Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nat Immunol ; 12(3): 239-46, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21278735

ABSTRACT

Colonic homeostasis entails epithelium-lymphocyte cooperation, yet many participants in this process are unknown. We show here that epithelial microRNAs mediate the mucosa-immune system crosstalk necessary for mounting protective T helper type 2 (T(H)2) responses. Abolishing the induction of microRNA by gut-specific deletion of Dicer1 (Dicer1(Δgut)), which encodes an enzyme involved in microRNA biogenesis, deprived goblet cells of RELMß, a key T(H)2 antiparasitic cytokine; this predisposed the host to parasite infection. Infection of Dicer1(Δgut) mice with helminths favored a futile T(H)1 response with hallmarks of inflammatory bowel disease. Interleukin 13 (IL-13) induced the microRNA miR-375, which regulates the expression of TSLP, a T(H)2-facilitating epithelial cytokine; this indicated a T(H)2-amplification loop. We found that miR-375 was required for RELMß expression in vivo; miR-375-deficient mice had significantly less intestinal RELMß, which possibly explains the greater susceptibility of Dicer1(Δgut) mice to parasites. Our findings indicate that epithelial microRNAs are key regulators of gut homeostasis and mucosal immunity.


Subject(s)
Immunity, Mucosal/immunology , MicroRNAs/immunology , T-Lymphocytes/immunology , Animals , Cell Communication , Epithelium/immunology , Gastrointestinal Tract/immunology , HT29 Cells , Humans , Immunohistochemistry , Interleukin-13/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
2.
Cancer Immunol Immunother ; 70(12): 3525-3540, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33903974

ABSTRACT

Immune checkpoint inhibitors (ICIs) have emerged as promising therapies for the treatment of cancer. However, existing ICIs, namely PD-(L)1 and CTLA-4 inhibitors, generate durable responses only in a subset of patients. TIGIT is a co-inhibitory receptor and member of the DNAM-1 family of immune modulating proteins. We evaluated the prevalence of TIGIT and its cognate ligand, PVR (CD155), in human cancers by assessing their expression in a large set of solid tumors. TIGIT is expressed on CD4+ and CD8+ TILs and is upregulated in tumors compared to normal tissues. PVR is expressed on tumor cells and tumor-associated macrophages from multiple solid tumors. We explored the therapeutic potential of targeting TIGIT by generating COM902, a fully human anti-TIGIT hinge-stabilized IgG4 monoclonal antibody that binds specifically to human, cynomolgus monkey, and mouse TIGIT, and disrupts the binding of TIGIT with PVR. COM902, either alone or in combination with a PVRIG (COM701) or PD-1 inhibitor, enhances antigen-specific human T cell responses in-vitro. In-vivo, a mouse chimeric version of COM902 in combination with an anti-PVRIG or anti-PD-L1 antibody inhibited tumor growth and increased survival in two syngeneic mouse tumor models. In summary, COM902 enhances anti-tumor immune responses and is a promising candidate for the treatment of advanced malignancies.


Subject(s)
Antibodies, Monoclonal/immunology , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology , Signal Transduction/immunology , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Female , Humans , Immunoglobulin G/immunology , Immunotherapy/methods , Jurkat Cells , Macaca fascicularis , Mice , Mice, Inbred BALB C
3.
J Immunol ; 200(6): 2025-2037, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29431694

ABSTRACT

The B7-like protein family members play critical immunomodulatory roles and constitute attractive targets for the development of novel therapies for human diseases. We identified Ig-like domain-containing receptor (ILDR)2 as a novel B7-like protein with robust T cell inhibitory activity, expressed in immune cells and in immune-privileged and inflamed tissues. A fusion protein, consisting of ILDR2 extracellular domain with an Fc fragment, that binds to a putative counterpart on activated T cells showed a beneficial effect in the collagen-induced arthritis model and abrogated the production of proinflammatory cytokines and chemokines in autologous synovial-like cocultures of macrophages and cytokine-stimulated T cells. Collectively, these findings point to ILDR2 as a novel negative regulator for T cells, with potential roles in the development of immune-related diseases, including autoimmunity and cancer.


Subject(s)
B7 Antigens/immunology , Membrane Proteins/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Cytokines/immunology , Humans , Immunoglobulin Domains/immunology , Immunoglobulin Fc Fragments/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C
4.
Genes Dev ; 24(5): 470-7, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20194439

ABSTRACT

beta-TrCP, the substrate recognition subunit of a Skp1-Cul1-F-box (SCF) ubiquitin ligase, is ubiquitously expressed from two distinct paralogs, targeting many regulatory proteins for proteasomal degradation. We generated inducible beta-TrCP hypomorphic mice and found that they are surprisingly healthy, yet have a severe testicular defect. We show that the two beta-TrCP paralogs have a nonredundant role in spermatogenesis. The testicular defect is tightly associated with cell adhesion failure within the seminiferous tubules and is fully reversible upon beta-TrCP restoration. Remarkably, testicular depletion of a single beta-TrCP substrate, Snail1, rescued the adhesion defect and restored spermatogenesis. Our studies highlight an unexpected functional reserve of this central E3, as well as a bottleneck in a specific tissue: a single substrate whose stabilization is incompatible with testicular differentiation.


Subject(s)
SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Spermatogenesis/physiology , Testis/embryology , Transcription Factors/deficiency , Animals , Intercellular Junctions/pathology , Male , Mice , Mice, Transgenic , Models, Animal , Phenotype , Protein Isoforms , Snail Family Transcription Factors , Testis/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nature ; 470(7334): 409-13, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21331045

ABSTRACT

The mature gut renews continuously and rapidly throughout adult life, often in a damage-inflicting micro-environment. The major driving force for self-renewal of the intestinal epithelium is the Wnt-mediated signalling pathway, and Wnt signalling is frequently hyperactivated in colorectal cancer. Here we show that casein kinase Iα (CKIα), a component of the ß-catenin-destruction complex, is a critical regulator of the Wnt signalling pathway. Inducing the ablation of Csnk1a1 (the gene encoding CKIα) in the gut triggers massive Wnt activation, surprisingly without causing tumorigenesis. CKIα-deficient epithelium shows many of the features of human colorectal tumours in addition to Wnt activation, in particular the induction of the DNA damage response and cellular senescence, both of which are thought to provide a barrier against malignant transformation. The epithelial DNA damage response in mice is accompanied by substantial activation of p53, suggesting that the p53 pathway may counteract the pro-tumorigenic effects of Wnt hyperactivation. Notably, the transition from benign adenomas to invasive colorectal cancer in humans is typically linked to p53 inactivation, underscoring the importance of p53 as a safeguard against malignant progression; however, the mechanism of p53-mediated tumour suppression is unknown. We show that the maintenance of intestinal homeostasis in CKIα-deficient gut requires p53-mediated growth control, because the combined ablation of Csnk1a1 and either p53 or its target gene p21 (also known as Waf1, Cip1, Sdi1 and Cdkn1a) triggered high-grade dysplasia with extensive proliferation. Unexpectedly, these ablations also induced non-proliferating cells to invade the villous lamina propria rapidly, producing invasive carcinomas throughout the small bowel. Furthermore, in p53-deficient gut, loss of heterozygosity of the gene encoding CKIα caused a highly invasive carcinoma, indicating that CKIα functions as a tumour suppressor when p53 is inactivated. We identified a set of genes (the p53-suppressed invasiveness signature, PSIS) that is activated by the loss of both p53 and CKIα and which probably accounts for the brisk induction of invasiveness. PSIS transcription and tumour invasion were suppressed by p21, independently of cell cycle control. Restraining tissue invasion through suppressing PSIS expression is thus a novel tumour-suppressor function of wild-type p53.


Subject(s)
Casein Kinase Ialpha/deficiency , Colorectal Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Adenoma/enzymology , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Casein Kinase Ialpha/genetics , Casein Kinase Ialpha/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Cellular Senescence , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Disease Progression , Female , Fibroblasts , Genes, APC , Genes, Tumor Suppressor , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Loss of Heterozygosity , Male , Mice , Mice, Knockout , Neoplasm Invasiveness/pathology , Signal Transduction , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
6.
Cancer Immunol Res ; 12(7): 876-890, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38752503

ABSTRACT

Cancers that are poorly immune infiltrated pose a substantial challenge, with current immunotherapies yielding limited clinical success. Stem-like memory T cells (TSCM) have been identified as a subgroup of T cells that possess strong proliferative capacity and that can expand and differentiate following interactions with dendritic cells (DCs). In this study, we explored the pattern of expression of a recently discovered inhibitory receptor poliovirus receptor-related immunoglobulin domain protein (PVRIG) and its ligand, poliovirus receptor-related ligand 2 (PVRL2), in the human tumor microenvironment. Using spatial and single-cell RNA transcriptomics data across diverse cancer indications, we found that among the T-cell checkpoints, PVRIG is uniquely expressed on TSCM and PVRL2 is expressed on DCs in immune aggregate niches in tumors. PVRIG blockade could therefore enhance TSCM-DC interactions and efficiently drive T-cell infiltration to tumors. Consistent with these data, following PVRIG blockade in patients with poorly infiltrated tumors, we observed immune modulation including increased tumor T-cell infiltration, T-cell receptor (TCR) clonality, and intratumoral T-cell expansion, all of which were associated with clinical benefit. These data suggest PVRIG blockade as a promising strategy to induce potent antitumor T-cell responses, providing a novel approach to overcome resistance to immunotherapy in immune-excluded tumors.


Subject(s)
Dendritic Cells , Neoplasms , Tumor Microenvironment , Humans , Dendritic Cells/immunology , Dendritic Cells/metabolism , Tumor Microenvironment/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Memory T Cells/immunology , Memory T Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Cancer Immunol Res ; 12(6): 687-703, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38592331

ABSTRACT

Recombinant cytokines have limited anticancer efficacy mostly due to a narrow therapeutic window and systemic adverse effects. IL18 is an inflammasome-induced proinflammatory cytokine, which enhances T- and NK-cell activity and stimulates IFNγ production. The activity of IL18 is naturally blocked by a high-affinity endogenous binding protein (IL18BP). IL18BP is induced in the tumor microenvironment (TME) in response to IFNγ upregulation in a negative feedback mechanism. In this study, we found that IL18 is upregulated in the TME compared with the periphery across multiple human tumors and most of it is bound to IL18BP. Bound IL18 levels were largely above the amount required for T-cell activation in vitro, implying that releasing IL18 in the TME could lead to potent T-cell activation. To restore the activity of endogenous IL18, we generated COM503, a high-affinity anti-IL18BP that blocks the IL18BP:IL18 interaction and displaces precomplexed IL18, thereby enhancing T- and NK-cell activation. In vivo, administration of a surrogate anti-IL18BP, either alone or in combination with anti-PD-L1, resulted in significant tumor growth inhibition and increased survival across multiple mouse tumor models. Moreover, the anti-IL18BP induced pronounced TME-localized immune modulation including an increase in polyfunctional nonexhausted T- and NK-cell numbers and activation. In contrast, no increase in inflammatory cytokines and lymphocyte numbers or activation state was observed in serum and spleen. Taken together, blocking IL18BP using an Ab is a promising approach to harness cytokine biology for the treatment of cancer.


Subject(s)
Interleukin-18 , Tumor Microenvironment , Animals , Humans , Interleukin-18/metabolism , Mice , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Cell Line, Tumor , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Neoplasms/immunology , Neoplasms/drug therapy , Lymphocyte Activation/immunology , Lymphocyte Activation/drug effects , Female , Mice, Inbred C57BL , Intercellular Signaling Peptides and Proteins/metabolism , Xenograft Model Antitumor Assays , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
8.
Nat Cell Biol ; 8(12): 1327-36, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17128265

ABSTRACT

The mechanisms by which commensal bacteria suppress inflammatory signalling in the gut are still unclear. Here, we present a cellular mechanism whereby the polarity of intestinal epithelial cells (IECs) has a major role in colonic homeostasis. TLR9 activation through apical and basolateral surface domains have distinct transcriptional responses, evident by NF-kappaB activation and cDNA microarray analysis. Whereas basolateral TLR9 signals IkappaBalpha degradation and activation of the NF-kappaB pathway, apical TLR9 stimulation invokes a unique response in which ubiquitinated IkappaB accumulates in the cytoplasm preventing NF-kappaB activation. Furthermore, apical TLR9 stimulation confers intracellular tolerance to subsequent TLR challenges. IECs in TLR9-deficient mice, when compared with wild-type and TLR2-deficient mice, display a lower NF-kappaB activation threshold and these mice are highly susceptible to experimental colitis. Our data provide a case for organ-specific innate immunity in which TLR expression in polarized IECs has uniquely evolved to maintain colonic homeostasis and regulate tolerance and inflammation.


Subject(s)
Cell Polarity , Colon/cytology , Enterocytes/cytology , Homeostasis , Signal Transduction , Toll-Like Receptor 9/metabolism , Animals , Caco-2 Cells , Chloroquine/pharmacology , Colon/drug effects , Colon/pathology , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Humans , Immune Tolerance/drug effects , Ligands , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 9/chemistry
9.
J Immunother Cancer ; 9(5)2021 05.
Article in English | MEDLINE | ID: mdl-34021032

ABSTRACT

BACKGROUND: Current therapy for osteosarcoma pulmonary metastases (PMs) is ineffective. The mechanisms that prevent successful immunotherapy in osteosarcoma are incompletely understood. We investigated the tumor microenvironment of metastatic osteosarcoma with the goal of harnessing the immune system as a therapeutic strategy. METHODS: 66 osteosarcoma tissue specimens were analyzed by immunohistochemistry (IHC) and immune markers were digitally quantified. Tumor-infiltrating lymphocytes (TILs) from 25 specimens were profiled by functional cytometry. Comparative transcriptomic studies of distinct tumor-normal lung 'PM interface' and 'PM interior' regions from 16 PMs were performed. Clinical follow-up (median 24 months) was available from resection. RESULTS: IHC revealed a statistically significantly higher concentration of TILs expressing immune checkpoint and immunoregulatory molecules in PMs compared with primary bone tumors (including programmed cell death 1 (PD-1), programmed death ligand 1 (PD-L1), lymphocyte-activation gene 3 (LAG-3), T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), and indoleamine 2,3-dioxygenase (IDO1). Remarkably, these lymphocytes are excluded at the PM interface compared with PM interior. TILs from PMs exhibited significantly higher amounts of PD-1 and LAG-3 and functional cytokines including interferon-γ (IFNγ) by flow cytometry. Gene expression profiling further confirmed the presence of CD8 and CD4 lymphocytes concentrated at the PM interface, along with upregulation of immunoregulatory molecules and IFNγ-driven genes in the same region. We further discovered a strong alternatively activated macrophage signature throughout the entire PMs along with a polymorphonuclear myeloid-derived suppressor cell signature focused at the PM interface. Expression of PD-L1, LAG-3, and colony-stimulating factor 1 receptor (CSF1R) at the PM interface was associated with significantly worse progression-free survival (PFS), while gene sets indicative of productive T cell immune responses (CD8 T cells, T cell survival, and major histocompatibility complex class 1 expression) were associated with significantly improved PFS. CONCLUSIONS: Osteosarcoma PMs exhibit immune exclusion characterized by the accumulation of TILs at the PM interface. These TILs produce effector cytokines, suggesting their capability of activation and recognition of tumor antigens. Our findings suggest cooperative immunosuppressive mechanisms in osteosarcoma PMs including immune checkpoint molecule expression and the presence of immunosuppressive myeloid cells. We identify cellular and molecular signatures that are associated with patient outcomes, which could be exploited for successful immunotherapy.


Subject(s)
Biomarkers, Tumor/analysis , Bone Neoplasms/therapy , Cytokines/analysis , Immune Checkpoint Proteins/analysis , Immunotherapy , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Osteosarcoma/therapy , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Cytokines/genetics , Electronic Health Records , Humans , Immune Checkpoint Proteins/genetics , Immunotherapy/adverse effects , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphocyte Activation , Macrophage Activation , Myeloid-Derived Suppressor Cells , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/secondary , Progression-Free Survival , Retrospective Studies , Transcriptome
10.
Cancer Immunol Res ; 8(7): 895-911, 2020 07.
Article in English | MEDLINE | ID: mdl-32312711

ABSTRACT

The immunoglobulin-like domain containing receptor 2 (ILDR2), a type I transmembrane protein belonging to the B7 family of immunomodulatory receptors, has been described to induce an immunosuppressive effect on T-cell responses. Besides its expression in several nonlymphoid tissue types, we found that ILDR2 was also expressed in fibroblastic reticular cells (FRC) in the stromal part of the lymph node. These immunoregulatory cells were located in the T-cell zone and were essential for the recruitment of naïve T cells and activated dendritic cells to the lymph nodes. Previously, it has been shown that an ILDR2-Fc fusion protein exhibits immunomodulatory effects in several models of autoimmune diseases, such as multiple sclerosis, rheumatoid arthritis, and type I diabetes. Herein, we report the generation and characterization of a human/mouse/monkey cross-reactive anti-ILDR2 hIgG2 antibody, BAY 1905254, developed to block the immunosuppressive activity of ILDR2 for cancer immunotherapy. BAY 1905254 was shown to promote T-cell activation in vitro and enhance antigen-specific T-cell proliferation and cytotoxicity in vivo in mice. BAY 1905254 also showed potent efficacy in various syngeneic mouse cancer models, and the efficacy was found to correlate with increasing mutational load in the cancer models used. Additive or even synergistic antitumor effects were observed when BAY 1905254 was administered in combination with anti-PD-L1, an immunogenic cell death-inducing chemotherapeutic, or with tumor antigen immunization. Taken together, our data showed that BAY 1905254 is a potential drug candidate for cancer immunotherapy, supporting its further evaluation.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immunoglobulin G/pharmacology , Lymphocyte Activation/immunology , Membrane Proteins/immunology , Neoplasms/drug therapy , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immune Tolerance , Immunoglobulin G/immunology , Immunotherapy/methods , Leukocytes, Mononuclear/immunology , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism
11.
Clin Cancer Res ; 26(15): 4018-4030, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32332015

ABSTRACT

PURPOSE: Clinical trials with immune checkpoint inhibition in sarcomas have demonstrated minimal response. Here, we interrogated the tumor microenvironment (TME) of two contrasting soft-tissue sarcomas (STS), rhabdomyosarcomas and undifferentiated pleomorphic sarcomas (UPS), with differing genetic underpinnings and responses to immune checkpoint inhibition to understand the mechanisms that lead to response. EXPERIMENTAL DESIGN: Utilizing fresh and formalin-fixed, paraffin-embedded tissue from patients diagnosed with UPS and rhabdomyosarcomas, we dissected the TME by using IHC, flow cytometry, and comparative transcriptomic studies. RESULTS: Our results demonstrated both STS subtypes to be dominated by tumor-associated macrophages and infiltrated with immune cells that localized near the tumor vasculature. Both subtypes had similar T-cell densities, however, their in situ distribution diverged. UPS specimens demonstrated diffuse intratumoral infiltration of T cells, while rhabdomyosarcomas samples revealed intratumoral T cells that clustered with B cells near perivascular beds, forming tertiary lymphoid structures (TLS). T cells in UPS specimens were comprised of abundant CD8+ T cells exhibiting high PD-1 expression, which might represent the tumor reactive repertoire. In rhabdomyosarcomas, T cells were limited to TLS, but expressed immune checkpoints and immunomodulatory molecules which, if appropriately targeted, could help unleash T cells into the rest of the tumor tissue. CONCLUSIONS: Our work in STS revealed an immunosuppressive TME dominated by myeloid cells, which may be overcome with activation of T cells that traffic into the tumor. In rhabdomyosarcomas, targeting T cells found within TLS may be key to achieve antitumor response.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Neoplasms, Complex and Mixed/immunology , Rhabdomyosarcoma/immunology , Tertiary Lymphoid Structures/immunology , Tumor-Associated Macrophages/immunology , Adolescent , Adult , Aged , Aged, 80 and over , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Drug Resistance, Neoplasm/immunology , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Neoplasms, Complex and Mixed/drug therapy , Neoplasms, Complex and Mixed/genetics , Neoplasms, Complex and Mixed/pathology , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Tertiary Lymphoid Structures/pathology , Tumor Escape , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Young Adult
12.
J Pharmacol Exp Ther ; 328(2): 426-34, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19023040

ABSTRACT

Activation of the formyl-peptide receptor-like (FPRL) 1 pathway has recently gained high recognition for its significance in therapy of inflammatory diseases. Agonism at FPRL1 affords a beneficial effect in animal models of acute inflammatory conditions, as well as in chronic inflammatory diseases. TIPMFVPESTSKLQKFTSWFM-amide (CGEN-855A) is a novel 21-amino acid peptide agonist for FPRL1 and also activates FPRL2. CGEN-855A was discovered using a computational platform designed to predict novel G protein-coupled receptor peptide agonists cleaved from secreted proteins by convertase proteolysis. In vivo, CGEN-855A displays anti-inflammatory activity manifested as 50% inhibition of polymorphonuclear neutrophil (PMN) recruitment to inflamed air pouch and provides protection against ischemia-reperfusion-mediated injury to the myocardium in both murine and rat models (36 and 25% reduction in infarct size, respectively). Both these activities are accompanied by inhibition of PMN recruitment to the injured organ. The secretion of inflammatory cytokines, including interleukin (IL)-6, IL-1beta, and tumor necrosis factor-alpha, was not affected upon incubation of human peripheral blood mononuclear cells with CGEN-855A, whereas IL-8 secretion was elevated up to 2-fold upon treatment with the highest CGEN-855A dose only. Collectively, these new data support a potential role for CGEN-855A in the treatment of reperfusion-mediated injury and in other acute and chronic inflammatory conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/prevention & control , Myocardial Infarction/prevention & control , Peptides/therapeutic use , Receptors, Formyl Peptide/agonists , Receptors, Lipoxin/agonists , Animals , CHO Cells , Cricetinae , Cricetulus , Cytokines/metabolism , Disease Models, Animal , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Mice , Myocardial Infarction/pathology , Peptides/pharmacology , Rats
13.
PLoS One ; 8(3): e57757, 2013.
Article in English | MEDLINE | ID: mdl-23469229

ABSTRACT

CGEN-856S is a novel Mas agonist. Herein, we examined the effects of this peptide on isoproterenol (ISO)-induced cardiac remodeling and myocardial infarction (MI) injury. We also sought to determine whether CGEN-856S activates the underlying mechanisms related to Mas receptor activation. Heart hypertrophy and fibrosis were induced by ISO (2 mg·kg(-1)·day(-1)) in Wistar rats. After a 7-day treatment period with CGEN-856S (90 µg·kg(-1)·day(-1)) or vehicle, the cardiomyocyte diameter was evaluated in left ventricular sections stained with hematoxylin and eosin, and immunofluorescence labeling and quantitative confocal microscopy were used to quantify the deposition of type I and III collagen and fibronectin in the left ventricles. MI was induced by coronary artery ligation, and CGEN-856S (90 µg·kg(-1)·day(-1)) or saline was administered for 14 days. The Langendorff technique was used to evaluate cardiac function, and left ventricular sections were stained with Masson's trichrome dye to quantify the infarct area. Using Chinese hamster ovary cells stably transfected with Mas cDNA, we evaluated whether CGEN-856S alters AKT and endothelial nitric oxide synthase (eNOS) phosphorylation. CGEN-856S reduced the degree of ISO-induced hypertrophy (13.91±0.17 µm vs. 12.41±0.16 µm in the ISO+CGEN-856S group). In addition, the Mas agonist attenuated the ISO-induced increase in collagen I, collagen III, and fibronectin deposition. CGEN-856S markedly attenuated the MI-induced decrease in systolic tension, as well as in +dT/dt and -dT/dt. Furthermore, CGEN-856S administration significantly decreased the infarct area (23.68±2.78% vs. 13.95±4.37% in the MI+CGEN-856S group). These effects likely involved the participation of AKT and NO, as CGEN-856S administration increased the levels of p-AKT and p-eNOS. Thus, our results indicate that CGEN-856S exerts cardioprotective effects on ISO-induced cardiac remodeling and MI-mediated heart failure in rats through a mechanism likely involving the eNOS/AKT pathway.


Subject(s)
Cardiomegaly/drug therapy , Cardiotonic Agents/pharmacology , Heart/drug effects , Myocardial Infarction/drug therapy , Peptides/pharmacology , Proto-Oncogene Proteins/agonists , Receptors, G-Protein-Coupled/agonists , Ventricular Remodeling/drug effects , Animals , CHO Cells , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cardiotonic Agents/chemical synthesis , Collagen/biosynthesis , Cricetinae , Cricetulus , Fibronectins/biosynthesis , Gene Expression/drug effects , Heart/physiopathology , Isoproterenol , Male , Myocardial Infarction/chemically induced , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Peptides/chemical synthesis , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
14.
PLoS One ; 5(2): e9060, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20140206

ABSTRACT

BACKGROUND: Prostate cancer is a common and heterogeneous disease, where androgen receptor (AR) signaling plays a pivotal role in development and progression. The initial treatment for advanced prostate cancer is suppression of androgen signaling. Later on, essentially all patients develop an androgen independent stage which does not respond to anti hormonal treatment. Thus, alternative strategies targeting novel molecular mechanisms are required. beta-TrCP is an E3 ligase that targets various substrates essential for many aspects of tumorigenesis. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that beta-TrCP depletion suppresses prostate cancer and identify a relevant growth control mechanism. shRNA targeted against beta-TrCP reduced prostate cancer cell growth and cooperated with androgen ablation in vitro and in vivo. We found that beta-TrCP inhibition leads to upregulation of the aryl hydrocarbon receptor (AhR) mediating the therapeutic effect. This phenomenon could be ligand independent, as the AhR ligand 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD) did not alter prostate cancer cell growth. We detected high AhR expression and activation in basal cells and atrophic epithelial cells of human cancer bearing prostates. AhR expression and activation is also significantly higher in tumor cells compared to benign glandular epithelium. CONCLUSIONS/SIGNIFICANCE: Together these observations suggest that AhR activation may be a cancer counteracting mechanism in the prostate. We maintain that combining beta-TrCP inhibition with androgen ablation could benefit advanced prostate cancer patients.


Subject(s)
Cell Proliferation , Prostatic Neoplasms/genetics , Receptors, Aryl Hydrocarbon/genetics , beta-Transducin Repeat-Containing Proteins/genetics , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Doxycycline/pharmacology , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Ovariectomy , Polychlorinated Dibenzodioxins/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , Receptors, Aryl Hydrocarbon/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Up-Regulation/drug effects , beta-Transducin Repeat-Containing Proteins/metabolism
15.
Hypertension ; 56(1): 112-20, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20479330

ABSTRACT

Mas stimulation with angiotensin (Ang)-(1-7) produces cardioprotective effects and vasorelaxation. Using a computational discovery platform for predicting novel naturally occurring peptides that may activate G protein-coupled receptors, we discovered a novel Mas agonist peptide, CGEN-856S. An endothelium- and NO-dependent vasodilating effect was observed for CGEN-856S in thoracic aorta rings of rats (maximal value for the relaxant effect: 39.99+/-5.034%), which was similar to that produced by Ang-(1-7) (10(-10) to 10(-6) mol/L). In addition, the vasodilator activity of this peptide depended on a functional Mas receptor, because it was abolished in aorta rings of Mas-knockout mice. CGEN-856S appears to bind the Mas receptor at the same binding domain as Ang-(1-7), as suggested by the blocking of its vasorelaxant effect with the Ang-(1-7) analogue d-Ala(7)-Ang-(1-7), and by its competitive inhibition of Ang-(1-7) binding to Mas-transfected cells. The effect of CGEN-856S on reperfusion arrhythmias and cardiac function was studied on ischemia reperfusion of isolated rat hearts. We found that picomolar concentration of CGEN-856S (0.04 nmol/L) had an antiarrhythmogenic effect, as demonstrated by a reduction in the incidence and duration of reperfusion arrhythmias. Furthermore, acute infusion of CGEN-856S produced a shallow dose-dependent decrease in mean arterial pressure of conscious spontaneously hypertensive rats. The maximum change during infusion was observed at the highest dose. Strikingly, blood pressure continued to drop in the postinfusion period. The results presented here indicate that the novel Mas agonist, CGEN-856S, might have a therapeutic value, because it induces vasorelaxing, antihypertensive, and cardioprotective effects.


Subject(s)
Angiotensin I/pharmacology , Aorta/physiopathology , Arrhythmias, Cardiac/prevention & control , Hypertension/physiopathology , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Vasodilation/physiology , Animals , Antihypertensive Agents/pharmacology , Aorta/drug effects , Aorta/metabolism , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Disease Models, Animal , Heart/drug effects , Heart/physiopathology , Hypertension/metabolism , Hypertension/pathology , Male , Membrane Proteins , Mice , Mice, Knockout , Proto-Oncogene Mas , Proto-Oncogene Proteins/agonists , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL