Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Front Immunol ; 11: 573405, 2020.
Article in English | MEDLINE | ID: mdl-33117369

ABSTRACT

The molecule "T cell immunoreceptor with immunoglobulin and ITIM domain," or TIGIT, has recently received much attention as a promising target in the treatment of various malignancies. In spite of the quick progression of anti-TIGIT antibodies into clinical testing both as monotherapy and in combination with programmed cell death-1 (PD-1)-directed immune checkpoint blockade, the molecular mechanism behind the observed therapeutic benefits remains poorly understood. Here we demonstrate, using mouse tumor models, that TIGIT blocking antibodies with functional Fc binding potential induce effective anti-tumor response. Our observations reveal that the anti-TIGIT therapeutic effect is not achieved by depletion of intratumoral regulatory T cells (Treg) or any cell population expressing TIGIT, but instead is mediated by possible "reverse activating signals" through FcγRs on myeloid cells, inducing expression of various mediators such as cytokines and chemokines. Furthermore, we discovered an induction of a robust and persistent granzyme B and perforin response, distinct from a predominantly interferon-γ (IFN-γ)-driven anti-PD-1 blockade. Our observations, for the first time, provide the basis for a mechanistic hypothesis involving the requirement of a functional Fc domain of anti-TIGIT monoclonal antibodies, of which various isotypes are currently under intense clinical investigation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Colonic Neoplasms/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Myeloid Cells/drug effects , Receptors, IgG/metabolism , Receptors, Immunologic/metabolism , Animals , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Granzymes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Signal Transduction , Tumor Burden/drug effects , Tumor Microenvironment
3.
J Immunother Cancer ; 8(2)2020 10.
Article in English | MEDLINE | ID: mdl-33127658

ABSTRACT

BACKGROUND: Programmed cell death protein 1 (PD-1) and CTLA4 combination blockade enhances clinical efficacy in melanoma compared with targeting either checkpoint alone; however, clinical response improvement is coupled with increased risk of developing immune-related adverse events (irAE). Delineating the mechanisms of checkpoint blockade-mediated irAE has been hampered by the lack of animal models that replicate these clinical events. METHODS: We have developed a mouse model of checkpoint blockade-mediated enterocolitis via prolonged administration of an Fc-competent anti-CTLA4 antibody. RESULTS: Sustained treatment with Fc-effector, but not Fc-mutant or Fc-null, anti-CTLA4 antagonist for 7 weeks resulted in enterocolitis. Moreover, combining Fc-null or Fc-mutant CTLA4 antagonists with PD-1 blockade results in potent antitumor combination efficacy indicating that Fc-effector function is not required for combination benefit. CONCLUSION: These data suggest that using CTLA4 antagonists with no Fc-effector function can mitigate gut inflammation associated with anti-CTLA4 antibody therapy yet retain potent antitumor activity in combination with PD-1 blockade.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Inflammation/physiopathology , Programmed Cell Death 1 Receptor/metabolism , Animals , Humans , Mice
4.
J Biotechnol ; 208: 13-21, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26015261

ABSTRACT

Monoclonal antibody (mAb) therapy has been successfully used for the treatment of B-cell lymphomas and is currently extended for the treatment of multiple myeloma (MM). New developments in MM therapeutics have achieved significant survival gains in patients but the disease still remains incurable. Elotuzumab (HuLuc63), an anti-CS1 monoclonal IgG1 antibody, is believed to induce anti-tumor activity and MM cytotoxicity through antibody dependent cellular cytotoxicity (ADCC) and inhibition of MM cell adhesion to bone marrow stromal cells (BMSCs). Modulations of the Fc glycan composition at the N297 site by selective mutations or afucosylation have been explored as strategies to develop bio-better therapeutics with enhanced ADCC activity. Afucosylated therapeutic antibodies with enhanced ADCC activity have been reported to possess greater efficacy in tumor growth inhibition at lower doses when compared to fucosylated therapeutic antibodies. The N-linked glycosylation pathway in Pichia pastoris has been engineered to produce human-like N-linked glycosylation with uniform afucosylated complex type glycans. The purpose of this study was to compare afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris with fucosylated anti-CS1 mAb expressed in mammalian HEK293 cells through in vitro ADCC and in vivo tumor inhibition models. Our results indicate that Fc glycosylation is critical for in vivo efficacy and afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris shows a better in vivo efficacy in tumor regression when compared to fucosylated anti-CS1 mAb expressed in HEK293 cells. Glycoengineered Pichia pastoris could provide an alternative platform for generating homogeneous afucosylated recombinant antibodies where Fc mediated immune effector function is important for efficacy.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neoplasm , Cell Engineering , Multiple Myeloma/drug therapy , Neoplasms, Experimental/drug therapy , Pichia , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Cell Line, Tumor , Glycosylation , HEK293 Cells , Humans , Mice, SCID , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Xenograft Model Antitumor Assays
5.
J Immunol ; 192(12): 6111-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24829407

ABSTRACT

Cancer progression is associated with inflammation, increased metabolic demand, infection, cachexia, and eventually death. Myeloid-derived suppressor cells (MDSCs) commonly expand during cancer and are associated with adaptive immune suppression and inflammatory metabolite production. We propose that cancer-induced cachexia is driven at least in part by the expansion of MDSCs. MDSC expansion in 4T1 mammary carcinoma-bearing hosts is associated with induction of a hepatic acute-phase protein response and altered host energy and fat metabolism, and eventually reduced survival to polymicrobial sepsis and endotoxemia. Similar results are also seen in mice bearing a Lewis lung carcinoma and a C26 colon adenocarcinoma. However, a similar cachexia response is not seen with equivalent growth of the 66C4 subclone of 4T1, in which MDSC expansion does not occur. Importantly, reducing MDSC numbers in 4T1-bearing animals can ameliorate some of these late responses and reduce susceptibility to inflammation-induced organ injury and death. In addition, administering MDSCs from both tumor- and nontumor-bearing mice can produce an acute-phase response. Thus, we propose a previously undescribed mechanism for the development of cancer cachexia, whereby progressive MDSC expansion contributes to changes in host protein and energy metabolism and reduced resistance to infection.


Subject(s)
Cachexia/immunology , Immune Tolerance , Myeloid Cells/immunology , Neoplasms, Experimental/immunology , Animals , Cachexia/etiology , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Myeloid Cells/pathology , Neoplasms, Experimental/pathology
6.
Rapid Commun Mass Spectrom ; 27(23): 2639-47, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24591025

ABSTRACT

RATIONALE: Human genetics studies in African Americans have shown a strong correlation between polymorphisms in the ApoL1 gene and chronic kidney disease (CKD). To gain further insight into the etiology of ApoL1-associated kidney diseases, the determination of circulating levels of both wild type as well as ApoL1 variants could be of significant use. To date, antibodies that discriminate between all three ApoL1 variant forms (wild type, G1 and G2) are not available. We aimed to develop a rapid method for detecting and quantifying ApoL1 variants and total levels in plasma. METHODS: Ultra-performance liquid chromatography (UPLC) and tandem mass spectrometry (MS/MS) in multiple-reaction monitoring acquisition mode was used to quantify ApoL1. RESULTS: We demonstrated that it is feasible to detect and quantify ApoL1 variants (wild type, G1 and G2), and total ApoL1 concentrations in plasma. ApoL1 genotypes determined by LC/MS agreed perfectly with the traditional method DNA sequencing for 74 human subjects. The method exhibited at least three orders of linearity with a lower limit of quantification of 10 nM. Moreover, the method can readily be multiplexed for the quantification of a panel of protein markers in a single sample. CONCLUSIONS: The method reported herein obviates the need to perform DNA genotyping of ApoL1 variants, which is of significant value in cases where stored samples are unsuitable for DNA analysis. More importantly, the method could potentially be of use in the early identification of individuals at risk of developing CKD, and for the stratification of patients for treatment with future ApoL1-modifying therapies.


Subject(s)
Apolipoproteins/blood , Apolipoproteins/genetics , Chromatography, High Pressure Liquid/methods , Genetic Variation , Kidney Diseases/blood , Lipoproteins, HDL/blood , Lipoproteins, HDL/genetics , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Animals , Apolipoprotein L1 , Genotype , Humans , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data
7.
Nat Med ; 18(7): 1069-76, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22772566

ABSTRACT

The spondyloarthropathies are a group of rheumatic diseases that are associated with inflammation at anatomically distal sites, particularly the tendon-bone attachments (entheses) and the aortic root. Serum concentrations of interleukin-23 (IL-23) are elevated and polymorphisms in the IL-23 receptor are associated with ankyosing spondylitis, however, it remains unclear whether IL-23 acts locally at the enthesis or distally on circulating cell populations. We show here that IL-23 is essential in enthesitis and acts on previously unidentified IL-23 receptor (IL-23R)(+), RAR-related orphan receptor γt (ROR-γt)(+)CD3(+)CD4(-)CD8(-), stem cell antigen 1 (Sca1)(+) entheseal resident T cells. These cells allow entheses to respond to IL-23 in vitro-in the absence of further cellular recruitment--and to elaborate inflammatory mediators including IL-6, IL-17, IL-22 and chemokine (C-X-C motif) ligand 1 (CXCL1). Notably, the in vivo expression of IL-23 is sufficient to phenocopy the human disease, with the specific and characteristic development of enthesitis and entheseal new bone formation in the initial complete absence of synovitis. As in the human condition, inflammation also develops in vivo at the aortic root and valve, which are structurally similar to entheses. The presence of these entheseal resident cells and their production of IL-22, which activates signal transducer and activator of transcription 3 (STAT3)-dependent osteoblast-mediated bone remodeling, explains why dysregulation of IL-23 results in inflammation at this precise anatomical site.


Subject(s)
Interleukin-23/immunology , Spondylarthropathies/immunology , T-Lymphocytes/immunology , Tendons/immunology , Animals , Antigens, CD/metabolism , Aorta/pathology , Arthritis, Experimental/complications , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Bone Remodeling , CD3 Complex/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Disease Models, Animal , Extremities/pathology , Flow Cytometry , Humans , Immunization, Passive , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Interleukin-17 , Interleukins , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Osteogenesis/immunology , Periosteum/growth & development , Receptors, Interleukin/metabolism , Spondylarthropathies/complications , Spondylarthropathies/pathology , Tendons/pathology , Th17 Cells , Interleukin-22
8.
Cancer Res ; 72(14): 3570-81, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22581824

ABSTRACT

The presence of activated intratumoral T cells correlates clinically with better prognosis in patients with cancer. Although tumor vaccines can increase the number of tumor-specific CD8(+) T cells in systemic circulation, they frequently fail to increase the number of active and tumor reactive T cells within the tumor. Here we show that treatment with the pleiotropic cytokine interleukin-10 (IL-10) induces specific activation of tumor-resident CD8(+) T cells as well as their intratumoral expansion in several mouse tumor models. We found that inhibition of T-cell trafficking from lymphoid organs did not impair IL-10-induced tumor rejection or the activation of tumor-resident CD8(+) T cells. Tumor-resident CD8(+) T cells expressed elevated levels of the IL-10 receptor and were directly activated by IL-10, resulting in prominent phosphorylation of STAT3 and STAT1. Although CD4(+) T cells, regulatory T cells, NK cells, and dendritic cells have been reported as prominent targets of IL-10 in the tumor microenvironment, we found that expression of the IL-10R was required only on CD8(+) T cells to facilitate IL-10-induced tumor rejection as well as in situ expansion and proliferation of tumor-resident CD8 T cells. Together, our findings indicate that IL-10 activates CD8(+) T-cell-mediated tumor control and suggest that IL-10 may represent a potential tumor immunotherapy in human patients with cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-10/pharmacology , Lymphocyte Activation/immunology , Neoplasms/immunology , Animals , Female , Humans , Lymphatic System/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin-10/metabolism , T-Lymphocytes, Regulatory/immunology
9.
Cancer Cell ; 20(6): 781-96, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22172723

ABSTRACT

Tumor immune surveillance and cancer immunotherapies are thought to depend on the intratumoral infiltration of activated CD8(+) T cells. Intratumoral CD8(+) T cells are rare and lack activity. IL-10 is thought to contribute to the underlying immune suppressive microenvironment. Defying those expectations we demonstrate that IL-10 induces several essential mechanisms for effective antitumor immune surveillance: infiltration and activation of intratumoral tumor-specific cytotoxic CD8(+) T cells, expression of the Th1 cytokine interferon-γ (IFNγ) and granzymes in CD8(+) T cells, and intratumoral antigen presentation molecules. Consequently, tumor immune surveillance is weakened in mice deficient for IL-10 whereas transgenic overexpression of IL-10 protects mice from carcinogenesis. Treatment with pegylated IL-10 restores tumor-specific intratumoral CD8(+) T cell function and controls tumor growth.


Subject(s)
Interferon-gamma/metabolism , Interleukin-10/metabolism , Neoplasms, Experimental/immunology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Granzymes/metabolism , Humans , Interferon-gamma/genetics , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10 Receptor alpha Subunit/genetics , Interleukin-10 Receptor alpha Subunit/metabolism , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Perforin/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Spleen/metabolism , Transplantation, Heterologous , Tumor Burden , Tumor Escape
10.
PLoS One ; 6(7): e22256, 2011.
Article in English | MEDLINE | ID: mdl-21799806

ABSTRACT

C17 was first described about ten years ago as a gene expressed in CD34+ cells. A more recent study has suggested a role for C17 in chondrogenesis and development of cartilage. However, based on sequence analysis, we believe that C17 has homology to IL-2 and hence we present the hypothesis that C17 is a cytokine possessing immune-regulatory properties. We provide evidence that C17 is a secreted protein preferentially expressed in chondrocytes, hence in cartilage-rich tissues. Systemic expression of C17 in vivo reduces disease in a collagen antibody-induced arthritis model in mice (CAIA). Joint protection is evident by delayed disease onset, minimal edema, bone protection and absence of diverse histological features of disease. Expression of genes typically associated with acute joint inflammation and erosion of cartilage or bone is blunted in the presence of C17. Consistent with the observed reduction in bone erosion, we demonstrate reduced levels of RANKL in the paws and sera of mice over-expressing C17. Administration of C17 at the peak of disease, however, had no effect on disease progression, indicating that C17's immune-regulatory activity must be most prominent prior to or at the onset of severe joint inflammation. Based on this data we propose C17 as a cytokine that s contributes to immune homeostasis systemically or in a tissue-specific manner in the joint.


Subject(s)
Arthritis/metabolism , Blood Proteins/metabolism , Cytokines/metabolism , Joints/metabolism , Joints/pathology , Amino Acid Sequence , Animals , Arthritis/immunology , Arthritis/pathology , Arthritis/therapy , Biomarkers/metabolism , Blood Proteins/chemistry , Blood Proteins/genetics , Bone Diseases/complications , Bone Diseases/metabolism , Cartilage/metabolism , Chondrocytes/metabolism , Cytokines/chemistry , Cytokines/genetics , Gene Expression Regulation , HEK293 Cells , Homeostasis/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Joints/immunology , Male , Mice , Molecular Sequence Data , RANK Ligand/blood
11.
J Immunol ; 187(2): 911-8, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21690321

ABSTRACT

Neutrophils are essential for successful host eradication of bacterial pathogens and for survival to polymicrobial sepsis. During inflammation, the bone marrow provides a large reserve of neutrophils that are released into the peripheral circulation where they traverse to sites of infection. Although neutrophils are essential for survival, few studies have investigated the mechanisms responsible for neutrophil mobilization from the bone marrow during polymicrobial sepsis. Using a cecal ligation and puncture model of polymicrobial sepsis, we demonstrated that neutrophil mobilization from the bone marrow is not dependent on TLR4, MyD88, TRIF, IFNARα/ß, or CXCR2 pathway signaling during sepsis. In contrast, we observed that bone marrow CXCL12 mRNA abundance and specific CXCL12 levels are sharply reduced, whereas splenic CXCR4 mRNA and cell surface expression are increased during sepsis. Blocking CXCL12 activity significantly reduced blood neutrophilia by inhibiting bone marrow release of granulocytes during sepsis. However, CXCL12 inhibition had no impact on the expansion of bone marrow neutrophil precursors and hematopoietic progenitors. Bone marrow neutrophil retention by CXCL12 blockade prevented blood neutrophilia, inhibited peritoneal neutrophil accumulation, allowed significant peritoneal bacterial invasion, and increased polymicrobial sepsis mortality. We concluded that changes in the pattern of CXCL12 signaling during sepsis are essential for neutrophil bone marrow mobilization and host survival but have little impact on bone marrow granulopoiesis.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Chemokine CXCL12/physiology , Neutrophil Infiltration/immunology , Sepsis/immunology , Sepsis/microbiology , Signal Transduction/immunology , Acute Disease , Animals , Bone Marrow Cells/microbiology , Chemokine CXCL12/genetics , Mice , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelopoiesis/genetics , Myelopoiesis/immunology , Neutrophil Infiltration/genetics , Sepsis/mortality , Signal Transduction/genetics , Survival Analysis
12.
J Immunol ; 187(2): 951-9, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21670317

ABSTRACT

The role of IL-23 in the development of arthritis and bone metabolism was studied using systemic IL-23 exposure in adult mice via hydrodynamic delivery of IL-23 minicircle DNA in vivo and in mice genetically deficient in IL-23. Systemic IL-23 exposure induced chronic arthritis, severe bone loss, and myelopoiesis in the bone marrow and spleen, which resulted in increased osteoclast differentiation and systemic bone loss. The effect of IL-23 was partly dependent on CD4(+) T cells, IL-17A, and TNF, but could not be reproduced by overexpression of IL-17A in vivo. A key role in the IL-23-induced arthritis was made by the expansion and activity of myeloid cells. Bone marrow macrophages derived from IL-23p19(-/-) mice showed a slower maturation into osteoclasts with reduced tartrate-resistant acid phosphatase-positive cells and dentine resorption capacity in in vitro osteoclastogenesis assays. This correlated with fewer multinucleated osteoclast-like cells and more trabecular bone volume and number in 26-wk-old male IL-23p19(-/-) mice compared with control animals. Collectively, our data suggest that systemic IL-23 exposure induces the expansion of a myeloid lineage osteoclast precursor, and targeting IL-23 pathway may combat inflammation-driven bone destruction as observed in rheumatoid arthritis and other autoimmune arthritides.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Bone Resorption/immunology , Cell Differentiation/immunology , Interleukin-23 Subunit p19/physiology , Osteoclasts/immunology , Osteoclasts/pathology , Animals , Arthritis, Experimental/genetics , Bone Resorption/genetics , Bone Resorption/pathology , CHO Cells , Cell Differentiation/genetics , Chronic Disease , Cricetinae , Cricetulus , DNA, Kinetoplast/biosynthesis , DNA, Kinetoplast/genetics , HEK293 Cells , Humans , Interleukin-23 Subunit p19/deficiency , Interleukin-23 Subunit p19/isolation & purification , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Severity of Illness Index , Spleen/immunology , Spleen/metabolism , Spleen/pathology
13.
Int Immunopharmacol ; 11(7): 780-2, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21315784

ABSTRACT

Recent investigations of regulatory myeloid cell mobilization and amplification in response to cancer and chronic inflammation associated with infectious disease and autoimmunity have provided numerous insights into the suppressive mechanisms, pathobiology and potential pharmaceutical modulation of MDSC. Controversies have emerged as hematology investigators, have suggested that regulatory myeloid cells, including MDSC merely reflect normal physiological responses to inflammatory and growth factor stimuli. A recurring theme at the recent "International Immunopharmacology Conference on Regulatory Myeloid cells" held in Arlington, Virginia, October 21-24, 2010, focused on efforts to more clearly elucidate molecular features of MDSC from the immature myeloid cells observed in the circulation in response to acute infection and inflammatory responses. Evidence for the potential contribution of regulatory myeloid cells to pathologic processes and sequelae associated with numerous inflammatory and malignant diseases also suggested that these cells may represent a significant deviation from the normal physiologic responses to acute inflammatory stimuli. The relevance of these research efforts was evident from the discussions by numerous clinical investigations regarding their efforts to modulate MDSC in cancer patients to augment immunotherapeutic treatments.


Subject(s)
Autoimmune Diseases/immunology , Immunotherapy , Infections/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Animals , Autoimmune Diseases/therapy , Biomarkers/metabolism , Cell Differentiation , Humans , Immunomodulation , Immunotherapy/trends , Infections/therapy , Inflammation , Mice , Neoplasms/therapy
14.
Mol Med ; 17(3-4): 281-92, 2011.
Article in English | MEDLINE | ID: mdl-21085745

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of immature myeloid cells whose numbers dramatically increase in chronic and acute inflammatory diseases, including cancer, autoimmune disease, trauma, burns and sepsis. Studied originally in cancer, these cells are potently immunosuppressive, particularly in their ability to suppress antigen-specific CD8(+) and CD4(+) T-cell activation through multiple mechanisms, including depletion of extracellular arginine, nitrosylation of regulatory proteins, and secretion of interleukin 10, prostaglandins and other immunosuppressive mediators. However, additional properties of these cells, including increased reactive oxygen species and inflammatory cytokine production, as well as their universal expansion in nearly all inflammatory conditions, suggest that MDSCs may be more of a normal component of the inflammatory response ("emergency myelopoiesis") than simply a pathological response to a growing tumor. Recent evocative data even suggest that the expansion of MDSCs in acute inflammatory processes, such as burns and sepsis, plays a beneficial role in the host by increasing immune surveillance and innate immune responses. Although clinical efforts are currently underway to suppress MDSC numbers and function in cancer to improve antineoplastic responses, such approaches may not be desirable or beneficial in other clinical conditions in which immune surveillance and antimicrobial activities are required.


Subject(s)
Lymphocyte Activation/immunology , Myeloid Cells/immunology , Sepsis/immunology , Wounds and Injuries/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Humans , Models, Immunological
15.
Arthritis Res Ther ; 12(1): R29, 2010.
Article in English | MEDLINE | ID: mdl-20167120

ABSTRACT

INTRODUCTION: The interaction between the immune and skeletal systems is evidenced by the bone loss observed in autoimmune diseases such as rheumatoid arthritis. In this paper we describe a new mechanism by which the immune cytokine IL-17A directly affects osteoclastogenesis. METHODS: Human CD14+ cells were isolated from healthy donors, cultured on dentine slices and coverslips and stimulated with IL-17A and/or receptor activator of NF-kappaB ligand (RANKL). Osteoclast differentiation was evaluated by gene expression, flow cytometry, tartrate-resistant acid phosphatase staining, fluorescence and electron microscopy. Physiologic bone remodelling was studied in wild-type (Wt) and IL-17A-/- mice using micro-computer tomography and serum RANKL/osteoprotegerin concentration. Functional osteoclastogenesis assays were performed using bone marrow macrophages isolated from IL-17A-/- and Wt mice. RESULTS: IL-17A upregulates the receptor activator for NF-kappaB receptor on human osteoclast precursors in vitro, leading to increased sensitivity to RANKL signalling, osteoclast differentiation and bone loss. IL-17A-/- mice have physiological bone homeostasis indistinguishable from Wt mice, and bone marrow macrophages isolated from these mice develop fully functional normal osteoclasts. CONCLUSIONS: Collectively our data demonstrate anti-IL-17A treatment as a selective therapeutic target for bone loss associated with autoimmune diseases.


Subject(s)
Bone Remodeling/genetics , Interleukin-17/metabolism , Osteoclasts/metabolism , RANK Ligand/biosynthesis , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Separation , Cells, Cultured , Flow Cytometry , Gene Expression , Gene Expression Regulation , Humans , Interleukin-17/genetics , Mice , Mice, Knockout , Osteoclasts/cytology , Tomography, X-Ray Computed , Up-Regulation
16.
Mol Ther ; 17(3): 508-15, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19127252

ABSTRACT

Intravascular delivery (1.5 x 10(9) particles and higher) of recombinant adenovirus (rAd) induces myeloid cell mediated, self-limiting hemodynamic responses in normal mice. However, we observed anaphylactoid-type reactions and exacerbated hemodynamic events following rAd injection in mice bearing malignant 4T1 mammary carcinoma. Because 4T1 tumors induce significant CD11b(+)Gr-1(+) myeloid cell expansion and activation, we set to determine whether this causes rAd-induced exaggerated responses. When treated with a single intravenous dose (1 x 10(10) particles) of rAd, mice implanted with 4T1 carcinoma succumbed due to the anaphylactoid-type reactions. In contrast, normal mice and mice implanted with a related mammary carcinoma (66cl4) that does not induce CD11b(+)Gr-1(+) cell expansion, showed minimal responses. Depletion of phagocytic CD11b(+)Gr-1(+) cells prior to rAd delivery protected 4T1 tumor-bearing animals, whereas passive transfer of CD11b(+)Gr-1(+) cells from 4T1 tumor-bearing animals was sufficient to convey susceptibility to anaphylactoid-type reactions in normal animals. We further show that there is upregulation of nitric oxide and leukotriene signaling pathways in the 4T1 tumor-induced CD11b(+)Gr-1(+) myeloid cells and that pretreating mice with inhibitors of nitric oxide synthetase and leukotrienes can attenuate the anaphylactoid-type reactions. These data show that malignant tumor growth can alter CD11b(+)Gr-1(+) myeloid cells, rendering hosts susceptible to anaphylactoid-type reactions upon intravascular treatment with rAd.


Subject(s)
Adenoviridae/immunology , Anaphylaxis/metabolism , CD11b Antigen/immunology , Cell Differentiation , Genetic Therapy/adverse effects , Neoplasms/pathology , Receptors, Chemokine/metabolism , Adenoviridae/genetics , Anaphylaxis/physiopathology , Animals , Cardiovascular System/metabolism , Cell Line, Tumor , Disease Susceptibility , Female , Heart Rate , Humans , Leukotriene Antagonists/pharmacology , Liposomes , Mice , Myeloid Cells/metabolism , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Phagocytes , Receptors, Chemokine/immunology , Receptors, Leukotriene/metabolism
17.
JPEN J Parenter Enteral Nutr ; 32(6): 651-5, 2008.
Article in English | MEDLINE | ID: mdl-18974247

ABSTRACT

Cachexia accompanies many chronic inflammatory diseases, including cancer. Lean tissue wasting is only one component of the cancer cachexia response, which also includes anemia, anorexia, a hepatic acute phase protein response, and increased susceptibility to secondary infections. The etiologies of cancer cachexia are multifactorial and include an overproduction of inflammatory mediators, including cytokines produced by inappropriate activation of innate immunity. However, anticytokine therapies have generally not been seriously considered for cancer cachexia, in large part because of the overlapping activities of several inflammatory cytokines and the inability to prospectively identify the contributions of individual mediators. In contrast, recent evidence has focused on an immature myeloid cell population that expands dramatically in the tumors and secondary lymphoid organs of animals with some actively growing tumors. These immature GR-1(+)CD11b(+) cells are metabolically active and secrete large quantities of inflammatory cytokines and chemokines with the potential to produce cachexia. Their expansion is temporally associated with the development of cachexia. Future studies are required to determine whether therapeutic efforts intended to block the expansion of these cells can prevent the lean tissue wasting that accompanies active tumor growth.


Subject(s)
Cachexia/etiology , Cytokines/physiology , Energy Metabolism/physiology , Inflammation/metabolism , Neoplasms/metabolism , Acute-Phase Proteins/metabolism , Acute-Phase Reaction/metabolism , Cachexia/metabolism , Humans , Inflammation/physiopathology , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Neoplasms/physiopathology
18.
J Exp Med ; 204(6): 1463-74, 2007 Jun 11.
Article in English | MEDLINE | ID: mdl-17548519

ABSTRACT

Polymicrobial sepsis alters the adaptive immune response and induces T cell suppression and Th2 immune polarization. We identify a GR-1(+)CD11b(+) population whose numbers dramatically increase and remain elevated in the spleen, lymph nodes, and bone marrow during polymicrobial sepsis. Phenotypically, these cells are heterogeneous, immature, predominantly myeloid progenitors that express interleukin 10 and several other cytokines and chemokines. Splenic GR-1(+) cells effectively suppress antigen-specific CD8(+) T cell interferon (IFN) gamma production but only modestly suppress antigen-specific and nonspecific CD4(+) T cell proliferation. GR-1(+) cell depletion in vivo prevents both the sepsis-induced augmentation of Th2 cell-dependent and depression of Th1 cell-dependent antibody production. Signaling through MyD88, but not Toll-like receptor 4, TIR domain-containing adaptor-inducing IFN-beta, or the IFN-alpha/beta receptor, is required for complete GR-1(+)CD11b(+) expansion. GR-1(+)CD11b(+) cells contribute to sepsis-induced T cell suppression and preferential Th2 polarization.


Subject(s)
CD11b Antigen/metabolism , Immune Tolerance/immunology , Myeloid Differentiation Factor 88/metabolism , Myeloid Progenitor Cells/immunology , Receptors, Chemokine/metabolism , Sepsis/immunology , Th2 Cells/immunology , Animals , Cell Proliferation , Colony-Forming Units Assay , Cytokines/metabolism , Flow Cytometry , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Signal Transduction/immunology
19.
Mol Ther ; 12(2): 254-63, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16043097

ABSTRACT

Intravascular administration of recombinant adenovirus (rAd) in cancer patients has been well tolerated. However, dose-limiting hemodynamic responses associated with suppression of cardiac output have been observed at doses of 7.5 x 10(13) particles. While analysis of hemodynamic responses induced by small-molecule pharmaceuticals is well established, little is known about the cardiovascular effects of rAd. Telemetric cardiovascular (CV) monitoring in mice was utilized to measure hemodynamic events following intravascular rAd administration. Electrocardiogram analysis revealed a block in the SA node 3-4 min postinfusion, resulting in secondary pacemaking initiated at the AV node. This was associated with acute bradycardia, reduced blood pressure, and hypothermia followed by gradual recovery. Adenovirus-primed murine sera with high neutralizing antibody (nAb) titers could inhibit CV responses, whereas human sera with equivalent nAb titers induced by natural infection were, surprisingly, not inhibitory. Interestingly, repeat dosing within 2-4 h of the primary injection resulted in desensitization, resembling tachyphylaxis, for subsequent CV responses. Last, depletion of Kupffer cells prior to rAd infusion precluded induction of CV responses. These inhibitory effects suggest that rAd interactions with certain cells of the reticular endothelial system are associated with induction of CV responses. Significantly, these studies may provide insight into management of acute adverse effects following rAd systemic delivery, enabling a broadening of therapeutic index.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Genetic Vectors/pharmacology , Heart Rate/drug effects , beta-Galactosidase/therapeutic use , Animals , Blood Pressure/drug effects , Carcinoma, Non-Small-Cell Lung , Cardiac Output/drug effects , Cardiovascular System/drug effects , Electrocardiography , Genetic Vectors/therapeutic use , Humans , Influenza A virus , Kupffer Cells/physiology , Mice , Mice, Inbred BALB C
20.
Shock ; 23(6): 507-15, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15897802

ABSTRACT

Control of dendritic cell (DC) function is critical for strategies to modulate innate and acquired immune responses. We examined whether transduction of murine DCs with adenoviral vectors (Adv) expressing interleukin (IL)-10 could alter their phenotype and T cell stimulatory function. Murine bone marrow-derived DCs were transduced with AdV encoding human IL-10 or green fluorescent protein (GFP). Whereas transduction of immature DCs with AdV/GFP resulted in dose-dependent maturation, DCs transduced with Adv/IL-10 maintained an immature state with low major histocompatibility complex (MHC) class II, CD86, and IL-12 expression. The Adv/IL-10 transduced DCs were phenotypically unique, characterized by suppression of IL-12 expression, failure to stimulate Th1 or Th2 cytokine responses, and retained capacity to endocytose antigen. Importantly, Adv/IL-10-transduced DCs were biologically active in vivo, in that administration of these DCs into mice before a generalized peritonitis significantly improved survival. We conclude that Adv/IL-10 transduction of DCs provides an efficient means to modulate DC function. The capacity to modify DCs by adenoviral expression of IL-10 may provide a novel ex vivo or in vivo approach to mitigate acute and chronic inflammatory diseases like sepsis.


Subject(s)
Adenoviridae/genetics , Dendritic Cells/cytology , Interleukin-10/genetics , Interleukin-10/therapeutic use , Sepsis/therapy , Adenoviridae/metabolism , Animals , Antigens, CD/biosynthesis , B7-2 Antigen , CD3 Complex/biosynthesis , CD40 Antigens/biosynthesis , Cell Survival , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Endocytosis , Female , Flow Cytometry , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Green Fluorescent Proteins/metabolism , Interleukin-12/metabolism , Lipopolysaccharides/metabolism , Lymph Nodes/pathology , Lymphocytes/cytology , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Phenotype , Sepsis/metabolism , T-Lymphocytes/cytology , Th1 Cells , Th2 Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...