Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Transpl Int ; 36: 11077, 2023.
Article in English | MEDLINE | ID: mdl-37908676

ABSTRACT

Islet delivery devices (IDDs) offer potential benefits for islet transplantation and stem cell-based replacement in type 1 diabetes. Little is known about patient preferences regarding islet delivery device characteristics and implantation strategies. Patient preferences for IDDs and implantation strategies remain understudied. We invited patients, parents and caregivers to fill in an online questionnaire regarding IDDs. An online survey gathered responses from 809 type 1 diabetes patients and 47 caregivers. We also assessed diabetes distress in a subgroup of 412 patients. A significant majority (97%) expressed willingness to receive an IDD. Preferred IDD attributes included a 3.5 cm diameter for 37.7% of respondents, while when provided with all options, 30.4% found dimensions unimportant. Respondents were open to approximately 4 implants, each with a 5 cm incision. Many favored a device functioning for 12 months (33.4%) or 24 months (24.8%). Younger participants (16-30) were more inclined to accept a 6 months functional duration (p < 0.001). Functional duration outweighed implant quantity and size (p < 0.001) in device importance. This emphasizes patients' willingness to accommodate burdens related to IDD features and implantation methods, crucial for designing future beta cell replacement strategies.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Humans , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation/methods , Patient Preference
2.
Transpl Int ; 36: 11633, 2023.
Article in English | MEDLINE | ID: mdl-37822447

ABSTRACT

The field of transplantation has witnessed the emergence of Advanced Therapy Medicinal Products (ATMPs) as highly promising solutions to address the challenges associated with organ and tissue transplantation. ATMPs encompass gene therapy, cell therapy, and tissue-engineered products, hold immense potential for breakthroughs in overcoming the obstacles of rejection and the limited availability of donor organs. However, the development and academic research access to ATMPs face significant bottlenecks that hinder progress. This opinion paper emphasizes the importance of addressing bottlenecks in the development and academic research access to ATMPs by implementing several key strategies. These include the establishment of streamlined regulatory processes, securing increased funding for ATMP research, fostering collaborations and partnerships, setting up centralized ATMP facilities, and actively engaging with patient groups. Advocacy at the policy level is essential to provide support for the development and accessibility of ATMPs, thereby driving advancements in transplantation and enhancing patient outcomes. By adopting these strategies, the field of transplantation can pave the way for the introduction of innovative and efficacious ATMP therapies, while simultaneously fostering a nurturing environment for academic research.


Subject(s)
Cell- and Tissue-Based Therapy , Tissue Engineering , Humans , Genetic Therapy
3.
J Mater Sci Mater Med ; 29(7): 91, 2018 Jun 25.
Article in English | MEDLINE | ID: mdl-29938334

ABSTRACT

The development of immune protective islet encapsulation devices could allow for islet transplantation in the absence of immunosuppression. However, the immune protective membrane / barrier introduced there could also impose limitations in transport of oxygen and nutrients to the encapsulated cells resulting to limited islet viability. In the last years, it is well understood that achieving prevascularization of the device in vitro could facilitate its connection to the host vasculature after implantation, and therefore could provide sufficient blood supply and oxygenation to the encapsulated islets. However, the microvascular networks created in vitro need to mimic well the highly organized vasculature of the native tissue. In earlier study, we developed a functional macroencapsulation device consisting of two polyethersulfone/polyvinylpyrrolidone (PES/PVP) membranes, where a bottom microwell membrane provides good separation of encapsulated islets and the top flat membrane acts as a lid. In this work, we investigate the possibility of creating early microvascular networks on the lid of this device by combining novel membrane microfabrication with co-culture of human umbilical vein endothelial cell (HUVEC) and fibroblasts. We create thin porous microstructured PES/PVP membranes with solid and intermittent line-patterns and investigate the effect of cell alignment and cell interconnectivity as a first step towards the development of a stable prevascularized layer in vitro. Our results show that, in contrast to non-patterned membranes where HUVECs form unorganized HUVEC branch-like structures, for the micropatterned membranes, we can achieve cell alignment and the co-culture of HUVECs on a monolayer of fibroblasts attached on the membranes with intermittent line-pattern allows for the creation of HUVEC branch-like structures over the membrane surface. This important step towards creating early microvascular networks was achieved without the addition of hydrogels, often used in angiogenesis assays, as gels could block the pores of the membrane and limit the transport properties of the islet encapsulation device.


Subject(s)
Islets of Langerhans Transplantation/instrumentation , Biocompatible Materials/chemistry , Cell Adhesion , Cells, Cultured , Cellular Microenvironment , Coculture Techniques , Endothelial Cells/cytology , Fibroblasts/cytology , Human Umbilical Vein Endothelial Cells , Humans , Materials Testing , Membranes, Artificial , Microscopy, Electron, Scanning , Neovascularization, Physiologic , Polymers , Polyvinyls , Pyrrolidines , Sulfones
4.
J Mater Sci Mater Med ; 29(11): 174, 2018 Nov 09.
Article in English | MEDLINE | ID: mdl-30413974

ABSTRACT

Extrahepatic transplantation of islets of Langerhans could aid in better survival of islets after transplantation. When islets are transfused into the liver 60-70% of them are lost immediately after transplantation. An important factor for a successful extrahepatic transplantation is a well-vascularized tissue surrounding the implant. There are many strategies known for enhancing vessel formation such as adding cells with endothelial potential, the combination with angiogenic factors and / or applying surface topography at the exposed surface of the device. Previously we developed porous, micropatterned membranes which can be applied as a lid for an islet encapsulation device and we showed that the surface topography induces human umbilical vein endothelial cell (HUVEC) alignment and interconnection. This was achieved without the addition of hydrogels, often used in angiogenesis assays. In this work, we went one step further towards clinical implementation of the device by combining this micropatterned lid with Mesenchymal Stem Cells (MSCs) to facilitate prevascularization in vivo. As for HUVECs, the micropatterned membranes induced MSC alignment and organization in vitro, an important contributor to vessel formation, whereas in vivo (subcutaneous rat model) they contributed to improved implant prevascularization. In fact, the combination of MSCs seeded on the micropatterned membrane induced the highest vessel formation score in 80% of the sections.


Subject(s)
Drug Compounding , Islets of Langerhans/growth & development , Membranes, Artificial , Mesenchymal Stem Cells , Tissue Scaffolds , Human Umbilical Vein Endothelial Cells , Humans , Islets of Langerhans/blood supply , Neovascularization, Physiologic
5.
Ann Surg ; 266(1): 149-157, 2017 07.
Article in English | MEDLINE | ID: mdl-27429018

ABSTRACT

OBJECTIVE: We aim on developing a polymeric ectopic scaffold in a readily accessible site under the skin. SUMMARY BACKGROUND DATA: The liver as transplantation site for pancreatic islets is associated with significant loss of islets. Several extrahepatic sites were tested in experimental animals, but many have practical limitations in the clinical setting and do not have the benefit of easy accessibility. METHODS AND RESULTS: Functional survival of rat islets was tested during 7 days of culture in the presence of poly(D,L-lactide-co-ε-caprolactone) (PDLLCL), poly(ethylene oxide terephthalate)/polybutylene terephthalate (PEOT/PBT) block copolymer, and polysulfone. Tissue responses were studied in vivo after subcutaneous implantation in rats. Culture on PEOT/PBT and polysulfone profoundly disturbed function of islets, and induced severe tissue responses in vivo. Modification of their hydrophilicity did not change the suitability of the polymers. PDLLCL was the only polymer that promoted functional survival of rat islets in vitro and was associated with minor tissue reactions after 28 days. Rat islets were transplanted in the PDLLCL scaffold in a diabetic rat model. Before islet seeding, the scaffold was allowed to engraft for 28 days to allow the tissue response to dampen and to allow blood vessel growth into the device. Islet transplantation into the scaffold resulted in normoglycemia within 3 days and for the duration of the study period of 16 weeks. CONCLUSIONS: In conclusion, we found that some polymers such as PEOT/PBT and polysulfone interfere with islet function. PDLLCL is a suitable polymer to create an artificial islet transplantation site under the skin and supports islet survival.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/methods , Polymers , Tissue Scaffolds , Animals , Biocompatible Materials , Cell Culture Techniques , Cell Survival , Polyesters , Polyethylene Glycols , Sulfones
6.
Int J Mol Sci ; 17(9)2016 Sep 12.
Article in English | MEDLINE | ID: mdl-27626415

ABSTRACT

In vitro research in the field of type I diabetes is frequently limited by the availability of a functional model for islets of Langerhans. This method shows that by the addition of theophylline to the glucose buffers, mouse insulinoma MIN6 and rat insulinoma INS1E pseudo-islets can serve as a model for islets of Langerhans for in vitro research. The effect of theophylline is dose- and cell line-dependent, resulting in a minimal stimulation index of five followed by a rapid return to baseline insulin secretion by reducing glucose concentrations after a first high glucose stimulation. This protocol solves issues concerning in vitro research for type I diabetes as donors and the availability of primary islets of Langerhans are limited. To avoid the limitations of using human donor material, cell lines represent a valid alternative. Many different ß cell lines have been reported, but the lack of reproducible responsiveness to glucose stimulation remains a challenge.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Insulin/metabolism , Islets of Langerhans/drug effects , Theophylline/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Insulin Secretion , Islets of Langerhans/cytology , Mice , Models, Biological , Rats
7.
J Cell Mol Med ; 19(8): 1836-46, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25782016

ABSTRACT

Clinical islet transplantation is a promising treatment for patients with type 1 diabetes. However, pancreatic islets vary in size and shape affecting their survival and function after transplantation because of mass transport limitations. To reduce diffusion restrictions and improve islet cell survival, the generation of islets with optimal dimensions by dispersion followed by reassembly of islet cells, can help limit the length of diffusion pathways. This study describes a microwell platform that supports the controlled and reproducible production of three-dimensional pancreatic cell clusters of human donor islets. We observed that primary human islet cell aggregates with a diameter of 100-150 µm consisting of about 1000 cells best resembled intact pancreatic islets as they showed low apoptotic cell death (<2%), comparable glucose-responsiveness and increasing PDX1, MAFA and INSULIN gene expression with increasing aggregate size. The re-associated human islet cells showed an a-typical core shell configuration with beta cells predominantly on the outside unlike human islets, which became more randomized after implantation similar to native human islets. After transplantation of these islet cell aggregates under the kidney capsule of immunodeficient mice, human C-peptide was detected in the serum indicating that beta cells retained their endocrine function similar to human islets. The agarose microwell platform was shown to be an easy and very reproducible method to aggregate pancreatic islet cells with high accuracy providing a reliable tool to study cell-cell interactions between insuloma and/or primary islet cells.


Subject(s)
Glucose/pharmacology , Islets of Langerhans/cytology , Animals , Cell Aggregation/drug effects , Cell Line, Tumor , Cell Size , Cell Survival , Cells, Cultured , Female , Humans , Insulinoma/pathology , Male , Mice, SCID , Middle Aged , Reproducibility of Results
8.
Adv Healthc Mater ; 12(32): e2300640, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37781993

ABSTRACT

Intra-portal islet transplantation is currently the only clinically approved beta cell replacement therapy, but its outcome is hindered by limited cell survival due to a multifactorial reaction against the allogeneic tissue in liver. Adipose-derived stromal cells (ASCs) can potentially improve the islet micro-environment by their immunomodulatory action. The challenge is to combine both islets and ASCs in a relatively easy and consistent long-term manner in a deliverable scaffold. Manufacturing the 3D bioprinted double-layered scaffolds with primary islets and ASCs using a mix of alginate/nanofibrillated cellulose (NFC) bioink is reported. The diffusion properties of the bioink and the supportive effect of human ASCs on islet viability, glucose sensing, insulin secretion, and reducing the secretion of pro-inflammatory cytokines are demonstrated. Diabetic mice transplanted with islet-ASC scaffolds reach normoglycemia seven days post-transplantation with no significant difference between this group and the group received islets under the kidney capsules. In addition, animals transplanted with islet-ASC scaffolds stay normoglycemic and show elevated levels of C-peptide compared to mice transplanted with islet-only scaffolds. The data present a functional 3D bioprinted scaffold for islets and ASCs transplanted to the extrahepatic site and suggest a possible role of ASCs on improving the islet micro-environment.


Subject(s)
Diabetes Mellitus, Experimental , Insulin-Secreting Cells , Islets of Langerhans Transplantation , Islets of Langerhans , Mice , Humans , Animals , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Experimental/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Stromal Cells/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism
9.
Small ; 8(24): 3823-31, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-22907803

ABSTRACT

A reproducible wafer-scale method to obtain 3D nanostructures is investigated. This method, called corner lithography, explores the conformal deposition and the subsequent timed isotropic etching of a thin film in a 3D shaped silicon template. The technique leaves a residue of the thin film in sharp concave corners which can be used as structural material or as an inversion mask in subsequent steps. The potential of corner lithography is studied by fabrication of functional 3D microfluidic components, in particular i) novel tips containing nano-apertures at or near the apex for AFM-based liquid deposition devices, and ii) a novel particle or cell trapping device using an array of nanowire frames. The use of these arrays of nanowire cages for capturing single primary bovine chondrocytes by a droplet seeding method is successfully demonstrated, and changes in phenotype are observed over time, while retaining them in a well-defined pattern and 3D microenvironment in a flat array.


Subject(s)
Microfluidics/methods , Nanostructures , Printing/methods , Animals , Cattle , Cell Separation/instrumentation , Cell Separation/methods , Chondrocytes/cytology , Microfluidics/instrumentation , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Nanotechnology/instrumentation , Nanotechnology/methods , Nanowires/ultrastructure , Printing/instrumentation
10.
Catheter Cardiovasc Interv ; 79(4): 644-53, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21805599

ABSTRACT

OBJECTIVES: To assess and quantify coating irregularities on unexpanded and expanded durable polymer-based drug-eluting stents (DES) to gain insights into the origin of coating irregularities. BACKGROUND: Previous scanning electron microscopy (SEM) studies in various expanded DES revealed differences in frequency and size of coating irregularities between DES types and specific distribution patterns, however, the origin of these irregularities is unclear. METHODS: We assessed at bench side a total of 1,200 SEM images obtained in 30 DES samples (15 expanded and 15 unexpanded) of Cypher Select Plus, Taxus Liberté, Endeavor, Xience V, and resolute. RESULTS: For most coating irregularities seen on expanded DES (72%; 23/32), a matching irregularity (n = 18/23) and/or its precursor (n = 11/23) was observed in unexpanded DES. Unexpanded Cypher select showed (small) crater lesions and cracks together with precursors of "peeling." On unexpanded Taxus Liberté, thinning of polymer, small bare metal areas, wrinkles, and one precursor type were found. Unexpanded endeavor showed cracks, small bare metal areas, crater lesions, and precursors of the latter. Unexpanded Xience V and resolute mainly revealed crater lesions and their precursors. On unexpanded versus expanded DES, there was no difference in measured frequency of coating irregularities and precursors (P = ns) with the exception of more bare metal areas on expanded Taxus Liberte (P = 0.01). CONCLUSIONS: Most coating irregularities, or the potential to develop them, are inherent to the unexpanded DES. Important determinants of the formation of coating irregularities may be the stent geometry and the physical properties of the coating, while stent-balloon interaction plays no major role.


Subject(s)
Coated Materials, Biocompatible , Drug-Eluting Stents , Microscopy, Electron, Scanning , Polymers/chemistry , Metals , Prosthesis Failure , Risk Assessment , Surface Properties
11.
J Interv Cardiol ; 24(2): 149-61, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21198852

ABSTRACT

BACKGROUND: Oversized postdilation of drug-eluting stents (DES) is often performed to avoid stent malapposition. In stents implanted in long lesion or major bifurcations, extremely oversized partial postdilation may be required, which exposes DES coating to extreme forces. This study aims to assess shape and incidence of coating irregularities on durable polymer-based DES following extremely oversized partial postdilatation. METHODS: Fifteen DES samples (3 3.5 mm stents of Cypher Select plus [Cordis Europa, Roden, the Netherlands], Taxus Liberté[Boston Scientific Corp., Natick, MA, USA], Endeavor Sprint [Medtronic Vascular, Santa Rosa, CA, USA], Endeavor Resolute [Medtronic Vascular, Santa Rosa, CA, USA], and Xience V [Abbott Vascular, Santa Clara, CA, USA]) were deployed in sterile water (37 °C) at 14 atm, followed by a proximal postdilation with noncompliant 5.0-mm balloons at 18 atm. Stents were then examined with scanning electron microscopy. RESULTS: Thorough examination of a total of 660 scanning electron microscopic images demonstrated that shape and incidence of coating irregularities in the postdilated and/or transitional DES regions differed only mildly from the nonpostdilated regions. Cypher Select plus showed more peeling without bare metal aspect in the postdilated and transitional regions, and cracks were wider (P < 0.001) in the postdilated and transitional regions; in Taxus Liberté one additional irregularity (torn webbing) and more wrinkles were observed (P < 0.05 for both); in Endeavor Resolute wider cracks were found in the extremely postdilated region only (P < 0.001). Endeavor Sprint and Xience V showed no differences in shape or incidence of coating irregularities between oversized and nonoversized stent regions. CONCLUSIONS: Bench side assessment of five contemporary durable polymer-based DES with scanning electron microscopy suggests that even very aggressive stent postdilatation results in no more than mild differences in coating irregularities between postdilated and nonpostdilated stent regions.


Subject(s)
Angioplasty, Balloon, Coronary/methods , Drug-Eluting Stents , Microscopy, Electron, Scanning , Polymers , Prosthesis Design , Humans , Microscopy, Electron, Scanning/methods
12.
J Biomed Mater Res B Appl Biomater ; 109(1): 117-127, 2021 01.
Article in English | MEDLINE | ID: mdl-32672384

ABSTRACT

To effectively apply microwell array cell delivery devices their biodegradation rate must be tailored towards their intended use and implantation location. Two microwell array devices with distinct degradation profiles, either suitable for the fabrication of retrievable systems in the case of slow degradation, or cell delivery systems capable of extensive remodeling using a fast degrading polymer, were compared in this study. Thin films of a poly(ethylene glycol)-poly(butylene terephthalate) (PEOT-PBT) and a poly(ester urethane) were evaluated for their in vitro degradation profiles over 34 weeks incubation in PBS at different pH values. The PEOT-PBT films showed minimal in vitro degradation over time, while the poly(ester urethane) films showed extensive degradation and fragmentation over time. Subsequently, microwell array cell delivery devices were fabricated from these polymers and intraperitoneally implanted in Albino Oxford rats to study their biocompatibility over a 12-week period. The PEOT-PBT implants shown to be capable to maintain the microwell structure over time. Implants provoked a foreign body response resulting in multilayer fibrosis that integrated into the surrounding tissue. The poly(ester urethane) implants showed a loss of the microwell structures over time, as well as a fibrotic response until the onset of fragmentation, at least 4 weeks post implantation. It was concluded that the PEOT-PBT implants could be used as retrievable cell delivery devices while the poly(ester urethane) implants could be used for cell delivery devices that require remodeling within a 4-12 week period.


Subject(s)
Biocompatible Materials/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Polyurethanes/chemistry , Tissue Scaffolds/chemistry , Animals , Biodegradation, Environmental , Humans , In Vitro Techniques , Mechanical Phenomena , Mechanical Tests , Models, Animal , Polyethylene Terephthalates/chemistry , Prostheses and Implants , Rats , Regeneration , Tensile Strength , Tissue Engineering
13.
Biomaterials ; 267: 120449, 2021 01.
Article in English | MEDLINE | ID: mdl-33129188

ABSTRACT

The clinical success rate of islet transplantation, namely independence from insulin injections, is limited by factors that lead to graft failure, including inflammation, acute ischemia, acute phase response, and insufficient vascularization. The ischemia and insufficient vascularization both lead to high levels of oxidative stress, which are further aggravated by islet encapsulation, inflammation, and undesirable cell-biomaterial interactions. To identify biomaterials that would not further increase damaging oxidative stress levels and that are also suitable for manufacturing a beta cell encapsulation device, we studied five clinically approved polymers for their effect on oxidative stress and islet (alpha and beta cell) function. We found that 300 poly(ethylene oxide terephthalate) 55/poly(butylene terephthalate) 45 (PEOT/PBT300) was more resistant to breakage and more elastic than other biomaterials, which is important for its immunoprotective function. In addition, it did not induce oxidative stress or reduce viability in the MIN6 beta cell line, and even promoted protective endogenous antioxidant expression over 7 days. Importantly, PEOT/PBT300 is one of the biomaterials we studied that did not interfere with insulin secretion in human islets.


Subject(s)
Insulin-Secreting Cells , Islets of Langerhans Transplantation , Islets of Langerhans , Biocompatible Materials/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Oxidative Stress
14.
Macromol Biosci ; 20(8): e2000021, 2020 08.
Article in English | MEDLINE | ID: mdl-32567161

ABSTRACT

Macroencapsulation of islets of Langerhans is a promising strategy for transplantation of insulin-producing cells in the absence of immunosuppression to treat type 1 diabetes. Hollow fiber membranes are of interest there because they offer a large surface-to-volume ratio and can potentially be retrieved or refilled. However, current available fibers have limitations in exchange of nutrients, oxygen, and delivery of insulin potentially impacting graft survival. Here, multibore hollow fibers for islets encapsulation are designed and tested. They consist of seven bores and are prepared using nondegradable polymers with high mechanical stability and low cell adhesion properties. Human islets encapsulated there have a glucose induced insulin response (GIIS) similar to nonencapsulated islets. During 7 d of cell culture in vitro, the GIIS increases with graded doses of islets demonstrating the suitability of the microenvironment for islet survival. Moreover, first implantation studies in mice demonstrate device material biocompatibility with minimal tissue responses. Besides, formation of new blood vessels close to the implanted device is observed, an important requirement for maintaining islet viability and fast exchange of glucose and insulin. The results indicate that the developed fibers have high islet bearing capacity and can potentially be applied for a clinically applicable bioartificial pancreas.


Subject(s)
Islets of Langerhans/cytology , Tissue Engineering/instrumentation , Tissue Engineering/methods , Biocompatible Materials/pharmacology , Blood Vessels/growth & development , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Humans , Islets of Langerhans/physiology , Membranes, Artificial , Neovascularization, Physiologic/drug effects , Water
15.
16.
Tissue Eng Part C Methods ; 24(11): 628-636, 2018 11.
Article in English | MEDLINE | ID: mdl-30306836

ABSTRACT

IMPACT STATEMENT: This research deals with finding a proper bioengineering strategy for the creation of improved ß-cell replacement therapy in type 1 diabetes. It specifically deals with the microenvironment of ß-cells and its relationship to their endocrine function.


