Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Front Oncol ; 11: 662723, 2021.
Article in English | MEDLINE | ID: mdl-35223446

ABSTRACT

Myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) contribute to cancer-related inflammation and tumor progression. While several myeloid molecules have been ascribed a regulatory function in these processes, the triggering receptors expressed on myeloid cells (TREMs) have emerged as potent modulators of the innate immune response. While various TREMs amplify inflammation, others dampen it and are emerging as important players in modulating tumor progression-for instance, soluble TREM-1 (sTREM-1), which is detected during inflammation, associates with disease progression, while TREM-2 expression is associated with tumor-promoting macrophages. We hypothesized that TREM-1 and TREM-2 might be co-expressed on tumor-infiltrating myeloid cells and that elevated sTREM-1 associates with disease outcomes, thus representing a possibility for mutual modulation in cancer. Using the 4T1 breast cancer model, we found TREM-1 and TREM-2 expression on MDSC and TAM and that sTREM-1 was elevated in tumor-bearing mice in multiple models and correlated with tumor volume. While TREM-1 engagement enhanced TNF, a TREM-2 ligand was detected on MDSC and TAM, suggesting that both TREM could be functional in the tumor setting. Similarly, we detected TREM-1 and Trem2 expression in myeloid cells in the RENCA model of renal cell carcinoma (RCC). We confirmed these findings in human disease by demonstrating the expression of TREM-1 on tumor-infiltrating myeloid cells from patients with RCC and finding that sTREM-1 was increased in patients with RCC. Finally, The Cancer Genome Atlas analysis shows that TREM1 expression in tumors correlates with poor outcomes in RCC. Taken together, our data suggest that manipulation of the TREM-1/TREM-2 balance in tumors may be a novel means to modulate tumor-infiltrating myeloid cell phenotype and function.

2.
J Natl Cancer Inst Monogr ; 2017(52)2017 11 01.
Article in English | MEDLINE | ID: mdl-29140488

ABSTRACT

Since 2007, the US National Cancer Institute (NCI) Office of Cancer Complementary and Alternative Medicine (OCCAM), together with the Cancer Institute of the China Academy of Chinese Medical Sciences (CICACMS), institutes at China Academy of Sciences and Chinese Academy of Medical Sciences, have engaged in collaborations on Chinese medicine (CM) and cancer research. Through these collaborations, CM drugs and compounds have been studied at NCI labs. This paper summarizes the discoveries and progress on these research projects, exploring the aspects of cancer prevention, botanical drug mechanisms of action and component analysis/quality control (QC), and anticancer activity screening. These and other related projects have been presented in various jointly convened workshops and have provided the backdrop for establishing a new organization, the International Consortium for CM and Cancer, to promote international collaborations in this field.


Subject(s)
Medicine, Chinese Traditional , Neoplasms/therapy , China , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Humans , Medicine, Chinese Traditional/methods , National Cancer Institute (U.S.) , Neoplasms/diagnosis , Neoplasms/prevention & control , Research , United States
3.
Biomaterials ; 53: 545-53, 2015.
Article in English | MEDLINE | ID: mdl-25890750

ABSTRACT

DNA that encodes tumor-specific antigens represents potential immunostimulatory agents. However, rapid enzymatic degradation and fragmentation of DNA during administration can result in limited vector expression and, consequently, poor efficacy. These challenges have necessitated the use of novel strategies for DNA delivery. Herein, we study the ability of cationic self-assembling peptide hydrogels to encapsulate plasmid DNA, and enhance its immunostimulatory potential in vivo. The effect of network charge on the gel's ability to retain the DNA was assessed employing three gel-forming peptides that vary systematically in formal charge. The peptide HLT2, having a formal charge of +5 at neutral pH, was optimal in encapsulating microgram quantities of DNA with little effect on its rheological properties, allowing its effective syringe delivery in vivo. The plasmid, DNA(TA), encapsulated within these gels encodes for a melanoma-specific gp100 antigen fused to the alarmin protein adjuvant HMGN1. Implantation of DNA(TA)-loaded HLT2 gels into mice resulted in an acute inflammatory response with the presence of polymorphonuclear cells, which was followed by infiltrating macrophages. These cellular infiltrates aid in the processing of encapsulated DNA, promoting increased lymphoproliferation and producing an enhanced immune response mediated by CD4+/IFNγ+ expressing Th1 cells, and complemented by the formation of gp100-specific antibodies.


Subject(s)
Adjuvants, Immunologic/pharmacology , DNA/chemistry , Hydrogels , Peptides/pharmacology , Adjuvants, Immunologic/chemistry , Amino Acid Sequence , Animals , Female , Inflammation/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptides/chemistry
4.
FASEB J ; 29(2): 443-54, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25376833

ABSTRACT

It was reported that TNF receptor type II signaling, which has the capacity to stimulate CD4+ forkhead box P3+ (Foxp3+) regulatory T cells (Tregs), activated the noncanonical NF-κB pathway in an IKKα-dependent manner. Therefore, we studied the role of IKKα in the homeostasis of Treg population. To this end, we generated a mouse strain with conditional knockout of IKKα in CD4 cells (Ikkα(f/f):CD4.Cre) that showed a >60% reduction in the number of Tregs in the thymus and peripheral lymphoid tissues, whereas the number of Foxp3- effector T cells (Teffs) remained at a normal level. The function of Tregs deficient in IKKα was examined using Rag1(-/-) mice cotransferred with naive CD4 cells (nCD4s). Although wild-type (WT) Tregs inhibited colitis induced by transfer of WT nCD4s, IKKα-deficient Tregs failed to do so, which was associated with their inability to reconstitute Rag1(-/-) mice. Furthermore, nCD4s deficient in IKKα also failed to reconstitute Rag1(-/-) mice and were defective in proliferative responses in vitro and in vivo. Thus, our study reveals a novel role of IKKα in the maintenance of a normal Treg population and in the control of expansion of CD4 T cells. These properties of IKKα may be exploited as therapeutic strategies in the treatment of major human diseases.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , I-kappa B Kinase/physiology , T-Lymphocytes, Regulatory/cytology , Animals , Cell Proliferation , Colitis/metabolism , Flow Cytometry , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeostasis , I-kappa B Kinase/metabolism , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Thymus Gland/metabolism
5.
Cancer Lett ; 355(2): 232-41, 2014 Dec 28.
Article in English | MEDLINE | ID: mdl-25242356

ABSTRACT

Cancer pain is a deleterious consequence of tumor growth and related inflammation. Opioids and anti-inflammatory drugs provide first line treatment for cancer pain, but both are limited by side effects. Fufang Kushen injection (FKI) is GMP produced, traditional Chinese medicine used alone or with chemotherapy to reduce cancer-associated pain. FKI limited mouse sarcoma growth both in vivo and in vitro, in part, by reducing the phosphorylation of ERK and AKT kinases and BAD. FKI inhibited TRPV1 mediated capsaicin-induced ERK phosphorylation and reduced tumor-induced proinflammatory cytokine production. Thus, FKI limited cancer pain both directly by blocking TRPV1 signaling and indirectly by reducing tumor growth.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hyperalgesia/drug therapy , MAP Kinase Signaling System/drug effects , Sarcoma/complications , Sarcoma/drug therapy , TRPV Cation Channels/metabolism , Animals , Capsaicin/metabolism , Cell Line, Tumor , Cytokines/metabolism , Female , Hyperalgesia/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Sarcoma/enzymology , Sarcoma/metabolism
6.
Cancer Res ; 74(21): 5989-98, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205103

ABSTRACT

Alarmins are endogenous mediators that are elicited rapidly in response to danger signals, enhancing innate and adaptive immune responses by promoting the recruitment and maturation of antigen-presenting cells (APC). The nucleosome-binding protein HMGN1 is a potent alarmin that binds TLR4 and induces antigen-specific Th1 immune responses, but its contributions to antitumor immunity have not been explored. We found that ovalbumin (OVA)-expressing EG7 mouse thymoma cells grew much faster in Hmgn1-deficient mice than littermate-matched controls. Tumor-bearing Hmgn1(-/-) mice generated fewer OVA-specific CD8 cells in the spleen than EG7-bearing Hmgn1(+/+) mice, suggesting that HMGN1 supported T cell-mediated antitumor immunity. In addition, EG7 tumors expressing HMGN1 grew more slowly than control EG7 tumors, suggesting greater resistance to HMGN1-expressing tumors. This resistance relied on T cell-mediated immunity because it was abolished by in vivo depletion of CD4(+) and CD8(+) T cells. Moreover, mice vaccinated with a DNA vector expressing an HMGN1-gp100 fusion protein manifested gp100-specific, Th1-polarized immune responses, acquiring resistance to challenge with mouse B16F1 melanoma. Overall, our findings show that HMGN1 contributes to antitumor immunity and it may offer an effective adjuvant to heighten responses to cancer vaccines.


Subject(s)
Antigen-Presenting Cells/immunology , HMGN1 Protein/genetics , Immunity, Cellular/genetics , Melanoma, Experimental/immunology , Adjuvants, Immunologic/genetics , Animals , Antigen-Presenting Cells/metabolism , CD8-Positive T-Lymphocytes/immunology , HMGN1 Protein/immunology , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Recombinant Fusion Proteins/therapeutic use , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Vaccination , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/immunology
7.
Cytokine ; 65(1): 56-64, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24080164

ABSTRACT

CD4(+) T cells stimulate immune responses through distinct patterns of cytokine produced by Th1, Th2 or Th17 cells, or inhibit immune responses through Foxp3-expressing regulatory T cells (Tregs). Paradoxically, effector T cells were recently shown to activate Tregs, however, it remains unclear which Th subset is responsible for this effect. In this study, we found that Th17 cells expressed the highest levels of TNF among in vitro generated Th subsets, and most potently promoted expansion and stabilized Foxp3 expression by Tregs when co-transferred into Rag1(-/-) mice. Both TNF and IL-2 produced by Th17 cells contributed to this effect. The stimulatory effect of Th17 cells on Tregs was largely abolished when co-transferred with TNFR2-deficient Tregs. Furthermore, Tregs deficient in TNFR2 also supported a much lower production of IL-17A and TNF expression by co-transferred Th17 cells. Thus, our data indicate that the TNF-TNFR2 pathway plays a crucial role in the reciprocal stimulatory effect of Th17 cells and Tregs. This bidirectional interaction should be taken into account when designing therapy targeting Th17 cells, Tregs, TNF and TNFR2.


Subject(s)
Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Adoptive Transfer , Animals , CD4 Antigens/immunology , Cytokines/immunology , Forkhead Transcription Factors/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-2/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Th1 Cells/immunology , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/biosynthesis
8.
J Immunol ; 191(2): 865-74, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23776172

ABSTRACT

Alarmins are a group of structurally diverse host defense antimicrobial peptides that are important immune activators. In this article, we present a novel role for two potent alarmins, human ß-defensin 2 and 3 (HBD2 and 3), in promoting IFN-α production by human plasmacytoid dendritic cells. We demonstrate that HBD2 and 3 activate pDCs by enhancing the intracellular uptake of CpG and self DNA and promote DNA-induced IFN-α production in a TLR9-dependent manner. Both CpG and host DNA form aggregates that resemble DNA nets when combined with HBD2 and 3. Isothermal titration calorimetry studies to elucidate the nature of HBD3/CpG complexes demonstrate involvement of enthalpy-driven interactions, in addition to hydrophobic interactions, with the formation of complexes at a molar ratio of 2:1 defensin/CpG. The i.v. administration of HBD3/CpG complexes induced proinflammatory cytokines like IL-12, IFN-γ, IL-6, IFN-α, and IL-10 in serum, associated with an increased recruitment of APCs in the spleen. Subcutaneous injections of these complexes showed enhanced infiltration of inflammatory cells at the injection site, indicating a potential pathophysiological role for alarmin/DNA complexes in contributing to inflammation. Intraperitoneal immunization of HBD3/CpG complexes with OVA enhanced both cellular and humoral responses to OVA, compared with OVA/HBD3 or OVA/CPG alone, indicative of a much more potent adjuvant effect of the HBD3/CpG complexes. Thus, the ability of defensins to enhance cellular uptake of nucleic acids can lead to improved vaccine formulations by promoting their uptake by various cells, resulting in an enhanced immune response.


Subject(s)
Dendritic Cells/metabolism , Inflammation/immunology , Interferon-alpha/biosynthesis , beta-Defensins/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/immunology , Biological Transport , Cells, Cultured , CpG Islands , DNA/metabolism , Dendritic Cells/immunology , Female , Humans , Hydrophobic and Hydrophilic Interactions , Interferon-alpha/blood , Interferon-gamma/blood , Interleukin-12/blood , Interleukin-6/blood , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Toll-Like Receptor 9/metabolism
9.
Eur J Immunol ; 43(6): 1412-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23619968

ABSTRACT

Alarmins are endogenous, constitutively available, damage-associated molecular patterns that upon release can mobilize and activate various leukocytes for the induction of innate and adaptive immune responses. For our immune system to function appropriately, it relies on navigating various leukocytes to distinct places at the right time. The direction of cell migration is determined by chemotactic factors that include classical chemoattractants, chemokines, certain growth factors, and alarmins. This viewpoint provides an overview of alarmin-induced cell migration. Alarmins are capable of inducing the migration of diverse types of leukocytes and nonleukocytes either directly by triggering specific receptors or indirectly by inducing production of chemokines through the activation of various leukocytes via pattern recognition receptors. The receptors used by alarmins to directly induce cell migration can either be Gαi protein-coupled receptors or receptors such as the receptor for advanced glycation end products; however, the intracellular signaling events responsible for the direct chemotactic activities of alarmins are, to date, only partially elucidated. Given that alarmins act in concert with chemokines to regulate the recruitment and trafficking of leukocytes, these damage-associated molecular patterns are potentially involved in diverse biological processes as discussed in this viewpoint.


Subject(s)
Chemotactic Factors/immunology , Leukocytes/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Pattern Recognition/immunology , Adaptive Immunity , Animals , Cell Communication/immunology , Cell Movement/immunology , Humans , Immunity, Innate , Immunomodulation , Signal Transduction/immunology
10.
PLoS One ; 8(3): e58791, 2013.
Article in English | MEDLINE | ID: mdl-23527025

ABSTRACT

MCP-1/CCL2 plays an important role in the initiation and progression of cancer. Since tumor cells produce MCP-1, they are considered to be the main source of this chemokine. Here, we examined whether MCP-1 produced by non-tumor cells affects the growth and lung metastasis of 4T1 breast cancer cells by transplanting them into the mammary pad of WT or MCP-1(-/-) mice. Primary tumors at the injected site grew similarly in both mice; however, lung metastases were markedly reduced in MCP-1(-/-) mice, with significantly longer mouse survival. High levels of MCP-1 mRNA were detected in tumors growing in WT, but not MCP-1(-/-) mice. Serum MCP-1 levels were increased in tumor-bearing WT, but not MCP-1(-/-) mice. Transplantation of MCP-1(-/-) bone marrow cells into WT mice did not alter the incidence of lung metastasis, whereas transplantation of WT bone marrow cells into MCP-1(-/-) mice increased lung metastasis. The primary tumors of MCP-1(-/-) mice consistently developed necrosis earlier than those of WT mice and showed decreased infiltration by macrophages and reduced angiogenesis. Interestingly, 4T1 cells that metastasized to the lung constitutively expressed elevated levels of MCP-1, and intravenous injection of 4T1 cells producing a high level of MCP-1 resulted in increased tumor foci in the lung of WT and MCP-1(-/-) mice. Thus, stromal cell-derived MCP-1 in the primary tumors promotes lung metastasis of 4T1 cells, but tumor cell-derived MCP-1 can also contribute once tumor cells enter the circulation. A greater understanding of the source and role of this chemokine may lead to novel strategies for cancer treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chemokine CCL2/genetics , Lung Neoplasms/secondary , Stromal Cells/metabolism , Animals , Blood Cells/metabolism , Bone Marrow Cells/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Chemokine CCL2/metabolism , Disease Models, Animal , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Leukocytes/immunology , Leukocytes/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Necrosis/genetics , Neovascularization, Pathologic/genetics
11.
J Immunol ; 190(3): 1076-84, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23277487

ABSTRACT

Several lines of evidence indicate the instability of CD4(+)Foxp3(+) regulatory T cells (Tregs). We have therefore investigated means of promoting the stability of Tregs. In this study, we found that the proportion of Tregs in mouse strains deficient in TNFR2 or its ligands was reduced in the thymus and peripheral lymphoid tissues, suggesting a potential role of TNFR2 in promoting the sustained expression of Foxp3. We observed that upon in vitro activation with plate-bound anti-CD3 Ab and soluble anti-CD28 Ab, Foxp3 expression by highly purified mouse Tregs was markedly downregulated. Importantly, TNF partially abrogated this effect of TCR stimulation and stabilized Foxp3 expression. This effect of TNF was blocked by anti-TNFR2 Ab, but not by anti-TNFR1 Ab. Furthermore, TNF was not able to maintain Foxp3 expression by TNFR2-deficient Tregs. In a mouse colitis model induced by transfer of naive CD4 cells into Rag1(-/-) mice, the disease could be inhibited by cotransfer of wild-type Tregs, but not by cotransfer of TNFR2-deficient Tregs. Furthermore, in the lamina propria of the colitis model, most wild-type Tregs maintained Foxp3 expression. In contrast, an increased number of TNFR2-deficient Tregs lost Foxp3 expression. Thus, our data clearly show that TNFR2 is critical for the phenotypic and functional stability of Tregs in the inflammatory environment. This effect of TNF should be taken into account when designing future therapy of autoimmunity and graft-versus-host disease by using TNF inhibitors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Animals, Congenic , CD4-Positive T-Lymphocytes/transplantation , Colitis/microbiology , Colitis/pathology , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation/immunology , Homeodomain Proteins/genetics , Intestinal Mucosa/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphopenia/etiology , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/transplantation , Thymus Gland/immunology , Thymus Gland/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology
12.
Immunol Lett ; 148(1): 34-8, 2012.
Article in English | MEDLINE | ID: mdl-22898052

ABSTRACT

To clarify controversies in the literature of the field, we have purified and characterized B16F1 melanoma cell derived exosomes (mcd-exosomes) then we attempted to dissect their immunological activities. We tested how mcd-exosomes influence CD4+ T cell proliferation induced by bone marrow derived dendritic cells; we quantified NF-κB activation in mature macrophages stimulated with mcd-exosomes, and we compared the cytokine profile of LPS-stimulated, IL-4 induced, and mcd-exosome treated macrophages. We observed that mcd-exosomes helped the maturation of dendritic cells, enhancing T cell proliferation induced by the treated dendritic cells. The exosomes also activated macrophages, as measured by NF-κB activation. The cytokine and chemokine profile of macrophages treated with tumor cell derived exosomes showed marked differences from those induced by either LPS or IL-4, and it suggested that exosomes may play a role in the tumor progression and metastasis formation through supporting tumor immune escape mechanisms.


Subject(s)
Dendritic Cells/immunology , Exosomes/immunology , Macrophages/immunology , Melanoma/immunology , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Chemokines/immunology , Chemokines/metabolism , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Exosomes/metabolism , Exosomes/ultrastructure , Female , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , NF-kappa B/immunology , NF-kappa B/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Mol Biochem Parasitol ; 185(1): 66-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22710390

ABSTRACT

A wide range of secondary biological functions have been documented for eukaryotic aminoacyl-tRNA synthetases including roles in transcriptional regulation, mitochondrial RNA splicing, cell growth, and chemokine-like activities. The asparaginyl-tRNA synthetase (AsnRS) of the filarial nematode, Brugia malayi, is a highly expressed excretory-secretory molecule which activates interleukin 8 (IL-8) receptors via extracellular domains that are different from those used by IL-8. Recent success in determining the complete atomic structure of the B. malayi AsnRS provided the opportunity to map its chemokine-like activity. Chemotaxis assays demonstrated that IL-8-like activity is localized in a novel 80 amino acid amino terminal substructure. Structural homology searches revealed similarities between that domain in B. malayi AsnRS and substructures involved in receptor binding by human IL-8. These observations provide important new insights into how parasite-derived molecules may play a role in the modulation of immune cell function.


Subject(s)
Aspartate-tRNA Ligase/immunology , Brugia malayi/enzymology , Helminth Proteins/immunology , Interleukin-8/immunology , RNA, Transfer, Amino Acyl/immunology , Amino Acid Sequence , Animals , Brugia malayi/genetics , Brugia malayi/immunology , Chemotaxis , Computational Biology/methods , Enzyme Activation , Humans , Immunologic Factors/immunology , Molecular Sequence Data , Neutrophils/immunology , Protein Structure, Tertiary , Receptors, Interleukin-8/immunology , Sequence Homology, Amino Acid
14.
Proc Natl Acad Sci U S A ; 109(16): 6130-5, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22474389

ABSTRACT

Nuclear translocation of cytosolic CLIC4 is an essential feature of its proapoptotic and prodifferentiation functions. Here we demonstrate that CLIC4 is induced concurrently with inducible nitric oxide synthase (iNOS) and S-nitrosylated in proinflammatory peritoneal macrophages. Chemical inhibition or genetic ablation of iNOS inhibits S-nitrosylation and nuclear translocation of CLIC4. In macrophages, iNOS-induced nuclear CLIC4 coincides with the pro- to anti-inflammatory transition of the cells because IL-1ß and CXCL1 mRNA remain elevated in CLIC4 and iNOS knockout macrophages at late time points, whereas TNFα mRNA is elevated only in the iNOS knockout macrophages. Active IL-1ß remains elevated in CLIC4 knockout macrophages and in macrophages in which CLIC4 nuclear translocation is prevented by the NOS inhibitor l-NAME. Moreover, overexpression of nuclear-targeted CLIC4 down-regulates IL-1ß in stimulated macrophages. In mice, genetically null for CLIC4, the number of phagocytosing macrophages stimulated by LPS is reduced. Thus, iNOS-induced nuclear CLIC4 is an essential part of the macrophage deactivation program.


Subject(s)
Cell Nucleus/metabolism , Chloride Channels/metabolism , Macrophages/metabolism , Mitochondrial Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Cells, Cultured , Chloride Channels/genetics , Gene Expression/drug effects , Immunoblotting , Interferon-gamma/pharmacology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Reverse Transcriptase Polymerase Chain Reaction
15.
Proc Natl Acad Sci U S A ; 108(24): 9798-803, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21628584

ABSTRACT

Significant efforts have been devoted to the development of nanoparticular delivering systems targeting tumors. However, clinical application of nanoparticles is hampered by insufficient size homogeneity, difficulties in reproducible synthesis and manufacturing, frequent high uptake in the liver, systemic toxicity of the carriers (particularly for inorganic nanoparticles), and insufficient selectivity for tumor cells. We have found that properly modified synthetic analogs of transmembrane domains of membrane proteins can self-assemble into remarkably uniform spherical nanoparticles with innate biological activity. Self-assembly is driven by a structural transition of the peptide that adopts predominantly a beta-hairpin conformation in aqueous solutions, but folds into an alpha-helix upon spontaneous fusion of the nanoparticles with cell membrane. A 24-amino acid peptide corresponding to the second transmembrane helix of the CXCR4 forms self-assembled particles that inhibit CXCR4 function in vitro and hamper CXCR4-dependent tumor metastasis in vivo. Furthermore, such nanoparticles can encapsulate hydrophobic drugs, thus providing a delivery system with the potential for dual biological activity.


Subject(s)
Cell Membrane/chemistry , Membrane Proteins/chemistry , Nanoparticles/chemistry , Peptides/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival/drug effects , Circular Dichroism , Dose-Response Relationship, Drug , Female , Humans , Magnetic Resonance Spectroscopy , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Membrane Proteins/metabolism , Mice , Mice, Nude , Microscopy, Confocal , Microscopy, Electron, Transmission , Molecular Sequence Data , Nanoparticles/ultrastructure , Particle Size , Peptides/metabolism , Peptides/pharmacology , Protein Binding , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/chemistry , Receptors, CXCR4/metabolism
16.
J Immunol ; 186(11): 6417-26, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21515789

ABSTRACT

We hypothesize that innate immune signals from infectious organisms and/or injured tissues may activate peripheral neuronal pain signals. In this study, we demonstrated that TLRs 3, 7, and 9 are expressed by human dorsal root ganglion neurons (DRGNs) and in cultures of primary mouse DRGNs. Stimulation of murine DRGNs with TLR ligands induced expression and production of proinflammatory chemokines and cytokines CCL5 (RANTES), CXCL10 (IP-10), IL-1α, IL-1ß, and PGE(2), which have previously been shown to augment pain. Further, TLR ligands upregulated the expression of a nociceptive receptor, transient receptor potential vanilloid type 1 (TRPV1), and enhanced calcium flux by TRPV1-expressing DRGNs. Using a tumor-induced temperature sensitivity model, we showed that in vivo administration of a TLR9 antagonist, known as a suppressive oligodeoxynucleotide, blocked tumor-induced temperature sensitivity. Taken together, these data indicate that stimulation of peripheral neurons by TLR ligands can induce nerve pain.


Subject(s)
Neurons/drug effects , Pain/physiopathology , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Aminoquinolines/pharmacology , Anilides/pharmacology , Animals , Arachidonic Acids/pharmacology , Blotting, Western , Calcium/metabolism , Capsaicin/pharmacology , Cells, Cultured , Cinnamates/pharmacology , Cytokines/genetics , Cytokines/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Endocannabinoids , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Imidazoles/pharmacology , Mice , Microscopy, Confocal , Neurons/metabolism , Poly I-C/pharmacology , Polyunsaturated Alkamides/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Toll-Like Receptors/agonists
17.
Sci Transl Med ; 2(61): 61ra90, 2010 Dec 08.
Article in English | MEDLINE | ID: mdl-21148126

ABSTRACT

Psoriasis is a common complex genetic disease characterized by hyperplasia and inflammation in the skin; however, the relative contributions of epidermal cells and the immune system to disease pathogenesis remain unclear. Linkage studies have defined a psoriasis susceptibility locus (PSORS4) on 1q21, the epidermal differentiation complex, which includes genes for small S100 calcium-binding proteins. These proteins are involved in extracellular and intracellular signaling during epithelial host defense, linking innate and adaptive immunity. Inflammation-prone psoriatic skin constitutively expresses elevated concentrations of S100A7 (psoriasin) and S100A15 (koebnerisin) in the epidermis. Here, we report that genetically modified mice expressing elevated amounts of doxycycline-regulated mS100a7a15 in skin keratinocytes demonstrated an exaggerated inflammatory response when challenged by exogenous stimuli such as abrasion (Koebner phenomenon). This immune response was characterized by immune cell infiltration and elevated concentrations of T helper 1 (T(H)1) and T(H)17 proinflammatory cytokines, which have been linked to the pathogenesis of psoriasis and were further amplified upon challenge. Both inflammation priming and amplification required mS100a7a15 binding to the receptor of advanced glycation end products (RAGE). mS100a7a15 potentiated inflammation by acting directly as a chemoattractant for leukocytes, further increasing the number of inflammatory cells infiltrating the skin. This study provides a pathogenetic psoriasis model using a psoriasis candidate gene to link the epidermis and innate immune system in inflammation priming, highlighting the S100A7A15-RAGE axis as a potential therapeutic target.


Subject(s)
Inflammation/immunology , Inflammation/pathology , Psoriasis/genetics , Psoriasis/pathology , S100 Proteins/genetics , Skin/pathology , Animals , Cells, Cultured , Cytokines/immunology , Genetic Predisposition to Disease , Humans , Keratinocytes/cytology , Keratinocytes/immunology , Mice , Mice, Transgenic , Psoriasis/immunology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , S100 Calcium Binding Protein A7 , S100 Proteins/immunology , Skin/immunology , Tumor Necrosis Factor-alpha/immunology
18.
J Immunol ; 185(1): 174-82, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20525892

ABSTRACT

Our previous study showed that TNFR2 is preferentially expressed by CD4(+)FoxP3(+) regulatory T cells (Tregs), and expression of this receptor identified maximally suppressive Tregs. TNFR2 is also expressed by a small fraction of CD4(+)FoxP3(-) conventional T cells (Tconvs) in normal mice, and its expression is upregulated by T cell activation. This raises questions about the role of TNFR2 signaling in the function of Tconv cells. In this study, by using FoxP3/gfp knock-in mice, we showed that TNFR2 signaling did not induce FoxP3(-) CD4 cells to become suppressive. Ki-67, a marker of proliferation, was concomitantly expressed with TNFR2 by CD4 cells, independent of forkhead box P3 expression, in normal mice and Lewis lung carcinoma-bearing mice. TNFR2 is associated with greater suppressive functions when expressed by Tregs and is associated with greater resistance to suppression when expressed by Tconv cells. In mice bearing 4T1 breast tumor or Lewis lung carcinoma, intratumoral Tconv cells expressing elevated levels of TNFR2 acquired the capacity to resist suppression by lymph node-derived Tregs. However, they remained susceptible to inhibition by more suppressive tumor-infiltrating Tregs, which expressed higher levels of TNFR2. Our data indicate that TNFR2 also costimulates Tconv cells. However, intratumoral Tregs expressing more TNFR2 are able to overcome the greater resistance to suppression of intratumoral Tconv cells, resulting in a dominant immunosuppressive tumor environment.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Forkhead Transcription Factors , Immunity, Innate , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/physiology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/prevention & control , Cell Line, Tumor , Cells, Cultured , Clonal Anergy/genetics , Clonal Anergy/immunology , Coculture Techniques , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/deficiency , Gene Knock-In Techniques , Immunity, Innate/genetics , Lymphocyte Activation/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor, Type II/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
19.
Eur J Immunol ; 40(4): 1099-106, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20127680

ABSTRACT

Previously, we found that co-expression of CD25 and TNFR2 identified the most suppressive subset of mouse Treg. In this study, we report that human peripheral blood (PB) FOXP3(+) cells present in CD25(high), CD25(low) and even CD25(-) subsets of CD4(+) cells expressed high levels of TNFR2. Consequently, TNFR2-expressing CD4(+)CD25(+) Treg included all of the FOXP3(+) cells present in the CD4(+)CD25(high) subset as well as a substantial proportion of the FOXP3(+) cells present in the CD4(+)CD25(low) subset. Flow cytometric analysis of PB identified five-fold more Treg, determined by FOXP3 expression, in the CD4(+)CD25(+)TNFR2(+) subset than in the CD4(+)CD25(high) subset. In addition, similar levels of FOXP3(+) cells were identified in both the CD4(+)CD25(+)TNFR2(+) and CD4(+)CD25(+)CD127(low/-) subsets. Furthermore, the CD4(+)CD25(+)TNFR2(+) subset expressed high levels of CTLA-4, CD45RO, CCR4 and low levels of CD45RA and CD127, a phenotype characteristic of Treg. Upon TCR stimulation, human PB CD4(+)CD25(+)TNFR2(+) cells were anergic and markedly inhibited the proliferation and cytokine production of co-cultured T-responder cells. In contrast, CD4(+)CD25(+)TNFR2(-) and CD4(+)CD25(-)TNFR2(+) T cells did not show inhibitory activity. As some non-Treg express TNFR2, the combination of CD25 and TNFR2 must be used to identify a larger population of human Treg, a population that may prove to be of diagnostic and therapeutic benefit in cancer and autoimmune diseases.


Subject(s)
Forkhead Transcription Factors/analysis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/classification , Adult , Antigen Presentation , Antigens, CD/analysis , CTLA-4 Antigen , Cells, Cultured/immunology , Cells, Cultured/metabolism , Coculture Techniques , Flow Cytometry , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-7 Receptor alpha Subunit/analysis , Leukocyte Common Antigens/analysis , Lymphocyte Activation/drug effects , Receptors, Antigen, T-Cell/immunology , Receptors, Tumor Necrosis Factor, Type II/analysis , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
20.
J Nat Prod ; 72(8): 1369-72, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19637889

ABSTRACT

A biological screen used to identify inhibitors of monocyte chemotactic protein-1 (CCL2)-induced chemotaxis was applied in the activity-guided fractionation of an extract from a fungus of the genus Leptoxyphium sp. Inhibition of CCL2-induced chemotaxis was traced to a new dichlorinated diketopiperazine, cyclo(13,15-dichloro-L-Pro-L-Tyr). A structure-activity relationship (SAR) study evaluating relative activities of cyclo(13,15-dichloro-L-Pro-L-Tyr) and a nonchlorinated homologue cyclo(L-Pro-L-Tyr) showed that the dichlorinated molecule was 10- to 20-fold more active than the nonchlorinated form, while no activity was observed for cyclo(D-N-methylLeu-L-Trp).


Subject(s)
Ascomycota/chemistry , Chemokine CCL2/antagonists & inhibitors , Dipeptides/isolation & purification , Dipeptides/pharmacology , Peptides, Cyclic/isolation & purification , Peptides, Cyclic/pharmacology , Dipeptides/chemistry , Ericaceae/microbiology , Molecular Structure , Peptides, Cyclic/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...