Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Oncogene ; 32(35): 4139-47, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-22986525

ABSTRACT

The miR-106b-25 microRNA (miRNA) cluster is a candidate oncogene in human prostate cancer. Here, we report that miRNAs encoded by miR-106b-25 are upregulated in both primary tumors and distant metastasis. Moreover, increased tumor miR-106b expression was associated with disease recurrence and the combination of high miR-106b and low CASP7 (caspase-7) expressions in primary tumors was an independent predictor of early disease recurrence (adjusted hazard ratio=4.1; 95% confidence interval: 1.6-12.3). To identify yet unknown oncogenic functions of miR-106b, we overexpressed it in LNCaP human prostate cancer cells to examine miR-106b-induced global expression changes among protein-coding genes. The approach revealed that CASP7 is a direct target of miR-106b, which was confirmed by western blot analysis and a 3'-untranslated region reporter assay. Moreover, selected phenotypes induced by miR-106b knockdown in DU145 human prostate cancer cells did not develop when both miR-106b and CASP7 expression were inhibited. Further analyses showed that CASP7 is downregulated in primary prostate tumors and metastatic lesions across multiple data sets and is by itself associated with disease recurrence and disease-specific survival. Using bioinformatics, we also observed that miR-106b-25 may specifically influence focal adhesion-related pathways. This observation was experimentally examined using miR-106b-25-transduced 22Rv1 human prostate cancer cells. After infection with a miR-106b-25 lentiviral expression construct, 22Rv1 cells showed increased adhesion to basement membrane- and bone matrix-related filaments and enhanced soft agar growth. In summary, miR-106b-25 was found to be associated with prostate cancer progression and disease outcome and may do so by altering apoptosis- and focal adhesion-related pathways.


Subject(s)
Caspase 7/genetics , Focal Adhesions , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Neoplasm Recurrence, Local/etiology , Prostatic Neoplasms/genetics , 3' Untranslated Regions , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Male , Neoplasm Metastasis , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology
2.
J Immunol ; 167(12): 6804-11, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739496

ABSTRACT

Increasing the long-term survival of memory T cells after immunization is key to a successful vaccine. In the past, the generation of large numbers of memory T cells in vivo has been difficult because Ag-stimulated T cells are susceptible to activation-induced cell death. Previously, we reported that OX40 engagement resulted in a 60-fold increase in the number of Ag-specific CD4(+) memory T cells that persisted 60 days postimmunization. In this report, we used the D011.10 adoptive transfer model to examine the kinetics of Ag-specific T cell entry into the peripheral blood, the optimal route of administration of Ag and alphaOX40, and the Ag-specific Ab response after immunization with soluble OVA and alphaOX40. Finally, we compared the adjuvant properties of alphaOX40 to those of alphaCTLA-4. Engagement of OX-40 in vivo was most effective when the Ag was administered s.c. Time course studies revealed that it was crucial for alphaOX40 to be delivered within 24-48 h after Ag exposure. Examination of anti-OVA Ab titers revealed a 10-fold increase in mice that received alphaOX40 compared with mice that received OVA alone. Both alphaOX40 and alphaCTLA-4 increased the percentage of OVA-specific CD4(+) T cells early after immunization (day 4), but alphaOX40-treated mice had much higher percentages of OVA-specific memory CD4(+) T cells from days 11 to 29. These studies demonstrate that OX40 engagement early after immunization with soluble Ag enhances long-term T cell and humoral immunity in a manner distinct from that provided by blocking CTLA-4.


Subject(s)
Antigens, Differentiation/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Immunoconjugates , Immunoglobulin G/biosynthesis , Immunologic Memory , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Abatacept , Adjuvants, Immunologic/administration & dosage , Adoptive Transfer , Animals , Antibodies/administration & dosage , Antigens/administration & dosage , Antigens/immunology , Antigens, CD , Antigens, Differentiation/immunology , Blood/immunology , CD4-Positive T-Lymphocytes/transplantation , CTLA-4 Antigen , Cell Survival , Cytokines/biosynthesis , Female , Kinetics , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, OX40 , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
3.
J Exp Med ; 194(4): 481-9, 2001 Aug 20.
Article in English | MEDLINE | ID: mdl-11514604

ABSTRACT

We have previously shown that small B16 melanomas can be successfully treated using a combination of anti-cytotoxic T lymphocyte antigen (CTLA)-4 monoclonal antibody with a granulocyte/macrophage colony-stimulating factor (GM-CSF) producing irradiated tumor cell vaccine. Regression of tumors results in long-lasting immunity and is frequently accompanied by autoimmune depigmentation. Here we examine the cellular and molecular mechanisms of this combined treatment. Histological examination of depigmented lesions revealed infiltration of polymorphonuclear cells and deposition of antibody. The combination therapy also induced tumor rejection and skin depigmentation in B cell-deficient and in CD4(+) T cell-depleted mice. Both effects of the treatment absolutely required CD8(+) T cells. Analysis of the response in successfully treated mice revealed elevated levels of CD8(+) T cells specific for a nonameric peptide consisting of residues 180-188 of the melanocyte differentiation antigen tyrosinase-related protein (TRP)2. There was no evidence of reactivity to the melanocyte antigens gp100, tyrosinase, Mart1/MelanA, or TRP1. Fas-FasL interactions and perforin played a role in mounting the effector response, whereas the tumor necrosis factor pathway was not required. The cellular requirements for tumor rejection in this therapeutic setting were strikingly different from those in a prophylactic setting. In particular, if mice received a prophylactic vaccine consisting of anti-CTLA-4 and B16-GM-CSF before tumor challenge, full protection was obtained even in the absence of CD8(+) T cells. Our data demonstrate that therapeutic autoreactive CD8(+) T cell responses can effectively be generated in tumor-bearing mice and stresses the value of studying tumor immunity in a therapeutic rather than a prophylactic setting.


Subject(s)
Antigens, Differentiation/immunology , Cancer Vaccines/immunology , Immunoconjugates , Melanoma, Experimental/therapy , Abatacept , Animals , Antigens, CD , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Cancer Vaccines/therapeutic use , Female , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL
4.
Curr Opin Immunol ; 12(5): 589-96, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11007364

ABSTRACT

Advances in our understanding of T cell costimulatory molecules have provided a vast array of novel approaches to tumor immunotherapy. In the past year, combinatorial immunotherapy based on earlier studies of CTLA-4 blockade, the identification of novel B7-family members, the modulation of CD40 to reverse tolerance to tumor-associated antigens and the use of OX40 to enhance antitumor responses of CD4+ T cells have all contributed to the development of more-powerful immunomodulatory cancer therapies.


Subject(s)
Immunoconjugates , Immunotherapy , Neoplasms/therapy , Receptors, Tumor Necrosis Factor , Abatacept , Antigens, CD , Antigens, Differentiation/physiology , B7-1 Antigen/physiology , CD28 Antigens/physiology , CD40 Antigens/physiology , CTLA-4 Antigen , Humans , Membrane Proteins/physiology , Receptors, OX40 , Tumor Necrosis Factor Ligand Superfamily Member 14 , Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology , Tumor Necrosis Factor-alpha/physiology
5.
Cancer Res ; 60(9): 2444-8, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10811122

ABSTRACT

We have previously shown that antibodies to CTLA-4, an inhibitory receptor on T cells, can be effective at inducing regression of transplantable murine tumors. In this study, we demonstrate that an effective immune response against primary prostate tumors in transgenic (TRAMP) mice can be elicited using a strategy that combines CTLA-4 blockade and an irradiated tumor cell vaccine. Treatment of TRAMP mice at 14 weeks of age resulted in a significant reduction in tumor incidence (15% versus control, 75%), as assessed 2 months after treatment. Histopathological analysis revealed that treated mice had a lower tumor grade with significant accumulation of inflammatory cells in interductal spaces when treated with anti-CTLA-4 and a granulocyte-macrophage colony-stimulating factor-expressing vaccine. Vaccination of nontransgenic mice with this regimen resulted in marked prostatitis accompanied by destruction of epithelium, indicating that the immune response was, at least in part, directed against normal prostate antigens. These findings demonstrate that this combinatorial treatment can elicit a potent antiprostate response and suggest potential of this approach for treatment of prostate cancer.


