Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Biochem Pharmacol ; 188: 114541, 2021 06.
Article in English | MEDLINE | ID: mdl-33812857

ABSTRACT

The acute phase response, as a component of the innate immune system, is part of the first line of defense against invading pathogens. The Stimulator of Interferon Genes (STING) pathway initiates innate immune responses upon recognition of exogenous bacterial and viral DNA. However, whether STING signaling pathway plays any roles in regulating acute phase response during bacterial infection remains unknown. In this study, we used STING-deficient (Tmem173gt) and wildtype mice to investigate acute phase responses to bacterial infection (Escherichia coli, E. coli) and test the effect of exogenous cyclic GMP-AMP (cGAMP, a STING agonist) treatment. Bacterial infection of STING-deficient mice resulted in an increase in mortality and bacterial dissemination. Also, inflammation-induced acute phase response was drastically reduced in STING-deficient mice, showing significant reduction in expression of cytokine TNF-α and acute phase proteins. In contrast, exogenous cGAMP treatment enhanced inflammation-induced acute phase response by increasing the expression of TNF-α and acute phase proteins. Also, cGAMP accelerated bacterial clearance and improved survival rate of wildtype mice, but not STING-deficient mice. Interestingly, cGAMP treatment mitigated bacterial infection induced liver injury in both wildtype and STING-deficient mice. Further in vitro evidence showed that cGAMP treatment retarded TNF-α-mediated hepatocyte apoptosis, potentially accelerating autophagy. Taken together, our results indicated that cGAMP/STING signaling pathway is critical for organism to initiate blood-borne innate immune-responses to defend bacterial infection, and cGAMP is envisaged as a drug candidate for further clinical trial.


Subject(s)
Acute-Phase Reaction/metabolism , Acute-Phase Reaction/prevention & control , Cyclic AMP/administration & dosage , Cyclic GMP/administration & dosage , Escherichia coli Infections/metabolism , Escherichia coli Infections/prevention & control , Membrane Proteins/deficiency , Acute-Phase Reaction/genetics , Animals , Escherichia coli , Escherichia coli Infections/genetics , Hepatocytes/metabolism , Hepatocytes/microbiology , Male , Membrane Proteins/agonists , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
2.
Biomed Res Int ; 2021: 8852233, 2021.
Article in English | MEDLINE | ID: mdl-33575350

ABSTRACT

BACKGROUND/AIM: Irreversible electroporation (IRE) showed promising results for small-size tumors and very early cancers. However, further development is needed to evolve this procedure into a more efficient ablation technique for long-term control of tumor growth. In this work, we show that it is possible to increase the antitumor efficiency of IRE by simmultaneously injecting c-di-GMP, a STING agonist, intratumorally. MATERIALS AND METHODS: Intratumoral administration of c-di-GMP simultaneously to IRE was evaluated in murine models of melanona (B16.OVA) and hepatocellular carcinoma (PM299L). RESULTS: The combined therapy increased the number of tumor-infiltrating IFN-γ/TNF-α-producing CD4 and CD8 T cells and delayed tumor growth, as compared to the effect observed in groups treated with c-di-GMP or IRE alone. CONCLUSION: These results can lead to the development of a new therapeutic strategy for the treatment of cancer patients refractory to other therapies.


Subject(s)
Ablation Techniques/methods , Carcinoma, Hepatocellular/therapy , Cyclic GMP/analogs & derivatives , Electroporation/methods , Liver Neoplasms/therapy , Membrane Proteins/agonists , Animals , Cell Line , Combined Modality Therapy/methods , Cyclic GMP/administration & dosage , Female , Liver Neoplasms, Experimental/therapy , Mice, Inbred C57BL
3.
Front Immunol ; 11: 1674, 2020.
Article in English | MEDLINE | ID: mdl-32849581

ABSTRACT

Cyclic dinucleotides (CDNs) are promising vaccine adjuvants inducing balanced, potent humoral, and cellular immune responses. How aging influences CDN efficacy is unclear. We examined the vaccine efficacy of 3',5'-cyclic diguanylic acid (cyclic di-GMP, CDG), the founding member of CDNs, in 1-year-old (middle-aged) and 2-year-old (aged) C57BL/6J mice. We found that 1- and 2-year-old C57BL/6J mice are defective in CDG-induced memory T helper (Th)1 and Th17 responses and high-affinity serum immunoglobulin (Ig)G, mucosal IgA production. Next, we generated two novel tumor necrosis factor (TNF) fusion proteins that target soluble TNF (solTNF) and transmembrane TNF (tmTNF) to monocyte-derived dendritic cells (moDCs) to enhance CDG vaccine efficacy in 1- and 2-year-old mice. The moDC-targeting TNF fusion proteins restored CDG-induced memory Th1, Th17, and high-affinity IgG, IgA responses in the 1- and 2-year-old mice. Together, the data suggested that aging negatively impacts CDG vaccine adjuvanticity. MoDC-targeting TNF fusion proteins enhanced CDG adjuvanticity in the aging mice.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cyclic GMP/analogs & derivatives , Dendritic Cells/drug effects , Immunoglobulin Fc Fragments/pharmacology , Lung/drug effects , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Age Factors , Aging/immunology , Aging/metabolism , Animals , Cells, Cultured , Cyclic GMP/administration & dosage , Cyclic GMP/pharmacology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity, Mucosal/drug effects , Immunization , Immunogenicity, Vaccine , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin Fc Fragments/administration & dosage , Lung/immunology , Lung/metabolism , Mice, Inbred C57BL , Recombinant Fusion Proteins/administration & dosage , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Tumor Necrosis Factor-alpha/administration & dosage
4.
Cancer Res ; 79(20): 5394-5406, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31431457

ABSTRACT

Effective cancer immunotherapy depends on the robust activation of tumor-specific antigen-presenting cells (APC). Immune agonists encapsulated within nanoparticles (NP) can be delivered to tumor sites to generate powerful antitumor immune responses with minimal off-target dissemination. Systemic delivery enables widespread access to the microvasculature and draining to the APC-rich perivasculature. We developed an immuno-nanoparticle (immuno-NP) coloaded with cyclic diguanylate monophosphate, an agonist of the stimulator of interferon genes pathway, and monophosphoryl lipid A, and a Toll-like receptor 4 agonist, which synergize to produce high levels of type I IFNß. Using a murine model of metastatic triple-negative breast cancer, systemic delivery of these immuno-NPs resulted in significant therapeutic outcomes due to extensive upregulation of APCs and natural killer cells in the blood and tumor compared with control treatments. These results indicate that NPs can facilitate systemic delivery of multiple immune-potentiating cargoes for effective APC-driven local and systemic antitumor immunity. SIGNIFICANCE: Systemic administration of an immuno-nanoparticle in a murine breast tumor model drives a robust tumor site-specific APC response by delivering two synergistic immune-potentiating molecules, highlighting the potential of nanoparticles for immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , Cyclic GMP/analogs & derivatives , Drug Delivery Systems/methods , Interferon-beta/physiology , Lipid A/analogs & derivatives , Mammary Neoplasms, Experimental/drug therapy , Melanoma, Experimental/drug therapy , Nanocapsules/administration & dosage , Toll-Like Receptor 4/agonists , Triple Negative Breast Neoplasms/drug therapy , Animals , Antigen-Presenting Cells/drug effects , Cyclic GMP/administration & dosage , Cyclic GMP/therapeutic use , Drug Screening Assays, Antitumor , Drug Synergism , Female , Killer Cells, Natural/immunology , Lipid A/administration & dosage , Lipid A/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microcirculation , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology
5.
Mol Pharm ; 15(11): 5437-5443, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30299105

ABSTRACT

Microneedle-based skin allergen-specific immunotherapy (AIT) can benefit from adjuvants that can stimulate a stronger Th1 response against the allergen. We evaluated two stimulator of interferon genes (STING) agonists, namely, cyclic diguanylate monophosphate (c-di-GMP) and cyclic diadenylate monophosphate (c-di-AMP), as skin adjuvants using coated microneedles (MNs). For comparison, the approved subcutaneous (SC) hypodermic injection containing alum was used. Ovalbumin (Ova) was used as a model allergen. Ova-specific IgG2a antibody in serum, which is a surrogate marker for Th1 type immune response was significantly higher when STING agonists were used with coated MNs as compared to SC injection of Ova+alum in mice. In contrast, IgG1 antibody, a surrogate marker for Th2 type immune response, was at comparable levels in the MN and SC groups. Restimulation of splenocytes with Ova produced higher levels of Th1 cytokines (IFN-γ and IL-2) in the STING agonists MN groups as compared to the SC group. In conclusion, delivery of STING agonists into the skin using coated MNs activated the Th1 pathway better than SC- and MN-based delivery of alum. Thus, STING agonists could fulfill the role of adjuvants for skin AIT and even for infectious disease vaccines, where stimulation of the Th1 pathway is of interest.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Allergens/administration & dosage , Desensitization, Immunologic/methods , Th1 Cells/immunology , Th2 Cells/immunology , Administration, Cutaneous , Alum Compounds/administration & dosage , Animals , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Dinucleoside Phosphates/administration & dosage , Female , Membrane Proteins/agonists , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Models, Animal , Needles , Ovalbumin/administration & dosage , Th1 Cells/drug effects , Th2 Cells/drug effects
6.
Nanoscale ; 10(19): 9311-9319, 2018 May 17.
Article in English | MEDLINE | ID: mdl-29737353

ABSTRACT

Nanotechnology has demonstrated tremendous clinical utility, with potential applications in cancer immunotherapy. Although nanoparticles with intrinsic cytotoxicity are often considered unsuitable for clinical applications, such toxicity may be harnessed in the fight against cancer. Nanoparticle-associated toxicity can induce acute necrotic cell death, releasing tumor-associated antigens which may be captured by antigen-presenting cells to initiate or amplify tumor immunity. To test this hypothesis, cytotoxic cationic silica nanoparticles (CSiNPs) were directly administered into B16F10 melanoma implanted in C57BL/6 mice. CSiNPs caused plasma membrane rupture and oxidative stress of tumor cells, inducing local inflammation, tumor cell death and the release of tumor-associated antigens. The CSiNPs were further complexed with bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP), a molecular adjuvant which activates the stimulator of interferon genes (STING) in antigen-presenting cells. Compared with unformulated c-di-GMP, the delivery of c-di-GMP with CSiNPs markedly prolonged its local retention within the tumor microenvironment and activated tumor-infiltrating antigen-presenting cells. The combination of CSiNPs and a STING agonist showed dramatically increased expansion of antigen-specific CD8+ T cells, and potent tumor growth inhibition in murine melanoma. These results demonstrate that cationic nanoparticles can be used as an effective in situ vaccine platform which simultaneously causes tumor destruction and immune activation.


Subject(s)
Cell Death , Melanoma, Experimental/immunology , Membrane Proteins/metabolism , Nanoparticles , Silicon Dioxide , Tumor Microenvironment , Animals , CD8-Positive T-Lymphocytes , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Female , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Oxidative Stress
7.
Proc Natl Acad Sci U S A ; 115(13): E2997-E3006, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29531030

ABSTRACT

Inherited retinal degeneration (RD) is a devastating and currently untreatable neurodegenerative condition that leads to loss of photoreceptor cells and blindness. The vast genetic heterogeneity of RD, the lack of "druggable" targets, and the access-limiting blood-retinal barrier (BRB) present major hurdles toward effective therapy development. Here, we address these challenges (i) by targeting cGMP (cyclic guanosine- 3',5'-monophosphate) signaling, a disease driver common to different types of RD, and (ii) by combining inhibitory cGMP analogs with a nanosized liposomal drug delivery system designed to facilitate transport across the BRB. Based on a screen of several cGMP analogs we identified an inhibitory cGMP analog that interferes with activation of photoreceptor cell death pathways. Moreover, we found liposomal encapsulation of the analog to achieve efficient drug targeting to the neuroretina. This pharmacological treatment markedly preserved in vivo retinal function and counteracted photoreceptor degeneration in three different in vivo RD models. Taken together, we show that a defined class of compounds for RD treatment in combination with an innovative drug delivery method may enable a single type of treatment to address genetically divergent RD-type diseases.


Subject(s)
Blood-Retinal Barrier/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/administration & dosage , Disease Models, Animal , Drug Delivery Systems , Retinal Degeneration/drug therapy , Animals , Blood-Retinal Barrier/drug effects , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Liposomes , Mice , Photoreceptor Cells/metabolism , Retina/drug effects , Retina/metabolism , Retinal Degeneration/metabolism , Signal Transduction/drug effects
8.
Braz J Med Biol Res ; 51(5): e6693, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29561954

ABSTRACT

Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 µM and 1 mM), L-arginine (10, 100, 300, and 500 µM), ODQ (300 µM), and 8-Br-cGMP (100 µM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 µM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO.


Subject(s)
Adenosine Triphosphate/physiology , Leydig Cells/physiology , Nitric Oxide/physiology , Receptors, Purinergic/metabolism , Action Potentials , Animals , Arginine/administration & dosage , Arginine/metabolism , Cells, Cultured , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Male , Mice , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/metabolism , Nitric Oxide/biosynthesis , Patch-Clamp Techniques , Thionucleotides/administration & dosage , Thionucleotides/metabolism
9.
Appl Environ Microbiol ; 84(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29427430

ABSTRACT

Pseudomonas aeruginosa exhibits flagellum-mediated swimming in liquid and swarming on hydrated surfaces under diverse nutrient conditions. Prior studies have implicated a phosphodiesterase, DipA, in regulating these flagellum-mediated motilities, but collectively, the necessity for DipA was unclear. In this study, we find that the medium composition conditionally constrains the influence of DipA on flagellar motility. We show that DipA exhibits more influence on minimal medium supplemented with glutamate or glucose, where flagellar motility was negated for the dipA mutant. Conversely, a dipA-deficient mutant exhibits flagellar motility when growing with LB Lennox broth and minimal medium supplemented with Casamino Acids. Swarming under these rich medium conditions occurs under elevated levels of c-di-GMP. We also demonstrate that the influence of DipA upon swimming often differs from that upon swarming, and we conclude that a direct comparison of the motility phenotypes is generally valid only when characterizing motility assay results from the same medium composition. Our results are consistent with the explanation that DipA is one of several phosphodiesterases responding to the nutrient environment sensed by P. aeruginosa On minimal medium with glutamate or glucose, DipA is dominant; however, on rich medium, the necessity of DipA is fully negated.IMPORTANCE Motile and ubiquitous bacteria such as Pseudomonas aeruginosa can quickly colonize surfaces and form biofilms in numerous environments such as water distribution systems, soil, and the human lung. To effectively disrupt bacterial colonization, it is imperative to understand how bacteria regulate motility in these different growth environments. Here, we show that the phosphodiesterase DipA is not required for flagellar motility under all nutrient conditions. Thus, the maintenance of intracellular c-di-GMP levels to promote flagellar motility or biofilm development must be conditionally regulated by differing phosphodiesterases in variation with select nutrient cues.


Subject(s)
Bacterial Proteins/genetics , Cyclic GMP/analogs & derivatives , Gene Expression Regulation, Bacterial/physiology , Phosphoric Diester Hydrolases/genetics , Pseudomonas aeruginosa/genetics , Bacterial Proteins/metabolism , Cyclic GMP/administration & dosage , Phosphoric Diester Hydrolases/metabolism , Pseudomonas aeruginosa/metabolism
10.
Braz. j. med. biol. res ; 51(5): e6693, 2018. graf
Article in English | LILACS | ID: biblio-889091

ABSTRACT

Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 μM and 1 mM), L-arginine (10, 100, 300, and 500 μM), ODQ (300 μM), and 8-Br-cGMP (100 μM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 μM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO.


Subject(s)
Animals , Male , Mice , Adenosine Triphosphate/physiology , Receptors, Purinergic/metabolism , Leydig Cells/physiology , Nitric Oxide/physiology , Arginine/administration & dosage , Arginine/metabolism , Thionucleotides/administration & dosage , Thionucleotides/metabolism , Action Potentials , Cells, Cultured , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Patch-Clamp Techniques , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/metabolism , Nitric Oxide/biosynthesis
11.
J Clin Invest ; 127(6): 2176-2191, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28436934

ABSTRACT

Therapies using T cells that are programmed to express chimeric antigen receptors (CAR T cells) consistently produce positive results in patients with hematologic malignancies. However, CAR T cell treatments are less effective in solid tumors for several reasons. First, lymphocytes do not efficiently target CAR T cells; second, solid tumors create an immunosuppressive microenvironment that inactivates T cell responses; and third, solid cancers are typified by phenotypic diversity and thus include cells that do not express proteins targeted by the engineered receptors, enabling the formation of escape variants that elude CAR T cell targeting. Here, we have tested implantable biopolymer devices that deliver CAR T cells directly to the surfaces of solid tumors, thereby exposing them to high concentrations of immune cells for a substantial time period. In immunocompetent orthotopic mouse models of pancreatic cancer and melanoma, we found that CAR T cells can migrate from biopolymer scaffolds and eradicate tumors more effectively than does systemic delivery of the same cells. We have also demonstrated that codelivery of stimulator of IFN genes (STING) agonists stimulates immune responses to eliminate tumor cells that are not recognized by the adoptively transferred lymphocytes. Thus, these devices may improve the effectiveness of CAR T cell therapy in solid tumors and help protect against the emergence of escape variants.


Subject(s)
Biopolymers/administration & dosage , Carcinoma, Pancreatic Ductal/therapy , Melanoma, Experimental/therapy , Pancreatic Neoplasms/therapy , Adoptive Transfer , Animals , Antigen-Presenting Cells/physiology , Antineoplastic Agents/administration & dosage , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Drug Carriers/administration & dosage , Female , Implants, Experimental , Melanoma, Experimental/immunology , Membrane Proteins/agonists , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Pancreatic Neoplasms/immunology , T-Lymphocytes/physiology
12.
Brain Behav Immun ; 57: 360-370, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27189036

ABSTRACT

Patients with hepatic encephalopathy (HE) show working memory and visuo-spatial orientation deficits. Hyperammonemia is a main contributor to cognitive impairment in HE. Hyperammonemic rats show impaired spatial learning and learning ability in the Y maze. Intracerebral administration of extracellular cGMP restores learning in the Y-maze. The underlying mechanisms remain unknown. It also remains unknown whether extracellular cGMP improves neuroinflammation or restores spatial learning in hyperammonemic rats and if it affects differently reference and working memory. The aims of this work were: Spatial working and reference memory were assessed using the radial and Morris water mazes and neuroinflammation by immunohistochemistry and Western blot. Membrane expression of NMDA and AMPA receptor subunits was analyzed using the BS3 crosslinker. Extracellular cGMP was administered intracerebrally using osmotic minipumps. Chronic hyperammonemia induces neuroinflammation in hippocampus, with astrocytes activation and increased IL-1ß, which are associated with increased NMDA receptors membrane expression and impaired working memory. This process is not affected by extracellular cGMP. Hyperammonemia also activates microglia and increases TNF-α, alters membrane expression of AMPA receptor subunits (increased GluA1 and reduced GluA2) and impairs reference memory. All these changes are reversed by extracellular cGMP. These results show that extracellular cGMP modulates spatial reference memory but not working memory. This would be mediated by modulation of TNF-α levels and of membrane expression of GluA1 and GluA2 subunits of AMPA receptors.


Subject(s)
Cognitive Dysfunction/metabolism , Cyclic GMP/pharmacology , Hippocampus/metabolism , Hyperammonemia/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Memory, Short-Term , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spatial Memory , Tumor Necrosis Factor-alpha/metabolism , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cyclic GMP/administration & dosage , Disease Models, Animal , Hippocampus/drug effects , Hyperammonemia/complications , Hyperammonemia/drug therapy , Inflammation/drug therapy , Inflammation/etiology , Interleukin-1beta/drug effects , Male , Memory, Short-Term/drug effects , Rats , Rats, Wistar , Receptors, AMPA/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Spatial Memory/drug effects , Tumor Necrosis Factor-alpha/drug effects
13.
Cancer Res ; 76(8): 2137-52, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26951929

ABSTRACT

Endoplasmic reticulum (ER) stress responses through the IRE-1/XBP-1 pathway are required for the function of STING (TMEM173), an ER-resident transmembrane protein critical for cytoplasmic DNA sensing, IFN production, and cancer control. Here we show that the IRE-1/XBP-1 pathway functions downstream of STING and that STING agonists selectively trigger mitochondria-mediated apoptosis in normal and malignant B cells. Upon stimulation, STING was degraded less efficiently in B cells, implying that prolonged activation of STING can lead to apoptosis. Transient activation of the IRE-1/XBP-1 pathway partially protected agonist-stimulated malignant B cells from undergoing apoptosis. In Eµ-TCL1 mice with chronic lymphocytic leukemia, injection of the STING agonist 3'3'-cGAMP induced apoptosis and tumor regression. Similarly efficacious effects were elicited by 3'3'-cGAMP injection in syngeneic or immunodeficient mice grafted with multiple myeloma. Thus, in addition to their established ability to boost antitumoral immune responses, STING agonists can also directly eradicate malignant B cells. Cancer Res; 76(8); 2137-52. ©2016 AACR.


Subject(s)
Apoptosis/physiology , B-Lymphocytes/metabolism , Membrane Proteins/agonists , Animals , B-Lymphocytes/cytology , Cell Line , Cyclic GMP/administration & dosage , Cyclic GMP/pharmacology , Injections, Intraperitoneal , Membrane Proteins/physiology , Mice
14.
Stem Cells ; 33(10): 2973-84, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26269433

ABSTRACT

Cardiac hypertrophy is an abnormal enlargement of heart muscle. It frequently results in congestive heart failure, which is a leading cause of human death. Previous studies demonstrated that the nitric oxide (NO), cyclic GMP (cGMP), and protein kinase G (PKG) signaling pathway can inhibit cardiac hypertrophy and thus improve cardiac function. However, the underlying mechanisms are not fully understood. Here, based on the human embryonic stem cell-derived cardiomyocyte (hESC-CM) model system, we showed that Orai1, the pore-forming subunit of store-operated Ca(2+) entry (SOCE), is the downstream effector of PKG. Treatment of hESC-CMs with an α-adrenoceptor agonist phenylephrine (PE) caused a marked hypertrophy, which was accompanied by an upregulation of Orai1. Moreover, suppression of Orai1 expression/activity using Orai1-siRNAs or a dominant-negative construct Orai1(G98A) inhibited the hypertrophy, suggesting that Orai1-mediated SOCE is indispensable for the PE-induced hypertrophy of hESC-CMs. In addition, the hypertrophy was inhibited by NO and cGMP via activating PKG. Importantly, substitution of Ala for Ser(34) in Orai1 abolished the antihypertrophic effects of NO, cGMP, and PKG. Furthermore, PKG could directly phosphorylate Orai1 at Ser(34) and thus prevent Orai1-mediated SOCE. Together, we conclude that NO, cGMP, and PKG inhibit the hypertrophy of hESC-CMs via PKG-mediated phosphorylation on Orai1-Ser-34. These results provide novel mechanistic insights into the action of cGMP-PKG-related antihypertrophic agents, such as NO donors and sildenafil.


Subject(s)
Calcium Channels/genetics , Cardiomegaly/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Heart Failure/genetics , Calcium/metabolism , Calcium Channels/biosynthesis , Calcium Channels/drug effects , Cardiomegaly/pathology , Cell Differentiation/genetics , Cyclic GMP/administration & dosage , Cyclic GMP/metabolism , Gene Expression Regulation/drug effects , Heart Failure/pathology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitric Oxide/administration & dosage , Nitric Oxide/metabolism , ORAI1 Protein , Phenylephrine/administration & dosage , Phosphorylation/drug effects , Signal Transduction/drug effects
15.
J Mol Cell Cardiol ; 87: 74-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26219954

ABSTRACT

The Exchange Protein directly Activated by cAMP (EPAC) participates to the pathological signaling of cardiac hypertrophy and heart failure, in which the role of Ca(2+) entry through the Transient Receptor Potential Canonical (TRPC) channels begin to be appreciated. Here we studied whether EPAC activation could influence the activity and/or expression of TRPC channels in cardiac myocytes. In adult rat ventricular myocytes treated for 4 to 6h with the selective EPAC activator, 8-pCPT (10µM), we observed by Fluo-3 confocal fluorescence a Store-Operated Ca(2+) Entry (SOCE) like-activity, which was blunted by co-incubation with EPAC inhibitors (ESI-05 and CE3F4 at 10 µM). This SOCE-like activity, which was very small in control incubated cells, was sensitive to 30-µM SKF-96365. Molecular screening showed a specific upregulation of TRPC3 and C4 protein isoforms after 8-pCPT treatment. Moreover, sustained EPAC activation favored proarrhythmic Ca(2+) waves, which were reduced either by co-incubation with EPAC inhibitors or bath perfusion with TRPC inhibitors. Our study provides the first evidence that sustained selective EPAC activation leads to an increase in TRPC3 and C4 protein expression and induces a proarrhythmic SOCE-like activity in adult rat ventricular cardiomyocytes, which might be of importance during the development of cardiac diseases.


Subject(s)
Cardiomegaly/genetics , Complement C4/biosynthesis , Guanine Nucleotide Exchange Factors/biosynthesis , Myocytes, Cardiac/metabolism , TRPC Cation Channels/genetics , Animals , Benzene Derivatives/administration & dosage , Calcium/metabolism , Calcium Signaling/drug effects , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Complement C4/genetics , Cyclic AMP/metabolism , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Guanine Nucleotide Exchange Factors/genetics , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Quinolines/administration & dosage , Rats , Sulfones/administration & dosage , TRPC Cation Channels/antagonists & inhibitors , Thionucleotides/administration & dosage
16.
Cancer Immunol Immunother ; 64(8): 1057-66, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986168

ABSTRACT

Therapeutic vaccines to induce anti-tumor CD8 T cells have been used in clinical trials for advanced melanoma patients, but the clinical response rate and overall survival time have not improved much. We believe that these dismal outcomes are caused by inadequate number of antigen-specific CD8 T cells generated by most vaccines. In contrast, huge CD8 T cell responses readily occur during acute viral infections. High levels of type-I interferon (IFN-I) are produced during these infections, and this cytokine not only exhibits anti-viral activity but also promotes CD8 T cell responses. The studies described here were performed to determine whether promoting the production of IFN-I could enhance the potency of a peptide vaccine. We report that cyclic diguanylate monophosphate (c-di-GMP), which activates the stimulator of interferon genes, potentiated the immunogenicity and anti-tumor effects of a peptide vaccine against mouse B16 melanoma. The synergistic effects of c-di-GMP required co-administration of costimulatory anti-CD40 antibody, the adjuvant poly-IC, and were mediated in part by IFN-I. These findings demonstrate that peptides representing CD8 T cell epitopes can be effective inducers of large CD8 T cell responses in vaccination strategies that mimic acute viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/therapeutic use , Cyclic GMP/analogs & derivatives , Immunotherapy/methods , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Animals , Antibodies, Monoclonal/administration & dosage , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cyclic GMP/administration & dosage , Cyclic GMP/adverse effects , Epitopes, T-Lymphocyte/administration & dosage , Humans , Interferon Type I/immunology , Lymphocyte Activation/drug effects , Melanoma, Experimental/immunology , Membrane Proteins/agonists , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/administration & dosage , Receptors, Interferon/genetics , Skin Neoplasms/immunology , Vaccines, Subunit/therapeutic use
17.
Cell Rep ; 11(1): 71-84, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25843711

ABSTRACT

Upon systemic bacterial infection, hematopoietic stem and progenitor cells (HSPCs) migrate to the periphery in order to supply a sufficient number of immune cells. Although pathogen-associated molecular patterns reportedly mediate HSPC activation, how HSPCs detect pathogen invasion in vivo remains elusive. Bacteria use the second messenger bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP) for a variety of activities. Here, we report that c-di-GMP comprehensively regulated both HSPCs and their niche cells through an innate immune sensor, STING, thereby inducing entry into the cell cycle and mobilization of HSPCs while decreasing the number and repopulation capacity of long-term hematopoietic stem cells. Furthermore, we show that type I interferon acted as a downstream target of c-di-GMP to inhibit HSPC expansion in the spleen, while transforming growth factor-ß was required for c-di-GMP-dependent splenic HSPC expansion. Our results define machinery underlying the dynamic regulation of HSPCs and their niches during bacterial infection through c-di-GMP/STING signaling.


Subject(s)
Cyclic GMP/analogs & derivatives , Hematopoietic Stem Cells/metabolism , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Membrane Proteins/genetics , Animals , Bacteria/metabolism , Bacteria/pathogenicity , Cyclic GMP/administration & dosage , Cyclic GMP/immunology , Cyclic GMP/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Interferon Regulatory Factor-3/immunology , Membrane Proteins/immunology , Mice , Signal Transduction/drug effects , Signal Transduction/immunology , Stem Cell Niche/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
18.
Hum Vaccin Immunother ; 11(5): 1235-43, 2015.
Article in English | MEDLINE | ID: mdl-25714901

ABSTRACT

Highly pathogenic avian influenza H5N1 infection remains a public health threat and vaccination is the best measure of limiting the impact of a potential pandemic. Mucosal vaccines have the advantage of eliciting immune responses at the site of viral entry, thereby preventing infection as well as further viral transmission. In this study, we assessed the protective efficacy of hemagglutinin (HA) from the A/Indonesia/05/05 (H5N1) strain of influenza virus that was produced by transient expression in plants. The plant-derived vaccine, in combination with the mucosal adjuvant (3',5')-cyclic dimeric guanylic acid (c-di-GMP) was used for intranasal immunization of mice and ferrets, before challenge with a lethal dose of the A/Indonesia/05/05 (H5N1) virus. Mice vaccinated with 15 µg or 5 µg of adjuvanted HA survived the viral challenge, while all control mice died within 10 d of challenge. Vaccinated animals elicited serum hemagglutination inhibition, IgG and IgA antibody titers. In the ferret challenge study, all animals vaccinated with the adjuvanted plant vaccine survived the lethal viral challenge, while 50% of the control animals died. In both the mouse and ferret models, the vaccinated animals were better protected from weight loss and body temperature changes associated with H5N1 infection compared with the non-vaccinated controls. Furthermore, the systemic spread of the virus was lower in the vaccinated animals compared with the controls. Results presented here suggest that the plant-produced HA-based influenza vaccine adjuvanted with c-di-GMP is a promising vaccine/adjuvant combination for the development of new mucosal influenza vaccines.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Disease Models, Animal , Female , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza Vaccines/isolation & purification , Male , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Plants, Genetically Modified , Survival Analysis , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
19.
Cancer Immunol Res ; 2(9): 901-10, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24913717

ABSTRACT

Cancer vaccination may be our best and most benign option for preventing or treating metastatic cancer. However, breakthroughs are hampered by immune suppression in the tumor microenvironment. In this study, we analyzed whether cyclic diguanylate (c-di-GMP), a ligand for stimulator of interferon genes (STING), could overcome immune suppression and improve vaccination against metastatic breast cancer. Mice with metastatic breast cancer (4T1 model) were therapeutically immunized with an attenuated Listeria monocytogenes (LM)-based vaccine, expressing tumor-associated antigen Mage-b (LM-Mb), followed by multiple low doses of c-di-GMP (0.2 µmol/L). This treatment resulted in a striking and near elimination of all metastases. Experiments revealed that c-di-GMP targets myeloid-derived suppressor cells (MDSC) and tumor cells. Low doses of c-di-GMP significantly increased the production of IL12 by MDSCs, in correlation with improved T-cell responses to Mage-b, whereas a high dose of c-di-GMP (range, 0.3-3 mmol/L) activated caspase-3 in the 4T1 tumor cells and killed the tumor cells directly. On the basis of these results, we tested one administration of high-dose c-di-GMP (3 mmol/L) followed by repeated administrations of low-dose c-di-GMP (0.2 µmol/L) in the 4T1 model, and found equal efficacy compared with the combination of LM-Mb and c-di-GMP. This finding correlated with a mechanism of improved CD8 T-cell responses to tumor-associated antigens (TAA) Mage-b and Survivin, most likely through cross-presentation of these TAAs from c-di-GMP-killed 4T1 tumor cells, and through c-di-GMP-activated TAA-specific T cells. Our results demonstrate that activation of STING-dependent pathways by c-di-GMP is highly attractive for cancer immunotherapy.


Subject(s)
Breast Neoplasms/secondary , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Cyclic GMP/analogs & derivatives , Membrane Proteins/genetics , Animals , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cross-Priming , Cyclic GMP/administration & dosage , Cyclic GMP/genetics , Female , HEK293 Cells , Humans , Immunotherapy , Listeria monocytogenes/genetics , Mammary Neoplasms, Experimental , Mice , Mice, Inbred BALB C , Tumor Microenvironment , Vaccines, Attenuated/therapeutic use
20.
J Cell Physiol ; 229(12): 2142-52, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24911927

ABSTRACT

Published data provide strong evidence that heparin treatment of proliferating vascular smooth muscle cells results in decreased signaling through the ERK pathway and decreases in cell proliferation. In addition, these changes have been shown to be mimicked by antibodies that block heparin binding to the cell surface. Here, we provide evidence that the activity of protein kinase G is required for these heparin effects. Specifically, a chemical inhibitor of protein kinase G, Rp-8-pCPT-cGMS, eliminates heparin and anti-heparin receptor antibody effects on bromodeoxyuridine incorporation into growth factor-stimulated cells. In addition, protein kinase G inhibitors decrease heparin effects on ERK activity, phosphorylation of the transcription factor Elk-1, and heparin-induced MKP-1 synthesis. Although transient, the levels of cGMP increase in heparin treated cells. Finally, knock down of protein kinase G also significantly decreases heparin effects in growth factor-activated vascular smooth muscle cells. Together, these data indicate that heparin effects on vascular smooth muscle cell proliferation depend, at least in part, on signaling through protein kinase G.


Subject(s)
Cell Proliferation/drug effects , Cyclic GMP-Dependent Protein Kinases/metabolism , Heparin/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Cyclic GMP/administration & dosage , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Gene Expression Regulation/drug effects , Heparin/administration & dosage , MAP Kinase Signaling System/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phosphorylation/drug effects , Rats , Receptors, Cell Surface/metabolism , Thionucleotides/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...