Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cureus ; 15(12): e50019, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38186456

ABSTRACT

The critical need for new diagnostic and prognostic methods is highlighted by the fact that breast cancer continues to be the top cause of cancer-related deaths globally. Due to the dysregulation of long non-coding RNAs (lncRNAs) in numerous malignancies, they have become potential biomarkers in cancer. Recent research has focused on the lncRNA HOTTIP (HOXA transcript at the distal tip), which has a function in breast cancer metastasis and carcinogenesis. Until recently, HOTTIP had only been measured in cancer tissues and specimens. The aim of this study is to assess the amounts of the lncRNA HOTTIP in the blood serum of 46 breast cancer patients using real-time PCR analysis and identify the relationships between HOTTIP expression and several known clinical and pathological factors, including tumor grade, stage, lymph node involvement, hormone receptor status, and cell proliferation. The results of the study confirmed a positive relation of HOTTIP expression and breast cancer aggressiveness and metastatic behavior. The analysis results showed elevated HOTTIP values in stage III and T3/T4 tumors with multifocal characteristics and in lymph node involvement. Our findings raise the possibility of HOTTIP serving as a future prognostic biomarker for breast cancer patients.

2.
Cancer Immunol Immunother ; 68(1): 57-70, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30276443

ABSTRACT

Breast cancer (BCa) is a heterogeneous disease with different histological, prognostic and clinical aspects. Therefore, the need for identification of novel biomarkers for diagnosis, prognosis and monitoring of disease, as well as treatment outcome prediction remains at the forefront of research. The search for circulating elements, obtainable by simple peripheral blood withdrawal, which may serve as possible biomarkers, constitutes still a challenge. In the present study, we have evaluated the expression of 6 circulating miRNAs, (miR-16, miR-21, miR-23α, miR-146α, miR-155 and miR-181α), in operable BCa patients, with non-metastatic, invasive ductal carcinoma, not receiving neoadjuvant chemotherapy. These miRNAs, known to be involved in both tumor cell progression and immune pathways regulation, were analyzed in relation to circulating cytokines, tumor immune-cell infiltration and established prognostic clinicopathological characteristics. We have identified three different clusters, with overall low (C1), moderate (C2) or high (C3) expression levels of these six circulating miRNAs, which define three distinct groups of non-metastatic BCa patients characterized by different clinicopathological and immune-related characteristics, with possibly different clinical outcomes. Our data provide the proof-of-principle to support the notion that, up- or down-regulation of the same circulating miRNA may reflect different prognosis in BCa. Nonetheless, the prognostic and/or predictive potential of these three "signatures" needs to be further evaluated in larger cohorts of BCa patients with an, at least, 5-year clinical follow-up.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Biomarkers, Tumor/blood , Breast Neoplasms/classification , Breast Neoplasms/immunology , Cytokines/blood , Humans , Leukocytes, Mononuclear/metabolism , MicroRNAs/blood , Prognosis
3.
Cancer Immunol Immunother ; 66(12): 1631-1642, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28871365

ABSTRACT

Melanoma, like most solid tumors, is highly heterogeneous in terms of invasive, proliferative, and tumor-initiating potential. This heterogeneity is the outcome of differential gene expression resulting from conditions in the tumor microenvironment and the selective pressure of the immune system. To investigate possible signatures combining immune-related gene expression and lymphocyte infiltration, we established a preclinical model using B16.F1-derived clones, in the context of melanoma aggressiveness. Combinatorial analyses revealed that tumors concomitantly expressing low levels of Tnf-a, Pd-1, Il-10, Il-1ra, Ccl5, Ido, high Il-9, and with low infiltration by CD45+, CD3+, CD4+ and CD8+ cells and a high CD4+:CD8+ T cell ratio exhibited the most aggressive growth characteristics. Overall, these results support the notion that the intratumoral immunologic network molds aggressive melanoma phenotypes.


