Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Br J Dermatol ; 177(3): 791-800, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28256712

ABSTRACT

BACKGROUND: MicroRNA (miR)-155 contributes to the proliferation of mycosis fungoides (MF) in vitro and is upregulated in tumours of MF compared with early MF lesions. OBJECTIVES: To investigate the contribution of miR-155 to the cancerous phenotype and drug resistance of MF/Sézary cell lines. METHODS: miR-155 was inhibited in MF cell lines (MyLa and MJ) by transduction of miRZip anti-miR-155, and overexpressed in Hut78 cells by transduction of miRVec-miR-155; empty plasmids served as controls. Cells were analysed for response to inducers of apoptosis and cell-cycle arrest, using fluorescence-activated cell sorting. Transduced MyLa cells were subcutaneously injected into severe combined immunodeficient mice, and tumours were analysed immunohistochemically and for final size. RESULT: MyLa and MJ cells expressed a high level of miR-155; Hut78 cells expressed a low level. MF cell lines stably expressing miR-155 inhibitor showed increased G2/M arrest in response to N-p-tolyl-2-(3,4,5-trimethoxyphenyl quinazolin-4-amine) (SL111), an inducer of cell-cycle arrest, followed by increased apoptosis. Additionally, they showed increased apoptosis in response to suberoylanilide hydroxamic acid (SAHA). Tumours formed in mice from injected anti-miR-155-expressing MyLa cells had a significantly lower volume and higher occurrence of apoptosis than controls. Stable overexpression of miR-155 in Hut78 cells had no effect. CONCLUSIONS: Oncogenic miR-155 appears to contribute to the cancerous phenotype of MyLa and MJ cells, but not of Hut78 cells, by interrupting activation of the G2/M checkpoint in response to SL111, and decreasing apoptosis in response to SL111 and SAHA, thereby facilitating tumour growth. These findings have implications for the potential development of novel therapeutic modalities for MF incorporating miR-155 inhibitors.


Subject(s)
MicroRNAs/physiology , Mycosis Fungoides/etiology , Skin Neoplasms/etiology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Female , Genes, cdc/drug effects , HEK293 Cells , Heterografts , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Lentivirus , Mice, SCID , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Phenotype , Quinazolines/pharmacology , Sezary Syndrome/etiology , Transduction, Genetic , Transplantation, Heterologous , Vorinostat
2.
Pharmacogenomics J ; 16(4): 393-8, 2016 08.
Article in English | MEDLINE | ID: mdl-26261062

ABSTRACT

Clinician attitudes toward multiplexed genomic testing may be vital to the success of translational programs. We surveyed clinicians at an academic medical center about their views on a large pharmacogenomics implementation, the PREDICT (Pharmacogenomic Resource for Enhanced Decisions in Care and Treatment) program. Participants were asked about test ordering, major factors influencing use of results, expectations of efficacy and responsibility for applying results to patient care. Virtually all respondents (99%) agreed that pharmacogenomics variants influence patients' response to drug therapy. The majority (92%) favored immediate, active notification when a clinically significant drug-genome interaction was present. However, clinicians were divided on which providers were responsible for acting on a result when a prescription change was indicated and whether patients should be directly notified of a significant result. We concluded genotype results were valued for tailoring prescriptions, but clinicians do not agree on how to appropriately assign clinical responsibility for actionable results from a multiplexed panel.The Pharmacogenomics Journal advance online publication, 11 August 2015; doi:10.1038/tpj.2015.57.


Subject(s)
Attitude of Health Personnel , Drug-Related Side Effects and Adverse Reactions/genetics , Health Knowledge, Attitudes, Practice , Pharmacogenetics , Pharmacogenomic Variants/genetics , Physicians/psychology , Adult , Aged , Female , Genotype , Humans , Male , Middle Aged , Perception , Pharmacogenomic Testing , Phenotype , Precision Medicine , Predictive Value of Tests , Risk Assessment , Risk Factors , Surveys and Questionnaires , Young Adult
3.
Genet Med ; 3(6): 405-10, 2001.
Article in English | MEDLINE | ID: mdl-11715004

ABSTRACT

PURPOSE: To evaluate genetics tests available for clinical, research, and public health purposes in terms of their public health impact as measured by the number of people who could potentially be tested. METHODS: Genetic tests for the 751 inherited diseases or conditions listed in the GeneTests database as of November 2000, were classified on the basis of their use for population-based testing and the prevalence of the disease or condition being tested. The GeneTests database divides the tests into two groups: those offered for clinical use and those available for research only. RESULTS: Of the 423 clinical tests, 51 had potentially greater impact on public health because of their use in statewide newborn screening programs, other population screening programs, or testing for common diseases with a prevalence over 1 in 2,000 people. Among the 328 tests performed for research purposes only, 18 met the criteria for potentially greater public health impact. CONCLUSIONS: Our classification scheme indicated that fewer than 10% of the genetic tests listed in the GeneTests database at the end of 2000 are highly relevant to public health. The majority of genetic tests are used in diagnosis and/or genetic counseling for rare, single-gene disorders in a limited number of people. However, as more tests are being considered for newborn screening, and associations between genes and common diseases are being discovered, the impact of genetic testing on public health is likely to increase.


Subject(s)
Genetic Diseases, Inborn/diagnosis , Genetic Testing/classification , Databases, Genetic , Genetic Counseling , Genetic Diseases, Inborn/epidemiology , Genetic Testing/statistics & numerical data , Genetics, Population , Humans , Infant, Newborn , Public Health/trends , United States
4.
Hepatology ; 32(3): 588-96, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10960454

ABSTRACT

Two human monoclonal antibodies (mAbs) against hepatitis B surface antigen (HBsAg) generated in the Trimera mouse system are described. Both mAbs 17.1.41 and 19.79.5 are of the IgG1 isotype and have high affinity constants for HBsAg binding in the range of 10(-10) mol/L. Monoclonal antibody 17.1.41 recognizes a conformational epitope on the a determinant of HBsAg whereas mAb 19.79.5 recognizes a linear one. The 2 mAbs bind to a panel of hepatitis B virus (HBV) subtypes with distinct patterns. The neutralizing activity of these antibodies was tested in 2 different animal model systems. Administration of each mAb to HBV-Trimera mice, a system that provides a mouse model for human hepatitis B infection, reduced the viral load and the percentage of HBV-DNA-positive mice in a dose-dependent manner. These 2 mAbs were more effective than a polyclonal antibody preparation (Hepatect; Biotest Pharma, Dreieich, Germany) in both inhibition of HBV liver infection and reduction of viral load. A single administration of a mixture of these mAbs into HBV chronic carrier chimpanzees resulted in immediate reduction in HBsAg levels followed by recurrence to initial levels within few days. Thus, these mAbs may be potential candidates for preventive therapy or in combination with other antiviral agents against HBV. Further studies in humans are needed to assess these mAbs in various clinical indications.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carrier State/drug therapy , Hepatitis B Surface Antigens/immunology , Hepatitis B/drug therapy , Animals , DNA, Viral/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Combinations , Hepatitis B/prevention & control , Hepatitis B/virology , Hepatitis B virus/genetics , Humans , Mice , Pan troglodytes , Viral Load
5.
Arch Pathol Lab Med ; 123(12): 1177-81, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10583922

ABSTRACT

CONTEXT: Hereditary hemochromatosis is recognized as one of the most common autosomal recessive disorders, with a prevalence of 1 in 200 to 400 in the white population. Early detection and treatment are completely effective in preventing pathology. It is anticipated that testing for hereditary hemochromatosis will increase, as will the need for a technology that can handle the demand. OBJECTIVE: To describe a high-throughput, single-tube, allele-specific multiplex polymerase chain reaction assay for identifying the 2 mutations in the HFE gene associated with hereditary hemochromatosis. DESIGN: Fluorescence-labeled polymerase chain reaction products from a multiplex polymerase chain reaction are analyzed by automated capillary electrophoresis. DATA ANALYSIS: The assay was validated by analysis of 25 blinded samples, and results were concordant with an established laboratory assay. CONCLUSION: The assay described offers a significant improvement over manual laboratory assays in throughput, reduced technologist time, and cost.


Subject(s)
Alleles , Electrophoresis, Capillary , HLA Antigens/genetics , Histocompatibility Antigens Class I/genetics , Membrane Proteins , Polymerase Chain Reaction/methods , Fluorescence , Genotype , Hemochromatosis Protein , Humans , Mutation/genetics , Single-Blind Method
6.
Hepatology ; 29(2): 553-62, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9918935

ABSTRACT

Previous studies have demonstrated the feasibility of implantation of human blood cells or tissues in lethally irradiated mice or rats, radioprotected with SCID mouse bone marrow cells: The Trimera system. In the present study, we describe the development of a mouse Trimera model for human hepatitis B virus (HBV) infection. In this model, viremia is induced by transplantation of ex vivo HBV-infected human liver fragments. Engraftment of the human liver fragments, evaluated by hematoxylin-eosin staining and human serum albumin mRNA expression, was observed in 85% of the transplanted animals 1 month postimplantation. Viremia levels were determined in these mice by measuring serum HBV DNA using polymerase chain reaction (PCR), followed by dot-blot hybridization. HBV DNA is first detected 8 days after liver transplantation. Viremia attains a peak between days 18 and 25 when HBV infection is observed in 85% of the transplanted animals. The HBV-Trimera model was used to evaluate the therapeutic effects of human polyclonal anti-HBs antibodies (Hepatect) and of two reverse-transcriptase inhibitors, lamivudine (3TC) and beta-L-5-fluoro-2',3'-dideoxycytidine (beta-L-5FddC). Treatment of HBV-Trimera mice with these drugs effectively reduced both the percentage of infected animals and the viral load in their sera. Treatment cessation resulted in rebound of viral load, indicating HBV replication upon drug withdrawal. These results show that the HBV-Trimera model represents a novel experimental tool for simulating human HBV infection and evaluating potential anti-HBV therapeutic agents.


Subject(s)
Antiviral Agents/therapeutic use , Disease Models, Animal , Hepatitis B/drug therapy , Animals , Antibodies, Viral/therapeutic use , DNA, Viral/blood , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Hepatitis B/virology , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/genetics , Humans , Kidney , Lamivudine/therapeutic use , Liver/virology , Liver Transplantation , Mice , Mice, Inbred NOD , Mice, SCID , Polymerase Chain Reaction , RNA, Messenger/analysis , Serum Albumin/genetics , Viremia
7.
Blood ; 92(10): 3701-9, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9808564

ABSTRACT

The human blood coagulation factor VIII C2 domain (Ser2173-Tyr2332) contains an epitope recognized by most polyclonal inhibitory anti-factor VIII alloantibodies and autoantibodies. We took advantage of the differential reactivity of inhibitory antibodies with human and porcine factor VIII and mapped a major determinant of the C2 epitope by using a series of active recombinant hybrid human/porcine factor VIII molecules. A series of five C2-specific human antibodies and a murine anti-factor VIII monoclonal antibody, NMC-VIII/5, inhibited a hybrid containing a substitution of porcine sequence for Glu2181-Val2243 significantly less than human factor VIII. In contrast, four of the five patient antibodies and NMC-VIII/5 inhibited a hybrid containing a substitution of porcine sequence for Thr2253-Tyr2332 equally well as human factor VIII. Thus, a major factor VIII inhibitor epitope determinant is bounded by Glu2181-Val2243 at the NH2-terminal end of the C2 domain. Because C2 inhibitors block the binding of factor VIII to phospholipid and von Willebrand factor, for which binding sites have been localized to Thr2303-Tyr2332, these results imply that the segment bounded by Glu2181-Val2243 also is involved in these macromolecular interactions.


Subject(s)
Epitopes/chemistry , Factor VIII/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cross Reactions , Epitopes/immunology , Factor VIII/antagonists & inhibitors , Factor VIII/immunology , Factor VIII/metabolism , Hemophilia A/blood , Humans , Macromolecular Substances , Molecular Sequence Data , Phospholipids/metabolism , Recombinant Fusion Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Structure-Activity Relationship , Swine , von Willebrand Factor/metabolism
8.
Immunology ; 93(2): 154-61, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9616363

ABSTRACT

An approach to develop fully human monoclonal antibodies in a human/mouse radiation chimera, the Trimera system, is described. In this system, functional human lymphocytes are engrafted in normal strains of mice which are rendered immuno-incompetent by lethal total body irradiation followed by radioprotection with severe combined immunodeficient (SCID) mouse bone marrow. Following transplantation, human lymphocytes colonize murine lymphatic organs and secrete human immunoglobulins. We have established this system as a tool to develop fully human monoclonal antibodies, and applied it for the generation of monoclonal antibodies specific for hepatitis B virus surface antigen. A strong memory response to hepatitis B surface antigen was elicited in Trimera engrafted with lymphocytes from human donors positive for antibodies to hepatitis B surface antigen. The human specific antibody fraction in the Trimera was 10(2)-10(3)-fold higher as compared with that found in the donors. Spleens were harvested from Trimera mice showing high specific-antibody titres and cells were fused to a human-mouse heteromyeloma fusion partner. Several stable hybridoma clones were isolated and characterized. These hybridomas produce high-affinity, IgG, anti-hepatitis B surface antigen antibodies demonstrating the potential of the Trimera system for generating fully human monoclonal antibodies. The biological function and the neutralizing activity of these antibodies are currently being tested.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Hepatitis B Antibodies/biosynthesis , Hepatitis B virus/immunology , Radiation Chimera/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Clone Cells/immunology , Hepatitis B Antibodies/chemistry , Hepatitis B Surface Antigens/immunology , Humans , Hybridomas/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data
9.
J Biol Chem ; 272(48): 30191-5, 1997 Nov 28.
Article in English | MEDLINE | ID: mdl-9374501

ABSTRACT

Antibodies directed to the A2 domain of factor VIII (fVIII) are usually an important component of the polyclonal response in patients who have clinically significant inhibitory antibodies to fVIII. A major determinant of the A2 epitope has been located by homolog scanning mutagenesis using recombinant hybrid human/porcine fVIII molecules to a sequence bounded by Arg484-Ile508 (Healey, J. F. , Lubin, I. M., Nakai, H., Saenko, E. L., Hoyer, L. W., Scandella, D. , and Lollar, P. (1995) J. Biol. Chem. 270, 14505-14509). Within this region, human residues Arg484, Pro485, Tyr487, Ser488, Arg489, Pro492, Val495, Phe501, and Ile508 differ from porcine fVIII. We stably expressed in mammalian cells nine active B-domainless human fVIII molecules containing single alanine substitutions at these sites. Their inhibition by a murine anti-A2 monoclonal antibody, monoclonal antibody (mAb) 413, and by three A2-specific alloimmune and two A2-specific autoimmune human inhibitor plasmas was measured by the Bethesda assay. The inhibition of Arg484 --> Ala, Tyr487 --> Ala, Arg489 --> Ala, and Arg492 --> Ala by mAb413 was reduced by greater than 90% compared with wild-type, B-domainless human fVIII. mAb413 inhibited the most severely affected mutant, Arg489 --> Ala, 0.01% as well as wild-type fVIII. For all five patient plasmas, the Tyr487 --> Ala mutant displayed the greatest reduction in inhibition. The inhibition of the Tyr487 --> Ala mutant by these antibodies ranged from 10% to 20% that of wild-type fVIII. The inhibition of the Ser488 --> Ala, Arg489 --> Ala, Pro492 --> Ala, Val495 --> Ala, Phe501 --> Ala, and Ile508 --> Ala mutants by most of the plasmas also was significantly reduced. In contrast, the Arg484 --> Ala and Pro485 --> Ala mutants were relatively unaffected. Thus, although mAb413 binds to the same region as human A2 inhibitors, it recognizes a different set of amino acid side chains. The side chains recognized by human A2 inhibitors appear to be similar, despite the differing immune settings that give rise to fVIII alloantibodies and autoantibodies.


Subject(s)
Factor VIII/immunology , Alanine/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal/immunology , Autoimmune Diseases/immunology , Epitope Mapping , Factor VIII/chemistry , Hemophilia A/immunology , Humans , Mice , Molecular Sequence Data , Mutagenesis , Recombinant Proteins , Swine
10.
J Virol ; 71(6): 4495-501, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9151841

ABSTRACT

Replication of human immunodeficiency virus type 1 (HIV-1) is regulated by virus-encoded regulatory proteins, as well as by a variety of cellular factors. Productive infection of human T lymphocytes by HIV-1 is dependent upon the activation status of the target cells. In general, short-term mitogenic stimulation of CD4 T cells is used to enhance infection of peripheral blood mononuclear cells (PBMC) in vitro. Recently, we demonstrated that adoptive transfer of human PBMC into lethally irradiated BALB/c mice, radioprotected with severe combined immunodeficiency (SCID) mouse bone marrow, leads to marked T-cell activation and proliferation. In the present study, we investigated the effect of such xenoactivation of human T cells on their susceptibility to HIV-1 infection. Human cells that were recovered from human/Balb radiation chimeras supported efficient replication of laboratory strains of HIV-1, as well as of HIV-1 clinical isolates. The multiplicity of infection required to attain effective virus replication in the recovered xenoactivated human cells was 10- to 100-fold lower than that needed for infection of short- or long-term phytohemagglutinin (PHA)-stimulated blasts or of various T-cell lines. Analysis of human cell surface activation markers has indicated that xenoactivation in the mouse, in contrast to in vitro stimulation with PHA, is associated with a marked downregulation of CD25 (interleukin 2 receptor). Our results demonstrate that human cells recovered from human/Balb radiation chimeras, which are hypersensitive to HIV-1 infection, differ from in vitro-stimulated cells in their activation status. Therefore, this system could be used to study host factors that participate in HIV-1 infection and replication in vitro and in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/microbiology , HIV-1/growth & development , Animals , Cell Line , Flow Cytometry , Humans , Immunophenotyping , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Radiation Chimera
11.
Transplantation ; 63(8): 1166-71, 1997 Apr 27.
Article in English | MEDLINE | ID: mdl-9133480

ABSTRACT

BACKGROUND: We recently described a new approach that enables the generation of human/mouse chimera by adoptive transfer of human peripheral blood mononuclear cells into lethally irradiated normal strains of mice or rats, radioprotected with bone marrow from donors with severe combined immune deficiency. In such human/mouse chimera, a marked humoral response to recall antigens, as well as a significant primary response to keyhole limpet hemocyanin, has been generated. METHODS: In the present study, the organ distribution of the engrafted human cells in the human/mouse and human/rat chimera was investigated by immunohistochemistry. RESULTS: Our results show that the T cells seem to be distributed throughout the reticular endothelial system, almost behaving like particles without any homing specificity. The B cells, however, can barely be found in internal organs, such as the liver or the pancreas, and are concentrated in the secondary lymphoid system (e.g., spleen, lymph node, and nonencapsulated lymphoid tissue). The B cells, together with the engrafted human T cells, form mixed lymphoid follicles. CONCLUSIONS: The different homing patterns exhibited by the T and B lymphocytes indicate that the homing receptors on human B cells might be cross-reactive with their mouse counterparts, in contrast to the human T cells, which seem to be unable to interact with the mouse homing receptors. The presence of human B and T lymphocytes in close proximity to each other in the lymphoid tissues is in accordance with the ability of human/BALB radiation chimera to mount significant primary human antibody responses.


Subject(s)
B-Lymphocytes/transplantation , Lymphoid Tissue/cytology , T-Lymphocytes/transplantation , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Humans , Immunoglobulin G/analysis , Immunoglobulin Heavy Chains/analysis , Immunoglobulin Light Chains/analysis , Immunoglobulin M/analysis , Immunohistochemistry , Lymph Nodes/cytology , Mice , Mice, Inbred BALB C , Mice, SCID , Radiation Chimera , Rats , Rats, Inbred Lew , Receptors, Antigen, B-Cell/analysis , Spleen/cytology , Staining and Labeling , T-Lymphocytes/immunology , Time Factors
12.
Blood ; 88(11): 4209-14, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8943856

ABSTRACT

The cDNA corresponding to 137 bp of the 5' untranslated region, the signal peptide, and the A1, A3, C1, and C2 domains of porcine factor VIII (fVIII) have been cloned and sequenced. Along with previously determined sequences of the porcine fVIII B domain and the A2 domain, this completes the sequence determination of the cDNA corresponding to the translated protein. Alignments of the derived amino acid sequence of porcine fVIII with human and murine fVIII indicate that the A1, A2, A3, C1, and C2 domains are more conserved than the B domains or the proteolytic cleavage peptides corresponding to residues 337-372 and 1649-1689. The knowledge of the porcine fVIII cDNA may be useful to understand functional and immunological differences between human and porcine fVIII and may lead to improved fVIII replacement products for hemophilia. A patients through the development of recombinant porcine fVIII or hybrid human/porcine fVIII derivatives.


Subject(s)
DNA, Complementary/genetics , Factor VIII/genetics , Swine/genetics , Amino Acid Sequence , Animals , Base Sequence , Epitopes/chemistry , Epitopes/immunology , Factor VIII/immunology , Genes , Humans , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Protein Sorting Signals/chemistry , Sequence Alignment , Species Specificity
13.
Blood ; 88(2): 721-30, 1996 Jul 15.
Article in English | MEDLINE | ID: mdl-8695821

ABSTRACT

Severe combined immunodeficient (SCID) mice are increasingly used as hosts for the adoptive transfer of human lymphocytes. Human antibody responses can be obtained in these xenogeneic chimeras, but information about the functionality of the human T cells in SCID mice is limited and controversial. Studies using human peripheral blood lymphocytes (PBL) injected intraperitoneally (IP) into SCID mice (hu-PBL-SCID mice) have shown that human T cells from these chimeras are anergic and have a defective signaling via the T-cell receptor. In addition, their antigenic repertoire is limited to xenoreactive clones. In the present study, we tested the functionality of human T cell in a recently described chimeric model. In this system, BALB/c mice are conditioned by irradiation and then transplanted with SCID bone marrow, followed by IP injection of human PBL. Our experiments demonstrated that human T cells, recovered from these hu-PBL-BALB mice within 1 month posttransplant, proliferated and expressed activation markers upon stimulation with anti-CD3 monoclonal antibody. A vigorous antiallogeneic human cytotoxic T-lymphocyte (CTL) response could be generated in these mice by immunizing them with irradiated allogeneic cells. Moreover, anti-human immunodeficiency virus type 1 (HIV-1) Net-specific human CTLs could be generated in vivo from naive lymphocytes by immunization of mouse-human chimeras with a recombinant vaccinia-nef virus. This model may be used to evaluate potential immunomodulatory drugs or cytokines, and could provide a relevant model for testing HIV vaccines, for production of antiviral T-cell clones for adoptive therapy, and for studying human T-cell responses in vivo.


Subject(s)
Gene Products, nef/immunology , HIV-1/immunology , Isoantigens/immunology , Mice, SCID/immunology , Radiation Chimera/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/transplantation , Transplantation, Heterologous/immunology , AIDS Vaccines/immunology , Animals , Bone Marrow Transplantation , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Cytotoxicity, Immunologic , Graft Rejection/immunology , Humans , Immunization , Immunotherapy, Adoptive , Injections, Intraperitoneal , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Muromonab-CD3/immunology , Neoplasm Transplantation/immunology , Receptors, Interleukin-2/analysis , Transplantation, Homologous , Tumor Cells, Cultured , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , nef Gene Products, Human Immunodeficiency Virus
14.
Immunol Lett ; 49(3): 155-61, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8739310

ABSTRACT

It has been shown that engraftment of human peripheral blood lymphocytes (PBL) from Epstein-Barr virus (EBV) seropositive donors in C.B-17/SCID mice is associated with a high incidence of human B cell tumors. More recently, we described a new approach enabling engraftment of human PBL in normal strains of mice or rats receiving lethal split-dose radiation and radioprotected with SCID bone marrow. We now demonstrate that, in contrast to SCID recipients of human PBL, Balb/c and C3H/HeJ recipients of 50-100 x 10(6) human PBL did not develop any EBV lymphoma during a 7-month follow-up period, but were successfully engrafted with human B and T cells. On the other hand, lymphoma developed in 90% of the C.B-17/SCID mice infused with 70 x 10(6) human PBL from the same donor. Likewise, 36% of beige/nude/xid (BNX) mice, exposed to 12 Gy TBI, radioprotected with SCID bone marrow and then transplanted with human PBL developed lymphoma. Similar results were obtained when different strains were infused with PBL of the same donor. Immunohistochemical analysis indicated that the tumor cells were of human B cell origin and expressed the EBV-encoded latent membrane protein-1 and nuclear antigen 2. While further studies are required to understand the mechanisms which suppressed outgrowth of EBV lymphoma in human --> mouse radiation chimera, compared to human --> C.B-17/SCID or human --> BNX chimera, this marked resistance offers new possibilities for transplantation of hematopoietic tissues or cells from EBV-positive donors.


Subject(s)
B-Lymphocytes/physiology , Herpesviridae Infections/immunology , Herpesvirus 4, Human/pathogenicity , Lymphoma/etiology , Tumor Virus Infections/etiology , Animals , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, SCID , Radiation Chimera , Species Specificity , Spleen/cytology , T-Lymphocytes/physiology , Time Factors
15.
Transplantation ; 60(7): 740-7, 1995 Oct 15.
Article in English | MEDLINE | ID: mdl-7570987

ABSTRACT

We have recently shown that lethally irradiated normal strains of mice, radioprotected with SCID bone marrow, can be engrafted with human peripheral blood mononuclear cells (PBMC). We now demonstrate that lethally irradiated Lewis rats can also be radioprotected with a transplant of SCID bone marrow cells, administered 1 day after total body irradiation. Split chimerism was found in PBMC, 30 days after transplantation, with predominance of SCID donor-type cells. The average percentages of CD4 and CD8 T cells, of mouse or rat origin, were < 1%. This chimerism status could be maintained for over 3 months. When human PBMC (300-1000 x 10(6) cells) were transplanted intraperitoneally 1 day after the administration of SCID bone marrow, prompt engraftment of human CD4 and human CD8 T cells, as well as human CD20 B cells, was found in the peritoneum and in internal organ (such as liver, lung, spleen, thymus, and lymph nodes). T cell activation was high: about 50% of the cells expressed HLA-DR and almost all expressed CD45RO. High titers of human Ig (> 1 mg/ml) were initially found after 2 weeks; these levels were similar to those found in the irradiated mouse model and in the SCID model. Likewise, marked human anti-tetanus response, predominantly of the IgG type, was recorded 2 weeks after the immunization, reaching maximal levels at 4 weeks. The triple-chimeric SCID-like rats, which accept as much as 1000 x 10(6) human PBMC, can potentially be used to elicit both antibody responses and T cell responses against specific antigens, with the advantages of a larger animal.


Subject(s)
Blood Component Transfusion , Bone Marrow Transplantation/immunology , Leukocytes, Mononuclear/immunology , Severe Combined Immunodeficiency/immunology , Transplantation Chimera/immunology , Transplantation, Heterologous/immunology , Animals , Antibody Formation/immunology , Humans , Immunologic Memory , Mice , Mice, SCID , Radiation Tolerance , Rats , Rats, Inbred Lew , Severe Combined Immunodeficiency/etiology , Species Specificity , T-Lymphocytes/immunology , Tissue Distribution , Whole-Body Irradiation
16.
Blood ; 86(1): 398-406, 1995 Jul 01.
Article in English | MEDLINE | ID: mdl-7795248

ABSTRACT

Lubin et al recently described a new approach that enables the generation of human/mouse chimera by adoptive transfer of human peripheral blood mononuclear cells (PBMC) into lethally irradiated normal strains of mice, radioprotected with bone marrow (BM) from donors with severe combined immune deficiency (SCID). In the present study, we demonstrate in such human/mouse chimera a marked humoral response to recall antigen, such as tetanus toxoid (TT) or hepatitis B surface antigen (HBsAg), as well as a significant primary response to keyhole limpet hemocyanin (KLH). Maximal anti-KLH response in human/Balb chimera was attained 2 to 4 weeks after the immunization and declined thereafter. One week after transplantation, the predominant anti-KLH subtype was IgM, while after 2 weeks, the dominance had shifted to IgG. Similar primary antibody response was also demonstrated against the human immunodeficiency virus (HIV) Nef protein. Comparison between human/Balb and human/SCID chimera showed a major difference in their ability to mount a primary response against KLH. In Balb/c recipients, more than half of the mice exhibited marked IgM titers against KLH, while there was hardly any anti-KLH IgM response in the SCID recipients. From the earliest time point onwards, when anti-KLH antibodies were found in the latter chimera, they were predominantly of the IgG type. We have previously shown that in human/Balb chimera, unlike in SCID recipients, dissemination of transplanted PBMC into the spleen and other internal organs occurs within 24 hours. Therefore, it is likely that the early seeding in the appropriate microenvironment of the lymphoid tissues, is crucial for the maintenance of virgin human B cells.


Subject(s)
Antibody Formation , Immunologic Memory , Immunotherapy, Adoptive , Leukocyte Transfusion , Radiation Chimera/immunology , Animals , Antibody Specificity , Bone Marrow Transplantation , Cell Movement , Hemocyanins/immunology , Hepatitis B Surface Antigens/immunology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Immunoglobulin M/biosynthesis , Immunoglobulin M/immunology , Lymphocyte Depletion , Lymphoid Tissue/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, SCID , Tetanus Toxoid/immunology , Transplantation, Heterologous
17.
J Infect Dis ; 172(1): 25-30, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7797923

ABSTRACT

Chimpanzees are currently the only nonhuman animal model for reproducible propagation of hepatitis C virus (HCV). A chimeric mouse model was used for the induction of hepatitis C viremia, using BNX (beige/nude/X-linked immunodeficient) mice preconditioned by total body irradiation and reconstituted with SCID mouse bone marrow cells. HCV-infected liver fragments from patients with HCV RNA-positive sera were transplanted under the kidney capsule of the chimeric mice. HCV-specific RNA sequences were detected by reverse transcriptase nested polymerase chain reaction (RT-PCR) in serum of approximately 50% of grafted animals. In addition, normal liver specimens were incubated with HCV serum and transplanted into chimeric mice, leading to viremia in approximately 25% of animals. Sequential histologic evaluation of the liver implants, from day 2 to week 14 after transplantation, revealed loss of lobular architecture within the implants. However, viremia persisted for 10-50 days after transplantation. These results offer a new HCV model.


Subject(s)
Bone Marrow Transplantation/immunology , Hepacivirus/isolation & purification , Hepatitis C/physiopathology , Hepatitis C/transmission , Liver Transplantation , Viremia/physiopathology , Animals , Base Sequence , Chimera , DNA Primers , Disease Models, Animal , Hepacivirus/genetics , Humans , Immunologic Deficiency Syndromes , In Situ Hybridization , Lymphocyte Depletion , Mice , Mice, Nude , Mice, SCID , Molecular Sequence Data , Pan troglodytes , Polymerase Chain Reaction , RNA, Viral/analysis , Rats , T-Lymphocytes , Time Factors , Transplantation, Heterologous , Whole-Body Irradiation
18.
J Biol Chem ; 270(24): 14505-9, 1995 Jun 16.
Article in English | MEDLINE | ID: mdl-7540171

ABSTRACT

The A2 domain (residues 373-740) of human blood coagulation factor VIII (fVIII) contains a major epitope for inhibitory alloantibodies and autoantibodies. We took advantage of the differential reactivity of inhibitory antibodies with human and porcine fVIII and mapped a major determinant of the A2 epitope by using a series of active recombinant hybrid human/porcine fVIII molecules. Hybrids containing a substitution of porcine sequence at segment 410-508, 445-508, or 484-508 of the human A2 domain were not inhibited by a murine monoclonal antibody A2 inhibitory, mAb 413, whereas hybrids containing substitutions at 387-403, 387-444, and 387-468 were inhibited by mAb 413. This indicates that the segment bounded by Arg484 and Ile508 contains a major determinant of the A2 epitope. mAb 413 did not inhibit two more hybrids that contained porcine substitutions at residues 484-488 and 489-508, indicating that amino acid side chains on both sides of the Ser488-Arg489 bond within the Arg484-Ile508 segment contribute to the A2 epitope. The 484-508, 484-488, and 489-508 porcine substitution hybrids displayed decreased inhibition by A2 inhibitors from four patient plasmas, suggesting that there is little variation in the structure of the A2 epitope in the inhibitor population.


Subject(s)
Epitopes/chemistry , Factor VIII/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Base Sequence , DNA Primers , Factor VIII/antagonists & inhibitors , Factor VIII/genetics , Humans , Immunoglobulin Heavy Chains/physiology , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry , Swine
19.
Immunol Lett ; 46(1-2): 9-14, 1995 May.
Article in English | MEDLINE | ID: mdl-7590935

ABSTRACT

Normal strains of mice are rendered sensitive to small amounts (3-10 micrograms) of staphylococcal enterotoxin B (SEB) by transplanting bone marrow cells of SCID donor mice to lethally irradiated recipients. Four to 12 weeks post-transplantation, SEB induces 56-100% lethality. Transplantation of normal mouse bone marrow cells, either alone or with the SCID mouse selected bone marrow cells, does not confer SEB sensitivity. These data imply that either irradiation ablates certain cell population(s), that confer resistance to SEB in normal mice (populations that are absent in the SCID donor mice) or that the donor cells selectively repopulate recipients with SEB-sensitive cells. This model will help elucidate the cells, cytokines and the SEB peptide fragments responsible for SEB toxicity and will be useful in identifying promising vaccine candidates and in developing preventive medicines to protect against this potent toxin.


Subject(s)
Bone Marrow Transplantation/adverse effects , Enterotoxins/toxicity , Radiation Chimera/immunology , Radiation Injuries, Experimental/immunology , Staphylococcus aureus/immunology , Whole-Body Irradiation/adverse effects , Animals , Dose-Response Relationship, Immunologic , Immunity, Innate , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, SCID , Molecular Sequence Data , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control
20.
J Immunol ; 153(6): 2562-7, 1994 Sep 15.
Article in English | MEDLINE | ID: mdl-8077666

ABSTRACT

After allogeneic bone marrow transplantation (BMT) for leukemia, beneficial graft-vs-leukemia (GVL) effects are usually accompanied by potentially serious graft-vs-host disease (GVHD). Because T cell depletion is the only effective way to prevent GVHD it seems important to understand whether effective GVL can develop after BMT with T cell depletion in GVHD-free recipients. Well-established C57BL/6-->BALB/c chimeras that were free of GVHD, reconstituted with T cell-depleted allogeneic bone marrow cells, and inoculated 3 mo after BMT with a high inoculation of murine B cell leukemia (BCL1) showed no evidence of disease, whereas all control mice developed leukemia and died within 58 days. Results from adoptive transfer experiments in secondary naive BALB/c recipients indicated that all BCL1 cells were eliminated in the chimeras within 14 days. Hence, complete resistance to BCL1 developed in the chimeras despite complete tolerance to host alloantigens. The GVL effects observed in tolerant chimeras were further amplified by administration of immunocompetent allogeneic C57BL/6 spleen cells, low dose rIL-2, or both for 5 days. Our data suggest that GVL effects can develop even after T cell depletion in the absence of clinically overt GVHD and that GVL can be further amplified by rIL-2, either with or without use of additional immunocompetent donor T cells. Our data may provide the basis for new approaches to induce effective GVL after allogeneic BMT with cell therapy and rIL-2 at the stage of minimal residual disease, while avoiding early GVHD induced by the BMT procedure.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease/immunology , Leukemia, B-Cell/therapy , Animals , Bone Marrow Cells , Female , Immune Tolerance/immunology , Immunotherapy, Adoptive , Interleukin-2/therapeutic use , Leukemia, B-Cell/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , Transplantation Chimera/immunology , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...