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










Publication year range
1.
Nat Commun ; 15(1): 4914, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851846

ABSTRACT

FOXA family proteins act as pioneer factors by remodeling compact chromatin structures. FOXA1 is crucial for the chromatin binding of the androgen receptor (AR) in both normal prostate epithelial cells and the luminal subtype of prostate cancer (PCa). Recent studies have highlighted the emergence of FOXA2 as an adaptive response to AR signaling inhibition treatments. However, the role of the FOXA1 to FOXA2 transition in regulating cancer lineage plasticity remains unclear. Our study demonstrates that FOXA2 binds to distinct classes of developmental enhancers in multiple AR-independent PCa subtypes, with its binding depending on LSD1. Moreover, we reveal that FOXA2 collaborates with JUN at chromatin and promotes transcriptional reprogramming of AP-1 in lineage-plastic cancer cells, thereby facilitating cell state transitions to multiple lineages. Overall, our findings underscore the pivotal role of FOXA2 as a pan-plasticity driver that rewires AP-1 to induce the differential transcriptional reprogramming necessary for cancer cell lineage plasticity.


Subject(s)
Cell Lineage , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-beta , Prostatic Neoplasms , Transcription Factor AP-1 , Male , Humans , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transcription Factor AP-1/metabolism , Transcription Factor AP-1/genetics , Cell Line, Tumor , Cell Lineage/genetics , Histone Demethylases/metabolism , Histone Demethylases/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Animals , Chromatin/metabolism , Chromatin/genetics , Cell Plasticity/genetics , Cellular Reprogramming/genetics , Mice , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-jun/genetics , Enhancer Elements, Genetic/genetics , Transcription, Genetic
2.
Cancer Res ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38657108

ABSTRACT

Solid tumors are highly reliant on lipids for energy, growth, and survival. In prostate cancer, the activity of the androgen receptor (AR) is associated with reprogramming of lipid metabolic processes. Here, we identified acyl-CoA synthetase medium chain family members 1 and 3 (ACSM1 and ACSM3) as AR-regulated mediators of prostate cancer metabolism and growth. ACSM1 and ACSM3 were upregulated in prostate tumors compared to non-malignant tissues and other cancer types. Both enzymes enhanced proliferation and protected prostate cancer cells from death in vitro, while silencing ACSM3 led to reduced tumor growth in an orthotopic xenograft model. ACSM1 and ACSM3 were major regulators of the prostate cancer lipidome and enhanced energy production via fatty acid oxidation. Metabolic dysregulation caused by loss of ACSM1/3 led to mitochondrial oxidative stress, lipid peroxidation and cell death by ferroptosis. Conversely, elevated ACSM1/3 activity enabled prostate cancer cells to survive toxic levels of medium chain fatty acids and promoted resistance to ferroptosis-inducing drugs and AR antagonists. Collectively, this study reveals a tumor-promoting function for medium chain acyl-CoA synthetases and positions ACSM1 and ACSM3 as key players in prostate cancer progression and therapy resistance.

3.
Cancer Cell ; 41(7): 1309-1326.e10, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37295428

ABSTRACT

The first step of oncogenesis is the acquisition of a repertoire of genetic mutations to initiate and sustain the malignancy. An important example of this initiation phase in acute leukemias is the formation of a potent oncogene by chromosomal translocations between the mixed lineage leukemia (MLL) gene and one of 100 translocation partners, known as the MLL recombinome. Here, we show that circular RNAs (circRNAs)-a family of covalently closed, alternatively spliced RNA molecules-are enriched within the MLL recombinome and can bind DNA, forming circRNA:DNA hybrids (circR loops) at their cognate loci. These circR loops promote transcriptional pausing, proteasome inhibition, chromatin re-organization, and DNA breakage. Importantly, overexpressing circRNAs in mouse leukemia xenograft models results in co-localization of genomic loci, de novo generation of clinically relevant chromosomal translocations mimicking the MLL recombinome, and hastening of disease onset. Our findings provide fundamental insight into the acquisition of chromosomal translocations by endogenous RNA carcinogens in leukemia.


Subject(s)
Leukemia , Translocation, Genetic , Animals , Mice , Humans , RNA, Circular/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Leukemia/genetics , Leukemia/pathology , DNA , Oncogene Proteins, Fusion/genetics
4.
Cancer Res Commun ; 2(7): 706-724, 2022 07.
Article in English | MEDLINE | ID: mdl-36923279

ABSTRACT

Inhibiting the androgen receptor (AR), a ligand-activated transcription factor, with androgen deprivation therapy is a standard-of-care treatment for metastatic prostate cancer. Paradoxically, activation of AR can also inhibit the growth of prostate cancer in some patients and experimental systems, but the mechanisms underlying this phenomenon are poorly understood. This study exploited a potent synthetic androgen, methyltestosterone (MeT), to investigate AR agonist-induced growth inhibition. MeT strongly inhibited growth of prostate cancer cells expressing AR, but not AR-negative models. Genes and pathways regulated by MeT were highly analogous to those regulated by DHT, although MeT induced a quantitatively greater androgenic response in prostate cancer cells. MeT potently downregulated DNA methyltransferases, leading to global DNA hypomethylation. These epigenomic changes were associated with dysregulation of transposable element expression, including upregulation of endogenous retrovirus (ERV) transcripts after sustained MeT treatment. Increased ERV expression led to accumulation of double-stranded RNA and a "viral mimicry" response characterized by activation of IFN signaling, upregulation of MHC class I molecules, and enhanced recognition of murine prostate cancer cells by CD8+ T cells. Positive associations between AR activity and ERVs/antiviral pathways were evident in patient transcriptomic data, supporting the clinical relevance of our findings. Collectively, our study reveals that the potent androgen MeT can increase the immunogenicity of prostate cancer cells via a viral mimicry response, a finding that has potential implications for the development of strategies to sensitize this cancer type to immunotherapies. Significance: Our study demonstrates that potent androgen stimulation of prostate cancer cells can elicit a viral mimicry response, resulting in enhanced IFN signaling. This finding may have implications for the development of strategies to sensitize prostate cancer to immunotherapies.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Male , Humans , Animals , Mice , Receptors, Androgen/genetics , Androgens/pharmacology , Prostatic Neoplasms/drug therapy , Androgen Antagonists/pharmacology , CD8-Positive T-Lymphocytes/metabolism , DNA
5.
Cell Rep ; 34(1): 108585, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406413

ABSTRACT

Potent therapeutic inhibition of the androgen receptor (AR) in prostate adenocarcinoma can lead to the emergence of neuroendocrine prostate cancer (NEPC), a phenomenon associated with enhanced cell plasticity. Here, we show that microRNA-194 (miR-194) is a regulator of epithelial-neuroendocrine transdifferentiation. In clinical prostate cancer samples, miR-194 expression and activity were elevated in NEPC and inversely correlated with AR signaling. miR-194 facilitated the emergence of neuroendocrine features in prostate cancer cells, a process mediated by its ability to directly target a suite of genes involved in cell plasticity. One such target was FOXA1, which encodes a transcription factor with a vital role in maintaining the prostate epithelial lineage. Importantly, a miR-194 inhibitor blocked epithelial-neuroendocrine transdifferentiation and inhibited the growth of cell lines and patient-derived organoids possessing neuroendocrine features. Overall, our study reveals a post-transcriptional mechanism regulating the plasticity of prostate cancer cells and provides a rationale for targeting miR-194 in NEPC.


Subject(s)
Cell Transdifferentiation , Hepatocyte Nuclear Factor 3-alpha/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Cell Culture Techniques/methods , Cell Line, Tumor , Cell Lineage , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Male , Mice , PC-3 Cells , Signal Transduction
6.
Sci Transl Med ; 12(559)2020 09 02.
Article in English | MEDLINE | ID: mdl-32878981

ABSTRACT

Intertissue communication is a fundamental feature of metabolic regulation, and the liver is central to this process. We have identified sparc-related modular calcium-binding protein 1 (SMOC1) as a glucose-responsive hepatokine and regulator of glucose homeostasis. Acute intraperitoneal administration of SMOC1 improved glycemic control and insulin sensitivity in mice without changes in insulin secretion. SMOC1 exerted its favorable glycemic effects by inhibiting adenosine 3',5'-cyclic monophosphate (cAMP)-cAMP-dependent protein kinase (PKA)-cAMP response element-binding protein (CREB) signaling in the liver, leading to decreased gluconeogenic gene expression and suppression of hepatic glucose output. Overexpression of SMOC1 in the liver or once-weekly intraperitoneal injections of a stabilized SMOC1-FC fusion protein induced durable improvements in glucose tolerance and insulin sensitivity in db/db mice, without adverse effects on adiposity, liver histopathology, or inflammation. Furthermore, circulating SMOC1 correlated with hepatic and systemic insulin sensitivity and was decreased in obese, insulin-resistant humans. Together, these findings identify SMOC1 as a potential pharmacological target for the management of glycemic control in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Blood Glucose , Glucose , Glycemic Control , Insulin , Liver , Mice , Mice, Inbred C57BL
7.
Cancer Res ; 77(13): 3417-3430, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28473532

ABSTRACT

Alteration to the expression and activity of androgen receptor (AR) coregulators in prostate cancer is an important mechanism driving disease progression and therapy resistance. Using a novel proteomic technique, we identified a new AR coregulator, the transcription factor Grainyhead-like 2 (GRHL2), and demonstrated its essential role in the oncogenic AR signaling axis. GRHL2 colocalized with AR in prostate tumors and was frequently amplified and upregulated in prostate cancer. Importantly, GRHL2 maintained AR expression in multiple prostate cancer model systems, was required for cell proliferation, enhanced AR's transcriptional activity, and colocated with AR at specific sites on chromatin to regulate genes relevant to disease progression. GRHL2 is itself an AR-regulated gene, creating a positive feedback loop between the two factors. The link between GRHL2 and AR also applied to constitutively active truncated AR variants (ARV), as GRHL2 interacted with and regulated ARVs and vice versa. These oncogenic functions of GRHL2 were counterbalanced by its ability to suppress epithelial-mesenchymal transition and cell invasion. Mechanistic evidence suggested that AR assisted GRHL2 in maintaining the epithelial phenotype. In summary, this study has identified a new AR coregulator with a multifaceted role in prostate cancer, functioning as an enhancer of the oncogenic AR signaling pathway but also as a suppressor of metastasis-related phenotypes. Cancer Res; 77(13); 3417-30. ©2017 AACR.


Subject(s)
DNA-Binding Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription Factors/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Chick Embryo , DNA-Binding Proteins/metabolism , Humans , Male , Oncogenes , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Transfection
8.
Cancer Res ; 77(4): 1021-1034, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28011622

ABSTRACT

Serum levels of miR-194 have been reported to predict prostate cancer recurrence after surgery, but its functional contributions to this disease have not been studied. Herein, it is demonstrated that miR-194 is a driver of prostate cancer metastasis. Prostate tissue levels of miR-194 were associated with disease aggressiveness and poor outcome. Ectopic delivery of miR-194 stimulated migration, invasion, and epithelial-mesenchymal transition in human prostate cancer cell lines, and stable overexpression of miR-194 enhanced metastasis of intravenous and intraprostatic tumor xenografts. Conversely, inhibition of miR-194 activity suppressed the invasive capacity of prostate cancer cell lines in vitro and in vivo Mechanistic investigations identified the ubiquitin ligase suppressor of cytokine signaling 2 (SOCS2) as a direct, biologically relevant target of miR-194 in prostate cancer. Low levels of SOCS2 correlated strongly with disease recurrence and metastasis in clinical specimens. SOCS2 downregulation recapitulated miR-194-driven metastatic phenotypes, whereas overexpression of a nontargetable SOCS2 reduced miR-194-stimulated invasion. Targeting of SOCS2 by miR-194 resulted in derepression of the oncogenic kinases FLT3 and JAK2, leading to enhanced ERK and STAT3 signaling. Pharmacologic inhibition of ERK and JAK/STAT pathways reversed miR-194-driven phenotypes. The GATA2 transcription factor was identified as an upstream regulator of miR-194, consistent with a strong concordance between GATA2 and miR-194 levels in clinical specimens. Overall, these results offer new insights into the molecular mechanisms of metastatic progression in prostate cancer. Cancer Res; 77(4); 1021-34. ©2016 AACR.


Subject(s)
MicroRNAs/physiology , Prostatic Neoplasms/pathology , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , GATA2 Transcription Factor/physiology , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/genetics , STAT3 Transcription Factor/physiology , Suppressor of Cytokine Signaling Proteins/physiology
9.
Methods Mol Biol ; 1024: 235-46, 2013.
Article in English | MEDLINE | ID: mdl-23719956

ABSTRACT

MicroRNAs (miRNAs) derived from the cell-free fractions of blood are emerging as useful noninvasive markers of cancer. However, many tumors display significant molecular heterogeneity, which is likely to be reflected in the circulating miRNA fingerprints associated with that pathology. One strategy to minimize such heterogeneity is to employ genetically engineered mouse models of human cancer. Here, we describe a method to profile miRNAs in the serum of a mouse model of prostate cancer, TRansgenic Adenocarcinoma of Mouse Prostate (TRAMP), and discuss practical considerations for translating these potential biomarkers into a clinical setting.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/blood , MicroRNAs/blood , Prostatic Neoplasms/diagnosis , Real-Time Polymerase Chain Reaction/standards , Reverse Transcriptase Polymerase Chain Reaction/standards , Adenocarcinoma/blood , Adenocarcinoma/genetics , Animals , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics
10.
Oncotarget ; 4(5): 691-704, 2013 May.
Article in English | MEDLINE | ID: mdl-23674566

ABSTRACT

The development of lethal, castration resistant prostate cancer is associated with adaptive changes to the androgen receptor (AR), including the emergence of mutant receptors and truncated, constitutively active AR variants. AR relies on the molecular chaperone HSP90 for its function in both normal and malignant prostate cells, but the requirement for HSP90 in environments with aberrant AR expression is largely unknown. Here, we investigated the efficacy of three HSP90 inhibitors, 17-AAG, HSP990 and AUY922, against clinically-relevant AR missense mutants and truncated variants. HSP90 inhibition effectively suppressed the signaling of wild-type AR and all AR missense mutants tested. By contrast, two truncated AR variants, AR-V7 and ARv567es, exhibited marked resistance to HSP90 inhibitors. Supporting this observation, nuclear localization of the truncated AR variants was not affected by HSP90 inhibition and AR variant:HSP90 complexes could not be detected in prostate cancer cells. Interestingly, HSP90 inhibition resulted in accumulation of AR-V7 and ARv567es in both cell lines and human tumor explants. Despite the apparent independence of AR variants from HSP90 and their treatment-associated induction, the growth of cell lines with endogenous or enforced expression of AR-V7 or ARv567es remained highly sensitive to AUY922. This study demonstrates that functional AR variant signaling does not confer resistance to HSP90 inhibition, yields insight into the interaction between AR and HSP90 and provides further impetus for the clinical application of HSP90 inhibitors in advanced prostate cancer.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Proliferation , Genetic Variation , Humans , Isoxazoles/pharmacology , Lactams, Macrocyclic/pharmacology , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/genetics , Resorcinols/pharmacology , Signal Transduction , Transcription, Genetic/drug effects
11.
Int J Cancer ; 131(3): 652-61, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22052531

ABSTRACT

Circulating microRNAs (miRNAs) are emerging as useful non-invasive markers of disease. The objective of this study was to use a mouse model of prostate cancer as a tool to discover serum miRNAs that could be assessed in a clinical setting. Global miRNA profiling identified 46 miRNAs at significantly altered levels (p ≤ 0.05) in the serum of TRansgenic Adenocarcinoma of Mouse Prostate (TRAMP) mice with advanced prostate cancer compared to healthy controls. A subset of these miRNAs with known human homologues were validated in an independent cohort of mice and then measured in serum from men with metastatic castration-resistant prostate cancer (mCRPC; n = 25) or healthy men (n = 25). Four miRNAs altered in mice, mmu-miR-141, mmu-miR-298, mmu-miR-346 and mmu-miR-375, were also found to be at differential levels in the serum of men with mCRPC. Three of these (hsa-miR-141, hsa-miR-298 and hsa-miR-375) were upregulated in prostate tumors compared with normal prostate tissue, suggesting that they are released into the blood as disease progresses. Moreover, the intra-tumoral expression of hsa-miR-141 and hsa-miR-375 were predictors of biochemical relapse after surgery. This study is the first to demonstrate that specific serum miRNAs are common between human prostate cancer and a mouse model of the disease, highlighting the potential of such models for the discovery of novel biomarkers.


Subject(s)
Biomarkers, Tumor/blood , MicroRNAs/blood , Prostatic Neoplasms/genetics , Animals , Biomarkers, Tumor/genetics , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Prognosis , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology
12.
Nat Commun ; 2: 287, 2011.
Article in English | MEDLINE | ID: mdl-21505443

ABSTRACT

The epithelial sodium channel (ENaC) is essential for sodium homoeostasis in many epithelia. ENaC activity is required for lung fluid clearance in newborn animals and for maintenance of blood volume and blood pressure in adults. In vitro studies show that the ubiquitin ligase Nedd4-2 ubiquitinates ENaC to regulate its cell surface expression. Here we show that knockout of Nedd4-2 in mice leads to increased ENaC expression and activity in embryonic lung. This increased ENaC activity is the likely reason for premature fetal lung fluid clearance in Nedd4-2(-/-) animals, resulting in a failure to inflate lungs and perinatal lethality. A small percentage of Nedd4-2(-/-) animals survive up to 22 days, and these animals also show increased ENaC expression and develop lethal sterile inflammation of the lung. Thus, we provide critical in vivo evidence that Nedd4-2 is essential for correct regulation of ENaC expression, fetal and postnatal lung function and animal survival.


Subject(s)
Endosomal Sorting Complexes Required for Transport/deficiency , Epithelial Sodium Channels/metabolism , Lung Diseases/genetics , Lung/pathology , Ubiquitin-Protein Ligases/deficiency , Animals , Blood Pressure , Blotting, Northern , Bronchoalveolar Lavage , Chemokines/metabolism , Cytokines/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endosomal Sorting Complexes Required for Transport/metabolism , Immunoblotting , Lung/embryology , Lung/metabolism , Lung Diseases/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Nedd4 Ubiquitin Protein Ligases , Ubiquitin-Protein Ligases/metabolism
13.
J Autoimmun ; 36(3-4): 278-87, 2011 May.
Article in English | MEDLINE | ID: mdl-21396797

ABSTRACT

The Class IA phosphoinositide 3-kinase delta (PI3Kδ) has been implicated in multiple signaling pathways involved in leukocyte activation and hence is an attractive target in many human autoimmune diseases, including multiple sclerosis (MS). Here, using mice expressing a catalytically inactive form of the PI3Kδ subunit p110δ, we show that signaling through PI3Kδ is required for full and sustained pathology of experimental autoimmune encephalomyelitis (EAE), a Th17-driven model of MS. In p110δ-inactivated mice, T cell activation and function during EAE was markedly reduced and fewer T cells were observed in the central nervous system (CNS). The decrease in T cell activation is unlikely to be due to defects in dendritic cell (DC) function, as p110δ-inactivated DCs migrate and present antigen normally. However, significant increases in the proportion of T cells undergoing apoptosis at early stages of EAE were evident in the absence of PI3Kδ activity. Furthermore, a profound defect in Th17 cellular responses during EAE was apparent in the absence of PI3Kδ activity while Th1 responses were less affected. A highly selective PI3Kδ inhibitor, IC87114, also had greater inhibitory effects on Th17 cell generation in vitro than it did on Th1 cell generation. Thus, PI3Kδ plays an important role in Th17 responses in EAE, suggesting that small molecule inhibitors of PI3Kδ may be useful therapeutics for treatment of MS and other autoimmune diseases.


Subject(s)
Apoptosis , Encephalomyelitis, Autoimmune, Experimental/etiology , Phosphatidylinositol 3-Kinases/physiology , T-Lymphocytes/physiology , Th17 Cells/cytology , Animals , Cell Differentiation , Class I Phosphatidylinositol 3-Kinases , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunologic Memory , Male , Mice , Mice, Inbred C57BL , Phosphoinositide-3 Kinase Inhibitors , T-Lymphocytes/cytology , Th1 Cells/cytology
14.
J Immunol ; 182(5): 3121-30, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234209

ABSTRACT

Chemokines are essential for homeostasis and activation of the immune system. The chemokine ligand/receptor pairing CCL20/CCR6 is interesting because these molecules display characteristics of both homeostatic and activation functions. These dual characteristics suggest a role for CCR6 in the priming and effector phases of the immune response. However, while CCR6 has been implicated in the effector phase in several models, a role in the priming phase is less clear. Herein we analyze the role of CCR6 in these two important arms of the immune response during experimental autoimmune encephalomyelitis (EAE). Both CCR6 and its chemokine ligand CCL20 were up-regulated in the draining lymph nodes and spinal cord during EAE, and CCR6 was up-regulated on CD4(+) T cells that had divided following induction of EAE. The functional role of this expression was demonstrated by impaired development of EAE in gene-targeted CCR6-deficient mice and in mice treated either with a neutralizing anti-CCR6 Ab or with a novel receptor antagonist. Inhibition of EAE was due to reduced priming of autoreactive CD4(+) T cells probably as a result of impaired late-stage influx of dendritic cells into draining lymph nodes. This was accompanied by reduced egress of activated lymphocytes from the lymph nodes. These results demonstrate a novel role for CCR6 in the mechanism of autoreactive lymphocyte priming and emigration to the efferent lymphatics.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Receptors, CCR6/antagonists & inhibitors , Receptors, CCR6/physiology , Amino Acid Sequence , Animals , Chemokine CCL20/biosynthesis , Chemokine CCL20/genetics , Chemokine CCL20/physiology , Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Vessels/immunology , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Receptors, CCR6/biosynthesis , Severity of Illness Index , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology
15.
Brain Pathol ; 18(4): 504-16, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18422759

ABSTRACT

Chemokines regulate lymphocyte trafficking under physiologic and pathologic conditions. In this study, we have investigated the role of CXCR3 and CXCR4 in the activation of T lymphocytes and their migration to the central nervous system (CNS) using novel mutant chemokines to antagonize CXCR3 and CXCR4 specifically. A series of truncation mutants of CXCL11, which has the highest affinity for CXCR3, were synthesized, and an antagonist, CXCL11((4-79)), was obtained. CXCL11((4-79)) strongly inhibited the migration of activated mouse T cells in response to all three high-affinity CXCR3 ligands, CXCL9, 10 and 11. CXCL12((P2G2)), while exhibiting minimal agonistic activity, potently inhibited the migration of activated mouse T cells in response to CXCL12. Interfering with the action of CXCR3 and CXCR4 with these synthetic receptor antagonists inhibited experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis and reduced the accumulation of CD4(+) T cells in the CNS. Further investigation demonstrated that CXCL12((P2G2)) inhibited the sensitization phase, whereas CXCL11((4-79)) inhibited the effector phase of the immune response. Our data suggest that simultaneous targeting of CXCR4 and CXCR3 may be of benefit in the treatment of the CNS autoimmune disease.


Subject(s)
Central Nervous System/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunologic Factors/pharmacology , Receptors, CXCR3/antagonists & inhibitors , Receptors, CXCR4/antagonists & inhibitors , Adoptive Transfer , Animals , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/physiopathology , Chemokine CXCL11/antagonists & inhibitors , Chemokine CXCL11/genetics , Chemokine CXCL11/immunology , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Chemokines/agonists , Chemokines/genetics , Chemokines/immunology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Immunologic Factors/therapeutic use , Immunosuppression Therapy/methods , Mice , Molecular Sequence Data , Peptides/genetics , Peptides/immunology , Peptides/pharmacology , Receptors, CXCR3/immunology , Receptors, CXCR4/immunology , Sequence Homology, Amino Acid , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Treatment Outcome
16.
Infect Immun ; 74(12): 6885-94, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16982830

ABSTRACT

CXCL16 is a recently discovered multifaceted chemokine that has been shown not only to recruit activated T lymphocytes but also to play a direct role in the binding and phagocytosis of bacteria by professional antigen-presenting cells. In this study, we investigated the role of CXCL16 in vivo in the regulation of the immune response using a murine model of Salmonella enterica serovar Enteritidis infection. The expression of CXCL16 was strongly upregulated in the spleens and livers of animals developing an immune response to a primary acute infection but not in the Peyer's patches. Animals developing a secondary response after reexposure to the bacteria displayed a similar pattern of expression. During the primary response, prior treatment with neutralizing antibodies to CXCL16 induced a significant increase in bacterial burden in the spleen and liver. The production of gamma interferon (IFN-gamma) by the lymphocytes in the spleen was decreased by anti-CXCL16 treatment. In comparison, during the secondary response, anti-CXCL16 treatment also significantly increased bacterial burden in both the spleen and liver but had no effect on IFN-gamma production. No role was found for CXCL16 in the production of antibody against SefA, a major surface antigen of S. enteritidis. Together, these results demonstrate a role for CXCL16 in the control of bacterial colonization of target organs and, more specifically, in the regulation of the cell-mediated arm of the primary response to S. enteritidis.


Subject(s)
Chemokines, CXC/physiology , Interferon-gamma/metabolism , Receptors, Scavenger/physiology , Salmonella Infections/immunology , Salmonella enteritidis , Animals , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Chemokine CXCL10 , Chemokine CXCL11 , Chemokine CXCL16 , Chemokine CXCL6 , Chemokine CXCL9 , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Female , Interferon-gamma/analysis , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Receptors, Scavenger/antagonists & inhibitors , Receptors, Scavenger/genetics , Salmonella Infections/genetics , Up-Regulation
17.
J Immunol ; 174(11): 6677-85, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905507

ABSTRACT

UVB irradiation of the shaved dorsal skin of mice can cause both local and systemic suppression of contact hypersensitivity responses; the former demonstrated by administration of the sensitizing Ag/hapten to the irradiated site and the latter by its administration at least 72 h later to distal unirradiated sites. The immunological basis of systemic immunomodulation is not clear. When haptens (trinitrochlorobenzene, FITC) were administered to the shaved ventral skin 4 days after irradiation (8 kJ/m(2)) to the shaved dorsum of BALB/c mice, CD11c(+)/FITC(+) cells in the skin-draining lymph nodes from control and irradiated mice produced on a per cell basis similar levels of IL-12 and PGE(2) were phenotypically mature and efficient at presenting FITC to lymphocytes from FITC-sensitized mice. Ag presentation by FACS-sorted CD11c(+) lymph node cells isolated 4 days after UVB irradiation was as efficient as were cells from unirradiated mice at presentation in vitro of an OVA peptide (OVA(323-339)) to CD4(+) cells from OVA-TCR-transgenic DO11.10 mice. Further, IFN-gamma levels were increased in the cultures containing CD11c(+) cells from UVB-irradiated mice, suggesting that inflammation may precede downstream immunosuppression. These results suggest that the primary cause of reduced contact hypersensitivity responses in mice in which UV irradiation and the sensitizing Ag are applied to different sites several days apart must originate from cells other than CD11c(+) APCs that directly or by production of soluble mediators (IL-12, PGE(2)) affect cellular responses in the nodes of UVB-irradiated mice.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/radiation effects , Cell Differentiation/immunology , Cell Differentiation/radiation effects , Immunologic Factors/biosynthesis , Immunologic Factors/radiation effects , Lymph Nodes/immunology , Ultraviolet Rays , Administration, Topical , Amino Acid Sequence , Animals , Antigen Presentation/radiation effects , Antigen-Presenting Cells/pathology , CD11c Antigen/biosynthesis , Cell Movement/immunology , Cell Movement/radiation effects , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/radiation effects , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Dinoprostone/biosynthesis , Dinoprostone/physiology , Dinoprostone/radiation effects , Female , Fluorescein-5-isothiocyanate/administration & dosage , Fluorescein-5-isothiocyanate/metabolism , Fluorescein-5-isothiocyanate/radiation effects , Haptens/administration & dosage , Haptens/biosynthesis , Haptens/radiation effects , Immunologic Factors/physiology , Interleukin-12/biosynthesis , Interleukin-12/physiology , Interleukin-12/radiation effects , Lymph Nodes/cytology , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Picryl Chloride/administration & dosage , Picryl Chloride/immunology , Skin/immunology , Skin/pathology , Skin/radiation effects
18.
Lab Invest ; 84(11): 1501-11, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15448707

ABSTRACT

While chemokines are clearly important in the generation of protective immunity, the role of individual chemokines in the control of bacterial infection is still poorly understood. In this study, we investigated the role of macrophage inflammatory protein (MIP)-3alpha/CCL20, a chemokine that attracts activated T and B lymphocytes and immature dendritic cells, in host responses to bacterial infection. CCL20 production was induced in subcutaneous tissue in the BALB/c mouse in response to Salmonella enteritidis, Staphylococcus aureus and zymosan, with S. enteritidis being the most potent. S. enteritidis induced CCL20 production in the spleen following either oral administration or injection into the peritoneal cavity. In contrast, no increase was observed in the Peyer's patches. In this model, following intraperitoneal injection, dose-dependent colonization of the spleen and Peyer's patches by S. enteritidis, expression of IFNgamma and IL-4, and production of antibodies against the S. enteritidis surface antigen SefA were observed. Prior treatment with neutralizing antibodies against CCL20 enhanced bacterial dissemination to the spleen and Peyer's patches and strongly biased the IFNgamma/IL-4 ratio towards a type 2 profile in the spleen, while the humoral response was unaffected. In contrast, treatment with neutralizing anti-MIP-1alpha/CCL3 antibodies enhanced the bacterial burden in the Peyer's patches but not in the spleen, had no significant effect on the cytokine ratio, but significantly inhibited anti-SefA production. Together, these results demonstrate an important role for CCL20 in the control of bacterial infection and more specifically in the regulation of cell-mediated immunity against intracellular bacteria such as S. enteritidis.


Subject(s)
Chemokines, CC/physiology , Macrophage Inflammatory Proteins/physiology , Salmonella Infections/microbiology , Salmonella enteritidis/growth & development , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Chemokine CCL20 , Chemokine CCL3 , Chemokine CCL4 , Chemokines, CC/immunology , Colony Count, Microbial , Dose-Response Relationship, Immunologic , Female , Fimbriae Proteins/immunology , Fimbriae Proteins/metabolism , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Macrophage Inflammatory Proteins/immunology , Macrophage Inflammatory Proteins/metabolism , Mice , Mice, Inbred BALB C , Neutralization Tests , Peritoneal Cavity/microbiology , Peyer's Patches/immunology , Peyer's Patches/microbiology , Salmonella Infections/immunology , Salmonella enteritidis/immunology , Spleen/immunology , Spleen/metabolism , Spleen/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology
19.
Methods ; 28(1): 79-89, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231191

ABSTRACT

There is a direct correlation between dermal mast cell prevalence in dorsal skin of different mouse strains and susceptibility to UVB-induced systemic immunosuppression; highly UV-susceptible C57BL/6 mice have a high dermal mast cell prevalence while BALB/c mice, which require considerable UV radiation for 50% immunosuppression, have a low mast cell prevalence. There is also a functional link between the prevalence of dermal mast cells and susceptibility to UVB- and cis-urocanic acid (UCA)-induced systemic immunosuppression. Mast cell-depleted mice are unresponsive to UVB or cis-UCA for systemic immunosuppression unless they are previously reconstituted at the irradiated or cis-UCA-administered site with bone marrow-derived mast cell precursors. cis-UCA does not stimulate mast cell degranulation directly. Instead, in support of studies showing that neither UVB nor cis-UCA was immunosuppressive in capsaicin-treated, neuropeptide-depleted mice, cis-UCA-stimulated neuropeptide release from sensory c-fibers which, in turn, could efficiently degranulate mast cells. Studies in mice suggested that histamine, and not tumor necrosis factor alpha (TNF-alpha), was the product from mast cells that stimulated downstream immunosuppression. Histamine receptor antagonists reduced by approximately 60% UVB and cis-UCA-induced systemic immunosuppression. Indomethacin administration to mice had a similar effect which was not cumulative with the histamine receptor antagonists. Histamine can stimulate keratinocyte prostanoid production. We propose that both histamine and prostaglandin E(2) are important in downstream immunosuppression; both are regulatory molecules supporting the development of T helper 2 cells and reduced expression of type 1 immune responses such as a contact hypersensitivity reaction.


Subject(s)
Histamine/physiology , Immune Tolerance/radiation effects , Mast Cells/physiology , Neuropeptides/physiology , Prostaglandins/physiology , Ultraviolet Rays , Animals , Cell Degranulation/drug effects , Dermatitis, Contact/prevention & control , Female , Indomethacin/pharmacology , Male , Mice , Mice, Inbred Strains , Microcirculation/drug effects , Nerve Growth Factor/physiology , Rats , Rats, Sprague-Dawley , Urocanic Acid/pharmacology
20.
J Invest Dermatol ; 118(3): 396-401, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11874476

ABSTRACT

The induction of systemic immunosuppression following ultraviolet B radiation exposure has been linked with the release of inflammatory and immunomodulatory mediators by cells of the epidermis and dermis. Nerve growth factor has not previously been linked with ultraviolet-B-induced immunosuppressive effects. Nerve growth factor antibodies abrogated ultraviolet-B-induced systemic suppression of contact hypersensitivity responses in BALB/C mice. Subcutaneous injection of nerve growth factor (20 microg per mouse) into dorsal skin 5 d before hapten sensitization on ventral skin suppressed contact hypersensitivity responses in mast-cell-replete but not Wf/Wf mast-cell-depleted mice. Nerve growth factor injected 24 h prior to challenge was not able to suppress the efferent phase of the contact hypersensitivity response. Subcutaneous injection of nerve growth factor (20 microg per mouse) did not suppress contact hypersensitivity responses in capsaicin-pretreated (neuropeptide-depleted) BALB/c mice, and thus sensory c-fibers are necessary for nerve-growth-factor-mediated systemic suppression of contact hypersensitivity responses. Increased concentrations of nerve growth factor within epidermal keratinocytes 8 h after ultraviolet B irradiation were confirmed immunohistochemically. These findings support a role for keratinocyte-derived nerve growth factor via its action on sensory c-fibers, and subsequent release of neuropeptides to mediate mast cell degranulation in systemic suppression of contact hypersensitivity responses in mice following ultraviolet B exposure.


Subject(s)
Dermatitis, Contact/radiotherapy , Keratinocytes/radiation effects , Mast Cells/physiology , Nerve Growth Factor/physiology , Neuropeptides/physiology , Ultraviolet Rays , Animals , Antibodies/immunology , Cell Degranulation/physiology , Immunosuppressive Agents/pharmacology , Keratinocytes/metabolism , Mice , Mice, Inbred BALB C , Nerve Fibers/physiology , Nerve Growth Factor/immunology , Nerve Growth Factor/pharmacology , Neurons, Afferent/physiology , Peritoneum/cytology , Skin/cytology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...