Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Type of study
Publication year range
2.
PLoS One ; 4(8): e6709, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19696931

ABSTRACT

In the human fetal kidney (HFK) self-renewing stem cells residing in the metanephric mesenchyme (MM)/blastema are induced to form all cell types of the nephron till 34(th) week of gestation. Definition of useful markers is crucial for the identification of HFK stem cells. Because wilms' tumor, a pediatric renal cancer, initiates from retention of renal stem cells, we hypothesized that surface antigens previously up-regulated in microarrays of both HFK and blastema-enriched stem-like wilms' tumor xenografts (NCAM, ACVRIIB, DLK1/PREF, GPR39, FZD7, FZD2, NTRK2) are likely to be relevant markers. Comprehensive profiling of these putative and of additional stem cell markers (CD34, CD133, c-Kit, CD90, CD105, CD24) in mid-gestation HFK was performed using immunostaining and FACS in conjunction with EpCAM, an epithelial surface marker that is absent from the MM and increases along nephron differentiation and hence can be separated into negative, dim or bright fractions. No marker was specifically localized to the MM. Nevertheless, FZD7 and NTRK2 were preferentially localized to the MM and emerging tubules (<10% of HFK cells) and were mostly present within the EpCAM(neg) and EpCAM(dim) fractions, indicating putative stem/progenitor markers. In contrast, single markers such as CD24 and CD133 as well as double-positive CD24(+)CD133(+) cells comprise >50% of HFK cells and predominantly co-express EpCAM(bright), indicating they are mostly markers of differentiation. Furthermore, localization of NCAM exclusively in the MM and in its nephron progenitor derivatives but also in stroma and the expression pattern of significantly elevated renal stem/progenitor genes Six2, Wt1, Cited1, and Sall1 in NCAM(+)EpCAM(-) and to a lesser extent in NCAM(+)EpCAM(+) fractions confirmed regional identity of cells and assisted us in pinpointing the presence of subpopulations that are putative MM-derived progenitor cells (NCAM(+)EpCAM(+)FZD7(+)), MM stem cells (NCAM(+)EpCAM(-)FZD7(+)) or both (NCAM(+)FZD7(+)). These results and concepts provide a framework for developing cell selection strategies for human renal cell-based therapies.


Subject(s)
Biomarkers/metabolism , Kidney/embryology , Stem Cells/cytology , Flow Cytometry , Humans , Kidney/metabolism , Reverse Transcriptase Polymerase Chain Reaction
3.
In Vivo ; 20(4): 543-8, 2006.
Article in English | MEDLINE | ID: mdl-16900787

ABSTRACT

Two-year-old mice of the long-living transgenic mice of the alphaMUPA strain were previously found to show higher tumor resistance than the their initial wild-type (WT) strain (Tirosh, 2003). To better understand the mechanism underlying the differences in tumorigenesis rates between the two mouse lines, the rate of tumorigenesis and survival effects were studied in alphaMUPA mice and parental WT mice exposed to dimethylbenz(a)anthracene (DMBA). Each animal received three intragastric feedings of DMBA, each one week apart, at doses of 2, 1, and 1 mg dissolved in 0.2 ml corn oil; thus, the total amount of the carcinogen was 4 mg/mouse. Control mice received corn oil. The alphaMUPA mice exhibited distinctly higher survival rates in experimental chemically-induced tumorigenesis compared to their WT counterparts: 93% vs. 67%, p =2.7. The rate of tumorigenesis differed between the mouse lines (yield was 1.5 and 2.1), owing to a distinct tendency toward decreased tumor frequency in the skin and forestomach in the alphaMUPA mice. The experimental duration was also significantly higher for transgenic mice: 35.9 +/- 1.2 weeks compared to 30.5 +/- 1.3 weeks in WT mice, p <0.01. The lungs, forestomach and skin were target organs for the carcinogenic effect of DMBA. Our observations suggest that aging promotes the rate of spontaneous and induced tumorigenesis.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/toxicity , Carcinogens/toxicity , Longevity , Neoplasms, Experimental/mortality , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , Administration, Oral , Animals , Carcinogens/administration & dosage , Lung Neoplasms/chemically induced , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Skin Neoplasms/chemically induced , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Stomach Neoplasms/chemically induced , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate , Time Factors
4.
Oncol Rep ; 15(2): 479-83, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16391872

