Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(18)2023 Sep 06.
Article in English | MEDLINE | ID: mdl-37762046

ABSTRACT

Radiation therapy (RT) has recently demonstrated promise at stimulating an enhanced immune response. The recent success of immunotherapies, such as checkpoint inhibitors, CART cells, and other immune modulators, affords new opportunities for combination with radiation. The aim of this study is to evaluate whether and to what extent blockade of VISTA, an immune checkpoint, can potentiate the tumor control ability of radiation therapy. Our study is novel in that it is the first comparison of two VISTA-blocking methods (antibody inhibition and genetic knockout) in combination with RT. VISTA was blocked either through genetic knockout (KO) or an inhibitory antibody and combined with RT in two syngeneic murine flank tumor models (B16 and MC38). Selected mRNA, immune cell infiltration, and tumor growth delay were used to assess the biological effects. When combined with a single 15Gy radiation dose, VISTA blockade via genetic knockout in the B16 model and via anti-VISTA antibodies in the MC38 model significantly improved survival compared to RT alone by an average of 5.5 days and 6.3 days, respectively (p < 0.05). The gene expression data suggest that the mechanism behind the enhanced tumor control is primarily a result of increased apoptosis and immune-mediated cytotoxicity. VISTA blockade significantly enhances the anti-tumor effect of a single dose of 15Gy radiation through increased expression and stimulation of cell-mediated apoptosis pathways. These results suggest that VISTA is a biologically relevant immune promoter that has the potential to enhance the efficacy of a large single radiation dose in a synergic manner.


Subject(s)
Adenocarcinoma , Melanoma , Animals , Mice , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Antibodies , Disease Models, Animal , Melanoma/drug therapy , Melanoma/radiotherapy , T-Lymphocytes , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
2.
Cancer Immunol Res ; 11(1): 38-55, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36260656

ABSTRACT

V domain immunoglobulin suppressor of T-cell activation (VISTA) is a premier target for cancer treatment due to its broad expression in many cancer types and enhanced expression upon development of adaptive immune checkpoint resistance. In the CT26 colorectal cancer model, monotherapy of small tumors with anti-VISTA resulted in slowed tumor growth. In a combination therapy setting, large CT26 tumors showed complete adaptive resistance to anti-PD-1/CTLA-4, but inclusion of anti-VISTA led to rejection of half the tumors. Mechanisms of enhanced antitumor immunity were investigated using single-cell RNA sequencing (scRNA-seq), multiplex image analysis, and flow cytometry of the tumor immune infiltrate. In both treatment models, anti-VISTA upregulated stimulated antigen presentation pathways and reduced myeloid-mediated suppression. Imaging revealed an anti-VISTA stimulated increase in contacts between T cells and myeloid cells, further supporting the notion of increased antigen presentation. scRNA-seq of tumor-specific CD8+ T cells revealed that anti-VISTA therapy induced T-cell pathways highly distinct from and complementary to those induced by anti-PD-1 therapy. Whereas anti-CTLA-4/PD-1 expanded progenitor exhausted CD8+ T-cell subsets, anti-VISTA promoted costimulatory genes and reduced regulators of T-cell quiescence. Notably, this is the first report of a checkpoint regulator impacting CD8+ T-cell quiescence, and the first indication that quiescence may be a target in the context of T-cell exhaustion and in cancer. This study builds a foundation for all future studies on the role of anti-VISTA in the development of antitumor immunity and provides important mechanistic insights that strongly support use of anti-VISTA to overcome the adaptive resistance seen in contemporary treatments involving PD-1 and/or CTLA-4. See related Spotlight by Wei, p. 3.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , B7 Antigens/immunology , Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Immunoglobulins
3.
J Immunol ; 205(11): 3058-3070, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33087405

ABSTRACT

RIG-I-like receptors (RLR) are cytosolic RNA sensors that signal through the MAVS adaptor to activate IFN responses against viruses. Whether the RLR family has broader effects on host immunity against other pathogen families remains to be fully explored. In this study, we demonstrate that MDA5/MAVS signaling was essential for host resistance against pulmonary Aspergillus fumigatus challenge through the regulation of antifungal leukocyte responses in mice. Activation of MDA5/MAVS signaling was driven by dsRNA from live A. fumigatus serving as a key vitality-sensing pattern recognition receptor. Interestingly, induction of type I IFNs after A. fumigatus challenge was only partially dependent on MDA5/MAVS signaling, whereas type III IFN expression was entirely dependent on MDA5/MAVS signaling. Ultimately, type I and III IFN signaling drove the expression of CXCL10. Furthermore, the MDA5/MAVS-dependent IFN response was critical for the induction of optimal antifungal neutrophil killing of A. fumigatus spores. In conclusion, our data broaden the role of the RLR family to include a role in regulating antifungal immunity against A. fumigatus.


