Nicoletta Zini
Istituto di Genetica Molecolare “Luigi Luca Cavalli-Sforza” – Sede di Bologna
c/o Istituto Ortopedico Rizzoli -via di Barbiano 1/10
40136 Bologna
Phone: +39 051 6366771
E-mail: nicoletta.zini@cnr.it
Curriculum Vitae – Download
Elenco completo delle pubblicazioni – Download
Attività di ricerca
Italiano
L’attività di ricerca riguarda studi sull’ ultrastruttura e funzioni del nucleo interfasico, l’analisi funzionale della distribuzione intranucleare dei diversi elementi del sistema di trasduzione del segnale inositide-dipendente.
Più recentemente sono stati valutati, mediante analisi in microscopia elettronica, il differenziamento osteogenico e condrogenico di progenitori cellulari cresciuti su materiali biocompatibili, al fine di chiarire alcuni aspetti dell’ interazione cellule/matrice extracellulare e biomateriali impiegabili nei processi riparativi dell’osso e della cartilagine.
Ulteriori studi sono stati condotti per analizzare il comportamento di cellule cresciute su scaffold biocompatibili 3-D che riproducono parte della morfologia degli organi adulti.
Inoltre sono stati caratterizzati esosomi da diversi modelli cellulari con il fine di dimostrare il loro ruolo nella comunicazione tra cellule.
Competenze: microscopia elettronica a trasmissione e relative metodiche di allestimento dei preparati, immunocitochimica ultrastrutturale, immunofluorescenza, microscopia ottica.
Progetti di ricerca
- Progetto europeo Horizon 2020-SCREENED EU 2019-2023, grant N° 825745: “A multistage model of thyroid gland function for screening endocrine-disrupting chemicals in a biologically sex-specific manner”. Responsabile presso IGM-CNR: Dott.ssa Nicoletta Zini, afferente all’Unità di Ricerca SCREENED coordinata dal Prof. Dott. Roberto Toni, Università di Parma.
Gruppo di ricerca
Despina Kiriakidu, tecnico di laboratorio IGM-CNR
Pubblicazioni Recenti
Toni R; Barbaro F; Di Conza G; Zini N; Remaggi G; Elviri L; Spaletta G; Quarantini E; Quarantini M; Mosca S; Caravelli S; Mosca M; Ravanetti F; Sprio S; Tampieri A A bioartificial and vasculomorphic bone matrix-based organoid mimicking microanatomy of flat and short bones Journal Article In: Journal of biomedical materials research. Part B, Applied biomaterials, vol. 112, iss. 1, pp. e35329, 2024. Barbaro F; Conza GD; Quartulli FP; Quarantini E; Quarantini M; Zini N; Fabbri C; Mosca S; Caravelli S; Mosca M; Vescovi P; Sprio S; Tampieri A; Toni R. 2024. Di Conza G; Barbaro F; Zini N; Spaletta G; Remaggi G; Elviri L; Mosca S; Caravelli S; Mosca M; Toni R. Woven bone formation and mineralization by rat mesenchymal stromal cells imply increased expression of the intermediate filament desmin Journal Article In: Frontiers in endocrinology, vol. 14, pp. 1234569, 2023. Perut F; Graziani G; Roncuzzi L; Zini N; Avnet S; Baldini N FT-IR Spectral Signature of Sensitive and Multidrug-Resistant Osteosarcoma Cell-Derived Extracellular Nanovesicles Journal Article In: Cells, vol. 11, iss. 5, pp. 778, 2022. Cavallo C; Merli G; Zini N; D'Adamo S; Cattini L; Guescini M; Grigolo B; Di Martino A; Santi S; Borzì RM; Filardo G In: Stem cells international, vol. 2022, pp. 9376338, 2022. Bolognesi F; Fazio N; Boriani F; Fabbri VP; Gravina D; Pedrini FA; Zini N; Greco M; Paolucci M; Re MC; Asioli S; Foschini MP; D'Errico A; Baldini N; Marchetti C Validation of a Cleanroom Compliant Sonication-Based Decellularization Technique: A New Concept in Nerve Allograft Production Journal Article In: International journal of molecular sciences, vol. 23, iss. 2, pp. 1530, 2022. Bottega R; Ravera S; Napolitano LMR; Chiappetta V; Zini N; Crescenzi B; Arniani S; Faleschini M; Cortone G; Faletra F; Medagli B; Sirchia F; Moretti M; de Lange J; Cappelli E; Mecucci C; Onesti S; Pisani FM; Savoia A In: Journal of cellular physiology, vol. 236, no 8, pp. 5664-5675, 2021. Costa R; Rodia MT; Zini N; Pegoraro V; Marozzo R; Capanni C; Angelini C; Lattanzi G; Santi S; Cenacchi G In: Molecular and cellular biochemistry, vol. 476, no 4, pp. 1797-1811, 2021. Sartore L; Manferdini C; Saleh Y; Dey K; Gabusi E; Ramorino G; Zini N; Almici C; Re F; Russo D; Mariani E; Lisignoli G Polysaccharides on gelatin-based hydrogels differently affect chondrogenic differentiation of human mesenchymal stromal cells Journal Article In: Materials science & engineering. C, Materials for biological applications, vol. 126, pp. 112175, 2021. Avnet S; Lemma S; Cortini M; Di Pompo G; Perut F; Lipreri MV; Roncuzzi L; Columbaro M; Errani C; Longhi A; Zini N; Heymann D; Dominici M; Grisendi G; Golinelli G; Consolino L; Longo DL; Nanni C; Righi A; Baldini N Release of Inflammatory Mediators from Acid-Stimulated Mesenchymal Stromal Cells Favours Tumour Invasiveness and Metastasis in Osteosarcoma. Journal Article In: Cancers (Basel), vol. 13, no 22, pp. 5855, 2021. Cavallo C; Merli G; Borzì RM; Zini N; D'Adamo S; Guescini M; Grigolo B; Di Martino A; Santi S; Filardo G In: Scientific Reports, vol. 11, no 1, pp. 1053, 2021. Perut F; Roncuzzi L; Avnet S; Massa A; Zini N; Sabbadini S; Giampieri F; Mezzetti B; Baldini N Strawberry-Derived Exosome-Like Nanoparticles Prevent Oxidative Stress in Human Mesenchymal Stromal Cells Journal Article In: Biomolecules, vol. 11, no 1, pp. e87, 2021. Toni R; Di Conza G; Barbaro F; Zini N; Consolini E; Dallatana D; Antoniel M; Quarantini E; Quarantini M; Maioli S; Bruni CA; Elviri L; Panseri S; Sprio S; Sandri M; Tampieri A Microtopography of Immune Cells in Osteoporosis and Bone Lesions by Endocrine Disruptors. Journal Article In: Frontiers in immunology, vol. 11, pp. 1737, 2020. Boriani F; Savarino L; Fazio N; Pedrini FA; Fini M; Nicoli Aldini N; Martini L; Zini N; Bernardini M; Bolognesi F; Marchetti C; Baldini N Auto-Allo Graft Parallel Juxtaposition for Improved Neuroregeneration in Peripheral Nerve Reconstruction Based on Acellular Nerve Allografts. Journal Article In: Annals of plastic surgery, vol. 83, no 3, pp. 318-325, 2019. Manferdini C; Gabusi E; Sartore L; Dey K; Agnelli S; Almici C; Bianchetti A; Zini N; Russo D; Re F; Mariani E; Lisignoli G Chitosan-based scaffold counteracts hypertrophic and fibrotic markers in chondrogenic differentiated mesenchymal stromal cells. Journal Article In: Journal of tissue engineering and regenerative medicine, vol. 13, no 10, pp. 1896-1911, 2019. Perut F; Roncuzzi L; Zini N; Massa A; Baldini N Extracellular Nanovesicles Secreted by Human Osteosarcoma Cells Promote Angiogenesis. Journal Article In: Cancers (Basel), vol. 11, no 6, pp. e779, 2019. Barbaro F; Consolini E; Toscani G; Zini N; Dallatana D; Setti P; Mosca S; Di Conza G; Bassi E; Quarantini E; Quarantini M; Raposio E; Gorreri M; Porro A; Toni R. The masks of Lorenzo Tenchini: their anatomy and surgical/bioengineering clues. Journal Article In: Journal of anatomy, vol. 235, no 6, pp. 1036-1044, 2019. Diomede F; Zini N; Pizzicannella J; Merciaro I; Pizzicannella G; D'Orazio M; Piattelli A; Trubiani O 5-Aza Exposure Improves Reprogramming Process Through Embryoid Body Formation in Human Gingival Stem Cells. Journal Article In: Frontiers in genetics, vol. 9, pp. 419, 2018. Baldini N; Torreggiani E; Roncuzzi L; Perut F; Zini N; Avnet S Exosome-like nanovesicles isolated from Citrus limon L. exert antioxidative effect. Journal Article In: Current pharmaceutical biotechnology, vol. 19, no 11, pp. 877-885, 2018. Manferdini C; Zini N; Gabusi E; Paolella F; Lambertini E; Penolazzi L; Piva R; Lisignoli G Immunoelectron microscopic localization of Collagen type XV during human mesenchymal stem cells mineralization. Journal Article In: Connective tissue research, vol. 59, no Suppl. 1, pp. 42-45, 2018. Boriani F; Fazio N; Fotia C; Savarino L; Nicoli Aldini N; Martini L; Zini N; Bernardini M; Baldini N A novel technique for decellularization of allogenic nerves and in vivo study of their use for peripheral nerve reconstruction. Journal Article In: Journal of biomedical materials research. Part A, vol. 105, no 8, pp. 2228-2240, 2017. Baglio SR; Lagerweij T; Pérez-Lanzón M; Ho XD; Léveillé N; Melo SA; Cleton-Jansen AM; Jordanova ES; Roncuzzi L; Greco M; van Eijndhoven MAJ; Grisendi G; Dominici M; Bonafede R; Lougheed SM; de Gruijl TD; Zini N; Cervo S; Steffan A; Canzonieri V; Martson A; Maasalu K; Köks S; Wurdinger T; Baldini N; Pegtel DM Blocking Tumor-Educated MSC Paracrine Activity Halts Osteosarcoma Progression. Journal Article In: Clinical cancer research, vol. 23, no 14, pp. 3721-3733, 2017. Avnet S; Lemma S; Cortini M; Pellegrini P; Perut F; Zini N; Kusuzaki K; Chano T; Grisendi G; Dominici M; De Milito A; Baldini N Altered pH gradient at the plasma membrane of osteosarcoma cells is a key mechanism of drug resistance. Journal Article In: Oncotarget, vol. 7, no 39, pp. 63408-63423, 2016. Manara MC; Terracciano M; Mancarella C; Sciandra M; Guerzoni C; Pasello M; Grilli A; Zini N; Picci P; Colombo MP; Morrione A; Scotlandi K CD99 triggering induces methuosis of Ewing sarcoma cells through IGF-1R/RAS/Rac1 signaling. Journal Article In: Oncotarget, vol. 7, no 48, pp. 79925-79942, 2016. Lemma S; Sboarina M; Porporato PE; Zini N; Sonveaux P; Di Pompo G; Baldini N; Avnet S Energy metabolism in osteoclast formation and activity. Journal Article In: International Journal of Biochemistry And Cell Biology, vol. 79, pp. 168-180, 2016. Diomede F; Zini N; Gatta V; Fulle S; Merciaro I; D'Aurora M; La Rovere RM; Traini T; Pizzicannella J; Ballerini P; Caputi S; Piattelli A; Trubiani O Human periodontal ligament stem cells cultured onto cortico-cancellous scaffold drive bone regenerative process. Journal Article In: European Cells & Materials, vol. 32, pp. 181-201, 2016. Cavallo C; Desando G; Ferrari A; Zini N; Mariani E; Grigolo B Hyaluronan scaffold supports osteogenic differentiation of bone marrow concentrate cells. Journal Article In: Journal of Biological Regulators and Homeostatic Agents, vol. 30, no 2, pp. 409-420, 2016. Torreggiani E; Roncuzzi L; Perut F; Zini N; Baldini N Multimodal transfer of MDR by exosomes in human osteosarcoma Journal Article In: International Journal of Oncology, vol. 49, no 1, pp. 189-196, 2016. Koppers-Lalic D; Hackenberg M; Menezes R; Misovic B; Wachalska M; Geldof A; Zini N; Reijke T; Wurdinger T; Vis A; Moorselaar JV; Pegtel M; Bijnsdorp I Non‑invasive prostate cancer detection by measuring miRNA variants (isomiRs) in urine extracellular vesicles. Journal Article In: Oncotarget, vol. 7, no 16, pp. 22566-22578, 2016. Manescu A; Giuliani A; Mohammadi S; Tromba G; Mazzoni S; Diomede F; Zini N; Piattelli A; Trubiani O Osteogenic potential of dualblocks cultured with human periodontal ligament stem cells: in vitro and synchrotron microtomography study. Journal Article In: Journal of Periodontal Research, vol. 51, no 1, pp. 112-124, 2016. Manferdini C; Cavallo C; Grigolo B; Fiorini M; Nicoletti A; Gabusi E; Zini N; Pressato D; Facchini A; Lisignoli G Specific inductive potential of a novel nanocomposite biomimetic biomaterial for osteochondral tissue regeneration Journal Article In: Journal of Tissue Engineering and Regenerative Medicine, vol. 10, pp. 374-391, 2016. Trubiani O; Piattelli A; Gatta V; Marchisio M; Diomede F; D'Aurora M; Merciaro I; Pierdomenico L; Maraldi NM; Zini N Assessment of an efficient xeno-free culture system for human periodontal ligament stem cells. Journal Article In: Tissue Engineering. Part C, Methods, vol. 21, no 1, pp. 52-64, 2015. Baglio SR; Rooijers K; Koppers-Lalic D; Verweij FJ; Perez Lanzon M; Zini N; Naaijkens B; Perut F; Niessen HW; Baldini N; Pegtel DM Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Journal Article In: Stem Cell Research & Therapy, vol. 6, pp. 127, 2015. Grigolo B; Cavallo C; Desando G; Manferdini C; Lisignoli G; Ferrari A; Zini N; Facchini A Novel nano-composite biomimetic biomaterial allows chondrogenic and osteogenic differentiation of bone marrow concentrate derived cells. Journal Article In: Journal of Materials Science. Materials in Medicine, vol. 26, no 4, pp. 173, 2015. Lambertini E; Penolazzi L; Morganti C; Lisignoli G; Zini N; Angelozzi M; Bonora M; Ferroni L; Pinton P; Zavan B; Piva R Osteogenic differentiation of human MSCs: Specific occupancy of the mitochondrial DNA by NFATc1 transcription factor. Journal Article In: International Journal of Biochemistry & Cell Biology, vol. 64, pp. 212-219, 2015.
2024
@article{%a1.%Y_144,
title = {A bioartificial and vasculomorphic bone matrix-based organoid mimicking microanatomy of flat and short bones},
author = {Toni R and Barbaro F and Di Conza G and Zini N and Remaggi G and Elviri L and Spaletta G and Quarantini E and Quarantini M and Mosca S and Caravelli S and Mosca M and Ravanetti F and Sprio S and Tampieri A},
url = {https://onlinelibrary.wiley.com/doi/10.1002/jbm.b.35329},
doi = {10.1002/jbm.b.35329},
year = {2024},
date = {2024-02-12},
journal = {Journal of biomedical materials research. Part B, Applied biomaterials},
volume = {112},
issue = {1},
pages = {e35329},
abstract = {We engineered an in vitro model of bioartificial 3D bone organoid consistent with an anatomical and vascular microenvironment common to mammalian flat and short bones. To achieve this, we chose the decellularized-decalcified matrix of the adult male rat scapula, implemented with the reconstruction of its intrinsic vessels, obtained through an original intravascular perfusion with polylevolactic (PLLA), followed by coating of the PLLA-fabricated vascularization with rat tail collagen. As a result, the 3D bone and vascular geometry of the native bone cortical and cancellous compartments was reproduced, and the rat tail collagen-PLLA biomaterial could in vitro act as a surrogate of the perivascular extracellular matrix (ECM) around the wall of the biomaterial-reconstituted cancellous vessels. As a proof-of-concept of cell compatibility and site-dependent osteoinductive properties of this bioartificial 3D construct, we show that it in vitro leads to a time-dependent microtopographic positioning of rat mesenchymal stromal cells (MSCs), initiating an osteogenic fate in relation to the bone compartment. In addition, coating of PLLA-reconstructed vessels with rat tail collagen favored perivascular attachment and survival of MSC-like cells (mouse embryonic fibroblasts), confirming its potentiality as a perivascular stroma for triggering competence of seeded MSCs. Finally, in vivo radiographic topography of bone lesions in the human jaw and foot tarsus of subjects with primary osteoporosis revealed selective bone cortical versus cancellous involvement, suggesting usefulness of a human 3D bone organoid engineered with the same principles of our rat organoid, to in vitro investigate compartment-dependent activities of human MSC in flat and short bones under experimental osteoporotic challenge. We conclude that our 3D bioartificial construct offers a reliable replica of flat and short bones microanatomy, and promises to help in building a compartment-dependent mechanistic perspective of bone remodeling, including the microtopographic dysregulation of osteoporosis.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@bachelorthesis{nokey,
title = {Correlation between tooth decay and insulin resistance in normal weight males prompts a role for myo-inositol as a regenerative factor in dentistry and oral surgery: a feasibility study},
author = {Barbaro F and Conza GD and Quartulli FP and Quarantini E and Quarantini M and Zini N and Fabbri C and Mosca S and Caravelli S and Mosca M and Vescovi P and Sprio S and Tampieri A and Toni R.},
url = {https://www.frontiersin.org/journals/bioengineering-and-biotechnology/articles/10.3389/fbioe.2024.1374135/full},
doi = {10.3389/fbioe.2024.1374135},
year = {2024},
date = {2024-08-19},
journal = {Frontiers in bioengineering and biotechnology},
volume = {12},
pages = {1374135},
abstract = {Background: In an era of precision and stratified medicine, homogeneity in population-based cohorts, stringent causative entry, and pattern analysis of datasets are key elements to investigate medical treatments. Adhering to these principles, we collected in vivo and in vitro data pointing to an insulin-sensitizing/insulin-mimetic effect of myo-inositol (MYO) relevant to cell regeneration in dentistry and oral surgery. Confirmation of this possibility was obtained by in silico analysis of the relation between in vivo and in vitro results (the so-called bed-to-benchside reverse translational approach). Results: Fourteen subjects over the 266 screened were young adult, normal weight, euglycemic, sedentary males having normal appetite, free diet, with a regular three-times-a-day eating schedule, standard dental hygiene, and negligible malocclusion/enamel defects. Occlusal caries were detected by fluorescence videoscanning, whereas body composition and energy balance were estimated with plicometry, predictive equations, and handgrip. Statistically significant correlations (Pearson r coefficient) were found between the number of occlusal caries and anthropometric indexes predicting insulin resistance (IR) in relation to the abdominal/visceral fat mass, fat-free mass, muscular strength, and energy expenditure adjusted to the fat and muscle stores. This indicated a role for IR in affecting dentin reparative processes. Consistently, in vitro administration of MYO to HUVEC and Swiss NIH3T3 cells in concentrations corresponding to those administered in vivo to reduce IR resulted in statistically significant cell replication (ANOVA/Turkey tests), suggesting that MYO has the potential to counteract inhibitory effects of IR on dental vascular and stromal cells turnover. Finally, in in silico experiments, quantitative evaluation (WOE and information value) of a bioinformatic Clinical Outcome Pathway confirmed that in vitro trophic effects of MYO could be transferred in vivo with high predictability, providing robust credence of its efficacy for oral health. Conclusion: Our reverse bed-to-benchside data indicate that MYO might antagonize the detrimental effects of IR on tooth decay. This provides feasibility for clinical studies on MYO as a regenerative factor in dentistry and oral surgery, including dysmetabolic/aging conditions, bone reconstruction in oral destructive/necrotic disorders, dental implants, and for empowering the efficacy of a number of tissue engineering methodologies in dentistry and oral surgery.},
keywords = {},
pubstate = {published},
tppubtype = {bachelorthesis}
}
2023
@article{%a1.%Yb_118,
title = {Woven bone formation and mineralization by rat mesenchymal stromal cells imply increased expression of the intermediate filament desmin},
author = {{Di Conza G} and Barbaro F and Zini N and Spaletta G and Remaggi G and Elviri L and Mosca S and Caravelli S and Mosca M and Toni R.},
url = {https://www.frontiersin.org/articles/10.3389/fendo.2023.1234569/full},
doi = {10.3389/fendo.2023.1234569},
year = {2023},
date = {2023-10-04},
journal = {Frontiers in endocrinology},
volume = {14},
pages = {1234569},
abstract = {Background: Disordered and hypomineralized woven bone formation by dysfunctional mesenchymal stromal cells (MSCs) characterize delayed fracture healing and endocrine -metabolic bone disorders like fibrous dysplasia and Paget disease of bone. To shed light on molecular players in osteoblast differentiation, woven bone formation, and mineralization by MSCs we looked at the intermediate filament desmin (DES) during the skeletogenic commitment of rat bone marrow MSCs (rBMSCs), where its bone-related action remains elusive. Results: Monolayer cultures of immunophenotypically- and morphologically - characterized, adult male rBMSCs showed co-localization of desmin (DES) with vimentin, F-actin, and runx2 in all cell morphotypes, each contributing to sparse and dense colonies. Proteomic analysis of these cells revealed a topologically-relevant interactome, focused on cytoskeletal and related enzymes//chaperone/signalling molecules linking DES to runx2 and alkaline phosphatase (ALP). Osteogenic differentiation led to mineralized woven bone nodules confined to dense colonies, significantly smaller and more circular with respect to controls. It significantly increased also colony-forming efficiency and the number of DES-immunoreactive dense colonies, and immunostaining of co-localized DES/runx-2 and DES/ALP. These data confirmed pre-osteoblastic and osteoblastic differentiation, woven bone formation, and mineralization, supporting DES as a player in the molecular pathway leading to the osteogenic fate of rBMSCs. Conclusion: Immunocytochemical and morphometric studies coupled with proteomic and bioinformatic analysis support the concept that DES may act as an upstream signal for the skeletogenic commitment of rBMSCs. Thus, we suggest that altered metabolism of osteoblasts, woven bone, and mineralization by dysfunctional BMSCs might early be revealed by changes in DES expression//levels. Non-union fractures and endocrine - metabolic bone disorders like fibrous dysplasia and Paget disease of bone might take advantage of this molecular evidence for their early diagnosis and follow-up.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2022
@article{%a1.%Yb_33,
title = {FT-IR Spectral Signature of Sensitive and Multidrug-Resistant Osteosarcoma Cell-Derived Extracellular Nanovesicles},
author = {Perut F and Graziani G and Roncuzzi L and Zini N and Avnet S and Baldini N},
url = {https://www.mdpi.com/2073-4409/11/5/778},
doi = {10.3390/cells11050778},
year = {2022},
date = {2022-08-26},
journal = {Cells},
volume = {11},
issue = {5},
pages = {778},
abstract = {Osteosarcoma (OS) is the most common primary bone cancer in children and adolescents. Despite aggressive treatment regimens, the outcome is unsatisfactory, and multidrug resistance (MDR) is a pivotal process in OS treatment failure. OS-derived extracellular vesicles (EVs) promote drug resistance to chemotherapy and target therapy through different mechanisms. The aim of this study was to identify subpopulations of osteosarcoma-EVs by Fourier transform infrared spectroscopy (FT-IR) to define a specific spectral signature for sensitive and multidrug-resistant OS-derived EVs. EVs were isolated from sensitive and MDR OS cells as well as from mesenchymal stem cells by differential centrifugation and ultracentrifugation. EVs size, morphology and protein expression were characterized. FT-IR/ATR of EVs spectra were acquired in the region of 400-4000 cm-1 (resolution 4 cm-1, 128 scans). The FT-IR spectra obtained were consistently different in the EVs compared to cells from which they originate. A specific spectral signature, characterized by a shift and a new band (1601 cm-1), permitted to clearly distinguish EVs isolated by sensitive and multidrug-resistant OS cells. Our data suggest that FT-IR spectroscopy allows to characterize and define a specific spectral signature for sensitive and MDR OS-derived EVs.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1.%Ybv,
title = {Small Extracellular Vesicles from Inflamed Adipose Derived Stromal Cells Enhance the NF-κB-Dependent Inflammatory/Catabolic Environment of Osteoarthritis. },
author = {Cavallo C and Merli G and Zini N and {D'Adamo} S and Cattini L and Guescini M and Grigolo B and Di Martino A and Santi S and Borzì RM and Filardo G},
url = {https://www.hindawi.com/journals/sci/2022/9376338/},
doi = {10.1155/2022/9376338},
year = {2022},
date = {2022-08-11},
urldate = {2022-08-11},
journal = {Stem cells international},
volume = {2022},
pages = {9376338},
abstract = {The last decade has seen exponentially growing efforts to exploit the effects of adipose derived stromal cells (ADSC) in the treatment of a wide range of chronic degenerative diseases, including osteoarthritis (OA), the most prevalent joint disorder. In the perspective of developing a cell-free advanced therapy medicinal product, a focus has been recently addressed to the ADSC secretome that lends itself to an allogeneic use and can be further dissected for the selective purification of small extracellular vesicles (sEVs). sEVs can act as "biological drug carriers" to transfer information that mirror the pathophysiology of the providing cells. This is important in the clinical perspective where many OA patients are also affected by the metabolic syndrome (MetS). ADSC from MetS OA patients are dysfunctional and "inflammatory" primed within the adipose tissue. To mimic this condition, we exposed ADSC to IL-1β, and then we investigated the effects of the isolated sEVs on chondrocytes and synoviocytes, either cultured separately or in co-culture, to tease out the effects of these "IL-1β primed sEVs" on gene and protein expression of major inflammatory and catabolic OA markers. In comparison with sEVs isolated from unstimulated ADSC, the IL-1β primed sEVs were able to propagate NF-κB activation in bystander joint cells. The effects were more prominent on synoviocytes, possibly because of a higher expression of binding molecules such as CD44. These findings call upon a careful characterization of the "inflammatory fingerprint" of ADSC to avoid the transfer of an unwanted message as well as the development of in vitro "preconditioning" strategies able to rescue the antiinflammatory/anticatabolic potential of ADSC-derived sEVs.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1.%Yb,
title = {Validation of a Cleanroom Compliant Sonication-Based Decellularization Technique: A New Concept in Nerve Allograft Production},
author = {Bolognesi F and Fazio N and Boriani F and Fabbri VP and Gravina D and Pedrini FA and Zini N and Greco M and Paolucci M and Re MC and Asioli S and Foschini MP and D'Errico A and Baldini N and Marchetti C},
url = {https://www.mdpi.com/1422-0067/23/3/1530},
doi = {10.3390/ijms23031530},
year = {2022},
date = {2022-02-25},
urldate = {2022-02-25},
journal = {International journal of molecular sciences},
volume = {23},
issue = {2},
pages = {1530},
abstract = {Defects of the peripheral nervous system are extremely frequent in trauma and surgeries and have high socioeconomic costs. If the direct suture of a lesion is not possible, i.e., nerve gap > 2 cm, it is necessary to use grafts. While the gold standard is the autograft, it has disadvantages related to its harvesting, with an inevitable functional deficit and further morbidity. An alternative to autografting is represented by the acellular nerve allograft (ANA), which avoids disadvantages of autograft harvesting and fresh allograft rejection. In this research, the authors intend to transfer to human nerves a novel technique, previously implemented in animal models, to decellularize nerves. The new method is based on soaking the nerve tissues in decellularizing solutions while associating ultrasounds and freeze-thaw cycles. It is performed without interrupting the sterility chain, so that the new graft may not require post-production γ-ray irradiation, which is suspected to affect the structural and functional quality of tissues. The new method is rapid, safe, and inexpensive if compared with available commercial ANAs. Histology and immunohistochemistry have been adopted to evaluate the new decellularized nerves. The study shows that the new method can be applied to human nerve samples, obtaining similar, and, sometimes better, results compared with the chosen control method, the Hudson technique.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2021
@article{%a1:%Y__490,
title = {Mitochondrial defect in Warsaw syndrome cells genomic integrity and mitochondrial metabolism defects in Warsaw syndrome cells: A comparison with Fanconi anemia. },
author = {Bottega R and Ravera S and Napolitano LMR and Chiappetta V and Zini N and Crescenzi B and Arniani S and Faleschini M and Cortone G and Faletra F and Medagli B and Sirchia F and Moretti M and de Lange J and Cappelli E and Mecucci C and Onesti S and Pisani FM and Savoia A},
url = {https://onlinelibrary.wiley.com/doi/10.1002/jcp.30265},
doi = {10.1002/jcp.30265},
year = {2021},
date = {2021-03-09},
journal = {Journal of cellular physiology},
volume = {236},
number = {8},
pages = {5664-5675},
abstract = {"Warsaw breakage syndrome (WABS), is caused by biallelic mutations of DDX11, a gene coding a DNA helicase. We have recently reported two affected sisters, compound heterozygous for a missense (p.Leu836Pro) and a frameshift (p.Lys303Glufs*22) variant. By investigating the pathogenic mechanism, we demonstrate the inability of the DDX11 p.Leu836Pro mutant to unwind forked DNA substrates, while retaining DNA binding activity. We observed the accumulation of patient-derived cells at the G2/M phase and increased chromosomal fragmentation after mitomycin C treatment. The phenotype partially overlaps with features of the Fanconi anemia cells, which shows not only genomic instability but also defective mitochondria. This prompted us to examine mitochondrial functionality in WABS cells and revealed an altered aerobic metabolism. This opens the door to the further elucidation of the molecular and cellular basis of an impaired mitochondrial phenotype and sheds light on this fundamental process in cell physiology and the pathogenesis of these diseases.
"},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
"@article{%a1:%Y__494,
title = {Morphological study of TNPO3 and SRSF1 interaction during myogenesis by combining confocal, structured illumination and electron microscopy analysis. },
author = {Costa R and Rodia MT and Zini N and Pegoraro V and Marozzo R and Capanni C and Angelini C and Lattanzi G and Santi S and Cenacchi G},
url = {https://link.springer.com/article/10.1007/s11010-020-04023-y},
doi = {10.1007/s11010-020-04023-y},
year = {2021},
date = {2021-03-09},
urldate = {2021-03-09},
journal = {Molecular and cellular biochemistry},
volume = {476},
number = {4},
pages = {1797-1811},
abstract = {Transportin3 (TNPO3) shuttles the SR proteins from the cytoplasm to the nucleus. The SR family includes essential splicing factors, such as SRSF1, that influence alternative splicing, controlling protein diversity in muscle and satellite cell differentiation. Given the importance of alternative splicing in the myogenic process and in the maintenance of healthy muscle, alterations in the splicing mechanism might contribute to the development of muscle disorders. Combining confocal, structured illumination and electron microscopy, we investigated the expression of TNPO3 and SRSF1 during myogenesis, looking at nuclear and cytoplasmic compartments. We investigated TNPO3 and its interaction with SRSF1 and we observed that SRSF1 remained mainly localized in the nucleus, while TNPO3 decreased in the cytoplasm and was strongly clustered in the nuclei of differentiated myotubes. In conclusion, combining different imaging techniques led us to describe the behavior of TNPO3 and SRSF1 during myogenesis, showing that their dynamics follow the myogenic process and could influence the proteomic network necessary during myogenesis. The combination of different high-, super- and ultra-resolution imaging techniques led us to describe the behavior of TNPO3 and its interaction with SRSF1, looking at nuclear and cytoplasmic compartments. These observations represent a first step in understanding the role of TNPO3 and SRFSF1 in complex mechanisms, such as myogenesis.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Yb,
title = {Polysaccharides on gelatin-based hydrogels differently affect chondrogenic differentiation of human mesenchymal stromal cells},
author = {Sartore L and Manferdini C and Saleh Y and Dey K and Gabusi E and Ramorino G and Zini N and Almici C and Re F and Russo D and Mariani E and Lisignoli G},
url = {https://www.sciencedirect.com/science/article/pii/S0928493121003143?via%3Dihub},
doi = {10.1016/j.msec.2021.112175},
year = {2021},
date = {2021-06-08},
journal = {Materials science & engineering. C, Materials for biological applications},
volume = {126},
pages = {112175},
abstract = {Selection of feasible hybrid-hydrogels for best chondrogenic differentiation of human mesenchymal stromal cells (hMSCs) represents an important challenge in cartilage regeneration. In this study, three-dimensional hybrid hydrogels obtained by chemical crosslinking of poly (ethylene glycol) diglycidyl ether (PEGDGE), gelatin (G) without or with chitosan (Ch) or dextran (Dx) polysaccharides were developed. The hydrogels, namely G-PEG, G-PEG-Ch and G-PEG-Dx, were prepared with an innovative, versatile and cell-friendly technique that involves two preparation steps specifically chosen to increase the degree of crosslinking and the physical-mechanical stability of the product: a first homogeneous phase reaction followed by directional freezing, freeze-drying and post-curing. Chondrogenic differentiation of human bone marrow mesenchymal stromal cells (hBM-MSC) was tested on these hydrogels to ascertain whether the presence of different polysaccharides could favor the formation of the native cartilage structure. We demonstrated that the hydrogels exhibited an open pore porous morphology with high interconnectivity and the incorporation of Ch and Dx into the G-PEG common backbone determined a slightly reduced stiffness compared to that of G-PEG hydrogels. We demonstrated that G-PEG-Dx showed a significant increase of its anisotropic characteristic and G-PEG-Ch exhibited higher and faster stress relaxation behavior than the other hydrogels. These characteristics were associated to absence of chondrogenic differentiation on G-PEG-Dx scaffold and good chondrogenic differentiation on G-PEG and G-PEG-Ch. Furthermore, G-PEG-Ch induced the minor collagen proteins and the formation of collagen fibrils with a diameter like native cartilage. This study demonstrated that both anisotropic and stress relaxation characteristics of the hybrid hydrogels were important features directly influencing the chondrogenic differentiation potentiality of hBM-MSC.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Yb_59,
title = {Release of Inflammatory Mediators from Acid-Stimulated Mesenchymal Stromal Cells Favours Tumour Invasiveness and Metastasis in Osteosarcoma.},
author = {Avnet S and Lemma S and Cortini M and Di Pompo G and Perut F and Lipreri MV and Roncuzzi L and Columbaro M and Errani C and Longhi A and Zini N and Heymann D and Dominici M and Grisendi G and Golinelli G and Consolino L and Longo DL and Nanni C and Righi A and Baldini N},
url = {https://www.mdpi.com/2072-6694/13/22/5855},
doi = {10.3390/cancers13225855},
year = {2021},
date = {2021-12-06},
urldate = {2021-12-06},
journal = {Cancers (Basel)},
volume = {13},
number = {22},
pages = {5855},
abstract = {Osteosarcoma is the most frequent primary malignant bone tumour with an impressive tendency to metastasise. Highly proliferative tumour cells release a remarkable amount of protons into the extracellular space that activates the NF-kB inflammatory pathway in adjacent stromal cells. In this study, we further validated the correlation between tumour glycolysis/acidosis and its role in metastases. In patients, at diagnosis, we found high circulating levels of inflammatory mediators (IL6, IL8 and miR-136-5p-containing extracellular vesicles). IL6 serum levels significantly correlated with disease-free survival and 18F-FDG PET/CT uptake, an indirect measurement of tumour glycolysis and, hence, of acidosis. In vivo subcutaneous and orthotopic models, co-injected with mesenchymal stromal (MSC) and osteosarcoma cells, formed an acidic tumour microenvironment (mean pH 6.86, as assessed by in vivo MRI-CEST pH imaging). In these xenografts, we enlightened the expression of both IL6 and the NF-kB complex subunit in stromal cells infiltrating the tumour acidic area. The co-injection with MSC also significantly increased lung metastases. Finally, by using 3D microfluidic models, we directly showed the promotion of osteosarcoma invasiveness by acidosis via IL6 and MSC. In conclusion, osteosarcoma-associated MSC react to intratumoural acidosis by triggering an inflammatory response that, in turn, promotes tumour invasiveness at the primary site toward metastasis development.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y__493,
title = {Small Extracellular Vesicles from adipose derived stromal cells significantly attenuate in vitro the NF-kappaB dependent inflammatory/catabolic environment of osteoarthritis. },
author = {Cavallo C and Merli G and Borzì RM and Zini N and D'Adamo S and Guescini M and Grigolo B and Di Martino A and Santi S and Filardo G},
url = {https://www.nature.com/articles/s41598-020-80032-7},
doi = {10.1038/s41598-020-80032-7},
year = {2021},
date = {2021-03-09},
journal = {Scientific Reports},
volume = {11},
number = {1},
pages = {1053},
abstract = {The therapeutic ability of Mesenchymal Stem/Stromal Cells to address osteoarthritis (OA) is mainly related to the secretion of biologically active factors, which can be found within their secreted Extracellular Vesicles including small Extracellular Vesicles (sEV). Aim of this study was to investigate the effects of sEV from adipose derived stromal cells (ADSC) on both chondrocytes and synoviocytes, in order to gain insights into the mechanisms modulating the inflammatory/catabolic OA environment. sEV, obtained by a combined precipitation and size exclusion chromatography method, were quantified and characterized, and administered to chondrocytes and synoviocytes stimulated with IL-1beta. Cellular uptake of sEV was evaluated from 1 to 12 h. Gene expression and protein release of cytokines/chemokines, catabolic and inflammatory molecules were analyzed at 4 and 15 h, when p65 nuclear translocation was investigated to study NF-kappaB pathway. This study underlined the potential of ADSC derived sEV to affect gene expression and protein release of both chondrocytes and synoviocytes, counteracting IL-1beta induced inflammatory effects, and provided insights into their mechanisms of action. sEV uptake was faster in synoviocytes, where it also elicited stronger effects, especially in terms of cytokine and chemokine modulation. The inflammatory/catabolic environment mediated by NF-kappaB pathway was significantly attenuated by sEV, which hold promise as new therapeutic strategy to address OA.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y__499,
title = {Strawberry-Derived Exosome-Like Nanoparticles Prevent Oxidative Stress in Human Mesenchymal Stromal Cells},
author = {Perut F and Roncuzzi L and Avnet S and Massa A and Zini N and Sabbadini S and Giampieri F and Mezzetti B and Baldini N},
url = {https://www.mdpi.com/2218-273X/11/1/87/html},
doi = {10.3390/biom11010087},
year = {2021},
date = {2021-03-09},
journal = {Biomolecules},
volume = {11},
number = {1},
pages = {e87},
abstract = {Plant-derived exosome-like nanovesicles (EPDENs) have recently been isolated and evaluated as potential bioactive nutraceutical biomolecules. It has been hypothesized that EPDENs may exert their activity on mammalian cells through their specific cargo. In this study, we isolated and purified EPDENs from the strawberry juice of Fragaria x ananassa (cv. Romina), a new cultivar characterized by a high content of anthocyanins, folic acid, flavonols, and vitamin C and an elevated antioxidant capacity. Fragaria-derived EPDENs were purified by a series of centrifugation and filtration steps. EPDENs showed size and morphology similar to mammalian extracellular nanovesicles. The internalization of Fragaria-derived EPDENs by human mesenchymal stromal cells (MSCs) did not negatively affect their viability, and the pretreatment of MSCs with Fragaria-derived EPDENs prevented oxidative stress in a dose-dependent manner. This is possibly due to the presence of vitamin C inside the nanovesicle membrane. The analysis of EPDEN cargo also revealed the presence of small RNAs and miRNAs. These findings suggest that Fragaria-derived EPDENs may be considered nanoshuttles contained in food, with potential health-promoting activity.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2020
@article{%a1:%Y_480,
title = {Microtopography of Immune Cells in Osteoporosis and Bone Lesions by Endocrine Disruptors. },
author = {Toni R and Di Conza G and Barbaro F and Zini N and Consolini E and Dallatana D and Antoniel M and Quarantini E and Quarantini M and Maioli S and Bruni CA and Elviri L and Panseri S and Sprio S and Sandri M and Tampieri A},
url = {https://www.frontiersin.org/articles/10.3389/fimmu.2020.01737/full},
doi = {10.3389/fimmu.2020.01737},
year = {2020},
date = {2020-01-01},
journal = {Frontiers in immunology},
volume = {11},
pages = {1737},
abstract = {Osteoporosis stems from an unbalance between bone mineral resorption and deposition. Among the numerous cellular players responsible for this unbalance bone marrow (BM) monocytes/macrophages, mast cells, T and B lymphocytes, and dendritic cells play a key role in regulating osteoclasts, osteoblasts, and their progenitor cells through interactions occurring in the context of the different bone compartments (cancellous and cortical). Therefore, the microtopography of immune cells inside trabecular and compact bone is expected to play a relevant role in setting initial sites of osteoporotic lesion. Indeed, in physiological conditions, each immune cell type preferentially occupies either endosteal, subendosteal, central, and/or perisinusoidal regions of the BM. However, in the presence of an activation, immune cells recirculate throughout these different microanatomical areas giving rise to a specific distribution. As a result, the trabeculae of the cancellous bone and endosteal free edge of the diaphyseal case emerge as the primary anatomical targets of their osteoporotic action. Immune cells may also transit from the BM to the depth of the compact bone, thanks to the efferent venous capillaries coursing in the Haversian and Volkmann canals. Consistently, the innermost parts of the osteons and the periosteum are later involved by their immunomodulatory action, becoming another site of mineral reabsorption in the course of an osteoporotic insult. The novelty of our updating is to highlight the microtopography of bone immune cells in the cancellous and cortical compartments in relation to the most consistent data on their action in bone remodeling, to offer a mechanist perspective useful to dissect their role in the osteoporotic process, including bone damage derived from the immunomodulatory effects of endocrine disrupting chemicals.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2019
@article{%a1:%Y%n,
title = {Auto-Allo Graft Parallel Juxtaposition for Improved Neuroregeneration in Peripheral Nerve Reconstruction Based on Acellular Nerve Allografts.},
author = {Boriani F and Savarino L and Fazio N and Pedrini FA and Fini M and Nicoli Aldini N and Martini L and Zini N and Bernardini M and Bolognesi F and Marchetti C and Baldini N},
url = {https://insights.ovid.com/crossref?an=00000637-900000000-96969},
doi = {doi: 10.1097/SAP.0000000000001900},
year = {2019},
date = {2019-02-21},
journal = {Annals of plastic surgery},
volume = {83},
number = {3},
pages = {318-325},
abstract = {INTRODUCTION: Nerve repair poses a significant surgical challenge, and much research on this topic for improvement in reconstruction of segmental defects is ongoing. The aims of the study were to reconfirm the accuracy and safety of a previously described nerve decellularization method on a larger experimental population of rabbits, as well as on human nerves, and to establish in vivo the efficacy of a new-concept mixed graft, comprising autologous and acellular nerve allograft components within the same graft. METHODS: Acellular nerve allografts were implanted into tibial nerve defects of 5 rabbits (group A), autografts were implanted, representing the criterion standard, in other 5 animals (group B), and the innovative technique was used in the remaining 5 (group C). Twelve weeks postoperatively, nerve conduction evaluations were performed; animals were euthanatized, and grafts were harvested and morphologically, histomorphometrically, and immunohistochemically analyzed. Eventually, a preliminary in vitro validation of the decellularization method was performed on human nerves from a cadaver. RESULTS: No clinical adverse effect was revealed during all the experimental times. No tissue reaction was observed, and in all groups, regenerated fascicles and bundles were shown by histology. However, both histology and histomorphometry demonstrated a better regenerative efficiency in group C. The morphological evaluation of the human nerve treated with the novel method showed complete decellularization. CONCLUSION: The microsurgical combined model demonstrated a better neuroregeneration than did pure autografts and acellular nerve allografts. The decellularization method seemed effective also on human nerves. Deeper investigations are necessary to further validate and transfer this new encouraging protocol to the clinical arena.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_52,
title = {Chitosan-based scaffold counteracts hypertrophic and fibrotic markers in chondrogenic differentiated mesenchymal stromal cells.},
author = {Manferdini C and Gabusi E and Sartore L and Dey K and Agnelli S and Almici C and Bianchetti A and Zini N and Russo D and Re F and Mariani E and Lisignoli G},
url = {https://onlinelibrary.wiley.com/doi/full/10.1002/term.2941},
doi = {10.1002/term.2941},
year = {2019},
date = {2019-02-24},
journal = {Journal of tissue engineering and regenerative medicine},
volume = {13},
number = {10},
pages = {1896-1911},
abstract = {Cartilage tissue engineering remains problematic because no systems are able to induce signals that contribute to native cartilage structure formation. Therefore, we tested the potentiality of gelatin-polyethylene glycol scaffolds containing three different concentrations of chitosan (CH; 0%, 8%, and 16%) on chondrogenic differentiation of human platelet lysate-expanded human bone marrow mesenchymal stromal cells (hBM-MSCs). Typical chondrogenic (SOX9, collagen type 2, and aggrecan), hypertrophic (collagen type 10), and fibrotic (collagen type 1) markers were evaluated at gene and protein level at Days 1, 28, and 48. We demonstrated that 16% CH scaffold had the highest percentage of relaxation with the fastest relaxation rate. In particular, 16% CH scaffold, combined with chondrogenic factor TGFβ3, was more efficient in inducing hBM-MSCs chondrogenic differentiation compared with 0% or 8% scaffolds. Collagen type 2, SOX9, and aggrecan showed the same expression in all scaffolds, whereas collagen types 10 and 1 markers were efficiently down-modulated only in 16% CH. We demonstrated that using human platelet lysate chronically during hBM-MSCs chondrogenic differentiation, the chondrogenic, hypertrophic, and fibrotic markers were significantly decreased. Our data demonstrate that only a high concentration of CH, combined with TGFβ3, creates an environment capable of guiding in vitro hBM-MSCs towards a phenotypically stable chondrogenesis. Copyrights 2019 John Wiley & Sons, Ltd.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_56,
title = {Extracellular Nanovesicles Secreted by Human Osteosarcoma Cells Promote Angiogenesis.},
author = {Perut F and Roncuzzi L and Zini N and Massa A and Baldini N},
url = {https://www.mdpi.com/2072-6694/11/6/779},
doi = {Cancers (Basel)},
year = {2019},
date = {2019-06-05},
journal = {Cancers (Basel)},
volume = {11},
number = {6},
pages = {e779},
abstract = {Angiogenesis involves a number of different players among which extracellular nanovesicles (EVs) have recently been proposed as an efficient cargo of pro-angiogenic mediators. Angiogenesis plays a key role in osteosarcoma (OS) development and progression. Acidity is a hallmark of malignancy in a variety of cancers, including sarcomas, as a result of an increased energetic metabolism. The aim of this study was to investigate the role of EVs derived from osteosarcoma cells on angiogenesis and whether extracellular acidity, generated by tumor metabolism, could influence EVs activity. For this purpose, we purified and characterized EVs from OS cells maintained at either acidic or neutral pH. The ability of EVs to induce angiogenesis was assessed in vitro by endothelial cell tube formation and in vivo using chicken chorioallantoic membrane. Our findings demonstrated that EVs derived from osteosarcoma cells maintained either in acidic or neutral conditions induced angiogenesis. The results showed that miRNA and protein content of EVs cargo are correlated with pro-angiogenic activity and this activity is increased by the acidity of tumor microenvironment. This study provides evidence that EVs released by human osteosarcoma cells act as carriers of active angiogenic stimuli that are able to promote endothelial cell functions relevant to angiogenesis.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y%f,
title = {The masks of Lorenzo Tenchini: their anatomy and surgical/bioengineering clues.},
author = {Barbaro F and Consolini E and Toscani G and Zini N and Dallatana D and Setti P and Mosca S and Di Conza G and Bassi E and Quarantini E and Quarantini M and Raposio E and Gorreri M and Porro A and Toni R.},
url = {https://onlinelibrary.wiley.com/doi/full/10.1111/joa.13069},
doi = {10.1111/joa.13069},
year = {2019},
date = {2019-12-17},
journal = {Journal of anatomy},
volume = {235},
number = {6},
pages = {1036-1044},
abstract = {An academic, anatomist, and Lombrosian psychiatrist active at the University of Parma in Italy at the end of the 19th century, Lorenzo Tenchini produced ceroplastic-like masks that are unique in the anatomical Western context. These were prepared from 1885 to 1893 with the aim of 'cataloguing' the behaviour of prison inmates and psychiatric patients based on their facial surface anatomy. Due to the lack of any reference to the procedure used to prepare the masks, studies were undertaken by our group using X-ray scans, infrared spectroscopy, bioptic sampling, and microscopy analysis of the mask constituents. Results showed that the masks were stratified structures including plaster, cotton gauze/human epidermis, and wax, leading to a fabrication procedure reminiscent of 'additive layer manufacturing'. Differences in the depths of these layers were observed in relation to the facial contours, suggesting an attempt to reproduce, at least partially, the three-dimensional features of the facial soft tissues. We conclude the Tenchini masks are the first historical antecedent of the experimental method for face reconstruction used in the early 2000s to test the feasibility of transferring a complete strip of face and scalp from a deceased donor to a living recipient, in preparation for a complete face transplant. In addition, the layering procedure adopted conceptually mimics that developed only in the late 20th century for computer-aided rapid prototyping, and recently applied to bioengineering with biomaterials for a number of human structures including parts of the skull and face. Finally, the masks are a relevant example of mixed ceroplastic-cutaneous preparations in the history of anatomical research for clinical purposes. Copyrights 2019 Anatomical Society.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2018
@article{%a1:%Y_133,
title = {5-Aza Exposure Improves Reprogramming Process Through Embryoid Body Formation in Human Gingival Stem Cells.},
author = {Diomede F and Zini N and Pizzicannella J and Merciaro I and Pizzicannella G and D'Orazio M and Piattelli A and Trubiani O},
url = {https://www.frontiersin.org/articles/10.3389/fgene.2018.00419/full},
doi = {10.3389/fgene.2018.00419},
year = {2018},
date = {2018-02-21},
journal = {Frontiers in genetics},
volume = {9},
pages = {419},
abstract = {Embryoid bodies (EBs) are three-dimensional aggregates formed by pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells. They are used as an in vitro model to evaluate early extraembryonic tissue formation and differentiation process. In the adult organisms, cell differentiation is controlled and realized through the epigenetic regulation of gene expression, which consists of various mechanisms including DNA methylation. One demethylating agent is represented by 5-Azacytidine (5-Aza), considered able to induce epigenetic changes through gene derepression. Human gingival mesenchymal stem cells (hGMSCs), an easily accessible stem cells population, migrated from neural crest. They are particularly apt as an in vitro study model in regenerative medicine and in systemic diseases. The ability of 5-Aza treatment to induce hGMSCs toward a dedifferentiation stage and in particular versus EBs formation was investigated. For this purpose hGMSCs were treated for 48 h with 5-Aza (5 μM). After treatment, hGMSCs are organized as round 3D structures (EBs-hGMSCs). At light and transmission electron microscopy, the cells at the periphery of EBs-hGMSCs appear elongated, while ribbon-shaped cells and smaller cells with irregular shape surrounded by extracellular matrix were present in the center. By RT-PCR, EBs-hGMSCs expressed specific transcription markers related to the three germ layers as MAP-2, PAX-6 (ectoderm), MSX-1, Flk-1 (mesoderm), GATA-4, and GATA-6 (endoderm). Moreover, in EB-hGMSCs the overexpression of DNMT1 and ACH3 other than the down regulation of p21 was detectable. Immunofluorescence staining also showed a positivity for specific etodermal and mesodermal markers. In conclusion, 5-Aza was able to induce the direct conversion of adult hGMSCs into cells of three embryonic lineages: endoderm, ectoderm, and mesoderm, suggesting their possible application in autologous cell therapy for clinical organ repair.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_110,
title = {Exosome-like nanovesicles isolated from Citrus limon L. exert antioxidative effect.},
author = {Baldini N and Torreggiani E and Roncuzzi L and Perut F and Zini N and Avnet S},
url = {http://www.eurekaselect.com/166345/article},
doi = {10.2174/1389201019666181017115755},
year = {2018},
date = {2018-02-16},
journal = {Current pharmaceutical biotechnology},
volume = {19},
number = {11},
pages = {877-885},
abstract = {Background: Exosome-like nanovesicles are biological nanostructures mediating cell-tocell communication and capable to load selected cargos also in the interaction among different species. Objective: We aimed to explore the content of exosome-like nanovesicles derived from Citrus limon L. and to analyze the effects of their uptake on human cells. Method: We isolated exosome-like nanovesicles from Citrus limon L. juice (EXO-CLs) by differential centrifugation. EXO-CLs were analyzed for short RNA content by advanced sequencing technologies, and for ascorbic acid (vitamin C) and citrate content by enzymatic assays. EXO-CLs anti-oxidant and pro-differentiative potential was evaluated in vitro on mesenchymal stromal cells (MSC), a common tool for regenerative strategies for several human tissues. Results: We showed that EXO-CLs carry detectable amounts of citrate and vitamin C and, although it was not possible to identify specific miRNAs, we detected short RNA sequences (20-30 bp) with unknown functions and with different distribution size in respect to whole Citrus limon L. juice. In vitro, EXO-CLs were uptaken by MSC and had a significant protective effect against oxidative stress. Furthermore, regarding the potential benefit for human bone health, we found that EXO-CLs modulate MSC differentiation versus the osteogenic lineage. Conclusion: We demonstrated that incubation with EXO-CLs exert antioxidant activity in human cells. This is most likely due to the direct delivery and uptake of micronutrients by human cells that are well preserved inside the nanovesicle membrane, including the unstable vitamin C. Based on our results, we speculate that fruit-derived nanovesicles have the potential to mediate interspecies influence after food intake.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_156,
title = {Immunoelectron microscopic localization of Collagen type XV during human mesenchymal stem cells mineralization.},
author = {Manferdini C and Zini N and Gabusi E and Paolella F and Lambertini E and Penolazzi L and Piva R and Lisignoli G},
url = {https://www.tandfonline.com/doi/full/10.1080/03008207.2017.1408600},
doi = {10.1080/03008207.2017.1408600},
year = {2018},
date = {2018-12-20},
journal = {Connective tissue research},
volume = {59},
number = {Suppl. 1},
pages = {42-45},
abstract = {Purpose/Aim of the study. Collagen type XV (ColXV) was identified, in our previews studies, as a novel component of bone extracellular matrix. The present study aims to investigate ColXV localization during mineralization of osteodifferentiated human mesenchymal stem cells (hMSCs). MATERIAL AND METHODS: hMSCs cultured in osteogenic medium have been analyzed at day 14 and 28 for mineral matrix deposition by alizarin red S staining, ultrastructural analysis and ColXV localization by immunogold electron microscopy. RESULTS: Our data show an intimate association between ColXV and fibrillar components in areas localized far from mineralized nodules. CONCLUSIONS: We have demonstrated the efficacy of ultrastructural analysis, combined with immunocytochemistry, to establish a temporal and spatial localization of ColXV. This data, added to previous evidences, contribute to validate the negative effects of calcium deposits on ColXV expression.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2017
@article{%a1:%Y_226,
title = {A novel technique for decellularization of allogenic nerves and in vivo study of their use for peripheral nerve reconstruction.},
author = {Boriani F and Fazio N and Fotia C and Savarino L and Nicoli Aldini N and Martini L and Zini N and Bernardini M and Baldini N},
url = {http://onlinelibrary.wiley.com/doi/10.1002/jbm.a.36090/abstract},
doi = {10.1002/jbm.a.36090},
year = {2017},
date = {2017-02-23},
journal = {Journal of biomedical materials research. Part A},
volume = {105},
number = {8},
pages = {2228-2240},
abstract = {Autografts represent the gold standard for peripheral nerve reconstruction but their limited availability, the discrepancy of nerve caliber and long surgical times are drawbacks. Allografts have therefore become a valid alternative option. In particular, acellular nerve allografts (ANAs) rather than fresh allografts do not need immunosuppression and appear to be safe and effective based on recent studies. An innovative method was conceived to obtain ANAs, so as to speed up nerve decellularization, without compromising nerve architecture, and without breaking the asepsis chain. Several detergent-based techniques, integrated with sonication and mechanical stirring, were tested in vitro on rabbit nerves, to identify, by microscopy and immunohistochemistry, the most effective protocol in terms of cell lysis and cellular debris clearance, while maintaining nerve architecture. Furthermore, a pilot in vivo study was performed: ANAs were implanted into tibial nerve defects of three rabbits, and autografts, representing the gold standard, in other three animals. Twelve weeks postoperatively, rabbits were clinically evaluated and euthanasized; grafts were harvested and microscopically and histomorphometrically analyzed. The method proved to be effective in vitro: the treatment removed axons, myelin and cells, without altering nerve architecture. The in vivo study did not reveal any adverse effect: animals maintained normal weight and function of posterior limb during the entire experimental time. A mild fibrotic reaction was observed, macrophages and leukocytes were rare or absent; ANAs regenerated fascicles and bundles were comparable versus autografts. Based on these results, this decellularization protocol is encouraging and deserves deeper investigations with further pre-clinical and clinical studies. This article is protected by copyright. All rights reserved.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_191,
title = {Blocking Tumor-Educated MSC Paracrine Activity Halts Osteosarcoma Progression.},
author = {Baglio SR and Lagerweij T and Pérez-Lanzón M and Ho XD and Léveillé N and Melo SA and Cleton-Jansen AM and Jordanova ES and Roncuzzi L and Greco M and van Eijndhoven MAJ and Grisendi G and Dominici M and Bonafede R and Lougheed SM and de Gruijl TD and Zini N and Cervo S and Steffan A and Canzonieri V and Martson A and Maasalu K and Köks S and Wurdinger T and Baldini N and Pegtel DM},
url = {http://clincancerres.aacrjournals.org/content/23/14/3721.long},
doi = {10.1158/1078-0432.CCR-16-2726},
year = {2017},
date = {2017-02-16},
journal = {Clinical cancer research},
volume = {23},
number = {14},
pages = {3721-3733},
abstract = {Purpose: Human osteosarcoma is a genetically heterogeneous bone malignancy with poor prognosis despite the employment of aggressive chemotherapy regimens. Because druggable driver mutations have not been established, dissecting the interactions between osteosarcoma cells and supporting stroma may provide insights into novel therapeutic targets.Experimental Design: By using a bioluminescent orthotopic xenograft mouse model of osteosarcoma, we evaluated the effect of tumor extracellular vesicle (EV)-educated mesenchymal stem cells (TEMSC) on osteosarcoma progression. Characterization and functional studies were designed to assess the mechanisms underlying MSC education. Independent series of tissue specimens were analyzed to corroborate the preclinical findings, and the composition of patient serum EVs was analyzed after isolation with size-exclusion chromatography.Results: We show that EVs secreted by highly malignant osteosarcoma cells selectively incorporate a membrane-associated form of TGFbeta, which induces proinflammatory IL6 production by MSCs. TEMSCs promote tumor growth, accompanied with intratumor STAT3 activation and lung metastasis formation, which was not observed with control MSCs. Importantly, intravenous administration of the anti-IL6 receptor antibody tocilizumab abrogated the tumor-promoting effects of TEMSCs. RNA-seq analysis of human osteosarcoma tissues revealed a distinct TGFbeta-induced prometastatic gene signature. Tissue microarray immunostaining indicated active STAT3 signaling in human osteosarcoma, consistent with the observations in TEMSC-treated mice. Finally, we isolated pure populations of EVs from serum and demonstrated that circulating levels of EV-associated TGFbeta are increased in osteosarcoma patients.Conclusions: Collectively, our findings suggest that TEMSCs promote osteosarcoma progression and provide the basis for testing IL6- and TGFbeta-blocking agents as new therapeutic options for osteosarcoma patients. Clin Cancer Res; 1-13. ©2017 AACR.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2016
@article{%a1:%Y_248,
title = {Altered pH gradient at the plasma membrane of osteosarcoma cells is a key mechanism of drug resistance.},
author = {Avnet S and Lemma S and Cortini M and Pellegrini P and Perut F and Zini N and Kusuzaki K and Chano T and Grisendi G and Dominici M and De Milito A and Baldini N},
url = {http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path[]=11503&pubmed-linkout=1},
doi = {10.18632/oncotarget.11503},
year = {2016},
date = {2016-02-12},
journal = {Oncotarget},
volume = {7},
number = {39},
pages = {63408-63423},
abstract = {Current therapy of osteosarcoma (OS), the most common primary bone malignancy, is based on a combination of surgery and chemotherapy. Multidrug resistance mediated by P-glycoprotein (P-gp) overexpression has been previously associated with treatment failure and progression of OS, although other mechanisms may also play a role. We considered the typical acidic extracellular pH (pHe) of sarcomas, and found that doxorubicin (DXR) cytotoxicity is reduced in P-gp negative OS cells cultured at pHe 6.5 compared to standard 7.4. Short-time (24-48 hours) exposure to low pHe significantly increased the number and acidity of lysosomes, and the combination of DXR with omeprazole, a proton pump inhibitor targeting lysosomal acidity, significantly enhanced DXR cytotoxicity. In OS xenografts, the combination treatment of DXR and omeprazole significantly reduced tumor volume and body weight loss. The impaired toxicity of DXR at low pHe was not associated with increased autophagy or lysosomal acidification, but rather, as shown by SNARF staining, with a reversal of the pH gradient at the plasma membrane (ΔpHcm), eventually leading to a reduced DXR intracellular accumulation. Finally, the reversal of ΔpHcm in OS cells promoted resistance not only to DXR, but also to cisplatin and methotrexate, and, to a lesser extent, to vincristine. Altogether, our findings show that, in OS cells, short-term acidosis induces resistance to different chemotherapeutic drugs by a reversal of ΔpHcm, suggesting that buffer therapies or regimens including proton pump inhibitors in combination to low concentrations of conventional anticancer agents may offer novel solutions to overcome drug resistance.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_295,
title = {CD99 triggering induces methuosis of Ewing sarcoma cells through IGF-1R/RAS/Rac1 signaling.},
author = {Manara MC and Terracciano M and Mancarella C and Sciandra M and Guerzoni C and Pasello M and Grilli A and Zini N and Picci P and Colombo MP and Morrione A and Scotlandi K},
url = {http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path[]=13160&pubmed-linkout=1},
doi = {10.18632/oncotarget.13160},
year = {2016},
date = {2016-02-18},
journal = {Oncotarget},
volume = {7},
number = {48},
pages = {79925-79942},
abstract = {CD99 is a cell surface molecule that has emerged as a novel target for Ewing sarcoma (EWS), an aggressive pediatric bone cancer. This report provides the first evidence of methuosis in EWS, a non-apoptotic form of cell death induced by an antibody directed against the CD99 molecule. Upon mAb triggering, CD99 induces an IGF-1R/RAS/Rac1 complex, which is internalized into RAB5-positive endocytic vacuoles. This complex is then dissociated, with the IGF-1R recycling to the cell membrane while CD99 and RAS/Rac1 are sorted into immature LAMP-1-positive vacuoles, whose excessive accumulation provokes methuosis. This process, which is not detected in CD99-expressing normal mesenchymal cells, is inhibited by disruption of the IGF-1R signaling, whereas enhanced by IGF-1 stimulation. Induction of IGF-1R/RAS/Rac1 was also observed in the EWS xenografts that respond to anti-CD99 mAb, further supporting the role of the IGF/RAS/Rac1 axis in the hyperstimulation of macropinocytosis and selective death of EWS cells. Thus, we describe a vulnerability of EWS cells, including those resistant to standard chemotherapy, to a treatment with anti-CD99 mAb, which requires IGF-1R/RAS signaling but bypasses the need for their direct targeting. Overall, we propose CD99 targeting as new opportunity to treat EWS patients resistant to canonical apoptosis-inducing agents.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_291,
title = {Energy metabolism in osteoclast formation and activity.},
author = {Lemma S and Sboarina M and Porporato PE and Zini N and Sonveaux P and Di Pompo G and Baldini N and Avnet S},
url = {10.1016/j.biocel.2016.08.034},
doi = {10.1016/j.biocel.2016.08.034},
year = {2016},
date = {2016-02-20},
journal = {International Journal of Biochemistry And Cell Biology},
volume = {79},
pages = {168-180},
abstract = {Osteoclastogenesis and osteolysis are energy-consuming processes supported by high metabolic activities. In human osteoclasts derived from the fusion of monocytic precursors, we found a substantial increase in the number of mitochondria with differentiation. In mature osteoclasts, mitochondria were also increased in size, rich of cristae and arranged in a complex tubular network. When compared with immature cells, fully differentiated osteoclasts showed higher levels of enzymes of the electron transport chain, a higher mitochondrial oxygen consumption rate and a lower glycolytic efficiency, as evaluated by extracellular flux analysis and by the quantification of metabolites in the culture supernatant. Thus, oxidative phosphorylation appeared the main bioenergetic source for osteoclast formation. Conversely, we found that bone resorption mainly relied on glycolysis. In fact, osteoclast fuelling with galactose, forcing cells to depend on Oxidative Phosphorylation by reducing the rate of glycolysis, significantly impaired Type I collagen degradation, whereas non-cytotoxic doses of rotenone, an inhibitor of the mitochondrial complex I, enhanced osteoclast activity. Furthermore, we found that the enzymes associated to the glycolytic pathway are localised close to the actin ring of polarised osteoclasts, where energy-demanding activities associated with bone degradation take place. In conclusion, we demonstrate that the energy required for osteoclast differentiation mainly derives from mitochondrial oxidative metabolism, whereas the peripheral cellular activities associated with bone matrix degradation are supported by glycolysis. A better understanding of human osteoclast energy metabolism holds the potential for future therapeutic interventions aimed to target osteoclast activity in different pathological conditions of bone.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_269,
title = {Human periodontal ligament stem cells cultured onto cortico-cancellous scaffold drive bone regenerative process.},
author = {Diomede F and Zini N and Gatta V and Fulle S and Merciaro I and D'Aurora M and La Rovere RM and Traini T and Pizzicannella J and Ballerini P and Caputi S and Piattelli A and Trubiani O},
url = {https://www.ecmjournal.org/papers/vol032/vol032a12.php},
year = {2016},
date = {2016-09-16},
journal = {European Cells & Materials},
volume = {32},
pages = {181-201},
abstract = {The purpose of this work was to test, in vitro and in vivo, a new tissue-engineered construct constituted by porcine cortico-cancellous scaffold (Osteobiol Dual Block) (DB) and xeno-free ex vivo culture of human Periodontal Ligament Stem Cells (hPDLSCs). hPDLSCs cultured in xeno-free media formulation preserved the stem cells' morphological features, the expression of stemness and pluripotency markers, and their ability to differentiate into mesenchymal lineage. Transmission electron microscopy analysis suggested that after one week of culture, both noninduced and osteogenic differentiation induced cells joined and grew on DB secreting extracellular matrix (ECM) that in osteogenic induced samples was hierarchically assembled in fibrils. Quantitative RT-PCR (qRT-PCR) showed the upregulation of key genes involved in the bone differentiation pathway in both differentiated and undifferentiated hPDLSCs cultured with DB (hPDLSCs/DB). Functional studies revealed a significant increased response of calcium transients in the presence of DB, both in undifferentiated and differentiated cells stimulated with calcitonin and parathormone, suggesting that the biomaterial could drive the osteogenic differentiation process of hPDLSCs. These data were confirmed by the increase of gene expression of L-type voltage-dependent Ca2+ (VDCCL), subunits α1C and α2D1 in undifferentiated cells in the presence of DB. In vivo implantation of the hPDLSCs/DB living construct in the mouse calvaria evidenced a precocious osteointegration and vascularisation process. Our results suggest consideration of DB as a biocompatible, osteoinductive and osteoconductive biomaterial, making it a promising tool to regulate cell activities in biological environments and for a potential use in the development of new custom-made tissue engineering.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_259,
title = {Hyaluronan scaffold supports osteogenic differentiation of bone marrow concentrate cells.},
author = {Cavallo C and Desando G and Ferrari A and Zini N and Mariani E and Grigolo B},
url = {https://www.biolifesas.org/biolife/jbrha-2/},
year = {2016},
date = {2016-02-20},
journal = {Journal of Biological Regulators and Homeostatic Agents},
volume = {30},
number = {2},
pages = {409-420},
abstract = {Osteochondral lesions are considered a challenge for orthopedic surgeons. Currently, the treatments available are often unsatisfactory and unable to stimulate tissue regeneration. Tissue engineering offers a new therapeutic strategy, taking into account the role exerted by cells, biomaterial and growth factors in restoring tissue damage. In this light, Mesenchymal Stem Cells (MSCs) have been indicated as a fascinating tool for regenerative medicine thanks to their ability to differentiate into bone, cartilage and adipose tissue. However, in vitro-cultivation of MSCs could be associated with some risks such as de-differentiation/reprogramming, infection and contaminations of the cells. To overcome these shortcomings, a new approach is represented by the use of Bone Marrow Concentrate (BMC), that could allow the delivery of cells surrounded by their microenvironment in injured tissue. For this purpose, cells require a tridimensional scaffold that can support their adhesion, proliferation and differentiation. This study is focused on the potentiality of BMC seeded onto a hyaluronan-based scaffold (Hyaff-11) to differentiate into osteogenic lineage. This process depends on the specific interaction between cells derived from bone marrow (surrounded by their niche) and scaffold, that create an environment able to support the regeneration of damaged tissue. The data obtained from the present study demonstrate that BMC grown onto Hyaff-11 are able to differentiate toward osteogenic sense, producing specific osteogenic genes and matrix proteins.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_315,
title = {Multimodal transfer of MDR by exosomes in human osteosarcoma},
author = {Torreggiani E and Roncuzzi L and Perut F and Zini N and Baldini N},
url = {https://www.spandidos-publications.com/ijo/49/1/189},
doi = {10.3892/ijo.2016.3509},
year = {2016},
date = {2016-07-19},
journal = {International Journal of Oncology},
volume = {49},
number = {1},
pages = {189-196},
abstract = {Exosomes are extracellular vesicles released by both normal and tumour cells which are involved in a new intercellular communication pathway by delivering cargo (e.g., proteins, microRNAs, mRNAs) to recipient cells. Tumour-derived exosomes have been shown to play critical roles in different stages of tumour growth and progression. In this study, we investigated the potential role of exosomes to transfer the multidrug resistance (MDR) phenotype in human osteosarcoma cells. Exosomes were isolated by differential centrifugation of culture media from multidrug resistant human osteosarcoma MG-63DXR30 (Exo/DXR) and MG-63 parental cells (Exo/S). Exosome purity was examined by transmission electron microscopy and confirmed by immunoblot analysis for the expression of specific exosomal markers. Our data showed that exosomes derived from doxorubicin-resistant osteosarcoma cells could be taken up into secondary cells and induce a doxorubicin-resistant phenotype. The incubation of osteosarcoma cells with Exo/DXR decreased the sensitivity of parental cells to doxorubicin, while exposure with Exo/S was ineffective. In addition, we demonstrated that Exo/DXR expressed higher levels of MDR-1 mRNA and P-glycoprotein compared to Exo/S (p=0.03). Interestingly, both MDR-1 mRNA and P-gp increased in MG-63 cells after incubation with Exo/DXR, suggesting this as the main mechanism of exosome-mediated transfer of drug resistance. Our findings suggest that multidrug resistant osteosarcoma cells are able to spread their ability to resist the effects of doxorubicin treatment on sensitive cells by transferring exosomes carrying MDR-1 mRNA and its product P-glycoprotein.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_289,
title = {Non‑invasive prostate cancer detection by measuring miRNA variants (isomiRs) in urine extracellular vesicles.},
author = {Koppers-Lalic D and Hackenberg M and Menezes R and Misovic B and Wachalska M and Geldof A and Zini N and Reijke T and Wurdinger T and Vis A and Moorselaar JV and Pegtel M and Bijnsdorp I},
url = {http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path[]=8124&pubmed-linkout=1},
doi = {10.18632/oncotarget.8124},
year = {2016},
date = {2016-04-19},
journal = {Oncotarget},
volume = {7},
number = {16},
pages = {22566-22578},
abstract = {In many cancer types, the expression and function of ~22 nucleotide-long microRNAs (miRNA) is deregulated. Mature miRNAs can be stably detected in extracellular vesicles (EVs) in biofluids, therefore they are considered to have great potential as biomarkers. In the present study, we investigated whether miRNAs have a distinct expression pattern in urine-EVs of prostate cancer (PCa) patients compared to control males. By next generation sequencing, we determined the miRNA expression in a discovery cohort of 4 control men and 9 PCa patients. miRNAs were validated by using a stemloop RT-PCR in an independent cohort of 74 patients (26 control and 48 PCa-patients). Whereas standard mapping protocols identified > 10 PCa associated miRNAs in urinary EVs, miR-21, miR-375 and miR-204 failed to robustly discriminate for disease in a validation study with RT-PCR-detection of mature miRNA sequences. In contrast, we observed that miRNA isoforms (isomiRs) with 3' end modifications were highly discriminatory between samples from control men and PCa patients. Highly differentially expressed isomiRs of miR-21, miR-204 and miR-375 were subsequently validated in an independent group of 74 patients. Receiver-operating characteristic analysis was performed to evaluate the diagnostic performance of three isomiRs, resulting in a 72.9% sensitivity with a high (88%) specificity and an area under the curve (AUC) of 0.866. In comparison, prostate specific antigen had an AUC of 0.707 and measuring the mature form of these miRNAs yielded a lower 70.8% sensitivity and 72% specificity (AUC 0.766). We propose that isomiRs may carry discriminatory information which is useful to generate stronger biomarkers.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_296,
title = {Osteogenic potential of dualblocks cultured with human periodontal ligament stem cells: in vitro and synchrotron microtomography study.},
author = {Manescu A and Giuliani A and Mohammadi S and Tromba G and Mazzoni S and Diomede F and Zini N and Piattelli A and Trubiani O},
url = {http://onlinelibrary.wiley.com/doi/10.1111/jre.12289/abstract},
doi = {10.1111/jre.12289},
year = {2016},
date = {2016-02-29},
journal = {Journal of Periodontal Research},
volume = {51},
number = {1},
pages = {112-124},
abstract = {"BACKGROUND AND OBJECTIVE: In the present study, the early stages of in vitro bone formation in collagenated porcine scaffolds cultured with human periodontal ligament cells were investigated. The comparison between the osteogenic potential of this structure in basal and differentiating culture media was explored to predict the mechanism of its biological behavior as graft in human defect. Results were validated by synchrotron radiation X-Ray phase contrast computed microtomography (micro-CT). As the periodontal disease plays a key role in systemic and oral diseases, it is crucial to find advanced therapeutic clinical interventions to repair periodontal defects. This has been recently explored using cells and tissues developed in vitro that should ideally be immunologically, functionally, structurally and mechanically identical to the native tissue. MATERIAL AND METHODS:
In vitro cultures of human periodontal ligament cells, easily obtained by scraping of alveolar crestal and horizontal fibers of the periodontal ligament, were seeded on to collagenated porcine blocks constituted by natural cancellous and cortical bone. 3D images were obtained by synchrotron radiation micro-CT and processed with a phase-retrieval algorithm based on the transport of intensity equation. RESULTS: Starting from the second week of culture, newly formed mineralized bone was detected in all the scaffolds, both in basal and differentiating media. Bone mineralization was proved to occur preferentially in the trabecular portion and in differentiating media. CONCLUSION: The chosen method, supported by phase contrast micro-CT analysis, successfully and quantitatively monitored the early stages of bone formation and the rate of the bioscaffold resorption in basal and differentiating culture media. 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd."},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
In vitro cultures of human periodontal ligament cells, easily obtained by scraping of alveolar crestal and horizontal fibers of the periodontal ligament, were seeded on to collagenated porcine blocks constituted by natural cancellous and cortical bone. 3D images were obtained by synchrotron radiation micro-CT and processed with a phase-retrieval algorithm based on the transport of intensity equation. RESULTS: Starting from the second week of culture, newly formed mineralized bone was detected in all the scaffolds, both in basal and differentiating media. Bone mineralization was proved to occur preferentially in the trabecular portion and in differentiating media. CONCLUSION: The chosen method, supported by phase contrast micro-CT analysis, successfully and quantitatively monitored the early stages of bone formation and the rate of the bioscaffold resorption in basal and differentiating culture media. 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd."@article{%a1:%Y_297,
title = {Specific inductive potential of a novel nanocomposite biomimetic biomaterial for osteochondral tissue regeneration},
author = {Manferdini C and Cavallo C and Grigolo B and Fiorini M and Nicoletti A and Gabusi E and Zini N and Pressato D and Facchini A and Lisignoli G},
url = {http://onlinelibrary.wiley.com/doi/10.1002/term.1723/abstract;jsessionid=0358FE4D72F098C66724266255F4F49A.f02t04},
year = {2016},
date = {2016-05-27},
journal = {Journal of Tissue Engineering and Regenerative Medicine},
volume = {10},
pages = {374-391},
abstract = {Osteochondral lesions require treatment to restore the biology and functionality of the joint. A novel nanostructured biomimetic gradient scaffold was developed to mimic the biochemical and biophysical properties of the different layers of native osteochondral structure. The present results show that the scaffold presents important physicochemical characteristics and can support the growth and differentiation of mesenchymal stromal cells (h-MSCs), which adhere and penetrate into the cartilaginous and bony layers. H-MSCs grown in chondrogenic or osteogenic medium decreased their proliferation during days 14-52 on both scaffold layers and in medium without inducing factors used as controls. Both chondrogenic and osteogenic differentiation of h-MSCs occurred from day 28 and were increased on day 52, but not in the control medium. Safranin O staining and collagen type II and proteoglycans immunostaining confirmed that chondrogenic differentiation was specifically induced only in the cartilaginous layer. Conversely, von Kossa staining, osteocalcin and osteopontin immunostaining confirmed that osteogenic differentiation occurred on both layers. This study shows the specific potential of each layer of the biomimetic scaffold to induce chondrogenic or osteogenic differentiation of h-MSCs. These processes depended mainly on the media used but not the biomaterial itself, suggesting that the local milieu is fundamental for guiding cell differentiation.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
2015
@article{%a1:%Y_338,
title = {Assessment of an efficient xeno-free culture system for human periodontal ligament stem cells.},
author = {Trubiani O and Piattelli A and Gatta V and Marchisio M and Diomede F and D'Aurora M and Merciaro I and Pierdomenico L and Maraldi NM and Zini N},
url = {https://www.liebertpub.com/doi/full/10.1089/ten.TEC.2014.0024?url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org&rfr_dat=cr_pub%3Dpubmed&},
doi = {10.1089/ten.TEC.2014.0024},
year = {2015},
date = {2015-02-11},
journal = {Tissue Engineering. Part C, Methods},
volume = {21},
number = {1},
pages = {52-64},
abstract = {The possibility of transplanting adult stem cells into damaged organs has opened new prospects for the treatment of several human pathologies. The purpose of this study was to develop a culture system for the expansion and production of human Periodontal Ligament Stem Cells (hPDLSCs) using a new xeno-free media formulation and ensuring the maintenance of the stem cells features comprising the multiple passage expansion, mesengenic lineage differentiation, cellular phenotype, and genomic stability, essential elements for conforming to translation to cell therapy. Somatic stem cells were isolated from the human periodontium using a minimally invasive periodontal access flap surgery in healthy donors. Expanded hPDLSCs in a xeno-free culture showed the morphological features of stem cells, expressed the markers associated with pluripotency, and a normal karyotype. Under appropriate culture conditions, hPDLSCs presented adipogenic and osteogenic potential; indeed, a very high accumulation of lipid droplets was evident in the cytoplasm of adipogenic-induced cells, and indisputable evidence of osteogenic differentiation, investigated by transmission electron microscopy, and analyzed for gene expression analysis has been shown. Based on these data, the novel xeno-free culture method might provide the basis for Good Manufacturing Procedure culture of autologous stem cells, readily accessible from human periodontium, and can be a resource to facilitate their use in human clinical studies for potential therapeutic regeneration.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_371,
title = {Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species.},
author = {Baglio SR and Rooijers K and Koppers-Lalic D and Verweij FJ and Perez Lanzon M and Zini N and Naaijkens B and Perut F and Niessen HW and Baldini N and Pegtel DM},
url = {https://stemcellres.biomedcentral.com/articles/10.1186/s13287-015-0116-z},
doi = {10.1186/s13287-015-0116-z},
year = {2015},
date = {2015-07-01},
journal = {Stem Cell Research & Therapy},
volume = {6},
pages = {127},
abstract = {INTRODUCTION: Administration of mesenchymal stem cells (MSCs) represents a promising treatment option for patients suffering from immunological and degenerative disorders. Accumulating evidence indicates that the healing effects of MSCs are mainly related to unique paracrine properties, opening opportunities for secretome-based therapies. Apart from soluble factors, MSCs release functional small RNAs via extracellular vesicles (EVs) that seem to convey essential features of MSCs. Here we set out to characterize the full small RNAome of MSC-produced exosomes. METHODS: We set up a protocol for isolating exosomes released by early passage adipose- (ASC) and bone marrow-MSCs (BMSC) and characterized them via electron microscopy, protein analysis and small RNA-sequencing. We developed a bioinformatics pipeline to define the exosome-enclosed RNA species and performed the first complete small RNA characterization of BMSCs and ASCs and their corresponding exosomes in biological replicates. RESULTS: Our analysis revealed that primary ASCs and BMSCs have highly similar small RNA expression profiles dominated by miRNAs and snoRNAs (together 64-71 %), of which 150-200 miRNAs are present at physiological levels. In contrast, the miRNA pool in MSC exosomes is only 2-5 % of the total small RNAome and is dominated by a minor subset of miRNAs. Nevertheless, the miRNAs in exosomes do not merely reflect the cellular content and a defined set of miRNAs are overrepresented in exosomes compared to the cell of origin. Moreover, multiple highly expressed miRNAs are precluded from exosomal sorting, consistent with the notion that these miRNAs are involved in functional repression of RNA targets. While ASC and BMSC exosomes are similar in RNA class distribution and composition, we observed striking differences in the sorting of evolutionary conserved tRNA species that seems associated with the differentiation status of MSCs, as defined by Sox2, POU5F1A/B and Nanog expression. CONCLUSIONS: We demonstrate that primary MSCs release small RNAs via exosomes, which are increasingly implicated in intercellular communications. tRNAs species, and in particular tRNA halves, are preferentially released and their specific sorting into exosomes is related to MSC tissue origin and stemness. These findings may help to understand how MSCs impact neighboring or distant cells with possible consequences for their therapeutic usage.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_389,
title = {Novel nano-composite biomimetic biomaterial allows chondrogenic and osteogenic differentiation of bone marrow concentrate derived cells.},
author = {Grigolo B and Cavallo C and Desando G and Manferdini C and Lisignoli G and Ferrari A and Zini N and Facchini A},
url = {https://link.springer.com/article/10.1007%2Fs10856-015-5500-9},
doi = {10.1007/s10856-015-5500-9},
year = {2015},
date = {2015-04-30},
journal = {Journal of Materials Science. Materials in Medicine},
volume = {26},
number = {4},
pages = {173},
abstract = {In clinical orthopedics suitable materials that induce and restore biological functions together with the right mechanical properties are particularly needed for the regeneration of osteochondral lesions. For this purpose, the ideal scaffold should possess the right properties with respect to degradation, cell binding, cellular uptake, non-immunogenicity, mechanical strength, and flexibility. In addition, it should be easy to handle and serve as a template for chondrocyte and bone cells guiding both cartilage and bone formation. The aim of the present study was to estimate the chondrogenic and osteogenic capability of bone marrow concentrated derived cells seeded onto a novel nano-composite biomimetic material. These properties have been evaluated by means of histological, immunohistochemical and electron microscopy analyses. The data obtained demonstrated that freshly harvested cells obtained from bone marrow were able, once seeded onto the biomaterial, to differentiate either down the chondrogenic and osteogenic pathways as evaluated by the expression and production of specific matrix molecules. These findings support the use, for the repair of osteochondral lesions, of this new nano-composite biomimetic material together with bone marrow derived cells in a "one step" transplantation procedure.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
@article{%a1:%Y_390,
title = {Osteogenic differentiation of human MSCs: Specific occupancy of the mitochondrial DNA by NFATc1 transcription factor.},
author = {Lambertini E and Penolazzi L and Morganti C and Lisignoli G and Zini N and Angelozzi M and Bonora M and Ferroni L and Pinton P and Zavan B and Piva R},
url = {https://www.sciencedirect.com/science/article/pii/S1357272515001120?via%3Dihub},
doi = {10.1016/j.biocel.2015.04.011},
year = {2015},
date = {2015-07-29},
journal = {International Journal of Biochemistry & Cell Biology},
volume = {64},
pages = {212-219},
abstract = {A substantial body of evidence indicates that mitochondrial morphology and function change during osteogenic differentiation. However, molecular mechanisms linking mitochondrial dynamics with the regulation of osteoblast functions are poorly understood. Amongst the molecules that influence the decision of human mesenchymal stem cells (hMSCs) to become osteoblasts are Slug and NFATc1 transcription factors (TFs). These molecules also interfere with different mitochondria-dependent pathways in response to a variety of cellular demands. The present study investigated the recruitment of Slug and NFATc1 at the D-loop regulatory region of mitochondrial DNA (mtDNA) in osteogenic differentiated hMSCs with the aim of exploring whether Slug and NFATc1 also act as mitoTFs in the mitochondrial pool of nuclear TFs. The results demonstrate that NFATc1, but not Slug, is localized in the mitochondria. Using chromatin immunoprecipitation assay, we found that NFATc1 is recruited at mtDNA, but this occurs only when the calcification process is at its highest in osteo-induced MSC and the maximum level of differentiation is reached. Occupancy of the mtDNA by NFATc1 is associated with a decreased expression of crucial mitochondrial genes such as Cytochrome B and NADH dehydrogenase 1. This suggests that NFATc1 acts as a negative regulator of mtDNA transcription during the calcification process and interruption of aerobic energy demand. The finding of NFATc1 participation in osteogenic differentiation through its direct involvement in the regulatory machinery of mitochondria suggests a new role for this TF and adds information on communication between mitochondrial and nuclear genomes. Copyright © 2015 Elsevier Ltd. All rights reserved.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}