Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Neurol ; 333: 113430, 2020 11.
Article in English | MEDLINE | ID: mdl-32745471

ABSTRACT

High-capacity mitochondrial calcium (Ca2+) uptake by the mitochondrial Ca2+ uniporter (MCU) is strategically positioned to support the survival and remyelination of axons in multiple sclerosis (MS) by undocking mitochondria, buffering Ca2+ and elevating adenosine triphosphate (ATP) synthesis at metabolically stressed sites. Respiratory chain deficits in MS are proposed to metabolically compromise axon survival and remyelination by suppressing MCU activity. In support of this hypothesis, clinical scores, mitochondrial dysfunction, myelin loss, axon damage and inflammation were elevated while remyelination was blocked in neuronal MCU deficient (Thy1-MCU Def) mice relative to Thy1 controls subjected to experimental autoimmune encephalomyelitis (EAE). At the first sign of walking deficits, mitochondria in EAE/Thy1 axons showed signs of activation. By contrast, cytoskeletal damage, fragmented mitochondria and large autophagosomes were seen in EAE/Thy1-MCU Def axons. As EAE severity increased, EAE/Thy1 axons were filled with massively swollen mitochondria with damaged cristae while EAE/Thy1-MCU Def axons were riddled with late autophagosomes. ATP concentrations and mitochondrial gene expression were suppressed while calpain activity, autophagy-related gene mRNA levels and autophagosome marker (LC3) co-localization in Thy1-expressing neurons were elevated in the spinal cords of EAE/Thy1-MCU Def compared to EAE/Thy1 mice. These findings suggest that MCU inhibition contributes to axonal damage that drives MS progression.


Subject(s)
Calcium Channels/deficiency , Encephalomyelitis, Autoimmune, Experimental/pathology , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Myelin Sheath/pathology , Neurons/metabolism , Adenosine Triphosphate/metabolism , Animals , Autophagy/genetics , Axons/pathology , Calcium Channels/genetics , Gait Disorders, Neurologic/genetics , Gait Disorders, Neurologic/pathology , Gene Expression/genetics , Male , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Swelling , Phagosomes/pathology , Spinal Cord/pathology
2.
J Transl Med ; 11: 42, 2013 Feb 18.
Article in English | MEDLINE | ID: mdl-23414419

ABSTRACT

BACKGROUND: Tumors of the head and neck present aggressive pathological behavior in patients due to high expression of CDK/CCND1 proteins. P276-00, a novel CDK inhibitor currently being tested in clinic, inhibits growth of several cancers in vitro and in vivo. The pre clinical activity of P276-00 in head and neck cancer and its potential mechanisms of action at molecular level are the focus of the current studies. METHOD: We have investigated the anti-cancer activity of P276-00 in head and neck tumors in vitro and in vivo. Candidate gene expression profiling and cell based proteomic approaches were taken to understand the pathways affected by P276-00 treatment. RESULTS: It was observed that P276-00 is cytotoxic across various HNSCC cell lines with an IC50 ranging from 1.0-1.5 µmoles/L and culminated in significant cell-cycle arrest in G1/S phase followed by apoptosis. P276-00 treatment suppressed cell proliferation through inhibition of CCND1 expression, reduced phosphorylation of retinoblastoma protein and abrogative transcription of E2F1 gene targets. Further, we observed that apoptosis was mediated through P53 activation leading to higher BAX/BCL-2 ratio and cleaved caspase-3 levels. It was also seen that P276-00 treatment reduced expression of tumor micro-environment proteins such as IL-6, secreted EGFR and HSPA8. Finally, P276-00 treatment resulted in significant tumor growth inhibition in xenograft tumor models via lowered proliferative activity of E2F1 and aggravated P53 mediated apoptosis. CONCLUSION: In summary, we have observed that P276-00 inhibits cyclin-D/CDK4/P16/pRB/E2F axis and induces apoptosis by increased P53 phosphorylation in HNSCC cells. These results suggest a novel indication for P276-00 in head and neck cancer with a potential role for IL-6 and HSPA8 as candidate serum biomarkers.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Flavones/pharmacology , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cyclin D/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , E2F1 Transcription Factor/metabolism , ErbB Receptors/metabolism , Gene Expression Profiling , HSC70 Heat-Shock Proteins/metabolism , Humans , Inhibitory Concentration 50 , Interleukin-6/metabolism , Mice , Mice, SCID , Neoplasm Transplantation , Phosphorylation , Retinoblastoma Protein/metabolism , Xenograft Model Antitumor Assays
3.
Eur J Pharmacol ; 657(1-3): 41-50, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21296061

ABSTRACT

Microarray technology can be used to study the molecular mechanisms of new chemical entities with the aim to develop effective therapeutics. 7-Hydroxyfrullanolide (7HF) is a sesquiterpene lactone that was found to be efficacious in multiple animal models of inflammation by suppression of pro-inflammatory cytokines; however, its molecular mechanism of action remains unclear. We investigated the effects of 7HF on lipopolysaccharide (LPS)-stimulated human peripheral blood mononuclear cells using microarray-based gene expression studies and explored the molecular targets affected. Gene expression profiles and pathway analysis revealed that 7HF potently suppressed multiple inflammatory pathways induced by LPS. More importantly, 7HF was found to inhibit NF-κB related transcripts. These transcripts were further validated using freshly isolated synovial cells from rheumatoid arthritis patients, thus clinically validating our findings. Cell-based imaging and subsequent Western blot analysis demonstrated that 7HF inhibited the translocation of NF-κB into the nucleus by directly inhibiting the phosphorylation of IKK-ß. Since the transcription of adhesion molecules is regulated by NF-κB, further investigation showed that 7HF dose-dependently suppressed ICAM-1, VCAM-1 and E-selectin expression on LPS-stimulated endothelial cells as well as inhibited the adhesion of monocytes to LPS-stimulated endothelial cells. Taken together, our results reveal that 7HF possesses NF-κB inhibitory potential and suggest a likely molecular mechanism of its anti-inflammatory activity.


Subject(s)
Anti-Inflammatory Agents/pharmacology , NF-kappa B/metabolism , Sesquiterpenes/pharmacology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression Profiling , Humans , I-kappa B Kinase/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , Monocytes/cytology , Monocytes/drug effects , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synovial Fluid/drug effects , Synovial Fluid/metabolism
4.
J Appl Toxicol ; 31(2): 117-30, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20623750

ABSTRACT

Several studies have characterized drug-induced toxicity in liver and kidney. However, the majority of these studies have been performed with 'individual' organs in isolation. Separately, little is known about the role of whole blood as a surrogate tissue in drug-induced toxicity. Accordingly, we investigated the 'concurrent' response of liver, kidney and whole blood during a toxic assault. Rats were acutely treated with therapeutics (acetaminophen, rosiglitazone, fluconazole, isoniazid, cyclophosphamide, amphotericin B, gentamicin and cisplatin) reported for their liver and/or kidney toxicity. Changes in clinical chemistry parameters (e.g. AST, urea) and/or observed microscopic tissue damage confirmed induced hepatotoxicity and/or nephrotoxicity by all drugs. Drug-induced toxicity was not confined to an 'individual' organ. Not all drugs elicited significant alterations in phenotypic parameters of toxicity (e.g. ALT, creatinine). Accordingly, the transcriptional profile of the organs was studied using a toxicity panel of 30 genes derived from literature. Each of the test drugs generated specific gene expression patterns which were unique for all three organs. Hierarchical cluster analyses of purported hepatotoxicants and nephrotoxicants each led to characteristic 'fingerprints' (e.g. decrease in Cyp3a1 indicative of hepatotoxicity; increase in Spp1 and decrease in Gstp1 indicative of nephrotoxicity). In whole blood cells, a set of genes was derived which closely correlated with individual drug-induced concomitant changes in liver or kidney. Collectively, these data demonstrate drug-induced multi-organ toxicity. Furthermore, our findings underscore the importance of transcriptional profiling during inadequate phenotypic anchorage and suggest that whole blood may be judiciously used as a surrogate for drug-induced extra-hematological organ toxicity.


Subject(s)
Blood Cells/drug effects , Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Kidney/drug effects , Animals , Biomarkers/blood , Biomarkers/metabolism , Blood Cells/metabolism , Blood Cells/pathology , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Drug-Related Side Effects and Adverse Reactions/blood , Drug-Related Side Effects and Adverse Reactions/metabolism , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Kidney/metabolism , Kidney/pathology , Organ Specificity , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Toxicity Tests, Acute/methods
5.
Eur J Pharmacol ; 644(1-3): 220-9, 2010 Oct 10.
Article in English | MEDLINE | ID: mdl-20621086

ABSTRACT

A promising therapeutic approach to reduce pathological inflammation is to inhibit the increased production of pro-inflammatory cytokines (e.g., TNF-alpha, IL-6). In this study, we investigated the anti-inflammatory potential of 7-hydroxyfrullanolide (7HF). 7HF is an orally bioavailable, small molecule sesquiterpene lactone isolated from the fruit of Sphaeranthus indicus. 7HF significantly and dose-dependently diminished induced and spontaneous production of TNF-alpha and IL-6 from freshly isolated human mononuclear cells, synovial tissue cells isolated from patients with active rheumatoid arthritis and BALB/c mice. Oral administration of 7HF significantly protected C57BL/6J mice against endotoxin-mediated lethality. In the dextran sulfate sodium (DSS) model of murine colitis, oral administration of 7HF prevented DSS-induced weight loss, attenuated rectal bleeding, improved disease activity index and diminished shortening of the colon of C57BL/6J mice. Histological analyses of colonic tissues revealed that 7HF attenuated DSS-induced colonic edema, leukocyte infiltration in the colonic mucosa and afforded significant protection against DSS-induced crypt damage. 7HF was also significantly efficacious in attenuating carrageenan-induced paw edema in Wistar rats after oral administration. In the collagen-induced arthritis in DBA/1J mice, 7HF significantly reduced disease associated increases in articular index and paw thickness, protected against bone erosion and joint space narrowing and prominently diminished joint destruction, hyperproliferative pannus formation and infiltration of inflammatory cells. Collectively, these results provide evidence that 7HF-mediated inhibition of pro-inflammatory cytokines functionally results in marked protection in experimental models of acute and chronic inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Asteraceae/chemistry , Inflammation/drug therapy , Sesquiterpenes/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/isolation & purification , Arthritis, Experimental/drug therapy , Arthritis, Experimental/physiopathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/physiopathology , Colitis/drug therapy , Colitis/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Inflammation/physiopathology , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Rats , Rats, Wistar , Sesquiterpenes/administration & dosage , Sesquiterpenes/isolation & purification , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...