Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Oncotarget ; 15: 175-198, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38484151

ABSTRACT

Our understanding of DNA G-quadruplexes (G4s) from in vitro studies has been complemented by genome-wide G4 landscapes from cultured cells. Conventionally, the formation of G4s is accepted to depend on G-repeats such that they form tetrads. However, genome-wide G4s characterized through high-throughput sequencing suggest that these structures form at a large number of regions with no such canonical G4-forming signatures. Many G4-binding proteins have been described with no evidence for any protein that binds to and stabilizes G4s. It remains unknown what fraction of G4s formed in human cells are protein-bound. The G4-chromatin immunoprecipitation (G4-ChIP) method hitherto employed to describe G4 landscapes preferentially reports G4s that get crosslinked to proteins in their proximity. Our current understanding of the G4 landscape is biased against representation of G4s which escape crosslinking as they are not stabilized by protein-binding and presumably transient. We report a protocol that captures G4s from the cells efficiently without any bias as well as eliminates the detection of G4s formed artifactually on crosslinked sheared chromatin post-fixation. We discover that G4s form sparingly at SINEs. An application of this method shows that depletion of a repeat-binding protein CGGBP1 enhances net G4 capture at CGGBP1-dependent CTCF-binding sites and regions of sharp interstrand G/C-skew transitions. Thus, we present an improved method for G4 landscape determination and by applying it we show that sequence property-specific constraints of the nuclear environment mitigate G4 formation.


Subject(s)
G-Quadruplexes , Humans , Chromatin , Genome , Antibodies , Protein Binding , DNA-Binding Proteins/genetics
2.
Nat Commun ; 14(1): 5313, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37658059

ABSTRACT

Transposable elements (TE) are repetitive genomic elements that harbor binding sites for human transcription factors (TF). A regulatory role for TEs has been suggested in embryonal development and diseases such as cancer but systematic investigation of their functions has been limited by their widespread silencing in the genome. Here, we utilize unbiased massively parallel reporter assay data using a whole human genome library to identify TEs with functional enhancer activity in two human cancer types of endodermal lineage, colorectal and liver cancers. We show that the identified TE enhancers are characterized by genomic features associated with active enhancers, such as epigenetic marks and TF binding. Importantly, we identify distinct TE subfamilies that function as tissue-specific enhancers, namely MER11- and LTR12-elements in colon and liver cancers, respectively. These elements are bound by distinct TFs in each cell type, and they have predicted associations to differentially expressed genes. In conclusion, these data demonstrate how different cancer types can utilize distinct TEs as tissue-specific enhancers, paving the way for comprehensive understanding of the role of TEs as bona fide enhancers in the cancer genomes.


Subject(s)
DNA Transposable Elements , Liver Neoplasms , Humans , DNA Transposable Elements/genetics , Liver Neoplasms/genetics , Regulatory Sequences, Nucleic Acid , Binding Sites , Biological Assay , Transcription Factors/genetics
3.
JAMA Netw Open ; 5(4): e226417, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35389497

ABSTRACT

Importance: Standard diabetic ketoacidosis care in the US includes intravenous insulin treatment in the intensive care unit. Subcutaneous (SQ) insulin could decrease intensive care unit need, but the data are limited. Objective: To assess outcomes after implementation of an SQ insulin protocol for treating diabetic ketoacidosis. Design, Setting, and Participants: This cohort study is a retrospective evaluation of a prospectively implemented SQ insulin protocol. The study was conducted at an integrated health care system in Northern California. Participants included hospitalized patients with diabetic ketoacidosis at 21 hospitals between January 1, 2010, and December 31, 2019. The preimplementation phase was 2010 to 2015, and the postimplementation phase was 2017 to 2019. Data analysis was performed from October 2020 to January 2022. Exposure: An SQ insulin treatment protocol for diabetic ketoacidosis. Main Outcomes and Measures: Difference-in-differences evaluation of the need for intensive care, mortality, readmission, and length of stay at a single intervention site using an SQ insulin protocol from 2017 onward compared with 20 control hospitals using standard care. Results: A total of 7989 hospitalizations for diabetic ketoacidosis occurred, with 4739 (59.3%) occurring before and 3250 (40.7%) occurring after implementation. The overall mean (SD) age was 42.3 (17.7) years, with 4137 hospitalizations (51.8%) occurring among female patients. Before implementation, SQ insulin was the first insulin used in 40 intervention (13.4%) and 651 control (14.7%) hospitalizations. After implementation, 98 hospitalizations (80.3%) received SQ insulin first at the intervention site compared with 402 hospitalizations (12.8%) at control sites. The adjusted rate ratio for intensive care unit admission was 0.43 (95% CI, 0.33-0.56) at the intervention sites, a 57% reduction compared with control sites, and was 0.50 (95% CI, 0.25-0.99) for 30-day hospital readmission, a 50% reduction. There were no significant changes in hospital length of stay and rates of death. Conclusions and Relevance: These findings suggest that a protocol based on SQ insulin for diabetic ketoacidosis treatment was associated with significant decreases in intensive care unit need and readmission, with no evidence of increases in adverse events.


Subject(s)
Diabetes Mellitus , Diabetic Ketoacidosis , Adult , Cohort Studies , Diabetic Ketoacidosis/drug therapy , Diabetic Ketoacidosis/epidemiology , Female , Hospitals , Humans , Insulin/therapeutic use , Insulin, Regular, Human , Length of Stay , Retrospective Studies
4.
Immunopharmacol Immunotoxicol ; 44(3): 316-325, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35225131

ABSTRACT

BACKGROUND: During Aspergillus fumigatus mediated lung inflammation, NLRP3 inflammasome is rapidly activated that aggravates IL-1ß production contributing to lung inflammation. Previously, we have shown the protective role of SYK-1 inhibition in inhibiting inflammasome activation during lung inflammation. In the current manuscript, we explored the protective role of direct caspase-1 inhibition during ß-glucan-induced lung inflammation. METHODS: We have mimicked the lung inflammation by administering intranasal ß-glucan in mice model. YVAD was used for caspase-1 inhibition. RESULTS: We have shown that caspase-1 inhibition by YVAD did not alter inflammasome independent inflammatory cytokines, while it significantly reduced inflammasome activation and IL-1ß secretion. Caspase-1 inhibited bone marrow derived dendritic cells (BMDCs), co-cultured with T cells showed decreased T-cell proliferation and direct them to secrete high TGF-ß and IL-10 compared to the T cells co-cultured with ß-glucan primed dendritic cells. Caspase-1 inhibition in BMDCs also induced IL-22 secretion from CD4+T cells. Caspase-1 inhibition in intranasal ß-glucan administered mice showed decreased tissue damage, immune cell infiltration and IgA secretion compared to control mice. Further, splenocytes challenged with ß-glucan show high IL-10 secretion and increased FOXp3 and Ahr indicating an increase in regulatory T cells on caspase-1 inhibition. CONCLUSION: Caspase-1 inhibition can thus be an attractive target to prevent inflammation mediated tissue damage during Aspergillus fumigatus mouse model and can be explored as an attractive therapeutic strategy.HIGHLIGHTSCaspase-1 inhibition protects lung damage from inflammation during ß-glucan exposureCaspase-1 inhibition in dendritic cells decreases IL-1ß production resulting in decreased pathogenic Th17Caspase-1 inhibition promotes regulatory T cells thereby inhibiting lung inflammation.


Subject(s)
Pneumonia , beta-Glucans , Animals , Caspase 1 , Inflammasomes , Inflammation/chemically induced , Inflammation/drug therapy , Interleukin-10 , Interleukin-17 , Interleukin-1beta , Interleukins , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , beta-Glucans/pharmacology , Interleukin-22
5.
Cureus ; 14(1): e21535, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35106259

ABSTRACT

Background Effective treatment for COVID-19 infection is still under evaluation. Remdesivir is an approved drug for COVID-19 treatment and major countries have released guidelines on the use of remdesivir. Still, many factors are under evaluation which can determine the future use of remdesivir. Aim To study the clinical outcome and healthcare modalities of COVID-19 patients treated with remdesivir. Materials and methods A retrospective study was conducted through the clinical records of patients admitted to the tertiary care hospital between August 2020 and December 2020. All the patients who were administered remdesivir intravenously as per standard protocol were included in the study. Data were analyzed for statistical association between health care modalities and patient characteristics. Results Among 166 patients included, the mean age of patient who received remdesivir was 57.51 ± 12.98 years (95% confidence interval (CI), 30-84). The mean duration of stay and duration of oxygen requirement were 12.80 ± 5.99 (95% CI, 5-30) and 9.41 ± 7.47 (95% CI, 0-39) days, respectively. A total of 12 (7.23%) required assisted ventilation and the cure rate was 89.76% (149/166). Out of 166 patients, 105 (63.25%) had comorbidities, among which hypertension and diabetes were the most common. Significantly >60 year age group had a higher duration of oxygen requirement (u=2,639.5, p=0.01), while ≤60 year age group had a higher cure rate (X2=4.23, p=0.03) and a higher requirement for assisted ventilation (X2=4.77, p=0.02). Differences and associations in the above-mentioned health care modalities were not statistically significant for gender and comorbidity except that non-comorbid had a higher cure rate (X2=3.97, p=0.04). The odds ratio of comorbidity and cure rate was 1.07, while the association of the number of comorbidities with the duration of stay (p=0.62) and duration of oxygen requirement (p=0.35) was not statistically significant. Conclusion In remdesivir-treated patients, age affects utilization of health care modalities. Female, non-comorbid, and younger patients have better clinical outcomes.

6.
Oncotarget ; 13: 136-155, 2022.
Article in English | MEDLINE | ID: mdl-35070079

ABSTRACT

Genomes of somatic cells in culture are prone to spontaneous mutations due to errors in replication and DNA repair. Some of these errors, such as chromosomal fusions, are not rectifiable and subject to selection or elimination in growing cultures. Somatic cell cultures are thus expected to generate background levels of potentially stable chromosomal chimeras. A description of the landscape of such spontaneously generated chromosomal chimeras in cultured cells will help understand the factors affecting somatic mosaicism. Here we show that short homology-associated non-homologous chromosomal chimeras occur in normal human fibroblasts and HEK293T cells at genomic repeats. The occurrence of chromosomal chimeras is enhanced by heat stress and depletion of a repeat regulatory protein CGGBP1. We also present evidence of homologous chromosomal chimeras between allelic copies in repeat-rich DNA obtained by methylcytosine immunoprecipitation. The formation of homologous chromosomal chimeras at Alu and L1 repeats increases upon depletion of CGGBP1. Our data are derived from de novo sequencing from three different cell lines under different experimental conditions and our chromosomal chimera detection pipeline is applicable to long as well as short read sequencing platforms. These findings present significant information about the generation, sensitivity and regulation of somatic mosaicism in human cell cultures.


Subject(s)
Chimera , DNA , Cell Culture Techniques , Chromosomes , DNA-Binding Proteins , Genomics , HEK293 Cells , Humans
7.
Cell Cycle ; 20(22): 2387-2401, 2021 11.
Article in English | MEDLINE | ID: mdl-34585631

ABSTRACT

Binding sites of the chromatin regulator protein CTCF function as important landmarks in the human genome. The recently characterized CTCF-binding sites at LINE-1 repeats depend on another repeat-regulatory protein CGGBP1. These CGGBP1-dependent CTCF-binding sites serve as potential barrier elements for epigenetic marks such as H3K9me3. Such CTCF-binding sites are associated with asymmetric H3K9me3 levels as well as RNA levels in their flanks. The functions of these CGGBP1-dependent CTCF-binding sites remain unknown. By performing targeted studies on candidate CGGBP1-dependent CTCF-binding sites cloned in an SV40 promoter-enhancer episomal system we show that these regions act as inhibitors of ectopic transcription from the SV40 promoter. CGGBP1-dependent CTCF-binding sites that recapitulate their genomic function of loss of CTCF binding upon CGGBP1 depletion and H3K9me3 asymmetry in immediate flanks are also the ones that show the strongest inhibition of ectopic transcription. By performing a series of strand-specific reverse transcription PCRs we demonstrate that this ectopic transcription results in the synthesis of RNA from the SV40 promoter in a direction opposite to the downstream reporter gene in a strand-specific manner. The unleashing of the bidirectionality of the SV40 promoter activity and a breach of the transcription barrier seems to depend on depletion of CGGBP1 and loss of CTCF binding proximal to the SV40 promoter. RNA-sequencing reveals that CGGBP1-regulated CTCF-binding sites act as barriers to transcription at multiple locations genome-wide. These findings suggest a role of CGGBP1-dependent binding sites in restricting ectopic transcription.


Subject(s)
CCCTC-Binding Factor , Chromatin , DNA-Binding Proteins , Transcription Factors , Binding Sites , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genome, Human , Humans , Promoter Regions, Genetic , Transcription Factors/metabolism
8.
BMC Cancer ; 20(1): 1016, 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33081720

ABSTRACT

BACKGROUND: Inhibition of DNA-binding of proteins by small-molecule chemicals holds immense potential in manipulating the activities of DNA-binding proteins. Such a chemical inhibition of DNA-binding of proteins can be used to modulate processes such as replication, transcription, DNA repair and maintenance of epigenetic states. This prospect is currently challenged with the absence of robust and generic protocols to identify DNA-protein interactions. Additionally, much of the current approaches to designing inhibitors requires structural information of the target proteins. METHODS: We have developed a simple dot blot and immunodetection-based assay to screen chemical libraries for inhibitors of DNA-protein interactions. The assay has been applied to a library of 1685 FDA-approved chemicals to discover inhibitors of CGGBP1, a multifunctional DNA-binding protein with no known structure. Additional in vitro and in cellulo assays have been performed to verify and supplement the findings of the screen. RESULTS: Our primary screen has identified multiple inhibitors of direct or indirect interactions between CGGBP1 and genomic DNA. Of these, one inhibitor, Givinostat, was found to inhibit direct DNA-binding of CGGBP1 in the secondary screen using purified recombinant protein as the target. DNA and chromatin immunoprecipitation assays reinforced the findings of the screen that Givinostat inhibits CGGBP1-DNA binding. CONCLUSIONS: The assay we have described successfully identifies verifiable inhibitors of DNA-binding of protein; in this example, the human CGGBP1. This assay is customizable for a wide range of targets for which primary antibodies are available. It works with different sources of the target protein, cell lysates or purified recombinant preparations and does not require special equipment, DNA modifications or protein structural data. This assay is scalable and highly adaptable with the potential to discover inhibitors of transcription factors with implications in cancer biology.


Subject(s)
Carbamates/pharmacology , DNA-Binding Proteins/metabolism , DNA/metabolism , DNA-Binding Proteins/chemistry , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Immunoblotting , Protein Binding/drug effects , Small Molecule Libraries/pharmacology
9.
BMC Genet ; 21(1): 84, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32727353

ABSTRACT

BACKGROUND: The human CGGBP1 binds to GC-rich regions and interspersed repeats, maintains homeostasis of stochastic cytosine methylation and determines DNA-binding of CTCF. Interdependence between regulation of cytosine methylation and CTCF occupancy by CGGBP1 remains unknown. RESULTS: By analyzing methylated DNA-sequencing data obtained from CGGBP1-depleted cells, we report that some transcription factor-binding sites, including CTCF, resist stochastic changes in cytosine methylation. By analysing CTCF-binding sites we show that cytosine methylation changes at CTCF motifs caused by CGGBP1 depletion resist stochastic changes. These CTCF-binding sites are positioned at locations where the spread of cytosine methylation in cis depends on the levels of CGGBP1. CONCLUSION: Our findings suggest that CTCF occupancy and functions are determined by CGGBP1-regulated cytosine methylation patterns.


Subject(s)
Binding Sites , Cytosine/chemistry , DNA Methylation , DNA-Binding Proteins/genetics , Alleles , Chromosome Mapping , HEK293 Cells , Humans , Sequence Analysis, DNA , Transduction, Genetic
10.
Epigenetics Chromatin ; 12(1): 57, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31547883

ABSTRACT

BACKGROUND: CGGBP1 is a repeat-binding protein with diverse functions in the regulation of gene expression, cytosine methylation, repeat silencing and genomic integrity. CGGBP1 has also been identified as a cooperator of histone-modifying enzymes and as a component of CTCF-containing complexes that regulate the enhancer-promoter looping. CGGBP1-CTCF cross talk in chromatin regulation has been hitherto unknown. RESULTS: Here, we report that the occupancy of CTCF at repeats depends on CGGBP1. Using ChIP-sequencing for CTCF, we describe its occupancy at repetitive DNA. Our results show that endogenous level of CGGBP1 ensures CTCF occupancy preferentially on repeats over canonical CTCF motifs. By combining CTCF ChIP-sequencing results with ChIP sequencing for three different kinds of histone modifications (H3K4me3, H3K9me3 and H3K27me3), we show that the CGGBP1-dependent repeat-rich CTCF-binding sites regulate histone marks in flanking regions. CONCLUSION: CGGBP1 affects the pattern of CTCF occupancy. Our results posit CGGBP1 as a regulator of CTCF and its binding sites in interspersed repeats.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Binding Sites , CCCTC-Binding Factor/chemistry , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Histones/metabolism , Humans , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/metabolism
11.
Cancer Inform ; 18: 1176935119843835, 2019.
Article in English | MEDLINE | ID: mdl-31037026

ABSTRACT

Genome-wide occupancy of several histone modifications in various cell types has been studied using chromatin immunoprecipitation (ChIP) sequencing. Histone occupancy depends on DNA sequence features like inter-strand symmetry of base composition and periodic occurrence of TT/AT. However, whether DNA sequence motifs act as an additional effector of histone occupancy is not known. We have analyzed the presence of DNA sequence motifs in publicly available ChIP-sequence datasets for different histone modifications. Our results show that DNA sequence motifs are associated with histone occupancy, some of which are different between primary and transformed cells. The motifs for primary and transformed cells showed different levels of GC-richness and proximity to transcription start sites (TSSs). The TSSs associated with transformed or primary cell-specific motifs showed different levels of TSS flank transcription in primary and transformed cells. Interestingly, TSSs with a motif-linked occupancy of H2AFZ, a component of positioned nucleosomes, showed a distinct pattern of RNA Polymerase II (POLR2A) occupancy and TSS flank transcription in primary and transformed cells. These results indicate that DNA sequence features dictate differential histone occupancy in primary and transformed cells, and the DNA sequence motifs affect transcription through regulation of histone occupancy.

12.
Perm J ; 232019.
Article in English | MEDLINE | ID: mdl-30939262

ABSTRACT

BACKGROUND: Optimizing preoperative anemia is a required component of the Joint Commission Patient Blood Management Certification and an important component of Enhanced Recovery After Surgery. OBJECTIVE: To describe a preoperative anemia protocol developed and implemented at the Kaiser Permanente San Jose Medical Center in California to facilitate preoperative identification and treatment of anemia. METHODS: The protocol included all operations at risk of causing substantial blood loss. It excluded emergent operations and those for which the patient had a normal last hemoglobin value within the prior 12 months unless newly developed anemia was suspected. Eligible patients were screened for laboratory evaluation, and those with anemia were treated for reversible causes. Consistency was ensured by physician, staff, and patient education, and by use of electronic health records. Administration of intravenous iron and erythropoietin and consultation with specialists were expedited as part of a management algorithm. RESULTS: Among 510 patients enrolled during 1 year, 442 (87%) received anemia screening laboratory tests. Half of those with laboratory results were eligible for further optimization: 207 had anemia and 21 had iron deficiency without anemia. Among the 228 patients eligible for optimization, 189 (83%) had anemia addressed preoperatively. Of 129 patients with iron deficiency anemia, 102 (79%) received intravenous iron preoperatively, with a mean preoperative increase in hemoglobin level by 0.98 g/dL (n = 79). CONCLUSION: Integration of specialty services, optimization of technology, and consistency across practitioners were crucial for successful implementation and sustainability of a preoperative anemia protocol developed to expedite and enhance best practices.


Subject(s)
Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/therapy , Delivery of Health Care, Integrated/methods , Erythropoietin/therapeutic use , Iron/therapeutic use , Preoperative Care/methods , Anemia, Iron-Deficiency/blood , Erythropoietin/blood , Feasibility Studies , Female , Humans , Iron/blood , Male , Middle Aged , Trace Elements/therapeutic use
13.
BMC Res Notes ; 11(1): 419, 2018 Jul 02.
Article in English | MEDLINE | ID: mdl-29966527

ABSTRACT

OBJECTIVES: Although CpG methylation is well studied, mechanisms of non-CpG methylation in mammals remains elusive. Studying proteins with non-CpG cytosine methylation-sensitive DNA-binding, such as human CGGBP1, can unveil cytosine methylation regulatory mechanisms. Here we have resequenced a published genome-wide bisulfite sequencing library and analyzed it at base level resolution. CpG, CHG and CHH (where H is any nucleotide other than G) methylation states in non-targeting or CGGBP1-targeting shmiR lentivirus-transduced cells have been analyzed to identify how CGGBP1 regulates CpG and non-CpG methylation. RESULTS: We report that CGGBP1 acts as a dynamic bimodal balancer of methylation. Both gain and loss of methylation observed upon CGGBP1 depletion were spatially overlapping at annotated functional regions and not identifiable with any sequence motifs but clearly associated with GC-skew. CGGBP1 depletion caused clustered methylation changes in cis, upstream of R-loop forming promoters. This was complemented by clustered occurrences of methylation changes in proximity of transcription start sites of known cytosine methylation regulatory genes, altered expression of which can regulate cytosine methylation in trans. Despite low coverage, our data provide reliable estimates of the spectrum of methylation changes regulated by CGGBP1 in all cytosine contexts genome-wide through a combination of cis and trans-acting mechanisms.


Subject(s)
CpG Islands , DNA Methylation , DNA-Binding Proteins/genetics , Cytosine , Genome , Humans , Loss of Function Mutation , Promoter Regions, Genetic , Sequence Analysis, DNA
14.
Int Immunopharmacol ; 54: 375-384, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29202301

ABSTRACT

Repeated exposure to the fungal pathogen Aspergillus fumigates triggers spleen tyrosine kinase (SYK) signalling through dectin-1 activation, which is associated with deleterious airway inflammation. ß-Glucan-induced dectin-1 signalling activates the NLRP3 inflammasome, which in turn rapidly produces IL-1ß, a master regulator of inflammation. IL-1ß expression results in Th17/Treg imbalance, pulmonary inflammation, and bystander tissue injury. This study reports that 3,4 methylenedioxy-ß-nitrostyrene (MNS), a potent SYK inhibitor, markedly decreased the expression of pro-inflammatory cytokines and increased the expression of anti-inflammatory cytokines in vitro. Furthermore, SYK inhibition markedly decreased ß-glucan-induced IL-1ß expression, suggesting that SYK is indispensable for NLRP3 inflammasome activation. Decreased IL-1ß expression correlated with reduced Th17 response and enhanced immunosuppressive Treg response. Notably, SYK inhibition ameliorated inflammation caused by repeated intranasal ß-glucan challenge in BALB/C mice. SYK inhibition also restored the Th17/Treg balance via decreased Th17 and increased Treg responses, as evidenced by decreased IL-17 and ror-γ levels. Additionally, inhibition of SYK increased IL-10 secreting CD4+FOXP3+ T cells that accompanied reduced T cell proliferation. Decreased IgA in the Bronchoalveolar lavage (BAL) fluid and serum also indicated the immunosuppressive potential of SYK inhibition. Histopathology data revealed that repeated ß-glucan challenge caused substantial pulmonary damage, as indicated by septal thickening and interstitial lymphocytic, neutrophil and granulocyte recruitment. These processes were effectively prevented by SYK inhibition, resulting in lung protection. Collectively, our findings suggest that SYK inhibition ameliorates dectin-1- mediated detrimental pulmonary inflammation and subsequent tissue damage. Therefore, SYK can be a new target gene in the therapeutic approach against fungal induced airway inflammation.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Lung/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Spleen/metabolism , Syk Kinase/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Dioxolanes/pharmacology , Immunosuppression Therapy , Inflammasomes/metabolism , Lectins, C-Type/metabolism , Lung/microbiology , Mice , Mice, Inbred BALB C , Signal Transduction , beta-Glucans/immunology
15.
Cell Mol Neurobiol ; 37(2): 351-359, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26961545

ABSTRACT

During acute brain injury and/or sterile inflammation, release of danger-associated molecular patterns (DAMPs) activates pattern recognition receptors (PRRs). Microglial toll-like receptor (TLR)-4 activated by DAMPs potentiates neuroinflammation through inflammasome-induced IL-1ß and pathogenic Th17 polarization which critically influences brain injury. TLR4 activation accompanies increased CD40, a cognate costimulatory molecule, involved in microglia-mediated immune responses in the brain. During brain injury, excessive release of extracellular ATP (DAMPs) is involved in promoting the damage. However, the regulatory role of CD40 in microglia during ATP-TLR4-mediated inflammasome activation has never been explored. We report that CD40, in the absence of ATP, synergizes TLR4-induced proinflammatory cytokines but not IL-1ß, suggesting that the response is independent of inflammasome. The presence of ATP during TLR4 activation leads to NLRP3 inflammasome activation and caspase-1-mediated IL-1ß secretion which was inhibited during CD40 activation, accompanied with inhibition of ERK1/2 and reactive oxygen species (ROS), and elevation in p38 MAPK phosphorylation. Experiments using selective inhibitors prove indispensability of ERK 1/2 and ROS for inflammasome activation. The ATP-TLR4-primed macrophages polarize the immune response toward pathogenic Th17 cells, whereas CD40 activation mediates Th1 response. Exogenous supplementation of IFN-γ (a Th1 cytokine and CD40 inducer) results in decreased IL-1ß, suggesting possible feedback loop mechanism of inflammasome inhibition, whereby IFN-γ-mediated increase in CD40 expression and activation suppress neurotoxic inflammasome activation required for Th17 response. Collectively, the findings indicate that CD40 is a novel negative regulator of ATP-TLR4-mediated inflammasome activation in microglia, thus providing a checkpoint to regulate excessive inflammasome activation and Th17 response during DAMP-mediated brain injury.


Subject(s)
Adenosine Triphosphate/pharmacology , CD40 Antigens/pharmacology , Inflammasomes/metabolism , Microglia/metabolism , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Microglia/drug effects
18.
Acta Pol Pharm ; 69(5): 879-91, 2012.
Article in English | MEDLINE | ID: mdl-23061284

ABSTRACT

A series of 1,3,5-triazine derivatives that contain aniline, 4-hydroxycoumarin and 7-hydroxy-4-methylcoumarin and different piperazines and piperidines as substituents on the carbon atoms of the triazine ring has been synthesized by a simple and efficient synthetic protocol. Comparative studies were performed on above compounds, which were synthesized with conventional and microwave heating method. The microwave method was observed to be more beneficial as it provides an increase of yield and 90-95% reduction time. The antimicrobial activity of the compounds was tested against four bacteria (Staphylococcus aureus MTCC 96, B. subtilis MTCC 441, Escherichia coli MTCC 739, Pseudomonas aeruginosa MTCC 741) and two fungi (Aspergillus niger MTCC 282, Candida albicans MTCC 183). The preliminary in vitro evaluation studies revealed that some of the compounds have promising antimicrobial activities.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Coumarins/chemistry , Piperazines/chemical synthesis , Piperidines/chemical synthesis , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Coumarins/pharmacology , Fungi/drug effects , Microwaves , Piperazines/pharmacology , Piperidines/pharmacology
19.
Z Naturforsch C J Biosci ; 67(3-4): 108-22, 2012.
Article in English | MEDLINE | ID: mdl-22624326

ABSTRACT

A novel series of thiazolidinone derivatives, namely 4-{4-dimethylamino-6-[4-oxo-2-phenyl-5-(4-pyridin-2-yl-piperazin-1-ylmethyl)-thiazolidin-3-yl]-[1,3,5]-triazin-2-yloxy}-1-methyl-1H-quinolin-2-ones, have been synthesized from the key intermediate 4-(4-amino-6-dimethylamino-[1,3,5]-triazin-2-yloxy)-1-methyl-1H-quinolin-2-one (5). Compound 5 was condensed with various aldehydes to give Schiff base derivatives, which after cyclization gave thiazolidinones that were linked with 1-pyridin-2-yl-piperazine to obtain the target compounds. The newly synthesized compounds were evaluated for their antimicrobial activity against eight bacteria (Staphylococcus aureus, Bacillus cereus, Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Salmonella typhi, Proteus vulgaris, Shigella flexneri) and four fungi (Aspergillus niger, Candida albicans, Aspergillus fumigatus, Aspergillus clavatus).


Subject(s)
Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/pharmacology , Thiazolidines/chemical synthesis , Thiazolidines/pharmacology , Triazines/chemistry , Anti-Infective Agents/chemistry , Bacteria/classification , Bacteria/drug effects , Fungi/classification , Fungi/drug effects , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Spectrophotometry, Infrared , Thiazolidines/chemistry
20.
Eur J Med Chem ; 48: 354-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22182927

ABSTRACT

A novel series of thiazolidinone derivatives namely 4-(4-dimethylamino-6-{4-[5-(4-ethylpiperazin-1-ylmethyl)-4-oxo-2-phenylthiazolidin-3-yl]-phenylamino}-[1,3,5]triazin-2-yloxy)-1-methyl-1H-quinolin-2-one have been synthesized from the key intermediate 4-[4-(4-aminophenylamino)-6-dimethylamino-[1,3,5]triazin-2-yloxy]-1-methyl-1H-quinolin-2-one (7). Condensation reaction of compound 7 with different aldehyde derivatives were performed to obtain Schiff base derivatives, which after cyclization gave thiazolidinones and finally they were reacted with N-ethylpiperazine to get target compounds. The newly synthesized compounds were evaluated for their antimicrobial activity against eight bacterial strains (Staphylococcus aureus, Bacillus cereus, Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Salmonella typhi, Proteus vulgaris, Shigella flexneri) and four fungal strains (Aspergillus niger, Candida albicans, Aspergillus fumigatus, Aspergillus clavatus).


Subject(s)
Anti-Infective Agents/chemical synthesis , Thiazolidines/chemical synthesis , Thiazolidines/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Bacteria/drug effects , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Spectroscopy, Fourier Transform Infrared , Structure-Activity Relationship , Thiazolidines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...