Subject(s)
Collagen Type IV/metabolism , Fibronectins/metabolism , Insulin Secretion , Insulin/metabolism , Insulinoma/metabolism , Pancreatic Neoplasms/metabolism , Animals , Collagen Type IV/chemistry , Fibronectins/chemistry , Insulinoma/pathology , Pancreatic Neoplasms/pathology , Printing, Three-Dimensional , Rats , Tumor Cells, Cultured
17.
J Gerontol A Biol Sci Med Sci ; 62(1): 34-40, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17301035

ABSTRACT

Although it has been established that premature aging trichothiodystrophy (TTD) mice display typical signs of osteoporosis, exact changes in physicochemical properties of these mice have not been elucidated. We used confocal Raman microscopy and histology to study femora of TTD mice. We measured femora isolated from xeroderma pigmentosum group A (XPA)/TTD double mutant mice to establish that Raman microscopy can be applied to measure differences in bone composition. Raman data from XPA/TTD mice showed remarkable changes in bone mineral composition. Moreover, we observed a severe form of osteoporosis, with strongly reduced cortical bone thickness. We used Raman microscopy to analyze bone composition in eight wild-type and eight TTD animals, and observed decreased levels of phosphate and carbonate in the cortex of femora isolated from TTD mice. In contrast, the bands representing the bone protein matrix were not affected in these mice.


Subject(s)
Aging, Premature , Carbonates/analysis , Collagen/analysis , Femur/chemistry , Hair Diseases/metabolism , Osteoporosis , Phosphates/analysis , Aging, Premature/etiology , Aging, Premature/metabolism , Aging, Premature/pathology , Animals , Bone Density , Bone Matrix/chemistry , Disease Models, Animal , Femur/pathology , Hair/abnormalities , Hair Diseases/complications , Hair Diseases/genetics , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Osteoporosis/complications , Osteoporosis/metabolism , Osteoporosis/pathology , Spectrum Analysis, Raman , Xeroderma Pigmentosum/complications , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/pathology
18.
Sci Rep ; 7(1): 9186, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28835662

ABSTRACT

Allogeneic islet transplantation into the liver in combination with immune suppressive drug therapy is widely regarded as a potential cure for type 1 diabetes. However, the intrahepatic system is suboptimal as the concentration of drugs and nutrients there is higher compared to pancreas, which negatively affects islet function. Islet encapsulation within semipermeable membranes is a promising strategy that allows for the islet transplantation outside the suboptimal liver portal system and provides environment, where islets can perform their endocrine function. In this study, we develop a macroencapsulation device based on thin microwell membranes. The islets are seeded in separate microwells to avoid aggregation, whereas the membrane porosity is tailored to achieve sufficient transport of nutrients, glucose and insulin. The non-degradable, microwell membranes are composed of poly (ether sulfone)/polyvinylpyrrolidone and manufactured via phase separation micro molding. Our results show that the device prevents aggregation and preserves the islet's native morphology. Moreover, the encapsulated islets maintain their glucose responsiveness and function after 7 days of culture (stimulation index above 2 for high glucose stimulation), demonstrating the potential of this novel device for islet transplantation.


Subject(s)
Biocompatible Materials , Islets of Langerhans , Membranes , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Biological Transport , Cell Line , Cell Survival , Diabetes Mellitus, Type 1/therapy , Glucose/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/immunology , Islets of Langerhans Transplantation , Membranes/chemistry , Membranes/metabolism , Membranes/ultrastructure , Mice , Permeability , Porosity , Tissue Culture Techniques , Tissue Scaffolds
19.
Biomaterials ; 135: 10-22, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28478326

