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










Publication year range
1.
Gene Ther ; 20(8): 853-60, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23303282

ABSTRACT

Genetic modification of human T cells to express transgene-encoded polypeptides, such as tumor targeting chimeric antigen receptors, is an emerging therapeutic modality showing promise in clinical trials. The development of simple and efficient techniques for purifying transgene(+) T cells is needed to facilitate the derivation of cell products with uniform potency and purity. Unlike selection platforms that utilize physical methods (immunomagnetic or sorting) that are technically cumbersome and limited by the expense and availability of clinical-grade components, we focused on designing a selection system on the basis of the pharmaceutical drug methotrexate (MTX), a potent allosteric inhibitor of human dihydrofolate reductase (DHFR). Here, we describe the development of self inactivating (SIN) lentiviral vectors that direct the coordinated expression of a CD19-specific chimeric antigen receptor (CAR), the human EGFRt tracking/suicide construct, and a methotrexate-resistant human DHFR mutein (huDHFR(FS); L22F, F31S). Our results demonstrate that huDHFR(FS) expression renders lentivirally transduced primary human CD45RO(+)CD62L(+) central memory T cells resistant to lymphotoxic concentrations of MTX up to 0.1 µM. Our modular complementary DNA (cDNA) design insures that selected MTX-resistant T cells co-express functionally relevant levels of the CD19-specific CAR and EGFRt. This selection system on the basis of huDHFR(FS) and MTX has considerable potential utility in the manufacturing of clinical-grade T cell products.


Subject(s)
Cell Engineering , Genetic Vectors , T-Lymphocytes/cytology , Tetrahydrofolate Dehydrogenase/genetics , Antigens, CD19/genetics , Folic Acid Antagonists/pharmacology , Gene Transfer Techniques , Humans , Lentivirus/genetics , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Methotrexate/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
2.
Cytotherapy ; 9(8): 771-84, 2007.
Article in English | MEDLINE | ID: mdl-17917884

ABSTRACT

Background The role of co-stimulation in CD4+ T cell activation by professional APC is well established, while less is known of the role co-stimulation plays when CD4+ T cells interact directly with tumor cells. Methods Through genetic engineering of human CD4+ T cells, we tested the hypothesis that integration of co-stimulatory signaling domains within a tumor-targeting chimeric Ag receptor (CAR), the IL-13Ralpha2-specific IL-13-zetakine (IL13zeta), would enhance CD4+ T cell mediated responses against tumors that fail to express ligands for co-stimulatory receptors. Results Compared with CD3zeta-mediated activation alone, CD4+ effector T cells expressing the IL13-CD28-41BBzeta CAR exhibited augmented/sustained MAPK and AKT activity, up-regulated Th1 cytokine production, and enhanced cytolytic potency against tumor targets. Moreover, upon recursive stimulation with tumor, the IL13-CD28-41BBzeta+ cells retained/recycled their lytic function, whereas IL-13zeta+ CD4+ cells became anergic/exhausted. These in vitro observations correlated with enhanced in vivo control of established orthotopic CNS glioma xenografts in immunodeficient mice mediated by adoptively transferred ex vivo-expanded CD4+ T cells expressing the co-stimulatory CAR. Discussion Together these studies demonstrate the importance of integrating co-stimulation with CD3zeta signaling events to activate fully CD4+ anti-tumor effector cells for sustained function in the tumor microenvironment.


Subject(s)
CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Lymphocyte Activation , Signal Transduction , Animals , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/metabolism , Glioma/immunology , Glioma/therapy , Humans , Immunoglobulin G/immunology , Immunotherapy, Adoptive , Interleukin-13/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/immunology , Neoplasms/therapy , Protein Structure, Tertiary , Recombinant Fusion Proteins/immunology , Transgenes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
3.
Int J Hyperthermia ; 19(5): 520-33, 2003.
Article in English | MEDLINE | ID: mdl-12944167

ABSTRACT

Dendritic cells (DCs) in the skin rapidly take up antigen and migrate out of the skin to draining lymph nodes for antigen presentation. As a result, these cells play an important role in generating specific immune responses against infectious agents that enter the skin and against antigens delivered as vaccines. Previous efforts revealed that fever-like elevations in body temperature enhance antigen-dependent immune responses initiated at the site of the skin and stimulate the migration of epidermal DCs to draining lymph nodes. Collectively, these data have led to the hypothesis that the activation of epidermal DCs is sensitive to physiological thermal stimuli. In this study, ear skin explants derived from BALB/c mice were either maintained at 37 degrees C or incubated at 40 degrees C for the first 6.5 h before being placed at 37 degrees C. This heating protocol altered the density and morphology of the epidermal DCs in a manner suggestive of an increased kinetics of activation-associated DC migration. Flow cytometric analysis of the emigrated cells also indicated that mild heating enhanced the migration kinetics of DCs and increased the DC expression of MHC class II and the activation marker CD86. Importantly, these migrated cells displayed higher stimulatory capacity in a mixed lymphocyte reaction compared to those of controls. Overall, these results suggest that mild thermal stimuli can enhance DC activation and function and that strategic applications of heat could enhance the potency of vaccines consisting of relatively weak antigens, such as cancer vaccines.


Subject(s)
Cell Movement/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Epidermal Cells , Hyperthermia, Induced , Animals , Cytokines/genetics , Epidermis/immunology , Female , Gene Expression , HSP70 Heat-Shock Proteins/genetics , Hot Temperature , Kinetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Organ Culture Techniques , RNA, Messenger/analysis , Skin Temperature/immunology , Vaccination
4.
Int J Hyperthermia ; 18(3): 253-66, 2002.
Article in English | MEDLINE | ID: mdl-12028640

ABSTRACT

Various studies in animal tumour models have revealed the potential of fever-range whole body hyperthermia (FR-WBH) to be used in cancer therapy. To determine the safety of FR-WBH treatment in the clinic, patients with advanced solid tumours were heated in the outpatient setting to 39-39.5 degrees C for 3 or 6h, or 39.5-40 degrees C for 6h using the Heckel-HT 2000 apparatus. These WBH treatments were well tolerated, with no significant adverse events related to cardiac, hepatic, renal or pulmonary systems. In the majority of patients, flow cytometric analysis of peripheral blood leukocyte populations indicated that there were transient decreases in the number of circulating T lymphocytes and a concomitant decrease in the number of L-selectin positive lymphocytes in the peripheral blood. These findings closely mimic the affects seen previously in pre-clinical murine studies in which this same fever-like treatment was shown to inhibit tumour growth. These studies have established the safety of this treatment and will allow for future clinical trials where application of FR-WBH treatment can be combined with other anti-cancer therapies, including immunotherapy and chemotherapy.


Subject(s)
Hyperthermia, Induced/methods , Neoplasms/therapy , Adult , Animals , Female , Humans , Hyperthermia, Induced/adverse effects , Lymphopenia/etiology , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasms, Experimental/therapy , Safety
5.
Int J Hyperthermia ; 18(6): 552-62, 2002.
Article in English | MEDLINE | ID: mdl-12537754

ABSTRACT

When exposed to environmental stress, cell survival is supported by the upregulation of stress proteins such as heat shock proteins (HSPs) or glucose regulated proteins (GRPs), which help prevent protein denaturation. To begin to characterize the ability of a physiologically relevant heat exposure to induce stress protein expression, the cerebellum, cerebrum, colon, heart, kidney, liver, lung, lymph nodes, muscle, serum and thymus were extracted from BALB/c mice at various times after fever-range whole body hyperthermia (FR-WBH, 39.5-40 degrees C for 6 h) treatment. The expression of three stress proteins, HSP70, HSP110 and GRP170, was determined in these tissues and serum and compared to constitutive levels in control tissues and serum using Western analysis. Constitutive expression of GRP170 was not affected by FR-WBH in any tissue. In contrast, FR-WBH did enhance HSP expression: HSP70 in heart, kidney, lung, lymph nodes and thymus; and HSP110 in lung, lymph nodes and thymus. The lymphoid tissues displayed the most consistent upregulation of both HSP70 and HSP110 upon FR-WBH treatment. The apparent sensitivity of immunologically relevant tissues to FR-WBH may relate to the enhanced immune responses that are observed during febrile temperatures.


Subject(s)
Fever/metabolism , Glycoproteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Hyperthermia, Induced , Stress, Physiological/metabolism , Animals , Blotting, Western , Brain/metabolism , Female , HSP110 Heat-Shock Proteins , Kidney/metabolism , Lung/metabolism , Lymph Nodes/metabolism , Mice , Mice, Inbred BALB C , Myocardium/metabolism , Organ Specificity , Thymus Gland/metabolism
6.
J Immunol ; 167(5): 2666-70, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11509609

ABSTRACT

The febrile response is one of the most common features of infection and inflammation. However, temperature is rarely a variable in experimental immunological investigations. To determine whether the thermal microenvironment has any immunoregulatory potential in an Ag-dependent response, we applied a mild fever-range whole body hyperthermia (FR-WBH) protocol to BALB/c mice experiencing the contact hypersensitivity (CHS) reaction. We observed that the timing of this FR-WBH treatment relative to the different phases of the CHS response was crucial to the outcome. FR-WBH treatment before sensitization with a 0.5% FITC solution resulted in a depressed CHS response. This appears to be due to direct effects of FR-WBH on epidermal Langerhans cell trafficking to the draining lymph nodes. In contrast, application of FR-WBH directly after application of the elicitation dose of FITC solution resulted in an enhanced reaction. This result correlates with increased homing of lymphocytes to the site of elicitation. Overall, these data have important implications regarding the role of thermal changes experienced during infection and the clinical use of FR-WBH relative to immunotherapeutic strategies.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Fever/immunology , Langerhans Cells/immunology , Lymphocytes/immunology , Animals , Cell Movement , Dermatitis, Contact , Female , Fever/pathology , Hyperthermia, Induced , Immunotherapy , Infections/immunology , Infections/pathology , Infections/therapy , Inflammation/immunology , Inflammation/pathology , Langerhans Cells/pathology , Lymphocytes/pathology , Mice , Mice, Inbred BALB C , Neoplasms/therapy , Time Factors
7.
Blood ; 97(9): 2727-33, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313264

ABSTRACT

Fever is associated with increased survival during acute infection, although its mechanism of action is largely unknown. This study found evidence of an unexpectedly integrated mechanism by which fever-range temperatures stimulate lymphocyte homing to secondary lymphoid tissues by increasing L-selectin and alpha4beta7 integrin-dependent adhesive interactions between circulating lymphocytes and specialized high endothelial venules (HEV). Exposure of splenic lymphocytes in vivo to fever-like whole-body hyperthermia (WBH; 39.8 +/- 0.2 degrees C for 6 hours) stimulated both L-selectin and alpha4beta7 integrin-dependent adhesion of lymphocytes to HEV under shear conditions in lymph nodes and Peyer patches. The adhesiveness of HEV ligands for L-selectin and alpha4beta7 integrin (ie, peripheral lymph node addressin and mucosal addressin cell adhesion molecule-1) also increased during WBH or febrile responses associated with lipopolysaccharide-induced or turpentine-induced inflammation. Similar increases in HEV adhesion occurred during hyperthermia treatment of lymph node and Peyer patch organ cultures in vitro, indicating that the local lymphoid tissue microenvironment is sufficient for the hyperthermia response. In contrast, WBH did not augment adhesion in squamous endothelium of nonlymphoid tissues. Analysis of homing of alpha4beta7(hi) L-selectin(lo) murine TK1 cells and L-selectin(hi) alpha4beta7 integrin-negative 300.19/L-selectin transfectant cells showed that fever-range temperatures caused a 3- to 4-fold increase in L-selectin and alpha4beta7 integrin-dependent trafficking to secondary lymphoid tissues. Thus, enhanced lymphocyte delivery to HEV by febrile temperatures through bimodal regulation of lymphocyte and endothelial adhesion provides a novel mechanism to promote immune surveillance.


Subject(s)
Cell Movement/immunology , Endothelium, Vascular/immunology , Fever/immunology , Lymphocytes/immunology , Animals , Endothelium, Vascular/pathology , Humans , Lymphocytes/pathology , Mice , Mice, Inbred BALB C
8.
J Leukoc Biol ; 68(6): 815-20, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11129648

ABSTRACT

The thermal component of fever is one of the most poorly understood aspects of inflammation. To evaluate the role of fever-range hyperthermia on acute inflammation, BALB/c and C57BL/6 mice were exposed to mild, long-duration whole-body hyperthermia (WBH), and serum concentrations of tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), IL-1beta, and the acute phase proteins (APPs) alpha1-acid glycoprotein and haptoglobin were analyzed. WBH alone did not affect serum concentrations of these cytokines or APPs when compared with controls. In contrast, when WBH was applied just after intraperitoneal administration of lipopolysaccharide (LPS), serum concentrations of TNF-alpha and IL-6 were greater than or equal to threefold higher in BALB/c mice compared with LPS-treated controls. LPS-induced IL-6 levels were also enhanced in WBH-treated C57BL/6 mice. However, APP levels were prolonged only in WBH-treated BALB/c mice. It is interesting that in vitro hyperthermia treatment of LPS-stimulated peritoneal cells resulted in decreased cytokine production compared with controls. These results suggest that fever-range hyperthermia regulates acute inflammation in a mouse strain-specific manner that is more complex than that observed in vitro.


Subject(s)
Acute-Phase Reaction/physiopathology , Fever/physiopathology , Inflammation/physiopathology , Acute-Phase Proteins/analysis , Animals , Female , Fever/blood , Fever/immunology , Haptoglobins/analysis , Hyperthermia, Induced , Inflammation/blood , Interleukin-1/blood , Interleukin-6/blood , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orosomucoid/analysis , Tumor Necrosis Factor-alpha/analysis
10.
Int J Hyperthermia ; 16(1): 29-43, 2000.
Article in English | MEDLINE | ID: mdl-10669315

ABSTRACT

Two predominant WBH protocols presently being used in clinical trials include a low temperature, long duration (LL) WBH, where core body temperature is raised to 39.5-40 degrees C for 6h or more, and a high temperature, short duration (HS) WBH, where core body temperature is raised to 41.8 degrees C for up to 2h. Here, the effects of LL-WBH and HS-WBH on leukocyte populations in the blood, spleen, lymph node (LN) and peritoneal cavity (PerC) of Balb/c mice were compared using flow cytometry. The total numbers of peripheral blood leukocytes decreased up to 2-fold immediately after LL-WBH, reflecting a decrease of lymphocyte numbers compared to controls. In contrast, the numbers of blood leukocytes are increased 2.7-fold immediately after HS-WBH compared to controls, reflecting an increase in lymphocytes, monocytes and granulocytes. After both LL- and HS-WBH treatment, leukocyte numbers in the spleen are decreased approximately 2-fold, again reflecting decreases in lymphocyte numbers. In the PerC, total numbers of leukocytes are also significantly decreased (2-fold) during LL-WBH but not HS-WBH. Total numbers of leukocytes in the LNs were unaffected by both LL- and HS-WBH. Overall, these data reveal differential effects of the LL- and HS-WBH protocols on leukocyte populations in the blood, spleen, LN and PerC of Balb/c mice.


Subject(s)
Hyperthermia, Induced , Leukocytes/physiology , Animals , Body Temperature , Cell Movement , Leukocytes/pathology , Mice , Organ Specificity
11.
Cell Stress Chaperones ; 5(5): 458-61, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11189452

ABSTRACT

Inflammation of the skin and systemic fever, both of which occur with injury or infection, include a hyperthermic component that many believe constitutes a physiological stress. Such increases in local or systemic body temperature may also have a regulatory effect on immune function. Langerhans cells (LCs), the dendritic cells of the skin, continuously monitor the extracellular matrix of the skin by taking up particles and microbes that they then carry to draining lymph nodes for presentation to T lymphocytes. We hypothesize that the thermal element of inflammation and/or fever may help regulate the activation and migration of LCs out of the epidermis. To test this hypothesis, Balb/ c mice were exposed to a mild (39.8 degrees C +/- 0.2 degrees C), long-duration (6 hours) whole body hyperthermia (WBH) treatment, which mimics the thermal component of fever. The number of LCs and their morphology were analyzed at various time points up to 7 days after the initiation of WBH. The LCs of the ear epidermis were visualized using a fluorescein isothiocyanate-conjugated antibody specific for the major histocompatibility complex (MHC) class II molecule and confocal microscopy. Although MHC class II staining was diffuse on the surface of the LC body and dendritic extensions of both WBH and control samples, the WBH-treated LCs exhibited a more punctate morphology with fewer dendritic processes compared with control LCs. A significant decrease in the number of LCs was also observed 1 to 5 days after WBH treatment. Furthermore, in vitro heating of Balb/c ear skin cultures at 40 degrees C for 6 to 8 hours enhanced the numbers of viable LCs that migrated into the culture wells. These results suggest that WBH treatment stimulates epidermal LCs in the absence of foreign antigen.


Subject(s)
Epidermis/immunology , Fever/immunology , Langerhans Cells/immunology , Animals , Cell Count , Ear , Epidermal Cells , Female , Fever/pathology , Langerhans Cells/cytology , Mice , Mice, Inbred BALB C , Organ Culture Techniques
12.
J Immunol ; 162(6): 3378-87, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10092792

ABSTRACT

Regional inflammation and systemic fever are hallmarks of host immune responses to pathogenic stimuli. Although the thermal element of fever is thought to enhance the activity of immune effector cells, it is unclear what the precise role of increased body temperatures is on the activation state and effector functions of lymphocytes. We report here that mild, fever-like whole body hyperthermia (WBH) treatment of mice results in a distinct increase in the numbers of tissue lymphocytes with polarized spectrin cytoskeletons and uropods, as visualized in situ. WBH also induces a coincident reorganization of protein kinase C (PKC) isozymes and increased PKC activity within T cells. These hyperthermia-induced cellular alterations are nearly identical with the previously described effects of Ag- and mitogen-induced activation on lymphocyte spectrin and PKC. Immunoprecipitation studies combined with dual staining and protein overlay assays confirmed the association of PKC beta and PKC theta with spectrin following its reorganization. The receptor for activated C kinase-1 was also found to associate with the spectrin-based cytoskeleton. Furthermore, all these molecules (spectrin, PKC beta, PKC theta, and receptor for activated C kinase-1) cotranslocate to the uropod. Enhanced intracellular spectrin phosphorylation upon WBH treatment of lymphocytes was also found and could be blocked by the PKC inhibitor bisindolylmaleimide I (GF109203X). These data suggest that the thermal element of fever, as mimicked by these studies, can modulate critical steps in the signal transduction pathways necessary for effective lymphocyte activation and function. Further work is needed to determine the cellular target(s) that transduces the signaling pathway(s) induced by hyperthermia.


Subject(s)
Cell Movement/immunology , Fever/immunology , Hyperthermia, Induced , Spectrin/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism , Animals , Enzyme Activation/immunology , Female , Fever/enzymology , Fever/metabolism , Intracellular Fluid/metabolism , Isoenzymes/metabolism , Mice , Mice, Inbred BALB C , Peptides/analysis , Phosphorylation , Protein Kinase C/metabolism , Protein Kinase C beta , Protein Kinase C-theta , Receptors for Activated C Kinase , Receptors, Cell Surface/analysis , T-Lymphocytes/physiology
14.
Eur J Immunol ; 27(9): 2152-9, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9341753

ABSTRACT

CD43 (leukosialin), a sialylated glycoprotein expressed on the surface of most hematopoietic cells, has been implicated in cell adhesion and signaling. However, its precise physiological function remains unclear. We used mouse CD43 (mCD43)-immunoglobulin enhancer-transgenic (TG) mice to study the role of mCD43 in vivo. Previous work revealed that mCD43 expression on mature B cells in these mice resulted in immunodeficiency to T-dependent (TD) antigens (Ag), possibly by impairing B-T cell interactions. In the present study we have immunized the TG mice with the T-independent (TI) Ag fluorescein-(Fl) lipopolysaccharide (LPS) (TI type 1 Ag) and Fl-Ficoll (TI type 2 Ag). Surprisingly, the mCD43-Ig enhancer expressing mice were impaired in their ability to mount humoral responses to both Fl-LPS and Fl-Ficoll, and had decreased numbers of cells responding to Ag in vivo. Flow cytometric analysis was performed on peritoneal B-1 cells, a population which often plays a major role in humoral responses to TI Ag such as bacterial Ag. This analysis revealed similar B220, IgM and CD5 expression patterns for the TG and nontransgenic (NTG) B-1 cells. In addition, purified peritoneal B-1 cells from TG and NTG mice were able to respond to LPS. Stimulation of splenic B cells in vitro with Fl-LPS and Fl-Ficoll revealed that, in contrast to NTG B cell responses, TG B cell responses could not be enhanced by co-culture with T cells. However, soluble T cell factor enhancement of the TG B cell responses was normal. These data suggest that the mCD43 expression on B cells may inhibit cell interactions that are important for enhanced TI Ag responses. The anti-adhesive forces of mucins in general may thus be critical in regulating both TD and TI humoral responses.


Subject(s)
Antigens, CD , Antigens, T-Independent/immunology , B-Lymphocytes/immunology , Sialoglycoproteins/physiology , Animals , Antibody Formation , Enhancer Elements, Genetic , Ficoll/immunology , Genes, Immunoglobulin , Leukosialin , Lipopolysaccharides/immunology , Lymphocyte Activation , Lymphocyte Cooperation , Mice , Mice, Transgenic , Peritoneal Cavity/cytology
15.
J Immunol ; 157(11): 4876-84, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8943391

ABSTRACT

Leukosialin (CD43 or sialophorin) is a cell surface sialoglycoprotein implicated in cell adhesion and proliferation whose tightly regulated expression in B lymphocytes is likely important for their normal development and/or function. To examine the physiologic role of mouse CD43 (mCD43) in vivo, we exploited transgenic (TG) mice whose developmental expression of mCD43 was extended during B cell differentiation so that mCD43 was now expressed on peripheral B cells. Despite having increased B cells, localization of lymphocytes in the TG spleens appeared normal by immunocytochemistry with anti-CD4, anti-CD8, and anti-B220 mAbs. However, the numbers of splenic germinal centers and the resting sera Ig levels were decreased in the TG mice compared with littermate controls. TG mice had decreased humoral responses to the T-dependent Ags keyhole limpet hemocyanin and OVA, as well as reduced Ag-specific B cell numbers. In contrast, in vitro LPS stimulation of purified TG or control B cells resulted in similar proliferation and IgM responses. Thus, the alteration of B cell mCD43 expression that resulted in profound immunodeficiency in vivo was not due to absolute defects in B cell development or Ab production. However, TG B cells had a decreased ability to homotypically aggregate and to present Ag to the T cell hybridoma B3Z. These data suggest that the immunodeficiency seen in vivo is due to the anti-adhesive forces of mCD43 preventing normal T-B cell interaction. This likely reflects a general property of mucins in regulating cell interactions.


Subject(s)
Antigens, CD/metabolism , B-Lymphocytes/immunology , Immunologic Deficiency Syndromes/etiology , Sialoglycoproteins/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD/genetics , B-Lymphocytes/pathology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Differentiation , Gene Expression , Hemocyanins/immunology , Immunoglobulin M/biosynthesis , Immunoglobulins/genetics , Immunoglobulins/metabolism , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Leukosialin , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Lymphocyte Cooperation/genetics , Lymphocyte Cooperation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Models, Biological , Ovalbumin/immunology , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Spleen/immunology , Spleen/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...