Subject(s)
Adenocarcinoma/therapy , Antigens, Differentiation/immunology , Cancer Vaccines/therapeutic use , Immunization, Passive , Immunoconjugates , Prostatic Neoplasms/therapy , Abatacept , Animals , Antibodies, Neoplasm/pharmacology , Antigens, CD , CTLA-4 Antigen , Cancer Vaccines/radiation effects , Immunohistochemistry , Inflammation , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/immunology
6.
Proc Natl Acad Sci U S A ; 96(26): 15074-9, 1999 Dec 21.
Article in English | MEDLINE | ID: mdl-10611340

ABSTRACT

Cancer relapse after surgery is a common occurrence, most frequently resulting from the outgrowth of minimal residual disease in the form of metastases. We examined the effectiveness of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade as an adjunctive immunotherapy to reduce metastatic relapse after primary prostate tumor resection. For these studies, we developed a murine model in which overt metastatic outgrowth of TRAMP-C2 (C2) prostate cancer ensues after complete primary tumor resection. Metastatic relapse in this model occurs reliably and principally within the draining lymph nodes in close proximity to the primary tumor, arising from established metastases present at the time of surgery. Using this model, we demonstrate that adjunctive CTLA-4 blockade administered immediately after primary tumor resection reduces metastatic relapse from 97.4 to 44%. Consistent with this, lymph nodes obtained 2 weeks after treatment reveal marked destruction or complete elimination of C2 metastases in 60% of mice receiving adjunctive anti-CTLA-4 whereas 100% of control antibody-treated mice demonstrate progressive C2 lymph node replacement. Our study demonstrates the potential of adjunctive CTLA-4 blockade immunotherapy to reduce cancer relapse emanating from minimal residual metastatic disease and may have broader implications for improving the capability of immunotherapy by combining such forms of therapy with other cytoreductive measures including surgery.


Subject(s)
Antibodies/therapeutic use , Antigens, Differentiation/immunology , Immunoconjugates , Immunotherapy/methods , Prostatic Neoplasms/therapy , Abatacept , Animals , Antigens, CD , CTLA-4 Antigen , Chemotherapy, Adjuvant/methods , Lymphatic Metastasis/prevention & control , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/mortality , Prostatic Neoplasms/surgery
7.
J Exp Med ; 190(3): 355-66, 1999 Aug 02.
Article in English | MEDLINE | ID: mdl-10430624

ABSTRACT

We examined the effectiveness of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade, alone or in combination with a granulocyte/macrophage colony-stimulating factor (GM-CSF)-expressing tumor cell vaccine, on rejection of the highly tumorigenic, poorly immunogenic murine melanoma B16-BL6. Recently established tumors could be eradicated in 80% (68/85) of the cases using combination treatment, whereas each treatment by itself showed little or no effect. Tumor rejection was dependent on CD8(+) and NK1.1(+) cells but occurred irrespective of the presence of CD4(+) T cells. Mice surviving a primary challenge rejected a secondary challenge with B16-BL6 or the parental B16-F0 line. The same treatment regimen was found to be therapeutically effective against outgrowth of preestablished B16-F10 lung metastases, inducing long-term survival. Of all mice surviving B16-BL6 or B16-F10 tumors after combination treatment, 56% (38/68) developed depigmentation, starting at the site of vaccination or challenge and in most cases progressing to distant locations. Depigmentation was found to occur in CD4-depleted mice, strongly suggesting that the effect was mediated by CTLs. This study shows that CTLA-4 blockade provides a powerful tool to enhance T cell activation and memory against a poorly immunogenic spontaneous murine tumor and that this may involve recruitment of autoreactive T cells.


Subject(s)
Antigens, Differentiation/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/immunology , Autoimmune Diseases/immunology , Cancer Vaccines/immunology , Graft Rejection/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Immunoconjugates , Melanoma, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Vitiligo/immunology , Abatacept , Adjuvants, Immunologic/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antigens/immunology , Antigens, CD , Antigens, Differentiation/immunology , Antigens, Ly , Antigens, Surface , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autoimmune Diseases/therapy , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Cancer Vaccines/therapeutic use , Cell Survival/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Hair Color/immunology , Immunosuppressive Agents/therapeutic use , Injections, Subcutaneous , Lectins, C-Type , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymphocyte Activation , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B , Neoplasm Transplantation , Proteins/immunology , Skin Pigmentation/immunology , Vitiligo/pathology , Vitiligo/therapy
8.
Proc Natl Acad Sci U S A ; 95(17): 10067-71, 1998 Aug 18.
Article in English | MEDLINE | ID: mdl-9707601

ABSTRACT

Generation of a T cell-mediated antitumor response depends on T cell receptor engagement by major histocompatibility complex/antigen as well as CD28 ligation by B7. CTLA-4 is a second B7 receptor expressed by T cells upon activation that, unlike CD28, appears to deliver an inhibitory signal to T cells. Recently, we and others demonstrated that administration of an anti-CTLA-4 antibody was sufficient to promote regression of several murine tumors. However, certain tumors, such as the SM1 mammary carcinoma, remain refractory to this type of immunotherapy. In the present study, we report that the combination of both CTLA-4 blockade and a vaccine consisting of granulocyte-macrophage colony-stimulating factor-expressing SM1 cells resulted in regression of parental SM1 tumors, despite the ineffectiveness of either treatment alone. This synergistic therapy resulted in long-lasting immunity to SM1 and depended on both CD4(+) and CD8(+) T cells. Interestingly, synergy was not observed between CTLA-4 and a B7-expressing SM1 vaccine. Given that granulocyte-macrophage colony-stimulating factor promotes differentiation and activation of dendritic cells as well as enhances cross-priming of T cells to tumor-derived antigens and that SM1 is major histocompatibility complex class II-negative, our findings suggest that CTLA-4 blockade acts at the level of a host-derived antigen-presenting cell. In addition, these results also support the idea that the most effective and synergistic vaccine strategy targets treatments that enhance T cell priming at the level of host-derived antigen-presenting cells.


Subject(s)
Antigens, Differentiation/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunoconjugates , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Abatacept , Animals , Antibodies, Monoclonal/administration & dosage , Antigen-Presenting Cells/immunology , Antigens, CD , B7-1 Antigen/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Cancer Vaccines/administration & dosage , Female , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunotherapy , Interferon-gamma/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Transduction, Genetic
9.
Int J Cancer ; 77(1): 107-13, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9639401

ABSTRACT

In recent years, tumor immunotherapy has begun to exploit the emerging knowledge of the mechanisms of T cell activation to enhance the immune responses to tumors. However, many tumors, despite genetic modification to express co-stimulatory molecules or cytokines, are not readily rejected due to their inherently poor immunogenicity. In the present study, we tested whether expression of the co-stimulatory ligand B7-1 and the immunostimulatory cytokines interferon gamma (IFN-gamma) and granulocyte-macrophage colony-stimulating factor (GM-CSF) by a mammary carcinoma (SM1) would sufficiently augment its immunogenicity to obtain rejection and immunity. Our findings demonstrate that expression of B7, IFN-gamma, or GM-CSF alone, or co-expression of B7 and GM-CSF did not result in rejection of SM1. However, co-expression of B7 and IFN-gamma was sufficient to result in regression of SM1 tumors by a CD8+ T cell-dependent mechanism. Rejection of the B7/IFN-gamma-expressing SM1 tumor resulted in protection from rechallenge not only with the unmodified SM1 tumor but with another syngeneic mammary tumor. Our data support the idea that although B7 expression alone may not be sufficient for rejection of certain tumors, the immune system may be stimulated to mount an effective anti-tumor immune response by the co-expression of both the co-stimulatory ligand and a cytokine.


Subject(s)
B7-1 Antigen/genetics , CD8-Positive T-Lymphocytes/immunology , Carcinoma/genetics , Carcinoma/immunology , Cytotoxicity, Immunologic/genetics , Interferon-gamma/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Animals , B7-1 Antigen/immunology , Female , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C
10.
Proc Natl Acad Sci U S A ; 94(15): 8099-103, 1997 Jul 22.
Article in English | MEDLINE | ID: mdl-9223321

ABSTRACT

The identification of potentially useful immune-based treatments for prostate cancer has been severely constrained by the scarcity of relevant animal research models for this disease. Moreover, some of the most critical mechanisms involved in complete and proper antitumoral T cell activation have only recently been identified for experimental manipulation, namely, components involved in the costimulatory pathway for T cell activation. Thus, we have established a novel syngeneic murine prostate cancer model that permits us to examine two distinct manipulations intended to elicit an antiprostate cancer response through enhanced T cell costimulation: (i) provision of direct costimulation by prostate cancer cells transduced to express the B7.1 ligand and (ii) in vivo antibody-mediated blockade of the T cell CTLA-4, which prevents T cell down-regulation. In the present study we found that a tumorigenic prostate cancer cell line, TRAMPC1 (pTC1), derived from transgenic mice, is rejected by syngeneic C57BL/6 mice, but not athymic mice, after this cell line is transduced to express the costimulatory ligand B7.1. Also, we demonstrated that in vivo antibody-mediated blockade of CTLA-4 enhances antiprostate cancer immune responses. The response raised by anti-CTLA-4 administration ranges from marked reductions in wild-type pTC1 growth to complete rejection of these cells. Collectively, these experiments suggest that appropriate manipulation of T cell costimulatory and inhibitory signals may provide a fundamental and highly adaptable basis for prostate cancer immunotherapy. Additionally, the syngeneic murine model that we introduce provides a comprehensive system for further testing of immune-based treatments for prostate cancer.


Subject(s)
B7-1 Antigen/immunology , CD28 Antigens/immunology , Immunoconjugates , Immunotherapy , Prostatic Neoplasms/therapy , Abatacept , Animals , Antigens, CD , Antigens, Differentiation/immunology , CTLA-4 Antigen , Histocompatibility Antigens Class I/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic
11.
J Neuroimmunol ; 73(1-2): 57-62, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9058759

ABSTRACT

In addition to an antigen-specific signal, T cell activation requires an antigen-independent costimulatory signal provided by interaction of CD28 with B7 (CD80 and CD86) on the APC. By blocking B7 interactions, previous studies demonstrated the requirement for costimulation in the induction of experimental allergic encephalomyelitis (EAE). Recent studies suggest that unlike CD28, CTLA-4 (a second B7 ligand) delivers an inhibitory signal. To address the regulatory role of CTLA-4 in EAE, we used an antibody directed against CTLA-4 administered at the time of disease induction. This resulted in a significantly more severe clinical course and more inflammatory and demyelinating lesions in the CNS of anti-CTLA-4-treated mice. These data suggest that CTLA-4-mediated inhibitory signals can regulate the clinical severity and histologic parameters of neuroautoimmune disease.


Subject(s)
Antigens, Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunoconjugates , Immunosuppressive Agents/immunology , Abatacept , Animals , Antibodies, Monoclonal/immunology , Antigens, CD , CTLA-4 Antigen , Cricetinae , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Mice , Mice, Inbred Strains
12.
Curr Opin Immunol ; 7(5): 682-6, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8573312

ABSTRACT

One of the major goals of tumor immunotherapy is the induction of tumor-specific T-cell responses that will be effective in eradicating disseminated tumors. Emerging information on the role of costimulatory molecules in T-cell activation offers several new strategies for enhancing antitumor responses, including the induction of expression of costimulatory molecules on tumor cells, enhancement of the presentation of transferred tumor antigen by host antigen-presenting cells, and ex vivo antigen priming of autologous antigen-presenting cells.


Subject(s)
Immunotherapy , Lymphocyte Activation/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antigen Presentation , Humans , Neoplasms/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy
13.
J Neuroimmunol ; 57(1-2): 193-8, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7706436

ABSTRACT

Astrocytes participate in the pathophysiology of central nervous system (CNS) inflammatory disease. Astrocyte expression of adhesion molecules, cytokines, and major histocompatibility complex antigens may contribute to these inflammatory processes. In addition, recent data suggested that astrocytes may be a source of monocyte chemoattractant protein-1 (MCP-1). MCP-1 is a member of the chemokine family of small cytokines and functions both as a chemoattractant as well as a stimulator of monocytes. To further characterize the role of astrocytes in CNS inflammation, we examined the effect of inflammatory cytokines on MCP-1 expression by astrocytes. Results of these studies demonstrate that the pro-inflammatory cytokine tumor necrosis factor alpha (TNF alpha) upregulates MCP-1 message and protein expression. The pleiotropic cytokine transforming growth factor beta (TGF beta) also stimulated MCP-1 expression. When astrocytes were exposed to both cytokines simultaneously, an additive effect on MCP-1 message, but not MCP-1 protein expression, was observed. These data suggest that TNF alpha and TGF beta, each present during CNS inflammatory disease, may upregulate the expression of MCP-1 which, in turn, may function to both recruit monocytes to the site of inflammation as well as to activate those monocytes already present in an inflammatory lesion.


Subject(s)
Astrocytes/metabolism , Chemotactic Factors/biosynthesis , Gene Expression Regulation/drug effects , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured , Chemokine CCL2 , Chemotactic Factors/genetics , Humans , Up-Regulation
14.
Adv Neuroimmunol ; 4(3): 249-56, 1994.
Article in English | MEDLINE | ID: mdl-7533041

ABSTRACT

The blood-brain barrier (BBB) functions to regulate the entry of macromolecules, microbial pathogens, and circulating leukocytes into the central nervous system (CNS). It consists, in part, of the microvascular endothelium and associated astrocyte foot processes, found in close apposition to the abluminal side of the vascular endothelial cells (EC). During the pathogenesis of certain nervous system diseases with inflammatory components, the BBB may function to facilitate the entry of leukocytes into the CNS parenchyma. A common histologic observation in human immunodeficiency virus type-1 (HIV) encephalitis is the localization of HIV proteins to multinucleated giant cells that co-immunolabel with antibodies specific for cells of the monocyte/macrophage lineage, suggesting that HIV can enter the CNS as cell-associated virus. We previously characterized a tissue culture model of the BBB that consists of the co-culture of autologous EC and astrocytes. In this presentation, we used this model to examine the expression of adhesion molecules by both the EC and astrocyte components of this BBB model, and to characterize the interactions between HIV-infected monocytes and EC. The data presented in this review of our work demonstrates that astrocytes upregulate the expression of intercellular adhesion molecule (ICAM-1) by EC. In a parallel study, western blot analysis demonstrated that ICAM-1 is also expressed in the developing human CNS. When exposed to the proinflammatory cytokine tumor necrosis factor alpha (TNF), both EC cocultured with astrocytes and astrocytes cultured alone expressed the adhesion proteins IG9, ICAM-1, vascular cell adhesion molecule 1 (VCAM) and E-selection.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Blood-Brain Barrier , Central Nervous System/virology , HIV Infections/physiopathology , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/physiology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cells, Cultured , Cytokines/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Gene Expression Regulation/drug effects , Giant Cells , HIV-1/physiology , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Monocytes/physiology , Monocytes/virology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1
15.
Brain Res ; 625(2): 238-43, 1993 Oct 22.
Article in English | MEDLINE | ID: mdl-7903899

ABSTRACT

The blood-brain barrier (BBB) is involved in many normal regulatory mechanisms as well as in pathologic conditions of the central nervous system. Previous studies examining the development and function of the BBB in vitro have primarily utilized cell lines or cultured tissues from non-human sources. In contrast, this study used a coculture system of human fetal astrocytes and autologous endothelial cells. Astrocytes and endothelial cells (EC) were isolated and cultured on the opposite sides of a synthetic permeable membrane. The cocultures were characterized by electron and light microscopy for morphology and by immunocytochemistry for cell-type specific markers. Using these coculture conditions, astrocytes displayed characteristic morphology and expressed glial fibrillary acidic protein. When cocultured with astrocytes, endothelial cells retained factor VIII expression and expressed the BBB-specific proteins, brain-type glucose transporter (GLUT-1) and gamma-glutamyl transpeptidase. This expression was dependent on EC being in close apposition to or in direct contact with astrocytes. The model presented in this study may permit further examination of the role of the BBB in both normal human neurodevelopment and neuropathologic conditions.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/physiology , Endothelium, Vascular/metabolism , Fetal Proteins/biosynthesis , Astrocytes/ultrastructure , Biomarkers/chemistry , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Monosaccharide Transport Proteins/biosynthesis , gamma-Glutamyltransferase/biosynthesis
16.
J Exp Med ; 176(6): 1631-6, 1992 Dec 01.
Article in English | MEDLINE | ID: mdl-1281214

ABSTRACT

Leukocyte adhesion molecules on endothelial cells of the blood-brain barrier may participate in the entry of leukocytes into the central nervous system. Because astrocytes are also a component of the blood-brain barrier and have been associated with inflammation, we studied the ability of astrocytes to express leukocyte adhesion molecules using Northern blot and immunocytochemical techniques. Astrocytes treated with the proinflammatory cytokine tumor necrosis factor alpha (TNF) expressed messenger RNA for the adhesion molecules E-selectin, vascular cell adhesion molecule 1, and intercellular adhesion molecule 1, as well as their corresponding proteins. In addition, TNF-treated astrocytes expressed a monocyte adhesion protein identified by our laboratory, recognized by the monoclonal antibody IG9. These results indicate that under inflammatory conditions in the central nervous system, such as multiple sclerosis and acquired immune deficiency syndrome, astrocyte expression of adhesion molecules may facilitate the migration of leukocytes and contribute to the disease process.


