Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 1150, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35241644

ABSTRACT

Huntington's Disease (HD) is a progressive neurodegenerative disorder caused by CAG trinucleotide repeat expansions in exon 1 of the huntingtin (HTT) gene. The mutant HTT (mHTT) protein causes neuronal dysfunction, causing progressive motor, cognitive and behavioral abnormalities. Current treatments for HD only alleviate symptoms, but cerebral spinal fluid (CSF) or central nervous system (CNS) delivery of antisense oligonucleotides (ASOs) or virus vectors expressing RNA-induced silencing (RNAi) moieties designed to induce mHTT mRNA lowering have progressed to clinical trials. Here, we present an alternative disease modifying therapy the orally available, brain penetrant small molecule branaplam. By promoting inclusion of a pseudoexon in the primary transcript, branaplam lowers mHTT protein levels in HD patient cells, in an HD mouse model and in blood samples from Spinal Muscular Atrophy (SMA) Type I patients dosed orally for SMA (NCT02268552). Our work paves the way for evaluating branaplam's utility as an  HD therapy, leveraging small molecule splicing modulators to reduce expression of dominant disease genes by driving pseudoexon inclusion.


Subject(s)
Huntington Disease , Animals , Brain/metabolism , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Oligonucleotides, Antisense/metabolism , Trinucleotide Repeat Expansion
3.
Nature ; 596(7871): 291-295, 2021 08.
Article in English | MEDLINE | ID: mdl-34321659

ABSTRACT

So far, gene therapies have relied on complex constructs that cannot be finely controlled1,2. Here we report a universal switch element that enables precise control of gene replacement or gene editing after exposure to a small molecule. The small-molecule inducers are currently in human use, are orally bioavailable when given to animals or humans and can reach both peripheral tissues and the brain. Moreover, the switch system, which we denote Xon, does not require the co-expression of any regulatory proteins. Using Xon, the translation of the desired elements for controlled gene replacement or gene editing machinery occurs after a single oral dose of the inducer, and the robustness of expression can be controlled by the drug dose, protein stability and redosing. The ability of Xon to provide temporal control of protein expression can be adapted for cell-biology applications and animal studies. Additionally, owing to the oral bioavailability and safety of the drugs used, the Xon switch system provides an unprecedented opportunity to refine and tailor the application of gene therapies in humans.


Subject(s)
Alternative Splicing/drug effects , Gene Editing/methods , Genetic Therapy/methods , Protein Biosynthesis/drug effects , Animals , Brain/drug effects , Brain/metabolism , CRISPR-Associated Protein 9/metabolism , Drug Delivery Systems/methods , Erythropoietin/biosynthesis , Erythropoietin/genetics , Erythropoietin/metabolism , Exons/genetics , Female , Frontotemporal Dementia/metabolism , HEK293 Cells , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscular Atrophy, Spinal/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Progranulins/biosynthesis , Progranulins/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/metabolism
5.
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
6.
J Mol Med (Berl) ; 92(12): 1283-92, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25120128

ABSTRACT

UNLABELLED: Intratumoral hypoxia and hypoxia inducible factor-1α (HIF-1-α)-dependent CD39/CD73 ectoenzymes may govern the accumulation of tumor-protecting extracellular adenosine and signaling through A2A adenosine receptors (A2AR) in tumor microenvironments (TME). Here, we explored the conceptually novel motivation to use supplemental oxygen as a treatment to inhibit the hypoxia/HIF-1α-CD39/CD73-driven accumulation of extracellular adenosine in the TME in order to weaken the tumor protection. We report that hyperoxic breathing (60 % O2) decreased the TME hypoxia, as well as levels of HIF-1α and downstream target proteins of HIF-1α in the TME according to proteomic studies in mice. Importantly, oxygenation also downregulated the expression of adenosine-generating ectoenzymes and significantly lowered levels of tumor-protecting extracellular adenosine in the TME. Using supplemental oxygen as a tool in studies of the TME, we also identified FHL-1 as a potentially useful marker for the conversion of hypoxic into normoxic TME. Hyperoxic breathing resulted in the upregulation of antigen-presenting MHC class I molecules on tumor cells and in the better recognition and increased susceptibility to killing by tumor-reactive cytotoxic T cells. Therapeutic breathing of 60 % oxygen resulted in the significant inhibition of growth of established B16.F10 melanoma tumors and prolonged survival of mice. Taken together, the data presented here provide proof-of principle for the therapeutic potential of systemic oxygenation to convert the hypoxic, adenosine-rich and tumor-protecting TME into a normoxic and extracellular adenosine-poor TME that, in turn, may facilitate tumor regression. We propose to explore the combination of supplemental oxygen with existing immunotherapies of cancer. KEY MESSAGES: Oxygenation decreases levels of tumor protecting hypoxia. Oxygenation decreases levels of tumor protecting extracellular adenosine. Oxygenation decreases expression of HIF-1alpha dependent tumor-protecting proteins. Oxygenation increases MHC class I expression and enables tumor regression.


Subject(s)
Adenosine/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/complications , Hypoxia/therapy , Neoplasms/complications , Neoplasms/therapy , Oxygen/therapeutic use , Animals , Cell Hypoxia , Cell Line, Tumor , Female , Hypoxia/metabolism , Mice, Inbred C57BL , Neoplasms/metabolism , Tumor Microenvironment
7.
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
8.
Expert Opin Biol Ther ; 14(4): 419-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24397825

ABSTRACT

INTRODUCTION: Sepsis is an extremely fast-paced disease, initiated by an infection that can progress to multiple organ dysfunction and death. The complexity associated with sepsis makes the therapies difficult to develop. Moreover, the 'one-fits-all' kind of therapy is far from being realistic. AREAS COVERED: This review provides a conspectus of the current results of sepsis therapies and their benefits, focusing on the development of small interfering RNA (siRNA) therapeutics for targeting immune cells and sepsis pathways. EXPERT OPINION: The question, 'When will an effective therapy for sepsis be available for patients?' remains unanswered. New RNA interference-mediated therapies are emerging as novel approaches for the treatment of sepsis by downregulating key inflammatory cytokine expression. Strategies that exploit multimodal gene silencing using siRNA and targeted delivery systems are discussed in this review. Some of these strategies have shown positive results in preclinical model of sepsis.


Subject(s)
RNA Interference , Sepsis/drug therapy , Animals , Cytokines/genetics , Cytokines/physiology , Drug Delivery Systems , Gene Silencing , Humans , Inflammation/genetics , Inflammation/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , Sepsis/physiopathology
9.
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
10.
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
11.
J Clin Invest ; 122(2): 693-710, 2012 02.
Article in English | MEDLINE | ID: mdl-22269324

ABSTRACT

A complex biologic network regulates kidney perfusion under physiologic conditions. This system is profoundly perturbed following renal ischemia, a leading cause of acute kidney injury (AKI) - a life-threatening condition that frequently complicates the care of hospitalized patients. Therapeutic approaches to prevent and treat AKI are extremely limited. Better understanding of the molecular pathways promoting postischemic reflow could provide new candidate targets for AKI therapeutics. Due to its role in adapting tissues to hypoxia, we hypothesized that extracellular adenosine has a regulatory function in the postischemic control of renal perfusion. Consistent with the notion that equilibrative nucleoside transporters (ENTs) terminate adenosine signaling, we observed that pharmacologic ENT inhibition in mice elevated renal adenosine levels and dampened AKI. Deletion of the ENTs resulted in selective protection in Ent1-/- mice. Comprehensive examination of adenosine receptor-knockout mice exposed to AKI demonstrated that renal protection by ENT inhibitors involves the A2B adenosine receptor. Indeed, crosstalk between renal Ent1 and Adora2b expressed on vascular endothelia effectively prevented a postischemic no-reflow phenomenon. These studies identify ENT1 and adenosine receptors as key to the process of reestablishing renal perfusion following ischemic AKI. If translatable from mice to humans, these data have important therapeutic implications.


Subject(s)
Acute Kidney Injury/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Ischemia/metabolism , Regional Blood Flow/physiology , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Adenosine/metabolism , Animals , Cell Line , Chimerism , Dipyridamole/therapeutic use , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Equilibrative Nucleoside Transporter 1/genetics , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , No-Reflow Phenomenon , Nucleoside Transport Proteins/antagonists & inhibitors , Nucleoside Transport Proteins/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism
12.
J Immunol ; 186(4): 2444-53, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242513

ABSTRACT

Antimicrobial treatment strategies must improve to reduce the high mortality rates in septic patients. In noninfectious models of acute inflammation, activation of A2B adenosine receptors (A2BR) in extracellular adenosine-rich microenvironments causes immunosuppression. We examined A2BR in antibacterial responses in the cecal ligation and puncture (CLP) model of sepsis. Antagonism of A2BR significantly increased survival, enhanced bacterial phagocytosis, and decreased IL-6 and MIP-2 (a CXC chemokine) levels after CLP in outbred (ICR/CD-1) mice. During the CLP-induced septic response in A2BR knockout mice, hemodynamic parameters were improved compared with wild-type mice in addition to better survival and decreased plasma IL-6 levels. A2BR deficiency resulted in a dramatic 4-log reduction in peritoneal bacteria. The mechanism of these improvements was due to enhanced macrophage phagocytic activity without augmenting neutrophil phagocytosis of bacteria. Following ex vivo LPS stimulation, septic macrophages from A2BR knockout mice had increased IL-6 and TNF-α secretion compared with wild-type mice. A therapeutic intervention with A2BR blockade was studied by using a plasma biomarker to direct therapy to those mice predicted to die. Pharmacological blockade of A2BR even 32 h after the onset of sepsis increased survival by 65% in those mice predicted to die. Thus, even the late treatment with an A2BR antagonist significantly improved survival of mice (ICR/CD-1) that were otherwise determined to die according to plasma IL-6 levels. Our findings of enhanced bacterial clearance and host survival suggest that antagonism of A2BRs offers a therapeutic target to improve macrophage function in a late treatment protocol that improves sepsis survival.


Subject(s)
Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/metabolism , Macrophages/immunology , Phagocytosis/immunology , Receptor, Adenosine A2B/metabolism , Sepsis/immunology , Up-Regulation/immunology , Animals , Antigens, CD1/biosynthesis , Cecum , Female , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Ligation , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Phagocytosis/genetics , Punctures , Receptor, Adenosine A2B/deficiency , Receptor, Adenosine A2B/genetics , Sepsis/genetics , Sepsis/mortality , Survival Rate , Up-Regulation/genetics
13.
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
14.
J Leukoc Biol ; 86(3): 545-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564571

ABSTRACT

Here, we attract attention to the possibility of iatrogenic exacerbation of immune-mediated tissue damage as a result of the unintended weakening of the tissue-protecting, hypoxia-adenosinergic pathway. These immunosuppressive, anti-inflammatory pathways play a critical and nonredundant role in the protection of normal tissues from collateral damage during an inflammatory response. We believe that it is the tissue hypoxia associated with inflammatory damage that leads to local inhibition of overactive immune cells by activating A2AR and A2BR and stabilizing HIF-1alpha. We show in an animal model of acute lung injury that oxygenation (i.e., inspiring supplemental oxygen) reverses tissue hypoxia and exacerbates ongoing inflammatory lung tissue damage. However, little has been done to carefully investigate and prevent this in a clinical setting. Similarly, the consumption of caffeine antagonizes A2ARs, resulting in exacerbation of ongoing acute inflammation. It is suggested that although the elimination of hypoxia-adenosinergic immunosuppression is desirable to improve vaccines, it is important to take into account the unintentional effects of supplemental oxygen and caffeine, which may increase collateral, inflammatory tissue damage.


Subject(s)
Acute Lung Injury/etiology , Hypoxia/physiopathology , Inflammation/pathology , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/metabolism , Adenosine/metabolism , Adenosine A2 Receptor Antagonists , Animals , Caffeine/pharmacology , Disease Models, Animal , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/immunology , Mice , Mice, Mutant Strains , Oxygen/toxicity , Oxygen Consumption , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/physiopathology , T-Lymphocytes, Regulatory/immunology
15.
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
16.
Hum Immunol ; 69(7): 421-5, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18638657

ABSTRACT

Hypoxia-inducible factor-1alpha (HIF-1alpha) is critical not only in the regulation of oxygen homeostasis but also in the regulation of innate and adaptive immune systems. We previously reported that T-cell receptor-mediated activation of T cells in mice leads to the expression of an alternative isoform of HIF-1alpha that inhibits activated T cells in a delayed negative feed-back manner. In this paper, we describe a novel mRNA isoform of human HIF-1alpha that is upregulated in peripheral T lymphocytes after T-cell receptor stimulation. This activation- inducible isoform is expressed using the alternative first exon I.3, and it encodes a protein that is 24 amino acids longer than the ubiquitous HIF-1alpha isoform. This mRNA isoform I.3 of HIF-1alpha is expressed in a tissue-specific manner with the highest expression found in peripheral blood leukocytes and the thymus.


Subject(s)
Alternative Splicing , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Lymphocyte Activation , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Exons , Gene Expression , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Molecular Sequence Data , Organ Specificity , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/analysis , T-Lymphocytes/immunology , Up-Regulation
17.
Mol Med ; 14(3-4): 116-23, 2008.
Article in English | MEDLINE | ID: mdl-18163162

ABSTRACT

Whole body exposure of wild type control littermates and A2A adenosine receptor (A2AR) gene deleted mice to low oxygen containing inspired gas mixture allowed the investigation of the mechanism that controls inflammatory liver damage and protects the liver using a mouse model of T cell-mediated viral and autoimmune hepatitis. We tested the hypothesis that the inflammatory tissue damage-associated hypoxia and extracellular adenosine --> A2AR signaling plays an important role in the physiological anti-inflammatory mechanism that limits liver damage during fulminant hepatitis. After induction of T cell-mediated hepatitis, mice were kept in modular chambers either under normoxic (21% oxygen) or hypoxic (10% oxygen) conditions for 8 h. It was shown that the whole body exposure to hypoxic atmosphere caused tissue hypoxia in healthy animals as evidenced by a decrease in the arterial blood oxygen tension and increase of the plasma adenosine concentration (P < 0.05). This "hypoxic" treatment resulted in significantly reduced hepatocellular damage and attenuated levels of serum cytokines in mice with acute liver inflammation. The anti-inflammatory effects of hypoxia were not observed in the absence of A2AR in studies of A2AR gene-deficient mice or when A2AR have been pharmacologically antagonized with synthetic antagonist. The presented data demonstrate that total body hypoxia-triggered pathway provides protection in acute hepatitis and that hypoxia (upstream) and A2AR (downstream) function in the same immunosuppressive and liver tissue-protecting pathway.


Subject(s)
Hepatitis , Hypoxia , Inflammation/metabolism , Liver , Receptor, Adenosine A2A/metabolism , Signal Transduction/physiology , Adenosine/metabolism , Adenosine A2 Receptor Antagonists , Animals , Concanavalin A/metabolism , Cytokines/blood , Hepatitis/metabolism , Hepatitis/pathology , Hepatitis/virology , Humans , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen/blood , Purines/blood , Receptor, Adenosine A2A/genetics , T-Lymphocytes/immunology , Triazines/metabolism , Triazoles/metabolism
18.
Diabetes ; 57(2): 484-93, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18003758

ABSTRACT

OBJECTIVES: Non-obese diabetic (NOD) mice develop spontaneous type 1 diabetes. We have shown that sphingosine-1-phosphate (S1P) reduces activation of NOD diabetic endothelium via the S1P1 receptor. In the current study, we tested the hypothesis that S1P could inhibit CD4(+) T-cell activation, further reducing inflammatory events associated with diabetes. RESEARCH DESIGN AND METHODS: CD4(+) T-cells were isolated from diabetic and nondiabetic NOD mouse splenocytes and treated in the absence or presence of S1P or the S1P1 receptor-specific agonist, SEW2871. Lymphocyte activation was examined using flow cytometry, cytokine bead assays, and a lymphocyte:endothelial adhesion assay. RESULTS: Diabetic T-cells secreted twofold more gamma-interferon (IFN-gamma) and interleukin-17 than nondiabetic lymphocytes. Pretreatment with either S1P or SEW2871 significantly reduced cytokine secretion by approximately 50%. Flow cytometry analysis showed increased expression of CD69, a marker of lymphocyte activation, on diabetic T-cells. Both S1P and SEW2871 prevented upregulation of CD69 on CD4(+) cells. Quantitative RT-PCR showed that lymphocytes from diabetic NOD mice had 2.5-fold lower hypoxia-inducible factor (HIF)-1alpha short isoform I.1 (HIF1alphaI.1) mRNA levels than control. HIF1alphaI.1 is a negative regulator of lymphocyte activation. S1P significantly increased HIF1alpha I.1 mRNA levels in both control and diabetic groups. IFN-gamma production and surface CD69 expression was significantly increased in lymphocytes of HIF1alphaI.1-deficient mice. S1P did not reduce either CD69 or IFN-gamma expression in lymphocytes from HIF1alphaI.1-deficient mice. CONCLUSIONS: S1P acts through the S1P1 receptor and HIF1alpha I.1 to negatively regulate T-cell activation, providing a potential therapeutic target for prevention of diabetes and its vascular complications.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Diabetes Mellitus, Type 1/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocyte Activation/drug effects , Lysophospholipids/therapeutic use , Sphingosine/analogs & derivatives , T-Lymphocytes/immunology , Animals , Antigens, CD/drug effects , Antigens, Differentiation, T-Lymphocyte/drug effects , Cytokines/immunology , Diabetes Mellitus, Type 1/prevention & control , Diabetic Angiopathies/prevention & control , Flow Cytometry , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Lectins, C-Type , Mice , Mice, Inbred NOD , Mice, Knockout , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/therapeutic use , Spleen/immunology , T-Lymphocytes/drug effects
19.
J Immunol ; 179(11): 7431-8, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18025187

ABSTRACT

The genetic elimination of A2A adenosine receptors (A2AR) was shown to disengage the critical immunosuppressive mechanism and cause the dramatic exacerbation of acute inflammatory tissue damage by T cells and myeloid cells. This prompted the evaluation of the proinflammatory vs the anti-inflammatory effects of the widely consumed behavioral drug caffeine, as the psychoactive effects of caffeine are mediated largely by its antagonistic action on A2AR in the brain. Because caffeine has other biochemical targets besides A2AR, it was important to test whether the consumption of caffeine during an acute inflammation episode would lead to the exacerbation of immune-mediated tissue damage. We examined acute and chronic treatment with caffeine for its effects on acute liver inflammation. It is shown that caffeine at lower doses (10 and 20 mg/kg) strongly exacerbated acute liver damage and increased levels of proinflammatory cytokines. Because caffeine did not enhance liver damage in A2AR-deficient mice, we suggest that the potentiation of liver inflammation was mediated by interference with the A2AR-mediated tissue-protecting mechanism. In contrast, a high dose of caffeine (100 mg/kg) completely blocked both liver damage and proinflammatory cytokine responses through an A2AR-independent mechanism. Furthermore, caffeine administration exacerbated liver damage even when mice consumed caffeine chronically, although the extent of exacerbation was less than in "naive" mice that did not consume caffeine before. This study suggests an unappreciated "man-made" immunological pathogenesis whereby consumption of the food-, beverage-, and medication-derived adenosine receptor antagonists may modify an individual's inflammatory status and lead to excessive organ damage during acute inflammation.


Subject(s)
Adenosine A2 Receptor Antagonists , Caffeine/toxicity , Chemical and Drug Induced Liver Injury/immunology , Liver/immunology , Receptor, Adenosine A2A/immunology , ADP Ribose Transferases , Acute Disease , Animals , Bacterial Toxins , Caffeine/blood , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Exotoxins , Female , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphodiesterase Inhibitors/blood , Phosphodiesterase Inhibitors/toxicity , Receptor, Adenosine A2A/genetics , Virulence Factors , Pseudomonas aeruginosa Exotoxin A
20.
Purinergic Signal ; 3(1-2): 129-34, 2007 Mar.
Article in English | MEDLINE | ID: mdl-18404426

ABSTRACT

Cancer therapy by endogenous or adoptively transferred anti-tumor T cells is considered complementary to conventional cancer treatment by surgery, radiotherapy or chemotherapy. However, the scope of promising immunotherapeutic protocols is currently limited because tumors can create a "hostile" immunosuppressive microenvironment that prevents their destruction by anti-tumor T cells. There is a possibility to develop better and more effective immunotherapies by inactivating mechanisms that inhibit anti-tumor T cells in the tumor microenvironment and thereby protect cancerous tissues from immune damage. This may be now possible because of the recent demonstration that genetic deletion of immunosuppressive A2A and A2B adenosine receptors (A2AR and A2BR) or their pharmacological inactivation can prevent the inhibition of anti-tumor T cells by the hypoxic tumor microenvironment and as a result facilitate full tumor rejection [Ohta A, Gorelik E, Prasad SJ et al (2006) Proc Natl Acad Sci USA 103(35):13132-13137]. This approach is based on in vivo genetic evidence that A2AR play a critical role in the protection of normal tissues from overactive immune cells in acutely inflamed and hypoxic areas. The observations of much improved T-cell-mediated rejection of tumors in mice with inactivated A2AR strongly suggest that A2AR also protects hypoxic cancerous tissues and that A2AR should be inactivated in order to improve tumor rejection by anti-tumor T cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...