Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 25(3): 496-511, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38356058

ABSTRACT

Visceral adipose tissue (VAT) is an energy store and endocrine organ critical for metabolic homeostasis. Regulatory T (Treg) cells restrain inflammation to preserve VAT homeostasis and glucose tolerance. Here, we show that the VAT harbors two distinct Treg cell populations: prototypical serum stimulation 2-positive (ST2+) Treg cells that are enriched in males and a previously uncharacterized population of C-X-C motif chemokine receptor 3-positive (CXCR3+) Treg cells that are enriched in females. We show that the transcription factors GATA-binding protein 3 and peroxisome proliferator-activated receptor-γ, together with the cytokine interleukin-33, promote the differentiation of ST2+ VAT Treg cells but repress CXCR3+ Treg cells. Conversely, the differentiation of CXCR3+ Treg cells is mediated by the cytokine interferon-γ and the transcription factor T-bet, which also antagonize ST2+ Treg cells. Finally, we demonstrate that ST2+ Treg cells preserve glucose homeostasis, whereas CXCR3+ Treg cells restrain inflammation in lean VAT and prevent glucose intolerance under high-fat diet conditions. Overall, this study defines two molecularly and developmentally distinct VAT Treg cell types with unique context- and sex-specific functions.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein , T-Lymphocytes, Regulatory , Female , Male , Humans , Intra-Abdominal Fat , Cytokines , Inflammation , Glucose
2.
Nat Immunol ; 17(10): 1187-96, 2016 10.
Article in English | MEDLINE | ID: mdl-27487330

ABSTRACT

During unresolved infections, some viruses escape immunological control and establish a persistant reservoir in certain cell types, such as human immunodeficiency virus (HIV), which persists in follicular helper T cells (TFH cells), and Epstein-Barr virus (EBV), which persists in B cells. Here we identified a specialized group of cytotoxic T cells (TC cells) that expressed the chemokine receptor CXCR5, selectively entered B cell follicles and eradicated infected TFH cells and B cells. The differentiation of these cells, which we have called 'follicular cytotoxic T cells' (TFC cells), required the transcription factors Bcl6, E2A and TCF-1 but was inhibited by the transcriptional regulators Blimp1, Id2 and Id3. Blimp1 and E2A directly regulated Cxcr5 expression and, together with Bcl6 and TCF-1, formed a transcriptional circuit that guided TFC cell development. The identification of TFC cells has far-reaching implications for the development of strategies to control infections that target B cells and TFH cells and to treat B cell-derived malignancies.


Subject(s)
Arenaviridae Infections/immunology , B-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , HIV/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Germinal Center/pathology , Germinal Center/virology , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Article in English | MEDLINE | ID: mdl-33875601

ABSTRACT

Leukocyte homing driven by the chemokine CCL21 is pivotal for adaptive immunity because it controls dendritic cell (DC) and T cell migration through CCR7. ACKR4 scavenges CCL21 and has been shown to play an essential role in DC trafficking at the steady state and during immune responses to tumors and cutaneous inflammation. However, the mechanism by which ACKR4 regulates peripheral DC migration is unknown, and the extent to which it regulates CCL21 in steady-state skin and lymph nodes (LNs) is contested. Specifically, our previous findings that CCL21 levels are increased in LNs of ACKR4-deficient mice [I. Comerford et al., Blood 116, 4130-4140 (2010)] were refuted [M. H. Ulvmar et al., Nat. Immunol. 15, 623-630 (2014)], and no differences in CCL21 levels in steady-state skin of ACKR4-deficient mice were reported despite compromised CCR7-dependent DC egress in these animals [S. A. Bryce et al., J. Immunol. 196, 3341-3353 (2016)]. Here, we resolve these issues and reveal that two forms of CCL21, full-length immobilized and cleaved soluble CCL21, exist in steady-state barrier tissues, and both are regulated by ACKR4. Without ACKR4, extracellular CCL21 gradients in barrier sites are saturated and nonfunctional, DCs cannot home directly to lymphatic vessels, and excess soluble CCL21 from peripheral tissues pollutes downstream LNs. The results identify the mechanism by which ACKR4 controls DC migration in barrier tissues and reveal a complex mode of CCL21 regulation in vivo, which enhances understanding of functional chemokine gradient formation.


