Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Exp Eye Res ; 242: 109861, 2024 May.
Article in English | MEDLINE | ID: mdl-38522635

ABSTRACT

Amyloid-beta (Aß), a family of aggregation-prone and neurotoxic peptides, has been implicated in the pathophysiology of age-related macular degeneration (AMD). We have previously shown that oligomeric and fibrillar species of Aß42 exerted retinal toxicity in rats, but while the consequences of exposure to amyloid were related to intracellular effects, the mechanism of Aß42 internalization in the retina is not well characterized. In the brain, the 67 kDa laminin receptor (67LR) participates in Aß-related neuronal cell death. A short peptide derived from pigment epithelium-derived factor (PEDF), formerly designated PEDF-335, was found to mitigate experimental models of ischemic retinopathy via targeting of 67LR. In the present study, we hypothesized that 67LR mediates the uptake of pathogenic Aß42 assemblies in the retina, and that targeting of this receptor by PEDF-335 may limit the internalization of Aß, thereby ameliorating its retinotoxicity. To test this assumption ARPE-19 cells in culture were incubated with PEDF-335 before treatment with fibrillar or oligomeric structures of Aß42. Immunostaining confirmed that PEDF-335 treatment substantially prevented amyloid internalization into ARPE-19 cells and maintained their viability in the presence of toxic oligomeric and fibrillar Aß42 entities in vitro. FRET competition assay was performed and confirmed the binding of PEDF-335 to 67LR in RPE-like cells. Wild-type rats were treated with intravitreal PEDF-335 in the experimental eye 2 days prior to administration of retinotoxic Aß42 oligomers or fibrils to both eyes. Retinal function was assessed by electroretinography through 6 weeks post injection. The ERG responses in rats treated with oligomeric or fibrillar Aß42 assemblies were near-normal in eyes previously treated with intravitreal PEDF-335, whereas those measured in the control eyes treated with injection of the Aß42 assemblies alone showed pathologic attenuation of the retinal function through 6 weeks. The retinal presence of 67LR was determined ex vivo by immunostaining and western blotting. Retinal staining demonstrated the constitutional expression of 67LR mainly in the retinal nuclear layers. In the presence of Aß42, the levels of 67LR were increased, although its retinal distribution remained largely unaltered. In contrast, no apparent differences in the retinal expression level of 67LR were noted following exposure to PEDF-335 alone, and its pattern of localization in the retina remained similarly concentrated primarily in the inner and outer nuclear layers. In summary, we found that PEDF-335 confers protection against Aß42-mediated retinal toxicity, with significant effects noted in cells as well as in vivo in rats. The effects of PEDF-335 in the retina are potentially mediated via binding to 67LR and by at least partial inhibition of Aß42 internalization. These results suggest that PEDF-335 may merit further consideration in the development of targeted inhibition of amyloid-related toxicity in the retina. More broadly, our observations provide evidence on the importance of extracellular versus intracellular Aß42 in the retina and suggest concepts on the molecular mechanism of Aß retinal pathogenicity.


Subject(s)
Amyloid beta-Peptides , Electroretinography , Eye Proteins , Nerve Growth Factors , Serpins , Animals , Serpins/metabolism , Eye Proteins/metabolism , Nerve Growth Factors/metabolism , Rats , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Peptide Fragments/toxicity , Disease Models, Animal , Receptors, Laminin/metabolism , Male , Retina/drug effects , Retina/metabolism , Humans , Intravitreal Injections , Blotting, Western , Retinal Diseases/prevention & control , Retinal Diseases/metabolism , Retinal Diseases/chemically induced , Cells, Cultured
2.
BMC Genomics ; 25(1): 119, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38281016

ABSTRACT

BACKGROUND: Organisms from many distinct evolutionary lineages acquired the capacity to enter a dormant state in response to environmental conditions incompatible with maintaining normal life activities. Most studied organisms exhibit seasonal or annual episodes of dormancy, but numerous less studied organisms enter long-term dormancy, lasting decades or even centuries. Intriguingly, many planktonic animals produce encased embryos known as resting eggs or cysts that, like plant seeds, may remain dormant for decades. Herein, we studied a rotifer Brachionus plicatilis as a model planktonic species that forms encased dormant embryos via sexual reproduction and non-dormant embryos via asexual reproduction and raised the following questions: Which genes are expressed at which time points during embryogenesis? How do temporal transcript abundance profiles differ between the two types of embryos? When does the cell cycle arrest? How do dormant embryos manage energy? RESULTS: As the molecular developmental kinetics of encased embryos remain unknown, we employed single embryo RNA sequencing (CEL-seq) of samples collected during dormant and non-dormant embryogenesis. We identified comprehensive and temporal transcript abundance patterns of genes and their associated enriched functional pathways. Striking differences were uncovered between dormant and non-dormant embryos. In early development, the cell cycle-associated pathways were enriched in both embryo types but terminated with fewer nuclei in dormant embryos. As development progressed, the gene transcript abundance profiles became increasingly divergent between dormant and non-dormant embryos. Organogenesis was suspended in dormant embryos, concomitant with low transcript abundance of homeobox genes, and was replaced with an ATP-poor preparatory phase characterized by very high transcript abundance of genes encoding for hallmark dormancy proteins (e.g., LEA proteins, sHSP, and anti-ROS proteins, also found in plant seeds) and proteins involved in dormancy exit. Surprisingly, this period appeared analogous to the late maturation phase of plant seeds. CONCLUSIONS: The study highlights novel divergent temporal transcript abundance patterns between dormant and non-dormant embryos. Remarkably, several convergent functional solutions appear during the development of resting eggs and plant seeds, suggesting a similar preparatory phase for long-term dormancy. This study accentuated the broad novel molecular features of long-term dormancy in encased animal embryos that behave like "animal seeds".


Subject(s)
Rotifera , Animals , Rotifera/genetics , Gene Expression Profiling , Transcriptome , Proteins/metabolism , Seeds , Plant Dormancy , Germination/genetics
3.
Sci Adv ; 7(41): eabj5435, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34613777

ABSTRACT

Neurons within the tumor microenvironment promote cancer progression; thus, their local targeting has potential clinical benefits. We designed PEGylated lipid nanoparticles loaded with a non-opioid analgesic, bupivacaine, to target neurons within breast cancer tumors and suppress nerve-to-cancer cross-talk. In vitro, 100-nm nanoparticles were taken up readily by primary neurons, trafficking from the neuronal body and along the axons. We demonstrate that signaling between triple-negative breast cancer cells (4T1) and neurons involves secretion of cytokines stimulating neurite outgrowth. Reciprocally, neurons stimulated 4T1 proliferation, migration, and survival through secretion of neurotransmitters. Bupivacaine curbs neurite growth and signaling with cancer cells, inhibiting cancer cell viability. In vivo, bupivacaine-loaded nanoparticles intravenously administered suppressed neurons in orthotopic triple-negative breast cancer tumors, inhibiting tumor growth and metastatic dissemination. Overall, our findings suggest that reducing nerve involvement in tumors is important for treating cancer.

4.
Cell Signal ; 86: 110085, 2021 10.
Article in English | MEDLINE | ID: mdl-34280495

ABSTRACT

Inflammation and cancer are intimately linked. A key mediator of inflammation is the transcription-factor NF-κB/RelA:p50. SEF (also known as IL-17RD) is a feedback antagonist of NF-κB/RelA:p50 that is emerging as an important link between inflammation and cancer. SEF acts as a buffer to prevent excessive NF-κB activity by sequestering NF-κB/RelA:p50 in the cytoplasm of unstimulated cells, and consequently attenuating the NF-κB response upon pro-inflammatory cytokine stimulation. SEF contributes to cancer progression also via modulating other signaling pathways, including those triggered by growth-factors. Despite its important role in human physiology and pathology, mechanisms that regulate SEF biochemical properties and inhibitory activity are unknown. Here we show that human SEF is an intrinsically labile protein that is stabilized via CK2-mediated phosphorylation, and identified the residues whom phosphorylation by CK2 stabilizes hSEF. Unlike endogenous SEF, ectopic SEF was rapidly degraded when overexpressed but was stabilized in the presence of excess CK2, suggesting a mechanism for limiting SEF levels depending upon CK2 processivity. Additionally, phosphorylation by CK2 potentiated hSef interaction with NF-κB in cell-free binding assays. Most importantly, we identified a CK2 phosphorylation site that was indispensable for SEF inhibition of pro-inflammatory cytokine signaling but was not required for SEF inhibition of growth-factor signaling. To our knowledge, this is the first demonstration of post-translational modifications that regulate SEF at multiple levels to optimize its inhibitory activity in a specific signaling context. These findings may facilitate the design of SEF variants for treating cytokine-dependent pathologies, including cancer and chronic inflammation.


Subject(s)
Casein Kinase II , Caseins , Casein Kinase II/metabolism , Caseins/metabolism , Humans , NF-kappa B/metabolism , Phosphorylation , Signal Transduction , Transcription Factor RelA/metabolism
5.
FASEB J ; 34(6): 7745-7758, 2020 06.
Article in English | MEDLINE | ID: mdl-32337805

ABSTRACT

The field of angiogenesis research provides deep understanding regarding this important process, which plays fundamental roles in tissue development and different abnormalities. In vitro models offer the advantages of low-cost high-throughput research of angiogenesis while sparing animal lives, and enabling the use of human cells. Nevertheless, prevailing in vitro models lack stability and are limited to a few days' assays. This study, therefore, examines the hypothesis that closely mimicking the vascular microenvironment can more reliably support longer angiogenesis processes in vitro. To this end, porcine arterial extracellular matrix (paECM)- a key component of blood vessels-was isolated and processed into a thermally induced hydrogel and characterized in terms of composition, structure, and mechanical properties, thus confirming the preservation of important characteristics of arterial extracellular matrix. This unique hydrogel was further tailored into a three-dimensional model of angiogenesis using endothelial cells and supporting cells, in a configuration that allows high-throughput quantitative analysis of cell viability and proliferation, cell migration, and apoptosis, thus revealing the advantages of paECM over frequently used biomaterials. Markedly, when applied with well-known effectors of angiogenesis, the model measures reflected the expected response, hence validating its efficacy and establishing its potential as a promising tool for the research of angiogenesis.


Subject(s)
Arteries/cytology , Extracellular Matrix/physiology , Hydrogels/pharmacology , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Animals , Apoptosis/drug effects , Biocompatible Materials/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells/cytology , Humans , Neovascularization, Physiologic/physiology , Swine , Tissue Engineering/methods , Tissue Scaffolds
6.
Nat Commun ; 11(1): 1381, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170063

ABSTRACT

Signaling through the insulin receptor governs central physiological functions related to cell growth and metabolism. Here we show by tandem native protein complex purification approach and super-resolution STED microscopy that insulin receptor activity requires association with the fundamental structural module in muscle, the dystrophin glycoprotein complex (DGC), and the desmosomal component plakoglobin (γ-catenin). The integrity of this high-molecular-mass assembly renders skeletal muscle susceptibility to insulin, because DGC-insulin receptor dissociation by plakoglobin downregulation reduces insulin signaling and causes atrophy. Furthermore, low insulin receptor activity in muscles from transgenic or fasted mice decreases plakoglobin-DGC-insulin receptor content on the plasma membrane, but not when plakoglobin is overexpressed. By masking ß-dystroglycan LIR domains, plakoglobin prevents autophagic clearance of plakoglobin-DGC-insulin receptor co-assemblies and maintains their function. Our findings establish DGC as a signaling hub, and provide a possible mechanism for the insulin resistance in Duchenne Muscular Dystrophy, and for the cardiomyopathies seen with plakoglobin mutations.


