Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Clin Microbiol Infect ; 24(6): 646-652, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29133154

ABSTRACT

OBJECTIVES: We aimed to report the first 54 cases of pregnant women infected by Zika virus (ZIKV) and their virologic and clinical outcomes, as well as their newborns' outcomes, in 2016, after the emergence of ZIKV in dengue-endemic areas of São Paulo, Brazil. METHODS: This descriptive study was performed from February to October 2016 on 54 quantitative real-time PCR ZIKV-positive pregnant women identified by the public health authority of São José do Rio Preto, São Paulo, Brazil. The women were followed and had clinical and epidemiologic data collected before and after birth. Adverse outcomes in newborns were analysed and reported. Urine or blood samples from newborns were collected to identify ZIKV infection by reverse transcription PCR (RT-PCR). RESULTS: A total of 216 acute Zika-suspected pregnant women were identified, and 54 had the diagnosis confirmed by RT-PCR. None of the 54 women miscarried. Among the 54 newborns, 15 exhibited adverse outcomes at birth. The highest number of ZIKV infections occurred during the second and third trimesters. No cases of microcephaly were reported, though a broad clinical spectrum of outcomes, including lenticulostriate vasculopathy, subependymal cysts, and auditory and ophthalmologic disorders, were identified. ZIKV RNA was detected in 18 of 51 newborns tested and in eight of 15 newborns with adverse outcomes. CONCLUSIONS: Although other studies have associated many newborn outcomes to ZIKV infection during pregnancy, these same adverse outcomes were rare or nonexistent in this study. The clinical presentation the newborns we studied was mild compared to other reports, suggesting that there is significant heterogeneity in congenital Zika infection.


Subject(s)
Fetal Diseases/virology , Pregnancy Complications, Infectious/virology , Zika Virus Infection/complications , Zika Virus/isolation & purification , Adult , Brazil , Female , Humans , Infant, Newborn , Phylogeny , Pregnancy , Young Adult , Zika Virus/classification , Zika Virus/genetics
2.
J Biomed Mater Res A ; 105(8): 2228-2240, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28426158

ABSTRACT

Autografts represent the gold standard for peripheral nerve reconstruction but their limited availability, the discrepancy of nerve caliber, and long surgical times are drawbacks. Allografts have therefore become a valid alternative option. In particular, acellular nerve allografts (ANAs) rather than fresh allografts do not need immunosuppression and appear to be safe and effective based on recent studies. An innovative method was conceived to obtain ANAs, so as to speed up nerve decellularization, without compromising nerve architecture, and without breaking the asepsis chain. Several detergent-based techniques, integrated with sonication and mechanical stirring, were tested in vitro on rabbit nerves, to identify, by microscopy and immunohistochemistry, the most effective protocol in terms of cell lysis and cellular debris clearance, while maintaining nerve architecture. Furthermore, a pilot in vivo study was performed: ANAs were implanted into tibial nerve defects of three rabbits, and autografts, representing the gold standard, in other three animals. Twelve weeks postoperatively, rabbits were clinically evaluated and euthanasized; grafts were harvested and microscopically and histomorphometrically analyzed. The method proved to be effective in vitro: the treatment removed axons, myelin and cells, without altering nerve architecture. The in vivo study did not reveal any adverse effect: animals maintained normal weight and function of posterior limb during the entire experimental time. A mild fibrotic reaction was observed, macrophages and leukocytes were rare or absent; ANAs regenerated fascicles and bundles were comparable versus autografts. Based on these results, this decellularization protocol is encouraging and deserves deeper investigations with further preclinical and clinical studies. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2228-2240, 2017.


Subject(s)
Guided Tissue Regeneration/methods , Nerve Regeneration , Peripheral Nerves/cytology , Peripheral Nerves/transplantation , Tissue Scaffolds , Allografts , Animals , Detergents/chemistry , Male , Peripheral Nerves/physiology , Peripheral Nerves/surgery , Rabbits , Sonication/methods , Tissue Scaffolds/chemistry , Transplantation, Homologous/methods
3.
Eur Cell Mater ; 32: 181-201, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27633707

ABSTRACT

The purpose of this work was to test, in vitro and in vivo, a new tissue-engineered construct constituted by porcine cortico-cancellous scaffold (Osteobiol Dual Block) (DB) and xeno-free ex vivo culture of human Periodontal Ligament Stem Cells (hPDLSCs). hPDLSCs cultured in xeno-free media formulation preserved the stem cells' morphological features, the expression of stemness and pluripotency markers, and their ability to differentiate into mesenchymal lineage. Transmission electron microscopy analysis suggested that after one week of culture, both noninduced and osteogenic differentiation induced cells joined and grew on DB secreting extracellular matrix (ECM) that in osteogenic induced samples was hierarchically assembled in fibrils. Quantitative RT-PCR (qRT-PCR) showed the upregulation of key genes involved in the bone differentiation pathway in both differentiated and undifferentiated hPDLSCs cultured with DB (hPDLSCs/DB). Functional studies revealed a significant increased response of calcium transients in the presence of DB, both in undifferentiated and differentiated cells stimulated with calcitonin and parathormone, suggesting that the biomaterial could drive the osteogenic differentiation process of hPDLSCs. These data were confirmed by the increase of gene expression of L-type voltage-dependent Ca2+ (VDCCL), subunits α1C and α2D1 in undifferentiated cells in the presence of DB. In vivo implantation of the hPDLSCs/DB living construct in the mouse calvaria evidenced a precocious osteointegration and vascularisation process. Our results suggest consideration of DB as a biocompatible, osteoinductive and osteoconductive biomaterial, making it a promising tool to regulate cell activities in biological environments and for a potential use in the development of new custom-made tissue engineering.


Subject(s)
Bone Regeneration/physiology , Periodontal Ligament/cytology , Stem Cells/cytology , Tissue Scaffolds/chemistry , Adult , Animals , Biomarkers/metabolism , Bone Regeneration/genetics , Calcification, Physiologic/genetics , Cell Proliferation , Cell Shape , Cells, Cultured , Flow Cytometry , Gene Expression Regulation , Humans , Male , Mice, Inbred C57BL , Osteogenesis/genetics , Stem Cells/ultrastructure , Sus scrofa , Young Adult
4.
J Biol Regul Homeost Agents ; 30(2): 409-20, 2016.
Article in English | MEDLINE | ID: mdl-27358127

ABSTRACT

Osteochondral lesions are considered a challenge for orthopedic surgeons. Currently, the treatments available are often unsatisfactory and unable to stimulate tissue regeneration. Tissue engineering offers a new therapeutic strategy, taking into account the role exerted by cells, biomaterial and growth factors in restoring tissue damage. In this light, Mesenchymal Stem Cells (MSCs) have been indicated as a fascinating tool for regenerative medicine thanks to their ability to differentiate into bone, cartilage and adipose tissue. However, in vitro-cultivation of MSCs could be associated with some risks such as de-differentiation/reprogramming, infection and contaminations of the cells. To overcome these shortcomings, a new approach is represented by the use of Bone Marrow Concentrate (BMC), that could allow the delivery of cells surrounded by their microenvironment in injured tissue. For this purpose, cells require a tridimensional scaffold that can support their adhesion, proliferation and differentiation. This study is focused on the potentiality of BMC seeded onto a hyaluronan-based scaffold (Hyaff-11) to differentiate into osteogenic lineage. This process depends on the specific interaction between cells derived from bone marrow (surrounded by their niche) and scaffold, that create an environment able to support the regeneration of damaged tissue. The data obtained from the present study demonstrate that BMC grown onto Hyaff-11 are able to differentiate toward osteogenic sense, producing specific osteogenic genes and matrix proteins.


Subject(s)
Bone Marrow Cells/cytology , Hyaluronic Acid/pharmacology , Osteogenesis/drug effects , Tissue Scaffolds , Adult , Cell Differentiation/drug effects , Collagen Type I/analysis , Female , Humans , Immunohistochemistry , Male , RNA, Messenger/analysis
5.
J Periodontal Res ; 51(1): 112-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26094874

ABSTRACT

BACKGROUND AND OBJECTIVE: In the present study, the early stages of in vitro bone formation in collagenated porcine scaffolds cultured with human periodontal ligament cells were investigated. The comparison between the osteogenic potential of this structure in basal and differentiating culture media was explored to predict the mechanism of its biological behavior as graft in human defect. Results were validated by synchrotron radiation X-Ray phase contrast computed microtomography (micro-CT). As the periodontal disease plays a key role in systemic and oral diseases, it is crucial to find advanced therapeutic clinical interventions to repair periodontal defects. This has been recently explored using cells and tissues developed in vitro that should ideally be immunologically, functionally, structurally and mechanically identical to the native tissue. MATERIAL AND METHODS: In vitro cultures of human periodontal ligament cells, easily obtained by scraping of alveolar crestal and horizontal fibers of the periodontal ligament, were seeded on to collagenated porcine blocks constituted by natural cancellous and cortical bone. 3D images were obtained by synchrotron radiation micro-CT and processed with a phase-retrieval algorithm based on the transport of intensity equation. RESULTS: Starting from the second week of culture, newly formed mineralized bone was detected in all the scaffolds, both in basal and differentiating media. Bone mineralization was proved to occur preferentially in the trabecular portion and in differentiating media. CONCLUSION: The chosen method, supported by phase contrast micro-CT analysis, successfully and quantitatively monitored the early stages of bone formation and the rate of the bioscaffold resorption in basal and differentiating culture media.


Subject(s)
Periodontal Ligament , Alveolar Process , Animals , Cells, Cultured , Humans , Osteogenesis , Stem Cells , Swine , Synchrotrons
6.
J Tissue Eng Regen Med ; 10(5): 374-91, 2016 05.
Article in English | MEDLINE | ID: mdl-23495253

ABSTRACT

Osteochondral lesions require treatment to restore the biology and functionality of the joint. A novel nanostructured biomimetic gradient scaffold was developed to mimic the biochemical and biophysical properties of the different layers of native osteochondral structure. The present results show that the scaffold presents important physicochemical characteristics and can support the growth and differentiation of mesenchymal stromal cells (h-MSCs), which adhere and penetrate into the cartilaginous and bony layers. H-MSCs grown in chondrogenic or osteogenic medium decreased their proliferation during days 14-52 on both scaffold layers and in medium without inducing factors used as controls. Both chondrogenic and osteogenic differentiation of h-MSCs occurred from day 28 and were increased on day 52, but not in the control medium. Safranin O staining and collagen type II and proteoglycans immunostaining confirmed that chondrogenic differentiation was specifically induced only in the cartilaginous layer. Conversely, von Kossa staining, osteocalcin and osteopontin immunostaining confirmed that osteogenic differentiation occurred on both layers. This study shows the specific potential of each layer of the biomimetic scaffold to induce chondrogenic or osteogenic differentiation of h-MSCs. These processes depended mainly on the media used but not the biomaterial itself, suggesting that the local milieu is fundamental for guiding cell differentiation. Copyright © 2013 John Wiley & Sons, Ltd.


Subject(s)
Biomimetic Materials/chemistry , Bone Regeneration , Cell Differentiation , Chondrogenesis , Mesenchymal Stem Cells/metabolism , Nanocomposites/chemistry , Antigens, Differentiation/biosynthesis , Humans , Mesenchymal Stem Cells/cytology
7.
Eur Cell Mater ; 28: 137-51; discussion 151, 2014 Sep 22.
Article in English | MEDLINE | ID: mdl-25241964

ABSTRACT

Despite the popularity of platelet-rich plasma (PRP) and platelet lysate (PL) in orthopaedic practice, the mechanism of action and the effectiveness of these therapeutic tools are still controversial. So far, the activity of PRP and PL has been associated with different growth factors (GF) released during platelet degranulation. This study, for the first time, identifies exosomes, nanosized vesicles released in the extracellular compartment by a number of elements, including platelets, as one of the effectors of PL activity. Exosomes were isolated from human PL by differential ultracentrifugation, and analysed by electron microscopy and Western blotting. Bone marrow stromal cells (MSC) treated with three different exosome concentrations (0.6 µg, 5 µg and 50 µg) showed a significant, dose-dependent increase in cell proliferation and migration compared to the control. In addition, osteogenic differentiation assays demonstrated that exosome concentration differently affected the ability of MSC to deposit mineralised matrix. Finally, the analysis of exosome protein content revealed a higher amount of basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF-BB) and transforming growth factor beta 1 (TGF-ß1) as compared to PL. In regards to RNA content, an enrichment of small RNAs in exosomes as compared to donor platelets has been found. These results suggest that exosomes consistently contribute to PL activity and could represent an advantageous nanodelivery system for cell-free regeneration therapies.


Subject(s)
Blood Platelets/chemistry , Cell Extracts/chemistry , Exosomes/chemistry , Mesenchymal Stem Cells/drug effects , Becaplermin , Cell Differentiation , Cell Extracts/pharmacology , Cell Proliferation , Exosomes/ultrastructure , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/pharmacology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , MicroRNAs/analysis , Proto-Oncogene Proteins c-sis/analysis , Proto-Oncogene Proteins c-sis/pharmacology , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/pharmacology , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/pharmacology
8.
J Biol Regul Homeost Agents ; 25(2 Suppl): S3-13, 2011.
Article in English | MEDLINE | ID: mdl-22051166

ABSTRACT

The requirements for a successful regeneration of an osteo-chondral defect could effectively be met by using a bi-layered composite scaffold, able to support proliferation and differentiation of mesenchymal stem cells, while providing a biochemical environment promoting the formations of the two distinct tissues. The novel strategy here presented consists of developing a bio-mimetic scaffolds obtained by the combination of two integrated organic compounds (type I collagen and chitosan) with or without bioactive Mg-doped hydroxyapatite (Mg-HA) nanocrystals, depending on the specific layer, reproducing cartilaginous or subchondral bone tissue. An innovative patented methodology for scaffolds production, called - pH-dependent 3-phasic assembling -, allowed to development of a highly homogenous and chemically stable scaffold, presenting a very good integration among all three components, as confirmed by extensive SEM and thermogravimetric analyses. A preliminary in vitro evaluation was also carried out by seeding bi-layered scaffold with human bone marrow stromal cells (h-MSCs), by giving particular emphasis to cell viability and distribution at day 0, 7 and 14. Cells were viable and uniformly colonized the whole scaffold until day 14, indicating that the scaffold contributed to the maintenance of cell behaviour.


Subject(s)
Biomimetic Materials/chemistry , Bone Marrow Cells/cytology , Bone Regeneration , Cartilage , Materials Testing , Tissue Scaffolds/chemistry , Bone Marrow Cells/metabolism , Bone Substitutes/chemistry , Cells, Cultured , Chitosan/chemistry , Collagen Type I/chemistry , Durapatite/chemistry , Humans , Stromal Cells/cytology , Stromal Cells/metabolism
9.
J Biol Regul Homeost Agents ; 25(2 Suppl): S53-62, 2011.
Article in English | MEDLINE | ID: mdl-22051171

ABSTRACT

Association of biomaterials with autologous cells can provide a new generation of implantable devices for cartilage repair. An ideal scaffold should possess a preformed three-dimensional shape, fix the cells to the damaged area and prevent their migration into the articular cavity. Furthermore, the constructs should have sufficient mechanical strength to facilitate handling in a clinical setting and stimulate the uniform spreading of cells and a phenotype re-differentiation process. The aim of this study was to verify the ability of an equine collagen membrane to support the growth of human chondrocytes and to allow the re-expression of their original phenotype. This ability was assessed by the evaluation of collagen type I, II and aggrecan mRNA expression by Real-Time PCR. Immunohistochemical analyses were performed to evaluate collagen type I, II and proteoglycans synthesis. Electron microscopy was utilized to highlight the structure of the biomaterial and its interactions with the cells. Our data indicate that human chondrocytes seeded onto a collagen membrane express and produce collagen type II and aggrecan and downregulate the production of collagen type I during the experimental times analyzed. These results provide an in vitro demonstration for the therapeutic potential of autologous chondrocyte transplantation by an equine collagen membrane as a delivery vehicle in a tissue-engineered approach towards the repair of articular cartilage defects.


Subject(s)
Cartilage, Articular , Chondrocytes/metabolism , Collagen Type II/chemistry , Collagen Type I/chemistry , Materials Testing , Membranes, Artificial , Tissue Scaffolds/chemistry , Adult , Aggrecans/biosynthesis , Animals , Cells, Cultured , Chondrocytes/cytology , Collagen Type I/biosynthesis , Collagen Type II/biosynthesis , Female , Humans , Male , RNA, Messenger/biosynthesis , Sheep , Tissue Engineering
10.
Curr Cancer Drug Targets ; 10(7): 649-59, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20578992

ABSTRACT

Bone metastases contribute to morbidity in patients with common cancers, and conventional therapy provides only palliation and can induce systemic side effects. The development of nanostructured delivery systems that combine carriers with bone-targeting molecules can potentially overcome the drawbacks presented by conventional approaches. We have recently developed biodegradable, biocompatible nanoparticles (NP) made of a conjugate between poly (D,L-lactide-co-glycolic) acid and alendronate, suitable for systemic administration, and directly targeting the site of tumor-induced osteolysis. Here, we loaded NP with doxorubicin (DXR), and analyzed the in vitro and in vivo activity of the drug encapsulated in the carrier system. After confirming the intracellular uptake of DXR-loaded NP, we evaluated the anti-tumor effects in a panel of human cell lines, representative for primary or metastatic bone tumors, and in an orthotopic mouse model of breast cancer bone metastases. In vitro, both free DXR and DXR-loaded NP, (58-580 ng/mL) determined a significant dose-dependent growth inhibition of all cell lines. Similarly, both DXR-loaded NP and free DXR reduced the incidence of metastases in mice. Unloaded NP were ineffective, although both DXR-loaded and unloaded NP significantly reduced the osteoclast number at the tumor site (P = 0.014, P = 0.040, respectively), possibly as a consequence of alendronate activity. In summary, NP may act effectively as a delivery system of anticancer drugs to the bone, and deserve further evaluation for the treatment of bone tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Carcinoma/secondary , Doxorubicin/administration & dosage , Nanocapsules , Acid Phosphatase/metabolism , Alendronate/chemistry , Alendronate/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biological Transport , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/ultrastructure , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/ultrastructure , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Female , Humans , Isoenzymes/metabolism , Mice , Mice, Nude , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteolysis/diagnostic imaging , Osteolysis/prevention & control , Radiography , Tartrate-Resistant Acid Phosphatase , Xenograft Model Antitumor Assays
11.
J Biomed Mater Res A ; 87(4): 986-93, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18257082

ABSTRACT

Recent studies have shown that mesenchymal stem cells obtained from periodontal ligament (PDL-MSCs) are multipotent cells that have similar features of the bone marrow and dental pulp MSCs and are capable of proliferating and producing different types of tissue such as bone and tooth associated-tissues. Human PDL-MSCs expanded ex vivo were induced to osteogenesis, seeded in three-dimensional biocompatible scaffolds (fibrin sponge, bovine-derived substitutes) and examined using light, scanning and transmission electron microscopy. Morphological observations showed extensive growth of cellular biomass partially covering the scaffolds after 4 weeks of incubation in mineralization medium. These findings indicate that periodontal ligament can be an easily and efficient autologous source of stem cells with a high expansion capacity and ability to differentiate in osteogenic cells that can colonize and grow connected to bio-compatible scaffold. It can be suggested that the use of PDL-MSCs for generating graft biomaterials is advantageous for bone tissue engineering in regenerative dentistry.


Subject(s)
Biocompatible Materials/chemistry , Periodontal Ligament/cytology , Regeneration/physiology , Stem Cells/physiology , Adult , Alkaline Phosphatase/metabolism , Animals , Cattle , Cell Differentiation/physiology , Cells, Cultured , Humans , Materials Testing , Periodontal Ligament/physiology , Stem Cells/cytology , Tissue Engineering/methods , Young Adult
12.
J Dent Res ; 84(8): 747-51, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16040734

ABSTRACT

One of the most commonly observed adverse effects of cyclosporin A (CsA) is the development of gingival overgrowth (GO). Fibroblasts are involved in GO, but the question why only a percentage of patients undergoing CsA treatment shows this side-effect remains unanswered. In a previous study, CsA has been demonstrated to induce over-expression of phospholipase C (PLC) beta(1) in fibroblasts of patients with clinical GO, in cells from both enlarged and clinically healthy gingival sites. In this work, we assessed the expression of PLCbeta isoforms to investigate whether the exaggerated fibroblast response to CsA related to increased PLCbeta(1) expression could also be detected in CsA-treated patients without clinical signs of GO. Our results support the hypothesis of a multi-factorial origin of gingival overgrowth, including specific changes within the gingival tissues orchestrating fibroblastic hyper-responsiveness as a consequence of a long-term in vivo exposure to cyclosporin A.


Subject(s)
Cell Nucleus/enzymology , Cyclosporine/adverse effects , Fibroblasts/enzymology , Gingival Overgrowth/enzymology , Immunosuppressive Agents/adverse effects , Isoenzymes/biosynthesis , Type C Phospholipases/biosynthesis , Adult , Blotting, Western , Case-Control Studies , Cells, Cultured , Enzyme Induction , Fibroblasts/drug effects , Genetic Predisposition to Disease , Gingiva/drug effects , Gingiva/enzymology , Gingival Overgrowth/chemically induced , Gingival Overgrowth/genetics , Heart Transplantation , Humans , Male , Microscopy, Electron, Transmission , Middle Aged , Phospholipase C beta , Statistics, Nonparametric
13.
Int J Oncol ; 22(5): 1147-53, 2003 May.
Article in English | MEDLINE | ID: mdl-12684684

ABSTRACT

Inositol lipid cycle, among the pletora of signalling events, is directly involved in cell growth. It is located both in the cytoplasm and in the nucleus. Disturbances may cause uncontrolled proliferation of the cell and ultimately cancer. The phosphatidyl inositol phospolipase C (PLC) is a key enzyme in the hydrolysis of polyphosphoinositides (PIs) and could be differently involved in the normal and pathological cell growth. We report immunochemical and immunocytochemical demonstrations that the PLC isoforms are present in both cytoplasmic and nuclear compartments of low and fast proliferating hepatoma cells. The PLC activity is increased in fast proliferating cells, in which PLC delta1 and to a greater extent PLC delta4 are more expressed at cytosolic level, suggesting an involvement of PI specific PLCs in the progression of cell cycle and in the control of cell proliferation and possibly of neoplastic cell growth.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cell Division/physiology , Liver Neoplasms/enzymology , Type C Phospholipases/metabolism , Animals , Biomarkers, Tumor , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/ultrastructure , Immunohistochemistry , Isoenzymes/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/ultrastructure , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/ultrastructure , Microscopy, Confocal , Phosphatidylinositol Diacylglycerol-Lyase , Rats , Substrate Specificity , Tumor Cells, Cultured
14.
Chir Organi Mov ; 88(4): 363-7, 2003.
Article in English, Italian | MEDLINE | ID: mdl-15259552

ABSTRACT

Various techniques are widely used to repair bone defects, association of hyaluronan-based biodegradable polymers (Hyaff-11) with bone marrow stromal cells (BMSC) promises to provide successful cell scaffolds for tissue-engineered repair of bone tissue. We evaluate in vitro and in vivo the potential of Hyaff-11 to facilitate mineralization of BMSC. Rat BMSC were seeded on Hyaff-11 and their differentiation were assessed at different time points. Osteogenic differentiation was investigated in vitro analysing the expression of alkaline phosphatase and osteocalcin. Mineralization of bone defects was evaluated also in vivo implanting Hyaff-11 scaffold combined with BMSC in large segmental radius defects. In vitro, we found a decrease expression of alkaline phosphatase and an increase of osteocalcin. In vivo, our data showed that mineralization was induced and basic fibroblast growth factor contributed to this process. These results provide a demonstration to therapeutic potential of Hyaff-11 as appropriate carrier vehicle for differentiation and mineralization of BMSC and for the repair of bone defects.


Subject(s)
Bone Marrow Cells , Calcification, Physiologic , Hyaluronic Acid/analogs & derivatives , Stromal Cells , Animals , Rats , Rats, Inbred F344
15.
Arch Virol ; 147(6): 1247-56, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12111433

ABSTRACT

Human Cytomegalovirus (HCMV) UL73 encodes for a polymorphic structural glycoprotein, gpUL73(gN), conserved among herpesviruses. This study analyzed the intracellular and intraviral localization of gpUL73 by immunoelectron-microscopy comparing the reactivity of two different antibodies. We found that gN is an envelope component of the mature viral particle with at least a portion exposed at the virus surface and another at the internal side of the envelope. Furthermore, gpUL73 is also present in the matrix of dense bodies and "black holes". These results, as well as immunoblotting analysis, suggest that the two antibodies recognize different forms, fully processed or unprocessed, of gpUL73-gN.


Subject(s)
Cytomegalovirus/chemistry , Cytoplasm/chemistry , Viral Envelope Proteins/analysis , Antibodies, Viral/immunology , Blotting, Western , Cell Line , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Cytomegalovirus/ultrastructure , Cytoplasm/ultrastructure , Cytoplasm/virology , Cytoplasmic Vesicles/chemistry , Cytoplasmic Vesicles/ultrastructure , Cytoplasmic Vesicles/virology , Humans , Microscopy, Immunoelectron , Viral Envelope Proteins/immunology , Virion/chemistry , Virion/ultrastructure
16.
J Gen Virol ; 83(Pt 5): 1005-1012, 2002 May.
Article in English | MEDLINE | ID: mdl-11961254

ABSTRACT

Human cytomegalovirus (HCMV) UL53 belongs to a family of conserved herpesvirus genes. In this work, the expression and localization of the UL53 gene product was analysed. Results obtained showed that pUL53 is a new structural protein. In infected human fibroblasts, pUL53 localizes in cytoplasmic perinuclear granular formations together with other structural viral proteins. In the nucleus, pUL53 forms patches at the nuclear periphery and co-localizes with lamin B at the internal nuclear membrane level. Immunoelectron microscopy studies have disclosed that nuclear pseudo-inclusions are labelled, whereas nucleocapsid formations within the intranuclear skein are negative. Furthermore, the mature virus particle maintains pUL53 at its tegumental level. These data suggest that pUL53 could be involved either in nucleocapsid maturation or in the egress of nucleocapsids from the nucleus to the cytoplasm through the nuclear membrane, a role compatible with the function hypothesized for UL31, its positional homologue in herpes simplex virus type 1.


Subject(s)
Cytomegalovirus/chemistry , Viral Structural Proteins/analysis , Humans , Microscopy, Immunoelectron , Viral Structural Proteins/physiology , Virion/chemistry
17.
Connect Tissue Res ; 42(1): 49-58, 2001.
Article in English | MEDLINE | ID: mdl-11696988

ABSTRACT

Bone marrow stromal cells (BMSCs) for osteoblast differentiation studies can be obtained by gradient isolation techniques or by directly plating a filtered cell suspension. We compared these two procedures to evaluate whether this step is critical in order to obtain a high number of differentiated colonies. Isolated primary rat BMSCs were cultured in vitro with or without insulin-like growth factor II (IGFII), basic fibroblast growth factor (b-FGF), epidermal growth factor (EGF) or transforming growth factor beta 1 (TGF beta 1), and histochemically and biochemically analysed at different time points. The gradient procedure produced a significantly higher number of colonies capable of osteoblastic differentiation. The growth factors had different effects. In particular, b-FGF and EGF significantly increased the number of Alizarin red S positive colonics, while IGFII and TGF beta I exerted inhibitory effects. Nodules obtained on day 21 showed some alkaline phosphatase positive cells and were Von Kossa-positive. These data demonstrate that more differentiated colonies are obtainable from BMSCs isolated by the gradient procedure.


Subject(s)
Bone Marrow Cells/cytology , Cell Separation/methods , Centrifugation, Density Gradient/methods , Osteoblasts/cytology , Stromal Cells/cytology , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/drug effects , Cell Count , Cell Differentiation , Cells, Cultured , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Insulin-Like Growth Factor II/pharmacology , Male , Rats , Rats, Inbred F344 , Stromal Cells/drug effects , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
18.
J Cell Physiol ; 189(1): 34-44, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11573202

ABSTRACT

It has been recently reported that retinoblastoma family proteins suppress cell growth by regulating not only E2F-dependent mRNA transcription but also rRNA and tRNA transcription and, through HDAC1 recruitment, chromatin packaging. In the present study we report data showing that these various control strategies are correlated, at least in part, with nuclear compartmentalization of retinoblastoma proteins. In a first series of experiments, we showed that pRb2/p130 and p107 are not evenly distributed within the nucleus and that cell cycle-dependent binding with E2F4 changes also as a function of their subnuclear localization. Namely, in the nucleoplasm pRb2/p130-E2F4 complexes are more numerous during G0/G1 while in the nucleolus they increase in S phase. Partially different functions for p107 are suggested since p107-E2F4 complexes in the nucleoplasm are more numerous is S phase with respect to G0/G1 and no cell cycle change is observed in the nucleolus. In a second series of experiments we showed that pRb2/p130, p107, E2F4, and pRb2/p130-HDAC1 complexes are all inner nuclear matrix-associated proteins and localize to sites different from pRb/p105 ones. We provide further evidence of multiple and partially distinct retinoblastoma protein family functional roles during cell cycle. Moreover, our data support emerging evidence for functional interrelationships between nuclear structure and gene expression.


Subject(s)
Cell Nucleus/metabolism , Nuclear Proteins/physiology , Phosphoproteins/physiology , Proteins , Cell Compartmentation , Cell Cycle , Cell Division , Cell Nucleus/ultrastructure , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , E2F4 Transcription Factor , Histone Deacetylase 1 , Histone Deacetylases/immunology , Histone Deacetylases/metabolism , Humans , Macromolecular Substances , Microscopy, Immunoelectron , Models, Biological , Nuclear Matrix/metabolism , Phosphoproteins/immunology , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Transcription Factors/immunology , Transcription Factors/metabolism , Tumor Cells, Cultured
19.
Arthritis Rheum ; 44(8): 1800-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11508432

ABSTRACT

OBJECTIVE: To investigate the in vitro effect of therapeutic hyaluronan (HA) of 500-730 kd on anti-Fas-induced apoptosis of chondrocytes from osteoarthritis (OA) patients, and to assess its mechanism of action by analyzing the role of the 2 HA receptors, CD44 and CD54 (intercellular adhesion molecule 1 [ICAM-1]). METHODS: Chondrocytes isolated from human OA knee cartilage were cultured and the effect of HA on both spontaneous and anti-Fas-induced apoptosis was evaluated. Apoptosis was analyzed by JAM test (for quantitative analysis of fragmented DNA), cell death detection immunoassay (for quantitative analysis of oligonucleosome), TUNEL assay, and electron microscopy. Blocking experiments with anti-CD44 and anti-CD54 alone or in combination were performed to investigate the HA mechanism of action. RESULTS: Both quantitative tests demonstrated that anti-Fas significantly induced apoptosis of isolated OA chondrocytes. HA at 1,000 microg/ml significantly reduced the anti-Fas-induced apoptosis of chondrocytes but did not affect spontaneous chondrocyte apoptosis. These data were also confirmed by TUNEL staining and by electron microscopy morphologic evaluation. The antiapoptotic effects of HA on anti-FAS-induced chondrocyte apoptosis were significantly decreased by both anti-CD44 (mean +/- SD 57 +/- 12% inhibition) and anti-ICAM-1 (31 +/- 22% inhibition). The mixture of the 2 antibodies had an additive effect, since the rate of inhibition increased to 87 +/- 13%. CONCLUSION: These data demonstrate that 500-730-kd HA exerts an antiapoptotic effect on anti-FAS-induced chondrocyte apoptosis by binding its specific receptors (CD44 and ICAM-1). Furthermore, this HA fraction may be able to slow down chondrocyte apoptosis in OA by regulating the processes of cartilage matrix degradation.


Subject(s)
Apoptosis/drug effects , Chondrocytes/pathology , Hyaluronan Receptors/physiology , Hyaluronic Acid/pharmacology , Intercellular Adhesion Molecule-1/physiology , Osteoarthritis/pathology , fas Receptor/physiology , Aged , Antibodies/immunology , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Chromatin/ultrastructure , Female , Humans , In Situ Nick-End Labeling , Male , fas Receptor/immunology
20.
Biomaterials ; 22(15): 2095-105, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11432589

ABSTRACT

A biodegradable non-woven hyaluronic acid polymer scaffold (Hyaff 11) was analysed in vitro as a carrier vehicle for differentiation and mineralization of rat bone marrow stromal cells (BMSC). BMSC were grown on Hyaff 11 in a mineralizing medium in the presence/absence of basic fibroblast growth factor (bFGF). Osteoblastic differentiation was investigated by light and electron microscopy analysing the expression of osteogenic markers: calcium, alkaline phosphatase (AP), osteopontin (OP), bone sialoprotein (BSP) and collagen type 1. We also measured proliferation, AP activity and mRNA expression of AP and osteocalcin (OC). Electron microscopy and Toluidine-blue staining demonstrated that bFGF accelerated (day 20 vs. day 40) and increased mineralization. With bFGF, calcium, OP and BSP were strongly enhanced at day 40, whereas AP decreased. Our in vitro results demonstrate that Hyaff 11 is a useful vehicle for growth, differentiation and mineralization of rat BMSC, and that it permits bone development.


Subject(s)
Bone Marrow Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Hyaluronic Acid/chemistry , Polymers/chemistry , Stromal Cells/cytology , Alkaline Phosphatase/metabolism , Animals , Biocompatible Materials/chemistry , Calcium/metabolism , Cell Culture Techniques/methods , Cell Division , Cells, Cultured , Collagen Type I/metabolism , Coloring Agents/pharmacology , Culture Media , Integrin-Binding Sialoprotein , Kinetics , Microscopy, Electron , Osteoblasts/cytology , Osteocalcin/metabolism , Osteopontin , Protein Binding , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Sialoglycoproteins/metabolism , Time Factors , Tolonium Chloride/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...