Subject(s)
Cell Movement , Chemokine CCL21/metabolism , Dendritic Cells/physiology , Lymph Nodes/metabolism , Receptors, CCR/metabolism , Animals , Mice, Inbred C57BL
4.
PLoS Pathog ; 17(10): e1010004, 2021 10.
Article in English | MEDLINE | ID: mdl-34695149

ABSTRACT

While Salmonella enterica is seen as an archetypal facultative intracellular bacterial pathogen where protection is mediated by CD4+ T cells, identifying circulating protective cells has proved very difficult, inhibiting steps to identify key antigen specificities. Exploiting a mouse model of vaccination, we show that the spleens of C57BL/6 mice vaccinated with live-attenuated Salmonella serovar Typhimurium (S. Typhimurium) strains carried a pool of IFN-γ+ CD4+ T cells that could adoptively transfer protection, but only transiently. Circulating Salmonella-reactive CD4+ T cells expressed the liver-homing chemokine receptor CXCR6, accumulated over time in the liver and assumed phenotypic characteristics associated with tissue-associated T cells. Liver memory CD4+ T cells showed TCR selection bias and their accumulation in the liver could be inhibited by blocking CXCL16. These data showed that the circulation of CD4+ T cells mediating immunity to Salmonella is limited to a brief window after which Salmonella-specific CD4+ T cells migrate to peripheral tissues. Our observations highlight the importance of triggering tissue-specific immunity against systemic infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Liver/immunology , Salmonella Infections, Animal/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Salmonella typhimurium/immunology
5.
PLoS Pathog ; 15(8): e1007957, 2019 08.
Article in English | MEDLINE | ID: mdl-31437249

ABSTRACT

Human zinc deficiency increases susceptibility to bacterial infection. Although zinc supplementation therapies can reduce the impact of disease, the molecular basis for protection remains unclear. Streptococcus pneumoniae is a major cause of bacterial pneumonia, which is prevalent in regions of zinc deficiency. We report that dietary zinc levels dictate the outcome of S. pneumoniae infection in a murine model. Dietary zinc restriction impacts murine tissue zinc levels with distribution post-infection altered, and S. pneumoniae virulence and infection enhanced. Although the activation and infiltration of murine phagocytic cells was not affected by zinc restriction, their efficacy of bacterial control was compromised. S. pneumoniae was shown to be highly sensitive to zinc intoxication, with this process impaired in zinc restricted mice and isolated phagocytic cells. Collectively, these data show how dietary zinc deficiency increases sensitivity to S. pneumoniae infection while revealing a role for zinc as a component of host antimicrobial defences.


Subject(s)
Dietary Supplements , Disease Models, Animal , Lung Diseases/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Virulence/drug effects , Zinc/administration & dosage , Animals , Female , Lung Diseases/drug therapy , Lung Diseases/microbiology , Mice , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/growth & development
6.
Immunol Cell Biol ; 97(8): 726-739, 2019 09.
Article in English | MEDLINE | ID: mdl-31050022

ABSTRACT

Existing capsular polysaccharide-based vaccines against pneumococcal disease are highly effective against vaccine-included serotypes, but they are unable to combat serotype replacement. We have developed a novel pneumococcal vaccine that confers serotype-independent protection, and could therefore constitute a "universal" vaccine formulation. This preparation is comprised of whole un-encapsulated pneumococci inactivated with gamma irradiation (γ-PN), and we have previously reported induction of cross-reactive immunity after nonadjuvanted intranasal vaccination. To further enhance vaccine immunogenicity and safety, we modified the pneumococcal vaccine strain to induce a stressed state during growth. Specifically, the substrate binding component of the psaBCA operon for manganese import was mutated to create a pneumococcal surface antigen A (psaA) defective vaccine strain. psaA mutation severely attenuated the growth of the vaccine strain in vitro without negatively affecting pneumococcal morphology, thereby enhancing vaccine safety. In addition, antibodies raised against vaccine preparations based on the modified strain [γ-PN(ΔPsaA)] showed more diversified reactivity to wild-type pneumococcal challenge strains compared to those induced by the original formulation. The modified vaccine also induced comparable protective TH 17 responses in the lung, and conferred greater protection against lethal heterologous pneumococcal challenge. Overall, the current study demonstrates successful refinement of a serotype-independent pneumococcal vaccine candidate to enhance safety and immunogenicity.


Subject(s)
Adhesins, Bacterial/immunology , Lipoproteins/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/immunology , Adhesins, Bacterial/genetics , Administration, Intranasal , Animals , Antigens, Surface/genetics , Antigens, Surface/immunology , Disease Models, Animal , Female , HEK293 Cells , Humans , Immunogenicity, Vaccine , Lipoproteins/genetics , Lung/cytology , Lung/immunology , Mice , Mutation , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/administration & dosage , Pneumococcal Vaccines/adverse effects , Pneumococcal Vaccines/genetics , Streptococcus pneumoniae/genetics , Th17 Cells/immunology , Vaccination/methods , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology
8.
Immunol Cell Biol ; 96(4): 347-357, 2018 04.
Article in English | MEDLINE | ID: mdl-29377354

ABSTRACT

Mesenchymal stromal cells or stem cells (MSCs) have been shown to participate in tissue repair and are immunomodulatory in neuropathological settings. Given this, their potential use in developing a new generation of personalized therapies for autoimmune and inflammatory diseases of the central nervous system (CNS) will be explored. To effectively exert these effector functions, MSCs must first gain entry into damaged neural tissues, a process that has been demonstrated to be a limiting factor in their therapeutic efficacy. In this review, we discuss approaches to maximize the therapeutic efficacy of MSCs by altering their intrinsic trafficking programs to effectively enter neuropathological sites. To this end, we explore the significant role of chemokine receptors and adhesion molecules in directing cellular traffic to the inflamed CNS and the capacity of MSCs to adopt these molecular mechanisms to gain entry to this site. We postulate that understanding and exploiting these migratory mechanisms may be key to the development of cell-based therapies tailored to respond to the migratory cues unique to the nature and stage of progression of individual CNS disorders.


Subject(s)
Adult Stem Cells/transplantation , Autoimmunity , Brain/pathology , Inflammation/immunology , Inflammation/therapy , Mesenchymal Stem Cells/cytology , Humans , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy
9.
Mol Cancer ; 16(1): 19, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28137279

ABSTRACT

BACKGROUND: Breast cancer is the major cause of cancer-related mortality in women. It is thought that quiescent stem-like cells within solid tumors are responsible for cancer maintenance, progression and eventual metastasis. We recently reported that the chemokine receptor CCR7, a multi-functional regulator of breast cancer, maintains the stem-like cell population. METHODS: This study used a combination of molecular and cellular assays on primary mammary tumor cells from the MMTV-PyMT transgenic mouse with or without CCR7 to examine the signaling crosstalk between CCR7 and Notch pathways. RESULTS: We show for the first time that CCR7 functionally intersects with the Notch signaling pathway to regulate mammary cancer stem-like cells. In this cell subpopulation, CCR7 stimulation activated the Notch signaling pathway, and deletion of CCR7 significantly reduced the levels of activated cleaved Notch1. Moreover, blocking Notch activity prevented specific ligand-induced signaling of CCR7 and augmentation of mammary cancer stem-like cell function. CONCLUSION: Crosstalk between CCR7 and Notch1 promotes stemness in mammary cancer cells and may ultimately potentiate mammary tumor progression. Therefore, dual targeting of both the CCR7 receptor and Notch1 signaling axes may be a potential therapeutic avenue to specifically inhibit the functions of breast cancer stem cells.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/metabolism , Receptor, Notch1/metabolism , Receptors, CCR7/genetics , Animals , Female , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/virology , Mice , Mice, Transgenic , Receptor, Notch1/genetics , Receptors, CCR7/metabolism , Signal Transduction , Tumor Cells, Cultured
10.
Blood ; 126(13): 1609-20, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26206951

ABSTRACT

IL-17-producing cells are important mediators of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Here we demonstrate that a distinct CD8(+) Tc17 population develops rapidly after SCT but fails to maintain lineage fidelity such that they are unrecognizable in the absence of a fate reporter. Tc17 differentiation is dependent on alloantigen presentation by host dendritic cells (DCs) together with IL-6. Tc17 cells express high levels of multiple prototypic lineage-defining transcription factors (eg, RORγt, T-bet) and cytokines (eg, IL-17A, IL-22, interferon-γ, granulocyte macrophage colony-stimulating factor, IL-13). Targeted depletion of Tc17 early after transplant protects from lethal acute GVHD; however, Tc17 cells are noncytolytic and fail to mediate graft-versus-leukemia (GVL) effects. Thus, the Tc17 differentiation program during GVHD culminates in a highly plastic, hyperinflammatory, poorly cytolytic effector population, which we term "inflammatory iTc17" (iTc17). Because iTc17 cells mediate GVHD without contributing to GVL, therapeutic inhibition of iTc17 development in a clinical setting represents an attractive approach for separating GVHD and GVL.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Graft vs Host Disease/pathology , Graft vs Leukemia Effect , Interleukin-17/immunology , Stem Cell Transplantation/adverse effects , Th17 Cells/pathology , Animals , Bone Marrow Transplantation/adverse effects , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Graft vs Host Disease/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Th17 Cells/immunology
11.
Clin Sci (Lond) ; 131(2): 169-180, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27885052

ABSTRACT

Streptococcus pneumoniae and influenza are the world's foremost bacterial and viral respiratory pathogens. We have previously described a γ-irradiated influenza A virus (γ-FLU) vaccine that provides cross-protective immunity against heterosubtypic infections. More recently, we reported a novel non-adjuvanted γ-irradiated S pneumoniae (γ-PN) vaccine that elicits serotype-independent protection. Considering the clinical synergism of both pathogens, combination of a serotype-independent pneumococcal vaccine with a broad-spectrum influenza vaccine to protect against both infections would have a considerable clinical impact. In the present study, we co-immunized C57BL/6 mice intranasally (IN) with a mixture of γ-PN (whole inactivated cells) and γ-FLU (whole inactivated virions) and examined protective efficacy. Co-immunization enhanced γ-PN vaccine efficacy against virulent pneumococcal challenge, which was dependent on CD4+ T-cell responses. In contrast, vaccination with γ-PN alone, co-immunization enhanced pneumococcal-specific effector T-helper 17 cell (Th17) and Th1 memory cell, promoted development of CD4+ tissue-resident memory (TRM) cells and enhanced Pneumococcus-specific antibody responses. Furthermore, co-immunization elicited significant protection against lethal influenza challenge, as well as against co-infection with both influenza and S pneumoniae. This is the first report showing the synergistic effect of combining whole cell and whole virion vaccines to both S pneumoniae and influenza as a single vaccine to protect against individual and co-infection, without compromising pathogen-specific immunity.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/prevention & control , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Administration, Intranasal , Animals , Antibody Formation , Humans , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/administration & dosage , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/immunology , Vaccination
12.
J Immunol ; 195(12): 5707-17, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26538396

ABSTRACT

Intracellular infections, such as those caused by the protozoan parasite Leishmania donovani, a causative agent of visceral leishmaniasis (VL), require a potent host proinflammatory response for control. IL-17 has emerged as an important proinflammatory cytokine required for limiting growth of both extracellular and intracellular pathogens. However, there are conflicting reports on the exact roles for IL-17 during parasitic infections and limited knowledge about cellular sources and the immune pathways it modulates. We examined the role of IL-17 in an experimental model of VL caused by infection of C57BL/6 mice with L. donovani and identified an early suppressive role for IL-17 in the liver that limited control of parasite growth. IL-17-producing γδ T cells recruited to the liver in the first week of infection were the critical source of IL-17 in this model, and CCR2(+) inflammatory monocytes were an important target for the suppressive effects of IL-17. Improved parasite control was independent of NO generation, but associated with maintenance of superoxide dismutase mRNA expression in the absence of IL-17 in the liver. Thus, we have identified a novel inhibitory function for IL-17 in parasitic infection, and our results demonstrate important interactions among γδ T cells, monocytes, and infected macrophages in the liver that can determine the outcome of parasitic infection.


Subject(s)
Interleukin-17/metabolism , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Liver/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Humans , Immunosuppression Therapy , Leishmania donovani/growth & development , Liver/parasitology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/parasitology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR2/metabolism , Superoxide Dismutase/metabolism , T-Lymphocytes/parasitology
13.
J Biol Chem ; 290(37): 22385-97, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26216880

ABSTRACT

The chemokine CXCL12 and its G protein-coupled receptors CXCR4 and ACKR3 are implicated in cancer and inflammatory and autoimmune disorders and are targets of numerous antagonist discovery efforts. Here, we describe a series of novel, high affinity CXCL12-based modulators of CXCR4 and ACKR3 generated by selection of N-terminal CXCL12 phage libraries on live cells expressing the receptors. Twelve of 13 characterized CXCL12 variants are full CXCR4 antagonists, and four have Kd values <5 nm. The new variants also showed high affinity for ACKR3. The variant with the highest affinity for CXCR4, LGGG-CXCL12, showed efficacy in a murine model for multiple sclerosis, demonstrating translational potential. Molecular modeling was used to elucidate the structural basis of binding and antagonism of selected variants and to guide future designs. Together, this work represents an important step toward the development of therapeutics targeting CXCR4 and ACKR3.


Subject(s)
Chemokine CXCL12/chemistry , Models, Molecular , Peptide Library , Receptors, CXCR4/chemistry , Receptors, CXCR/chemistry , Animals , Chemokine CXCL12/genetics , Chemokine CXCL12/pharmacology , Disease Models, Animal , HEK293 Cells , Humans , Jurkat Cells , Mice , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Protein Engineering , Receptors, CXCR/genetics , Receptors, CXCR4/genetics
14.
PLoS Pathog ; 10(2): e1003905, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586147

ABSTRACT

Differentiation of naïve CD4⁺ cells into functionally distinct effector helper T cell subsets, characterised by distinct "cytokine signatures," is a cardinal strategy employed by the mammalian immune system to efficiently deal with the rapidly evolving array of pathogenic microorganisms encountered by the host. Since the T(H)1/T(H)2 paradigm was first described by Mosmann and Coffman, research in the field of helper T cell biology has grown exponentially with seven functionally unique subsets having now been described. In this review, recent insights into the molecular mechanisms that govern differentiation and function of effector helper T cell subsets will be discussed in the context of microbial infections, with a focus on how these different helper T cell subsets orchestrate immune responses tailored to combat the nature of the pathogenic threat encountered.


Subject(s)
Adaptive Immunity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation/immunology , Humans , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Helper-Inducer/cytology
15.
Clin Sci (Lond) ; 130(9): 697-710, 2016 May.
Article in English | MEDLINE | ID: mdl-26831937

ABSTRACT

Generating a pneumococcal vaccine that is serotype independent and cost effective remains a global challenge. γ-Irradiation has been used widely to sterilize biological products. It can also be utilized as an inactivation technique to generate whole-cell bacterial and viral vaccines with minimal impact on pathogen structure and antigenic determinants. In the present study, we utilized γ-irradiation to inactivate an un-encapsulated Streptococcus pneumoniae strain Rx1 with an unmarked deletion of the autolysin gene lytA and with the pneumolysin gene ply replaced with an allele encoding a non-toxic pneumolysoid (PdT) (designated γ-PN vaccine). Intranasal vaccination of C57BL/6 mice with γ-PN was shown to elicit serotype-independent protection in lethal challenge models of pneumococcal pneumonia and sepsis. Vaccine efficacy was shown to be reliant on B-cells and interleukin (IL)-17A responses. Interestingly, immunization promoted IL-17 production by innate cells not T helper 17 (Th17) cells. These data are the first to report the development of a non-adjuvanted intranasal γ-irradiated pneumococcal vaccine that generates effective serotype-independent protection, which is mediated by both humoral and innate IL-17 responses.


Subject(s)
B-Lymphocytes/immunology , Gamma Rays , Immunity, Innate , Interleukin-17/metabolism , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/radiation effects , Vaccination , Administration, Intranasal , Animals , CD4-Positive T-Lymphocytes/immunology , Immunity, Innate/immunology , Immunologic Memory , Interferon-gamma/metabolism , Mice, Inbred C57BL , Pneumococcal Infections/complications , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Sepsis/complications , Sepsis/immunology , Sepsis/microbiology , Sepsis/prevention & control , Serotyping , Streptococcus pneumoniae/classification , T-Lymphocytes/immunology , Treatment Outcome
16.
Breast Cancer Res ; 17: 46, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25887862

ABSTRACT

INTRODUCTION: Podocalyxin (gene name PODXL) is a CD34-related sialomucin implicated in the regulation of cell adhesion, migration and polarity. Upregulated expression of podocalyxin is linked to poor patient survival in epithelial cancers. However, it is not known if podocalyxin has a functional role in tumor progression. METHODS: We silenced podocalyxin expression in the aggressive basal-like human (MDA-MB-231) and mouse (4T1) breast cancer cell lines and also overexpressed podocalyxin in the more benign human breast cancer cell line, MCF7. We evaluated how podocalyxin affects tumorsphere formation in vitro and compared the ability of podocalyxin-deficient and podocalyxin-replete cell lines to form tumors and metastasize using xenogenic or syngeneic transplant models in mice. Finally, in an effort to develop therapeutic treatments for systemic cancers, we generated a series of antihuman podocalyxin antibodies and screened these for their ability to inhibit tumor progression in xenografted mice. RESULTS: Although deletion of podocalyxin does not alter gross cell morphology and growth under standard (adherent) culture conditions, expression of PODXL is required for efficient formation of tumorspheres in vitro. Correspondingly, silencing podocalyxin resulted in attenuated primary tumor growth and invasiveness in mice and severely impaired the formation of distant metastases. Likewise, in competitive tumor engraftment assays where we injected a 50:50 mixture of control and shPODXL (short-hairpin RNA targeting PODXL)-expressing cells, we found that podocalyxin-deficient cells exhibited a striking decrease in the ability to form clonal tumors in the lung, liver and bone marrow. Finally, to validate podocalyxin as a viable target for immunotherapy, we screened a series of novel antihuman podocalyxin antibodies for their ability to inhibit tumor progression in vivo. One of these antibodies, PODOC1, potently blocked tumor growth and metastasis. CONCLUSIONS: We show that podocalyxin plays a key role in the formation of primary tumors and distant tumor metastasis. In addition, we validate podocalyxin as potential target for monoclonal antibody therapy to inhibit primary tumor growth and systemic dissemination.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Sialoglycoproteins/antagonists & inhibitors , Sialoglycoproteins/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Humans , Mammary Neoplasms, Animal , Mice , Neoplasm Metastasis , RNA Interference , RNA, Small Interfering/genetics , Sialoglycoproteins/genetics , Spheroids, Cellular , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Mol Cancer ; 14: 115, 2015 Jun 06.
Article in English | MEDLINE | ID: mdl-26047945

ABSTRACT

BACKGROUND: The expression of the chemokine receptor CCR6 has been previously correlated with higher grades and stages of breast cancer and decreased relapse-free survival. Also, its cognate chemokine ligand CCL20 has been reported to induce proliferation of cultured human breast epithelial cells. METHODS: To establish if CCR6 plays a functional role in mammary tumorigenesis, a bigenic MMTV-PyMT CCR6-null mouse was generated and mammary tumor development was assessed. Levels of tumor-infiltrating immune cells within tumor-bearing mammary glands from MMTV-PyMT Ccr6 (WT) and Ccr6 (-/-) mice were also analyzed. RESULTS: Deletion of CCR6 delayed tumor onset, significantly reduced the extent of initial hyperplastic outgrowth, and decreased tumor incidence in PyMT transgenic mice. CCR6 was then shown to promote the recruitment of pro-tumorigenic macrophages to the tumor site, facilitating the onset of neoplasia. CONCLUSIONS: This study delineated for the first time a role for CCR6 in the development of breast cancer, and demonstrated a critical function for this receptor in maintaining the pro-tumorigenic cancer microenvironment.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Macrophages/metabolism , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse/metabolism , Receptors, CCR6/metabolism , Animals , Carcinogenesis/pathology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Deletion , Macrophages/pathology , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Mice, Inbred C57BL , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment
18.
Immunol Cell Biol ; 93(8): 735-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25753393

ABSTRACT

Phosphoinositide 3-kinase γ (PI3Kγ) consists of the catalytic subunit p110γ that forms a mutually exclusive heterodimer with one of the two adaptor subunits, p101 or p84. Although activation of PI3Kγ is necessary for cell migration downstream of G-protein-coupled receptor engagement, particularly within the immune system, aberrant PI3Kγ signalling has been associated with transformation, increased migration and the progression of multiple cancer types. Regulation of PI3Kγ signal activation and duration is critical to controlling and maintaining coordinated cellular migration; however, the mechanistic basis for this is not well understood. We have recently demonstrated that, in contrast to the tumour-promoting potential of p110γ and p101, p84 possesses tumour-suppressor activity, suggesting a negative regulatory role within PI3Kγ signalling. The present study investigated the role of p84 phosphorylation in the context of PI3Kγ signalling, cell migration and p84-mediated tumour suppression. Two putative phosphorylation sites were characterised within p84, Ser358 and Thr607. Expression of wild-type p84 reduced the oncogenic potential of MDA.MB.231 cells and inhibited metastatic lung colonisation in vivo, effects that were dependent on Thr607. Furthermore, loss of Thr607 enhanced migration of MDA.MB.231 cells in vitro and prevented the induction of p84/p110γ dimers. The dimerisation of wild-type p84 with p110γ was not detected at the plasma membrane, indicating an inhibitory interaction preventing PI3Kγ lipid-kinase activity. In contrast, Ser358 phosphorylation was not determined to be critical for p84 activity in the context of migration. Our findings suggest that p84 binding to p110γ may represent a novel negative feedback signal that terminates PI3Kγ activity.


Subject(s)
Cell Movement , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Multiprotein Complexes/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Cell Line , Chemokine CXCL12/pharmacology , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/genetics , Female , Gene Expression , Humans , Immunoprecipitation , Mice , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , Protein Multimerization , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled , Recombinant Fusion Proteins , Signal Transduction/drug effects
19.
Blood ; 121(1): 118-28, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23152546

ABSTRACT

The atypical chemokine receptor CCX-CKR regulates bioavailability of CCL19, CCL21, and CCL25, homeostatic chemokines that play crucial roles in thymic lymphopoiesis. Deletion of CCX-CKR results in accelerated experimental autoimmunity induced by immunization. Here we show that CCX-CKR deletion also increases incidence of a spontaneous Sjögren's syndrome-like pathology, characterized by lymphocytic infiltrates in salivary glands and liver of CCX-CKR(-/-) mice, suggestive of a defect in self-tolerance when CCX-CKR is deleted. This prompted detailed examination of the thymus in CCX-CKR(-/-) mice. Negatively selected mature SP cells were less abundant in CCX-CKR(-/-) thymi, yet expansion of both DP and immature SP cells was apparent. Deletion of CCX-CKR also profoundly reduced proportions of DN3 thymocyte precursors and caused DN2 cells to accumulate within the medulla. These effects are likely driven by alterations in thymic stroma as CCX-CKR(-/-) mice have fewer cTECs per thymocyte, and cTECs express the highest level of CCX-CKR in the thymus. A profound decrease in CCL25 within the thymic cortex was observed in CCX-CKR(-/-) thymi, likely accounting for their defects in thymocyte distribution and frequency. These findings identify a novel role for CCX-CKR in regulating cTEC biology, which promotes optimal thymocyte development and selection important for self-tolerant adaptive immunity.


Subject(s)
Autoimmunity , Lymphopoiesis , Receptors, Chemokine/deficiency , Thymocytes/pathology , Thymus Gland/pathology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Chemokines/metabolism , Chemokines, CC/biosynthesis , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Immunophenotyping , Kidney/pathology , Liver/pathology , Lymphopoiesis/genetics , Male , Mice , Mice, Knockout , Receptors, CCR7/deficiency , Receptors, CCR7/genetics , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Receptors, Chemokine/physiology , Self Tolerance/genetics , Self Tolerance/immunology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/pathology , Stem Cells/classification , Stem Cells/metabolism , Stem Cells/pathology , Submandibular Gland/pathology
20.
Blood ; 121(22): 4555-66, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23550035

ABSTRACT

Thrombosis promotes leukocyte infiltration into inflamed tissues, leading to organ injury in a broad range of diseases; however, the mechanisms by which thrombi guide leukocytes to sites of vascular injury remain ill-defined. Using mouse models of endothelial injury (traumatic or ischemia reperfusion), we demonstrate a distinct process of leukocyte recruitment, termed "directed intravascular migration," specifically mediated by platelet thrombi. Single adherent platelets and platelet aggregates stimulated leukocyte shape change at sites of endothelial injury; however, only thrombi were capable of inducing directed intravascular leukocyte migration. Leukocyte recruitment and migration induced by platelet thrombi occurred most prominently in veins but could also occur in arteries following ischemia-reperfusion injury. In vitro studies demonstrated a major role for platelet-derived NAP-2 (CXCL-7) and its CXCR1/2 receptor in regulating leukocyte polarization and motility. In vivo studies demonstrated the presence of an NAP-2 chemotactic gradient within the thrombus body. Pharmacologic blockade of CXCR1/2 as well as genetic deletion of NAP-2 markedly reduced leukocyte shape change and intrathrombus migration. These studies define a distinct process of leukocyte migration that is initiated by homotypic adhesive interactions between platelets, leading to the development of an NAP-2 chemotactic gradient within the thrombus body that guides leukocytes to sites of vascular injury.


Subject(s)
Blood Platelets/cytology , Chemokines, CXC/metabolism , Leukocytes/cytology , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Thrombosis/immunology , Animals , Blood Platelets/immunology , Blood Platelets/metabolism , Cell Adhesion/immunology , Cell Movement/immunology , Cell Polarity/immunology , Green Fluorescent Proteins/genetics , Leukocytes/immunology , Mesenteric Arteries/immunology , Mesenteric Arteries/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Needlestick Injuries/immunology , Needlestick Injuries/pathology , Neutrophils/cytology , Neutrophils/immunology , Reperfusion Injury/immunology , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL