Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Open Ophthalmol J ; 12: 110-120, 2018.
Article in English | MEDLINE | ID: mdl-30123376

ABSTRACT

BACKGROUND: The study aimed to evaluate iris thickness changes in patients with Primary Open Angle Glaucoma (POAG) or Ocular Hypertension (OHT) under treatment with Prostaglandin Analogues (PG). OBJECTIVES: Primary outcome measures were iris thickness at the region of Dilator Muscle Region (DMR) and Sphincter Muscle Region (SMR). DMR/SMR ratio was also evaluated. The secondary outcome was the correlation between PG treatment length and iris parameters. METHODS: The charts of patients with POAG or OHT who underwent Visante OCT were retrospectively selected. The patients were divided in a group using PG for at least 6 months and a group using hypotensive drops not including PG or alpha-adrenergic agonists. A third group included healthy subjects. RESULT: 98 subjects were selected. Patients with POAG or OHT using PG eyedrops showed a significant iris thickness reduction at DMR compared to healthy subjects and to patients using hypotensive eyedrops not containing PG. Significantly higher SMR thickness values were found in PG group compared to both control groups. DMR/SMR ratio significantly reduced in PG group. No correlation was found between PG treatment length and iris parameters. CONCLUSION: The present data indicate that PG treatment induced DMR thickness reduction and an increase in SMR thickness. These changes were not related to the duration of PG exposure.

2.
Nanoscale ; 6(14): 7924-33, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-24898567

ABSTRACT

Nanoparticles (NPs) emitting in the second biological near infrared (NIR) window of the electromagnetic spectrum have been successfully synthesized by growing a silica shell on the hydrophobic surface of OLEA/TOP PbS nanocrystals (NCs), by means of a reverse microemulsion approach, and subsequently decorated with biotin molecules. The fabrication of very uniform and monodisperse NPs, formed of SiO2 shell coated single core PbS NCs, has been demonstrated by means of a set of complementary optical and structural techniques (Vis-NIR absorption and photoluminescence spectroscopy, transmission electron microscopy) that have highlighted how experimental parameters, such as PbS NC and silica precursor concentration, are crucial to direct the morphology and optical properties of silica coated PbS NPs. Subsequently, the silica surface of the core-shell NPs has been grafted with amino groups, in order to achieve covalent binding of biotin to NIR emitting silica coated NPs. Finally the successful reaction with a green-fluorescent labelled streptavidin has verified the molecular recognition response of the biotin molecules decorating the PbS@SiO2 NP surface. Dynamic light scattering (DLS) and ζ-potential techniques have been used to monitor the hydrodynamic diameter and colloidal stability of both PbS@SiO2 and biotin decorated NPs, showing their high colloidal stability in physiological media, as needed for biomedical applications. Remarkably the obtained biotinylated PbS@SiO2 NPs have been found to retain emission properties in the 'second optical window' of the NIR region of the electromagnetic spectrum, thus representing attractive receptor-targeted NIR fluorescent probes for in vivo tumour imaging.


Subject(s)
Biotin/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Amines/chemistry , Fluorescein-5-isothiocyanate/chemistry , Humans , Lead/chemistry , Nanoparticles/ultrastructure , Neoplasms/diagnosis , Particle Size , Spectroscopy, Near-Infrared , Streptavidin/chemistry , Streptavidin/metabolism , Sulfides/chemistry
3.
Leukemia ; 26(2): 225-35, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21844875

ABSTRACT

Childhood acute myeloid leukemia (AML) is a hematological malignancy in which tumor burden is continuously replenished by leukemic-initiating cells (ICs), which proliferate slowly and are refractory to chemotherapeutic agents. We investigated whether interleukin (IL)-12, an immuno-modulatory cytokine with anti-tumor activity, may target AML blasts (CD45(+)CD33(+)) and populations known to contain leukemia ICs (that is, CD34(+)CD38(-), CD33(+)CD38(+) and CD44(+)CD38(-) cells). We demonstrate for the first time that: i) AML blasts and their CD34(+)CD38(-), CD33(+)CD38(+), CD44(+)CD38(-) subsets express the heterodimeric IL-12 receptor (IL-12R), ii) AML cells injected subcutaneously into NOD/SCID/Il2rg(-/-) (NSG) mice developed a localized tumor mass containing leukemic ICs and blasts that were virtually eliminated by IL-12 treatment, iii) AML cells injected intravenously into NSG mice engrafted within the first month in the spleen, but not in bone marrow or peripheral blood. At this time, IL-12 dramatically dampened AML CD45(+)CD33(+), CD34(+)CD38(-), CD33(+)CD38(+) and CD44(+)CD38(-) populations, only sparing residual CD33(+)CD38(+) cells that did not express IL-12Rß2. From 30 to 60 days after the initial inoculum, these IL-12-unresponsive cells expanded and metastasized in both control and IL-12-treated NSG mice. Our data indicate that the absence of IL-12Rß2 in pediatric AML cells favours leukemia progression in NOD/SCID/IL2Rγc-deficient mice.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Interleukin-12 Receptor beta 2 Subunit/metabolism , Leukemia, Myeloid, Acute/pathology , Adolescent , Adult , Animals , Cell Division , Child , Child, Preschool , Disease Progression , Female , Flow Cytometry , Humans , Infant , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Real-Time Polymerase Chain Reaction , Sialic Acid Binding Ig-like Lectin 3
4.
Leukemia ; 26(6): 1365-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22193967

ABSTRACT

Interleukin (IL)-23 and IL-27 are pro-inflammatory cytokines that share functional and structural similarities and may exert anti-tumor activities against solid and hematological malignancies. Here, we asked whether IL-23 and IL-27, alone or in combination, may act directly against human follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL) cells. In this study, we demonstrated for the first time that human primary FL and DLBCL cells expressed complete and functional IL-23 and IL-27 receptors (R) and that IL-23 and IL-27 exerted anti-tumor activities in vitro and in vivo through different and complementary mechanisms. In vivo studies using severe combined immunodeficiency /non-obese diabetic mice-injected subcutaneously with human SU-DHL-4 cell line revealed that IL-23 inhibited directly tumor-cell proliferation, whereas IL-27 impaired the angiogenic program of lymphoma cells resulting in strong reduction of cell growth. In addition, combined treatment of IL-23 and IL-27 amplified the anti-tumor effects in vivo as compared with administration of each cytokine alone. These anti-tumor mechanisms were confirmed by in vitro experiments performed with primary lymphoma cells and cell lines. Our results strongly encourage the development of future clinical trials to evaluate the toxicity and efficacy of the IL-23 and IL-27 in lymphoma patients.


Subject(s)
Interleukin-17/therapeutic use , Interleukin-23/therapeutic use , Lymphoma, Follicular/prevention & control , Lymphoma, Large B-Cell, Diffuse/prevention & control , Aged , Aged, 80 and over , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Chickens , Chorioallantoic Membrane , DNA, Neoplasm/genetics , Drug Synergism , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
5.
Leukemia ; 25(12): 1815-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21701492

ABSTRACT

B-acute lymphoblastic leukemia (B-ALL) represents the most common pediatric hematological tumor that derives from the aberrant proliferation of early B lymphocytes in the bone marrow. Although most of the B-ALL children take advantage from current therapeutic protocols, some patients relapse and need alternative therapies. With this background, we investigated whether interleukin (IL)-27, an immunomodulatory cytokine with antitumor properties, may function as an antitumor agent against pediatric B-ALL cells. Here we show for the first time that pediatric B-ALL cells functional IL-27R and that IL-27 dampens directly tumor growth in vivo and in vitro through mechanisms elucidated in this study. The novelty of these results deals with the first demonstration that (1) B-ALL cells from pediatric patients injected intravenously (i.v.) into NOD/SCID/Il2rg(-/-) (NSG) mice gave rise to leukemic spreading that was severely hampered by IL-27; (2) IL-27-treated mice, compared with controls, showed significant reduction of putative B-ALL-initiating cells and blasts in the peripheral blood (PB), bone marrow (BM) and spleen; and that (3) IL-27 reduced in vitro B-ALL cell proliferation and angiogenesis, induced apoptosis and downregulated miR-155. Our results strongly encourage the development of future clinical trials to evaluate the toxicity and efficacy of IL-27 in childhood B-ALL patients.


Subject(s)
Apoptosis , Interleukin-17/therapeutic use , Leukemia, B-Cell/pathology , Leukemia, B-Cell/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Adolescent , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation , Chickens , Child , Child, Preschool , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , Female , Flow Cytometry , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Infant , Leukemia, B-Cell/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Am J Transplant ; 7(1): 201-10, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17061985

ABSTRACT

Quilty effect (QE) is a frequent, yet enigmatic feature of cardiac allograft, since it is apparently devoid of clinical significance, though its association with acute (A) rejection (R) is strongly suspected. It was observed in 126/379 biopsies from 22 patients during the first posttransplant year. Most grade (G)2R biopsies displayed a concomitant QE. The following features typical of QE were identified: (a) focal angiogenesis and lymphangiogenesis associated with bFGF, VEGF-C and VEGF-A expression, (b) marked infiltrate of CD4(+)T and CD20(+)B followed by CD8(+)T lymphocytes arranged around PNAd(+)HEV-like vessels. Most QE appear as distinct B-T-cell-specific areas with lymphoid follicles sometimes endowed with germinal center-like structures containing VCAM-1(+)CD21(+)FDC and CD68(+)macrophages, which frequently expressed CXCL13. These cells were also found in mantle-like zones, where small lymphocytes expressed CXCR5, otherwise in the whole area of not clustered lymphoid aggregates. CXCL13 was also expressed, in association with CD20(+)B lymphocyte recruitment, in G2R biopsies obtained from patients with recurrent AR. QE has features of a tertiary lymphoid tissue suggesting an attempt, by the heart allograft, to mount a local response to a persistent alloantigen stimulation resulting in aberrant CXCL13 production, as also occurs in recurrent AR. CXCL13-CXCR5 emerge as a common molecular pathway for QE and recurrent episodes of AR.


Subject(s)
Chemokines, CXC/metabolism , Graft Rejection/pathology , Heart Transplantation/adverse effects , Lymphangiogenesis , Myocardium/pathology , Receptors, Chemokine/metabolism , Aged , Angiogenic Proteins/analysis , Biopsy , Cell Movement , Chemokine CXCL13 , Female , Graft Rejection/etiology , Humans , Lymphocytes , Macrophages , Male , Middle Aged , Neovascularization, Pathologic , Receptors, CXCR5 , Recurrence
7.
J Pathol ; 209(3): 400-10, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16583358

ABSTRACT

Heart allograft outcome is unpredictable and acute rejection episodes still occur despite the improvement of immunosuppressive regimens. We therefore investigated whether the immunopathological profile of endomyocardial biopsies might underlie the variations in the clinical course of a graft. Biopsies from transplanted patients were analysed by histology, immunohistochemistry (associated with digital image analysis), confocal and electron microscopy to determine the type and the functional state of leukocytes infiltrating the myocardium, together with their ultrastructural features and those of the graft itself. In comparison with biopsies with grade 0R or grade 1R rejection, those from patients with grade 2R rejection displayed significant infiltration of macrophages, T lymphocytes, and CD83+ and DC-SIGN+ dendritic cells. Fifty-seven per cent were invaded by CD20+B lymphocytes, most of which expressed CD69 activation marker and cooperated in interleukin-12 production, and by CD69+CD94+NK cells expressing interferon-gamma. Ultrastructural signs of myocyte degeneration and microvessel rupture by NK cells were frequent. These patients developed recurrent episodes of acute allograft rejection. Endomyocardial B and NK infiltrates are involved in the dynamics of allograft rejection and are associated with a high risk of its recurrence. Immunopathological assessment of endomyocardial biopsies may thus serve to forecast the probable outcome of a heart allograft.


Subject(s)
B-Lymphocytes/immunology , Endocardium/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Killer Cells, Natural/immunology , Adult , Aged , Biopsy , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Endocardium/pathology , Endocardium/ultrastructure , Female , Humans , Immunoenzyme Techniques , Immunophenotyping , Interleukin-12/biosynthesis , Male , Microscopy, Confocal , Microscopy, Electron , Middle Aged , Myocardial Ischemia/immunology , Recurrence
9.
Int J Immunopathol Pharmacol ; 17(2): 171-80, 2004.
Article in English | MEDLINE | ID: mdl-15171818

