Change search
Link to record
Permanent link

Direct link
Publications (10 of 21) Show all publications
Österberg, K., Bogestål, Y., Jenndahl, L., Gustafsson-Hedberg, T., Synnergren, J., Holmgren, G., . . . Håkansson, J. (2023). Personalized tissue-engineered veins - long term safety, functionality and cellular transcriptome analysis in large animals. Biomaterials Science, 11(11), 3860-3877
Open this publication in new window or tab >>Personalized tissue-engineered veins - long term safety, functionality and cellular transcriptome analysis in large animals
Show others...
2023 (English)In: Biomaterials Science, ISSN 2047-4830, E-ISSN 2047-4849, Vol. 11, no 11, p. 3860-3877Article in journal (Refereed) Published
Abstract [en]

Tissue engineering is a promising methodology to produce advanced therapy medicinal products (ATMPs). We have developed personalized tissue engineered veins (P-TEV) as an alternative to autologous or synthetic vascular grafts utilized in reconstructive vein surgery. Our hypothesis is that individualization through reconditioning of a decellularized allogenic graft with autologous blood will prime the tissue for efficient recellularization, protect the graft from thrombosis, and decrease the risk of rejection. In this study, P-TEVs were transplanted to vena cava in pig, and the analysis of three veins after six months, six veins after 12 months and one vein after 14 months showed that all P-TEVs were fully patent, and the tissue was well recellularized and revascularized. To confirm that the ATMP product had the expected characteristics one year after transplantation, gene expression profiling of cells from P-TEV and native vena cava were analyzed and compared by qPCR and sequencing. The qPCR and bioinformatics analysis confirmed that the cells from the P-TEV were highly similar to the native cells, and we therefore conclude that P-TEV is functional and safe in large animals and have high potential for use as a clinical transplant graft.

Place, publisher, year, edition, pages
NLM (Medline), 2023
Keywords
animal, endothelium cell, gene expression profiling, pig, procedures, tissue engineering, transplantation, vein, Animals, Endothelial Cells, Swine, Veins
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:ri:diva-65415 (URN)10.1039/d2bm02011d (DOI)2-s2.0-85160870522 (Scopus ID)
Note

This study was supported by Vinnova project CAMP (contract no. 2017-02130), a common call by VINNOVA and Vetenskapsrådet: Biologcal pharmaseuticals (Dnr 2017-02983),by University of Skövde under grants from the Swedish Knowledge Foundation [#2016-0330, #2020-0014] and Västra Götalandsregionen (consultant check).

Available from: 2023-06-15 Created: 2023-06-15 Last updated: 2023-12-22Bibliographically approved
Jenndahl, L., Österberg, K., Bogestål, Y., Simsa, R., Gustafsson-Hedberg, T., Stenlund, P., . . . Håkansson, J. (2022). Personalized tissue-engineered arteries as vascular graft transplants: A safety study in sheep. Regenerative Therapy, 21, 331-341
Open this publication in new window or tab >>Personalized tissue-engineered arteries as vascular graft transplants: A safety study in sheep
Show others...
2022 (English)In: Regenerative Therapy, ISSN 2352-3204, Vol. 21, p. 331-341Article in journal (Refereed) Published
Abstract [en]

Patients with cardiovascular disease often need replacement or bypass of a diseased blood vessel. With disadvantages of both autologous blood vessels and synthetic grafts, tissue engineering is emerging as a promising alternative of advanced therapy medicinal products for individualized blood vessels. By reconditioning of a decellularized blood vessel with the recipient's own peripheral blood, we have been able to prevent rejection without using immunosuppressants and prime grafts for efficient recellularization in vivo. Recently, decellularized veins reconditioned with autologous peripheral blood were shown to be safe and functional in a porcine in vivo study as a potential alternative for vein grafting. In this study, personalized tissue engineered arteries (P-TEA) were developed using the same methodology and evaluated for safety in a sheep in vivo model of carotid artery transplantation. Five personalized arteries were transplanted to carotid arteries and analyzed for safety and patency as well as with histology after four months in vivo. All grafts were fully patent without any occlusion or stenosis. The tissue was well cellularized with a continuous endothelial cell layer covering the luminal surface, revascularized adventitia with capillaries and no sign of rejection or infection. In summary, the results indicate that P-TEA is safe to use and has potential as clinical grafts. 

Place, publisher, year, edition, pages
Japanese Society of Regenerative Medicine, 2022
Keywords
ATMP, Blood vessels, Recellularization, Regenerative medicine, Scaffold, Tissue engineering, amoxicillin, attane, buprenorphine, carprofen, clopidogrel, dalteparin, dexmedetomidine, heparin, isoflurane, norocarp, poliglecaprone, promea, propofol, receptor type tyrosine protein phosphatase C, vetergesic, vetrimoxin, adventitia, anatomical concepts, angiography, animal experiment, animal model, animal tissue, antibiotic prophylaxis, arterial stiffness, artery anastomosis, artery transplantation, Article, biomechanics, biopsy technique, blood flow, blood vessel graft, blood vessel parameters, blood vessel wall, burst pressure, capillary, carotid artery transplantation, confocal laser scanning microscopy, controlled study, decellularization, decellularized blood vessel, device safety, DNA extraction, end to end anastomosis, endothelial cell layer, evening dosage, extracellular matrix, failure strain, female, fluorescence microscopy, full luminal endothelialization, histology, hyperplasia, immunohistochemistry, intimal hyperplasia, luminal surface, medical procedures, morning dosage, nonhuman, reconditioning of vascular graft, revascularization, scanning electron microscopy
National Category
Clinical Medicine
Identifiers
urn:nbn:se:ri:diva-60256 (URN)10.1016/j.reth.2022.08.005 (DOI)2-s2.0-85137671619 (Scopus ID)
Note

Funding details: Horizon 2020 Framework Programme, H2020; Funding details: H2020 Marie Skłodowska-Curie Actions, MSCA, 722779; Funding details: VINNOVA, Dnr 2017-01413, Dnr 2017–02983; Funding text 1: We want to acknowledge the staff at the Department of Experimental Biomedicine at Gothenburg University. This study was partly performed by funding from VINNOVA (Dnr 2017–02983 and Dnr 2017-01413) and Region Västra Götalandsregionen (consultant check) as well as from the European Union's Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement No 722779 conducted within the “Training 4 Cell Regenerative Medicine” (T4CRM) network. The company VERIGRAFT AB holds a patent on peripheral whole blood perfusion of decellularized tissues and did also finance the project.; Funding text 2: We want to acknowledge the staff at the Department of Experimental Biomedicine at Gothenburg University. This study was partly performed by funding from VINNOVA ( Dnr 2017–02983 and Dnr 2017-01413 ) and Region Västra Götalandsregionen (consultant check) as well as from the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement No 722779 conducted within the “Training 4 Cell Regenerative Medicine” (T4CRM) network. The company VERIGRAFT AB holds a patent on peripheral whole blood perfusion of decellularized tissues and did also finance the project.

Available from: 2022-10-10 Created: 2022-10-10 Last updated: 2023-10-05Bibliographically approved
Rosendahl, J., Svanström, A., Berglin, M., Petronis, S., Bogestål, Y., Stenlund, P., . . . Håkansson, J. (2021). 3D Printed Nanocellulose Scaffolds as a Cancer Cell Culture Model System. Bioengineering, 8(7), Article ID 97.
Open this publication in new window or tab >>3D Printed Nanocellulose Scaffolds as a Cancer Cell Culture Model System
Show others...
2021 (English)In: Bioengineering, E-ISSN 2306-5354, Vol. 8, no 7, article id 97Article in journal (Refereed) Published
Abstract [en]

Current conventional cancer drug screening models based on two-dimensional (2D) cell culture have several flaws and there is a large need of more in vivo mimicking preclinical drug screening platforms. The microenvironment is crucial for the cells to adapt relevant in vivo characteristics and here we introduce a new cell culture system based on three-dimensional (3D) printed scaffolds using cellulose nanofibrils (CNF) pre-treated with 2,2,6,6-tetramethylpyperidine-1-oxyl (TEMPO) as the structural material component. Breast cancer cell lines, MCF7 and MDA-MB-231, were cultured in 3D TEMPO-CNF scaffolds and were shown by scanning electron microscopy (SEM) and histochemistry to grow in multiple layers as a heterogenous cell population with different morphologies, contrasting 2D cultured mono-layered cells with a morphologically homogenous cell population. Gene expression analysis demonstrated that 3D TEMPO-CNF scaffolds induced elevation of the stemness marker CD44 and the migration markers VIM and SNAI1 in MCF7 cells relative to 2D control. T47D cells confirmed the increased level of the stemness marker CD44 and migration marker VIM which was further supported by increased capacity of holoclone formation for 3D cultured cells. Therefore, TEMPO-CNF was shown to represent a promising material for 3D cell culture model systems for cancer cell applications such as drug screening.

Keywords
nanocellulose, 3D printing, cancer, 3D cell culture, CNF, cancer stemness
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:ri:diva-55646 (URN)10.3390/bioengineering8070097 (DOI)
Available from: 2021-08-05 Created: 2021-08-05 Last updated: 2023-06-07Bibliographically approved
Håkansson, J., Simsa, R., Bogestål, Y., Jenndahl, L., Gustafsson-Hedberg, T., Petronis, S., . . . Österberg, K. (2021). Individualized tissue-engineered veins as vascular grafts: a proof of concept study in pig.. Journal of Tissue Engineering and Regenerative Medicine, 15(10), 818
Open this publication in new window or tab >>Individualized tissue-engineered veins as vascular grafts: a proof of concept study in pig.
Show others...
2021 (English)In: Journal of Tissue Engineering and Regenerative Medicine, ISSN 1932-6254, E-ISSN 1932-7005, Vol. 15, no 10, p. 818-Article in journal (Refereed) Published
Abstract [en]

Personalized tissue engineered vascular grafts are a promising advanced therapy medicinal product (ATMP) alternative to autologous or synthetic vascular grafts utilized in blood vessel bypass or replacement surgery. We hypothesized that an individualized tissue engineered vein (P-TEV) would make the body recognize the transplanted blood vessel as autologous, decrease the risk of rejection and thereby avoid lifelong treatment with immune suppressant medication as is standard with allogenic organ transplantation. To individualize blood vessels, we decellularized vena cava from six deceased donor pigs and tested them for cellular removal and histological integrity. A solution with peripheral blood from the recipient pigs was used for individualized reconditioning in a perfusion bioreactor for seven days prior to transplantation. To evaluate safety and functionality of the individualized vascular graft in vivo, we transplanted reconditioned porcine vena cava into six pigs and analyzed histology and patency of the graft at different time points, with three pigs at the final endpoint 4-5 weeks after surgery. Our results showed that the P-TEV was fully patent in all animals, did not induce any occlusion or stenosis formation and we did not find any signs of rejection. The P-TEV showed rapid recellularization in vivo with the luminal surface covered with endothelial cells. In summary, the results indicate that P-TEV is functional and have potential for use as clinical transplant grafts. 

Keywords
ATMP, Blood vessels, Recellularization, Regenerative medicine, Scaffold, Tissue engineering, decellularization, recellularization, tissue engineering, vascular grafts
National Category
Surgery
Identifiers
urn:nbn:se:ri:diva-55773 (URN)10.1002/term.3233 (DOI)34318614 (PubMedID)
Available from: 2021-08-09 Created: 2021-08-09 Last updated: 2023-06-07Bibliographically approved
Karazisis, D., Omar, O., Petronis, S., Thomsen, P. & Rasmusson, L. (2021). Molecular Response to Nanopatterned Implants in the Human Jaw Bone. ACS Biomaterials Science & Engineering, 7(12), 5878-5889
Open this publication in new window or tab >>Molecular Response to Nanopatterned Implants in the Human Jaw Bone
Show others...
2021 (English)In: ACS Biomaterials Science & Engineering, E-ISSN 2373-9878, Vol. 7, no 12, p. 5878-5889Article in journal (Refereed) Published
Abstract [en]

Implant surface modification by nanopatterning is an interesting route for enhancing osseointegration in humans. Herein, the mol. response to an intentional, controlled nanotopog. pattern superimposed on screw-shaped titanium implants is investigated in human bone. When clin. implants are installed, addnl. two mini-implants, one with a machined surface (M) and one with a machined surface superimposed with a hemispherical nanopattern (MN), are installed in the posterior maxilla. In the second-stage surgery, after 6-8 wk, the mini-implants are retrieved by unscrewing, and the implant-adherent cells are subjected to gene expression anal. using quant. polymerase chain reaction (qPCR). Compared to those adherent to the machined (M) implants, the cells adherent to the nanopatterned (MN) implants demonstrate significant upregulation (1.8- to 2-fold) of bone-related genes (RUNX2, ALP, and OC). No significant differences are observed in the expression of the analyzed inflammatory and remodeling genes. Correlation anal. reveals that older patient age is associated with increased expression of proinflammatory cytokines (TNF-α and MCP-1) on the machined implants and decreased expression of pro-osteogenic factor (BMP-2) on the nanopatterned implants. Controlled nanotopog., in the form of hemispherical 60 nm protrusions, promotes gene expressions related to early osteogenic differentiation and osteoblastic activity in implant-adherent cells in the human jaw bone.

Place, publisher, year, edition, pages
American Chemical Society, 2021
Keywords
gene expression, human jaw, nanomedicine, nanotopography, osseointegration, osteogenic activities
National Category
Biomaterials Science
Identifiers
urn:nbn:se:ri:diva-57561 (URN)10.1021/acsbiomaterials.1c00861 (DOI)
Available from: 2022-01-07 Created: 2022-01-07 Last updated: 2023-06-07Bibliographically approved
Svanström, A., Rosendahl, J., Salerno, S., Leiva, M., Gregersson, P., Berglin, M., . . . Landberg, G. (2021). Optimized alginate-based 3D printed scaffolds as a model of patient derived breast cancer microenvironments in drug discovery. Biomedical Materials, 16(4), Article ID 045046.
Open this publication in new window or tab >>Optimized alginate-based 3D printed scaffolds as a model of patient derived breast cancer microenvironments in drug discovery
Show others...
2021 (English)In: Biomedical Materials, ISSN 1748-6041, E-ISSN 1748-605X, Vol. 16, no 4, article id 045046Article in journal (Refereed) Published
Abstract [en]

The cancer microenvironment influences tumor progression and metastasis and is pivotal to consider when designing in vivo-like cancer models. Current preclinical testing platforms for cancer drug development are mainly limited to 2D cell culture systems that poorly mimic physiological environments and traditional, low throughput animal models. The aim of this work was to produce a tunable testing platform based on 3D printed scaffolds (3DPS) with a simple geometry that, by extracellular components and response of breast cancer reporter cells, mimics patient-derived scaffolds (PDS) of breast cancer. Here, the biocompatible polysaccharide alginate was used as base material to generate scaffolds consisting of a 3D grid containing periostin and hydroxyapatite. Breast cancer cell lines (MCF7 and MDA-MB-231) produced similar phenotypes and gene expression levels of cancer stem cell, epithelial-mesenchymal transition, differentiation and proliferation markers when cultured on 3DPS and PDS, contrasting conventional 2D cultures. Importantly, cells cultured on 3DPS and PDS showed scaffold-specific responses to cytotoxic drugs (doxorubicin and 5-fluorouracil) that were different from 2D cultured cells. In conclusion, the data presented support the use of a tunable alginate-based 3DPS as a tumor model in breast cancer drug discovery. © 2021 The Author(s).

Place, publisher, year, edition, pages
IOP Publishing Ltd, 2021
Keywords
Alginate, Animal cell culture, Biocompatibility, Diseases, Drug products, Gene expression, Hydroxyapatite, Physiological models, Scaffolds (biology), Stem cells, Tumors, Biocompatible polysaccharides, Breast cancer cells, Differentiation and proliferations, Epithelial-mesenchymal transition, Gene expression levels, Physiological environment, Pre-clinical testing, Tumor progressions, 3D printers
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:ri:diva-55478 (URN)10.1088/1748-605X/ac0451 (DOI)2-s2.0-85109424226 (Scopus ID)
Available from: 2021-08-06 Created: 2021-08-06 Last updated: 2023-06-07Bibliographically approved
Knutsen, M., Agrenius, K., Ugland, H., Petronis, S., Haglerod, C., Håkansson, J. & Chinga-Carrasco, G. (2021). Oxygenated Nanocellulose - A Material Platform for Antibacterial Wound Dressing Devices. ACS Applied Bio Materials, 4(10), 7554-7562
Open this publication in new window or tab >>Oxygenated Nanocellulose - A Material Platform for Antibacterial Wound Dressing Devices
Show others...
2021 (English)In: ACS Applied Bio Materials, E-ISSN 2576-6422, Vol. 4, no 10, p. 7554-7562Article in journal (Refereed) Published
Abstract [en]

Both carboxylated cellulose nanofibrils (CNF) and dissolved oxygen (DO) have been reported to possess antibacterial properties. However, the combination for use as wound dressings against biofilm infections in chronic wounds is less known. The present study reports the development of oxygenated CNF dispersions that exhibit strong antibacterial effect. Carboxylated CNF dispersions with different oxidation levels were oxygenated by the OXY BIO System and tested for antibacterial activity against Pseudomonas aeruginosa and Staphylococcus aureus. The results reveal that the higher oxidation level of the CNFs, the better antibacterial effect. Scanning electron microscopy of bacterial biofilms revealed that a potential mechanism of action of the CNFs is the formation of a network surrounding and entrapping the bacteria. This effect is further potentiated by the oxygenation process. A CNF sample (concentration 0.6 wt %) that was oxygenated to a DO level of 46.4 mg/L demonstrated a strong antibacterial effect against S. aureus in vivo using a mouse model of surgical site infection. The oxygenated CNF dispersion reduced the bacterial survival by 71%, after 24 h treatment. The potent antibacterial effect indicates that oxygenated nanocellulose is a promising material for antibacterial wound dressings. © 2021 The Authors.

Place, publisher, year, edition, pages
American Chemical Society, 2021
Keywords
antibacterial properties, biofilm infections, hyperoxia, nanocellulose, topical dressings, wound healing, Biofilms, Dispersions, Dissolved oxygen, Scanning electron microscopy, Antibacterial effects, Antibacterials, Cellulose nanofibrils, Nano-cellulose, Topical dressing, Wound dressings, Bacteria, Diseases, Formation, Oxidation, Processes, Surgical Dressings, Survival
National Category
Biomaterials Science
Identifiers
urn:nbn:se:ri:diva-56894 (URN)10.1021/acsabm.1c00819 (DOI)2-s2.0-85117321748 (Scopus ID)
Note

 Funding details: Norges Forskningsråd, 283895, 309178; Funding text 1: This work was partly funded by the MANUNET III program (Project No. MNET17/NMCS-1204) and Research Council of Norway Grant No. 283895, MedIn project (“New functionalized medical devices for surgical interventions in the pelvic cavity”, Grant No. 283895), and the OxyPol project (“Oxygenated biopolymers for biomedical applications”, Grant No. 309178).

Available from: 2021-11-22 Created: 2021-11-22 Last updated: 2023-11-21Bibliographically approved
Karazisis, D., Rasmusson, L., Petronis, S., Palmquist, A., Shah, F., Agheli, H., . . . Thomsen, P. (2021). The effects of controlled nanotopography, machined topography and their combination on molecular activities, bone formation and biomechanical stability during osseointegration. Acta Biomaterialia, 136, 279-290
Open this publication in new window or tab >>The effects of controlled nanotopography, machined topography and their combination on molecular activities, bone formation and biomechanical stability during osseointegration
Show others...
2021 (English)In: Acta Biomaterialia, ISSN 1742-7061, E-ISSN 1878-7568, Vol. 136, p. 279-290Article in journal (Refereed) Published
Abstract [en]

The initial cellular and molecular activities at the bone interface of implants with controlled nanoscale topography and microscale roughness have previously been reported. However, the effects of such surface modifications on the development of osseointegration have not yet been determined. This study investigated the molecular events and the histological and biomechanical development of the bone interface in implants with nanoscale topography, microscale roughness or a combination of both. Polished and machined titanium implants with and without controlled nanopatterning (75 nm protrusions) were produced using colloidal lithography and coated with a thin titanium layer to unify the chemistry. The implants were inserted in rat tibiae and subjected to removal torque (RTQ) measurements, molecular analyses and histological analyses after 6, 21 and 28 days. The results showed that nanotopography superimposed on microrough, machined, surfaces promoted an early increase in RTQ and hence produced greater implant stability at 6 and 21 days. Two-way MANOVA revealed that the increased RTQ was influenced by microscale roughness and the combination of nanoscale and microscale topographies. Furthermore, increased bone-implant contact (BIC) was observed with the combined nanopatterned machined surface, although MANOVA results implied that the increased BIC was mainly dependent on microscale roughness. At the molecular level, the nanotopography, per se, and in synergy with microscale roughness, downregulated the expression of the proinflammatory cytokine tumor necrosis factor alpha (TNF-α). In conclusion, controlled nanotopography superimposed on microrough machined implants promoted implant stability during osseointegration. Nanoscale-driven mechanisms may involve attenuation of the inflammatory response at the titanium implant site. Statement of Significance: The role of combined implant microscale and nanotopography features for osseointegration is incompletely understood. Using colloidal lithography technique, we created an ordered nanotopography pattern superimposed on screwshaped implants with microscale topography. The midterm and late molecular, bone-implant contact and removal torque responses were analysed in vivo. Nanotopography superimposed on microrough, machined, surfaces promoted the implant stability, influenced by microscale topography and the combination of nanoscale and microscale topographies. Increased bone-implant contact was mainly dependent on microscale roughness whereas the nanotopography, per se, and in synergy with microscale roughness, attenuated the proinflammatory tumor necrosis factor alpha (TNF-α) expression. It is concluded that microscale and nanopatterns provide individual as well as synergistic effects on molecular, morphological and biomechanical implant-tissue processes in vivo. © 2021 The Author(s)

Place, publisher, year, edition, pages
Acta Materialia Inc, 2021
Keywords
Colloidal lithography, Cytokines, Gene expression, Implant, Microroughness, Nanotopography, Osseointegration, Removal torque, Titanium
National Category
Biomaterials Science
Identifiers
urn:nbn:se:ri:diva-56947 (URN)10.1016/j.actbio.2021.10.001 (DOI)2-s2.0-85117085120 (Scopus ID)
Note

 Funding details: ALFGBG-725641; Funding details: Västra Götalandsregionen; Funding details: Stiftelsen Handlanden Hjalmar Svenssons; Funding details: Svenska Sällskapet för Medicinsk Forskning, SSMF; Funding details: IngaBritt och Arne Lundbergs Forskningsstiftelse; Funding details: Vetenskapsrådet, VR, 2018-02891; Funding details: Tokyo University of Agriculture, TUA; Funding text 1: Financial support was provided by the Swedish Research Council ( 2018-02891 ), the BIOMATCELL VINN Excellence Center of Biomaterials and Cell Therapy , the Västra Götaland Region , the Swedish state under the agreement between the Swedish government and the county councils, the ALF agreement ( ALFGBG-725641 ), the T UA/Region Västra Götaland research grant , the Stiftelsen Handlanden Hjalmar Svensson , the IngaBritt and Arne Lundberg Foundation , the Eivind o Elsa K: son Sylvan Foundation and the Area of Advance Materials of Chalmers and GU Biomaterials within the Strategic Research Area initiative launched by the Swedish Government . F.A.S was supported by the Svenska Sällskapet för Medicinsk Forskning (SSMF) postdoctoral scholarship. The sponsors were not involved in the study design; data acquisition; or interpretation, writing or submission of the article.; Funding text 2: Financial support was provided by the Swedish Research Council (2018-02891), the BIOMATCELL VINN Excellence Center of Biomaterials and Cell Therapy, the V?stra G?taland Region, the Swedish state under the agreement between the Swedish government and the county councils, the ALF agreement (ALFGBG-725641), the TUA/Region V?stra G?taland research grant, the Stiftelsen Handlanden Hjalmar Svensson, the IngaBritt and Arne Lundberg Foundation, the Eivind o Elsa K: son Sylvan Foundation and the Area of Advance Materials of Chalmers and GU Biomaterials within the Strategic Research Area initiative launched by the Swedish Government. F.A.S was supported by the Svenska S?llskapet f?r Medicinsk Forskning (SSMF) postdoctoral scholarship. The sponsors were not involved in the study design; data acquisition; or interpretation, writing or submission of the article.

Available from: 2021-11-19 Created: 2021-11-19 Last updated: 2023-06-07Bibliographically approved
Landberg, G., Jonasson, E., Gustafsson, A., Fitzpatrick, P., Isakson, P., Karlsson, J., . . . Ståhlberg, A. (2020). Characterization of cell-free breast cancer patient-derived scaffolds using liquid chromatography-mass spectrometry/mass spectrometry data and RNA sequencing data. Data in Brief, 31, Article ID 105860.
Open this publication in new window or tab >>Characterization of cell-free breast cancer patient-derived scaffolds using liquid chromatography-mass spectrometry/mass spectrometry data and RNA sequencing data
Show others...
2020 (English)In: Data in Brief, E-ISSN 2352-3409, Vol. 31, article id 105860Article in journal (Refereed) Published
Abstract [en]

Patient-derived scaffolds (PDSs) generated from primary breast cancer tumors can be used to model the tumor microenvironment in vitro. Patient-derived scaffolds are generated by repeated detergent washing, removing all cells. Here, we analyzed the protein composition of 15 decellularized PDSs using liquid chromatography-mass spectrometry/mass spectrometry. One hundred forty-three proteins were detected and their relative abundance was calculated using a reference sample generated from all PDSs. We performed heatmap analysis of all the detected proteins to display their expression patterns across different PDSs together with pathway enrichment analysis to reveal which processes that were connected to PDS protein composition. This protein dataset together with clinical information is useful to investigators studying the microenvironment of breast cancers. Further, after repopulating PDSs with either MCF7 or MDA-MB-231 cells, we quantified their gene expression profiles using RNA sequencing. These data were also compared to cells cultured in conventional 2D conditions, as well as to cells cultured as xenografts in immune-deficient mice. We investigated the overlap of genes regulated between these different culture conditions and performed pathway enrichment analysis of genes regulated by both PDS and xenograft cultures compared to 2D in both cell lines to describe common processes associated with both culture conditions. Apart from our described analyses of these systems, these data are useful when comparing different experimental model systems. Downstream data analyses and interpretations can be found in the research article “Patient-derived scaffolds uncover breast cancer promoting properties of the microenvironment” [1]. © 2020 The Authors

Place, publisher, year, edition, pages
Elsevier Inc., 2020
Keywords
3d cell culture, Breast cancer, Extracellular matrix, Liquid chromatography-mass spectrometry/mass spectrometry, Patient-derived scaffolds, RNA sequencing
National Category
Natural Sciences
Identifiers
urn:nbn:se:ri:diva-45151 (URN)10.1016/j.dib.2020.105860 (DOI)2-s2.0-85086837473 (Scopus ID)
Available from: 2020-07-13 Created: 2020-07-13 Last updated: 2023-06-07Bibliographically approved
Newman, D. K., New, P. W., Heriseanu, R., Petronis, S., Håkansson, J., Håkansson, M. Å. & Lee, B. B. (2020). Intermittent catheterization with single- or multiple-reuse catheters: clinical study on safety and impact on quality of life. International Urology and Nephrology, 19(1), 1-153
Open this publication in new window or tab >>Intermittent catheterization with single- or multiple-reuse catheters: clinical study on safety and impact on quality of life
Show others...
2020 (English)In: International Urology and Nephrology, ISSN 0301-1623, E-ISSN 1573-2584, Vol. 19, no 1, p. 1-153Article in journal (Refereed) Published
Abstract [en]

PURPOSE: Intermittent catheterization (IC) is a proven effective long-term bladder management strategy for individuals who have lower urinary tract dysfunction. This study provides clinical evidence about multiple-reuse versus single-use catheterization techniques and if catheter choice can have an impact on health-related quality of life (HRQoL).

METHOD: A prospective, multi-center, clinical trial studied patients who currently practiced catheter reuse, and who agreed to prospectively evaluate single-use hydrophilic-coated (HC) (i.e. LoFric) catheters for 4 weeks. A validated Intermittent Self-Catheterization Questionnaire (ISC-Q) was used to obtain HRQoL. Reused catheters were collected and studied with regard to microbial and debris contamination.

RESULTS: The study included 39 patients who had practiced IC for a mean of 10 years, 6 times daily. At inclusion, all patients reused catheters for a mean of 21 days (SD = 48) per catheter. 36 patients completed the prospective test period and the mean ISC-Q score increased from 58.0 (SD = 22.6) to 67.2 (SD = 17.7) when patients switched to the single-use HC catheters (p = 0.0101). At the end of the study, 83% (95% CI [67-94%]) preferred to continue using single-use HC catheters. All collected reused catheters (100%) were contaminated by debris and 74% (95% CI [58-87%]) were contaminated by microorganisms, some with biofilm.

CONCLUSION: Single-use HC catheters improved HRQoL and were preferred over catheter reuse among people practicing IC. Catheter multiple-reuse may pose a potential safety concern due to colonization by microorganisms as well as having reduced acceptance compared to single use.

TRIAL REGISTRY NUMBER: ClinicalTrials.gov NCT02129738.

Keywords
Catheter-associated urinary tract infection (CAUTI), Clean intermittent catheterization, Lower urinary tract dysfunction (LUTD), Neurogenic lower urinary tract dysfunction (NLUTD), Urinary catheter
National Category
Natural Sciences
Identifiers
urn:nbn:se:ri:diva-44561 (URN)10.1007/s11255-020-02435-9 (DOI)32172456 (PubMedID)
Available from: 2020-03-24 Created: 2020-03-24 Last updated: 2023-06-07Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0003-4592-5851

Search in DiVA

Show all publications