Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Breast Cancer Res ; 21(1): 103, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31488209

ABSTRACT

BACKGROUND: Solid tumors produce proteins that can induce the accumulation of bone marrow-derived cells in various tissues, and these cells can enhance metastatic tumor growth by several mechanisms. 4T1 murine mammary tumors are known to produce granulocyte colony-stimulating factor (G-CSF) and increase the numbers of immunosuppressive CD11b+Gr1+ myeloid-derived suppressor cells (MDSCs) in tissues such as the spleen and lungs of tumor-bearing mice. While surgical resection of primary tumors decreases MDSC levels in the spleen, the longevity and impact of MDSCs and other immune cells in the lungs after tumor resection have been less studied. METHODS: We used mass cytometry time of flight (CyTOF) and flow cytometry to quantify MDSCs in the spleen, peripheral blood, and lungs of mice bearing orthotopic murine mammary tumors. We also tested the effect of primary tumor resection and/or gemcitabine treatment on the levels of MDSCs, other immune suppressor and effector cells, and metastatic tumor cells in the lungs. RESULTS: We have found that, similar to mice with 4T1 tumors, mice bearing metastatic 4T07 tumors also exhibit accumulation of CD11b+Gr1+ MDSCs in the spleen and lungs, while tissues of mice with non-metastatic 67NR tumors do not contain MDSCs. Mice with orthotopically implanted 4T1 tumors have increased granulocytic (G-) MDSCs, monocytic (M-) MDSCs, macrophages, eosinophils, and NK cells in the lungs. Resection of primary 4T1 tumors decreases G-MDSCs, M-MDSCs, and macrophages in the lungs within 48 h, but significant numbers of functional immunosuppressive G-MDSCs persist in the lungs for 2 weeks after tumor resection, indicative of an environment that can promote metastatic tumor growth. The chemotherapeutic agent gemcitabine depletes G-MDSCs, M-MDSCs, macrophages, and eosinophils in the lungs of 4T1 tumor-bearing mice, and we found that treating mice with gemcitabine after primary tumor resection decreases residual G-MDSCs in the lungs and decreases subsequent metastatic growth. CONCLUSIONS: Our data support the development of therapeutic strategies to target MDSCs and to monitor MDSC levels before and after primary tumor resection to enhance the effectiveness of immune-based therapies and improve the treatment of metastatic breast cancer in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mammary Neoplasms, Experimental/pathology , Mastectomy , Myeloid-Derived Suppressor Cells/drug effects , Animals , Antigens, Ly/metabolism , CD11b Antigen/metabolism , Cell Line, Tumor , Combined Modality Therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Eosinophils/pathology , Female , Killer Cells, Natural/pathology , Lung Neoplasms/immunology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/immunology , Gemcitabine
3.
Am J Respir Crit Care Med ; 193(2): 116-30, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26583808

ABSTRACT

Lung cancer is the leading cause of cancer mortality worldwide, and at only 18%, it has one of the lowest 5-year survival rates of all malignancies. With its highly complex mutational landscape, treatment strategies against lung cancer have proved largely ineffective. However with the recent success of immunotherapy trials in lung cancer, there is renewed enthusiasm in targeting the immune component of tumors. Macrophages make up the majority of the immune infiltrate in tumors and are a key cell type linking inflammation and cancer. Although the mechanisms through which inflammation promotes cancer are not fully understood, two connected hypotheses have emerged: an intrinsic pathway, driven by genetic alterations that lead to neoplasia and inflammation, and an extrinsic pathway, driven by inflammatory conditions that increase cancer risk. Here, we discuss the contribution of macrophages to these pathways and subsequently their roles in established tumors. We highlight studies investigating the association of macrophages with lung cancer prognosis and discuss emerging therapeutic strategies for targeting macrophages in the tumor microenvironment.


Subject(s)
Immunotherapy/methods , Inflammation/etiology , Lung Neoplasms/therapy , Macrophages/immunology , Biomarkers, Tumor/analysis , Clinical Trials as Topic , Disease Progression , Humans , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Phenotype , Prognosis , Survival Rate
4.
Oncotarget ; 7(4): 3677-91, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26683227

ABSTRACT

SH2-containing-inositol-5'-phosphatase (SHIP) is a negative regulator of the phosphatidylinositol-3-kinase pathway in hematopoietic cells and limits the development of leukemias and lymphomas. The potential role of SHIP in solid tumor development and metastasis remains unknown. While SHIP restricts the aberrant development of myeloid cells in C57BL/6 mice, there are conflicting reports regarding the effect of SHIP deletion in BALB/c mice with important consequences for determining the influence of SHIP in different model tumor systems. We generated SHIP-/- BALB/c mice and challenged them with syngeneic non-metastatic 67NR or metastatic 4T1 mammary tumors. We demonstrate that SHIP restricts the development, alternative-activation, and immunosuppressive function of myeloid cells in tumor-free and tumor-bearing BALB/c mice. Tumor-free SHIP-/- BALB/c mice exhibited pulmonary inflammation, myeloid hyperplasia, and M2-polarized macrophages and this phenotype was greatly exacerbated by 4T1, but not 67NR, tumors. 4T1-bearing SHIP-/- mice rapidly lost weight and died from necrohemorrhagic inflammatory pulmonary disease, characterized by massive infiltration of pulmonary macrophages and myeloid-derived suppressor cells that were more M2-polarized and immunosuppressive than wild-type cells. Importantly, while SHIP loss did not affect primary tumor growth, 4T1-bearing SHIP-/- mice had 7.5-fold more metastatic tumor cells in their lungs than wild-type mice, consistent with the influence of immunosuppressive myeloid cells on metastatic growth. Our findings identify the hematopoietic cell-restricted protein SHIP as an intriguing target to influence the development of solid tumor metastases, and support development of SHIP agonists to prevent the accumulation of immunosuppressive myeloid cells and tumor metastases in the lungs to improve treatment of metastatic breast cancer.


Subject(s)
Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Phosphoric Monoester Hydrolases/physiology , Pneumonia/prevention & control , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Female , Humans , Immunoenzyme Techniques , Inositol Polyphosphate 5-Phosphatases , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Macrophages/metabolism , Macrophages/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/pathology , Pneumonia/genetics , Pneumonia/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Cancer Res ; 75(6): 996-1008, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25623234

ABSTRACT

The mobilization of bone marrow-derived cells (BMDC) to distant tissues before the arrival of disseminated tumor cells has been shown preclinically to facilitate metastasis through the establishment of metastatic niches. Primary tumor hypoxia has been demonstrated to play a pivotal role in the production of chemokines and cytokines responsible for the mobilization of these BMDCs, especially in breast cancer. Carbonic anhydrase IX (CAIX, CA9) expression is highly upregulated in hypoxic breast cancer cells through the action of hypoxia-inducible factor-1 (HIF1). Preclinical evidence has demonstrated that CAIX is required for breast tumor growth and metastasis; however, the mechanism by which CAIX exerts its prometastatic function is not well understood. Here, we show that CAIX is indispensable for the production of granulocyte colony-stimulating factor (G-CSF) by hypoxic breast cancer cells and tumors in an orthotopic model. Furthermore, we demonstrate that tumor-expressed CAIX is required for the G-CSF-driven mobilization of granulocytic myeloid-derived suppressor cells (MDSC) to the breast cancer lung metastatic niche. We also determined that CAIX expression is required for the activation of NF-κB in hypoxic breast cancer cells and constitutive activation of the NF-κB pathway in CAIX-depleted cells restored G-CSF secretion. Together, these findings identify a novel hypoxia-induced CAIX-NF-κB-G-CSF cellular signaling axis culminating in the mobilization of granulocytic MDSCs to the breast cancer lung metastatic niche.


Subject(s)
Carbonic Anhydrases/physiology , Cell Movement , Granulocyte Colony-Stimulating Factor/biosynthesis , Myeloid Cells/physiology , Neoplasms, Experimental/pathology , Animals , CD11b Antigen/analysis , Carbonic Anhydrase IX , Cell Hypoxia , Chemokine CXCL10/physiology , Female , Mice , Mice, Inbred BALB C , NF-kappa B/physiology , Neoplasm Metastasis
6.
Carcinogenesis ; 35(10): 2291-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25023988

ABSTRACT

We recently demonstrated that both murine and human carcinomas grow significantly slower in mice on low carbohydrate (CHO), high protein diets than on isocaloric Western diets and that a further reduction in tumor growth rates occur when the low CHO diets are combined with the cyclooxygenase-2 inhibitor, celecoxib. Following upon these studies, we asked herein what effect low CHO, high protein diets, with or without celecoxib, might have on tumor metastasis. In the highly metastatic 4T1 mouse mammary tumor model, a 15% CHO, high protein diet supplemented with celecoxib (1 g/kg chow) markedly reduced lung metastases. Moreover, in longer-term studies using male Transgenic Adenocarcinoma of the Mouse Prostate mice, which are predisposed to metastatic prostate cancer, the 15% CHO diet, with and without celecoxib (0.3 g/kg chow), gave the lowest incidence of metastases, but a more moderate 25% CHO diet containing celecoxib led to the best survival. Metabolic studies with 4T1 tumors suggested that the low CHO, high protein diets may be forcing tumors to become dependent on amino acid catabolism for survival/growth. Taken together, our results suggest that a combination of a low CHO, high protein diet with celecoxib substantially reduces metastasis.


Subject(s)
Diet, Carbohydrate-Restricted , Dietary Proteins/pharmacology , Neoplasm Metastasis/drug therapy , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Celecoxib , Diet Therapy/methods , Disease Models, Animal , Lung Neoplasms/diet therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis/therapy , Prostatic Neoplasms/diet therapy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
7.
J Immunol ; 192(1): 512-22, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24285836

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are emerging as potential promoters of metastatic tumor growth, and there is interest in targeting immature MDSCs by inducing their differentiation into more mature myeloid cells. We used all-trans retinoic acid (ATRA) to differentiate MDSCs in mice bearing metastatic 4T1 or 4TO7 murine mammary tumors, and assessed the immune-suppressive mechanisms and potencies of different myeloid cell subpopulations. Metastatic mammary tumors induced the accumulation of distinct populations of immature CD11b(+)Gr1(+)F4/80(-)Ly6C(mid)Ly6G(+) MDSCs ("Gr1(+) cells") and mature CD11b(+)Gr1(-)F4/80(+) cells ("F4/80(+) cells") in metastatic target organs. ATRA triggered the differentiation of Gr1(+) cells into F4/80(+) cells in the lungs and, unexpectedly, enhanced pulmonary metastatic tumor growth. We found that F4/80(+)Ly6C(-)Ly6G(-) mature macrophages (Ms) were up to 30-fold more potent immune suppressors than Gr1(+) cells on a per-cell basis, which we postulate may contribute to the increased metastatic growth observed with ATRA treatment. F4/80(+) cells and Gr1(+) cells used different reactive oxygen species (ROS)-mediated mechanisms of immunosuppression ex vivo, with F4/80(+) cells producing higher levels of ROS, which is consistent with their superior immunosuppressive abilities. These data highlight the potent immunosuppressive functions of Ms, reveal that Ms can suppress T cell responses via ROS production, and suggest that ROS inhibitors may be useful in promoting antitumor immune responses. Our findings also caution against using ATRA to modulate myeloid cell differentiation and function to treat breast cancer metastases in the lung, and support the development of therapeutic strategies to enhance antitumor immunity by targeting myeloid cells as a collective group.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/pathology , Macrophages/immunology , Myeloid Cells/immunology , Animals , Cell Differentiation/drug effects , Disease Models, Animal , Female , Immunophenotyping , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Transgenic , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Metastasis , Phenotype , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tretinoin/pharmacology
8.
Cancer Immunol Immunother ; 61(5): 643-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22021068

ABSTRACT

As more groups investigate the role of myeloid-derived suppressor cells (MDSCs) in promoting the growth of primary tumors and distant tumor metastases, it is imperative to ensure the accurate detection and quantification of MDSC immunosuppression ex vivo. MDSCs are defined by their ability to suppress immune responses. Although different in vitro culture conditions have been used to study MDSCs, the effect of different culture conditions on MDSC immunosuppression is unknown. We therefore isolated MDSCs from the lungs and spleens of 4T1 murine mammary tumor-bearing mice and assayed MDSC-mediated suppression of T cell responses under different culture conditions. We found that 4T1-induced MDSCs effectively suppressed T cell proliferation under serum-free conditions, but not when fetal calf serum (FCS) was present. FCS neither altered the immunosuppressive activities of other myeloid cell types (i.e., peritoneal or tumor-associated macrophages) nor modified the susceptibility of T cells to myeloid cell-mediated suppression, but instead acted directly on 4T1-induced MDSCs to significantly reduce their immunosuppressive function. Importantly, we found that bovine serum albumin was a major contributor to the antagonistic effects of FCS on 4T1-induced MDSC immunosuppression by inhibiting reactive oxygen species production from MDSCs. This work reveals that in vitro culture conditions influence the immunosuppressive properties of MDSCs and highlights the importance of testing different culture conditions on MDSC phenotype to ensure that MDSC immunosuppression is not being masked. These data have important implications for the accurate detection and identification of MDSCs, as well as for determining the influence of MDSC-mediated immunosuppression on primary and metastatic tumor growth.


Subject(s)
Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/immunology , Myeloid Cells/immunology , Animals , Cattle , Cell Culture Techniques , Cell Growth Processes/immunology , Female , Immunosuppression Therapy , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Myeloid Cells/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Serum Albumin, Bovine/immunology , Serum Albumin, Bovine/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
Cancer Res ; 71(14): 5050-1; author reply 5052-3, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21750177

ABSTRACT

4T1 murine mammary carcinoma cells implanted in syngeneic Balb/c mice are increasingly being used in metastasis research, with some groups using this model to study tumor-induced accumulation of bone marrow-derived cells in metastatic target organs. Bone marrow-derived cells (including CD11b(+)Gr-1(+) myelomonocytic cells) are thought to modify the local lung microenvironment to facilitate subsequent colonization by metastatic tumor cells. While quantification of metastatic 4T1 tumor cells in various tissues can be done using ex vivo colony-forming assays, detection of metastatic 4T1 cells is often facilitated by expressing fluorescent proteins in the tumor cells prior to implantation. We found that Balb/c mice mount a potent immune response against 4T1 cells expressing green fluorescent protein (GFP) that includes the generation of anti-GFP antibodies in the circulation. Importantly, the number of bone marrow-derived CD11b(+)Gr-1(+) cells and metastatic tumor cells that accumulate in the lungs is significantly decreased in mice implanted with 4T1 cells expressing GFP compared with mice bearing wild-type 4T1 tumors. Taken together, our data caution against the use of GFP-expressing tumor cells in the Balb/c mouse strain, particularly for studying the influence of immunomodulatory cells on tumor cell metastasis.


Subject(s)
Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung/immunology , Mammary Neoplasms, Experimental/immunology , Myeloid Cells/immunology , Animals , CD11b Antigen , Female , Green Fluorescent Proteins/immunology , Mice , Mice, Inbred BALB C , Tumor Microenvironment/immunology
10.
Exp Hematol ; 39(1): 2-13, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21056081

ABSTRACT

The SH2-containing inositol-5'-phosphatase, SHIP (or SHIP1), is a hematopoietic-restricted phosphatidylinositide phosphatase that translocates to the plasma membrane after extracellular stimulation and hydrolyzes the phosphatidylinositol-3-kinase-generated second messenger PI-3,4,5-P(3) to PI-3,4-P(2). As a result, SHIP dampens down PI-3,4,5-P(3)-mediated signaling and represses the proliferation, differentiation, survival, activation, and migration of hematopoietic cells. There are multiple lines of evidence suggesting that SHIP may act as a tumor suppressor during leukemogenesis and lymphomagenesis. Because of its ability to skew macrophage progenitors toward M1 macrophages and naïve T cells toward T helper 1 and T helper 17 cells, SHIP may play a critical role in activating the immune system to eradicate solid tumors. In this review, we will discuss the role of SHIP in hematopoietic cells and its therapeutic potential in terms of suppressing leukemias and lymphomas and manipulating the immune system to combat cancer.


Subject(s)
Neoplasms/physiopathology , Phosphoric Monoester Hydrolases/physiology , Animals , Humans , Inositol Polyphosphate 5-Phosphatases , Mice , Mice, Knockout , Neoplasms/enzymology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/chemistry , Protein Conformation
11.
J Immunol ; 185(8): 4545-53, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20844190

ABSTRACT

Resident tissue macrophages (Mφs) continually survey the microenvironment, ingesting Ags and presenting them on their surface for recognition by T cells. Because these Ags can be either host cell- or pathogen-derived, Mφs must be able to distinguish whether a particular Ag should provoke an immune response or be tolerated. However, the mechanisms that determine whether Mφs promote or inhibit T cell activation are not well understood. To investigate this, we first determined the mechanism by which murine resident peritoneal Mφs suppress in vitro T cell proliferation in the absence of pathogens and then explored the effects of different pathogen-derived molecules on Mφ immunosuppression. Our results suggest that, in response to IFN-γ, which is secreted by TCR-activated T cells, resident peritoneal Mφs acquire immunosuppressive properties that are mediated by NO. However, pretreatment of Mφs with LPS or dsRNA, but not CpG or peptidoglycan, eliminates their suppressive properties, in part via the induction of autocrine-acting IFN-ß. These results suggest TLR agonists that activate TRIF, and consequently induce IFN-ß, but not those that exclusively signal through MyD88, abrogate the immunosuppressive properties of Mφs, and thus promote T cell expansion and elimination of invading microorganisms.


Subject(s)
Immune Tolerance/immunology , Interferon-beta/biosynthesis , Lymphocyte Activation/immunology , Macrophages/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Blotting, Western , Cell Proliferation , Cell Separation , Flow Cytometry , Interferon-beta/immunology , Lipopolysaccharides/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Double-Stranded/immunology , T-Lymphocytes/metabolism , Toll-Like Receptors/agonists
12.
J Immunol ; 183(2): 975-83, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19542365

ABSTRACT

Maintaining an appropriate balance between subsets of CD4(+) Th and T regulatory cells (Tregs) is critical to maintain immune homeostasis and prevent autoimmunity. Through a common requirement for TGF-beta, the development of peripherally induced Tregs is intimately linked to that of Th17 cells, with the resulting lineages depending on the presence of proinflammatory cytokines such as IL-6. Currently very little is known about the molecular signaling pathways that control the development of Tregs vs Th17 cells. Reduced activity of the PI3K pathway is required for TGF-beta-mediated induction of Foxp3 expression and the suppressive activity of Tregs. To investigate how negative regulators of the PI3K pathway impact Treg development, we investigated whether SHIP, a lipid phosphatase that regulates PI3K activity, also plays a role in the development and function of Tregs. SHIP-deficient Tregs maintained suppressive capacity in vitro and in a T cell transfer model of colitis. Surprisingly, SHIP-deficient Th cells were significantly less able to cause colitis than were wild-type Th cells due to a profound deficiency in Th17 cell differentiation, both in vitro and in vivo. The inability of SHIP-deficient T cells to develop into Th17 cells was accompanied by decreased IL-6-stimulated phosphorylation of STAT3 and an increased capacity to differentiate into Treg cells under the influence of TGF-beta and retinoic acid. These data indicate that SHIP is essential for normal Th17 cell development and that this lipid phosphatase plays a key role in the reciprocal regulation of Tregs and Th17 cells.


Subject(s)
Homeostasis/immunology , Interleukin-17 , Phosphoric Monoester Hydrolases/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Regulatory/cytology , Animals , Cell Differentiation , Colitis/etiology , Inositol Polyphosphate 5-Phosphatases , Interleukin-6/pharmacology , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/deficiency , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/transplantation
13.
Blood ; 113(13): 2945-54, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19139077

ABSTRACT

Gram-negative bacterial infections, unlike viral infections, do not typically protect against subsequent viral infections. This is puzzling given that lipopolysaccharide (LPS) and double-stranded (ds) RNA both activate the TIR domain-containing adaptor-inducing interferon beta (TRIF) pathway and, thus, are both capable of eliciting an antiviral response by stimulating type I interferon (IFN) production. We demonstrate herein that SH2-containing inositol-5'-phosphatase (SHIP) protein levels are dramatically increased in murine macrophages via the MyD88-dependent pathway, by up-regulating autocrine-acting transforming growth factor-beta (TGFbeta). The increased SHIP then mediates, via inhibition of the phosphatidylinositol-3-kinase (PI3K) pathway, cytosine-phosphate-guanosine (CPG)- and LPS-induced tolerance and cross-tolerance and restrains IFN-beta production induced by a subsequent exposure to LPS or dsRNA. Intriguingly, we found, using isoform-specific PI3K inhibitors, that LPS- or cytosine-phosphate-guanosine-induced interleukin-6 (IL-6) is positively regulated by p110alpha, -gamma, and -delta but negatively regulated by p110beta. This may explain some of the controversy concerning the role of PI3K in Toll-like receptor-induced cytokine production. Consistent with our in vitro findings, SHIP(-/-) mice overproduce IFN-beta in response to LPS, and this leads to antiviral hypothermia. Thus, up-regulation of SHIP in response to Gram-negative bacterial infections probably explains the inability of such infections to protect against subsequent viral infections.


Subject(s)
Immunity, Innate/drug effects , Immunity, Innate/genetics , Lipopolysaccharides/pharmacology , Phosphoric Monoester Hydrolases/genetics , Viruses/immunology , Animals , Cells, Cultured , CpG Islands/immunology , CpG Islands/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Hypothermia/genetics , Hypothermia/immunology , Immune Tolerance/drug effects , Immune Tolerance/genetics , Inositol Polyphosphate 5-Phosphatases , Interferon-beta/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/physiology , Phosphoric Monoester Hydrolases/metabolism , RNA, Double-Stranded/immunology , RNA, Double-Stranded/pharmacology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL