Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Transplant ; 15(7): 1948-57, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25809545

ABSTRACT

Successful lung transplantation (LTx) depends on multiple components of healthcare delivery and performance. Therefore, we conducted an international registry analysis to compare post-LTx outcomes for cystic fibrosis (CF) patients using the UNOS registry in the United States and the National Health Service (NHS) Transplant Registry in the United Kingdom. Patients with CF who underwent lung or heart-lung transplantation in the United States or United Kingdom between January 1, 2000 and December 31, 2011 were included. The primary outcome was all-cause mortality. Kaplan-Meier analysis and Cox proportional hazards regression evaluated the effect of healthcare system and insurance on mortality after LTx. 2,307 US LTx recipients and 451 individuals in the United Kingdom were included. 894 (38.8%) US LTx recipients had publically funded Medicare/Medicaid insurance. US private insurance and UK patients had improved median predicted survival compared with US Medicare/Medicaid recipients (p < 0.001). In multivariable Cox regression, US Medicare/Medicaid insurance was associated with worse survival after LTx (US private: HR0.78,0.68-0.90,p = 0.001 and UK: HR0.63,0.41-0.97, p = 0.03). This study in CF patients is the largest comparison of LTx in two unique health systems. Both the United States and United Kingdom have similar early survival outcomes, suggesting important dissemination of best practices internationally. However, the performance of US public insurance is significantly worse and may put patients at risk.


Subject(s)
Cystic Fibrosis/mortality , Cystic Fibrosis/surgery , Delivery of Health Care, Integrated/organization & administration , Graft Rejection/mortality , Lung Transplantation/mortality , National Health Programs/organization & administration , Postoperative Complications , Adult , Cohort Studies , Delivery of Health Care, Integrated/standards , Female , Follow-Up Studies , Humans , International Agencies , Male , National Health Programs/standards , Prognosis , Quality of Health Care , Registries , Risk Factors , Survival Rate , United Kingdom , United States
2.
Osteoarthritis Cartilage ; 22(2): 264-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24315792

ABSTRACT

OBJECTIVE: Meniscus injury increases the risk of osteoarthritis; however, the biologic mechanism remains unknown. We hypothesized that pro-inflammatory stimulation of meniscus would increase production of matrix-degrading enzymes, cytokines and chemokines which cause joint tissue destruction and could contribute to osteoarthritis development. DESIGN: Meniscus and cartilage tissue from healthy tissue donors and total knee arthroplasties (TKAs) was cultured. Primary cell cultures were stimulated with pro-inflammatory factors [IL-1ß, IL-6, or fibronectin fragments (FnF)] and cellular responses were analyzed by real-time PCR, protein arrays and immunoblots. To determine if NF-κB was required for MMP production, meniscus cultures were treated with inflammatory factors with and without the NF-κB inhibitor, hypoestoxide. RESULTS: Normal and osteoarthritic meniscus cells increased their MMP secretion in response to stimulation, but specific patterns emerged that were unique to each stimulus with the greatest number of MMPs expressed in response to FnF. Meniscus collagen and connective tissue growth factor (CTGF) gene expression was reduced. Expression of cytokines (IL-1α, IL-1ß, IL-6), chemokines (IL-8, CXCL1, CXCL2, CSF1) and components of the NF-κB and tumor necrosis factor (TNF) family were significantly increased. Cytokine and chemokine protein production was also increased by stimulation. When primary cell cultures were treated with hypoestoxide in conjunction with pro-inflammatory stimulation, p65 activation was reduced as were MMP-1 and MMP-3 production. CONCLUSIONS: Pro-inflammatory stimulation of meniscus cells increased matrix metalloproteinase production and catabolic gene expression. The meniscus could have an active biologic role in osteoarthritis development following joint injury through increased production of cytokines, chemokines, and matrix-degrading enzymes.


Subject(s)
Cytokines/biosynthesis , Inflammation Mediators/pharmacology , Matrix Metalloproteinases/biosynthesis , Menisci, Tibial/metabolism , Osteoarthritis, Knee/metabolism , Adult , Aged , Aged, 80 and over , Cells, Cultured , Chemokines/biosynthesis , Chemokines/genetics , Culture Media, Conditioned , Cytokines/genetics , Diterpenes/pharmacology , Gene Expression Regulation/drug effects , Humans , Menisci, Tibial/drug effects , Menisci, Tibial/pathology , Middle Aged , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Osteoarthritis, Knee/pathology , Protein Array Analysis/methods , Real-Time Polymerase Chain Reaction/methods , Signal Transduction/physiology
3.
J Cardiothorac Surg ; 19(1): 108, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409178

ABSTRACT

Artificial intelligence (AI) is a transformative technology with many benefits, but also risks when applied to healthcare and cardiac surgery in particular. Surgeons must be aware of AI and its application through generative pre-trained transformers (GPT/ChatGPT) to fully understand what this offers to clinical care, decision making, training, research and education. Clinicians must appreciate that the advantages and potential for transformative change in practice is balanced by risks typified by validation, ethical challenges and medicolegal concerns. ChatGPT should be seen as a tool to support and enhance the skills of surgeons, rather than a replacement for their experience and judgment. Human oversight and intervention will always be necessary to ensure patient safety and to make complex decisions that may require a refined understanding of individual patient circumstances.


Subject(s)
Cardiac Surgical Procedures , Heart Transplantation , Humans , Artificial Intelligence , Educational Status , Patient Safety
4.
J Exp Med ; 166(5): 1436-46, 1987 Nov 01.
Article in English | MEDLINE | ID: mdl-3119759

ABSTRACT

Recombinant granulocyte/macrophage colony-stimulating factor (rGM-CSF) obtained from cloned complementary Mo cell DNA and expressed in COS-1 cells activates cultured peripheral blood monocyte-derived macrophages in vitro to become cytotoxic for intracellular L. donovani. The antileishmanial effect of rGM-CSF, which can be completely neutralized by anti-rGM-CSF antiserum, is maximal after 36 h preincubation with the cultured macrophages, compared with that of rIFN-gamma, which reaches its maximum at 72 h of preincubation. The antileishmanial effect of GM-CSF as well as IFN-gamma is independent of detectable amounts of LPS and is not augmented by the addition of 10 or 50 ng/ml of LPS. Simultaneous administration of suboptimal doses of rGM-CSF and rIFN-gamma to monocyte-derived macrophages results in greater antileishmanial activity by these cells than administration of either lymphokine alone, although no enhancement of antileishmanial activity is observed when optimal doses of these two lymphokines are applied together.


Subject(s)
Colony-Stimulating Factors/pharmacology , Granulocytes , Growth Substances/pharmacology , Leishmania donovani/immunology , Macrophage Activation , Macrophages/immunology , Animals , Cells, Cultured , Drug Interactions , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Interferon-gamma/pharmacology , Kinetics , Lipopolysaccharides/pharmacology , Recombinant Proteins
5.
J Exp Med ; 160(4): 1147-69, 1984 Oct 01.
Article in English | MEDLINE | ID: mdl-6434688

ABSTRACT

The present study shows that recombinant interleukin 2 (IL-2) purified to homogeneity induces a rapid and potent enhancement of spontaneous cytotoxicity of human peripheral blood lymphocytes. The cells mediating cytotoxicity after 18-h treatment with IL-2 have surface markers of natural killer (NK) cells and are generated from the peripheral blood subset containing spontaneous cytotoxic cells. A parallel production of gamma interferon (IFN-gamma) is induced by recombinant IL-2 (rIL-2), and NK cells appear to be the major producer cells, whereas T cells are unable to produce IFN-gamma under these experimental conditions. However, the kinetics of the enhancement of cytotoxicity are faster than those of IFN-gamma production, and monoclonal anti-IFN-gamma antibodies do not suppress this effect, making it unlikely that the IFN-gamma produced is responsible for the enhancement. The enhancement of NK cell activity induced by rIL-2 precedes any proliferative response of the lymphocytes, which is instead observed in longer-term cultures of both NK and T cells.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-2/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation , Adjuvants, Immunologic/physiology , Antibodies, Monoclonal/immunology , Antigens, Surface/analysis , Growth Substances/physiology , Humans , Interferon-gamma/biosynthesis , Interleukin-2/isolation & purification , Interphase , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Kinetics , Phenotype
6.
J Exp Med ; 178(4): 1175-88, 1993 Oct 01.
Article in English | MEDLINE | ID: mdl-8104229

ABSTRACT

Post 5-fluorouracil-treated murine bone marrow cells infected with a recombinant retrovirus (murine stem cell virus-interleukin 11 [MSCV-IL-11]) bearing a human IL-11 gene were transplanted into lethally irradiated syngeneic mice. Analysis of proviral integration sites in DNA prepared from hematopoietic tissues and purified cell populations of long-term reconstituted primary and secondary recipients demonstrated polyclonal engraftment by multipotential stem cells. High levels (100-1,500 U/ml) of IL-11 were detected in the plasma of the MSCV-IL-11 mice. Systemic effects of chronic IL-11 exposure included loss of body fat, thymus atrophy, some alterations in plasma protein levels, frequent inflammation of the eyelids, and often a hyperactive state. A sustained rise in peripheral platelet levels (approximately 1.5-fold) was seen throughout the observation period (4-17 wk). No changes were observed in the total number of circulating leukocytes in the majority of the transplanted animals (including 10 primary and 18 secondary recipients) despite a > 20-fold elevation in myeloid progenitor cell content in the spleen. The exceptions were members of one transplant pedigree which presented with myeloid leukemia during the secondary transplant phase. A clonal origin of the disease was determined, with significant expansion of the MSCV-IL-11-marked clone having occurred in the spleen of the primary host. Culturing of leukemic spleen cells from a quaternary recipient led to the establishment of a permanent cell line (denoted PGMD1). IL-11-producing PGMD1 myeloid leukemic cells are dependent on IL-3 for continuous growth in vitro and they differentiate into granulocytes and macrophages in response to granulocyte/macrophage colony-stimulating factor. The inability of autogenously produced IL-11 to support autonomous growth of PGMD1 cells argues against a mechanism of transformation involving a classical autocrine loop.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cells/pathology , Interleukin-11/physiology , Leukemia, Myeloid/etiology , Animals , B-Lymphocytes/microbiology , Bone Marrow/microbiology , Bone Marrow/pathology , Bone Marrow Transplantation , Chimera , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/microbiology , Humans , Hyperplasia , Interleukin-11/genetics , Interleukin-11/metabolism , Interleukin-6/physiology , Leukemia, Myeloid/pathology , Mice , Mice, Inbred BALB C , Platelet Count , Retroviridae/genetics
7.
J Exp Med ; 168(2): 463-74, 1988 Aug 01.
Article in English | MEDLINE | ID: mdl-3261773

ABSTRACT

IL-1 is released by activated monocytes and is thought to be a key mediator of the host immune response. The availability of the purified and, more recently, recombinant IL-1 has allowed the characterization of other biological properties of this molecule. Thus, IL-1 is thought to have the same properties as hemopoietic 1, a growth factor that has been shown to act on primitive murine hemopoietic cells. Here we report that rIL-1 acts synergistically with granulocyte/macrophage CSF (GM-CSF) or granulocyte CSF in the stimulation of clonogenic cells from many patients with acute myeloblastic leukemia (AML). Although IL-1 by itself has no effect on AML blasts, it can support colony formation under conditions where there is detectable production of endogenous GM-CSF. IL-1 also promotes the growth of multipotential progenitors from normal human bone marrow cells in the presence of GM-CSF. These observations support the hypothesis that in the hemopoietic system, IL-1 has a selective effect on primitive precursors.


Subject(s)
Colony-Forming Units Assay , Colony-Stimulating Factors/pharmacology , Hematopoietic Stem Cells/cytology , Interleukin-1/physiology , Leukemia, Myeloid, Acute/pathology , Recombinant Proteins/pharmacology , Tumor Cells, Cultured/cytology , Tumor Stem Cell Assay , Bone Marrow Cells , Cell Division/drug effects , Cells, Cultured , Colony-Stimulating Factors/immunology , Drug Synergism , Hematopoietic Stem Cells/drug effects , Humans , Immune Sera , Interleukin-1/genetics , Kinetics , Nucleic Acid Hybridization , Tumor Cells, Cultured/drug effects
8.
J Exp Med ; 169(2): 569-83, 1989 Feb 01.
Article in English | MEDLINE | ID: mdl-2521357

ABSTRACT

We have analyzed the ability of highly purified preparations of human NK cells to produce CSF. NK cells, purified by negative selection from 10-d cultures of PBMC incubated with irradiated B-lymphoblastoid cell lines, were stimulated with rIL-2, FcR(CD16) ligands (particulate immune complexes or anti-CD16 antibodies bound to Sepharose), a combination of CD16 ligands and rIL-2, or the phorbol diester phorbol dibutyrate (PDBu) together with the Ca2+ ionophore A23187. Both rIL-2 and CD16 ligands induce accumulation of GM-CSF mRNA in NK cells and the combined effect of the two stimuli is synergistic. Maximal accumulation of GM-CSF mRNA is observed after PDBu/A23187 stimulation. The participation of contaminant T cells in the observed expression of the GM-CSF gene is excluded because CD16 ligands do not stimulate T cells and CD3 ligands, powerful stimulators of T cells, are inactive on NK cells. Accumulation of CSF-1 mRNA is observed only in NK cells stimulated with both CD16 ligands and rIL-2, whereas accumulation of IL-3 mRNA is observed only in NK cells stimulated with PDBu/A23187. Transcripts of the G-CSF, IL-1 alpha, and IL-1 beta genes were never detected in NK cells in these experiments. The kinetics of accumulation of GM-CSF and CSF-1 mRNA in NK cells stimulated with CD16 ligands and rIL-2 peaked at 2-4 h and was slower than that of TNF and IFN-gamma mRNA, which peak at 1 h. GM-CSF was precipitated from the supernatant fluids of NK cells stimulated with PDBu/A23187 and its biological activity was demonstrated by the ability of the supernatants to sustain proliferation of the TALL-101 cell line or CML blasts. Biological activity of IL-3 and CSF-1 was demonstrable in supernatant fluids of NK cells stimulated with PDBu/A23187 and CD16 ligands/rIL-2, respectively.


Subject(s)
Colony-Stimulating Factors/biosynthesis , Growth Substances/biosynthesis , Interleukin-3/biosynthesis , Killer Cells, Natural/physiology , Antigens, Differentiation/physiology , Antigens, Differentiation, T-Lymphocyte/physiology , Biological Assay , Blotting, Northern , CD3 Complex , Calcimycin/pharmacology , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Interleukin-1/biosynthesis , Interleukin-2/pharmacology , Ligands , Phorbol Esters/pharmacology , Precipitin Tests , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/physiology , Receptors, Fc/physiology , Receptors, IgG
9.
J Exp Med ; 185(11): 1939-50, 1997 Jun 02.
Article in English | MEDLINE | ID: mdl-9166423

ABSTRACT

The receptor for granulocyte/macrophage colony-stimulating factor (GM-CSF) is composed of two chains, alpha and betac. Both chains belong to the superfamily of cytokine receptors characterized by a common structural feature, i.e., the presence of at least two fibronectin-like folds in the extracellular domain, which was first identified in the growth hormone receptor. The GM-CSF receptor (GMR)-alpha chain confers low affinity binding only (5-10 nM), whereas the other chain, betac, does not bind GM-CSF by itself but confers high affinity binding when associated with GMR-alpha (25-100 pM). The present study was designed to define the assembly of the GMR complex at the molecular level through site-directed mutagenesis guided by homology modeling with the growth hormone receptor complex. In our three-dimensional model, R280 of GMR-alpha, located in the F'-G' loop and close to the WSSWS motif, is in the vicinity of the ligand Asp112, suggesting the possibility of electrostatic interaction between these two residues. Through site directed mutagenesis, we provide several lines of evidence indicating the importance of electrostatic interaction in ligand-receptor recognition. First, mutagenesis of GMR-alphaR280 strikingly ablated ligand binding in the absence of beta common (betac); ligand binding was restored in the presence of betac with, nonetheless, a significant shift from high (26 pM) toward low affinity (from 2 to 13 nM). The rank order of the dissociation constant for the different GMR-alphaR280 mutations where Lys > Gln > Met > Asp, suggesting the importance of the charge at this position. Second, a mutant GM-CSF with charge reversal mutation at position Asp112 exhibited a 1,000-fold decrease in affinity in receptor binding, whereas charge ablation or conservative mutations were the least affected (10-20-fold). Third, removal of the charge at position R280 of GMR-alpha introduced a 10-fold decrease in the association rate constant and only a 2-fold change in the dissociation rate constant, suggesting that R280 is implicated in ligand recognition, possibly through interaction with Asp112 of GM-CSF. For all R280 mutants, the half-efficient concentrations of GM-CSF required for membrane (receptor binding) to nuclear events (c-fos promoter activation) and cell proliferation (thymidine incorporation) were in the same range, indicating that the threshold for biologic activity is governed mainly by the affinity of ligand-receptor interaction. Furthermore, mutation of other residues in the immediate vicinity of R280 was less drastic. Sequence alignment and modeling of interleukin (IL)-3R and IL-5R identified an arginine residue at the tip of a beta turn in a highly divergent context at the F'-G' loop, close to a conserved structural element, the WSXWS motif, suggesting the possibility of a ligand association mechanism similar to the one described herein for GMR.


Subject(s)
Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , 3T3 Cells , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , CHO Cells , Cricetinae , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Kinetics , Ligands , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Sequence Homology, Amino Acid , Software , Transfection/genetics
10.
J Exp Med ; 174(4): 925-9, 1991 Oct 01.
Article in English | MEDLINE | ID: mdl-1717634

ABSTRACT

We have used antisense oligonucleotides to study the roles of transforming growth factor beta (TGF-beta) and the two antioncogenes, retinoblastoma susceptibility (Rb) and p53, in the negative regulation of proliferation of early hematopoietic cells in culture. The antisense TGF-beta sequence significantly enhanced the frequency of colony formation by multi-lineage, early erythroid, and granulomonocytic progenitors, but did not affect colony formation by late progenitors. Single cell culture and limiting dilution analysis indicated that autocrine TGF-beta is produced by a subpopulation of early progenitors. Antisense Rb but not antisense p53 yielded similar results in releasing multipotential progenitors (colony-forming unit-granulocyte/erythroid/macrophage/megakaryocyte) from quiescence. Rb antisense could partially reverse the inhibitory effect of exogenous TGF-beta. Anti-TGF-beta blocking antibodies, antisense TGF-beta, or Rb oligonucleotides all had similar effects. No additive effects were observed when these reagents were combined, suggesting a common pathway of action. Our results are consistent with the model that autocrine production of TGF-beta negatively regulates the cycling status of early hematopoietic progenitors through interaction with the Rb gene product.


Subject(s)
Cell Division/drug effects , Genes, Retinoblastoma , Genes, p53 , Hematopoietic Stem Cells/cytology , Oligonucleotides, Antisense/pharmacology , Thionucleotides/pharmacology , Transforming Growth Factor beta/genetics , Antigens, CD/analysis , Antigens, CD34 , Base Sequence , Colony-Forming Units Assay , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Kinetics , Molecular Sequence Data
11.
J Exp Med ; 170(3): 827-45, 1989 Sep 01.
Article in English | MEDLINE | ID: mdl-2504877

ABSTRACT

We have identified and purified a novel cytokine, NK cell stimulatory factor (NKSF), from the cell-free supernatant fluid of the phorbol diester-induced EBV-transformed human B lymphoblastoid cell line RPMI 8866. NKSF activity is mostly associated to a 70-kD anionic glycoprotein. The purified 70-kD protein, isolated from an SDS-PAGE gel, yields upon reduction two small species of molecular masses of 40 and 35 kD, suggesting that this cytokine is a heterodimer. When added to human PBL, purified NKSF preparations induce IFN-gamma production and synergize with rIL-2 in this activity, augment the NK cell-mediated cytotoxicity of PBL preparations against both NK-sensitive and NK-resistant target cell lines, and enhance the mitogenic response of T cells to mitogenic lectins and phorbol diesters. The three activities remain associated through different purification steps resulting in a 9,200-fold purification, and purified NKSF mediates the three biological activities at concentrations in the range of 0.1-10 pM. These data strongly suggest that the same molecule mediates these three activities, although the presence of traces of contaminant peptides even in the most purified NKSF preparations does not allow us to exclude the possibility that distinct biologically active molecules have been co-purified. The absence of other known cytokines in the purified NKSF preparations, the unusual molecular conformation of NKSF, the high specific activity of the purified protein, and the spectrum of biological activities distinguish NKSF from other previously described cytokines.


Subject(s)
Biological Factors/isolation & purification , Killer Cells, Natural/drug effects , Biological Factors/analysis , Biological Factors/pharmacology , Cell Line , Cytokines , Cytotoxicity, Immunologic/drug effects , Interferon-gamma/biosynthesis , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocytes/immunology , Molecular Weight , Phorbol 12,13-Dibutyrate/pharmacology
12.
J Exp Med ; 173(4): 869-79, 1991 Apr 01.
Article in English | MEDLINE | ID: mdl-1672545

ABSTRACT

We previously reported that natural killer cell stimulatory factor (NKSF), a heterodimeric lymphokine purified from the conditioned medium of human B lymphoblastoid cell lines, induces interferon gamma (IFN-gamma) production from resting peripheral blood lymphocytes (PBL) and synergizes with interleukin 2 in this activity. In this study, we show that human NKSF induces IFN-gamma production from both resting and activated human PBL and from freshly isolated murine splenocytes. Human T and NK cells produce IFN-gamma in response to NKSF, but resting PBL require the presence of nonadherent human histocompatibility leukocyte antigens DR+ (HLA-DR+) accessory cells to respond to NKSF. The mechanism(s) by which NKSF induces IFN-gamma production results in accumulation of IFN-gamma mRNA, is insensitive to cyclosporin A, and synergizes with those mediated by phytohemagglutinin, phorbol diesters, anti-CD3 antibodies, and allogeneic antigens, but not by Ca2+ ionophores. The ability of NKSF to directly induce IFN-gamma production and to synergize with other physiological IFN-gamma inducers, joined with the previously described ability to enhance lymphocyte cytotoxicity and proliferation, indicates that this lymphokine is a powerful immunopotentiating agent.


Subject(s)
Cytokines/pharmacology , Interferon-gamma/biosynthesis , Interleukins/pharmacology , Killer Cells, Natural/metabolism , Killer Cells, Natural/physiology , T-Lymphocytes/metabolism , Animals , Antigen-Presenting Cells/physiology , Blotting, Northern , Cells, Cultured , Cyclosporins/pharmacology , Gene Expression , HLA-DR Antigens/analysis , Humans , In Vitro Techniques , Interferon-gamma/genetics , Interleukin-12 , Lymphocyte Culture Test, Mixed , Mice , RNA, Messenger/genetics , Recombinant Proteins
13.
Science ; 236(4806): 1229-37, 1987 Jun 05.
Article in English | MEDLINE | ID: mdl-3296190

ABSTRACT

The complementary DNAs and genes encoding the four major human myeloid growth factors--granulocyte colony-stimulating factor, macrophage colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, and interleukin-3--have all been molecularly cloned. These DNA clones have proved valuable for studying the molecular biology of these important regulatory molecules as well as for the large-scale production of the recombinant growth factor proteins. These advances have led to a much better understanding of the role of the myeloid growth factors in regulating hematopoiesis in vivo that should soon find practical application in clinical medicine.


Subject(s)
Interleukin-3 , Cloning, Molecular , Gene Expression Regulation , Granulocytes/cytology , Humans , Interleukin-3/genetics , Interleukin-3/physiology , Interleukin-3/therapeutic use , Macrophages/cytology , Recombinant Proteins
14.
Science ; 226(4671): 177-9, 1984 Oct 12.
Article in English | MEDLINE | ID: mdl-6091270

ABSTRACT

Human T-cell leukemia viruses (HTLV) are closely associated with some human T-cell leukemias and lymphomas. A unique 3' region of the HTLV genome is believed to be involved in HTLV-induced cellular transformation, although the function of this region has yet to be determined. A subgenomic messenger RNA transcribed from this region of HTLV has now been characterized. These results provide direct evidence for the expression of a novel gene in HTLV.


Subject(s)
Deltaretrovirus/genetics , Genes, Viral , RNA, Viral/genetics , Viral Proteins/genetics , Base Sequence , Cell Line , Cell Transformation, Viral , Deltaretrovirus/physiology , Humans , Nucleic Acid Hybridization , RNA Splicing , RNA, Messenger/genetics , T-Lymphocytes , Transcription, Genetic
15.
Science ; 228(4707): 1534-7, 1985 Jun 28.
Article in English | MEDLINE | ID: mdl-2990032

ABSTRACT

The human T-cell leukemia viruses (HTLV) are replication-competent retroviruses whose genomes contain gag, pol, and env genes as well as a fourth gene, termed x, which is believed to be the transforming gene of HTLV. The product of the x gene is now shown to be encoded by a 2.1-kilobase messenger RNA derived by splicing of at least two introns. By means of S1 nuclease mapping of this RNA and nucleic acid sequence analysis of a complementary DNA clone, the complete primary structure of the x-gene product has been determined. It is encoded by sequences containing the env initiation codon and one nucleotide of the next codon spliced to the major open reading frame of the HTLV-I and HTLV-II x gene.


