163 results on '"Ornella, Parolini"'
Search Results
2. Mapping of the Human Amniotic Membrane: Detection of Microvesicles Secreted by Amniotic Epithelial Cells
- Author
-
Mariangela Basile, Lucia Centurione, Francesca Passaretta, Gianmarco Stati, Olga Soritau, Sergiu Susman, Florelle Gindraux, Antonietta Silini, Ornella Parolini, and Roberta Di Pietro
- Subjects
Medicine - Abstract
The potential clinical applications of human amniotic membrane (hAM) and human amniotic epithelial cells (hAECs) in the field of regenerative medicine have been known in literature since long. However, it has yet to be elucidated whether hAM contains different anatomical regions with different plasticity and differentiation potential. Recently, for the first time, we highlighted many differences in terms of morphology, marker expression, and differentiation capabilities among four distinct anatomical regions of hAM, demonstrating peculiar functional features in hAEC populations. The aim of this study was to investigate in situ the ultrastructure of the four different regions of hAM by means of transmission electron microscopy (TEM) to deeply understand their peculiar characteristics and to investigate the presence and localization of secretory products because to our knowledge, there are no similar studies in the literature. The results of this study confirm our previous observations of hAM heterogeneity and highlight for the first time that hAM can produce extracellular vesicles (EVs) in a heterogeneous manner. These findings should be considered to increase efficiency of hAM applications within a therapeutic context.
- Published
- 2023
- Full Text
- View/download PDF
3. A machine-learning parsimonious multivariable predictive model of mortality risk in patients with Covid-19
- Author
-
Rita Murri, Jacopo Lenkowicz, Carlotta Masciocchi, Chiara Iacomini, Massimo Fantoni, Andrea Damiani, Antonio Marchetti, Paolo Domenico Angelo Sergi, Giovanni Arcuri, Alfredo Cesario, Stefano Patarnello, Massimo Antonelli, Rocco Bellantone, Roberto Bernabei, Stefania Boccia, Paolo Calabresi, Andrea Cambieri, Roberto Cauda, Cesare Colosimo, Filippo Crea, Ruggero De Maria, Valerio De Stefano, Francesco Franceschi, Antonio Gasbarrini, Ornella Parolini, Luca Richeldi, Maurizio Sanguinetti, Andrea Urbani, Maurizio Zega, Giovanni Scambia, Vincenzo Valentini, and The Gemelli against Covid Group
- Subjects
Medicine ,Science - Abstract
Abstract The COVID-19 pandemic is impressively challenging the healthcare system. Several prognostic models have been validated but few of them are implemented in daily practice. The objective of the study was to validate a machine-learning risk prediction model using easy-to-obtain parameters to help to identify patients with COVID-19 who are at higher risk of death. The training cohort included all patients admitted to Fondazione Policlinico Gemelli with COVID-19 from March 5, 2020, to November 5, 2020. Afterward, the model was tested on all patients admitted to the same hospital with COVID-19 from November 6, 2020, to February 5, 2021. The primary outcome was in-hospital case-fatality risk. The out-of-sample performance of the model was estimated from the training set in terms of Area under the Receiving Operator Curve (AUROC) and classification matrix statistics by averaging the results of fivefold cross validation repeated 3-times and comparing the results with those obtained on the test set. An explanation analysis of the model, based on the SHapley Additive exPlanations (SHAP), is also presented. To assess the subsequent time evolution, the change in paO2/FiO2 (P/F) at 48 h after the baseline measurement was plotted against its baseline value. Among the 921 patients included in the training cohort, 120 died (13%). Variables selected for the model were age, platelet count, SpO2, blood urea nitrogen (BUN), hemoglobin, C-reactive protein, neutrophil count, and sodium. The results of the fivefold cross-validation repeated 3-times gave AUROC of 0.87, and statistics of the classification matrix to the Youden index as follows: sensitivity 0.840, specificity 0.774, negative predictive value 0.971. Then, the model was tested on a new population (n = 1463) in which the case-fatality rate was 22.6%. The test model showed AUROC 0.818, sensitivity 0.813, specificity 0.650, negative predictive value 0.922. Considering the first quartile of the predicted risk score (low-risk score group), the case-fatality rate was 1.6%, 17.8% in the second and third quartile (high-risk score group) and 53.5% in the fourth quartile (very high-risk score group). The three risk score groups showed good discrimination for the P/F value at admission, and a positive correlation was found for the low-risk class to P/F at 48 h after admission (adjusted R-squared = 0.48). We developed a predictive model of death for people with SARS-CoV-2 infection by including only easy-to-obtain variables (abnormal blood count, BUN, C-reactive protein, sodium and lower SpO2). It demonstrated good accuracy and high power of discrimination. The simplicity of the model makes the risk prediction applicable for patients in the Emergency Department, or during hospitalization. Although it is reasonable to assume that the model is also applicable in not-hospitalized persons, only appropriate studies can assess the accuracy of the model also for persons at home.
- Published
- 2021
- Full Text
- View/download PDF
4. Shaping modern human skull through epigenetic, transcriptional and post-transcriptional regulation of the RUNX2 master bone gene
- Author
-
Lorena Di Pietro, Marta Barba, Daniela Palacios, Federica Tiberio, Chiara Prampolini, Mirko Baranzini, Ornella Parolini, Alessandro Arcovito, and Wanda Lattanzi
- Subjects
Medicine ,Science - Abstract
Abstract RUNX2 encodes the master bone transcription factor driving skeletal development in vertebrates, and playing a specific role in craniofacial and skull morphogenesis. The anatomically modern human (AMH) features sequence changes in the RUNX2 locus compared with archaic hominins’ species. We aimed to understand how these changes may have contributed to human skull globularization occurred in recent evolution. We compared in silico AMH and archaic hominins’ genomes, and used mesenchymal stromal cells isolated from skull sutures of craniosynostosis patients for in vitro functional assays. We detected 459 and 470 nucleotide changes in noncoding regions of the AMH RUNX2 locus, compared with the Neandertal and Denisovan genomes, respectively. Three nucleotide changes in the proximal promoter were predicted to alter the binding of the zinc finger protein Znf263 and long-distance interactions with other cis-regulatory regions. By surface plasmon resonance, we selected nucleotide substitutions in the 3’UTRs able to affect miRNA binding affinity. Specifically, miR-3150a-3p and miR-6785-5p expression inversely correlated with RUNX2 expression during in vitro osteogenic differentiation. The expression of two long non-coding RNAs, AL096865.1 and RUNX2-AS1, within the same locus, was modulated during in vitro osteogenic differentiation and correlated with the expression of specific RUNX2 isoforms. Our data suggest that RUNX2 may have undergone adaptive phenotypic evolution caused by epigenetic and post-transcriptional regulatory mechanisms, which may explain the delayed suture fusion leading to the present-day globular skull shape.
- Published
- 2021
- Full Text
- View/download PDF
5. Human acellular amniotic membrane implantation for lower third nasal reconstruction: a promising therapy to promote wound healing
- Author
-
Si-Liang Xue, Kai Liu, Ornella Parolini, Yue Wang, Li Deng, and Yong-Can Huang
- Subjects
Acellular amniotic membrane ,Nasal reconstruction ,Wound healing ,Repair ,Medicine - Abstract
Abstract Background The lower third of the nose is one of the most important cosmetic units of the face, and its reconstructive techniques remain a big challenge. As an alternative approach to repair or regenerate the nasal tissue, the biomaterial-based strategy has been extensively investigated. The aim of this study is to determine the safety and efficacy of human acellular amniotic membrane (HAAM) to repair the full-thickness defects in the lower third of the nose in humans. Methods In this study, 180 patients who underwent excision of skin lesions of the lower third of the nose from 2012 to 2016 were included; of the patients, 92 received HAAM and Vaseline gauze treatments, and the other 88 patients received Vaseline gauze treatment only. The haemostasis time and the duration of operation were recorded during surgery; after surgery, the time to pain disappearance, scab formation and wound healing, and the wound healing rate were measured. Results Immediately after the HAAM implantation, a reduction of the haemostasis time and an accelerated disappearance of pain were observed. Compared with the control group, the formation and detachment of scab in patients who received the HAAM implantation were notably accelerated, postoperatively. When the diameter of the lesion exceeded 5 mm, the HAAM implantation was found to enhance the wound healing, although this enhancement was not seen when the diameter was less than 5 mm. Additionally, the HAAM implantation significantly reduced bleeding, wound infection and scar formation, postoperatively. Conclusions HAAM-assisted healing is a promising therapy for lower third nasal reconstruction leading to rapid wound healing and fewer complications and thus has considerable potential for extensive clinical application in repairing skin wounds. Trial registration ChiCTR1800017618, retrospectively registered on July 08, 2018.
- Published
- 2018
- Full Text
- View/download PDF
6. Human amniotic mesenchymal stromal cells support the ex vivo expansion of cord blood hematopoietic stem cells
- Author
-
Luciana Teofili, Pietro Romele, Patrizia Bonassi Signoroni, Andrea Papait, Marta Magatti, Ornella Parolini, Valentina Orticelli, Antonietta Silini, Lorena Di Pietro, and Elsa Vertua
- Subjects
Medicine (General) ,CD34 ,Placenta‐Derived Stem Cells ,Antigens, CD34 ,Fetal and Neonatal Stem Cells ,Biology ,R5-920 ,Pregnancy ,Bone Marrow Stem Cells ,medicine ,Humans ,Settore BIO/13 - BIOLOGIA APPLICATA ,Amnion ,Progenitor cell ,Amniotic Stem Cells ,Cells, Cultured ,Cell Proliferation ,QH573-671 ,Mesenchymal stem cell ,Hematopoietic stem cell ,Mesenchymal Stem Cells ,bone marrow mesenchymal stromal cells ,human amniotic mesenchymal stromal cells ,Cell Biology ,General Medicine ,ex vivo expansion ,Fetal Blood ,Coculture Techniques ,Cell biology ,hematopoietic stem cells ,medicine.anatomical_structure ,Hematopoietic Stem/Progenitor Cells ,Cord blood ,cord blood ,Female ,Bone marrow ,Stromal Cells ,Stem cell ,Cytology ,Ex vivo ,Developmental Biology - 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., Ex vivo expansion of cord blood hematopoietic stem cell (HSC) and progenitor cell is required to improve efficacy of transplantation in adults. Bone marrow mesenchymal stromal cells are the gold standard for HSC expansion, but they can be difficult to obtain. Amniotic membrane‐derived MSC can be a valid alternative that could help meet the demand of HSC for transplants.
- Published
- 2021
7. Immunological and Differentiation Properties of Amniotic Cells Are Retained After Immobilization in Pectin Gel
- Author
-
Antonietta R. Silini, Valentina Spoldi, Silvia De Munari, Elsa Vertua, Fabiola Munarin, Paola Petrini, Silvia Farè, and Ornella Parolini
- Subjects
Medicine - Abstract
Mesenchymal stromal cells from the human amniotic membrane (i.e., human amniotic mesenchymal stromal cells [hAMSCs]) of term placenta are increasingly attracting attention for their applications in regenerative medicine. Osteochondral defects represent a major clinical problem with lifelong chronic pain and compromised quality of life. Great promise for osteochondral regeneration is held in hydrogel-based constructs that have a flexible composition and mimic the physiological structure of cartilage. Cell loading within a hydrogel represents an advantage for regenerative purposes, but the encapsulation steps can modify cell properties. As pectin gels have also been explored as cell vehicles on 3D scaffolds, the aim of this study was to explore the possibility to include hAMSCs in pectin gel. Immobilization of hAMSCs into pectin gels could expand their application in cell-based bioengineering strategies. hAMSCs were analyzed for their viability and recovery from the pectin gel and for their ability to differentiate toward the osteogenic lineage and to maintain their immunological characteristics. When treated with a purposely designed pectin/hydroxyapatite gel biocomposite, hAMSCs retained their ability to differentiate toward the osteogenic lineage, did not induce an immune response, and retained their ability to reduce T cell proliferation. Taken together, these results suggest that hAMSCs could be used in combination to pectin gels for the study of novel osteochondral regeneration strategies.
- Published
- 2018
- Full Text
- View/download PDF
8. Mapping of the Human Placenta
- Author
-
Lucia Centurione, Francesca Passaretta, Maria Antonietta Centurione, Silvia De Munari, Elsa Vertua, Antonietta Silini, Marco Liberati, Ornella Parolini, and Roberta Di Pietro
- Subjects
Medicine - Abstract
The human placenta is an important source of stem cells that can be easily collected without ethical concerns since it is usually discarded after childbirth. In this study, we analyzed the amniotic membrane (AM) from the human placenta with the aim of mapping different regions with respect to their morpho-functional features and regenerative potential. AMs were obtained from 24 healthy women, undergoing a caesarean section, and mapped into 4 different regions according to their position in relation to the umbilical cord: the central, intermediate, peripheral, and reflected areas. We carried out a multiparametric analysis focusing our attention on amniotic epithelial cells (AECs). Our results revealed that AECs, isolated from the different areas, are a heterogeneous cell population with different pluripotency and proliferation marker expression (octamer-binding transcription factor 4 [OCT-4], tyrosine-protein kinase KIT [c-KIT], sex determining region Y-box 2 [SOX-2], α-fetoprotein, cyclic AMP response element binding [CREB] protein, and phosphorylated active form of CREB [p-CREB]), proliferative ability, and osteogenic potential. Our investigation discloses interesting findings that could be useful for increasing the efficiency of AM isolation and application for therapeutic purposes.
- Published
- 2018
- Full Text
- View/download PDF
9. Proliferation and survival of human amniotic epithelial cells during their hepatic differentiation.
- Author
-
Julieta L Maymó, Rodrigo Riedel, Antonio Pérez-Pérez, Marta Magatti, Bernardo Maskin, José Luis Dueñas, Ornella Parolini, Víctor Sánchez-Margalet, and Cecilia L Varone
- Subjects
Medicine ,Science - Abstract
Stem cells derived from placental tissues are an attractive source of cells for regenerative medicine. Amniotic epithelial cells isolated from human amnion (hAECs) have desirable and competitive characteristics that make them stand out between other stem cells. They have the ability to differentiate toward all three germ layers, they are not tumorigenic and they have immunosuppressive properties. Although liver transplantation is the best way to treat acute and chronic hepatic failure patients, there are several obstacles. Recently, stem cells have been spotlighted as alternative source of hepatocytes because of their potential for hepatogenic differentiation. In this work, we aimed to study the proliferation and survival of the hAECs during their hepatic differentiation. We have also analyzed the changes in pluripotency and hepatic markers. We differentiated amniotic cells applying a specific hepatic differentiation (HD) protocol. We determined by qRT-PCR that hAECs express significant levels of SOX-2, OCT-4 and NANOG during at least 15 days in culture and these pluripotent markers diminish during HD. SSEA-4 expression was reduced during HD, measured by immunofluorescence. Morphological characteristics became more similar to hepatic ones in differentiated cells and representative hepatic markers significantly augmented their expression, measured by qRT-PCR and Western blot. Cells achieved a differentiation efficiency of 75%. We observed that HD induced proliferation and promoted survival of hAECs, during 30 days in culture, evaluated by 3H-thymidine incorporation and MTT assay. HD also promoted changes in hAECs cell cycle. Cyclin D1 expression increased, while p21 and p53 levels were reduced. Immunofluorescence analysis showed that Ki-67 expression was upregulated during HD. Finally, ERK 1/2 phosphorylation, which is intimately linked to proliferation and cell survival, augmented during all HD process and the inhibition of this signaling pathway affected not only proliferation but also differentiation. Our results suggest that HD promotes proliferation and survival of hAECs, providing important evidence about the mechanisms governing their hepatic differentiation. We bring new knowledge concerning some of the optimal transplantation conditions for these hepatic like cells.
- Published
- 2018
- Full Text
- View/download PDF
10. The Immunomodulatory Properties of Amniotic Cells
- Author
-
Marta Magatti, Elsa Vertua, Anna Cargnoni, Antonietta Silini, and Ornella Parolini
- Subjects
Medicine - Abstract
Among the many cell types useful in developing therapeutic treatments, human amniotic cells from placenta have been proposed as valid candidates. Both human amniotic epithelial and mesenchymal stromal cells, and the conditioned medium generated from their culture, exert multiple immunosuppressive activities. Indeed, they inhibit T and B cell proliferation, suppress inflammatory properties of monocytes, macrophages, dendritic cells, neutrophils, and natural killer cells, while promoting induction of cells with regulatory functions such as regulatory T cells and anti-inflammatory M2 macrophages. These properties have laid the foundation for their use for the treatment of inflammatory-based diseases, and encouraging results have been obtained in different preclinical disease models where exacerbated inflammation is present. Moreover, an immune-privileged status of amniotic cells has been often highlighted. However, even if long-term engraftment of amniotic cells has been reported into immunocompetent animals, only few cells survive after infusion. Furthermore, amniotic cells have been shown to be able to induce immune responses in vivo and, under specific culture conditions, they can stimulate T cell proliferation in vitro. Although immunosuppressive properties are a widely recognized characteristic of amniotic cells, immunogenic and stimulatory activities appear to be less reported, sporadic events. In order to improve therapeutic outcome, the mechanisms responsible for the suppressive versus stimulatory activity need to be carefully addressed. In this review, both the immunosuppressive and immunostimulatory activity of amniotic cells will be discussed.
- Published
- 2018
- Full Text
- View/download PDF
11. Placenta-Derived Cells for Acute Brain Injury
- Author
-
Francesca Pischiutta, Eliana Sammali, Ornella Parolini, Hilary V. O. Carswell, and Elisa R. Zanier
- Subjects
Medicine - Abstract
Acute brain injury resulting from ischemic/hemorrhagic or traumatic damage is one of the leading causes of mortality and disability worldwide and is a significant burden to society. Neuroprotective options to counteract brain damage are very limited in stroke and traumatic brain injury (TBI). Given the multifaceted nature of acute brain injury and damage progression, several therapeutic targets may need to be addressed simultaneously to interfere with the evolution of the injury and improve the patient’s outcome. Stem cells are ideal candidates since they act on various mechanisms of protection and repair, improving structural and functional outcomes after experimental stroke or TBI. Stem cells isolated from placenta offer advantages due to their early embryonic origin, ease of procurement, and ethical acceptance. We analyzed the evidence for the beneficial effects of placenta-derived stem cells in acute brain injury, with the focus on experimental studies of TBI and stroke, the engineering strategies pursued to foster cell potential, and characterization of the bioactive molecules secreted by placental cells, known as their secretome, as an alternative cell-free strategy. Results from the clinical application of placenta-derived stem cells for acute brain injury and ongoing clinical trials are summarily discussed.
- Published
- 2018
- Full Text
- View/download PDF
12. Placental Cells and Derivatives
- Author
-
Antonietta R. Silini and Ornella Parolini
- Subjects
Medicine - Published
- 2018
- Full Text
- View/download PDF
13. Is Immune Modulation the Mechanism Underlying the Beneficial Effects of Amniotic Cells and Their Derivatives in Regenerative Medicine?
- Author
-
Antonietta R. Silini, Marta Magatti, Anna Cargnoni, and Ornella Parolini Ph.D.
- Subjects
Medicine - Abstract
Regenerative medicine aims to repair and regenerate damaged cells, tissues, and organs in order to restore function. Regeneration can be obtained either by cell replacement or by stimulating the body's own repair mechanisms. Importantly, a favorable environment is required before any regenerative signal can stimulate resident stem/stromal cells, and regeneration is possible only after the resolution of injury-induced inflammation. An exacerbated immune response is often present in cases of degenerative, inflammatory-based diseases. Here we discuss how amniotic membrane cells, and their derivatives, can contribute to the resolution of many diseases with altered immune response by acting on different inflammatory mediators.
- Published
- 2017
- Full Text
- View/download PDF
14. Amniotic membrane-mesenchymal stromal cells secreted factors and extracellular vesicle-miRNAs: Anti-inflammatory and regenerative features for musculoskeletal tissues
- Author
-
Laura de Girolamo, Francesca Libonati, Marco Viganò, Enrico Ragni, Alessandra Colombini, Ornella Parolini, Antonietta Rosa Silini, Carlotta Perucca Orfei, and Andrea Papait
- Subjects
0301 basic medicine ,Medicine (General) ,Chemokine ,Inflammation ,Fetal and Neonatal Stem Cells ,Extracellular matrix ,Extracellular Vesicles ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,Humans ,Regeneration ,Amnion ,mesenchymal stem/stromal cells ,Musculoskeletal System ,Secretome ,QH573-671 ,biology ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Chemotaxis ,Cell Biology ,General Medicine ,Extracellular vesicle ,In vitro ,Extracellular Matrix ,Cell biology ,MicroRNAs ,osteoarthritis ,030104 developmental biology ,inflammation ,Tendinopathy ,biology.protein ,Cytokines ,Chemokines ,medicine.symptom ,Cytology ,030217 neurology & neurosurgery ,Developmental Biology - 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., The secretome from human amniotic membrane‐derived mesenchymal stromal cells offers tremendous potential for treatment of musculoskeletal disorders. Combined analysis of soluble factors and extracellular vesicle‐miRNAs show their teno‐/chondro‐protective features, as well as ability to induce M2 macrophage polarization, inhibit inflammatory T cells, and promote Treg. The fingerprint of key regulatory molecules will define the potency of future off‐the‐shelf and cell‐free regenerative therapies.
- Published
- 2021
- Full Text
- View/download PDF
15. Antifibrotic Effects of Human Amniotic Membrane Transplantation in Established Biliary Fibrosis Induced in Rats
- Author
-
Luciana B. Sant'anna, Raduan Hage, Maria Angélica G. Cardoso, Emilia A. L. Arisawa, Maria Martin Cruz, Ornella Parolini, Anna Cargnoni, and Nilson Sant'anna
- Subjects
Medicine - Abstract
Liver fibrosis is characterized by excessive accumulation of extracellular matrix components in the liver parenchyma that distorts the normal architecture and hepatic function. Progressive fibrosis could end in the advanced stage known as cirrhosis, resulting in the need to resort to liver transplantation. Amniotic membrane (AM) has emerged as an innovative therapeutic approach for chronic liver diseases due to its anti-inflammatory, antiscarring, and wound-healing effects. We have recently shown that AM can be used as a patch on the liver surface at the same time of fibrosis induction, resulting in significantly reduced progression and severity of biliary fibrosis. Here we investigated the effects of human AM on the established rat model of liver fibrosis, induced by the bile duct ligation (BDL). We also explored the effect of AM on the expression of transforming growth factor-β1 (TGF-β1), the main profibrogenic factor in hepatic fibrosis, and the proinflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and anti-inflammatory cytokine IL-10. Two weeks after BDL, the liver was covered with a fragment of AM or left untreated. Six weeks later, the fibrosis was first assessed by the semiquantitative Knodell and the METAVIR scoring systems and, thereafter, by CellProfiler digital image analysis to quantify the area occupied by collagen deposition, ductular reactions (DRs), activated myofibroblasts, and TGF-β1. The hepatic cytokines were determined by ELISA. AM-treated rats showed a significantly lower score compared to the control BDL rats (2.5 ± 0.9 vs. 3.5 ± 0.3, respectively; p < 0.05). The collagen deposition, DRs, number of activated myofibroblasts, and TGF-β1 were all reduced to about 50% of levels observed in untreated BDL rats. These findings suggest that AM, when applied as a patch onto the liver surface, is useful for treating well-established cholestatic fibrosis, and the mechanism was partly by means of downregulating the profibrotic factor TGF-β1 and IL-6.
- Published
- 2016
- Full Text
- View/download PDF
16. Human amniotic stem cells improve hepatic microvascular dysfunction and portal hypertension in cirrhotic rats
- Author
-
Angelo Luca, Ornella Parolini, Jordi Gracia-Sancho, Juan José Lozano, Jaime Bosch, Pier G. Conaldi, Laia Abad-Jordà, Giada Pietrosi, Mariangela Pampalone, Anabel Fernández-Iglesias, Giovanni Vizzini, Héctor García-Calderó, Martí Ortega-Ribera, Pietrosi G., Fernandez-Iglesias A., Pampalone M., Ortega-Ribera M., Lozano J.J., Garcia-Caldero H., Abad-Jorda L., Conaldi P.G., Parolini O., Vizzini G., Luca A., Bosch J., and Gracia-Sancho J.
- Subjects
Liver Cirrhosis ,Pathology ,medicine.medical_specialty ,hAEC ,Cirrhosis ,placenta ,Portal venous pressure ,03 medical and health sciences ,0302 clinical medicine ,Hypertension, Portal ,Settore BIO/13 - BIOLOGIA APPLICATA ,Animals ,Humans ,Medicine ,Amnion ,610 Medicine & health ,Hepatology ,medicine.diagnostic_test ,business.industry ,Microcirculation ,Stem Cells ,chronic liver disease ,portal hypertension ,Endothelial Cells ,Amniotic stem cells ,medicine.disease ,Rats ,Liver ,030220 oncology & carcinogenesis ,Hepatic stellate cell ,Portal hypertension ,Vascular Resistance ,030211 gastroenterology & hepatology ,Stem cell ,business ,Hepatic fibrosis ,Liver function tests ,hAMSC - Abstract
BACKGROUND AND AIMS Portal hypertension is the main consequence of cirrhosis, responsible for the complications defining clinical decompensation. The only cure for decompensated cirrhosis is liver transplantation, but it is a limited resource and opens the possibility of regenerative therapy. We investigated the potential of primary human amniotic membrane-derived mesenchymal stromal (hAMSCs) and epithelial (hAECs) stem cells for the treatment of portal hypertension and decompensated cirrhosis. METHODS In vitro: Primary liver sinusoidal endothelial cells (LSECs) and hepatic stellate cells (HSCs) from cirrhotic rats (chronic CCl4 inhalation) were co-cultured with hAMSCs, hAECs or vehicle for 24 hours, and their RNA profile was analysed. In vivo: CCl4-cirrhotic rats received 4x106 hAMSCs, 4x106 hAECs, or vehicle (NaCl 0.9%) (intraperitoneal). At 2-weeks we analysed: a) portal pressure (PP) and hepatic microvascular function; b) LSECs and HSCs phenotype; c) hepatic fibrosis and inflammation. RESULTS In vitro experiments revealed sinusoidal cell phenotype amelioration when co-cultured with stem cells. Cirrhotic rats receiving stem cells, particularly hAMSCs, had significantly lower PP than vehicle-treated animals, together with improved liver microcirculatory function. This hemodynamic amelioration was associated with improvement in LSECs capillarization and HSCs de-activation, though hepatic collagen was not reduced. Rats that received amnion derived stem cells had markedly reduced hepatic inflammation and oxidative stress. Finally, liver function tests significantly improved in rats receiving hAMSCs. CONCLUSIONS This preclinical study shows that infusion of human amniotic stem cells effectively decreases PP by ameliorating liver microcirculation, suggesting that it may represent a new treatment option for advanced cirrhosis with portal hypertension.
- Published
- 2020
- Full Text
- View/download PDF
17. Human Amniotic Membrane-Derived Mesenchymal and Epithelial Cells Exert Different Effects on Monocyte-Derived Dendritic Cell Differentiation and Function
- Author
-
Marta Magatti, Maddalena Caruso, Silvia De Munari, Elsa Vertua, Debashree De, Ursula Manuelpillai, and Ornella Parolini Ph.D.
- Subjects
Medicine - Abstract
We previously demonstrated that mesenchymal cells from human amniotic membrane (hAMTCs) inhibit the generation and maturation of monocyte-derived dendritic cells (DCs) in vitro. Considering the crucial role of DCs in the immune response and that epithelial cells of the human amniotic membrane (hAECs) share some of the immunoregulatory properties of hAMTCs, we investigated whether hAECs also modulate monocyte-derived DCs. We compared hAECs with hAMTCs in a cell-to-cell contact setting and their secreted factors in modulating DC differentiation and function. First, we demonstrated that primary and expanded hAMTCs strongly inhibited the differentiation of DCs and induced a shift toward M2-like macrophages. This was observed when hAMTCs were cultured in contact (hAMTC-DC cont ) or in Transwells (hAMTC-DC tw ) with monocytes and even when medium conditioned by hAMTCs was used instead of hAMTCs. hAECs also prevented DC development, but to a lesser extent than hAMTCs. hAECs were more effective when cultured in contact with monocytes (hAEC-DC cont ) rather than in Transwells (hAEC-DC tw ). The modulatory capacity of hAECs changed during passaging unlike the hAMSCs. The ability to stimulate CD4 + and CD8 + T-cell proliferation was almost completely abolished by hAMTC-DC cont , whereas hAMTC-DC tw and hAEC-DC cont displayed only a reduced ability to stimulate CD8 + T cells. Furthermore, monocytes cocultured with hAMTCs and hAECs showed some similarities, but also differences in cytokine/chemokine secretion. Similarities were observed in the inhibition of IL-12p70 and TNF-α and the increase in IL-10 in supernatants taken from monocyte-DCs cocultured with hAMTCs and hAECs in contact and Transwell settings. The inflammatory factors IL-8, CXCL9, and MIP-1α were significantly lower in hAMTC-DC cont , hAMTC-DC tw , and hAEC-DC cont conditions. In contrast, only hAMTCs (in both contact and Transwell conditions) were able to significantly increase IL-1β and CCL2. Altogether, we demonstrated that hAMTCs and hAECs affect DC differentiation, but that hAMTCs exerted a stronger inhibitory effect, abolished T-cell proliferation, and also induced more changes in cytokine/ chemokine production.
- Published
- 2015
- Full Text
- View/download PDF
18. Distinct In Vitro Properties of Embryonic and Extraembryonic Fibroblast-Like Cells are Reflected in their in Vivo Behavior following Grafting in the Adult Mouse Brain
- Author
-
Roberta Costa, Irene Bergwerf, Eva Santermans, Nathalie De Vocht, Jelle Praet, Jasmijn Daans, Debbie Le Blon, Chloé Hoornaert, Kristien Reekmans, Niel Hens, Herman Goossens, Zwi Berneman, Ornella Parolini, Francesco Alviano, and Peter Ponsaerts
- Subjects
Medicine - Abstract
Although intracerebral transplantation of various fibroblast(-like) cell populations has been shown feasible, little is known about the actual in vivo remodeling of these cellular grafts and their environment. In this study, we aimed to compare the in vitro and in vivo behavior of two phenotypically similar—but developmentally distinct—fibroblast-like cell populations, namely, mouse embryonic fibroblasts (mEFs) and mouse fetal membrane-derived stromal cells (mFMSCs). While both mEFs and mFMSCs are readily able to reduce TNF-α secretion by LPS/IFN-γ-activated BV-2 microglia, mFMSCs and mEFs display strikingly opposite behavior with regard to VEGF production under normal and inflammatory conditions. Whereas mFMSCs downregulate VEGF production upon coculture with LPS/IFN-γ-activated BV-2 microglia, mEFs upregulate VEGF production in the presence of LPS/IFN-γ-activated BV-2 microglia. Subsequently, in vivo grafting of mFMSCs and mEFs revealed no difference in microglial and astroglial responses toward the cellular grafts. However, mFMSC grafts displayed a lower degree of neoangiogenesis compared to mEF grafts, thereby potentially explaining the lower cell number able to survive in mFMSC grafts. In summary, our results suggest that physiological differences between fibroblast-like cell populations might lie at the basis of variations in histopathological and/or clinical outcome following cell grafting in mouse brain.
- Published
- 2015
- Full Text
- View/download PDF
19. CM from intact hAM: an easily obtained product with relevant implications for translation in regenerative medicine
- Author
-
Antonietta Silini, Giorgio Pagani, Ornella Parolini, Andrea Papait, Marta Magatti, Alice Masserdotti, Patrizia Bonassi Signoroni, Anna Cargnoni, Sara Rota Nodari, Mario Bignardi, Silvia De Munari, Anna Pasotti, Pietro Romele, and Elsa Vertua
- Subjects
Medicine (General) ,Mononuclear ,Cell ,Amniotic membrane ,Medicine (miscellaneous) ,QD415-436 ,Regenerative Medicine ,Biochemistry ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Regenerative medicine ,Immunomodulation ,R5-920 ,Leukocytes ,Conditioned medium ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,Amnion ,Secretome ,Chemistry ,Research ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Translation (biology) ,Immune functional assays ,Cell Biology ,Lyophilization ,In vitro ,Cell biology ,medicine.anatomical_structure ,Monocyte differentiation ,Leukocytes, Mononuclear ,Molecular Medicine ,Stem cell - 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
20. Immunohistochemical detection of 'ex novo' HLA-DR in tumor cells determines clinical outcome in laryngeal cancer patients
- Author
-
Marta Barba, Ornella Parolini, Gaetano Paludetti, Davide Bonvissuto, Jacopo Galli, Giovanni Almadori, Stefania Boccia, Stefano Settimi, Luca Giraldi, Gabriella Cadoni, and Chiara Prampolini
- Subjects
Oncology ,medicine.medical_specialty ,Immunology ,Disease ,MHC II ,Internal medicine ,Genetics ,medicine ,HLA-DR ,Immunology and Allergy ,Humans ,tumor immunology ,Settore MED/42 - IGIENE GENERALE E APPLICATA ,Laryngeal Neoplasms ,Alleles ,Retrospective Studies ,MHC class II ,Univariate analysis ,biology ,Proportional hazards model ,business.industry ,Cancer ,Retrospective cohort study ,personalized medicine ,HLA-DR Antigens ,medicine.disease ,laryngeal squamous cell carcinoma ,immunohistochemistry ,biology.protein ,Carcinoma, Squamous Cell ,Immunohistochemistry ,Settore MED/31 - OTORINOLARINGOIATRIA ,Neoplasm Recurrence, Local ,business - Abstract
There are controversial results about the role of "ex novo" HLA-DR expression by tumor cells and its correlation with the oncological outcomes. Unfortunately, little is known about HLA-DR expression in laryngeal cancer tumor cells. The main purpose of this retrospective study is to strengthen the usefulness of studying "ex novo" HLA-DR expression on tumor cells from primary laryngeal squamous cell carcinoma (LSCC) patients and investigate its correlation with clinical outcome. We analyzed HLA-DR expression by immunohistochemical analysis in 56 patients with LSCC. The "ex novo" HLA-DR expression on laryngeal cancer tumor cells, assessing non-neoplastic LSCC - adjacent tissue, and the association of HLA-DR expression (HLA-DR+) with clinical outcomes were investigated. HLA-DR+ tumor cells were detected in 18/56 LSCC patients (32.1%). All specimens of non-neoplastic laryngeal carcinoma-adjacent tissue resulted HLA-DR negative (HLA-DR-). A statistically significant association was observed between HLA-DR + and well differentiated tumors (G1) (p
- Published
- 2021
21. Ciliary Signalling and Mechanotransduction in the Pathophysiology of Craniosynostosis
- Author
-
Federica Tiberio, Wanda Lattanzi, and Ornella Parolini
- Subjects
0301 basic medicine ,Mesenchymal stromal cells ,Review ,Biology ,QH426-470 ,Ciliopathies ,Mechanotransduction, Cellular ,Craniosynostosis ,craniofacial malformations ,Craniofacial Abnormalities ,03 medical and health sciences ,Craniosynostoses ,0302 clinical medicine ,Osteogenesis ,suture ossification ,medicine ,Genetics ,Humans ,Settore BIO/13 - BIOLOGIA APPLICATA ,Cilia ,Mechanotransduction ,Craniofacial ,Ciliary membrane ,Genetics (clinical) ,mechanotransduction ,Cilium ,Neural crest ,Cranial Sutures ,medicine.disease ,Phenotype ,craniosynostosis ,030104 developmental biology ,Neural Crest ,ciliopathies ,Osteogenic pathways ,Neuroscience ,030217 neurology & neurosurgery ,Signal Transduction ,primary cilium - Abstract
Craniosynostosis (CS) is the second most prevalent inborn craniofacial malformation; it results from the premature fusion of cranial sutures and leads to dimorphisms of variable severity. CS is clinically heterogeneous, as it can be either a sporadic isolated defect, more frequently, or part of a syndromic phenotype with mendelian inheritance. The genetic basis of CS is also extremely heterogeneous, with nearly a hundred genes associated so far, mostly mutated in syndromic forms. Several genes can be categorised within partially overlapping pathways, including those causing defects of the primary cilium. The primary cilium is a cellular antenna serving as a signalling hub implicated in mechanotransduction, housing key molecular signals expressed on the ciliary membrane and in the cilioplasm. This mechanical property mediated by the primary cilium may also represent a cue to understand the pathophysiology of non-syndromic CS. In this review, we aimed to highlight the implication of the primary cilium components and active signalling in CS pathophysiology, dissecting their biological functions in craniofacial development and in suture biomechanics. Through an in-depth revision of the literature and computational annotation of disease-associated genes we categorised 18 ciliary genes involved in CS aetiology. Interestingly, a prevalent implication of midline sutures is observed in CS ciliopathies, possibly explained by the specific neural crest origin of the frontal bone.
- Published
- 2021
22. Basic and Preclinical Research for Personalized Medicine
- Author
-
Marta Barba, Federica Iavarone, Cristian Ripoli, Andrea Urbani, Ornella Parolini, Wanda Lattanzi, Viviana Greco, Claudio Grassi, and Angelo Minucci
- Subjects
0301 basic medicine ,Actionable knowledge ,Computer science ,induced pluripotent stem cells ,Medicine (miscellaneous) ,Genomics ,Review ,Regenerative medicine ,neuroscience ,03 medical and health sciences ,Preclinical research ,0302 clinical medicine ,proteomics ,stem cells ,genomics ,Settore BIO/13 - BIOLOGIA APPLICATA ,Profiling (information science) ,mesenchymal stem cells ,business.industry ,Scientific progress ,personalized medicine ,Proteogenomics ,Data science ,030104 developmental biology ,proteogenomics ,Medicine ,Personalized medicine ,business ,030217 neurology & neurosurgery - Abstract
Basic and preclinical research founded the progress of personalized medicine by providing a prodigious amount of integrated profiling data and by enabling the development of biomedical applications to be implemented in patient-centered care and cures. If the rapid development of genomics research boosted the birth of personalized medicine, further development in omics technologies has more recently improved our understanding of the functional genome and its relevance in profiling patients’ phenotypes and disorders. Concurrently, the rapid biotechnological advancement in diverse research areas enabled uncovering disease mechanisms and prompted the design of innovative biological treatments tailored to individual patient genotypes and phenotypes. Research in stem cells enabled clarifying their role in tissue degeneration and disease pathogenesis while providing novel tools toward the development of personalized regenerative medicine strategies. Meanwhile, the evolving field of integrated omics technologies ensured translating structural genomics information into actionable knowledge to trace detailed patients’ molecular signatures. Finally, neuroscience research provided invaluable models to identify preclinical stages of brain diseases. This review aims at discussing relevant milestones in the scientific progress of basic and preclinical research areas that have considerably contributed to the personalized medicine revolution by bridging the bench-to-bed gap, focusing on stem cells, omics technologies, and neuroscience fields as paradigms.
- Published
- 2021
23. 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
0301 basic medicine ,Histology ,Amniotic fluid ,lcsh:Biotechnology ,Biomedical Engineering ,Bioengineering ,Review ,tissue regeneration ,Biology ,immunomodulation ,Umbilical cord ,Regenerative medicine ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,perinatal derivatives ,Placenta ,lcsh:TP248.13-248.65 ,microRNA ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,Fetus ,Regeneration (biology) ,Bioengineering and Biotechnology ,Cell biology ,secretome ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,extracellular vesicles ,Biotechnology - 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
24. Mesenchymal Stromal Cells and Their Secretome: New Therapeutic Perspectives for Skeletal Muscle Regeneration
- Author
-
Martina Sandonà, Lorena Di Pietro, Federica Esposito, Alessia Ventura, Antonietta Rosa Silini, Ornella Parolini, and Valentina Saccone
- Subjects
0301 basic medicine ,muscular dystrophy ,Histology ,muscle ,Biomedical Engineering ,atrophy ,extracellular vesicles ,mesenchymal stromal cells ,muscle regeneration ,secretome ,Bioengineering ,Review ,Biology ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Myocyte ,Settore BIO/13 - BIOLOGIA APPLICATA ,Muscular dystrophy ,Myogenesis ,Regeneration (biology) ,Mesenchymal stem cell ,Skeletal muscle ,Bioengineering and Biotechnology ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Bone marrow ,TP248.13-248.65 ,Biotechnology - Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells found in different tissues: bone marrow, peripheral blood, adipose tissues, skeletal muscle, perinatal tissues, and dental pulp. MSCs are able to self-renew and to differentiate into multiple lineages, and they have been extensively used for cell therapy mostly owing to their anti-fibrotic and immunoregulatory properties that have been suggested to be at the basis for their regenerative capability. MSCs exert their effects by releasing a variety of biologically active molecules such as growth factors, chemokines, and cytokines, either as soluble proteins or enclosed in extracellular vesicles (EVs). Analyses of MSC-derived secretome and in particular studies on EVs are attracting great attention from a medical point of view due to their ability to mimic all the therapeutic effects produced by the MSCs (i.e., endogenous tissue repair and regulation of the immune system). MSC-EVs could be advantageous compared with the parental cells because of their specific cargo containing mRNAs, miRNAs, and proteins that can be biologically transferred to recipient cells. MSC-EV storage, transfer, and production are easier; and their administration is also safer than MSC therapy. The skeletal muscle is a very adaptive tissue, but its regenerative potential is altered during acute and chronic conditions. Recent works demonstrate that both MSCs and their secretome are able to help myofiber regeneration enhancing myogenesis and, interestingly, can be manipulated as a novel strategy for therapeutic interventions in muscular diseases like muscular dystrophies or atrophy. In particular, MSC-EVs represent promising candidates for cell free-based muscle regeneration. In this review, we aim to give a complete picture of the therapeutic properties and advantages of MSCs and their products (MSC-derived EVs and secreted factors) relevant for skeletal muscle regeneration in main muscular diseases.
- Published
- 2021
25. Editorial: Perinatal Derivatives and the Road to Clinical Translation, Volume I
- Author
-
Antonietta R. Silini, Peter Ponsaerts, and Ornella Parolini
- Subjects
medicine.medical_specialty ,in vitro characterisation ,Histology ,Biomedical Engineering ,regenerative medicine ,Bioengineering ,Biology ,Translation (geometry) ,secretome ,consensus ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,Medical physics ,Engineering sciences. Technology ,TP248.13-248.65 ,perinatal ,Biotechnology ,Volume (compression) - Published
- 2021
26. The role of b cells in pe pathophysiology: A potential target for perinatal cell-based therapy?
- Author
-
Anna Cargnoni, Antonietta Silini, Ornella Parolini, Francesca Romana Stefani, Alice Masserdotti, Andrea Papait, and Marta Magatti
- Subjects
Amniotic fluid ,Regulatory B cells ,Placenta ,Review ,Perinatal cells ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,Immune system ,Pre-Eclampsia ,Immunity ,Pregnancy ,Medicine ,Animals ,Humans ,Settore BIO/13 - BIOLOGIA APPLICATA ,Amnion ,Physical and Theoretical Chemistry ,Umbilical cord ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,B cell ,Embryonic Stem Cells ,Autoantibodies ,B-Lymphocytes ,B cells ,business.industry ,Organic Chemistry ,General Medicine ,Preeclampsia ,Computer Science Applications ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer research ,Chorionic villi ,Female ,business ,Stem Cell Transplantation - Abstract
The pathophysiology of preeclampsia (PE) is poorly understood; however, there is a large body of evidence that suggests a role of immune cells in the development of PE. Amongst these, B cells are a dominant element in the pathogenesis of PE, and they have been shown to play an important role in various immune-mediated diseases, both as pro-inflammatory and regulatory cells. Perinatal cells are defined as cells from birth-associated tissues isolated from term placentas and fetal annexes and more specifically from the amniotic membrane, chorionic membrane, chorionic villi, umbilical cord (including Wharton’s jelly), the basal plate, and the amniotic fluid. They have drawn particular attention in recent years due to their ability to modulate several aspects of immunity, making them promising candidates for the prevention and treatment of various immune-mediated diseases. In this review we describe main findings regarding the multifaceted in vitro and in vivo immunomodulatory properties of perinatal cells, with a focus on B lymphocytes. Indeed, we discuss evidence on the ability of perinatal cells to inhibit B cell proliferation, impair B cell differentiation, and promote regulatory B cell formation. Therefore, the findings discussed herein unveil the possibility to modulate B cell activation and function by exploiting perinatal immunomodulatory properties, thus possibly representing a novel therapeutic strategy in PE.
- Published
- 2021
27. 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
0301 basic medicine ,medicine.medical_specialty ,ARDS ,Histology ,lcsh:Biotechnology ,Biomedical Engineering ,Bioengineering ,Context (language use) ,Review ,PLacental eXpanded ,law.invention ,Sepsis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Fibrosis ,law ,coronavirus-induced disease 2019 ,lcsh:TP248.13-248.65 ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,Intensive care medicine ,perinatal ,Disseminated intravascular coagulation ,business.industry ,Bioengineering and Biotechnology ,medicine.disease ,Intensive care unit ,030104 developmental biology ,030220 oncology & carcinogenesis ,severe acute respiratory distress syndrome coronavirus-2 ,business ,Cytokine storm ,mesenchymal stromal cells ,Biotechnology - 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
28. Anti-inflammatory effects of adult stem cells in sustained lung injury: a comparative study.
- Author
-
Yuben Moodley, Vijesh Vaghjiani, James Chan, Svetlana Baltic, Marisa Ryan, Jorge Tchongue, Chrishan S Samuel, Padma Murthi, Ornella Parolini, and Ursula Manuelpillai
- Subjects
Medicine ,Science - Abstract
Lung diseases are a major cause of global morbidity and mortality that are treated with limited efficacy. Recently stem cell therapies have been shown to effectively treat animal models of lung disease. However, there are limitations to the translation of these cell therapies to clinical disease. Studies have shown that delayed treatment of animal models does not improve outcomes and that the models do not reflect the repeated injury that is present in most lung diseases. We tested the efficacy of amnion mesenchymal stem cells (AM-MSC), bone marrow MSC (BM-MSC) and human amniotic epithelial cells (hAEC) in C57BL/6 mice using a repeat dose bleomycin-induced model of lung injury that better reflects the repeat injury seen in lung diseases. The dual bleomycin dose led to significantly higher levels of inflammation and fibrosis in the mouse lung compared to a single bleomycin dose. Intravenously infused stem cells were present in the lung in similar numbers at days 7 and 21 post cell injection. In addition, stem cell injection resulted in a significant decrease in inflammatory cell infiltrate and a reduction in IL-1 (AM-MSC), IL-6 (AM-MSC, BM-MSC, hAEC) and TNF-α (AM-MSC). The only trophic factor tested that increased following stem cell injection was IL-1RA (AM-MSC). IL-1RA levels may be modulated by GM-CSF produced by AM-MSC. Furthermore, only AM-MSC reduced collagen deposition and increased MMP-9 activity in the lung although there was a reduction of the pro-fibrogenic cytokine TGF-β following BM-MSC, AM-MSC and hAEC treatment. Therefore, AM-MSC may be more effective in reducing injury following delayed injection in the setting of repeated lung injury.
- Published
- 2013
- Full Text
- View/download PDF
29. The Cells and Extracellular Matrix of Human Amniotic Membrane Hinder the Growth and Invasive Potential of Bladder Urothelial Cancer Cells
- Author
-
Taja Železnik Ramuta, Urška Dragin Jerman, Larisa Tratnjek, Aleksandar Janev, Marta Magatti, Elsa Vertua, Patrizia Bonassi Signoroni, Antonietta Rosa Silini, Ornella Parolini, and Mateja Erdani Kreft
- Subjects
0301 basic medicine ,Histology ,lcsh:Biotechnology ,Biomedical Engineering ,regenerative medicine ,Bioengineering ,02 engineering and technology ,Biology ,anticancer ,urologic and male genital diseases ,Regenerative medicine ,Extracellular matrix ,03 medical and health sciences ,Tissue engineering ,immune system diseases ,lcsh:TP248.13-248.65 ,hemic and lymphatic diseases ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,Original Research ,Bladder cancer ,amniotic membrane ,Bioengineering and Biotechnology ,virus diseases ,food and beverages ,amniotic epithelial cells ,021001 nanoscience & nanotechnology ,medicine.disease ,Epithelium ,030104 developmental biology ,medicine.anatomical_structure ,tissue engineering ,Amniotic epithelial cells ,amniotic mesenchymal stromal cells ,Cancer research ,bladder cancer ,urothelial cancer cells ,Basal lamina ,0210 nano-technology ,Wound healing ,Biotechnology - Abstract
Bladder cancer is one of the most common cancers among men in industrialized countries and on the global level incidence and mortality rates are increasing. In spite of progress in surgical treatment and chemotherapy, the prognosis remains poor for patients with muscle-invasive bladder cancer. Therefore, there is a great need for the development of novel therapeutic approaches. The human amniotic membrane (hAM) is a multi-layered membrane that comprises the innermost part of the placenta. It has unique properties that make it suitable for clinical use, such as the ability to promote wound healing and decrease scarring, low immunogenicity, and immunomodulatory, antimicrobial and anticancer properties. This study aimed to investigate the effect of (i) hAM-derived cells and (ii) hAM scaffolds on the growth dynamics, proliferation rate, and invasive potential of muscle-invasive bladder cancer T24 cells. Our results show that 24 and 48 h of co-culturing T24 cells with hAM-derived cells (at 1:1 and 1:4 ratios) diminished the proliferation rate of T24 cells. Furthermore, when seeded on hAM scaffolds, namely (1) epithelium of hAM (e-hAM), (2) basal lamina of hAM (denuded; d-hAM), and (3) stroma of hAM (s-hAM), the growth dynamic of T24 cells was altered and proliferation was reduced, even more so by the e-hAM scaffolds. Importantly, despite their muscle-invasive potential, the T24 cells did not disrupt the basal lamina of hAM scaffolds. Furthermore, we observed a decrease in the expression of epithelial-mesenchymal transition (EMT) markers N-cadherin, Snail and Slug in T24 cells grown on hAM scaffolds and individual T24 cells even expressed epithelial markers E-cadherin and occludin. Our study brings new knowledge on basic mechanisms of hAM affecting bladder carcinogenesis and the results serve as a good foundation for further research into the potential of hAM-derived cells and the hAM extracellular matrix to serve as a novel bladder cancer treatment.
- Published
- 2020
- Full Text
- View/download PDF
30. GLI1 and AXIN2 Are Distinctive Markers of Human Calvarial Mesenchymal Stromal Cells in Nonsyndromic Craniosynostosis
- Author
-
Lorena Di Pietro, Luca Massimi, Marta Barba, Chiara Prampolini, Alessia Vita, Paolo Frassanito, Enrico Guadagni, Davide Bonvissuto, Wanda Lattanzi, Gianpiero Tamburrini, Ornella Parolini, and Sabrina Ceccariglia
- Subjects
Male ,Pathology ,GLI1 ,lcsh:Chemistry ,Cranial vault ,Settore BIO/13 - BIOLOGIA APPLICATA ,lcsh:QH301-705.5 ,Spectroscopy ,Cells, Cultured ,Cultured ,personalised medicine ,Cell Differentiation ,General Medicine ,Computer Science Applications ,bone development ,Intramembranous ossification ,nonsyndromic craniosynostosis ,Female ,medicine.symptom ,Stem cell ,mesenchymal stromal cells ,medicine.medical_specialty ,Cells ,Primary Cell Culture ,AXIN2 ,Down-Regulation ,regenerative medicine ,Context (language use) ,Biology ,Zinc Finger Protein GLI1 ,Catalysis ,Article ,Craniosynostosis ,osteogenesis ,Inorganic Chemistry ,Craniosynostoses ,Axin Protein ,medicine ,Humans ,stem cell niche ,Physical and Theoretical Chemistry ,Molecular Biology ,Endochondral ossification ,Ossification ,Organic Chemistry ,Mesenchymal stem cell ,Skull ,Infant, Newborn ,Infant ,Mesenchymal Stem Cells ,Newborn ,medicine.disease ,lcsh:Biology (General) ,lcsh:QD1-999 ,cranial suture ,Biomarkers - Abstract
All skeletal bones house osteogenic stem cell niches, in which mesenchymal stromal cells (MSC) provide progenitors for tissue growth and regeneration. They have been widely studied in long bones formed through endochondral ossification. Limited information is available on the composition of the osteogenic niche in flat bones (i.e., skull vault bones) that develop through direct membranous ossification. Craniosynostosis (CS) is a congenital craniofacial defect due to the excessive and premature ossification of skull vault sutures. This study aimed at analysing the expression of GLI1, AXIN2 and THY1 in the context of the human skull vault, using nonsyndromic forms of CS (NCS) as a model to test their functional implication in the aberrant osteogenic process. The expression of selected markers was studied in NCS patients&rsquo, calvarial bone specimens, to assess the in vivo location of cells, and in MSC isolated thereof. The marker expression profile was analysed during in vitro osteogenic differentiation to validate the functional implication. Our results show that GLI1 and AXIN2 are expressed in periosteal and endosteal locations within the osteogenic niche of human calvarial bones. Their expression is higher in MSC isolated from calvarial bones than in those isolated from long bones and tends to decrease upon osteogenic commitment and differentiation. In particular, AXIN2 expression was lower in cells isolated from prematurely fused sutures than in those derived from patent sutures of NCS patients. This suggests that AXIN2 could reasonably represent a marker for the stem cell population that undergoes depletion during the premature ossification process occurring in CS.
- Published
- 2020
31. B Lymphocytes as Targets of the Immunomodulatory Properties of Human Amniotic Mesenchymal Stromal Cells
- Author
-
Marta Magatti, Alice Masserdotti, Patrizia Bonassi Signoroni, Elsa Vertua, Francesca Romana Stefani, Antonietta Rosa Silini, and Ornella Parolini
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,placenta ,CD14 ,Immunology ,Lymphocyte Activation ,immunomodulation ,plasma cells ,03 medical and health sciences ,0302 clinical medicine ,PRDM1 ,medicine ,Humans ,Settore BIO/13 - BIOLOGIA APPLICATA ,Immunology and Allergy ,Amnion ,Cells, Cultured ,B cell ,Original Research ,B-Lymphocytes ,B cells ,amniotic membrane ,Chemistry ,Mesenchymal stem cell ,TLR9 ,Cell Differentiation ,Mesenchymal Stem Cells ,Acquired immune system ,In vitro ,Cell biology ,conditioned medium ,030104 developmental biology ,medicine.anatomical_structure ,Culture Media, Conditioned ,plasmablast ,mesenchymal stromal cells ,lcsh:RC581-607 ,030215 immunology ,IRF4 - Abstract
Mesenchymal stromal cells (MSC) from the amniotic membrane of human term placenta (hAMSC), and the conditioned medium generated from their culture (CM-hAMSC) offer significant tools for their use in regenerative medicine mainly due to their immunomodulatory properties. Interestingly, hAMSC and their CM have been successfully exploited in preclinical disease models of inflammatory and autoimmune diseases where depletion or modulation of B cells have been indicated as an effective treatment, such as inflammatory bowel disease, lung fibrosis, would healing, collagen-induced arthritis, and multiple sclerosis. While the interactions between hAMSC or CM-hAMSC and T lymphocytes, monocytes, dendritic cells, and macrophages has been extensively explored, how they affect B lymphocytes remains unclear. Considering that B cells are key players in the adaptive immune response and are a central component of different diseases, in this study we investigated the in vitro properties of hAMSC and CM-hAMSC on B cells. We provide evidence that both hAMSC and CM-hAMSC strongly suppressed CpG-activated B-cell proliferation. Moreover, CM-hAMSC blocked B-cell differentiation, with an increase of the proportion of mature B cells, and a reduction of antibody secreting cell formation. We observed the strong inhibition of B cell terminal differentiation into CD138+ plasma cells, as further shown by a significant decrease of the expression of interferon regulatory factor 4 (IRF-4), PR/SET domain 1(PRDM1), and X-box binding protein 1 (XBP-1) genes. Our results point out that the mechanism by which CM-hAMSC impacts B cell proliferation and differentiation is mediated by secreted factors, and prostanoids are partially involved in these actions. Factors contained in the CM-hAMSC decreased the CpG-uptake sensors (CD205, CD14, and TLR9), suggesting that B cell stimulation was affected early on. CM-hAMSC also decreased the expression of interleukin-1 receptor-associated kinase (IRAK)-4, consequently inhibiting the entire CpG-induced downstream signaling pathway. Overall, these findings add insight into the mechanism of action of hAMSC and CM-hAMSC and are useful to better design their potential therapeutic application in B-cell mediated diseases.
- Published
- 2020
- Full Text
- View/download PDF
32. miRNA Reference Genes in Extracellular Vesicles Released from Amniotic Membrane-Derived Mesenchymal Stromal Cells
- Author
-
Ornella Parolini, Carlotta Perucca Orfei, Alessandra Colombini, Enrico Ragni, Laura de Girolamo, Marco Viganò, and Antonietta Silini
- Subjects
placenta ,Tendon disorders ,Pharmaceutical Science ,lcsh:RS1-441 ,reference genes ,Biology ,Extracellular vesicles ,Regenerative medicine ,Article ,lcsh:Pharmacy and materia medica ,03 medical and health sciences ,0302 clinical medicine ,Amniotic membrane ,Delivery platforms ,Mesenchymal stromal cells ,MiRNAs ,Nanocarriers ,Placenta ,Reference genes ,Tendinopathy ,microRNA ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,030304 developmental biology ,0303 health sciences ,amniotic membrane ,Mesenchymal stem cell ,Cell biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,miRNAs ,mesenchymal stromal cells ,extracellular vesicles ,Small nuclear RNA - Abstract
Human amniotic membrane and amniotic membrane-derived mesenchymal stromal cells (hAMSCs) have produced promising results in regenerative medicine, especially for the treatment of inflammatory-based diseases and for different injuries including those in the orthopedic field such as tendon disorders. hAMSCs have been proposed to exert their anti-inflammatory and healing potential via secreted factors, both free and conveyed within extracellular vesicles (EVs). In particular, EV miRNAs are considered privileged players due to their impact on target cells and tissues, and their future use as therapeutic molecules is being intensely investigated. In this view, EV-miRNA quantification in either research or future clinical products has emerged as a crucial paradigm, although, to date, largely unsolved due to lack of reliable reference genes (RGs). In this study, a panel of thirteen putative miRNA RGs (let-7a-5p, miR-16-5p, miR-22-5p, miR-23a-3p, miR-26a-5p, miR-29a-5p, miR-101-3p, miR-103a-3p, miR-221-3p, miR-423-5p, miR-425-5p, miR-660-5p and U6 snRNA) that were identified in different EV types was assessed in hAMSC-EVs. A validated experimental pipeline was followed, sifting the output of four largely accepted algorithms for RG prediction (geNorm, NormFinder, BestKeeper and &Delta, Ct method). Out of nine RGs constitutively expressed across all EV isolates, miR-101-3p and miR-22-5p resulted in the most stable RGs, whereas miR-423-5p and U6 snRNA performed poorly. miR-22-5p was also previously reported to be a reliable RG in adipose-derived MSC-EVs, suggesting its suitability across samples isolated from different MSC types. Further, to shed light on the impact of incorrect RG choice, the level of five tendon-related miRNAs (miR-29a-3p, miR-135a-5p, miR-146a-5p, miR-337-3p, let-7d-5p) was compared among hAMSC-EVs isolates. The use of miR-423-5p and U6 snRNA did not allow a correct quantification of miRNA incorporation in EVs, leading to less accurate fingerprinting and, if used for potency prediction, misleading indication of the most appropriate clinical batch. These results emphasize the crucial importance of RG choice for EV-miRNAs in hAMSCs studies and contribute to the identification of reliable RGs such as miR-101-3p and miR-22-5p to be validated in other MSC-EVs related fields.
- Published
- 2020
33. Redirecting T cells to Ewing's sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection.
- Author
-
Manfred Lehner, Gabriel Götz, Julia Proff, Niels Schaft, Jan Dörrie, Florian Full, Armin Ensser, Yves A Muller, Adelheid Cerwenka, Hinrich Abken, Ornella Parolini, Peter F Ambros, Heinrich Kovar, and Wolfgang Holter
- Subjects
Medicine ,Science - Abstract
We explored the possibility to target Ewing's sarcoma family of tumors (ESFT) by redirecting T cells. To this aim, we considered NKG2D-ligands (NKG2D-Ls) as possible target antigens. Detailed analysis of the expression of MICA, MICB, ULBP-1, -2, and -3 in fourteen ESFT cell lines revealed consistent expression of at least one NKG2D-L. Thus, for redirecting T cells, we fused a CD3ζ/CD28-derived signaling domain to the ectodomain of NKG2D, however, opposite transmembrane orientation of this signaling domain and NKG2D required inverse orientation fusion of either of them. We hypothesized that the particularly located C-terminus of the NKG2D ectodomain should allow reengineering of the membrane anchoring from a native N-terminal to an artificial C-terminal linkage. Indeed, the resulting chimeric NKG2D receptor (chNKG2D) was functional and efficiently mediated ESFT cell death triggered by activated T cells. Notably, ESFT cells with even low NKG2D-L expression were killed by CD8(pos) and also CD4(pos) cells. Both, mRNA transfection and lentiviral transduction resulted in high level surface expression of chNKG2D. However, upon target-cell recognition receptor surface levels were maintained by tranfected RNA only during the first couple of hours after transfection. Later, target-cell contact resulted in strong and irreversible receptor down-modulation, whereas lentivirally mediated expression of chNKG2D remained constant under these conditions. Together, our study defines NKG2D-Ls as targets for a CAR-mediated T cell based immunotherapy of ESFT. A comparison of two different methods of gene transfer reveals strong differences in the susceptibility to ligand-induced receptor down-modulation with possible implications for the applicability of RNA transfection.
- Published
- 2012
- Full Text
- View/download PDF
34. Characterization of the conditioned medium from amniotic membrane cells: prostaglandins as key effectors of its immunomodulatory activity.
- Author
-
Daniele Rossi, Stefano Pianta, Marta Magatti, Peter Sedlmayr, and Ornella Parolini
- Subjects
Medicine ,Science - Abstract
We previously demonstrated that cells isolated from the mesenchymal region of the human amniotic membrane (human amniotic mesenchymal tissue cells, hAMTC) possess immunoregulatory roles, such as inhibition of lymphocyte proliferation and cytokine production, and suppression of generation and maturation of monocyte-derived dendritic cells, as reported for MSC from other sources. The precise factors and mechanisms responsible for the immunoregulatory roles of hAMTC remain unknown. In this study, we aimed to identify the soluble factors released by hAMTC and responsible for the anti-proliferative effect on lymphocytes, and the mechanisms underlying their actions, in vitro. Conditioned medium (CM) was prepared under routine culture conditions from hAMTC (CM-hAMTC) and also from fragments of the whole human amniotic membrane (CM-hAM). We analyzed the thermostability, chemical nature, and the molecular weight of the factors likely responsible for the anti-proliferative effects. We also evaluated the participation of cytokines known to be involved in the immunomodulatory actions of MSC from other sources, and attempted to block different synthetic pathways. We demonstrate that the inhibitory factors are temperature-stable, have a small molecular weight, and are likely of a non-proteinaceous nature. Only inhibition of cyclooxygenase pathway partially reverted the anti-proliferative effect, suggesting prostaglandins as key effector molecules. Factors previously documented to take part in the inhibitory effects of MSCs from other sources (HGF, TGF-β, NO and IDO) were not involved. Furthermore, we prove for the first time that the anti-proliferative effect is intrinsic to the amniotic membrane and cells derived thereof, since it is manifested in the absence of stimulating culture conditions, as opposed to MSC derived from the bone marrow, which possess an anti-proliferative ability only when cultured in the presence of activating stimuli. Finally, we show that the amniotic membrane could be an interesting source of soluble factors, without referring to extensive cell preparation.
- Published
- 2012
- Full Text
- View/download PDF
35. Incorporating placental tissue in cord blood banking for stem cell transplantation
- Author
-
Caterina Giovanna Valentini, Maria Bianchi, Luciana Teofili, Ornella Parolini, and Antonietta Silini
- Subjects
umbilical blood ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Stromal cell ,placenta ,Cellular differentiation ,regenerative medicine ,Tissue Banks ,Regenerative medicine ,Hematopoietic stem cells ,individualized therapy ,mesenchymal stem/stromal cells ,personalized medicine ,03 medical and health sciences ,Pregnancy ,Humans ,Settore BIO/13 - BIOLOGIA APPLICATA ,Medicine ,Precision Medicine ,business.industry ,Mesenchymal stem cell ,food and beverages ,Cell Differentiation ,Mesenchymal Stem Cells ,Hematology ,Transplantation ,Haematopoiesis ,030104 developmental biology ,Cord blood ,Blood Banks ,Female ,Stem cell ,business ,Stem Cell Transplantation - Abstract
Human term placenta is comprised of various tissues from which different cells can be obtained, including hematopoietic stem cells and mesenchymal stem/stromal cells (MSCs). Areas covered: This review will discuss the possibility to incorporate placental tissue cells in cord blood banking. It will discuss general features of human placenta, with a brief review of the immune cells at the fetal-maternal interface and the different cell populations isolated from placenta, with a particular focus on MSCs. It will address the question as to why placenta-derived MSCs should be banked with their hematopoietic counterparts. It will discuss clinical trials which are studying safety and efficacy of placenta tissue-derived MSCs in selected diseases, and preclinical studies which have proven their therapeutic properties in other diseases. It will discuss banking of umbilical cord blood and raise several issues for improvement, and the applications of cord blood cells in non-malignant disorders. Expert commentary: Umbilical cord blood banking saves lives worldwide. The concomitant banking of non-hematopoietic cells from placenta, which could be applied therapeutically in the future, alone or in combination to their hematopoietic counterparts, could exploit current banking processes while laying the foundation for clinical trials exploring placenta-derived cell therapies in regenerative medicine.
- Published
- 2018
- Full Text
- View/download PDF
36. Amniotic Membrane Application Reduces Liver Fibrosis in a Bile Duct Ligation Rat Model
- Author
-
Luciana B. Sant'anna, Anna Cargnoni, Lorenzo Ressel, Graziella Vanosi, and Ornella Parolini Ph.D.
- Subjects
Medicine - Abstract
Biliary fibrosis and resultant cirrhosis are among the most common outcomes of chronic liver diseases. Currently, liver transplantation remains the only effective treatment. In seeking alternative therapeutic approaches, we focused on the potential use of the human amniotic membrane (AM). Indeed, AM has gained increasing importance for its antiscarring, anti-inflammatory, and wound-healing properties, as well as for the multipotent differentiation ability and immunomodulatory features of AM-derived cells. Intriguingly, we have recently demonstrated that placenta-derived cells reduce lung fibrosis in bleomycin-treated mice, and that AM patches reduce postischemic cardiac injury in rats. Hence, we have now investigated the effects of human AM on biliary fibrosis induced in rats through the bile duct ligation (BDL) procedure. A fragment of human AM was applied onto the liver surface after BDL and the effects on fibrosis establishment and progression were evaluated at different time points in comparison with fibrosis progression in control BDL rats. The degree of liver fibrosis was first assessed by the semiquantitative Knodell scoring system and, thereafter, by digital image morphometric analysis to quantify the area occupied by ductular reaction, activated myofibroblasts, and collagen deposition. We demonstrated a significant reduction in the severity of BDL-induced fibrosis in AM-treated rats. Indeed, while fibrosis progressed rapidly in control BDL rats, leading to cirrhosis within 6 weeks, AM-treated rats showed confined fibrosis at the portal/periportal area with no signs of cirrhosis, and a reduction in collagen deposition to about 50% of levels observed in control BDL rats. In addition, the AM was able to significantly slow the gradual progression of the ductular reaction and reduce, at all time points, the area occupied by activated myofibroblasts. These findings suggest that human AM, when applied as a patch onto the liver surface, might inhibit fibrosis progression in BDL-injured livers, and could protect against hepatic damage associated with fibrotic degeneration.
- Published
- 2011
- Full Text
- View/download PDF
37. International Placenta Stem Cell Society: Planting the Seed for Placenta Stem Cell Research
- Author
-
Cesar V. Borlongan and Ornella Parolini
- Subjects
Medicine - Published
- 2010
- Full Text
- View/download PDF
38. Amniotic Membrane Patching Promotes Ischemic Rat Heart Repair
- Author
-
Anna Cargnoni, Marco Di Marcello, Marino Campagnol, Claudia Nassuato, Alberto Albertini, and Ornella Parolini Ph.D.
- Subjects
Medicine - Abstract
The amniotic membrane has long been applied for wound healing and treatment of ophthalmological disorders, even though the mechanisms underlying its actions remain to be clarified. Recently, cells derived from fetal membranes of human term placenta have raised strong interest in regenerative medicine for their stem cell potential and immunomodulatory features. Our study aimed to investigate the possible utility of amniotic membrane to limit postischemic cardiac injury. A fragment of human amniotic membrane was applied onto the left ventricle of rats that had undergone ischemia through left anterior descending coronary artery ligation. Echocardiographic assessment of morphological and functional cardiac parameters was then performed over a 3-month period. We demonstrated that application of an amniotic membrane fragment onto ischemic rat hearts could significantly reduce postischemic cardiac dysfunction. The amniotic membrane-treated rats showed higher preservation of cardiac dimensions and improved cardiac contractile function in terms of higher left ventricle ejection fraction, fractional shortening, and wall thickening. These improvements were apparent by day 7 after application of the amniotic membrane, persisted for at least 2 months, and occurred independently of cardiac injury severity. No engraftment of amniotic cells was detected into host cardiac tissues. Our results suggest that use of amniotic membrane may constitute a convenient vehicle for supplying cells that produce cardioprotective soluble factors, and reinforce the notion that this tissue constitutes a cell source with clinical potential that has yet to be completely revealed.
- Published
- 2009
- Full Text
- View/download PDF
39. Amniotic Mesenchymal Tissue Cells Inhibit Dendritic Cell Differentiation of Peripheral Blood and Amnion Resident Monocytes
- Author
-
Marta Magatti, Silvia De Munari, Elsa Vertua, Claudia Nassauto, Alberto Albertini, Georg S. Wengler, and Ornella Parolini Ph.D.
- Subjects
Medicine - Abstract
Cells derived from the amniotic membranes of human term placenta have drawn much interest for their characteristics of multipotency and low immunogenicity, supporting a variety of possible clinical applications in the field of cell transplantation and regenerative medicine. We have previously shown that cells derived from the mesenchymal region of human amnion (AMTC) can strongly inhibit T-lymphocyte proliferation. In this study, we demonstrate that AMTC can block differentiation and maturation of monocytes into dendritic cells (DC), preventing the expression of the DC marker CD1a and reducing the expression of HLA-DR, CD80, and CD83. The monocyte maturation block resulted in impaired allostimulatory ability of these cells on allogeneic T cells. In attempting to define the mechanisms responsible for these findings, we have observed that the presence of AMTC in differentiating DC cultures results in the arrest of the cells to the G 0 phase and abolishes the production of inflammatory cytokines such as TNF-α, CXCL10, CXCL9, and CCL5. Finally, we also demonstrate that the monocytic cells present in the amniotic mesenchymal region fail to differentiate toward the DC lineage. Taken together, our data suggest that the mechanisms by which AMTC exert immumodulatory effects do not only relate directly to T cells, but also include inhibition of the generation and maturation of antigen-presenting cells. In this context, AMTC represent a very attractive source of multipotent allogeneic cells that promise to be remarkably valuable for cell transplantation approaches, not only due to their low immunogenicity, but also because of the added potential of modulating immune responses, which could be fundamental both for controlling graft rejection after transplantation and also for controlling diseases characterized by inflammatory processes.
- Published
- 2009
- Full Text
- View/download PDF
40. Transplantation of Allogeneic and Xenogeneic Placenta-Derived Cells Reduces Bleomycin-Induced Lung Fibrosis
- Author
-
Anna Cargnoni, Lucia Gibelli, Alessandra Tosini, Patrizia Bonassi Signoroni, Claudia Nassuato, Davide Arienti, Guerino Lombardi, Alberto Albertini, Georg S. Wengler, and Ornella Parolini Ph.D.
- Subjects
Medicine - Abstract
Fetal membranes (amnion and chorion) have recently raised significant attention as potential sources of stem cells. We have recently demonstrated that cells derived from human term placenta show stem cell phenotype, high plasticity, and display low immunogenicity both in vitro and in vivo. Moreover, placenta-derived cells, after xenotransplantation, are able to engraft in solid organs including the lung. On these bases, we studied the effects of fetal membrane-derived cells on a mouse model of bleomycin-induced lung fibrosis. Fetal membrane-derived cells were infused 15 min after intratracheal bleomycin instillation. Different delivery routes were used: intraperitoneal or intratracheal for both xenogeneic and allogeneic cells, and intravenous for allogeneic cells. The effects of the transplanted cells on bleomycin-induced inflammatory and fibrotic processes were then scored and compared between transplanted and control animals at different time points. By PCR and immunohistochemistry analyses, we demonstrated the presence of transplanted cells 3, 7, 9, and 14 days after transplantation. Concomitantly, we observed a clear decrease in neutrophil infiltration and a significant reduction in the severity of bleomycin-induced lung fibrosis in mice treated with placenta-derived cells, irrespective of the source (allogeneic or xenogeneic) or delivery route. Our findings constitute further evidence in support of the hypothesis that placenta-derived cells could be useful for clinical application, and warrant further studies toward the use of these cells for the repair of tissue damage associated with inflammatory and fibrotic degeneration.
- Published
- 2009
- Full Text
- View/download PDF
41. Amnion: A Potent Graft Source for Cell Therapy in Stroke
- Author
-
Seong Jin Yu, Maddalena Soncini, Yuji Kaneko, David C. Hess, Ornella Parolini, and Cesar V. Borlongan Ph.D.
- Subjects
Medicine - Abstract
Regenerative medicine is a new field primarily based on the concept of transplanting exogenous or stimulating endogenous stem cells to generate biological substitutes and improve tissue functions. Recently, amnion-derived cells have been reported to have multipotent differentiation ability, and these cells have attracted attention as a novel cell source for cell transplantation therapy. Cells isolated from amniotic membrane can differentiate into all three germ layers, have low immunogenicity and anti-inflammatory function, and do not require the destruction of human embryos for their isolation, thus circumventing the ethical debate commonly associated with the use of human embryonic stem cells. Accumulating evidence now suggests that the amnion, which had been discarded after parturition, is a highly potent transplant material in the field of regenerative medicine. In this report, we review the current progress on the characterization of MSCs derived from the amnion as a remarkable transplantable cell population with therapeutic potential for multiple CNS disorders, especially stroke.
- Published
- 2009
- Full Text
- View/download PDF
42. The dichotomy of placenta-derived cells in cancer growth
- Author
-
Ornella Parolini, Sara Cancelli, Marta Magatti, Antonietta Silini, Anna Cargnoni, and Patrizia Bonassi Signoroni
- Subjects
0301 basic medicine ,Placenta ,Cell- and Tissue-Based Therapy ,Mesenchymal stromal cells ,Amniotic membrane ,Context (language use) ,Biology ,Bioinformatics ,Regenerative medicine ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,Neoplasms ,Decidua ,medicine ,Animals ,Humans ,Settore BIO/13 - BIOLOGIA APPLICATA ,Umbilical cord ,Cancer ,Chorion ,Reproductive Medicine ,Obstetrics and Gynecology ,Developmental Biology ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Female ,Bone marrow - Abstract
Placenta-derived mesenchymal stromal cells (MSC) have often been considered to linger behind their equivalents from other tissues, such as MSC from bone marrow, in many aspects including their therapeutic potential in regenerative medicine. Nowadays however, it is clear that certain aspects make placental MSC attractive as a cellular therapy, such as their lack of ethical concerns and ease of isolation from human term placenta, a material long regarded as biological waste. Moreover, placental MSC virtually lack expression of human leukocyte antigens and co-stimulatory molecules, making them very attractive for transplantation in allogeneic settings. In the context of cancer, cell therapy remains an area of intense investigation whereby MSC have been shown to play opposing roles, and placental MSC are no exception. In this review, we will discuss dichotomy of placental MSC that underscores the challenges in understanding their therapeutic potential in oncology.
- Published
- 2017
- Full Text
- View/download PDF
43. Amniotic MSCs reduce pulmonary fibrosis by hampering lung B-cell recruitment, retention, and maturation
- Author
-
Elsa Vertua, Marta Magatti, Ornella Parolini, I. Toschi, Francesca Romana Stefani, Antonietta Silini, Patrizia Bonassi Signoroni, Pietro Romele, V. Cesari, Anna Cargnoni, and Serafina Farigu
- Subjects
0301 basic medicine ,Pulmonary Fibrosis ,T-Lymphocytes ,Mice ,0302 clinical medicine ,Pulmonary fibrosis ,Settore BIO/13 - BIOLOGIA APPLICATA ,Lung ,Cell Aggregation ,lcsh:R5-920 ,B-Lymphocytes ,medicine.diagnostic_test ,bleomycin ,lcsh:Cytology ,FOXP3 ,Cell Differentiation ,General Medicine ,Lung Injury ,respiratory system ,medicine.anatomical_structure ,medicine.symptom ,Chemokines ,lcsh:Medicine (General) ,Antigen-Presenting Cells ,Inflammation ,Fetal and Neonatal Stem Cells ,Mesenchymal Stem Cell Transplantation ,03 medical and health sciences ,Immune system ,medicine ,Animals ,Humans ,Amnion ,lcsh:QH573-671 ,Antigen-presenting cell ,B cell ,business.industry ,Mesenchymal stem cell ,lung fibrosis ,Mesenchymal Stem Cells ,Cell Biology ,medicine.disease ,Lymphocyte Subsets ,030104 developmental biology ,Bronchoalveolar lavage ,amniotic mesenchymal stromal cells ,Cancer research ,business ,030217 neurology & neurosurgery ,Developmental Biology ,B lymphocytes - Abstract
Growing evidence suggests a mechanistic link between inflammation and the development and progression of fibrotic processes. Mesenchymal stromal cells derived from the human amniotic membrane (hAMSCs), which display marked immunomodulatory properties, have been shown to reduce bleomycin‐induced lung fibrosis in mice, possibly by creating a microenvironment able to limit the evolution of chronic inflammation to fibrosis. However, the ability of hAMSCs to modulate immune cells involved in bleomycin‐induced pulmonary inflammation has yet to be elucidated. Herein, we conducted a longitudinal study of the effects of hAMSCs on alveolar and lung immune cell populations upon bleomycin challenge. Immune cells collected through bronchoalveolar lavage were examined by flow cytometry, and lung tissues were used to study gene expression of markers associated with different immune cell types. We observed that hAMSCs increased lung expression of T regulatory cell marker Foxp3, increased macrophage polarization toward an anti‐inflammatory phenotype (M2), and reduced the antigen‐presentation potential of macrophages and dendritic cells. For the first time, we demonstrate that hAMSCs markedly reduce pulmonary B‐cell recruitment, retention, and maturation, and counteract the formation and expansion of intrapulmonary lymphoid aggregates. Thus, hAMSCs may hamper the self‐maintaining inflammatory condition promoted by B cells that continuously act as antigen presenting cells for proximal T lymphocytes in injured lungs. By modulating B‐cell response, hAMSCs may contribute to blunting of the chronicization of lung inflammatory processes with a consequent reduction of the progression of the fibrotic lesion., This study demonstrates that amniotic mesenchymal stromal cells reduce pulmonary fibrosis in bleomycin‐challenged mice by creating an anti‐inflammatory microenvironment mediated by their ability to control B‐cell recruitment, retention, maturation, and to reduce the formation/expansion of lymphoid aggregates in diseased lungs.
- Published
- 2020
44. 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, Ornella Parolini, Silini A.R., Di Pietro R., Lang-Olip I., Alviano F., Banerjee A., Basile M., Borutinskaite V., Eissner G., Gellhaus A., Giebel B., Huang Y.-C., Janev A., Kreft M.E., Kupper N., Abadia-Molina A.C., Olivares E.G., Pandolfi A., Papait A., Pozzobon M., Ruiz-Ruiz C., Soritau O., Susman S., Szukiewicz D., Weidinger A., Wolbank S., Huppertz B., and Parolini O.
- Subjects
0301 basic medicine ,Histology ,placenta ,lcsh:Biotechnology ,Cell ,Biomedical Engineering ,Medizin ,Consensus criteria ,Bioengineering ,Review ,cells ,consensus nomenclature ,derivatives ,fetal annexes ,perinatal ,tissues ,Biology ,Bioinformatics ,Cell morphology ,03 medical and health sciences ,0302 clinical medicine ,lcsh:TP248.13-248.65 ,Placenta ,medicine ,derivative ,Settore BIO/13 - BIOLOGIA APPLICATA ,fetal annexe ,Bioengineering and Biotechnology ,Human placenta ,cell ,3. Good health ,Clinical trial ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Identification (biology) ,Biotechnology - 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., Austrian Science Fund (FWF) DOC 31-B26, Medical University Graz, Universita Cattolica del Sacro Cuore, PRIN 2017 program of Italian Ministry of Research and University (MIUR) 2017RSAFK7, Ministry of Health, Italy GR-2018-12366992, Slovenian Research Agency - Slovenia P3-0108, MRIC UL IP-0510, Plan Estatal de Investigacion Cientifica y Tecnica y de Innovacion, ISCIII Subdireccion General de Evaluacion y Fomento de la Investigacion, Ministerio de Economia y Competitividad, Spain PI16/01642, European Union (EU), European Community (EC), German Research Foundation (DFG) GE-2223/2-1
- Published
- 2020
45. Assessment of the in vivo biofunctionality of a biomimetic hybrid scaffold for osteochondral tissue regeneration
- Author
-
Serena Bertoldi, Matilde Tschon, Silvia Brogini, Ornella Parolini, Annapaola Parrilli, Lucia Martini, Antonietta Silini, Milena Fini, Silvia Farè, Francesca Veronesi, and Gianluca Giavaresi
- Subjects
0106 biological sciences ,0301 basic medicine ,Cartilage, Articular ,Male ,Scaffold ,Bone Regeneration ,osteochondral regeneration ,Bioengineering ,Mesenchymal Stem Cell Transplantation ,01 natural sciences ,Applied Microbiology and Biotechnology ,calcium phosphate ,03 medical and health sciences ,Tissue engineering ,In vivo ,Biomimetic Materials ,010608 biotechnology ,Highly porous ,medicine ,Animals ,Humans ,Settore BIO/13 - BIOLOGIA APPLICATA ,Femur ,Bone regeneration ,pectin ,Tissue Scaffolds ,Chemistry ,Cartilage ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,human amniotic mesenchymal stromal cells ,Cells, Immobilized ,polyurethane scaffold ,030104 developmental biology ,medicine.anatomical_structure ,Heterografts ,Implant ,Rabbits ,Biotechnology ,Biomedical engineering - Abstract
Chondral and osteochondral lesions represent one of the most challenging problems in the orthopedic field, as these types of injuries lead to disability and worsened quality of life for patients and have an economic impact on the healthcare system. The aim of this in vivo study was to develop a new tissue engineering approach through a hybrid scaffold for osteochondral tissue regeneration made of porous polyurethane foam (PU) coated under vacuum with calcium phosphates (PU/VAC). Scaffold characterization showed a highly porous and interconnected structure. Human amniotic mesenchymal stromal cells (hAMSCs) were loaded into scaffolds using pectin (PECT) as a carrier. Osteochondral defects in medial femoral condyles of rabbits were created and randomly allocated in one of the following groups: plain scaffold (PU/VAC), scaffold with hAMSCs injected in the implant site (PU/VAC/hAMSC), scaffold with hAMSCs loaded in pectin (PU/VAC/PECT/hAMSC), and no treated defects (untreated). The therapeutic efficacy was assessed by macroscopic, histological, histomorphometric, microtomographic, and ultrastructural analyses at 3, 6, 12, and 24 weeks. Histological results showed that the scaffold was permissive to tissue growth and penetration, an immature osteocartilaginous tissue was observed at early experimental times, with a more accentuated bone regeneration in comparison with the cartilage layer in the absence of any inflammatory reaction.
- Published
- 2020
46. The Multifaceted Roles of MSCs in the Tumor Microenvironment: Interactions With Immune Cells and Exploitation for Therapy
- Author
-
Antonietta Rosa Silini, Andrea Papait, Marta Magatti, Anna Cargnoni, Ornella Parolini, and Francesca Romana Stefani
- Subjects
0301 basic medicine ,placenta ,Inflammation ,immunosurveillance ,Review ,medicine.disease_cause ,immunoediting ,Cell and Developmental Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,cancer ,tumor microenvironment ,Settore BIO/13 - BIOLOGIA APPLICATA ,lcsh:QH301-705.5 ,perinatal ,Tumor microenvironment ,business.industry ,Mesenchymal stem cell ,Cancer ,Cell Biology ,medicine.disease ,Immunosurveillance ,030104 developmental biology ,lcsh:Biology (General) ,Immunoediting ,inflammation ,030220 oncology & carcinogenesis ,Cancer research ,sense organs ,medicine.symptom ,Carcinogenesis ,business ,mesenchymal stromal cells ,Developmental Biology - 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
47. Autophagy is Activated In Vivo during Trimethyltin-Induced Apoptotic Neurodegeneration: A Study in the Rat Hippocampus
- Author
-
Aurora Del Fa, Ornella Parolini, Carlo Gangitano, Sabrina Ceccariglia, Fabrizio Michetti, and Alessandra Alvino
- Subjects
Autophagosome ,autophagy ,hippocampus ,Hippocampal formation ,Trimethyltin ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,TUNEL assay ,biology ,Chemistry ,Organic Chemistry ,Neurodegeneration ,Autophagy ,apoptosis ,neurodegeneration ,General Medicine ,medicine.disease ,Computer Science Applications ,Astrogliosis ,Cell biology ,Apoptosis ,Hippocampus ,lcsh:Biology (General) ,lcsh:QD1-999 ,nervous system ,biology.protein ,NeuN - Abstract
Trimethyltin (TMT) is an organotin compound known to produce significant and selective neuronal degeneration and reactive astrogliosis in the rodent central nervous system. Autophagy is the main cellular mechanism for degrading and recycling protein aggregates and damaged organelles, which in different stress conditions, such as starvation, generally improves cell survival. Autophagy is documented in several pathologic conditions, including neurodegenerative diseases. This study aimed to investigate the autophagy and apoptosis signaling pathways in hippocampal neurons of TMT-treated (Wistar) rats to explore molecular mechanisms involved in toxicant-induced neuronal injury. The microtubule-associated protein light chain (LC3, autophagosome marker) and sequestosome1 (SQSTM1/p62) (substrate of autophagy-mediated degradation) expressions were examined by Western blotting at different time points after intoxication. The results demonstrate that the LC3 II/I ratio significantly increased at 3 and 5 days, and that p62 levels significantly decreased at 7 and 14 days. Immunofluorescence images of LC3/neuronal nuclear antigen (NeuN) showed numerous strongly positive LC3 neurons throughout the hippocampus at 3 and 5 days. The terminal deoxynucleotidyltransferase dUTP nick end labeling (TUNEL) assay indicated an increase in apoptotic cells starting from 5 days after treatment. In order to clarify apoptotic pathway, immunofluorescence images of apoptosis-inducing factor (AIF)/NeuN did not show nuclear translocation of AIF in neurons. Increased expression of cleaved Caspase-3 was revealed at 5&ndash, 14 days in all hippocampal regions by Western blotting and immunohistochemistry analyses. These data clearly demonstrate that TMT intoxication induces a marked increase in both autophagy and caspase-dependent apoptosis, and that autophagy occurring just before apoptosis could have a potential role in neuronal loss in this experimental model of neurodegeneration.
- Published
- 2019
- Full Text
- View/download PDF
48. Perinatal Mesenchymal Stromal Cells and Their Possible Contribution to Fetal-Maternal Tolerance
- Author
-
Anna Cargnoni, Ornella Parolini, Francesca Romana Stefani, Andrea Papait, Marta Magatti, Antonietta Silini, and Alice Masserdotti
- Subjects
placenta ,Uterus ,Review ,Biology ,immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Fetus ,Antigen ,Pregnancy ,Placenta ,medicine ,Immune Tolerance ,Settore BIO/13 - BIOLOGIA APPLICATA ,Animals ,Humans ,lcsh:QH301-705.5 ,030304 developmental biology ,0303 health sciences ,Mechanism (biology) ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,General Medicine ,medicine.disease ,medicine.anatomical_structure ,lcsh:Biology (General) ,fetal-maternal tolerance ,Immunology ,mesenchymal stromal cells ,pregnancy ,Female ,030215 immunology - Abstract
During pregnancy, a successful coexistence between the mother and the semi-allogenic fetus occurs which requires a dynamic immune system to guarantee an efficient immune protection against possible infections and tolerance toward fetal antigens. The mechanism of fetal-maternal tolerance is still an open question. There is growing in vitro and in vivo evidence that mesenchymal stromal cells (MSC) which are present in perinatal tissues have a prominent role in generating a functional microenvironment critical to a successful pregnancy. This review highlights the immunomodulatory properties of perinatal MSC and their impact on the major immune cell subsets present in the uterus during pregnancy, such as natural killer cells, antigen-presenting cells (macrophages and dendritic cells), and T cells. Here, we discuss the current understanding and the possible contribution of perinatal MSC in the establishment of fetal-maternal tolerance, providing a new perspective on the physiology of gestation.
- Published
- 2019
49. Shaping the Future of Perinatal Cells: Lessons From the Past and Interpretations of the Present
- Author
-
Antonietta R. Silini, Alice Masserdotti, Andrea Papait, and Ornella Parolini
- Subjects
0301 basic medicine ,Histology ,lcsh:Biotechnology ,Cell ,human term placenta ,Biomedical Engineering ,regenerative medicine ,Bioengineering ,02 engineering and technology ,Biology ,immunomodulation ,Regenerative medicine ,Amniotic membrane ,Human term placenta ,Immunomodulation ,Perinatal tissues ,03 medical and health sciences ,Immune system ,lcsh:TP248.13-248.65 ,Placenta ,medicine ,Settore BIO/13 - BIOLOGIA APPLICATA ,perinatal tissues ,amniotic membrane ,Regeneration (biology) ,Mesenchymal stem cell ,Bioengineering and Biotechnology ,021001 nanoscience & nanotechnology ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,Perspective ,0210 nano-technology ,Neuroscience ,Function (biology) ,Biotechnology - Abstract
Since their discovery and characterization, mesenchymal stromal cells (MSC) have been a topic of great interest in regenerative medicine. Over the last 10 years, detailed studies investigated the properties of MSC from perinatal tissues and have indicated that these cells may represent important tools for restoring tissue damage or promoting regeneration and repair of the tissue microenvironment. At first, perinatal tissue-derived MSC drew attention due to their potential differentiation capacities suggested by their early embryological origin. It is nowadays accepted that perinatal tissue-derived MSC are promising for a wide range of regenerative medicine applications because of their unique immune modulatory properties, rather than their differentiation ability. As a matter of fact, the activation and function of various cells of the innate and adaptive immune systems are suppressed and modulated by MSC from different perinatal tissues, such as human term placenta. However, the mechanisms by which they act on immune cells to facilitate tissue repair during pathological processes remain to be thoroughly elucidated to develop safe and efficient therapeutic approaches. In addition to immune modulatory ability, several other peculiar characteristics of placenta MSC, less explored and/or more debated, are being investigated. These include an understanding of the anti-microbial properties and the role of placental MSC in tumor progression. Moreover, a thorough investigation on preparation methods, bioactive factors, mechanisms of action of the cell secretome, and the development of potency assays to predict clinical efficacy of placenta MSC and their products, are necessary to provide a solid basis for their clinical application.
- Published
- 2019
50. Antifibrotic Effects of Human Amniotic Membrane Transplantation in Established Biliary Fibrosis Induced in Rats
- Author
-
Ornella Parolini, Maria Martin Cruz, Raduan Hage, Luciana Barros Sant'Anna, Emilia Angela Lo Schiavo Arisawa, Anna Cargnoni, Maria Angélica Gargione Cardoso, and Nilson Sant'Anna
- Subjects
Liver Cirrhosis ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Cirrhosis ,medicine.medical_treatment ,Biomedical Engineering ,lcsh:Medicine ,Enzyme-Linked Immunosorbent Assay ,Bile Duct Diseases ,Liver transplantation ,Biology ,Proinflammatory cytokine ,Transforming Growth Factor beta1 ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,medicine ,Animals ,Humans ,Settore BIO/13 - BIOLOGIA APPLICATA ,Amnion ,Rats, Wistar ,Transplantation ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Liver Diseases ,lcsh:R ,Cell Biology ,Amniotic Membrane ,Bile Duct Ligation ,Digital image analysis ,ELISA ,Immunohistochemistry ,medicine.disease ,Interleukin-10 ,Rats ,030104 developmental biology ,Cytokine ,Liver ,030220 oncology & carcinogenesis ,Female ,Tumor necrosis factor alpha ,Bile Ducts ,Hepatic fibrosis - Abstract
Liver fibrosis is characterized by excessive accumulation of extracellular matrix components in the liver parenchyma that distorts the normal architecture and hepatic function. Progressive fibrosis could end in the advanced stage known as cirrhosis, resulting in the need to resort to liver transplantation. Amniotic membrane (AM) has emerged as an innovative therapeutic approach for chronic liver diseases due to its anti-inflammatory, antiscarring, and wound-healing effects. We have recently shown that AM can be used as a patch on the liver surface at the same time of fibrosis induction, resulting in significantly reduced progression and severity of biliary fibrosis. Here we investigated the effects of human AM on the established rat model of liver fibrosis, induced by the bile duct ligation (BDL). We also explored the effect of AM on the expression of transforming growth factor-β1 (TGF-β1), the main profibrogenic factor in hepatic fibrosis, and the proinflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and anti-inflammatory cytokine IL-10. Two weeks after BDL, the liver was covered with a fragment of AM or left untreated. Six weeks later, the fibrosis was first assessed by the semiquantitative Knodell and the METAVIR scoring systems and, thereafter, by CellProfiler digital image analysis to quantify the area occupied by collagen deposition, ductular reactions (DRs), activated myofibroblasts, and TGF-β1. The hepatic cytokines were determined by ELISA. AM-treated rats showed a significantly lower score compared to the control BDL rats (2.5 ± 0.9 vs. 3.5 ± 0.3, respectively; p < 0.05). The collagen deposition, DRs, number of activated myofibroblasts, and TGF-β1 were all reduced to about 50% of levels observed in untreated BDL rats. These findings suggest that AM, when applied as a patch onto the liver surface, is useful for treating well-established cholestatic fibrosis, and the mechanism was partly by means of downregulating the profibrotic factor TGF-β1 and IL-6.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.