ABSTRACT

Despite the clinical success of intrahepatic islet transplantation in treating type 1 diabetes, factors specific to this transplantation site hinder long-term insulin independence. The adoption of alternative, extravascular sites likely improve islet survival and function, but few locations are able to sufficiently confine islets in order to facilitate engraftment. This work describes a porous microwell scaffold with a well-defined pore size and spacing designed to guarantee islet retention at an extrahepatic transplantation site and facilitate islet revascularization. Three techniques to introduce pores were characterized: particulate leaching; solvent casting on pillared wafers; and laser drilling. Our criteria of a maximum pore diameter of 40 µm were best achieved via laser drilling. Transplantation studies in the epididymal fat of diabetic mice elucidated the potential of this porous scaffold platform to restore blood glucose levels and facilitate islet engraftment. Six out of eight mice reverted to stable normoglycemia with a mean time to remission of 6.2 ± 3.2 days, which was comparable to that of the gold standard of renal subcapsular islet grafts. In contrast, when islets were transplanted in the epididymal fat pad without a microwell scaffold, only two out of seven mice reverted to stable normoglycemia. Detailed histological evaluation four weeks after transplantation found a comparable vascular density in scaffold-seeded islets, renal subcapsular islets and native pancreatic islets. However, the vascularization pattern in scaffold-seeded islets was more inhomogeneous compared to native pancreatic islets with a higher vascular density in the outer shell of the islets compared to the inner core. We also observed a corresponding decrease in the beta-cell density in the islet core. Despite this, our data indicated that islets transplanted in the microwell scaffold platform were able to maintain a viable beta-cell population and restore glycemic control. Furthermore, we demonstrated that the microwell scaffold platform facilitated detailed analysis at a subcellular level to correlate design parameters with functional physiological observations.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/methods , Tissue Scaffolds , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Graft Survival , Insulin/blood , Male , Mice
20.
Transplantation ; 101(4): e112-e119, 2017 04.
Article in English | MEDLINE | ID: mdl-28207637

ABSTRACT

BACKGROUND: The liver as transplantation site for human pancreatic islets is a harsh microenvironment for islets and it lacks the ability to retrieve the graft. A retrievable, extrahepatic transplantation site that mimics the pancreatic environment is desired. Ideally, this transplantation site should be located subdermal for easy surgical-access but this never resulted in normoglycemia. Here, we describe the design and efficacy of a novel prevascularized, subcutaneously implanted, retrievable poly (D,L-lactide-co-ε-caprolactone) scaffold. METHOD: Three dosages of rat islets, that is, 400, 800, and 1200, were implanted in immune compromised mice to test the efficacy (n = 5). Islet transplantation under the kidney capsule served as control (n = 5). The efficacy was determined by nonfasting blood glucose measurements and glucose tolerance tests. RESULTS: Transplantation of 800 (n = 5) and 1200 islets (n = 5) into the scaffold reversed diabetes in respectively 80 and 100% of the mice within 6.8 to 18.5 days posttransplant. The marginal dose of 400 islets (n = 5) induced normoglycemia in 20%. The glucose tolerance test showed major improvement of the glucose clearance in the scaffold groups compared to diabetic controls. However, the kidney capsule was slightly more efficacious because all 800 (n = 5) and 1200 islets (n = 5) recipients and 40% of the 400 islets (n = 5) recipients became normoglycemic within 8 days. Removal of the scaffolds or kidney grafts resulted in immediate return to hyperglycemia. Normoglycemia was not achieved with 1200 islets in the unmodified skin group. CONCLUSIONS: Our findings demonstrate that the prevascularized poly (D,L-lactide-co-ε-caprolactone) scaffold maintains viability and function of islets in the subcutaneous site.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/methods , Islets of Langerhans/blood supply , Islets of Langerhans/surgery , Polyesters/chemistry , Subcutaneous Tissue/blood supply , Subcutaneous Tissue/surgery , Tissue Engineering/methods , Tissue Scaffolds , Animals , Biomarkers/blood , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/diagnosis , Disease Models, Animal , Glucose Tolerance Test , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice, Nude , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL