Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Polymers (Basel) ; 14(22)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36432947

ABSTRACT

Analysing the composition and organisation of the fibrous capsule formed as a result of the Foreign Body Response (FBR) to medical devices, is imperative for medical device improvement and biocompatibility. Typically, analysis is performed using histological techniques which often involve random sampling strategies. This method is excellent for acquiring representative values but can miss the unique spatial distribution of features in 3D, especially when analysing devices used in large animal studies. To overcome this limitation, we demonstrate a non-destructive method for high-resolution large sample imaging of the fibrous capsule surrounding human-sized implanted devices using diffusion tensor imaging (DTI). In this study we analyse the fibrous capsule surrounding two unique macroencapsulation devices that have been implanted in a porcine model for 21 days. DTI is used for 3D visualisation of the microstructural organisation and validated using the standard means of fibrous capsule investigation; histological analysis and qualitative micro computed tomography (microCT) and scanning electron microscopy (SEM) imaging. DTI demonstrated the ability to distinguish microstructural differences in the fibrous capsules surrounding two macroencapsulation devices made from different materials and with different surface topographies. DTI-derived metrics yielded insight into the microstructural organisation of both capsules which was corroborated by microCT, SEM and histology. The non-invasive characterisation of the integration of implants in the body has the potential to positively influence analysis methods in pre-clinical studies and accelerate the clinical translation of novel implantable devices.

2.
Adv Healthc Mater ; 10(14): e2100229, 2021 07.
Article in English | MEDLINE | ID: mdl-34165264

ABSTRACT

Medical devices, such as silicone-based prostheses designed for soft tissue implantation, often induce a suboptimal foreign-body response which results in a hardened avascular fibrotic capsule around the device, often leading to patient discomfort or implant failure. Here, it is proposed that additive manufacturing techniques can be used to deposit durable coatings with multiscale porosity on soft tissue implant surfaces to promote optimal tissue integration. Specifically, the "liquid rope coil effect", is exploited via direct ink writing, to create a controlled macro open-pore architecture, including over highly curved surfaces, while adapting atomizing spray deposition of a silicone ink to create a microporous texture. The potential to tailor the degree of tissue integration and vascularization using these fabrication techniques is demonstrated through subdermal and submuscular implantation studies in rodent and porcine models respectively, illustrating the implant coating's potential applications in both traditional soft tissue prosthetics and active drug-eluting devices.


Subject(s)
Prostheses and Implants , Silicones , Animals , Humans , Materials Testing , Porosity , Swine
3.
Adv Healthc Mater ; 9(11): e2000305, 2020 06.
Article in English | MEDLINE | ID: mdl-32339411

ABSTRACT

Regenerative medicine approaches, specifically stem cell technologies, have demonstrated significant potential to treat a diverse array of pathologies. However, such approaches have resulted in a modest clinical benefit, which may be attributed to poor cell retention/survival at the disease site. A delivery system that facilitates regional and repeated delivery to target tissues can provide enhanced clinical efficacy of cell therapies when localized delivery of high doses of cells is required. In this study, a new regenerative reservoir platform (Regenervoir) is described for use in large animal models, with relevance to cardiac, abdominal, and soft tissue pathologies. Regenervoir incorporates multiple novel design features essential for clinical translation, with a focus on scalability, mechanism of delivery, fixation to target tissue, and filling/refilling with a therapeutic cargo, and is demonstrated in an array of clinical applications that are easily translated to human studies. Regenervoir consists of a porous reservoir fabricated from a single material, a flexible thermoplastic polymer, capable of delivering cargo via fill lines to target tissues. A radiopaque shear thinning hydrogel can be delivered to the therapy reservoir and multiple fixation methods (laparoscopic tacks and cyanoacrylate bioadhesive) can be used to secure Regenervoir to target tissues through a minimally invasive approach.


Subject(s)
Hydrogels , Regenerative Medicine , Animals , Humans , Models, Animal , Polymers , Prostheses and Implants
4.
Mater Sci Eng C Mater Biol Appl ; 103: 109751, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31349422

ABSTRACT

The limited regenerative capacity of the heart after a myocardial infarct results in remodeling processes that can progress to congestive heart failure (CHF). Several strategies including mechanical stabilization of the weakened myocardium and regenerative approaches (specifically stem cell technologies) have evolved which aim to prevent CHF. However, their final performance remains limited motivating the need for an advanced strategy with enhanced efficacy and reduced deleterious effects. An epicardial carrier device enabling a targeted application of a biomaterial-based therapy to the infarcted ventricle wall could potentially overcome the therapy and application related issues. Such a device could play a synergistic role in heart regeneration, including the provision of mechanical support to the remodeling heart wall, as well as providing a suitable environment for in situ stem cell delivery potentially promoting heart regeneration. In this study, we have developed a novel, single-stage concept to support the weakened myocardial region post-MI by applying an elastic, biodegradable patch (SPREADS) via a minimal-invasive, closed chest intervention to the epicardial heart surface. We show a significant increase in %LVEF 14 days post-treatment when GS (clinical gold standard treatment) was compared to GS + SPREADS + Gel with and without cells (p ≤ 0.001). Furthermore, we did not find a significant difference in infarct quality or blood vessel density between any of the groups which suggests that neither infarct quality nor vascularization is the mechanism of action of SPREADS. The SPREADS device could potentially be used to deliver a range of new or previously developed biomaterial hydrogels, a remarkable potential to overcome the translational hurdles associated with hydrogel delivery to the heart.


Subject(s)
Absorbable Implants , Cell- and Tissue-Based Therapy/instrumentation , Hydrogels/administration & dosage , Mesenchymal Stem Cells , Myocardial Infarction/therapy , Adipose Tissue/cytology , Animals , Biocompatible Materials , Cell Movement/drug effects , Cell- and Tissue-Based Therapy/methods , Equipment Design , Female , Humans , Hyaluronic Acid , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Myocardial Infarction/physiopathology , Pericardium , Swine , Viscosity
5.
J Biomater Appl ; 33(5): 681-692, 2018 11.
Article in English | MEDLINE | ID: mdl-30354912

ABSTRACT

Injectable hydrogels that aim to mechanically stabilise the weakened left ventricle wall to restore cardiac function or to deliver stem cells in cardiac regenerative therapy have shown promising data. However, the clinical translation of hydrogel-based therapies has been limited due to difficulties injecting them through catheters. We have engineered a novel catheter, Advanced Materials Catheter (AMCath), that overcomes translational hurdles associated with delivering fast-gelling covalently cross-linked hyaluronic acid hydrogels to the myocardium. We developed an experimental technique to measure the force required to inject such hydrogels and determined the mechanical/viscoelastic properties of the resulting hydrogels. The preliminary in vivo feasibility of delivering fast-gelling hydrogels through AMCath was demonstrated by accessing the porcine left ventricle and showing that the hydrogel was retained in the myocardium post-injection (three 200 µL injections delivered, 192, 204 and 183 µL measured). However, the mechanical properties of the hydrogels were reduced by passage through AMCath (≤20.62% reduction). We have also shown AMCath can be used to deliver cardiopoietic adipose-derived stem cell-loaded hydrogels without compromising the viability (80% viability) of the cells in vitro. Therefore, we show that hydrogel/catheter compatibility issues can be overcome as we have demonstrated the minimally invasive delivery of a fast-gelling covalently cross-linked hydrogel to the beating myocardium.


Subject(s)
Biocompatible Materials/administration & dosage , Cardiac Catheters , Drug Delivery Systems/instrumentation , Hyaluronic Acid/administration & dosage , Hydrogels/administration & dosage , Animals , Cell Line , Cells, Immobilized/cytology , Cells, Immobilized/transplantation , Cross-Linking Reagents/administration & dosage , Equipment Design , Humans , Injections , Myocardial Infarction/therapy , Stem Cell Transplantation , Stem Cells/cytology , Swine
6.
Int J Cardiol ; 212: 324-35, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-27057951

ABSTRACT

BACKGROUND: Critical limb ischemia (CLI) is a major health problem with no adequate treatment. Since CLI is characterized by insufficient tissue vascularization, efforts have focused on the discovery of novel angiogenic factors. Cyclophilin A (CyPA) is an immunophilin that has been shown to promote angiogenesis in vitro and to enhance bone marrow (BM) cell mobilization in vivo. However, its potential as an angiogenic factor in CLI is still unknown. Thus, this study aimed to evaluate whether CyPA might induce neo-angiogenesis in ischemic tissues. METHODS AND RESULTS: Wild-type C57Bl/6j mice underwent acute hind-limb ischemia (HLI) and received a single intramuscular administration of recombinant CyPA or saline. Limb perfusion, capillary density and arteriole number in adductor muscles were significantly increased after CyPA treatment. Interestingly, BM-derived CD117(+) cell recruitment was significantly higher in ischemic adductor tissue of mice treated with CyPA versus saline. Therefore, the effect of CyPA on isolated BM-derived CD117(+) cells in vitro was evaluated. Low concentrations of CyPA stimulated CD117(+) cell proliferation while high concentrations promoted cell death. Moreover, CyPA enhanced CD117(+) cell adhesion and migration in a dose-dependent manner. Mechanistic studies revealed that CyPA up-regulated CXCR4 in CD117(+) cells and in adductor muscles after ischemia. Additionally, SDF-1/CXCR4 axis inhibition by the CXCR4 antagonist AMD3100 decreased CyPA-mediated CD117(+) cell recruitment in the ischemic limb. CONCLUSION: CyPA induces neo-angiogenesis by recruiting BM-derived CD117(+) cell into ischemic tissues, at least in part, through SDF-1/CXCR4 axis.


Subject(s)
Cyclophilin A/administration & dosage , Hindlimb/blood supply , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Animals , Bone Marrow Cells/drug effects , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL12/metabolism , Cyclophilin A/pharmacology , Disease Models, Animal , Hindlimb/drug effects , Humans , Ischemia/metabolism , Mice , Mice, Inbred C57BL , Receptors, CXCR4/metabolism , Signal Transduction/drug effects
7.
Cytotherapy ; 17(9): 1302-13, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26038175

ABSTRACT

BACKGROUND AIMS: The Pall Celeris system is a filtration-based point-of-care device designed to obtain a high concentrate of peripheral blood total nucleated cells (PB-TNCs). We have characterized the Pall Celeris-derived TNCs for their in vitro and in vivo angiogenic potency. METHODS: PB-TNCs isolated from healthy donors were characterized through the use of flow cytometry and functional assays, aiming to assess migratory capacity, ability to form capillary-like structures, endothelial trans-differentiation and paracrine factor secretion. In a hind limb ischemia mouse model, we evaluated perfusion immediately and 7 days after surgery, along with capillary, arteriole and regenerative fiber density and local bio-distribution. RESULTS: Human PB-TNCs isolated by use of the Pall Celeris filtration system were shown to secrete a panel of angiogenic factors and migrate in response to vascular endothelial growth factor and stromal-derived factor-1 stimuli. Moreover, after injection in a mouse model of hind limb ischemia, PB-TNCs induced neovascularization by increasing capillary, arteriole and regenerative fiber numbers, with human cells detected in murine tissue up to 7 days after ischemia. CONCLUSIONS: The Pall Celeris system may represent a novel, effective and reliable point-of-care device to obtain a PB-derived cell product with adequate potency for therapeutic angiogenesis.


Subject(s)
Ischemia/therapy , Neovascularization, Physiologic , Peripheral Arterial Disease/therapy , Point-of-Care Systems , Animals , Blood Component Removal , Cell Differentiation , Cell Movement , Cell Separation/methods , Chemokine CXCL12/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Filtration , Flow Cytometry , Hindlimb/blood supply , Humans , Leukocytes/immunology , Mice , Reperfusion , Vascular Endothelial Growth Factor A/metabolism
8.
Commun Integr Biol ; 6(5): e25466, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24265859

ABSTRACT

In physiopathological conditions, such as diabetes, wound healing is significantly compromised and chronic complications, including ulcers, may occur. In a mouse model of skin repair, we recently reported that wound treatment with Sirtuin activators and class I HDAC inhibitors induced keratinocyte proliferation and enhanced healing via a nitric oxide (NO) dependent mechanism. We observed an increase in total protein acetylation in the wound area, as determined by acetylation of α-tubulin and histone H3 Lysine 9. We reasoned that this process activated cell function as well as regulated gene expression to foster tissue repair. We report here that the direct activation of P300/CBP-associated factor (PCAF) by the histone acetylase activator pentadecylidenemalonate 1b (SPV-106) induced Lysine acetylation in the wound area. This intervention was sufficient to enhance repair process by a NO-independent mechanism. Hence, an impairment of PCAF and/or other GCN5 family acetylases may delay skin repair in physiopathological conditions.

9.
Arterioscler Thromb Vasc Biol ; 33(12): 2867-76, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24072696

ABSTRACT

OBJECTIVE: Dystrophin, the missing or defective protein in Duchenne muscular dystrophy, is expressed not only in muscle cells but also in vascular endothelial cells (ECs). In this study, we assessed the effects of dystrophin deficiency on the angiogenic capacities of ECs. APPROACH AND RESULTS: We isolated vascular ECs from mdx mice, the murine equivalent of Duchenne muscular dystrophy in humans, and wild-type controls, and we found that mdx-derived ECs have impaired angiogenic properties, in terms of migration, proliferation, and tube formation. They also undergo increased apoptosis in vitro compared with wild-type cells and have increased senescence-associated ß-galactosidase activity. Mdx-derived ECs also display reduced ability to support myoblast proliferation when cocultured with satellite cell-derived primary myoblasts. These endothelial defects are mirrored by systemic impairment of angiogenesis in vivo, both on induction of ischemia, stimulation with growth factors in the corneal model and matrigel plug assays, and tumor growth. We also found that dystrophin forms a complex with endothelial NO synthase and caveolin-1 in ECs, and that NO production and cGMP formation are compromised in ECs isolated from mdx mice. Interestingly, treatment with aspirin enhances production of both cGMP and NO in dystrophic ECs, whereas low-dose aspirin improves the dystrophic phenotype of mdx mice in vivo, in terms of resistance to physical exercise, muscle fiber permeability, and capillary density. CONCLUSIONS: These findings demonstrate that impaired angiogenesis is a novel player and potential therapeutic target in Duchenne muscular dystrophy.


Subject(s)
Dystrophin/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Muscular Dystrophy, Duchenne/metabolism , Neovascularization, Physiologic , Animals , Apoptosis , Aspirin/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Caveolin 1/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Cellular Senescence , Coculture Techniques , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Corneal Neovascularization/physiopathology , Cyclic GMP/metabolism , Disease Models, Animal , Dystrophin/genetics , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Ischemia/metabolism , Ischemia/pathology , Ischemia/physiopathology , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Mutation , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Neovascularization, Pathologic , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Time Factors
10.
J Biol Chem ; 288(32): 22915-29, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23836913

ABSTRACT

Histone deacetylase inhibitors (DIs) are promising drugs for the treatment of several pathologies including ischemic and failing heart where they demonstrated efficacy. However, adverse side effects and cardiotoxicity have also been reported. Remarkably, no information is available about the effect of DIs during tissue regeneration following acute peripheral ischemia. In this study, mice made ischemic by femoral artery excision were injected with the DIs MS275 and MC1568, selective for class I and IIa histone deacetylases (HDACs), respectively. In untreated mice, soon after damage, class IIa HDAC phosphorylation and nuclear export occurred, paralleled by dystrophin and neuronal nitric-oxide synthase (nNOS) down-regulation and decreased protein phosphatase 2A activity. Between 14 and 21 days after ischemia, dystrophin and nNOS levels recovered, and class IIa HDACs relocalized to the nucleus. In this condition, the MC1568 compound increased the number of newly formed muscle fibers but delayed their terminal differentiation, whereas MS275 abolished the early onset of the regeneration process determining atrophy and fibrosis. The selective DIs had differential effects on the vascular compartment: MC1568 increased arteriogenesis whereas MS275 inhibited it. Capillarogenesis did not change. Chromatin immunoprecipitations revealed that class IIa HDAC complexes bind promoters of proliferation-associated genes and of class I HDAC1 and 2, highlighting a hierarchical control between class II and I HDACs during tissue regeneration. Our findings indicate that class-selective DIs interfere with normal mouse ischemic hindlimb regeneration and suggest that their use could be limited by alteration of the regeneration process in peripheral ischemic tissues.


Subject(s)
Benzamides/adverse effects , Hindlimb/blood supply , Histone Deacetylase Inhibitors/adverse effects , Hydroxamic Acids/adverse effects , Ischemia , Muscle, Skeletal , Pyridines/adverse effects , Pyrroles/adverse effects , Regeneration/drug effects , Animals , Benzamides/pharmacology , Dystrophin/metabolism , Hindlimb/metabolism , Hindlimb/pathology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Ischemia/drug therapy , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Nitric Oxide Synthase Type I/metabolism , Protein Phosphatase 2/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Time Factors
11.
J Biol Chem ; 288(16): 11004-12, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23463510

ABSTRACT

In a mouse model of skin repair we found that the class I-IIa histone deacetylase inhibitor trichostatin A accelerated tissue regeneration. Unexpectedly, this effect was suppressed by Sirtinol, a class III histone deacetylase (HDAC) (sirtuin)-selective inhibitor. The role of sirtuins (SIRTs) was then investigated by using resveratrol and a novel SIRT1-2-3 activator, the MC2562 compound we synthesized recently. Both resveratrol and MC2562 were effective in accelerating wound repair. The local administration of natural or synthetic SIRT activators, in fact, significantly accelerated skin regeneration by increasing keratinocyte proliferation. In vitro experiments revealed that the activation of SIRTs stimulated keratinocyte proliferation via endothelial NO synthase phosphorylation and NO production. In this condition, the class I member HDAC2 was found S-nitrosylated on cysteine, a post-transduction modification associated with loss of activity and DNA binding capacity. After deacetylase inhibitor or SIRT activator treatment, ChIP showed, in fact, a significant HDAC2 detachment from the promoter region of insulin growth factor I (IGF-I), fibroblast growth factor 10 (FGF-10), and Epithelial Growth Factor (EGF), which may be the final recipients and effectors of the SIRT-NO-HDAC signaling cascade. Consistently, the effect of SIRT activators was reduced in the presence of NG-nitro-L-arginine methyl ester (L-NAME), a general inhibitor of NO synthesis. In conclusion, the NO-dependent cross-talk among class III and I histone deacetylases suggests an unprecedented signaling pathway important for skin repair.


Subject(s)
Group III Histone Deacetylases/metabolism , Histone Deacetylase 2/metabolism , Nitric Oxide/metabolism , Skin/enzymology , Skin/injuries , Wound Healing/physiology , Animals , Cell Line, Transformed , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 10/metabolism , Group III Histone Deacetylases/antagonists & inhibitors , Humans , Insulin-Like Growth Factor I/metabolism , Male , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Wound Healing/drug effects
12.
Eur Heart J ; 34(26): 2007-16, 2013 Jul.
Article in English | MEDLINE | ID: mdl-21784762

ABSTRACT

AIMS: The chemokine receptor CXCR4 modulates endothelial progenitor cell migration, homing, and differentiation, and plays a key role in cardiovascular regeneration. Here we examined the effect of ex vivo acidic preconditioning (AP) on CXCR4 expression and on the regenerative potential of mouse bone marrow (BM) ckit(+) cells. METHODS AND RESULTS: Acidic preconditioning was achieved by exposing BM ckit(+) cells to hypercarbic acidosis (pH 7.0) for 24 h; control cells were kept at pH 7.4. Acidic preconditioning enhanced CXCR4 and stromal cell-derived factor 1 (SDF-1) mRNA levels, as well as CXCR4 phosphorylation. Acidic preconditioning ability to modulate CXCR4 expression depended on cytosolic calcium [Ca(2+)]i mobilization and on nitric oxide (NO), as determined by [Ca(2+)]i buffering with BAPTA, and by treatment with the NO donor (DETA/NO) and the NO synthase inhibitor (L-NAME). Further, AP increased SDF-1-driven chemotaxis, transendothelial migration, and differentiation toward the endothelial lineage in vitro. In a mouse model of hindlimb ischaemia, control and AP ckit(+) cells were transplanted into the ischaemic muscle; AP cells accelerated blood flow recovery, increased capillary, and arteriole number as well as the number of regenerating muscle fibres vs. control. These effects were abolished by treating AP cells with L-NAME. CONCLUSION: Acidic preconditioning represents a novel strategy to enhance BM ckit(+) cell therapeutic potential via NO-dependent increase in CXCR4 expression.


Subject(s)
Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Proto-Oncogene Proteins c-kit/metabolism , Receptors, CXCR4/metabolism , Regeneration/physiology , Animals , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Proliferation , Chelating Agents/pharmacology , Chemokine CXCL12/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endothelial Cells/cytology , Endothelial Cells/physiology , Hindlimb/blood supply , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/prevention & control , Ischemic Preconditioning/methods , Male , Mice , Nitric Oxide Donors/metabolism
13.
J Vasc Res ; 49(5): 425-31, 2012.
Article in English | MEDLINE | ID: mdl-22739401

ABSTRACT

We have previously demonstrated that sonic hedgehog (Shh) gene transfer improves angiogenesis in the setting of ischemia by upregulating the expression of multiple growth factors and enhancing the incorporation of endogenous bone marrow (BM)-derived endothelial progenitor cells (EPCs). In this study, we hypothesized that combined therapy with Shh gene transfer and BM-derived EPCs is more effective than Shh gene therapy alone in an experimental model of peripheral limb ischemia. We used old mice, which have a significantly reduced angiogenic response to ischemia, and compared the ability of Shh gene transfer, exogenous EPCs, or both to improve regeneration after ischemia. We found a significantly higher capillary density in the Shh + EPC-treated muscles compared to the other experimental groups. We also found that Shh gene transfer increases the incorporation and survival of transplanted EPCs. Finally, we found a significantly higher number of regenerating myofibers in the ischemic muscles of mice receiving combined treatment with Shh and BM-derived EPCs. In summary, the combination of Shh gene transfer and BM-derived EPCs more effectively promotes angiogenesis and muscle regeneration than each treatment individually and merits further investigation for its potential beneficial effects in ischemic diseases.


Subject(s)
Bone Marrow Transplantation , Genetic Therapy/methods , Hedgehog Proteins/genetics , Ischemia/therapy , Animals , Bone Marrow Cells/metabolism , Endothelial Cells/metabolism , Hindlimb/blood supply , Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Muscle Development , Muscle, Skeletal/blood supply , Muscle, Skeletal/physiology , Neovascularization, Physiologic/physiology , Regeneration
14.
Mol Biol Cell ; 23(8): 1467-74, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22379106

ABSTRACT

In vivo imaging involving the use of genetically engineered animals is an innovative powerful tool for the noninvasive assessment of the molecular and cellular events that are often targets of therapy. On the basis of the knowledge that the activity of the nuclear factor-Y (NF-Y) transcription factor is restricted in vitro to proliferating cells, we have generated a transgenic reporter mouse, called MITO-Luc (for mitosis-luciferase), in which an NF-Y-dependent promoter controls luciferase expression. In these mice, bioluminescence imaging of NF-Y activity visualizes areas of physiological cell proliferation and regeneration during response to injury. Using this tool, we highlight for the first time a role of NF-Y activity on hepatocyte proliferation during liver regeneration. MITO-Luc reporter mice should facilitate investigations into the involvement of genes in cell proliferation and provide a useful model for studying aberrant proliferation in disease pathogenesis. They should be also useful in the development of new anti/proproliferative drugs and assessment of their efficacy and side effects on nontarget tissues.


Subject(s)
CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Cell Proliferation , Liver Regeneration , Molecular Imaging , Transcription, Genetic , Animals , Cell Cycle/genetics , Cell Line , Cyclin B2/genetics , DNA-Binding Proteins/genetics , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic
15.
J Invest Dermatol ; 132(7): 1908-17, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22437320

ABSTRACT

We aimed at identifying novel regulators of skin wound healing (WH), in an epidermal scratch WH assay, by a small interfering RNA (siRNA) silencing approach. Several transcription factors have been previously reported to affect wound repair. We here show that gene silencing of the transcription factor CAAT enhancer-binding protein γ (C/EBPγ), STAT3, REL, RELA, RELB, SP1, and NFkB impaired WH in vitro, in keratinocytes, whereas E2F and CREBBP silencing accelerated the WH process. We further characterized C/EBPγ, as its silencing yielded the maximal impairment (52.2 ± 12.5%) of scratch wounding (SW). We found that C/EBPγ silencing inhibited both EGF- and serum-induced keratinocyte migration, whereas C/EBPγ overexpression enhanced cell migration to EGF and to serum via the EGFR. Further, C/EBPγ silencing impaired scratch-induced Y1068 and Y1173 EGFR phosphorylation, as well as Y118 paxillin phosphorylation, key molecules regulating cell migration and epidermal WH. Moreover, C/EBPγ levels were induced in keratinocytes, following both SW and EGF stimulation. C/EBPγ siRNA silencing in vivo impaired WH at 3, 5, 7, and 14 days following excisional wounding in mice inhibited both re-epithelialization and granulation tissue formation, and induced a decrease of arteriole number. In conclusion, we here report that C/EBPγ positively regulates wound repair both in vitro and in vivo, at least in part, by affecting EGFR signaling.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , ErbB Receptors/physiology , Signal Transduction/physiology , Wound Healing , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/genetics , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , NF-kappa B/physiology , Paxillin/metabolism , Phosphorylation , RNA, Small Interfering/genetics
16.
Proc Natl Acad Sci U S A ; 108(7): 2795-800, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282606

ABSTRACT

Wanting to explore the epigenetic basis of Duchenne cardiomyopathy, we found that global histone acetylase activity was abnormally elevated and the acetylase P300/CBP-associated factor (PCAF) coimmunoprecipitated with connexin 43 (Cx43), which was N(ε)-lysine acetylated and lateralized in mdx heart. This observation was paralleled by Cx43 dissociation from N-cadherin and zonula occludens 1, whereas pp60-c-Src association was unaltered. In vivo treatment of mdx with the pan-histone acetylase inhibitor anacardic acid significantly reduced Cx43 N(ε)-lysine acetylation and restored its association to GAP junctions (GJs) at intercalated discs. Noteworthy, in normal as well as mdx mice, the class IIa histone deacetylases 4 and 5 constitutively colocalized with Cx43 either at GJs or in the lateralized compartments. The class I histone deacetylase 3 was also part of the complex. Treatment of normal controls with the histone deacetylase pan-inhibitor suberoylanilide hydroxamic acid (MC1568) or the class IIa-selective inhibitor 3-{4-[3-(3-fluorophenyl)-3-oxo-1-propen-1-yl]-1-methyl-1H-pyrrol-2-yl}-N-hydroxy-2-propenamide (MC1568) determined Cx43 hyperacetylation, dissociation from GJs, and distribution along the long axis of ventricular cardiomyocytes. Consistently, the histone acetylase activator pentadecylidenemalonate 1b (SPV106) hyperacetylated cardiac proteins, including Cx43, which assumed a lateralized position that partly reproduced the dystrophic phenotype. In the presence of suberoylanilide hydroxamic acid, cell to cell permeability was significantly diminished, which is in agreement with a Cx43 close conformation in the consequence of hyperacetylation. Additional experiments, performed with Cx43 acetylation mutants, revealed, for the acetylated form of the molecule, a significant reduction in plasma membrane localization and a tendency to nuclear accumulation. These results suggest that Cx43 N(ε)-lysine acetylation may have physiopathological consequences for cell to cell coupling and cardiac function.


Subject(s)
Cardiomyopathies/metabolism , Connexin 43/metabolism , Gap Junctions/metabolism , Lysine/metabolism , Muscular Dystrophy, Duchenne/complications , Myocytes, Cardiac/metabolism , Acetylation/drug effects , Anacardic Acids/pharmacology , Animals , Cardiomyopathies/etiology , Histone Acetyltransferases/metabolism , Hydroxamic Acids , Immunoprecipitation , Mice , Mice, Inbred mdx , Microscopy, Fluorescence , Vorinostat , p300-CBP Transcription Factors/metabolism
17.
Mol Ther ; 19(4): 658-66, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21224834

ABSTRACT

We have previously shown that the signaling pathway of the embryonic morphogen Sonic hedgehog (Shh) is recapitulated in the postnatal skeletal muscle in response to ischemia. We have also demonstrated that Shh is an indirect angiogenic agent upregulating various families of angiogenic growth factors and that Shh gene therapy improves angiogenesis and heart function in experimental models of myocardial ischemia. Based on these findings, we hypothesized that Shh gene therapy is beneficial in an experimental model of peripheral ischemia. We found that intramuscular (i.m.) treatment with a plasmid encoding the Shh human gene (phShh) increased blood flow, capillary density, and arteriole density in mice in which peripheral circulation of the hindlimb was disrupted by removal of the common femoral artery. Shh gene therapy also enhanced vasculogenesis, by increasing the number of circulating bone marrow (BM)-derived endothelial precursors and improving the contribution of these cells to the process of neovascularization. Finally, phShh treatment induced upregulation of prototypical angiogenic, arteriogenic, and vasculogenic factors, such as vascular endothelial growth factor (VEGF), angiopoietin 1 (Ang-1), and stromal cell-derived factor-1 (SDF-1α). These data suggest that Shh gene therapy merits further investigation for its ability to trigger the expression of potent trophic factors and stimulate pleiotropic aspects of neovascularization in the setting of ischemia.


Subject(s)
Genetic Therapy/methods , Hedgehog Proteins/metabolism , Hindlimb/blood supply , Ischemia/therapy , Angiopoietin-1/metabolism , Animals , Chemokine CXCL12/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hedgehog Proteins/genetics , Ischemia/genetics , Ischemia/metabolism , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism
18.
Eur Heart J ; 31(22): 2765-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20534597

ABSTRACT

AIMS: Circulating microRNAs (miRNAs) may represent a novel class of biomarkers; therefore, we examined whether acute myocardial infarction (MI) modulates miRNAs plasma levels in humans and mice. METHODS AND RESULTS: Healthy donors (n = 17) and patients (n = 33) with acute ST-segment elevation MI (STEMI) were evaluated. In one cohort (n = 25), the first plasma sample was obtained 517 ± 309 min after the onset of MI symptoms and after coronary reperfusion with percutaneous coronary intervention (PCI); miR-1, -133a, -133b, and -499-5p were ~15- to 140-fold control, whereas miR-122 and -375 were ~87-90% lower than control; 5 days later, miR-1, -133a, -133b, -499-5p, and -375 were back to baseline, whereas miR-122 remained lower than control through Day 30. In additional patients (n = 8; four treated with thrombolysis and four with PCI), miRNAs and troponin I (TnI) were quantified simultaneously starting 156 ± 72 min after the onset of symptoms and at different times thereafter. Peak miR-1, -133a, and -133b expression and TnI level occurred at a similar time, whereas miR-499-5p exhibited a slower time course. In mice, miRNAs plasma levels and TnI were measured 15 min after coronary ligation and at different times thereafter. The behaviour of miR-1, -133a, -133b, and -499-5p was similar to STEMI patients; further, reciprocal changes in the expression levels of these miRNAs were found in cardiac tissue 3-6 h after coronary ligation. In contrast, miR-122 and -375 exhibited minor changes and no significant modulation. In mice with acute hind-limb ischaemia, there was no increase in the plasma level of the above miRNAs. CONCLUSION: Acute MI up-regulated miR-1, -133a, -133b, and -499-5p plasma levels, both in humans and mice, whereas miR-122 and -375 were lower than control only in STEMI patients. These miRNAs represent novel biomarkers of cardiac damage.


Subject(s)
MicroRNAs/metabolism , Myocardial Infarction/diagnosis , Adult , Aged , Analysis of Variance , Animals , Biomarkers/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Hindlimb/blood supply , Humans , Ischemia , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle, Skeletal/metabolism , Troponin I/metabolism
19.
Cardiovasc Res ; 87(1): 73-82, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20164117

ABSTRACT

AIMS: The effect of histone deacetylase inhibitors on dystrophic heart function is not established. To investigate this aspect, dystrophic mdx mice and wild-type (WT) animals were treated 90 days either with suberoylanilide hydroxamic acid (SAHA, 5 mg/kg/day) or with an equivalent amount of vehicle. METHODS AND RESULTS: The following parameters were evaluated: (i) number of ventricular arrhythmias in resting and stress conditions (restraint test) or after aconitine administration; (ii) cardiac excitability, conduction velocity, and refractoriness; (iii) expression and distribution of connexins (Cxs) and Na(v)1.5 sodium channel. Ventricular arrhythmias were negligible in all resting animals. During restraint, however, an increase in the number of arrhythmias was detected in vehicle-treated mdx mice (mdx-V) when compared with SAHA-treated mdx (mdx-SAHA) mice or normal control (WT-V). Interestingly, aconitine, a sodium channel pharmacologic opener, induced ventricular arrhythmias in 83% of WT-V mice, 11% of mdx-V, and in 57% of mdx-SAHA. Epicardial multiple lead recording revealed a prolongation of the QRS complex in mdx-V mice in comparison to WT-V and WT-SAHA mice, paralleled by a significant reduction in impulse propagation velocity. These alterations were efficiently counteracted by SAHA. Molecular analyses revealed that in mdx mice, SAHA determined Cx remodelling of Cx40, Cx37 and Cx32, whereas expression levels of Cx43 and Cx45 were unaltered. Remarkably, Cx43 lateralization observed in mdx control animals was reversed by SAHA treatment which also re-induced Na(v)1.5 expression. CONCLUSION: SAHA attenuates arrhythmias in mdx mice by a mechanism in which Cx remodelling and sodium channel re-expression could play an important role.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/prevention & control , Heart Conduction System/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Aconitine , Action Potentials , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Connexins/metabolism , Disease Models, Animal , Electrocardiography, Ambulatory , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Heart Rate/drug effects , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , NAV1.5 Voltage-Gated Sodium Channel , Restraint, Physical , Sodium Channels/drug effects , Sodium Channels/metabolism , Telemetry , Time Factors , Vorinostat
20.
Stem Cells ; 28(3): 431-42, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20073046

ABSTRACT

In human endothelial cells, nitric oxide (NO) results in class IIa histone deacetylases (HDACs) activation and marked histone deacetylation. It is unknown whether similar epigenetic events occur in embryonic stem cells (ESC) exposed to NO and how this treatment could influence ESC therapeutic potential during tissue regeneration.This study reports that the NO-dependent class IIa HDACs subcellular localization and activity decreases the global acetylation level of H3 histones in ESC and that this phenomenon is associated with the inhibition of Oct4, Nanog, and KLF4 expression. Further, a NO-induced formation of macromolecular complexes including HDAC3, 4, 7, and protein phosphatase 2A (PP2A) have been detected. These processes correlated with the expression of the mesodermal-specific protein brachyury (Bry) and the appearance of several vascular and skeletal muscle differentiation markers. These events were abolished by the class IIa-specific inhibitor MC1568 and by HDAC4 or HDAC7 short interfering RNA (siRNA). The ability of NO to induce mesodermic/cardiovascular gene expression prompted us to evaluate the regenerative potential of these cells in a mouse model of hindlimb ischemia. We found that NO-treated ESCs injected into the cardiac left ventricle selectively localized in the ischemic hindlimb and contributed to the regeneration of muscular and vascular structures. These findings establish a key role for NO and class IIa HDACs modulation in ESC mesodermal commitment and enhanced regenerative potential in vivo.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/enzymology , Histone Deacetylase 2/metabolism , Ischemia/therapy , Mesoderm/enzymology , Nitric Oxide/metabolism , Animals , Biomarkers/metabolism , Cell Line , Cell Proliferation , Disease Models, Animal , Embryonic Stem Cells/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Graft Survival/drug effects , Graft Survival/genetics , Histone Deacetylase 2/drug effects , Histone Deacetylase 2/genetics , Histones/drug effects , Histones/metabolism , Kruppel-Like Factor 4 , Macromolecular Substances/metabolism , Male , Mesoderm/drug effects , Mice , Mice, Inbred C57BL , Nitric Oxide/pharmacology , Recovery of Function/drug effects , Recovery of Function/genetics , Regeneration/drug effects , Regeneration/genetics , Stem Cell Transplantation/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...