Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Clin Invest ; 133(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38099496

ABSTRACT

Cell therapies such as tumor-infiltrating lymphocyte (TIL) therapy have shown promise in the treatment of patients with refractory solid tumors, with improvement in response rates and durability of responses nevertheless sought. To identify targets capable of enhancing the antitumor activity of T cell therapies, large-scale in vitro and in vivo clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screens were performed, with the SOCS1 gene identified as a top T cell-enhancing target. In murine CD8+ T cell-therapy models, SOCS1 served as a critical checkpoint in restraining the accumulation of central memory T cells in lymphoid organs as well as intermediate (Texint) and effector (Texeff) exhausted T cell subsets derived from progenitor exhausted T cells (Texprog) in tumors. A comprehensive CRISPR tiling screen of the SOCS1-coding region identified sgRNAs targeting the SH2 domain of SOCS1 as the most potent, with an sgRNA with minimal off-target cut sites used to manufacture KSQ-001, an engineered TIL therapy with SOCS1 inactivated by CRISPR/Cas9. KSQ-001 possessed increased responsiveness to cytokine signals and enhanced in vivo antitumor function in mouse models. These data demonstrate the use of CRISPR/Cas9 screens in the rational design of T cell therapies.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Humans , Animals , Mice , RNA, Guide, CRISPR-Cas Systems , Lymphocytes, Tumor-Infiltrating , Immunotherapy, Adoptive , Neoplasms/genetics , Gene Editing , Suppressor of Cytokine Signaling 1 Protein/genetics
2.
Mol Cancer Res ; 19(7): 1196-1207, 2021 07.
Article in English | MEDLINE | ID: mdl-33771882

ABSTRACT

Nuclear envelope proteins play an important role in regulating nuclear size and structure in cancer. Altered expression of nuclear lamins are found in many cancers and its expression is correlated with better clinical outcomes. The nucleus is the largest organelle in the cell with a diameter between 10 and 20 µm. Nuclear size significantly impacts cell migration. Nuclear structural changes are predicted to impact cancer metastasis by regulating cancer cell migration. Here we show emerin regulates nuclear structure in invasive breast cancer cells to impact cancer metastasis. Invasive breast cancer cells had 40% to 50% less emerin than control cells, which resulted in decreased nuclear size. Overexpression of GFP-emerin in invasive breast cancer cells rescued nuclear size and inhibited migration through 3.0 and 8.0 µm pores. Mutational analysis showed emerin binding to nucleoskeletal proteins was important for its regulation of nuclear structure, migration, and invasion. Importantly, emerin expression inhibited lung metastasis by 91% in orthotopic mouse models of breast cancer. Emerin nucleoskeleton-binding mutants failed to inhibit metastasis. These results support a model whereby emerin binding to the nucleoskeleton regulates nuclear structure to impact metastasis. In this model, emerin plays a central role in metastatic transformation, because decreased emerin expression during transformation causes the nuclear structural defects required for increased cell migration, intravasation, and extravasation. IMPLICATIONS: Modulating emerin expression and function represents new targets for therapeutic interventions of metastasis, because increased emerin expression rescued cancer metastasis.


Subject(s)
Breast Neoplasms/genetics , Cell Movement/genetics , Cell Nucleus/genetics , Membrane Proteins/genetics , Nuclear Matrix/genetics , Nuclear Proteins/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mice, Nude , Microscopy, Confocal/methods , Neoplasm Metastasis , Nuclear Matrix/metabolism , Nuclear Proteins/metabolism , Protein Binding , Transplantation, Heterologous
3.
Aging (Albany NY) ; 12(14): 15104-15120, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32633727

ABSTRACT

Ductal carcinoma in situ (DCIS) is one of the earliest stages of breast cancer (BCa). The mechanisms by which DCIS lesions progress to an invasive state while others remain indolent are yet to be fully characterized and both diagnosis and treatment of this pre-invasive disease could benefit from better understanding the pathways involved. While a decreased expression of Caveolin-1 (Cav-1) in the tumor microenvironment of patients with DCIS breast cancer was linked to progression to invasive breast cancer (IBC), the downstream effector(s) contributing to this process remain elusive. The current report shows elevated expression of Signal Transducer and Activator of Transcription 5a (STAT5a) within the DCIS-like lesions in Cav-1 KO mice following estrogen treatment and inhibition of STAT5a expression prevented the formation of these mammary lesions. In addition, STAT5a overexpression in a human DCIS cell line (MCF10DCIS.com) promoted their invasion, a process accelerated by estrogen treatment and associated with increased levels of the matrix metalloproteinase-9 (MMP-9) precursor. In sum, our results demonstrate a novel regulatory axis (Cav-1♦STAT5a♦MMP-9) in DCIS that is fully activated by the presence of estrogen. Our sudies suggest to further study phosphorylated STAT5a (Y694) as a potential biomarker to guide and predict outcome of DCIS patient population.


Subject(s)
Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Caveolin 1/metabolism , Estrogens , Neoplasm Invasiveness , STAT5 Transcription Factor/metabolism , Animals , Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Disease Progression , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/prevention & control , Phosphorylation , Tumor Microenvironment
4.
Cancer Biol Ther ; 21(6): 506-521, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32164483

ABSTRACT

Triple-negative breast cancer (TNBC) displays an aggressive clinical course, heightened metastatic potential, and is linked to poor survival rates. Through its lack of expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), this subtype remains unresponsive to traditional targeted therapies. Undesirable and sometimes life-threatening side effects associated with current chemotherapeutic agents warrant the development of more targeted treatment options. Targeting signal transducer and activator of transcription 3 (STAT3), a transcription factor implicated in breast cancer (BCa) progression, has proven to be an efficient approach to halt cancer growth in vitro and in vivo. Currently, there are no FDA-approved STAT3 inhibitors for TNBC. Although pimozide, a FDA-approved antipsychotic drug, has been attributed a role as a STAT3 inhibitor in several cancers, its role on this pathway remains unexplored in TNBC. As a "one size fits all" approach cannot be applied to TNBC therapies due to the heterogeneous nature of this aggressive cancer, we hypothesized that STAT3 could be a novel biomarker of response to guide pimozide therapy. Using human cell lines representative of four TNBC subtypes (basal-like 1, basal-like 2, mesenchymal-like, mesenchymal stem-like), our current report demonstrates that pimozide significantly reduced their invasion and migration, an effect that was predicted by STAT3 phosphorylation on tyrosine residue 705 (Tyr705). Mechanistically, phosphorylated STAT3 (Tyr705) inhibition resulting from pimozide treatment caused a downregulation of downstream transcriptional targets such as matrix metalloproteinase-9 (MMP-9) and vimentin, both implicated in invasion and migration. The identification of biomarkers of response to TNBC treatments is an active area of research in the field of precision medicine and our results propose phosphorylated STAT3 (Tyr705) as a novel biomarker to guide pimozide treatment as an inhibitor of invasion and migration.


Subject(s)
Biomarkers, Tumor/metabolism , Dopamine Antagonists/pharmacology , Pimozide/pharmacology , STAT3 Transcription Factor/metabolism , Triple Negative Breast Neoplasms/pathology , Apoptosis , Cell Movement , Cell Proliferation , Female , Humans , Phosphorylation , STAT3 Transcription Factor/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured
5.
Cell Cycle ; 19(4): 432-447, 2020 02.
Article in English | MEDLINE | ID: mdl-31931653

ABSTRACT

Triple negative breast cancer (TNBC) is a heterogeneous disease, which lacks expression of the estrogen receptor (ER), progesterone receptor (PR) and the human epidermal growth factor 2 receptor (HER2). This subtype of breast cancer has the poorest prognosis with limited therapies currently available, and hence additional options are needed. CAPER is a coactivator of the activator protein-1 (AP-1) (interacting specifically with the c-Jun component) and the ER and is known to be involved in human breast cancer pathogenesis. Recent published data have demonstrated a role for CAPER in TNBC and, as such, disrupting the function of CAPER with c-Jun could be a novel approach to treat TNBC patients. The data presented here shows the development and in vitro testing of CAPER-derived peptides that inhibit the coactivator activity of CAPER with c-Jun. These CAPER peptides result in a decrease in cell number and an increase in apoptosis in two TNBC cell lines, MDA-MB-231 and BT-549, while having no effect on the non-tumorigenic cell line MCF 10A. Additionally, two modes of action were demonstrated which appear to be cell line dependent: 1) a modulation of phosphorylated c-Jun leading to a decrease in Bcl-2 in MDA-MB-231 cells and a decrease in p21 in BT-549 cells and 2) a decrease in DNA repair proteins, leading to impaired DNA repair function in MDA-MB-231 cells. The data presented here supports further development of CAPER-derived peptides for the treatment of TNBC.


Subject(s)
Peptides/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Amino Acid Sequence , Apoptosis/drug effects , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Count , Cell Cycle/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Histones/metabolism , Humans , Neoplasm Proteins/metabolism , Peptides/chemistry , Peptides/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Recombinant Proteins/metabolism , Triple Negative Breast Neoplasms/pathology
6.
Science ; 367(6475)2020 01 17.
Article in English | MEDLINE | ID: mdl-31949051

ABSTRACT

Negative checkpoint regulators (NCRs) temper the T cell immune response to self-antigens and limit the development of autoimmunity. Unlike all other NCRs that are expressed on activated T lymphocytes, V-type immunoglobulin domain-containing suppressor of T cell activation (VISTA) is expressed on naïve T cells. We report an unexpected heterogeneity within the naïve T cell compartment in mice, where loss of VISTA disrupted the major quiescent naïve T cell subset and enhanced self-reactivity. Agonistic VISTA engagement increased T cell tolerance by promoting antigen-induced peripheral T cell deletion. Although a critical player in naïve T cell homeostasis, the ability of VISTA to restrain naïve T cell responses was lost under inflammatory conditions. VISTA is therefore a distinctive NCR of naïve T cells that is critical for steady-state maintenance of quiescence and peripheral tolerance.


Subject(s)
B7 Antigens/physiology , Membrane Proteins/physiology , Peripheral Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , B7 Antigens/genetics , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Tolerance/genetics , Receptors, Antigen, T-Cell/physiology
7.
Cancer Immunol Res ; 7(7): 1079-1090, 2019 07.
Article in English | MEDLINE | ID: mdl-31088847

ABSTRACT

Tumor hypoxia is a negative prognostic factor that is implicated in oncogenic signal activation, immune escape, and resistance to treatment. Identifying the mechanistic role of hypoxia in immune escape and resistance to immune-checkpoint inhibitors may aid the identification of therapeutic targets. We and others have shown that V-domain Ig suppressor of T-cell activation (VISTA), a negative checkpoint regulator in the B7 family, is highly expressed in the tumor microenvironment in tumor models and primary human cancers. In this study, we show that VISTA and HIF1α activity are correlated in a cohort of colorectal cancer patients. High VISTA expression was associated with worse overall survival. We used the CT26 colon cancer model to investigate the regulation of VISTA by hypoxia. Compared with less hypoxic tumor regions or draining lymph nodes, regions of profound hypoxia in the tumor microenvironment were associated with increased VISTA expression on tumor-infiltrating myeloid-derived suppressor cells (MDSC). Using chromatin immunoprecipitation and genetic silencing, we show that hypoxia-inducible factor (HIF)-1α binding to a conserved hypoxia response element in the VISTA promoter upregulated VISTA on myeloid cells. Further, antibody targeting or genetic ablation of VISTA under hypoxia relieved MDSC-mediated T-cell suppression, revealing VISTA as a mediator of MDSC function. Collectively, these data suggest that targeting VISTA may mitigate the deleterious effects of hypoxia on antitumor immunity.


Subject(s)
Adenocarcinoma/immunology , B7 Antigens/metabolism , Colorectal Neoplasms/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/physiopathology , Myeloid-Derived Suppressor Cells/immunology , Tumor Microenvironment/immunology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis , B7 Antigens/genetics , Case-Control Studies , Cell Proliferation , Cohort Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Prognosis , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured
8.
Oncotarget ; 9(54): 30340-30354, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30100993

ABSTRACT

Breast cancers (BCas) that lack expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) are referred to as triple negative breast cancers (TNBCs) and have the poorest clinical outcome. Once these aggressive tumors progress to distant organs, the median survival decreases to 12 months. With endocrine therapies being ineffective in this BCa subtype, highly toxic chemo- and radiation therapies are the only options. A better understanding of the functional role(s) of molecular targets contributing to TNBC progression could help in the design and development of new treatments that are more targeted with less toxicity. CAPER (Co-activator of AP-1 and ER) is a nuclear transcriptional co-activator that was recently involved in ER-positive BCa progression, however its role in hormone-independent cancers remains unknown. Our current report demonstrates that CAPER expression is upregulated in human TNBC specimens compared to normal breast tissue and that its selective downregulation through a lentiviral-mediated shRNA knockdown approach resulted in decreased cell numbers in MDA-MB-231 and BT549 TNBC cell lines without affecting the growth of non-tumorigenic cell line MCF-10A. Concordant with these observations, CAPER knockdown was also associated with a decrease in DNA repair proteins leading to a marked increase in apoptosis, through caspase-3/7 activation without any changes in cell cycle. Collectively, we propose CAPER as an important signaling molecule in the development of TNBC linked to DNA repair mechanisms, which could lead to new therapeutic modalities for the treatment of this aggressive cancer.

9.
Immunol Rev ; 276(1): 66-79, 2017 03.
Article in English | MEDLINE | ID: mdl-28258694

ABSTRACT

Utilization of negative checkpoint regulators (NCRs) for cancer immunotherapy has garnered significant interest with the completion of clinical trials demonstrating efficacy. While the results of monotherapy treatments are compelling, there is increasing emphasis on combination treatments in an effort to increase response rates to treatment. One of the most recently discovered NCRs is VISTA (V-domain Ig-containing Suppressor of T cell Activation). In this review, we describe the functions of this molecule in the context of cancer immunotherapy. We also discuss factors that may influence the use of anti-VISTA antibody in combination therapy and how genomic analysis may assist in providing indications for treatment.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7 Antigens/metabolism , Immunotherapy/methods , Neoplasms/therapy , Animals , B7 Antigens/genetics , B7 Antigens/immunology , Combined Modality Therapy , Genome , Humans , Lymphocyte Activation , Neoplasms/immunology
10.
J Immunother Cancer ; 4: 86, 2016.
Article in English | MEDLINE | ID: mdl-28031817

ABSTRACT

Negative checkpoint regulators function to restrain T cell responses to maintain tolerance and limit immunopathology. However, in the setting of malignancy, these pathways work in concert to promote immune-mediate escape leading to the development of a clinically overt cancer. In the recent years, clinical trials demonstrating the efficacy of blocking antibodies against these molecules have invigorated the field of immunotherapy. In this review, we discuss the current understanding on established NCR blockade and how strategic combination therapy with anti-VISTA antibody can be used to target multiple non-redundant NCR pathways.

11.
Front Immunol ; 6: 418, 2015.
Article in English | MEDLINE | ID: mdl-26347741

ABSTRACT

In the last two years, clinical trials with blocking antibodies to the negative checkpoint regulators CTLA-4 and PD-1 have rekindled the hope for cancer immunotherapy. Multiple negative checkpoint regulators protect the host against autoimmune reactions but also restrict the ability of T cells to effectively attack tumors. Releasing these brakes has emerged as an exciting strategy for cancer treatment. Conversely, these pathways can be manipulated to achieve durable tolerance for treatment of autoimmune diseases and transplantation. In the future, treatment may involve combination therapy to target multiple cell types and stages of the adaptive immune responses. In this review, we describe the current knowledge on the recently discovered negative checkpoint regulators, future targets for immunotherapy.

12.
Methods Mol Biol ; 1220: 461-86, 2015.
Article in English | MEDLINE | ID: mdl-25388268

ABSTRACT

Mast cells (MCs) play an important role in both inflammatory and immunosuppressive responses [1]. The importance of MCs in maintaining peripheral tolerance was discovered in a FoxP3(+) regulatory T-cell (Treg)-mediated skin transplant model [2]. MCs can directly mediate tolerance by releasing anti-inflammatory mediators (reviewed in ref. 3) or by interacting with other immune cells in the graft. Here we will present protocols used to study the role of MCs in peripheral tolerance with the emphasis on how MCs can regulate T-cell functionality. First we will introduce the skin transplant model followed by reconstitution of mast cell-deficient mice (B6.Cg-Kit (W-sh) ). This includes the preparation of MCs from the bone marrow. Finally the methods used to study the influence of MCs on T-cell responses and Treg functionality will be presented by modulating the balance between tolerance and inflammation.


Subject(s)
Mast Cells/immunology , Peripheral Tolerance , Skin Transplantation , Adoptive Transfer , Animals , Antibodies/immunology , Bone Marrow Cells/cytology , Cell Degranulation , Cytokines/metabolism , Graft Rejection , Mast Cells/cytology , Mice , Microscopy, Fluorescence , Models, Animal , Skin Transplantation/adverse effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tissue and Organ Harvesting
13.
Cell Tissue Res ; 358(3): 821-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25322709

ABSTRACT

Atherosclerosis is a complex disease initiated by the vascular accumulation of lipoproteins in the sub-endothelial space, followed by the infiltration of monocytes into the arterial intima. Caveolin-1 (Cav-1) plays an essential role in the regulation of cellular cholesterol metabolism and of various signaling pathways. In order to study specifically the role of macrophage Cav-1 in atherosclerosis, we used Cav-1 (-/-) Apoe (-/-) mice and transplanted them with bone marrow (BM) cells obtained from Cav-1 (+/+) Apoe (-/-) or Cav-1 (-/-) Apoe (-/-) mice and vice versa. We found that Cav-1 (+/+) mice harboring Cav-1 (-/-) BM-derived macrophages developed significantly larger lesions than Cav-1 (+/+) mice harboring Cav-1 (+/+) BM-derived macrophages. Cav-1 (-/-) macrophages were more susceptible to apoptosis and more prone to induce inflammation. The present study provides clear evidence that the absence of Cav-1 in macrophage is pro-atherogenic, whereas its absence in endothelial cells protects against atherosclerotic lesion formation. These findings demonstrate the cell-specific role of Cav-1 during the development of this disease.


Subject(s)
Atherosclerosis/pathology , Caveolin 1/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Animals , Apoptosis/drug effects , Atherosclerosis/blood , Bone Marrow Transplantation , Caveolin 1/deficiency , Cytokines/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Lipoproteins/blood , Macrophages, Peritoneal/drug effects , Mice, Inbred C57BL , Up-Regulation/drug effects
14.
Proc Natl Acad Sci U S A ; 111(41): 14846-51, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25267631

ABSTRACT

V domain-containing Ig suppressor of T-cell activation (VISTA) is a negative checkpoint regulator that suppresses T cell-mediated immune responses. Previous studies using a VISTA-neutralizing monoclonal antibody show that VISTA blockade enhances T-cell activation. The current study describes a comprehensive characterization of mice in which the gene for VISTA has been deleted. Despite the apparent normal hematopoietic development in young mice, VISTA genetic deficiency leads to a gradual accumulation of spontaneously activated T cells, accompanied by the production of a spectrum of inflammatory cytokines and chemokines. Enhanced T-cell responsiveness was also observed upon immunization with neoantigen. Despite the presence of multiorgan chronic inflammation, aged VISTA-deficient mice did not develop systemic or organ-specific autoimmune disease. Interbreeding of the VISTA-deficient mice with 2D2 T-cell receptor transgenic mice, which are predisposed to the development of experimental autoimmune encephalomyelitis, drastically enhanced disease incidence and intensity. Disease development is correlated with the increase in the activation of encephalitogenic T cells in the periphery and enhanced infiltration into the CNS. Taken together, our data suggest that VISTA is a negative checkpoint regulator whose loss of function lowers the threshold for T-cell activation, allowing for an enhanced proinflammatory phenotype and an increase in the frequency and intensity of autoimmunity under susceptible conditions.


Subject(s)
Autoimmunity/genetics , Autoimmunity/immunology , B7 Antigens/genetics , Genetic Predisposition to Disease , Inflammation/pathology , Aging/pathology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , B7 Antigens/deficiency , B7 Antigens/metabolism , Chemokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Hematopoiesis , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Th1 Cells/immunology , Th17 Cells/immunology
15.
Cancer Res ; 74(7): 1933-44, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24691994

ABSTRACT

V-domain Ig suppressor of T-cell activation (VISTA) is a novel negative checkpoint ligand that is homologous to PD-L1 and suppresses T-cell activation. This study demonstrates the multiple mechanisms whereby VISTA relieves negative regulation by hematopoietic cells and enhances protective antitumor immunity. VISTA is highly expressed on myeloid cells and Foxp3(+)CD4(+) regulatory cells, but not on tumor cells within the tumor microenvironment (TME). VISTA monoclonal antibody (mAb) treatment increased the number of tumor-specific T cells in the periphery and enhanced the infiltration, proliferation, and effector function of tumor-reactive T cells within the TME. VISTA blockade altered the suppressive feature of the TME by decreasing the presence of monocytic myeloid-derived suppressor cells and increasing the presence of activated dendritic cells within the tumor microenvironment. In addition, VISTA blockade impaired the suppressive function and reduced the emergence of tumor-specific Foxp3(+)CD4(+) regulatory T cells. Consequently, VISTA mAb administration as a monotherapy significantly suppressed the growth of both transplantable and inducible melanoma. Initial studies explored a combinatorial regimen using VISTA blockade and a peptide-based cancer vaccine with TLR agonists as adjuvants. VISTA blockade synergized with the vaccine to effectively impair the growth of established tumors. Our study therefore establishes a foundation for designing VISTA-targeted approaches either as a monotherapy or in combination with additional immune-targeted strategies for cancer immunotherapy.


Subject(s)
B7 Antigens/physiology , Immunoglobulins/physiology , Lymphocyte Activation , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/therapeutic use , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment
16.
Cell Cycle ; 13(8): 1256-64, 2014.
Article in English | MEDLINE | ID: mdl-24621503

ABSTRACT

CAPER is an estrogen receptor (ER) co-activator that was recently shown to be involved in human breast cancer pathogenesis. Indeed, we reported increased expression of CAPER in human breast cancer specimens. We demonstrated that CAPER was undetectable or expressed at relatively low levels in normal breast tissue and assumed a cytoplasmic distribution. In contrast, CAPER was expressed at higher levels in ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC) specimens, where it assumed a predominantly nuclear distribution. However, the functional role of CAPER in human breast cancer initiation and progression remained unknown. Here, we used a lentiviral-mediated gene silencing approach to reduce the expression of CAPER in the ER-positive human breast cancer cell line MCF-7. The proliferation and tumorigenicity of MCF-7 cells stably expressing control or human CAPER shRNAs was then determined via both in vitro and in vivo experiments. Knockdown of CAPER expression significantly reduced the proliferation of MCF-7 cells in vitro. Importantly, nude mice injected with MCF-7 cells harboring CAPER shRNAs developed smaller tumors than mice injected with MCF-7 cells harboring control shRNAs. Mechanistically, tumors derived from mice injected with MCF-7 cells harboring CAPER shRNAs displayed reduced expression of the cell cycle regulators PCNA, MCM7, and cyclin D1, and the protein synthesis marker 4EBP1. In conclusion, knockdown of CAPER expression markedly reduced human breast cancer cell proliferation in both in vitro and in vivo settings. Mechanistically, knockdown of CAPER abrogated the activity of proliferative and protein synthesis pathways.


Subject(s)
Breast Neoplasms/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Breast Neoplasms/pathology , Cell Cycle Checkpoints , Cell Proliferation , Disease Progression , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells , Mice, Nude , Neoplasm Transplantation , Nuclear Proteins/genetics , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Protein Biosynthesis , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics
17.
Breast Cancer Res ; 15(5): R87, 2013.
Article in English | MEDLINE | ID: mdl-24060386

ABSTRACT

INTRODUCTION: Previous studies have identified cholesterol as an important regulator of breast cancer development. High-density lipoprotein (HDL) and its cellular receptor, the scavenger receptor class B type I (SR-BI) have both been implicated in the regulation of cellular cholesterol homeostasis, but their functions in cancer remain to be established. METHODS: In the present study, we have examined the role of HDL and SR-BI in the regulation of cellular signaling pathways in breast cancer cell lines and in the development of tumor in a mouse xenograft model. RESULTS: Our data show that HDL is capable of stimulating migration and can activate signal transduction pathways in the two human breast cancer cell lines, MDA-MB-231 and MCF7. Furthermore, we also show that knockdown of the HDL receptor, SR-BI, attenuates HDL-induced activation of the phosphatidylinositol 3-kinase (PI3K)/protein Kinase B (Akt) pathway in both cell lines. Additional investigations show that inhibition of the PI3K pathway, but not that of the mitogen-activated protein kinase (MAPK) pathway, could lead to a reduction in cellular proliferation in the absence of SR-BI. Importantly, whereas the knockdown of SR-BI led to decreased proliferation and migration in vitro, it also led to a significant reduction in tumor growth in vivo. Most important, we also show that pharmacological inhibition of SR-BI can attenuate signaling and lead to decreased cellular proliferation in vitro. Taken together, our data indicate that both cholesteryl ester entry via HDL-SR-BI and Akt signaling play an essential role in the regulation of cellular proliferation and migration, and, eventually, tumor growth. CONCLUSIONS: These results identify SR-BI as a potential target for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , CD36 Antigens/metabolism , Cell Transformation, Neoplastic , Cholesterol/metabolism , Signal Transduction , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD36 Antigens/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Cholesterol/pharmacology , Cholesterol, HDL/metabolism , Cholesterol, HDL/pharmacology , Disease Models, Animal , Enzyme Activation/drug effects , Female , Gene Knockdown Techniques , Heterografts , Humans , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , MCF-7 Cells , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Tumor Burden/genetics
18.
Cancer Res ; 73(15): 4629-40, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23722543

ABSTRACT

Plasmacytoid dendritic cells (pDC) are key regulators of antiviral immunity. In previous studies, we reported that pDC-infiltrating human primary breast tumors represent an independent prognostic factor associated with poor outcome. To understand this negative impact of tumor-associated pDC (TApDC), we developed an orthotopic murine mammary tumor model that closely mimics the human pathology, including pDC and regulatory T cell (Treg) infiltration. We showed that TApDC are mostly immature and maintain their ability to internalize antigens in vivo and to activate CD4(+) T cells. Most importantly, TApDC were specifically altered for cytokine production in response to Toll-like receptor (TLR)-9 ligands in vitro while preserving unaltered response to TLR7 ligands (TLR7L). In vivo pDC depletion delayed tumor growth, showing that TApDC provide an immune-subversive environment, most likely through Treg activation, thus favoring tumor progression. However, in vivo intratumoral administration of TLR7L led to TApDC activation and displayed a potent curative effect. Depletion of pDC and type I IFN neutralization prevented TLR7L antitumoral effect. Our results establish a direct contribution of TApDC to primary breast tumor progression and rationalize the application of TLR7 ligands to restore TApDC activation in breast cancer. Cancer Res; 73(15); 4629-40. ©2013 AACR.


Subject(s)
Dendritic Cells/immunology , Lymphocyte Activation/immunology , Mammary Neoplasms, Experimental/immunology , Membrane Glycoproteins/immunology , Toll-Like Receptor 7/immunology , Animals , Dendritic Cells/metabolism , Disease Models, Animal , Female , Ligands , Lymphocyte Culture Test, Mixed , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mammary Neoplasms, Experimental/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 7/metabolism
19.
Cell Cycle ; 12(10): 1510-20, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23598719

ABSTRACT

Caveolin-1 (Cav-1) is a critical regulator of tumor progression in a variety of cancers where it has been shown to act as either a tumor suppressor or tumor promoter. In glioblastoma multiforme, it has been previously demonstrated to function as a putative tumor suppressor. Our studies here, using the human glioblastoma-derived cell line U-87MG, further support the role of Cav-1 as a negative regulator of tumor growth. Using a lentiviral transduction approach, we were able to stably overexpress Cav-1 in U-87MG cells. Gene expression microarray analyses demonstrated significant enrichment in gene signatures corresponding to downregulation of MAPK, PI3K/AKT and mTOR signaling, as well as activation of apoptotic pathways in Cav-1-overexpressing U-87MG cells. These same gene signatures were later confirmed at the protein level in vitro. To explore the ability of Cav-1 to regulate tumor growth in vivo, we further show that Cav-1-overexpressing U-87MG cells display reduced tumorigenicity in an ectopic xenograft mouse model, with marked hypoactivation of MAPK and PI3K/mTOR pathways. Finally, we demonstrate that Cav-1 overexpression confers sensitivity to the most commonly used chemotherapy for glioblastoma, temozolomide. In conclusion, Cav-1 negatively regulates key cell growth and survival pathways and may be an effective biomarker for predicting response to chemotherapy in glioblastoma.


Subject(s)
Apoptosis/drug effects , Caveolin 1/metabolism , Dacarbazine/analogs & derivatives , Animals , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caveolin 1/genetics , Cell Line, Tumor , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Down-Regulation , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Lentivirus/genetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Temozolomide , Transplantation, Heterologous
20.
J Exp Med ; 209(11): 2127-35, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23008335

ABSTRACT

Nutrient deprivation based on the loss of essential amino acids by catabolic enzymes in the microenvironment is a critical means to control inflammatory responses and immune tolerance. Here we report the novel finding that Tph-1 (tryptophan hydroxylase-1), a synthase which catalyses the conversion of tryptophan to serotonin and exhausts tryptophan, is a potent regulator of immunity. In models of skin allograft tolerance, tumor growth, and experimental autoimmune encephalomyelitis, Tph-1 deficiency breaks allograft tolerance, induces tumor remission, and intensifies neuroinflammation, respectively. All of these effects of Tph-1 deficiency are independent of its downstream product serotonin. Because mast cells (MCs) appear to be the major source of Tph-1 and restoration of Tph-1 in the MC compartment in vivo compensates for the defect, these experiments introduce a fundamentally new mechanism of MC-mediated immune suppression that broadly impacts multiple arms of immunity.


Subject(s)
Immune Tolerance/immunology , Inflammation/immunology , Mast Cells/immunology , Tryptophan Hydroxylase/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Flow Cytometry , Gene Expression , Immune Tolerance/genetics , Inflammation/genetics , Male , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Reverse Transcriptase Polymerase Chain Reaction , Skin Transplantation/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Transplantation, Homologous , Tryptophan/blood , Tryptophan/metabolism , Tryptophan Hydroxylase/deficiency , Tryptophan Hydroxylase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...