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










Publication year range
1.
Int Immunopharmacol ; 7(6): 845-57, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17466918

ABSTRACT

Dendritic cells (DCs) play a key role as potent biological adjuvants in anti-tumor host responses. However, DC-based vaccination does not always eradicate tumors effectively. Clinical evidence suggests that a strategy to recruit a substantial number of DCs into the tumor mass might provoke proficient anti-tumor immune responses. Here we describe that myeloid DCs (mDCs) efficiently accumulate in tumor sites after intravenous injection of recombinant macrophage inflammatory protein (MIP)-1alpha when pretreated locally with adjuvants like Propionibacterium acnes. Combined treatment of tumor-bearing mice with MIP-1alpha and P. acnes also recruited a large number of natural killer cells (NK cells) to both tumor sites and regional lymph nodes (LNs) and induced a strong T helper 1 immunity at an early time. This early response later led to accumulation of CD8(+) T cells, retraction of tumors and survival of animals treated with P. acnes/MIP-1alpha. In vivo depletion of NK cells or CD8(+) T cells impaired anti-tumor effects, suggesting that activation of NK cells and CD8(+) T cells contributes to anti-tumor immunity in this model. Therefore, this study provides a novel therapeutic strategy for cancer treatment using MIP-1alpha and certain adjuvants.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Lewis Lung/therapy , Dendritic Cells/drug effects , Macrophage Inflammatory Proteins/administration & dosage , Propionibacterium acnes , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line , Cell Movement/drug effects , Chemokine CCL3 , Chemokine CCL4 , Combined Modality Therapy , Dendritic Cells/immunology , Female , Injections, Intravenous , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Tumor Burden/drug effects
2.
J Allergy Clin Immunol ; 116(4): 820-6, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16210056

ABSTRACT

BACKGROUND: Little is known about the mechanisms that regulate the selective recruitment of basophils to sites of allergic inflammation. OBJECTIVE: Here we examine the role of stem cell factor (SCF) in the regulation of basophil function. METHODS: Human basophils were isolated from peripheral blood, and their migration was investigated in chemotaxis assays. Apoptosis was detected by means of annexin V and propidium iodide staining. The expression of cell-surface molecules was measured by means of flow cytometry. RESULTS: SCF amplified the chemotactic responsiveness of human peripheral blood basophils to the chemoattractants eotaxin, monocyte chemotactic protein 2 and macrophage inflammatory protein 1alpha, and C5a, without being chemotactic or chemokinetic by itself. SCF synergized with chemoattractants in causing basophil upregulation of the integrin CD11b, and this effect was inhibited by a c-kit antibody, the tyrosine kinase inhibitor imatinib mesylate (STI-571), and a phosphatidylinositol 3 kinase inhibitor but not by inhibitors of p38 mitogen-activated protein kinase or mitogen-activated protein kinase/extracellular signal-regulated kinase kinase. Basophils bound fluorescence-labeled SCF and expressed its receptor, c-kit, which was markedly upregulated in culture for 24 to 48 hours in the presence of IL-3. Moreover, SCF prolonged basophil survival in concert with IL-3 by delaying apoptosis. These effects of SCF were selective for basophils because chemotaxis and CD11b upregulation of eosinophils or neutrophils were unchanged. CONCLUSION: SCF might be an important selective modulator of basophil function through a phosphatidylinositol 3 kinase-dependent pathway.


Subject(s)
Basophils/drug effects , Integrins/metabolism , Stem Cell Factor/pharmacology , Basophils/cytology , Basophils/physiology , CD11b Antigen/metabolism , Cell Survival/drug effects , Chemokine CCL11 , Chemokine CCL4 , Chemokine CCL8 , Chemokines, CC/administration & dosage , Chemotactic Factors/administration & dosage , Chemotaxis, Leukocyte/drug effects , Complement C5a/administration & dosage , Drug Synergism , Humans , In Vitro Techniques , Interleukin-3/administration & dosage , Macrophage Inflammatory Proteins/administration & dosage , Monocyte Chemoattractant Proteins/administration & dosage , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Stem Cell Factor/administration & dosage , Up-Regulation/drug effects
3.
Gut ; 54(8): 1114-20, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16009684

ABSTRACT

INTRODUCTION: Exacerbations of inflammatory bowel disease are thought to be related to concurrent infections. As infections are associated with elevated local and serum concentrations of chemokines, we have determined whether systemic administration of the CC chemokine macrophage inflammatory protein 1alpha (MIP-1alpha) exacerbates colitis in a mouse model. METHODS: Colitis was induced in Balb/c mice using trinitrobenzene sulfonic acid (TNBS). Starting four days later, animals received daily intraperitoneal injections of recombinant MIP-1alpha. On day 7, mice were killed and pieces of colon taken for immunohistology and polymerase chain reaction analysis. The direct effects of MIP-1alpha on mucosal T cells and fibroblasts in vitro were also investigated. RESULTS: Systemic administration of MIP-1alpha markedly enhanced colitis with mice developing large transmural ulcers filled with granulation tissue. Treatment resulted in increased numbers of CD4 cells infiltrating the colonic lamina propria, increased interferon gamma (IFN-gamma) levels, and increased transcripts for tumour necrosis factor alpha (TNF-alpha) and matrix metalloproteinase 3 (MMP3). Isolated lamina propria lymphocytes from mice with TNBS colitis contained increased numbers of IFN-gamma and TNF-alpha transcripts when stimulated with MIP-1alpha in vitro. Colonic lamina propria fibroblasts also responded to MIP-1alpha with increased proliferation and decreased collagen 1 synthesis but fibroblast proliferation was not seen in vivo. CONCLUSIONS: These experiments show that increasing serum concentrations of a chemokine, MIP-1alpha, exacerbates immune mediated colitis. The effect seems to be due to the ability of MIP-1alpha to boost Th1 responses in the gut wall. Our findings also suggest a potential pathway by which peripheral infections can exacerbate inflammatory bowel disease.


Subject(s)
Colitis, Ulcerative/immunology , Macrophage Inflammatory Proteins/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Colon/immunology , Disease Models, Animal , Female , Fibroblasts/immunology , Immunohistochemistry/methods , Injections, Intraperitoneal , Interferon-gamma/analysis , Intestinal Mucosa/immunology , Matrix Metalloproteinase 3/analysis , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/analysis
4.
J Immunol ; 174(9): 5766-73, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843579

ABSTRACT

Strategies that generate tumor Ag-specific effector cells do not necessarily cure established tumors. We hypothesized that the relative efficiency with which tumor-specific effector cells reach the tumor is critical for therapy. We demonstrate in this study that activated T cells respond to the chemokine CCL3, both in vitro and in vivo, and we further demonstrate that expression of CCL3 within tumors increases the effector T cell infiltrate in those tumors. Importantly, we show that adenoviral gene transfer to cause expression of CCL3 within B16ova tumors in vivo increases the efficacy of adoptive transfer of tumor-specific effector OT1 T cells. We additionally demonstrate that such therapies result in endogenous immune responses to tumor Ags that are capable of protecting animals against subsequent tumor challenge. Strategies that modify the "visibility" of tumors have the potential to significantly enhance the efficacy of both vaccine and adoptive transfer therapies currently in development.


Subject(s)
Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Genetic Therapy , Immunotherapy, Adoptive , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Adjuvants, Immunologic/biosynthesis , Adjuvants, Immunologic/physiology , Animals , Antigens, Neoplasm/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Line, Tumor , Chemokine CCL3 , Chemokine CCL4 , Chemokines/physiology , Chemokines, CC/administration & dosage , Chemokines, CC/biosynthesis , Chemokines, CC/physiology , Genetic Therapy/methods , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Immunotherapy, Adoptive/methods , Inflammation Mediators/physiology , Injections, Subcutaneous , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/physiology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR5/biosynthesis , T-Lymphocytes, Regulatory/metabolism
5.
Biomaterials ; 26(24): 5048-63, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15769541

ABSTRACT

Chemotaxis, cell migration directed by spatial concentration gradients of chemoattractant molecules, is critical for proper function of the immune system. Materials capable of generating defined chemoattractant gradients via controlled release may be useful for the design of improved vaccines and immunotherapies that draw specific cells to an immunization site. To this end, we encapsulated formyl-Nle-Leu-Phe-Nle-Tyr-Lys (fN'LFN'YK) peptides or macrophage inflammatory protein-3alpha (MIP-3alpha or CCL20) in degradable poly(lactide-co-glycolide) microspheres that provided sustained release for more than 2 weeks in vitro. fN'LFN'YK and MIP-3alpha chemoattract dendritic cells (DCs), the key antigen-presenting cells involved in generation of primary immune responses, and their precursors, monocytes. Using an in vitro videomicroscopy migration assay, we detected strong chemotaxis of human monocytes and monocyte-derived DCs through 3D collagen gels toward microspheres releasing fN'LFN'YK. Similarly, microparticles releasing MIP-3alpha were able to attract mouse bone marrow-derived dendritic cells. Strikingly, prolonged attraction of DCs from distances up to 500 microm from the source to the point of contact with individual microspheres was observed. Such microspheres could be of general interest for the design of vaccines that promote adaptive immunity and as a platform for studying the biology of chemotaxis in vitro and in vivo.


Subject(s)
Absorbable Implants , Cell Movement/drug effects , Chemotactic Factors/administration & dosage , Chemotactic Factors/chemistry , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Dendritic Cells/physiology , Monocytes/physiology , Animals , Cell Movement/physiology , Cells, Cultured , Chemokine CCL20 , Chemokines, CC/administration & dosage , Chemokines, CC/chemistry , Coated Materials, Biocompatible/chemistry , Dendritic Cells/drug effects , Diffusion , Humans , Kinetics , Lactic Acid/chemistry , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/chemistry , Materials Testing , Mice , Microspheres , Monocytes/drug effects , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/chemistry
6.
J Neurooncol ; 66(1-2): 147-54, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15015780

ABSTRACT

Subcutaneous vaccination using granulocyte-macrophage colony-stimulating factor (GM-CSF)-transduced glioma cells substantially prolongs survival in the mouse GL261 glioma model. To potentiate the efficacy of GM-CSF-based vaccination, syngeneic C57BL/6 mice bearing pre-implanted intracerebral GL261 gliomas were vaccinated twice subcutaneously with various combinations of glioma cells retrovirally engineered to release GM-CSF, interleukin (IL)-4 or macrophage inflammatory protein (MIP)-1alpha. More than 80% of the animals vaccinated with GM-CSF-secreting or GM-CSF- and IL-4-secreting cells were long-term survivors (> 120 days). Their survival was significantly prolonged compared with animals vaccinated with wild-type cells, which died after a median survival time of 30 days. The combination of IL-4 with GM-CSF did not provide a survival advantage over GM-CSF alone, regardless of whether the animals carried a small or large intracranial tumor load. Further, when the animals were vaccinated with a mixture of GM-CSF-, IL-4- and MIP-1alpha-secreting cells, the median survival was 37 days, and only 22% of the animals in this group were long-term survivors, similar to the vaccination effect of non-modified glioma cells. Thus, unexpectedly, the co-expression of MIP-1alpha, which was meant to attract T cells for stimulation by GM-CSF- and IL-4-stimulated dendritic cells, nullified the induction of an immune response against the GL261 glioma by a GM-CSF- and IL-4-expressing subcutaneous vaccine.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Cancer Vaccines/antagonists & inhibitors , Glioma/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Macrophage Inflammatory Proteins/administration & dosage , Vaccination , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Chemokine CCL3 , Chemokine CCL4 , Drug Synergism , Genetic Vectors , Glioma/pathology , Immunohistochemistry , Immunosuppression Therapy , Injections, Subcutaneous , Interleukin-4/administration & dosage , Mice , Mice, Inbred C57BL , Retroviridae/genetics , Survival Analysis , Transduction, Genetic , Treatment Failure
7.
J Natl Cancer Inst ; 96(3): 201-9, 2004 Feb 04.
Article in English | MEDLINE | ID: mdl-14759987

ABSTRACT

BACKGROUND: Dendritic cells (DCs) play a central role in immune responses and may be useful adjuvants for tumor vaccine therapy. We previously reported that F4/80(-)B220(-)CD11c(+) DC precursors expressing the CC chemokine receptors CCR1 and CCR5 are mobilized rapidly into the circulation in mice injected with Propionibacterium acnes and are recruited into inflammatory tissue by macrophage inflammatory protein 1alpha (MIP-1alpha), which binds to CCR1 and CCR5. Here we investigate the mechanisms of DC precursor mobilization and the antitumor effect of these cells in mice. METHODS: Numbers of DC precursors in peripheral blood were determined in P. acnes-treated mice (groups of 10 C57BL/B6 [B6] wild-type mice, CCR1(-/-) mice, CCR5(-/-) mice, and B6 mice treated with antibody to MIP-1alpha or control antibody) and in B6 mice injected with recombinant MIP-1alpha. MIP-1alpha-mobilized DC precursors matured by treatment with granulocyte-macrophage colony-stimulating factor, interleukin 4, and tumor necrosis factor-alpha and pulsed with B16 melanoma lysates were assayed for their ability to confer protective immunity against tumor challenge in vivo and to induce cytotoxic T lymphocytes against B16 tumor cells in vitro. RESULTS: The recruitment of DC precursors into the circulation by P. acnes administration was higher in B6 mice (12.6%, 95% confidence interval [CI] = 9.1% to 16.1%) than in CCR1(-/-) (9.0%, 95% CI = 7.5% to 10.5%), CCR5(-/-) (6.3%, 95% CI = 5.2% to 7.3%), or anti-MIP-1alpha antibody-treated (6.6%, 95% CI = 5.7% to 7.5%) mice. Injection of MIP-1alpha also mobilized DC precursors into the circulation (13.1%, 95% CI = 10.8% to 15.6%). Matured MIP-1alpha-mobilized-DC precursors pulsed with B16 tumor lysates elicited B16-specific antitumor immunity in vitro and in vivo. CONCLUSIONS: MIP-1alpha and its receptors are important in recruiting DC precursors into the circulation. DC precursors mobilized rapidly by MIP-1alpha may provide sufficient useful DC precursors for DC-based vaccination in cancer treatment.


Subject(s)
Dendritic Cells/metabolism , Immunotherapy/methods , Macrophage Inflammatory Proteins/pharmacology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Receptors, CCR5/genetics , Receptors, Chemokine/genetics , Animals , Cell Line, Tumor , Chemokine CCL3 , Chemokine CCL4 , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunophenotyping , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/metabolism , Melanoma, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Monocytes/metabolism , Receptors, CCR1 , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , T-Lymphocytes, Cytotoxic
8.
Brain Res ; 992(1): 76-84, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14604775

ABSTRACT

This study was undertaken to determine whether cyclooxygenase (COX)-2, the critical enzyme in the production of febrigenic prostaglandin (PG) E(2), may be involved centrally in the fever induced in mice by homologous interleukin (IL)-6, macrophage inflammatory protein (MIP)-1 beta, and interleukin (IL)-18, a member of the pyrogenic IL-1 beta family. To this end, the core temperatures (Tc) of COX-1 and COX-2 gene-ablated mice and of their normal wild-type (WT) counterparts were recorded after intracerebroventricular (i.c.v.) challenge with recombinant murine (rm) IL-6 (10 ng/mouse), rmMIP-1 beta (20 pg/mouse), rmIL-18 (0.01-1 microgram/mouse), rmIL-1 beta (positive control; 0.1 microgram/mouse), or their vehicle (0.1% bovine serum albumin [BSA] in sterile phosphate-buffered saline [PBS]; 5 microl/mouse). rmIL-6 caused a approximately 1 degrees C T(c) rise in WT mice that peaked at approximately 120 min and gradually recovered over the next 3 h; COX-1(-/-) mice exhibited a relatively faster (peak at 45 min) and shorter (recovery at 150 min) febrile course, whereas COX-2(-/-) mice did not develop fever. rmMIP-1 beta induced a 1 degrees C fever (peak at 60 min) with a long time course (recovery incomplete at 300 min) in both WT and COX-2(-/-) mice; COX-1(-/-) mice displayed a quick-onset (peak at 40 min) and shorter (recovery at approximately 240 min) fever. rmIL-18 did not cause any thermal response at any dose whether administered intraperitoneally (i.p.) or i.c.v. in WT mice; COX gene-ablated mice, therefore, were not tested. These data indicate that COX-2-dependent PGE(2) is critical for the febrile response to IL-6, but not to MIP-1 beta. IL-18 i.p. or i.c.v. is not pyrogenic.


Subject(s)
Brain/drug effects , Fever/chemically induced , Interleukin-18/pharmacology , Interleukin-6/pharmacology , Isoenzymes/physiology , Macrophage Inflammatory Proteins/pharmacology , Prostaglandin-Endoperoxide Synthases/physiology , Animals , Body Temperature/drug effects , Brain/enzymology , Chemokine CCL4 , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Fever/enzymology , Injections, Intraperitoneal , Injections, Intraventricular , Interleukin-18/administration & dosage , Interleukin-6/administration & dosage , Macrophage Inflammatory Proteins/administration & dosage , Mice , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
9.
Mol Ther ; 8(4): 629-36, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14529836

ABSTRACT

Circulating lymphocytes are important target cells for the treatment of HIV-related and autoimmune diseases and for stimulating anti-tumor immunity. To date, gene transfection of these nonactivated cells after intravenous delivery of viral or nonviral vectors remains low although these circulating cells are highly accessible. Optimized lentiviral vectors currently can transduce less than 10% of nonactivated circulating lymphocytes. Here we report transfection of up to 15% of these nonactivated cells using liposomes directed to human CCR5 displayed on the surface of helper T cells and macrophages in transgenic mice. Attachment of modified MIP-1 beta to the surface of DNA-liposome complexes increased gene delivery and expression in nonactivated circulating lymphocytes approximately sixfold. In vitro data using these complexes to transfect PM1 cells that have elevated levels of CCR5 supported our data obtained in vivo. Therefore, ligands that bind to cell surface receptors on circulating lymphocytes can be used with optimized systemic liposomes to increase transfection and gene expression in these cells without activation.


Subject(s)
DNA/pharmacology , Gene Expression/drug effects , Liposomes/pharmacology , Lymphocytes/drug effects , Macrophage Inflammatory Proteins/pharmacology , Animals , Chemokine CCL4 , DNA/administration & dosage , Liposomes/administration & dosage , Macrophage Inflammatory Proteins/administration & dosage , Mice , Transfection
10.
Blood ; 102(1): 311-9, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12649140

ABSTRACT

Recent data have implicated macrophage inflammatory protein-1alpha (MIP-1alpha) in multiple myeloma (MM)-associated osteolysis. However, it is unclear whether the chemokine's effects are direct, to enhance osteolysis, or indirect and mediated through a reduction in tumor burden, or both. It is also unclear whether MIP-1alpha requires other factors such as receptor activator of nuclear factor-kappaB ligand (RANKL) for its effects on bone. In murine 5TGM1 (Radl) myeloma-bearing mice, administration of neutralizing anti-MIP-1alpha antibodies reduced tumor load assessed by monoclonal paraprotein titers, prevented splenomegaly, limited development of osteolytic lesions, and concomitantly reduced tumor growth in bone. To determine the effects of MIP-1alpha on bone in vivo, Chinese hamster ovary (CHO) cells secreting human MIP-1alpha (CHO/MIP-1alpha) were inoculated into athymic mice. Mice bearing intramuscular CHO/MIP-1alpha tumors developed lytic lesions at distant skeletal sites, which occurred earlier and were larger than those in mice with CHO/empty vector (EV) tumors. When experimental metastases were induced via intracardiac inoculation, mice bearing CHO/MIP-1alpha tumors developed hypercalcemia and significantly more osteolytic lesions than mice bearing CHO/EV tumors, with intramedullary CHO/MIP-1alpha tumors associated with significantly more tartrate-resistant acid phosphatase-positive (TRAP+) osteoclasts. Injection of recombinant MIP-1alpha over calvariae of normal mice evoked a striking increase in osteoclast formation, an effect dependent on RANK/RANKL signaling because MIP-1alpha had no effect in RANK-/- mice. Together, these results establish that MIP-1alpha is sufficient to induce MM-like destructive lesions in bone in vivo. Because, in the 5TGM1 model, blockade of osteoclastic resorption in other situations does not decrease tumor burden, we conclude that MIP-1alpha exerts a dual effect in myeloma, on osteoclasts, and tumor cells.


Subject(s)
Macrophage Inflammatory Proteins/pharmacology , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Osteolysis/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Bone Resorption/chemically induced , Chemokine CCL3 , Chemokine CCL4 , Disease Models, Animal , Hypercalcemia , Injections, Intralesional , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/immunology , Mice , Multiple Myeloma/pathology , Osteoclasts/cytology , Osteoclasts/drug effects , Osteolysis/etiology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Skull/cytology
11.
Blood ; 101(3): 807-14, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12393512

ABSTRACT

Macrophage inflammatory protein-1alpha (MIP-1alpha) and MIP-1beta are distinct but highly homologous CC chemokines produced by a variety of host cells in response to various external stimuli and share affinity for CCR5. To better elucidate the role of these CC chemokines in adaptive immunity, we have characterized the affects of MIP-1alpha and MIP-1beta on cellular and humoral immune responses. MIP-1alpha stimulated strong antigen (Ag)-specific serum immunoglobulin G (IgG) and IgM responses, while MIP-1beta promoted lower IgG and IgM but higher serum IgA and IgE antibody (Ab) responses. MIP-1alpha elevated Ag-specific IgG1 and IgG2b followed by IgG2a and IgG3 subclass responses, while MIP-1beta only stimulated IgG1 and IgG2b subclasses. Correspondingly, MIP-1beta produced higher titers of Ag-specific mucosal secretory IgA Ab levels when compared with MIP-1alpha. Splenic T cells from MIP-1alpha- or MIP-1beta-treated mice displayed higher Ag-specific Th1 (interferon-gamma [IFN-gamma]) as well as selective Th2 (interleukin-5 [IL-5] and IL-6) cytokine responses than did T cells from control groups. Interestingly, mucosally derived T cells from MIP-1beta-treated mice displayed higher levels of IL-4 and IL-6 compared with MIP-1alpha-treated mice. However, MIP-1alpha effectively enhanced Ag-specific cell-mediated immune responses. In correlation with their selective effects on humoral and cellular immune responses, these chemokines also differentially attract CD4(+) versus CD8(+) T cells and modulate CD40, CD80, and CD86 expressed by B220(+) cells as well as CD28, 4-1BB, and gp39 expression by CD4(+) and CD8(+) T cells in a dose-dependent fashion. Taken together, these studies suggest that these CC chemokines differentially enhance mucosal and serum humoral as well as cellular immune responses.


Subject(s)
Immunity, Mucosal/drug effects , Macrophage Inflammatory Proteins/immunology , Administration, Intranasal , Animals , Antibody Formation/drug effects , Antigens, CD/drug effects , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL3 , Chemokine CCL4 , Cytokines/metabolism , Female , Immunity, Cellular/drug effects , Immunoglobulin Isotypes , Lymphocyte Activation/drug effects , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Ovalbumin/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Brain Res Bull ; 59(3): 181-7, 2002 Nov 30.
Article in English | MEDLINE | ID: mdl-12431747

ABSTRACT

It has been proposed that prostaglandin (PG)E(2) production via a process catalyzed by the inducible isoform of cyclooxygenase (COX)-2 and activation of specific PGE(2) receptor subtypes within the preoptic/anterior hypothalamus (AH/POA) is the last step and unique pathway in the induction of a fever. However, many data support the existence of a PG-independent pathway. That is, other more rapid mechanisms, which involve the constitutive COX-1 isozyme, may be more critical for a PG-dependent fever. Thus, we examined the role of both COX isoforms in the AH/POA in fevers induced by macrophage inflammatory protein (MIP)-1beta, a PG-independent pyrogen, and RANTES (regulated on activation, normal T-cells expressed and secreted), a PG-dependent pyrogen. In freely moving rats, two independent polyclonal antibodies were used which neutralize COX-1 and COX-2. The microinjection of either MIP-1beta or RANTES into the pyrogen-sensitive region of the AH/POA induced an intense fever of rapid onset. Peripheral pretreatment with an antipyretic dose of dexamethasone which prevents COX-2 expression, or the microinjections into the AH/POA of either anti-COX-1 or anti-COX-2, blocked the febrile response induced by RANTES but not that induced by MIP-1beta. These results provide strong evidence for the existence of rapid mechanisms in the AH/POA which involve both COX isozymes during the fever induced by RANTES, and further support the existence of an alternative PG-independent pathway in the febrile response.


Subject(s)
Fever/chemically induced , Isoenzymes/antagonists & inhibitors , Pyrogens/metabolism , Animals , Antibodies/administration & dosage , Antibodies/immunology , Chemokine CCL4 , Chemokine CCL5/administration & dosage , Chemokine CCL5/metabolism , Cyclooxygenase 1 , Cyclooxygenase 2 , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Dinoprostone/metabolism , Isoenzymes/drug effects , Isoenzymes/immunology , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/metabolism , Male , Membrane Proteins , Microinjections , Preoptic Area/metabolism , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/immunology , Pyrogens/administration & dosage , Pyrogens/pharmacology , Rats , Rats, Wistar , Time Factors
13.
J Immunol ; 167(1): 524-31, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418691

ABSTRACT

The understanding of the mechanisms underlying eosinophil recruitment in vivo may aid in the development of novel strategies for the treatment of allergic disorders. In this study, we investigated the role of chemokines in the cascade of events leading to eosinophil recruitment in a stem cell factor (SCF)- and leukotriene B(4) (LTB(4))-dependent allergic pleurisy model in mice. The intrapleural administration of the eosinophil-active chemokines eotaxin, RANTES, and macrophage-inflammatory protein 1alpha (MIP-1alpha) induced a time- and dose-dependent eosinophil recruitment. Pretreatment with anti-eotaxin, but not anti-RANTES or anti-MIP-1alpha, blocked the recruitment of eosinophils following Ag challenge of sensitized animals, and significant eotaxin immunoreactivity was detected in the pleural cavity of these animals. Similarly, only the anti-eotaxin inhibited the eosinophil recruitment induced by injection of SCF in naive animals. However, blockade of SCF did not inhibit the release of eotaxin after Ag challenge of sensitized mice. Akin to its effects on SCF and in the allergic reaction, eotaxin-induced eosinophil recruitment was blocked by the LTB(4) receptor antagonist CP105696. Nevertheless, SCF, but not eotaxin, appeared to regulate the endogenous release of LTB(4) after Ag challenge. Finally, we show that low doses of eotaxin synergized with LTB(4) to induce eosinophil recruitment in the pleural cavity. Overall, the present results show that eotaxin and SCF-induced LTB(4) cooperate to induce eosinophil recruitment into sites of allergic inflammation. Cooperation between inflammatory mediators must be an important phenomenon in vivo, explaining both the ability of lower concentrations of mediators to induce a full-blown functional response and the effectiveness of different strategies at inhibiting these responses.


Subject(s)
Cell Movement/immunology , Chemokines, CC , Cytokines/physiology , Leukotriene B4/biosynthesis , Pleurisy/immunology , Respiratory Hypersensitivity/immunology , Stem Cell Factor/administration & dosage , Animals , Chemokine CCL11 , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/administration & dosage , Chemokine CCL5/analogs & derivatives , Cytokines/administration & dosage , Injections, Intraperitoneal , Injections, Subcutaneous , Leukotriene B4/physiology , Macrophage Inflammatory Proteins/administration & dosage , Male , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Pleura/immunology , Pleura/pathology , Pleurisy/pathology , Receptors, Chemokine/antagonists & inhibitors , Respiratory Hypersensitivity/pathology
14.
J Immunol ; 164(6): 3392-401, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10706735

ABSTRACT

Macrophage inflammatory protein (MIP-1 alpha), a member of the CC chemokine subfamily, has been shown to attract T cells and monocytes in vitro and to be expressed at sites of inflammation. Although the in vitro activities of MIP-1 alpha have been well documented, the in vivo biological activities of MIP-1 alpha in humans have not been studied. To address this, we challenged human subjects by intradermal injection with up to 1000 pmol of MIP-1 alpha and performed biopsies 2, 10, and 24 h later. Although no acute cutaneous or systemic reactions were noted, endothelial cell activation, as indicated by the expression of E-selectin, was observed. In agreement with its in vitro activity, monocyte, lymphocyte, and, to a lesser degree, eosinophil infiltration was observed, peaking at 10-24 h. Surprisingly, in contrast to its reported lack of in vitro neutrophil-stimulating activity, a rapid infiltration of neutrophils was observed in vivo. This neutrophil infiltration occurred as early as 2 h, preceding the appearance of other cells, and peaked at 10 h. Interestingly, we found that neutrophils in whole blood, but not after isolation, expressed CCR1 on their cell surface. This CCR1 was thought to be functional as assessed by neutrophil CD11b up-regulation following whole-blood MIP-1 alpha stimulation. These studies substantiate the biological effects of MIP-1 alpha on monocytes and lymphocytes and uncover the previously unrecognized activity of MIP-1 alpha to induce neutrophil infiltration and endothelial cell activation, underscoring the need to evaluate chemokines in vivo in humans.


Subject(s)
Cell Movement/immunology , Macrophage Inflammatory Proteins/administration & dosage , Monocytes/immunology , Neutrophils/immunology , Adolescent , Adult , Cell Line , Chemokine CCL4 , E-Selectin/biosynthesis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Humans , Injections, Intradermal , Macrophage Inflammatory Proteins/pharmacology , Macrophage Inflammatory Proteins/physiology , Male , Middle Aged , Neutrophils/metabolism , Receptors, CCR1 , Receptors, CCR5/biosynthesis , Receptors, Chemokine/biosynthesis , Skin/cytology
15.
J Immunol ; 164(5): 2650-9, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10679105

ABSTRACT

The role of the CC chemokines, macrophage inflammatory protein-1 beta (MIP-1 beta), monocyte chemotactic peptide-1 (MCP-1), and RANTES, in acute lung inflammatory injury induced by intrapulmonary deposition of IgG immune complexes injury in rats was determined. Rat MIP-1 beta, MCP-1, and RANTES were cloned, the proteins were expressed, and neutralizing Abs were developed. mRNA and protein expression for MIP-1 beta and MCP-1 were up-regulated during the inflammatory response, while mRNA and protein expression for RANTES were constitutive and unchanged during the inflammatory response. Treatment of rats with anti-MIP-1 beta Ab significantly decreased vascular permeability by 37% (p = 0.012), reduced neutrophil recruitment into lung by 65% (p = 0.047), and suppressed levels of TNF-alpha in bronchoalveolar lavage fluids by 61% (p = 0.008). Treatment of rats with anti-rat MCP-1 or anti-rat RANTES had no effect on the development of lung injury. In animals pretreated intratracheally with blocking Abs to MCP-1, RANTES, or MIP-1 beta, significant reductions in the bronchoalveolar lavage content of these chemokines occurred, suggesting that these Abs had reached their targets. Conversely, exogenously MIP-1 beta, but not RANTES or MCP-1, caused enhancement of the lung vascular leak. These data indicate that MIP-1 beta, but not MCP-1 or RANTES, plays an important role in intrapulmonary recruitment of neutrophils and development of lung injury in the model employed. The findings suggest that in chemokine-dependent inflammatory responses in lung CC chemokines do not necessarily demonstrate redundant function.


Subject(s)
Chemokine CCL2/physiology , Chemokine CCL5/physiology , Chemokines, CC/physiology , Lung/immunology , Lung/pathology , Macrophage Inflammatory Proteins/physiology , Acute Disease , Animals , Antibodies, Blocking/administration & dosage , Antigen-Antibody Complex/toxicity , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL2/administration & dosage , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/genetics , Chemokine CCL4 , Chemokine CCL5/administration & dosage , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Chemokines, CC/administration & dosage , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/genetics , Chemotaxis, Leukocyte/immunology , Cloning, Molecular , Immune Sera/administration & dosage , Immunoglobulin G/toxicity , Intubation, Intratracheal , Lung/metabolism , Macrophage Inflammatory Proteins/administration & dosage , Macrophage Inflammatory Proteins/antagonists & inhibitors , Macrophage Inflammatory Proteins/genetics , Male , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , RNA, Messenger/biosynthesis , Rats , Rats, Long-Evans , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology
16.
Dig Dis Sci ; 45(12): 2306-12, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11258549

ABSTRACT

BB10010/MIP-1 alpha reduces the number of proliferating cells in the small intestine, strongly suggesting a radioprotective potential in this organ. This study was designed to optimize BB10010 administration for maximal radioprotection. In single administration protocols 1 or 4 mg/kg of BB10010 was injected into mice 2, 4 or 10 hr before death. In double administration protocols an initial dose of either 0.4 or 200 microg/kg, and a second dose (2.5 hr apart) of 200 microg/kg 4 hr before death were administered. The number of vincristine-arrested metaphases were counted on individually microdissected crypts from the midpoint of the small intestine. When compared to the smaller doses of BB 10010 used in our previous studies, the higher doses used in these experiments did not result in any further reduction in the number of proliferating cells under any of the protocols assessed. Furthermore, some values were found to be above not only those observed with the smaller doses, but also above untreated controls. It is concluded that a single dose of 200 microg/kg of BB10010 offers the most consistent reduction of mitotic cells, and is, therefore, considered optimal for assessment of radioprotection.


Subject(s)
Intestine, Small/cytology , Macrophage Inflammatory Proteins/administration & dosage , Animals , Chemokine CCL3 , Chemokine CCL4 , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelium/drug effects , Intestine, Small/drug effects , Male , Metaphase , Mice
17.
Eur J Cancer ; 34(7): 1023-9, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9849450

ABSTRACT

BB-10010 is a genetically engineered variant of human macrophage inflammatory protein-1 alpha (hMIP-1 alpha) with improved pharmaceutical formulation properties. Although initially described as a pro-inflammatory cytokine, it is now recognised that hMIP-1 alpha has additional effects on haemopoietic stem cell cycling and on human immunodeficiency virus uptake by macrophages. In view of the potential clinical utility of the molecule, we have embarked on a clinical trials programme to evaluate the safety, tolerability and haematological effects of BB-10010. We now report the results of two phase I clinical studies in which 49 subjects (9 patients with advanced breast carcinoma and 40 normal healthy volunteers) received escalating doses of BB-10010, from 0.1 to 300 micrograms/kg using the subcutaneous (s.c.) or intravenous route (i.v.) of administration. Treatment was associated with a dose-related increase in monocyte count which peaked at 200% of steady-state levels and was preceded by an acute, short-lived, monocytopenia, 50-100% of baseline. no measurable effects were noted on other leucocyte subsets or on circulating progenitor cell numbers. In all cases, BB-10010 was extremely well tolerated with no significant toxicity observed at any dose level and a maximum tolerated dose was not defined. Pharmacokinetic analysis revealed that serum concentrations of BB-10010 were detectable using doses of > or = 10 micrograms/kg i.v. or > or = 30 micrograms/kg s.c., and that a single s.c. injection resulted in sustained plasma levels over a 24 h period. These preliminary studies have confirmed the safety and tolerability of BB-10010 using a dose range up to 300 micrograms/kg. Further clinical studies are ongoing to determine the biological effects and to investigate the potential myeloprotective properties using a variable dose range and schedule of BB-10010 in combination with cytotoxic chemotherapy.


Subject(s)
Breast Neoplasms/therapy , Macrophage Inflammatory Proteins/administration & dosage , Adult , Aged , Chemokine CCL3 , Chemokine CCL4 , Dose-Response Relationship, Drug , Female , Hematopoietic Stem Cells/physiology , Humans , Infusions, Intravenous , Injections , Leukocyte Count , Leukocytes/physiology , Macrophage Inflammatory Proteins/adverse effects , Macrophage Inflammatory Proteins/pharmacokinetics , Male , Middle Aged
18.
Neuroreport ; 9(11): 2519-22, 1998 Aug 03.
Article in English | MEDLINE | ID: mdl-9721925

ABSTRACT

It has been hypothesized that endogenous glucocorticoids represent an important negative feedback system involved in the modulation of cytokine-induced fever through the inhibition of prostaglandins (PG) production in the preoptic anterior hypothalamus (AH/POA). The purpose of this study was to determinate whether glucocorticoids modulate the PG-independent febrile response induced by macrophage inflammatory protein-1beta (MIP-1beta) in a manner similar to other pyrogenic cytokines. Subcutaneous pretreatment with dexamethasone (1, 2 and 4 mg/kg; 1 h) had no effect on fever induced by microinjection of 50 pg MIP-1beta into the rat's AH/POA. It is demonstrated for the first time that, unlike other cytokines, fever induced by MIP-1beta is independent of glucocorticoid modulation. Finally, these results offer new perspectives about the pathogenesis of glucocorticoid-unresponsive pyrexia.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Fever/chemically induced , Macrophage Inflammatory Proteins/pharmacology , Animals , Body Temperature/drug effects , Chemokine CCL4 , Fever/drug therapy , Macrophage Inflammatory Proteins/administration & dosage , Male , Microinjections , Preoptic Area , Rats , Rats, Wistar
19.
Br J Cancer ; 75(12): 1715-20, 1997.
Article in English | MEDLINE | ID: mdl-9192972

ABSTRACT

Macrophage inflammatory protein 1alpha (MIP-1alpha) inhibits haemopoietic stem cell proliferation. This property has been exploited in a murine chemotherapy model and has been shown to ameliorate cytotoxic-induced myelosuppression after S-phase-specific cytotoxic therapy. We have now shown that BB-10010, a stable mutant of MIP-1alpha, (a) is more effective when administered as a continuous infusion than when bolus injected and (b), when administered via a 7-day infusion during and after cyclophosphamide treatment, results in an earlier recovery of leucocyte numbers. This effect was accompanied by progenitor cell mobilization into the peripheral blood and included primitive cells with marrow-repopulating ability (MRA). Maximal mobilization and recovery of leucocytes occurred when MIP-1alpha was combined with granulocyte colony-stimulating factor (G-CSF) therapy. The findings suggest that MIP1-alpha used alone or in combination with G-CSF may allow delivery of a greater chemotherapy dose intensity as a consequence of both accelerated leucocyte recovery and maintenance of high-quality mobilized progenitor cells for harvesting and peripheral blood stem cell transplantation.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Bone Marrow/drug effects , Cyclophosphamide/pharmacology , Hematopoietic Stem Cells/drug effects , Leukocytes/drug effects , Macrophage Inflammatory Proteins/administration & dosage , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/therapeutic use , Bone Marrow/radiation effects , Chemokine CCL3 , Chemokine CCL4 , Colony-Forming Units Assay , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Data Interpretation, Statistical , Drug Therapy, Combination , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Transplantation , Infusions, Parenteral , Leukocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Radiation Dosage , Recombinant Proteins , Time Factors
20.
J Physiol ; 491 ( Pt 1): 209-17, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-9011612

ABSTRACT

1. The microinjection of macrophage inflammatory protein-1 (MIP-1 alpha; 5.0 and 25 pg) into the anterior hypothalamic, preoptic area (AHPOA) induced a slow onset; monophasic fever in rats that persisted for a long period. Microinjection of 25 pg MIP-1 beta into the AHPOA induced a fever of rapid onset, whereas 5.0 pg MIP-1 beta did not alter body temperature (Tb) significantly. When either MIP-1 alpha or MIP-1 beta was heated to 70 degrees C for 30 min prior to their injection, no pyrexic response was produced. 2. The concurrent microinjection of 25 pg MIP-1 alpha and 25 pg MIP-1 beta into the AHPOA attenuated the effects on Tb of either cytokine alone. However, pretreatment with either 5.0 pg MIP-1 beta or 5.0 pg MIP-1 alpha suppressed the febrile response induced by 25 pg MIP-1 alpha or 25 pg MIP-1 beta, given at the same site, respectively. 3. The present experiments show that MIP-1 alpha and MIP-1 beta are active individually and possess distinct differences in their evocation of a febrile response. Further, our results suggest a functional antagonism between MIP-1 alpha and MIP-1 beta that could represent a new level in the development of fever.


Subject(s)
Fever/physiopathology , Hypothalamus/physiopathology , Macrophage Inflammatory Proteins/pharmacology , Animals , Body Temperature/drug effects , Body Temperature/physiology , Chemokine CCL4 , Fever/chemically induced , Hypothalamus/anatomy & histology , Kinetics , Macrophage Inflammatory Proteins/administration & dosage , Male , Microinjections , Preoptic Area/anatomy & histology , Rats , Rats, Wistar , Telemetry
SELECTION OF CITATIONS
SEARCH DETAIL
...