Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ACS Chem Neurosci ; 11(24): 4152-4168, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33225679

ABSTRACT

The accumulation and deposition of ß-amyloid (Aß) is one postulated cause of Alzheimer's disease (AD). In addition to its direct toxicity on neurons, Aß may induce neuroinflammation through the concomitant activation of microglia. Emerging evidence suggests that microglia-mediated neuroinflammation plays an important role in the pathogenesis of AD. As brain macrophages, microglia engulf misfolded-Aß by phagocytosis. However, the accumulated toxic Aß may paradoxically "hyper-activate" microglia into a neurotoxic proinflammatory and less phagocytotic phenotype, contributing to neuronal death. This study reports that the known drug furosemide is a potential probe molecule for reducing AD-neuroinflammation. Our data demonstrate that furosemide inhibits the secretion of proinflammatory TNF-α, IL-6, and nitric oxide; downregulates the mRNA level of Cd86 and the protein expression of COX-2, iNOS; promotes phagocytic activity; and enhances the expression of anti-inflammatory IL-1RA and arginase. Our mechanism of action studies further demonstrate that furosemide reduces LPS-induced upregulation of endoplasmic reticulum (ER) stress marker genes, including Grp78, Atf4, Chop, tXbp1, and sXbp1. These data support the observation that furosemide is a known drug with the capacity to downregulate the proinflammatory microglial M1 phenotype and upregulate the anti-inflammatory M2 phenotype, a potentially powerful and beneficial pharmacologic effect for inflammatory diseases such as AD.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Endoplasmic Reticulum Chaperone BiP , Furosemide/pharmacology , Humans , Microglia , Phagocytosis
2.
PeerJ ; 8: e9533, 2020.
Article in English | MEDLINE | ID: mdl-32704455

ABSTRACT

The novel coronavirus SARS-CoV-2 has become a global health concern. The morbidity and mortality of the potentially lethal infection caused by this virus arise from the initial viral infection and the subsequent host inflammatory response. The latter may lead to excessive release of pro-inflammatory cytokines, IL-6 and IL-8, as well as TNF-α ultimately culminating in hypercytokinemia ("cytokine storm"). To address this immuno-inflammatory pathogenesis, multiple clinical trials have been proposed to evaluate anti-inflammatory biologic therapies targeting specific cytokines. However, despite the obvious clinical utility of such biologics, their specific applicability to COVID-19 has multiple drawbacks, including they target only one of the multiple cytokines involved in COVID-19's immunopathy. Therefore, we set out to identify a small molecule with broad-spectrum anti-inflammatory mechanism of action targeting multiple cytokines of innate immunity. In this study, a library of small molecules endogenous to the human body was assembled, subjected to in silico molecular docking simulations and a focused in vitro screen to identify anti-pro-inflammatory activity via interleukin inhibition. This has enabled us to identify the loop diuretic furosemide as a candidate molecule. To pre-clinically evaluate furosemide as a putative COVID-19 therapeutic, we studied its anti-inflammatory activity on RAW264.7, THP-1 and SIM-A9 cell lines stimulated by lipopolysaccharide (LPS). Upon treatment with furosemide, LPS-induced production of pro-inflammatory cytokines was reduced, indicating that furosemide suppresses the M1 polarization, including IL-6 and TNF-α release. In addition, we found that furosemide promotes the production of anti-inflammatory cytokine products (IL-1RA, arginase), indicating M2 polarization. Accordingly, we conclude that furosemide is a reasonably potent inhibitor of IL-6 and TNF-α that is also safe, inexpensive and well-studied. Our pre-clinical data suggest that it may be a candidate for repurposing as an inhaled therapy against COVID-19.

3.
Immunobiology ; 225(2): 151886, 2020 03.
Article in English | MEDLINE | ID: mdl-31812341

ABSTRACT

The objective of this study was to investigate the effect of EF24, an NF-κB-inhibitor, on the expression of negative regulators in IL-1R pathway, namely ST2 and SIGIRR. Murine JAWS II dendritic cells (DC) were challenged with lipopolysaccharide (LPS, 100 ng/ml) for 4 h, followed by treatment with 10 µM EF24 for 1 h. ST2 and SIGIRR expression was monitored by qRT-PCR and immunoblotting. ST2L and MyD88 interaction was studied by co-immunoprecipitation, and IL-33, a ST2L ligand, was assayed by ELISA. Activation of transcription factor SP1 was examined by confocal microscopy, immunoblotting, and EMSA. The effect of EF24 on accumulation of ubiquitinated proteins in DCs and proteolysis of fluorogenic peptides by purified proteasome was studied. We found that EF24 upregulated the expression of ST2 and SIGIRR and decreased the interaction of the membrane-bound ST2 (ST2L) with MyD88, and significantly reduced IL-33 levels in LPS-stimulated DCs. Simultaneously it increased the activation of transcription factor SP1and restored the basal level of ubiquitinated proteins in LPS-stimulated DCs. Moreover, EF24 inhibited trypsin- and chymotrypsin-like activity of proteasome by directly interacting with 26S proteasome. The results suggest that EF24 activates endogenous anti-inflammatory arm of IL-1R signaling, most likely by stabilizing SP1 against proteasomal degradation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzylidene Compounds/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Inflammation Mediators/antagonists & inhibitors , Interleukin-1 Receptor-Like 1 Protein/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Piperidones/pharmacology , Receptors, Interleukin-1/antagonists & inhibitors , Animals , Cell Line , Interleukin-33/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Interleukin-1/metabolism , Sp1 Transcription Factor/metabolism , Up-Regulation/drug effects
4.
Vaccine ; 37(12): 1685-1691, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30795939

ABSTRACT

Valley Fever, or coccidioidomycosis, is caused by a soil-borne, highly virulent fungal pathogen, Coccidioides spp. Infection with Coccidioides can be life-threatening. Since an effective treatment is not available and the T cell-mediated immune response is protective, vaccine development is of interest. In this study, a primary dendritic cell (DC)-vaccine was evaluated for its ability to stimulate Coccidioides antigen-specific immune response in an extremely susceptible BALB/c mouse model. The DC-vaccine (Ag2-DC) was prepared by non-virally transfecting the primary bone marrow-derived DCs with a plasmid DNA encoding Ag2/PRA (protective epitope of Coccidioides). Mice were intranasally immunized with Ag2-DC on days 2 and 10. Immunized mice were necropsied on days 8, 32, and 44. Major organs and blood samples were harvested. The most common indicators of injury (protein, lactate, and albumin), Ag/PRA-specific cytokine-secreting cells, and IgG and its isotypes were determined by biochemical and immunologic assays, respectively. No signs of sickness were noted. Similarly, no significant changes were observed in the levels of total lung protein, lactate, and albumin, in immunized mice compared with healthy control mice. Interferon (IFN-γ), and interleukin (IL)-4 and IL-17 cytokine-secreting cells were observed in lung and lymph nodes upon Ag2-DC immunization. Our results showed that the levels of serum IgG and its isotypes were increased in Ag2-DC-immunized mice. This report provides evidence of DC immunization-stimulated Ag2/PRA-specific immune responses.


Subject(s)
Antigens, Fungal/immunology , Coccidioides/immunology , Coccidioidomycosis/immunology , Dendritic Cells/immunology , Fungal Proteins/immunology , Glycoproteins/immunology , Host-Pathogen Interactions/immunology , Animals , Antibodies, Fungal/immunology , Coccidioidomycosis/microbiology , Cytokines/biosynthesis , Dendritic Cells/metabolism , Disease Models, Animal , Female , Immunization , Lymphocytes/immunology , Lymphocytes/metabolism , Mice
5.
Can J Neurol Sci ; 46(1): 23-34, 2019 01.
Article in English | MEDLINE | ID: mdl-30688198

ABSTRACT

In recent decades, clinical trials in Alzheimer's disease (AD) have failed at an unprecedented rate. The etiology of AD has since come under renewed scrutiny, both to elucidate the underlying pathologies and to identify novel therapeutic strategies. Here, diet has emerged as a potential causative/protective agent. A variety of nutrients, including lipids, minerals, vitamins, antioxidants and sugars as well as broader dietary patterns and microbiotal interactions have demonstrated associations with AD. Although clinical trials have yet to definitively implicate any singular dietary element as therapeutic or causative, it is apparent that dietary preferences, likely in complex synergies, may influence the risk, onset and course of AD. This review catalogs the impact of major dietary elements on AD. It further examines an unexplored reciprocal association where AD may modulate diet, as well as how potential therapeutics may complicate these interactions. In doing so, we observe diet may have profound effects on the outcome of a clinical trial, either as a confounder of a drug/disease interaction or as a generally disruptive covariate. We therefore conclude that future clinical trials in AD should endeavor to control for diet, either in study design or subsequent analyses.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/therapy , Antipsychotic Agents/therapeutic use , Diet/methods , Nutrients , Alzheimer Disease/metabolism , Humans , Insulin/metabolism , Lipids , Microbiota , Sugars/metabolism
6.
J Inflamm (Lond) ; 12: 55, 2015.
Article in English | MEDLINE | ID: mdl-26401121

ABSTRACT

BACKGROUND: Unresolved and prolonged inflammation is a pathological basis of many disorders such as cancer and multiple organ failure in shock. Interleukin-1 receptor (IL-1R) superfamily consists of IL-1R1 and pathogen pattern recognition receptor toll-like receptor-4 (TLR4) which, upon ligand binding, initiate pro-inflammatory signaling. The study objective was to investigate the effect of a diphenyldifluoroketone EF24 on the expression of IL-1R1 and TLR4 in lipopolysaccharide (LPS)-stimulated dendritic cells (DCs). METHODS: Immortalized murine bone marrow-derived JAWS II dendritic cells (DC) were challenged with LPS (100 ng/ml) for 4 h. The LPS-stimulated DCs were treated with 10 µM of EF24 for 1 h. The expression levels of IL-1R1 and TLR4 were monitored by RT-PCR, immunoblotting, and confocal microscopy. The effect of EF24 on the viability and cell cycle of DCs was examined by lactate dehydrogenase assay and flow cytometry, respectively. RESULTS: EF24 treatment suppressed the LPS-induced TLR4 and IL-1R1 expression in DCs. However, the expression levels of IL-1RA and IL-1R2 were not influenced by either LPS or EF24 treatments. These effects of EF24 were associated with a decrease in LPS-induced expression of phospho-NF-kB p65, indicative of its role in the transcriptional control of IL-1R superfamily members. We did not find any significant effect of EF24 on the proliferation or cell cycle of DCs. CONCLUSIONS: The results suggest that EF24 influences IL-1R superfamily signaling pathway in ways that could have salutary effects in inflammation. The pluripotent anti-inflammatory actions of EF24 warrant further investigation of EF24 in inflammatory conditions of systemic nature.

7.
Artif Organs ; 38(8): 675-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24749913

ABSTRACT

Severe blood loss in victims of trauma creates an exaggerated inflammatory background that contributes to the development of intravascular coagulopathy and multiple organ dysfunction syndrome. We hypothesized that treatment with diphenyldifluoroketone EF24, an inhibitor of nuclear factor kappa-B, would have salutary effects in hemorrhagic shock. The objective of this study was to investigate the effect of EF24 on the expression of the interleukin-1 receptor (IL-1R) superfamily in a rat model of hypovolemic shock. Hypovolemia was induced by gradually withdrawing approximately 50% of circulating blood, and EF24 was administered intraperitoneally (0.2 mg/kg) in 50 µL of saline. After 6 h of shock, lung tissue was probed immunohistochemically and by immunoblotting to study the expression of Toll-like receptor 4 (TLR4), IL-1R, suppression of tumorigenicity 2 (ST2), and single immunoglobulin IL-1R-related (SIGIRR). The tissue-associated pro-inflammatory cytokines, tumor necrosis factor alpha (TNF-α) and IL-6, were measured by enzyme-linked immunosorbent assay. We observed a reduction in immunoreactive TLR4 and IL-1R1 in lung tissue of rats treated with EF24. Simultaneously, the pulmonary expression of ST2 and SIGIRR (the putative down-regulators of the pro-inflammatory IL-1R pathway) was increased in EF24-treated hemorrhaged rats. The concentration of hemorrhage-induced TNF-α and IL-6 in lung tissue homogenates was also reduced by EF24 treatment. These results confirm our previous in vitro observations in lipopolysaccharide-stimulated dendritic cells that EF24 beneficially modulates the IL-1R pathway and suggest that it could be investigated as an adjunct therapeutic in managing inflammation associated with hemorrhagic shock.


Subject(s)
Benzylidene Compounds/pharmacology , Lung/drug effects , Piperidones/pharmacology , Receptors, Interleukin-1/metabolism , Shock, Hemorrhagic/drug therapy , Signal Transduction/drug effects , Animals , Benzylidene Compounds/therapeutic use , Disease Models, Animal , Lung/metabolism , Male , NF-kappa B/metabolism , Piperidones/therapeutic use , Rats , Rats, Sprague-Dawley , Shock, Hemorrhagic/metabolism , Toll-Like Receptor 4/metabolism
8.
Appl Biochem Biotechnol ; 172(7): 3363-73, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24532443

ABSTRACT

Curcumin is well known for its antimicrobial and anti-inflammatory properties. However, since systemic absorption and bioavailability of curcumin from gastrointestinal tract is considerably poor, synthetic curcuminoids are being developed as better alternatives. Two curcumin derivatives: 3,5-bis(benzylidene)-4-piperidone (EF24) and EF24-dimer linked via diethylenetriaminepentacetic acid (EF2DTPA), were included in this study. We investigated the antibacterial activity of EF24 and EF2DTPA against Gram-negative (Escherichia coli) and Gram-positive (Enterococcus faecalis, Staphylococcus aureus) bacteria. We also studied the effects of EF24 and EF2DTPA on uptake and localization of pHrodo-labeled E. coli in the acidic compartments (phagolysosomes) of dendritic cells (DCs) under in vitro conditions. Our results demonstrate that treatment with EF24 and EF2DTPA directly suppresses the bacterial growth. However, these compounds do not affect the bacterial uptake or localization in the DCs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Curcumin/chemical synthesis , Curcumin/pharmacology , Pentetic Acid/chemistry , Piperidones/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacteria/drug effects , Cell Line , Curcumin/chemistry , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dimerization , Mice , Phagocytosis/drug effects , Piperidones/chemical synthesis , Piperidones/chemistry
9.
Int Immunol ; 24(7): 455-64, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22378503

ABSTRACT

Synthetic curcuminoid EF24 was studied for its effect on the maturation and inflammatory response in murine bone marrow derived immortalized JAWS II dendritic cells (DCs). EF24 reduced the expression of LPS-induced MHC class II, CD80 and CD86 molecules. It also abrogated the appearance of dendrites, a typical characteristic of mature DCs. These effects were accompanied by the inhibition of LPS-induced activation of transcription factor nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB). Simultaneous reduction of pro-inflammatory cytokines [tumor necrosis factor (TNF)-α, IL-6] both at the mRNA and secreted levels was also observed. To investigate the dependency of LPS effects on MyD88 adaptor protein, we transfected JAWS II DCs with dominant negative MyD88 plasmid construct (MyD88-DN). EF24 reduced NF-κB activity and TNF-α secretion in a MyD88-dependent manner. These results suggest that EF24 modulates DCs by suppressing their maturation and reducing the secretion of inflammatory cytokines. Further, it appears that EF24 acts at or upstream of MyD88 in the LPS-TLR4/MyD88/NF-κB pathway.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzylidene Compounds/pharmacology , Dendritic Cells/drug effects , Piperidones/pharmacology , Animals , Bone Marrow Cells/immunology , Cell Differentiation/drug effects , Cell Line, Transformed , Cells, Cultured , Curcumin/analogs & derivatives , Cytokines/metabolism , Dendritic Cells/immunology , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Transcriptional Activation/drug effects
10.
BMC Immunol ; 11: 60, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21143974

ABSTRACT

BACKGROUND: Coccidioidomycosis or Valley fever is caused by a highly virulent fungal pathogen: Coccidioides posadasii or immitis. Vaccine development against Coccidioides is of contemporary interest because a large number of relapses and clinical failures are reported with antifungal agents. An efficient Th1 response engenders protection. Thus, we have focused on developing a dendritic cell (DC)-based vaccine for coccidioidomycosis. In this study, we investigated the immunostimulatory characteristics of an intranasal primary DC-vaccine in BALB/c mouse strain that is most susceptible to coccidioidomycosis. The DCs were transfected nonvirally with Coccidioides-Ag2/PRA-cDNA. Expression of DC-markers, Ag2/PRA and cytokines were studied by flow cytometry, dot-immunoblotting and cytometric bead array methods, respectively. The T cell activation was studied by assessing the upregulation of activation markers in a DC-T cell co-culture assay. For trafficking, the DCs were co-transfected with a plasmid DNA encoding HSV1 thymidine kinase (TK) and administered intranasally into syngeneic mice. The trafficking and homing of TK-expressing DCs were monitored with positron emission tomography (PET) using 18F-FIAU probe. Based on the PET-probe accumulation in vaccinated mice, selected tissues were studied for antigen-specific response and T cell phenotypes using ELISPOT and flow cytometry, respectively. RESULTS: We found that the primary DCs transfected with Coccidioides-Ag2/PRA-cDNA were of immature immunophenotype, expressed Ag2/PRA and activated naïve T cells. In PET images and subsequent biodistribution, intranasally-administered DCs were found to migrate in blood, lung and thymus; lymphocytes showed generation of T effector memory cell population (T(EM)) and IFN-γ release. CONCLUSIONS: In conclusion, our results demonstrate that the intranasally-administered primary DC vaccine is capable of inducing Ag2/PRA-specific T cell response. Unique approaches utilized in our study represent an attractive and novel means of producing and evaluating an autologous DC-based vaccine.


Subject(s)
Dendritic Cells/immunology , Fungal Vaccines/administration & dosage , Fungal Vaccines/immunology , Immunization/methods , Administration, Intranasal , Animals , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Shape , Cells, Cultured , Coccidioides/immunology , Cytokines/metabolism , DNA, Complementary/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , Epitopes/immunology , Immunologic Memory , Immunophenotyping , Lymphocyte Activation/immunology , Mice , Models, Animal , Molecular Imaging , Plasmids/genetics , Thymidine Kinase/metabolism , Tissue Distribution , Transfection , Transgenes/genetics
11.
Bioorg Med Chem ; 18(16): 6109-20, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20638855

ABSTRACT

3,5-Bis(benzylidene)-4-piperidones are being advanced as synthetic analogs of curcumin for anti-cancer and anti-inflammatory properties. We performed structure-activity relationship studies, by testing several synthesized 3,5-bis(benzylidene)-4-piperidones for anti-proliferative activity in lung adenocarcinoma H441 cells. Compared to the lead compound 1, or 3,5-bis(2-fluorobenzylidene)-4-piperidone, five compounds were found to be more potent (IC(50) < 30 microM), and 16 compounds possessed reduced cell-killing efficacy (IC(50) > 50 microM). Based on the observations, we synthesized 4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid] (29 or CLEFMA) as a novel analog of 1. CLEFMA was evaluated for anti-proliferative activity in H441 cells, and was found to be several folds more potent than compound 1. We did not find apoptotic cell population in flow cytometry, and the absence of apoptosis was confirmed by the lack of caspase cleavage. The electron microscopy of H441cells indicated that CLEFMA and compound 1 induce autophagic cell death that was inhibited by specific autophagy inhibitor 3-methyladenine. The results suggest that the potent and novel curcuminoid, CLEFMA, offers an alternative mode of cell death in apoptosis-resistant cancers.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzylidene Compounds/chemistry , Benzylidene Compounds/pharmacology , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Piperidones/chemistry , Piperidones/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Humans , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 19(16): 4764-7, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19574048

ABSTRACT

N4-Modified, novel Ara-C conjugate capable of radiolabeling with gamma ray-emitting ((99m)Tc) as well as positron emitting ((18)F) radionuclides, that is, N4-hydrazine derivative was synthesized. The radiolabeling of N4-(hydrazinonicotinyl)-1-beta-arabinofuranosyl cytosine (HAra-C) with (99m)Tc was performed with over 95% labeling yield. To label HAra-C with (18)F, 4-fluoro((18)F)-benzaldehyde was synthesized from 4-formyl-N,N,N-trimethylanilinium triflate in 30% radiochemical yield; it quantitatively formed hydrazone derivative with HAra-C within 45min. The radiolabeled conjugates were analyzed by radio-UV-RP-HPLC. The cold precursors were characterized by (1)H, (13)C NMR. Additionally, HAra-C was evaluated for cytotoxicity in lung adenocarcinoma (H441) cells and found to be comparable in cell killing efficiency to that of Ara-C. Uptake of (99m)Tc-HAra-C in cultures of H441 cells and sensitive pancreatic cancer cells (MIAPaCa-2) was inhibited by nucleoside transporter inhibitor nitrobenzylthioinosine. The results suggest that (99m)Tc-labeled HAra-C is a substrate for the membrane nucleoside transporters, and that it may be used in molecular imaging of nucleoside transporter expression for the verification of potential anticancer efficacy of nucleoside drugs, such as Ara-C and gemcitabine.


Subject(s)
Cytarabine/analogs & derivatives , Cytarabine/chemistry , Organotechnetium Compounds/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Cell Line, Tumor , Cytarabine/chemical synthesis , Cytarabine/toxicity , Humans , Isotope Labeling , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/toxicity , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/toxicity , Technetium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...