ABSTRACT

We analyzed the morphological changes accompanying the development of cancer in the mouse forestomach. The main aim of the study was to evaluate whether cancer in this area of the stomach arises de novo or undergoes a series of precancerous changes. Tumors were induced by the 1,2-dimethylbenz(a)antracene (DMBA) at a total dose of 4 mg/mouse. The suspected areas of the stomach were studied morphologically in 79 mice. Benign tumors (squamous-cell papillomas) and malignant tumors (squamous-cell carcinomas) were found in 40 mice. Tumors arose in all cases together with differential changes in the forestomach epithelium. These changes were seen as irregular diffuse hyperplasia or focal proliferation, with or without differential signs of dysplasia. Destruction of the basal epithelial membrane indicated transformation of the process into malignant invasive carcinoma. Thus, in chemically induced cancer of the forestomach, squamous-cell carcinoma develops as the final stage of morphologically recognizable precancerous changes in the epithelial layer. De novo formation of such tumors in the forestomach was not observed.


Subject(s)
Carcinoma, Squamous Cell/pathology , Precancerous Conditions/pathology , Stomach Neoplasms/pathology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Carcinogens/toxicity , Carcinoma, Squamous Cell/chemically induced , Female , Hyperplasia/pathology , Mice , Stomach Neoplasms/chemically induced
5.
Oncol Rep ; 14(6): 1625-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16273267

ABSTRACT

In our previous studies, we showed that soluble tumor-associated antigens (sTAA) of 66 kDa and 51 kDa have distinct tumor-preventive effects on chemically induced mammary cancer in rats and are able to repair the damage caused by tumorigenesis in its early stages. In the present study, we investigated whether these proteins can prevent the development of chemically induced tumors in mice. The study was performed on C3H/He mice which have the ability to develop many spontaneous tumors with age. Forty-four, 6-week-old mice were exposed twice at a 2-week interval to the carcinogen 9,10-dimethyl-1,2-benz(alpha)anthracene (DMBA), at a dose of 2 mg/mouse administered intragastrically. Two months later, the mice were divided into two groups. One group received sterile saline twice a week at a dose of 0.2 ml/mouse, intraperitoneally (i.p.). The other group received sTAA twice a week at a dose of about 10 microl in 0.2 ml of sterile saline/mouse, i.p. Periodically, all mice were checked for the presence of tumors. The experiment was terminated at week 35. Vaccination with sTAA increased the time of involvement of mice in the experiment, prevented the tumorigenic effect of DMBA, and inhibited further development of existing tumors.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Neoplasms, Experimental/immunology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred C3H , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/prevention & control , Ovarian Neoplasms/chemically induced , Ovarian Neoplasms/immunology , Ovarian Neoplasms/prevention & control , Stomach Neoplasms/chemically induced , Stomach Neoplasms/immunology , Stomach Neoplasms/prevention & control , Time Factors , Uterine Neoplasms/chemically induced , Uterine Neoplasms/immunology , Uterine Neoplasms/prevention & control
6.
Oncol Rep ; 14(5): 1317-21, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16211303

ABSTRACT

Previously, we showed that the 66 and 51 kDa soluble tumor-associated antigens (sTAAs) have distinct suppressive effects on chemically induced mammary cancer in rats, both alone and in combination with the hormone-related anticancer drug tamoxifen. Here, we describe the effects of both sTAA and tamoxifen on the histological structure of ovaries in mammary tumor-bearing 30- to 34-week-old rats. Central ovary sections were pooled, the number of the healthy and degenerated follicles were counted, and the size of the corpora lutea was estimated. In follicular development primordial, primary, preantral and antral stages were recognized. Only healthy follicles with visible nuclei were counted. Follicular degeneration was estimated as the number of atretic follicles with follicular remnants. Treatment with tamoxifen alone or in combination with sTAA significantly increased the number of primordial follicles and atretic follicles in the ovaries, and promoted the formation of small follicular cysts. Total area of the corpora lutea decreased. sTAA participated in this process by increasing apoptosis in degenerated follicles.


Subject(s)
Antigens, Neoplasm , Antineoplastic Agents, Hormonal/pharmacology , Mammary Neoplasms, Animal/physiopathology , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Tamoxifen/pharmacology , Animals , Apoptosis , Female , Ovarian Follicle/pathology , Rats , Rats, Wistar
7.
Int J Mol Med ; 16(3): 401-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16077946

ABSTRACT

We studied the effect of ascending infections of the birth canal on the transport of maternal immunoglobulins (Igs) through the placental barrier in humans. The study was performed on 41 human placentas obtained from embryos (n=21) and fetuses (n=20) who had died from different causes, including those connected with ascending infections of the birth canal, and seven placentas obtained after normal delivery at term. Different morphological and immunohistochemical methods were used. The transfer of Igs through the placental barrier is a complex process that involves tissues (trophoblast, stroma of the trophoblastic villi, and capillaries), cells (monocytes and erythroblasts) and molecular components (at least six types of transfer receptors and biologically active components). We found that the intensification of transfer of different types of maternal Igs (IgG, IgA, IgM) is accompanied by certain morphological and functional changes in the placental barrier. In normal development without infection, the transfer of IgG is steady and the process most intensive, while the transfer of IgA was evaluated in 75% of the cases, and of IgM in only 10%. Inflammation of the birth canal was accompanied by an increase in the transport of IgG in early embryogenesis, which was maintained throughout intrauterine development. In cases with moderate infection, transfer of IgG and IgA was found in all cases studied, while transfer of IgM was seen in 45% of the cases. In cases with massive infection, transfer of all three types of Igs was seen, the most intensive being of IgG and the least of IgM. Ascending infection of the birth canal changes dramatically the transport of Igs through the placenta and can be dangerous and even fatal for the embryo or fetus.


Subject(s)
Immunoglobulins/metabolism , Inflammation/metabolism , Placenta/metabolism , Acute Disease , Antigens, CD/analysis , Female , Humans , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Immunohistochemistry , Infections/metabolism , Infections/physiopathology , Inflammation/physiopathology , Placenta/blood supply , Placenta/chemistry , Pregnancy , Protein Transport , Receptors, Fc/analysis , Time Factors , Trophoblasts/chemistry , Trophoblasts/metabolism
8.
Int J Mol Med ; 16(1): 127-33, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15942689

ABSTRACT

The role of protein components of the secretory immune system (SIS), such as the polymeric immunoglobulin receptor/secretory component (pIgR/SC), immunoglobulins (Igs) and joining (J) chain, in human intrauterine development was reviewed. These components are already present in 3.5- to 4-week-old embryos, and found in all tissues and organs of epithelial origin. The SIS is made up of two parts: the SIS of mucous membranes and their derivatives (mucosal or secretory immune system), and the SIS of barrier structures (barrier immune system). During organogenesis, SC disappears from the cells of organs that lose their exocrine Ig-secretion function, such as the hypophysis, pancreatic islands and adrenal glands. In cells and tissues of mesenchymal origin, SC is absent from the start, i.e. during their initial development. As examples of the barrier immune system, blood-tissue and tissue-tissue barriers, such as the chorion of the placenta, the epithelium of the choroid plexuses in the brain, as well as other barrier structures to Ig transfer were considered. Besides the SC and J chain, Fc receptors, cellular and tissue structures participate in this process. Three stages were described in Ig transfer: i) passing from the maternal blood into intervillous spaces and the trophoblast, ii) shifting in the intravillous stroma and its cells, and iii) excretion into embryonic (fetal) blood through the endothelium of the trophoblastic villous capillaries. Igs of maternal origin, mainly IgG and least abundant IgA, pass through the placental barrier in healthy embryos. Following a massive antigenic attack, the increased exocrine secretion of IgG, IgA, and IgM to a lesser extent, are already seen in embryos, reflecting increased functional activity of the SIS. Thus, in human intrauterine development, the SIS is a very early immune defensive system, which presents and acts before the appearance of the common lymphoid system.


Subject(s)
Embryo, Mammalian/embryology , Embryo, Mammalian/immunology , Immunity, Mucosal/immunology , Placenta/immunology , Uterus/immunology , Uterus/pathology , Female , Humans , Immunoglobulins/immunology , Immunoglobulins/metabolism , Uterus/metabolism
9.
Stem Cells ; 23(4): 561-74, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15790777

ABSTRACT

The mechanism of human stem cell expansion ex vivo is not fully understood. Furthermore, little is known about the mechanisms of human stem cell homing/repopulation and the role that differentiating progenitor cells may play in these processes. We report that 2- to 3-day in vitro cytokine stimulation of human cord blood CD34(+)-enriched cells induces the production of short-term repopulating, cycling G1 CD34(+)/CD38(+) cells with increased matrix metalloproteinase (MMP)-9 secretion as well as increased migration capacity to the chemokine stromal cell-derived factor-1 (SDF-1) and homing to the bone marrow of irradiated nonobese diabetic severe/combined immunodeficiency (NOD/SCID) mice. These cycling G1 cells enhance SDF-1-mediated in vitro migration and in vivo homing of quiescent G0 CD34(+) cells, which is partially abrogated after inhibition of MMP-2/-9 activity. Moreover, the engraftment potential of quiescent G0 SCID repopulating cells (SRCs) is also increased by the cycling G1 CD34(+)/CD38(+) cells. This effect is significantly abrogated after incubation of cycling G1 cells with a neutralizing anti-CXCR4 antibody. Our data suggest synergistic interactions between accessory cycling G1 CD34(+)/CD38(+) committed progenitor cells and quiescent, primitive G0 CD34(+)/CD38(-/low) SRC/stem cells, the former increasing the motility and engraftment potential of the latter, partly via secretion of MMP-9.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , G1 Phase , Resting Phase, Cell Cycle , Stem Cells/cytology , Animals , Bone Marrow Cells/physiology , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/metabolism , Chemotaxis , Colony-Forming Units Assay , Cytokines/metabolism , Fetal Blood/cytology , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, CXCR4/metabolism , Stem Cell Transplantation , Stem Cells/physiology
10.
Oncol Rep ; 13(4): 585-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15756427

ABSTRACT

This study examined whether soluble 66 and 51 kDa tumor-associated antigens (sTAA), isolated from the serum of rats with mammary cancer, possess specific suppressive effects on chemically-induced mammary tumorigenesis in syngeneic counterparts. Dimethylbenzanthracene (DMBA, 10 mg/rat, two administrations) was used to induce mammary tumors in 8-week-old Sprague Dawley rats. After the appearance of numerous tumors, preparations of sTAA (50 to 60 microg/rat in 0.5 ml sterile PBS) obtained from breast cancer patients (heterologous sTAA) or from syngeneic mammary tumor-bearing rats (syngeneic sTAA) were administered weekly for 12 weeks. The following groups of mammary tumor-bearing rats were studied: groups 1 and 3, control rats treated with saline; group 2, rats treated with heterologous sTAA; and group 4, rats treated with syngeneic sTAA. The experiment was terminated when tumors in 50% of the rats became ulcerous. The treatment with both types of sTAA significantly decreased, compared to controls and initial values, the yield and total area of the tumors. We conclude that syngeneic sTAA have tumor-suppressive properties, which are very similar to those in heterologous sTAA.


Subject(s)
Antigens, Neoplasm/biosynthesis , Immunotherapy, Active/methods , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/prevention & control , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/prevention & control , 9,10-Dimethyl-1,2-benzanthracene , Animals , Antigens, Neoplasm/immunology , Carcinogens/pharmacology , Electrophoresis, Polyacrylamide Gel , Female , Humans , Rats , Rats, Sprague-Dawley , Time Factors
11.
Stem Cells ; 22(6): 1085-100, 2004.
Article in English | MEDLINE | ID: mdl-15536198

ABSTRACT

A major problem after clinical hematopoietic stem cell transplantations is poor T-cell reconstitution. Studying the mechanisms underlying this concern is hampered, because experimental transplantation of human stem and progenitor cells into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice usually results in low T-lymphocyte reconstitution. Because tumor necrosis factor alpha (TNFalpha) has been proposed to play a role in T-lineage commitment and differentiation in vitro, we investigated its potential to augment human T-cell development in vivo. Administration of TNF to irradiated NOD/SCID mice before transplantation of human mononuclear cells from either cord blood or adult G-CSF-mobilized peripheral blood (MPBL) led 2-3 weeks after transplantation to the emergence of human immature CD4(+)CD8(+) double-positive T-cells in the bone marrow (BM), spleen, and thymus, and in this organ, the human cells also express CD1a marker. One to 2 weeks later, single-positive CD4(+) and CD8(+) cells expressing heterogenous T-cell receptor alpha beta were detected in all three organs. These cells were also capable of migrating through the blood circulation. Interestingly, human T-cell development in these mice was associated with a significant reduction in immature lymphoid human CD19(+) B cells and natural killer progenitors in the murine BM. The human T cells were mostly derived from the transplanted immature CD34(+) cells. This study demonstrates the potential of TNF to rapidly augment human T lymphopoiesis in vivo and also provides clinically relevant evidence for this process with adult MPBL progenitors.