Subject(s)
Astrocytes/physiology , Cell Adhesion Molecules/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Blotting, Northern , Cell Adhesion Molecules/genetics , Cells, Cultured , Central Nervous System/physiology , E-Selectin , Female , Fetus , Gene Expression/drug effects , Gestational Age , Glial Fibrillary Acidic Protein/analysis , Humans , Immunohistochemistry , Intercellular Adhesion Molecule-1 , Kinetics , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular Cell Adhesion Molecule-1
17.
Hum Immunol ; 32(4): 241-5, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1686025

ABSTRACT

Lymphocytes propagated from allografts have shown a wide spectrum of activity during rejection including cytotoxicity, proliferation, and lymphokine production. It is necessary to correlate these activities to the rejection process to understand the in vivo immune response. The frequent need to obtain a biopsy of human cardiac allografts permits the evaluation of the function of the graft-infiltrating lymphocytes (GIL) as related to development of the rejection process. Lymphocyte cultures established from biopsies taken before, during, and after rejection episodes of grade 1.0 or greater were assayed for surface antigen expression using flow cytometry, proliferative activity using a primed lymphocyte test (PLT), and cytotoxicity using a cell-mediated lympholysis assay. Fifteen rejection episodes were followed from 10 patients. Two patients were followed through two different rejection episodes and one patient through four rejection episodes. CD8+ cells usually predominated during the rejection episode. Following the rejection episodes the GIL showed a shift toward higher proportion of CD4+ cells. Most cultures taken prior to and during rejection episodes (8/9 and 12/13 assayed, respectively) demonstrated greater than 30% killing of targets bearing donor-related HLA antigens. Seven of 15 cultures remained cytotoxic after a rejection episode whereas 8 of 15 lost cytotoxicity. The patients whose cultures remained cytotoxic after a rejection episode went on to further rejection episodes at 6, 7, 11, 20, 37, or 118 days later. Those patients whose cultures were no longer cytotoxic did not experience any subsequent rejection episode until at least 257 days later.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Graft Rejection/immunology , Heart Transplantation/immunology , T-Lymphocytes, Cytotoxic/immunology , Biopsy , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Humans , Immunophenotyping , Lymphocyte Activation/immunology
18.
Hum Immunol ; 28(2): 208-15, 1990 Jun.
Article in English | MEDLINE | ID: mdl-2141012

ABSTRACT

Graft-infiltrating lymphocytes from both human renal and cardiac allografts were propagated in interleukin 2 in order to evaluate rearrangements in the T-cell receptor (TcR) beta-chain genes. Individual biopsies from renal allografts during episodes of cellular rejection were examined as well as multiple biopsies of heart transplant patients from whom endomyocardial samples were taken prior to, during, and after episodes of rejection. TcR beta-chain rearrangements were evaluated in Southern blots using DNA extracted from interleukin 2-propagated cells and digested with restriction endonucleases permitting assessment of rearrangements to both C beta 1 and C beta 2. Rearrangements shared among greater than 5% of the "bulk" culture appear as nongermline bands when hybridized with a C beta probe. Single-cell progeny were generated from limiting dilution, and the rearrangements among the cloned progeny compared to the "bulk" of the cultured progeny of graft-infiltrating lymphocytes. The results indicate that "dominant" rearrangements are a common feature of renal allograft-infiltrating lymphocytes (14 of 15 cases examined). Since the number of cells which can be recovered from a given cardiac biopsy may be limiting, evaluation of clonal dominance from these cultures is more difficult to evaluate. However, sharing of "dominant" rearrangements among multiple biopsies from the same cardiac allograft patient indicates an in vivo selection for T cells with the same receptor rearrangement. Analysis of individual clones showed 3/33 clones from a renal allograft sharing the "dominant" rearrangement noted in the bulk culture, but none of these "dominant" clones showed antidonor specificity.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Gene Rearrangement, T-Lymphocyte/immunology , Heart Transplantation/immunology , Kidney Transplantation/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Biopsy , Clone Cells , Graft Rejection/immunology , Humans , Receptors, Antigen, T-Cell, alpha-beta
SELECTION OF CITATIONS
SEARCH DETAIL