Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 15(2): 169, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395995

ABSTRACT

Phosphodiesterase 2A (Pde2A) is a dual-specific PDE that breaks down both cAMP and cGMP cyclic nucleotides. We recently highlighted a direct relationship between Pde2A impairment, a consequent increase of cAMP, and the appearance of mouse congenital heart defects (CHDs). Here we aimed to characterize the pathways involved in the development of CHDs and in their prevention by pharmacological approaches targeting cAMP and cGMP signaling. Transcriptome analysis revealed a modulation of more than 500 genes affecting biological processes involved in the immune system, cardiomyocyte development and contractility, angiogenesis, transcription, and oxidative stress in hearts from Pde2A-/- embryos. Metoprolol and H89 pharmacological administration prevented heart dilatation and hypertabeculation in Pde2A-/- embryos. Metoprolol was also able to partially impede heart septum defect and oxidative stress at tissue and molecular levels. Amelioration of cardiac defects was also observed by using the antioxidant NAC, indicating oxidative stress as one of the molecular mechanisms underpinning the CHDs. In addition, Sildenafil treatment recovered cardiac defects suggesting the requirement of cAMP/cGMP nucleotides balance for the correct heart development.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2 , Heart Defects, Congenital , Mice , Animals , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Metoprolol , Signal Transduction , Cyclic GMP/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/prevention & control , Oxidative Stress
2.
Nucleic Acids Res ; 52(8): 4167-4184, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38324473

ABSTRACT

Sam68 and SLM2 are paralog RNA binding proteins (RBPs) expressed in the cerebral cortex and display similar splicing activities. However, their relative functions during cortical development are unknown. We found that these RBPs exhibit an opposite expression pattern during development. Sam68 expression declines postnatally while SLM2 increases after birth, and this developmental pattern is reinforced by hierarchical control of Sam68 expression by SLM2. Analysis of Sam68:Slm2 double knockout (Sam68:Slm2dko) mice revealed hundreds of exons that respond to joint depletion of these proteins. Moreover, parallel analysis of single and double knockout cortices indicated that exons regulated mainly by SLM2 are characterized by a dynamic splicing pattern during development, whereas Sam68-dependent exons are spliced at relatively constant rates. Dynamic splicing of SLM2-sensitive exons is completely suppressed in the Sam68:Slm2dko developing cortex. Sam68:Slm2dko mice die perinatally with defects in neurogenesis and in neuronal differentiation, and develop a hydrocephalus, consistent with splicing alterations in genes related to these biological processes. Thus, our study reveals that developmental control of separate Sam68 and Slm2 paralog genes encoding homologous RBPs enables the orchestration of a dynamic splicing program needed for brain development and viability, while ensuring a robust redundant mechanism that supports proper cortical development.


Subject(s)
Cerebral Cortex , Mice, Knockout , RNA Splicing , RNA-Binding Proteins , Animals , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Mice , Exons/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Neurogenesis/genetics , Gene Expression Regulation, Developmental , Neurons/metabolism
4.
EBioMedicine ; 89: 104453, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36736132

ABSTRACT

BACKGROUND: Keratitis ichthyosis deafness (KID) syndrome is a rare disorder caused by hemichannel (HC) activating gain-of-function mutations in the GJB2 gene encoding connexin (Cx) 26, for which there is no cure, or current treatments based upon the mechanism of disease causation. METHODS: We applied Adeno Associated Virus (AAV) mediated mAb gene transfer (AAVmAb) to treat the epidermal features of KID syndrome with a well-characterized HC blocking antibody using male mice of a murine model that replicates the skin pathology of the human disease. FINDINGS: We demonstrate that in vivo AAVmAb treatment significantly reduced the size and thickness of KID lesions, in addition to blocking activity of mutant HCs in the epidermis in vivo. We also show that AAVmAb treatment eliminated abnormal keratinocyte proliferation and enlarged cell size, decreased apoptosis, and restored the normal distribution of keratin expression. INTERPRETATION: Our findings reinforce the critical role played by increased HC activity in the skin pathology associated with KID syndrome. They also underscore the clinical potential of anti-HC mAbs coupled with genetic based delivery systems for treating the underlying mechanistic basis of this disorder. Inhibition of HC activity is an ideal therapeutic target in KID syndrome, and the genetic delivery of mAbs targeted against mutant HCs could form the basis of new therapeutic interventions to treat this incurable disease. FUNDING: Fondazione Telethon grant GGP19148 and University of Padova grant Prot. BIRD187130 to FM; Foundation for Ichthyosis and Related Skin Types (FIRST) and National Institutes of Health grant EY 026911 to TWW.


Subject(s)
Connexins , Deafness , Ichthyosis , Keratitis , Animals , Male , Mice , Antibodies , Connexins/genetics , Deafness/genetics , Epidermis/metabolism , Gene Transfer Techniques , Ichthyosis/genetics , Ichthyosis/metabolism , Ichthyosis/pathology , Keratitis/genetics , Keratitis/metabolism , Keratitis/pathology , Mutation
5.
Cancers (Basel) ; 14(24)2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36551679

ABSTRACT

Glioblastoma (GBM), the most malignant primary brain tumor in adults. Although not frequent, it has a relevant social impact because the peak incidence coincides with the age of professional maturity. A number of novel treatments have been proposed, yet clinical trials have been disappointing. Recently, a phase II clinical trial (REGOMA) demonstrated that the multikinase inhibitor regorafenib significantly increased the median overall survival (OS) of GBM patients when compared to lomustine-treated patients. On this basis, the National Comprehensive Cancer Network (NCCN) 2020 Guidelines included regorafenib as a preferred regimen in relapsed GBM treatment. Despite the use in GBM patients' therapy, little is known about the molecular mechanisms governing regorafenib effectiveness on the GBM tumor. Here we report an in vitro characterization of GBM tumor cells' response to regorafenib, performed both on cell lines and on patient-derived glioma stem cells (GSCs). Overall, regorafenib significantly reduced cell growth of 2D tumor cell cultures and of 3D tumor spheroids. Strikingly, this effect was accompanied by transcriptional regulation of epithelial to mesenchymal transition (EMT) genes and by an increased ability of surviving tumor cells to invade the surrounding matrix. Taken together, our data suggest that regorafenib limits cell growth, however, it might induce an invasive phenotype.

6.
Sci Rep ; 12(1): 22535, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36581671

ABSTRACT

Autism Spectrum Disorder (ASD) is a pervasive neurodevelopmental disorder emerging in early life characterized by impairments in social interaction, poor verbal and non-verbal communication, and repetitive patterns of behaviors. Among the best-known genetic risk factors for ASD, there are mutations causing the loss of the Fragile X Messenger Ribonucleoprotein 1 (FMRP) leading to Fragile X syndrome (FXS), a common form of inherited intellectual disability and the leading monogenic cause of ASD. Being a pivotal regulator of motor activity, motivation, attention, and reward processing, dopaminergic neurotransmission has a key role in several neuropsychiatric disorders, including ASD. Fmr1 Δexon 8 rats have been validated as a genetic model of ASD based on FMR1 deletion, and they are also a rat model of FXS. Here, we performed behavioral, biochemical and in vivo SPECT neuroimaging experiments to investigate whether Fmr1 Δexon 8 rats display ASD-like repetitive behaviors associated with changes in striatal dopamine transporter (DAT) availability assessed through in vivo SPECT neuroimaging. At the behavioral level, Fmr1 Δexon 8 rats displayed hyperactivity in the open field test in the absence of repetitive behaviors in the hole board test. However, these behavioral alterations were not associated with changes in striatal DAT availability as assessed by non-invasive in vivo SPECT and Western blot analyses.


Subject(s)
Autism Spectrum Disorder , Fragile X Mental Retardation Protein , Animals , Rats , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/genetics , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Fragile X Mental Retardation Protein/genetics
7.
Cancers (Basel) ; 13(20)2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34680212

ABSTRACT

In this study, we used B16-F10 cells grown in the dorsal skinfold chamber (DSC) preparation that allowed us to gain optical access to the processes triggered by photodynamic therapy (PDT). Partial irradiation of a photosensitized melanoma triggered cell death in non-irradiated tumor cells. Multiphoton intravital microscopy with genetically encoded fluorescence indicators revealed that bystander cell death was mediated by paracrine signaling due to adenosine triphosphate (ATP) release from connexin (Cx) hemichannels (HCs). Intercellular calcium (Ca2+) waves propagated from irradiated to bystander cells promoting intracellular Ca2+ transfer from the endoplasmic reticulum (ER) to mitochondria and rapid activation of apoptotic pathways. Combination treatment with S-nitrosoglutathione (GSNO), an endogenous nitric oxide (NO) donor that biases HCs towards the open state, greatly potentiated anti-tumor bystander killing via enhanced Ca2+ signaling, leading to a significant reduction of post-irradiation tumor mass. Our results demonstrate that HCs can be exploited to dramatically increase cytotoxic bystander effects and reveal a previously unappreciated role for HCs in tumor eradication promoted by PDT.

8.
Brain Sci ; 11(6)2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34199780

ABSTRACT

Acquisition of detailed anatomical and molecular knowledge from intact biological samples while preserving their native three-dimensional structure is still a challenging issue for imaging studies aiming to unravel a system's functions. Three-dimensional micro-CT X-ray imaging with a high spatial resolution in minimally perturbed naive non-transparent samples has recently gained increased popularity and broad application in biomedical research. Here, we describe a novel X-ray-based methodology for analysis of ß-galactosidase (lacZ) reporter-driven gene expression in an intact murine brain ex vivo by micro-CT. The method relies on detection of bromine molecules in the product of the enzymatic ß-galactosidase reaction. Enhancement of the X-ray signal is observed specifically in the regions of the murine brain where expression of the lacZ reporter gene is also detected histologically. We performed quantitative analysis of the expression levels of lacZ reporter activity by relative radiodensity estimation of the ß-galactosidase/X-gal precipitate in situ. To demonstrate the feasibility of the method, we performed expression analysis of the Tsen54-lacZ reporter gene in the murine brain in a semi-quantitative manner. Human mutations in the Tsen54 gene cause pontocerebellar hypoplasia (PCH), a group of severe neurodegenerative disorders with both mental and motor deficits. Comparing relative levels of Tsen54 gene expression, we demonstrate that the highest Tsen54 expression is observed in anatomical brain substructures important for the normal motor and memory functions in mice.

9.
Int J Mol Sci ; 22(3)2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33535482

ABSTRACT

Background-the graphene-doping procedure represents a useful procedure to improve the mechanical, physical and biological response of several Polymethyl methacrylate (PMMA)-derived polymers and biomaterials for dental applications. The aim of this study was to evaluate osseointegration of Graphene doped Poly(methyl methacrylate) (GD-PMMA) compared with PMMA as potential materials for dental implant devices. Methods-eighteen adult New Zealand white male rabbits with a mean weight of approx. 3000 g were used in this research. A total of eighteen implants of 3.5 mm diameter and 11 mm length in GD-PMMA and eighteen implants in PMMA were used. The implants were placed into the articular femoral knee joint. The animals were sacrificed after 15, 30 and 60 days and the specimens were evaluated by µCT and histomorphometry. Results-microscopically, all 36 implants, 18 in PMMA and 18 in DG-PMMA were well-integrated into the bone. The implants were in contact with cortical bone along the upper threads, while the lower threads were in contact with either newly formed bone or with marrow spaces. The histomorphometry and µCT evaluation showed that the GP-PMMA and PMMA implants were well osseointegrated and the bone was in direct contact with large portions of the implant surfaces, including the space in the medullary canal. Conclusions-in conclusion, the results suggest that GD-PMMA titanium surfaces enhance osseointegration in rabbit femurs. This encourages further research to obtain GD-PMMA with a greater radiopacity. Also, further in vitro and vivo animal studies are necessary to evaluate a potential clinical usage for dental implant applications.


Subject(s)
Dental Implants , Graphite/chemistry , Polymethyl Methacrylate/chemistry , Titanium/chemistry , Animals , Biocompatible Materials , Implants, Experimental , Male , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Osseointegration , Polymers , Prosthesis Design , Rabbits , Surface Properties , X-Ray Microtomography
10.
Toxins (Basel) ; 12(8)2020 07 31.
Article in English | MEDLINE | ID: mdl-32751937

ABSTRACT

Botulinum neurotoxin type A (BoNT/A) is a major therapeutic agent that has been proven to be a successful treatment for different neurological disorders, with emerging novel therapeutic indications each year. BoNT/A exerts its action by blocking SNARE complex formation and vesicle release through the specific cleavage of SNAP-25 protein; the toxin is able to block the release of pro-inflammatory molecules for months after its administration. Here we demonstrate the extraordinary capacity of BoNT/A to neutralize the complete paralysis and pain insensitivity induced in a murine model of severe spinal cord injury (SCI). We show that the toxin, spinally administered within one hour from spinal trauma, exerts a long-lasting proteolytic action, up to 60 days after its administration, and induces a complete recovery of muscle and motor function. BoNT/A modulates SCI-induced neuroglia hyperreactivity, facilitating axonal restoration, and preventing secondary cells death and damage. Moreover, we demonstrate that BoNT/A affects SCI-induced neuropathic pain after moderate spinal contusion, confirming its anti-nociceptive action in this kind of pain, as well. Our results provide the intriguing and real possibility to identify in BoNT/A a therapeutic tool in counteracting SCI-induced detrimental effects. Because of the well-documented BoNT/A pharmacology, safety, and toxicity, these findings strongly encourage clinical translation.


Subject(s)
Analgesics/therapeutic use , Botulinum Toxins, Type A/therapeutic use , Muscular Atrophy/drug therapy , Neuralgia/drug therapy , Neuromuscular Agents/therapeutic use , Neuroprotective Agents/therapeutic use , Paralysis/drug therapy , Spinal Cord Injuries/drug therapy , Animals , Cell Proliferation/drug effects , Cicatrix/prevention & control , Female , Mice , Neuroglia/drug effects , Neurons/drug effects
11.
Materials (Basel) ; 13(16)2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32824141

ABSTRACT

BACKGROUND: Various surface treatments have been tested for titanium implants aiming at increasing their surface biocompatibility and their biological characteristics, but also the efficiency of the implant surface will have to be improved to drastically decrease peri-implantite and mucosite. In fact, the peri-implantitis and peri-implant mucositis have a high incidence in clinical practice. The nanofabrication techniques that offer the possibility to achieve the implant surface that reduces bacterial colonization could influence the osteointegration. The aim of this research was to evaluate the bone response to titanium implants coated with a bifunctional molecule with antimicrobic activity consisting of a combination of silver ions covalently bound to titanium dioxide nanoparticles. METHODS: A total of 36 implants were inserted into 18 older New Zealand white male rabbits. They had two different surfaces. The implants Control group was characterized by an acid-etched and sandblasted surface treatment, and the Test implants had an acid-etched and sandblasted surface coated with a silver ion covalently bound to titanium dioxide nanoparticles in the solution. RESULTS: No statistically significant difference of the bone density was evidenced between Control and Test implants at two weeks (p-value = 0.623), four weeks (p-value = 0.339), and eight weeks (p-value = 0.461). Moreover, no statistically significant difference of the bone-implant contact percentage was evidenced between Control and Test implants at two weeks (p-value = 0.938), four weeks (p-value = 0.307), and eight weeks (p-value = 0.294). The effectiveness of the present investigation demonstrated no adverse effects on osseointegration, and no statistically significant differences were observed in the bone density and percentage of bone-implant contact between Test and Control implants at all the experimental time points (two, four, and eight weeks). CONCLUSIONS: Titanium implants coated with the silver-anatase solution bind very well to the bone and did not have an adverse effect on the bone tissue in a rabbit model. These facts suggest possible clinical applications for the silver composition.

12.
Int J Mol Sci ; 21(8)2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32326334

ABSTRACT

Phosphodiesterase 2A (PDE2A) is a cAMP-cGMP hydrolyzing enzyme essential for mouse development and the PDE2A knockout model (PDE2A-/-) is embryonic lethal. Notably, livers of PDE2A-/- embryos at embryonic day 14.5 (E14.5) have extremely reduced size. Morphological, cellular and molecular analyses revealed loss of integrity in the PDE2A-/- liver niche that compromises the hematopoietic function and maturation. Hematopoietic cells isolated from PDE2A-/- livers are instead able to differentiate in in vitro assays, suggesting the absence of blood cell-autonomous defects. Apoptosis was revealed in hepatoblasts and at the endothelial and stromal compartments in livers of PDE2A-/- embryos. The increase of the intracellular cAMP level and of the inducible cAMP early repressor (ICER) in liver of PDE2A-/- embryos might explain the impairment of liver development by downregulating the expression of the anti-apoptotic gene Bcl2. In summary, we propose PDE2A as an essential gene for integrity maintenance of liver niche and the accomplishment of hematopoiesis.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Hematopoiesis/genetics , Liver/embryology , Liver/metabolism , Organogenesis/genetics , Animals , Apoptosis/genetics , Biomarkers , Cell Differentiation , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Genotype , Immunohistochemistry , Mice , Mice, Transgenic , Mutation , Stem Cells/cytology , Stem Cells/metabolism , Stromal Cells/metabolism
14.
Dis Model Mech ; 12(8)2019 08 02.
Article in English | MEDLINE | ID: mdl-31383820

ABSTRACT

Primary ciliary dyskinesia (PCD) is a genetically heterogeneous disorder affecting normal structure and function of motile cilia, phenotypically manifested as chronic respiratory infections, laterality defects and infertility. Autosomal recessive mutations in genes encoding for different components of the ciliary axoneme have been associated with PCD in humans and in model organisms. The CCDC151 gene encodes for a coiled-coil axonemal protein that ensures correct attachment of outer dynein arm (ODA) complexes to microtubules. A correct arrangement of dynein arm complexes is required to provide the proper mechanical force necessary for cilia beat. Loss-of-function mutations in CCDC151 in humans leads to PCD disease with respiratory distress and defective left-right body asymmetry. In mice with the Ccdc151Snbl loss-of-function mutation (Snowball mutant), left-right body asymmetry with heart defects have been observed. Here, we demonstrate that loss of Ccdc151 gene function via targeted gene deletion in mice leads to perinatal lethality and congenital hydrocephalus. Microcomputed tomography (microCT) X-ray imaging of Ccdc151-ß-galactosidase reporter expression in whole-mount brain and histological analysis show that Ccdc151 is expressed in ependymal cells lining the ventricular brain system, further confirming the role of Ccdc151 dysfunction in hydrocephalus development. Analyzing the features of hydrocephalus in the Ccdc151-knockout animals by microCT volumetric imaging, we observe continuity of the aqueduct of Sylvius, indicating the communicating nature of hydrocephalus in the Ccdc151-knockout animals. Congenital defects in left-right asymmetry and male infertility have been also observed in Ccdc151-null animals. Ccdc151 gene deletion in adult animals results in abnormal sperm counts and defective sperm motility.This article has an associated First Person interview with the joint first authors of the paper.


Subject(s)
Carrier Proteins/metabolism , Ciliary Motility Disorders/pathology , Hydrocephalus/pathology , Animals , Animals, Newborn , Body Patterning , Ciliary Motility Disorders/diagnostic imaging , Ciliary Motility Disorders/genetics , Disease Models, Animal , Ependyma/diagnostic imaging , Ependyma/pathology , Gene Expression Regulation , Hydrocephalus/diagnostic imaging , Hydrocephalus/genetics , Imaging, Three-Dimensional , Male , Mice, Inbred C57BL , Mice, Knockout , Spermatogenesis , Testis/metabolism , X-Ray Microtomography
15.
Int J Mol Sci ; 20(13)2019 Jul 02.
Article in English | MEDLINE | ID: mdl-31269731

ABSTRACT

Bone tissue regeneration strategies require approaches that provide an osteogenic and angiogenic microenvironment able to drive the bone growth. Recently, the development of 3D printing biomaterials, including poly(lactide) (3D-PLA), enriched with mesenchymal stem cells (MSCs) and/or their derivatives, such as extracellular vesicles (EVs) has been achieving promising results. In this study, in vitro results showed an increased expression of osteogenic and angiogenic markers, as RUNX2, VEGFA, OPN and COL1A1 in the living construct 3D-PLA/human Gingival MSCs (hGMSCs)/EVs. Considering that EVs carry and transfer proteins, mRNA and microRNA into target cells, we evaluated miR-2861 and miR-210 expression related to osteoangiogenesis commitment. Histological examination of rats implanted with 3D-PLA/hGMSCs/EVs evidenced the activation of bone regeneration and of the vascularization process, confirmed also by MicroCT. In synthesis, an upregulation of miR-2861 and -210 other than RUNX2, VEGFA, OPN and COL1A1 was evident in cells cultured in the presence of the biomaterial and EVs. Then, these results evidenced that EVs may enhance bone regeneration in calvaria defects, in association with an enhanced vascularization offering a novel regulatory system in the osteoangiogenesis evolution. The application of new strategies to improve biomaterial engraftment is of great interest in the regenerative medicine and can represent a way to promote bone regeneration.


Subject(s)
Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Polyesters/chemistry , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Extracellular Vesicles/genetics , Extracellular Vesicles/transplantation , Gingiva/cytology , Humans , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic , Osteogenesis , Printing, Three-Dimensional , Rats, Wistar , Up-Regulation
16.
Sci Rep ; 9(1): 8883, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31222077

ABSTRACT

Traumatic spinal cord injury has dramatic consequences and a huge social impact. We propose a new mouse model of spinal trauma that induces a complete paralysis of hindlimbs, still observable 30 days after injury. The contusion, performed without laminectomy and deriving from the pressure exerted directly on the bone, mimics more closely many features of spinal injury in humans. Spinal cord was injured at thoracic level 10 (T10) in adult anesthetized female CD1 mice, mounted on stereotaxic apparatus and connected to a precision impactor device. Following severe injury, we evaluated motor and sensory functions, and histological/morphological features of spinal tissue at different time points. Moreover, we studied the effects of early and subchronic administration of Docosahexaenoic acid, investigating functional responses, structural changes proximal and distal to the lesion in primary and secondary injury phases, proteome modulation in injured spinal cord. Docosahexaenoic acid was able i) to restore behavioural responses and ii) to induce pro-regenerative effects and neuroprotective action against demyelination, apoptosis and neuroinflammation. Considering the urgent health challenge represented by spinal injury, this new and reliable mouse model together with the positive effects of docosahexaenoic acid provide important translational implications for promising therapeutic approaches for spinal cord injuries.


Subject(s)
Disease Models, Animal , Docosahexaenoic Acids/therapeutic use , Spinal Cord Injuries/pathology , Acute Disease , Animals , Chronic Disease , Female , Humans , Mice , Spinal Cord Injuries/drug therapy
17.
Front Physiol ; 10: 512, 2019.
Article in English | MEDLINE | ID: mdl-31114512

ABSTRACT

Bone regeneration represents still a challenge, in particular for calvarium defects. Recently, the development of biomaterials with the addiction of stem cells is giving promising results for the treatment of bone defects. In particular, it was demonstrated that scaffolds enriched with mesenchymal stem cells (MSCs) and/or their derivatives, such as conditioned medium (CM) and extracellular vesicles (EVs), may improve bone regeneration. Moreover, given the deep link between osteogenesis and angiogenesis, a successful approach must also take into consideration the development of vascularization. In this work we evaluated the bone regeneration capacity of a collagen membrane (3D-COL) enriched with human periodontal-ligament stem cells (hPDLSCs) and CM or EVs or EVs engineered with polyethylenimine (PEI-EVs) in rats subjected to a calvarial defect. We evaluated also their capacity to induce angiogenic factors. At first, in vitro results showed an increased expression of osteogenic markers in hPDLSCs cultured with the 3D-COL and PEI-EVs, associated also with the increased protein levels of Vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2). The increased expression of these proteins was confirmed also in vivo in rats implanted with the 3D-COL enriched with hPDLSCs and PEI-EVs. Moreover, histological examination evidenced in this group of rats the activation of bone regeneration and of the vascularization process. Also MicroCT imaging with morphometric analysis confirmed in rats transplanted with 3D-COL enriched with hPDLSCs and PEI-EVs an important regenerative process and a better integration level. All together, these results evidenced that the 3D-COL enriched with hPDLSCs and PEI-EVs may promote bone regeneration of calvaria defects, associated also with an increased vascularization.

18.
Int J Mol Sci ; 20(3)2019 Feb 08.
Article in English | MEDLINE | ID: mdl-30744023

ABSTRACT

BACKGROUND: The process of osseointegration of dental implants is characterized by healing phenomena at the level of the interface between the surface and the bone. Implant surface modification has been introduced in order to increase the level of osseointegration. The purpose of this study is to evaluate the influence of biofunctional coatings for dental implants and the bone healing response in a rabbit model. The implant surface coated with collagen type I was analyzed through X-ray Photoelectron Spectroscopy (XPS), Atomic Force Microscopy (AFM), micro-CT and histologically. METHODS: The sandblasted and double acid etched surface coated with collagen type I, and uncoated sandblasted and double acid etched surface were evaluated by X-ray Photoelectron Spectroscopy (XPS) and Atomic Force Microscopy (AFM) analysis in order evaluate the different morphology. In vivo, a total of 36 implants were positioned in rabbit articular femoral knee-joint, 18 fixtures for each surface. Micro-CT scans, histological and histomorphometrical analysis were conducted at 15, 30 and 60 days. RESULTS: A histological statistical differences were evident at 15, 30 and 60 days (p < 0.001). Both implant surfaces showed a close interaction with newly formed bone. Mature bone appeared in close contact with the surface of the fixture. The AFM outcome showed a similar roughness for both surfaces. CONCLUSION: However, the final results showed that a coating of collagen type I on the implant surface represents a promising procedure able to improve osseointegration, especially in regions with a low bone quality.


Subject(s)
Biomimetic Materials , Biomimetics , Coated Materials, Biocompatible , Collagen Type I , Animals , Biomimetic Materials/chemistry , Biomimetics/methods , Coated Materials, Biocompatible/chemistry , Collagen Type I/chemistry , Histocytochemistry , Microscopy, Atomic Force , Photoelectron Spectroscopy , Rabbits , Surface Properties , Time Factors , X-Ray Microtomography
19.
Int J Mol Sci ; 19(4)2018 Mar 29.
Article in English | MEDLINE | ID: mdl-29596323

ABSTRACT

Bone tissue engineering is based on bone grafting to repair bone defects. Bone graft substitutes can contribute to the addition of mesenchymal stem cells (MSCs) in order to enhance the rate and the quality of defect regeneration. The stem cell secretome contains many growth factors and chemokines, which could affect cellular characteristics and behavior. Conditioned medium (CM) could be used in tissue regeneration avoiding several problems linked to the direct use of MSCs. In this study, we investigated the effect of human periodontal ligament stem cells (hPDLSCs) and their CM on bone regeneration using a commercially available membrane scaffold Evolution (EVO) implanted in rat calvarias. EVO alone or EVO + hPDLSCs with or without CM were implanted in Wistar male rats subjected to calvarial defects. The in vivo results revealed that EVO membrane enriched with hPDLSCs and CM showed a better osteogenic ability to repair the calvarial defect. These results were confirmed by acquired micro-computed tomography (CT) images and the increased osteopontin levels. Moreover, RT-PCR in vitro revealed the upregulation of three genes (Collagen (COL)5A1, COL16A1 and transforming growth factor (TGF)ß1) and the down regulation of 26 genes involved in bone regeneration. These results suggest a promising potential application of CM from hPDLSCs and scaffolds for bone defect restoration and in particular for calvarial repair in case of trauma.


Subject(s)
Bone Regeneration , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Periodontal Ligament , Skull , Tissue Scaffolds/chemistry , Animals , Antigens, Differentiation/biosynthesis , Female , Heterografts , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Periodontal Ligament/metabolism , Periodontal Ligament/pathology , Rats , Rats, Wistar , Skull/injuries , Skull/metabolism , Skull/pathology , Tissue Engineering
20.
Mamm Genome ; 29(3-4): 245-259, 2018 04.
Article in English | MEDLINE | ID: mdl-29170794

ABSTRACT

In this work, we applied three-dimensional microCT imaging to study murine embryogenesis in the range from immediate post-implantation period (embryonic day 5.5) to mid-gestation (embryonic day 12.5) with the resolution up to 1.4 µm/voxel. Also, we introduce an imaging procedure for non-invasive volumetric estimation of an entire litter of embryos within the maternal uterine structures. This method allows for an accurate, detailed and systematic morphometric analysis of both embryonic and extra-embryonic components during embryogenesis. Three-dimensional imaging of unperturbed embryos was performed to visualize the egg cylinder, primitive streak, gastrulation and early organogenesis stages of murine development in the C57Bl6/N mouse reference strain. Further, we applied our microCT imaging protocol to determine the earliest point when embryonic development is arrested in a mouse line with knockout for tRNA splicing endonuclease subunit Tsen54 gene. Our analysis determined that the embryonic development in Tsen54 null embryos does not proceed beyond implantation. We demonstrated that application of microCT imaging to entire litter of non-perturbed embryos greatly facilitate studies to unravel gene function during early embryogenesis and to determine the precise point at which embryonic development is arrested in mutant animals. The described method is inexpensive, does not require lengthy embryos dissection and can be applicable for detailed analysis of mutant mice at laboratory scale as well as for high-throughput projects.


Subject(s)
Embryo Implantation/genetics , Embryo Loss/genetics , Embryo Loss/pathology , Imaging, Three-Dimensional , Mutation/genetics , Organogenesis/genetics , X-Ray Microtomography , Animals , Embryo Loss/diagnostic imaging , Embryo, Mammalian/diagnostic imaging , Female , Gastrulation , Mice, Inbred C57BL , Phenotype , Uterus/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...