Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
1.
Mucosal Immunol ; 10(6): 1468-1479, 2017 11.
Article in English | MEDLINE | ID: mdl-28422186

ABSTRACT

The development of obesity-associated insulin resistance is associated with B-lymphocyte accumulation in visceral adipose tissue (VAT) and is prevented by B-cell ablation. To characterize potentially pathogenic B-cell repertoires in this disorder, we performed high-throughput immunoglobulin (Ig) sequencing from multiple tissues of mice fed high-fat diet (HFD) and regular diet (RD). HFD significantly changed the biochemical properties of Ig heavy-chain complementarity-determining region-3 (CDRH3) sequences, selecting for IgA antibodies with shorter and more hydrophobic CDRH3 in multiple tissues. A set of convergent antibodies of highly similar sequences found in the VAT of HFD mice but not RD mice showed significant somatic mutation, suggesting a response shared between mice to a common antigen or antigens. These findings indicate that a simple high-fat dietary intervention has a major impact on mouse B-cell repertoires, particularly in adipose tissues.


Subject(s)
B-Lymphocytes/immunology , Complementarity Determining Regions/genetics , Immunoglobulin A/genetics , Inflammation/immunology , Intra-Abdominal Fat/metabolism , Obesity/immunology , Receptors, Antigen, B-Cell/genetics , Animals , Cell Movement , Cells, Cultured , Diet, High-Fat , High-Throughput Nucleotide Sequencing , Immunoglobulin A/metabolism , Insulin Resistance , Intra-Abdominal Fat/immunology , Male , Mice , Mice, Inbred C57BL , Somatic Hypermutation, Immunoglobulin , Transcriptome
2.
Am J Transplant ; 15(3): 695-704, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25693475

ABSTRACT

Thirty-eight HLA matched and mismatched patients given combined living donor kidney and enriched CD34(+) hematopoietic cell transplants were enrolled in tolerance protocols using posttransplant conditioning with total lymphoid irradiation and anti-thymocyte globulin. Persistent chimerism for at least 6 months was associated with successful complete withdrawal of immunosuppressive drugs in 16 of 22 matched patients without rejection episodes or kidney disease recurrence with up to 5 years follow up thereafter. One patient is in the midst of withdrawal and five are on maintenance drugs. Persistent mixed chimerism was achieved in some haplotype matched patients for at least 12 months by increasing the dose of T cells and CD34(+) cells infused as compared to matched recipients in a dose escalation study. Success of drug withdrawal in chimeric mismatched patients remains to be determined. None of the 38 patients had kidney graft loss or graft versus host disease with up to 14 years of observation. In conclusion, complete immunosuppressive drug withdrawal could be achieved thus far with the tolerance induction regimen in HLA matched patients with uniform long-term graft survival in all patients.


Subject(s)
Chimerism , Graft Survival , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/administration & dosage , Kidney Transplantation , Living Donors , Adult , Cohort Studies , Female , Humans , Male , Middle Aged , Young Adult
3.
Am J Transplant ; 14(11): 2467-77, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25311657

ABSTRACT

The goal of the study was to elucidate the cellular and molecular mechanisms by which a clinically applicable immune tolerance regimen of combined bone marrow and heart transplants in mice results in mixed chimerism and graft acceptance. The conditioning regimen of lymphoid irradiation and anti-T cell antibodies changed the balance of cells in the lymphoid tissues to create a tolerogenic microenvironment favoring the increase of natural killer T (NKT) cells, CD4+ CD25+ regulatory T cells and Gr-1+ CD11b+ myeloid-derived suppressor cells (MDSCs), over conventional T cells (Tcons). The depletion of MDSCs abrogated chimerism and tolerance, and add back of these purified cells was restorative. The conditioning regimen activated the MDSCs as judged by the increased expression of arginase-1, IL-4Rα and programmed death ligand 1, and the activated cells gained the capacity to suppress the proliferation of Tcons to alloantigens in the mixed leukocyte reaction. MDSC activation was dependent on the presence of host invariant NKT cells. The conditioning regimen polarized the host invariant NKT cells toward IL-4 secretion, and MDSC activation was dependent on IL-4. In conclusion, there was a requirement for MDSCs for chimerism and tolerance, and their suppressive function was dependent on their interactions with NKT cells and IL-4.


Subject(s)
Bone Marrow Transplantation , Heart Transplantation , Immune Tolerance , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
Am J Transplant ; 12(5): 1133-45, 2012 May.
Article in English | MEDLINE | ID: mdl-22405058

ABSTRACT

Sixteen patients conditioned with total lymphoid irradiation (TLI) and antithymocyte globulin (ATG) were given kidney transplants and an injection of CD34+ hematopoietic progenitor cells and T cells from HLA-matched donors in a tolerance induction protocol. Blood cell monitoring included changes in chimerism, balance of T-cell subsets and responses to donor alloantigens. Fifteen patients developed multilineage chimerism without graft-versus-host disease (GVHD), and eight with chimerism for at least 6 months were withdrawn from antirejection medications for 1-3 years (mean, 28 months) without subsequent rejection episodes. Four chimeric patients have just completed or are in the midst of drug withdrawal, and four patients were not withdrawn due to return of underlying disease or rejection episodes. Blood cells from all patients showed early high ratios of CD4+CD25+ regulatory T cells and NKT cells versus conventional naive CD4+ T cells, and those off drugs showed specific unresponsiveness to donor alloantigens. In conclusion, TLI and ATG promoted the development of persistent chimerism and tolerance in a cohort of patients given kidney transplants and hematopoietic donor cell infusions. All 16 patients had excellent graft function at the last observation point with or without maintenance drugs.


Subject(s)
Graft Survival/immunology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/therapeutic use , Kidney Transplantation , Transplantation Immunology , Adult , Antilymphocyte Serum/therapeutic use , Blood Group Incompatibility , Female , Fluorescent Antibody Technique , Graft vs Host Disease/immunology , Humans , Immune Tolerance , Lymphatic Irradiation , Male , Middle Aged , Transplantation Chimera , Treatment Outcome , Young Adult
5.
Int J Obes Suppl ; 2(Suppl 1): S4-S7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-25089193

ABSTRACT

Obesity is associated with chronic inflammation of various tissues including visceral adipose tissue (VAT), which contributes to insulin resistance. T cells and macrophages infiltrate VAT in obesity and orchestrate this inflammation. Recently, we made the surprising discovery that B cells are important contributors to this process. Thus, some B cells and the antibodies they produce can promote VAT-associated and systemic inflammation, leading to insulin resistance. This report will focus on the properties of these B cells, and how they contribute to insulin resistance through T-cell modulation and production of pathogenic autoantibodies. Understanding the mechanisms by which B cells contribute to insulin resistance should lead to new antibody-based diagnostics and B-cell modulating therapeutics to manage this increasingly prevalent disease.

7.
J Immunol ; 167(12): 7150-6, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739538

ABSTRACT

Many tumor-associated Ags represent tissue differentiation Ags that are poorly immunogenic. Their weak immunogenicity may be due to immune tolerance to self-Ags. Prostatic acid phosphatase (PAP) is just such an Ag that is expressed by both normal and malignant prostate tissue. We have previously demonstrated that PAP can be immunogenic in a rodent model. However, generation of prostate-specific autoimmunity was seen only when a xenogeneic homolog of PAP was used as the immunogen. To explore the potential role of xenoantigen immunization in cancer patients, we performed a phase I clinical trial using dendritic cells pulsed with recombinant mouse PAP as a tumor vaccine. Twenty-one patients with metastatic prostate cancer received two monthly vaccinations of xenoantigen-loaded dendritic cells with minimal treatment-associated side effects. All patients developed T cell immunity to mouse PAP following immunization. Eleven of the 21 patients also developed T cell proliferative responses to the homologous self-Ag. These responses were associated with Ag-specific IFN-gamma and/or TNF-alpha secretion, but not IL-4, consistent with induction of Th1 immunity. Finally, 6 of 21 patients had clinical stabilization of their previously progressing prostate cancer. All six of these patients developed T cell immunity to human PAP following vaccination. These results demonstrate that xenoantigen immunization can break tolerance to a self-Ag in humans, resulting in a clinically significant antitumor effect.


Subject(s)
Antigens, Heterophile/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/transplantation , Immunotherapy, Adoptive , Prostatic Neoplasms/therapy , Acid Phosphatase , Animals , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Humans , Kinetics , Male , Mice , Middle Aged , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/immunology , Protein Tyrosine Phosphatases/immunology , Th1 Cells/immunology , Treatment Outcome
8.
Biol Blood Marrow Transplant ; 7(9): 517-22, 2001.
Article in English | MEDLINE | ID: mdl-11669219

ABSTRACT

Vaccination with the idiotype (Id) protein derived from B-cell malignancies can produce Id-specific immune responses that correlate with improved remission duration and survival rates in patients with follicular non-Hodgkin's lymphoma (NHL). A state of minimal or no residual disease correlates strongly with the laboratory detection of a cellular or humoral immune response. High-dose cytotoxic therapy (HDCT) with autologous stem cell support (autologous bone marrow transplantation [ABMT]) can provide profound cytoreduction of B-cell NHL, but the potential immune suppression associated with myeloablative therapy may compromise a patient's ability to mount a specific immune response. To determine whether patients with NHL could mount detectable immuneresponses following ABMT, Id vaccines were administered at 2 to 12 months following myeloablative therapy to a series of patients with relapsed or resistant B-cell NHL. Two different vaccination strategies produced robust immune responses against KLH in all patients, supporting the capacity of the reconstituted immune system following HDCT to react against a strong antigen. Combining the results from both vaccination strategies, 10 of 12 patients mounted Id-specific humoral or cellular responses. Vaccinations were consistently well tolerated. Of the 12 patients, 7 have experienced prolonged remissions with a follow-up from HDCT ranging from 3 to more than 11 years. Our experience serves to document the ability of the recovering immune system to react against both self and xenotypic antigens and supports the feasibility and safety of antigen-specific vaccination following myeloablative therapy in patients with B-cell NHL.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/analogs & derivatives , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Neoplasm/immunology , Bone Marrow Transplantation/immunology , Immunoglobulin Idiotypes/immunology , Lymphoma, B-Cell/therapy , Squalene/analogs & derivatives , Vaccination , Acetylmuramyl-Alanyl-Isoglutamine/administration & dosage , Adjuvants, Immunologic , Antibodies, Anti-Idiotypic/immunology , Antibodies, Neoplasm/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carmustine/administration & dosage , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Dendritic Cells/immunology , Dendritic Cells/transplantation , Disease-Free Survival , Drug Resistance, Neoplasm , Etoposide/administration & dosage , Feasibility Studies , Female , Follow-Up Studies , Hemocyanins/administration & dosage , Humans , Ifosfamide/administration & dosage , Immunity, Cellular , Immunoglobulin Idiotypes/administration & dosage , Lymphocyte Activation , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/immunology , Male , Polysorbates/administration & dosage , Receptors, Antigen, B-Cell/immunology , Safety , Squalene/administration & dosage , Transplantation Conditioning , Transplantation, Autologous , Treatment Outcome , Whole-Body Irradiation
9.
Ann N Y Acad Sci ; 938: 167-73; discussion 173-4, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11458504

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells which both initiate adaptive immune responses and control tolerance to self-antigens. It has been suggested that these different effects on responder cells depend on subsets of DCs arising from either myeloid or lymphoid hematopoietic origins. In this model, CD8 alpha+ Mac-1- DCs are supposed to be of lymphoid while CD8 alpha- Mac-1+ DCs are supposed to be of myeloid origin. Here we summarize our findings that both CD8 alpha+ and CD8 alpha- DCs can arise from clonogenic common myeloid progenitors (CMPs) in both thymus and spleen. Therefore CD8 alpha expression DCs does not indicate a lymphoid origin and differences among CD8 alpha+ and CD8 alpha- DCs might rather reflect maturation status than ontogeny. On the basis of transplantation studies, it seems likely that most of the DCs in secondary lymphoid organs and a substantial fraction of thymic DCs are myeloid-derived.


Subject(s)
Dendritic Cells/cytology , Hematopoietic Stem Cells/cytology , Myeloid Cells/cytology , Animals , Antigens, Differentiation/analysis , Bone Marrow Cells/cytology , CD8 Antigens/analysis , Cell Differentiation , Cell Lineage , Clone Cells/cytology , Dendritic Cells/classification , Hematopoietic Stem Cell Transplantation , Macrophage-1 Antigen/analysis , Mice , Radiation Chimera , Thymus Gland/cytology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology
10.
Proc Natl Acad Sci U S A ; 98(15): 8809-14, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11427731

ABSTRACT

Most tumor-associated antigens represent self-proteins and as a result are poorly immunogenic due to immune tolerance. Here we show that tolerance to carcinoembryonic antigen (CEA), which is overexpressed by the majority of lethal malignancies, can be reversed by immunization with a CEA-derived peptide. This peptide was altered to make it a more potent T cell antigen and loaded onto dendritic cells (DCs) for delivery as a cellular vaccine. Although DCs are rare in the blood, we found that treatment of advanced cancer patients with Flt3 ligand, a hematopoietic growth factor, expanded DCs 20-fold in vivo. Immunization with these antigen-loaded DCs induced CD8 cytotoxic T lymphocytes that recognized tumor cells expressing endogenous CEA. Staining with peptide-MHC tetramers demonstrated the expansion of CD8 T cells that recognize both the native and altered epitopes and possess an effector cytotoxic T lymphocyte phenotype (CD45RA(+)CD27(-)CCR7(-)). After vaccination, two of 12 patients experienced dramatic tumor regression, one patient had a mixed response, and two had stable disease. Clinical response correlated with the expansion of CD8 tetramer(+) T cells, confirming the role of CD8 T cells in this treatment strategy.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Colonic Neoplasms/immunology , Dendritic Cells/immunology , Lung Neoplasms/immunology , Membrane Proteins/immunology , Vaccines, Synthetic/immunology , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/physiopathology , Carcinoma, Non-Small-Cell Lung/therapy , Colonic Neoplasms/physiopathology , Colonic Neoplasms/therapy , Female , HLA-A2 Antigen/immunology , Humans , Immunotherapy , Ligands , Lung Neoplasms/physiopathology , Lung Neoplasms/therapy , Male , Middle Aged , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination
11.
Mol Ther ; 3(2): 186-96, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11237675

ABSTRACT

Angiostatin and endostatin are potent endothelial cell growth inhibitors that have been shown to inhibit angiogenesis in vivo and tumor growth in mice. However, tumor shrinkage requires chronic delivery of large doses of these proteins. Here we report synergistic antitumor activity and survival of animals when these factors are delivered in combination to tumors by retroviral gene transfer. We have demonstrated this efficacy in both murine leukemia and melanoma models. Complete loss of tumorigenicity was seen in 40% of the animals receiving tumors transduced by the combination of angiostatin and endostatin in the leukemia model. The synergy was also demonstrated in vitro on human umbilical vein endothelial cell differentiation and this antiangiogenic activity may suggest a mechanism for the antitumor activity in vivo. These findings imply separate pathways by which angiostatin and endostatin mediate their antiangiogenic effects. Together, these data suggest that a combination of antiangiogenic factors delivered by retroviral gene transfer may produce synergistic antitumor effects in both leukemia and solid tumors, thus avoiding long-term administration of recombinant proteins. The data also suggest that novel combinations of antiangiogenic factors delivered into tumors require further investigation as therapeutic modalities.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Collagen/therapeutic use , Gene Transfer Techniques , Genetic Therapy/methods , Leukemia/therapy , Melanoma/therapy , Neovascularization, Pathologic , Peptide Fragments/therapeutic use , Plasminogen/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Angiostatins , Animals , Antineoplastic Agents/administration & dosage , Blotting, Western , Cell Division/drug effects , Cell Separation , Cell Survival , Collagen/administration & dosage , Collagen/metabolism , Down-Regulation , Drug Combinations , Endostatins , Female , Flow Cytometry , Genetic Vectors , Green Fluorescent Proteins , Humans , Immunohistochemistry , Laminin/metabolism , Luminescent Proteins/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Genetic , Moloney murine leukemia virus/genetics , Peptide Fragments/administration & dosage , Plasminogen/administration & dosage , Precipitin Tests , Proteoglycans/metabolism , Retroviridae/genetics , Time Factors , Transduction, Genetic , Transfection
12.
J Immunol ; 166(6): 4254-9, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238679

ABSTRACT

Dendritic cells (DC) represent potent APCs that are capable of generating tumor-specific immunity. We performed a pilot clinical trial using Ag-pulsed DC as a tumor vaccine. Twenty-one patients with metastatic prostate cancer received two monthly injections of DC enriched and activated from their PBMC. DC were cocultured ex vivo with recombinant mouse prostatic acid phosphatase as the target neoantigen. Following enrichment, DC developed an activated phenotype with up-regulation of CD80, CD86, and CD83 expression. During culture, the DC maintained their levels of various adhesion molecules, including CD44, LFA-1, cutaneous lymphocyte-associated Ag, and CD49d, up-regulated CCR7, but lost CD62 ligand and CCR5. In the absence of CD62 ligand, such cells would not be expected to prime T cells efficiently if administered i.v. due to their inability to access lymphoid tissue via high endothelial venules. To assess this possibility, three patient cohorts were immunized with Ag-pulsed DC by i.v., intradermal (i.d.), or intralymphatic (i.l.) injection. All patients developed Ag-specific T cell immune responses following immunization, regardless of route. Induction of IFN-gamma production, however, was seen only with i.d. and i.l. routes of administration, and no IL-4 responses were seen regardless of route, consistent with the induction of Th1-type immunity. Five of nine patients who were immunized by the i.v. route developed Ag-specific Abs compared with one of six for i.d. and two of six for i.l. routes. These results suggest that while activated DC can prime T cell immunity regardless of route, the quality of this response and induction of Ag-specific Abs may be affected by the route of administration.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Dendritic Cells/transplantation , Prostatic Neoplasms/immunology , Acid Phosphatase/immunology , Antibody Specificity , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Cell Adhesion Molecules/biosynthesis , Cells, Cultured , Cytokines/biosynthesis , Flow Cytometry , Humans , Injections, Intradermal , Injections, Intralymphatic , Injections, Intravenous , Lymphocyte Activation , Male , Pilot Projects , Prostate/enzymology , Prostate/immunology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/therapy , Receptors, Chemokine/biosynthesis , Receptors, Lymphocyte Homing/biosynthesis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Angiogenesis ; 4(4): 263-8, 2001.
Article in English | MEDLINE | ID: mdl-12197471

ABSTRACT

The combination of angiostatin and endostatin has been shown to have synergistic antiangiogenic and antitumor effects when the genes for these proteins are delivered to tumor cells by retroviral gene transfer. Here we report the construction of a murine angiostatin-endostatin fusion gene (Statin-AE) which shows enhanced antiangiogenic activity on human umbilical vein endothelial cell (HUVEC) tube formation in vitro compared with angiostatin or endostatin alone. Similarly, the fusion gene demonstrates antiangiogenic effects in vivo and antitumor activity in a B16F10 melanoma model when co-delivered by retroviral packaging cell inoculation in mice. The fusion gene demonstrates significantly greater inhibition of tumor growth compared with angiostatin, endostatin or the combination of genes.


Subject(s)
Collagen/physiology , Neovascularization, Pathologic , Peptide Fragments/physiology , Plasminogen/physiology , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Angiostatins , Animals , Cells, Cultured , Collagen/chemistry , Endostatins , Humans , Melanoma, Experimental/blood supply , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neovascularization, Pathologic/genetics , Peptide Fragments/chemistry , Plasminogen/chemistry , Recombinant Fusion Proteins/chemistry
14.
Biol Blood Marrow Transplant ; 6(6): 621-7, 2000.
Article in English | MEDLINE | ID: mdl-11128812

ABSTRACT

The idiotype (Id) determinants on the multiple myeloma immunoglobulin can serve as tumor-specific antigens. An anti-Id immune response may stem the growth of the malignant clone. We report on 26 patients treated at our institution with high-dose chemotherapy and peripheral blood progenitor cell transplantation (PBPCT) and vaccinated with the Id protein. The patients received chemotherapy and PBPCT to establish a minimal residual disease state. After high-dose therapy, the patients received a series of monthly immunizations consisting of 2 intravenous infusions of dendritic cells (DCs) pulsed with either Id protein or Id coupled with keyhole limpet hemocyanin (KLH) as an immunogenic carrier protein, followed by subcutaneous boosts of Id-KLH conjugates. DCs were obtained in all patients from a leukapheresis product 3 to 9 months after PBPCT. Patients were observed for toxicity, immune responses, and tumor status. The DC infusions and the administration of Id-KLH boosts were well tolerated, with patients experiencing only minor and transient side effects. Of the patients, 24 of 26 generated a KLH-specific cellular proliferative immune response. Only 4 patients developed an Id-specific proliferative immune response. Three of these immune responders were in complete remission at the time of vaccination. A total of 17 patients are alive at a median follow-up of 30 months after transplantation. Id vaccination with autologous DCs is feasible for myeloma patients after transplantation. Id-specific cellular responses can be induced in patients who are in complete remission. Further studies are needed to increase the rate of anti-Id immune responses in patients who do not achieve complete remission.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Dendritic Cells/transplantation , Hematopoietic Stem Cell Transplantation , Multiple Myeloma/therapy , Adult , Combined Modality Therapy , Dendritic Cells/immunology , Female , Humans , Immunoglobulin Idiotypes/administration & dosage , Immunoglobulin Idiotypes/immunology , Immunotherapy , Male , Middle Aged , Multiple Myeloma/immunology , Transplantation, Autologous , Vaccination
15.
Science ; 290(5499): 2152-4, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11118150

ABSTRACT

Dendritic cells (DCs) are critical in both initiating adaptive immune responses and maintaining tolerance to self antigens. These apparently contradictory roles have been suggested to depend on different subsets of DCs that arise from either myeloid or lymphoid hematopoietic origins, respectively. Although DC expression of CD8alpha is attributed to a lymphoid origin, here we show that both CD8alpha+ and CD8alpha- DCs can arise from clonogenic common myeloid progenitors in both thymus and spleen. Thus, expression of CD8alpha is not indicative of a lymphoid origin, and phenotypic and functional differences among DC subsets are likely to reflect maturation status rather than ontogeny.


Subject(s)
CD8 Antigens/analysis , Dendritic Cells/cytology , Dendritic Cells/immunology , Myeloid Progenitor Cells/cytology , Spleen/cytology , Thymus Gland/cytology , Animals , Antigens, CD/analysis , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Lineage , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Immunophenotyping , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/transplantation , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/immunology
16.
Blood ; 96(5): 1865-72, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10961888

ABSTRACT

Bone marrow-derived dendritic cells (DC) represent a family of antigen-presenting cells (APC) with varying phenotypes. For example, in mice, CD8alpha(+) and CD8alpha(-) DC are thought to represent cells of lymphoid and myeloid origin, respectively. Langerhans cells (LC) of the epidermis are typical myeloid DC; they do not express CD8alpha, but they do express high levels of myeloid antigens such as CD11b and FcgammaR. By contrast, thymic DC, which derive from a lymphoid-related progenitor, express CD8alpha but only low levels of myeloid antigens. CD8alpha(+) DC are also found in the spleen and lymph nodes (LN), but the origin of these cells has not been determined. By activating and labeling CD8alpha(-) epidermal LC in vivo, it was found that these cells expressed CD8alpha on migration to the draining LN. Similarly, CD8alpha(-) LC generated in vitro from a CD8 wild-type mouse and injected into the skin of a CD8alphaKO mouse expressed CD8alpha when they reached the draining LN. The results also show that CD8alpha(+) LC are potent APC. After migration from skin, they localized in the T-cell areas of LN, secreted high levels of interleukin-12, interferon-gamma, and chemokine-attracting T cells, and they induced antigen-specific T-cell activation. These results demonstrate that myeloid DC in the periphery can express CD8alpha when they migrate to the draining LN. CD8alpha expression on these DC appears to reflect a state of activation, mobilization, or both, rather than lineage. (Blood. 2000;96:1865-1872)


Subject(s)
CD8 Antigens/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , CD3 Complex/genetics , CD8 Antigens/genetics , Cell Movement , Chemokines/metabolism , Cytokines/metabolism , Dendritic Cells/cytology , Female , Gene Expression , Langerhans Cells/immunology , Langerhans Cells/metabolism , Langerhans Cells/transplantation , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR6 , Receptors, CCR7 , Receptors, Chemokine/genetics , Skin/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Th1 Cells/immunology
17.
Transplantation ; 69(8): 1549-54, 2000 Apr 27.
Article in English | MEDLINE | ID: mdl-10836360

ABSTRACT

BACKGROUND: Previous studies showed the feasibility of inducing transplantation tolerance to cadaveric renal allografts in patients given pretransplant total lymphoid irradiation (TLI). Microchimerism has been theorized to be an important or necessary factor in long-term graft acceptance and tolerance in humans. METHODS: A cadaveric renal transplant recipient given pretransplant total lymphoid irradiation and withdrawn from immunosuppressive drugs more than 12 years ago was tested for microchimerism using a sensitive nested polymerase chain reaction technique, and for anti-donor reactivity using the mixed leukocyte reaction and an ELISA screen for anti-HLA antibodies. Donor and recipient were mismatched for all HLA-A, B, and DR antigens. RESULTS: The "tolerant" recipient had good graft function, no detectable donor-type cells in the blood by polymerase chain reaction analysis, vigorous reactivity to donor stimulator cells in the mixed leukocyte reaction, and no detectable serum anti-HLA antibodies. CONCLUSIONS: Operational tolerance to HLA-A, B, and DR mismatched organ allografts can be induced prospectively in humans for at least 12 years after withdrawal of immunosuppressive drugs. The allograft can be maintained in the absence of detectable donor microchimerism and in the presence of anti-donor reactivity in the mixed leukocyte reaction, suggesting that neither chimerism nor clonal deletion or anergy of recipient T cells to alloantigens presented by donor Class II HLA molecules is required for persistence of the tolerant state using this total lymphoid irradiation protocol.


Subject(s)
Immune Tolerance , Immunosuppressive Agents/administration & dosage , Kidney Transplantation/immunology , Alleles , Chimera/immunology , Enzyme-Linked Immunosorbent Assay , HLA-DR Antigens/analysis , HLA-DR Antigens/genetics , HLA-DRB1 Chains , Humans , Immune Tolerance/physiology , Immunosuppressive Agents/therapeutic use , Kidney/immunology , Lymphocyte Culture Test, Mixed , Male , Middle Aged , Polymerase Chain Reaction , Prospective Studies , Tissue Donors
18.
Annu Rev Immunol ; 18: 245-73, 2000.
Article in English | MEDLINE | ID: mdl-10837059

ABSTRACT

The potential to harness the potency and specificity of the immune system underlies the growing interest in cancer immunotherapy. One such approach uses bone marrow-derived dendritic cells, phenotypically distinct and extremely potent antigen-presenting cells, to present tumor-associated antigens and thereby generate tumor-specific immunity. Support for this strategy comes from animal studies that have demonstrated that dendritic cells, when loaded ex vivo with tumor antigens and administered to tumor-bearing hosts, can elicit T cell-mediated tumor destruction. These observations have led to clinical trials designed to investigate the immunologic and clinical effects of antigen-loaded dendritic cells administered as a therapeutic vaccine to patients with cancer. In the design and conduct of such trials, important considerations include antigen selection, methods for introducing the antigen into MHC class I and II processing pathways, methods for isolating and activating dendritic cells, and route of administration. Although current dendritic cell-based vaccination methods are cumbersome, promising results from clinical trials in patients with malignant lymphoma, melanoma, and prostate cancer suggest that immunotherapeutic strategies that take advantage of the antigen presenting properties of dendritic cells may ultimately prove both efficacious and widely applicable to human tumors.


Subject(s)
Dendritic Cells/immunology , Immunotherapy , Neoplasms/therapy , Animals , Humans
19.
Pediatr Res ; 46(6): 755-9, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10590035

ABSTRACT

Neonates are unusually susceptible to herpes simplex virus infection, which may be explained in part by defects in killing of herpes simplex virus-infected cells by natural killer (NK) cell cytotoxicity and antibody-dependent cellular cytotoxicity. The mechanism for these defects remains poorly defined. We have for the first time used immunomagnetically enriched NK cells to explore neonatal NK cell phenotype and target cell adhesion. CD56-positive neonatal NK cells had markedly lower CD57 expression, but adult level expression of adhesive glycoproteins (CD18, CD44) and Fc receptor for IgG (CD16). Although the cells conjugated normally with target cells in the absence of antibody, antibody-mediated conjugation was significantly lower than that of NK cells from adults (p < 0.002). These results demonstrate intact adhesion in neonatal NK cell cytotoxicity. In contrast, defective neonatal antibody-dependent cellular cytotoxicity is caused, in part, by an adhesion defect in the presence of antibody.


Subject(s)
Antibodies/immunology , Fetal Blood , Herpes Simplex/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Simplexvirus , Adult , CD56 Antigen/immunology , Cell Adhesion/immunology , Cytotoxicity, Immunologic , Herpes Simplex/pathology , Humans , Immunoglobulin G/immunology , Killer Cells, Natural/virology , Receptors, Fc/immunology
20.
Clin Immunol ; 92(1): 67-75, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10413654

ABSTRACT

Recombinant adeno-associated virus (rAAV) is a replication-defective parvovirus which is being explored as a vector for gene therapy because of its broad host range, excellent safety profile, and durable transgene expression in infected hosts. rAAV has also been reported by several groups to induce little or no immune response to its encoded transgene products. In this study we examined the immunogenicity of rAAV by studying the immune response of C57BL/6 mice to a single dose of rAAV-encoding ovalbumin (AAV-Ova) administered by a variety of routes. Mice injected with AAV-Ova intraperitoneally (ip), intravenously, or subcutaneously developed potent ovalbumin-specific cytotoxic T lymphocytes (CTL) as well as anti-ovalbumin antibodies and antibodies to AAV. In contrast, mice injected with AAV-Ova intramuscularly developed a humoral response to the virus and the transgene but minimal ovalbumin-specific CTLs. The induced CTL response after ip administration of AAV-Ova protected mice against a subsequent tumor challenge with an ovalbumin-transfected B16 melanoma cell line. Studies of the mechanism by which AAV-Ova induces CTL confirmed that the virus delivers the transgene product into the classical MHC class I pathway of antigen processing. Mice that previously had been exposed to rAAV vectors failed to develop ovalbumin-specific CTL following administration of AAV-Ova. Analysis of these mice revealed the presence of circulating anti-AAV antibodies that blocked rAAV transduction in vitro and inhibited CTL induction in vivo. These results suggest a possible role for rAAV in the immunotherapy of malignancies and viral infections, although induced antibody responses to AAV may limit its ability to be administered for repeated vaccinations.


Subject(s)
Antigens, Viral/administration & dosage , Dependovirus/immunology , Animals , Antibody Formation , Drug Administration Routes , Female , Immunity, Cellular/immunology , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Recombinant Proteins/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...