396 results on '"Papait A"'
Search Results
2. Exploring perinatal mesenchymal stromal cells as a potential therapeutic strategy for rheumatoid arthritis
- Author
-
Stefano Alivernini, Alice Masserdotti, Marta Magatti, Anna Cargnoni, Andrea Papait, Antonietta R. Silini, Jacopo Romoli, Sara Ficai, Clara Di Mario, Elisa Gremese, Barbara Tolusso, and Ornella Parolini
- Subjects
Rheumatoid arthritis ,Mesenchymal stromal cells ,Autoimmune disease ,Perinatal cells ,Immune cells ,Fibroblast-like synoviocytes ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by inflammation in the synovial tissue, driven by aberrant activation of both the innate and adaptive immune systems, which can lead to irreversible disability. Despite the increasing therapeutic approaches for RA, only a low percentage of patients achieve sustained disease remission, and the persistence of immune dysregulation is likely responsible for disease recurrence once remission is attained. Cell therapy is an attractive, wide-spectrum strategy to modulate inflammation, and mesenchymal stromal cells (MSC) derived from perinatal tissues provide valuable tools for their use in regenerative medicine, mainly due to their immunomodulatory properties. Several in vitro studies have shown that perinatal MSC modulate the proliferation, maturation, and cytokine secretion profile of both innate and adaptive immune cells. Moreover, different beneficial effects have also been described when perinatal MSC were used to treat animal models of diseases associated with inflammatory conditions and degenerative processes. Specifically, in experimental models of RA, treatment with perinatal MSC resulted in a strong reduction of articular damage, which was associated with the modulation of both inflammation and activation of stromal resident cells in the synovial tissue. Here, we present in vitro and in vivo evidence supporting the use of perinatal MSC in RA. We also highlight the promising results from the few published clinical trials, which demonstrate the safety of perinatal MSC.
- Published
- 2025
- Full Text
- View/download PDF
3. Proteomic analysis of the human amniotic mesenchymal stromal cell secretome by integrated approaches via filter-aided sample preparation
- Author
-
Muntiu, Alexandra, Papait, Andrea, Vincenzoni, Federica, Rossetti, Diana Valeria, Romele, Pietro, Cargnoni, Anna, Silini, Antonietta, Parolini, Ornella, and Desiderio, Claudia
- Published
- 2025
- Full Text
- View/download PDF
4. Defining the immunological compatibility of graphene oxide-loaded PLGA scaffolds for biomedical applications
- Author
-
Papait, Andrea, Perini, Giordano, Palmieri, Valentina, Cargnoni, Anna, Vertua, Elsa, Pasotti, Anna, Rosa, Enrico, De Spirito, Marco, Silini, Antonietta Rosa, Papi, Massimiliano, and Parolini, Ornella
- Published
- 2024
- Full Text
- View/download PDF
5. Unveiling the human fetal-maternal interface during the first trimester: biophysical knowledge and gaps
- Author
-
Alice Masserdotti, Michael Gasik, Regina Grillari-Voglauer, Johannes Grillari, Anna Cargnoni, Paola Chiodelli, Andrea Papait, Marta Magatti, Jacopo Romoli, Sara Ficai, Lorena Di Pietro, Wanda Lattanzi, Antonietta Rosa Silini, and Ornella Parolini
- Subjects
placenta ,fetal-maternal interactions ,first trimester ,fetal membranes ,metabolite exchanges ,Biology (General) ,QH301-705.5 - Abstract
The intricate interplay between the developing placenta and fetal-maternal interactions is critical for pregnancy outcomes. Despite advancements, gaps persist in understanding biomechanics, transport processes, and blood circulation parameters, all of which are crucial for safe pregnancies. Moreover, the complexity of fetal-maternal interactions led to conflicting data and methodological variations. This review presents a comprehensive overview of current knowledge on fetal-maternal interface structures, with a particular focus on the first trimester. More in detail, the embryological development, structural characteristics, and physiological functions of placental chorionic plate and villi, fetal membranes and umbilical cord are discussed. Furthermore, a description of the main structures and features of maternal and fetal fluid dynamic exchanges is provided. However, ethical constraints and technological limitations pose still challenges to studying early placental development directly, which calls for sophisticated in vitro, microfluidic organotypic models for advancing our understanding. For this, knowledge about key in vivo parameters are necessary for their design. In this scenario, the integration of data from later gestational stages and mathematical/computational simulations have proven to be useful tools. Notwithstanding, further research into cellular and molecular mechanisms at the fetal-maternal interface is essential for enhancing prenatal care and improving maternal and fetal health outcomes.
- Published
- 2024
- Full Text
- View/download PDF
6. Native characterization and QC profiling of human amniotic mesenchymal stromal cell vesicular fractions for secretome-based therapy
- Author
-
Marassi, Valentina, La Rocca, Giampiero, Placci, Anna, Muntiu, Alexandra, Vincenzoni, Federica, Vitali, Alberto, Desiderio, Claudia, Maraldi, Tullia, Beretti, Francesca, Russo, Eleonora, Miceli, Vitale, Conaldi, Pier Giulio, Papait, Andrea, Romele, Pietro, Cargnoni, Anna, Silini, Antonietta Rosa, Alviano, Francesco, Parolini, Ornella, Giordani, Stefano, Zattoni, Andrea, Reschiglian, Pierluigi, and Roda, Barbara
- Published
- 2024
- Full Text
- View/download PDF
7. Slow and steady wins the race: Fractionated near-infrared treatment empowered by graphene-enhanced 3D scaffolds for precision oncology
- Author
-
Perini, Giordano, Palmieri, Valentina, Papait, Andrea, Augello, Alberto, Fioretti, Daniela, Iurescia, Sandra, Rinaldi, Monica, Vertua, Elsa, Silini, Antonietta, Torelli, Riccardo, Carlino, Angela, Musarra, Teresa, Sanguinetti, Maurizio, Parolini, Ornella, De Spirito, Marco, and Papi, Massimiliano
- Published
- 2024
- Full Text
- View/download PDF
8. Disclosing the molecular profile of the human amniotic mesenchymal stromal cell secretome by filter-aided sample preparation proteomic characterization
- Author
-
Alexandra Muntiu, Andrea Papait, Federica Vincenzoni, Alberto Vitali, Wanda Lattanzi, Pietro Romele, Anna Cargnoni, Antonietta Silini, Ornella Parolini, and Claudia Desiderio
- Subjects
Proteomics ,Secretome ,Human amniotic mesenchymal stromal cells ,Filter-aided sample preparation, Immunomodulation, Regenerative medicine ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background The secretome of mesenchymal stromal cells isolated from the amniotic membrane (hAMSCs) has been extensively studied for its in vitro immunomodulatory activity as well as for the treatment of several preclinical models of immune-related disorders. The bioactive molecules within the hAMSCs secretome are capable of modulating the immune response and thus contribute to stimulating regenerative processes. At present, only a few studies have attempted to define the composition of the secretome, and several approaches, including multi-omics, are underway in an attempt to precisely define its composition and possibly identify key factors responsible for the therapeutic effect. Methods In this study, we characterized the protein composition of the hAMSCs secretome by a filter-aided sample preparation (FASP) digestion and liquid chromatography-high resolution mass spectrometry (LC–MS) approach. Data were processed for gene ontology classification and functional protein interaction analysis by bioinformatics tools. Results Proteomic analysis of the hAMSCs secretome resulted in the identification of 1521 total proteins, including 662 unique elements. A number of 157 elements, corresponding to 23.7%, were found as repeatedly characterizing the hAMSCs secretome, and those that resulted as significantly over-represented were involved in immunomodulation, hemostasis, development and remodeling of the extracellular matrix molecular pathways. Conclusions Overall, our characterization enriches the landscape of hAMSCs with new information that could enable a better understanding of the mechanisms of action underlying the therapeutic efficacy of the hAMSCs secretome while also providing a basis for its therapeutic translation.
- Published
- 2023
- Full Text
- View/download PDF
9. Muscle fibrosis as a prognostic biomarker in facioscapulohumeral muscular dystrophy: a retrospective cohort study
- Author
-
Elvira Ragozzino, Sara Bortolani, Lorena Di Pietro, Andrea Papait, Ornella Parolini, Mauro Monforte, Giorgio Tasca, and Enzo Ricci
- Subjects
Muscle fibrosis ,Skeletal muscle ,Facioscapulohumeral muscular dystrophy ,Neuromuscular disease ,Biomarker ,Muscle magnetic resonance imaging ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant epigenetic disorder with highly variable muscle involvement and disease progression. Ongoing clinical trials, aimed at counteracting muscle degeneration and disease progression in FSHD patients, increase the need for reliable biomarkers. Muscle magnetic resonance imaging (MRI) studies showed that the appearance of STIR-positive (STIR+) lesions in FSHD muscles represents an initial stage of muscle damage, preceding irreversible adipose changes. Our study aimed to investigate fibrosis, a parameter of muscle degeneration undetectable by MRI, in relation to disease activity and progression of FSHD muscles. We histologically evaluated collagen in FSHD1 patients’ (STIR+ n = 27, STIR− n = 28) and healthy volunteers’ (n = 12) muscles by picrosirius red staining. All patients (n = 55) performed muscle MRI before biopsy, 45 patients also after 1 year and 36 patients also after 2 years. Fat content (T1 signal) and oedema/inflammation (STIR signal) were evaluated at baseline and at 1- and 2-year MRI follow-up. STIR+ muscles showed significantly higher collagen compared to both STIR− (p = 0.001) and healthy muscles (p
- Published
- 2023
- Full Text
- View/download PDF
10. Systemic immune response in young and elderly patients after traumatic brain injury
- Author
-
Magatti, Marta, Pischiutta, Francesca, Ortolano, Fabrizio, Pasotti, Anna, Caruso, Enrico, Cargnoni, Anna, Papait, Andrea, Capuzzi, Franco, Zoerle, Tommaso, Carbonara, Marco, Stocchetti, Nino, Borsa, Stefano, Locatelli, Marco, Erba, Elisa, Prati, Daniele, Silini, Antonietta R, Zanier, Elisa R, and Parolini, Ornella
- Published
- 2023
- Full Text
- View/download PDF
11. Adipose mesenchymal stem cell-derived soluble factors, produced under hypoxic condition, efficiently support in vivo angiogenesis
- Author
-
Barone, Ludovica, Palano, Maria Teresa, Gallazzi, Matteo, Cucchiara, Martina, Rossi, Federica, Borgese, Marina, Raspanti, Mario, Zecca, Piero Antonio, Mortara, Lorenzo, Papait, Roberto, Bernardini, Giovanni, Valdatta, Luigi, Bruno, Antonino, and Gornati, Rosalba
- Published
- 2023
- Full Text
- View/download PDF
12. Amniotic MSC affect CD8 naive polarization toward SLEC/MPEC subsets by down-modulating IL-12Rβ1 and IL-2Rα signaling pathways
- Author
-
Papait, Andrea, Vertua, Elsa, Signoroni, Patrizia Bonassi, Cargnoni, Anna, Magatti, Marta, Stefani, Francesca Romana, Romoli, Jacopo, Silini, Antonietta Rosa, and Parolini, Ornella
- Published
- 2023
- Full Text
- View/download PDF
13. The evolution of in vitro models of lung fibrosis: promising prospects for drug discovery
- Author
-
Emanuel Kolanko, Anna Cargnoni, Andrea Papait, Antonietta Rosa Silini, Piotr Czekaj, and Ornella Parolini
- Subjects
Diseases of the respiratory system ,RC705-779 - Abstract
Lung fibrosis is a complex process, with unknown underlying mechanisms, involving various triggers, diseases and stimuli. Different cell types (epithelial cells, endothelial cells, fibroblasts and macrophages) interact dynamically through multiple signalling pathways, including biochemical/molecular and mechanical signals, such as stiffness, affecting cell function and differentiation. Idiopathic pulmonary fibrosis (IPF) is the most common fibrosing interstitial lung disease (fILD), characterised by a notably high mortality. Unfortunately, effective treatments for advanced fILD, and especially IPF and non-IPF progressive fibrosing phenotype ILD, are still lacking. The development of pharmacological therapies faces challenges due to limited knowledge of fibrosis pathogenesis and the absence of pre-clinical models accurately representing the complex features of the disease. To address these challenges, new model systems have been developed to enhance the translatability of preclinical drug testing and bridge the gap to human clinical trials. The use of two- and three-dimensional in vitro cultures derived from healthy or diseased individuals allows for a better understanding of the underlying mechanisms responsible for lung fibrosis. Additionally, microfluidics systems, which replicate the respiratory system's physiology ex vivo, offer promising opportunities for the development of effective therapies, especially for IPF.
- Published
- 2024
- Full Text
- View/download PDF
14. Contributors
- Author
-
Ahmad, Muhammad Shaheez, primary, Ahmad, Saba, additional, Aitani, Abdullah, additional, Alcántara, Andres R., additional, Ameen, Ayesha, additional, Armenia, Ilaria, additional, Asghar, Asma, additional, Asharf, Warda, additional, Bari Khan, Maham Abdul, additional, Basel, Muhammad Ehtisham, additional, Berenguer-Murcia, Ángel, additional, Bernardini, Giovanni, additional, Bilal, Muhammad, additional, Borgese, Marina, additional, Bussolari, Francesca, additional, Cassinelli, Nicolas, additional, Cipolatti, Eliane, additional, Cruz-Cruz, Angelica, additional, Cárdenas-Alcaide, María Fernanda, additional, Fernández-Lafuente, Roberto, additional, Fernández-Lucas, Jesús, additional, Fuente, J.M. de la, additional, Gallo-Cordova, Alvaro, additional, Gamberoni, Federica, additional, González-González, Reyna Berenice, additional, Gornati, Rosalba, additional, Grazu, Valeria, additional, Guimarães, José R., additional, Guisán-Seijas, J.M., additional, Hafeez, Shahzar, additional, Hameed, Sidra, additional, Hanif, Maha, additional, Hassan Gilliani, Muhammad Rehan, additional, Hussain, Asim, additional, Hussain, Dilshad, additional, Hussain, Nazim, additional, Intisar, Azeem, additional, Iqbal, Rimsha, additional, Iqbal, Hafiz M.N., additional, Ismail, Fatima, additional, Jabbar, Samahar Abdullah, additional, Javed, Kashif, additional, Javed, Sana, additional, Jesionowski, Teofil, additional, Kanwal, Faiza, additional, Khalid, Maham, additional, Khan, Faisal, additional, Kumar, Adarsh, additional, Kumar, Saroj, additional, Liaqat, Sana, additional, Lopez-Gallego, Fernando, additional, Macedo de Melo, Eduardo, additional, Mahmood, Tehreem, additional, Majeed, Saadat, additional, Maqsood, Salman, additional, Mumtaz, Ayesha, additional, Munir, Shahid, additional, Naqvi, Wajahat Zahra, additional, Naseem, Amina, additional, Naz, Maimuna, additional, Ovejero, Jesús G., additional, Papait, Roberto, additional, Parra-Saldívar, Roberto, additional, Puerto Morales, M., additional, Qaisar, Uzma, additional, Rafeeq, Hamza, additional, Rafique, Sadia, additional, Ramzan, Rehana, additional, Ratna, Sheel, additional, Rios, Nathália S., additional, Rivas-Sanchez, Andrea, additional, Rizwan, Komal, additional, Rocha-Martin, Javier, additional, Rodrigues, Rafael C., additional, Rother, Doerte, additional, Safdar, Ayesha, additional, Sanchez, Manu, additional, Serio, Simone, additional, Shafi, Sameera, additional, Shahbaz, Areej, additional, Siddique, Sara, additional, Tardioli, Paulo W., additional, Zafar, Saba, additional, Zahid, Mahnoor, additional, Zdarta, Jakub, additional, and Zeshan, Muhammad, additional
- Published
- 2023
- Full Text
- View/download PDF
15. Contributors
- Author
-
Agrawala, Paban K., primary, Alvarado, Christopher, additional, Amir, Mohammed, additional, Bernau, Ksenija, additional, Bhattacharya, Aditi, additional, Biswas, Biswajit, additional, Block, Megan, additional, Boosani, Anvitha, additional, Boosani, Chandra S., additional, Brasier, Allan R., additional, Brown, Julia A., additional, Chakraborty, Rajasree, additional, Das, Abhijit, additional, Dasari, Nikhitha, additional, DiStefano, Johanna K., additional, Dwarakanath, B.S., additional, Fukai, Tohru, additional, Gaikwad, Anil Bhanudas, additional, Gong, Fangyan, additional, Gornati, Rosalba, additional, Goswami, Ritobrata, additional, Gunasekar, Palanikumar, additional, Gupta, Noopur, additional, Hassan, Ebrahim Bani, additional, Jadhav, Gopal P., additional, Jain, Natasha Victoria, additional, Jiang, Fang, additional, Jiang, Wanlin, additional, Kreger, Jonathan, additional, Kulkarni, Yogesh A., additional, Le, Patricia N., additional, Liu, Lei, additional, Maini, Jayant, additional, Mondal, Papiya, additional, Nakai, Kenta, additional, Natarajan, Umamaheswari, additional, Olson, Michael T., additional, Pagiatakis, Christina, additional, Pandkar, Madhura R., additional, Pant, Amit M., additional, Papait, Roberto, additional, Patnaik, Srinivas, additional, Pelham, Christopher J., additional, Prakash, Anu, additional, Rathinavelu, Appu, additional, Sandbo, Nathan, additional, Sarkar, Subhajit, additional, Sharma, Ajay, additional, Sharma, Nisha, additional, Sharma, Vineeta, additional, Shiva, Niharika, additional, Shukla, Sanjeev, additional, Singh, Lakshman, additional, Singh, Sukhveer, additional, Singhal, Saurabh, additional, Srivastava, Vikas, additional, Sudhahar, Varadarajan, additional, Tiffon, Céline, additional, Ushio-Fukai, Masuko, additional, Vandenbon, Alexis, additional, Weng, Judy, additional, Woo, Jesse, additional, Wu, Xiumei, additional, Yadav, Pooja, additional, and Zeng, Melody Y., additional
- Published
- 2023
- Full Text
- View/download PDF
16. Epigenetics of aging
- Author
-
Pagiatakis, Christina, primary, Gornati, Rosalba, additional, and Papait, Roberto, additional
- Published
- 2023
- Full Text
- View/download PDF
17. A chromEM-staining protocol optimized for cardiac tissue
- Author
-
Elettra Musolino, Christina Pagiatakis, Federica Pierin, Daniele Sabatino, Giovanna Finzi, Rosalba Gornati, Giovanni Bernardini, and Roberto Papait
- Subjects
chromEM ,chromatin structure ,heterochromatin ,cardiac tissue ,aging ,Biology (General) ,QH301-705.5 - Abstract
Three-dimensional (3D) chromatin organization has a key role in defining the transcription program of cells during development. Its alteration is the cause of gene expression changes responsible for several diseases. Thus, we need new tools to study this aspect of gene expression regulation. To this end, ChromEM was recently developed: this is an electron-microscopy staining technique that selectively marks nuclear DNA without altering its structure and, thus, allows better visualization of 3D chromatin conformation. However, despite increasingly frequent application of this staining technique on cells, it has not yet been applied to visualize chromatin ultrastructure in tissues. Here, we provide a protocol to carry out ChromEM on myocardial tissue harvested from the left ventricles of C57BL/6J mice and use this in combination with transmission electron microscopy (TEM) to measure some morphological parameters of peripheral heterochromatin in cardiomyocytes. This protocol could also be used, in combination with electron tomography, to study 3D chromatin organization in cardiomyocytes in different aspects of heart pathobiology (e.g., heart development, cardiac aging, and heart failure) as well as help to set-up ChromEM in other tissues.
- Published
- 2023
- Full Text
- View/download PDF
18. Stable Housekeeping Genes in Bone Marrow, Adipose Tissue, and Amniotic Membrane-Derived Mesenchymal Stromal Cells for Orthopedic Regenerative Medicine Approaches
- Author
-
Ragni, Enrico, Piccolo, Simona, Papait, Andrea, De Luca, Paola, Taiana, Michela, Grieco, Giulio, Silini, Antonietta Rosa, Parolini, Ornella, de Girolamo, Laura, Papait, Andrea (ORCID:0000-0003-1229-9671), Parolini, Ornella (ORCID:0000-0002-5211-6430), Ragni, Enrico, Piccolo, Simona, Papait, Andrea, De Luca, Paola, Taiana, Michela, Grieco, Giulio, Silini, Antonietta Rosa, Parolini, Ornella, de Girolamo, Laura, Papait, Andrea (ORCID:0000-0003-1229-9671), and Parolini, Ornella (ORCID:0000-0002-5211-6430)
- Abstract
The therapeutic effect of mesenchymal stromal cells (MSCs) has been described for a variety of disorders, including those affecting musculoskeletal tissues. In this context, the literature reports several data about the regenerative effectiveness of MSCs derived from bone marrow, adipose tissue, and an amniotic membrane (BMSCs, ASCs, and hAMSCs, respectively), either when expanded or when acting as clinical-grade biologic pillars of products used at the point of care. To date, there is no evidence about the superiority of one source over the others from a clinical perspective. Therefore, a reliable characterization of the tissue-specific MSC types is mandatory to identify the most effective treatment, especially when tailored to the target disease. Because molecular characterization is a crucial parameter for cell definition, the need for reliable normalizers as housekeeping genes (HKGs) is essential. In this report, the stability levels of five commonly used HKGs (ACTB, EF1A, GAPDH, RPLP0, and TBP) were sifted into BMSCs, ASCs, and hAMSCs. Adult and fetal/neonatal MSCs showed opposite HKG stability rankings. Moreover, by analyzing MSC types side-by-side, comparison-specific HKGs emerged. The effect of less performant HKG normalization was also demonstrated in genes coding for factors potentially involved in and predicting MSC therapeutic activity for osteoarthritis as a model musculoskeletal disorder, where the choice of the most appropriate normalizer had a higher impact on the donors rather than cell populations when compared side-by-side. In conclusion, this work confirms HKG source-specificity for MSCs and suggests the need for cell-type specific normalizers for cell source or condition-tailored gene expression studies.
- Published
- 2024
19. The evolution of in vitro models of lung fibrosis: promising prospects for drug discovery
- Author
-
Kolanko, Emanuel, Cargnoni, Anna, Papait, Andrea, Silini, Antonietta Rosa, Czekaj, Piotr, Parolini, Ornella, Papait, Andrea (ORCID:0000-0003-1229-9671), Parolini, Ornella (ORCID:0000-0002-5211-6430), Kolanko, Emanuel, Cargnoni, Anna, Papait, Andrea, Silini, Antonietta Rosa, Czekaj, Piotr, Parolini, Ornella, Papait, Andrea (ORCID:0000-0003-1229-9671), and Parolini, Ornella (ORCID:0000-0002-5211-6430)
- Abstract
Lung fibrosis is a complex process, with unknown underlying mechanisms, involving various triggers, diseases and stimuli. Different cell types (epithelial cells, endothelial cells, fibroblasts and macrophages) interact dynamically through multiple signalling pathways, including biochemical/molecular and mechanical signals, such as stiffness, affecting cell function and differentiation. Idiopathic pulmonary fibrosis (IPF) is the most common fibrosing interstitial lung disease (fILD), characterised by a notably high mortality. Unfortunately, effective treatments for advanced fILD, and especially IPF and non-IPF progressive fibrosing phenotype ILD, are still lacking. The development of pharmacological therapies faces challenges due to limited knowledge of fibrosis pathogenesis and the absence of pre -clinical models accurately representing the complex features of the disease. To address these challenges, new model systems have been developed to enhance the translatability of preclinical drug testing and bridge the gap to human clinical trials. The use of two- and three-dimensional in vitro cultures derived from healthy or diseased individuals allows for a better understanding of the underlying mechanisms responsible for lung fibrosis. Additionally, microfluidics systems, which replicate the respiratory system's physiology ex vivo, offer promising opportunities for the development of effective therapies, especially for IPF.
- Published
- 2024
20. Extracellular vesicles of different cellular origin feature distinct biomolecular corona dynamics.
- Author
-
Musicò, Angelo, Zendrini, Andrea, Reyes, Santiago Gimenez, Mangolini, Valentina, Paolini, Lucia, Romano, Miriam, Papait, Andrea, Silini, Antonietta Rosa, Di Gianvincenzo, Paolo, Neva, Arabella, Cretich, Marina, Parolini, Ornella, Almici, Camillo, Moya, Sergio E., Radeghieri, Annalisa, and Bergese, Paolo
- Published
- 2025
- Full Text
- View/download PDF
21. Human Dental Pulp Mesenchymal Stem Cell-Derived Soluble Factors Combined with a Nanostructured Scaffold Support the Generation of a Vascular Network In Vivo
- Author
-
Ludovica Barone, Matteo Gallazzi, Federica Rossi, Roberto Papait, Mario Raspanti, Piero Antonio Zecca, Luca Buonarrivo, Barbara Bassani, Giovanni Bernardini, Antonino Bruno, and Rosalba Gornati
- Subjects
dental pulp mesenchymal stem cells ,nanostructured scaffold ,angiogenesis ,M2 polarization ,Chemistry ,QD1-999 - Abstract
Among all strategies directed at developing new tools to support re-vascularization of damaged tissues, the use of pro-angiogenic soluble factors, derived from mesenchymal stem cells (MSCs), appears a promising approach for regenerative medicine. Here, we compared the feasibility of two devices, generated by coupling soluble factors of human dental pulp mesenchymal stem cells (DPSCs), with a nanostructured scaffold, to support angiogenesis once transplanted in mice. DPSCs were obtained from impacted wisdom tooth removal, usually considered surgical waste material. After 28 days, we verified the presence of active blood vessels inside the scaffold through optical and scansion electron microscopy. The mRNA expression of surface antigens related to macrophage polarization (CD68, CD80, CD86, CD163, CD206), as well as pro-angiogenic markers (CD31, CD34, CD105, Angpt1, Angpt2, CDH5) was evaluated by real-time PCR. Our results demonstrate the capability of DPSC–scaffold and DPSC soluble factors–scaffold to support angiogenesis, similarly to adipose stem cells, whereas the absence of blood vessels was found in the scaffold grafted alone. Our results provide evidence that DPSC-conditioned medium can be proposed as a cell-free preparation able to support angiogenesis, thus, providing a relevant tool to overcome the issues and restrictions associated with the use of cells.
- Published
- 2023
- Full Text
- View/download PDF
22. Human amniotic mesenchymal stromal cells support the ex vivo expansion of cord blood hematopoietic stem cells
- Author
-
Valentina Orticelli, Andrea Papait, Elsa Vertua, Patrizia Bonassi Signoroni, Pietro Romele, Lorena Di Pietro, Marta Magatti, Luciana Teofili, Antonietta Rosa Silini, and Ornella Parolini
- Subjects
bone marrow mesenchymal stromal cells ,cord blood ,ex vivo expansion ,hematopoietic stem cells ,human amniotic mesenchymal stromal cells ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Abstract Currently, more than 30 000 allogeneic hematopoietic stem cell (HSC) transplantations have been performed for the treatment of hematological and nonhematological diseases using HSC from umbilical cord blood (CB). However, the wide utilization of CB as a source of HSC is limited by the low number of cells recovered. One strategy to expand ex vivo CB‐HSC is represented by the use of bone marrow mesenchymal stromal cells (BM‐MSCs) as a feeder to enhance HSC proliferation while maintaining HSC stemness. Indeed, BM‐MSCs have been recognized as one of the most relevant players in the HSC niche. Thus, it has been hypothesized that they can support the ex vivo expansion of HSC by mimicking the physiological microenvironment present in the hematopoietic niche. Due to the role of placenta in supporting fetal hematopoiesis, MSC derived from the amniotic membrane (hAMSC) of human term placenta could represent an interesting alternative to BM‐MSC as a feeder layer to enhance the proliferation and maintain HSC stemness. Therefore, in this study we investigated if hAMSC could support the ex vivo expansion of HSC and progenitor cells. The capacity of hAMSCs to support the ex vivo expansion of CB‐HSC was evaluated in comparison to the control condition represented by the CB‐CD34+ cells without a feeder layer. The coculture was performed at two different CD34+:MSC ratios (1:2 and 1:8) in both cell‐to‐cell contact and transwell setting. After 7 days, the cells were collected and analyzed for phenotype and functionality. Our results suggest that hAMSCs represent a valuable alternative to BM‐MSC to support: (a) the ex vivo expansion of CB‐HSC in both contact and transwell systems, (b) the colony forming unit ability, and (c) long‐term culture initiating cells ability. Overall, these findings may contribute to address the unmet need of high HSC content in CB units available for transplantation.
- Published
- 2021
- Full Text
- View/download PDF
23. CM from intact hAM: an easily obtained product with relevant implications for translation in regenerative medicine
- Author
-
Antonietta Rosa Silini, Andrea Papait, Anna Cargnoni, Elsa Vertua, Pietro Romele, Patrizia Bonassi Signoroni, Marta Magatti, Silvia De Munari, Alice Masserdotti, Anna Pasotti, Sara Rota Nodari, Giorgio Pagani, Mario Bignardi, and Ornella Parolini
- Subjects
Amniotic membrane ,Secretome ,Conditioned medium ,Lyophilization ,Immunomodulation ,Immune functional assays ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background It is now well established that factors (free or in extracellular vesicles) secreted by mesenchymal stromal cells (MSC) are important mediators of MSC regenerative actions. Herein we produced the secretome (conditioned medium, CM) from MSC isolated from the amniotic membrane (hAMSC) and CM from the intact amniotic membrane (hAM, no manipulation or enzymatic digestion) in order to potentially identify an effective, easy and less expensive secretome to produce for potential applications in regenerative medicine. Given that immunomodulation is a key mechanism of action through which hAMSC contributes to tissue regeneration, we used a comprehensive panel of in vitro immunomodulatory tests to compare the CMs. Methods Amniotic membranes were either cut into fragments or used for hAMSC isolation. CMs from hAMSC at passages 0 and 2 were collected after a standard 5-day culture while CM from hAM was collected after a 2- and 5-day culture. Immunomodulation was assessed in terms of PBMC and T-cell proliferation, T-cell subset polarization, T-regulatory cell induction, cell cytotoxicity and monocyte differentiation toward antigen-presenting cells. Furthermore, we performed a comparison between CM obtained from single donors and pooled CM. We also assessed the impact of lyophilization on the immunomodulatory properties of CM. Results We demonstrate that CM from hAM has comparable immunomodulatory properties to CM from hAMSC at passages 0 and 2. Furthermore, we demonstrate that pooled CMs have similar effects when compared to CM from single donors used separately. Finally, we demonstrate that lyophilization does not alter the in vitro immunomodulatory properties of CM from hAM and hAMSC. Conclusions The results presented herein support the possibility to produce secretome from intact hAM and open the prospect to highly improve the scalability of the GMP production process while reducing the costs and time related to the process of cell isolation and expansion. Moreover, the possibility of having a lyophilized secretome that maintains its original properties would allow for a ready-to-use product with easier handling, shipping and storage. The use of a lyophilized product will also facilitate clinicians by permitting customized reconstitution volumes and methods according to the most suitable formula required by the clinical application.
- Published
- 2021
- Full Text
- View/download PDF
24. Perinatal derivatives: How to best validate their immunomodulatory functions
- Author
-
Andrea Papait, Antonietta Rosa Silini, Maria Gazouli, Ricardo Malvicini, Maurizio Muraca, Lorraine O’Driscoll, Natalia Pacienza, Wei Seong Toh, Gustavo Yannarelli, Peter Ponsaerts, Ornella Parolini, Günther Eissner, Michela Pozzobon, Sai Kiang Lim, and Bernd Giebel
- Subjects
perinatal derivatives ,mesenchymal stromal cells ,functional assays ,mechanisms of action ,extracellular vesicles ,exosomes ,Biotechnology ,TP248.13-248.65 - Abstract
Perinatal tissues, mainly the placenta and umbilical cord, contain a variety of different somatic stem and progenitor cell types, including those of the hematopoietic system, multipotent mesenchymal stromal cells (MSCs), epithelial cells and amnion epithelial cells. Several of these perinatal derivatives (PnDs), as well as their secreted products, have been reported to exert immunomodulatory therapeutic and regenerative functions in a variety of pre-clinical disease models. Following experience with MSCs and their extracellular vesicle (EV) products, successful clinical translation of PnDs will require robust functional assays that are predictive for the relevant therapeutic potency. Using the examples of T cell and monocyte/macrophage assays, we here discuss several assay relevant parameters for assessing the immunomodulatory activities of PnDs. Furthermore, we highlight the need to correlate the in vitro assay results with preclinical or clinical outcomes in order to ensure valid predictions about the in vivo potency of therapeutic PnD cells/products in individual disease settings.
- Published
- 2022
- Full Text
- View/download PDF
25. Amniotic membrane‐mesenchymal stromal cells secreted factors and extracellular vesicle‐miRNAs: Anti‐inflammatory and regenerative features for musculoskeletal tissues
- Author
-
Enrico Ragni, Andrea Papait, Carlotta Perucca Orfei, Antonietta Rosa Silini, Alessandra Colombini, Marco Viganò, Francesca Libonati, Ornella Parolini, and Laura deGirolamo
- Subjects
amnion ,extracellular vesicles ,inflammation ,mesenchymal stem/stromal cells ,osteoarthritis ,tendinopathy ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Abstract Human amniotic membrane‐derived mesenchymal stromal cells (hAMSCs) are easily obtained in large quantities and free from ethical concerns. Promising therapeutic results for both hAMSCs and their secreted factors (secretome) were described by several in vitro and preclinical studies, often for treatment of orthopedic disorders such as osteoarthritis (OA) and tendinopathy. For clinical translation of the hAMSC secretome as cell‐free therapy, a detailed characterization of hAMSC‐secreted factors is mandatory. Herein, we tested the presence of 200 secreted factors and 754 miRNAs in extracellular vesicles (EVs). Thirty‐seven cytokines/chemokines were identified at varying abundance, some of which involved in both chemotaxis and homeostasis of inflammatory cells and in positive remodeling of extracellular matrix, often damaged in tendinopathy and OA. We also found 336 EV‐miRNAs, 51 of which accounted for more than 95% of the genetic message. A focused analysis based on miRNAs related to OA and tendinopathy showed that most abundant EV‐miRNAs are teno‐ and chondro‐protective, able to induce M2 macrophage polarization, inhibit inflammatory T cells, and promote Treg. Functional analysis on IL‐1β treated tenocytes and chondrocytes resulted in downregulation of inflammation‐associated genes. Overall, presence of key regulatory molecules and miRNAs explain the promising therapeutic results of hAMSCs and their secretome for treatment of musculoskeletal conditions and are a groundwork for similar studies in other pathologies. Furthermore, identified molecules will pave the way for future studies aimed at more sharply predicting disease‐targeted clinical efficacy, as well as setting up potency and release assays to fingerprint clinical‐grade batches of whole secretome or purified components.
- Published
- 2021
- Full Text
- View/download PDF
26. Comparison of EV-free fraction, EVs, and total secretome of amniotic mesenchymal stromal cells for their immunomodulatory potential: a translational perspective
- Author
-
Andrea Papait, Enrico Ragni, Anna Cargnoni, Elsa Vertua, Pietro Romele, Alice Masserdotti, Carlotta Perucca Orfei, Patrizia Bonassi Signoroni, Marta Magatti, Antonietta R. Silini, Laura De Girolamo, and Ornella Parolini
- Subjects
amnion ,secretome/conditioned medium ,extracellular vesicles ,mesenchymal stem/stromal cells ,immune modulation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Amniotic mesenchymal stromal cells (hAMSCs) have unique immunomodulatory properties demonstrated in vitro and in vivo in various diseases in which the dysregulated immune system plays a major role. The immunomodulatory and pro-regenerative effects of MSCs, among which hAMSCs lie in the bioactive factors they secrete and in their paracrine activity, is well known. The mix of these factors (i.e., secretome) can be either freely secreted or conveyed by extracellular vesicles (EV), thus identifying two components in the cell secretome: EV-free and EV fractions. This study aimed to discern the relative impact of the individual components on the immunomodulatory action of the hAMSC secretome in order to obtain useful information for implementing future therapeutic approaches using immunomodulatory therapies based on the MSC secretome. To this aim, we isolated EVs from the hAMSC secretome (hAMSC-CM) by ultracentrifugation and validated the vesicular product according to the International Society for Extracellular Vesicles (ISEV) criteria. EVs were re-diluted in serum-free medium to maintain the EV concentration initially present in the original CM. We compared the effects of the EV-free and EV fractions with those exerted by hAMSC-CM in toto on the activation and differentiation of immune cell subpopulations belonging to both the innate and adaptive immune systems.We observed that the EV-free fraction, similar to hAMSC-CM in toto, a) decreases the proliferation of activated peripheral blood mononuclear cells (PBMC), b) reduces the polarization of T cells toward inflammatory Th subsets, and induces the induction of regulatory T cells; c) affects monocyte polarization to antigen-presenting cells fostering the acquisition of anti-inflammatory macrophage (M2) markers; and d) reduces the activation of B lymphocytes and their maturation to plasma cells. We observed instead that all investigated EV fractions, when used in the original concentrations, failed to exert any immunomodulatory effect, even though we show that EVs are internalized by various immune cells within PBMC. These findings suggest that the active component able to induce immune regulation, tested at original concentrations, of the hAMSC secretome resides in factors not conveyed in EVs. However, EVs isolated from hAMSC could exert actions on other cell types, as reported by others.
- Published
- 2022
- Full Text
- View/download PDF
27. Divergent Transcription of the Nkx2-5 Locus Generates Two Enhancer RNAs with Opposing Functions
- Author
-
Salamon, Irene, Serio, Simone, Bianco, Simona, Pagiatakis, Christina, Crasto, Silvia, Chiariello, Andrea M., Conte, Mattia, Cattaneo, Paola, Fiorillo, Luca, Felicetta, Arianna, di Pasquale, Elisa, Kunderfranco, Paolo, Nicodemi, Mario, Papait, Roberto, and Condorelli, Gianluigi
- Published
- 2020
- Full Text
- View/download PDF
28. Extracellular vesicles derived from mesenchymal stromal cells and red blood cells feature distinct biomolecular corona dynamics
- Author
-
Musicò, Angelo, primary, Zendrini, Andrea, additional, Gimenez Reyes, Santiago, additional, Mangolini, Valentina, additional, Paolini, Lucia, additional, Romano, Miriam, additional, Papait, Andrea, additional, Silini, Antonietta Rosa, additional, Di Gianvincenzo, Paolo, additional, Neva, Arabella, additional, Cretich, Marina, additional, Parolini, Ornella, additional, Almici, Camillo, additional, Moya, Sergio, additional, Radeghieri, Annalisa, additional, and Bergese, Paolo, additional
- Published
- 2024
- Full Text
- View/download PDF
29. Epigenetics of aging and disease: a brief overview
- Author
-
Pagiatakis, Christina, Musolino, Elettra, Gornati, Rosalba, Bernardini, Giovanni, and Papait, Roberto
- Published
- 2021
- Full Text
- View/download PDF
30. This title is unavailable for guests, please login to see more information.
- Author
-
Papait, Andrea, Vertua, E., Signoroni, P. B., Cargnoni, A., Magatti, M., Stefani, F. R., Romoli, Jacopo, Silini, A. R., Parolini, Ornella, Papait A. (ORCID:0000-0003-1229-9671), Romoli J., Parolini O. (ORCID:0000-0002-5211-6430), Papait, Andrea, Vertua, E., Signoroni, P. B., Cargnoni, A., Magatti, M., Stefani, F. R., Romoli, Jacopo, Silini, A. R., Parolini, Ornella, Papait A. (ORCID:0000-0003-1229-9671), Romoli J., and Parolini O. (ORCID:0000-0002-5211-6430)
- Abstract
Mesenchymal stromal cells (MSCs) are known for their immunomodulatory activity. Here, we report that MSCs isolated from the amniotic membrane of human term placenta (hAMSCs) impact CD8 T cell fate through a multifaceted mechanism. We observed that hAMSCs are able to impact the metabolism of naive CD8 lymphocytes by downregulating the phosphorylation of mTOR and AKT, thus blocking cell differentiation. This effect is due to the ability of hAMSCs to reduce the expression of two receptors, IL-12R beta 1 and IL-2RA, resulting in reduced phosphorylation of STAT4 and STAT5. In addition, hAMSCs reduce the expression of two transcriptional factors, Tbet and Eomes, directly involved in early effector cell commitment. Our results unravel an unknown feature of MSCs, offering alternative mechanistic insights into the effects of MSCs for the treatment of diseases characterized by an altered activation of memory subsets, such as autoimmune diseases and graft versus host disease.
- Published
- 2023
31. Systemic immune response in young and elderly patients after traumatic brain injury
- Author
-
Magatti, M., Pischiutta, F., Ortolano, F., Pasotti, A., Caruso, E., Cargnoni, A., Papait, Andrea, Capuzzi, F., Zoerle, T., Carbonara, M., Stocchetti, N., Borsa, S., Locatelli, M., Erba, E., Prati, D., Silini, A. R., Zanier, E. R., Parolini, Ornella, Papait A. (ORCID:0000-0003-1229-9671), Parolini O. (ORCID:0000-0002-5211-6430), Magatti, M., Pischiutta, F., Ortolano, F., Pasotti, A., Caruso, E., Cargnoni, A., Papait, Andrea, Capuzzi, F., Zoerle, T., Carbonara, M., Stocchetti, N., Borsa, S., Locatelli, M., Erba, E., Prati, D., Silini, A. R., Zanier, E. R., Parolini, Ornella, Papait A. (ORCID:0000-0003-1229-9671), and Parolini O. (ORCID:0000-0002-5211-6430)
- Abstract
BackgroundTraumatic brain injury (TBI) is a leading cause of death and long-term disability worldwide. In addition to primary brain damage, systemic immune alterations occur, with evidence for dysregulated immune responses in aggravating TBI outcome and complications. However, immune dysfunction following TBI has been only partially understood, especially in the elderly who represent a substantial proportion of TBI patients and worst outcome. Therefore, we aimed to conduct an in-depth immunological characterization of TBI patients, by evaluating both adaptive (T and B lymphocytes) and innate (NK and monocytes) immune cells of peripheral blood mononuclear cells (PBMC) collected acutely (< 48 h) after TBI in young (18-45 yo) and elderly (> 65 yo) patients, compared to age-matched controls, and also the levels of inflammatory biomarkers.ResultsOur data show that young respond differently than elderly to TBI, highlighting the immune unfavourable status of elderly compared to young patients. While in young only CD4 T lymphocytes are activated by TBI, in elderly both CD4 and CD8 T cells are affected, and are induced to differentiate into subtypes with low cytotoxic activity, such as central memory CD4 T cells and memory precursor effector CD8 T cells. Moreover, TBI enhances the frequency of subsets that have not been previously investigated in TBI, namely the double negative CD27- IgD- and CD38-CD24- B lymphocytes, and CD56dim CD16- NK cells, both in young and elderly patients. TBI reduces the production of pro-inflammatory cytokines TNF-& alpha; and IL-6, and the expression of HLA-DM, HLA-DR, CD86/B7-2 in monocytes, suggesting a compromised ability to drive a pro-inflammatory response and to efficiently act as antigen presenting cells.ConclusionsWe described the acute immunological response induced by TBI and its relation with injury severity, which could contribute to pathologic evolution and possibly outcome. The focus on age-related immunological differences
- Published
- 2023
32. Nano-Immunomodulation: A New Strategy for Skeletal Muscle Diseases and Aging?
- Author
-
Millozzi, Francesco, Papait, Andrea, Bouché, Marina, Parolini, Ornella, Palacios, Daniela, Papait, Andrea (ORCID:0000-0003-1229-9671), Parolini, Ornella (ORCID:0000-0002-5211-6430), Palacios, Daniela (ORCID:0000-0002-2207-2369), Millozzi, Francesco, Papait, Andrea, Bouché, Marina, Parolini, Ornella, Palacios, Daniela, Papait, Andrea (ORCID:0000-0003-1229-9671), Parolini, Ornella (ORCID:0000-0002-5211-6430), and Palacios, Daniela (ORCID:0000-0002-2207-2369)
- Abstract
The skeletal muscle has a very remarkable ability to regenerate upon injury under physiological conditions; however, this regenerative capacity is strongly diminished in physio-pathological conditions, such as those present in diseased or aged muscles. Many muscular dystrophies (MDs) are characterized by aberrant inflammation due to the deregulation of both the lymphoid and myeloid cell populations and the production of pro-inflammatory cytokines. Pathological inflammation is also observed in old muscles due to a systemic change in the immune system, known as "inflammaging". Immunomodulation represents, therefore, a promising therapeutic opportunity for different skeletal muscle conditions. However, the use of immunomodulatory drugs in the clinics presents several caveats, including their low stability in vivo, the need for high doses to obtain therapeutically relevant effects, and the presence of strong side effects. Within this context, the emerging field of nanomedicine provides the powerful tools needed to control the immune response. Nano-scale materials are currently being explored as biocarriers to release immunomodulatory agents in the damaged tissues, allowing therapeutic doses with limited off-target effects. In addition, the intrinsic immunomodulatory properties of some nanomaterials offer further opportunities for intervention that still need to be systematically explored. Here we exhaustively review the state-of-the-art regarding the use of nano-sized materials to modulate the aberrant immune response that characterizes some physio-pathological muscle conditions, such as MDs or sarcopenia (the age-dependent loss of muscle mass). Based on our learnings from cancer and immune tolerance induction, we also discuss further opportunities, challenges, and limitations of the emerging field of nano-immunomodulation.
- Published
- 2023
33. Disclosing the molecular profile of the human amniotic mesenchymal stromal cell secretome by filter-aided sample preparation proteomic characterization
- Author
-
Muntiu, Alexandra, Papait, Andrea, Vincenzoni, Federica, Vitali, Alberto, Lattanzi, Wanda, Romele, Pietro, Cargnoni, Anna, Silini, Antonietta, Parolini, Ornella, Desiderio, Claudia, Papait, Andrea (ORCID:0000-0003-1229-9671), Lattanzi, Wanda (ORCID:0000-0003-3092-4936), Parolini, Ornella (ORCID:0000-0002-5211-6430), Muntiu, Alexandra, Papait, Andrea, Vincenzoni, Federica, Vitali, Alberto, Lattanzi, Wanda, Romele, Pietro, Cargnoni, Anna, Silini, Antonietta, Parolini, Ornella, Desiderio, Claudia, Papait, Andrea (ORCID:0000-0003-1229-9671), Lattanzi, Wanda (ORCID:0000-0003-3092-4936), and Parolini, Ornella (ORCID:0000-0002-5211-6430)
- Abstract
Background: The secretome of mesenchymal stromal cells isolated from the amniotic membrane (hAMSCs) has been extensively studied for its in vitro immunomodulatory activity as well as for the treatment of several preclinical models of immune-related disorders. The bioactive molecules within the hAMSCs secretome are capable of modulating the immune response and thus contribute to stimulating regenerative processes. At present, only a few studies have attempted to define the composition of the secretome, and several approaches, including multi-omics, are underway in an attempt to precisely define its composition and possibly identify key factors responsible for the therapeutic effect. Methods: In this study, we characterized the protein composition of the hAMSCs secretome by a filter-aided sample preparation (FASP) digestion and liquid chromatography-high resolution mass spectrometry (LC-MS) approach. Data were processed for gene ontology classification and functional protein interaction analysis by bioinformatics tools. Results: Proteomic analysis of the hAMSCs secretome resulted in the identification of 1521 total proteins, including 662 unique elements. A number of 157 elements, corresponding to 23.7%, were found as repeatedly characterizing the hAMSCs secretome, and those that resulted as significantly over-represented were involved in immunomodulation, hemostasis, development and remodeling of the extracellular matrix molecular pathways. Conclusions: Overall, our characterization enriches the landscape of hAMSCs with new information that could enable a better understanding of the mechanisms of action underlying the therapeutic efficacy of the hAMSCs secretome while also providing a basis for its therapeutic translation.
- Published
- 2023
34. Muscle fibrosis as a prognostic biomarker in facioscapulohumeral muscular dystrophy: a retrospective cohort study
- Author
-
Ragozzino, Elvira, Bortolani, S., Di Pietro, Lorena, Papait, Andrea, Parolini, Ornella, Monforte, Mauro, Tasca, Giorgio, Ricci, Enzo, Ragozzino E., Di Pietro L. (ORCID:0000-0001-5723-2169), Papait A. (ORCID:0000-0003-1229-9671), Parolini O. (ORCID:0000-0002-5211-6430), Monforte M., Tasca G., Ricci E. (ORCID:0000-0003-3092-3597), Ragozzino, Elvira, Bortolani, S., Di Pietro, Lorena, Papait, Andrea, Parolini, Ornella, Monforte, Mauro, Tasca, Giorgio, Ricci, Enzo, Ragozzino E., Di Pietro L. (ORCID:0000-0001-5723-2169), Papait A. (ORCID:0000-0003-1229-9671), Parolini O. (ORCID:0000-0002-5211-6430), Monforte M., Tasca G., and Ricci E. (ORCID:0000-0003-3092-3597)
- Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant epigenetic disorder with highly variable muscle involvement and disease progression. Ongoing clinical trials, aimed at counteracting muscle degeneration and disease progression in FSHD patients, increase the need for reliable biomarkers. Muscle magnetic resonance imaging (MRI) studies showed that the appearance of STIR-positive (STIR+) lesions in FSHD muscles represents an initial stage of muscle damage, preceding irreversible adipose changes. Our study aimed to investigate fibrosis, a parameter of muscle degeneration undetectable by MRI, in relation to disease activity and progression of FSHD muscles. We histologically evaluated collagen in FSHD1 patients' (STIR+ n = 27, STIR- n = 28) and healthy volunteers' (n = 12) muscles by picrosirius red staining. All patients (n = 55) performed muscle MRI before biopsy, 45 patients also after 1 year and 36 patients also after 2 years. Fat content (T1 signal) and oedema/inflammation (STIR signal) were evaluated at baseline and at 1- and 2-year MRI follow-up. STIR+ muscles showed significantly higher collagen compared to both STIR- (p = 0.001) and healthy muscles (p < 0.0001). STIR- muscles showed a higher collagen content compared to healthy muscles (p = 0.0194). FSHD muscles with a worsening in fatty infiltration during 1- (P = 0.007) and 2-year (P < 0.0001) MRI follow-up showed a collagen content of 3.6- and 3.7-fold higher compared to FSHD muscles with no sign of progression. Moreover, the fibrosis was significantly higher in STIR+ muscles who showed a worsening in fatty infiltration in a timeframe of 2 years compared to both STIR- (P = 0.0006) and STIR+ muscles with no sign of progression (P = 0.02). Fibrosis is a sign of muscle degeneration undetectable at MRI never deeply investigated in FSHD patients. Our data show that 23/27 of STIR+ and 12/28 STIR- muscles have a higher amount of collagen deposition compared to healthy muscles. Fibrosis is h
- Published
- 2023
35. Stable Housekeeping Genes in Bone Marrow, Adipose Tissue, and Amniotic Membrane-Derived Mesenchymal Stromal Cells for Orthopedic Regenerative Medicine Approaches
- Author
-
Ragni, Enrico, primary, Piccolo, Simona, additional, Papait, Andrea, additional, De Luca, Paola, additional, Taiana, Michela, additional, Grieco, Giulio, additional, Silini, Antonietta Rosa, additional, Parolini, Ornella, additional, and de Girolamo, Laura, additional
- Published
- 2024
- Full Text
- View/download PDF
36. The evolution ofin vitromodels of lung fibrosis: promising prospects for drug discovery
- Author
-
Kolanko, Emanuel, primary, Cargnoni, Anna, additional, Papait, Andrea, additional, Silini, Antonietta Rosa, additional, Czekaj, Piotr, additional, and Parolini, Ornella, additional
- Published
- 2024
- Full Text
- View/download PDF
37. Unveiling the human fetal-maternal interface during the first trimester: biophysical knowledge and gaps.
- Author
-
Masserdotti, Alice, Gasik, Michael, Grillari-Voglauer, Regina, Grillari, Johannes, Cargnoni, Anna, Chiodelli, Paola, Papait, Andrea, Magatti, Marta, Romoli, Jacopo, Ficai, Sara, Di Pietro, Lorena, Lattanzi, Wanda, Silini, Antonietta Rosa, and Parolini, Ornella
- Subjects
AMNIOTIC liquid ,EMBRYOLOGY ,CHORIONIC villi ,UMBILICAL cord ,FETAL membranes - Abstract
The intricate interplay between the developing placenta and fetal-maternal interactions is critical for pregnancy outcomes. Despite advancements, gaps persist in understanding biomechanics, transport processes, and blood circulation parameters, all of which are crucial for safe pregnancies. Moreover, the complexity of fetal-maternal interactions led to conflicting data and methodological variations. This review presents a comprehensive overview of current knowledge on fetal-maternal interface structures, with a particular focus on the first trimester. More in detail, the embryological development, structural characteristics, and physiological functions of placental chorionic plate and villi, fetal membranes and umbilical cord are discussed. Furthermore, a description of the main structures and features of maternal and fetal fluid dynamic exchanges is provided. However, ethical constraints and technological limitations pose still challenges to studying early placental development directly, which calls for sophisticated in vitro, microfluidic organotypic models for advancing our understanding. For this, knowledge about key in vivo parameters are necessary for their design. In this scenario, the integration of data from later gestational stages and mathematical/computational simulations have proven to be useful tools. Notwithstanding, further research into cellular and molecular mechanisms at the fetal-maternal interface is essential for enhancing prenatal care and improving maternal and fetal health outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. ANÁLISE DO DESEMPENHO TÉRMICO DE CONTAINERS COM E SEM TRATAMENTO
- Author
-
Souza, Paula Cristina de, primary, Halmeman, Maria Cristina Rodrigues, additional, Zavatin, Daiane Cristina Pereira, additional, and Nascimento, Glacielle A. Papait do, additional
- Published
- 2021
- Full Text
- View/download PDF
39. Genome-wide analysis of histone marks identifying an epigenetic signature of promoters and enhancers underlying cardiac hypertrophy
- Author
-
Papait, Roberto, Cattaneo, Paola, Kunderfranco, Paolo, Greco, Carolina, Carullo, Pierluigi, Guffanti, Alessandro, Viganò, Valentina, Stirparo, Giuliano Giuseppe, Latronico, Michael VG, Hasenfuss, Gerd, Chen, Ju, and Condorelli, Gianluigi
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Cardiovascular ,Human Genome ,Heart Disease ,Biotechnology ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Aetiology ,Underpinning research ,Acetylation ,Animals ,Cardiomegaly ,Enhancer Elements ,Genetic ,Epigenesis ,Genetic ,Gene Expression Regulation ,Developmental ,Histones ,Methylation ,Mice ,Promoter Regions ,Genetic ,Transcription Factors ,epigenetic regulation ,histone acetylation ,histone methylation - Abstract
Cardiac hypertrophy, initially an adaptive response of the myocardium to stress, can progress to heart failure. The epigenetic signature underlying this phenomenon is poorly understood. Here, we report on the genome-wide distribution of seven histone modifications in adult mouse cardiomyocytes subjected to a prohypertrophy stimulus in vivo. We found a set of promoters with an epigenetic pattern that distinguishes specific functional classes of genes regulated in hypertrophy and identified 9,207 candidate active enhancers whose activity was modulated. We also analyzed the transcriptional network within which these genetic elements act to orchestrate hypertrophy gene expression, finding a role for myocyte enhancer factor (MEF)2C and MEF2A in regulating enhancers. We propose that the epigenetic landscape is a key determinant of gene expression reprogramming in cardiac hypertrophy and provide a basis for understanding the role of chromatin in regulating this phenomenon.
- Published
- 2013
40. Extracellular Vesicles From Perinatal Cells for Anti-inflammatory Therapy
- Author
-
Anna Cargnoni, Andrea Papait, Alice Masserdotti, Anna Pasotti, Francesca Romana Stefani, Antonietta Rosa Silini, and Ornella Parolini
- Subjects
perinatal derivatives ,secretome ,extracellular vesicles ,immunomodulation ,tissue regeneration ,Biotechnology ,TP248.13-248.65 - Abstract
Perinatal cells, including cells from placenta, fetal annexes (amniotic and chorionic membranes), umbilical cord, and amniotic fluid display intrinsic immunological properties which very likely contribute to the development and growth of a semiallogeneic fetus during pregnancy. Many studies have shown that perinatal cells can inhibit the activation and modulate the functions of various inflammatory cells of the innate and adaptive immune systems, including macrophages, neutrophils, natural killer cells, dendritic cells, and T and B lymphocytes. These immunological properties, along with their easy availability and lack of ethical concerns, make perinatal cells very useful/promising in regenerative medicine. In recent years, extracellular vesicles (EVs) have gained great interest as a new therapeutic tool in regenerative medicine being a cell-free product potentially capable, thanks to the growth factors, miRNA and other bioactive molecules they convey, of modulating the inflammatory microenvironment thus favoring tissue regeneration. The immunomodulatory actions of perinatal cells have been suggested to be mediated by still not fully identified factors (secretoma) secreted either as soluble proteins/cytokines or entrapped in EVs. In this review, we will discuss how perinatal derived EVs may contribute toward the modulation of the immune response in various inflammatory pathologies (acute and chronic) by directly targeting different elements of the inflammatory microenvironment, ultimately leading to the repair and regeneration of damaged tissues.
- Published
- 2021
- Full Text
- View/download PDF
41. Perinatal Cells: A Promising COVID-19 Therapy?
- Author
-
Andrea Papait, Anna Cargnoni, Michal Sheleg, Antonietta R. Silini, Gilad Kunis, Racheli Ofir, and Ornella Parolini
- Subjects
coronavirus-induced disease 2019 ,severe acute respiratory distress syndrome coronavirus-2 ,mesenchymal stromal cells ,PLacental eXpanded ,perinatal ,Biotechnology ,TP248.13-248.65 - Abstract
The COVID-19 pandemic has become a priority in the health systems of all nations worldwide. In fact, there are currently no specific drugs or preventive treatments such as vaccines. The numerous therapies available today aim to counteract the symptoms caused by the viral infection that in some subjects can evolve causing acute respiratory distress syndromes (ARDS) with consequent admission to intensive care unit. The exacerbated response of the immune system, through cytokine storm, causes extensive damage to the lung tissue, with the formation of edema, fibrotic tissues and susceptibility to opportunistic infections. The inflammatory picture is also aggravated by disseminated intravascular coagulation which worsens the damage not only to the respiratory system, but also to other organs. In this context, perinatal cells represent a valid strategy thanks to their strong immunomodulatory potential, their safety profile, the ability to reduce fibrosis and stimulate reparative processes. Furthermore, perinatal cells exert antibacterial and antiviral actions. This review therefore provides an overview of the characteristics of perinatal cells with a particular focus on the beneficial effects that they could have in patients with COVID-19, and more specifically for their potential use in the treatment of ARDS and sepsis.
- Published
- 2021
- Full Text
- View/download PDF
42. Perinatal Derivatives: Where Do We Stand? A Roadmap of the Human Placenta and Consensus for Tissue and Cell Nomenclature
- Author
-
Antonietta Rosa Silini, Roberta Di Pietro, Ingrid Lang-Olip, Francesco Alviano, Asmita Banerjee, Mariangela Basile, Veronika Borutinskaite, Günther Eissner, Alexandra Gellhaus, Bernd Giebel, Yong-Can Huang, Aleksandar Janev, Mateja Erdani Kreft, Nadja Kupper, Ana Clara Abadía-Molina, Enrique G. Olivares, Assunta Pandolfi, Andrea Papait, Michela Pozzobon, Carmen Ruiz-Ruiz, Olga Soritau, Sergiu Susman, Dariusz Szukiewicz, Adelheid Weidinger, Susanne Wolbank, Berthold Huppertz, and Ornella Parolini
- Subjects
perinatal ,derivatives ,tissues ,placenta ,fetal annexes ,cells ,Biotechnology ,TP248.13-248.65 - Abstract
Progress in the understanding of the biology of perinatal tissues has contributed to the breakthrough revelation of the therapeutic effects of perinatal derivatives (PnD), namely birth-associated tissues, cells, and secreted factors. The significant knowledge acquired in the past two decades, along with the increasing interest in perinatal derivatives, fuels an urgent need for the precise identification of PnD and the establishment of updated consensus criteria policies for their characterization. The aim of this review is not to go into detail on preclinical or clinical trials, but rather we address specific issues that are relevant for the definition/characterization of perinatal cells, starting from an understanding of the development of the human placenta, its structure, and the different cell populations that can be isolated from the different perinatal tissues. We describe where the cells are located within the placenta and their cell morphology and phenotype. We also propose nomenclature for the cell populations and derivatives discussed herein. This review is a joint effort from the COST SPRINT Action (CA17116), which broadly aims at approaching consensus for different aspects of PnD research, such as providing inputs for future standards for the processing and in vitro characterization and clinical application of PnD.
- Published
- 2020
- Full Text
- View/download PDF
43. The Multifaceted Roles of MSCs in the Tumor Microenvironment: Interactions With Immune Cells and Exploitation for Therapy
- Author
-
Andrea Papait, Francesca Romana Stefani, Anna Cargnoni, Marta Magatti, Ornella Parolini, and Antonietta Rosa Silini
- Subjects
perinatal ,placenta ,mesenchymal stromal cells ,cancer ,tumor microenvironment ,inflammation ,Biology (General) ,QH301-705.5 - Abstract
The tumor microenvironment (TME) plays a critical role in tumorigenesis and is composed of different cellular components, including immune cells and mesenchymal stromal cells (MSCs). In this review, we will discuss MSCs in the TME setting and more specifically their interactions with immune cells and how they can both inhibit (immunosurveillance) and favor (immunoediting) tumor growth. We will also discuss how MSCs are used as a therapeutic strategy in cancer. Due to their unique immunomodulatory properties, MSCs isolated from perinatal tissues are intensely explored as therapeutic interventions in various inflammatory-based disorders with promising results. However, their therapeutic applications in cancer remain for the most part controversial and, importantly, the interactions between administered perinatal MSC and immune cells in the TME remain to be clearly defined.
- Published
- 2020
- Full Text
- View/download PDF
44. Targeting of the A2A adenosine receptor counteracts immunosuppression in vivo in a mouse model of chronic lymphocytic leukemia
- Author
-
Francesca Arruga, Sara Serra, Nicoletta Vitale, Giulia Guerra, Andrea Papait, Benjamin Baffour Gyau, Francesco Tito, Dimitar Efremov, Tiziana Vaisitti, and Silvia Deaglio
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Tumor immunosuppression is a major cause for treatment failure and disease relapse, both in solid tumors and leukemia. Local hypoxia is among the conditions that cause immunosuppression, acting at least in part through the upregulation of extracellular adenosine levels, which potently suppress T cell responses and skew macrophages towards an M2 phenotype. Hence, there is intense investigation to identify drugs that target this axis. By using the TCL1 adoptive transfer CLL mouse model, we show that adenosine production and signaling are upregulated in the hypoxic lymphoid niches, where intense colonization of leukemic cells occurs. This leads to a progressive remodeling of the immune system towards tolerance, with expansion of T regulatory cells (Tregs), loss of CD8+ T cell cytotoxicity and differentiation of murine macrophages towards the patrolling (M2-like) subset. In vivo administration of SCH58261, an inhibitor the A2A adenosine receptor, re-awakens T cell responses, while limiting Tregs expansion, and re-polarizes monocytes towards the inflammatory (M1-like) phenotype. These results show for the first time the in vivo contribution of adenosine signaling to immune tolerance in CLL, and the translational implication of drugs interrupting this pathway. Although the effects of SCH58261 on leukemic cells are limited, interfering with adenosine signaling may represent an appealing strategy for combination-based therapeutic approaches.
- Published
- 2020
- Full Text
- View/download PDF
45. Human Adipose-Derived Stem Cell-Conditioned Medium Promotes Vascularization of Nanostructured Scaffold Transplanted into Nude Mice
- Author
-
Ludovica Barone, Federica Rossi, Luigi Valdatta, Mario Cherubino, Roberto Papait, Giorgio Binelli, Nicla Romano, Giovanni Bernardini, and Rosalba Gornati
- Subjects
in vivo experiment ,regenerative medicine ,tissue engineering ,cell culture ,angiogenesis ,cell-free therapy ,Chemistry ,QD1-999 - Abstract
Several studies have been conducted on the interaction between three-dimensional scaffolds and mesenchymal stem cells for the regeneration of damaged tissues. Considering that stem cells do not survive for sufficient time to directly sustain tissue regeneration, it is essential to develop cell-free systems to be applied in regenerative medicine. In this work, by in vivo experiments, we established that a collagen-nanostructured scaffold, loaded with a culture medium conditioned with mesenchymal stem cells derived from adipose tissue (hASC-CM), exerts a synergic positive effect on angiogenesis, fundamental in tissue regeneration. To this aim, we engrafted athymic BALB-C nude mice with four different combinations: scaffold alone; scaffold with hASCs; scaffold with hASC crude protein extract; scaffold with hASC-CM. After their removal, we verified the presence of blood vessels by optical microscopy and confirmed the vascularization evaluating, by real-time PCR, several vascular growth factors: CD31, CD34, CD105, ANGPT1, ANGPT2, and CDH5. Our results showed that blood vessels were absent in the scaffold grafted alone, while all the other systems appeared vascularized, a finding supported by the over-expression of CD31 and CDH5 mRNA. In conclusion, our data sustain the capability of hASC-CM to be used as a therapeutic cell-free approach for damaged tissue regeneration.
- Published
- 2022
- Full Text
- View/download PDF
46. Disclosing the molecular profile of the human amniotic mesenchymal stromal cell secretome by filter-aided sample preparation proteomic characterization
- Author
-
Muntiu, Alexandra, primary, Papait, Andrea, additional, Vincenzoni, Federica, additional, Vitali, Alberto, additional, Lattanzi, Wanda, additional, Romele, Pietro, additional, Cargnoni, Anna, additional, Silini, Antonietta, additional, Parolini, Ornella, additional, and Desiderio, Claudia, additional
- Published
- 2023
- Full Text
- View/download PDF
47. Muscle fibrosis as a prognostic biomarker in facioscapulohumeral muscular dystrophy: a retrospective cohort study
- Author
-
Ragozzino, Elvira, primary, Bortolani, Sara, additional, Di Pietro, Lorena, additional, Papait, Andrea, additional, Parolini, Ornella, additional, Monforte, Mauro, additional, Tasca, Giorgio, additional, and Ricci, Enzo, additional
- Published
- 2023
- Full Text
- View/download PDF
48. Cardiac Aging Is Promoted by Pseudohypoxia Increasing p300-Induced Glycolysis
- Author
-
Serio, Simone, primary, Pagiatakis, Christina, additional, Musolino, Elettra, additional, Felicetta, Arianna, additional, Carullo, Pierluigi, additional, Laura Frances, Javier, additional, Papa, Laura, additional, Rozzi, Giacomo, additional, Salvarani, Nicolò, additional, Miragoli, Michele, additional, Gornati, Rosalba, additional, Bernardini, Giovanni, additional, Condorelli, Gianluigi, additional, and Papait, Roberto, additional
- Published
- 2023
- Full Text
- View/download PDF
49. Human Dental Pulp Mesenchymal Stem Cell-Derived Soluble Factors Combined with a Nanostructured Scaffold Support the Generation of a Vascular Network In Vivo
- Author
-
Barone, Ludovica, primary, Gallazzi, Matteo, additional, Rossi, Federica, additional, Papait, Roberto, additional, Raspanti, Mario, additional, Zecca, Piero Antonio, additional, Buonarrivo, Luca, additional, Bassani, Barbara, additional, Bernardini, Giovanni, additional, Bruno, Antonino, additional, and Gornati, Rosalba, additional
- Published
- 2023
- Full Text
- View/download PDF
50. Np95 Is Regulated by E1A during Mitotic Reactivation of Terminally Differentiated Cells and Is Essential for S Phase Entry
- Author
-
Bonapace, Ian Marc, Latella, Lucia, Papait, Roberto, Nicassio, Francesco, Sacco, Alessandra, Muto, Masahiro, Crescenzi, Marco, and Di Fiore, Pier Paolo
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.