Subject(s)
Deltaretrovirus/genetics , Methionine/genetics , Viral Proteins/analysis , Amino Acid Sequence , Animals , Cell Transformation, Viral , Codon , Electrophoresis, Polyacrylamide Gel , Humans , Rats
16.
Science ; 226(4680): 1339-42, 1984 Dec 14.
Article in English | MEDLINE | ID: mdl-6390681

ABSTRACT

Neutrophil migration inhibition factor from T lymphocytes (NIF-T) is a lymphokine that acts to localize granulocytes. Medium conditioned by the Mo human T-lymphoblast cell line was used to purify NIF-T, a glycoprotein with a molecular weight of 22,000. The NIF-T was found to potently stimulate the growth of granulocyte and macrophage colonies from human bone marrow and colony formation by the KG-1 myeloid leukemia cell line. Thus a human lymphokine (NIF-T) that modulates the activities of mature neutrophilic granulocytes is also a colony-stimulating factor acting on precursors to induce growth and differentiation of new effector cells.


Subject(s)
Colony-Stimulating Factors/isolation & purification , Granulocytes/cytology , Leukocyte Migration-Inhibitory Factors/pharmacology , Lymphokines/pharmacology , Macrophages/cytology , Bone Marrow Cells , Cell Division , Cell Line , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Molecular Weight
17.
Science ; 230(4730): 1171-3, 1985 Dec 06.
Article in English | MEDLINE | ID: mdl-3877981

ABSTRACT

Human recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) was tested for its ability to induce colony formation in human bone marrow that had been enriched for progenitor cells. In addition to its expected granulocyte-monocyte colony-stimulating activity, the recombinant GM-CSF had burst-promoting activity for erythroid burst-forming units and also stimulated colonies derived from multipotent (mixed) progenitors. In contrast, recombinant erythroid-potentiating activity did not stimulate erythroid progenitors. The experiments prove that human GM-CSF has multilineage colony-stimulating activity.


Subject(s)
Bone Marrow/drug effects , Colony-Stimulating Factors/pharmacology , Granulocytes/drug effects , Macrophages/drug effects , Recombinant Proteins/pharmacology , Animals , Colony-Stimulating Factors/biosynthesis , DNA/genetics , Dose-Response Relationship, Drug , Erythroblasts/drug effects , Humans , Mice , Stem Cells/drug effects , T-Lymphocytes/drug effects
18.
Heart Surg Forum ; 11(1): E21-3, 2008.
Article in English | MEDLINE | ID: mdl-18270133

ABSTRACT

Reoperative cardiac surgery is associated with substantial morbidity and mortality due to technical problems at sternal reentry, which can result in laceration of the right ventricle, innominate vein injury, or embolization from patent grafts. To minimize the risk associated with reentry, we adopted the method of assisted venous drainage in the cardiopulmonary bypass circuit with peripheral cannulation for cardiac reoperations. From March 1999 to May 2003, a series of 52 patients (38 males; mean age 48.7 years, range 4 months to 78 years) underwent cardiac reoperations performed with centrifugal pump venous-assisted cardiopulmonary bypass. EuroSCORE was 7.34 +/- 3.9 (range, 4-19). The reoperations were coronary artery bypass graft (25 patients), valve replacement/repair (18 patients), and complex pediatric procedures (11 patients). The studied adverse events were structural damage at reentry, mortality, blood loss, stroke, and hemolysis. Complications at sternotomy were damage to the innominate vein (1 patient) and aorta (1 patient) with blood loss of 625 and 225 mL, respectively. Four patients required intraaortic balloon pump or extracorporeal membrane oxygenation (n = 1) for hemodynamic support on weaning off cardiopulmonary bypass. Three patients died in the postoperative period. Our experience with centrifugal pump-assisted venous drainage in cardiac reoperations has shown excellent results, with reduced risk of damage to vital structures on sternal reentry. In cases in which structural damage did occur, blood loss was minimal.


Subject(s)
Cardiac Surgical Procedures/instrumentation , Cardiopulmonary Bypass/methods , Reoperation , Suction/methods , Treatment Failure , Vacuum Curettage/instrumentation , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Health Status Indicators , Humans , Infant , Male , Middle Aged , Retrospective Studies , Risk Factors , Sternum/surgery , Suction/instrumentation , Treatment Outcome , Vacuum Curettage/methods
19.
J Clin Invest ; 94(3): 1076-84, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8083348

ABSTRACT

Advanced human malignancies cannot be permanently cured by adoptive transfer of autologous lymphokine-activated killer (LAK) cells. Moreover, administration of high doses of IL-2 to maintain LAK activity in vivo is associated with severe toxicity. In this study, we tested the anti-tumor efficacy of a uniquely potent MHC non-restricted human killer T cell clone (TALL-104) in humanized severe combined immunodeficient (SCID) mice bearing acute myelogenous leukemia (AML). We show that, in appropriate experimental conditions, TALL-104 cells could effectively reverse the aggressive growth of the myelomonocytic leukemia cell line U937 in SCID mouse tissues, leading to complete abrogation of AML. A single transfer of TALL-104 cells at the time of tumor challenge in combination with recombinant human (rh) IL-12 (1 microgram/d) prolonged significantly the life of the mice. However, eradication of leukemia, as monitored both clinically and by PCR, was achieved by repeated injection of the effectors at close intervals. Complete cure was obtained also upon transfer of lethally irradiated (non-proliferating) TALL-104 cells together with low doses of rh IL-2 (100 U/d). Most notably, of the mice that received multiple transfers of TALL-104 cells without cytokines in an advanced disease stage, 50% were clinically cured, and 50% survived significantly longer. The potential of TALL-104 cells as an effective and safe leukemia purging agent is discussed.


Subject(s)
Immunotherapy, Adoptive , Killer Cells, Lymphokine-Activated , Leukemia, Myeloid, Acute/therapy , Lymphocyte Transfusion , Animals , Cell Line , Clone Cells , Cytotoxicity, Immunologic , Etoposide/therapeutic use , Gamma Rays , Humans , Immunosuppression Therapy , Killer Cells, Natural/immunology , Killer Cells, Natural/radiation effects , Mice , Mice, SCID , Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Cells, Cultured
20.
J Clin Invest ; 78(2): 597-601, 1986 Aug.
Article in English | MEDLINE | ID: mdl-3090106

ABSTRACT

Human granulocyte-macrophage colony-stimulating factor (GM-CSF) has been shown to inhibit migration of mature granulocytes and to enhance their antibody-dependent cellular cytotoxicity. We found that human recombinant GM-CSF also enhanced granulocyte-granulocyte adhesion and increased by two- to threefold the surface expression of Mo1 and LeuM5 (P150, 95), two members of a family of leukocyte adhesion molecules (Leu-CAM). Increased Mo1 surface expression occurred within 15 min at 37 degrees C and was maximal at the migration inhibitory concentration of 500 pM. One-half maximal rise in the expression of Mo1 on the cell surface occurred at 5 pM. The chemotactic peptide f-Met-Leu-Phe produced a comparable rise in surface Mo1 with one-half maximal expression occurring at 7 nM. Both GM-CSF and f-Met-Leu-Phe produced optimal granulocyte-granulocyte adhesion at 500 pM and 100 nM, respectively. This adhesion-promoting effect induced by either stimulus was inhibited by a mouse monoclonal antibody directed against Mo1 antigen. These data indicate that GM-CSF promotes cell-to-cell adhesion, presumably through enhanced expression of leukocyte adhesion molecules. This mechanism may explain, in part, the known effects of GM-CSF on the function of mature granulocytes.


Subject(s)
Antigens, Surface/biosynthesis , Colony-Stimulating Factors/pharmacology , Glycoproteins/biosynthesis , Granulocytes/metabolism , Recombinant Proteins/pharmacology , Animals , Antigens, Differentiation, T-Lymphocyte , Cell Adhesion/drug effects , Cell Adhesion Molecules , Cell Aggregation/drug effects , Cell Differentiation , Cricetinae , Cricetulus , Granulocytes/physiology , Humans , Lymphocyte Function-Associated Antigen-1
SELECTION OF CITATIONS
SEARCH DETAIL