Subject(s)
Melanoma/immunology , Animals , Cell Line, Tumor , Female , Humans , Melanoma/pathology , Mice, Inbred C57BL , Tumor Microenvironment
4.
Anticancer Res ; 37(1): 143-148, 2017 01.
Article in English | MEDLINE | ID: mdl-28011484

ABSTRACT

Numerous studies have revealed a variety of pathways involved in the development of melanoma, however, the molecular and genetic divergence of underlying mechanisms remain vague. In a mouse model, we studied the expression pattern of insulin-like growth factor 2 mRNA-binding protein 1 (Igf2bp1) and target genes microphthalmia-associated transcription factor (Mitf), v-myc avian myelocytomatosis viral oncogene homolog (Myc), B-cell lymphoma 2 (Bcl2), prothymosin alpha (Ptma) and melan-A (Mlana) in relation to tumor-growth characteristics. The in vivo expression of the aforementioned genes was assessed by quantitative Real Time-Polymerase Chain Reaction (RT-PCR) in tumors established by B16-F1-derived clones. Gene expression was correlated with tumor growth characteristics. Simultaneous expression of elevated levels of Myc, Igf2bp1, Ptma and Mitf characterizes tumors with a more aggressive phenotype. Our findings introduce a tumor-specific molecular signature possibly associated with melanoma heterogeneity. The concomitant overexpression of key molecules such as IGF2BP1, PTMA, MYC and MITF could serve as prognostic or predictive marker.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Melanoma, Experimental/genetics , Skin Neoplasms/genetics , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genotype , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Phenotype , Protein Precursors/genetics , Protein Precursors/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Thymosin/analogs & derivatives , Thymosin/genetics , Thymosin/metabolism , Time Factors , Tumor Burden
5.
Stem Cells Dev ; 23(20): 2501-12, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24915579

ABSTRACT

Mesenchymal stem cells (MSCs) are a population of cells harboring in many tissues with the ability to differentiate toward many different lineages. Unraveling the molecular profile of MSCs is of great importance due to the fact that these cells are very often used in preclinical and clinical studies. We have previously reported the expression of insulin-like growth factor 2 mRNA binding protein 1 (IGF2BP1) an oncofetal mRNA-binding protein-in different stem cell types such as bone marrow (BM)-MSC and umbilical cord blood (UCB)-hematopoietic stem cells. Here, we demonstrate that MSCs of adipose tissue, BM, and UC origin have a differential pattern of IGF2BP1 and ten-eleven-translocate 1/2 (TET1/2) expression that could correlate with their proliferation potential. Upon IGF2BP1 interference, a significant reduction of cell proliferation is observed, accompanied by reduced expression of c-MYC and GLI1 and increased p21. We also present, for the first time, evidence that IGF2BP1 is epigenetically regulated by TET1 and TET2 demethylases. Specifically, we show that TET1 directly binds to the promoter of IGF2BP1 gene and affects the hydroxymethylation status of its promoter. These results indicate that IGF2BP1 and TET1/2 contribute to the stemness of MSCs, at least regarding their proliferative potential.


Subject(s)
Cell Proliferation/physiology , DNA-Binding Proteins/biosynthesis , Epigenesis, Genetic/physiology , Gene Expression Regulation, Enzymologic/physiology , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins/biosynthesis , RNA-Binding Proteins/biosynthesis , Cells, Cultured , DNA-Binding Proteins/genetics , Dioxygenases , Female , Humans , Male , Mesenchymal Stem Cells/cytology , Mixed Function Oxygenases , Proto-Oncogene Proteins/genetics , RNA-Binding Proteins/genetics
6.
Int J Radiat Biol ; 89(5): 319-25, 2013 May.
Article in English | MEDLINE | ID: mdl-23311575

ABSTRACT

PURPOSE: Overexpression of human epidermal growth factor receptor-2 (HER-2/neu) in breast cancer patients is a prerequisite for treatment with trastuzumab. In the present study, we demonstrate by fluorescence in situ hybridization (FISH) analysis that HER-2/neu gene amplification and chromosome 17 (CEP17) polysomy can be induced by irradiation in human breast cancer cell lines with low basal level of HER-2/neu. MATERIALS AND METHODS: The irradiation-induced HER-2/neu gene amplification and CEP17 polysomy enhanced HER-2/neu at the protein level in both human MDA-MB-231 and MDA-MB-435 breast cancer cell lines which was determined by immunohistochemistry and fluorescence analysis and was correlated with mRNA levels. RESULTS: Irradiation affected to a high degree the responsiveness of both cell lines to in vitro treatment with trastuzumab. The direct antiproliferative effect of trastuzumab, as well as its capacity to induce natural killer (NK) cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC), was considerably higher in the irradiated tumor cells compared to their non-irradiated counterparts. CONCLUSION: Our data demonstrate that irradiation induces HER-2/neu gene amplification and CEP17 polysomy thereby enhancing expression of this protein in breast cancer cell lines rendering them susceptible to treatment with trastuzumab. They also suggest that patients with HER-2/neu negative inoperable tumors undergoing local radiation therapy may benefit from treatment with trastuzumab.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/pathology , Combined Modality Therapy/methods , Gamma Rays/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chromosome Aberrations/drug effects , Chromosome Aberrations/radiation effects , Chromosomes, Human, Pair 17/drug effects , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 17/radiation effects , Drug Resistance, Neoplasm/radiation effects , Humans , In Situ Hybridization, Fluorescence , Trastuzumab
7.
Eur J Pharmacol ; 666(1-3): 173-82, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21620827

ABSTRACT

The labdane diterpene sclareol has demonstrated significant cytotoxicity against human tumor cell lines and human colon cancer xenografts. Therefore, there is need to elucidate the mode of action of this compound as very little information is known for the anticancer activity of sclareol and other labdane diterpenes, in general. COMPARE analysis of GI(50) values for a number of human cancer cell lines was initially implicated in an effort to assign a putative mechanism of action to the compound. Sclareol-induced cell cycle arrest and apoptosis were assessed by flow cytometry and Western blot analyses. Finally, the anticancer ability of sclareol in vivo was assessed by using human colon cancer xenograft/mouse models. Sclareol arrested in vitro the growth of p53-deficient (HCT116(p53-/-)) human colon cancer cells and subsequently induced apoptosis by activating both caspases-8 and -9. Intraperitoneal administration of liposome-encapsulated sclareol at the maximum tolerated dose induced a marked growth suppression of HCT116(p53-/-) tumors established as xenografts in immunodeficient NOD/SCID mice. In conclusion, we demonstrate herein that sclareol kills human tumor cells by inducing arrest at the G(1)-phase of the cell cycle followed by apoptosis that involves activation of caspases-8, -9 and -3 via a p53-independent mechanism. These findings suggest that liposome-encapsulated sclareol possesses chemotherapeutic potential for the treatment of colorectal and other types of human cancer regardless of the p53-status.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Diterpenes/pharmacology , Xenograft Model Antitumor Assays , Animals , Caspases/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Cyclic AMP/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Diterpenes/administration & dosage , Enzyme Activation/drug effects , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Liposomes , Male , Mice , Tumor Suppressor Protein p53/metabolism
8.
Stem Cells Dev ; 20(9): 1549-61, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21204633

ABSTRACT

Mesenchymal stem cells (MSCs) are a very important adult stem cell population with a multitude of potential applications in regenerative medicine. The thorough characterization of the bone marrow MSC (BM-MSC) population derived from the BALB/c species was essential, considering the significance of the murine model amongst animal models. In the present study, we examined the effect of gender, age, and in vitro culture on the basic properties (proliferation, differentiation, and immunosuppressive potential) of BM-MSCs. We found a decline in the progenitor frequencies from the BM of adult mice, lower MSC frequencies in all female donors, and an increase in the BM-MSC proliferation rate upon in vitro propagation. We also examined BM-MSCs for the expression of the 3 major embryonic stem cell transcription factors, Oct3/4, Sox-2, and Nanog, as well as 2 mRNA binding proteins, coding region determinant binding protein/insulin-like growth factor 2 mRNA binding protein 1 (Crd-bp/Imp1) and Deleted in azoospermia-like (Dazl), which are expressed in primitive stem cells, umbilical cord blood-hematopoietic stem cells and amniotic fluid stem cells, respectively. Further, it has been reported that these 2 genes are critical for embryonic development. In this study, therefore, we report, for the first time, the expression of Crd-bp/Imp1 and Dazl in BM-MSCs. Dazl, Oct3/4, and Sox2 were detected in relatively low levels in contrast to Crd-bp/Imp1, its major target c-Myc, as well as Nanog, which were expressed redundantly, irrespective of sex, donor age, or in vitro passaging. These findings could further support the extrinsic theory of aging of the MSC population and the potential implication of embryonic genes in adult stem cell physiology.


Subject(s)
Bone Marrow Cells/cytology , Cellular Senescence , Mesenchymal Stem Cells/physiology , Age Factors , Animals , Antigens, Differentiation/metabolism , Cell Differentiation , Cell Proliferation , Cell Size , Cells, Cultured , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
9.
Int J Cancer ; 121(9): 2031-2041, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17634957

ABSTRACT

The Ii-Key fragment from the MHC class II-associated invariant chain (or Ii protein) has been shown to facilitate direct charging of MHC class II epitopes to the peptide binding groove. The purpose of the present study was to test the potential of a series of Ii-Key/HER-2/neu776-790 hybrid peptides to generate increased frequencies of peptide-specific CD4+ T cells over the native peptide in mice transgenic (Tg) for a chimeric human mouse class II molecule (DR4-IE) (H-2b) as well as their antitumor potency. Following in vivo priming, such hybrid peptides induced increased proliferation and frequencies of IFN-gamma producing CD4+ T cells in response to either syngeneic dendritic cells pulsed with native peptide, or HLA-DR4+ human tumor cell lines expressing HER-2/neu. Hybrid peptides were more stable in an off-rate kinetics assay compared to the native peptide. In addition, antigen-specific CD4+ T cells from hybrid peptide immunized DR4-IE Tg mice synergized with HER-2/neu(435-443)-specific CD8+ T cells from HLA-A2.1 Tg HHD (H-2b) mice in producing antitumor immunity into SCID mice xenografted with the HER-2/neu+, HLA-A2.1+ and HLA-DR4+ FM3 human melanoma cell line. High proportions of these adoptively transferred HER-2/neu peptide-specific CD4+ and CD8+ T cells infiltrated FM3-induced tumors (tumor infiltrating lymphocytes; TIL) in SCID mice. CD8+ TIL exhibited long-lasting antitumor activity when cotransferred with CD4+ TIL, inducing regression of FM3 tumors in a group of untreated, tumor-bearing SCID mice, following adoptive transfer. Our data show that Ii-Key modified HER-2/neu776-790 hybrid peptides are sufficiently potent to provide antigen-specific CD4+ TH cells with therapeutic antitumor activity.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Neoplasms/immunology , Neoplasms/metabolism , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/chemistry , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Kinetics , Mice , Mice, SCID , Mice, Transgenic , Neoplasms/pathology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Xenograft Model Antitumor Assays
10.
Cancer Res ; 66(10): 5452-60, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707474

ABSTRACT

HER-2/neu is a self-antigen expressed by tumors and nonmalignant epithelial tissues. The possibility of self-tolerance to HER-2/neu-derived epitopes has raised questions concerning their utility in antitumor immunotherapy. Altered HER-2/neu peptide ligands capable of eliciting enhanced immunity to tumor-associated HER-2/neu epitopes may circumvent this problem. The human CTL peptide HER-2/neu (435-443) [hHER-2(9(435))] represents a xenogeneic altered peptide ligand of its mouse homologue, differing by one amino acid residue at position 4. In contrast to mHER-2(9(435)), vaccination of HLA-A*0201 transgenic (HHD) mice with hHER-2(9(435)) significantly increased the frequency of mHER-2(9(435))-specific CTL and also induced strong protective and therapeutic immunity against the transplantable ALC tumor cell line transfected to coexpress HLA-A*0201 and hHER-2/neu or rHER-2/neu. Similar results were also obtained with wild-type C57BL/6 mice inoculated with HER-2/neu transfectants of ALC. Adoptive transfer of CD8(+) CTL from mice immunized with hHER-2(9(435)) efficiently protected naive syngeneic mice inoculated with ALC tumors. In conclusion, our results show that HER-2(9(435)) serves as a tumor rejection molecule. They also propose a novel approach for generating enhanced immunity against a self-HER-2/neu CTL epitope by vaccinating with xenogeneic altered peptide ligands and provide useful insights for the design of improved peptide-based vaccines for the treatment of patients with HER-2/neu-overexpressing tumors.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Lymphoma/prevention & control , Ovarian Neoplasms/prevention & control , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA-A Antigens/genetics , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Immunotherapy, Active/methods , Lymphoma/genetics , Lymphoma/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Receptor, ErbB-2/genetics , Transfection
11.
Int Immunol ; 18(1): 49-58, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16291655

ABSTRACT

IL-21 plays a role in the proliferation and maturation of NK cells developed from hematopoietic stem cells. In this study, we found that IL-21, in the presence of physiological concentration of hydrocortisone (HC), has a significant impact on the functions of NK cells derived from umbilical cord blood (CB) populations. We demonstrate that IL-21, in combination with Flt3-ligand, IL-15 and HC, induces high proliferative responses and, apart from enhancing NK-mediated cytotoxicity, it also induces a significant increase in lymphokine-activated killer activity of CB/CD34+-derived CD56+ cells. In addition, IL-21 induced changes in the CD56+ cell cytokine secretion profile. Thus, we observed increased levels of IL-10 and granulocyte macrophage colony-stimulating factor, whereas tumor necrosis factor-alpha levels decreased. IFN-gamma production was also modified by IL-21, depending on the presence or absence of IL-18. CB/CD34+ cells did not express the IL-21R ex vivo, but receptor expression was induced during their commitment to differentiation into CD56+ cells. Our data ascribe to IL-21 an essential role on NK cell development and function under conditions similar to the in vivo CB microenvironment.


Subject(s)
Cell Differentiation/immunology , Fetal Blood/immunology , Hematopoietic Stem Cells/immunology , Interleukins/immunology , Killer Cells, Natural/immunology , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/pharmacology , Antigens, CD34/immunology , CD56 Antigen/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/immunology , Fetal Blood/cytology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Hematopoietic Stem Cells/cytology , Humans , Hydrocortisone/immunology , Hydrocortisone/pharmacology , Interleukin-21 Receptor alpha Subunit , Interleukins/pharmacology , Killer Cells, Natural/cytology , Membrane Proteins/immunology , Receptors, Interleukin/immunology , Receptors, Interleukin-21
12.
Cell Immunol ; 234(1): 67-75, 2005 Mar.
Article in English | MEDLINE | ID: mdl-16024007

ABSTRACT

Monokines IL12, IL15, and IL18 have been shown to activate NK cell function, however with high apoptosis induced by their combination within 48 h. Here, we demonstrate for the first time that CD56+ cells incubated for only 18 h with the combination of IL15/IL12 or IL15/IL18, then washed, and further cultured in plain medium, exhibit low levels of apoptosis. These shortly activated CD56+ cells show high killer activity against NK- and LAK-sensitive tumor targets that persists over a culture period of 18 days after two additional 6 h cycles of exposure to the monokines applied every 8 days and also retain their ability for high cytokine production during each exposure. Moreover, these repetitive short-term exposures of CD56+ cells to the monokine combinations result in long-lived CD56+ cells with slower rates of FcgammaRIII receptor (CD16) decline, therefore exhibiting higher antibody depended cytotoxicity, as opposed to the continuous incubation with the monokine combinations. In conclusion, short-term exposure of CD56+ cells to IL15/IL12 or IL15/IL18 at 8-day intervals may hold a promise for improved clinical results in cellular adoptive cancer immunotherapy and for the in vivo injections of the monokines.


Subject(s)
Apoptosis/drug effects , Interleukin-12/pharmacology , Interleukin-15/pharmacology , Interleukin-18/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , CD56 Antigen/metabolism , Cell Line, Tumor , Chemokines/metabolism , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Kinetics , Receptors, IgG/metabolism , Time Factors
13.
J Biol Chem ; 280(20): 20086-93, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15769738

ABSTRACT

The coding region determinant-binding protein/insulin-like growth factor II mRNA-binding protein (CRD-BP/IMP1) is an RNA-binding protein specifically recognizing c-myc, leader 3' IGF-II and tau mRNAs, and the H19 RNA. CRD-BP/IMP1 is predominantly expressed in embryonal tissues but is de novo activated and/or overexpressed in various human neoplasias. To address the question of whether CRD-BP/IMP1 expression characterizes certain cell types displaying distinct proliferation and/or differentiation properties (i.e. stem cells), we isolated cell subpopulations from human bone marrow, mobilized peripheral blood, and cord blood, all sources known to contain stem cells, and monitored for its expression. CRD-BP/IMP1 was detected only in cord blood-derived CD34(+) stem cells and not in any other cell type of either adult or cord blood origin. Adult BM CD34(+) cells cultured in the presence of 5'-azacytidine expressed de novo CRD-BP/IMP1, suggesting that epigenetic modifications may be responsible for its silencing in adult non-expressing cells. Furthermore, by applying the short interfering RNA methodology in MCF-7 cells, we observed, subsequent to knocking down CRD-BP/IMP1, decreased c-myc expression, increased IGF-II mRNA levels, and reduced cell proliferation rates. These data 1) suggest a normal role for CRD-BP/IMP1 in pluripotent stem cells with high renewal capacity, like the CB CD34(+) cells, 2) indicate that altered methylation may directly or indirectly affect its expression in adult cells, 3) imply that its de novo activation in cancer cells may affect the expression of c-Myc and insulin-like growth factor II, and 4) indicate that the inhibition of CRD-BP/IMP1 expression might affect cancer cell proliferation.


Subject(s)
Breast Neoplasms/genetics , Fetal Blood/cytology , Genes, myc , Insulin-Like Growth Factor II/genetics , RNA-Binding Proteins/metabolism , Adult , Antigens, CD34/blood , Azacitidine/pharmacology , Base Sequence , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , DNA, Neoplasm/biosynthesis , Female , Fetal Blood/immunology , Fetal Blood/metabolism , Gene Expression/drug effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , In Vitro Techniques , Pluripotent Stem Cells/immunology , Pluripotent Stem Cells/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Transfection
14.
Blood ; 106(1): 158-66, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15755904

ABSTRACT

Although glucocorticoids (GCs) have been described as acting mainly as anti-inflammatory and immunosuppressive drugs, they may also positively influence the immune system. In the present study, we demonstrate for the first time that hydrocortisone (HC), in synergy with interleukin-15 (IL-15), induces a dramatic increase in the expansion of peripheral blood-derived CD56+ cells, favoring the preferential outgrowth of classical natural killer (CD56+CD3- NK) over CD56+CD3+ natural killer T (NKT) cells. HC plus IL-15-driven CD56+ cells exhibited an increased potential for cytokine production with no impairment in their NK- and lymphokine-activated killer (LAK) activities. Elevated levels of GC-induced leucine zipper protein (GILZ) messenger RNA (mRNA) were detected in both NK and NKT cells cultured with HC and IL-15, in comparison to IL-15 alone. Phosphorylation status of signal transducer and activator of transcription 5 (STAT5) was not affected by the presence of HC in either of the populations. On the contrary, HC differentially affected the IL-2/IL-15R beta- and gamma-chain surface expression and the phosphorylation levels of extracellular signal-regulated kinases 1/2 (ERK1/2) in IL-15-activated NK and NKT cells. Our data ascribe a novel role to GCs on mature NK-cell expansion and function and open new perspectives for their use in cellular adoptive cancer immunotherapy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hydrocortisone/pharmacology , Interleukin-15/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Adoptive Transfer/methods , Burkitt Lymphoma , CD3 Complex/metabolism , CD56 Antigen/metabolism , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA-Binding Proteins/metabolism , Humans , Interleukin-2/metabolism , K562 Cells , Killer Cells, Natural/metabolism , Milk Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA, Messenger/analysis , Receptors, Interleukin-15 , Receptors, Interleukin-2/genetics , STAT5 Transcription Factor , Trans-Activators/metabolism , Transcription Factors/genetics
15.
Blood ; 101(9): 3444-50, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12506032

ABSTRACT

Natural killer (NK) cell differentiation from pluripotent CD34(+) human hematopoietic stem cells or oligopotent lymphoid progenitors has already been reported. In the present study, long-term cultures of the CD56(-)/CD34(-) myeloid-like adherent cell fraction (ACF) from umbilical cord blood (UCB), characterized by the expression of CD14(+) as well as other myeloid markers, were set up with flt3 ligand (FL) and interleukin-15 (IL-15). The UCB/ACF gradually expressed the CD56 marker, which reached fairly high levels (approximately 90% of the cells were CD56(+)) by day 15. FL plus IL-15-driven ACF/CD56(+) cells progressively expressed a mature NK functional program lysing both NK- and lymphokine-activate killer (LAK)-sensitive tumor targets and producing high levels of interferon-gamma (IFN-gamma), granulocyte-macrophage colony-stimulating factor, tumor necrosis factor alpha, and IL-10 upon stimulation with IL-12 and IL-18. Similar results were obtained when highly purified CD14(+) cells from UCB were cultured with FL and IL-15. In contrast, UCB/CD34(+) cells cultured under the same conditions showed a delayed expression of CD56 and behaved functionally differently in that they exhibited NK but not LAK cytotoxicity and produced significantly fewer cytokines. Kinetic studies on the phenotype of UCB/ACF or UCB/CD14(+) cells cultured in the presence of FL and IL-15 showed a rapid decrease in CD14 expression after day 5, which reached levels of zero by day 20. Approximately 60% of the CD56(+) derived from the UCB/ACF or the UCB/CD14(+) cells coexpressed CD14 by day 5. Taken together, our data support the role of CD14(+) myeloid-like cells within UCB as a novel progenitor for lymphoid NK cells.


Subject(s)
Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/cytology , Myeloid Cells/classification , Animals , Burkitt Lymphoma/pathology , CD56 Antigen/biosynthesis , Cell Differentiation/drug effects , Cells, Cultured/cytology , Cells, Cultured/metabolism , Cytotoxicity, Immunologic , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Infant, Newborn , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-15/pharmacology , K562 Cells , Lipopolysaccharide Receptors/biosynthesis , Membrane Proteins/pharmacology , Mice , Multiple Myeloma/pathology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...