Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Adv Funct Mater ; 27(3)2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28713230

ABSTRACT

Developing in vivo cell tracking is an important prerequisite for further development of cell-based therapy. So far, few computed tomography (CT) cell tracking studies have been described due to its notoriously low sensitivity and lack of efficient labeling protocols. We present a simple method to render human mesenchymal stem cells (hMSCs) sufficiently radiopaque by complexing 40 nm citrate-stabilized gold nanoparticles (AuNPs) with poly-L-lysine (PLL) and rhodamine B isothiocyanate (RITC). AuNP-PLL-RITC labeling did not affect cellular viability, proliferation, or downstream cell differentiation into adipocytes and osteocytes. Labeled hMSCs could be clearly visualized in vitro and in vivo with a micro-CT scanner, with a detection limit of approximately 2×104 cells/µl in vivo. Calculated HU values were 2.27 /pg of intracellular Au as measured with inductively coupled plasma mass spectrophotometry (ICP-MS), and were linear over a wide range of cell concentrations. This linear CT attenuation was observed for both naked AuNPs and those that were taken up by hMSCs, indicating that the number of labeled cells can be quantified similar to the use of radioactive or fluorine tracers. This approach for CT cell tracking may find applications in CT image-guided interventions and fluoroscopic procedures commonly used for the injection of cellular therapeutics.

2.
Xenotransplantation ; 23(3): 211-21, 2016 05.
Article in English | MEDLINE | ID: mdl-27225644

ABSTRACT

BACKGROUND: The fate of magnetically labeled, barium-gelled alginate/protamine sulfate/alginate microcapsules (APSA magnetocapsules) following xenotransplantation was assessed by magnetic resonance imaging (MRI) and histopathology. METHODS: Magnetocapsules with and without human islets were transplanted into five different clinically accessible sites: portal vein, subcutaneous tissue, skeletal muscle, the liver and the kidney subcapsular space. The surface of APSA magnetocapsules was modified using clinical-grade heparin to mitigate an instant blood-mediated inflammatory reaction. RESULTS: The accuracy of site-specific delivery was confirmed using a clinical 1.5T MRI setup, where the magnetocapsules appeared as distinct hypointense entities after transplantation. As proven by the Lee-White blood coagulation test, heparin-treated APSA magnetocapsules did not induce blood clotting for more than 48 h in vitro. Heparinized magnetocapsules induced innate and adaptive immune responses in vivo regardless of the transplantation sites. CONCLUSION: We have demonstrated the feasibility of using a clinical 1.5T MRI to non-invasively detect the accuracy of APSA magnetocapsule injection into various clinically accessible transplantation sites. Among the investigated transplantation sites, the liver and kidney subcapsular space were found to be the least immuno-responsive toward xenografted magneto-encapsulated human islets.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans/surgery , Transplantation, Heterologous , Animals , Blood Coagulation/drug effects , Capsules/pharmacology , Heterografts/drug effects , Heterografts/immunology , Humans , Islets of Langerhans Transplantation/methods , Liver/blood supply , Portal Vein/drug effects , Transplantation, Heterologous/methods
3.
Nat Mater ; 12(3): 268-75, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23353626

ABSTRACT

Biocompatible nanomaterials and hydrogels have become an important tool for improving cell-based therapies by promoting cell survival and protecting cell transplants from immune rejection. Although their potential benefit has been widely evaluated, at present it is not possible to determine, in vivo, if and how long cells remain viable following their administration without the use of a reporter gene. Here, we report a pH-nanosensor-based magnetic resonance imaging (MRI) technique that can monitor cell death in vivo non-invasively. We demonstrate that specific MRI parameters that change on cell death of microencapsulated hepatocytes are associated with the measured bioluminescence imaging radiance. Moreover, the readout from this pH-sensitive nanosensor can be directly co-registered with high-resolution anatomical images. All of the components of these nanosensors are clinical grade and hence this approach should be a translatable and universal modification of hydrogels.


Subject(s)
Biocompatible Materials , Cell Transplantation/methods , Magnetic Resonance Imaging/methods , Nanostructures , Animals , Cell Survival , Contrast Media/chemistry , Hepatocytes/transplantation , Hydrogels , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C
4.
ACS Appl Mater Interfaces ; 16(9): 11159-11171, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38385360

ABSTRACT

For the improved delivery of cancer therapeutics and imaging agents, the conjugation of cell-penetrating peptides (CPPs) increases the cellular uptake and water solubility of agents. Among the various CPPs, arginine-rich peptides have been the most widely used. Combining CPPs with enzyme-responsive peptides presents an innovative strategy to target specific intracellular enzymes in cancer cells and when combined with the appropriate click chemistry can enhance theranostic drug delivery through the formation of intracellular self-assembled nanostructures. However, one drawback of CPPs is their high positive charge which can cause nonspecific binding, leading to off-target accumulation and potential toxicity. Hence, balancing cell-specific penetration, toxicity, and biocompatibility is essential for future clinical efficacy. We synthesized six cancer-specific, legumain-responsive RnAANCK peptides containing one to six arginine residues, with legumain being an asparaginyl endopeptidase that is overexpressed in aggressive prostate tumors. When conjugated to Alexa Fluor 488, R1-R6AANCK peptides exhibited a concentration- and time-dependent cell penetration in prostate cancer cells, which was higher for peptides with higher R values, reaching a plateau after approximately 120 min. Highly aggressive DU145 prostate tumor cells, but not less aggressive LNCaP cells, self-assembled nanoparticles in the cytosol after the cleavage of the legumain-specific peptide. The in vivo biocompatibility was assessed in mice after the intravenous injection of R1-R6AANCK peptides, with concentrations ranging from 0.0125 to 0.4 mmol/kg. The higher arginine content in R4-6 peptides showed blood and urine indicators for the impairment of bone marrow, liver, and kidney function in a dose-dependent manner, with instant hemolysis and morbidity in extreme cases. These findings underscore the importance of designing peptides with the optimal arginine residue length for a proper balance of cell-specific penetration, toxicity, and in vivo biocompatibility.


Subject(s)
Cell-Penetrating Peptides , Neoplasms , Animals , Mice , Arginine/chemistry , Cell-Penetrating Peptides/chemistry , Neoplasms/drug therapy
5.
NMR Biomed ; 26(7): 850-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23225358

ABSTRACT

Microencapsulation of therapeutic cells has been widely pursued to achieve cellular immunoprotection following transplantation. Initial clinical studies have shown the potential of microencapsulation using semi-permeable alginate layers, but much needs to be learned about the optimal delivery route, in vivo pattern of engraftment, and microcapsule stability over time. In parallel with noninvasive imaging techniques for 'naked' (i.e. unencapsulated) cell tracking, microcapsules have now been endowed with contrast agents that can be visualized by (1) H MRI, (19) F MRI, X-ray/computed tomography and ultrasound imaging. By placing the contrast agent formulation in the extracellular space of the hydrogel, large amounts of contrast agents can be incorporated with negligible toxicity. This has led to a new generation of imaging biomaterials that can render cells visible with multiple imaging modalities.


Subject(s)
Cell Tracking/methods , Drug Compounding/methods , Animals , Capsules/chemistry , Contrast Media/chemistry , Dextrans/chemistry , Humans , Magnetic Phenomena , Magnetite Nanoparticles/chemistry
6.
Methods Mol Biol ; 2592: 75-88, 2023.
Article in English | MEDLINE | ID: mdl-36507986

ABSTRACT

We describe step-by-step methods to label human pancreatic islet cells and murine insulinoma cells and their subsequent transplantation into type I diabetic mouse models with a focus on in vivo imaging using clinically applicable scanners. We also cover islets that are microencapsulated within alginate hydrogels loaded with imaging agents. By following these methods, it is possible to image cell grafts using T1-weighted and T2/T2*-weighted 1H magnetic resonance imaging (MRI), 19F MRI, computed tomography, ultrasound imaging, and bioluminescence imaging in vivo. Considering a myriad of factors that may affect the outcome of proper in vivo detection, we discuss potential issues that may be encountered during and after the process of labeling. The ultimate goal is to use these in vivo imaging approaches to determine and optimize naked and encapsulated islet cell survival, therapeutic function, and engraftment procedures.


Subject(s)
Diabetes Mellitus , Insulinoma , Islets of Langerhans Transplantation , Islets of Langerhans , Pancreatic Neoplasms , Mice , Humans , Animals , Islets of Langerhans Transplantation/methods , Islets of Langerhans/diagnostic imaging , Islets of Langerhans/metabolism , Diabetes Mellitus/metabolism , Insulinoma/metabolism , Pancreatic Neoplasms/metabolism
7.
Adv Sci (Weinh) ; 10(34): e2304164, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37715297

ABSTRACT

Inspired by the principle of in situ self-assembly, the development of enzyme-activated molecular nanoprobes can have a profound impact on targeted tumor detection. However, despite their intrinsic promise, obtaining an optical readout of enzyme activity with high specificity in native milieu has proven to be challenging. Here, a fundamentally new class of Raman-active self-assembling bioorthogonal enzyme recognition (nanoSABER) probes for targeted tumor imaging is reported. This class of Raman probes presents narrow spectral bands reflecting their vibrational fingerprints and offers an attractive solution for optical imaging at different bio-organization levels. The optical beacon harnesses an enzyme-responsive peptide sequence, unique tumor-penetrating properties, and vibrational tags with stretching frequencies in the cell-silent Raman window. The design of nanoSABER is tailored and engineered to transform into a supramolecular structure exhibiting distinct vibrational signatures in presence of target enzyme, creating a direct causality between enzyme activity and Raman signal. Through the integration of substrate-specific for tumor-associated enzyme legumain, unique capabilities of nanoSABER for imaging enzyme activity at molecular, cellular, and tissue levels in combination with machine learning models are shown. These results demonstrate that the nanoSABER probe may serve as a versatile platform for Raman-based recognition of tumor aggressiveness, drug accumulation, and therapeutic response.


Subject(s)
Neoplasms , Humans , Neoplasms/diagnostic imaging , Optical Imaging
8.
J Extracell Vesicles ; 11(7): e12241, 2022 07.
Article in English | MEDLINE | ID: mdl-35844061

ABSTRACT

Extracellular vesicles (EVs) are lipid-bilayer delimited vesicles released by nearly all cell types that serve as mediators of intercellular signalling. Recent evidence has shown that EVs play a key role in many normal as well as pathological cellular processes. EVs can be exploited as disease biomarkers and also as targeted, cell-free therapeutic delivery and signalling vehicles for use in regenerative medicine and other clinical settings. Despite this potential, much remains unknown about the in vivo biodistribution and pharmacokinetic profiles of EVs after administration into living subjects. The ability to non-invasively image exogeneous EVs, especially in larger animals, will allow a better understanding of their in vivo homing and retention patterns, blood and tissue half-life, and excretion pathways, all of which are needed to advance clinical diagnostic and/or therapeutic applications of EVs. We present the current state-of-the-art methods for labeling EVs with various diagnostic contrast agents and tracers and the respective imaging modalities that can be used for their in vivo visualization: magnetic resonance imaging (MRI), X-ray computed tomography (CT) imaging, magnetic particle imaging (MPI), single-photon emission computed tomography (SPECT), positron emission tomography (PET), and optical imaging (fluorescence and bioluminescence imaging). We review here the strengths and weaknesses of each of these EV imaging approaches, with special emphasis on clinical translation.


Subject(s)
Extracellular Vesicles , Animals , Extracellular Vesicles/metabolism , Humans , Optical Imaging , Tissue Distribution
9.
Nat Biomed Eng ; 6(5): 658-666, 2022 05.
Article in English | MEDLINE | ID: mdl-35132228

ABSTRACT

The tracking of the in vivo biodistribution of transplanted human mesenchymal stromal cells (hMSCs) relies on reporter genes or on the addition of exogenous imaging agents. However, reporter genes and exogenous labels may require bespoke manufacturing and regulatory processes if used in cell therapies, and the labels may alter the cells' properties and are diluted on cellular division. Here we show that high-mannose N-linked glycans, which are abundantly expressed on the surface of hMSCs, can serve as a biomarker for the label-free tracking of transplanted hMSCs by mannose-weighted chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI). For live mice with luciferase-transfected hMSCs transplanted into their brains, post-mortem fluorescence staining with a mannose-specific lectin showed that increases in the CEST MRI signal, which correlated well with the bioluminescence intensity of viable hMSCs for 14 days, corresponded to the presence of mannose. In vitro, osteogenically differentiated hMSCs led to lower CEST MRI signal intensities owing to the concomitantly reduced expression of mannose. The label-free imaging of hMSCs may facilitate the development and testing of cell therapies.


Subject(s)
Mannose , Mesenchymal Stem Cells , Animals , Cell Differentiation , Magnetic Resonance Imaging/methods , Mannose/metabolism , Mice , Tissue Distribution
10.
Radiology ; 260(3): 790-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21734156

ABSTRACT

PURPOSE: To develop microcapsules that immunoprotect pancreatic islet cells for treatment of type I diabetes and enable multimodal cellular imaging of transplanted islet cells. MATERIALS AND METHODS: All animal experiments were approved by the institutional animal care and use committee. Gold nanoparticles functionalized with DTDTPA (dithiolated diethylenetriaminepentaacetic acid):gadolinium chelates (GG) were coencapsulated with pancreatic islet cells by using protamine sulfate as a clinical-grade alginate cross linker. Conventional poly-l-lysine-cross-linked microcapsules and unencapsulated islets were included as controls. The viability and glucose responsiveness of islet cells were assessed in vitro, and in vivo insulin (C-peptide) secretion was monitored for 6 weeks in (streptozotocin-induced) diabetic mice with (n = 7) or without (n = 8) intraabdominally engrafted islet cells. Five nondiabetic mice were included as controls. Differences between samples were calculated by using a nonparametric Wilcoxon Mann-Whitney method. To adjust for multiple comparisons, a significance level of P < .01 was chosen. Generalized estimating equations were used to model cell function over time. Three mice with engrafted capsules were imaged in vivo with high-field-strength (9.4-T) magnetic resonance (MR) imaging, micro-computed tomography (CT), and 40-MHz ultrasonography (US). RESULTS: Encapsulated human pancreatic islets were functional in vitro for at least 2 weeks after encapsulation. Blood glucose levels in the diabetic mice transplanted with GG-labeled encapsulated mouse ßTC6 insulinoma cells returned to normal within 1 week after transplantation, and normoglycemia was sustained for at least 6 weeks without the use of immunosuppressive drugs. GG microcapsules could be readily visualized with positive-contrast high-field-strength MR imaging, micro-CT, and US both in vitro and in vivo. CONCLUSION: Cell encapsulation with GG provides a means of trimodal noninvasive tracking of engrafted cells.


Subject(s)
Cell Tracking/methods , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Experimental/surgery , Gadolinium , Gold , Islets of Langerhans Transplantation/methods , Animals , Capsules , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Tomography, X-Ray Computed/methods , Treatment Outcome , Ultrasonography/methods
11.
Diabetes Metab Res Rev ; 27(8): 761-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22069256

ABSTRACT

At present, the onset and progress of diabetes, and the efficacy of potential treatments, can only be assessed through indirect means, i.e. blood glucose, insulin, or C-peptide measurements. The development of non-invasive and reliable methods for (1) quantification of pancreatic beta islet cell mass in vivo, (2) determining endogenous islet function and survival, and (3) visualizing the biodistribution, survival, and function of transplanted exogenous islets are critical to further advance both basic science research and islet cell therapy in diabetes. Islet cell imaging using magnetic resonance, bioluminescence, positron emission tomography, or single photon emission computed tomography may provide us with a direct means to interrogate islet cell distribution, survival, and function. Current state-of-the-art strategies for beta-cell imaging are discussed and reviewed here in context of their clinical relevance.


Subject(s)
Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Animals , Diagnostic Imaging/methods , Humans , Islets of Langerhans/diagnostic imaging , Islets of Langerhans Transplantation , Luminescent Measurements , Magnetic Resonance Imaging , Positron-Emission Tomography , Tomography, Emission-Computed, Single-Photon
12.
Front Endocrinol (Lausanne) ; 12: 640117, 2021.
Article in English | MEDLINE | ID: mdl-33737913

ABSTRACT

Transplantation of pancreatic islets has potential to offer life-long blood glucose management in type I diabetes and severe type II diabetes without the need of exogenous insulin administration. However, islet cell therapy suffers from autoimmune and allogeneic rejection as well as non-immune related factors. Non-invasive techniques to monitor and evaluate the fate of cell implants in vivo are essential to understand the underlying causes of graft failure, and hence to improve the precision and efficacy of islet therapy. This review describes how imaging technology has been employed to interrogate the distribution, number or volume, viability, and function of islet implants in vivo. To date, fluorescence imaging, PET, SPECT, BLI, MRI, MPI, and ultrasonography are the many imaging modalities being developed to fulfill this endeavor. We outline here the advantages, limitations, and clinical utility of each particular imaging approach.


Subject(s)
Diabetes Mellitus/diagnostic imaging , Diabetes Mellitus/therapy , Diagnostic Imaging/methods , Islets of Langerhans Transplantation/diagnostic imaging , Pancreas/diagnostic imaging , Animals , Autoimmune Diseases/immunology , Graft Rejection , Humans , Magnetic Resonance Imaging , Mice , Microscopy, Fluorescence , Multimodal Imaging , Positron-Emission Tomography , Tomography, Emission-Computed, Single-Photon , Ultrasonography
13.
Front Robot AI ; 8: 702566, 2021.
Article in English | MEDLINE | ID: mdl-34368238

ABSTRACT

Small soft robotic systems are being explored for myriad applications in medicine. Specifically, magnetically actuated microrobots capable of remote manipulation hold significant potential for the targeted delivery of therapeutics and biologicals. Much of previous efforts on microrobotics have been dedicated to locomotion in aqueous environments and hard surfaces. However, our human bodies are made of dense biological tissues, requiring researchers to develop new microrobotics that can locomote atop tissue surfaces. Tumbling microrobots are a sub-category of these devices capable of walking on surfaces guided by rotating magnetic fields. Using microrobots to deliver payloads to specific regions of sensitive tissues is a primary goal of medical microrobots. Central nervous system (CNS) tissues are a prime candidate given their delicate structure and highly region-specific function. Here we demonstrate surface walking of soft alginate capsules capable of moving on top of a rat cortex and mouse spinal cord ex vivo, demonstrating multi-location small molecule delivery to up to six different locations on each type of tissue with high spatial specificity. The softness of alginate gel prevents injuries that may arise from friction with CNS tissues during millirobot locomotion. Development of this technology may be useful in clinical and preclinical applications such as drug delivery, neural stimulation, and diagnostic imaging.

14.
Biomaterials ; 221: 119410, 2019 11.
Article in English | MEDLINE | ID: mdl-31421313

ABSTRACT

Clinical trials that have used encapsulated islet cell therapy have been few and overall disappointing. This is due in part to the lack of suitable methods to monitor the integrity vs. rupture of transplanted microcapsules over time. Fluorocapsules were synthesized by embedding emulsions of perfluoro-15-crown-5-ether (PFC), a bioinert compound detectable by 19F MRI, into dual-alginate layer, Ba2+-gelled alginate microcapsules. Fluorocapsules were spherical with an apparent smooth surface and an average diameter of 428 ±â€¯52 µm. After transplantation into mice, the 19F MRI signal of capsules remained stable for up to 90 days, corresponding to the total number of intact fluorocapsules. When single-alginate layer capsules were ruptured with alginate lyase, the 19F MRI signal dissipated within 4 days. For fluoroencapsulated luciferase-expressing mouse ßTC6 insulinoma cells implanted into autoimmune NOD/ShiLtJ mice and subjected to alginate-lyase induced capsule rupture in vivo, the 19F MRI signal decreased sharply over time along with a decrease in bioluminescence imaging signal used as a measure of cell viability in vivo. These results indicate that maintenance of capsule integrity is essential for preserving transplanted cell survival, where a decrease in 19F MRI signal may serve as a predictive imaging surrogate biomarker for impending failure of encapsulated islet cell therapy.


Subject(s)
Capsules/chemistry , Fluorine-19 Magnetic Resonance Imaging/methods , Alginates/chemistry , Animals , Islets of Langerhans/cytology , Islets of Langerhans Transplantation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD
15.
Micromachines (Basel) ; 10(4)2019 Mar 31.
Article in English | MEDLINE | ID: mdl-30935105

ABSTRACT

Soft, untethered microrobots composed of biocompatible materials for completing micromanipulation and drug delivery tasks in lab-on-a-chip and medical scenarios are currently being developed. Alginate holds significant potential in medical microrobotics due to its biocompatibility, biodegradability, and drug encapsulation capabilities. Here, we describe the synthesis of MANiACs-Magnetically Aligned Nanorods in Alginate Capsules-for use as untethered microrobotic surface tumblers, demonstrating magnetically guided lateral tumbling via rotating magnetic fields. MANiAC translation is demonstrated on tissue surfaces as well as inclined slopes. These alginate microrobots are capable of manipulating objects over millimeter-scale distances. Finally, we demonstrate payload release capabilities of MANiACs during translational tumbling motion.

17.
Nat Commun ; 6: 6719, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25813863

ABSTRACT

Alterations in mucin expression and glycosylation are associated with cancer development. Underglycosylated mucin-1 (uMUC1) is overexpressed in most malignant adenocarcinomas of epithelial origin (for example, colon, breast and ovarian cancer). Its counterpart MUC1 is a large polymer rich in glycans containing multiple exchangeable OH protons, which is readily detectable by chemical exchange saturation transfer (CEST) MRI. We show here that deglycosylation of MUC1 results in >75% reduction in CEST signal. Three uMUC1(+) human malignant cancer cell lines overexpressing uMUC1 (BT20, HT29 and LS174T) show a significantly lower CEST signal compared with the benign human epithelial cell line MCF10A and the uMUC1(-) tumour cell line U87. Furthermore, we demonstrate that in vivo CEST MRI is able to make a distinction between LS174T and U87 tumour cells implanted in the mouse brain. These results suggest that the mucCEST MRI signal can be used as a label-free surrogate marker to non-invasively assess mucin glycosylation and tumour malignancy.


Subject(s)
Carcinoma/metabolism , Glycosylation , Magnetic Resonance Imaging/methods , Mucin-1/metabolism , Animals , Carcinoma/pathology , Cell Line, Tumor , HT29 Cells , Humans , Mice , Mice, SCID , Molecular Imaging/methods , Neoplasm Transplantation
18.
Biotechnol Prog ; 19(6): 1798-811, 2003.
Article in English | MEDLINE | ID: mdl-14656159

ABSTRACT

In this study, we investigated the size distribution, encapsulation efficiency, and oxygen affinity of liposome-encapsulated tetrameric hemoglobin (LEHb) dispersions and correlated the data with the variation in extruder membrane pore size, ionic strength of the extrusion buffer, and hemoglobin (Hb) concentration. Asymmetric flow field-flow fractionation (AFFF) in series with multi-angle static light scattering (MASLS) was used to study the LEHb size distribution. We also introduced a novel method to measure the encapsulation efficiency using a differential interferometric refractive index (DIR) detector coupled to the AFFF-MASLS system. This technique was nondestructive toward the sample and easy to implement. LEHbs were prepared by extrusion using a lipid combination of dimyristoyl-phosphatidylcholine, cholesterol, and dimyristoyl-phosphatidylglycerol in a 10:9:1 molar ratio. Five initial Hb concentrations (50, 100, 150, 200, and 300 mg Hb per mL of buffer) extruded through five different membrane pore diameters (400, 200, 100, 80, and 50 nm) were studied. Phosphate buffered saline (PBS) and phosphate buffer (PB) both at pH 7.3 were used as extrusion buffers. Despite the variation, extrusion through 400-nm pore diameter membranes produced LEHbs smaller than the pore size, extrusion through 200-nm membranes produced LEHbs with diameters close to the pore diameter, and extrusion through 100-, 80-, and 50-nm membranes produced LEHbs larger than the pore sizes. We found that the choice of extrusion buffer had the greatest effect on the LEHb size distribution compared to either Hb concentration or extruder membrane pore size. Extrusion in PBS produced larger LEHbs and more monodisperse LEHb dispersions. However, LEHbs extruded in PB generally had higher Hb encapsulation efficiencies and lower methemoglobin (metHb) levels. The choice of extrusion buffer also affected how the encapsulation efficiency correlated with Hb concentration, extruder pore size, and the metHb level. The most optimum encapsulation efficiency and amount of Hb entrapped were achieved at the highest Hb concentration and the largest pore size for both extrusion buffers (62.38% and 187.14 mg Hb/mL of LEHb dispersion extruded in PBS, and 69.98% and 209.94 mg Hb/mL of LEHb dispersion extruded in PB). All LEHbs displayed good oxygen-carrying properties as indicated by their P(50) and cooperativity coefficients. LEHbs extruded in PB had an average P(50) of 23.04 mmHg and an average Hill number of 2.29, and those extruded in PBS had average values of 27.25 mmHg and 2.49. These oxygen-binding properties indicate that LEHbs possess strong potential as artificial blood substitutes. In addition, the metHb levels in PB-LEHb dispersions are significantly low even in the absence of antioxidants such as N-acetyl-L-cysteine.


Subject(s)
Blood Substitutes/chemistry , Coated Materials, Biocompatible/chemistry , Drug Delivery Systems/methods , Fractionation, Field Flow/methods , Hemoglobins/chemistry , Liposomes/chemistry , Materials Testing , Photometry/methods , Oxygen/chemistry , Particle Size , Porosity
19.
Biomaterials ; 33(18): 4681-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22444642

ABSTRACT

Microencapsulation is a commonly used technique for immunoprotection of engrafted therapeutic cells. We investigated a library of capsule formulations to determine the most optimal formulation for pancreatic beta islet cell transplantation, using barium as the gelating ion and clinical-grade protamine sulfate (PS) as a new cationic capsule cross-linker. Barium-gelated alginate/PS/alginate microcapsules (APSA, diameter = 444 ± 21 µm) proved to be mechanically stronger and supported a higher cell viability as compared to conventional alginate/poly-l-lysine/alginate (APLLA) capsules. Human pancreatic islets encapsulated inside APSA capsules, gelated with 20 mm barium as optimal concentration, exhibited a sustained morphological integrity, viability, and functionality for at least 3-4 weeks in vitro, with secreted human C-peptide levels of 0.2-160 pg/ml/islet. Unlike APLLA capsules that are gelled with calcium, barium-APSA capsules are intrinsically radiopaque and, when engrafted into mice, could be readily imaged in vivo with micro-computed tomography (CT). Without the need of adding contrast agents, these capsules offer a clinically applicable alternative for simultaneous immunoprotection and real-time, non-invasive X-ray/CT monitoring of engrafted cells during and after in vivo administration.


Subject(s)
Barium/chemistry , Capsules/chemistry , Islets of Langerhans/diagnostic imaging , Tomography, X-Ray Computed/methods , Alginates/chemistry , Animals , Cell Line, Tumor , Cells, Cultured , Humans , In Vitro Techniques , Mice , Polylysine/analogs & derivatives , Polylysine/chemistry
20.
Cell Med ; 4(2): 77-84, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-23293747

ABSTRACT

Hepatocyte transplantation is currently being considered as a new paradigm for treatment of fulminant liver failure. Xeno- and allotransplantation studies have shown considerable success but the long-term survival and immunorejection of engrafted cells needs to be further evaluated. Using novel alginate-protamine sulfate-alginate microcapsules, we have co-encapsulated luciferase-expressing HepG2 human hepatocytes with superparamagnetic iron oxide nanoparticles to create magnetocapsules that are visible on MRI as discrete hypointensities. Magnetoencapsulated cells survive and secrete albumin for at least 5 weeks in vitro. When transplanted i.p. in immunocompetent mice, encapsulated hepatocytes survive for at least 4 weeks as determined using bioluminescent imaging, which is in stark contrast to naked, unencapsulated hepatocytes, that died within several days after transplantation. However, in vivo human albumin secretion did not follow the time course of magnetoencapsulated cell survival, with plasma levels returning to baseline values already at 1 week post-transplantation. The present results demonstrate that encapsulation can dramatically prolong survival of xenotransplanted hepatocytes, leading to sustained albumin secretion with a duration that may be long enough for use as a temporary therapeutic bridge to liver transplantation.

SELECTION OF CITATIONS
SEARCH DETAIL