42 results on '"Bellahcene M"'
Search Results
2. Myocardial function and energy metabolism in carnitine-deficient rats
- Author
-
Zaugg, C. E., Spaniol, M., Kaufmann, P., Bellahcene, M., Barbosa, V., Tolnay, M., Buser, P. T., and Krähenbühl, S.
- Published
- 2003
- Full Text
- View/download PDF
3. Systemic autoimmunity induced by Toll-like receptor 7/8 agonist Resiquimod causes myocarditis and dilated cardiomyopathy: a new model of autoimmune heart disease
- Author
-
Hasham, MG, Baxan, N, Stuckey, D, Dent, O, Branca, J, Perkins, BRA, Duffy, T, Hameed, TS, Stella, SE, Bellahcene, M, Schneider, MD, Harding, SE, Rosenthal, N, Sattler, S, British Heart Foundation, Wellcome Trust, and Medical Research Council (MRC)
- Subjects
Toll-like receptor 7/8 ,Science & Technology ,Resiquimod ,PATHOGENESIS ,Dilated cardiomyopathy ,Autoimmunity ,Cell Biology ,CARDIAC INVOLVEMENT ,11 Medical And Health Sciences ,Heart disease ,DERMATOMYOSITIS ,06 Biological Sciences ,RHEUMATOID-ARTHRITIS ,Myocarditis ,MICE ,POLYMYOSITIS ,B-CELLS ,Pathology ,LUPUS-ERYTHEMATOSUS ,T-CELLS ,AUTOANTIBODIES ,Life Sciences & Biomedicine ,Model ,Developmental Biology - Abstract
Systemic autoimmune diseases such as Systemic Lupus Erythematosus (SLE) and Rheumatoid Arthritis (RA) show significant heart involvement and cardiovascular morbidity, which can be due to systemically increased levels of inflammation or direct autoreactivity targeting cardiac tissue. Despite high clinical relevance, cardiac damage secondary to systemic autoimmunity lacks inducible rodent models. Here we characterize immune-mediated cardiac tissue damage in a new model of SLE induced by topical application of the TLR-7/8 agonist Resiquimod. We observe a cardiac phenotype reminiscent of autoimmune-mediated dilated cardiomyopathy, and identify auto-antibodies as major contributors to cardiac tissue damage. Resiquimod-induced heart disease is a highly relevant mouse model for mechanistic and therapeutic studies aiming to protect the heart during autoimmunity.
- Published
- 2017
4. Chronic activation of γ2 AMPK induces obesity and reduces β cell function
- Author
-
Yavari, A, Stocker, CJ, Ghaffari, S, Wargent, ET, Steeples, V, Czibki, G, Pinter, K, Bellahcene, M, Oliver, PL, Stockenhuber, A, Nguyen, C, Lazdam, M, Kyriakou, T, Parnis, J, Sarma, D, Katritsis, G, Wortmann, DJ, Harper, AR, Brown, LA, Peirson, SN, Redwood, C, Watkins, H, and Ashrafian, H
- Abstract
Despite significant advances in our understanding of the biology determining systemic energy homeostasis, the treatment of obesity remains a medical challenge. Activation of AMP-activated protein kinase (AMPK) has been proposed as an attractive strategy for the treatment of obesity and its complications. AMPK is a conserved, ubiquitously expressed, heterotrimeric serine/threonine kinase whose short-term activation has multiple beneficial metabolic effects. Whether these translate into long-term benefits for obesity and its complications is unknown. Here, we observe that mice with chronic AMPK activation, resulting from mutation of the AMPK γ2 subunit, exhibit ghrelin signalling-dependent hyperphagia, obesity and impaired pancreatic islet insulin secretion. Humans bearing the homologous mutation manifest a congruent phenotype. Our studies highlight that longterm AMPK activation throughout all tissues can have adverse metabolic consequences, with implications for pharmacological strategies seeking to chronically activate AMPK systemically to treat metabolic disease.
- Published
- 2016
- Full Text
- View/download PDF
5. Vegetative compatibility of Verticillium dahlia isolated from olive trees (Olea europea L.) in Algeria
- Author
-
Bellahcene, M., Fortas, Z., Diana Fernandez, and Nicole, M.
- Subjects
MUTANT NIT ,ETUDE COMPARATIVE ,PATHOLOGIE VEGETALE ,OLIVIER ,Nit mutants, VCG, Verticillium dahliae, Olive tree ,STRUCTURE DE POPULATION ,CHAMPIGNON PARASITE ,MUTATION ,COMPATIBILITE VEGETATIVE ,ORIGINE GEOGRAPHIQUE - Abstract
25 isolates of Verticillium dahliae obtained from olive trees: 18 of them originating from two regions of Algeria (Nord-ouest and Kabylie), 4 isolates from France and 3 from Syria. They were investigated using complementation tests with nitrate-nonutilizing (Nit) mutants to know their vegetative compatibility. Among 250 chlorate-resistant sectors obtained, only 187 were Nit mutants. Three types of Nit mutants were obtained (Nit1, Nit3 and NitM) on the basis of the fungal phenotype. Nit1 mutants were the most frequent (71.6%), followed by NitM (16.6%) and Nit3 (11.8%). Based on their ability to formheterokaryons, all olive pathogenic isolates were grouped into a single vegetative compatibility groups (VCG). This is a good indication of the homogeneity of the Algerian V. dahliae population. The results also suggest the absence of a relationship between geographical origin of strains and VCG.
- Published
- 2011
6. Genetic diversity of Verticillium dahliae isolates from olive trees in Algeria
- Author
-
Bellahcene, M., Assigbetsé, K., Fortas, Z., Geiger, J. -P, Nicole, M., and Diana Fernandez
- Abstract
Verticillium wilt of olive trees (Olea europaea L.), a wilt caused by the soil-borne fungus Verticillium dahliae (Kleb), is one of the most serious diseases in Algerian olive groves. To assess the pathogenic and genetic diversity of olive-infecting V. dahliae populations in Algeria, orchards from the two main olive-producing regions (north-western Algeria and Kabylia) were sampled and 27 V. dahliae isolates were recovered. For purposes of comparison, V. dahliae strains from France and Syria were added to the analysis. By means of PCR primers that specifically discriminate between defoliating (D) and non-defoliating (ND) V. dahliae pathotypes it was shown that all V. dahliae isolates belonged to the ND pathotype. The amount of genetic variation between the 43 isolates was assessed by random amplification of polymorphic DNA (RAPD). A total of 16 RAPD haplotypes were found on the basis of the presence or absence of 25 polymorphic DNA fragments. Genotypic diversity between the 27 Algerian isolates was low, with two RAPD haplotypes accounting for 70% of all isolates. Genotypic diversity was however greater between isolates from Kabylia than between isolates from north-western Algeria. Cluster analysis showed that most of the Algerian V. dahliae isolates grouped together with the French and Syrian isolates. On the basis of their ability to form heterokaryons with each other, a subset of 25 olive-pathogenic isolates was grouped into a single vegetative compatibility group (VCG). These results suggest that the olive-infecting V. dahliae populations in Algeria show limited diversity and that caution should be taken to prevent introduction of the D pathotype.
- Published
- 2005
7. Antimycin A induced cardioprotection is dependent on pre-ischemic p38-MAPK activation but independent of MKK3
- Author
-
KABIR, A, primary, CAO, X, additional, GOROG, D, additional, TANNO, M, additional, BASSI, R, additional, BELLAHCENE, M, additional, QUINLAN, R, additional, DAVIS, R, additional, FLAVELL, R, additional, and SHATTOCK, M, additional
- Published
- 2005
- Full Text
- View/download PDF
8. 101 No acute antiarrhythmic effects of losartan and enalapril in hypertrophied rat hearts
- Author
-
BUTZ, S, primary, REMONDINO, A, additional, DRIAMOV, S, additional, BELLAHCENE, M, additional, TRAUB, D, additional, BUSER, P, additional, and ZAUGG, C, additional
- Published
- 2003
- Full Text
- View/download PDF
9. 93 Bradykinin is a mediator but not a trigger of antiarrhythmic effects of ischemic preconditioning
- Author
-
DRIAMOV, S, primary, BELLAHCENE, M, additional, BUTZ, S, additional, TRAUB, D, additional, BUSER, P, additional, and ZAUGG, C, additional
- Published
- 2003
- Full Text
- View/download PDF
10. Cardioprotection mediated by urocortin is dependent on PKCε activation.
- Author
-
Lawrence, K. M., Kabir, A. M. N., Bellahcene, M., Davidson, S., Cao, X. B., McCormick, J., Mesquita, R. A., Carroll, C. J., Chanalaris, A., Townsend, P. A., Hubank, M., Stephanou, A., Knight, R. A., Marber, M. S., and Latchman, D. S.
- Subjects
PROTEIN kinase C ,ISCHEMIA ,HEART cells ,HEART cytology ,PHARMACODYNAMICS - Abstract
Examines the role of protein kinase C epsilon (PKC∈) in the cardioprotective effect of urocortin (Ucn). Description of Ucn; Effect of Ucn on PKC∈ Mechanism of Ucn treatment during ischaemia and reperfusion in cardiomyocytes.
- Published
- 2005
- Full Text
- View/download PDF
11. 93 Bradykinin is a mediator but not a trigger of antiarrhythmic effects of ischemic preconditioning
- Author
-
Driamov, S., Bellahcene, M., Butz, S., Traub, D., Buser, P.T., and Zaugg, C.E.
- Subjects
- *
BRADYKININ - Abstract
An abstract of the study "Bradykinin is a mediator but not a trigger of antiarrhythmic effects of ischemic preconditioning," by S. Driamov et al, is presented.
- Published
- 2004
12. 101 No acute antiarrhythmic effects of losartan and enalapril in hypertrophied rat hearts
- Author
-
Butz, S., Remondino, A., Driamov, S., Bellahcene, M., Traub, D., Buser, P.T., and Zaugg, C.E.
- Subjects
LOSARTAN ,ENALAPRIL - Abstract
An abstract of the study "No acute antiarrhythmic effects of losartan and enalapril in hypertrophied rat hearts," by S. Butz et al, is presented.
- Published
- 2004
13. MAPKAPK-2 AND SERINE PHOSPHORYLATION OF H5P25/27 AND αB-CRYSTALLIN DO NOT INCREASE MYOCARDIAL RESISTANCE TO INFARCTION.
- Author
-
Gorog, D. A., Tanno, M., Fisher, S. G., Cao, X. B., Bellahcene, M., Dighe, K., Kabir, A. M. N., Quinlan, R. A., Kato, K., Gaestel, M., and Marber, M. S.
- Subjects
PHOSPHORYLATION ,CHEMICAL reactions ,PHOSPHORYLASES ,ISCHEMIA ,CYTOSKELETAL proteins ,AMINO acids - Abstract
This article focuses on a study related to MAPKAPK-2 and serine phosphorylation of HSP25/27 and αB-crystallin do not increase myocardial resistance to infarction. Ischaemic activation of MAPKAPK-2 and the phosphoryiation of its downstream targets H5P25/27 (pHSP25/27) and αB-crystallin may be protective since pHSP25/27 and pαBC contribute to cytoskeletal stabilisation during ischaernia. MAPKAPK-2 does not influence the sensitivity to ischaemia, with or without IP.
- Published
- 2004
14. Cross-Priming Dendritic Cells Exacerbate Immunopathology After Ischemic Tissue Damage in the Heart.
- Author
-
Forte E, Perkins B, Sintou A, Kalkat HS, Papanikolaou A, Jenkins C, Alsubaie M, Chowdhury RA, Duffy TM, Skelly DA, Branca J, Bellahcene M, Schneider MD, Harding SE, Furtado MB, Ng FS, Hasham MG, Rosenthal N, and Sattler S
- Subjects
- Animals, Antigen Presentation, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Dendritic Cells metabolism, Disease Models, Animal, Female, Heart Failure pathology, Humans, Lectins, C-Type deficiency, Lectins, C-Type genetics, Lymph Nodes immunology, Lymph Nodes metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Myocardial Infarction immunology, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardium immunology, Myocardium metabolism, Receptors, Chemokine metabolism, Receptors, Immunologic deficiency, Receptors, Immunologic genetics, Cross-Priming, Dendritic Cells immunology, Myocardium pathology
- Abstract
Background: Ischemic heart disease is a leading cause of heart failure and despite advanced therapeutic options, morbidity and mortality rates remain high. Although acute inflammation in response to myocardial cell death has been extensively studied, subsequent adaptive immune activity and anti-heart autoimmunity may also contribute to the development of heart failure. After ischemic injury to the myocardium, dendritic cells (DC) respond to cardiomyocyte necrosis, present cardiac antigen to T cells, and potentially initiate a persistent autoimmune response against the heart. Cross-priming DC have the ability to activate both CD4
+ helper and CD8+ cytotoxic T cells in response to necrotic cells and may thus be crucial players in exacerbating autoimmunity targeting the heart. This study investigates a role for cross-priming DC in post-myocardial infarction immunopathology through presentation of self-antigen from necrotic cardiac cells to cytotoxic CD8+ T cells., Methods: We induced type 2 myocardial infarction-like ischemic injury in the heart by treatment with a single high dose of the β-adrenergic agonist isoproterenol. We characterized the DC population in the heart and mediastinal lymph nodes and analyzed long-term cardiac immunopathology and functional decline in wild type and Clec9a -depleted mice lacking DC cross-priming function., Results: A diverse DC population, including cross-priming DC, is present in the heart and activated after ischemic injury. Clec9a-/- mice deficient in DC cross-priming are protected from persistent immune-mediated myocardial damage and decline of cardiac function, likely because of dampened activation of cytotoxic CD8+ T cells., Conclusion: Activation of cytotoxic CD8+ T cells by cross-priming DC contributes to exacerbation of postischemic inflammatory damage of the myocardium and corresponding decline in cardiac function. Importantly, this provides novel therapeutic targets to prevent postischemic immunopathology and heart failure.- Published
- 2021
- Full Text
- View/download PDF
15. Neutrophils Enable Local and Non-Invasive Liposome Delivery to Inflamed Skeletal Muscle and Ischemic Heart.
- Author
-
Che J, Najer A, Blakney AK, McKay PF, Bellahcene M, Winter CW, Sintou A, Tang J, Keane TJ, Schneider MD, Shattock RJ, Sattler S, and Stevens MM
- Subjects
- Animals, Humans, Inflammation metabolism, Liposomes, Mice, Muscle, Skeletal metabolism, Myocardial Ischemia metabolism, Neutrophils metabolism
- Abstract
Uncontrolled inflammation is a major pathological factor underlying a range of diseases including autoimmune conditions, cardiovascular disease, and cancer. Improving localized delivery of immunosuppressive drugs to inflamed tissue in a non-invasive manner offers significant promise to reduce severe side effects caused by systemic administration. Here, a neutrophil-mediated delivery system able to transport drug-loaded nanocarriers to inflamed tissue by exploiting the inherent ability of neutrophils to migrate to inflammatory tissue is reported. This hybrid system (neutrophils loaded with liposomes ex vivo) efficiently migrates in vitro following an inflammatory chemokine gradient. Furthermore, the triggered release of loaded liposomes and reuptake by target macrophages is studied. The migratory behavior of liposome-loaded neutrophils is confirmed in vivo by demonstrating the delivery of drug-loaded liposomes to an inflamed skeletal muscle in mice. A single low-dose injection of the hybrid system locally reduces inflammatory cytokine levels. Biodistribution of liposome-loaded neutrophils in a human-disease-relevant myocardial ischemia reperfusion injury mouse model after i.v. injection confirms the ability of injected neutrophils to carry loaded liposomes to inflammation sites. This strategy shows the potential of nanocarrier-loaded neutrophils as a universal platform to deliver anti-inflammatory drugs to promote tissue regeneration in inflammatory diseases., (© 2020 The Authors. Published by Wiley-VCH GmbH.)
- Published
- 2020
- Full Text
- View/download PDF
16. Human pluripotent stem cell-derived cardiomyocytes as a target platform for paracrine protection by cardiac mesenchymal stromal cells.
- Author
-
Constantinou C, Miranda AMA, Chaves P, Bellahcene M, Massaia A, Cheng K, Samari S, Rothery SM, Chandler AM, Schwarz RP, Harding SE, Punjabi P, Schneider MD, and Noseda M
- Subjects
- Animals, Coculture Techniques, Culture Media, Conditioned, Humans, Mice, Mesenchymal Stem Cells cytology, Myocytes, Cardiac cytology, Pluripotent Stem Cells cytology, Stromal Cells cytology
- Abstract
Ischemic heart disease remains the foremost cause of death globally, with survivors at risk for subsequent heart failure. Paradoxically, cell therapies to offset cardiomyocyte loss after ischemic injury improve long-term cardiac function despite a lack of durable engraftment. An evolving consensus, inferred preponderantly from non-human models, is that transplanted cells benefit the heart via early paracrine signals. Here, we tested the impact of paracrine signals on human cardiomyocytes, using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) as the target of mouse and human cardiac mesenchymal stromal cells (cMSC) with progenitor-like features. In co-culture and conditioned medium studies, cMSCs markedly inhibited human cardiomyocyte death. Little or no protection was conferred by mouse tail tip or human skin fibroblasts. Consistent with the results of transcriptomic profiling, functional analyses showed that the cMSC secretome suppressed apoptosis and preserved cardiac mitochondrial transmembrane potential. Protection was independent of exosomes under the conditions tested. In mice, injecting cMSC-conditioned media into the infarct border zone reduced apoptotic cardiomyocytes > 70% locally. Thus, hPSC-CMs provide an auspicious, relevant human platform to investigate extracellular signals for cardiac muscle survival, substantiating human cardioprotection by cMSCs, and suggesting the cMSC secretome or its components as potential cell-free therapeutic products.
- Published
- 2020
- Full Text
- View/download PDF
17. MAP4K4 Inhibition Promotes Survival of Human Stem Cell-Derived Cardiomyocytes and Reduces Infarct Size In Vivo.
- Author
-
Fiedler LR, Chapman K, Xie M, Maifoshie E, Jenkins M, Golforoush PA, Bellahcene M, Noseda M, Faust D, Jarvis A, Newton G, Paiva MA, Harada M, Stuckey DJ, Song W, Habib J, Narasimhan P, Aqil R, Sanmugalingam D, Yan R, Pavanello L, Sano M, Wang SC, Sampson RD, Kanayaganam S, Taffet GE, Michael LH, Entman ML, Tan TH, Harding SE, Low CMR, Tralau-Stewart C, Perrior T, and Schneider MD
- Published
- 2020
- Full Text
- View/download PDF
18. Evaluation of antimicrobial activity of Terfezia arenaria extracts collected from Saharan desert against bacteria and filamentous fungi.
- Author
-
Harir M, Bendif H, Yahiaoui M, Bellahcene M, Zohra F, and Rodríguez-Couto S
- Abstract
Different desert truffles, collected from Algerian Saharan soils, were identified and their capacity to produce bioactive substances with antimicrobial activity was analyzed. Based on morphological characterization using Melzer's reagent staining, the collected strains were identified as Terfezia arenaria . The bioactive substances from T. arenaria were extracted using the following techniques: maceration with methanol and Soxhlet with dichloromethane. The former led to a yield much higher than that of the latter (i.e., 15% and 0.48%, respectively). Both extracts presented antifungal activities against all the tested strains (i.e., A. niger , Penicillium sp., and C. albicans ). However, the dichloromethane extracts showed much higher antibacterial activities against all the tested bacteria (i.e., S. aureus, E. faecalis, E. coli, and P. aeruginosa ) than the methanol extracts. The thin layer chromatography of both extracts confirmed the presence of polyphenols and flavonoids., Competing Interests: Conflict of interestThe authors declare no competing financial interest.
- Published
- 2019
- Full Text
- View/download PDF
19. MAP4K4 Inhibition Promotes Survival of Human Stem Cell-Derived Cardiomyocytes and Reduces Infarct Size In Vivo.
- Author
-
Fiedler LR, Chapman K, Xie M, Maifoshie E, Jenkins M, Golforoush PA, Bellahcene M, Noseda M, Faust D, Jarvis A, Newton G, Paiva MA, Harada M, Stuckey DJ, Song W, Habib J, Narasimhan P, Aqil R, Sanmugalingam D, Yan R, Pavanello L, Sano M, Wang SC, Sampson RD, Kanayaganam S, Taffet GE, Michael LH, Entman ML, Tan TH, Harding SE, Low CMR, Tralau-Stewart C, Perrior T, and Schneider MD
- Subjects
- Animals, Cell Survival drug effects, Cells, Cultured, Dose-Response Relationship, Drug, Female, Humans, Hydrogen Peroxide pharmacology, Induced Pluripotent Stem Cells cytology, Infarction metabolism, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Myocytes, Cardiac metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Structure-Activity Relationship, Doxorubicin pharmacology, Infarction drug therapy, Infarction pathology, Intracellular Signaling Peptides and Proteins antagonists & inhibitors, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Heart disease is a paramount cause of global death and disability. Although cardiomyocyte death plays a causal role and its suppression would be logical, no clinical counter-measures target the responsible intracellular pathways. Therapeutic progress has been hampered by lack of preclinical human validation. Mitogen-activated protein kinase kinase kinase kinase-4 (MAP4K4) is activated in failing human hearts and relevant rodent models. Using human induced-pluripotent-stem-cell-derived cardiomyocytes (hiPSC-CMs) and MAP4K4 gene silencing, we demonstrate that death induced by oxidative stress requires MAP4K4. Consequently, we devised a small-molecule inhibitor, DMX-5804, that rescues cell survival, mitochondrial function, and calcium cycling in hiPSC-CMs. As proof of principle that drug discovery in hiPSC-CMs may predict efficacy in vivo, DMX-5804 reduces ischemia-reperfusion injury in mice by more than 50%. We implicate MAP4K4 as a well-posed target toward suppressing human cardiac cell death and highlight the utility of hiPSC-CMs in drug discovery to enhance cardiomyocyte survival., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
20. Mutant Muscle LIM Protein C58G causes cardiomyopathy through protein depletion.
- Author
-
Ehsan M, Kelly M, Hooper C, Yavari A, Beglov J, Bellahcene M, Ghataorhe K, Poloni G, Goel A, Kyriakou T, Fleischanderl K, Ehler E, Makeyev E, Lange S, Ashrafian H, Redwood C, Davies B, Watkins H, and Gehmlich K
- Subjects
- Animals, Cardiomyopathy, Hypertrophic physiopathology, Disease Models, Animal, Gene Knock-In Techniques, Humans, Mice, Mutation, Sarcomeres genetics, Adaptor Proteins, Signal Transducing genetics, Apoptosis Regulatory Proteins genetics, Cardiomyopathy, Hypertrophic genetics, Heart physiopathology, LIM Domain Proteins genetics, Muscle Proteins genetics
- Abstract
Cysteine and glycine rich protein 3 (CSRP3) encodes Muscle LIM Protein (MLP), a well-established disease gene for Hypertrophic Cardiomyopathy (HCM). MLP, in contrast to the proteins encoded by the other recognised HCM disease genes, is non-sarcomeric, and has important signalling functions in cardiomyocytes. To gain insight into the disease mechanisms involved, we generated a knock-in mouse (KI) model, carrying the well documented HCM-causing CSRP3 mutation C58G. In vivo phenotyping of homozygous KI/KI mice revealed a robust cardiomyopathy phenotype with diastolic and systolic left ventricular dysfunction, which was supported by increased heart weight measurements. Transcriptome analysis by RNA-seq identified activation of pro-fibrotic signalling, induction of the fetal gene programme and activation of markers of hypertrophic signalling in these hearts. Further ex vivo analyses validated the activation of these pathways at transcript and protein level. Intriguingly, the abundance of MLP decreased in KI/KI mice by 80% and in KI/+ mice by 50%. Protein depletion was also observed in cellular studies for two further HCM-causing CSRP3 mutations (L44P and S54R/E55G). We show that MLP depletion is caused by proteasome action. Moreover, MLP C58G interacts with Bag3 and results in a proteotoxic response in the homozygous knock-in mice, as shown by induction of Bag3 and associated heat shock proteins. In conclusion, the newly generated mouse model provides insights into the underlying disease mechanisms of cardiomyopathy caused by mutations in the non-sarcomeric protein MLP. Furthermore, our cellular experiments suggest that protein depletion and proteasomal overload also play a role in other HCM-causing CSPR3 mutations that we investigated, indicating that reduced levels of functional MLP may be a common mechanism for HCM-causing CSPR3 mutations., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
21. Isolation and characterization of a novel tyrosinase produced by Sahara soil actinobacteria and immobilization on nylon nanofiber membranes.
- Author
-
Harir M, Bellahcene M, Baratto MC, Pollini S, Rossolini GM, Trabalzini L, Fatarella E, and Pogni R
- Subjects
- Africa, Northern, Amino Acid Sequence, DNA, Bacterial analysis, Enzymes, Immobilized chemistry, Enzymes, Immobilized metabolism, Levodopa metabolism, Membranes, Artificial, Microscopy, Electron, Scanning, Monophenol Monooxygenase chemistry, Monophenol Monooxygenase metabolism, Nanofibers, Nylons, Soil Microbiology, Streptomyces genetics, Streptomyces ultrastructure, Monophenol Monooxygenase isolation & purification, Streptomyces metabolism
- Abstract
In the present study different actinomycete strains were collected and isolated from Algerian Sahara soil with the aim to select novel enzymes with promising features for biotechnological applications. The Ms1 strain was selected, amongst the others, for its capability to produce melanin in different solid media. Ms1 chromosomal DNA was sequenced and the strain assigned to Streptomyces cyaneofuscatus sp. A tyrosinase (MW∼30kD) encoding sequence was identified and the corresponding enzyme was isolated and biochemically characterized. The tyrosinase showed the highest activity and stability at neutral and alkaline pH and it was able to oxidize l-DOPA at T=55°C and pH 7. The enzyme showed variable stability in presence of various water-miscible organic solvents, while it was inactivated by reducing agents. The tyrosinase activity was unaffected by NaCl and enhanced by different cations. Furthermore, the enzyme showed a higher specificity for diphenols than monophenols showing a higher diphenolase than monophenolase activity. Finally, tyrosinase was stabilized by immobilization on nylon nanofiber membranes with a payload of 82% when 1% glutaraldeyde was used. Taken all together, these results show that the enzyme displays interesting properties for biotechnological purposes., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
22. Mammalian γ2 AMPK regulates intrinsic heart rate.
- Author
-
Yavari A, Bellahcene M, Bucchi A, Sirenko S, Pinter K, Herring N, Jung JJ, Tarasov KV, Sharpe EJ, Wolfien M, Czibik G, Steeples V, Ghaffari S, Nguyen C, Stockenhuber A, Clair JRS, Rimmbach C, Okamoto Y, Yang D, Wang M, Ziman BD, Moen JM, Riordon DR, Ramirez C, Paina M, Lee J, Zhang J, Ahmet I, Matt MG, Tarasova YS, Baban D, Sahgal N, Lockstone H, Puliyadi R, de Bono J, Siggs OM, Gomes J, Muskett H, Maguire ML, Beglov Y, Kelly M, Dos Santos PPN, Bright NJ, Woods A, Gehmlich K, Isackson H, Douglas G, Ferguson DJP, Schneider JE, Tinker A, Wolkenhauer O, Channon KM, Cornall RJ, Sternick EB, Paterson DJ, Redwood CS, Carling D, Proenza C, David R, Baruscotti M, DiFrancesco D, Lakatta EG, Watkins H, and Ashrafian H
- Subjects
- Adult, Animals, Bradycardia metabolism, Electrocardiography, Ambulatory, Exercise, Heart diagnostic imaging, Humans, Magnetic Resonance Imaging, Cine, Magnetic Resonance Spectroscopy, Mice, Microscopy, Electron, Transmission, Mutation, Myocardium metabolism, Myocardium pathology, Myocardium ultrastructure, Physical Conditioning, Animal, Physical Endurance, Ryanodine Receptor Calcium Release Channel metabolism, Sinoatrial Node pathology, AMP-Activated Protein Kinases genetics, Bradycardia genetics, Calcium metabolism, Heart Rate genetics, Sarcolemma metabolism, Sinoatrial Node metabolism
- Abstract
AMPK is a conserved serine/threonine kinase whose activity maintains cellular energy homeostasis. Eukaryotic AMPK exists as αβγ complexes, whose regulatory γ subunit confers energy sensor function by binding adenine nucleotides. Humans bearing activating mutations in the γ2 subunit exhibit a phenotype including unexplained slowing of heart rate (bradycardia). Here, we show that γ2 AMPK activation downregulates fundamental sinoatrial cell pacemaker mechanisms to lower heart rate, including sarcolemmal hyperpolarization-activated current (I
f ) and ryanodine receptor-derived diastolic local subsarcolemmal Ca2+ release. In contrast, loss of γ2 AMPK induces a reciprocal phenotype of increased heart rate, and prevents the adaptive intrinsic bradycardia of endurance training. Our results reveal that in mammals, for which heart rate is a key determinant of cardiac energy demand, AMPK functions in an organ-specific manner to maintain cardiac energy homeostasis and determines cardiac physiological adaptation to exercise by modulating intrinsic sinoatrial cell behavior.- Published
- 2017
- Full Text
- View/download PDF
23. Systemic autoimmunity induced by the TLR7/8 agonist Resiquimod causes myocarditis and dilated cardiomyopathy in a new mouse model of autoimmune heart disease.
- Author
-
Hasham MG, Baxan N, Stuckey DJ, Branca J, Perkins B, Dent O, Duffy T, Hameed TS, Stella SE, Bellahcene M, Schneider MD, Harding SE, Rosenthal N, and Sattler S
- Subjects
- Adaptive Immunity drug effects, Adoptive Transfer, Animals, Autoantibodies blood, Cardiomyopathy, Dilated complications, Cardiomyopathy, Dilated immunology, Cardiomyopathy, Dilated physiopathology, Disease Models, Animal, Female, Genetic Variation, Heart Function Tests, Immunity, Cellular drug effects, Inflammation pathology, Lymph Nodes drug effects, Lymph Nodes pathology, Male, Mutation genetics, Myocarditis complications, Myocarditis immunology, Myocarditis physiopathology, Myocardium pathology, Spleen pathology, Toll-Like Receptor 7 metabolism, Toll-Like Receptor 8 metabolism, Autoimmunity drug effects, Cardiomyopathy, Dilated chemically induced, Imidazoles adverse effects, Myocarditis chemically induced, Toll-Like Receptor 7 agonists, Toll-Like Receptor 8 agonists
- Abstract
Systemic autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) show significant heart involvement and cardiovascular morbidity, which can be due to systemically increased levels of inflammation or direct autoreactivity targeting cardiac tissue. Despite high clinical relevance, cardiac damage secondary to systemic autoimmunity lacks inducible rodent models. Here, we characterise immune-mediated cardiac tissue damage in a new model of SLE induced by topical application of the Toll-like receptor 7/8 (TLR7/8) agonist Resiquimod. We observe a cardiac phenotype reminiscent of autoimmune-mediated dilated cardiomyopathy, and identify auto-antibodies as major contributors to cardiac tissue damage. Resiquimod-induced heart disease is a highly relevant mouse model for mechanistic and therapeutic studies aiming to protect the heart during autoimmunity., (© 2017. Published by The Company of Biologists Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
24. Resistance of dynamin-related protein 1 oligomers to disassembly impairs mitophagy, resulting in myocardial inflammation and heart failure.
- Author
-
Cahill TJ, Leo V, Kelly M, Stockenhuber A, Kennedy NW, Bao L, Cereghetti GM, Harper AR, Czibik G, Liao C, Bellahcene M, Steeples V, Ghaffari S, Yavari A, Mayer A, Poulton J, Ferguson DJ, Scorrano L, Hettiarachchi NT, Peers C, Boyle J, Hill RB, Simmons A, Watkins H, Dear TN, and Ashrafian H
- Published
- 2016
- Full Text
- View/download PDF
25. Benzimidazole derivative small-molecule 991 enhances AMPK activity and glucose uptake induced by AICAR or contraction in skeletal muscle.
- Author
-
Bultot L, Jensen TE, Lai YC, Madsen AL, Collodet C, Kviklyte S, Deak M, Yavari A, Foretz M, Ghaffari S, Bellahcene M, Ashrafian H, Rider MH, Richter EA, and Sakamoto K
- Subjects
- AMP-Activated Protein Kinases drug effects, Aminoimidazole Carboxamide pharmacology, Animals, Antibodies, Blocking pharmacology, Humans, In Vitro Techniques, Isoenzymes, Mice, Mice, Knockout, Muscle Contraction drug effects, AMP-Activated Protein Kinases metabolism, Aminoimidazole Carboxamide analogs & derivatives, Benzimidazoles pharmacology, Benzoates pharmacology, Enzyme Activators pharmacology, Glucose metabolism, Hypoglycemic Agents pharmacology, Muscle, Skeletal drug effects, Ribonucleotides pharmacology
- Abstract
AMP-activated protein kinase (AMPK) plays diverse roles and coordinates complex metabolic pathways for maintenance of energy homeostasis. This could be explained by the fact that AMPK exists as multiple heterotrimer complexes comprising a catalytic α-subunit (α1 and α2) and regulatory β (β1 and β2)- and γ (γ1, γ2, γ3)-subunits, which are uniquely distributed across different cell types. There has been keen interest in developing specific and isoform-selective AMPK-activating drugs for therapeutic use and also as research tools. Moreover, establishing ways of enhancing cellular AMPK activity would be beneficial for both purposes. Here, we investigated if a recently described potent AMPK activator called 991, in combination with the commonly used activator 5-aminoimidazole-4-carboxamide riboside or contraction, further enhances AMPK activity and glucose transport in mouse skeletal muscle ex vivo. Given that the γ3-subunit is exclusively expressed in skeletal muscle and has been implicated in contraction-induced glucose transport, we measured the activity of AMPKγ3 as well as ubiquitously expressed γ1-containing complexes. We initially validated the specificity of the antibodies for the assessment of isoform-specific AMPK activity using AMPK-deficient mouse models. We observed that a low dose of 991 (5 μM) stimulated a modest or negligible activity of both γ1- and γ3-containing AMPK complexes. Strikingly, dual treatment with 991 and 5-aminoimidazole-4-carboxamide riboside or 991 and contraction profoundly enhanced AMPKγ1/γ3 complex activation and glucose transport compared with any of the single treatments. The study demonstrates the utility of a dual activator approach to achieve a greater activation of AMPK and downstream physiological responses in various cell types, including skeletal muscle., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
26. Mutation of Fnip1 is associated with B-cell deficiency, cardiomyopathy, and elevated AMPK activity.
- Author
-
Siggs OM, Stockenhuber A, Deobagkar-Lele M, Bull KR, Crockford TL, Kingston BL, Crawford G, Anzilotti C, Steeples V, Ghaffari S, Czibik G, Bellahcene M, Watkins H, Ashrafian H, Davies B, Woods A, Carling D, Yavari A, Beutler B, and Cornall RJ
- Subjects
- AMP-Activated Protein Kinases genetics, Animals, B-Lymphocytes enzymology, B-Lymphocytes metabolism, Cardiomyopathies genetics, Carrier Proteins metabolism, Cell Count, Humans, Mice, Mice, Inbred C57BL, Mutation, Proto-Oncogene Proteins genetics, Tumor Suppressor Proteins genetics, AMP-Activated Protein Kinases metabolism, B-Lymphocytes cytology, Cardiomyopathies metabolism, Carrier Proteins genetics, Proto-Oncogene Proteins metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Folliculin (FLCN) is a tumor-suppressor protein mutated in the Birt-Hogg-Dubé (BHD) syndrome, which associates with two paralogous proteins, folliculin-interacting protein (FNIP)1 and FNIP2, forming a complex that interacts with the AMP-activated protein kinase (AMPK). Although it is clear that this complex influences AMPK and other metabolic regulators, reports of its effects have been inconsistent. To address this issue, we created a recessive loss-of-function variant of Fnip1 Homozygous FNIP1 deficiency resulted in profound B-cell deficiency, partially restored by overexpression of the antiapoptotic protein BCL2, whereas heterozygous deficiency caused a loss of marginal zone B cells. FNIP1-deficient mice developed cardiomyopathy characterized by left ventricular hypertrophy and glycogen accumulation, with close parallels to mice and humans bearing gain-of-function mutations in the γ2 subunit of AMPK. Concordantly, γ2-specific AMPK activity was elevated in neonatal FNIP1-deficient myocardium, whereas AMPK-dependent unc-51-like autophagy activating kinase 1 (ULK1) phosphorylation and autophagy were increased in FNIP1-deficient B-cell progenitors. These data support a role for FNIP1 as a negative regulator of AMPK.
- Published
- 2016
- Full Text
- View/download PDF
27. Resistance of Dynamin-related Protein 1 Oligomers to Disassembly Impairs Mitophagy, Resulting in Myocardial Inflammation and Heart Failure.
- Author
-
Cahill TJ, Leo V, Kelly M, Stockenhuber A, Kennedy NW, Bao L, Cereghetti GM, Harper AR, Czibik G, Liao C, Bellahcene M, Steeples V, Ghaffari S, Yavari A, Mayer A, Poulton J, Ferguson DJ, Scorrano L, Hettiarachchi NT, Peers C, Boyle J, Hill RB, Simmons A, Watkins H, Dear TN, and Ashrafian H
- Subjects
- Animals, Biopolymers genetics, Biopolymers metabolism, Cells, Cultured, Dynamins genetics, Dynamins metabolism, Heart Failure physiopathology, Mice, Mutation, Myocarditis physiopathology, Oxidative Phosphorylation, Biopolymers physiology, Dynamins physiology, Heart Failure etiology, Mitophagy, Myocarditis etiology
- Abstract
We have reported previously that a missense mutation in the mitochondrial fission gene Dynamin-related protein 1 (Drp1) underlies the Python mouse model of monogenic dilated cardiomyopathy. The aim of this study was to investigate the consequences of the C452F mutation on Drp1 protein function and to define the cellular sequelae leading to heart failure in the Python monogenic dilated cardiomyopathy model. We found that the C452F mutation increased Drp1 GTPase activity. The mutation also conferred resistance to oligomer disassembly by guanine nucleotides and high ionic strength solutions. In a mouse embryonic fibroblast model, Drp1 C452F cells exhibited abnormal mitochondrial morphology and defective mitophagy. Mitochondria in C452F mouse embryonic fibroblasts were depolarized and had reduced calcium uptake with impaired ATP production by oxidative phosphorylation. In the Python heart, we found a corresponding progressive decline in oxidative phosphorylation with age and activation of sterile inflammation. As a corollary, enhancing autophagy by exposure to a prolonged low-protein diet improved cardiac function in Python mice. In conclusion, failure of Drp1 disassembly impairs mitophagy, leading to a downstream cascade of mitochondrial depolarization, aberrant calcium handling, impaired ATP synthesis, and activation of sterile myocardial inflammation, resulting in heart failure., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
28. Changes in the cardiac metabolome caused by perhexiline treatment in a mouse model of hypertrophic cardiomyopathy.
- Author
-
Gehmlich K, Dodd MS, Allwood JW, Kelly M, Bellahcene M, Lad HV, Stockenhuber A, Hooper C, Ashrafian H, Redwood CS, Carrier L, and Dunn WB
- Subjects
- Animals, Cardiomyopathy, Hypertrophic diagnostic imaging, Cardiomyopathy, Hypertrophic pathology, Chromatography, High Pressure Liquid, Disease Models, Animal, Male, Mass Spectrometry, Metabolomics, Mice, Inbred C57BL, Myocardium pathology, Perhexiline pharmacology, Phenotype, Principal Component Analysis, Ultrasonography, Cardiomyopathy, Hypertrophic drug therapy, Cardiomyopathy, Hypertrophic metabolism, Metabolome drug effects, Myocardium metabolism, Perhexiline therapeutic use
- Abstract
Energy depletion has been highlighted as an important contributor to the pathology of hypertrophic cardiomyopathy (HCM), a common inherited cardiac disease. Pharmacological reversal of energy depletion appears an attractive approach and the use of perhexiline has been proposed as it is thought to shift myocardial metabolism from fatty acid to glucose utilisation, increasing ATP production and myocardial efficiency. We used the Mybpc3-targeted knock-in mouse model of HCM to investigate changes in the cardiac metabolome following perhexiline treatment. Echocardiography indicated that perhexiline induced partial improvement of some, but not all hypertrophic parameters after six weeks. Non-targeted metabolomics, applying ultra-high performance liquid chromatography-mass spectrometry, described a phenotypic modification of the cardiac metabolome with 272 unique metabolites showing a statistically significant change (p < 0.05). Changes in fatty acids and acyl carnitines indicate altered fatty acid transport into mitochondria, implying reduction in fatty acid beta-oxidation. Increased glucose utilisation is indirectly implied through changes in the glycolytic, glycerol, pentose phosphate, tricarboxylic acid and pantothenate pathways. Depleted reduced glutathione and increased production of NADPH suggest reduction in oxidative stress. These data delineate the metabolic changes occurring during improvement of the HCM phenotype and indicate the requirements for further targeted interventions.
- Published
- 2015
- Full Text
- View/download PDF
29. Male mice that lack the G-protein-coupled receptor GPR41 have low energy expenditure and increased body fat content.
- Author
-
Bellahcene M, O'Dowd JF, Wargent ET, Zaibi MS, Hislop DC, Ngala RA, Smith DM, Cawthorne MA, Stocker CJ, and Arch JR
- Subjects
- Adipose Tissue drug effects, Animals, Bacteria metabolism, Body Fluid Compartments drug effects, Body Fluid Compartments metabolism, Diet, Fat-Restricted, Diet, High-Fat, Dietary Fats metabolism, Female, Gastrointestinal Tract metabolism, Gastrointestinal Tract microbiology, Glucose Intolerance genetics, Heart drug effects, Insulin metabolism, Insulin Secretion, Leptin metabolism, Male, Mice, Mice, Knockout, Muscle, Skeletal drug effects, Obesity etiology, Obesity metabolism, Obesity prevention & control, Organ Size, Receptors, G-Protein-Coupled metabolism, Sex Factors, Adipose Tissue metabolism, Body Composition genetics, Dietary Fats pharmacology, Energy Metabolism genetics, Fatty Acids, Volatile metabolism, Obesity genetics, Receptors, G-Protein-Coupled genetics
- Abstract
SCFA are produced in the gut by bacterial fermentation of undigested carbohydrates. Activation of the Gαi-protein-coupled receptor GPR41 by SCFA in β-cells and sympathetic ganglia inhibits insulin secretion and increases sympathetic outflow, respectively. A possible role in stimulating leptin secretion by adipocytes is disputed. In the present study, we investigated energy balance and glucose homoeostasis in GPR41 knockout mice fed on a standard low-fat or a high-fat diet. When fed on the low-fat diet, body fat mass was raised and glucose tolerance was impaired in male but not female knockout mice compared to wild-type mice. Soleus muscle and heart weights were reduced in the male mice, but total body lean mass was unchanged. When fed on the high-fat diet, body fat mass was raised in male but not female GPR41 knockout mice, but by no more in the males than when they were fed on the low-fat diet. Body lean mass and energy expenditure were reduced in male mice but not in female knockout mice. These results suggest that the absence of GPR41 increases body fat content in male mice. Gut-derived SCFA may raise energy expenditure and help to protect against obesity by activating GPR41.
- Published
- 2013
- Full Text
- View/download PDF
30. AMP-activated protein kinase phosphorylates cardiac troponin I and alters contractility of murine ventricular myocytes.
- Author
-
Oliveira SM, Zhang YH, Solis RS, Isackson H, Bellahcene M, Yavari A, Pinter K, Davies JK, Ge Y, Ashrafian H, Walker JW, Carling D, Watkins H, Casadei B, and Redwood C
- Subjects
- AMP-Activated Protein Kinases antagonists & inhibitors, AMP-Activated Protein Kinases genetics, Aminoimidazole Carboxamide analogs & derivatives, Aminoimidazole Carboxamide pharmacology, Animals, Calcium Signaling, Enzyme Activation, Enzyme Activators pharmacology, Heart Ventricles enzymology, Humans, Male, Mice, Mice, Inbred C57BL, Myocytes, Cardiac drug effects, Myosins drug effects, Myosins metabolism, Phosphorylation, Protein Kinase Inhibitors pharmacology, Pyrazoles pharmacology, Pyrimidines pharmacology, Ribonucleotides pharmacology, Serine, Time Factors, Troponin I genetics, Two-Hybrid System Techniques, AMP-Activated Protein Kinases metabolism, Myocardial Contraction, Myocytes, Cardiac enzymology, Troponin I metabolism, Ventricular Function, Left drug effects
- Abstract
Rationale: AMP-activated protein kinase (AMPK) is an important regulator of energy balance and signaling in the heart. Mutations affecting the regulatory γ2 subunit have been shown to cause an essentially cardiac-restricted phenotype of hypertrophy and conduction disease, suggesting a specific role for this subunit in the heart., Objective: The γ isoforms are highly conserved at their C-termini but have unique N-terminal sequences, and we hypothesized that the N-terminus of γ2 may be involved in conferring substrate specificity or in determining intracellular localization., Methods and Results: A yeast 2-hybrid screen of a human heart cDNA library using the N-terminal 273 residues of γ2 as bait identified cardiac troponin I (cTnI) as a putative interactor. In vitro studies showed that cTnI is a good AMPK substrate and that Ser150 is the principal residue phosphorylated. Furthermore, on AMPK activation during ischemia, Ser150 is phosphorylated in whole hearts. Using phosphomimics, measurements of actomyosin ATPase in vitro and force generation in demembraneated trabeculae showed that modification at Ser150 resulted in increased Ca(2+) sensitivity of contractile regulation. Treatment of cardiomyocytes with the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) resulted in increased myocyte contractility without changing the amplitude of Ca(2+) transient and prolonged relaxation despite shortening the time constant of Ca(2+) transient decay (tau). Compound C prevented the effect of AICAR on myocyte function. These results suggest that AMPK activation increases myocyte contraction and prolongs relaxation by increasing myofilament Ca(2+) sensitivity., Conclusions: We conclude that cTnI phosphorylation by AMPK may represent a novel mechanism of regulation of cardiac function.
- Published
- 2012
- Full Text
- View/download PDF
31. Fumarate is cardioprotective via activation of the Nrf2 antioxidant pathway.
- Author
-
Ashrafian H, Czibik G, Bellahcene M, Aksentijević D, Smith AC, Mitchell SJ, Dodd MS, Kirwan J, Byrne JJ, Ludwig C, Isackson H, Yavari A, Støttrup NB, Contractor H, Cahill TJ, Sahgal N, Ball DR, Birkler RI, Hargreaves I, Tennant DA, Land J, Lygate CA, Johannsen M, Kharbanda RK, Neubauer S, Redwood C, de Cabo R, Ahmet I, Talan M, Günther UL, Robinson AJ, Viant MR, Pollard PJ, Tyler DJ, and Watkins H
- Subjects
- Animals, Dimethyl Fumarate, Fumarate Hydratase deficiency, Fumarate Hydratase genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Models, Biological, Myocardial Infarction genetics, Myocardial Infarction prevention & control, NF-E2-Related Factor 2 genetics, Signal Transduction genetics, Signal Transduction physiology, Antioxidants metabolism, Fumarates therapeutic use, NF-E2-Related Factor 2 metabolism
- Abstract
The citric acid cycle (CAC) metabolite fumarate has been proposed to be cardioprotective; however, its mechanisms of action remain to be determined. To augment cardiac fumarate levels and to assess fumarate's cardioprotective properties, we generated fumarate hydratase (Fh1) cardiac knockout (KO) mice. These fumarate-replete hearts were robustly protected from ischemia-reperfusion injury (I/R). To compensate for the loss of Fh1 activity, KO hearts maintain ATP levels in part by channeling amino acids into the CAC. In addition, by stabilizing the transcriptional regulator Nrf2, Fh1 KO hearts upregulate protective antioxidant response element genes. Supporting the importance of the latter mechanism, clinically relevant doses of dimethylfumarate upregulated Nrf2 and its target genes, hence protecting control hearts, but failed to similarly protect Nrf2-KO hearts in an in vivo model of myocardial infarction. We propose that clinically established fumarate derivatives activate the Nrf2 pathway and are readily testable cytoprotective agents., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
32. Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids.
- Author
-
Zaibi MS, Stocker CJ, O'Dowd J, Davies A, Bellahcene M, Cawthorne MA, Brown AJ, Smith DM, and Arch JR
- Subjects
- Animals, Fatty Acids, Volatile metabolism, Mice, Mice, Knockout, Pertussis Toxin metabolism, Signal Transduction, Acetates metabolism, Adipocytes metabolism, Butyrates metabolism, Leptin metabolism, Propionates metabolism
- Abstract
GPR41 is reportedly expressed in murine adipose tissue and mediates short chain fatty acid (SCFA)-stimulated leptin secretion by activating Galpha(i). Here, we agree with a contradictory report in finding no expression of GPR41 in murine adipose tissue. Nevertheless, in the presence of adenosine deaminase to minimise Galpha(i) signalling via the adenosine A1 receptor, SCFA stimulated leptin secretion by adipocytes from wild-type but not GPR41 knockout mice. Expression of GPR43 was reduced in GPR41 knockout mice. Acetate but not butyrate stimulated leptin secretion in wild-type mesenteric adipocytes, consistent with mediation of the response by GPR43 rather than GPR41. Pertussis toxin prevented stimulation of leptin secretion by propionate in epididymal adipocytes, implicating Galpha(i) signalling mediated by GPR43 in SCFA-stimulated leptin secretion., (Copyright 2010 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
33. MAPKAPK-2 modulates p38-MAPK localization and small heat shock protein phosphorylation but does not mediate the injury associated with p38-MAPK activation during myocardial ischemia.
- Author
-
Gorog DA, Jabr RI, Tanno M, Sarafraz N, Clark JE, Fisher SG, Cao XB, Bellahcene M, Dighe K, Kabir AM, Quinlan RA, Kato K, Gaestel M, Marber MS, and Heads RJ
- Subjects
- Animals, Enzyme Activation drug effects, Imidazoles pharmacology, Intracellular Signaling Peptides and Proteins deficiency, Mice, Mice, Knockout, Phosphorylation, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases deficiency, Protein Transport, Pyridines pharmacology, Signal Transduction, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, Heat-Shock Proteins, Small metabolism, Intracellular Signaling Peptides and Proteins metabolism, Myocardial Ischemia metabolism, Protein Serine-Threonine Kinases metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
MAPKAPK-2 (MK2) is a protein kinase activated downstream of p38-MAPK which phosphorylates the small heat shock proteins HSP27 and alphaB crystallin and modulates p38-MAPK cellular distribution. p38-MAPK activation is thought to contribute to myocardial ischemic injury; therefore, we investigated MK2 effects on ischemic injury and p38 cellular localization using MK2-deficient mice (KO). Immunoblotting of extracts from Langendorff-perfused hearts subjected to aerobic perfusion or global ischemia or reperfusion showed that the total and phosphorylated p38 levels were significantly lower in MK2(-/-) compared to MK2(+/+) hearts at baseline, but the ratio of phosphorylated/total p38 was similar. These results were confirmed by cellular fractionation and immunoblotting for both cytosolic and nuclear compartments. Furthermore, HSP27 and alphaB crsytallin phosphorylation were reduced to baseline in MK2(-/-) hearts. On semiquantitative immunofluorescence laser confocal microscopy of hearts during aerobic perfusion, the mean total p38 fluorescence was significantly higher in the nuclear compared to extranuclear (cytoplasmic, sarcomeric, and sarcolemmal compartments) in MK2(+/+) hearts. However, although the increase in phosphorylated p38 fluorescence intensity in all compartments following ischemia in MK2(+/+) hearts was lost in MK2(-/-) hearts, it was basally elevated in nuclei of MK2(-/-) hearts and was similar to that seen during ischemia in MK2(+/+) hearts. Despite these differences, similar infarct volumes were recorded in wild-type MK2(+/+) and MK2(-/-) hearts, which were decreased by the p38 inhibitor SB203580 (1 microM) in both genotypes. In conclusion, p38 MAPK-induced myocardial ischemic injury is not modulated by MK2. However, the absence of MK2 perturbs the cellular distribution of p38. The preserved nuclear distribution of active p38 MAPK in MK2(-/-) hearts and the conserved response to SB203580 suggests that activation of p38 MAPK may contribute to injury independently of MK2.
- Published
- 2009
- Full Text
- View/download PDF
34. Bradykinin is a mediator, but unlikely a trigger, of antiarrhythmic effects of ischemic preconditioning.
- Author
-
Driamov SV, Bellahcene M, Butz S, Buser PT, and Zaugg CE
- Subjects
- Animals, Bradykinin analogs & derivatives, Bradykinin drug effects, Bradykinin pharmacology, Bradykinin Receptor Antagonists, Disease Models, Animal, Disease Progression, Electrocardiography, Heart Rate physiology, Heart Ventricles drug effects, Heart Ventricles physiopathology, Male, Pilot Projects, Prognosis, Rats, Rats, Sprague-Dawley, Tachycardia, Ventricular metabolism, Tachycardia, Ventricular prevention & control, Ventricular Fibrillation metabolism, Ventricular Fibrillation prevention & control, Bradykinin metabolism, Heart Ventricles metabolism, Ischemic Preconditioning, Myocardial methods, Tachycardia, Ventricular etiology, Ventricular Fibrillation etiology
- Abstract
Objective: Brief reversible ischemic episodes (ischemic preconditioning, IPC) protect the heart against arrhythmias during a subsequent prolonged low-flow ischemia. We have recently shown that this protection involves release of bradykinin, activation of bradykinin B2 receptors followed by opening of sarcolemmal, but not mitochondrial ATP-sensitive K+ channels. The goal of this study was to clarify a trigger and/or mediator role of bradykinin in the antiarrhythmic effects of IPC during low-flow ischemia., Methods: Isolated perfused rat hearts underwent 60 minutes of low-flow ischemia induced by reducing perfusion pressure followed by 60 minutes of reperfusion. Preconditioning was induced by 2 x 5 minutes episodes of zero-flow ischemia. In yet other groups, preconditioned or non-preconditioned hearts were treated either with bradykinin (10 nmol/L) or with HOE 140 (bradykinin B2 receptor antagonist, 100 nmol/L)., Results: IPC reduced the number of ventricular premature beats, as well as the incidence of ventricular tachycardia and of ventricular fibrillation during low-flow ischemia. In addition, this protection was abolished by HOE 140 given during low-flow ischemia. Pharmacological preconditioning using short bradykinin perfusion instead of IPC did not show antiarrhythmic effects. However, bradykinin administered during low-flow ischemia and reperfusion reduced the number of ventricular premature beats and the incidence of ventricular tachycardia and of ventricular fibrillation during low-flow ischemia., Conclusion: Bradykinin is a mediator, but unlikely a trigger, of antiarrhythmic effects of IPC during low-flow ischemia.
- Published
- 2007
- Full Text
- View/download PDF
35. Cardioprotection initiated by reactive oxygen species is dependent on activation of PKCepsilon.
- Author
-
Kabir AM, Clark JE, Tanno M, Cao X, Hothersall JS, Dashnyam S, Gorog DA, Bellahcene M, Shattock MJ, and Marber MS
- Subjects
- Animals, Anti-Bacterial Agents pharmacology, Antimycin A pharmacology, Cells, Cultured, Enzyme Activation physiology, Female, Gene Expression Regulation, Enzymologic, Ischemic Preconditioning, Myocardial, Male, Mice, Mice, Knockout, Mitochondria, Heart metabolism, Myocytes, Cardiac metabolism, Protein Kinase C-epsilon genetics, Random Allocation, Rats, Protein Kinase C-epsilon metabolism, Reactive Oxygen Species metabolism, Reperfusion Injury metabolism, Reperfusion Injury prevention & control
- Abstract
To examine whether cardioprotection initiated by reactive oxygen species (ROS) is dependent on protein kinase Cepsilon (PKCepsilon), isolated buffer-perfused mouse hearts were randomized to four groups: 1) antimycin A (AA) (0.1 microg/ml) for 3 min followed by 10 min washout and then 30 min global ischemia (I) and 2 h reperfusion (R); 2) controls of I/R alone; 3) AA bracketed with 13 min of N-2-mercaptopropionyl- glycine (MPG) followed by I/R; and 4) MPG (200 microM) alone, followed by I/R. Isolated adult rat ventricular myocytes (ARVM) were exposed to AA (0.1 microg/ml), and lucigenin was used to measure ROS production. Murine hearts and ARVM were exposed to AA (0.1 microg/ml) with or without MPG, and PKCepsilon translocation was measured by cell fractionation and subsequent Western blot analysis. Finally, the dependence of AA protection on PKCepsilon was determined by the use of knockout mice (-/-) lacking PKCepsilon. AA exposure caused ROS production, which was abolished by the mitochondrial uncoupler mesoxalonitrile 4-trifluoromethoxyphenylhydrazone. In addition, AA significantly reduced the percent infarction-left ventricular volume compared with control I/R (26 +/- 4 vs. 43 +/- 2%; P < 0.05). Bracketing AA with MPG caused a loss of protection (52 +/- 7 vs. 26 +/- 4%; P < 0.05). AA caused PKCepsilon translocation only in the absence of MPG, and protection was lost on the pkcepsilon(-/-) background (38 +/- 3 vs. 15 +/- 4%; P < 0.001). AA causes ROS production, on which protection and PKCepsilon translocation depend. In addition, protection is absent in PKCepsilon null hearts. Our results imply that, in common with ischemic preconditioning, PKCepsilon is crucial to ROS-mediated protection.
- Published
- 2006
- Full Text
- View/download PDF
36. Activation of p38 mitogen-activated protein kinase contributes to the early cardiodepressant action of tumor necrosis factor.
- Author
-
Bellahcene M, Jacquet S, Cao XB, Tanno M, Haworth RS, Layland J, Kabir AM, Gaestel M, Davis RJ, Flavell RA, Shah AM, Avkiran M, and Marber MS
- Subjects
- Animals, Enzyme Activation physiology, Enzyme Inhibitors pharmacology, Imidazoles pharmacology, In Vitro Techniques, Intracellular Signaling Peptides and Proteins, MAP Kinase Kinase 3 deficiency, Male, Mice, Mice, Knockout, Myocardium enzymology, Myocytes, Cardiac drug effects, Myocytes, Cardiac enzymology, Protein Kinases deficiency, Protein Serine-Threonine Kinases, Pyridines pharmacology, Stroke Volume drug effects, Myocardial Contraction drug effects, Myocardial Contraction physiology, Tumor Necrosis Factor-alpha pharmacology, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
Objectives: The purpose of this study was to determine whether p38 mitogen-activated protein kinase (p38-MAPK) contributes to tumor necrosis factor-alpha (TNFalpha)-induced contractile depression., Background: Tumor necrosis factor has both beneficial and detrimental consequences that may result from the activation of different downstream pathways. Tumor necrosis factor activates p38-MAPK, a stress-responsive kinase implicated in contractile depression and cardiac injury., Methods: In isolated hearts from mice lacking the p38-MAPK activator, MAPK kinase 3 (MKK3), perfused at constant coronary pressure or flow, we measured the left ventricular developed pressure (LVDP) and the relationship between end-diastolic volume and LVDP in the presence and absence of 10 ng/ml TNFalpha., Results: Within 15 min at constant pressure, TNFalpha significantly reduced LVDP and coronary flow in outbred and mkk3(+/+) mice. This early negative inotropic effect was associated with a marked phosphorylation of both p38-MAPK and its indirect substrate, HSP27. In hearts lacking MKK3, TNFalpha failed to activate p38-MAPK or to cause significant contractile dysfunction. The actions of TNFalpha were similarly attenuated in MAPK-activated protein kinase 2 (MK2)-deficient hearts, which have a marked reduction in myocardial p38-MAPK protein content, and by the p38-MAPK catalytic site inhibitor SB203580 (1 micromol/l). Under conditions of constant coronary flow, the p38-MAPK activation and contractile depression induced by TNFalpha, though attenuated, remained sensitive to the absence of MKK3 or the presence of SB203580. The role of p38-MAPK in TNFalpha-induced contractile depression was confirmed in isolated murine cardiac myocytes exposed to SB203580 or lacking MKK3., Conclusions: Tumor necrosis factor activates p38-MAPK in the intact heart and in isolated cardiac myocytes through MKK3. This activation likely contributes to the early cardiodepressant action of TNFalpha.
- Published
- 2006
- Full Text
- View/download PDF
37. Antiarrhythmic effect of ischemic preconditioning during low-flow ischemia. The role of bradykinin and sarcolemmal versus mitochondrial ATP-sensitive K(+) channels.
- Author
-
Driamov S, Bellahcene M, Ziegler A, Barbosa V, Traub D, Butz S, Buser PT, and Zaugg CE
- Subjects
- Animals, Anti-Arrhythmia Agents pharmacology, Arrhythmias, Cardiac metabolism, Male, Myocardial Ischemia metabolism, Myocardial Reperfusion, Potassium Channel Blockers pharmacology, Rats, Rats, Sprague-Dawley, Receptor, Bradykinin B2 drug effects, Arrhythmias, Cardiac physiopathology, Bradykinin metabolism, Ischemic Preconditioning, Myocardial, Mitochondria metabolism, Myocardial Ischemia physiopathology, Potassium Channels metabolism, Sarcolemma metabolism
- Abstract
Short episodes of ischemia (ischemic preconditioning) protect the heart against ventricular arrhythmias during zero-flow ischemia and reperfusion. However, in clinics, many episodes of ischemia present a residual flow (low-flow ischemia). Here we examined whether ischemic preconditioning protects against ventricular arrhythmias during and after a low-flow ischemia and, if so, by what mechanism(s). Isolated rat hearts were subjected to 60 min of low-flow ischemia (12% residual coronary flow) followed by 60 min of reperfusion. Ischemic preconditioning was induced by two cycles of 5 min of zero-flow ischemia followed by 5 and 15 min of reperfusion, respectively. Arrhythmias were evaluated as numbers of ventricular premature beats (VPBs) as well as incidences of ventricular tachycardia (VT) and ventricular fibrillation (VF) during low-flow ischemia and reperfusion. Ischemic preconditioning significantly reduced the number of VPBs and the incidence of VT and of VF during low-flow ischemia. This antiarrhythmic effect of preconditioning was abolished by HOE 140 (100 nM), a bradykinin B(2) receptor blocker. Similar to preconditioning, exogenous bradykinin (10 nM) reduced the number of VPBs and the incidence of VT and of VF during low-flow ischemia. Furthermore, the antiarrhythmic effects of both ischemic preconditioning and bradykinin were abolished by glibenclamide (1 microM), a non-specific blocker of ATP-sensitive K(+) (K(ATP)) channels. Finally, the antiarrhythmic effects of both ischemic preconditioning and bradykinin were abolished by HMR 1098 (10 microM), a sarcolemmal K(ATP) channel blocker but not by 5-hydroxydecanoate (100 microM), a mitochondrial K(ATP) channel blocker. In conclusion, ischemic preconditioning protects against ventricular arrhythmias induced by low-flow ischemia, and this protection involves activation of bradykinin B(2) receptors and subsequent opening of sarcolemmal but not of mitochondrial K(ATP) channels.
- Published
- 2004
- Full Text
- View/download PDF
38. Losartan but not enalaprilat acutely reduces reperfusion ventricular tachyarrhythmias in hypertrophied rat hearts after low-flow ischaemia.
- Author
-
Butz S, Driamov S, Remondino A, Bellahcene M, Beier K, Ziegler A, Buser PT, and Zaugg CE
- Subjects
- Action Potentials, Animals, Anti-Arrhythmia Agents administration & dosage, Dose-Response Relationship, Drug, Enalaprilat administration & dosage, Heart drug effects, Heart physiopathology, Hypertrophy, Left Ventricular complications, In Vitro Techniques, Losartan administration & dosage, Male, Rats, Rats, Sprague-Dawley, Reperfusion Injury complications, Reperfusion Injury physiopathology, Tachycardia, Ventricular etiology, Ventricular Fibrillation etiology, Ventricular Fibrillation physiopathology, Anti-Arrhythmia Agents pharmacology, Enalaprilat pharmacology, Losartan pharmacology, Reperfusion Injury drug therapy, Tachycardia, Ventricular prevention & control, Ventricular Fibrillation prevention & control
- Abstract
Based on clinical and experimental studies, angiotensin II receptor blockers and angiotensin converting enzyme inhibitors have been proposed to exert acute anti-arrhythmic effects in heart failure patients. Therefore, the goal of this study was to assess acute anti-arrhythmic effects of losartan and enalaprilat in hypertrophied rat hearts during low-flow ischaemia and reperfusion. In dose-finding experiments in non-hypertrophied isolated perfused hearts, we performed dose-response curves of losartan and enalaprilat studying monophasic action potential duration at 90% repolarisation (MAPD(90%)) and ventricular fibrillation (VF) threshold. Subsequently, we determined the effects of losartan and enalaprilat (in therapeutically relevant concentrations) on ventricular tachyarrhythmias induced by low-flow ischaemia/reperfusion in hearts demonstrating left ventricular (LV) hypertrophy 70 days after aortic banding. We found that neither drug significantly affected MAPD(90%) (1 nM-1 mM) or VF threshold (1 microM losartan and 10 microM enalaprilat) in non-hypertrophied hearts. Similarly in hypertrophied hearts, neither drug significantly affected the incidence or the duration of ventricular tachyarrhythmias (ventricular tachycardia and VF) during low-flow ischaemia. However, 1 microM losartan significantly reduced the duration of ventricular tachyarrhythmias during reperfusion. In conclusion, neither losartan nor enalaprilat is acutely anti-arrhythmic in hypertrophied rat hearts during low-flow ischaemia. During reperfusion, however, losartan but not enalaprilat exerts acute anti-arrhythmic effects.
- Published
- 2004
- Full Text
- View/download PDF
39. Inhibition of p38 MAPK activity fails to attenuate contractile dysfunction in a mouse model of low-flow ischemia.
- Author
-
Gorog DA, Tanno M, Cao X, Bellahcene M, Bassi R, Kabir AM, Dighe K, Quinlan RA, and Marber MS
- Subjects
- Animals, Blotting, Western methods, Enzyme Activation drug effects, Male, Mice, Mice, Inbred C57BL, Mitogen-Activated Protein Kinases analysis, Myocardium enzymology, Perfusion, p38 Mitogen-Activated Protein Kinases, Imidazoles pharmacology, Mitogen-Activated Protein Kinases antagonists & inhibitors, Myocardial Contraction drug effects, Myocardial Stunning physiopathology, Pyridines pharmacology, Ventricular Dysfunction, Left physiopathology
- Abstract
Objective: The basal activity of p38 MAPK has recently been shown to impair myocardial contractility. This kinase is activated by ischemia and short-term hibernation. We hypothesized that p38 MAPK activation may contribute to the contractile deficit that characterizes low-flow ischemia., Methods: In Langendorff-perfused isolated C57BL/6 mouse hearts, perfusion pressure was reduced from 85 to 15 or 30 mm Hg for 120 min to induce ischemic left ventricular dysfunction. The effect of the p38 MAPK inhibitor SB203580 (1 microM/l) on contractile function and p38 MAPK activation was assessed., Results: Reduction in perfusion pressure to 15 or 30 mm Hg was accompanied by stable reductions in coronary flow (83+/-2% and 66+/-2%, respectively) and developed pressure (84+/-2% and 61+/-3%), with minimal infarction (15.6+/-0.69% and 10.6+/-0.98% of LV myocardium, respectively), but marked activation of p38 MAPK (reflected in pHSP27 1092+/-326% basal and 996+/-301% basal, respectively). The p38 MAPK inhibitor SB203580, present during the last 60 min of reduced pressure perfusion, prevented p38 MAPK activation (pHSP27 281+/-92% basal, p=0.01 and 186+/-72% basal, p=0.01) but, despite the presence of a contractile reserve, had no effect on developed pressure. Similarly, early treatment with SB203580 started 5 min after the onset of reduced flow also failed to attenuate contractile dysfunction., Conclusion: The p38 MAPK activation that accompanies short-term hibernation does not appear to contribute to the contractile deficit.
- Published
- 2004
- Full Text
- View/download PDF
40. Tumor necrosis factor-induced protection of the murine heart is independent of p38-MAPK activation.
- Author
-
Tanno M, Gorog DA, Bellahcene M, Cao X, Quinlan RA, and Marber MS
- Subjects
- Animals, Blotting, Western, Enzyme Activation drug effects, Enzyme Inhibitors pharmacology, Imidazoles pharmacology, Ischemic Preconditioning, Myocardial, MAP Kinase Signaling System drug effects, Male, Mice, Myocardial Infarction etiology, Myocardial Infarction pathology, Myocardial Ischemia metabolism, Myocardial Ischemia pathology, Myocardial Reperfusion, Phosphorylation, Pyridines pharmacology, Time Factors, p38 Mitogen-Activated Protein Kinases, Mitogen-Activated Protein Kinases metabolism, Myocardial Infarction drug therapy, Myocardium enzymology, Tumor Necrosis Factor-alpha pharmacology, Tumor Necrosis Factor-alpha therapeutic use
- Abstract
Brief exposure to tumor necrosis factor (TNF) is known to trigger subsequent cardioprotection. TNF activates multiple downstream signaling cascades including p38-MAPK, a kinase known to initiate ischemic preconditioning. However, it is not known whether this kinase is similarly involved in TNF-induced cardioprotection. In isolated perfused murine hearts, subjected to 30-min global ischemia/2-h reperfusion, infarction/risk volume was significantly reduced by pretreatment with TNF for 15 min at 0.5 ng/ml, but not at 5 or 10 ng/ml, followed by 10-min washout vs. control (% I/R = 31 +/- 3, 46 +/- 5 or 54 +/- 3 vs. 48 +/- 5; P = 0.01, 0.80 and 0.25, respectively). This was in direct contrast to the concentration dependence of myocardial p38-MAPK phosphorylation, as measured by dual phosphorylated p38-MAPK, which was apparent at TNF concentrations of 5 and 10 ng/ml but not at 0.5 ng/ml vs. time-matched control (as % basal 315 +/- 25, 422 +/- 94 and 97 +/- 25 vs. 95 +/- 10; P < 0.01, 0.01 and =0.86, respectively). However, phosphorylation of p38-MAPK at 10 min of ischemia was similar among groups (as % basal 393 +/- 98, 410 +/- 67 and 369 +/- 49 for time-matched control, 0.5 and 5 ng/ml, respectively). These patterns were also reflected in the phosphorylation of the downstream substrate HSP27. Furthermore, the effects of TNF on infarct size were not affected by SB203580 (1 micromol/l). These findings suggest that the pre-ischemic activation of p38-MAPK by TNF does not contribute to cardioprotection afforded by this agent.
- Published
- 2003
- Full Text
- View/download PDF
41. Reversible protein kinase C activation in PC12 cells: effect of NGF treatment.
- Author
-
Dupont JL, Janoshazi A, Bellahcene M, Mykita S, and de Barry J
- Subjects
- Animals, Carbachol pharmacology, Enzyme Activation, Fluoresceins, Fluorescent Dyes, Fura-2 analogs & derivatives, Immunohistochemistry, Indoles, Kinetics, Microscopy, Fluorescence, PC12 Cells, Potassium Chloride pharmacology, Protein Kinase C beta, Protein Kinase C-alpha, Rats, Tetradecanoylphorbol Acetate pharmacology, Calcium metabolism, Isoenzymes metabolism, Nerve Growth Factors pharmacology, Protein Kinase C metabolism
- Abstract
Although protein kinase C (PKC) is a key enzyme in the signal transduction process, there is little information on the mechanism leading to PKC activation in living cells. Using a new fluorescence imaging method, we studied this mechanism and correlated PKC conformational changes with intracellular Ca2+ concentration. PC12 cells were simultaneously loaded with Fura-2-AM and Fim-1, two fluorescent probes, which recognize Ca2+ and PKC, respectively. KCl and carbachol (an agonist to muscarinic receptors) applications induced dose-dependent increases of fluorescence for both probes. Both Ca2+ and PKC responses were observed within seconds following KCl or carbachol application, and were reversible upon stimulus withdrawal. PKC activation kinetics was slightly more rapid than the Ca2+ response after KCl application. After nerve growth factor (NGF) treatment of the cells, the amplitude of the KCl-induced PKC responses was larger indicating an increase in the activated PKC-pool in these cells. This difference between control and NGF-treated cells was not observed following carbachol application, suggesting the involvement of different PKC pools. While the Ca2+ response uniformly occurred in the cytosol, the PKC response displayed a patch pattern with higher intensities in the peripheral zone near the plasma membrane. This heterogeneous distribution of PKC activation sites was similar to the immunocytological localization of Ca2+-dependent and independent PKC isoforms, which suggested that at least several PKC isoforms interacted with intracellular elements. Upon repeated stimulation, the PKC response rapidly desensitized.
- Published
- 2000
- Full Text
- View/download PDF
42. [Arguments for the follicular origin of Verneuil's disease (chronic suppurative hidradenitis)].
- Author
-
Grosshans E, Bouffioux B, and Toufik-Bellahcene M
- Subjects
- Chronic Disease, Folliculitis etiology, Folliculitis pathology, Humans, Inflammation, Suppuration, Sweat Gland Diseases pathology, Sweat Gland Diseases etiology
- Published
- 1991
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.