Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Adv Funct Mater ; 31(37)2021 Sep 09.
Article in English | MEDLINE | ID: mdl-37745940

ABSTRACT

Solid tumors are protected from antitumor immune responses due to their hypoxic microenvironments. Weakening hypoxia-driven immunosuppression by hyperoxic breathing of 60% oxygen has shown to be effective in unleashing antitumor immune cells against solid tumors. However, efficacy of systemic oxygenation is limited against solid tumors outside of lungs and has been associated with unwanted side effects. As a result, it is essential to develop targeted oxygenation alternatives to weaken tumor hypoxia as novel approaches to restore immune responses against cancer. Herein, we report on injectable oxygen-generating cryogels (O2-cryogels) to reverse tumor-induced hypoxia. These macroporous biomaterials were designed to locally deliver oxygen, inhibit the expression of hypoxia-inducible genes in hypoxic melanoma cells, and reduce the accumulation of immunosuppressive extracellular adenosine. Our data show that O2-cryogels enhance T cell-mediated secretion of cytotoxic proteins, restoring the killing ability of tumor-specific CTLs, both in vitro and in vivo. In summary, O2-cryogels provide a unique and safe platform to supply oxygen as a co-adjuvant in hypoxic tumors and have the potential to improve cancer immunotherapies.

2.
J Clin Invest ; 130(11): 5629-5637, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32870821

ABSTRACT

Hypoxia/HIF-1α- and extracellular adenosine/A2 adenosine receptor-mediated immunosuppression protects tissues from collateral damage by antipathogen immune cells. However, this mechanism also protects cancerous tissues by inhibiting antitumor immune cells in hypoxic and extracellular adenosine-rich tumors that are the most resistant to current therapies. Here, we explain a potentially novel, antiimmunosuppressive reasoning to justify strategies using respiratory hyperoxia and oxygenation agents in cancer treatment. Earlier attempts to use oxygenation of tumors as a monotherapy or to improve radiotherapy have failed because oxygenation protocols were not combined with immunotherapies of cancer. In contrast, the proposal for therapeutic use of antihypoxic oxygenation described here was motivated by the need to prevent the hypoxia/HIF-1α-driven accumulation of extracellular adenosine to (a) unleash antitumor immune cells from inhibition by intracellular cAMP and (b) prevent immunosuppressive transcription of cAMP response element- and hypoxia response element-containing immunosuppressive gene products (e.g., TGF-ß). Use of oxygenation agents together with inhibitors of the A2A adenosine receptor may be required to enable the most effective cancer immunotherapy. The emerging outcomes of clinical trials of cancer patients refractory to all other treatments provide support for the molecular and immunological mechanism-based approach to cancer immunotherapy described here.


Subject(s)
Antineoplastic Agents/therapeutic use , Hyperoxia , Immunotherapy , Neoplasms , Oxygen/therapeutic use , Animals , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy
3.
Curr Opin Pharmacol ; 53: 98-100, 2020 08.
Article in English | MEDLINE | ID: mdl-32861959

ABSTRACT

The anti-hypoxia-A2-Adenosinergic immunotherapies of cancer emerged as the only available now approach to enable the tumor rejection in those progressing cancer patients that are refractory to all other current treatments. Several different classes of drugs are offered to inhibit the Hypoxia-HIF-1alpha-mediated and extracellular adenosine-A2A adenosine receptor-mediated immunosuppressive signaling in tumor microenvironment. It is suggested that the most promising treatments must include the blockade of cAMP-elevating A2A adenosine receptors and the elimination of hypoxia in tumors by oxygenation agents and hyperoxic breathing. The observations in ongoing clinical trials support this conclusion.


Subject(s)
Adenosine A2 Receptor Antagonists/therapeutic use , Adenosine/immunology , Immunotherapy , Neoplasms/therapy , Receptor, Adenosine A2A/immunology , T-Lymphocytes/immunology , Tumor Hypoxia/drug effects , Animals , Humans , Neoplasms/immunology
4.
Cancer Discov ; 10(1): 16-19, 2020 01.
Article in English | MEDLINE | ID: mdl-31919119

ABSTRACT

In this issue of Cancer Discovery, Fong and colleagues describe the encouraging observations of tumor regression, disease control, and survival of patients with otherwise refractory renal cell cancer with progressive disease after treatment with the conceptually novel oral antagonist of the A2A adenosine receptor (A2AR), ciforadenant. A2AR antagonists may represent the until now missing but critically important part of more effective immunotherapies of cancer, because they prevent the inhibition of tumor-reactive T and natural killer cells by blocking the immunosuppressive hypoxia-A2A-adenosinergic signaling, which represents an emerging immunosuppressive hallmark of tumors that are the most resistant to therapies.See related article by Fong et al., p. 40.


Subject(s)
Carcinoma, Renal Cell , Adenosine , Humans , Immunotherapy , Purinergic P1 Receptor Antagonists , Receptor, Adenosine A2A
5.
Cell Rep ; 21(13): 3672-3680, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29281817

ABSTRACT

Rapidly evolving pathogens such as HIV or influenza can quickly mutate their antigenic profiles, reducing the efficacy of conventional vaccines. Despite this challenge, functionally required epitopes are highly conserved among heterologous viral strains and represent a key vulnerability that could be targeted during vaccine development. As the antigenicity of these conserved epitopes is frequently subdominant, there is a critical need for innovative vaccination strategies designed to target these neutralizing epitopes. Here, we immunized mice with antigens containing discrete immunodominant and subdominant moieties and show that treatment with soluble heterologous antigen bearing only the immunodominant epitope selectively suppresses these germinal center (GC) B cells. By exploiting this intrinsic tolerance mechanism, we promote the expansion of subdominant B cells in the GC and the subsequent long-lived components of the humoral response. We propose that this strategy may be applied to elicit preferential expansion of subdominant B cells that recognize weakly immunogenic epitopes on microbial pathogens.


Subject(s)
B-Lymphocytes/metabolism , Germinal Center/metabolism , Immunodominant Epitopes/metabolism , Animals , Antibody Formation , Cell Count , Clone Cells , Mice, Inbred C57BL , Nitrophenols/chemistry , Ovalbumin/immunology , Phenylacetates/chemistry , Plasma Cells/metabolism , Solubility
6.
PLoS One ; 12(11): e0187314, 2017.
Article in English | MEDLINE | ID: mdl-29155844

ABSTRACT

Human cancers are known to downregulate Major Histocompatibility Complex (MHC) class I expression thereby escaping recognition and rejection by anti-tumor T cells. Here we report that oxygen tension in the tumor microenvironment (TME) serves as an extrinsic cue that regulates antigen presentation by MHC class I molecules. In support of this view, hypoxia is shown to negatively regulate MHC expression in a HIF-dependent manner as evidenced by (i) lower MHC expression in the hypoxic TME in vivo and in hypoxic 3-dimensional (3D) but not 2-dimensional (2D) tumor cell cultures in vitro; (ii) decreased MHC in human renal cell carcinomas with constitutive expression of HIF due to genetic loss of von Hippel-Lindau (VHL) function as compared with isogenically paired cells with restored VHL function, and iii) increased MHC in tumor cells with siRNA-mediated knockdown of HIF. In addition, hypoxia downregulated antigen presenting proteins like TAP 1/2 and LMP7 that are known to have a dominant role in surface display of peptide-MHC complexes. Corroborating oxygen-dependent regulation of MHC antigen presentation, hyperoxia (60% oxygen) transcriptionally upregulated MHC expression and increased levels of TAP2, LMP2 and 7. In conclusion, this study reveals a novel mechanism by which intra-tumoral hypoxia and HIF can potentiate immune escape. It also suggests the use of hyperoxia to improve tumor cell-based cancer vaccines and for mining novel immune epitopes. Furthermore, this study highlights the advantage of 3D cell cultures in reproducing hypoxia-dependent changes observed in the TME.


Subject(s)
Cell Hypoxia/immunology , Genes, MHC Class I/immunology , Hypoxia-Inducible Factor 1/genetics , Kidney Neoplasms/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 2/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1/immunology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Oxygen/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , T-Lymphocytes/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/immunology
7.
Cancer Res ; 76(11): 3319-31, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26988986

ABSTRACT

Proteasome-regulated NF-κB has been shown to be important for cell survival in T-cell lymphoma and Hodgkin lymphoma models. Several new small-molecule proteasome inhibitors are under various stages of active preclinical and clinical development. We completed a comprehensive preclinical examination of the efficacy and associated biologic effects of a second-generation proteasome inhibitor, ixazomib, in T-cell lymphoma and Hodgkin lymphoma cells and in vivo SCID mouse models. We demonstrated that ixazomib induced potent cell death in all cell lines at clinically achievable concentrations. In addition, it significantly inhibited tumor growth and improved survival in T-cell lymphoma and Hodgkin lymphoma human lymphoma xenograft models. Through global transcriptome analyses, proteasomal inhibition showed conserved overlap in downregulation of cell cycle, chromatin modification, and DNA repair processes in ixazomib-sensitive lymphoma cells. The predicted activity for tumor suppressors and oncogenes, the impact on "hallmarks of cancer," and the analysis of key significant genes from global transcriptome analysis for ixazomib strongly favored tumor inhibition via downregulation of MYC and CHK1, its target genes. Furthermore, in ixazomib-treated lymphoma cells, we identified that CHK1 was involved in the regulation of MYC expression through chromatin modification involving histone H3 acetylation via chromatin immunoprecipitation. Finally, using pharmacologic and RNA silencing of CHK1 or the associated MYC-related mechanism, we demonstrated synergistic cell death in combination with antiproteasome therapy. Altogether, ixazomib significantly downregulates MYC and induces potent cell death in T-cell lymphoma and Hodgkin lymphoma, and we identified that combinatorial therapy with anti-CHK1 treatment represents a rational and novel therapeutic approach. Cancer Res; 76(11); 3319-31. ©2016 AACR.


Subject(s)
Apoptosis/drug effects , Boron Compounds/pharmacology , Checkpoint Kinase 1/metabolism , Glycine/analogs & derivatives , Hodgkin Disease/pathology , Lymphoma, T-Cell/pathology , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation/drug effects , Checkpoint Kinase 1/genetics , Chromatin Immunoprecipitation , Gene Expression Profiling , Glycine/pharmacology , Hodgkin Disease/drug therapy , Hodgkin Disease/metabolism , Humans , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/metabolism , Mice , Mice, SCID , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Sci Transl Med ; 7(277): 277ra30, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25739764

ABSTRACT

Antitumor T cells either avoid or are inhibited in hypoxic and extracellular adenosine-rich tumor microenvironments (TMEs) by A2A adenosine receptors. This may limit further advances in cancer immunotherapy. There is a need for readily available and safe treatments that weaken the hypoxia-A2-adenosinergic immunosuppression in the TME. Recently, we reported that respiratory hyperoxia decreases intratumoral hypoxia and concentrations of extracellular adenosine. We show that it also reverses the hypoxia-adenosinergic immunosuppression in the TME. This, in turn, stimulates (i) enhanced intratumoral infiltration and reduced inhibition of endogenously developed or adoptively transfered tumor-reactive CD8 T cells, (ii) increased proinflammatory cytokines and decreased immunosuppressive molecules, such as transforming growth factor-ß (TGF-ß), (iii) weakened immunosuppression by regulatory T cells, and (iv) improved lung tumor regression and long-term survival in mice. Respiratory hyperoxia also promoted the regression of spontaneous metastasis from orthotopically grown breast tumors. These effects are entirely T cell- and natural killer cell-dependent, thereby justifying the testing of supplemental oxygen as an immunological coadjuvant to combine with existing immunotherapies for cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Oxygen/therapeutic use , Adenosine/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Hyperoxia/complications , Hyperoxia/pathology , Hypoxia/complications , Hypoxia/immunology , Hypoxia/pathology , Immunosuppression Therapy , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms/pathology , Oxygen/pharmacology , Remission Induction , Respiration/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects
9.
Cancer Immunol Res ; 2(7): 598-605, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24990240

ABSTRACT

Hypoxia-driven, A2A adenosine receptor (A2AR)-mediated (hypoxia-A2-adenosinergic), T-cell-autonomous immunosuppression was first recognized as critical and nonredundant in protecting normal tissues from inflammatory damage and autoimmunity. However, this immunosuppressive mechanism can be highjacked by bacteria and tumors to provide misguided protection for pathogens and cancerous tissues. Inhibitors of the hypoxia-A2-adenosinergic pathway represent a conceptually novel type of immunologic coadjuvants that could be combined with cancer vaccines, adoptive cell transfer, and/or blockade of negative immunologic regulators to further prolong patient survival and to minimize treatment-related side effects. In support of this approach are preclinical studies and findings that some human cancers are resistant to chemotherapies and immunotherapies due to the tumor-generated extracellular adenosine and A2AR on antitumor T and natural killer (NK) cells. Among the coadjuvants are (i) antagonists of A2AR, (ii) extracellular adenosine-degrading drugs, (iii) inhibitors of adenosine generation by CD39/CD73 ectoenzymes, and (iv) inhibitors of hypoxia-HIF-1α signaling. Combining these coadjuvants with CTLA-4 and/or PD-1 blockade is expected to have additive or even synergistic effects of targeting two different antitumor protective mechanisms. It is expected that even after multicombinatorial blockade of negative immunologic regulators, the antitumor T and NK cells would still be vulnerable to inhibition by hypoxia and A2AR. Yet to be tested is the potential capacity of coadjuvants to minimize the side effects of CTLA-4 and/or PD-1 blockade by decreasing the dose of blocking antibodies or by eliminating the need for dual blockade.


Subject(s)
Neoplasms/immunology , Receptor, Adenosine A2A/immunology , Cell Hypoxia/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immune Tolerance/immunology , Immunotherapy , Neoplasms/therapy , Signal Transduction/immunology , T-Lymphocytes/immunology
11.
Eur J Immunol ; 43(3): 655-66, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23208786

ABSTRACT

Hypoxia-adenosinergic suppression and redirection of the immune response has been implicated in the regulation of antipathogen and antitumor immunity, with hypoxia-inducible factor 1α (HIF-1α) playing a major role. In this study, we investigated the role of isoform I.1, a quantitatively minor alternative isoform of HIF-1α, in antibacterial immunity and sepsis survival. By using the cecal ligation and puncture model of bacterial peritonitis, we studied the function of I.1 isoform in T cells using mice with total I.1 isoform deficiency and mice with T-cell-targeted I.1 knockdown. We found that genetic deletion of the I.1 isoform resulted in enhanced resistance to septic lethality, significantly reduced bacterial load in peripheral blood, increased M1 macrophage polarization, augmented levels of proinflammatory cytokines in serum, and significantly decreased levels of the anti-inflammatory cytokine IL-10. Our data suggest a previously unrecognized immunosuppressive role for the I.1 isoform in T cells during bacterial sepsis. We interpret these data as indicative that the activation-inducible isoform I.1 hinders the contribution of T cells to the antibacterial response by affecting M1/M2 macrophage polarization and microbicidal function.


Subject(s)
Bacterial Infections/genetics , Bacterial Infections/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Peritonitis/genetics , Peritonitis/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Bacterial Infections/mortality , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Lymphocyte Activation/immunology , Macrophages/immunology , Male , Mice , Mice, Knockout , Organ Specificity/genetics , Peritonitis/mortality , Protein Isoforms , Receptors, Antigen, T-Cell/metabolism , Sepsis/genetics , Sepsis/immunology , Sepsis/mortality , Transcriptional Activation
13.
Transplantation ; 94(9): 894-902, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23073466

ABSTRACT

BACKGROUND: Liver ischemia-reperfusion injury (IRI) is a known risk factor for the postoperative outcome of patients undergoing liver surgery/transplantation. Attempts to protect from organ damage require multidisciplinary strategies and are of emerging interest in view of patients with higher age and American Society of Anesthesiology status. Ischemic preconditioning has been successfully applied to prevent from IRI during liver resection/transplantation. Because even short periods of ischemia during preconditioning inevitably lead to hypoxia and formation of anti-inflammatory/cytoprotective acting adenosine, we reasoned that short nonischemic hypoxia also protects against hepatic IRI. METHODS: Mice underwent hypoxic preconditioning (HPC) by breathing 10% oxygen for 10 min followed by 10 min of 21% oxygen before left liver lobe ischemia (45 min) and reperfusion (4 hr). The interactions of hypoxia→adenosine→adenosine receptors were tested by pharmacologic antagonism at adenosine receptor (AR) sites in wild-type mice and in mice with genetic deletions at the A1, A2A, A2B, and A3 ARs. Hepatocellular damage, inflammation, and metabolic effects were quantified by enzyme activities, cytokines, liver myeloperoxidase, blood adenosine, and tissue AMP, respectively. RESULTS: Hepatoprotection by HPC was significant in wild-type and A1, A2A, and A3 AR knockout mice as quantified by lower alanine aminotransferase serum activities, cytokine levels, histologic damage scores, tissue myeloperoxidase concentrations, and preserved AMP concentrations. Protection by HPC was blunted in mice pretreated with the A2B AR antagonist MRS1754 or in A2B AR knockout mice. CONCLUSIONS: Because liver protective effects of HPC are negated when the A2B receptor is nonfunctional, the hypoxia→adenosine→A2B receptor pathway plays a critical role in the prevention of warm IRI in vivo. Hypoxic activation of this pathway warrants use of selective A2B AR agonists or even intermittent hypoxia (e.g., in deceased organ donors) to protect from liver IRI.


Subject(s)
Hypoxia/physiopathology , Ischemic Preconditioning , Liver/blood supply , Receptor, Adenosine A2B/physiology , Reperfusion Injury/prevention & control , Warm Ischemia , Acetamides/pharmacology , Adenosine/physiology , Animals , Hepatocytes/pathology , Hepatocytes/physiology , Liver/pathology , Liver/physiopathology , Liver Transplantation/pathology , Liver Transplantation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Purines/pharmacology , Receptor, Adenosine A2B/deficiency , Receptor, Adenosine A2B/drug effects , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction/physiology
14.
J Biol Chem ; 285(50): 39271-88, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20926384

ABSTRACT

The A2A and A2B adenosine receptors (A2AR and A2BR) are implicated in many physiological processes. However, the mechanisms of their intracellular maturation and trafficking are poorly understood. In comparative studies of A2AR versus A2BR expression in transfected cells, we noticed that the levels of cell surface expression of A2BR were significantly lower than those of A2AR. A large portion of the A2BR was degraded by the proteasome. Studies of cell surface expression of A2BR chimeric molecules in transfectants suggested that A2BR does not have the dominant forward transport signal for export from the endoplasmic reticulum to the cell surface. A2BR surface expression was increased in A2BR chimeras where the A2BR carboxyl terminus (CT) was replaced or fused with the A2AR CT. Co-transfection of A2AR with A2BR enhanced surface expression of A2BR though the F(X)(6)LL motif in the A2AR CT. The requirements of A2AR expression for better A2BR cell surface expression was not only established in transfectants but also confirmed by observations of much lower levels of A2BR-induced intracellular cAMP accumulation in response to A2BR-activating ligand in splenocytes from A2AR(-/-) mice than in wild type mice. The results of mechanistic studies suggested that poor A2BR expression at the cell surface might be accounted for mainly by the lack of a dominant forward transport signal from the endoplasmic reticulum to the plasma membrane; it is likely that A2BR forms a hetero-oligomer complex for better function.


Subject(s)
Gene Expression Regulation , Receptor, Adenosine A2A/physiology , Receptor, Adenosine A2B/physiology , Animals , Biological Transport , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Receptor, Adenosine A2A/biosynthesis , Receptor, Adenosine A2B/biosynthesis , Signal Transduction
15.
Neoplasia ; 12(7): 539-49, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20651983

ABSTRACT

Toll-like receptors (TLRs) recognize microbial/viral-derived components that trigger innate immune response and conflicting data implicate TLR agonists in cancer, either as protumor or antitumor agents. We previously demonstrated that TLR3 activation mediated by its agonist poly(I:C) induces antitumor signaling, leading to apoptosis of prostate cancer cells LNCaP and PC3 with much more efficiency in the former than in the second more aggressive line. The transcription factor hypoxia-inducible factor 1 (HIF-1) regulates several cellular processes, including apoptosis, in response to hypoxia and to other stimuli also in normoxic conditions. Here we describe a novel protumor machinery triggered by TLR3 activation in PC3 cells consisting of increased expression of the specific I.3 isoform of HIF-1 alpha and nuclear accumulation of HIF-1 complex in normoxia, resulting in reduced apoptosis and in secretion of functional vascular endothelial growth factor (VEGF). Moreover, we report that, in the less aggressive LNCaP cells, TLR3 activation fails to induce nuclear accumulation of HIF-1 alpha. However, the transfection of I.3 isoform of hif-1 alpha in LNCaP cells allows poly(I:C)-induced HIF-1 activation, resulting in apoptosis protection and VEGF secretion. Altogether, our findings demonstrate that differences in the basal level of HIF-1 alpha expression in different prostate cancer cell lines underlie their differential response to TLR3 activation, suggesting a correlation between different stages of malignancy, hypoxic gene expression, and beneficial responsiveness to TLR agonists.


Subject(s)
Adenocarcinoma/genetics , Apoptosis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Neovascularization, Pathologic/genetics , Prostatic Neoplasms/genetics , Toll-Like Receptor 3/physiology , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Poly I-C/pharmacology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
J Immunol ; 184(1): 154-63, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19949104

ABSTRACT

Hypoxia-inducible factor (HIF)-1alpha plays a central role in oxygen homeostasis and energy supply by glycolysis in many cell types. We previously reported that an HIF-1alpha gene deficiency caused abnormal B cell development and autoimmunity. In this study we show that HIF-1alpha-enabled glycolysis during B cell development is required in a developmental stage-specific manner. Supporting this conclusion are observations that the glycolytic pathway in HIF-1alpha-deficient B220(+) bone marrow cells is much less functionally effective than in wild-type control cells. The expression of genes encoding the glucose transporters and the key glycolytic enzyme, 6-phosphofructo-2-kinase/fructose-2,6-bishosphatase 3, was greatly reduced in HIF-1alpha-deficient cells. The compensatory adaptation to the defect of glycolysis was reflected in higher levels of expression of respiratory chain-related genes and TCA cycle-related genes in HIF-1alpha-deficient cells than in wild-type cells. In agreement with these findings, HIF-1alpha-deficient cells used pyruvate more efficiently than wild-type cells. The key role of HIF-1alpha-enabled glycolysis in bone marrow B cells was also demonstrated by glucose deprivation during in vitro bone marrow cell culture and by using a glycolysis inhibitor in the bone marrow cell culture. Taken together, these findings indicate that glucose dependency differs at different B cell developmental stages and that HIF-1alpha plays an important role in B cell development.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation/physiology , Glycolysis/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Animals , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Flow Cytometry , Gene Expression , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
17.
J Leukoc Biol ; 86(3): 681-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19477908

ABSTRACT

Adenosine A(2A)R and TLR agonists synergize to induce an "angiogenic switch" in macrophages, down-regulating TNF-alpha and up-regulating VEGF expression. This switch involves transcriptional regulation of VEGF by HIF-1, transcriptional induction of HIF-1alpha by LPS (TLR4 agonist), and A(2A)R-dependent post-transcriptional regulation of HIF-1alpha stability. Murine HIF-1alpha is expressed as two mRNA isoforms: HIF-1alphaI.1 and -I.2, which contain alternative first exons and promoters. HIF-1alphaI.2 is expressed ubiquitously, and HIF-1alphaI.1 is tissue-specific. We investigated the regulation of these isoforms in macrophages by TLR4 and A(2A)R agonists. HIF-1alphaI.1 is induced strongly compared with HIF-1alphaI.2 upon costimulation with LPS and A(2A)R agonists (NECA or CGS21680). In unstimulated cells, the I.1 isoform constituted approximately 4% of HIF-1alpha transcripts; in LPS and NECA- or CGS21680-treated macrophages, this level was approximately 15%, indicating a substantial contribution of HIF-1alphaI.1 to total HIF-1alpha expression. The promoters of both isoforms were induced by LPS but not enhanced further by NECA, suggesting A(2A)R-mediated post-transcriptional regulation. LPS/NECA-induced expression of HIF-1alphaI.1 was down-regulated by Bay 11-7085 (NF-kappaB inhibitor) and ZM241385 (A(2A)R antagonist). Although VEGF and IL-10 expression by HIF-1alphaI.1-/- macrophages was equivalent to that of wild-type macrophages, TNF-alpha, MIP-1alpha, IL-6, IL-12p40, and IL-1beta expression was significantly greater, suggesting a role for HIF-1alphaI.1 in modulating expression of these cytokines. A(2A)R expression in unstimulated macrophages was low but was induced rapidly by LPS in a NF-kappaB-dependent manner. LPS-induced expression of A(2A)Rs and HIF-1alpha and A(2A)R-dependent HIF-1alpha mRNA and protein stabilization provide mechanisms for the synergistic effects of LPS and A(2A)R agonists on macrophage VEGF expression.


Subject(s)
Adenosine A2 Receptor Agonists , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages, Peritoneal/metabolism , Toll-Like Receptor 4/agonists , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Antagonists , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Cells, Cultured , Drug Combinations , Female , Genes, Reporter , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lipopolysaccharides/pharmacology , Luciferases, Renilla/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Mice, Congenic , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Phenethylamines/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Purinergic P1 Receptor Agonists , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sulfones/pharmacology , Toll-Like Receptor 4/metabolism , Transfection , Triazines/pharmacology , Triazoles/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
Trends Immunol ; 30(3): 102-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19201652

ABSTRACT

T regulatory cells (Treg cells) suppress immune responses to maintain self tolerance, but they also protect cancerous tissues. I propose a model to potentially unify the diverse functions of Treg cells. This assumes that Treg cells provide a complementary immunological arm to a physiological tissue-protecting mechanism, driven by low oxygen tension (i.e. hypoxia) in inflamed or cancerous tissues. The cAMP-elevating A2A and A2B adenosine receptors, hypoxia inducible transcription factor 1alpha (HIF), the cAMP response element (CRE)- and hypoxia response element (HRE)-mediated transcription in Treg and effector cells have key roles in this model. Both the T cell receptor (TCR)-triggered- and HRE- and CRE-driven activities of Treg cells are required to achieve a maximal level of immune suppression.


Subject(s)
Adenosine/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/metabolism , T-Lymphocytes, Regulatory/immunology , Adenosine/analogs & derivatives , Animals , Cell Hypoxia/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Receptor, Adenosine A2A/immunology , Receptor, Adenosine A2B/immunology , T-Lymphocytes, Regulatory/metabolism
19.
Clin Cancer Res ; 14(19): 5947-52, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829471

ABSTRACT

Cancerous tissue protection from tumor-recognizing CD8(+) and CD4(+) T cells (antitumor T cells) limits the therapeutic potential of immunotherapies. We propose that tumor protection is to a large extent due to (a) inhibition of antitumor T cells by hypoxia-driven accumulation of extracellular adenosine in local tumor microenvironment and due to (b) T regulatory cell-produced extracellular adenosine. The adenosine triggers the immunosuppressive signaling via intracellular cyclic AMP-elevating A2A adenosine receptors (A2AR) on antitumor T cells. In addition, the activated antitumor T cells in hypoxic tumor microenvironment could be inhibited by elevated levels of immunosuppressive hypoxia-inducible factor-1alpha. Complete rejection or tumor growth retardation was observed when A2AR has been genetically eliminated or antagonized with synthetic drug or with natural A2AR antagonist 1,3,7-trimethylxanthine (caffeine). The promising strategy may be in combining the anti-hypoxia-adenosinergic treatment that prevents inhibition of antitumor T cells by tumor-produced and T regulatory cell-produced adenosine with targeting of other negative regulators, such as CTL antigen-4 blockade. Observations of tumor rejection in mice and massive prospective epidemiologic studies support the feasibility of anti-hypoxia-adenosinergic combined immunotherapy.


Subject(s)
Hypoxia , Immunosuppressive Agents/pharmacology , Neoplasms/metabolism , Receptor, Adenosine A2A/metabolism , T-Lymphocytes, Regulatory/metabolism , Adenosine/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Caffeine/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunotherapy/methods , Mice , Models, Biological , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/metabolism
20.
Blood ; 111(12): 5424-5, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18544695
SELECTION OF CITATIONS
SEARCH DETAIL
...