Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Blood ; 97(10): 3146-51, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342442

ABSTRACT

During the innate immune response to infection, monocyte-derived cytokines (monokines), stimulate natural killer (NK) cells to produce immunoregulatory cytokines that are important to the host's early defense. Human NK cell subsets can be distinguished by CD56 surface density expression (ie, CD56(bright) and CD56(dim)). In this report, it is shown that CD56(bright) NK cells produce significantly greater levels of interferon-gamma, tumor necrosis factor-beta, granulocyte macrophage-colony-stimulating factor, IL-10, and IL-13 protein in response to monokine stimulation than do CD56(dim) NK cells, which produce negligible amounts of these cytokines. Further, qualitative differences in CD56(bright) NK-derived cytokines are shown to be dependent on the specific monokines present. For example, the monokine IL-15 appears to be required for type 2 cytokine production by CD56(bright) NK cells. It is proposed that human CD56(bright) NK cells have a unique functional role in the innate immune response as the primary source of NK cell-derived immunoregulatory cytokines, regulated in part by differential monokine production.


Subject(s)
CD56 Antigen/analysis , Homeostasis , Immunity , Killer Cells, Natural/immunology , Lectins, C-Type , Antigens, CD/analysis , Cell Division , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-2/pharmacology , Interleukins/biosynthesis , Ionomycin/pharmacology , Lipopolysaccharides/pharmacology , Lymphotoxin-alpha/biosynthesis , Macrophages/metabolism , Membrane Glycoproteins/analysis , NK Cell Lectin-Like Receptor Subfamily D , RNA, Messenger/analysis , Receptors, IgG/analysis , Receptors, Immunologic/analysis , Receptors, Interleukin-2/analysis , Receptors, Interleukin-2/physiology , Receptors, KIR , Tetradecanoylphorbol Acetate/pharmacology
2.
FASEB J ; 14(9): 1083-92, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10834929

ABSTRACT

Apoptosis is a cell suicide mechanism that requires the activation of cellular death proteases for its induction. We examined whether the progress of apoptosis involves cleavage of phospholipase C-gamma1 (PLC-gamma1), which plays a pivotal role in mitogenic signaling pathway. Pretreatment of T leukemic Molt-4 cells with PLC inhibitors such as U-73122 or ET-18-OCH(3) potentiated etoposide-induced apoptosis in these cells. PLC-gamma1 was fragmented when Molt-4 cells were treated with several apoptotic stimuli such as etoposide, ceramides, and tumor necrosis factor alpha. Cleavage of PLC-gamma1 was blocked by overexpression of Bcl-2 and by specific inhibitors of caspases such as Z-DEVD-CH(2)F and YVAD-cmk. Purified caspase-3 and caspase-7, group II caspases, cleaved PLC-gamma1 in vitro and generated a cleavage product of the same size as that observed in vivo, suggesting that PLC-gamma1 is cleaved by group II caspases in vivo. From point mutagenesis studies, Ala-Glu-Pro-Asp(770) was identified to be a cleavage site within PLC-gamma1. Epidermal growth factor receptor (EGFR) -induced tyrosine phosphorylation of PLC-gamma1 resulted in resistance to cleavage by caspase-3 in vitro. Furthermore, cleaved PLC-gamma1 could not be tyrosine-phosphorylated by EGFR in vitro. In addition, tyrosine-phosphorylated PLC-gamma1 was not significantly cleaved during etoposide-induced apoptosis in Molt-4 cells. This suggests that the growth factor-induced tyrosine phosphorylation may suppress apoptosis-induced fragmentation of PLC-gamma1. We provide evidence for the biochemical relationship between PLC-gamma1-mediated signal pathway and apoptotic signal pathway, indicating that the defect of PLC-gamma1-mediated signaling pathway can facilitate an apoptotic progression.


Subject(s)
Apoptosis , Isoenzymes/metabolism , Type C Phospholipases/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Apoptosis/drug effects , Caspase 3 , Caspase 7 , Caspase Inhibitors , Caspases/metabolism , Ceramides/pharmacology , ErbB Receptors/metabolism , Etoposide/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Mice , Molecular Weight , Mutation/genetics , Phospholipase C gamma , Phosphorylation , Phosphotyrosine/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/genetics
3.
J Biol Chem ; 273(21): 13119-28, 1998 May 22.
Article in English | MEDLINE | ID: mdl-9582351

ABSTRACT

Although the available evidence suggests that whereas the caspase family plays a major role in apoptosis, they are not the sole stimulators of death. A random yeast two-hybrid screen of a lymphocyte cDNA library (using caspase-3 as the bait) found an interaction between caspase-3 and the regulatory subunit Aalpha of protein phosphatase 2A. This protein was found to be a substrate for caspase-3, but not caspase-1, and could compete effectively against either a protein or synthetic peptide substrate. In Jurkat cells induced to undergo apoptosis with anti-Fas antibody, protein phosphatase 2A (PP2A) activity increased 4.5-fold after 6 h. By 12 h, the regulatory Aalpha subunit could no longer be detected in cell lysates. There was no change in the amount of the catalytic subunit. The effects on PP2A could be prevented by the caspase family inhibitors acetyl-Asp-Glu-Val-Asp (DEVD) aldehyde or Ac-DEVD fluoromethyl ketone. The mitogen-activated protein (MAP) kinase pathway is regulated by PP2A. At 12 h after the addition of anti-Fas antibody, a decrease in the amount of the phosphorylated forms of MAP kinase was observed. Again, this loss of activated MAP kinase could be prevented by the addition of DEVD-cho or DEVD-fmk. These data are consistent with a pathway whereby induction of apoptosis activates caspase-3. This enzyme then cleaves the regulatory Aalpha subunit of PP2A, increasing its activity. These data show that the activated PP2A will then effect a change in the phosphorylation state of the cell. These data provide a link between the caspases and signal transduction pathways.


Subject(s)
Apoptosis , Caspases , Cysteine Endopeptidases/metabolism , Phosphoprotein Phosphatases/metabolism , Caspase 3 , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Humans , Hydrolysis , Jurkat Cells , Oligopeptides/pharmacology , Protein Phosphatase 2 , Saccharomyces cerevisiae/genetics , Substrate Specificity , Tumor Necrosis Factor-alpha/pharmacology
4.
J Biol Chem ; 272(45): 28315-20, 1997 Nov 07.
Article in English | MEDLINE | ID: mdl-9353287

ABSTRACT

PS2, the chromosome 1 familial Alzheimer's disease gene, has been shown to be involved in programmed cell death by three complementary experimental approaches. Reduction of PS2 protein levels by antisense RNA protects from apoptosis, whereas overexpression of an Alzheimer's PS2 mutant increases cell death induced by several stimuli. In addition, ALG-3, a truncated PS2 cDNA, encodes an artificial COOH-terminal PS2 segment that dominantly inhibits apoptosis. Here we describe a physiological COOH-terminal PS2 polypeptide (PS2s, Met298-Ile448) generated by both an alternative PS2 transcript and proteolytic cleavage. We find that PS2s protects transfected cells from Fas- and tumor necrosis factor alpha (TNFalpha)-induced apoptosis. Furthermore, a similar anti-apoptotic COOH-terminal PS2 polypeptide (PS2Ccas) is generated by caspase-3 cleavage at Asp329. These results suggest that caspase-3 not only activates pro-apoptotic substrates but also generates a negative feedback signal in which PS2Ccas antagonizes the progression of cell death. Thus, whereas PS2 is required for apoptosis, PS2s and PS2Ccas oppose this process, and the balance between PS2 and these COOH-terminal fragments may dictate the cell fate.


Subject(s)
Alternative Splicing , Apoptosis , Caspases , Cysteine Endopeptidases/metabolism , Membrane Proteins/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Caspase 3 , Consensus Sequence , Enzyme Precursors/metabolism , Fas Ligand Protein , HeLa Cells , Humans , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Presenilin-2 , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
J Exp Med ; 186(8): 1323-31, 1997 Oct 20.
Article in English | MEDLINE | ID: mdl-9334372

ABSTRACT

We report that the serine protease granzyme B (GrB), which is crucial for granule-mediated cell killing, initiates apoptosis in target cells by first maturing caspase-10. In addition, GrB has a limited capacity to mature other caspases and to cause cell death independently of the caspases. Compared with other members, GrB in vitro most efficiently processes caspase-7 and -10. In a human cell model, full maturation of caspase-7 does not occur unless caspase-10 is present. Furthermore, GrB matured caspase-3 with less efficiency than caspase-7 or caspase-10. With the caspases fully inactivated by peptidic inhibitors, GrB induced in Jurkat cells growth arrest and, over a delayed time period, cell death. Thus, the primary mechanism by which GrB initiates cell death is activation of the caspases through caspase-10. However, under circumstances where caspase-10 is absent or dysfunctional, GrB can act through secondary mechanisms including activation of other caspases and direct cell killing by cleavage of noncaspase substrates. The redundant functions of GrB ensure the effectiveness of granule-mediated cell killing, even in target cells that lack the expression or function (e.g., by mutation or a viral serpin) of one or more of the caspases, providing the host with overlapping safeguards against aberrantly replicating, nonself or virally infected cells.


Subject(s)
Apoptosis/immunology , Caspases , Cytoplasmic Granules/enzymology , Cytoplasmic Granules/immunology , Cytotoxicity, Immunologic , Serine Endopeptidases/pharmacology , Apoptosis/drug effects , Breast Neoplasms , Caspase 3 , Caspase 7 , Cell Death/drug effects , Cell Death/immunology , Cysteine Endopeptidases/metabolism , Cytotoxicity, Immunologic/drug effects , Enzyme Activation/drug effects , Granzymes , Humans , Jurkat Cells , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/metabolism , Substrate Specificity , Tumor Cells, Cultured
6.
J Biol Chem ; 272(15): 9677-82, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9092497

ABSTRACT

The caspase family represents a new class of intracellular cysteine proteases with known or suspected roles in cytokine maturation and apoptosis. These enzymes display a preference for Asp in the P1 position of substrates. To clarify differences in the biological roles of the interleukin-1beta converting enzyme (ICE) family proteases, we have examined in detail the specificities beyond the P1 position of caspase-1, -2, -3, -4, -6, and -7 toward minimal length peptide substrates in vitro. We find differences and similarities between the enzymes that suggest a functional subgrouping of the family different from that based on overall sequence alignment. The primary specificities of ICE homologs explain many observed enzyme preferences for macromolecular substrates and can be used to support predictions of their natural function(s). The results also suggest the design of optimal peptidic substrates and inhibitors.


Subject(s)
Caspases , Cysteine Endopeptidases/metabolism , Caspase 1 , Caspase 3 , Caspase 6 , Caspase 7 , Chromatography, High Pressure Liquid , Cysteine Endopeptidases/chemistry , Fluorescent Dyes/metabolism , Kinetics , Oligopeptides/metabolism , Signal Transduction , Substrate Specificity
7.
Nature ; 386(6625): 619-23, 1997 Apr 10.
Article in English | MEDLINE | ID: mdl-9121587

ABSTRACT

Interferon-gamma-inducing factor (IGIF, interleukin-18) is a recently described cytokine that shares structural features with the interleukin-1 (IL-1) family of proteins and functional properties with IL-12. Like IL-12, IGIF is a potent inducer of interferon (IFN)-gamma from T cells and natural killer cells. IGIF is synthesized as a biologically inactive precursor molecule (proIGIF). The cellular production of IL-1beta, a cytokine implicated in a variety of inflammatory diseases, requires cleavage of its precursor (proIL-1beta) at an Asp-X site by interleukin-1beta-converting enzyme (ICE, recently termed caspase-1). The Asp-X sequence at the putative processing site in proIGIF suggests that a protease such as caspase-1 might be involved in the maturation of IGIF. Here we demonstrate that caspase-1 processes proIGIF and proIL-1beta with equivalent efficiencies in vitro. A selective caspase-1 inhibitor blocks both lipopolysaccharide-induced IL-1beta and IFN-gamma production from human mononuclear cells. Furthermore, caspase-1-deficient mice are defective in lipopolysaccharide-induced IFN-gamma production. Our results thus implicate caspase-1 in the physiological production of IGIF and demonstrate that it plays a critical role in the regulation of multiple proinflammatory cytokines. Specific caspase-1 inhibitors would provide a new class of anti-inflammatory drugs with multipotent action.


Subject(s)
Cysteine Endopeptidases/metabolism , Cytokines/metabolism , Interferon Inducers/metabolism , Interferon-gamma/biosynthesis , Protein Processing, Post-Translational , Animals , Binding Sites , COS Cells , Caspase 1 , Cloning, Molecular , Cysteine Endopeptidases/genetics , Cysteine Proteinase Inhibitors/pharmacology , Humans , Interferon-gamma/blood , Interferon-gamma/genetics , Interleukin-1/blood , Interleukin-12/blood , Interleukin-18 , Interleukin-6/blood , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Lipopolysaccharides/pharmacology , Mice , Mutagenesis, Site-Directed , Oligopeptides/pharmacology , Protein Precursors/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
Apoptosis ; 2(3): 289-303, 1997.
Article in English | MEDLINE | ID: mdl-14646542

ABSTRACT

CTLL cells undergo apoptosis when cultured in the absence of IL-2. The IL-1beta-converting-enzyme (ICE)/ caspase family has been implicated as an integral component of some forms of apoptosis. Numerous members of the caspase family have been identified, and it appears as if caspase-3/CPP32 plays a critical role. Previously we demonstrated that ICE/caspase-1 expression increases in CTLL cells during apoptosis; however, inhibition of ICE activity did not abrogate apoptotic death. The purpose of this report is to determine if other members of the caspase family are involved in T cell apoptosis induced by growth factor starvation. We show that cytosolic CPP32-like activity, as measured by the cleavage of DEVD-pNA and poly(ADP-ribose) polymerase (PARP), increases during apoptosis following growth factor deprivation. Cytosolic CPP32-like activity is inhibited in cells treated with the broad spectrum ICE family inhibitor boc-aspartyl(OMe)-fluoromethylketone (D-FMK) and by VAD-FMK and DEVD-FMK which have greater specificity for CPP32-like ICE homologs; however, only the broad spectrum ICE inhibitor D-FMK inhibited apoptosis. Our results suggest that apoptosis induced by growth factor deprivation involves the caspase family, but increased CPP32-like activity is not sufficient to mediate apoptosis induced by IL-2 starvation.

9.
J Exp Med ; 184(6): 2399-404, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8976194

ABSTRACT

Recent studies have shown that protein kinase C (PKC) delta is proteolytically activated at the onset of apoptosis induced by DNA-damaging agents, tumor necrosis factor, and anti-Fas antibody. However, the relationship of PKC delta cleavage to induction of apoptosis is unknown. The present studies demonstrate that full-length PKC delta is cleaved at DMQD330N to a catalytically active fragment by the cysteine protease CPP32. The results also demonstrate that overexpression of the catalytic kinase fragment in cells is associated with chromatin condensation, nuclear fragmentation, induction of sub-G1 phase DNA and lethality. By contrast, overexpression of full-length PKC delta or a kinase inactive PKC delta fragment had no detectable effect. The findings suggest that proteolytic activation of PKC delta by a CPP32-like protease contributes to phenotypic changes associated with apoptosis.


Subject(s)
Apoptosis , Caspases , Cysteine Endopeptidases/metabolism , Helminth Proteins/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Caenorhabditis elegans Proteins , Caspase 1 , Caspase 3 , Enzyme Activation , HeLa Cells , Humans , Isoenzymes/chemistry , Kinetics , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Polymerase Chain Reaction , Protein Kinase C/chemistry , Protein Kinase C-delta , Recombinant Proteins/metabolism , Substrate Specificity , Transfection
10.
J Biol Chem ; 271(7): 3517-22, 1996 Feb 16.
Article in English | MEDLINE | ID: mdl-8631956

ABSTRACT

The Caenorhabditis elegans cell death gene, ced-3, encodes one of the two proteins required for apoptosis in this organism. The primary sequence similarities between Ced-3 and the mammalian interleukin-1beta converting enzyme (ICE) suggest that these two proteins may have functionally similar active sites and that Ced-3 may function as a cysteine protease. Here we report that in vitro transcribed and translated Ced-3 protein (p56) underwent rapid processing to smaller fragments. Replacement of the predicted active site cysteine of Ced-3 with serine (C364S) prevented the generation of smaller proteolytic fragments, suggesting that the processing might be an autocatalytic process. Peptide aldehydes with aspartic acid at the P1 position blocked Ced-3 autocatalysis. Furthermore, the protease inhibition profile of Ced-3 was similar to the profile reported for ICE. These functional data demonstrate that Ced-3 is an Asp-dependent cysteine protease with substrate specificity similar to that of ICE. Aurintricarboxylic acid, an inhibitor of apoptosis in mammalian cells, blocked Ced-3 autocatalytic activity, suggesting that an aurintricarboxylic acid-sensitive Ced-3/ICE-related protease might be involved in the apoptosis pathway(s) in mammalian cells.


Subject(s)
Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Caspases , Cysteine Endopeptidases/metabolism , Helminth Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis , Aurintricarboxylic Acid/pharmacology , Base Sequence , Binding Sites , Caenorhabditis elegans Proteins , Caspase 1 , Cysteine , Cysteine Endopeptidases/chemistry , DNA Primers , Helminth Proteins/biosynthesis , Helminth Proteins/chemistry , Kinetics , Mammals , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Polymerase Chain Reaction , Protein Biosynthesis , Recombinant Proteins/metabolism , Serine , Transcription, Genetic
11.
EMBO J ; 14(24): 6148-56, 1995 Dec 15.
Article in English | MEDLINE | ID: mdl-8557034

ABSTRACT

These studies demonstrate that treatment of human U-937 cells with ionizing radiation (IR) is associated with activation of a cytoplasmic myelin basic protein (MBP) kinase. Characterization of the kinase by gel filtration and in-gel kinase assays support activation of a 40 kDa protein. Substrate and inhibitor studies further support the induction of protein kinase C (PKC)-like activity. The results of N-terminal amino acid sequencing of the purified protein demonstrate identity of the kinase with an internal region of PKC delta. Immunoblot analysis was used to confirm proteolytic cleavage of intact 78 kDa PKC delta in control cells to the 40 kDa C-terminal fragment after IR exposure. The finding that both IR-induced proteolytic activation of PKC delta and endonucleolytic DNA fragmentation are blocked by Bcl-2 and Bcl-xL supports an association with physiological cell death (PCD). Moreover, cleavage of PKC delta occurs adjacent to aspartic acid at a site (QDN) similar to that involved in proteolytic activation of interleukin-1 beta converting enzyme (ICE). The specific tetrapeptide ICE inhibitor (YVAD) blocked both proteolytic activation of PKC delta and internucleosomal DNA fragmentation in IR-treated cells. These findings demonstrate that PCD is associated with proteolytic activation of PKC delta by an ICE-like protease.


Subject(s)
Apoptosis/physiology , Cysteine Endopeptidases/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Apoptosis/genetics , Apoptosis/radiation effects , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspase 1 , Cell Line , Cysteine Endopeptidases/genetics , DNA Damage , Enzyme Activation/radiation effects , Glycogen Synthase Kinase 3 , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Molecular Sequence Data , Peptides/chemistry , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Kinase C-delta , Sequence Homology, Amino Acid , Signal Transduction , Substrate Specificity
12.
J Immunol ; 155(7): 3433-42, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7561038

ABSTRACT

Clonal T cells undergo programmed cell death (PCD) or apoptosis when cultured without the appropriate cytokines. The cysteine protease, IL-1 beta converting enzyme (ICE), is implicated in apoptosis based on its structural similarity to the PCD gene, ced-3, in Caenorhabditis elegans and the induction of PCD in fibroblasts transfected with recombinant ICE. We show that the murine IL-2-dependent CTLL T cell line expresses ICE but not IL-1 beta. Interestingly, ICE mRNA and protein levels increase during apoptosis. Yet inhibition of ICE enzymatic activity (> 90%) with either of two cell-permeable ICE inhibitors does not abrogate or delay apoptosis following IL-2 deprivation, as measured by DNA fragmentation and viability. Our results suggest that ICE is not required for apoptosis in lymphokine-deprived T cells.


Subject(s)
Apoptosis/drug effects , Cysteine Endopeptidases/metabolism , Interleukin-2/metabolism , T-Lymphocytes/physiology , Amino Acid Sequence , Animals , Caspase 1 , Cell Line , Cysteine Endopeptidases/chemistry , DNA/analysis , Interleukin-1/metabolism , Mice , Molecular Sequence Data
13.
Science ; 269(5232): 1885-8, 1995 Sep 29.
Article in English | MEDLINE | ID: mdl-7569933

ABSTRACT

The baculovirus antiapoptotic protein p35 inhibited the proteolytic activity of human interleukin-1 beta converting enzyme (ICE) and three of its homologs in enzymatic assays. Coexpression of p35 prevented the autoproteolytic activation of ICE from its precursor form and blocked ICE-induced apoptosis. Inhibition of enzymatic activity correlated with the cleavage of p35 and the formation of a stable ICE-p35 complex. The ability of p35 to block apoptosis in different pathways and in distantly related organisms suggests a central and conserved role for ICE-like proteases in the induction of apoptosis.


Subject(s)
Apoptosis , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Caspase 1 , Cell Line , Cysteine Proteinase Inhibitors/genetics , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation/drug effects , Humans , Inhibitor of Apoptosis Proteins , Molecular Sequence Data , Recombinant Proteins/pharmacology , Transfection , Viral Proteins/genetics , Viral Proteins/pharmacology
14.
Blood ; 84(11): 3965-73, 1994 Dec 01.
Article in English | MEDLINE | ID: mdl-7949152

ABSTRACT

The nature of the effector cell(s) responsible for the depression of B-cell genesis in the bone marrow of mice undergoing systemic graft-versus-host disease (GVHD) has been examined. Donor C57BL/6 (B6) mice were treated in vivo with either a single injection of anti-asialo GM1 antibody (anti-ASGM1) to eliminate naturally occurring (endogenous) ASGM1+ cells or B6xAF1 (B6AF1) lymphoid cells followed by anti-ASGM1 to eliminate both endogenous and "induced" ASGM1+ cells. Lymphoid cells from donor mice after the elimination of endogenous ASGM1+ cells produced severe GVHD and concomitant depression of B-cell genesis when injected into B6AF1 recipients. In contrast, cells from donors depleted of both the endogenous and inducible ASGM1+ populations did not cause GVHD or depletion of B lineage cells in B6AF1 recipients but did depress B-cell genesis in B6C3F1 mice. The "induced" ASGM1+ cells were Thy 1+, but their elimination did not significantly alter either overall T-cell function or specific cytotoxic T-cell (CTL) reactivity against the sensitizing (B6AF1) strain. The results suggest that the effector cell responsible for the depression of B-cell genesis during systemic GVHD can be induced to express ASGM1, is strain-specific and Thy 1+; but is not a conventional CTL.


Subject(s)
B-Lymphocytes , Bone Marrow Transplantation/pathology , G(M1) Ganglioside/analysis , Graft vs Host Disease/pathology , Hematopoiesis , Killer Cells, Natural/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Female , G(M1) Ganglioside/antagonists & inhibitors , G(M1) Ganglioside/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/transplantation , Lymph Nodes/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Spleen/pathology , T-Lymphocyte Subsets/immunology
15.
Proc Natl Acad Sci U S A ; 91(24): 11393-7, 1994 Nov 22.
Article in English | MEDLINE | ID: mdl-7972071

ABSTRACT

Tumor necrosis factor alpha (TNF-alpha) is a cytokine that is responsible, in part, for several aspects of the acute-phase response to inflammation, including the generation of fever. TNF-alpha has direct effects on central nervous system neurons deep within the hypothalamus that are involved in producing the febrile response, but the blood-brain barrier prevents circulating TNF-alpha from having access to these sites. We therefore have hypothesized that TNF-alpha may be produced in the brain and used as a mediator in the cerebral components of the acute-phase response. We used in situ hybridization to determine the distribution of production of TNF-alpha mRNA in the mouse brain after systemic administration of lipopolysaccharide. During the initial phase of fever, hybridization was observed in perivascular cells and neurons in circumventricular organs, including the vascular organ of the lamina terminalis, median eminence, and area postrema, as well as along the ventral surface of the medulla; hybridization was also prominent over many cell in the meninges. During the late phase of the response, hybridization was observed over neurons in the pericircumventricular nuclei such as the anteroventral periventricular and arcuate nuclei of the hypothalamus and the nucleus of the solitary tract. TNF-alpha produced by a cascade of neurons within the brain may participate in the complex autonomic, neuroendocrine, metabolic, and behavioral responses to infection and inflammation.


Subject(s)
Acute-Phase Reaction , Brain/metabolism , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Base Sequence , DNA Primers/chemistry , Gene Expression , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA, Messenger/genetics , Salmonella typhimurium , Tumor Necrosis Factor-alpha/genetics
16.
Cell ; 78(2): 343-52, 1994 Jul 29.
Article in English | MEDLINE | ID: mdl-8044845

ABSTRACT

Interleukin-1 beta-converting enzyme (ICE) proteolytically cleaves pro-IL-1 beta to its mature, active form. The crystal structure at 2.5 A resolution of a recombinant human ICE-tetrapeptide chloromethylketone complex reveals that the holoenzyme is a homodimer of catalytic domains, each of which contains a p20 and a p10 subunit. The spatial separation of the C-terminus of p20 and the N-terminus of p10 in each domain suggests two alternative pathways of assembly and activation in vivo. ICE is homologous to the C. elegans cell death gene product, CED-3, and these may represent a novel class of cytoplasmic cysteine proteases that are important in programmed cell death (apoptosis). Conservation among members of the ICE/CED-3 family of the amino acids that form the active site region of ICE supports the hypothesis that they share functional similarities.


Subject(s)
Protein Conformation , Serpins/chemistry , Viral Proteins , Amino Acid Chloromethyl Ketones/metabolism , Amino Acid Sequence , Apoptosis , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serpins/genetics , Serpins/metabolism
17.
Scand J Immunol ; 34(4): 497-508, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1833814

ABSTRACT

In previous studies we demonstrated that an induced asialo-GM1 positive (ASGM1+) cell of donor origin that exerts natural killer cell-like activity (NK activity+) plays a crucial role in the development of graft-versus-host (GVH)-associated tissue damage and severe immunosuppression. This study examined whether the ASGM1+ (NK activity+) GVH effector cells were activated by non-specific signals or whether these cells were triggered by specific alloantigens and displayed antigenic specificity. C57B1/6 (B6) donor mice were treated with either B6 x AF1 (B6AF1) lymphoid cells and anti-asialo GM1 antibodies (anti-ASGM1) to induce and eliminate specifically activated B6-anti-B6AF1 ASGM1+ (NK activity+) cells or with polyinosinic: polycytidylic acid (poly I:C), and anti-ASGM1 to eliminate non-specifically activated ASGM1+ (NK activity+) cells. Donor spleen and lymph node cells depleted of the specific allo-induced ASGM1+ NK reactive cells showed near normal numbers of L3T4+ and Lyt-2+ cells and retained T- and B-cell functions as measured by mitogen responses (to PHA, Con A and LPS), mixed lymphocyte responses (MLR) (to B6AF1) and the generation of cytotoxic T cells (CTL) (to B6AF1 blasts). Anti-ASGM1 treatment almost completely abrogated NK activity in all donor inocula. GVH reactions were induced by injecting treated donor cells into B6AF1, B6 x C3HejF1 (B6C3HF1) and B6 x SJLF1 (B6SJLF1) hybrids and monitored by splenomegaly, suppression of T-cell mitogen responses and the development of histopathological lesions in the thymus, liver and pancreas. Cells from donors depleted of non-specifically (poly I:C) induced ASGM1+ cells induced severe histological lesions, marked immunosuppression and splenomegaly in all three F1 hybrid combinations. When the donor cells were depleted of specifically induced (B6-anti-B6AF1) ASGM1+ cells and injected into the three F1 combinations they induced splenomegaly in all three but caused severe tissue injury and intense immunosuppression only in B6C3HF1 and B6SJLF1 mice and not in B6AF1 mice. Genetic analysis suggests that the H-2D (or a closely related) region of the H-2 complex plays an important role in the activation of the specific GVH effector cells. These results suggest that the cell(s) responsible for splenomegaly are different from the ones that cause severe GVH-associated tissue damage and immunosuppression although there may be cells and/or lymphokines common to both processes.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
G(M1) Ganglioside , Glycosphingolipids/biosynthesis , Graft vs Host Disease/immunology , Killer Cells, Natural/immunology , Animals , Antibody Specificity , Cell Division/drug effects , Cytotoxicity, Immunologic , Female , Flow Cytometry , Fluorescent Antibody Technique , Glycosphingolipids/immunology , Immune Tolerance , Lipopolysaccharides , Lymph Nodes/immunology , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred C57BL , Poly I-C/pharmacology , Spleen/immunology
18.
J Immunol ; 146(4): 1130-3, 1991 Feb 15.
Article in English | MEDLINE | ID: mdl-1825104

ABSTRACT

The injection of mature T cells into a tolerant or immunocompromised allogeneic host animal produces a graft versus host response (GVHR) that can result in splenomegaly, immunosuppression and death of the host animal. We demonstrate here that lymphocytes from T cell receptor beta-chain (TCR-beta) transgenic mice, in which the expression of the transgene inhibits endogenous beta- and gamma-gene rearrangements and thus causes abnormal T cell development, are unable to mediate a GVHR. The GVHR was measured after the injection of lymphocytes from transgenic mice into normal F1 mice and also after transplantation of bone marrow and lymphocytes from transgenic mice into lethally irradiated F1 recipients. In both systems, cells from transgenic mice failed to produce a significant GVHR. Cells from the transgenic mice were able to recognize the foreign histocompatibility Ag of the host in vitro and in vivo although the transgenic mice rejected skin grafts more slowly than controls. Thus, lymphocytes from transgenic mice were unable to produce a GVHR despite the presence of alloreactive T cells. These results suggest that lymphocytes from TCR-beta transgenic mice fail to mediate a GVHR either because lymphocytes with a single transgenic TCR-beta chain have a limited ability to recognize allogeneic cells in vivo or because the transgenic mice lack lymphocyte subsets that are important for the mediation of a GVHR.


Subject(s)
Graft vs Host Reaction/immunology , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , Animals , Female , Histocompatibility Antigens/immunology , Isoantigens , Male , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocytes/transplantation , Whole-Body Irradiation
19.
Transplantation ; 49(5): 882-6, 1990 May.
Article in English | MEDLINE | ID: mdl-2186521

ABSTRACT

The LFA-1 molecule is a member of the leukocyte adhesion complex (CD11/CD18) that is critical to adhesion and effector function of T cells and NK cells. Both T cells and NK cells play important roles in bone marrow graft rejection, and anti-LFA-1 MAbs inhibit in vitro T cell and NK cell cytotoxicity. We here describe the in vivo use of a monoclonal antibody to the alpha chain of LFA-1 (CD11a) to prevent rejection of T cell-depleted allogeneic bone marrow. Animals receiving anti-LFA-1 and 1100 cGy total-body irradiation showed improved hemopoietic and immunologic reconstitution one month after BMT when compared with animals receiving TBI alone. Animals in the anti-LFA-1 group showed significantly improved long-term survival, and had no deficits in long-term lymphohemopoietic reconstitution.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Differentiation/immunology , Bone Marrow Transplantation/immunology , Graft Survival , Receptors, Leukocyte-Adhesion/immunology , Animals , Cell Separation , Hematopoiesis , Immunity , Lymphocyte Function-Associated Antigen-1 , Mice , Mice, Inbred Strains , T-Lymphocytes/immunology , Time Factors
20.
Am J Pathol ; 133(1): 39-46, 1988 Oct.
Article in English | MEDLINE | ID: mdl-3263049

ABSTRACT

This study presents the sequential morphologic regeneration of graft-vs.-host (GVH)-induced dysplastic thymuses in long-term survivors of GVH reactions. GVH reactions were induced in adult C57BL/6xAF1 (B6AF1) hybrids by injecting 20 x 10(6) A strain parental lymphoid cells (PLC). Starting on day 30 after GVH induction, five to ten animals were randomly selected from a pool of GVH-reactive mice and killed at various times. Each animal was tested for thymic histology and T cell functions. Thymuses taken on day 30 after GVH induction displayed severe dysplasia as characterized by lymphocytic depletion, complete effacement of cortico-medullary demarcation, and reduction and total loss of medullary epithelial cells or both. Starting by days 60-70 after GVH induction, at least four stages of thymic regeneration were identified. Day 60-70 thymuses displayed cortical regeneration and the reappearance of cortico-medullary demarcation. The medulla of these thymuses, although containing dark individual epithelial cells and numerous lymphocytes, was devoid of pale epithelial cells (stage 1). The medulla of thymuses on day 100 after GVH induction displayed a few sparcely distributed pale epithelial cells and numerous lymphocytes as well as dark epithelial cells (stage 2). The medulla of thymuses examined 130 days after GVH induction displayed numerous pale individual epithelial cells and a few pale epithelial cell clusters. Such thymuses also showed a reduction in the number of medullary lymphocytes (stage 3). Finally, the medulla of thymuses 150-160 days after GVH induction displayed numerous pale epithelial cell clusters and Hassall's bodies. These thymuses were indistinguishable from normal adult thymuses (stage 4). All of the animals tested up to day 130 after GVH induction showed no significant T cell function. Animals displaying stage 4 of thymic regeneration showed significant proliferative responses to T cell mitogen, concanavalin A (conA), and six of ten animals also displayed a few plaque forming cells (PFC) to sheep red blood cells (SRBC) in their spleens. Furthermore, all animals (10 of 10) killed on day 180 after GVH induction displayed significant T cell functions.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Graft vs Host Disease/physiopathology , Regeneration , Thymus Gland/physiopathology , Animals , Epithelium/pathology , Graft vs Host Disease/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...