Subject(s)
Aspergillus fumigatus/immunology , Interferon-Induced Helicase, IFIH1/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Female , Interferons/immunology , Interferons/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction/immunology
4.
Front Immunol ; 10: 2641, 2019.
Article in English | MEDLINE | ID: mdl-31803182

ABSTRACT

The role of negative checkpoint regulators (NCRs) in human health and disease cannot be overstated. V-domain Ig-containing Suppressor of T-cell Activation (VISTA) is an Ig superfamily protein predominantly expressed within the hematopoietic compartment and has been studied for its role in the negative regulation of T cell responses. The findings presented in this study show that, unlike all other NCRs, VISTA deficiency dramatically impacts on macrophage cytokine and chemokine production, as well as the chemotactic response of VISTA-deficient macrophages. A select group of inflammatory chemokines, including CCL2, CCL3, CCL4, and CCL5, was strikingly elevated in culture supernatants from VISTA KO macrophages. VISTA deficiency also altered chemokine receptor recycling and profoundly disrupted myeloid chemotaxis. The impact of VISTA deficiency on chemotaxis in vivo was apparent with the reduced ability of both KO macrophages and MDSCs to migrate to the tumor microenvironment. This is the first demonstration of an NCR impacting on myeloid mediator production and chemotaxis, and will guide the use of anti-VISTA therapeutics to manipulate the chemotaxis of inflammatory macrophages or immunosuppressive MDSCs in inflammatory diseases and cancer.


Subject(s)
Chemokines/physiology , Chemotaxis/physiology , Macrophages/physiology , Membrane Proteins/physiology , Myeloid-Derived Suppressor Cells/physiology , Animals , Cell Line, Tumor , Female , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Tumor Microenvironment
5.
Semin Immunol ; 42: 101308, 2019 04.
Article in English | MEDLINE | ID: mdl-31604531

ABSTRACT

V-domain Ig suppressor of T cell activation (VISTA) is a novel checkpoint regulator with limited homology to other B7 family members. The constitutive expression of VISTA on both the myeloid and T lymphocyte lineages coupled to its important role in regulating innate and adaptive immune responses, qualifies VISTA to be a promising target for immunotherapeutic intervention. Studies have shown differential impact of agonistic and antagonistic targeting of VISTA, providing a unique landscape for influencing the outcome of cancer and inflammatory diseases.


Subject(s)
B7 Antigens/immunology , Neoplasms/immunology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Immunotherapy , Neoplasms/therapy
6.
Endocrinology ; 157(3): 1276-88, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26727108

ABSTRACT

Timely and appropriate levels of thyroid hormone (TH) signaling are necessary to ensure normal developmental outcomes in many tissues. Studies using pharmacological models of altered TH status have revealed an influence of these hormones on testis development and size, but little is known about the role of endogenous determinants of TH action in the developing male gonads. Using a genetic approach, we demonstrate that the type 3 deiodinase (D3), which inactivates TH and protects developing tissues from undue TH action, is a key factor. D3 is highly expressed in the developing testis, and D3-deficient (D3KO) mice exhibit thyrotoxicosis and cell proliferation arrest in the neonatal testis, resulting in an approximately 75% reduction in testis size. This is accompanied by larger seminiferous tubules, impaired spermatogenesis, and a hormonal profile indicative of primary hypogonadism. A deficiency in the TH receptor-α fully normalizes testis size and adult testis gene expression in D3KO mice, indicating that the effects of D3 deficiency are mediated through this type of receptor. Similarly, genetic deficiencies in the D2 or in the monocarboxylate transporter 8 partially rescue the abnormalities in testis size and gonadal axis gene expression featured in the D3KO mice. Our study highlights the testis as an important tissue in which determinants of TH action coordinately converge to ensure normal development and identifies D3 as a critical factor in testis development and in testicular protection from thyrotoxicosis.


Subject(s)
Hypogonadism/genetics , Iodide Peroxidase/genetics , RNA, Messenger/metabolism , Testis/metabolism , Thyrotoxicosis/genetics , Thyroxine/metabolism , Animals , Animals, Newborn , Immunohistochemistry , Iodide Peroxidase/metabolism , Male , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Monocarboxylic Acid Transporters , Reverse Transcriptase Polymerase Chain Reaction , Seminiferous Tubules/embryology , Seminiferous Tubules/metabolism , Seminiferous Tubules/pathology , Spermatogenesis/genetics , Symporters , Testis/embryology , Thyroid Hormone Receptors alpha/genetics , Transcriptome , Iodothyronine Deiodinase Type II
7.
Cancer Immunol Res ; 3(8): 956-67, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26048575

ABSTRACT

Despite immunogenicity, melanoma-specific vaccines have demonstrated minimal clinical efficacy in patients with established disease but enhanced survival when administered in the adjuvant setting. Therefore, we hypothesized that organs bearing metastatic-like melanoma may differentially produce T-cell chemotactic proteins over the course of tumor development. Using an established model of metastatic-like melanoma in lungs, we assessed the production of specific cytokines and chemokines over a time course of tumor growth, and we correlated chemokine production with chemokine receptor-specific T-cell infiltration. We observed that the interferon (IFN)-inducible CXCR3-cognate chemokines (CXCL9 and CXCL10) were significantly increased in lungs bearing minimal metastatic lesions, but chemokine production was at or below basal levels in lungs with substantial disease. Chemokine production was correlated with infiltration of the organ compartment by adoptively transferred CD8(+) tumor antigen-specific T cells in a CXCR3- and host IFNγ-dependent manner. Adenosine signaling in the tumor microenvironment (TME) suppressed chemokine production and T-cell infiltration in the advanced metastatic lesions, and this suppression could be partially reversed by administration of the adenosine receptor antagonist aminophylline. Collectively, our data demonstrate that CXCR3-cognate ligand expression is required for efficient T-cell access of tumor-infiltrated lungs, and these ligands are expressed in a temporally restricted pattern that is governed, in part, by adenosine. Therefore, pharmacologic modulation of adenosine activity in the TME could impart therapeutic efficacy to immunogenic but clinically ineffective vaccine platforms.


Subject(s)
Adenosine/metabolism , Chemokines/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/metabolism , Receptors, CXCR3/metabolism , T-Lymphocytes/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Disease Models, Animal , Humans , Ligands , Lung Neoplasms/secondary , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/pathology , Melanoma, Experimental , Mice, Knockout , Neoplasm Metastasis , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Burden
8.
Matrix Biol ; 48: 66-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25989506

ABSTRACT

Vertical growth phase (VGP) melanoma is frequently metastatic, a process mediated by changes in gene expression, which are directed by signal transduction pathways in the tumor cells. A prominent signaling pathway is the Ras-Raf-Mek-Erk MAPK pathway, which increases expression of genes that promote melanoma progression. Many melanomas harbor a mutation in this pathway, BRAF(V600E), which constitutively activates MAPK signaling and expression of downstream target genes that facilitate tumor progression. In BRAF(V600E) melanoma, the small molecule inhibitor, vemurafenib (PLX4032), has revolutionized therapy for melanoma by inducing rapid tumor regression. This compound down-regulates the expression of many genes. However, in this study, we document that blocking the Ras-Raf-Mek-Erk MAPK pathway, either with an ERK (PLX4032) or a MEK (U1026) signaling inhibitor, in BRAF(V600E) human and murine melanoma cell lines increases collagen synthesis in vitro and collagen deposition in vivo. Since TGFß signaling is a major mediator of collagen synthesis, we examined whether blocking TGFß signaling with a small molecule inhibitor would block this increase in collagen. However, there was minimal reduction in collagen synthesis in response to blocking TGFß signaling, suggesting additional mechanism(s), which may include activation of the p38 MAPK pathway. Presently, it is unclear whether this increased collagen synthesis and deposition in melanomas represent a therapeutic benefit or an unwanted "off target" effect of inhibiting the Ras-Raf-Erk-Mek pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Collagen Type I/agonists , Gene Expression Regulation, Neoplastic , Indoles/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Collagen Type I/genetics , Collagen Type I/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vemurafenib , Xenograft Model Antitumor Assays , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/genetics , ras Proteins/metabolism
9.
J Cell Physiol ; 228(4): 773-80, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23001823

ABSTRACT

We used vertical growth phase (VGP) human VMM5 melanoma cells to ask whether the tumor microenvironment could induce matrix metalloproteinase-1 (MMP-1) in vivo, and whether this induction correlated with metastasis. We isolated two clones from parental VMM5 cells: a low MMP-1 producing clone (C4) and high producing clone (C9). When these clones were injected orthotopically (intradermally) into nude mice, both were equally tumorigenic and produced equivalent and abundant amounts of MMP-1. However, the tumors from the C4 clones displayed different growth kinetics and distinct profiles of gene expression from the C9 population. The C4 tumors, which had low MMP-1 levels in vitro, appeared to rely on growth factors and cytokines in the microenvironment to increase MMP-1 expression in vivo, while MMP-1 levels remained constant in the C9 tumors. C9 cells, but not C4 cells, grew as spheres in culture and expressed higher levels of JARID 1B, a marker associated with melanoma initiating cells. We conclude that VMM5 melanoma cells exhibit striking intra-tumor heterogeneity, and that the tumorigenicity of these clones is driven by different molecular pathways. Our data suggest that there are multiple mechanisms for melanoma progression within a tumor, which may require different therapeutic strategies.


Subject(s)
Melanoma/pathology , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/pathology , Animals , Clone Cells , Cytokines/metabolism , Disease Progression , Female , Gene Expression , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Nude , Neoplasm Metastasis/genetics , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , Tumor Cells, Cultured , Tumor Microenvironment/genetics
10.
Proc Natl Acad Sci U S A ; 103(15): 6043-8, 2006 Apr 11.
Article in English | MEDLINE | ID: mdl-16595628

ABSTRACT

Thyroid hormone (TH) is critical for cardiac development and heart function. In heart disease, TH metabolism is abnormal, and many biochemical and functional alterations mirror hypothyroidism. Although TH therapy has been advocated for treating heart disease, a clear benefit of TH has yet to be established, possibly because of peripheral actions of TH. To assess the potential efficacy of TH in treating heart disease, type 2 deiodinase (D2), which converts the prohormone thyroxine to active triiodothyronine (T3), was expressed transiently in mouse hearts by using the tetracycline transactivator system. Increased cardiac D2 activity led to elevated cardiac T3 levels and to enhanced myocardial contractility, accompanied by increased Ca(2+) transients and sarcoplasmic reticulum (SR) Ca(2+) uptake. These phenotypic changes were associated with up-regulation of sarco(endo)plasmic reticulum calcium ATPase (SERCA) 2a expression as well as decreased Na(+)/Ca(2+) exchanger, beta-myosin heavy chain, and sarcolipin (SLN) expression. In pressure overload, targeted increases in D2 activity could not block hypertrophy but could completely prevent impaired contractility and SR Ca(2+) cycling as well as altered expression patterns of SERCA2a, SLN, and other markers of pathological hypertrophy. Our results establish that elevated D2 activity in the heart increases T3 levels and enhances cardiac contractile function while preventing deterioration of cardiac function and altered gene expression after pressure overload.


Subject(s)
Heart Diseases/physiopathology , Heart/physiology , Iodide Peroxidase/genetics , Myocardial Contraction/physiology , Thyroxine/physiology , Triiodothyronine/physiology , Animals , Blood Pressure/physiology , Calcium Signaling , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Gene Expression Regulation, Enzymologic , Genotype , Homeostasis , Iodide Peroxidase/metabolism , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Iodothyronine Deiodinase Type II
11.
Genomics ; 83(3): 413-24, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14962667

ABSTRACT

The human DIO3 gene and its mouse homolog, Dio3, map to chromosomes 14q32 and 12F1, respectively, and code for the type 3 deiodinase, an enzyme that inactivates thyroid hormones and is highly expressed during pregnancy and development. Mouse Dio3 is imprinted and preferentially expressed from the paternal allele in the fetus. We analyzed the human DIO3 genomic region and identified a gene (DIO3OS) that is transcribed in the antisense orientation. Multiple DIO3OS transcripts are expressed in most tissues. The structure of several DIO3OS cDNAs obtained by RT-PCR-based techniques reveals the occurrence of numerous splice variants. The exon-intron structures of DIO3OS are similar in mouse and human, but the homology of the exonic sequence is very low, except for the first exon, and no conserved open reading frame is present. We also detected DIO3 transcripts containing additional 5' untranslated sequence and a potential alternative upstream promoter for mouse Dio3. Exonic sequence of a Dio3os cDNA overlaps with the Dio3 promoter and strong promoter activity in the antisense orientation is detected in a genomic fragment located 3' of mouse and human DIO3 but not in the DIO3 promoter region. These results suggest that the DIO3 gene may lie within the structure of the antisense gene, a complex arrangement often observed in imprinted loci.


Subject(s)
Gene Expression , Genomic Imprinting , Iodide Peroxidase/genetics , RNA, Antisense/genetics , Alternative Splicing , Animals , Base Composition , Base Sequence , COS Cells , Chromosomes, Human, Pair 14 , Conserved Sequence , Humans , Mice , Molecular Sequence Data , Physical Chromosome Mapping , Promoter Regions, Genetic , Sequence Homology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...