Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
PeerJ ; 11: e16481, 2023.
Article in English | MEDLINE | ID: mdl-38077444

ABSTRACT

Background: Exosomes, microvesicles, carry and release several vital molecules across cells, tissues, and organs. Epicardial adipose tissue exosomes are critical in the development and progression of coronary artery disease (CAD). It is hypothesized that exosomes may transport causative molecules from inflamed tissue and deliver to the target tissue and progress CAD. Thus, identifying and inhibiting the CAD-associated proteins that are being transported to other cells via exosomes will help slow the progression of CAD. Methods: This study uses a systems biological approach that integrates differential gene expression in the CAD, exosomal cargo assessment, protein network construction, and functional enrichment to identify the crucial exosomal cargo protein target. Meanwhile, absorption, distribution, metabolism, and excretion (ADME) screening of Panax ginseng-derived compounds was conducted and then docked against the protein target to identify potential inhibitors and then subjected to molecular dynamics simulation (MDS) to understand the behavior of the protein-ligand complex till 100 nanoseconds. Finally, density functional theory (DFT) calculation was performed on the ligand with the highest affinity with the target. Results: Through the systems biological approach, Mothers against decapentaplegic homolog 2 protein (SMAD2) was determined as a potential target that linked with PI3K-Akt signaling, Ubiquitin mediated proteolysis, and the focal adhesion pathway. Further, screening of 190 Panax ginseng compounds, 27 showed drug-likeness properties. Inermin, a phytochemical showed good docking with -5.02 kcal/mol and achieved stability confirmation with SMAD2 based on MDS when compared to the known CAD drugs. Additionally, DFT analysis of inermin showed high chemical activity that significantly contributes to effective target binding. Overall, our computational study suggests that inermin could act against SMAD2 and may aid in the management of CAD.


Subject(s)
Coronary Artery Disease , Panax , Molecular Dynamics Simulation , Ligands , Phosphatidylinositol 3-Kinases
2.
Molecules ; 28(19)2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37836798

ABSTRACT

Contact lens-mediated microbial keratitis caused by Pseudomonas aeruginosa and Streptococcus pneumoniae provokes corneal damage and vision loss. Recently, natural phytochemicals have become complementary medicines for corneal destruction. Herein, we aimed to identify multi-targeting Aloe vera-derived phytochemicals capable of inhibiting bacterial and host targets of keratitis through ADME (absorption, distribution, metabolism, and excretion), docking, molecular dynamics (MD) simulation, MMGBSA (molecular mechanics generalized Born surface area) and density functional theory (DFT) investigations. An extensive literature search revealed ExoU, ExoS, ExoT, ExoY, and PLY as virulent bacterial targets. Simultaneously, differential gene expression (DGE) and pathway enrichment analysis-specified host transcription factor (SPI1) influences keratitis pathogenesis. Molecular docking analysis uncovered aloeresin-A as a promising inhibitor against bacterial and host targets, demonstrating strong binding energies ranging from -7.59 to -6.20 kcal/mol. Further, MMGBSA and MD simulation analysis reflect higher binding free energies and stable interactions of aloeresin-A with the targets. In addition, DFT studies reveal the chemical reactiveness of aloeresin-A through quantum chemical calculations. Hence, our findings show aloeresin-A to be a promising candidate for effectively inhibiting keratitis. However, additional research is imperative for potential integration into lens care solutions.


Subject(s)
Contact Lenses , Keratitis , Humans , Molecular Docking Simulation , Multiomics , Keratitis/microbiology , Contact Lenses/adverse effects , Transcription Factors/metabolism , Pseudomonas aeruginosa
3.
Pharmacol Ther ; 250: 108522, 2023 10.
Article in English | MEDLINE | ID: mdl-37661054

ABSTRACT

Metastatic progression combined with non-responsiveness towards systemic therapy often shapes the course of disease for cancer patients and commonly determines its lethal outcome. The complex molecular events that promote metastasis are a combination of both, the acquired pro-metastatic properties of cancer cells and a metastasis-permissive or -supportive tumor micro-environment (TME). Yet, dissemination is a challenging process for cancer cells that requires a series of events to enable cancer cell survival and growth. Metastatic cancer cells have to initially detach themselves from primary tumors, overcome the challenges of their intravasal journey and colonize distant sites that are suited for their metastases. The implicated obstacles including anoikis and immune surveillance, can be overcome by intricate intra- and extracellular signaling pathways, which we will summarize and discuss in this review. Further, emerging modulators of metastasis, like the immune-microenvironment, microbiome, sublethal cell death engagement, or the nervous system will be integrated into the existing working model of metastasis.


Subject(s)
Neoplasms , Humans , Neoplasms/metabolism , Signal Transduction , Anoikis , Neoplasm Metastasis , Epithelial-Mesenchymal Transition , Tumor Microenvironment
5.
Sci Rep ; 12(1): 5052, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35322026

ABSTRACT

Lysosomal biogenesis plays a vital role in cell fate. Under certain conditions, excessive lysosomal biogenesis leads to susceptibility for lysosomal membrane permeabilization resulting in various pathological conditions including cell death. In cancer cells apoptosis machinery becomes dysregulated during the course of treatment, thus allows cancer cells to escape apoptosis. So it is therefore imperative to identify cytotoxic agents that exploit non-apoptotic mechanisms of cell death. Our study showed that pancreatic cancer cells treated with SDS-203 triggered an incomplete autophagic response and a nuclear translocation of transcriptional factor TFEB. This resulted in abundant biosynthesis and accumulation of autophagosomes and lysosomes into the cells leading to their death. It was observed that the silencing of autophagy genes didn't alter the cell fate, whereas siRNA-mediated silencing of TFEB subdued SDS-203 mediated lysosomal biogenesis and associated cell death. Further mouse tumors treated with SDS-203 showed a significant reduction in tumor burden and increased expression of lysosomal markers. Taken together this study demonstrates that SDS-203 treatment triggers non-apoptotic cell death in pancreatic cancer cells through a mechanism of lysosome over accumulation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Pancreatic Neoplasms , Animals , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Death , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism
6.
Autophagy ; 17(12): 4502-4503, 2021 12.
Article in English | MEDLINE | ID: mdl-34632910

ABSTRACT

VCP (valosin containing protein), a member of the AAA+ protein family, is critical for many cellular processes and functions. Dominant VCP mutations cause a rare neurodegenerative disease known as multisystem proteinopathy (MSP). The spectrum of mechanisms causing fronto-temporal dementia with TARDBP/TDP-43 inclusions (FTLD-TARDBP) by VCP disease mutations remains unclear. Our recent work identified VCP activity as a mediator of FTLD-TARDBP. Specifically, brain atrophy, behavioral changes, neuronal loss, gliosis, and TARDBP pathology were observed in vcp conditional knockout (cKO) mice. We also found that autophago-lysosomal dysfunction, TARDBP inclusions, and ubiquitin-proteasome impairment precede neuronal loss. We further studied conditional expression of the disease-associated mutation VCPR155C in vcp-null mice. We observed features similar to those of VCP inactivation, suggesting that VCP mutation is hypomorphic. Furthermore, proteomic, and transcriptomic signatures in vcp cKO mice resemble those of GRN/Progranulin carriers. Therefore, VCP is essential for neuronal survival by several mechanisms and could be a therapeutic target aimed at restoring protein homeostasis in patients with FTLD-TARDBP.


Subject(s)
Frontotemporal Lobar Degeneration , Neurodegenerative Diseases , Valosin Containing Protein , Animals , Autophagy/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Humans , Lysosomes/metabolism , Mice , Mutation/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Proteomics , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
7.
Mol Carcinog ; 60(10): 671-683, 2021 10.
Article in English | MEDLINE | ID: mdl-34324743

ABSTRACT

Rohitukine, a chromone alkaloid extracted from Dysoxylum binectariferum, has a propitious anticancer activity. Our previous study shows that a new Rohitukine derivative IIIM-290 restricts the growth of pancreatic cancer in vivo and in vitro. In the present findings, we report the mechanism of cell death induced by IIIM-290 in MOLT-4 cells (acute lymphoblastic leukemia) and its anticancer potential against various murine leukemic tumor models in vivo. We found that IIIM-290 induced apoptosis through upregulation of different apoptotic proteins like PUMA, BAX, cytochrome c, cleaved (active) caspase-3, and cleaved PARP in MOLT-4 cells. Moreover, IIIM-290 abated mitochondrial membrane potential, elevated calcium levels, reactive oxygen species, and arrested growth of MOLT-4 cells in the synthesis (S) phase of the cell cycle. Interestingly, the elevation in proapoptotic markers was p53 dependent-the silencing of p53 abrogated apoptosis (programmed cell death) triggered by IIIM-290 in MOLT-4 cells. Furthermore, IIIM-290 significantly enhanced the survival of animals with P388 and L1210 leukemia. Thus, our results put IIIM-290 as a potential candidate for the anticancer lead.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Chromones/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Piperidines/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Chromones/chemistry , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , Piperidines/chemistry , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
8.
Front Pharmacol ; 12: 695712, 2021.
Article in English | MEDLINE | ID: mdl-34248643

ABSTRACT

Aberrant activation of NLRP3 inflammasome has been implicated in several inflammatory diseases. Autophagy is one of the primary mechanisms that regulate NLRP3 inflammasome activity. In this study, we attempted to target NLRP3 inflammasome activity by a synthetic compound IIIM-941. We found that IIIM-941 inhibits ATP induced NLRP3 inflammasome by induction of autophagy through AMPK pathway in bone marrow derived macrophages (BMDMs) and J774A.1 cells. It was interesting to observe that IIIM-941 did not show any inhibitory activity against LPS induced pro-inflammatory cytokines TNF-α and IL-6. The anti-NLRP3 activity of IIIM-941 was significantly reversed when we attempted to block autophagy by using either pharmacological inhibitor bafilomycin A1or by using siRNA against AMPK. Further, we found that IIIM-941 downregulated the expression of NLRP3 and prevented the oligomerization of ASC to exert its anti-NLRP3 inflammasome effect in J774A.1 cells. We validated inhibitory activity of IIIM-941 against NLRP3 in three different mice models. The anti-inflammatory effect of IIIM-941 was highly significant in ATP induced peritoneal inflammation model. IIIM-941 was similarly effective in suppressing MSU induced IL-1ß in the air pouch model of inflammation without affecting the levels of TNF-α and IL-6. Finally, oral efficacy of IIIM-941 was also proved in MSU indued foot paw edema model of inflammation in mice at 10 and 20 mg/kg (b.w.). The compounds like IIIM-941 can be explored further for the development of therapies against diseases such as Alzheimer's disease and Parkinson's disease, where hampered autophagy and NLRP3 activation play a crucial role in the pathological development.

9.
Cell Rep ; 36(3): 109399, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289347

ABSTRACT

The pathogenic mechanism by which dominant mutations in VCP cause multisystem proteinopathy (MSP), a rare neurodegenerative disease that presents as fronto-temporal lobar degeneration with TDP-43 inclusions (FTLD-TDP), remains unclear. To explore this, we inactivate VCP in murine postnatal forebrain neurons (VCP conditional knockout [cKO]). VCP cKO mice have cortical brain atrophy, neuronal loss, autophago-lysosomal dysfunction, and TDP-43 inclusions resembling FTLD-TDP pathology. Conditional expression of a single disease-associated mutation, VCP-R155C, in a VCP null background similarly recapitulates features of VCP inactivation and FTLD-TDP, suggesting that this MSP mutation is hypomorphic. Comparison of transcriptomic and proteomic datasets from genetically defined patients with FTLD-TDP reveal that progranulin deficiency and VCP insufficiency result in similar profiles. These data identify a loss of VCP-dependent functions as a mediator of FTLD-TDP and reveal an unexpected biochemical similarity with progranulin deficiency.


Subject(s)
DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/pathology , Neurons/metabolism , Valosin Containing Protein/metabolism , Aged , Alleles , Animals , Atrophy , Autophagosomes/metabolism , Behavior, Animal , Brain/pathology , Frontotemporal Lobar Degeneration/genetics , Gliosis/pathology , Humans , Lysosomes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Nerve Degeneration/pathology , Neurons/pathology , Proteomics , Transcriptome/genetics
10.
Toxicol Appl Pharmacol ; 423: 115582, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34019860

ABSTRACT

NLRP3 inflammasome is involved in several chronic inflammatory diseases. The inflammatory effect of the NLRP3 inflammasome is executed through IL-1ß and IL-18. Therefore, IL-1ß is one of the primary targets in chronic inflammatory conditions. However, current treatment regimens are dependent on anti- IL-1ß biologicals. The therapies targeting IL-1ß through inhibition of NLRP3 inflammasome are thus being actively explored. We identified safranal, a small molecule responsible for the essence of saffron as a potential inhibitor of the NLRP3 inflammasome. Safranal significantly suppressed the release of IL-1ß from ATP stimulated J774A.1 and bone marrow-derived macrophages (BMDMs) by regulating CASP1 and CASP8 dependent cleavage of pro-IL-1ß. Safranal markedly suppressed the expression of NLRP3 and its ATPase activity. Safranal treatment enhanced the expression of NRF2, whereas, si-RNA mediated silencing of Nrf2 abrogated the anti-NLRP3 effect of safranal. Furthermore, safranal inhibited ASC oligomerization and formation of ASC specks. Safranal also displayed anti-NLRP3 activity in multiple mice models. Treatment of animals with safranal reduced the production of IL-1ß in ATP elicited peritoneal inflammation, MSU induced air pouch inflammation, and MSU injected foot paw edema in mice. Thus, our data projects safranal as a potential preclinical drug candidate against NLRP3 inflammasome triggered chronic inflammation.


Subject(s)
CARD Signaling Adaptor Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins/metabolism , Cyclohexenes/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Terpenes/pharmacology , Animals , Cell Line , Cells, Cultured , Cyclohexenes/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Terpenes/therapeutic use
11.
Autophagy ; 17(11): 3813-3832, 2021 11.
Article in English | MEDLINE | ID: mdl-33404280

ABSTRACT

Alzheimer disease (AD) is usually accompanied by two prominent pathological features, cerebral accumulation of amyloid-ß (Aß) plaques and presence of MAPT/tau neurofibrillary tangles. Dysregulated clearance of Aß largely contributes to its accumulation and plaque formation in the brain. Macroautophagy/autophagy is a lysosomal degradative process, which plays an important role in the clearance of Aß. Failure of autophagic clearance of Aß is currently acknowledged as a contributing factor to increased accumulation of Aß in AD brains. In this study, we have identified crocetin, a pharmacologically active constituent from the flower stigmas of Crocus sativus, as a potential inducer of autophagy in AD. In the cellular model, crocetin induced autophagy in N9 microglial and primary neuron cells through STK11/LKB1 (serine/threonine kinase 11)-mediated AMP-activated protein kinase (AMPK) pathway activation. Autophagy induction by crocetin significantly increased Aß clearance in N9 cells. Moreover, crocetin crossed the blood-brain barrier and induced autophagy in the brains' hippocampi of wild-type male C57BL/6 mice. Further studies in transgenic male 5XFAD mice, as a model of AD, revealed that one-month treatment with crocetin significantly reduced Aß levels and neuroinflammation in the mice brains and improved memory function by inducing autophagy that was mediated by AMPK pathway activation. Our findings support further development of crocetin as a pharmacological inducer of autophagy to prevent, slow down progression, and/or treat AD.Abbreviations: Aß: amyloid-ß; ABCB1/P-gp/P-glycoprotein: ATP-binding cassette, subfamily B (MDR/TAP), member 1; AD: Alzheimer disease; AMPK/PRKAA: AMP-activated protein kinase; APP: amyloid beta (A4) precursor protein; ATG: autophagy related; BBB: blood-brain barrier; BECN1: beclin 1, autophagy related; CAMKK2/CaMKKß: calcium/calmodulin-dependent protein kinase kinase 2, beta; CSE: Crocus sativus extract; CTSB: cathepsin B; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; GFAP: glial fibrillary acidic protein; GSK3B/GSK3ß: glycogen synthase kinase 3 beta; Kp: brain partition coefficient; LRP1: low density lipoprotein receptor-related protein 1; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MAP2: microtubule-associated protein 2; MAPK/ERK: mitogen-activated protein kinase; MAPT/tau: microtubule-associated protein tau; MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; MTOR: mechanistic target of rapamycin kinase; MWM: Morris water maze; NFKB/NF-κB: nuclear factor of kappa light polypeptide gene enhancer in B cells; NMDA: N-methyl-d-aspartic acid; RPTOR: regulatory associated protein of MTOR; RPS6KB1/p70S6K: ribosomal protein S6 kinase 1; SQSTM1: sequestosome 1; SRB: sulforhodamine B; STK11/LKB1: serine/threonine kinase 11; TFEB: transcription factor EB; TSC2: TSC complex subunit 2; ULK1: unc-51 like kinase 1.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Autophagy/drug effects , Carotenoids/pharmacology , MAP Kinase Signaling System/drug effects , Vitamin A/analogs & derivatives , Amyloid beta-Peptides/metabolism , Animals , Cell Line , Female , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Vitamin A/pharmacology
12.
Mol Neurobiol ; 57(8): 3589-3602, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32542594

ABSTRACT

Deposition of an amyloid-ß peptide is one of the first events in the pathophysiology of Alzheimer's disease (AD) and is clinically characterized by Aß plaques, tau tangles, and behavioral impairments that lead to neuronal death. A substantial number of studies encourage targeting the skewness in the production and degradation of amyloid-ß could be among the promising therapies in the disease. Neuronal autophagy has emerged for an essential role in the degradation of such toxic aggregate-prone proteins in various neurodegenerative diseases. We profiled a small library of common dietary compounds and identified those that can enhance autophagy in neuronal cells. Here we noted naringenin in silico exhibits a robust affinity with AMP-activated protein kinase (AMPK) and upregulated AMPK-mediated autophagy signaling in neurons. Naringenin can induce autophagy promoting proteins such as ULK1, Beclin1, ATG5, and ATG7 in Neuro2a cells and primary mouse neurons as well. The knockdown of AMPK by siRNA-AMPK was complemented by naringenin that restored transcript levels of AMPK. Further, naringenin can reduce the levels of Aß at a nontoxic concentration from neuronal cells. Moreover, it maintained the mitochondrial membrane potential and resisted reactive oxygen species production, which led to the protection against Aß1-42 evoked neurotoxicity. This highlights the neuroprotective potential of naringenin that can be developed as an anti-amyloidogenic nutraceutical.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Alzheimer Disease/metabolism , Autophagy/drug effects , Flavanones/pharmacology , Neurons/pathology , Amyloid beta-Peptides/metabolism , Animals , Apoptosis/drug effects , Mice , Neurons/metabolism , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects
13.
Autophagy ; 15(10): 1810-1828, 2019 10.
Article in English | MEDLINE | ID: mdl-30894052

ABSTRACT

Imbalance in production and clearance of amyloid beta (Aß) is the primary reason for its deposition in Alzheimer disease. Macroautophagy/autophagy is one of the important mechanisms for clearance of both intracellular and extracellular Aß. Here, through screening, we identified alborixin, an ionophore, as a potent inducer of autophagy. We found that autophagy induced by alborixin substantially cleared Aß in microglia and primary neuronal cells. Induction of autophagy was accompanied by up regulation of autophagy proteins BECN1/Beclin 1, ATG5, ATG7 and increased lysosomal activities. Autophagy induced by alborixin was associated with inhibition of the phosphoinositide 3-kinase (PI3K)-AKT pathway. A knock down of PTEN and consistent, constitutive activation of AKT inhibited alborixin-induced autophagy and consequent clearance of Aß. Furthermore, clearance of Aß by alborixin led to significant reduction of Aß-mediated cytotoxicity in primary neurons and differentiated N2a cells. Thus, our findings put forward alborixin as a potential anti-Alzheimer therapeutic lead. Abbreviations: Aß: amyloid beta; ALB: alborixin; ATG: autophagy-related; BECN1: beclin 1; DAPI: 4, 6-diamidino-2-phenylindole; DCFH-DA: 2,7-dichlorodihydrofluorescein diacetate; fAß: fibrillary form of amyloid beta; GFAP: glial fibrillary acidic protein; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MAP2: microtubule-associated protein 2; MTOR: mechanistic target of rapamycin kinase; PTEN: phosphatase and tensin homolog; ROS: reactive oxygen species; SQSTM1: sequestosome 1; TMRE: tetramethylrhodamine, ethyl ester.


Subject(s)
Amyloid beta-Peptides/metabolism , Autophagy/drug effects , PTEN Phosphohydrolase/physiology , Proto-Oncogene Proteins c-akt/metabolism , Amyloid beta-Peptides/drug effects , Animals , Autophagy/physiology , Cells, Cultured , Embryo, Mammalian , Humans , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/genetics , Proteolysis/drug effects , Pyrans/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
15.
Cell Mol Neurobiol ; 39(3): 387-399, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30725250

ABSTRACT

Alzheimer disease is a complex neurodegenerative disorder. It is the common form of dementia in elderly people. The etiology of this disease is multifactorial, pathologically it is accompanied with accumulation of amyloid beta and neurofibrillary tangles. Accumulation of amyloid beta and mitochondrial dysfunction leads to oxidative stress. In this study, neuroprotective effect of Artemisiaamygdalina against H2O2-induced death was studied in differentiated N2a and SH-SY5Ycells. Cells were treated with H2O2 to induce toxicity which was attenuated by Artemisia amygdalina. The nuclear factor erythroid 2-related factor 2 (Nrf2) is an emerging regulator of cellular resistance to oxidants. It controls the basal and induced expression of antioxidant response element-dependent genes. Further, we demonstrated that Artemisia amygdalina protects neurons through upregulation of Nrf2 pathway. Moreover, reactive oxygen species and mitochondrial membrane potential loss formed by H2O2 was attenuated by Artemisia amygdalina. Thus, Artemisia amygdalina may have the possibility to be a therapeutic agent for Alzheimer disease.


Subject(s)
Alzheimer Disease/drug therapy , Artemisia/chemistry , NF-E2-Related Factor 2/genetics , Neurons/pathology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Plant Extracts/therapeutic use , Up-Regulation , Animals , Biphenyl Compounds/chemistry , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Shape/drug effects , Cell Survival/drug effects , Free Radical Scavengers/chemistry , Heme Oxygenase-1/metabolism , Humans , Hydrogen Peroxide/toxicity , Hydroxyl Radical/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/genetics , Phenols/analysis , Picrates/chemistry , Plant Extracts/pharmacology , Superoxides/metabolism , Up-Regulation/drug effects
16.
Basic Clin Pharmacol Toxicol ; 124(4): 351-359, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29719125

ABSTRACT

Murrayanine (MK) is the main compound isolated from Murraya koenigii, an aromatic plant belonging to the Rutaceae family, also known as curry leaf tree. Murrayanine was reported to possess potential antioxidant, antimycobacterial and antifungal effects. However, its effect in sepsis remains unclear. This study was designed to investigate the anti-inflammatory effect of MK using both in vitro and in vivo assay. Results of this study indicated that MK decreased NO, TNF-α and IL-6 production in both lipopolysaccharide (LPS)-stimulated RAW 264.7 cells and murine peritoneal macrophages. Moreover, iNOS and COX-2 protein expression as well as their downstream product, PGE2, was also decreased effectively in RAW 264.7 cells. Furthermore, MK decreased the phosphorylation of IKB and repressed NF-kB activity in LPS-activated RAW 264.7 cells. Additionally, we evaluated MK efficacy in vivo using LPS-induced sepsis, a systemic inflammation model in mice. Administration of MK inhibits pro-inflammatory cytokines (TNF-α and IL-6) secretion; decreases AST, ALT, BUN and CRE level in mouse sera; mitigates lung, liver and kidney injuries; and also increases LPS-challenged mice survival rate. Collectively, our results suggest that MK exerts potential as a new anti-inflammatory and immunosuppressive drug in sepsis treatment.


Subject(s)
Carbazoles/pharmacology , Inflammation/prevention & control , Multiple Organ Failure/prevention & control , Sepsis/drug therapy , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Carbazoles/isolation & purification , Cytokines/metabolism , Disease Models, Animal , Female , Immunosuppressive Agents/isolation & purification , Immunosuppressive Agents/pharmacology , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred BALB C , Multiple Organ Failure/etiology , Murraya/chemistry , NF-kappa B/metabolism , RAW 264.7 Cells , Sepsis/complications
17.
Cytokine ; 111: 84-87, 2018 11.
Article in English | MEDLINE | ID: mdl-30125779

ABSTRACT

Deposition of amyloid-ß in Alzheimer's disease is accompanied by chronic inflammation, which involves raised levels of pro-inflammatory cytokines TNF-α, IL-6 and IL-1ß. However, the role of Aß1-42 in the inflammatory process, before it gets deposited into aggregates has not been investigated thoroughly. Through this study, we are illustrating the dual role of soluble Aß1-42 (sAß1-42) in activating the NLRP3 inflammasome and simultaneously inhibiting TNF-α secretion. Our data suggested that the treatment of chronically induced THP-1 macrophages and N9 microglial cells with sAß1-42 can suppress the major inflammatory cytokine TNF-α without affecting the level of IL-6. However, the activation of NLRP3 inflammasome was well evidenced by secretion of IL-1ß, increased expression of NLRP3 and caspase-1, implicating sAß1-42 in enhancing and suppressing one or other type of inflammation. Further investigation revealed that sAß1-42 was able to severely abrogate the expression of NF-κB, p50 and restricting the translocation of NF-κB, p65 to nucleus by inhibiting phosphorylation of IκB-α in THP-1 macrophages. These data indicate that the sAß1-42 may play a dual role during inflammatory process, wherein, it may be involved in protecting the cells from inflammatory damage due to TNF-α. This ability of sAß1-42 might be playing some role in protecting the brain cells during the process of aging and Alzheimer's disease, where, chronic inflammatory environment plays a vital role.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Inflammasomes/immunology , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Peptide Fragments/immunology , Tumor Necrosis Factor-alpha/immunology , Alzheimer Disease/pathology , Humans , Interleukin-1beta/immunology , Interleukin-6/immunology , Macrophages/pathology , Microglia/immunology , Microglia/pathology , NF-kappa B p50 Subunit/immunology , Signal Transduction/immunology , Solubility , THP-1 Cells , Transcription Factor RelA/immunology
18.
Cytokine ; 107: 93-104, 2018 07.
Article in English | MEDLINE | ID: mdl-29229421

ABSTRACT

Natural product derivatives have proven to be cutting edge window for drug discovery and development. BA-25 (3-α-o-acetoxy-4ß-amino-11-oxo-24-norurs-12-ene) an amino analogue of ß-boswellic acid exhibited inhibition of TNF-α and IL-6 in THP-1 cells as demonstrated previously, however, the effect on principal inflammatory mediators such as cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and the pathways that mediate this function remains unknown. This study was designed to examine the comparative anti-inflammatory activity of BA-25 with its parent compound, ß boswellic acid both in vitro and in vivo. The effect of BA and BA-25 on suppression of NO, PGE2, LTB4, COX-2 in LPS-stimulated RAW 264.7 cells was determined by ELISA, RT-PCR and ROS by flow cytometry. Phosphorylation of NF-kBp65, IKB degradation was determined by western blotting and also the nuclear localization of NF-kBp65 was assessed by immunofluorescence. Furthermore, this study was extended on Carrageenan induced paw oedema modelled BALB/c mice. A novel derivative BA-25, reported first time notably decreased the LPS (1 µg/mL) induced upregulation in the transcription of TNF-α, IL-6, iNOS and COX-2. Also the protein expression of iNOS and COX-2 as well as their downstream products NO and PGE2 respectively, were also decreased efficiently at a concentration of 10 µM than BA. Moreover, LPS upregulated NF-kB p65 expression and IκB degradation was significantly decreased after BA-25 treatment. In addition, the treatment of BA-25 also restored the paw oedema and decreased the magnitude of histopathological alterations. Our data together suggested that BA-25 might be regarded as prospective therapeutic anti-inflammatory alternative and demands further investigation in pharmacological studies.


Subject(s)
Inflammation Mediators/metabolism , Inflammation/drug therapy , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Triterpenes/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Edema/drug therapy , Edema/metabolism , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Up-Regulation/drug effects
19.
Chem Biol Interact ; 274: 35-49, 2017 Aug 25.
Article in English | MEDLINE | ID: mdl-28690100

ABSTRACT

Arginase is a therapeutic enzyme for arginine-auxotrophic cancers but their low anticancer activity, less proteolytic tolerance and shorter serum half-life are the major shortcomings. In this study, arginase from Pseudomonas aeruginosa IH2 was purified to homogeneity and estimated as 75 kDa on native-PAGE and 37 kDa on SDS-PAGE. Arginase showed optimum activity at pH 8 and temperature 35 °C. Mn2+ and Mg2+ ions enhanced arginase activity while, Li+, Cu2+, and Al3+ ions reduced arginase activity. In-vitro serum half-life of arginase was 36 h and proteolytic half-life against trypsin and proteinase-K was 25 and 29 min, respectively. Anticancer activity of arginase was evaluated against colon, breast, leukemia, and prostate cancer cell lines and lowest IC50 (0.8 IU ml-1) was found against leukemia cell line HL-60. Microscopic studies and flow cytometric analysis of Annexin V/PI staining of HL-60 cells revealed that arginase induced apoptosis in dose-dependent manner. Cell cycle analysis suggested that arginase induced cell cycle arrest in G0/G1 phase. The increasing level of MMP loss, ROS generation and decreasing level of SOD, CAT, GPx and GSH suggested that arginase treatment triggered dysfunctioning of mitochondria. The cleavage of caspase-3, PARP-1, activations of caspase-8, 9 and high expression of proapoptotic protein Bax, low expression of anti-apoptotic protein Bcl-2 indicated that arginase treatment activates mitochondrial pathway of apoptosis. Purified arginase did not exert cytotoxic effects on human noncancer cells. Our study strongly supports that arginase could be used as potent anticancer agent but further studies are required which are underway in our lab.


Subject(s)
Apoptosis/drug effects , Arginase/toxicity , Bacterial Proteins/toxicity , G1 Phase Cell Cycle Checkpoints/drug effects , Membrane Potential, Mitochondrial/drug effects , Pseudomonas aeruginosa/enzymology , Arginase/chemistry , Arginase/isolation & purification , Arginase/pharmacokinetics , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/pharmacokinetics , Caspases/metabolism , Catalase/metabolism , Cell Line, Tumor , DNA Damage/drug effects , Down-Regulation/drug effects , HL-60 Cells , Humans , Hydrogen-Ion Concentration , Microscopy, Phase-Contrast , Molecular Weight , Protein Stability , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Substrate Specificity , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein/metabolism
20.
Eur J Med Chem ; 122: 744-755, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27497733

ABSTRACT

N-(2,2,2-Trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1-(trifluoromethyl)ethyl]phenyl]-benzenesulfonamide (T0901317, 6) is a potent activator of pregnane-X-receptor (PXR), which is a nuclear receptor controlling P-gp expression. Herein, we aimed to investigate P-gp induction activity of T0901317 and establish its structure-activity relationship. T0901317 along with a series of N-triazolyl-methylene-linked benzenesulfonamides were synthesized and screened for P-gp induction activity using a rhodamine-123 based efflux assay in the P-gp overexpressing human adenocarcinoma LS-180 cells, wherein several compounds showed potent P-gp induction activity at 5 µM. Treatment with benzene sulphonamides led to the decrease in intracellular accumulation of a fluorescent P-gp substrate rhodamine-123 up to 48% (control 100%). In the western-blot studies, T0901317 (6) and its triazole linked analog 26e at 5 µM displayed induction of P-gp expression in LS180 cells. These compounds were non-toxic in LS-180 and human neuroblastoma SH-SY5Y cells (IC50 > 50 µM). The compound 26e showed significant P-gp induction even at 0.3 µM, indicating an excellent therapeutic window. These results clearly indicate promise of this class of compounds as potential agents to enhance amyloid-ß clearance in Alzheimers patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Drug Design , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/pharmacology , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chemistry Techniques, Synthetic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrocarbons, Fluorinated/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Benzenesulfonamides
SELECTION OF CITATIONS
SEARCH DETAIL
...