Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 882: 173255, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32553737

ABSTRACT

The anticancer prodrug butyroyloxymethyl diethylphosphate (AN-7), upon metabolic hydrolysis, releases the histone deacetylase inhibitor butyric acid and imparts histone hyperacetylation. We have shown previously that AN-7 increases doxorubicin-induced cancer cell death and reduces doxorubicin toxicity and hypoxic damage to the heart and cardiomyocytes. The cardiofibroblasts remain unprotected against both insults. Herein we examined the selective effect of AN-7 on hypoxic cardiomyocytes and cardiofibroblasts and investigated mechanisms underlying the cell specific response. Hypoxic cardiomyocytes and cardiofibroblasts or H2O2-treated H9c2 cardiomyoblasts, were treated with AN-7 and cell damage and death were evaluated as well as cell signaling pathways and the expression levels of heme oxygenase-1 (HO-1). AN-7 diminished hypoxia-induced mitochondrial damage and cell death in hypoxic cardiomyocytes and reduced hydrogen peroxide damage in H9c2 cells while increasing cell injury and death in hypoxic cardiofibroblasts. In the cell line, AN-7 induced Akt and ERK survival pathway activation in a kinase-specific manner including phosphorylation of the respective downstream targets, GSK-3ß and BAD. Hypoxic cardiomyocytes responded to AN-7 treatment by enhanced phosphorylation of Akt, ERK, GSK-3ß and BAD and a significant 6-fold elevation in HO-1 levels. In hypoxic cardiofibroblasts, AN-7 did not activate Akt and ERK beyond the effect of hypoxia alone and induced a limited (~1.5-fold) increase in HO-1. The cell specific differences in kinase activation and in heme oxygenase-1 upregulation may explain, at least in part, the disparate outcome of AN-7 treatment in hypoxic cardiomyocytes and hypoxic cardiofibroblasts.


Subject(s)
Antineoplastic Agents/pharmacology , Butyrates/pharmacology , Cardiotonic Agents/pharmacology , Fibroblasts/drug effects , Myocytes, Cardiac/drug effects , Organophosphorus Compounds/pharmacology , Prodrugs/pharmacology , Animals , Butyric Acid , Cell Hypoxia/drug effects , Cell Line , Cell Survival/drug effects , Fibroblasts/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Histone Deacetylase Inhibitors , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats
2.
Reprod Fertil Dev ; 29(10): 1982-1994, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28143632

ABSTRACT

The possibility of maturing human primordial follicles in vitro would assist fertility restoration without the danger of reseeding malignancies. Leukaemia inhibitory factor (LIF) and certain culture matrices may promote human follicular growth. The present study compared human primordial follicular growth on novel culture matrices, namely human recombinant vitronectin (hrVit), small intestine submucosa (SIS), alginate scaffolds and human recombinant virgin collagen bioengineered in tobacco plant lines (CollPlant). The frozen-thawed ovarian samples that were used had been obtained from girls or young women undergoing fertility preservation. In the first part of the study, 20 samples were cultured for 6 days on hrVit or SIS with basic culture medium alone or supplemented with one of two concentrations of LIF (10ngmL-1 and 100ngmL-1), with and without LIF-neutralising antibody. In the second part of the study, 15 samples were cultured for 6 days on alginate scaffolds or CollPlant matrices with basic culture medium. Follicular development was assessed by follicular counts and classification, Ki67 immunohistochemistry and 17ß-oestradiol and anti-Müllerian hormone measurements in spent media samples. Primordial follicular growth was not enhanced by LIF. Despite some significant differences among the four matrices, none appeared to have a clear advantage, apart from significantly more Ki67-stained follicles on alginate and CollPlant matrices. Further studies of other culture matrices and medium supplements are needed to obtain an optimal system.


Subject(s)
Cell Culture Techniques/methods , Extracellular Matrix , Leukemia Inhibitory Factor , Ovarian Follicle/growth & development , Anti-Mullerian Hormone/metabolism , Culture Media , Estradiol/metabolism , Female , Fertility Preservation/methods , Humans
3.
Reprod Biomed Online ; 34(1): 104-114, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27815062

ABSTRACT

How chemotherapy affects dormant ovarian primordial follicles is unclear. The 'burnout' theory, studied only in mice, suggests cyclophosphamide enhances primordial follicle activation. Using 4-hydroperoxycyclophosphamide (4hc) and phosphoramide mustard (PM), this study assessed how the active cyclophosphamide metabolites 4-hydroxycyclophosphamide (4-OHC) and PM, affect human primordial follicles. Frozen-thawed human ovarian samples were sliced and cultured with basic culture medium (cultured controls) or with 4hc/PM (3 µmol/l/10 µmol/l) (treated samples) for 24-48 h. Follicular counts and classification, Ki67 and anti-Müllerian hormone (AMH) immunohistochemistry and an apoptosis assay were used for evaluation, and 17ß-oestradiol and AMH were measured in spent media samples. Generally, there was primordial follicle decrease and elevated developing follicle rates in treated samples compared with cultured (P = 0.04 to P < 0.0005) and uncultured controls (P < 0.05 to P < 0.0001). No traces of apoptosis were found. There were almost twicethe levels of AMH and 17ß-oestradiol in treated compared with untreated samples (AMH with 4hc 3 µmol/l; P = 0.04). All follicles stained positively for AMHincluded treated samples. Ki67 positive staining was noted in all samples. Cyclophosphamide metabolites seem to enhance human primordial follicle activation to developing follicles, in vitro. Study findings support the 'burnout' theory as the mechanism of chemotherapy-induced ovarian toxicity.


Subject(s)
Cyclophosphamide/therapeutic use , Ovarian Follicle/drug effects , Adolescent , Anti-Mullerian Hormone/therapeutic use , Child , Cryopreservation , Culture Media , Cyclophosphamide/analogs & derivatives , Embryo Culture Techniques , Estradiol/metabolism , Female , Freezing , Humans , Immunosuppressive Agents/therapeutic use , Ki-67 Antigen/metabolism , Ovary/metabolism , Phosphoramide Mustards/therapeutic use , Time Factors
4.
Dermatoendocrinol ; 8(1): e1137399, 2016.
Article in English | MEDLINE | ID: mdl-27195054

ABSTRACT

Several inflammatory mediators increase calcitriol production by epidermal keratinocytes. In turn calcitriol attenuates the keratinocyte inflammatory response. Since the effect of the in-situ generated calcitriol depends also on the sensitivity to the hormone we studied the effect of inflammatory cytokines on the response of HaCaT human keratinocytes to calcitriol by examining the expression and transcriptional activity of VDR. Treatment with TNF, but not with IL-1ß or interferon γ, increased VDR protein level, while decreasing the level of its heterodimerization partner RXRα. This was associated with increased VDR mRNA levels. c-Jun N-terminal kinase, but not P38 MAPK or NFκB, was found to participate in the upregulation of VDR by TNF. The functional significance of the modulation of VDR and RXRα levels by TNF is manifested by increased induction of VDR target gene CYP24A1 by calcitriol. Calcitriol, in turn, inhibited the enhanced expression of VDR by TNF. In conclusion, the inflammatory cytokine TNF increases the response of keratinocytes to calcitriol through upregulation of its receptor VDR, which in turn is subject to negative feedback by the hormone accelerating the return of the keratinocyte vitamin D system to its basal activity. We surmise that the increased generation and sensitivity to calcitriol in keratinocytes play a role in the resolution of epidermal inflammation.

5.
Neoplasia ; 16(9): 741-50, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25246274

ABSTRACT

Immune tolerance mechanisms supporting normal human pregnancy are exploited by breast cancer and other malignancies. We cloned from human placenta and breast cancer cells the novel human immunomodulator named placenta immunosuppressive ferritin (PLIF). PLIF is composed of a ferritin heavy chain-like domain and a novel cytokine-like domain, named C48. Both intact PLIF and C48 inhibit T cell proliferation. Blocking PLIF by specific antibodies in a tolerant breast cancer model in nude mice resulted in tumor cell apoptosis and rejection. This prompted us to study active immune preventive strategies targeting PLIF activity. Currently, we report on the design and synthesis of the novel C24D polypeptide, which inhibits the binding of PLIF to T cells and therefore inhibits the immune suppressive effect of PLIF. The effect of C24D on the generation of anti-breast cancer cytotoxic T lymphocytes (CTLs) was studied in vitro in cultures of MCF-7 (HLA-A2(+)) or T47D (HLA-A2(-)) breast cancer cells incubated with peripheral blood mononuclear cells (PBMCs) from healthy blood donors. We found that C24D treatment exclusively induced development of CTLs. On reactivation by their specific target cells, the CTLs secreted interferon-γ and induced target apoptosis. Anti-MCF-7 CTLs were cross-cytotoxic to MDA-MB-231 (HLA-A2(+)) triple-negative breast cancer but not to T47D. Moreover, C24D treatment in vivo inhibited the growth of MCF-7 tumors engrafted in immune-compromised nude mice transfused with naïve allogeneic human PBMCs. Our results demonstrate that C24D treatment breakdown breast cancer induced tolerance enabling the initiation of effective anti-tumor immune response.


Subject(s)
Breast Neoplasms/immunology , Ferritins/metabolism , Immunomodulation/drug effects , Peptides/pharmacology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Humans , Interferon-gamma/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation , Nerve Tissue Proteins/metabolism , Oxidoreductases , Peptides/administration & dosage , Peptides/chemical synthesis , Protein Binding/drug effects , T-Lymphocytes, Cytotoxic/immunology , Xenograft Model Antitumor Assays
6.
PLoS One ; 8(9): e71100, 2013.
Article in English | MEDLINE | ID: mdl-24066022

ABSTRACT

Popeye domain containing1 (Popdc1), also named Bves, is an evolutionary conserved membrane protein. Despite its high expression level in the heart little is known about its membrane localization and cardiac functions. The study examined the hypothesis that Popdc1 might be associated with the caveolae and play a role in myocardial ischemia tolerance. To address these issues, we analyzed hearts and cardiomyocytes of wild type and Popdc1-null mice. Immunoconfocal microscopy revealed co-localization of Popdc1 with caveolin3 in the sarcolemma, intercalated discs and T-tubules and with costameric vinculin. Popdc1 was co-immunoprecipitated with caveolin3 from cardiomyocytes and from transfected COS7 cells and was co-sedimented with caveolin3 in equilibrium density gradients. Caveolae disruption by methyl-ß-cyclodextrin or by ischemia/reperfusion (I/R) abolished the cellular co-localization of Popdc1 with caveolin3 and modified their density co-sedimentation. The caveolin3-rich fractions of Popdc1-null hearts redistributed to fractions of lower buoyant density. Electron microscopy showed a statistically significant 70% reduction in caveolae number and a 12% increase in the average diameter of the remaining caveolae in the mutant hearts. In accordance with these changes, Popdc1-null cardiomyocytes displayed impaired [Ca(+2)]i transients, increased vulnerability to oxidative stress and no pharmacologic preconditioning. In addition, induction of I/R injury to Langendorff-perfused hearts indicated a significantly lower functional recovery in the mutant compared with wild type hearts while their infarct size was larger. No improvement in functional recovery was observed in Popdc1-null hearts following ischemic preconditioning. The results indicate that Popdc1 is a caveolae-associated protein important for the preservation of caveolae structural and functional integrity and for heart protection.


Subject(s)
Caveolae/metabolism , Membrane Proteins/metabolism , Myocardial Ischemia/metabolism , Animals , Blotting, Western , COS Cells , Calcium/metabolism , Caveolin 3/metabolism , Cells, Cultured , Chlorocebus aethiops , Immunoprecipitation , In Vitro Techniques , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Myocardial Ischemia/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Polymerase Chain Reaction , Protein Binding , Rats
7.
Exp Hematol ; 37(9): 1121-30, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19539693

ABSTRACT

OBJECTIVE: Cloned placenta immunoregulatory ferritin (PLIF) contains a novel, nonferritin bioactive domain (C-48) with immunodulatory activity. We documented that treatment of whole human bone marrow cells with PLIF and its subcloned C48 proteins resulted in myeloid progenitor cell growth and differentiation and T-cell suppression via an effect on the cytokine network. We tested whether this differential effect supports allogeneic bone marrow transplantation with long-lasting tolerance without any further treatments. MATERIALS AND METHODS: Splenocyte-enriched C3H (H2(k)) whole bone marrow was transplanted into C57Bl (H2(b)) recipients after total body irradiation. Recipients were injected with recombinant C48 (3 mg/kg, intraperitoneal) for 21 days or with glutathione S-transferase. Animals were monitored for survival, chimerism, and clinical signs of graft-vs-host disease (GVHD). Next, chimera whole bone marrow was transplanted to secondary myeloablated C57Bl (H2(b)) hosts without treatment. RESULTS: Mice that received C48 treatment following allogeneic splenocyte-enriched bone marrow transplantation demonstrated full-donor chimerism without GVHD mortality, and normal blood cell counts in 75% of recipients. Secondary transplants from the full chimera to myeloblated C57Bl hosts showed 100% engraftment, no GVHD mortality, and no impairment in the long-term hematopoietic reconstitution potential. Allogeneic response of spleen cells from secondary chimeras against donor C3H (H2(k)) and recipient C57Bl (H2(b)) were similar to syngeneic response, whereas reactivity to third party (DBA H2(d)) was significantly enhanced. CONCLUSIONS: Findings of this study provide the proof of concept that C48-a novel, single, bifunctional therapeutic modality enabled successful allogeneic, unmanipulated bone marrow transplantation without GVHD, and with lasting specific tolerance.


Subject(s)
Bone Marrow Transplantation , Ferritins/pharmacology , Graft vs Host Disease/prevention & control , Immunologic Factors/pharmacology , Myeloid Progenitor Cells/metabolism , Animals , Female , Graft vs Host Disease/metabolism , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred DBA , Oxidoreductases , Protein Structure, Tertiary , Recombinant Proteins/pharmacology , Transplantation Chimera , Transplantation, Homologous
8.
Semin Cancer Biol ; 18(4): 260-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18467123

ABSTRACT

Tumor progression is substantially dependent on network of multiple factors, including adhesion and homing molecules, which support the malignant metastatic spread. CD44, one of the adhesion/homing molecules, has attracted much attention not only because it is expressed on many types of tumors, but also owing to its numerous functions, such as supporting cell migration and transmitting survival signals, thereby being pro-oncogenic by nature. We have used the mouse malignant LB lymphoma cell line as a model for comprehensive in vitro and in vivo analyses of the interaction between CD44 and hyaluronic acid (HA), and its relevance to tumor dissemination. The in vitro studies revealed that LB cells could not bind HA, either under static or dynamic (i.e., shear flow) conditions, unless their CD44 is activated by phorbol ester, deglycosylated (to increase the CD44 positive net charge) or transfected with CD44 variants. In parallel, in vivo experiments showed that LB cell dissemination could be controlled by injection of anti-CD44 monoclonal antibodies or hyaluronidase. Furthermore, LB cells transfected with CD44v4-v10 variant, rather than standard CD44, displayed enhanced invasion of the peripheral lymph nodes. This effect was completely lost if the HA binding site of CD44 were mutated. LB cell accumulation in the lymph nodes is caused by enhanced migration via the afferent lymphatics rather than by accelerated proliferation within the lymph node. This information can be exploited to tailor a "therapeutic suit" that should be maximally effective in inducing tumor resistance, while minimizing destructive side effects.


Subject(s)
Hyaluronan Receptors/physiology , Lymphoma/metabolism , Animals , Humans , Hyaluronic Acid/metabolism
9.
Neoplasia ; 9(6): 487-94, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17603631

ABSTRACT

The recently cloned human gene named "placental immunoregulatory ferritin" (PLIF) is a pregnancy-related immunomodulator. Recombinant PLIF and its bioactive domain C48 are immune-suppressive and induce pronounced IL-10 production by immune cells. PLIF is expressed in the placenta and breast cancer cells. Blocking PLIF in pregnant mice by anti-C48 antibodies inhibited placental and fetal growth and modulated the cytokine network. It has been revealed that anti-C48 treatment inhibited MCF-7 tumor growth in nude mice. However, this significant effect was observed only in those transfused with human peripheral blood mononuclear cells. Blocking PLIF in tumor-engrafted human immune cell transfused mice resulted in massive infiltration of human CD45+ cells (mainly CD8+ T cells), both intratumorally and in the tumor periphery, and a significant number of caspase-3+ cells. In vitro, anti-C48 treatment of MCF-7 tumor cells cocultured with human lymphocytes induced a significant increase in interferon-gamma secretion. We conclude that blocking PLIF inhibits breast cancer growth, possibly by an effect on the cytokine network in immune cells and on breakdown of immunosuppression.


Subject(s)
Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Ferritins/immunology , Immunoglobulin G/therapeutic use , Pregnancy Proteins/immunology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cells, Cultured , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunity, Cellular , Immunophenotyping , Immunotherapy , Interferon-gamma/metabolism , Interleukin-10/metabolism , Leukocyte Common Antigens/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Nude , Rabbits
10.
Exp Hematol ; 34(2): 159-66, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16459184

ABSTRACT

OBJECTIVE: Placenta immunomodulator ferritin (PLIF) is a cloned human chimeric ferritin H chain with a novel non-ferritin C-terminal 48 amino acid sequence (C48). Recombinant PLIF-C48 exhibited cell-mediated immunosuppression. The aim of the current study was to investigate the regulatory effects of native placental ferritin (PLF), recombinant PLIF, and C48 on hematopoiesis of human bone marrow (BM). METHODS: BM mononuclear cells (BM-MNCs) and CD34(+) selected cells were treated in vitro with either PLF, PLIF, or C48 without and in combination with granulocyte (G)-monocyte (M) colony-stimulating factor (GM-CSF) and subjected to hematopoietic progenitor cell assay. Cytokines and chemokines secreted by the treated cells were evaluated in culture supernatant using antibody array assays to determine mechanism of action. RESULTS: In vitro treatment of BM-MNCs with PLF, PLIF, or C48 induced significant growth of myeloid colonies and when mixed with GM-CSF or Granulocyte-Colony Stimulating Factor (G-CSF) exhibited additive enhanced colony forming units-granulocyte monocyte growth. Yet, C48 treatment of selected CD34(+) cells did not yield colony formation and did not affect their response to GM-CSF. Treatment of BM-MNCs with C48 for 48 hours induced secretion of marked levels of GM-CSF, interleukin (IL)-6, IL-1, and IL-10. These cytokines were secreted primarily by C48-treated BM adherent cells and partly by nonadherent cells, whereas the CD34(+) selected cells secreted IL-6 only. CONCLUSION: C48-PLIF enhancement of myelopoiesis resulted from cross talk between BM accessory cells and progenitor cells. The differential PLIF-C48 effects (i.e., myeloid progenitor cell growth and T-cell suppression) are due to their effect on the cytokine-chemokine networks.


Subject(s)
Bone Marrow Cells/drug effects , Chemokines/metabolism , Cytokines/metabolism , Ferritins/pharmacology , Myelopoiesis/drug effects , T-Lymphocytes/drug effects , Adjuvants, Immunologic/pharmacology , Amino Acid Sequence , Antigens, CD34/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cloning, Molecular , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , In Vitro Techniques , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Recombinant Proteins/pharmacology , T-Lymphocytes/metabolism
11.
Int Immunol ; 16(2): 275-81, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14734613

ABSTRACT

Allicin, the main organic allyl sulfur component in garlic, exhibits immune-stimulatory and antitumor properties. Allicin stimulated [(3)H]thymidine incorporation in mouse splenocytes and enhanced cell-mediated cytotoxicity in human peripheral mononuclear cells. Multiple administration (i.p.) of allicin elicited a marked antitumor effect in mice inoculated with B-16 melanoma and MCA-105 fibrosarcoma. The immune-stimulatory and antitumor effects of allicin are characterized by a bell-shaped curve, i.e. allicin at high, supra-optimal concentrations is less effective or inhibitory. Allicin induced activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in human peripheral mononuclear cells, and also in wild-type Jurkat T-cells. Allicin failed to activate ERK1/2 in Jurkat T cells that express p21(ras), in which Cys118 was replaced by Ser. These cells are not susceptible to redox-stress modification and activation. We postulate that the immune stimulatory effect of allicin is mediated by redox-sensitive signaling such as activation of p21(ras). It is suggested that the antitumor effect of allicin is related to its immune-stimulatory properties.


Subject(s)
Anti-Infective Agents/pharmacology , Lymphocyte Activation/drug effects , Neoplasms, Experimental/drug therapy , Oncogene Protein p21(ras)/metabolism , Sulfinic Acids/pharmacology , T-Lymphocytes/immunology , Amino Acid Substitution , Animals , Cytotoxicity, Immunologic , Disulfides , Enzyme Activation/drug effects , Humans , Jurkat Cells , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Oncogene Protein p21(ras)/genetics , Oxidation-Reduction/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , T-Lymphocytes/drug effects
12.
FASEB J ; 17(8): 955-7, 2003 May.
Article in English | MEDLINE | ID: mdl-12670872

ABSTRACT

Recently, we reported the cloning and preliminary characterization of a novel human immunomodulator named PLIF (placenta immunomodulatory ferritin). PLIF has a unique molecular structure, which is composed of a ferritin heavy chain-like domain and a novel cytokine-like domain called C48. Both intact molecule and C48 inhibit T cell proliferation following allogeneic or anti-CD3 stimuli. PLIF is localized at the fetal-maternal interface of human placenta and might play a role in down-modulating the maternal immune reaction toward the embryo. The inhibitory effect of PLIF on T cell activation can be direct, indirect through cytokine mediators, or both. In the present study we investigated the possible indirect effects of PLIF by using its bioactive domain C48. Measurement of various cytokines revealed that C48, predominantly, induce pronounced and rapid IL-10 production in monocytes, which is immune activation-independent. Further, we discovered that C48-induced IL-10 production is mediated through a calcium/calmodulin-p38 mitogen-activated protein (MAP) kinase signaling pathway. However, extracellular signal-related kinases1,2 (ERK1,2), also activated by C48 stimulation, exhibited a limiting effect on IL-10 production.


Subject(s)
Ferritins/pharmacology , Interleukin-10/biosynthesis , Monocytes/drug effects , Pregnancy Proteins/pharmacology , Binding Sites/genetics , Calcium/metabolism , Calmodulin/metabolism , Coculture Techniques , Egtazic Acid/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Ferritins/chemistry , Ferritins/genetics , Humans , Indoles/pharmacology , Isoquinolines/pharmacology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Maleimides/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Monocytes/cytology , Monocytes/metabolism , Oxidoreductases , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Pregnancy Proteins/chemistry , Pregnancy Proteins/genetics , Recombinant Proteins/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , p38 Mitogen-Activated Protein Kinases
13.
Arthritis Rheum ; 48(3): 846-53, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12632441

ABSTRACT

OBJECTIVE: To determine the effect of treatment with C48, the recombinant cytokine-like domain of the novel human placental immunomodulator ferritin (PLIF) immunoregulator, on zymosan-induced arthritis (ZIA) in mice and on adjuvant-induced arthritis (AIA) in rats. METHODS: The in vitro effect of PLIF/C48 was tested in mixed lymphocyte cultures (MLCs) of allogeneic mouse splenocytes. Arthritis was induced by intraarticular injection of zymosan into naive mice and by subcutaneous injection of Mycobacterium tuberculosis into rats. C48 was injected intraperitoneally daily from day 3 to day 9 or from day 7 to day 13 after induction of synovitis by zymosan, and every other day from day 2 to day 14 after induction of AIA. Swelling of the joints and histologic features of the synovium were assessed. Th1 and Th2 cytokines were quantified by enzyme-linked immunosorbent assay. RESULTS: Both PLIF and C48 significantly inhibited the in vitro immunoreactivity of mouse splenocytes in MLCs. Treatment of ZIA mice and AIA rats with C48 effectively reduced joint swelling. C48 treatment reduced synovial lining thickening, numbers of mononuclear cells and histiocytes, as well as cartilage destruction and bone erosions. In vitro, activated splenocytes from C48-treated ZIA and AIA animals produced significantly higher levels of interleukin-10 (IL-10). In animals with ZIA, this was accompanied by lower levels of tumor necrosis factor and IL-2. CONCLUSION: Human PLIF and C48 were shown to exert cross-species immunosuppressive activity in vitro. The in vivo suppression of articular inflammation in the experimental models of ZIA and AIA was the result of treatment with the antiinflammatory human C48. These results suggest that treatment with C48 may offer an effective immunotherapeutic means of controlling inflammatory polyarthritis.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Arthritis, Experimental/prevention & control , Carrier Proteins/therapeutic use , Ferritins , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins/therapeutic use , Pregnancy Proteins , Adjuvants, Immunologic/administration & dosage , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Carrier Proteins/administration & dosage , Cell Cycle Proteins , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Ferritins/administration & dosage , Ferritins/immunology , Ferritins/pharmacology , Hindlimb/drug effects , Hindlimb/pathology , Injections, Intraperitoneal , Joints/drug effects , Joints/pathology , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Nerve Tissue Proteins/administration & dosage , Oxidoreductases , Pregnancy Proteins/administration & dosage , Pregnancy Proteins/immunology , Pregnancy Proteins/pharmacology , Rats , Rats, Inbred Lew , Spleen/cytology , Spleen/metabolism , Zymosan
14.
J Biol Chem ; 277(15): 12901-5, 2002 Apr 12.
Article in English | MEDLINE | ID: mdl-11821435

ABSTRACT

Ferritin is a ubiquitous iron storage protein existing in multiple isoforms composed of 24 heavy and light chain subunits. We describe here a third ferritin-related subunit cloned from human placenta cDNA library and named PLIF (placental immunomodulatory ferritin). The PLIF coding region is composed of ferritin heavy chain (FTH) sequence lacking the 65 C-terminal amino acids, which are substituted with a novel 48 amino acid domain (C48). In contrast to FTH, PLIF mRNA does not include the iron response element in the 5'-untranslated region, suggesting that PLIF synthesis is not regulated by iron. The linkage between the FTH and C48 domains created a restriction site for EcoRI. PLIF protein was found to localize in syncytiotrophoblasts of placentas (8 weeks of gestation) at the fetal-maternal interface. Increased levels of PLIF transcript and protein were also detected in the breast carcinoma cell lines T47D and MCF-7 but not in the benign corresponding cell line HBL-100. In vitro, PLIF was shown to down-modulate mixed lymphocyte reactions and to inhibit the proliferation of peripheral blood mononuclear cells stimulated with OKT3. The accumulated data indicate that PLIF is an embryonic immune factor involved in down-modulating the maternal immune recognition of the embryo toward anergy. This mechanism may have been adapted by breast cancer cells over expressing PLIF.


Subject(s)
Ferritins/isolation & purification , Immune Tolerance , Placenta/metabolism , Pregnancy Proteins/isolation & purification , Amino Acid Sequence , Base Sequence , Breast/cytology , Breast/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary , Female , Ferritins/chemistry , Ferritins/genetics , Ferritins/physiology , Humans , Molecular Sequence Data , Oxidoreductases , Pregnancy Proteins/chemistry , Pregnancy Proteins/genetics , Pregnancy Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...