Subject(s)
Dystrophin/metabolism , Glycoproteins/metabolism , Muscle, Skeletal/metabolism , Receptor, Insulin/metabolism , Signal Transduction , gamma Catenin/metabolism , Animals , Autophagy , Cell Membrane/metabolism , Disease Models, Animal , Dystroglycans/metabolism , Dystrophin/genetics , Male , Mice , Mice, Transgenic , Muscular Dystrophy, Duchenne/metabolism , Protein Domains
7.
Cell Signal ; 59: 110-121, 2019 07.
Article in English | MEDLINE | ID: mdl-30862497

ABSTRACT

The classical NF-κB transcription factor (RelA:p50) and the tumor suppressor Sef axis constitute a negative regulatory loop in which Sef, a target of NF-κB/RelA:p50, fine-tunes NF-κB/RelA:p50 transcriptional-activation in response to inflammatory stimuli trough binding to p50. Similar to the inhibitor IκBα, Sef sequesters NF-κB/RelA:p50 in the cytoplasm of unstimulated cells. Despite its key roles in regulating multiple cellular processes and its potential role as mediator between inflammation and cancer, Sef structural domains required to fulfill its tasks are poorly characterized, and how Sef specificity towards RelA:p50 is achieved is unknown. In-vitro binding assays using bacterially expressed Sef and Co-IP experiments, revealed that in addition to p50, Sef directly interacts with IκBα, and the IKKß subunit of the IKK complex which mediates RelA:p50 induction by inflammatory stimuli. These interactions are ligand-independent and do not require Sef post-translational modifications. Deletion mutagenesis mapped binding site to IKKß in a 74- residue segment juxtaposing Sef transmembrane domain, whereas several Sef regions seem to interact with IκBα. Moreover, we identified two new sites which together with the previously identified conserved tyrosine constitute three discontinuous Sef regions each indispensable for Sef binding to RelA:p50 and inhibiting its cytokine induced transcriptional activation. Contrary to IκBα, endogenous Sef is not degraded upon cytokine-stimulation, and its targeting in different cell types markedly enhances cytokine-induced NF-κB nuclear translocation. These results reveal Sef as the first scaffold that brings together the components of NF-κB/RelA:p50 signaling-module. Sef scaffolding function explains the basis for Sef specificity towards inhibiting inflammatory cytokine-induction of NF-κB/RelA:p50.


Subject(s)
NF-kappa B p50 Subunit/metabolism , Receptors, Interleukin/chemistry , Receptors, Interleukin/metabolism , Transcription Factor RelA/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Binding Sites , Cell Nucleus/metabolism , Cytoplasm/metabolism , HEK293 Cells , Humans , I-kappa B Kinase/metabolism , NF-KappaB Inhibitor alpha/metabolism , Protein Binding
8.
J Control Release ; 296: 1-13, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30615983

ABSTRACT

Acidic pH in the tumor microenvironment is associated with cancer metabolism and creates a physiological barrier that prevents from drugs to penetrate cells. Specifically, ionizable weak-base drugs, such as doxorubicin, freely permeate membranes in their uncharged form, however, in the acidic tumor microenvironment these drugs become charged and their cellular permeability is retarded. In this study, 100-nm liposomes loaded with sodium bicarbonate were used as adjuvants to elevate the tumor pH. Combined treatment of triple-negative breast cancer cells (4T1) with doxorubicin and sodium-bicarbonate enhanced drug uptake and increased its anti-cancer activity. In vivo, mice bearing orthotropic 4T1 breast cancer tumors were administered either liposomal or free bicarbonate intravenously. 3.7 ±â€¯0.3% of the injected liposomal dose was detected in the tumor after twenty-four hours, compared to 0.17% ±â€¯0.04% in the group injected free non-liposomal bicarbonate, a 21-fold increase. Analyzing nanoparticle biodistribution within the tumor tissue revealed that 93% of the PEGylated liposomes accumulated in the extracellular matrix, while 7% were detected intracellularly. Mice administered bicarbonate-loaded liposomes reached an intra-tumor pH value of 7.38 ±â€¯0.04. Treating tumors with liposomal bicarbonate combined with a sub-therapeutic dose of doxorubicin achieved an improved therapeutic outcome, compared to mice treated with doxorubicin or bicarbonate alone. Interestingly, analysis of the tumor microenvironment demonstrated an increase in immune cell' population (T-cell, B-cell and macrophages) in tumors treated with liposomal bicarbonate. This study demonstrates that targeting metabolic adjuvants with nanoparticles to the tumor microenvironment can enhance anticancer drug activity and improve treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Nanoparticles/administration & dosage , Neoplasms , Sodium Bicarbonate/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Biological Transport/drug effects , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacokinetics , Female , Humans , Hydrogen-Ion Concentration , Liposomes , Mice, Inbred BALB C , Neoplasms/chemistry , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Sodium Bicarbonate/pharmacokinetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
9.
Langmuir ; 34(44): 13263-13273, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30350695

ABSTRACT

Colloidal particles combined with a polymer can be used to stabilize an oil-water interface forming stable emulsions. Here, we described a novel liquid crystal (LC)-in-LC emulsion composed of a nematic oil phase and a cholesteric or nematic aqueous cellulose nanocrystal (CNC) continuous phase. The guest oil droplets were stabilized and suspended in liquid-crystalline CNCs, inducing distortions and topological defects inside the host LC phase. These emulsions exhibited anisotropic interactions between the two LCs that depended on the diameter-to-pitch ratio of suspended guest droplets and the host CNC cholesteric phase. When the ratio was high, oil droplets were embedded into a cholesteric shell with a concentric packing of CNC layers and took on a radial orientation of the helical axis. Otherwise, discrete surface-trapped LC droplet assemblies with long-range ordering were obtained, mimicking the fingerprint configuration of the cholesteric phase. Thus, the LC-in-LC emulsions presented here define a new class of ordered soft matter in which both nematic and cholesteric LC ordering can be well-manipulated.

10.
Tissue Eng Part A ; 23(1-2): 69-79, 2017 01.
Article in English | MEDLINE | ID: mdl-27784199

ABSTRACT

Effective cellularization is a key approach to prevent small-caliber (<4 mm) tissue-engineered vascular graft (TEVG) failure and maintain patency and contractility following implantation. To achieve this goal, however, improved biomimicking designs and/or relatively long production times (typically several months) are required. We previously reported on porcine carotid artery decellularization yielding biomechanically stable and cell supportive small-caliber (3-4 mm diameter, 5 cm long) arterial extracellular matrix (scaECM) vascular grafts. In this study, we aimed to study the scaECM graft patency in vivo and possibly improve that patency by graft pre-endothelialization with the recipient porcine autologous cells using our previously reported custom-designed dynamic perfusion bioreactor system. Decellularized scaECM vascular grafts were histologically characterized, their immunoreactivity studied in vitro, and their biocompatibility profile evaluated as a xenograft subcutaneous implantation in a mouse model. To study the scaECM cell support and remodeling ability, pig autologous endothelial and smooth muscle cells (SMCs) were seeded and dynamically cultivated within the scaECM lumen and externa/media, respectively. Finally, endothelialized-only scaECMs-hypothesized as a prerequisite for maintaining graft patency and controlling intimal hyperplasia-were transplanted as an interposition carotid artery graft in a porcine model. Graft patency was evaluated through angiography online and endpoint pathological assessment for up to 6 weeks. Our results demonstrate the scaECM-TEVG biocompatibility preserving a structurally and mechanically stable vascular wall not just following decellularization and recellularization but also after implantation. Using our dynamic perfusion bioreactor, we successfully demonstrated the ability of this TEVG to support in vitro recellularization and remodeling by primary autologous endothelial and SMCs, which were seeded on the lumen and the externa/media layers, respectively. Following transplantation, dynamically endothelialized scaECM-TEVGs remained patent for 6 weeks in a pig carotid interposition bypass model. When compared with nonrevitalized control grafts, reendothelialized grafts provided excellent antithrombogenic activity, inhibited intimal hyperplasia formation, and encouraged media wall infiltration and reorganization with recruited host SMCs. We thus demonstrate that readily available decellularized scaECM can be promptly revitalized with autologous cells in a 3-week period before implantation, indicating applicability as a future platform for vascular reconstructive procedures.


Subject(s)
Blood Vessel Prosthesis Implantation , Blood Vessel Prosthesis , Carotid Arteries/surgery , Extracellular Matrix , Animals , Autografts , Bioprosthesis , Carotid Arteries/physiopathology , Mice , Swine
11.
Nat Commun ; 7: 13325, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830705

ABSTRACT

Personalized medicine promises to revolutionize cancer therapy by matching the most effective treatment to the individual patient. Using a nanoparticle-based system, we predict the therapeutic potency of anticancer medicines in a personalized manner. We carry out the diagnostic stage through a multidrug screen performed inside the tumour, extracting drug activity information with single cell sensitivity. By using 100 nm liposomes, loaded with various cancer drugs and corresponding synthetic DNA barcodes, we find a correlation between the cell viability and the drug it was exposed to, according to the matching barcodes. Based on this screen, we devise a treatment protocol for mice bearing triple-negative breast-cancer tumours, and its results confirm the diagnostic prediction. We show that the use of nanotechnology in cancer care is effective for generating personalized treatment protocols.


Subject(s)
DNA/chemistry , Nanoparticles/chemistry , Precision Medicine/methods , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Base Sequence , Cell Line, Tumor , DNA/genetics , Drug Carriers/chemistry , Female , Humans , Kaplan-Meier Estimate , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/genetics , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
12.
Tissue Eng Part A ; 21(9-10): 1507-19, 2015 May.
Article in English | MEDLINE | ID: mdl-25602926

ABSTRACT

Functional vascularization is a prerequisite for cardiac tissue engineering of constructs with physiological thicknesses. We previously reported the successful preservation of main vascular conduits in isolated thick acellular porcine cardiac ventricular ECM (pcECM). We now unveil this scaffold's potential in supporting human cardiomyocytes and promoting new blood vessel development ex vivo, providing long-term cell support in the construct bulk. A custom-designed perfusion bioreactor was developed to remodel such vascularization ex vivo, demonstrating, for the first time, functional angiogenesis in vitro with various stages of vessel maturation supporting up to 1.7 mm thick constructs. A robust methodology was developed to assess the pcECM maximal cell capacity, which resembled the human heart cell density. Taken together these results demonstrate feasibility of producing physiological-like constructs such as the thick pcECM suggested here as a prospective treatment for end-stage heart failure. Methodologies reported herein may also benefit other tissues, offering a valuable in vitro setting for "thick-tissue" engineering strategies toward large animal in vivo studies.


Subject(s)
Extracellular Matrix/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bioreactors , Coculture Techniques , Feasibility Studies , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Myocardium/cytology , Sus scrofa
13.
Tissue Eng Part A ; 18(19-20): 2125-37, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22663095

ABSTRACT

The decellularization of porcine heart tissue offers many opportunities for the production of physiologically relevant myocardial mimetic scaffolds. Earlier, we reported the successful isolation of a thin porcine cardiac extracellular matrix (pcECM) exhibiting relevant bio-mechanical properties for myocardial tissue engineering. Nevertheless, since native cardiac tissue is much thicker, such thin scaffolds may offer limited regeneration capacity. However, generation of thicker myocardial mimetic tissue constructs is hindered by diffusion limitations (~100 µm), and the lack of a proper vascular-like network within these constructs. In our present work, we focused on optimizing the decellularization procedure for thicker tissue slabs (10-15 mm), while retaining their inherent vasculature, and on characterizing the resulting pcECM. The trypsin/Triton-based perfusion procedure that resulted in a nonimmunogenic and cell-supportive pcECM was found to be more effective in cell removal and in the preservation of fiber morphology and structural characteristics than stirring, sonication, or sodium dodecyl sulfate/Triton-based procedures. Mass spectroscopy revealed that the pcECM is mainly composed of ECM proteins with no apparent cellular protein remains. Mechanical testing indicated that the obtained pcECM is viscoelastic in nature and possesses the typical stress-strain profile of biological materials. It is stiffer than native tissue yet exhibits matched mechanical properties in terms of energy dissipation, toughness, and ultimate stress behavior. Vascular network functionality was maintained to the first three-four branches from the main coronary vessels. Taken together, these results reaffirm the efficiency of the decellularization procedure reported herein for yielding thick nonimmunogenic cell-supportive pcECM scaffolds, preserving both native tissue ultra-structural properties and an inherent vascular network. When reseeded with the appropriate progenitor cells, these scaffolds can potentially serve as ex vivo screening platforms for new therapeutics, as models for human cardiac ECM, or as biomedical constructs for patch or transmural transplantation strategies.


Subject(s)
Extracellular Matrix/chemistry , Myocardium/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Adhesion/physiology , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Microscopy, Electron, Scanning , Rats , Swine
14.
Tissue Eng Part A ; 18(3-4): 411-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21919798

ABSTRACT

Patients with small caliber artery disorders, often lack the suitable autologous tissue needed for bypassing diseased vessels or for other vascular reconstructive procedures. We propose to decellularize small caliber porcine carotid artery, then recellularize it with vascular cells and function as scaffold for tissue engineering vascular graft replacements. Based on a modified decellularization method developed in our laboratory, the cellular contents of small caliber (<4 mm) arteries were carefully removed using an enzymatic and detergent decellularization procedure. Decellularization efficiency was evaluated using histology and scanning electron microscopy, which demonstrated the absence of cellular remains in the artery wall. Proteomic analysis of the scaffold revealed that the decellularized vessels retained their major extracellular matrix protein composition. Mechanical analyses revealed no significant change in the extracellular matrix (ECM) properties versus the native artery. The decellularized artery was reseeded with human umbilical vein endothelial cells (HUVECs) and smooth muscle cells (SMCs) and cultured under static or dynamic conditions in a perfusion bioreactor designed and developed in our laboratory for these studies. Dynamic co-culturing of SMC and HUVEC, in this custom-made perfusion bioreactor, led to a higher infiltration, migration and proliferation of SMC toward the media and to a more confluent endothelium formation on the luminal surface when compared with static culturing. In addition, vascular media remodeling by SMC correlated to the expression of remodeling related genes assessed by real-time reverse transcription-polymerase chain reaction and HUVEC cultivation contributed to the remodeling of several basement membrane proteins stained using immunohistochemistry. All together, these findings indicate the potential of such decellularized arterial ECM for future small caliber vascular graft reconstruction therapies.


Subject(s)
Blood Vessel Prosthesis , Carotid Arteries/anatomy & histology , Endothelium, Vascular/growth & development , Extracellular Matrix/metabolism , Tissue Engineering/methods , Tunica Media/physiology , Animals , Biomechanical Phenomena , Bioreactors , Cell Survival , Extracellular Matrix/genetics , Extracellular Matrix/ultrastructure , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Humans , Myocytes, Smooth Muscle/cytology , Perfusion , Proteomics , Reproducibility of Results , Sheep , Sus scrofa , Tissue Culture Techniques , Tissue Scaffolds
15.
FASEB J ; 24(1): 22-31, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19726759

ABSTRACT

Cell encapsulation is a promising approach for long-term delivery of therapeutic agents. Nonetheless, this system has failed to reach clinical settings, as the entrapped cells provoke a host immune reaction. Mesenchymal stem cells (MSCs), however, potentially may overcome this impediment and serve as a promising platform for cell-based microencapsulation. They are known to be hypoimmunogenic and can be genetically modified to express a variety of therapeutic factors. We have designed alginate-PLL microcapsules that can encapsulate human MSCs (hMSCs) for extended periods, as demonstrated by fluorescence and H(3)-thymidine assays. The encapsulated hMSCs maintained their mesenchymal surface markers and differentiated to all the typical mesoderm lineages. In vitro and in vivo immunogenicity studies revealed that encapsulated hMSCs were significantly hypoimmunogenic, leading to a 3-fold decrease in cytokine expression compared to entrapped cell lines. The efficacy of such systems was demonstrated by genetically modifying the cells to express the hemopexin-like protein (PEX), an inhibitor of angiogenesis. Live imaging and tumor measurements showed that encapsulated hMSC-PEX injected adjacent to glioblastoma tumors in nude mice led to a significant reduction in tumor volume (87%) and weight (83%). We clearly demonstrate that hMSCs are the cell of choice for microencapsulation cell based-therapy, thus bringing this technology closer to clinical application.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Adipogenesis , Alginates , Animals , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , Chondrogenesis , Drug Compounding/methods , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , NIH 3T3 Cells , Osteogenesis , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Polylysine/analogs & derivatives , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
16.
Tissue Eng Part C Methods ; 16(4): 671-83, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19780649

ABSTRACT

We have developed an efficient decellularization process for the isolation of extracellular matrix (ECM) from native cardiac tissue. The isolated ECM exhibited desirable mechanical properties in terms of elasticity, strength and durability-properties required from scaffolds used for cardiac tissue repair. This study further investigates the potential use of this scaffold for cardiac tissue engineering in terms of interactions with seeded cells and biocompatibility. We used the commonly studied fibroblasts, cardiomyocytes, and mesenchymal stem cells, which were isolated and seeded onto the scaffold. Cell density and distribution were followed by 3,3'-dioctadecyloxacarbocyanine perchlorate staining, and their proliferation and viability were assessed by AlamarBlue assay and fluorecein-diacetate/propidium iodide staining. Fibroblast-seeded scaffolds shrank to 1-2 mm(3) spheroids, and their glycosaminoglycans significantly increased by 23%. The expression of ECM remodeling-related mRNAs of collagens I and III, matrix metalloproteinase 2, and type 1 tissue inhibitor of metalloproteinases was quantified by real-time polymerase chain reaction, and was found significantly elevated in fibroblast-seeded scaffold, compared with the control cells on plates. Fibroblast-seeded scaffolds lost some flexibility, yet gained strength compared with the acellular scaffolds, as shown by mechanical testing. Scaffold seeded with cardiomyocyte began to beat in concert few days after seeding, and the myocytes expressed typical functional cardiac markers such as alpha-actinin, troponin I, and connexin43. The cells revealed aligned elongated morphology, as presented by immunofluorescent staining and scanning electron microscopy. Mesenchymal stem cell-seeded scaffolds maintained viability over 24 days in culture. These findings further strengthen the potential use of acellular cardiac ECM as a biomaterial for heart regeneration.


Subject(s)
Biocompatible Materials/pharmacology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Biological Assay , Biomechanical Phenomena/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Collagen/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/ultrastructure , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibronectins/metabolism , Immunohistochemistry , Immunophenotyping , Macrophage Activation/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Myocardial Contraction/drug effects , Myocardium/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Reproducibility of Results , Sheep , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...