Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 12(1): 290, 2019 01.
Article in English | MEDLINE | ID: mdl-30504789

ABSTRACT

The original version of this Article omitted the author Dr Mathias Chamaillard from the l'Institut de Pasteur, Lille, France. This has been corrected in both the PDF and HTML versions of the Article.

2.
Parasite Immunol ; 40(2)2018 02.
Article in English | MEDLINE | ID: mdl-29117626

ABSTRACT

Cerebral malaria (CM) is one complication of Plasmodium parasite infection that can lead to strong inflammatory immune responses in the central nervous system (CNS), accompanied by lung inflammation and anaemia. Here, we focus on the role of the innate immune response in experimental cerebral malaria (ECM) caused by blood-stage murine Plasmodium berghei ANKA infection. While T cells are important for ECM pathogenesis, the role of innate lymphoid cells (ILCs) is only emerging. The role of ILCs and non-lymphoid cells, such as neutrophils and platelets, contributing to the host immune response and leading to ECM and human cerebral malaria (HCM) is reviewed.


Subject(s)
Cytokines/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Animals , Humans , Killer Cells, Natural/immunology , Macrophages/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Protein Kinase C-theta/immunology
3.
Trends Microbiol ; 25(11): 906-918, 2017 11.
Article in English | MEDLINE | ID: mdl-28625530

ABSTRACT

The STING (Stimulator of Interferon Genes) protein connects microorganism cytosolic sensing with effector functions of the host cell by sensing directly cyclic dinucleotides (CDNs), originating from pathogens or from the host upon DNA recognition. Although STING activation favors effective immune responses against viral infections, its role during bacterial diseases is controversial, ranging from protective to detrimental effects for the host. In this review, we summarize important features of the STING activation pathway and recent highlights about the role of STING in bacterial infections by Chlamydia, Listeria, Francisella, Brucella, Shigella, Salmonella, Streptococcus, and Neisseria genera, with a special focus on mycobacteria.


Subject(s)
Bacteria/genetics , Bacterial Infections/immunology , Bacterial Infections/microbiology , Membrane Proteins/immunology , Animals , Cytosol/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Membrane Proteins/genetics , Mice , Mycobacterium/immunology , Mycobacterium Infections/immunology
4.
Tuberculosis (Edinb) ; 88(3): 221-34, 2008 May.
Article in English | MEDLINE | ID: mdl-17950671

ABSTRACT

TNF is required for protection against virulent and non-virulent mycobacterial infections. Here we compared the effect of Tm-TNF and sTNF, two different molecular forms of TNF, in virulent and non-virulent murine challenge models. Using non-virulent Mycobacterium bovis BCG intranasal infection we established that immunity is durably compromised in Tm-TNF mice, with augmented bacilli burden, leading to chronic but non-lethal infection. Acute infection by a virulent Mycobacterium tuberculosis low-dose aerosol challenge was controlled in Tm-TNF mice with bacilli burdens equivalent to that in WT mice and pulmonary pathology characterised by the formation of well-defined, bactericidal granulomas. Protective immunity was however compromised in Tm-TNF mice during the chronic phase of M. tuberculosis infection, with increased lung bacterial growth and inflammatory cell activation, dissolution of granulomas associated with dispersed iNOS expression, increased pulmonary IFNgamma and IL-10 expression but decreased IL-12 production, followed by death. In conclusion, membrane TNF is sufficient to control non-virulent, M. bovis BCG infection, and acute but not chronic infection with virulent M. tuberculosis.


Subject(s)
Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/prevention & control , Tumor Necrosis Factor-alpha/immunology , Acute Disease , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Cytokines/biosynthesis , Disease Susceptibility , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mycobacterium bovis/growth & development , Mycobacterium bovis/isolation & purification , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/isolation & purification , Nitric Oxide Synthase Type II/biosynthesis , Tuberculoma/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology , Virulence
5.
Biochem Soc Trans ; 35(Pt 6): 1449-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18031243

ABSTRACT

A role for PRRs (pattern-recognition receptors) in immune cell function is now well established. In macrophages and other immune cells, activation of TLRs (Toll-like receptors) and cytosolic NLRs [NOD (nucleotide oligomerization domain) proteins containing a leucine-rich repeat] results in the induction of genes and release of imunoregulator hormones including cytokines and NO (nitric oxide). In addition to immune cells, structural cells of the cardiovascular system including endothelial cells, vascular smooth muscle and cardiac myocytes express functional PRRs and sense PAMPs (pathogen-associated molecular patterns). Furthermore, bacteria and PAMPs activate the coagulation system and platelets. TLRs are now implicated in a range of cardiovascular diseases and syndromes including atherosclerosis and sepsis. Our group is working on the hypotheses that differences exist in how tissues of the cardiovascular system, including vessels, endothelium, heart and blood, sense pathogens compared with immune cells (principally macrophages) and that identifying such differences will reveal new therapeutic targets for the treatment of cardiovascular disease. We have identified examples of similarities and differences in how cardiovascular tissues and macrophages sense PAMPs. These findings will be discussed together with our interpretation of how this information may lead to new treatments.


Subject(s)
Cardiovascular Diseases/immunology , Cardiovascular System/immunology , Intracellular Signaling Peptides and Proteins/immunology , Toll-Like Receptors/immunology , Humans , Immunity, Innate , Macrophages/immunology
6.
Cancer Gene Ther ; 14(9): 802-14, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17589432

ABSTRACT

Immunization with mRNA encoding tumor antigen is an emerging vaccine strategy for cancer. In this paper, we demonstrate that mice receiving systemic injections of MART1 mRNA histidylated lipopolyplexes were specifically and significantly protected against B16F10 melanoma tumor progression. The originality of this work concerns the use of a new tumor antigen mRNA formulation as vaccine, which allows an efficient protection against the growth of a highly aggressive tumor model after its delivery by intravenous route. Synthetic melanoma-associated antigen MART1 mRNA was formulated with a polyethylene glycol (PEG)ylated derivative of histidylated polylysine and L-histidine-(N,N-di-n-hexadecylamine)ethylamide liposomes (termed histidylated lipopolyplexes). Lipopolyplexes comprised mRNA/polymer complexes encapsulated by liposomes. The tumor protective effect was induced with MART1 mRNA carrying a poly(A) tail length of 100 adenosines at an optimal dose of 12.5 microg per mouse. MART1 mRNA lipopolyplexes elicited a cellular immune response characterized by the production of interferon-gamma and the induction of cytotoxic T lymphocytes. Finally, the anti-B16 response was enhanced using a formulation containing both MART1 mRNA and MART1-LAMP1 mRNA encoding the antigen targeted to the major histocompatibility complex class II compartments by the lysosomal sorting signal of LAMP1 protein. Our results provide a basis for the development of mRNA histidylated lipopolyplexes for cancer vaccine.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/administration & dosage , Histidine/metabolism , Melanoma, Experimental/pathology , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/genetics , RNA, Messenger/genetics , Animals , Antigens, Neoplasm/metabolism , Cancer Vaccines/genetics , Disease Progression , MART-1 Antigen , Melanoma, Experimental/immunology , Mice , Microscopy, Electron, Transmission , Neoplasm Proteins/metabolism , RNA, Messenger/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Transcription, Genetic
7.
J Immunol ; 166(12): 7165-71, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390463

ABSTRACT

We report here on the characterization of the novel immunosuppressant Sanglifehrin A (SFA). SFA is a representative of a class of macrolides produced by actinomycetes that bind to cyclophilin A (CypA), the binding protein of the fungal cyclic peptide cyclosporin A (CsA). SFA interacts with high affinity with the CsA binding side of CypA and inhibits its peptidyl-prolyl isomerase activity. The mode of action of SFA is different from known immunosuppressive drugs. It has no effect on the phosphatase activity of calcineurin, the target of the immunosuppressants CsA and FK506 when complexed to their binding proteins CypA and FK binding protein, respectively. Moreover, its effects are independent of binding of cyclophilin. SFA inhibits alloantigen-stimulated T cell proliferation but acts at a later stage than CsA and FK506. In contrast to these drugs, SFA does not affect IL-2 transcription or secretion. However, it blocks IL-2-dependent proliferation and cytokine production of T cells, in this respect resembling rapamycin. SFA inhibits the proliferation of mitogen-activated B cells, but, unlike rapamycin, it has no effect on CD154/IL-4-induced Ab synthesis. The activity of SFA is also different from that of other known late-acting immunosuppressants, e.g., mycophenolate mofetil or brequinar, as it does not affect de novo purine and pyrimidine biosynthesis. In summary, we have identified a novel immunosuppressant, which represents, in addition to CsA, FK506 and rapamycin, a fourth class of immunophilin-binding metabolites with a new, yet undefined mechanism of action.


Subject(s)
Cyclophilin A/metabolism , Immunosuppressive Agents/metabolism , Lactones/metabolism , Spiro Compounds/metabolism , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Cycle/drug effects , Cell Cycle/immunology , Clone Cells , Cyclophilin A/antagonists & inhibitors , Cytokines/biosynthesis , Dihydroorotase/antagonists & inhibitors , Dihydroorotase/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Immunosuppressive Agents/pharmacology , Jurkat Cells , Lactones/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred CBA , Monocytes/drug effects , Monocytes/immunology , Protein Binding/immunology , Spiro Compounds/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tacrolimus Binding Protein 1A/metabolism
8.
Transpl Immunol ; 8(3): 177-87, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11147698

ABSTRACT

OBJECTIVE: FTY720 is a new immunosuppressant active in transplantation models, which modulates lymphocyte recirculation, leading to transient peripheral lymphopenia and increased lymphocytes in lymph nodes and Peyer's patches. Here, we investigated the susceptibility of cynomolgus monkeys to FTY720 given orally either alone or in combination with two other immunosuppressants, Cyclosporin Neoral or RAD, as an introductory study to transplantation protocols. METHODS: Each of the three phases of the study comprised a 3-week treatment period with FTY720 administered daily orally at 0.3, 0.1 or 0.03 mg/kg/day, respectively, followed by a 3-week recovery. FTY720 was given as single compound during the first week and in combination with Neoral at 20 mg/kg/day p.o. or RAD at 0.5 mg/kg/day p.o. during the subsequent 2 weeks. MAIN FINDINGS: These treatment regimen were well tolerated, except for some body weight loss at high FTY720 dose (0.3 mg/kg/day). FTY720 treatment resulted in a rapid decrease of white blood cell counts which reached a plateau after 3 days. A decrease in both T- and B-lymphocyte counts by up to 80-90% was seen with FTY720 doses of 0.1 and 0.3 mg/kg/day. FTY720 blood levels, both trough levels and AUC(0-24 h), showed a linear relationship with FTY720 dose. The reduction in lymphocyte counts was not directly proportional to FTY720 blood levels. The exposure to FTY720 significantly increased upon coadministration of Neoral. This pharmacokinetic interaction was not observed for coadministration of RAD. However, the peripheral lymphodepletion was slightly increased after coadministration of RAD but not of Neoral. This may be related to the intrinsic effects of RAD on hematopoietic cells. CONCLUSIONS: FTY720 given orally was effective in terms of peripheral T- and B-lymphodepletion and was well tolerated in cynomolgus monkeys even in combination with Cyclosporine Neoral or RAD, indicating that such combination protocols could be used in allo- and xenotransplantation in this species. However, the data indicate a potentiation of FTY720 exposure by CsA coadministration and additional lymphodepletion by coadministration of FTY720 and RAD which should be carefully monitored.


Subject(s)
B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Immunosuppressive Agents/pharmacology , Lymphocyte Depletion/methods , Propylene Glycols/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Administration, Oral , Animals , Cyclosporine/administration & dosage , Dose-Response Relationship, Drug , Drug Tolerance , Everolimus , Fingolimod Hydrochloride , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , Lymphocyte Count , Macaca fascicularis , Propylene Glycols/administration & dosage , Propylene Glycols/blood , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sphingosine/analogs & derivatives , Transplantation Immunology
9.
Antimicrob Agents Chemother ; 44(1): 143-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10602736

ABSTRACT

Cyclosporine (CsA) is an immunosuppressive and antimicrobial drug which, in complex with cyclophilin A, inhibits the protein phosphatase calcineurin. We recently found that Cryptococcus neoformans growth is resistant to CsA at 24 degrees C but sensitive at 37 degrees C and that calcineurin is required for growth at 37 degrees C and pathogenicity. Here CsA analogs were screened for toxicity against C. neoformans in vitro. In most cases, antifungal activity was correlated with cyclophilin A binding in vitro and inhibition of the mixed-lymphocyte reaction and interleukin 2 production in cell culture. Two unusual nonimmunosuppressive CsA derivatives, (gamma-OH) MeLeu(4)-Cs (211-810) and D-Sar (alpha-SMe)(3) Val(2)-DH-Cs (209-825), which are also toxic to C. neoformans were identified. These CsA analogs inhibit C. neoformans via fungal cyclophilin A and calcineurin homologs. Our findings identify calcineurin as a novel antifungal drug target and suggest nonimmunosuppressive CsA analogs warrant investigation as antifungal agents.


Subject(s)
Calcineurin Inhibitors , Cryptococcus neoformans/drug effects , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Peptidylprolyl Isomerase/physiology , Animals , Cyclosporine/metabolism , Drug Resistance, Microbial , Fluconazole/pharmacology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Temperature
11.
Int Immunol ; 10(1): 37-48, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9488154

ABSTRACT

To define the immunological functions of tumor necrosis factor (TNF) in Candida albicans infection, TNF/lymphotoxin (LT)-alpha double-deficient mice were assessed for susceptibility to systemic or gastrointestinal infection and parameters of innate and adaptive Th immunity. When compared to wild-type mice, TNF/LT-alpha-deficient mice were more susceptible to either type of infection caused by virulent or low-virulence C. albicans cells. Susceptibility to infection correlated with impaired development of protective Th1 responses, in spite of the production of bioactive IL-12. The occurrence of predominant Th2 responses was associated with both impaired antifungal effector functions of neutrophils and a defective expression of co-stimulatory molecules on macrophages. All functions were improved upon administration of recombinant TNF-alpha, also resulting in increased resistance to infection. These findings indicate that the protective effect of TNF-alpha in candidiasis relies on the induction of antifungal Th1 responses, possibly occurring through stimulation of antifungal effector functions and co-stimulatory activities of phagocytic cells.


Subject(s)
Candidiasis/immunology , Lymphocyte Activation/immunology , Lymphotoxin-alpha/immunology , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/deficiency , Animals , Disease Susceptibility , Female , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Phagocytes/immunology , Spleen/cytology , Spleen/immunology , Tumor Necrosis Factor-alpha/immunology
12.
J Immunol ; 158(5): 2126-33, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9036957

ABSTRACT

TNF is implicated in in vitro Ig production, but its role in vivo is not clearly defined. Our previous studies had shown that TNF-LT alpha double-deficient mice have defective IgM and IgG primary Ab responses to the T cell-dependent (TD) Ag SRBC. We now extend these studies to secondary responses and to T cell-independent (TI) B cell responses. Injections of the TD Ag SRBC did not induce germinal center formation in the spleen of TNF-LT alpha-deficient mice. Associated with the morphologic defect, there was a defective IgG Ab response and a secondary hyper-IgM response to the TD Ag in TNF-LT alpha-deficient mice. The response to the TI Ag type 2 DNP-alanyl-glycyl-glycyl-Ficoll was essentially absent in TNF-LT alpha-deficient mice, while that to the TI Ag type 1 TNP-LPS was significantly reduced only for IgG2b isotype. Transplantation of bone marrow cells from wild-type mice into irradiated TNF-LT alpha-deficient mice restored the formation of splenic germinal centers and corrected the IgM and IgG responses to both TD and TI Ags. These data suggest that TNF and/or LT alpha signaling are critically required for germinal center formation and for the IgM and IgG responses to both TD and TI type 2 Ags.


Subject(s)
Antigens, T-Independent/immunology , B-Lymphocytes/immunology , Ficoll/analogs & derivatives , Immunoglobulin Class Switching/immunology , Lymphotoxin-alpha/genetics , Tumor Necrosis Factor-alpha/deficiency , Animals , Antibody Formation/genetics , B-Lymphocytes/metabolism , Bone Marrow Transplantation , Erythrocytes/immunology , Ficoll/immunology , Germinal Center/metabolism , Immunization , Mice , Mice, Inbred C57BL , Mice, Knockout , Sheep , Tumor Necrosis Factor-alpha/genetics
13.
J Antibiot (Tokyo) ; 50(11): 893-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9592559

ABSTRACT

Two novel metabolites, cymbimicins A and B, were isolated from the culture broth of a strain of Micromonospora sp. by screening for cyclophilin binding metabolites from actinomycete strains. Cymbimicin A binds to cyclophilin A with a high affinity six fold lower than to that of cyclosporin A. The binding affinity of cymbimicin B is about 100 times lower. The taxonomy of the producing strain, fermentation, isolation, physical and biological properties and structure elucidation are described.


Subject(s)
Immunosuppressive Agents/isolation & purification , Lactones/isolation & purification , Micromonospora/chemistry , Binding, Competitive/drug effects , Dose-Response Relationship, Drug , Fermentation , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Lactones/chemistry , Lactones/pharmacology , Magnetic Resonance Spectroscopy , Micromonospora/metabolism , Peptidylprolyl Isomerase/metabolism
14.
Ann N Y Acad Sci ; 795: 189-95, 1996 Oct 31.
Article in English | MEDLINE | ID: mdl-8958929

ABSTRACT

In vivo administration of interleukin-12 decreases peripheral blood counts and bone marrow hematopoiesis, although in vitro IL-12 was shown to synergize with other cytokines to stimulate the proliferation and differentiation of early hematopoietic progenitors. We investigated whether the in vivo inhibition of hematopoiesis by IL-12 is indirectly mediated by IL-12-induced IFN-gamma. IL-12 was administered to wild-type or IFN-gamma receptor deficient (IFN gamma R-/-) mice. IL-12 treatment resulted in lower peripheral blood counts and a twofold decrease in bone marrow cellularity and hematopoietic progenitors in wild-type mice, but not in IFN gamma R-/- mice. Splenic weight and cellularity were dramatically increased after IL-12 administration in wild-type mice and somewhat less in IFN gamma R-/- mice. The increase was predominantly due to NK cell and macrophage infiltration in wild-type mice, and strong extramedullary hematopoiesis in IFN gamma R-/- mice. Thus, the reduction in total bone marrow cells and hematopoietic progenitor numbers following IL-12 treatment are largely indirect, mediated by IL-12-induced IFN gamma. In the absence of IFN-gamma signaling, IL-12 promotes both bone marrow and splenic hematopoiesis, consistent with its in vitro activities.


Subject(s)
Hematopoiesis , Interferon-gamma/physiology , Interleukin-12/physiology , Animals , Antigens, CD/physiology , Bone Marrow Cells , Humans , Mice , Mice, Knockout , Receptors, Interferon/physiology , Spleen/cytology , Interferon gamma Receptor
15.
Mol Med ; 2(2): 247-55, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8726467

ABSTRACT

BACKGROUND: Mice with inactivated tumor necrosis factor (TNF) and lymphotoxin alpha (LT alpha) genes have profound abnormalities of the immune system including lymphocytosis, lack of lymph nodes, undifferentiated spleen, hypoimmunoglobulinaemia, and defective Ig class switch. Here, we asked whether this phenotype is due to incompetent lymphohemopoietic progenitors or to a defective environment. MATERIALS AND METHODS: Lethally irradiated TNF-LT alpha-deficient and wild-type mice received bone marrow cells from either TNF-LT alpha-deficient or wild-type mice. The reconstitution and transfer of the phenotype was followed by morphological and functional analyses. RESULTS: Bone marrow cells from wild-type mice restored the synthesis of TNF and LT alpha, corrected the splenic microarchitecture, normalized the lymphocyte counts in the circulation, and repopulated the lamina propria with IgA-producing plasma cells of TNF-LT alpha-deficient mice. Furthermore, the formation of germinal centers in the spleen and the defective Ig class switch in response to a T-cell dependent antigen was corrected, while no lymph nodes were formed. Conversely, the TNF-LT alpha phenotype could be transferred to wild-type mice by bone marrow transplantation after lethal irradiation. CONCLUSIONS: These data demonstrate that most TNF- and LT alpha-producing cells are bone marrow derived and radiosensitive, and that the immunodeficiency due to TNF-LT alpha deletion can be corrected to a large extent by normal bone marrow cell transplantation. The genotype of the donor bone marrow cells determines the functional and structural phenotype of the TNF-LT alpha-deficient adult murine host, with the exception of lymph node formation. These findings may have therapeutic implications for the restoration of genetically defined immunodeficiencies in humans.


Subject(s)
Bone Marrow Transplantation/immunology , Gene Deletion , Immunologic Deficiency Syndromes/immunology , Lymphotoxin-alpha/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Bone Marrow/immunology , Crosses, Genetic , Immunologic Deficiency Syndromes/genetics , Lymphocytes/immunology , Lymphocytosis/immunology , Lymphotoxin-alpha/biosynthesis , Lymphotoxin-alpha/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Spleen/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/deficiency , Whole-Body Irradiation
16.
J Exp Med ; 181(5): 1893-8, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7722464

ABSTRACT

Interleukin (IL)-12 synergizes with other cytokines to stimulate the proliferation and differentiation of early hematopoietic progenitors in vitro. However, in vivo administration of IL-12 decreases peripheral blood counts and bone marrow hematopoiesis. Here, we used interferon (IFN) gamma receptor-deficient (IFN gamma R-/-) mice to investigate whether the in vivo inhibition of hematopoiesis by IL-12 is indirectly mediated by IL-12-induced IFN-gamma. IL-12 administered for 4 d (1 microgram/mouse per day) resulted in lower peripheral blood counts and a 2-fold decrease in bone marrow cellularity in wild-type mice, but not in IFN gamma R-/- mice. Bone marrow hematopoietic progenitors were decreased after IL-12 treatment in wild-type mice, but rather increased in IFN gamma R-/- mice. Splenic cellularity was 2.3-fold higher after IL-12 administration in wild-type mice, largely due to natural killer (NK) cell and macrophage infiltration together with some extramedullary hematopoiesis. In IFN gamma R-/- mice, spleen cellularity was less increased, there were fewer infiltrating NK cells, but a strong extramedullary hematopoiesis. Thus, alterations mediated by IL-12-induced IFN-gamma include reduction in bone marrow cellularity and hematopoietic progenitors, as well as pronounced splenomegaly, largely caused by NK cell infiltration. In the absence of IFN-gamma signaling, IL-12 promotes hematopoiesis, consistent with its in vitro activities.


Subject(s)
Hematopoiesis/drug effects , Interferon-gamma/physiology , Interleukin-12/antagonists & inhibitors , Animals , Bone Marrow/drug effects , Mice , Receptors, Interferon/analysis , Interferon gamma Receptor
18.
Blood ; 84(5): 1543-52, 1994 Sep 01.
Article in English | MEDLINE | ID: mdl-7520778

ABSTRACT

The immunosuppressive drug rapamycin suppresses T-cell activation by impairing the T-cell response to lymphokines such as interleukin-2 (IL-2) and interleukin-4 (IL-4). In addition, rapamycin blocks the proliferative response of cell lines to a variety of hematopoietic growth factors, including interleukin-3 (IL-3), interleukin-6 (IL-6), granulocyte-colony stimulating factor (G-CSF), granulocyte macrophage-colony stimulating factor (GM-CSF), and kit ligand (KL), suggesting that it should be a strong inhibitor of hematopoiesis. In this report, we studied the effects of rapamycin on different hematopoietic cell populations in vitro and in vivo. In vitro, rapamycin inhibited the proliferation of primary bone marrow cells induced by IL-3, GM-CSF, KL, or a complex mixture of factors present in cell-conditioned media. Rapamycin also inhibited the multiplication of colony-forming cells in suspension cultures containing IL-3 plus interleukin-1 (IL-1) or interleukin-11 (IL-11) plus KL. In vivo, treatment for 10 to 28 days with high doses of rapamycin (50 mg/kg/d, orally) had no effect on myelopoiesis in normal mice, as measured by bone marrow cellularity, proliferative capacity, and number of colony-forming progenitors. In contrast, the same treatment strongly suppressed the hematopoietic recovery normally seen 10 days after an injection of 5-fluorouracil (5-FU; 150 mg/kg, intravenously [i.v.]). Thus, rapamycin may be detrimental in myelocompromised individuals. In addition, the results suggest that the rapamycin-sensitive cytokine-driven pathways are essential for hematopoietic recovery after myelodepression, but not for steady-state hematopoiesis.


Subject(s)
Bone Marrow Cells , Cell Division/drug effects , Cytokines/pharmacology , Heart Transplantation/immunology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Immunosuppressive Agents/pharmacology , Polyenes/pharmacology , Animals , Bone Marrow/drug effects , Cell Line , Culture Media, Conditioned , Cytokines/antagonists & inhibitors , Female , Fluorouracil/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-1/pharmacology , Interleukin-3/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Reference Values , Sirolimus , Stem Cell Factor , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous
19.
Biochemistry ; 33(27): 8218-24, 1994 Jul 12.
Article in English | MEDLINE | ID: mdl-8031755

ABSTRACT

A complementary DNA (cDNA) for human cyclophilin C (Cyp-C) was isolated from a human kidney cDNA library. Northern blot experiments with several human tissues and cell lines revealed that Cyp-C is less abundant than Cyp-A. The amount of Cyp-C mRNA was 10-fold lower than that of Cyp-A in kidney. Expression of human Cyp-C in the kidney is not significantly elevated compared to pancreas, skeletal muscle, heart, lung, and liver. This argues against a previously postulated specific role for Cyp-C in the nephrotoxic effects of CsA in humans, based on the studies of its relative abundance in murine kidney. It is present in extremely low concentrations in brain and in the Jurkat T cell line. The binding of recombinant human Cyp-A, -B, and -C to cyclosporin A (CsA) was studied by immunochemical methods. The relative affinity of Cyp-C for CsA is lower by a factor of 2 than that of Cyp-A, which itself is 10-fold lower than that of Cyp-B. Cross-reactivity studies with a series of Cs derivatives showed that Cyp-C binds CsA with a fine specificity similar to that of Cyp-A and Cyp-B. Cs amino acid residues 1, 2, 10, and 11 seemed essential for the interaction with all three Cyp subtypes. However, Cyp-C tolerates a greater variety of structures on Cs at position 2 than Cyp-A does, suggesting that this residue of CsA might not be in tight contact with Cyp-C. This was confirmed by modeling of human Cyp-C on the structure of the complex formed by Cyp-A and CsA. The knowledge of the fine specificity of human Cyps for CsA and of their expression levels may provide better insights into how CsA acts on its different target proteins in vivo.


Subject(s)
Amino Acid Isomerases/chemistry , Carrier Proteins/chemistry , Cyclophilins , Cyclosporine/metabolism , Amino Acid Isomerases/genetics , Amino Acid Isomerases/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , Cyclophilin C , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Escherichia coli , Humans , Kidney/chemistry , Models, Molecular , Molecular Sequence Data , Peptidylprolyl Isomerase , RNA, Messenger/analysis , Recombinant Proteins/metabolism , Sequence Homology , Tissue Distribution
20.
Leuk Lymphoma ; 14(3-4): 241-9, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7950912

ABSTRACT

IL-11 is a pleiotropic cytokine originally isolated from a bone marrow stromal cell line. It has been shown to share many activities with IL-6, namely to stimulate T cell-dependent B cell maturation, megakaryopoiesis and various stages of myeloid differentiation, but to inhibit adipogenesis. However, the activity of IL-11 on different stages of erythropoiesis in vitro clearly sets it apart form IL-6. IL-11 has little hematopoietic colony stimulatory activity of its own although it sustains terminal differentiation of the late erythroid progenitors CFU-E. In combination with IL-3, IL-11 has profound stimulatory effects on early multipotent hematopoietic progenitors (pre-CFCmulti), on multilineage colony-forming cells (CFCmulti), as well as on erythroid progenitors. The combination of IL-11 with the ligand for c-kit (KL) preferentially acts on early cells since it promotes the multiplication of pre-CFCmulti and stimulates highly proliferative erythroid progenitors that yields remarkable macroscopic erythroblast colonies in culture. The synergistic activity of IL-11 and KL, two stromal factors present in the bone marrow microenvironment, points to a pivotal role of IL-11 in early hematopoiesis. In vivo administration of recombinant human IL-11 elevates the number of circulating neutrophils and platelets and increased megakaryopoiesis in normal mice and primates.


Subject(s)
Interleukin-11/pharmacology , Interleukin-11/physiology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...