, endostatin and IFN-alpha(1) were generated, characterized, and used to transduce human breast cancer cell lines (MCF7 and MDA-MB435). Secretion of the recombinant proteins was confirmed by biological and Western blotting assays. Their production did not impair in vitro growth of these breast cancer cells nor their viability, and did not interfere with the expression of angiogenic factors. However, primary endothelial cell proliferation and migration in vitro were inhibited by supernatants of the transduced cells containing angiostatin, endostatin, and IFN-alpha(1). Stable gene transfer of the IFN-alpha(1) cDNA by retroviral vectors in both MCF7 and MDA-MB435 cells resulted in a marked and long-lasting inhibition of tumor growth in nude mice that was associated with reduced vascularization. Endostatin reduced the in vivo growth of MDA-MB435, but not MCF7 cells, despite similar levels of in vivo production, and angiostatin did not impair the in vivo growth of either cell line. These findings indicate heterogeneity in the therapeutic efficacy of angiostatic molecules delivered by viral vectors and suggest that gene therapy with IFN-alpha(1) and endostatin might be useful for treatment of breast cancer.


Subject(s)
Angiogenesis Inhibitors/genetics , Breast Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Moloney murine leukemia virus/genetics , Angiostatins , Animals , Breast Neoplasms/blood supply , Collagen/genetics , Endostatins , Female , Humans , Interferon-alpha/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Peptide Fragments/genetics , Plasminogen/genetics , Transduction, Genetic/methods , Tumor Cells, Cultured
20.
Mol Cell Biol Res Commun ; 4(6): 374-80, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11703097

ABSTRACT

The tuberous sclerosis 2 (TSC2) gene has been genetically mapped to a disease characterized by abnormal cell proliferation that results in the production of tumorous lesions in a variety of tissues. The molecular mechanism for TSC2 mediation of tuberous sclerosis is unclear but it appears to be related to its ability to cytoplasmically interact with a second gene, TSC1, mapping to the disease. These proteins are linked to constraints on cell cycle signaling pathways and therefore envisioned to function as tumor suppressor genes. In previous studies we have demonstrated TSC2 associations with steroid receptor family members and modulation of their gene expression capabilities. Here we provide evidence for TSC2 translocation to the nucleus and a possible role for phosphorylation in both TSC2 translocation and TSC2 modulation of steroid receptor-mediated transcription.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Phosphorylation , Repressor Proteins/biosynthesis , Animals , Blotting, Western , Cell Cycle , Cell Division , Cell Line , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Microscopy, Fluorescence , Plasmids/metabolism , Protein Binding , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Repressor Proteins/physiology , Signal Transduction , Staurosporine/pharmacology , Subcellular Fractions , Time Factors , Transcription, Genetic , Transfection , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...