Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 106(1): 117-24, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10880055

ABSTRACT

The continuous, in vivo infusion of low-dose IL-2 selectively expands the absolute number of human natural killer (NK) cells after 4-6 weeks of therapy. The mechanism responsible for this expansion is unknown and was examined in this study. NK cells cultured at low concentrations of IL-2, comparable to those found during in vivo therapy, proliferate for 6 days and then exit the cell cycle. However, NK cells in vivo did not traverse the S/G(2)/M phase of the cell cycle during low-dose IL-2 therapy. Low concentrations of IL-2 delay programmed cell death of NK cells but have the same effect on resting T cells that do not expand in vivo. When CD34(+) bone marrow hematopoietic progenitor cells are cultured for 21 days with low concentrations of IL-2, they differentiate into CD56(+)CD3(-) NK cells, not T cells. Thus, the selective expansion of human NK cells during continuous in vivo infusion of low-dose IL-2 likely results from enhanced NK-cell differentiation from bone marrow progenitors, combined with an IL-2-dependent delay in NK-cell death, rather than proliferation of mature NK cells in the periphery.


Subject(s)
Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Animals , Antigens, CD34/analysis , CD56 Antigen/analysis , Humans , Interleukin-2/therapeutic use , Killer Cells, Natural/physiology , Lymphocyte Activation/drug effects , Rabbits , Recombinant Proteins/pharmacology
2.
Radiat Res ; 152(5): 487-92, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10521925

ABSTRACT

We have investigated the mechanisms by which radiation inhibits proliferation of human breast cancer cells in culture. Radiation, within the dose range used for treatment of humans, decreased the rate of proliferation of estrogen-independent MDA-MB-231 cells more effectively than it did that of estrogen-dependent MCF-7 cells. The rate of proliferation of MDA-MB-231 cells was also inhibited to a greater extent than that of MCF-7 cells by purified TGFB1. Using an ELISA specific for activated TGFB1, we found that conditioned medium from irradiated MDA-MB-231 or MCF-7 cells contained twofold more TGFB1 than that from nonirradiated cells. Conditioned medium from irradiated breast cancer cells, but not from nonirradiated cells, inhibited the growth of untreated MDA-MB-231 cells. The inhibitory activity was blocked by an anti-TGFB1 neutralizing antibody. An approximately twofold increase in the TGFB1 mRNA in irradiated cells compared to controls was found using semiquantitative reverse-transcriptase PCR. Last, the mRNA for insulin-like growth factor binding protein 3, a reported target of the cell inhibitory activity of TGFB1, was increased threefold upon irradiation. Our results demonstrate that the TGFB1 is increased after irradiation and that the activation of the TGFB1 signaling pathway may sensitize cells to the effects of radiation.


Subject(s)
Breast Neoplasms/pathology , Transforming Growth Factor beta/radiation effects , Base Sequence , Cell Division/radiation effects , DNA Primers , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
3.
Blood ; 88(10): 3887-93, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8916954

ABSTRACT

The allogeneic mixed lymphocyte reaction (MLR) is a complex in vitro assay of T-cell recognition and responsiveness in which interleukin-2 (IL-2) plays a central role. We have previously demonstrated that c-kit ligand (KL) can enhance IL-2-induced proliferation in a subset of human natural killer cells expressing the c-kit tyrosine kinase receptor. In the present study, we asked whether KL could enhance IL-2-mediated T-cell proliferation in the allogeneic MLR. We demonstrate that the vast majority of activated human T-cell clones express the c-kit mRNA transcript. Binding studies performed on activated T cells with radioiodinated KL were consistent with the expression of a single class of c-kit receptors. The addition of exogenous KL to the MLR led to an increase in tritiated thymidine (3[H]-TdR) incorporation in the absence of other exogenous cytokines, and did so in a dose-dependent fashion. A reproducible increase in 3[H]-TdR incorporation was noted at concentrations of KL, which approximate those normally found in vivo. Antibody blocking of KL binding to c-kit, T-cell depletion and sorting experiments suggest that the action of KL is mediated at least in part by a direct effect on both CD4+ and CD8+ T-cells. KL's enhancement of the MLR also requires the binding of IL-2 to its high-affinity IL-2 receptor. Given the abundance of KL normally found in human serum, these data suggest that this cytokine may have a role during T-cell activation in vivo.


Subject(s)
Lymphocyte Culture Test, Mixed , Stem Cell Factor/pharmacology , T-Lymphocyte Subsets/immunology , Animals , DNA Replication , Histocompatibility , Humans , Immunity, Cellular , Interleukin-2/physiology , Isoantigens/immunology , Lymphocyte Activation , Mice , Proto-Oncogene Proteins c-kit/biosynthesis , Proto-Oncogene Proteins c-kit/classification , RNA, Messenger/biosynthesis , Receptors, Interleukin-2/drug effects , Receptors, Interleukin-2/physiology , Stem Cell Factor/biosynthesis , Stem Cell Factor/genetics , T-Lymphocyte Subsets/metabolism
4.
J Clin Invest ; 97(12): 2722-7, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8675682

ABSTRACT

Once infected by obligate intracellular pathogens, monocytes/macrophages release cytokines that activate natural killer (NK) cells. NK cells in turn produce and secrete monocyte/macrophage activating factors such as interferongamma (IFN-gamma), which are important in the early control of these infections. Here we demonstrate that human NK cells are potent producers of another monocyte/macrophage-activating factor, macrophage inflammatory protein-1 alpha (MIP-1 alpha). Fresh NK cells produce negligible amounts of MIP-1 alpha after stimulation with the monocyte-derived cytokines IL-12, TNF-alpha, IL-1 beta, or IL-10, while stimulation with IL-15 alone results in modest MIP-1 alpha production. Abundant NK cell production MIP-1 alpha is seen after costimulation with IL-12 and IL-15, and is dose-dependent. Combinations of IL-12, with TNF-alpha, IL-1 beta, or IL-10 are substantially less effective inducers of MIP-1 alpha production by NK cells. NK cell MIP-1 alpha mRNA transcripts were detectable within 1 h after costimulation with IL-12 plus IL-15 and steadily increased over 24 h, with a concomitant increase in protein production detectable at 12 h. Resting NK cells constitutively express mRNA transcript for a MIP-1 alpha receptor, and costimulation with IL-12 and IL-15 upregulates its level of expression. Equilibrium binding studies with radioiodinated MIP-1 alpha were consistent with the induction of a single class of high affinity MIP-1 alpha receptors on NK cells costimulated with IL-12 and IL-15. Addition of exogenous MIP-1 alpha to resting NK cells did not enhance cytokine production, but did increase NK cytotoxic activity. The requirement for IL-15 as a critical cofactor for NK cell production MIP-1 alpha suggests a potentially unique role for this monocyte-derived cytokine in combination with IL-12. As MIP-1 alpha is known to potentiate the action of IFN-gamma on monocytes and to suppress human immunodeficiency virus replication, the NK cell's production of MIP-1 alpha may be important during the innate immune response to infection.


Subject(s)
Cytokines/pharmacology , Killer Cells, Natural/drug effects , Monokines/biosynthesis , Cell Line , Chemokine CCL4 , Humans , Interleukin-1/pharmacology , Interleukin-15 , Interleukin-2/pharmacology , Interleukins/pharmacology , Killer Cells, Natural/metabolism , Macrophage Inflammatory Proteins , Monokines/genetics , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/pharmacology
5.
Blood ; 86(9): 3287-94, 1995 Nov 01.
Article in English | MEDLINE | ID: mdl-7579429

ABSTRACT

Ten adult patients with human immunodeficiency virus (HIV)-associated malignancies (five with lymphoma and five with Kaposi's Sarcoma) were treated with a daily subcutaneous injection of interleukin-2 (IL-2) for 90 consecutive days in a phase I dose-escalation study. Seven patients had absolute CD4 counts below 200/mm3 at the time malignancy was diagnosed. Each lymphoma patient had obtained a complete or partial remission with standard chemotherapy before initiating IL-2. The daily dose of IL-2 did not change during the 90-day course of therapy. Seventeen courses of IL-2 therapy were completed at doses ranging from 0.4 x 10(6) U/m2/d to 1.2 x 10(6) U/m2/d without significant (grade III) toxicity. Two of two patients experienced grade III toxicity within 21 days of initiating IL-2 at a dose of 1.4 x 10(6) U/m2/d, but both patients subsequently completed 90 days of therapy at the maximum tolerated dose (MTD) of 1.2 x 10(6) U/m2/d. Although there were no significant increases or decreases in T-cell subsets at any dose level, there was an increase in absolute natural killer (NK) cell number at the three highest doses of IL-2 (mean percent increase 247; 95% confidence interval, 124 to 369) that was statistically significant (Wilcoxon one-sample signed rank test, P = .015). One patient developed an anti-IL-2 antibody titer that correlated with minimal NK cell expansion in vitro and in vivo. An increase in eosinophils was noted during 9 of 17 courses of IL-2 therapy without correlation to IL-2 dose, prior course of IL-2, or NK cell expansion. At the MTD, there was no consistent increase in the plasma HIV RNA level over time. Three of 10 patients had progressive disease while on study. During 50 months of IL-2 therapy, no patient was treated for an opportunistic infection. We conclude that daily low dose subcutaneous IL-2 can be self-administered safely with good compliance for prolonged periods of time to patients with HIV-associated malignancies, including those with profound immune deficiency. The majority of patients show selective expansion of innate immune effectors, ie, NK cells and/or eosinophils, in the absence of significant clinical toxicity or increased viral burden. These results suggest that low-dose IL-2 therapy should be studied further in phase II clinical trials for evidence of activity against malignancy and opportunistic infection in this patient population.


Subject(s)
HIV Infections/complications , Immunologic Factors/therapeutic use , Immunotherapy , Interleukin-2/therapeutic use , Lymphoma, AIDS-Related/therapy , Sarcoma, Kaposi/therapy , Adult , Drug Administration Schedule , Eosinophils/immunology , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Injections, Subcutaneous , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Killer Cells, Natural/immunology , Lymphoma, AIDS-Related/immunology , Male , Middle Aged , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Sarcoma, Kaposi/etiology , Sarcoma, Kaposi/immunology , Treatment Outcome
6.
J Biol Chem ; 270(14): 8298-310, 1995 Apr 07.
Article in English | MEDLINE | ID: mdl-7713938

ABSTRACT

The gene regulatory functions of the human IL-2 receptor (IL-2R) were reconstituted in transiently transfected hepatoma cells. The combination of IL-2R beta and -gamma mediated a strong stimulation via the cytokine response element of the alpha 1-acid glycoprotein gene and the hematopoietin receptor response element, but none via the IL-6 response element or the sis-inducible element. IL-2R alpha enhanced 10-fold the sensitivity of the IL-2R beta.gamma complex to respond to IL-2 or IL-15, but did not modify the specificity or the magnitude of maximal gene regulation. A homodimerizing chimeric receptor G-CSFR-IL-2R beta could mimic the IL-2R action. The IL-2R-mediated gene regulation was similar to that seen with receptors for IL-4 and IL-7, but differed from that for IL-6 type cytokines, thrombopoietin, erythropoietin, and growth hormone. The activation of STAT proteins by the IL-2R was assessed in transfected L-cells and COS-1 cells. Although IL-2R subunits were highly expressed in these cells, no STAT protein activation was detectable. Transient overexpression of JAK3 was unable to change the signaling specificity of the hematopoietin receptors in rat hepatoma, L-, and COS cells, but established a prominent activation of the IL-6 response elements by the IL-2R and IL-4R in HepG2 cells. The data support the model that the IL-2R and related hematopoietin receptors produce at least two separate signals which control gene expression.


Subject(s)
Liver/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Receptors, Interleukin-2/metabolism , Signal Transduction , Animals , Base Sequence , Cell Line , Cytoplasm/metabolism , DNA Primers , Humans , Interleukin-6/metabolism , Liver/cytology , Mice , Molecular Sequence Data , Rats , Receptors, Interleukin-2/chemistry , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...