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory disease of unknown aetiology and pathogenesis. The presence in the colonic mucosa of reactive cells expressing proinflammatory cytokines and chemokines is associated with high levels of IL-10, an anti-inflammatory cytokine. Our aim was to investigate the role of IL-10 and the beta chemokine LEC/CCL16 selectively up-regulated by IL-10 in inflammatory cell recruitment and cytokine and chemokine production during UC. We studied histologically, immunohistochemically and ultrastructurally colonic biopsies from 20 active UC patients and 10 control specimens taken far from any macroscopically detectable lesion in age and sex-matched patients with noninflammatory bowel disease. In active UC, immature dendritic cells (DCs) in the LP are associated with IL-10 in the T cell rich area. Furthermore, most of the LP-infiltrating macrophages strongly expressed LEC/CCL16, a chemokine upregulated by IL-10. To evaluate if LEC/CCL16 plays a role in the inflammatory reaction present in UC, we performed morphological studies in mice injected s.c. with syngeneic tumor cells engineered to produce LEC/CCL16. We found that the LEC protein locally released by LEC-gene-transfected tumor cells is a potent proinflammatory chemokine that induces the recruitment of a reactive infiltrate, and an angiogenic process mirroring that in human UC.


Subject(s)
Chemokines, CC/biosynthesis , Colitis, Ulcerative/metabolism , Animals , Cell Division/physiology , Cell Line, Tumor , Clone Cells , Colitis, Ulcerative/pathology , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Interleukin-10/biosynthesis , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Mice , Mice, Inbred BALB C , Microscopy, Electron , RNA, Messenger/biosynthesis , Transfection , Up-Regulation/physiology
10.
Br J Cancer ; 90(11): 2210-8, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15150552

ABSTRACT

Interferon-gamma (IFN-gamma) directs T helper-1 cell differentiation and mediates antitumour effects in preclinical models. However, high-dose IFN-gamma is toxic in vivo, and IFN-gamma-transfected neuroblastoma (NB) cells secreting high amounts of the cytokine may be lost due to cell apoptosis or differentiation. Two human NB cell lines (ACN and SK-N-BE2(c)) differing as to genetic and phenotypic features were transfected with the human IFN-gamma gene and selected on the grounds of the low concentrations of IFN-gamma produced. In both IFN-gamma-transfected cell lines, autocrine and paracrine activation of IFN-gamma-mediated pathways occurred, leading to markedly reduced proliferation rate, to increased expression of surface HLA and CD40 molecules and of functional TNF binding sites. ACN/IFN-gamma cells showed a significantly delayed tumorigenicity in nude mice as compared to parental cells. ACN/IFN-gamma tumours were smaller, with extensive necrotic area as a result of a damaged and defective microvascular network. In addition, a significant reduction in the proliferation index was observed. This is the first demonstration that IFN-gamma inhibits in vivo proliferation of NB cell by acting on the tumour cell itself. This effect adds to the immunoregulatory and antiangiogenic activities operated by IFN-gamma in syngeneic tumour-bearing hosts.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Division/drug effects , Interferon-gamma/pharmacology , Neuroblastoma/immunology , Neuroblastoma/pathology , Animals , DNA Primers , Female , Flow Cytometry , Genetic Therapy , Humans , Interferon-gamma/biosynthesis , Mice , Mice, Nude , Neoplasms, Experimental , Neovascularization, Pathologic , Phenotype , Receptors, Tumor Necrosis Factor/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
11.
Buenos Aires; Proyecto Ambientes Libres de Humo de Tabaco. Organización Panamericana de la Salud; 2004. 80 p. (115004).
Monography in Spanish | BINACIS | ID: bin-115004
12.
Buenos Aires; Proyecto Ambientes Libres de Humo de Tabaco. Organización Panamericana de la Salud; 2004. 80 p.
Monography in Spanish | BINACIS, MINSALCHILE | ID: biblio-1217372
14.
Scand J Immunol ; 55(5): 484-92, 2002 May.
Article in English | MEDLINE | ID: mdl-11975760

ABSTRACT

Cytokine-immunoglobulin (Ig)-fusion proteins have attracted increasing interest as antitumour agents. Here, we have investigated the antimetastatic and antitumour responses elicited in vivo by mammary adenocarcinoma cells (TS/A) engineered to secrete interleukin (IL)-2-IgG fusion proteins. TS/A cells were transfected with DNA coding for IL-2-IgG2b, IgG2b or IL-2, and injected subcutaneously into syngeneic mice. Animals injected with TS/A-IL-2 or TS/A-IL-2-IgG2b both efficiently rejected tumours, whereas treatment with parental cells or TS/A-IgG2b was lethal. Interestingly, only mice vaccinated with IL-2-IgG2b fusion protein-secreting cells showed a long-lasting protective immunity against a later challenge with parental tumour cells. Moreover, the metastatic potential of TS/A-IL-2-IgG2b-transfected cells was dramatically decreased compared with TS/A-IL-2-cells, with a virtual absence of lung metastases after intravenous injection. Adenocarcinomas secreting IL-2-IgG2b exhibited a more prominent, early and persistent infiltration of CD4+, CD8+ and natural killer (NK) cells than TS/A-IL-2 cells. Therefore, upon transfection into adenocarcinoma cells, the IgG2b part of IL-2 fusion protein exerts intriguing added antitumour properties over IL-2 alone, thus contributing to a long-lasting tumour immunity, probably by the recruitment of specific immune effector cells. These findings suggest a promising new oncotherapeutic strategy for poorly immunogenic tumours: vaccination with tumour cells engineered to secrete IL-2-IgG2b fusion protein.


Subject(s)
Adenocarcinoma/immunology , Immunoglobulin G/immunology , Interleukin-2/immunology , Mammary Neoplasms, Experimental/immunology , Recombinant Fusion Proteins/immunology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Growth Inhibitors/genetics , Growth Inhibitors/immunology , Immunoglobulin G/genetics , Immunohistochemistry , Immunotherapy , Interleukin-2/genetics , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Transfection , Tumor Cells, Cultured
15.
J Exp Med ; 194(9): 1195-205, 2001 Nov 05.
Article in English | MEDLINE | ID: mdl-11696586

ABSTRACT

Transgenic Balb/c mice expressing the transforming rat HER-2/neu oncogene develop early and multifocal mammary carcinomas. Within the first 5 months of life the tissue-specific expression of HER-2/neu causes a progression in all their 10 mammary glands from atypical hyperplasia to invasive carcinoma. It was previously observed that chronic administration of interleukin (IL)-12 increased tumor latency, but every mouse eventually succumbed to multiple carcinomas. A significant improvement in tumor prevention was sought by administering allogeneic mammary carcinoma cells expressing HER-2/neu combined with systemic IL-12. This treatment reduced tumor incidence by 90% and more than doubled mouse lifetime. For the maximum prevention p185(neu) antigen must be expressed by allogeneic cells. IL-12 treatment strongly increased the cell vaccine efficacy. The mammary glands of mice receiving the combined treatment displayed a markedly reduced epithelial cell proliferation, angiogenesis, and HER-2/neu expression, while the few hyperplastic foci were heavily infiltrated by granulocytes, macrophages, and CD8(+) lymphocytes. Specific anti-HER-2/neu antibodies were produced and a nonpolarized activation of CD4(+) and CD8(+) cells secreting IL-4 and interferon (IFN)-gamma were evident. A central role for IFN-gamma in the preventive effect was proven by the lack of efficacy of vaccination in IFN-gamma gene knockout HER-2/neu transgenic Balb/c mice. A possible requirement for IFN-gamma is related to its effect on antibody production, in particular on IgG2a and IgG2b subclasses, that were not induced in IFN-gamma knockout HER-2/neu mice. In conclusion, our data show that an allogeneic HER-2/neu-expressing cell vaccine combined with IL-12 systemic treatment can prevent the onset of genetically determined tumors.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/immunology , Interleukin-12/immunology , Mammary Neoplasms, Experimental/prevention & control , Receptor, ErbB-2/physiology , Animals , Breast/pathology , CD4-Positive T-Lymphocytes/immunology , Cell Transplantation , Female , Immunity, Cellular/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/administration & dosage , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Rats , Receptor, ErbB-2/genetics , Transplantation, Homologous , Tumor Cells, Cultured , Vaccination/methods
16.
Gene Ther ; 8(1): 75-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11402305

ABSTRACT

The HER-2/neu proto-oncogene is overexpressed in 20-30% of human breast cancers and is associated with high recurrence risk. The oncogenic potential of HER-2/neu, together with its elevated expression in tumors, cell surface localization, and immunogenicity in some patients, make this oncoprotein an ideal target for immunotherapeutic approaches. To test the efficacy of immune-based strategies in eliciting an antitumor response, we used the N#202 transgenic mouse model engineered to overexpress the rat neu proto-oncogene under the control of the mouse mammary tumor virus promoter; females of this line develop spontaneous focal mammary tumors by 6 months of age. Transgenic mice immunized intramuscularly with a HER-2 cDNA ligated into the VR1012 (VICAL) expression vector under the control of the cytomegalovirus promoter developed significantly fewer spontaneous tumors as compared with mice injected with the empty vector (P < 0.0001) or not injected (P = 0.0006). However, this protection was observed only when immunization was started in 3-month-old but not in 6-month-old mice. These data suggest that the xenogeneic HER-2 DNA sequence can break immune tolerance to rat neu in transgenic N#202 mice and induce protective immunity that impairs the neu oncogene-driven progression of mammary carcinogenesis. The preventive effect achieved by our immunological approach appeared not to be based on anti-neu specific B and T cell immune attacks but was more possibly based on different mechanisms including aspecific and inflammatory immunological responses.


Subject(s)
Cancer Vaccines , Mammary Neoplasms, Experimental/prevention & control , Neoplasm Proteins/metabolism , Receptor, ErbB-2/metabolism , Vaccines, DNA , Age Factors , Animals , Cytotoxicity, Immunologic , Female , Fluorescent Antibody Technique, Indirect , Immunization Schedule , Lymphocyte Culture Test, Mixed , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Neoplasm Proteins/immunology , Proto-Oncogene Mas , Rats , Receptor, ErbB-2/immunology
17.
Kidney Int ; 59(6): 2299-308, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11380834

ABSTRACT

BACKGROUND: Several alterations of protein structure and function have been reported in uremia. Impairment of a transmethylation-dependent protein repair mechanism possibly related to a derangement in homocysteine metabolism is also present in this condition, causing erythrocyte membrane protein damage. Homocysteine may affect proteins via the accumulation of its parent compound S-adenosylhomocysteine (AdoHcy), a powerful in vivo methyltransferase inhibitor. However, since plasma homocysteine is mostly protein bound, a direct influence on protein structures cannot be ruled out. We measured the levels of L-isoaspartyl residues in plasma proteins of uremic patients on hemodialysis. These damaged residues are markers of molecular age, which accumulate when transmethylation-dependent protein repair is inhibited and/or protein instability is increased. METHODS: L-isoaspartyl residues in plasma proteins were quantitated using human recombinant protein carboxyl methyl transferase (PCMT). Plasma concentrations of homocysteine metabolites were also measured under different experimental conditions in hemodialysis patients. RESULTS: The concentration of damaged plasma proteins was increased almost twofold compared to control (controls 147.83 +/- 17.75, uremics 282.80 +/- 26.40 pmol of incorporated methyl groups/mg protein, P < 0.003). The major protein involved comigrated with serum albumin. Although hyperhomocysteinemia caused a redistribution of thiols bound to plasma proteins, this mechanism did not significantly contribute to the increase in isoaspartyl residues. The S-adenosylmethionine (AdoMet)/AdoHcy concentration ratio, an indicator of the flux of methyl group transfer, was altered. This ratio was partially corrected by folate treatment (0.385 +/- 0.046 vs. 0.682 +/- 0.115, P < 0.01), but protein L-isoaspartate content was not. CONCLUSIONS: Plasma protein damage, as determined by protein L-isoaspartyl content, is increased in uremia. This alteration is to be ascribed to an increased protein structural instability, rather than the effect of hyperhomocysteinemia.


Subject(s)
Aspartic Acid/blood , Blood Proteins/metabolism , Homocysteine/blood , S-Adenosylhomocysteine/blood , Uremia/blood , Aspartic Acid/analysis , Biomarkers , Blood Proteins/chemistry , Chromatography, High Pressure Liquid , Folic Acid/administration & dosage , Hematinics/administration & dosage , Humans , Hyperhomocysteinemia/metabolism , Methylation , Protein D-Aspartate-L-Isoaspartate Methyltransferase , Protein Methyltransferases , Renal Dialysis , Serum Albumin/metabolism , Uremia/drug therapy
18.
Gene Ther ; 8(6): 447-52, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11313823

ABSTRACT

An assessment was made of the effectiveness of DNA vaccination in prevention of the mammary adenocarcinomas of BALB/c female mice transgenic for the activated rat Her-2/neu oncogene. Atypical hyperplasia is evident in their mammary glands when they are 6 weeks old and in situ carcinoma by the 13th week. Palpable invasive carcinomas appear around the 17th week and are always evident in all 10 glands by the 33rd week. Intramuscular vaccinations with 100 microg plasmid DNA encoding the extracellular domain of the Her-2/neu p185 (ECD) performed at the 6th, 12th, 18th and 24th week provided no significant protection, whereas those ECD plasmids in which the DNA coding for the immunomodulatory 163-171 (VQGEESNDK) nonapeptide of human IL1beta (ECD-IL1betap) had been inserted both delayed carcinogenesis and reduced tumor multiplicity. This reduction was associated with a marked immune-inflammatory reaction and a conspicuous leukocyte infiltrate located in the stroma surrounding the hyperplastic mammary ductul-alveolar structures. It was also directly correlated with a high anti-p185(neu) antibody production and an immunoglobulin switch to IgG2a and IgA. No anti-p185(neu) cytotoxic response was found. No significant protection was obtained when the DNA coding for the non-active peptide 189-197 of IL1beta was inserted.


Subject(s)
Genes, erbB-2 , Genetic Therapy/methods , Interleukin-1/genetics , Mammary Neoplasms, Experimental/prevention & control , Vaccines, DNA/administration & dosage , Animals , Antibodies/blood , Female , Genes, erbB-2/immunology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Rats , Statistics, Nonparametric
19.
Cancer Res ; 61(7): 2809-12, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306448

ABSTRACT

With a slight asynchronous but consistent progression, all of the mammary glands of female BALB/c mice transgenic for the transforming rat HER-2/neu oncogene progress to atypical hyperplasia and to invasive carcinoma. Previous studies have shown that chronic administration of interleukin (IL) 12 started at the 2nd week of age hampers this progression because of its ability to inhibit tumor angiogenesis and activate a nonspecific immune response. Here we show that a similar inhibition is achieved when 7-week-old mice with fully blown atypical hyperplasia receive a weekly injection of 100 ng IL-12 for 16 times. This lower-dose and later IL-12 administration induces high and sustained levels of serum IFN-gamma equivalent to those elicited by more frequent administrations. A lower-dose and less toxic treatment may thus be envisaged as a possible option in the management of preneoplastic mammary lesions.


Subject(s)
Genes, erbB-2/genetics , Interleukin-12/pharmacology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/prevention & control , Animals , Dose-Response Relationship, Drug , Female , Hyperplasia/pathology , Interferon-gamma/biosynthesis , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic
20.
Cancer Res ; 61(8): 3518-23, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309316

ABSTRACT

T-lymphocytes (LYs) from normal and IFN-gamma knockout mice were activated by anti-CD3 and anti-CD28 antibodies and cultured in inserts in the presence of interleukin (IL)-12 (IL-12-activated LYs) or not (activated LYs). Their ability to modulate the genetic programs of two tumor lines growing at the bottom of transwells was evaluated. cDNA gene expression array, reverse transcription-PCR, and protein expression showed that LPS, transcription termination factor 1, transforming growth factor, and fibroblast growth factor genes were up-modulated by factors other than IFN-gamma released by activated LYS: The high levels of IFN-gamma released by normal IL-12-activated LYs up-modulated the expression of STAT1, IRF-1, LMP2, LMP7, monokine induced by IFN-gamma, monocyte chemoattractant protein 1, and angiopoietin 2 genes but down-modulated the expression of vascular endothelial growth factor. PA28, IFN-inducible protein 10, inducible NO synthetase, and macrophage-inhibitory protein 2 genes were up-modulated by factors released only by IL-12-activated LYs apart from IFN-gamma. The opposite modulations of vascular endothelial growth factor expression and of angiopoietin 2, monokine induced by IFN-gamma, IFN-inducible protein 10, and inducible NO synthetase by IL-12-activated LYs fit in well with the inhibition of angiogenesis that characterizes the antitumor activity of IL-12. T-LYs thus modify a tumor's behavior so that it becomes a party to its own inhibition.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/immunology , Interleukin-12/immunology , Lymphocyte Activation/immunology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD28 Antigens/immunology , CD3 Complex/immunology , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...