Subject(s)
T-Lymphocytes/cytology , Tumor Necrosis Factor-alpha/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , CD4 Antigens/biosynthesis , CD4-Positive T-Lymphocytes/cytology , CD8 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/cytology , Cell Lineage , Cell Movement , Cell Transplantation , DNA/metabolism , Flow Cytometry , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Kinetics , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Lymphocytes/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Phenotype , Polymerase Chain Reaction , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Stem Cell Transplantation , T-Lymphocytes/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
12.
Oncol Rep ; 12(6): 1329-33, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15547759

ABSTRACT

We showed previously that soluble tumor-associated antigens (sTAA) isolated from breast cancer patients could suppress chemically-induced tumorigenesis in rats in comparison to the effect of commercial human albumin (CHA). Herein we analyze the possible mechanism of those findings. The following groups of mammary tumor-bearing rats were used in the studies: i) control rats treated with saline; ii) rats treated with CHA; and iii) rats treated with human sTAA. Different zones of the spleen, regional lymph nodes and tumors and their cellular content (B and T cells) were analyzed using the methods of morphometry and immunohistochemistry. Treatment of tumor-bearing rats with CHA resulted in a significant decrease in the size of the germinal center of the follicles. The number of B lymphocytes in the mantle layer of the follicles, the marginal zone and red pulp decreased significantly. The number of CD8+ T cells also decreased in the marginal zone and red pulp, whereas the number of CD4+ T cells increased in the periarterial lymph sheath (PALS) and the red pulp. Reaction of the spleen to vaccination with sTAA manifested in a significant increase in the size of most areas of the white pulp and in the number of B lymphocytes. In lymph nodes from control rats or those treated with CHA, CD8+ lymphocytes mainly accumulated in the paracortical zone. In rats treated with sTAA, CD8+ lymphocytes accumulated also in the medulla. The number of CD4+ T cells in these rats sharply increased and accumulated mainly in the medulla around the vessels. The total number of lymphocytes was changed differently in different areas of tumors (peripheral vs. at depth). The number of CD8+ cells significantly increased at depth of tumors, and also the ratio in the number of these cells at depth of tumors compared to a periphery increased. No difference was found in response of lymph cells to different types of treatment. All findings indicated a strict antitumor effect of vaccination with the sTAA, which prevents the development of insufficiency of the immune system when an intensive immune reaction takes place.


Subject(s)
Albumins/pharmacology , Antigens, Neoplasm/pharmacology , B-Lymphocytes/drug effects , Cancer Vaccines/immunology , Mammary Neoplasms, Experimental/immunology , T-Lymphocytes/drug effects , Animals , B-Lymphocytes/immunology , Humans , Lymph Nodes/drug effects , Lymph Nodes/immunology , Rats , Rats, Sprague-Dawley , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/immunology
13.
Oncol Rep ; 12(1): 181-5, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15201981

ABSTRACT

We analyzed the role of T- and B-lymphocytes in the antitumor effects of the anticancer drug tamoxifen and soluble tumor-associated antigens (sTAA) on rat mammary carcinogenesis. Studies were performed on the spleen from the following groups of mammary tumor-bearing rats. i) Rats in group 1 were not exposed to DMBA and served as age-related controls. Rats in other groups were exposed to DMBA and received different types of treatment; ii) rats in group 2, received no additional treatment, and served as carcinogen-related controls; iii) rats in group 3 were treated with the commercial hormone-dependent anticancer drug tamoxifen by weekly subcutaneous (s.c.) injections of 10 mg dissolved in 0.5 ml distilled water per rat; iv) rats in group 4 were vaccinated s.c. weekly with a preparation of sTAA (50 micro l/rat) dissolved in 0.5 ml of phosphate-buffered saline; v) rats in group 5 were treated with tamoxifen and were also vaccinated with a preparation of sTAA. Different zones of the spleen were measured and their T- and B-cell contents were analyzed immunohistochemically. The treatment with tamoxifen significantly increased the total number of lymphocytes in the follicles, PALS (periarterial lymph sheath) and red pulp relative to all other groups. The combined treatment with tamoxifen and sTAA increased the areas of white pulp, the PALS, and marginal zone. The number of B-cells was higher in the marginal zone of spleens from age-related controls, as well as from rats treated with sTAA and those treated with tamoxifen and sTAA. The number of CD4+ lymphocytes in the PALS was higher in rats treated with sTAA and tamoxifen, and notably so in those treated with sTAA alone. The number of CD8+ lymphocytes was significantly lower in the PALS of spleens from all tumor-bearing rat groups compared to the unexposed age-related control rats. We suggest that the tumor-suppressive effect of sTAA and tamoxifen is accompanied by the activation of B- and T-lymphocyte production.


Subject(s)
Antigens, Neoplasm/therapeutic use , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Spleen/immunology , T-Lymphocytes/immunology , Tamoxifen/therapeutic use , Animals , Female , Mammary Neoplasms, Experimental/drug therapy , Rats , Rats, Sprague-Dawley
14.
Int J Mol Med ; 14(1): 35-42, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15202014

ABSTRACT

The role of joining (J) chain, one of the protein components of the secretory immune system (SIS), in the immune reactions of the human embryo and fetus was analyzed on the basis of data from the literature and our previous studies. All organs and structures, including extra-corporeal ones, of 18 embryos (4-8 weeks of development) and 45 fetuses (9-38 weeks) were studied using methods of pathomorphology, immunohistochemistry and morphometry. This approach enabled us to analyze the problem in the whole organism throughout its embryonic and fetal development. J chain, as well as polymeric immunoglobulin (Ig) receptor-secretory component (pIgR/SC) and Igs, are already widely distributed in 4-week-old embryos before the appearance of the common immune system. The whole complex of protein components of the SIS was seen in mucous layers, and in blood-tissue and tissue-tissue barrier structures. Therefore, we can consider two parts of the SIS: mucosal and barrier. Already in embryos, an increase in the functional activity of the SIS following massive antigenic attack in cases of acute chorioamnionitis reflects the increased exocrine secretion of Igs. The J chain appears to participate in the endocytosis but not exocytosis of Igs. J chain and Igs, but not pIgR/SC, were present in cells of the heart, endocrine glands, gonads and some other organs. The exocrine secretion of Igs, the main function of the SIS, is absent in these organs, and, they are therefore, not considered part of the SIS.


Subject(s)
Embryonic Development/immunology , Fetal Development/immunology , Immune System/metabolism , Immunoglobulin J-Chains/metabolism , Immunoglobulins/metabolism , Humans , Immunoglobulin J-Chains/analysis , Organogenesis/immunology , Protein Transport , Secretory Component/analysis , Secretory Component/metabolism
15.
Blood ; 103(8): 2981-9, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15070674

ABSTRACT

Trafficking of human CD34+ stem/progenitor cells (HSCs/HPCs) is regulated by chemokines, cytokines, proteolytic enzymes, and adhesion molecules. We report that the adhesion receptor CD44 and its major ligand, hyaluronic acid (HA), are essential for homing into the bone marrow (BM) and spleen of nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice and engraftment by human HSCs. Homing was blocked by anti-CD44 monoclonal antibodies (mAbs) or by soluble HA, and it was significantly impaired after intravenous injection of hyaluronidase. Furthermore, stromal cell-derived factor-1 (SDF-1) was found to be a rapid and potent stimulator of progenitor adhesion to immobilized HA, leading to formation of actin-containing protrusions with CD44 located at their tips. HPCs migrating on HA toward a gradient of SDF-1 acquired spread and polarized morphology with CD44 concentrating at the pseudopodia at the leading edge. These morphologic alterations were not observed when the progenitors were first exposed to anti-CD44 mAbs, demonstrating a crosstalk between CD44 and CXCR4 signaling. Unexpectedly, we found that HA is expressed on human BM sinusoidal endothelium and endosteum, the regions where SDF-1 is also abundant. Taken together, our data suggest a key role for CD44 and HA in SDF-1-dependent transendothelial migration of HSCs/HPCs and their final anchorage within specific niches of the BM.


Subject(s)
Antigens, CD34/metabolism , Chemokines, CXC/pharmacology , Hematopoietic Stem Cells/physiology , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Movement/drug effects , Chemokine CXCL12 , Chemotaxis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Heterologous
16.
Oncol Rep ; 11(2): 487-91, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14719088

ABSTRACT

This study examined whether the soluble 66- and 51 kDa tumor-associated antigens (sTAA), isolated from the serum of breast cancer patients, possess specific suppressive effects on chemically-induced rat mammary tumorigenesis in comparison to commercial human albumin. Dimethylbenzanthracene (DMBA, 10 mg/rat, 2 administrations) was used to induce mammary tumors in 8-week-old Sprague Dawley rats. After the appearance of many large tumors, preparations of sTAA (50-60 micro g/rat in 0.5 ml sterile PBS) or commercial human albumin (HA, in the same doses as sTAA) were administered weekly, for 10-14 more weeks. The following groups of mammary tumor-bearing rats were studied: i) control non-treated rats, ii) rats treated with HA, iii) rats treated with sTAA. The experiment was terminated when tumors in 70% of the rats became ulcerous. The treatment with sTAA significantly decreased, compared to controls, the yield and total area of the tumors. In rats treated with sTAA, the appearance of new tumors stopped at week 5 as compared to week 7 in rats treated with HA and week 10 in control rats. In rats treated with sTAA, the time of appearance of ulcerous tumors increased to 8 weeks, as compared to 6 weeks in controls and in rats treated with HA. Duration of the experiment increased from 11 weeks in controls to 12 weeks in rats treated with HA and to 14 weeks in rats treated with sTAA. We conclude that sTAA have tumor-suppressive properties, which are well-defined if the treatment is begun on small tumors.


Subject(s)
Antigens, Neoplasm/isolation & purification , Antigens, Neoplasm/therapeutic use , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Serum Albumin/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Anticarcinogenic Agents/therapeutic use , Female , Humans , Mammary Neoplasms, Experimental/chemically induced , Molecular Weight , Rats
17.
Int J Mol Med ; 12(5): 797-801, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14533012

ABSTRACT

This study examined whether the soluble 66 and 51 kDa tumor-associated antigens (sTAA) could promote suppression by the anticancer drug 5-fluorouracil (5-Fu) of chemically induced mammary tumorigenesis, and which, if any, morphological changes in the immune organs accompany this treatment. Dimethylbenzanthracene (DMBA, 8 mg/rat, twice) was used to induce mammary tumors. After the appearance of many large tumors, the preparations of sTAA and 5-Fu, alone or in combination, were administered in weekly doses, for 4 weeks. The following groups of mammary tumor-bearing rats were studied: 1) control non treated rats, 2) rats treated with sTAA, 3) rats treated with 5-Fu, 4) rats treated with 5-Fu and sTAA. The experiment was terminated when tumors in 70% of control rats became ulcerous. Treatment with sTAA alone significantly decreased tumor yield and their total area relative to controls. Both of these parameters showed an even larger significant decrease after treatment with 5-Fu, and the most marked decrease was obtained after the combined treatment with 5-Fu and sTAA. Results demonstrated that not only do sTAA have tumor-suppressive properties, they also enhance the anticancer effects of 5-Fu and prevent its toxic side effects. Morphologically, the treatment with sTAA was manifested in a significant increase in the size of the spleen follicles and mantle layer compared to control rats with large tumors. The treatment with 5-Fu decreased the sizes of almost all areas of the spleen compared to control rats, whereas the combined treatment with 5-Fu and sTAA increased all these parameters to the levels found in rats treated with sTAA alone. The total areas of the cortex and paracortex in the lymph nodes increased after treatment with sTAA. Treatment with 5-Fu alone resulted in a significant decrease of these areas which, as seen in the spleen, increased after combined treatment with 5-Fu and sTAA. Similar changes were seen in the areas of the separate lymph node zones. We concluded that the addition of sTAA to conventional tumor chemotherapy regimens has a remarkable synergistic effect on mammary tumors leading to curative antitumor responses of the host's immune organs.


Subject(s)
Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/therapeutic use , Fluorouracil/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , Animals , Antigens, Neoplasm/pharmacology , Female , Fluorouracil/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/pathology , Rats , Rats, Sprague-Dawley , Solubility , Spleen/drug effects , Spleen/pathology
18.
Oncol Rep ; 10(6): 2059-61, 2003.
Article in English | MEDLINE | ID: mdl-14534743

ABSTRACT

In this communication, we report for the first time, that immunization of cancer patients with autologous soluble tumor-associated antigens (sTAA) isolated from their own serum prevents the toxic side effects of chemotherapy, improves the patients' clinical status, and has therapeutic effects without chemotherapy. In 2001 and 2002, two cancer patients were treated, during chemotherapy, with autologous sTAA. Another benign tumor-bearing patient was treated with a medicinal herb and autologous sTAA. Doses for subcutaneous injections varied between 2.5 and 3 mg of sTAA in 0.5 ml of sterile distilled water. Injections were performed twice a week or at weekly intervals. In each case, the clinical status of the patient became more stable and healthier. Toxic side effects caused by chemotherapy decreased or even disappeared. No additional toxic side effects were observed after vaccination with sTAA. In the studied cases, a polyp disappeared and a metastatic brain tumor began to encapsulate. No metastases were seen in the case with colon adenocarcinoma. We concluded that vaccination of patients with autologous sTAA prevents the toxic side effects of chemotherapy in cancer patients and improves their clinical status. In the case with the benign tumor, this vaccination activated the host's immune system, prevented progress of the disease and even promoted tumor disappearance. We suggest that immunotherapy with autologous sTAA provides significant clinical benefits in cancer patients and appears to be an important new adjuvant treatment of cancer.


Subject(s)
Adjuvants, Pharmaceutic/therapeutic use , Antigens, Neoplasm/biosynthesis , Antineoplastic Agents/pharmacology , Cancer Vaccines , Immunotherapy, Active/methods , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Colon, Sigmoid/surgery , Colonic Diseases/therapy , Female , Humans , Male , Melanoma/pathology , Melanoma/surgery , Middle Aged , Polyps/therapy , Vaginal Diseases/therapy
19.
Int J Mol Med ; 12(4): 473-7, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12964022

ABSTRACT

The effect of the synthetic pineal peptide Epitalon (Ala-Glu-Asp-Gly) on proliferative activity in colon tumors, and in mucosal epithelial cells adjacent to and located far from tumors was studied in rats. To evaluate the effect of Epitalon on different stages of carcinogenesis, different treatment regimens were used: during the tumor initiation stage, during the tumor-promotion stage, or during the entire process of tumor development. Eighty 2-month-old male LIO rats were exposed weekly to five subcutaneous injections of 1,2-dimethylhydrazine (DMH) at a single dose of 21 mg/kg body weight. Rats were divided into four groups. Control rats (group 1) received saline at a dose of 0.1 ml during the entire experiment. Rats in group 2 were treated with Epitalon at a dose of 1 micro g, five times a week, for 6 months, from the first injection of DMH till the end of the experiment. Rats in group 3 were treated with Epitalon after termination of the carcinogen injections. Rats in group 4 were treated with Epitalon only during the period of DMH exposure (for the first 5 weeks of the experiment). DMH induced proliferation of the secretory epithelium, and this phenomenon was accompanied by a decrease in the size of the stromal area and the area of lymph infiltration in colon tumors and in the colon mucosa adjacent to the tumors (group 1). Epitalon attenuated this effect, especially when the treatment was continued throughout the experiment (group 2). It increased the stromal areas, as well as that of lymphoid infiltration in the colon mucosa adjacent to the tumors. The intensity of lymphoid infiltration was activated in both the colon mucosa adjacent to a tumor and in the tumor. Mitotic activity of tumor cells was significantly inhibited by Epitalon when the treatment was given throughout the experiment (group 2). In parallel, a high level of apoptosis was seen in the same group. Thus, the strongest inhibitory effect of Epitalon on carcinogenesis in the colon mucosa was manifested when the treatment was continued throughout the experiment.


Subject(s)
Apoptosis , Colonic Neoplasms/pathology , Oligopeptides/pharmacology , 1,2-Dimethylhydrazine , Animals , Carcinogens , Cell Division , In Situ Nick-End Labeling , Lymphatic Metastasis , Male , Mitosis , Mucous Membrane/pathology , Peptides/chemistry , Rats
20.
Am J Reprod Immunol ; 50(1): 13-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14506924

ABSTRACT

PROBLEM: We analyzed the presence and distribution of components of the secretory immune system (SIS) in human fetal membranes (amnion, yolk sac, chorion) and decidua from the first trimester of pregnancy. MATERIALS AND METHODS: Specimens from 17 embryos (4-8 weeks of pregnancy) and nine fetuses (9-12 weeks) were divided into those that had not been exposed to massive foreign antigenic effects (Group I, n = 18) and those that had suffered acute chorioamnionitis (Group II, n = 8). RESULTS: Positive immunostaining for secretory component (SC), joining (J) chain, IgG, IgA, and macrophages was seen from 4 to 5 weeks of development and then during the whole first trimester of pregnancy in the syncytio- and cytotrophoblasts, the amniotic epithelium, the yolk sac endoderm and decidual cells. Macrophages with J chain, IgG and IgA were found in embryonic tissues on week 4, whereas lymphocytes, including those synthesizing IgA and IgM, appeared only at the end of the first trimester of pregnancy. In the decidua, lymphocytes and macrophages were recognized during the whole period of study. In cases with chorioamnionitis (Group II), reactivity of IgG and IgA in the mentioned cells of fetuses decreased sharply while the rate of immunoreactivity of SC and J chain as well as the number of T and B lymphocytes did not change. In the decidua, the number of immune reactive cells sharply increased with the appearance of plasma cells. CONCLUSIONS: In the fetal membranes and decidua all the SIS components are present. We suggest that two SIS, maternal and fetal, participate in the formation of the barrier between a mother and the fetus. Both these systems have different origin and cellular content as well as different immune reactions.


Subject(s)
Decidua/immunology , Extraembryonic Membranes/immunology , Immune System/metabolism , Immunoglobulin J-Chains , Pregnancy Trimester, First/immunology , B-Lymphocytes/immunology , Chorioamnionitis/physiopathology , Decidua/cytology , Extraembryonic Membranes/cytology , Female , Humans , Immune System/cytology , Immune System/embryology , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Immunoglobulin J-Chains/analysis , Immunoglobulin M/analysis , Immunohistochemistry , Macrophages/immunology , Pregnancy , Secretory Component/analysis , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...