Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Toxicol Sci ; 199(2): 246-260, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38310335

ABSTRACT

Particulate matter (PM) containing environmentally persistent free radicals (EPFR) is formed by the incomplete combustion of organic wastes, resulting in the chemisorption of pollutants to the surface of PM containing redox-active transition metals. In prior studies in mice, EPFR inhalation impaired endothelium-dependent vasodilation. These findings were associated with aryl hydrocarbon receptor (AhR) activation in the alveolar type-II (AT-II) cells that form the air-blood interface in the lung. We thus hypothesized that AhR activation in AT-II cells promotes the systemic release of mediators that promote endothelium dysfunction peripheral to the lung. To test our hypothesis, we knocked down AhR in AT-II cells of male and female mice and exposed them to 280 µg/m3 EPFR lo (2.7e + 16 radicals/g) or EPFR (5.5e + 17 radicals/g) compared with filtered air for 4 h/day for 1 day or 5 days. AT-II-AhR activation-induced EPFR-mediated endothelial dysfunction, reducing endothelium-dependent vasorelaxation by 59%, and eNOS expression by 50%. It also increased endothelin-1 mRNA levels in the lungs and peptide levels in the plasma in a paracrine fashion, along with soluble vascular cell adhesion molecule-1 and iNOS mRNA expression, possibly via NF-kB activation. Finally, AhR-dependent increases in antioxidant response signaling, coupled to increased levels of 3-nitrotyrosine in the lungs of EPFR-exposed littermate control but not AT-II AhR KO mice suggested that ATII-specific AhR activation promotes oxidative and nitrative stress. Thus, AhR activation at the air-blood interface mediates endothelial dysfunction observed peripheral to the lung, potentially via release of systemic mediators.


Subject(s)
Mice, Inbred C57BL , Particulate Matter , Receptors, Aryl Hydrocarbon , Animals , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Male , Particulate Matter/toxicity , Female , Free Radicals/metabolism , Air Pollutants/toxicity , Mice , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Oxidative Stress/drug effects , Inhalation Exposure , Lung/drug effects , Lung/metabolism , Lung/blood supply , Endothelin-1/metabolism , Vasodilation/drug effects , Nitric Oxide Synthase Type III/metabolism , Basic Helix-Loop-Helix Transcription Factors
2.
BMC Vet Res ; 17(1): 104, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33663494

ABSTRACT

BACKGROUND: Domestic cats rarely develop hepatocellular carcinoma. The reason for the low prevalence is unknown. Reductions in hepatocellular ploidy have been associated with hepatic carcinogenesis. Recent work in mice has shown that livers with more polyploid hepatocytes are protected against the development of hepatocellular carcinoma. Hepatocyte ploidy in the domestic cat has not been evaluated. We hypothesized that ploidy would be reduced in peri-tumoral and neoplastic hepatocytes compared to normal feline hepatocytes. Using integrated fluorescence microscopy, we quantified the spectra of ploidy in hepatocellular carcinoma and healthy control tissue from paraffin embedded tissue sections. RESULTS: Feline hepatocytes are predominantly mononuclear and the number of nuclei per hepatocyte did not differ significantly between groups. Normal cats have a greater number of tetraploid hepatocytes than cats with hepatocellular carcinoma. CONCLUSIONS: Total hepatocellular polyploidy in normal cat liver is consistent with values reported in humans, yet cellular ploidy (nuclei per cell) is greater in humans than in cats. Tetraploid cat hepatocytes are predominantly mononuclear.


Subject(s)
Carcinoma, Hepatocellular/veterinary , Cat Diseases/genetics , Hepatocytes/cytology , Ploidies , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cat Diseases/pathology , Cats , Female , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/veterinary , Male
3.
Nanomedicine (Lond) ; 11(13): 1653-69, 2016 07.
Article in English | MEDLINE | ID: mdl-27347781

ABSTRACT

AIM: Quantify the biodistribution and assess the toxicity of PLGA (poly-lactic-co-glycolic acid) and surface-modified PLGA chitosan (PLGA/Chi) nanoparticles (NPs) orally administered for 7, 14 and 21 days to F344 rats. MATERIALS & METHODS: Fluorescent NPs were tracked in F344 rat tissues, and toxicity was evaluated by alkaline phosphatase and alanine transaminase levels, and by histologic examination of tissue samples. RESULTS: Biodistribution of PLGA and PLGA/Chi were similar, with highest amounts found in the intestine and liver. Alkaline phosphatase increased significantly in treated rats. Mild histological differences were detected in the intestine and liver. CONCLUSION: PLGA and PLGA/Chi NPs behaved similarly presenting minimal toxicity in the liver and intestine, but not in kidney, lung and brain.


Subject(s)
Lactic Acid/chemistry , Lactic Acid/pharmacokinetics , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacokinetics , Administration, Oral , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Chitosan/chemistry , Drug Carriers , Humans , Lactic Acid/toxicity , Nanoparticles/toxicity , Particle Size , Polyglycolic Acid/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Inbred F344 , Surface Properties , Tissue Distribution , Toxicity Tests, Subacute
4.
J Virol ; 88(13): 7618-27, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24760889

ABSTRACT

UNLABELLED: The herpes simplex virus type 1 (HSV-1) UL20 gene encodes a 222-amino-acid nonglycosylated envelope protein which forms a complex with viral glycoprotein K (gK) that functions in virion envelopment, egress, and virus-induced cell fusion. To investigate the role of the carboxyl terminus of the UL20 protein (UL20p) in cytoplasmic virion envelopment, a cadre of mutant viruses was constructed and characterized. The deletion of six amino acids from the carboxyl terminus of UL20p caused an approximately 1-log reduction in infectious virus production compared to that of the wild-type virus. Surprisingly, a phenylalanine-to-alanine replacement at amino acid position 210 caused a gain-of-function phenotype, increasing infectious virus production up to 1 log more than in the wild-type virus. In contrast, the replacement of two membrane-proximal phenylalanines with alanines caused drastic inhibition of infectious virion production and cytoplasmic virion envelopment. Prediction of the membrane topology of UL20p revealed that these two amino acid changes cause retraction of the carboxyl terminus of UL20p from the intracellular space. Confocal microscopy revealed that none of the engineered UL20 mutations affected intracellular transport of UL20p to trans-Golgi network membranes. In addition, a proximity ligation assay showed that none of the UL20 mutations affected UL20p colocalization and potential interactions with the UL37 protein recently found to interact with the gK/UL20 protein complex. Collectively, these studies show that phenylalanine residues within the carboxyl terminus of UL20p are involved in the regulation of cytoplasmic virion envelopment and infectious virus production. IMPORTANCE: We have shown previously that the UL20/gK protein complex serves crucial roles in cytoplasmic virion envelopment and that it interacts with the UL37 tegument protein to facilitate cytoplasmic virion envelopment. In this study, we investigated the role of phenylalanine residues within the carboxyl terminus of UL20p, since aromatic and hydrophobic amino acids are known to be involved in protein-protein interactions through stacking of their aromatic structures. Characterization of mutant viruses carrying phenylalanine (Phe)-to-alanine (Ala) mutations revealed that the two membrane-proximal Phe residues were critical for the proper UL20p membrane topology and efficient virion envelopment and infectious virus production. Surprisingly, a Phe-to-Ala change located approximately in the middle of the UL20p carboxyl terminus substantially enhanced cytoplasmic envelopment and overall production of infectious virions. This work revealed that Phe residues within the UL20p carboxyl terminus are involved in the regulation of cytoplasmic virion envelopment and infectious virus production.


Subject(s)
Cytoplasm/virology , Glycoproteins/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/growth & development , Phenylalanine/metabolism , Viral Proteins/metabolism , Virion/metabolism , Animals , Cell Fusion , Chlorocebus aethiops , Herpes Simplex/metabolism , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Microscopy, Electron , Mutation/genetics , Phenotype , Phenylalanine/genetics , Vero Cells , Viral Proteins/genetics , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Virion/pathogenicity , trans-Golgi Network
5.
Infect Immun ; 82(6): 2329-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24664505

ABSTRACT

Previous in vitro work demonstrated that Edwardsiella ictaluri produces an acid-activated urease that can modulate environmental pH through the production of ammonia from urea. Additional work revealed that expression of the E. ictaluri type III secretion system (T3SS) is upregulated by acidic pH. Both the urease and the T3SS were previously shown to be essential to intracellular replication. In this work, fluorescence microscopy with LysoTracker Red DND-99 (LTR) indicated that E. ictaluri-containing vacuoles (ECV) became acidified following ingestion by head kidney-derived macrophages (HKDM). In vivo ratiometric imaging demonstrated a lowered ECV pH, which fell to as low as pH 4 but subsequently increased to pH 6 or greater. Inhibition of vacuolar H(+)-ATPases by use of the specific inhibitor bafilomycin A1 abrogated both ECV acidification and intracellular replication in HKDM. Failure of an E. ictaluri urease knockout mutant to increase the ECV pH in the in vivo ratiometric assay suggests that ammonia produced by the urease reaction mediates the pH increase. Additionally, when the specific arginase inhibitor l-norvaline was used to treat E. ictaluri-infected HKDM, the ECV failed to neutralize and E. ictaluri was unable to replicate. This indicates that the HKDM-encoded arginase enzyme produces the urea used by the E. ictaluri urease enzyme. Failure of the ECV to acidify would prevent both upregulation of the T3SS and activation of the urease enzyme, either of which would prevent E. ictaluri from replicating in HKDM. Failure of the ECV to neutralize would result in a vacuolar pH too low to support E. ictaluri replication.


Subject(s)
Edwardsiella ictaluri/pathogenicity , Enterobacteriaceae Infections/microbiology , Fish Diseases/microbiology , Hydrogen-Ion Concentration , Macrophages/microbiology , Vacuoles/physiology , Analysis of Variance , Animals , Arginase/metabolism , Disease Models, Animal , Edwardsiella ictaluri/drug effects , Edwardsiella ictaluri/growth & development , Enzyme Inhibitors/pharmacology , Ictaluridae , Microbial Sensitivity Tests , Microscopy, Fluorescence
6.
J Neuroinflammation ; 6: 23, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19706181

ABSTRACT

BACKGROUND: Lyme neuroborreliosis (LNB) may present as meningitis, cranial neuropathy, acute radiculoneuropathy or, rarely, as encephalomyelitis. We hypothesized that glia, upon exposure to Borrelia burgdorferi, the Lyme disease agent, produce inflammatory mediators that promote the acute cellular infiltration of early LNB. This inflammatory context could potentiate glial and neuronal apoptosis. METHODS: We inoculated live B. burgdorferi into the cisterna magna of rhesus macaques and examined the inflammatory changes induced in the central nervous system (CNS), and dorsal root nerves and ganglia (DRG). RESULTS: ELISA of the cerebrospinal fluid (CSF) showed elevated IL-6, IL-8, CCL2, and CXCL13 as early as one week post-inoculation, accompanied by primarily lymphocytic and monocytic pleocytosis. In contrast, onset of the acquired immune response, evidenced by anti-B. burgdorferi C6 serum antibodies, was first detectable after 3 weeks post-inoculation. CSF cell pellets and CNS tissues were culture-positive for B. burgdorferi. Histopathology revealed signs of acute LNB: severe multifocal leptomeningitis, radiculitis, and DRG inflammatory lesions. Immunofluorescence staining and confocal microscopy detected B. burgdorferi antigen in the CNS and DRG. IL-6 was observed in astrocytes and neurons in the spinal cord, and in neurons in the DRG of infected animals. CCL2 and CXCL13 were found in microglia as well as in endothelial cells, macrophages and T cells. Importantly, the DRG of infected animals showed significant satellite cell and neuronal apoptosis. CONCLUSION: Our results support the notion that innate responses of glia to B. burgdorferi initiate/mediate the inflammation seen in acute LNB, and show that neuronal apoptosis occurs in this context.


Subject(s)
Encephalitis/physiopathology , Lyme Neuroborreliosis/physiopathology , Meningitis/physiopathology , Neuroglia/immunology , Radiculopathy/physiopathology , Spinal Cord Diseases/physiopathology , Animals , Antibodies/blood , Apoptosis/immunology , Brain/immunology , Brain/pathology , Brain/physiopathology , Chemokines/metabolism , Encephalitis/immunology , Encephalitis/microbiology , Ganglia, Spinal/immunology , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Gliosis/immunology , Gliosis/microbiology , Gliosis/physiopathology , Leukocytosis/immunology , Leukocytosis/microbiology , Leukocytosis/physiopathology , Lyme Neuroborreliosis/immunology , Lyme Neuroborreliosis/pathology , Macaca mulatta , Meningitis/immunology , Meningitis/microbiology , Nerve Degeneration/immunology , Nerve Degeneration/microbiology , Nerve Degeneration/physiopathology , Neuroglia/microbiology , Neurons/immunology , Neurons/microbiology , Neurons/pathology , Radiculopathy/immunology , Radiculopathy/microbiology , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Diseases/immunology , Spinal Cord Diseases/microbiology
7.
Cancer Res ; 67(18): 8900-5, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875732

ABSTRACT

B7-H4 is a recently identified B7 family member. We previously showed that ovarian tumor and associated macrophages expressed B7-H4; tumor B7-H4+ macrophages and CD4+CD25+FOXP3+ regulatory T cells (Treg cells) suppressed tumor-associated antigen-specific T-cell immunity. To determine the pathologic relationship between B7-H4, macrophages, and Treg cells in the tumor environment, in addition to Treg cell numbers, we quantified B7-H4 expression in the tumor and tumor-associated macrophages in 103 patients with ovarian carcinoma. We observed that the intensity of B7-H4 expression in macrophages was significantly correlated with Treg cell numbers in the tumor. Further, both Treg cells and macrophage B7-H4, but not tumor B7-H4, were negatively associated with patient outcome. Tumor Treg cells enabled macrophages to spontaneously produce interleukin (IL)-10 and IL-6. Tumor macrophages stimulated B7-H4 expression in an autocrine manner through IL-10 and IL-6. Our previous work showed that tumor-associated macrophages spontaneously produced chemokine CCL22 to mediate Treg cell trafficking into tumor, and Treg cells induced B7-H4 on antigen-presenting cells (APC) including macrophages. Altogether, our data support the concept that there is a mechanistic interaction between Treg cells and macrophage, and that Treg cells may convey the suppressive activity to APCs through B7-H4 induction in human ovarian cancer.


Subject(s)
B7-1 Antigen/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Antigen-Presenting Cells/immunology , B7-1 Antigen/biosynthesis , Female , Humans , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Macrophages/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1
8.
J Immunol ; 177(1): 40-4, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785496

ABSTRACT

Multiple modes of suppressive mechanisms including IL-10 are thought to be implicated in CD4+CD25+ regulatory T (Treg) cell-mediated suppression. However, the cellular source, role, and molecular mechanism of IL-10 in Treg cell biology remain controversial. We now studied the interaction between Treg cells and APCs. We demonstrate that Treg cells, but not conventional T cells, trigger high levels of IL-10 production by APCs, stimulate APC B7-H4 expression, and render APCs immunosuppressive. Initial blockade of B7-H4 reduces the suppressive activity mediated by Treg cell-conditioned APCs. Further, APC-derived, rather than Treg cell-derived, IL-10 is responsible for APC B7-H4 induction. Therefore, Treg cells convey suppressive activity to APCs by stimulating B7-H4 expression through IL-10. Altogether, our data provide a novel cellular and molecular mechanism for Treg cell-mediated immunosuppression at the level of APCs, and suggest a plausible mechanism for the suppressive effect of IL-10 in Treg cell-mediated suppression.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B7-1 Antigen/biosynthesis , Down-Regulation/immunology , Interleukin-10/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antibodies, Blocking/pharmacology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-1 Antigen/physiology , Cells, Cultured , Coculture Techniques , Down-Regulation/genetics , Female , Humans , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Up-Regulation/genetics , Up-Regulation/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1
9.
J Exp Med ; 203(4): 871-81, 2006 Apr 17.
Article in English | MEDLINE | ID: mdl-16606666

ABSTRACT

Tumor-associated macrophages are a prominent component of ovarian cancer stroma and contribute to tumor progression. B7-H4 is a recently identified B7 family molecule. We show that primary ovarian tumor cells express intracellular B7-H4, whereas a fraction of tumor macrophages expresses surface B7-H4. B7-H4+ tumor macrophages, but not primary ovarian tumor cells, suppress tumor-associated antigen-specific T cell immunity. Blocking B7-H4-, but not arginase-, inducible nitric oxide synthase or B7-H1 restored the T cell stimulating capacity of the macrophages and contributes to tumor regression in vivo. Interleukin (IL)-6 and IL-10 are found in high concentrations in the tumor microenvironment. These cytokines stimulate macrophage B7-H4 expression. In contrast, granulocyte/macrophage colony-stimulating factor and IL-4, which are limited in the tumor microenvironment, inhibit B7-H4 expression. Ectopic expression of B7-H4 makes normal macrophages suppressive. Thus, B7-H4+ tumor macrophages constitute a novel suppressor cell population in ovarian cancer. B7-H4 expression represents a critical checkpoint in determining host responses to dysfunctional cytokines in ovarian cancer. Blocking B7-H4 or depleting B7-H4+ tumor macrophages may represent novel strategies to enhance T cell tumor immunity in cancer.


Subject(s)
B7-1 Antigen/genetics , Carcinoma/metabolism , Macrophages/immunology , Macrophages/metabolism , Ovarian Neoplasms/metabolism , Animals , Antigens, CD/physiology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , B7-1 Antigen/biosynthesis , B7-H1 Antigen , Biomarkers , Carcinoma/immunology , Female , Humans , Immunophenotyping , Interleukin-10/physiology , Interleukin-6/physiology , Macrophages/classification , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Tumor Cells, Cultured , V-Set Domain-Containing T-Cell Activation Inhibitor 1
10.
Cancer Res ; 65(12): 5020-6, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958543

ABSTRACT

To directly dissect the role of each immune component in human tumor immunopathogenesis, we have studied the interaction between dendritic cells and T cells in the tumor environment of patients with ovarian carcinoma. We previously reported that functional plasmacytoid dendritic cells, but not functionally mature myeloid dendritic cells, accumulated in tumor microenvironments. We now show that tumor ascites macrophage-derived dendritic cells induced tumor-associated antigen-specific CD8+ T cells with effector functions. Strikingly, tumor ascites plasmacytoid dendritic cells induced interleukin-10+ CCR7+ CD45RO+ CD8+ regulatory T cells. Four characteristics have been identified in tumor plasmacytoid dendritic cell-induced CD8+ regulatory T cells: (a) induction of CD8+ regulatory T cells is independent of CD4+ CD25+ T cells; (b) CD8+ regulatory T cells significantly suppress myeloid dendritic cell-mediated tumor-associated antigen-specific T cell effector functions through interleukin-10; (c) repetitive myeloid dendritic cell stimulation can recover CD8+ regulatory T cell-mediated poor T cell proliferation, but not T cell effector function; (d) CD8+ regulatory T cells express functional CCR7, and efficiently migrate with lymphoid homing chemokine MIP-3beta. Primary suppressive CCR7+ CD45RO+ CD8+ T cells are found in the tumor environment of patients with ovarian cancers. Thus, tumor-associated plasmacytoid dendritic cells contribute to the tumor environmental immunosuppressive network. Collectively, tumors manipulate tumor microenvironmental dendritic cell subset distribution and function to subvert tumor immunity. The data are relevant to understanding tumor immunopathology as well as reevaluating tumor immunotherapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Dendritic Cells/immunology , Ovarian Neoplasms/immunology , Amino Acid Sequence , Cell Movement/immunology , Dendritic Cells/cytology , Female , Humans , Interleukin-10/immunology , Leukocyte Common Antigens/immunology , Lymphocyte Activation , Molecular Sequence Data , Myeloid Cells/immunology , Plasma Cells/immunology , Receptors, CCR7 , Receptors, Chemokine/immunology
11.
Cancer Res ; 65(2): 465-72, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15695388

ABSTRACT

Ovarian carcinomas have a poor prognosis, often associated with multifocal i.p. dissemination accompanied by intense neovascularization. To examine tumor angiogenesis in the tumor microenvironment, we studied malignant ascites and tumors of patients with untreated ovarian carcinoma. We observed that malignant ascites fluid induced potent in vivo neovascularization in Matrigel assay. We detected a sizable amount of vascular endothelial cell growth factor (VEGF) in malignant ascites. However, pathologic concentration of VEGF is insufficient to induce in vivo angiogenesis. We show that ovarian tumors strongly express CXC chemokine stromal-derived factor (SDF-1/CXCL12). High concentration of CXCL12, but not the pathologic concentration of CXCL12 induces in vivo angiogenesis. Strikingly, pathologic concentrations of VEGF and CXCL12 efficiently and synergistically induce in vivo angiogenesis. Migration, expansion, and survival of vascular endothelial cells (VEC) form the essential functional network of angiogenesis. We further provide a mechanistic basis for explaining the interaction between CXCL12 and VEGF. We show that VEGF up-regulates the receptor for CXCL12, CXCR4 expression on VECs, and synergizes CXCL12-mediated VEC migration. CXCL12 synergizes VEGF-mediated VEC expansion and synergistically protects VECs from sera starvation-induced apoptosis with VEGF. Finally, we show that hypoxia synchronously induces tumor CXCL12 and VEGF production. Therefore, hypoxia triggered tumor CXCL12 and VEGF form a synergistic angiogenic axis in vivo. Hypoxia-induced signals would be the important factor for initiating and maintaining an active synergistic angiogeneic pathway mediated by CXCL12 and VEGF. Thus, interrupting this synergistic axis, rather than VEGF alone, will be a novel efficient antiangiogenesis strategy to treat cancer.


Subject(s)
Chemokines, CXC/pharmacology , Ovarian Neoplasms/blood supply , Vascular Endothelial Growth Factor A/pharmacology , Animals , Ascites/metabolism , Ascites/pathology , Cell Hypoxia/physiology , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CXCL12 , Chemokines, CXC/biosynthesis , Chemokines, CXC/physiology , Drug Synergism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Humans , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/metabolism , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/physiology
12.
Cancer Res ; 64(22): 8451-5, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15548717

ABSTRACT

CD4(+)CD25(+) regulatory T cells (Tregs) mediate peripheral T-cell homeostasis and contribute to self-tolerance. Their homeostatic and pathologic trafficking is poorly understood. Under homeostatic conditions, we show a relatively high prevalence of functional Tregs in human bone marrow. Bone marrow strongly expresses functional stromal-derived factor (CXCL12), the ligand for CXCR4. Human Tregs traffic to and are retained in bone marrow through CXCR4/CXCL12 signals as shown in chimeric nonobese diabetic/severe combined immunodeficient mice. Granulocyte colony-stimulating factor (G-CSF) reduces human bone marrow CXCL12 expression in vivo, associated with mobilization of marrow Tregs to peripheral blood in human volunteers. These findings show a mechanism for homeostatic Treg trafficking and indicate that bone marrow is a significant reservoir for Tregs. These data also suggest a novel mechanism explaining reduced acute graft-versus-host disease and improvement in autoimmune diseases following G-CSF treatment.


Subject(s)
Bone Marrow Cells/metabolism , CD4 Antigens/immunology , Chemokines, CXC/metabolism , Receptors, CXCR4/metabolism , Receptors, Interleukin-2/immunology , Signal Transduction , T-Lymphocytes/immunology , Base Sequence , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Chemokine CXCL12 , DNA Primers , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Reverse Transcriptase Polymerase Chain Reaction
13.
Nat Med ; 10(9): 942-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15322536

ABSTRACT

Regulatory T (T(reg)) cells mediate homeostatic peripheral tolerance by suppressing autoreactive T cells. Failure of host antitumor immunity may be caused by exaggerated suppression of tumor-associated antigen-reactive lymphocytes mediated by T(reg) cells; however, definitive evidence that T(reg) cells have an immunopathological role in human cancer is lacking. Here we show, in detailed studies of CD4(+)CD25(+)FOXP3(+) T(reg) cells in 104 individuals affected with ovarian carcinoma, that human tumor T(reg) cells suppress tumor-specific T cell immunity and contribute to growth of human tumors in vivo. We also show that tumor T(reg) cells are associated with a high death hazard and reduced survival. Human T(reg) cells preferentially move to and accumulate in tumors and ascites, but rarely enter draining lymph nodes in later cancer stages. Tumor cells and microenvironmental macrophages produce the chemokine CCL22, which mediates trafficking of T(reg) cells to the tumor. This specific recruitment of T(reg) cells represents a mechanism by which tumors may foster immune privilege. Thus, blocking T(reg) cell migration or function may help to defeat human cancer.


Subject(s)
Cell Movement/immunology , Chemokines, CC/metabolism , Immunity, Cellular/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Ascites/immunology , CD4-Positive T-Lymphocytes , Chemokine CCL22 , Chemokines, CC/immunology , DNA-Binding Proteins , Dendritic Cells/immunology , Female , Forkhead Transcription Factors , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Mice , Microscopy, Confocal/methods , Receptors, Interleukin-2
14.
Cancer Res ; 64(16): 5535-8, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313886

ABSTRACT

Angiogenesis is essential for both primary and metastatic tumor growth. Tumor blood vessel formation is complex and regulated by many factors. Ovarian carcinomas have a poor prognosis, often associated with multifocal intraperitoneal dissemination accompanied by intense neovascularization. To examine tumor angiogenesis in the tumor microenvironment, we studied malignant ascites of patients with untreated ovarian carcinoma. We observed high numbers of plasmacytoid dendritic cells (PDCs) and significant stromal-derived factor (CXCL-12/SDF)-1 in their malignant ascites, attracting PDCs into the tumor environment. We now show that tumor-associated PDCs induced angiogenesis in vivo through production of tumor necrosis factor alpha and interleukin 8. By contrast, myeloid dendritic cells (MDCs) were absent from malignant ascites. MDCs derived in vitro suppressed angiogenesis in vivo through production of interleukin 12. Thus, the tumor may attract PDCs to augment angiogenesis while excluding MDCs to prevent angiogenesis inhibition, demonstrating a novel mechanism for modulating tumor neovascularization. Because dendritic cells (DCs) have long been known to affect tumor immunity, our data also implicate DCs in regulation of tumor neoangiogenesis, suggesting a novel role of DCs in tumor pathology.


Subject(s)
Dendritic Cells/physiology , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/blood supply , Animals , Ascites/metabolism , Ascites/pathology , Dendritic Cells/classification , Dendritic Cells/metabolism , Female , Humans , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Interleukin-8/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid Cells/metabolism , Myeloid Cells/pathology , Myeloid Cells/physiology , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/pathology , Tumor Necrosis Factor-alpha/biosynthesis
15.
Nat Med ; 9(5): 562-7, 2003 May.
Article in English | MEDLINE | ID: mdl-12704383

ABSTRACT

Suppression of dendritic cell function in cancer patients is thought to contribute to the inhibition of immune responses and disease progression. Molecular mechanisms of this suppression remain elusive, however. Here, we show that a fraction of blood monocyte-derived myeloid dendritic cells (MDCs) express B7-H1, a member of the B7 family, on the cell surface. B7-H1 could be further upregulated by tumor environmental factors. Consistent with this finding, virtually all MDCs isolated from the tissues or draining lymph nodes of ovarian carcinomas express B7-H1. Blockade of B7-H1 enhanced MDC-mediated T-cell activation and was accompanied by downregulation of T-cell interleukin (IL)-10 and upregulation of IL-2 and interferon (IFN)-gamma. T cells conditioned with the B7-H1-blocked MDCs had a more potent ability to inhibit autologous human ovarian carcinoma growth in non-obese diabetic-severe combined immunodeficient (NOD-SCID) mice. Therefore, upregulation of B7-H1 on MDCs in the tumor microenvironment downregulates T-cell immunity. Blockade of B7-H1 represents one approach for cancer immunotherapy.


Subject(s)
B7-1 Antigen/physiology , Blood Proteins , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Myeloid Cells/immunology , Ovarian Neoplasms/therapy , Peptides , Animals , Antigens, CD , B7-H1 Antigen , Female , Humans , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Lymphocyte Activation , Membrane Glycoproteins , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL