1
|
Lupidi G, Catalani E, Buonanno F, Gentili D, Giorgi S, Ponnarassery Aravindakshan V, Gabrielli S, Brunetti K, Fausto AM, Picchietti S, Ortenzi C, Marcantoni E, Cervia D. Chemical modification for improving drug-like molecular properties of climacostol, a natural resorcinolic lipid. Bioorg Med Chem 2025; 121:118113. [PMID: 39961231 DOI: 10.1016/j.bmc.2025.118113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025]
Abstract
Small organic molecules are compounds that are manufactured through chemical synthesis. One of the key advantages of small molecules is that they have a low molecular weight and simple chemical structures. This allows more predictability to their pharmacokinetics and pharmacodynamics, which means that dosing is simpler. To use small molecules as a useful tool to address human health issues, the collaboration between disciplines, especially chemistry and biology, is essential. In recent years in our laboratories, we have demonstrated that climacostol, a 5-alkenyl resorcinolic produced by eukaryotic microorganisms as secondary metabolite and obtained by our synthetic strategy too, it shows important biological and pharmacological activities. These ones are highly dependent on the 5-alkenyl chain, and chemical modifications to the resorcinolic moiety can be exploited to achieve higher toxicity against pathogen microbes and protists than climacostol. In this study, we have designed and made a synthetic strategy for a new analogue of climacostol (AN3), and evaluated how the new hydroxyl group at position four in the aromatic ring influences its biological effects on prokaryotic and free-living protists and on non-target cells/organisms, especially with regard to cytotoxic properties.
Collapse
Affiliation(s)
- Gabriele Lupidi
- School of Science and Technology, Section of Chemistry, University of Camerino, 62032 Camerino, Italy
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Federico Buonanno
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage, and Tourism (ECHT), University of Macerata, 62100 Macerata, Italy
| | - Dario Gentili
- School of Science and Technology, Section of Chemistry, University of Camerino, 62032 Camerino, Italy
| | - Simone Giorgi
- School of Science and Technology, Section of Chemistry, University of Camerino, 62032 Camerino, Italy
| | | | - Serena Gabrielli
- School of Science and Technology, Section of Chemistry, University of Camerino, 62032 Camerino, Italy
| | - Kashi Brunetti
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Anna Maria Fausto
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Simona Picchietti
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Claudio Ortenzi
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage, and Tourism (ECHT), University of Macerata, 62100 Macerata, Italy
| | - Enrico Marcantoni
- School of Science and Technology, Section of Chemistry, University of Camerino, 62032 Camerino, Italy.
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy.
| |
Collapse
|
2
|
Titov VY, Dolgorukova AM, Osipov AN, Kochish II. DNIC is a Structure Providing Specificity of Physiological Effects of NO. Bull Exp Biol Med 2023; 176:160-164. [PMID: 38194076 DOI: 10.1007/s10517-024-05987-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Indexed: 01/10/2024]
Abstract
Metabolism of nitric oxide (NO) donors: dinitrosyl iron complexes (DNIC), nitrosothiols (RSNO), and nitroprusside was studied on a chick embryo model. The obtained results give reason to assume that DNIC constituting the main pool of nitroso compounds in the vast majority of tissues are NO donors immediately interacting with the physiological target of NO, and other NO donors can perform this function after their transformation into DNIC. NO is released from DNIC not spontaneously, but under a joint influence of a factor destroying the complex and a target having chemical affinity for NO. A similar mechanism is apparently implicated in NO passage through the cell membrane.
Collapse
Affiliation(s)
- V Yu Titov
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
- Federal Scientific Center "All-Russian Research and Technological Poultry Institute, Russian Academy of Sciences, Sergiev Posad, Moscow region, Russia.
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia.
| | - A M Dolgorukova
- Federal Scientific Center "All-Russian Research and Technological Poultry Institute, Russian Academy of Sciences, Sergiev Posad, Moscow region, Russia
| | - A N Osipov
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - I I Kochish
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| |
Collapse
|
3
|
Catalani E, Del Quondam S, Brunetti K, Cherubini A, Bongiorni S, Taddei AR, Zecchini S, Giovarelli M, De Palma C, Perrotta C, Clementi E, Prantera G, Cervia D. Neuroprotective role of plumbagin on eye damage induced by high-sucrose diet in adult fruit fly Drosophila melanogaster. Biomed Pharmacother 2023; 166:115298. [PMID: 37597318 DOI: 10.1016/j.biopha.2023.115298] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/21/2023] Open
Abstract
The natural compound plumbagin has a wide range of pharmacological and potential therapeutic activities, although its role in neuroretina degeneration is unknown. Here we evaluated the effects of plumbagin on retina homeostasis of the fruit fly Drosophila melanogaster fed with high glucose diet, a model of hyperglycemia-induced eye impairment to study the pathophysiology of diabetic retinopathy at the early stages. To this aim, the visual system of flies orally administered with plumbagin has been analyzed at structural, functional, and molecular/cellular level as for instance neuronal apoptosis/autophagy dysregulation and oxidative stress-related signals. Our results demonstrated that plumbagin ameliorates the visual performance of hyperglycemic flies. Drosophila eye-structure, clearly altered by hyperglycemia, i.e. defects of the pattern of ommatidia, irregular rhabdomeres, vacuoles, damaged mitochondria, and abnormal phototransduction units were rescued, at least in part, by plumbagin. In addition, it reactivated autophagy, decreased the presence of cell death/apoptotic features, and exerted antioxidant effects in the retina. In terms of mechanisms favoring death/survival ratio, Nrf2 signaling activation may be one of the strategies by which plumbagin reduced redox unbalance mainly increasing the levels of glutathione-S-transferase. Likewise, plumbagin may act additively and/or synergistically inhibiting the mitochondrial-endoplasmic reticulum stress and unfolded protein response pathways, which prevented neuronal impairment and eye damage induced by reactive oxygen species. These results provide an avenue for further studies, which may be helpful to develop novel therapeutic candidates and drug targets against eye neurotoxicity by high glucose, a key aspect in retinal complications of diabetes.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Simona Del Quondam
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Kashi Brunetti
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Agnese Cherubini
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Silvia Bongiorni
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Anna Rita Taddei
- Section of Electron Microscopy, Great Equipment Center, Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, via L. Vanvitelli 32, 20129 Milano, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy; Scientific Institute IRCCS "Eugenio Medea", via Don Luigi Monza 20, 23842 Bosisio Parini, Italy
| | - Giorgio Prantera
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy.
| |
Collapse
|
4
|
Angove J, Willson NL, Barekatain R, Rosenzweig D, Forder R. In ovo corticosterone exposure does not influence yolk steroid hormone relative abundance or skeletal muscle development in the embryonic chicken. Poult Sci 2023; 102:102735. [PMID: 37209653 DOI: 10.1016/j.psj.2023.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 05/22/2023] Open
Abstract
In ovo corticosterone (CORT) exposure reportedly reduces growth and alters body composition traits in meat-type chickens. However, the mechanisms governing alterations in growth and body composition remain unclear but could involve myogenic stem cell commitment, and/or the presence of yolk steroid hormones. This study investigated whether in ovo CORT exposure influenced yolk steroid hormone content, as well as embryonic myogenic development in meat-type chickens. Fertile eggs were randomly divided at embryonic day (ED) 11 and administered either a control (CON; 100 µL of 10 mM PBS) or CORT solution (100 µL of 10 mM PBS containing 1 µg CORT) into the chorioallantoic membrane. Yolk samples were collected at ED 0 and ED 5. At ED 15 and hatch, embryos were humanely killed, and yolk and breast muscle (BM) samples were collected. The relative abundance of 15 steroid hormones, along with total lipid content was measured in yolk samples collected at ED 0, ED 5, ED 15, and ED 21. Muscle fiber number, cross-sectional area, and fascicle area occupied by muscle fibers were measured in BM samples collected at hatch. Relative expression of MyoD, MyoG, Pax7, PPARγ, and CEBP/β, and the sex steroid receptors were measured in BM samples collected at hatch. The administration of CORT had a limited effect on yolk steroid hormones. In ovo CORT significantly reduced fascicle area occupied by muscle fibers and CEBP/β expression was increased in CORT exposed birds at hatch. In addition, the quantity of yolk lipid was significantly reduced in CORT-treated birds. In conclusion, in ovo exposure to CORT does not appear to influence early muscle development through yolk steroid hormones in embryonic meat-type chickens however, the results provide a comprehensive analysis of the composition of yolk steroid hormones in ovo at different developmental time points. The findings may suggest increased mesenchymal stem cell commitment to the adipogenic lineage during differentiation and requires further investigation.
Collapse
Affiliation(s)
- J Angove
- School of Animal and Veterinary Sciences, the University of Adelaide, Roseworthy, SA, Australia
| | - N-L Willson
- School of Animal and Veterinary Sciences, the University of Adelaide, Roseworthy, SA, Australia
| | - R Barekatain
- South Australian Research and Development Institute, Roseworthy, SA, Australia
| | - D Rosenzweig
- School of Animal and Veterinary Sciences, the University of Adelaide, Roseworthy, SA, Australia
| | - R Forder
- School of Animal and Veterinary Sciences, the University of Adelaide, Roseworthy, SA, Australia.
| |
Collapse
|
5
|
Kochish II, Brazhnik EA, Vorobyov NI, Nikonov IN, Korenyuga MV, Myasnikova OV, Griffin DK, Surai PF, Romanov MN. Features of Fractal Conformity and Bioconsolidation in the Early Myogenesis Gene Expression and Their Relationship to the Genetic Diversity of Chicken Breeds. Animals (Basel) 2023; 13:521. [PMID: 36766410 PMCID: PMC9913260 DOI: 10.3390/ani13030521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/28/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Elements of fractal analysis are widely used in scientific research, including several biological disciplines. In this study, we hypothesized that chicken breed biodiversity manifests not only at the phenotypic level, but also at the genetic-system level in terms of different profiles of fractal conformity and bioconsolidation in the early myogenesis gene expression. To demonstrate this effect, we developed two mathematical models that describe the fractal nature of the expression of seven key genes in the embryonic breast and thigh muscles in eight breeds of meat, dual purpose, egg and game types. In the first model, we produced breed-specific coefficients of gene expression conformity in each muscle type using the slopes of regression dependencies, as well as an integral myogenesis gene expression index (MGEI). Additionally, breed fractal dimensions and integral myogenesis gene expression fractal dimension index (MGEFDI) were determined. The second gene expression model was based on plotting fractal portraits and calculating indices of fractal bioconsolidation. The bioconsolidation index of myogenesis gene expression correlated with the chick growth rate and nitric oxide (NO) oxidation rate. The proposed fractal models were instrumental in interpreting the genetic diversity of chickens at the level of gene expression for early myogenesis, NO metabolism and the postnatal growth of chicks.
Collapse
Affiliation(s)
- Ivan I. Kochish
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, 109472 Moscow, Russia
| | | | - Nikolai I. Vorobyov
- All-Russia Institute for Agricultural Microbiology, Pushkin, 196608 St. Petersburg, Russia
| | - Ilya N. Nikonov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, 109472 Moscow, Russia
| | - Maxim V. Korenyuga
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, 109472 Moscow, Russia
| | - Olga V. Myasnikova
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, 109472 Moscow, Russia
| | | | - Peter F. Surai
- Vitagene and Health Research Centre, Bristol BS4 2RS, UK
- Department of Microbiology and Biochemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
- Department of Animal Nutrition, Faculty of Agricultural and Environmental Sciences, Szent Istvan University, H-2103 Gödöllő, Hungary
| | - Michael N. Romanov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, 109472 Moscow, Russia
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| |
Collapse
|
6
|
Kochish II, Titov VY, Nikonov IN, Brazhnik EA, Vorobyov NI, Korenyuga MV, Myasnikova OV, Dolgorukova AM, Griffin DK, Romanov MN. Unraveling signatures of chicken genetic diversity and divergent selection in breed-specific patterns of early myogenesis, nitric oxide metabolism and post-hatch growth. Front Genet 2023; 13:1092242. [PMID: 36712856 PMCID: PMC9874007 DOI: 10.3389/fgene.2022.1092242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/27/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction: Due to long-term domestication, breeding and divergent selection, a vast genetic diversity in poultry currently exists, with various breeds being characterized by unique phenotypic and genetic features. Assuming that differences between chicken breeds divergently selected for economically and culturally important traits manifest as early as possible in development and growth stages, we aimed to explore breed-specific patterns and interrelations of embryo myogenesis, nitric oxide (NO) metabolism and post-hatch growth rate (GR). Methods: These characteristics were explored in eight breeds of different utility types (meat-type, dual purpose, egg-type, game, and fancy) by incubating 70 fertile eggs per breed. To screen the differential expression of seven key myogenesis associated genes (MSTN, GHR, MEF2C, MYOD1, MYOG, MYH1, and MYF5), quantitative real-time PCR was used. Results: We found that myogenesis associated genes expressed in the breast and thigh muscles in a coordinated manner showing breed specificity as a genetic diversity signature among the breeds studied. Notably, coordinated ("accord") expression patterns of MSTN, GHR, and MEFC2 were observed both in the breast and thigh muscles. Also, associated expression vectors were identified for MYOG and MYOD1 in the breast muscles and for MYOG and MYF5 genes in the thigh muscles. Indices of NO oxidation and post-hatch growth were generally concordant with utility types of breeds, with meat-types breeds demonstrating higher NO oxidation levels and greater GR values as compared to egg-type, dual purpose, game and fancy breeds. Discussion: The results of this study suggest that differences in early myogenesis, NO metabolism and post-hatch growth are breed-specific; they appropriately reflect genetic diversity and accurately capture the evolutionary history of divergently selected chicken breeds.
Collapse
Affiliation(s)
- Ivan I. Kochish
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | - Vladimir Yu. Titov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
- Federal Scientific Center “All-Russian Poultry Research and Technological Institute” of the Russian Academy of Sciences, Sergiev Posad, Moscow Oblast, Russia
| | - Ilya N. Nikonov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | | | - Nikolai I. Vorobyov
- All-Russia Institute for Agricultural Microbiology, Pushkin, St. Petersburg, Russia
| | - Maxim V. Korenyuga
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | - Olga V. Myasnikova
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | - Anna M. Dolgorukova
- Federal Scientific Center “All-Russian Poultry Research and Technological Institute” of the Russian Academy of Sciences, Sergiev Posad, Moscow Oblast, Russia
| | - Darren K. Griffin
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Michael N. Romanov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
7
|
Catalani E, Zecchini S, Giovarelli M, Cherubini A, Del Quondam S, Brunetti K, Silvestri F, Roux-Biejat P, Napoli A, Casati SR, Ceci M, Romano N, Bongiorni S, Prantera G, Clementi E, Perrotta C, De Palma C, Cervia D. RACK1 is evolutionary conserved in satellite stem cell activation and adult skeletal muscle regeneration. Cell Death Dis 2022; 8:459. [PMID: 36396939 PMCID: PMC9672362 DOI: 10.1038/s41420-022-01250-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
Skeletal muscle growth and regeneration involves the activity of resident adult stem cells, namely satellite cells (SC). Despite numerous mechanisms have been described, different signals are emerging as relevant in SC homeostasis. Here we demonstrated that the Receptor for Activated C-Kinase 1 (RACK1) is important in SC function. RACK1 was expressed transiently in the skeletal muscle of post-natal mice, being abundant in the early phase of muscle growth and almost disappearing in adult mature fibers. The presence of RACK1 in interstitial SC was also detected. After acute injury in muscle of both mouse and the fruit fly Drosophila melanogaster (used as alternative in vivo model) we found that RACK1 accumulated in regenerating fibers while it declined with the progression of repair process. To note, RACK1 also localized in the active SC that populate recovering tissue. The dynamics of RACK1 levels in isolated adult SC of mice, i.e., progressively high during differentiation and low compared to proliferating conditions, and RACK1 silencing indicated that RACK1 promotes both the formation of myotubes and the accretion of nascent myotubes. In Drosophila with depleted RACK1 in all muscle cells or, specifically, in SC lineage we observed a delayed recovery of skeletal muscle after physical damage as well as the low presence of active SC in the wound area. Our results also suggest the coupling of RACK1 to muscle unfolded protein response during SC activation. Collectively, we provided the first evidence that transient levels of the evolutionarily conserved factor RACK1 are critical for adult SC activation and proper skeletal muscle regeneration, favoring the efficient progression of SC from a committed to a fully differentiated state.
Collapse
|
8
|
L-Arginine/nitric oxide regulates skeletal muscle development via muscle fibre-specific nitric oxide/mTOR pathway in chickens. ANIMAL NUTRITION 2022; 10:68-85. [PMID: 35647326 PMCID: PMC9125674 DOI: 10.1016/j.aninu.2022.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/29/2021] [Accepted: 04/24/2022] [Indexed: 11/21/2022]
Abstract
L-Arginine (L-Arg), the precursor of nitric oxide (NO), plays an important role in muscle function. Fast-twitch glycolytic fibres are more susceptible to age-related atrophy than slow-twitch oxidative fibres. The effect of L-Arg/NO on protein metabolism of fast- and slow-twitch muscle fibres was evaluated in chickens. In Exp. 1, 48 chicks at 1 day old were divided into 4 groups of 12 birds and subjected to 4 treatments: basal diet without supplementation or supplemented with 1% L-Arg, and water supplemented with or without L-nitro-arginine methyl ester (L-NAME, 18.5 mM). In Exp. 2, 48 chicks were divided into 4 groups of 12 birds fed with the basal diet and subjected to the following treatments: tap water (control), tap water supplemented with L-NAME (18.5 mM), or molsidomine (MS, 0.1 mM), or 18.5 mM L-NAME + 0.1 mM MS (NAMS). The regulatory effect of L-Arg/NO was further investigated in vitro with myoblasts obtained from chicken embryo pectoralis major (PM) and biceps femoris (BF). In vivo, dietary L-Arg supplementation increased breast (+14.94%, P < 0.05) and thigh muscle mass (+23.40%, P < 0.05); whereas, MS treatment had no detectable influence. However, L-NAME treatment blocked the beneficial influence of L-Arg on muscle development. L-Arg decreased (P < 0.05) protein synthesis rate, phosphorylated mTOR and ribosomal protein S6 kinase beta-1 (p70S6K) levels in breast muscle, which was recovered by L-NAME treatment. In vitro, L-Arg or sodium nitroprusside (SNP) reduced protein synthesis rate, suppressed phosphorylated mTOR/p70S6K and decreased atrogin-1 and muscle RING finger 1 (MuRF1) in myoblasts from PM muscle (P < 0.05). L-NAME abolished the inhibitory effect of L-Arg on protein synthesis and the mTOR/p70S6K pathway. However, myoblasts from BF muscle showed the weak influence. Moreover, blocking the mTOR/p70S6K pathway with rapamycin suppressed protein synthesis of the 2 types of myoblasts; whereas, the protein expression of atrogin-1 and MuRF1 levels were restricted only in myoblasts from PM muscle. In conclusion, L-Arg/NO/mTOR/p70S6K pathway enhances protein accumulation and muscle development in fast-twitch glycolytic muscle in chickens. L-Arg/NO regulates protein turnover in a muscle fibre specific way, which highlights the potential clinical application in fast-twitch glycolytic muscle fibres.
Collapse
|
9
|
Roux-Biejat P, Coazzoli M, Marrazzo P, Zecchini S, Di Renzo I, Prata C, Napoli A, Moscheni C, Giovarelli M, Barbalace MC, Catalani E, Bassi MT, De Palma C, Cervia D, Malaguti M, Hrelia S, Clementi E, Perrotta C. Acid Sphingomyelinase Controls Early Phases of Skeletal Muscle Regeneration by Shaping the Macrophage Phenotype. Cells 2021; 10:3028. [PMID: 34831250 PMCID: PMC8616363 DOI: 10.3390/cells10113028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle regeneration is a complex process involving crosstalk between immune cells and myogenic precursor cells, i.e., satellite cells. In this scenario, macrophage recruitment in damaged muscles is a mandatory step for tissue repair since pro-inflammatory M1 macrophages promote the activation of satellite cells, stimulating their proliferation and then, after switching into anti-inflammatory M2 macrophages, they prompt satellite cells' differentiation into myotubes and resolve inflammation. Here, we show that acid sphingomyelinase (ASMase), a key enzyme in sphingolipid metabolism, is activated after skeletal muscle injury induced in vivo by the injection of cardiotoxin. ASMase ablation shortens the early phases of skeletal muscle regeneration without affecting satellite cell behavior. Of interest, ASMase regulates the balance between M1 and M2 macrophages in the injured muscles so that the absence of the enzyme reduces inflammation. The analysis of macrophage populations indicates that these events depend on the altered polarization of M1 macrophages towards an M2 phenotype. Our results unravel a novel role of ASMase in regulating immune response during muscle regeneration/repair and suggest ASMase as a supplemental therapeutic target in conditions of redundant inflammation that impairs muscle recovery.
Collapse
Affiliation(s)
- Paulina Roux-Biejat
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Marco Coazzoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Pasquale Marrazzo
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Ilaria Di Renzo
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy;
| | - Alessandra Napoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Maria Teresa Bassi
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy;
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milano, Italy;
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy;
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| |
Collapse
|
10
|
Titov VY, Dolgorukova AM, Osipov AN, Kochish II. Putative Role of Ligands of DNIC in the Physiological Action of the Complex. Bull Exp Biol Med 2021; 171:606-610. [PMID: 34617179 DOI: 10.1007/s10517-021-05278-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Indexed: 10/20/2022]
Abstract
In a relatively isolated system of avian embryo, the metabolism of NO, a component of the dinitrosyl iron complexes (DNIC), the main NO donor in most tissues, depends on the ligands that make up the complex. This fact corroborates the earlier hypothesis that these ligands perform a regulatory function in NO metabolism. It is also shown that nitrite injected into the embryo is not oxidized to nitrate like NO in DNIC, but is accumulated outside the amniotic sac. Normally, nitrite is present in an embryo in trace amounts. These facts suggest that NO in the embryo is transferred from the donor molecule to a target in the embryo tissues further transformed with minimum oxidation to nitrite.
Collapse
Affiliation(s)
- V Yu Titov
- All-Russian Research and Technological Institute of Poultry, Russian Academy of Sciences, Sergiev Posad, Moscow region, Russia. .,N. I. Pirogov Russian National Research Medical University, Ministry of the Health of the Russian Federation, Moscow, Russia. .,K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia.
| | - A M Dolgorukova
- All-Russian Research and Technological Institute of Poultry, Russian Academy of Sciences, Sergiev Posad, Moscow region, Russia
| | - A N Osipov
- N. I. Pirogov Russian National Research Medical University, Ministry of the Health of the Russian Federation, Moscow, Russia
| | - I I Kochish
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| |
Collapse
|
11
|
Nutraceutical Strategy to Counteract Eye Neurodegeneration and Oxidative Stress in Drosophila melanogaster Fed with High-Sugar Diet. Antioxidants (Basel) 2021; 10:antiox10081197. [PMID: 34439445 PMCID: PMC8388935 DOI: 10.3390/antiox10081197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022] Open
Abstract
Aberrant production of reactive oxygen species (ROS) is a common feature of damaged retinal neurons in diabetic retinopathy, and antioxidants may exert both preventive and therapeutic action. To evaluate the beneficial and antioxidant properties of food supplementation with Lisosan G, a powder of bran and germ of grain (Triticum aestivum) obtained by fermentation with selected lactobacillus and natural yeast strains, we used an in vivo model of hyperglycemia-induced retinal damage, the fruit fly Drosophila melanogaster fed with high-sucrose diet. Lisosan G positively affected the visual system of hyperglycemic flies at structural/functional level, decreased apoptosis, and reactivated protective autophagy at the retina internal network. Also, in high sucrose-fed Drosophila, Lisosan G reduced the levels of brain ROS and retina peroxynitrite. The analysis of oxidative stress-related metabolites suggested 7,8-dihydrofolate, uric acid, dihydroorotate, γ-L-glutamyl-L-cysteine, allantoin, cysteinyl-glycine, and quinolate as key mediators of Lisosan G-induced inhibition of neuronal ROS, along with the upregulation of glutathione system. Of note, Lisosan G may impact oxidative stress and the ensuing retinal cell death, also independently from autophagy, although the autophagy-ROS cross-talk is critical. This study demonstrated that the continuous supplementation with the alimentary integrator Lisosan G exerts a robust and multifaceted antioxidant effect on retinal neurons, thus providing efficacious neuroprotection of hyperglycemic eye.
Collapse
|
12
|
Catalani E, Silvestri F, Bongiorni S, Taddei AR, Fanelli G, Rinalducci S, De Palma C, Perrotta C, Prantera G, Cervia D. Retinal damage in a new model of hyperglycemia induced by high-sucrose diets. Pharmacol Res 2021; 166:105488. [PMID: 33582248 DOI: 10.1016/j.phrs.2021.105488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022]
Abstract
Loss of retinal neurons may precede clinical signs of diabetic retinopathy (DR). We studied for the first time the effects of hyperglycemia on the visual system of the fruit fly Drosophila melanogaster to characterize a model for glucose-induced retinal neurodegeneration, thus complementing more traditional vertebrate systems. Adult flies were fed with increased high-sucrose regimens which did not modify the locomotion ability, muscle phenotype and mobility after 10 days. The increased availability of dietary sucrose induced hyperglycemia and phosphorylation of Akt in fat tissue, without significant effects on adult growth and viability, consistent with the early phase of insulin signaling and a low impact on the overall metabolic profile of flies at short term. Noteworthy, high-sucrose diets significantly decreased Drosophila responsiveness to the light as a consequence of vision defects. Hyperglycemia did not alter the gross anatomical architecture of the external eye phenotype although a progressive damage of photosensitive units was observed. Appreciable levels of cleaved caspase 3 and nitrotyrosine were detected in the internal retina network as well as punctate staining of Light-Chain 3 and p62, and accumulated autophagosomes, indicating apoptotic features, peroxynitrite formation and autophagy turnover defects. In summary, our results in Drosophila support the view that hyperglycemia induced by high-sucrose diets lead to eye defects, apoptosis/autophagy dysregulation, oxidative stress, and visual dysfunctions which are evolutionarily conserved, thus offering a meaningful opportunity of using a simple in vivo model to study the pathophysiology of neuroretinal alterations that develop in patients at the early stages of DR.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Federica Silvestri
- Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Silvia Bongiorni
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Anna Rita Taddei
- Section of Electron Microscopy, Great Equipment Center, Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Giuseppina Fanelli
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Sara Rinalducci
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, via L. Vanvitelli 32, 20129 Milano, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Giorgio Prantera
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy.
| |
Collapse
|
13
|
Alami-Durante H, Cluzeaud M, Bazin D, Vachot C, Kaushik S. Variable impacts of L-arginine or L-NAME during early life on molecular and cellular markers of muscle growth mechanisms in rainbow trout. Comp Biochem Physiol A Mol Integr Physiol 2020; 242:110652. [DOI: 10.1016/j.cbpa.2020.110652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/23/2019] [Accepted: 01/06/2020] [Indexed: 10/25/2022]
|
14
|
Coazzoli M, Napoli A, Roux-Biejat P, De Palma C, Moscheni C, Catalani E, Zecchini S, Conte V, Giovarelli M, Caccia S, Procacci P, Cervia D, Clementi E, Perrotta C. Acid Sphingomyelinase Downregulation Enhances Mitochondrial Fusion and Promotes Oxidative Metabolism in a Mouse Model of Melanoma. Cells 2020; 9:cells9040848. [PMID: 32244541 PMCID: PMC7226741 DOI: 10.3390/cells9040848] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/20/2020] [Accepted: 03/28/2020] [Indexed: 02/07/2023] Open
Abstract
Melanoma is the most severe type of skin cancer. Its unique and heterogeneous metabolism, relying on both glycolysis and oxidative phosphorylation, allows it to adapt to disparate conditions. Mitochondrial function is strictly interconnected with mitochondrial dynamics and both are fundamental in tumour progression and metastasis. The malignant phenotype of melanoma is also regulated by the expression levels of the enzyme acid sphingomyelinase (A-SMase). By modulating at transcriptional level A-SMase in the melanoma cell line B16-F1 cells, we assessed the effect of enzyme downregulation on mitochondrial dynamics and function. Our results demonstrate that A-SMase influences mitochondrial morphology by affecting the expression of mitofusin 1 and OPA1. The enhanced expression of the two mitochondrial fusion proteins, observed when A-SMase is expressed at low levels, correlates with the increase of mitochondrial function via the stimulation of the genes PGC-1alpha and TFAM, two genes that preside over mitochondrial biogenesis. Thus, the reduction of A-SMase expression, observed in malignant melanomas, may determine their metastatic behaviour through the stimulation of mitochondrial fusion, activity and biogenesis, conferring a metabolic advantage to melanoma cells.
Collapse
Affiliation(s)
- Marco Coazzoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Alessandra Napoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
- Unit of Clinical Pharmacology, University Hospital “Luigi Sacco”-ASST Fatebenefratelli Sacco, 20157 Milano, Italy
| | - Paulina Roux-Biejat
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milano, Italy;
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Vincenzo Conte
- Department of Biomedical Sciences for Health (SCIBIS), Università degli Studi di Milano, 20133 Milano, Italy; (V.C.); (P.P.)
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Patrizia Procacci
- Department of Biomedical Sciences for Health (SCIBIS), Università degli Studi di Milano, 20133 Milano, Italy; (V.C.); (P.P.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy
- Correspondence: (E.C.); (C.P.)
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
- Correspondence: (E.C.); (C.P.)
| |
Collapse
|
15
|
Omidi S, Ebrahimi M, Janmohammadi H, Moghaddam G, Rajabi Z, Hosseintabar-Ghasemabad B. The impact of in ovo injection of l-arginine on hatchability, immune system and caecum microflora of broiler chickens. J Anim Physiol Anim Nutr (Berl) 2019; 104:178-185. [PMID: 31587369 DOI: 10.1111/jpn.13222] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/08/2019] [Accepted: 09/10/2019] [Indexed: 02/01/2023]
Abstract
The present article was conducted to evaluate the effect of in ovo injection of arginine on hatchability, immune system and caecum microflora of broiler chickens. For this reason, 300 fertile eggs were used in a completely randomized design with three experimental treatments. The experimental groups included: 1%-0.5% l-arginine (100 eggs), 2%-1% l-arginine (100 eggs), 3- control [included both sham control (injection of distilled water; 50 eggs) and control (no injection; 50 eggs)], which were injected on d 14 of incubation. After hatching, chicks of each experimental group (0.5% l-arginine, 1% l-arginine, and control groups) were randomly divided into four equal groups (as replicates) and reared for 30 days. Weight and feeding of chickens were recorded. Next, blood samples of chickens were collected on day 30 to evaluate antibody titre. Also, chickens were slaughtered on 24 and 30 days of the experiment to evaluate immune system organs and caecum microflora. Based on the results, in ovo injection of l-arginine had no significant effect on hatchability, body weight, antibody titre, spleen, bursa of Fabricius and thymus weight (p > .05). On the other hand, treatments significantly affected feed intake and feed conversion ratio (p < .05). As a novel finding, in ovo injection of l-arginine increased caecal Lactobacillus (p < .01), while decreasing Coliform and Escherichia Coli bacteria (p < .01). However, treatments did not influence caecal Enterococcus (p > .05). The overall results indicated that in ovo injection of 0.5% l-arginine had a better improving effect on caecal microflora and then considered as a recommended level of the present experiment.
Collapse
Affiliation(s)
- Somayeh Omidi
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Marziyeh Ebrahimi
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Hossein Janmohammadi
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Gholamali Moghaddam
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Zolfaghar Rajabi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | | |
Collapse
|
16
|
|
17
|
XIAP as a Target of New Small Organic Natural Molecules Inducing Human Cancer Cell Death. Cancers (Basel) 2019; 11:cancers11091336. [PMID: 31505859 PMCID: PMC6770071 DOI: 10.3390/cancers11091336] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022] Open
Abstract
X-linked inhibitor of apoptosis protein (XIAP) is an emerging crucial therapeutic target in cancer. We report on the discovery and characterisation of small organic molecules from Piper genus plants exhibiting XIAP antagonism, namely erioquinol, a quinol substituted in the 4-position with an alkenyl group and the alkenylphenols eriopodols A–C. Another isolated compound was originally identified as gibbilimbol B. Erioquinol was the most potent inhibitor of human cancer cell viability when compared with gibbilimbol B and eriopodol A was listed as intermediate. Gibbilimbol B and eriopodol A induced apoptosis through mitochondrial permeabilisation and caspase activation while erioquinol acted on cell fate via caspase-independent/non-apoptotic mechanisms, likely involving mitochondrial dysfunctions and aberrant generation of reactive oxygen species. In silico modelling and molecular approaches suggested that all molecules inhibit XIAP by binding to XIAP-baculoviral IAP repeat domain. This demonstrates a novel aspect of XIAP as a key determinant of tumour control, at the molecular crossroad of caspase-dependent/independent cell death pathway and indicates molecular aspects to develop tumour-effective XIAP antagonists.
Collapse
|
18
|
Catalani E, Buonanno F, Lupidi G, Bongiorni S, Belardi R, Zecchini S, Giovarelli M, Coazzoli M, De Palma C, Perrotta C, Clementi E, Prantera G, Marcantoni E, Ortenzi C, Fausto AM, Picchietti S, Cervia D. The Natural Compound Climacostol as a Prodrug Strategy Based on pH Activation for Efficient Delivery of Cytotoxic Small Agents. Front Chem 2019; 7:463. [PMID: 31316972 PMCID: PMC6609918 DOI: 10.3389/fchem.2019.00463] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/11/2019] [Indexed: 12/20/2022] Open
Abstract
We synthesized and characterized MOMO as a new small molecule analog of the cytotoxic natural product climacostol efficiently activated in mild extracellular acidosis. The synthesis of MOMO had a key step in the Wittig olefination for the construction of the carbon-carbon double bond in the alkenyl moiety of climacostol. The possibility of obtaining the target (Z)-alkenyl MOMO derivative in very good yield and without presence of the less active (E)-diastereomer was favored from the methoxymethyl ether (MOM)-protecting group of hydroxyl functions in aromatic ring of climacostol aldehyde intermediate. Of interest, the easy removal of MOM-protecting group in a weakly acidic environment allowed us to obtain a great quantity of climacostol in biologically active (Z)-configuration. Results obtained in free-living ciliates that share the same micro-environment of the climacostol natural producer Climacostomum virens demonstrated that MOMO is well-tolerated in a physiological environment, while its cytotoxicity is rapidly and efficiently triggered at pH 6.3. In addition, the cytostatic vs. cytotoxic effects of acidified-MOMO can be modulated in a dose-dependent manner. In mouse melanoma cells, MOMO displayed a marked pH-sensitivity since its cytotoxic and apoptotic effects become evident only in mild extracellular acidosis. Data also suggested MOMO being preferentially activated in the unique extra-acidic microenvironment that characterizes tumoural cells. Finally, the use of the model organism Drosophila melanogaster fed with an acidic diet supported the efficient activity and oral delivery of MOMO molecule in vivo. MOMO affected oviposition of mating adults and larvae eclosion. Reduced survival of flies was due to lethality during the larval stages while emerging larvae retained their ability to develop into adults. Interestingly, the gut of eclosed larvae exhibited an extended damage (cell death by apoptosis) and the brain tissue was also affected (reduced mitosis), demonstrating that orally activated MOMO efficiently targets different tissues of the developing fly. These results provided a proof-of-concept study on the pH-dependence of MOMO effects. In this respect, MOM-protection emerges as a potential prodrug strategy which deserves to be further investigated for the generation of efficient pH-sensitive small organic molecules as pharmacologically active cytotoxic compounds.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems, Università degli Studi della Tuscia, Viterbo, Italy
| | - Federico Buonanno
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage and Tourism, Università degli Studi di Macerata, Macerata, Italy
| | - Gabriele Lupidi
- School of Sciences and Technologies, Section of Chemistry, Università degli Studi di Camerino, Camerino, Italy
| | - Silvia Bongiorni
- Department of Ecological and Biological Sciences, Università degli Studi della Tuscia, Viterbo, Italy
| | - Riccardo Belardi
- Department for Innovation in Biological, Agro-Food and Forest Systems, Università degli Studi della Tuscia, Viterbo, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Marco Coazzoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Clara De Palma
- Unit of Clinical Pharmacology, University Hospital “Luigi Sacco”-ASST Fatebenefratelli Sacco, Milan, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Milan, Italy
- Scientific Institute IRCCS “Eugenio Medea”, Bosisio Parini, Italy
| | - Giorgio Prantera
- Department of Ecological and Biological Sciences, Università degli Studi della Tuscia, Viterbo, Italy
| | - Enrico Marcantoni
- School of Sciences and Technologies, Section of Chemistry, Università degli Studi di Camerino, Camerino, Italy
| | - Claudio Ortenzi
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage and Tourism, Università degli Studi di Macerata, Macerata, Italy
| | - Anna Maria Fausto
- Department for Innovation in Biological, Agro-Food and Forest Systems, Università degli Studi della Tuscia, Viterbo, Italy
| | - Simona Picchietti
- Department for Innovation in Biological, Agro-Food and Forest Systems, Università degli Studi della Tuscia, Viterbo, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems, Università degli Studi della Tuscia, Viterbo, Italy
| |
Collapse
|
19
|
Buonanno F, Catalani E, Cervia D, Proietti Serafini F, Picchietti S, Fausto AM, Giorgi S, Lupidi G, Rossi FV, Marcantoni E, Petrelli D, Ortenzi C. Bioactivity and Structural Properties of Novel Synthetic Analogues of the Protozoan Toxin Climacostol. Toxins (Basel) 2019; 11:toxins11010042. [PMID: 30650514 PMCID: PMC6356496 DOI: 10.3390/toxins11010042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/31/2022] Open
Abstract
Climacostol (5-[(2Z)-non-2-en-1-yl]benzene-1,3-diol) is a resorcinol produced by the protozoan Climacostomum virens for defence against predators. It exerts a potent antimicrobial activity against bacterial and fungal pathogens, inhibits the growth of several human and rodent tumour cells, and is now available by chemical synthesis. In this study, we chemically synthesized two novel analogues of climacostol, namely, 2-methyl-5 [(2Z)-non-2-en-1-yl]benzene-1,3-diol (AN1) and 5-[(2Z)-non-2-en-1-yl]benzene-1,2,3-triol (AN2), with the aim to increase the activity of the native toxin, evaluating their effects on prokaryotic and free-living protists and on mammalian tumour cells. The results demonstrated that the analogue bearing a methyl group (AN1) in the aromatic ring exhibited appreciably higher toxicity against pathogen microbes and protists than climacostol. On the other hand, the analogue bearing an additional hydroxyl group (AN2) in the aromatic ring revealed its ability to induce programmed cell death in protistan cells. Overall, the data collected demonstrate that the introduction of a methyl or a hydroxyl moiety to the aromatic ring of climacostol can effectively modulate its potency and its mechanism of action.
Collapse
Affiliation(s)
- Federico Buonanno
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage, and Tourism (ECHT), Università degli Studi di Macerata, 62100 Macerata, Italy.
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy.
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy.
| | - Francesca Proietti Serafini
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy.
| | - Simona Picchietti
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy.
| | - Anna Maria Fausto
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy.
| | - Simone Giorgi
- School of Sciences and Technologies, Section of Chemistry, Università degli Studi di Camerino, 62032 Camerino, Italy.
| | - Gabriele Lupidi
- School of Sciences and Technologies, Section of Chemistry, Università degli Studi di Camerino, 62032 Camerino, Italy.
| | - Federico Vittorio Rossi
- School of Sciences and Technologies, Section of Chemistry, Università degli Studi di Camerino, 62032 Camerino, Italy.
| | - Enrico Marcantoni
- School of Sciences and Technologies, Section of Chemistry, Università degli Studi di Camerino, 62032 Camerino, Italy.
| | - Dezemona Petrelli
- School of Biosciences and Veterinary Medicine, Università degli Studi di Camerino, 62032 Camerino, Italy.
| | - Claudio Ortenzi
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage, and Tourism (ECHT), Università degli Studi di Macerata, 62100 Macerata, Italy.
| |
Collapse
|
20
|
Dysfunctional autophagy induced by the pro-apoptotic natural compound climacostol in tumour cells. Cell Death Dis 2018; 10:10. [PMID: 30584259 PMCID: PMC6315039 DOI: 10.1038/s41419-018-1254-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/06/2018] [Accepted: 12/03/2018] [Indexed: 01/22/2023]
Abstract
Autophagy occurs at a basal level in all eukaryotic cells and may support cell survival or activate death pathways. Due to its pathophysiologic significance, the autophagic machinery is a promising target for the development of multiple approaches for anti-neoplastic agents. We have recently described the cytotoxic and pro-apoptotic mechanisms, targeting the tumour suppressor p53, of climacostol, a natural product of the ciliated protozoan Climacostomum virens. We report here on how climacostol regulates autophagy and the involvement of p53-dependent mechanisms. Using both in vitro and in vivo techniques, we show that climacostol potently and selectively impairs autophagy in multiple tumour cells that are committed to die by apoptosis. In particular, in B16-F10 mouse melanomas climacostol exerts a marked and sustained accumulation of autophagosomes as the result of dysfunctional autophagic degradation. We also provide mechanistic insights showing that climacostol affects autophagosome turnover via p53-AMPK axis, although the mTOR pathway unrelated to p53 levels plays a role. In particular, climacostol activated p53 inducing the upregulation of p53 protein levels in the nuclei through effects on p53 stability at translational level, as for instance the phosphorylation at Ser15 site. Noteworthy, AMPKα activation was the major responsible of climacostol-induced autophagy disruption in the absence of a key role regulating cell death, thus indicating that climacostol effects on autophagy and apoptosis are two separate events, which may act independently on life/death decisions of the cell. Since the activation of p53 system is at the molecular crossroad regulating both the anti-autophagic action of climacostol and its role in the apoptosis induction, it might be important to explore the dual targeting of autophagy and apoptosis with agents acting on p53 for the selective killing of tumours. These findings also suggest the efficacy of ciliate bioactive molecules to identify novel lead compounds in drug discovery and development.
Collapse
|
21
|
Dolgorukova AM, Titov VY, Petrov VA, Osipov AN, Slesarenko NA, Kochish II. Mechanisms of Specific Embryonic Effects of Nitrogen Oxide. Bull Exp Biol Med 2018; 165:635-639. [PMID: 30225706 DOI: 10.1007/s10517-018-4230-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Indexed: 11/28/2022]
Abstract
The study of NO metabolism in chicken embryos showed that the intensity of oxidation of both endogenous and exogenous for the embryo NO donors to nitrate is determined by the presence or state of NO targets, rather than donor concentration. The mechanism of this oxidation and its physiological role are discussed. It was also shown that oxidation product nitrate is actively eliminated from the amnionic sac.
Collapse
Affiliation(s)
- A M Dolgorukova
- Federal Research Centre All-Russian Research and Technology Institute of Poultry Industry, Russian Academy of Sciences, Moscow, Russia
| | - V Yu Titov
- Federal Research Centre All-Russian Research and Technology Institute of Poultry Industry, Russian Academy of Sciences, Moscow, Russia. .,N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - V A Petrov
- N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A N Osipov
- N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N A Slesarenko
- K. I. Skryabin Moscow State Academy of Veterinary and Biotechnology, Moscow, Russia
| | - I I Kochish
- K. I. Skryabin Moscow State Academy of Veterinary and Biotechnology, Moscow, Russia
| |
Collapse
|
22
|
Cervia D, Assi E, De Palma C, Giovarelli M, Bizzozero L, Pambianco S, Di Renzo I, Zecchini S, Moscheni C, Vantaggiato C, Procacci P, Clementi E, Perrotta C. Essential role for acid sphingomyelinase-inhibited autophagy in melanoma response to cisplatin. Oncotarget 2018; 7:24995-5009. [PMID: 27107419 PMCID: PMC5041885 DOI: 10.18632/oncotarget.8735] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/28/2016] [Indexed: 01/03/2023] Open
Abstract
The sphingolipid metabolising enzyme Acid Sphingomyelinase (A-SMase) has been recently shown to inhibit melanoma progression and correlate inversely to tumour grade. In this study we have investigated the role of A-SMase in the chemo-resistance to anticancer treatmentusing mice with melanoma allografts and melanoma cells differing in terms of expression/activity of A-SMase. Since autophagy is emerging as a key mechanism in tumour growth and chemo-resistance, we have also investigated whether an action of A-SMase in autophagy can explain its role. Melanoma sensitivity to chemotherapeutic agent cisplatin in terms of cell viability/apoptosis, tumour growth, and animal survival depended directly on the A-SMase levels in tumoural cells. A-SMase action was due to inhibition of autophagy through activation of Akt/mammalian target of rapamycin (mTOR) pathway. Treatment of melanoma-bearing mice with the autophagy inhibitor chloroquine restored sensitivity to cisplatin of tumours expressing low levels of A-SMase while no additive effects were observed in tumours characterised by sustained A-SMase levels. The fact that A-SMase in melanomas affects mTOR-regulated autophagy and plays a central role in cisplatin efficacy encourages pre-clinical testing on the modulation of A-SMase levels/activity as possible novel anti-neoplastic strategy.
Collapse
Affiliation(s)
- Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Emma Assi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy.,Present address: Division of Experimental Oncology, San Raffaele Scientific Institute, Milano, Italy
| | - Clara De Palma
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy.,Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, University Hospital "Luigi Sacco", Milano, Italy
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Laura Bizzozero
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy.,Present address: Department of Oncology, Università degli Studi di Torino and Laboratory of Neurovascular Biology, Candiolo Cancer Institute, Candiolo, Italy
| | - Sarah Pambianco
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Ilaria Di Renzo
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| | | | - Patrizia Procacci
- Department of Biomedical Sciences for Health (SCIBIS), Università degli Studi di Milano, Milano, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy.,Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy.,Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, University Hospital "Luigi Sacco", Milano, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
23
|
Cammalleri M, Locri F, Catalani E, Filippi L, Cervia D, Dal Monte M, Bagnoli P. The Beta Adrenergic Receptor Blocker Propranolol Counteracts Retinal Dysfunction in a Mouse Model of Oxygen Induced Retinopathy: Restoring the Balance between Apoptosis and Autophagy. Front Cell Neurosci 2017; 11:395. [PMID: 29375312 PMCID: PMC5770647 DOI: 10.3389/fncel.2017.00395] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/28/2017] [Indexed: 12/29/2022] Open
Abstract
In a mouse model of oxygen induced retinopathy (OIR), beta adrenergic receptor (BAR) blockade has been shown to recover hypoxia-associated retinal damages. Although the adrenergic signaling is an important regulator of apoptotic and autophagic processes, the role of BARs in retinal cell death remains to be elucidated. The present study was aimed at investigating whether ameliorative effects of BAR blockers may occur through their coordinated action on apoptosis and autophagy. To this aim, retinas from control and OIR mice untreated or treated with propranolol, a non-selective BAR1/2 blocker, were characterized in terms of expression and localization of apoptosis and autophagy markers. The effects of propranolol on autophagy signaling were also evaluated and specific autophagy modulators were used to get functional information on the autophagic effects of BAR antagonism. Finally, propranolol effects on neurodegenerative processes were associated to an electrophysiological investigation of retinal function by recording electroretinogram (ERG). We found that retinas of OIR mice are characterized by increased apoptosis and decreased autophagy, while propranolol reduces apoptosis and stimulates autophagy. In particular, propranolol triggers autophagosome formation in bipolar, amacrine and ganglion cells that are committed to die by apoptosis in response to hypoxia. Also our data argue that propranolol, through the inhibition of the Akt-mammalian target of rapamycin pathway, activates autophagy which decreases retinal cell death. At the functional level, propranolol recovers dysfunctional ERG by recovering the amplitude of a- and b-waves, and oscillatory potentials, thus indicating an efficient restoring of retinal transduction. Overall, our results demonstrate that BAR1/2 are key regulators of retinal apoptosis/autophagy, and that BAR1/2 blockade leads to autophagy-mediated neuroprotection. Reinstating the balance between apoptotic and autophagic machines may therefore be viewed as a future goal in the treatment of retinopathies.
Collapse
Affiliation(s)
| | - Filippo Locri
- Department of Biology, University of Pisa, Pisa, Italy
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, Viterbo, Italy
| | - Luca Filippi
- Neonatal Intensive Care Unit, Medical Surgical Fetal-Neonatal Department, Meyer University Children's Hospital, Florence, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, Viterbo, Italy
| | | | - Paola Bagnoli
- Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
24
|
Yamashita AMS, Ancillotti MTC, Rangel LP, Fontenele M, Figueiredo-Freitas C, Possidonio AC, Soares CP, Sorenson MM, Mermelstein C, Nogueira L. Balance between S-nitrosylation and denitrosylation modulates myoblast proliferation independently of soluble guanylyl cyclase activation. Am J Physiol Cell Physiol 2017; 313:C11-C26. [PMID: 28381519 DOI: 10.1152/ajpcell.00140.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 03/13/2017] [Accepted: 03/31/2017] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) contributes to myogenesis by regulating the transition between myoblast proliferation and fusion through cGMP signaling. NO can form S-nitrosothiols (RSNO), which control signaling pathways in many different cell types. However, neither the role of RSNO content nor its regulation by the denitrosylase activity of S-nitrosoglutathione reductase (GSNOR) during myogenesis is understood. Here, we used primary cultures of chick embryonic skeletal muscle cells to investigate whether changes in intracellular RSNO alter proliferation and fusion of myoblasts in the presence and absence of cGMP. Cultures were grown to fuse most of the myoblasts into myotubes, with and without S-nitrosocysteine (CysNO), 8-Br-cGMP, DETA-NO, or inhibitors for NO synthase (NOS), GSNOR, soluble guanylyl cyclase (sGC), or a combination of these, followed by analysis of GSNOR activity, protein expression, RSNO, cGMP, and cell morphology. Although the activity of GSNOR increased progressively over 72 h, inhibiting GSNOR (by GSNOR inhibitor - GSNORi - or by knocking down GSNOR with siRNA) produced an increase in RSNO and in the number of myoblasts and fibroblasts, accompanied by a decrease in myoblast fusion index. This was also detected with CysNO supplementation. Enhanced myoblast number was proportional to GSNOR inhibition. Effects of the GSNORi and GSNOR knockdown were blunted by NOS inhibition, suggesting their dependence on NO synthesis. Interestingly, GSNORi and GSNOR knockdown reversed the attenuated proliferation obtained with sGC inhibition in myoblasts, but not in fibroblasts. Hence myoblast proliferation is enhanced by increasing RSNO, and regulated by GSNOR activity, independently of cGMP production and signaling.
Collapse
Affiliation(s)
- Aline M S Yamashita
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maryana T C Ancillotti
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana P Rangel
- Departamento de Análise Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; and
| | - Marcio Fontenele
- Laboratório de Biologia Molecular do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cicero Figueiredo-Freitas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana C Possidonio
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina P Soares
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martha M Sorenson
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia Mermelstein
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Nogueira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil;
| |
Collapse
|
25
|
Tsao J, Kovanecz I, Awadalla N, Gelfand R, Sinha-Hikim I, White RA, Gonzalez-Cadavid NF. Muscle Derived Stem Cells Stimulate Muscle Myofiber Repair and Counteract Fat Infiltration in a Diabetic Mouse Model of Critical Limb Ischemia. ACTA ACUST UNITED AC 2016; 6. [PMID: 28217409 PMCID: PMC5313052 DOI: 10.4172/2157-7633.1000370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Critical Limb Ischemia (CLI) affects patients with Type 2 Diabetes (T2D) and obesity, with high risk of amputation and post-surgical mortality, and no effective medical treatment. Stem cell therapy, mainly with bone marrow mesenchymal, adipose derived, endothelial, hematopoietic, and umbilical cord stem cells, is promising in CLI mouse and rat models and is in clinical trials. Their general focus is on angiogenic repair, with no reports on the alleviation of necrosis, lipofibrosis, and myofiber regeneration in the ischemic muscle, or the use of Muscle Derived Stem Cells (MDSC) alone or in combination with pharmacological adjuvants, in the context of CLI in T2D. Methods Using a T2D mouse model of CLI induced by severe unilateral femoral artery ligation, we tested: a) the repair efficacy of MDSC implanted into the ischemic muscle and the effects of concurrent intraperitoneal administration of a nitric oxide generator, molsidomine; and b) whether MDSC may partially counteract their own repair effects by stimulating the expression of myostatin, the main lipofibrotic agent in the muscle and inhibitor of muscle mass. Results MDSC: a) reduced mortality, and b) in the ischemic muscle, increased stem cell number and myofiber central nuclei, reduced fat infiltration, myofibroblast number, and myofiber apoptosis, and increased smooth muscle and endothelial cells, as well as neurotrophic factors. The content of myosin heavy chain 2 (MHC-2) myofibers was not restored and collagen was increased, in association with myostatin overexpression. Supplementation of MDSC with molsidomine failed to stimulate the beneficial effects of MDSC, except for some reduction in myostatin overexpression. Molsidomine given alone was rather ineffective, except for inhibiting apoptosis and myostatin overexpression. Conclusions MDSC improved CLI muscle repair, but molsidomine did not stimulate this process. The combination of MDSC with anti-myostatin approaches should be explored to restore myofiber MHC composition.
Collapse
Affiliation(s)
- J Tsao
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | - I Kovanecz
- Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - N Awadalla
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | - R Gelfand
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA; Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - I Sinha-Hikim
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | - R A White
- Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - N F Gonzalez-Cadavid
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA; Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA; Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
26
|
Tirone M, Conti V, Manenti F, Nicolosi PA, D’Orlando C, Azzoni E, Brunelli S. Nitric Oxide Donor Molsidomine Positively Modulates Myogenic Differentiation of Embryonic Endothelial Progenitors. PLoS One 2016; 11:e0164893. [PMID: 27760216 PMCID: PMC5070765 DOI: 10.1371/journal.pone.0164893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 10/03/2016] [Indexed: 01/07/2023] Open
Abstract
Embryonic VE-Cadherin-expressing progenitors (eVE-Cad+), including hemogenic endothelium, have been shown to generate hematopoietic stem cells and a variety of other progenitors, including mesoangioblasts, or MABs. MABs are vessel-associated progenitors with multilineage mesodermal differentiation potential that can physiologically contribute to skeletal muscle development and regeneration, and have been used in an ex vivo cell therapy setting for the treatment of muscular dystrophy. There is currently a therapeutic need for molecules that could improve the efficacy of cell therapy protocols; one such good candidate is nitric oxide. Several studies in animal models of muscle dystrophy have demonstrated that nitric oxide donors provide several beneficial effects, including modulation of the activity of endogenous cell populations involved in muscle repair and the delay of muscle degeneration. Here we used a genetic lineage tracing approach to investigate whether the therapeutic effect of nitric oxide in muscle repair could derive from an improvement in the myogenic differentiation of eVE-Cad+ progenitors during embryogenesis. We show that early in vivo treatment with the nitric oxide donor molsidomine enhances eVE-Cad+ contribution to embryonic and fetal myogenesis, and that this effect could originate from a modulation of the properties of yolk sac hemogenic endothelium.
Collapse
Affiliation(s)
- Mario Tirone
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Conti
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Manenti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Cristina D’Orlando
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Emanuele Azzoni
- MRC Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- * E-mail: (SB); (EA)
| | - Silvia Brunelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- * E-mail: (SB); (EA)
| |
Collapse
|
27
|
Catalani E, Proietti Serafini F, Zecchini S, Picchietti S, Fausto AM, Marcantoni E, Buonanno F, Ortenzi C, Perrotta C, Cervia D. Natural products from aquatic eukaryotic microorganisms for cancer therapy: Perspectives on anti-tumour properties of ciliate bioactive molecules. Pharmacol Res 2016; 113:409-420. [PMID: 27650755 DOI: 10.1016/j.phrs.2016.09.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/12/2016] [Accepted: 09/16/2016] [Indexed: 11/27/2022]
Abstract
Several modern drugs, including those for cancer therapy, have been isolated from natural sources, are based on natural products and its derivatives, or mime natural products. Some of them are in clinical use, others in clinical trials. The success of natural products in drug discovery is related to their biochemical characteristics and to the technologic methods used to study their feature. Natural compounds may acts as chemo-preventive agents and as factors that increase therapeutic efficacy of existing drugs, thus overcoming cancer cell drug resistance that is the main factor determining the failure in conventional chemotherapy. Water environment, because of its physical and chemical conditions, shows an extraordinary collection of natural biological substances with an extensive structural and functional diversity. The isolation of bioactive molecules has been reported from a great variety of aquatic organisms; however, the therapeutic application of molecules from eukaryotic microorganisms remains inadequately investigated and underexploited on a systematic basis. Herein we describe the biological activities in mammalian cells of selected substances isolated from ciliates, free-living protozoa common almost everywhere there is water, focusing on their anti-tumour actions and their possible therapeutic activity. In particular, we unveil the cellular and molecular machine mediating the effects of cell type-specific signalling protein pheromone Er-1 and secondary metabolites, i.e. euplotin C and climacostol, in cancer cells. To support the feasibility of climacostol-based approaches, we also present novel findings and report additional mechanisms of action using both in vitro and in vivo models of mouse melanomas, with the scope of highlighting new frontiers that can be explored also in a therapeutic perspective. The high skeletal chemical difference of ciliate compounds, their sustainability and availability, also through the use of new organic synthesis/modifications processes, and the results obtained so far in biological studies provide a rationale to consider some of them a potential resource for the design of new anti-cancer drugs.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Francesca Proietti Serafini
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Silvia Zecchini
- Unit of Clinical Pharmacology, University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco, Milano, Italy
| | - Simona Picchietti
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Anna Maria Fausto
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Enrico Marcantoni
- School of Sciences and Technologies, Section of Chemistry, Università degli Studi di Camerino, Italy
| | - Federico Buonanno
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage and Tourism, Università degli Studi di Macerata, Italy
| | - Claudio Ortenzi
- Laboratory of Protistology and Biology Education, Department of Education, Cultural Heritage and Tourism, Università degli Studi di Macerata, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Italy.
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy; Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Italy.
| |
Collapse
|
28
|
Climacostol reduces tumour progression in a mouse model of melanoma via the p53-dependent intrinsic apoptotic programme. Sci Rep 2016; 6:27281. [PMID: 27271364 PMCID: PMC4895139 DOI: 10.1038/srep27281] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022] Open
Abstract
Climacostol, a compound produced by the ciliated protozoan Climacostomum virens, displayed cytotoxic properties in vitro. This study demonstrates that it has anti-tumour potential. Climacostol caused a reduction of viability/proliferation of B16-F10 mouse melanoma cells, a rapidly occurring DNA damage, and induced the intrinsic apoptotic pathway characterised by the dissipation of the mitochondrial membrane potential, the translocation of Bax to the mitochondria, the release of Cytochrome c from the mitochondria, and the activation of Caspase 9-dependent cleavage of Caspase 3. The apoptotic mechanism of climacostol was found to rely on the up-regulation of p53 and its targets Noxa and Puma. In vivo analysis of B16-F10 allografts revealed a persistent inhibition of tumour growth rate when melanomas were treated with intra-tumoural injections of climacostol. In addition, it significantly improved the survival of transplanted mice, decreased tumour weight, induced a remarkable reduction of viable cells inside the tumour, activated apoptosis and up-regulated the p53 signalling network. Importantly, climacostol toxicity was more selective against tumour than non-tumour cells. The anti-tumour properties of climacostol and the molecular events associated with its action indicate that it is a powerful agent that may be considered for the design of pro-apoptotic drugs for melanoma therapy.
Collapse
|
29
|
Cai C, Qin X, Wu Z, Shen Q, Yang W, Zhang S, Duan J, Liang F, Liu C. Inhibitory effect of MyoD on the proliferation of breast cancer cells. Oncol Lett 2016; 11:3589-3596. [PMID: 27284360 PMCID: PMC4887810 DOI: 10.3892/ol.2016.4448] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 02/24/2016] [Indexed: 12/31/2022] Open
Abstract
Skeletal muscle is rich in lymphatic vessels, with an abundant blood supply, and it is an infrequent site of cancer metastasis. Previous studies have demonstrated that enhanced secretion of MyoD may occur when skeletal muscle is injured or becomes cancerous. It was hypothesized that MyoD may act as an endogenous cytokine to inhibit the proliferation of cancer cells. To verify the possible effect of this protein on tumor cell proliferation, C2C12 mouse skeletal muscle cells and 4T1 mouse breast cancer cells were co-cultured using embedded Transwell plates. Following co-culture, cell cycle analysis revealed that C2C12 muscle cells were able to inhibit the proliferation of the breast cancer cells. Subsequently, MyoD was silenced in C2C12 cells to assess its effect on 4T1 cell proliferation. Following co-culture with MyoD-silenced cells, a 5-ethynyl-20-deoxyuridine assay indicated that MyoD silencing prevented the reduction in proliferation of 4T1 cells induced by untransfected C2C12 cells. In summary, the results indicated that MyoD inhibits the proliferation of breast cancer cells and may be a tumor suppressor factor.
Collapse
Affiliation(s)
- Changjing Cai
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Xiaoqun Qin
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Ziyi Wu
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Qixia Shen
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Wenqian Yang
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Shujun Zhang
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Jinling Duan
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Fenglan Liang
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Chi Liu
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
30
|
S-Nitrosylated fetal hemoglobin in neonatal human blood. Biochem Biophys Res Commun 2016; 473:1084-1089. [PMID: 27060546 DOI: 10.1016/j.bbrc.2016.04.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/05/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND Nitric oxide (NO) and its derivatives play important roles in the cardiopulmonary transition upon birth and in other oxygen-sensitive developmental milestones. One mechanism for the coupling of oxygen sensing and signaling by NO species is via the formation of an S-nitrosothiol (SNO) moiety on hemoglobin (Hb, forming SNO-Hb) and its release from the red blood cell in hypoxia. Although SNO-Hb formed on adult-type Hb (HbA, forming SNO-HbA) has been documented in physiological and pathophysiological human states, the fetal variant, SNO-HbF, has thus far not been isolated or characterized in human blood. METHODS AND RESULTS We developed a technique capable of separating Hbs A and F under conditions that preserve SNO. We then measured SNO-HbF in the blood of healthy and premature or otherwise ill neonates using the gold standard for SNO measurement, mercury-coupled photolysis-chemiluminescence. SNO-HbF levels were in the range of those previously reported for HbA in adults. We found that SNO-HbF was more abundant at earlier gestational age (<30 weeks), even when accounting for the absolute HbF level. CONCLUSIONS The ability to monitor SNO-HbF could provide new insights into fetal development and the perinatal transition, and has potential as a biomarker relevant to the management of neonatal diseases.
Collapse
|
31
|
Titov VY, Kosenko OV, Starkova ES, Kondratov GV, Borkhunova EN, Ivanova AV. Effect of Green Light on Nitric Oxide Metabolism in Chick Embryos. A Possible Physiological Role. Bull Exp Biol Med 2015; 159:700-3. [PMID: 26519269 DOI: 10.1007/s10517-015-3052-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Indexed: 12/27/2022]
Abstract
The exposure to green light, which serves as a well-known activating factor for myogenesis during incubation of chicken eggs, contributes to intensification of embryonic metabolism of NO. A metabolic product, nitrate, is mainly accumulated in the muscles. These data suggest that light induces a NO-dependent activation of the factor, which intensifies muscle tissue development.
Collapse
Affiliation(s)
- V Yu Titov
- All-Russian Research and Technology Poultry Institute, Sergiev Posad, Moscow region, Russia.
| | - O V Kosenko
- All-Russian Research and Technology Poultry Institute, Sergiev Posad, Moscow region, Russia
| | - E S Starkova
- All-Russian Research and Technology Poultry Institute, Sergiev Posad, Moscow region, Russia
| | - G V Kondratov
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Ministry of Agriculture of the Russian Federation, Moscow, Russia
| | - E N Borkhunova
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Ministry of Agriculture of the Russian Federation, Moscow, Russia
| | - A V Ivanova
- N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
32
|
Perrotta C, De Palma C, Clementi E, Cervia D. Hormones and immunity in cancer: are thyroid hormones endocrine players in the microglia/glioma cross-talk? Front Cell Neurosci 2015; 9:236. [PMID: 26157361 PMCID: PMC4477169 DOI: 10.3389/fncel.2015.00236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 06/11/2015] [Indexed: 11/16/2022] Open
Abstract
Accumulating evidence indicates that the endocrine and immune systems engage in complex cross-talks in which a prominent role is played by thyroid hormones (THs). The increase of resident vs. monocyte recruited macrophages was shown to be an important effector of the TH 3,3′,5′-Triiodo-L-thyronine (T3)-induced protection against inflammation and a key role of T3 in inhibiting the differentiation of peripheral monocytes into macrophages was observed. Herein, we report on the role of T3 as a modulator of microglia, the specialized macrophages of the central nervous system (CNS). Mounting evidence supports a role of microglia and macrophages in the growth and invasion of malignant glioma. In this respect, we unveil the putative involvement of T3 in the microglia/glioma cell communication. Since THs are known to cross the blood-brain barrier, we suggest that T3 not only exerts a direct modulation of brain cancer cell itself but also indirectly promotes glioma growth through a modulation of microglia. Our observations expand available information on the role of TH system in glioma and its microenvironment and highlight the endocrine modulation of microglia as an important target for future therapeutic development of glioma treatments.
Collapse
Affiliation(s)
- Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, University Hospital "Luigi Sacco", Università di Milano Milano, Italy
| | - Clara De Palma
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, University Hospital "Luigi Sacco", Università di Milano Milano, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, University Hospital "Luigi Sacco", Università di Milano Milano, Italy ; Scientific Institute IRCCS Eugenio Medea Bosisio Parini, Italy
| | - Davide Cervia
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, University Hospital "Luigi Sacco", Università di Milano Milano, Italy ; Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università della Tuscia, Largo dell'Università snc Viterbo, Italy
| |
Collapse
|
33
|
Modulation of Acid Sphingomyelinase in Melanoma Reprogrammes the Tumour Immune Microenvironment. Mediators Inflamm 2015; 2015:370482. [PMID: 26101462 PMCID: PMC4460251 DOI: 10.1155/2015/370482] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 12/30/2022] Open
Abstract
The inflammatory microenvironment induces tumours to acquire an aggressive and immunosuppressive behaviour. Since acid sphingomyelinase (A-SMase) downregulation in melanoma was shown to determine a malignant phenotype, we aimed here to elucidate the role of A-SMase in the regulation of tumour immunogenic microenvironment using in vivo melanoma models in which A-SMase was either downregulated or maintained at constitutively high levels. We found high levels of inflammatory factors in low A-SMase expressing tumours, which also displayed an immunosuppressive/protumoural microenvironment: high levels of myeloid-derived suppressor cells (MDSCs) and regulatory T lymphocytes (Tregs), as well as low levels of dendritic cells (DCs). In contrast, the restoration of A-SMase in melanoma cells not only reduced tumour growth and immunosuppression, but also induced a high recruitment at tumour site of effector immune cells with an antitumoural function. Indeed, we observed a poor homing of MDSCs and Tregs and the increased recruitment of CD8+ and CD4+ T lymphocytes as well as the infiltration of DCs and CD8+/CD44high T lymphocytes. This study demonstrates that change of A-SMase expression in cancer cells is sufficient per se to tune in vivo melanoma growth and that A-SMase levels modulate immune cells at tumour site. This may be taken into consideration in the setting of therapeutic strategies.
Collapse
|
34
|
Titov VY, Kondratov GV, Ivanova AV. Specific role of nitric oxide (NO) in avian embryonic myogenesis. Bull Exp Biol Med 2015; 158:508-12. [PMID: 25708336 DOI: 10.1007/s10517-015-2796-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Indexed: 11/25/2022]
Abstract
NO plays a specific role in avian embryogenesis stimulating the development of muscle tissue. The main consumer of NO synthesized at the initial stage of avian embryogenesis is presumably a factor stimulating myogenesis.
Collapse
Affiliation(s)
- V Yu Titov
- All-Russian Research and Technological Institute of Poultry Farming, The Russian Academy of Agricultural Sciences, Sergiev Posad, Moscow Region, Russia,
| | | | | |
Collapse
|
35
|
De Palma C, Morisi F, Pambianco S, Assi E, Touvier T, Russo S, Perrotta C, Romanello V, Carnio S, Cappello V, Pellegrino P, Moscheni C, Bassi MT, Sandri M, Cervia D, Clementi E. Deficient nitric oxide signalling impairs skeletal muscle growth and performance: involvement of mitochondrial dysregulation. Skelet Muscle 2014; 4:22. [PMID: 25530838 PMCID: PMC4272808 DOI: 10.1186/s13395-014-0022-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/18/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Nitric oxide (NO), generated in skeletal muscle mostly by the neuronal NO synthases (nNOSμ), has profound effects on both mitochondrial bioenergetics and muscle development and function. The importance of NO for muscle repair emerges from the observation that nNOS signalling is defective in many genetically diverse skeletal muscle diseases in which muscle repair is dysregulated. How the effects of NO/nNOSμ on mitochondria impact on muscle function, however, has not been investigated yet. METHODS In this study we have examined the relationship between the NO system, mitochondrial structure/activity and skeletal muscle phenotype/growth/functions using a mouse model in which nNOSμ is absent. Also, NO-induced effects and the NO pathway were dissected in myogenic precursor cells. RESULTS We show that nNOSμ deficiency in mouse skeletal muscle leads to altered mitochondrial bioenergetics and network remodelling, and increased mitochondrial unfolded protein response (UPR(mt)) and autophagy. The absence of nNOSμ is also accompanied by an altered mitochondrial homeostasis in myogenic precursor cells with a decrease in the number of myonuclei per fibre and impaired muscle development at early stages of perinatal growth. No alterations were observed, however, in the overall resting muscle structure, apart from a reduced specific muscle mass and cross sectional areas of the myofibres. Investigating the molecular mechanisms we found that nNOSμ deficiency was associated with an inhibition of the Akt-mammalian target of rapamycin pathway. Concomitantly, the Akt-FoxO3-mitochondrial E3 ubiquitin protein ligase 1 (Mul-1) axis was also dysregulated. In particular, inhibition of nNOS/NO/cyclic guanosine monophosphate (cGMP)/cGMP-dependent-protein kinases induced the transcriptional activity of FoxO3 and increased Mul-1 expression. nNOSμ deficiency was also accompanied by functional changes in muscle with reduced muscle force, decreased resistance to fatigue and increased degeneration/damage post-exercise. CONCLUSIONS Our results indicate that nNOSμ/NO is required to regulate key homeostatic mechanisms in skeletal muscle, namely mitochondrial bioenergetics and network remodelling, UPR(mt) and autophagy. These events are likely associated with nNOSμ-dependent impairments of muscle fibre growth resulting in a deficit of muscle performance.
Collapse
Affiliation(s)
- Clara De Palma
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Federica Morisi
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Sarah Pambianco
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Emma Assi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Thierry Touvier
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Stefania Russo
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Cristiana Perrotta
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Vanina Romanello
- Dulbecco Telethon Institute at Venetian Institute of Molecular Medicine, Padova, Italy
| | - Silvia Carnio
- Dulbecco Telethon Institute at Venetian Institute of Molecular Medicine, Padova, Italy
| | - Valentina Cappello
- National Research Council-Institute of Neuroscience, Department of Medical Biotechnology and Translational Medicine, Università di Milano, Milano, Italy ; CNI@NEST, Italian Institute of Technology, Pisa, Italy
| | - Paolo Pellegrino
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy
| | - Claudia Moscheni
- Unit of Morphology, Department of Biomedical and Clinical Sciences "Luigi Sacco", Università di Milano, Milano, Italy
| | | | - Marco Sandri
- Dulbecco Telethon Institute at Venetian Institute of Molecular Medicine, Padova, Italy ; Department of Biomedical Science, Università di Padova, Padova, Italy
| | - Davide Cervia
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy ; Department for Innovation in Biological, Agro-food and Forest Systems, Università della Tuscia, Viterbo, Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences "Luigi Sacco", University Hospital "Luigi Sacco", Università di Milano, Milano, Italy ; Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| |
Collapse
|
36
|
Cossu MV, Cattaneo D, Fucile S, Pellegrino P, Baldelli S, Cozzi V, Capetti A, Clementi E. Combined isosorbide dinitrate and ibuprofen as a novel therapy for muscular dystrophies: evidence from Phase I studies in healthy volunteers. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:411-9. [PMID: 24851040 PMCID: PMC4018313 DOI: 10.2147/dddt.s58803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We designed two Phase I studies that assessed healthy volunteers in order to evaluate the safety and to optimize the dosing of the combination of the drugs isosorbide dinitrate, a nitric oxide donor, and ibuprofen, a nonsteroidal antiinflammatory drug. We designed these studies with the aim of designing a Phase II trial to evaluate the drugs’ efficacy in patients affected by Duchenne muscular dystrophy. For the first trial, ISOFEN1, a single-dose, randomized-sequence, open-label, active control, three-treatment cross-over study, was aimed at comparing the pharmacokinetics of ibuprofen 200 mg and isosorbide dinitrate 20 mg when given alone and concomitantly. The pharmacokinetics of ibuprofen given alone versus ibuprofen given concomitantly with isosorbide dinitrate were similar, as documented by the lack of statistically significant differences in the main drug’s pharmacokinetic parameters (time to maximal concentration [Tmax], maximal concentration [Cmax], area under the curve [AUC]0–t, and AUC0–∞). Similarly, we found that the coadministration of ibuprofen did not significantly affect the pharmacokinetics of isosorbide dinitrate. No issues of safety were detected. The second trial, ISOFEN2, was a single-site, dose titration study that was designed to select the maximum tolerated dose for isosorbide dinitrate when coadministered with ibuprofen. Eighteen out of the 19 enrolled subjects tolerated the treatment well, and they completed the study at the highest dose of isosorbide dinitrate applied (80 mg/day). One subject voluntarily decided to reduce the dose of isosorbide dinitrate from 80 mg to 60 mg. The treatment-related adverse events recorded during the study were, for the large majority, episodes of headache that remitted spontaneously in 0.5–1 hour – a known side effect of isosorbide dinitrate. These studies demonstrate that the combination of isosorbide dinitrate and ibuprofen does not lead to pharmacokinetic interactions between the two drugs; they also demonstrate that the combination of isosorbide dinitrate and ibuprofen has optimal tolerability and safety profiles that are similar to those previously reported for isosorbide dinitrate and ibuprofen given alone.
Collapse
Affiliation(s)
- Maria Vittoria Cossu
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Serena Fucile
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Paolo Pellegrino
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Sara Baldelli
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Valeria Cozzi
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Amedeo Capetti
- Unit of Infectious Diseases, University Hospital "Luigi Sacco", Milan, Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy ; Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Lecco, Italy
| |
Collapse
|