Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
2.
J Appl Gerontol ; : 7334648231214940, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38037740

ABSTRACT

To understand health and well-being in later life, it is vital to consider the meaning of subjective aging. This study aimed to explore how perceived health, self-perceptions of aging, and participation in social activities relate to each other among older persons in the Bothnia region and Åland islands in Finland. Data were analyzed using Spearman's and polychoric correlation and multinomial logistic regression analyses. The perceived good health and the younger physical, psychological, and social dimensions of subjective age were found to be associated with each other and with participation in social activities outside one's home.

8.
Curr Stem Cell Res Ther ; 11(8): 666-675, 2016.
Article in English | MEDLINE | ID: mdl-26423295

ABSTRACT

Regenerative medicine is a multidisciplinary field where continued progress relies on the incorporation of a diverse set of technologies from a wide range of disciplines within medicine, science and engineering. This review describes how one such technique, mathematical modelling, can be utilised to improve the tissue engineering of organs and stem cell therapy. Several case studies, taken from research carried out by our group, ACTREM, demonstrate the utility of mechanistic mathematical models to help aid the design and optimisation of protocols in regenerative medicine.


Subject(s)
Models, Biological , Organ Specificity , Stem Cell Transplantation , Tissue Engineering/methods , Animals , Humans , Regenerative Medicine , Tissue Scaffolds/chemistry
9.
Biomaterials ; 77: 320-35, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26618750

ABSTRACT

The currently available surgical options to repair the diaphragm are associated with significant risks of defect recurrence, lack of growth potential and restored functionality. A tissue engineered diaphragm has the potential to improve surgical outcomes for patients with congenital or acquired disorders. Here we show that decellularized diaphragmatic tissue reseeded with bone marrow mesenchymal stromal cells (BM-MSCs) facilitates in situ regeneration of functional tissue. A novel bioreactor, using simultaneous perfusion and agitation, was used to rapidly decellularize rat diaphragms. The scaffolds retained architecture and mechanical properties and supported cell adhesion, proliferation and differentiation. Biocompatibility was further confirmed in vitro and in vivo. We replaced 80% of the left hemidiaphragm with reseeded diaphragmatic scaffolds. After three weeks, transplanted animals gained 32% weight, showed myography, spirometry parameters, and histological evaluations similar to native rats. In conclusion, our study suggested that reseeded decellularized diaphragmatic tissue appears to be a promising option for patients in need of diaphragmatic reconstruction.


Subject(s)
Diaphragm/transplantation , Mesenchymal Stem Cell Transplantation/methods , Tissue Engineering/methods , Tissue Scaffolds , Absorbable Implants , Allografts , Animals , Bioreactors , Cell Adhesion , Cell Differentiation , Diaphragm/blood supply , Diaphragm/diagnostic imaging , Diaphragm/immunology , Electromyography , Graft Survival , Hernias, Diaphragmatic, Congenital , Macrophages/immunology , Male , Neovascularization, Physiologic , Radiography , Rats , Rats, Inbred Lew , Tissue Engineering/instrumentation , Transplantation, Heterotopic , Transplants/blood supply , Transplants/immunology , Transplants/physiology , Wound Healing
10.
Respiration ; 90(6): 481-92, 2015.
Article in English | MEDLINE | ID: mdl-26613253

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a devastating disorder. Despite enormous efforts in clinical research, effective treatment options are lacking, and mortality rates remain unacceptably high. OBJECTIVES: A male patient with severe ARDS showed no clinical improvement with conventional therapies. Hence, an emergent experimental intervention was performed. METHODS: We performed intratracheal administration of autologous peripheral blood-derived mononuclear cells (PBMCs) and erythropoietin (EPO). RESULTS: We found that after 2 days of initial PBMC/EPO application, lung function improved and extracorporeal membrane oxygenation (ECMO) support was reduced. Bronchoscopy and serum inflammatory markers revealed reduced inflammation. Additionally, serum concentration of miR-449a, b, c and miR-34a, a transient upregulation of E-cadherin and associated chromatin marks in PBMCs indicated airway epithelial differentiation. Extracellular vesicles from PBMCs demonstrated anti-inflammatory capacity in a TNF-α-mediated nuclear factor-x03BA;B in vitro assay. Despite improving respiratory function, the patient died of multisystem organ failure on day 38 of ECMO treatment. CONCLUSIONS: This case report provides initial encouraging evidence to use locally instilled PBMC/EPO for treatment of severe refractory ARDS. The observed clinical improvement may partially be due to the anti-inflammatory effects of PBMC/EPO to promote tissue regeneration. Further studies are needed for more in-depth understanding of the underlying mechanisms of in vivo regeneration.


Subject(s)
Leukocytes, Mononuclear/transplantation , Respiratory Distress Syndrome/therapy , Cadherins/blood , Cytokines/blood , Down-Regulation , Erythropoietin/administration & dosage , Extracorporeal Membrane Oxygenation , Fatal Outcome , Humans , Male , MicroRNAs/blood , Multiple Organ Failure/etiology , Snail Family Transcription Factors , Transcription Factors/blood , Transplantation, Autologous , Up-Regulation , Young Adult
11.
J Extracell Vesicles ; 4: 26316, 2015.
Article in English | MEDLINE | ID: mdl-25899407

ABSTRACT

Extracellular vesicles (EVs) have emerged as important mediators of intercellular communication in a diverse range of biological processes. For future therapeutic applications and for EV biology research in general, understanding the in vivo fate of EVs is of utmost importance. Here we studied biodistribution of EVs in mice after systemic delivery. EVs were isolated from 3 different mouse cell sources, including dendritic cells (DCs) derived from bone marrow, and labelled with a near-infrared lipophilic dye. Xenotransplantation of EVs was further carried out for cross-species comparison. The reliability of the labelling technique was confirmed by sucrose gradient fractionation, organ perfusion and further supported by immunohistochemical staining using CD63-EGFP probed vesicles. While vesicles accumulated mainly in liver, spleen, gastrointestinal tract and lungs, differences related to EV cell origin were detected. EVs accumulated in the tumour tissue of tumour-bearing mice and, after introduction of the rabies virus glycoprotein-targeting moiety, they were found more readily in acetylcholine-receptor-rich organs. In addition, the route of administration and the dose of injected EVs influenced the biodistribution pattern. This is the first extensive biodistribution investigation of EVs comparing the impact of several different variables, the results of which have implications for the design and feasibility of therapeutic studies using EVs.

12.
PLoS One ; 9(9): e107712, 2014.
Article in English | MEDLINE | ID: mdl-25229469

ABSTRACT

Stem cells contribute to regeneration of tissues and organs. Cells with stem cell-like properties have been identified in tumors from a variety of origins, but to our knowledge there are yet no reports on tumor-related stem cells in the human upper respiratory tract. In the present study, we show that a tracheal mucoepidermoid tumor biopsy obtained from a 6 year-old patient contained a subpopulation of cells with morphology, clonogenicity and surface markers that overlapped with bone marrow mesenchymal stromal cells (BM-MSCs). These cells, designated as MEi (mesenchymal stem cell-like mucoepidermoid tumor) cells, could be differentiated towards mesenchymal lineages both with and without induction, and formed spheroids in vitro. The MEi cells shared several multipotent characteristics with BM-MSCs. However, they displayed differences to BM-MSCs in growth kinectics and gene expression profiles relating to cancer pathways and tube development. Despite this, the MEi cells did not possess in vivo tumor-initiating capacity, as proven by the absence of growth in situ after localized injection in immunocompromised mice. Our results provide an initial characterization of benign tracheal cancer-derived niche cells. We believe that this report could be of importance to further understand tracheal cancer initiation and progression as well as therapeutic development.


Subject(s)
Mucoepidermoid Tumor/pathology , Neoplastic Stem Cells/pathology , Tracheal Neoplasms/pathology , Animals , Cell Separation , Child , Female , Gene Expression Profiling , Genomics , Humans , Male , Mesenchymal Stem Cells/pathology , Mice , Mucoepidermoid Tumor/diagnosis , Mucoepidermoid Tumor/genetics , Tracheal Neoplasms/diagnosis , Tracheal Neoplasms/genetics
13.
Nat Protoc ; 9(9): 2164-79, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25122525

ABSTRACT

Tissue-engineered tracheal transplants have been successfully performed clinically. However, before becoming a routine clinical procedure, further preclinical studies are necessary to determine the underlying mechanisms of in situ tissue regeneration. Here we describe a protocol using a tissue engineering strategy and orthotopic transplantation of either natural decellularized donor tracheae or artificial electrospun nanofiber scaffolds into a rat model. The protocol includes details regarding how to assess the scaffolds' biomechanical properties and cell viability before implantation. It is a reliable and reproducible model that can be used to investigate the crucial aspects and pathways of in situ tracheal tissue restoration and regeneration. The model can be established in <6 months, and it may also provide a means to investigate cell-surface interactions, cell differentiation and stem cell fate.


Subject(s)
Guided Tissue Regeneration/methods , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Trachea/physiology , Animals , Biomechanical Phenomena , Colorimetry , Nanofibers/therapeutic use , Rats , Trachea/transplantation
14.
Nat Commun ; 5: 3562, 2014 04 15.
Article in English | MEDLINE | ID: mdl-24736316

ABSTRACT

A tissue-engineered oesophageal scaffold could be very useful for the treatment of pediatric and adult patients with benign or malignant diseases such as carcinomas, trauma or congenital malformations. Here we decellularize rat oesophagi inside a perfusion bioreactor to create biocompatible biological rat scaffolds that mimic native architecture, resist mechanical stress and induce angiogenesis. Seeded allogeneic mesenchymal stromal cells spontaneously differentiate (proven by gene-, protein and functional evaluations) into epithelial- and muscle-like cells. The reseeded scaffolds are used to orthotopically replace the entire cervical oesophagus in immunocompetent rats. All animals survive the 14-day study period, with patent and functional grafts, and gain significantly more weight than sham-operated animals. Explanted grafts show regeneration of all the major cell and tissue components of the oesophagus including functional epithelium, muscle fibres, nerves and vasculature. We consider the presented tissue-engineered oesophageal scaffolds a significant step towards the clinical application of bioengineered oesophagi.


Subject(s)
Esophagus/transplantation , Mesenchymal Stem Cells , Tissue Engineering/methods , Tissue Scaffolds , Animals , Cell Differentiation , Esophagus/pathology , Immunocompetence , Myocytes, Smooth Muscle/pathology , Rats , Regeneration
15.
Biomaterials ; 35(20): 5307-5315, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24703872

ABSTRACT

The development of tracheal scaffolds fabricated based on electrospinning technique by applying different ratios of polyethylene terephthalate (PET) and polyurethane (PU) is introduced here. Prior to clinical implantation, evaluations of biomechanical and morphological properties, as well as biocompatibility and cell adhesion verifications are required and extensively performed on each scaffold type. However, the need for bioreactors and large cell numbers may delay the verification process during the early assessment phase. Hence, we investigated the feasibility of performing biocompatibility verification using static instead of dynamic culture. We performed bioreactor seeding on 3-dimensional (3-D) tracheal scaffolds (PET/PU and PET) and correlated the quantitative and qualitative results with 2-dimensional (2-D) sheets seeded under static conditions. We found that an 8-fold reduction for 2-D static seeding density can essentially provide validation on the qualitative and quantitative evaluations for 3-D scaffolds. In vitro studies revealed that there was notably better cell attachment on PET sheets/scaffolds than with the polyblend. However, the in vivo outcomes of cell seeded PET/PU and PET scaffolds in an orthotopic transplantation model in rodents were similar. They showed that both the scaffold types satisfied biocompatibility requirements and integrated well with the adjacent tissue without any observation of necrosis within 30 days of implantation.


Subject(s)
Biocompatible Materials/chemistry , Polymers/chemistry , Tissue Scaffolds/chemistry , Trachea , Animals , Bioreactors , Cell Adhesion , Cell Count , Male , Mesenchymal Stem Cells/cytology , Microscopy, Electron, Scanning , Polyethylene Terephthalates/chemistry , Polyurethanes/chemistry , Rats , Rats, Sprague-Dawley , Tissue Engineering/methods
16.
J Math Biol ; 68(4): 785-813, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23412665

ABSTRACT

A mathematical model, in the form of an integro-partial differential equation, is presented to describe the dynamics of cells being deposited, attaching and growing in the form of a monolayer across an adherent surface. The model takes into account that the cells suspended in the media used for the seeding have a distribution of sizes, and that the attachment of cells restricts further deposition by fragmenting the parts of the domain unoccupied by cells. Once attached the cells are assumed to be able to grow and proliferate over the domain by a process of infilling of the interstitial gaps; it is shown that without cell proliferation there is a slow build up of the monolayer but if the surface is conducive to cell spreading and proliferation then complete coverage of the domain by the monolayer can be achieved more rapidly. Analytical solutions of the model equations are obtained for special cases, and numerical solutions are presented for parameter values derived from experiments of rat mesenchymal stromal cells seeded onto thin layers of collagen-coated polyethylene terephthalate electrospun fibers. The model represents a new approach to describing the deposition, attachment and growth of cells over adherent surfaces, and should prove useful for studying the dynamics of the seeding of biomaterials.


Subject(s)
Cell Adhesion/physiology , Mesenchymal Stem Cells/physiology , Models, Biological , Animals , Cell Proliferation , Numerical Analysis, Computer-Assisted , Rats
17.
Biomaterials ; 35(6): 1907-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24321707

ABSTRACT

Aortic valve degeneration and dysfunction is one of the leading causes for morbidity and mortality. The conventional heart-valve prostheses have significant limitations with either life-long anticoagulation therapeutic associated bleeding complications (mechanical valves) or limited durability (biological valves). Tissue engineered valve replacement recently showed encouraging results, but the unpredictable outcome of tissue degeneration is likely associated to the extensive tissue processing methods. We believe that optimized decellularization procedures may provide aortic valve/root grafts improved durability. We present an improved/innovative decellularization approach using a detergent-enzymatic perfusion method, which is both quicker and has less exposure of matrix degenerating detergents, compared to previous protocols. The obtained graft was characterized for its architecture, extracellular matrix proteins, mechanical and immunological properties. We further analyzed the engineered aortic root for biocompatibility by cell adhesion and viability in vitro and heterotopic implantation in vivo. The developed decellularization protocol was substantially reduced in processing time whilst maintaining tissue integrity. Furthermore, the decellularized aortic root remained bioactive without eliciting any adverse immunological reaction. Cell adhesion and viability demonstrated the scaffold's biocompatibility. Our optimized decellularization protocol may be useful to develop the next generation of clinical valve prosthesis with a focus on improved mechanical properties and durability.


Subject(s)
Aortic Valve/cytology , Tissue Engineering/methods , Animals , Cell Adhesion/physiology , Cell Survival/physiology , Cells, Cultured , Detergents , Immunohistochemistry , Mesenchymal Stem Cells/cytology
18.
Biomaterials ; 34(16): 4057-4067, 2013 May.
Article in English | MEDLINE | ID: mdl-23473965

ABSTRACT

The clinical outcome of transplantations of bioartificial tissues and organs depends on the presence of living cells. There are still no standard operative protocols that are simple, fast and reliable for confirming the presence of viable cells on bioartificial scaffolds prior to transplantation. By using mathematical modeling, we have developed a colorimetric-based system (colorimetric scale bar) to predict the cell viability and density for sufficient surface coverage. First, we refined a method which can provide information about cell viability and numbers in an in vitro setting: i) immunohistological staining by Phalloidin/DAPI and ii) a modified colorimetric cell viability assay. These laboratory-based methods and the developed colorimetric-based system were then validated in rat transplantation studies of unseeded and seeded tracheal grafts. This was done to provide critical information on whether the graft would be suitable for transplantation or if additional cell seeding was necessary. The potential clinical impact of the colorimetric scale bar was confirmed using patient samples. In conclusion, we have developed a robust, fast and reproducible colorimetric tool that can verify and warrant viability and integrity of an engineered tissue/organ prior to transplantation. This should facilitate a successful transplantation outcome and ensure patient safety.


Subject(s)
Bioartificial Organs , Bioengineering/methods , Transplantation , Animals , Cell Adhesion/drug effects , Cell Count , Cell Survival/drug effects , Female , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Microscopy, Electron, Scanning , Polyurethanes/pharmacology , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Tissue Scaffolds/chemistry , Trachea/physiology , Translational Research, Biomedical , Young Adult
19.
Biomaterials ; 33(32): 8094-103, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22901964

ABSTRACT

In 2011, the first in-man successful transplantation of a tissue engineered trachea-bronchial graft, using a synthetic POSS-PCU nanocomposite construct seeded with autologous stem cells, was performed. To further improve this technology, we investigated the feasibility of using polymers with a three dimensional structure more closely mimicking the morphology and size scale of native extracellular matrix (ECM) fibers. We therefore investigated the in vitro biocompatibility of electrospun polyethylene terephthalate (PET) and polyurethane (PU) scaffolds, and determined the effects on cell attachment by conditioning the fibers with adhesion proteins. Rat mesenchymal stromal cells (MSCs) were seeded on either PET or PU fiber-layered culture plates coated with laminin, collagen I, fibronectin, poly-D-lysine or gelatin. Cell density, proliferation, viability, morphology and mRNA expression were evaluated. MSC cultures on PET and PU resulted in similar cell densities and amounts of proliferating cells, with retained MSC phenotype compared to data obtained from tissue culture plate cultures. Coating the scaffolds with adhesion proteins did not increase cell density or cell proliferation. Our data suggest that both PET and PU mats, matching the dimensions of ECM fibers, are biomimetic scaffolds and, because of their high surface area-to-volume provided by the electrospinning procedure, makes them per se suitable for cell attachment and proliferation without any additional coating.


Subject(s)
Coated Materials, Biocompatible/metabolism , Mesenchymal Stem Cells/cytology , Polyethylene Terephthalates/metabolism , Polyurethanes/metabolism , Tissue Scaffolds/chemistry , Animals , Cell Adhesion , Cell Proliferation , Cell Survival , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Male , Mesenchymal Stem Cells/metabolism , Polyethylene Terephthalates/chemistry , Polyurethanes/chemistry , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL