1
|
Bhuin S, Chakraborty P, Yogeeswari P, Chakravarty M. Twisted Molecular Core Conjugated Oxo-Ether as a Fluorescent Probe for Lipid-Droplets Bioimaging and Live Cancer Cell Discrimination. ACS APPLIED BIO MATERIALS 2025; 8:2985-3001. [PMID: 40053476 DOI: 10.1021/acsabm.4c01817] [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] [Indexed: 03/09/2025]
Abstract
In quest of a new working design for a photostable lipid-droplets (LDs) bioimaging probe, we herein unveil and demonstrate a twisted donor(naphthalene)-π-acceptor(dicyano) architecture linked with oxo/thioether functionality, where the probes' emission, hydrophobicity, cytotoxicity, and cell permeability are altered by replacing the present chalcogen/s. In this class of molecules, an "oxanthrene"-based compound, "OXNCN", was realized as the noncytotoxic and cell-permeable probe, displaying intense fluorescence in a nonpolar solvent, aggregates, and viscous medium. Time-dependent density functional theory (TD-DFT) investigations revealed that OXNCN holds a favorable extent of excited-state planarity to bring out considerable emission only in a nonpolar solvent, resulting in polarity-dependent emission. Outcomes of the concentration- and time-dependent colocalization investigations, cholesterol depletion/repletion studies, and oleic acid treatment-based experiments validated its LD specificity. Strong twisted intramolecular charge transfer (TICT) culminated in weak emission in the polar medium, which helped the probe reduce the cytoplasmic signal. Moreover, the results of time-dependent kinetic acquisitional photophysical studies, fluorescence recovery after photobleaching (FRAP), and intracellular emission investigations testified to the probe's photostability. Assiduous analysis and quantification of confocal laser scanning microscopy (CLSM) images by two-way analysis of variance (ANOVA), followed by Sidak's multiple comparison statistics, could provide insights into the probe's better performance in robust cancer cells (FaDu) than in normal ones (HEK-293). A precise discrimination between oral and normal cancer cells could be established by quantifying the deposited lipid droplets from the CLSM-captured cellular images and applying Student's t test with the quantified values.
Collapse
|
2
|
Cano-Sarabia M, Aydin F, Meng L, Gil-Bonillo M, Fonseca J, Dietrich M, Renner S, Amenitsch H, Falcaro P, Imaz I, Maspoch D. Lipid/ZIF-8 Biocomposites Based on Liposomes or Vesicles: In Situ Formation, and Preliminary Evaluation as Delivery Vehicles for Hydrophobic Drugs. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407051. [PMID: 39981973 DOI: 10.1002/smll.202407051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Integrating lipid self-assemblies with metal-organic frameworks (MOFs) creates biocomposites ideal for encapsulation, protection, and delivery of functional species. This can be achieved using preformed MOFs or through in situ MOF formation. Herein, the one-pot formation of ZIF-8 MOF particles in the presence of two lipid self-assemblies (vesicles or liposomes) is reported, generating two types of hybrid lipid/ZIF-8 biocomposites. Each lipid assembly can be used to encapsulate hydrophobic actives into the hybrid lipid/ZIF-8 biocomposites, demonstrated with Nile Red and Astaxanthin (ATX) as representative cargo. In vitro digestion of ATX-loaded hybrid lipid/ZIF-8 particles in simulated intestinal fluid (SIF) shows distinct release kinetics: liposome-based particles offer a more sustained release compared to vesicle-based biocomposites. Intriguingly, in various media (water, simulated gastric fluid, bicarbonate, and SIF), the sodalite ZIF-8 topology in liposome-based lipid/ZIF-8 particles undergoes a crystalline phase transition to the denser, more-stable phase ZIF-C. This phase transition, along with a deeper internalization of ATX in liposome-based particles, accounts for the differences in release kinetics. In summary, the study provides valuable insights for the synthesis of hybrid lipid/ZIF-8 biocomposites, the encapsulation of hydrophobic molecules, the importance of investigating potential crystalline phase transitions of MOFs in different media, and their potential as drug delivery vehicles.
Collapse
Affiliation(s)
- Mary Cano-Sarabia
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and The Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, 08193, Spain
| | - Funda Aydin
- Department of Basic Sciences, Faculty of Pharmacy, Van Yüzüncü Yıl University, Van, 65080, Turkey
| | - Lingxin Meng
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and The Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, 08193, Spain
- Departament de Química, Facultat de Ciències, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Marta Gil-Bonillo
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and The Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, 08193, Spain
- Departament de Química, Facultat de Ciències, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Javier Fonseca
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and The Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, 08193, Spain
| | - Manuela Dietrich
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and The Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, 08193, Spain
| | - Simon Renner
- Institute of Physical and Theoretical Chemistry, Graz University of Technology, Graz, 8010, Austria
| | - Heinz Amenitsch
- Institute of Physical and Theoretical Chemistry, Graz University of Technology, Graz, 8010, Austria
- Institute of Inorganic Chemistry, Graz University of Technology, Graz, 8010, Austria
| | - Paolo Falcaro
- Institute of Physical and Theoretical Chemistry, Graz University of Technology, Graz, 8010, Austria
| | - Inhar Imaz
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and The Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, 08193, Spain
- Departament de Química, Facultat de Ciències, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and The Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, 08193, Spain
- Departament de Química, Facultat de Ciències, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
- ICREA, Pg. Lluis Companys 23, Barcelona, 08010, Spain
| |
Collapse
|
3
|
Reader KL, Pratt IG, Lawson GL, Woolley RJ. Identifying the composition of large vesicles in the cytoplasm of oocytes. Reprod Fertil Dev 2024; 37:RD24131. [PMID: 39688945 DOI: 10.1071/rd24131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Context Oocyte vesicles, or vacuoles, have been described using transmission electron microscopy in most species. In sheep and cow oocytes, vesicles constitute up to 30% of the cytoplasm, their volume decreases during maturation and is lower in poorer quality oocytes, suggesting they are important for oocyte competence. However, the composition and function of these organelles is unknown. Aim This study aimed to ascertain the content of oocyte vesicles and examine the effect of different fixation methods on the size and preservation of these organelles. Methods Sheep oocytes were centrifuged to segregate organelles then stained with organelle-specific fluorescent dyes (Nile Red, LysoTracker, Fluo-4-AM and TMRM) and imaged by live cell confocal microscopy. The oocytes were fixed with either glutaraldehyde or paraformaldehyde and prepared for electron microscopy to confirm the distribution of organelles and compare ultrastructure and organelle size. Key results Nile Red staining has identified that vesicles contain lipid that is different to that in the osmium-stained lipid droplets observed by electron microscopy. Lipid droplets and vesicles were significantly smaller when prepared for electron microscopy compared to live cell imaging. Organelles were less likely to be fully segregated following centrifugation in oocytes prior to maturation (20%) compared to oocytes after maturation (77%; P Conclusions Oocyte vesicles are lipid storing organelles that may be important for oocyte quality. Implications This study highlights the importance of lipid for oocyte quality and the need for further research to identify the optimal fatty acid content for in vitro maturation media and oocyte competence.
Collapse
Affiliation(s)
- Karen L Reader
- Department of Pathology, University of Otago, Dunedin, New Zealand
| | - Isabella G Pratt
- Department of Pathology, University of Otago, Dunedin, New Zealand
| | - Georgia L Lawson
- Department of Pathology, University of Otago, Dunedin, New Zealand
| | - Robert J Woolley
- Confocal Microscopy Unit, Research Infrastructure Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
4
|
Cravero BH, Prez G, Lombardo VA, Guastaferri FV, Delprato CB, Altabe S, de Mendoza D, Binolfi A. A high-resolution 13C NMR approach for profiling fatty acid unsaturation in lipid extracts and in live Caenorhabditiselegans. J Lipid Res 2024; 65:100618. [PMID: 39127170 PMCID: PMC11418130 DOI: 10.1016/j.jlr.2024.100618] [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: 02/28/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024] Open
Abstract
Unsaturated fatty acids (UFA) play a crucial role in central cellular processes in animals, including membrane function, development, and disease. Disruptions in UFA homeostasis can contribute to the onset of metabolic, cardiovascular, and neurodegenerative disorders. Consequently, there is a high demand for analytical techniques to study lipid compositions in live cells and multicellular organisms. Conventional analysis of UFA compositions in cells, tissues, and organisms involves solvent extraction procedures coupled with analytical techniques such as gas chromatography, MS and/or NMR spectroscopy. As a nondestructive and nontargeted technique, NMR spectroscopy is uniquely capable of characterizing the chemical profiling of living cells and multicellular organisms. Here, we use NMR spectroscopy to analyze Caenorhabditis elegans, enabling the determination of their lipid compositions and fatty acid unsaturation levels both in cell-free lipid extracts and in vivo. The NMR spectra of lipid extracts from WT and fat-3 mutant C. elegans strains revealed notable differences due to the absence of Δ-6 fatty acid desaturase activity, including the lack of arachidonic and eicosapentaenoic acyl chains. Uniform 13C-isotope labeling and high-resolution 2D solution-state NMR of live worms confirmed these findings, indicating that the signals originated from fast-tumbling lipid molecules within lipid droplets. Overall, this strategy permits the analysis of lipid storage in intact worms and has enough resolution and sensitivity to identify differences between WT and mutant animals with impaired fatty acid desaturation. Our results establish methodological benchmarks for future investigations of fatty acid regulation in live C. elegans using NMR.
Collapse
Affiliation(s)
- Bruno Hernández Cravero
- Institute of Molecular and Cellular Biology of Rosario (IBR-CONICET-UNR), Ocampo y Esmeralda, Rosario, Argentina
| | - Gastón Prez
- Institute of Molecular and Cellular Biology of Rosario (IBR-CONICET-UNR), Ocampo y Esmeralda, Rosario, Argentina
| | - Verónica A Lombardo
- Institute of Molecular and Cellular Biology of Rosario (IBR-CONICET-UNR), Ocampo y Esmeralda, Rosario, Argentina; Center of Interdisciplinary Studies (CEI), National University of Rosario (UNR), Rosario, Argentina
| | - Florencia V Guastaferri
- Institute of Molecular and Cellular Biology of Rosario (IBR-CONICET-UNR), Ocampo y Esmeralda, Rosario, Argentina
| | - Carla B Delprato
- Institute of Molecular and Cellular Biology of Rosario (IBR-CONICET-UNR), Ocampo y Esmeralda, Rosario, Argentina
| | - Silvia Altabe
- Institute of Molecular and Cellular Biology of Rosario (IBR-CONICET-UNR), Ocampo y Esmeralda, Rosario, Argentina; Department of Microbiology, Faculty of Biochemical and Pharmaceutical Sciences (FBIOyF), National University of Rosario (UNR) Suipacha 598, Rosario, Argentina
| | - Diego de Mendoza
- Institute of Molecular and Cellular Biology of Rosario (IBR-CONICET-UNR), Ocampo y Esmeralda, Rosario, Argentina; Department of Microbiology, Faculty of Biochemical and Pharmaceutical Sciences (FBIOyF), National University of Rosario (UNR) Suipacha 598, Rosario, Argentina.
| | - Andres Binolfi
- Institute of Molecular and Cellular Biology of Rosario (IBR-CONICET-UNR), Ocampo y Esmeralda, Rosario, Argentina; Argentinian Platform of Structural Biology and Metabolomics (PLABEM), Ocampo y Esmeralda, Rosario, Argentina.
| |
Collapse
|
5
|
Chowdhury RR, Grosso MF, Gadara DC, Spáčil Z, Vidová V, Sovadinová I, Babica P. Cyanotoxin cylindrospermopsin disrupts lipid homeostasis and metabolism in a 3D in vitro model of the human liver. Chem Biol Interact 2024; 397:111046. [PMID: 38735451 DOI: 10.1016/j.cbi.2024.111046] [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: 08/20/2023] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
Cylindrospermopsin, a potent hepatotoxin produced by harmful cyanobacterial blooms, poses environmental and human health concerns. We used a 3D human liver in vitro model based on spheroids of HepG2 cells, in combination with molecular and biochemical assays, automated imaging, targeted LC-MS-based proteomics, and lipidomics, to explore cylindrospermopsin effects on lipid metabolism and the processes implicated in hepatic steatosis. Cylindrospermopsin (1 μM, 48 h) did not significantly affect cell viability but partially reduced albumin secretion. However, it increased neutral lipid accumulation in HepG2 spheroids while decreasing phospholipid levels. Simultaneously, cylindrospermopsin upregulated genes for lipogenesis regulation (SREBF1) and triacylglycerol synthesis (DGAT1/2) and downregulated genes for fatty acid synthesis (ACLY, ACCA, FASN, SCD1). Fatty acid uptake, oxidation, and lipid efflux genes were not significantly affected. Targeted proteomics revealed increased levels of perilipin 2 (adipophilin), a major hepatocyte lipid droplet-associated protein. Lipid profiling quantified 246 lipid species in the spheroids, with 28 significantly enriched and 15 downregulated by cylindrospermopsin. Upregulated species included neutral lipids, sphingolipids (e.g., ceramides and dihexosylceramides), and some glycerophospholipids (phosphatidylethanolamines, phosphatidylserines), while phosphatidylcholines and phosphatidylinositols were mostly reduced. It suggests that cylindrospermopsin exposures might contribute to developing and progressing towards hepatic steatosis or metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- Riju Roy Chowdhury
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | - Marina Felipe Grosso
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | | | - Zdeněk Spáčil
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | - Veronika Vidová
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | - Iva Sovadinová
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic.
| |
Collapse
|
6
|
Dzobo KE, Cupido AJ, Mol BM, Stiekema LC, Versloot M, Winkelmeijer M, Peter J, Pennekamp AM, Havik SR, Vaz FM, van Weeghel M, Prange KH, Levels JH, de Winther MP, Tsimikas S, Groen AK, Stroes ES, de Kleijn DP, Kroon J. Diacylglycerols and Lysophosphatidic Acid, Enriched on Lipoprotein(a), Contribute to Monocyte Inflammation. Arterioscler Thromb Vasc Biol 2024; 44:720-740. [PMID: 38269588 PMCID: PMC10880937 DOI: 10.1161/atvbaha.123.319937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Oxidized phospholipids play a key role in the atherogenic potential of lipoprotein(a) (Lp[a]); however, Lp(a) is a complex particle that warrants research into additional proinflammatory mediators. We hypothesized that additional Lp(a)-associated lipids contribute to the atherogenicity of Lp(a). METHODS Untargeted lipidomics was performed on plasma and isolated lipoprotein fractions. The atherogenicity of the observed Lp(a)-associated lipids was tested ex vivo in primary human monocytes by RNA sequencing, ELISA, Western blot, and transendothelial migratory assays. Using immunofluorescence staining and single-cell RNA sequencing, the phenotype of macrophages was investigated in human atherosclerotic lesions. RESULTS Compared with healthy individuals with low/normal Lp(a) levels (median, 7 mg/dL [18 nmol/L]; n=13), individuals with elevated Lp(a) levels (median, 87 mg/dL [218 nmol/L]; n=12) demonstrated an increase in lipid species, particularly diacylglycerols (DGs) and lysophosphatidic acid (LPA). DG and the LPA precursor lysophosphatidylcholine were enriched in the Lp(a) fraction. Ex vivo stimulation with DG(40:6) demonstrated a significant upregulation in proinflammatory pathways related to leukocyte migration, chemotaxis, NF-κB (nuclear factor kappa B) signaling, and cytokine production. Functional assessment showed a dose-dependent increase in the secretion of IL (interleukin)-6, IL-8, and IL-1β after DG(40:6) and DG(38:4) stimulation, which was, in part, mediated via the NLRP3 (NOD [nucleotide-binding oligomerization domain]-like receptor family pyrin domain containing 3) inflammasome. Conversely, LPA-stimulated monocytes did not exhibit an inflammatory phenotype. Furthermore, activation of monocytes by DGs and LPA increased their transendothelial migratory capacity. Human atherosclerotic plaques from patients with high Lp(a) levels demonstrated colocalization of Lp(a) with M1 macrophages, and an enrichment of CD68+IL-18+TLR4+ (toll-like receptor) TREM2+ (triggering receptor expressed on myeloid cells) resident macrophages and CD68+CASP1+ (caspase) IL-1B+SELL+ (selectin L) inflammatory macrophages compared with patients with low Lp(a). Finally, potent Lp(a)-lowering treatment (pelacarsen) resulted in a reduction in specific circulating DG lipid subspecies in patients with cardiovascular disease with elevated Lp(a) levels (median, 82 mg/dL [205 nmol/L]). CONCLUSIONS Lp(a)-associated DGs and LPA have a potential role in Lp(a)-induced monocyte inflammation by increasing cytokine secretion and monocyte transendothelial migration. This DG-induced inflammation is, in part, NLRP3 inflammasome dependent.
Collapse
Affiliation(s)
- Kim E. Dzobo
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
| | - Arjen J. Cupido
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Barend M. Mol
- Department of Vascular Surgery, University Medical Centre Utrecht, the Netherlands (B.M.M., D.P.V.d.K.)
| | - Lotte C.A. Stiekema
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Miranda Versloot
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
| | - Maaike Winkelmeijer
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Jorge Peter
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Anne-Marije Pennekamp
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Stefan R. Havik
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Frédéric M. Vaz
- Core Facility Metabolomics (F.M.V., M.v.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Michel van Weeghel
- Core Facility Metabolomics (F.M.V., M.v.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Koen H.M. Prange
- Department of Medical Biochemistry, Amsterdam Infection and Immunity (K.H.M.P., M.P.J.d.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Johannes H.M. Levels
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam Infection and Immunity (K.H.M.P., M.P.J.d.W.), Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla (S.T.)
| | - Albert K. Groen
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Erik S.G. Stroes
- Vascular Medicine (A.J.C., L.C.A.S., E.S.G.S.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
| | - Dominique P.V. de Kleijn
- Department of Vascular Surgery, University Medical Centre Utrecht, the Netherlands (B.M.M., D.P.V.d.K.)
| | - Jeffrey Kroon
- Departments of Experimental Vascular Medicine (K.E.D., M.V., M.W., J.P., A.-M.P., S.R.H., J.H.M.L., A.K.G., J.K.), Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, the Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, the Netherlands (K.E.D., M.V., J.K.)
- Laboratory of Angiogenesis and Vascular Metabolism, Flanders Institute for Biotechnology (VIB)-KU Leuven Center for Cancer Biology, VIB, Belgium (J.K.)
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute, Belgium (J.K.)
| |
Collapse
|
7
|
Kalampounias G, Gardeli C, Alexis S, Anagnostopoulou E, Androutsopoulou T, Dritsas P, Aggelis G, Papanikolaou S, Katsoris P. Poly-Unsaturated Fatty Acids (PUFAs) from Cunninghamella elegans Grown on Glycerol Induce Cell Death and Increase Intracellular Reactive Oxygen Species. J Fungi (Basel) 2024; 10:130. [PMID: 38392802 PMCID: PMC10890652 DOI: 10.3390/jof10020130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
Cunninghamella elegans NRRL-1393 is an oleaginous fungus able to synthesize and accumulate unsaturated fatty acids, amongst which the bioactive gamma-linolenic acid (GLA) has potential anti-cancer activities. C. elegans was cultured in shake-flask nitrogen-limited media with either glycerol or glucose (both at ≈60 g/L) employed as the sole substrate. The assimilation rate of both substrates was similar, as the total biomass production reached 13.0-13.5 g/L, c. 350 h after inoculation (for both instances, c. 27-29 g/L of substrate were consumed). Lipid production was slightly higher on glycerol-based media, compared to the growth on glucose (≈8.4 g/L vs. ≈7.0 g/L). Lipids from C. elegans grown on glycerol, containing c. 9.5% w/w of GLA, were transformed into fatty acid lithium salts (FALS), and their effects were assessed on both human normal and cancerous cell lines. The FALS exhibited cytotoxic effects within a 48 h interval with an IC50 of about 60 μg/mL. Additionally, a suppression of migration was shown, as a significant elevation of oxidative stress levels, and the induction of cell death. Elementary differences between normal and cancer cells were not shown, indicating a generic mode of action; however, oxidative stress level augmentation may increase susceptibility to anticancer drugs, improving chemotherapy effectiveness.
Collapse
Affiliation(s)
- Georgios Kalampounias
- Laboratory of Cell Biology, Division of Genetics, Cell and Developmental Biology, Department of Biology, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| | - Chrysavgi Gardeli
- Laboratory of Food Chemistry and Analysis, Department of Food Science and Human Nutrition, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece
| | - Spyridon Alexis
- Hematology Division, Faculty of Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Elena Anagnostopoulou
- Laboratory of Food Microbiology and Biotechnology, Department of Food Science and Human Nutrition, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece
| | - Theodosia Androutsopoulou
- Laboratory of Cell Biology, Division of Genetics, Cell and Developmental Biology, Department of Biology, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| | - Panagiotis Dritsas
- Unit of Microbiology, Division of Genetics, Cell and Developmental Biology, Department of Biology, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| | - George Aggelis
- Unit of Microbiology, Division of Genetics, Cell and Developmental Biology, Department of Biology, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| | - Seraphim Papanikolaou
- Laboratory of Food Microbiology and Biotechnology, Department of Food Science and Human Nutrition, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece
| | - Panagiotis Katsoris
- Laboratory of Cell Biology, Division of Genetics, Cell and Developmental Biology, Department of Biology, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| |
Collapse
|
8
|
Yang X, Chen J, Lu Z, Huang S, Zhang S, Cai J, Zhou Y, Cao G, Yu J, Qin Z, Zhao W, Zhang B, Zhu L. Enterovirus A71 utilizes host cell lipid β-oxidation to promote its replication. Front Microbiol 2022; 13:961942. [PMID: 36246276 PMCID: PMC9554258 DOI: 10.3389/fmicb.2022.961942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Enterovirus A71 (EV-A71) is a major pathogen that causes severe and fatal cases of hand-foot-and-mouth disease (HFMD), which is an infectious disease that endangers children’s health. However, the pathogenic mechanisms underlying these severe clinical and pathological features remain incompletely understood. Metabolism and stress are known to play critical roles in multiple stages of the replication of viruses. Lipid metabolism and ER stress is an important characterization post viral infection. EV-A71 infection alters the perturbations of intracellular lipid homeostasis and induces ER stress. The characterizations induced by viral infections are essential for optimal virus replication and may be potential antiviral targets. In this study, we found that the addition of the chemical drug of ER stress, PKR IN, an inhibitor, or Tunicamycin, an activator, could significantly reduce viral replication with the decrease of lipid. The replication of viruses was reduced by Chemical reagent TOFA, an inhibitor of acetyl-CoA carboxylase (ACC) or C75, an inhibitor of fatty acid synthase (FASN), while enhanced by oleic acid (OA), which is a kind of exogenous supplement of triacylglycerol. The pharmacochemical reagent of carnitine palmitoyltransferase 1 (CPT1) called Etomoxir could knock down CPT1 to induce EV-A71 replication to decrease. This suggests that lipid, rather than ER stress, is the main factor affecting EV-A71 replication. In conclusion, this study revealed that it is the β-oxidation of lipid that plays a core role, not ER stress, which is only a concomitant change without restrictive effect, on virus replication.
Collapse
Affiliation(s)
- Xiuwen Yang
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiayi Chen
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zixin Lu
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shan Huang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shihao Zhang
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jintai Cai
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yezhen Zhou
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guanhua Cao
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jianhai Yu
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhiran Qin
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- *Correspondence: Wei Zhao,
| | - Bao Zhang
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Bao Zhang,
| | - Li Zhu
- BSL-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Li Zhu,
| |
Collapse
|
9
|
Dutta T, Pal K, Koner AL. Intracellular Physical Properties with Small Organic Fluorescent Probes: Recent Advances and Future Perspectives. CHEM REC 2022; 22:e202200035. [PMID: 35801859 DOI: 10.1002/tcr.202200035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/22/2022] [Indexed: 11/09/2022]
Abstract
The intracellular physical parameters i. e., polarity, viscosity, fluidity, tension, potential, and temperature of a live cell are the hallmark of cellular health and have garnered immense interest over the past decade. In this context, small molecule organic fluorophores exhibit prominent useful properties including easy functionalizability, environmental sensitivity, biocompatibility, and fast yet efficient cellular uptakability which has made them a popular tool to understand intra-cellular micro-environmental properties. Throughout this discussion, we have outlined the basic design strategies of small molecules for specific organelle targeting and quantification of physical properties. The values of these parameters are indicative of cellular homeostasis and subtle alteration may be considered as the onset of disease. We believe this comprehensive review will facilitate the development of potential future probes for superior insight into the physical parameters that are yet to be quantified.
Collapse
Affiliation(s)
- Tanoy Dutta
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, INDIA (TD) (ALK
| | - Kaushik Pal
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, INDIA (TD) (ALK.,Department of Physics and Astronomy, Iowa State University, Ames, Iowa, 50011, USA
| | - Apurba Lal Koner
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, INDIA (TD) (ALK
| |
Collapse
|
10
|
Aass KR, Mjelle R, Kastnes MH, Tryggestad SS, van den Brink LM, Aass Roseth I, Westhrin M, Zahoor M, Moen SH, Vikene Nedal TM, Buene G, Misund K, Sponaas AM, Ma Q, Sundan A, Groen RW, Slørdahl TS, Waage A, Standal T. Intracellular IL-32 regulates mitochondrial metabolism, proliferation, and differentiation of malignant plasma cells. iScience 2022; 25:103605. [PMID: 35005550 PMCID: PMC8717606 DOI: 10.1016/j.isci.2021.103605] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/13/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Interleukin-32 (IL-32) is a nonclassical cytokine expressed in cancers, inflammatory diseases, and infections. Its expression is regulated by two different oxygen sensing systems; HIF1α and cysteamine dioxygenase (ADO), indicating that IL-32 may be involved in the response to hypoxia. We here demonstrate that endogenously expressed, intracellular IL-32 interacts with components of the mitochondrial respiratory chain and promotes oxidative phosphorylation. Knocking out IL-32 in three myeloma cell lines reduced cell survival and proliferation in vitro and in vivo. High-throughput transcriptomic and MS-metabolomic profiling of IL-32 KO cells revealed that cells depleted of IL-32 had perturbations in metabolic pathways, with accumulation of lipids, pyruvate precursors, and citrate. IL-32 was expressed in a subgroup of myeloma patients with inferior survival, and primary myeloma cells expressing IL-32 had a gene signature associated with immaturity, proliferation, and oxidative phosphorylation. In conclusion, we demonstrate a previously unrecognized role of IL-32 in the regulation of plasma cell metabolism. Intracellular IL-32 is an endogenous growth factor for malignant plasma cells IL-32 interacts with components of the electron transport chain IL-32 promotes oxidative phosphorylation IL-32 is expressed by immature, CD45 + highly proliferating malignant plasma cells
Collapse
Affiliation(s)
- Kristin Roseth Aass
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Robin Mjelle
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Martin H Kastnes
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Synne S Tryggestad
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Luca M van den Brink
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Ingrid Aass Roseth
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Marita Westhrin
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Muhammad Zahoor
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo 0372, Norway
| | - Siv H Moen
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Tonje M Vikene Nedal
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Glenn Buene
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Kristine Misund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Anne-Marit Sponaas
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Qianli Ma
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Anders Sundan
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Richard Wj Groen
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam 1081, the Netherlands
| | - Tobias S Slørdahl
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Hematology, St.Olavs University Hospital, Trondheim 7491, Norway
| | - Anders Waage
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Hematology, St.Olavs University Hospital, Trondheim 7491, Norway
| | - Therese Standal
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Hematology, St.Olavs University Hospital, Trondheim 7491, Norway
| |
Collapse
|
11
|
Antioxidant and lipid-reducing effects of Rosa rugosa root extract in 3T3-L1 cell. Food Sci Biotechnol 2021; 31:121-129. [DOI: 10.1007/s10068-021-01018-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
|
12
|
Cytotoxic activity of cholesterol oxidase produced by Streptomyces sp. AKHSS against cancerous cell lines: mechanism of action in HeLa cells. World J Microbiol Biotechnol 2021; 37:141. [PMID: 34287712 DOI: 10.1007/s11274-021-03076-5] [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: 09/04/2020] [Accepted: 05/20/2021] [Indexed: 11/27/2022]
Abstract
Re-occurrence of cancer is the major drawback for the currently available anticancer therapies. Therefore, study of an efficient enzyme, cholesterol oxidase produced by various kinds of microbes especially obtained from unexplored marine actinobacterial species against human cancer cell lines and understanding its mechanism of action helps to identify an irreversible and potent anticancer agent. The cytotoxic potential of cholesterol oxidase produced by a marine Streptomyces sp. AKHSS against four different human cancer cell lines was demonstrated through MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] assay. Fluorescent confocal microscopy and flow cytometry based experiments were performed to understand the efficiency of the enzymatic action on HeLa cells. Further, the apoptotic related proteins were detected through western blotting. Interestingly, the enzyme exhibited potent cytotoxicity at very low concentrations (0.093-0.327 µM) against all the cells tested. Fluorescent confocal microscopy revealed the morphological variations induced by the enzyme on cancer cell lines such as the formation of lipid droplets and condensation of nuclei. The enzyme treated cell-free extracts of HeLa cells analyzed through gas chromatography mass spectrometry showed the depletion of membrane cholesterol and the presence of substituted enzyme oxidized product, cholest-4-ene-3-one. The enzyme had induced significant inhibitory effects on the cell viability such as cell cycle arrest (G1 phase), apoptosis and rise of reactive oxygen species as evident through flow cytometry. Besides, hyperpolarization of mitochondrial membrane, reduced rates of phosphorylation of pAkt and the expression of apoptotic death markers like Fas, Fas L, caspases (8 and 3) and PARP-1 were recorded in the enzyme treated HeLa cells. Thus, cholesterol oxidase purified from a marine Streptomyces sp. AKHSS exhibits potent cytotoxicity at very low concentrations against human cancer cell lines. All the ex vivo experiments portrayed the substantial inhibitory effect of the enzyme on HeLa cells suggesting that cholesterol oxidase of Streptomyces sp. AKHSS could be a prominent cancer chemotherapeutic agent.
Collapse
|
13
|
Poels K, Schnitzler JG, Waissi F, Levels JHM, Stroes ESG, Daemen MJAP, Lutgens E, Pennekamp AM, De Kleijn DPV, Seijkens TTP, Kroon J. Inhibition of PFKFB3 Hampers the Progression of Atherosclerosis and Promotes Plaque Stability. Front Cell Dev Biol 2020; 8:581641. [PMID: 33282864 PMCID: PMC7688893 DOI: 10.3389/fcell.2020.581641] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Aims 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase (PFKFB)3-mediated glycolysis is pivotal in driving macrophage- and endothelial cell activation and thereby inflammation. Once activated, these cells play a crucial role in the progression of atherosclerosis. Here, we analyzed the expression of PFKFB3 in human atherosclerotic lesions and investigated the therapeutic potential of pharmacological inhibition of PFKFB3 in experimental atherosclerosis by using the glycolytic inhibitor PFK158. Methods and Results PFKFB3 expression was higher in vulnerable human atheromatous carotid plaques when compared to stable fibrous plaques and predominantly expressed in plaque macrophages and endothelial cells. Analysis of advanced plaques of human coronary arteries revealed a positive correlation of PFKFB3 expression with necrotic core area. To further investigate the role of PFKFB3 in atherosclerotic disease progression, we treated 6-8 weeks old male Ldlr -/- mice. These mice were fed a high cholesterol diet for 13 weeks, of which they were treated for 5 weeks with the glycolytic inhibitor PFK158 to block PFKFB3 activity. The incidence of fibrous cap atheroma (advanced plaques) was reduced in PFK158-treated mice. Plaque phenotype altered markedly as both necrotic core area and intraplaque apoptosis decreased. This coincided with thickening of the fibrous cap and increased plaque stability after PFK158 treatment. Concomitantly, we observed a decrease in glycolysis in peripheral blood mononuclear cells compared to the untreated group, which alludes that changes in the intracellular metabolism of monocyte and macrophages is advantageous for plaque stabilization. Conclusion High PFKFB3 expression is associated with vulnerable atheromatous human carotid and coronary plaques. In mice, high PFKFB3 expression is also associated with a vulnerable plaque phenotype, whereas inhibition of PFKFB3 activity leads to plaque stabilization. This data implies that inhibition of inducible glycolysis may reduce inflammation, which has the ability to subsequently attenuate atherogenesis.
Collapse
Affiliation(s)
- Kikkie Poels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Johan G Schnitzler
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Farahnaz Waissi
- Division of Surgical Specialties, Department of Vascular Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands.,Department of Cardiology Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Johannes H M Levels
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Erik S G Stroes
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Mat J A P Daemen
- Department of Pathology, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Anne-Marije Pennekamp
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Dominique P V De Kleijn
- Division of Surgical Specialties, Department of Vascular Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands.,Department of Vascular Surgery, Netherlands and Netherlands Heart Institute, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | - Tom T P Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| |
Collapse
|
14
|
Wasiak S, Dzobo KE, Rakai BD, Kaiser Y, Versloot M, Bahjat M, Stotz SC, Fu L, Sweeney M, Johansson JO, Wong NCW, Stroes ESG, Kroon J, Kulikowski E. BET protein inhibitor apabetalone (RVX-208) suppresses pro-inflammatory hyper-activation of monocytes from patients with cardiovascular disease and type 2 diabetes. Clin Epigenetics 2020; 12:166. [PMID: 33172487 PMCID: PMC7657365 DOI: 10.1186/s13148-020-00943-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background Patients with cardiovascular disease (CVD) and type 2 diabetes (DM2) have a high residual risk for experiencing a major adverse cardiac event. Dysregulation of epigenetic mechanisms of gene transcription in innate immune cells contributes to CVD development but is currently not targeted by therapies. Apabetalone (RVX-208) is a small molecule inhibitor of bromodomain and extra-terminal (BET) proteins—histone acetylation readers that drive pro-inflammatory and pro-atherosclerotic gene transcription. Here, we assess the impact of apabetalone on ex vivo inflammatory responses of monocytes from DM2 + CVD patients. Results Monocytes isolated from DM2 + CVD patients and matched controls were treated ex vivo with apabetalone, interferon γ (IFNγ), IFNγ + apabetalone or vehicle and phenotyped for gene expression and protein secretion. Unstimulated DM2 + CVD monocytes had higher baseline IL-1α, IL-1β and IL-8 cytokine gene expression and Toll-like receptor (TLR) 2 surface abundance than control monocytes, indicating pro-inflammatory activation. Further, DM2 + CVD monocytes were hyper-responsive to stimulation with IFNγ, upregulating genes within cytokine and NF-κB pathways > 30% more than control monocytes (p < 0.05). Ex vivo apabetalone treatment countered cytokine secretion by DM2 + CVD monocytes at baseline (GROα and IL-8) and during IFNγ stimulation (IL-1β and TNFα). Apabetalone abolished pro-inflammatory hyper-activation by reducing TLR and cytokine gene signatures more robustly in DM2 + CVD versus control monocytes. Conclusions Monocytes isolated from DM2 + CVD patients receiving standard of care therapies are in a hyper-inflammatory state and hyperactive upon IFNγ stimulation. Apabetalone treatment diminishes this pro-inflammatory phenotype, providing mechanistic insight into how BET protein inhibition may reduce CVD risk in DM2 patients.
Collapse
Affiliation(s)
- Sylwia Wasiak
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Kim E Dzobo
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Brooke D Rakai
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Yannick Kaiser
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Miranda Versloot
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Mahnoush Bahjat
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Stephanie C Stotz
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Li Fu
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Michael Sweeney
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Jan O Johansson
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Norman C W Wong
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Ewelina Kulikowski
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada.
| |
Collapse
|
15
|
Schnitzler JG, Hoogeveen RM, Ali L, Prange KHM, Waissi F, van Weeghel M, Bachmann JC, Versloot M, Borrelli MJ, Yeang C, De Kleijn DPV, Houtkooper RH, Koschinsky ML, de Winther MPJ, Groen AK, Witztum JL, Tsimikas S, Stroes ESG, Kroon J. Atherogenic Lipoprotein(a) Increases Vascular Glycolysis, Thereby Facilitating Inflammation and Leukocyte Extravasation. Circ Res 2020; 126:1346-1359. [PMID: 32160811 PMCID: PMC7208285 DOI: 10.1161/circresaha.119.316206] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: Patients with elevated levels of lipoprotein(a) [Lp(a)] are hallmarked by increased metabolic activity in the arterial wall on positron emission tomography/computed tomography, indicative of a proinflammatory state. Objective: We hypothesized that Lp(a) induces endothelial cell inflammation by rewiring endothelial metabolism. Methods and Results: We evaluated the impact of Lp(a) on the endothelium and describe that Lp(a), through its oxidized phospholipid content, activates arterial endothelial cells, facilitating increased transendothelial migration of monocytes. Transcriptome analysis of Lp(a)-stimulated human arterial endothelial cells revealed upregulation of inflammatory pathways comprising monocyte adhesion and migration, coinciding with increased 6-phophofructo-2-kinase/fructose-2,6-biphosphatase (PFKFB)-3–mediated glycolysis. ICAM (intercellular adhesion molecule)-1 and PFKFB3 were also found to be upregulated in carotid plaques of patients with elevated levels of Lp(a). Inhibition of PFKFB3 abolished the inflammatory signature with concomitant attenuation of transendothelial migration. Conclusions: Collectively, our findings show that Lp(a) activates the endothelium by enhancing PFKFB3-mediated glycolysis, leading to a proadhesive state, which can be reversed by inhibition of glycolysis. These findings pave the way for therapeutic agents targeting metabolism aimed at reducing inflammation in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Johan G Schnitzler
- From the Experimental Vascular Medicine (J.G.S., L.A., J.C.B., M.V., A.K.G., J.K.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Renate M Hoogeveen
- Vascular Medicine (R.M.H., E.S.G.S.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Lubna Ali
- From the Experimental Vascular Medicine (J.G.S., L.A., J.C.B., M.V., A.K.G., J.K.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Koen H M Prange
- Medical Biochemistry (K.H.M.P., M.P.J.d.W.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Farahnaz Waissi
- Vascular Surgery, Netherlands (F.W., D.P.V.D.K.), UMC Utrecht, University Utrecht, the Netherlands.,Cardiology (F.W.), UMC Utrecht, University Utrecht, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism (M.v.W., R.H.H.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands.,Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, the Netherlands (M.v.W.)
| | - Julian C Bachmann
- From the Experimental Vascular Medicine (J.G.S., L.A., J.C.B., M.V., A.K.G., J.K.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Miranda Versloot
- From the Experimental Vascular Medicine (J.G.S., L.A., J.C.B., M.V., A.K.G., J.K.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Matthew J Borrelli
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada (M.J.B., M.L.K.)
| | - Calvin Yeang
- Vascular Medicine Program, Division of Cardiology, Department of Medicine, Sulpizio Cardiovascular Center (C.Y., S.T.), University of California San Diego, La Jolla
| | - Dominique P V De Kleijn
- Vascular Surgery, Netherlands (F.W., D.P.V.D.K.), UMC Utrecht, University Utrecht, the Netherlands.,Netherlands Heart Institute (D.P.V.D.K.), UMC Utrecht, University Utrecht, the Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism (M.v.W., R.H.H.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada (M.J.B., M.L.K.)
| | - Menno P J de Winther
- Medical Biochemistry (K.H.M.P., M.P.J.d.W.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands.,Institute for Cardiovascular Prevention, Munich, Germany (M.P.J.d.W.)
| | - Albert K Groen
- From the Experimental Vascular Medicine (J.G.S., L.A., J.C.B., M.V., A.K.G., J.K.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands.,Pediatrics, Laboratory of Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands (A.K.G.)
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, Department of Medicine (J.L.W.), University of California San Diego, La Jolla
| | - Sotirios Tsimikas
- Vascular Medicine Program, Division of Cardiology, Department of Medicine, Sulpizio Cardiovascular Center (C.Y., S.T.), University of California San Diego, La Jolla
| | - Erik S G Stroes
- Vascular Medicine (R.M.H., E.S.G.S.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Jeffrey Kroon
- From the Experimental Vascular Medicine (J.G.S., L.A., J.C.B., M.V., A.K.G., J.K.), Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands
| |
Collapse
|
16
|
Elgegren M, Kim S, Cordova D, Silva C, Noro J, Cavaco-Paulo A, Nakamatsu J. Ultrasound-Assisted Encapsulation of Sacha Inchi ( Plukenetia volubilis Linneo.) Oil in Alginate-Chitosan Nanoparticles. Polymers (Basel) 2019; 11:E1245. [PMID: 31357570 PMCID: PMC6723186 DOI: 10.3390/polym11081245] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022] Open
Abstract
Sacha inchi oil is rich in essential and non-essential fatty acids and other types of bioactive agents like tocopherols and polyphenolic compounds, which are very well-known antioxidants. In this study, the encapsulation of sacha inchi oil in alginate (AL) and chitosan (CS) nanoparticles was achieved with the assistance of high-intensity ultrasound. Nanoemulsion is the most effective delivery and high stability system for lipophilic bioactive agents. Chitosan and surfactant concentrations were varied to study their effect on particle formulations. Size, zeta-potential, polydispersity, and stability of particles were determined in time to optimize the preparation conditions. Sacha inchi oil encapsulated in AL-CS nanoparticles showed a higher loading efficiency and stability for short and long periods compared with other vegetable oils such as olive and soybean. Also, because of the types of tocopherols present in sacha inchi oil (γ- and δ-tocopherols), a much higher antioxidant activity (95% of radical reduction in 15 min) was found in comparison with nanocapsules with olive oil, which contain α-tocopherols. The particles showed high efficiency of protein loading at high concentration of bovine serum albumin (BSA) and a low rate of leaching profiles in various testing media like simulated gastric and intestinal fluids with/without enzymes, that is, pepsin 0.1% (w/v) and pancreatin 0.1% (w/v), respectively.
Collapse
Affiliation(s)
- Mariela Elgegren
- Department of Science, Chemistry Division, Pontificia Universidad Católica del Perú PUCP, Av. Universitaria 1801, Lima 32, Peru
| | - Suyeon Kim
- Department of Engineering, Pontificia Universidad Católica del Perú PUCP, Av. Universitaria 1801, Lima 32, Peru.
| | - Diego Cordova
- Department of Science, Chemistry Division, Pontificia Universidad Católica del Perú PUCP, Av. Universitaria 1801, Lima 32, Peru
| | - Carla Silva
- Centre of Biological Engineering, University of Minho, Campus De Gualtar, 4710-057 Braga, Portugal
| | - Jennifer Noro
- Centre of Biological Engineering, University of Minho, Campus De Gualtar, 4710-057 Braga, Portugal
| | - Artur Cavaco-Paulo
- Centre of Biological Engineering, University of Minho, Campus De Gualtar, 4710-057 Braga, Portugal
| | - Javier Nakamatsu
- Department of Science, Chemistry Division, Pontificia Universidad Católica del Perú PUCP, Av. Universitaria 1801, Lima 32, Peru
| |
Collapse
|
17
|
Exner T, Beretta CA, Gao Q, Afting C, Romero-Brey I, Bartenschlager R, Fehring L, Poppelreuther M, Füllekrug J. Lipid droplet quantification based on iterative image processing. J Lipid Res 2019; 60:1333-1344. [PMID: 30926625 DOI: 10.1194/jlr.d092841] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/28/2019] [Indexed: 12/15/2022] Open
Abstract
Lipid droplets (LDs) are ubiquitous and highly dynamic subcellular organelles required for the storage of neutral lipids. LD number and size distribution are key parameters affected not only by nutrient supply but also by lipotoxic stress and metabolic regulation. Current methods for LD quantification lack general applicability and are either based on time consuming manual evaluation or show limitations if LDs are high in numbers or closely clustered. Here, we present an ImageJ-based approach for the detection and quantification of LDs stained by neutral lipid dyes in images acquired by conventional wide-field fluorescence microscopy. The method features an adjustable preprocessing procedure that resolves LD clusters. LD identification is based on their circular edges and central fluorescence intensity maxima. Adaptation to different cell types is mediated by a set of interactive parameters. Validation was done for three different cell lines using manual evaluation of LD numbers and volume measurement by 3D rendering of confocal datasets. In an application example, we show that overexpression of the acyl-CoA synthetase, FATP4/ACSVL5, in oleate-treated COS7 cells increased the size of LDs but not their number.
Collapse
Affiliation(s)
- Tarik Exner
- Molecular Cell Biology Laboratory Internal Medicine IV, Heidelberg University, Heidelberg, Germany
| | - Carlo A Beretta
- CellNetworks Math-Clinic Core Facility, BioQuant Heidelberg University, Heidelberg, Germany
| | - Qi Gao
- CellNetworks Math-Clinic Core Facility, BioQuant Heidelberg University, Heidelberg, Germany
| | - Cassian Afting
- Molecular Cell Biology Laboratory Internal Medicine IV, Heidelberg University, Heidelberg, Germany
| | - Inés Romero-Brey
- Department of Infectious Diseases, Molecular Virology Heidelberg University, Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology Heidelberg University, Heidelberg, Germany.,Department of Virus-Associated Carcinogenesis, German Cancer Research Center, Heidelberg, Germany
| | - Leonard Fehring
- Molecular Cell Biology Laboratory Internal Medicine IV, Heidelberg University, Heidelberg, Germany
| | - Margarete Poppelreuther
- Molecular Cell Biology Laboratory Internal Medicine IV, Heidelberg University, Heidelberg, Germany
| | - Joachim Füllekrug
- Molecular Cell Biology Laboratory Internal Medicine IV, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
18
|
Fam TK, Klymchenko AS, Collot M. Recent Advances in Fluorescent Probes for Lipid Droplets. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E1768. [PMID: 30231571 PMCID: PMC6163203 DOI: 10.3390/ma11091768] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 09/14/2018] [Accepted: 09/14/2018] [Indexed: 12/15/2022]
Abstract
Lipid droplets (LDs) are organelles that serve as the storage of intracellular neutral lipids. LDs regulate many physiological processes. They recently attracted attention after extensive studies showed their involvement in metabolic disorders and diseases such as obesity, diabetes, and cancer. Therefore, it is of the highest importance to have reliable imaging tools. In this review, we focus on recent advances in the development of selective fluorescent probes for LDs. Their photophysical properties are described, and their advantages and drawbacks in fluorescence imaging are discussed. At last, we review the reported applications using these probes including two-photon excitation, in vivo and tissue imaging, as well as LDs tracking.
Collapse
Affiliation(s)
- Tkhe Kyong Fam
- Nanochemistry and Bioimaging Group, Laboratoire de Bioimagerie et Pathologies, CNRS UMR 7021, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France.
| | - Andrey S Klymchenko
- Nanochemistry and Bioimaging Group, Laboratoire de Bioimagerie et Pathologies, CNRS UMR 7021, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France.
| | - Mayeul Collot
- Nanochemistry and Bioimaging Group, Laboratoire de Bioimagerie et Pathologies, CNRS UMR 7021, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France.
| |
Collapse
|
19
|
Crociati M, Di Giacinto F, Manuali E, Stradaioli G, Sylla L, Monaci M, Maulucci G, De Spirito M. Systemic profiling of ectopic fat deposits in the reproductive tract of dairy cows. Theriogenology 2018; 114:46-53. [PMID: 29597123 DOI: 10.1016/j.theriogenology.2018.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/11/2018] [Accepted: 03/16/2018] [Indexed: 11/25/2022]
Abstract
During the transition period, high-yielding dairy cows suffer from negative energy balance, intense lipomobilization and impaired lipid metabolism; this metabolic condition can lead to overburdened triglycerides accumulation in the liver, known as liver lipidosis, which has been associated to impaired fertility in dairy cows. The mechanisms of this impairment can be in principle correlated with the presence and the extent of ectopic fat depots. However, current methods for evaluating fat accumulation in liver and in the reproductive tract suffer from low resolution, sensitivity, and specificity. Confocal microscopes are equipped with Gallium arsenide phosphide detectors, thus enabling the acquisition of intense signals from tissue biopsies. This method could differentiate whether fat deposition occurred without requiring sample sectioning. Here, we examined with this technique liver, uterine and ovarian samples of heifers and regularly slaughtered repeat breeder and overconditioned dairy cows, to quantify lipid droplets and depots at a submicrometer scale with high specificity. With the aid of this technique, we found lipid depots in uterine and ovarian specimens. Moreover, we found that the size and number of depots increased with the degree of liver lipidosis. Further studies are needed to elucidate the relationship between the severity and extent of these deposits and the fertility of lactating dairy cows. Since tissues other than liver display different characteristic lipid droplet distributions, this technique can be potentially employed to shed new light on the pathogenesis of lipidosis and to assess new risk factors for infertility.
Collapse
Affiliation(s)
- Martina Crociati
- Dipartimento di Medicina Veterinaria, Università di Perugia, Via S. Costanzo, 4, Perugia, Italy
| | - Flavio Di Giacinto
- Istituto di Fisica, Università Cattolica del Sacro cuore, Largo Francesco Vito 1, Rome, Italy
| | - Elisabetta Manuali
- Istituto Zooprofilattico Sperimentale Umbria e Marche, Via Gaetano Salvemini, 1, Perugia, Italy
| | - Giuseppe Stradaioli
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali, Università di Udine, via Palladio 8, Udine, Italy
| | - Lakamy Sylla
- Dipartimento di Medicina Veterinaria, Università di Perugia, Via S. Costanzo, 4, Perugia, Italy
| | - Maurizio Monaci
- Dipartimento di Medicina Veterinaria, Università di Perugia, Via S. Costanzo, 4, Perugia, Italy
| | - Giuseppe Maulucci
- Istituto di Fisica, Università Cattolica del Sacro cuore, Largo Francesco Vito 1, Rome, Italy.
| | - Marco De Spirito
- Istituto di Fisica, Università Cattolica del Sacro cuore, Largo Francesco Vito 1, Rome, Italy
| |
Collapse
|