Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Diabetes ; 72(6): 758-768, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36929171

ABSTRACT

Intrahepatic islet transplantation for type 1 diabetes is limited by the need for multiple infusions and poor islet viability posttransplantation. The development of alternative transplantation sites is necessary to improve islet survival and facilitate monitoring and retrieval. We tested a clinically proven biodegradable temporizing matrix (BTM), a polyurethane-based scaffold, to generate a well-vascularized intracutaneous "neodermis" within the skin for islet transplantation. In murine models, BTM did not impair syngeneic islet renal-subcapsular transplant viability or function, and it facilitated diabetes cure for over 150 days. Furthermore, BTM supported functional neonatal porcine islet transplants into RAG-1-/- mice for 400 days. Hence, BTM is nontoxic for islets. Two-photon intravital imaging used to map vessel growth through time identified dense vascular networks, with significant collagen deposition and increases in vessel mass up to 30 days after BTM implantation. In a preclinical porcine skin model, BTM implants created a highly vascularized intracutaneous site by day 7 postimplantation. When syngeneic neonatal porcine islets were transplanted intracutaneously, the islets remained differentiated as insulin-producing cells, maintained normal islet architecture, secreted c-peptide, and survived for over 100 days. Here, we show that BTM facilitates formation of an islet-supportive intracutaneous neodermis in a porcine preclinical model, as an alternative islet-transplant site. ARTICLE HIGHLIGHTS: Human and porcine pancreatic islets were transplanted into a fully vascularized biodegradable temporizing matrix (Novosorb) that creates a unique intracutaneous site outside of the liver in a large-animal preclinical model. The intracutaneous prevascularized site supported pancreatic islet survival for 3 months in a syngeneic porcine-transplant model. Pancreatic (human and porcine) islet survival and function were demonstrated in an intracutaneous site outside of the liver for the first time in a large-animal preclinical model.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Swine , Humans , Animals , Mice , Islets of Langerhans Transplantation/methods , Graft Survival , Islets of Langerhans/blood supply , Diabetes Mellitus, Type 1/surgery , Collagen
2.
Diabetologia ; 61(9): 2016-2029, 2018 09.
Article in English | MEDLINE | ID: mdl-29971529

ABSTRACT

AIMS/HYPOTHESIS: Islet transplantation is a treatment option that can help individuals with type 1 diabetes become insulin independent, but inefficient oxygen and nutrient delivery can hamper islet survival and engraftment due to the size of the islets and loss of the native microvasculature. We hypothesised that size-controlled pseudoislets engineered via centrifugal-forced-aggregation (CFA-PI) in a platform we previously developed would compare favourably with native islets, even after taking into account cell loss during the process. METHODS: Human islets were dissociated and reaggregated into uniform, size-controlled CFA-PI in our microwell system. Their performance was assessed in vitro and in vivo over a range of sizes, and compared with that of unmodified native islets, as well as islet cell clusters formed by a conventional spontaneous aggregation approach (in which dissociated islet cells are cultured on ultra-low-attachment plates). In vitro studies included assays for membrane integrity, apoptosis, glucose-stimulated insulin secretion assay and total DNA content. In vivo efficacy was determined by transplantation under the kidney capsule of streptozotocin-treated Rag1-/- mice, with non-fasting blood glucose monitoring three times per week and IPGTT at day 60 for glucose response. A recovery nephrectomy, removing the graft, was conducted to confirm efficacy after completing the IPGTT. Architecture and composition were analysed by histological assessment via insulin, glucagon, pancreatic polypeptide, somatostatin, CD31 and von Willebrand factor staining. RESULTS: CFA-PI exhibit markedly increased uniformity over native islets, as well as substantially improved glucose-stimulated insulin secretion (8.8-fold to 11.1-fold, even after taking cell loss into account) and hypoxia tolerance. In vivo, CFA-PI function similarly to (and potentially better than) native islets in reversing hyperglycaemia (55.6% for CFA-PI vs 20.0% for native islets at 500 islet equivalents [IEQ], and 77.8% for CFA-PI vs 55.6% for native islets at 1000 IEQ), and significantly better than spontaneously aggregated control cells (55.6% for CFA-PI vs 0% for spontaneous aggregation at 500 IEQ, and 77.8% CFA-PI vs 33.4% for spontaneous aggregation at 1000 IEQ; p < 0.05). Glucose clearance in the CFA-PI groups was improved over that in the native islet groups (CFA-PI 18.1 mmol/l vs native islets 29.7 mmol/l at 60 min; p < 0.05) to the point where they were comparable with the non-transplanted naive normoglycaemic control mice at a low IEQ of 500 IEQ (17.2 mmol/l at 60 min). CONCLUSIONS/INTERPRETATION: The ability to efficiently reformat dissociated islet cells into engineered pseudoislets with improved properties has high potential for both research and therapeutic applications.


Subject(s)
Diabetes Mellitus/therapy , Insulin/blood , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Tissue Engineering , Animals , Apoptosis , Cell Survival , DNA/analysis , Diabetes Mellitus, Experimental/therapy , Female , Gene Expression Profiling , Glucose/metabolism , Graft Survival , Humans , Hyperglycemia , Hypoxia , Insulin/metabolism , Male , Mice , Mice, Transgenic
4.
Am J Respir Crit Care Med ; 180(11): 1131-42, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19713449

ABSTRACT

RATIONALE: Bronchopulmonary dysplasia (BPD) and emphysema are characterized by arrested alveolar development or loss of alveoli; both are significant global health problems and currently lack effective therapy. Bone marrow-derived mesenchymal stem cells (BMSCs) prevent adult lung injury, but their therapeutic potential in neonatal lung disease is unknown. OBJECTIVES: We hypothesized that intratracheal delivery of BMSCs would prevent alveolar destruction in experimental BPD. METHODS: In vitro, BMSC differentiation and migration were assessed using co-culture assays and a modified Boyden chamber. In vivo, the therapeutic potential of BMSCs was assessed in a chronic hyperoxia-induced model of BPD in newborn rats. MEASUREMENTS AND MAIN RESULTS: In vitro, BMSCs developed immunophenotypic and ultrastructural characteristics of type II alveolar epithelial cells (AEC2) (surfactant protein C expression and lamellar bodies) when co-cultured with lung tissue, but not with culture medium alone or liver. Migration assays revealed preferential attraction of BMSCs toward oxygen-damaged lung versus normal lung. In vivo, chronic hyperoxia in newborn rats led to air space enlargement and loss of lung capillaries, and this was associated with a decrease in circulating and resident lung BMSCs. Intratracheal delivery of BMSCs on Postnatal Day 4 improved survival and exercise tolerance while attenuating alveolar and lung vascular injury and pulmonary hypertension. Engrafted BMSCs coexpressed the AEC2-specific marker surfactant protein C. However, engraftment was disproportionately low for cell replacement to account for the therapeutic benefit, suggesting a paracrine-mediated mechanism. In vitro, BMSC-derived conditioned medium prevented O(2)-induced AEC2 apoptosis, accelerated AEC2 wound healing, and enhanced endothelial cord formation. CONCLUSIONS: BMSCs prevent arrested alveolar and vascular growth in part through paracrine activity. Stem cell-based therapies may offer new therapeutic avenues for lung diseases that currently lack efficient treatments.


Subject(s)
Lung Injury/prevention & control , Mesenchymal Stem Cells , Pulmonary Alveoli/growth & development , Animals , Animals, Newborn , Bone Marrow , Cell Culture Techniques , Disease Models, Animal , Exercise Tolerance , Hyperoxia , Hypertension, Pulmonary/prevention & control , Pulmonary Alveoli/ultrastructure , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
5.
J Thorac Cardiovasc Surg ; 132(5): 1156-61, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17059938

ABSTRACT

OBJECTIVE: Cryopreserved allograft cardiovascular tissue elicits a strong cellular and humoral response in recipients; this may accelerate the deterioration of the allograft and complicate future heart transplantation. Juvenile sheep are the standard model for in vivo valve research and have been used to investigate the allogeneic immune response to cardiac valve and vascular tissue transplantation. Studies to date have not considered the extent of allogenicity of sheep used in transplantation studies. METHODS: Functional allogenicity was assessed by standard one-way mixed lymphocyte reaction assay using peripheral blood mononuclear cells. Responder cells were stimulated with irradiated stimulator cells and cultured at 37 degrees C in 95% air and 5% carbon dioxide for 3, 4, 5, and 6 days. Cultures were pulsed with tritiated thymidine for 24 hours and harvested onto filtermats. RESULTS: The allogeneic response, measured as counts per minute, demonstrated a bimodal distribution. Fifty-nine (36.9%) of 160 pairs fell within the first peak (counts per minute < 10,000) and were defined as weak responders. The remaining 101 (63.1%) of 160 pairs of animals demonstrated a strong allogeneic response (counts per minute > or = 10,000) that followed a normal distribution. CONCLUSIONS: More than 1 in 3 pairs of sheep is too closely related to elicit an immune response when cross-reacted. This finding may alter the interpretation of studies that base their findings on allogeneic transplantations in sheep without ascertaining the genetic similarities of the animals. Valve transplantation studies in a sheep model should assess the extent of allogenicity of donor and recipient sheep.


Subject(s)
Lymphocyte Culture Test, Mixed , Transplantation Immunology/immunology , Transplantation, Homologous/immunology , Animals , Cryopreservation , Heart Transplantation , Heart Valves , Models, Animal , Sheep
6.
J Immunol ; 177(8): 5051-8, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015688

ABSTRACT

Sertoli cells have long since been recognized for their ability to suppress the immune system and protect themselves as well as other cell types from harmful immune reaction. However, the exact mechanism or product produced by Sertoli cells that affords this immunoprotection has never been fully elucidated. We examined the effect of mouse Sertoli cell-conditioned medium on human granzyme B-mediated killing and found that there was an inhibitory effect. We subsequently found that a factor secreted by Sertoli cells inhibited killing through the inhibition of granzyme B enzymatic activity. SDS-PAGE analysis revealed that this factor formed an SDS-insoluble complex with granzyme B. Immunoprecipitation and mass spectroscopic analysis of the complex identified a proteinase inhibitor, serpina3n, as a novel inhibitor of human granzyme B. We cloned serpina3n cDNA, expressed it in Jurkat cells, and confirmed its inhibitory action on granzyme B activity. Our studies have led to the discovery of a new inhibitor of granzyme B and have uncovered a new mechanism used by Sertoli cells for immunoprotection.


Subject(s)
Acute-Phase Proteins/isolation & purification , Granzymes/antagonists & inhibitors , Serpins/isolation & purification , Sertoli Cells/metabolism , Acute-Phase Proteins/immunology , Acute-Phase Proteins/metabolism , Animals , Biological Factors/immunology , Biological Factors/isolation & purification , Biological Factors/metabolism , Cells, Cultured , Cloning, Molecular , Humans , Immune System , Jurkat Cells , Male , Mice , Protein Binding , Serpins/immunology , Serpins/metabolism , Sertoli Cells/cytology , Sertoli Cells/immunology
7.
Transplantation ; 82(7): 945-52, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17038911

ABSTRACT

BACKGROUND: Despite the success of the Edmonton protocol for human islet transplantation, an alternate source of islet tissue must be developed if beta-cell replacement therapy is to see widespread application. Neonatal porcine islets (NPI) represent one potential source of tissue. When human or rodent islets are transplanted, the majority of cells undergo hypoxia-induce apoptosis soon after the grafts are placed in the recipient. In the present study, we investigated whether NPI were similarly sensitive to hypoxia. METHODS: NPI were exposed to hypoxia and hypoxia/reoxygenation using an in vitro hypoxic chamber. Afterwards, viability, frequency of apoptosis, and beta-cell function were evaluated. NPI and adult porcine islets were transplanted into chemically diabetic, immunodeficient mice and graft apoptosis was assessed 24 hours and seven days posttransplant. RESULTS: NPI demonstrated a remarkable capacity to resist apoptosis and maintain insulin secretion despite severe stresses such as hypoxia/reoxygenation. One day after transplantation, NPI grafts showed limited apoptosis, confined to rare strongly insulin positive cells. In contrast, adult porcine islet grafts underwent widespread apoptosis. Western blotting revealed that NPI express high levels of at least one potent endogenous antiapoptotic protein (XIAP). CONCLUSIONS: The majority of cells within transplanted human islets undergo apoptosis soon after portal infusion. In contrast, NPI have the capacity to resist this early posttransplant apoptosis, with likely reduced antigen release and diminished immune stimulation. NPI appear to contain a population of insulin-low to insulin-negative pre-beta-cells, which are resistant to hypoxia-induced apoptosis and still capable of differentiating into mature beta-cells.


Subject(s)
Apoptosis/physiology , Insulin-Secreting Cells/physiology , Aging , Animals , Animals, Newborn , Glucose/pharmacology , Hypoxia , Immunity, Innate , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Mice , Mice, Transgenic , Oxygen Consumption , Swine , X-Linked Inhibitor of Apoptosis Protein/genetics
8.
Circulation ; 114(4): 318-27, 2006 Jul 25.
Article in English | MEDLINE | ID: mdl-16831988

ABSTRACT

BACKGROUND: Glutaraldehyde fixation (G-F) decreases but likely does not eliminate the antigenicity of bioprosthetic heart valves. Rejection (with secondary dystrophic calcification) may be why G-F xenograft valves fail, especially in young patients, who are more immunocompetent than the elderly. Therefore, we sought to determine whether rejection of G-F xenograft occurs and to correlate this with graft calcification. METHODS AND RESULTS: Ascending aortas/valves (from rats [syngeneic] or guinea pigs [xenogeneic]) were transplanted (fresh or after 48 hour of G-F) into the infrarenal aortas of young rat recipients for 20 days. A xenogeneic group was also treated with steroids until graft harvest. The valves and media/adventitia were scored blindly for inflammation (0 to 4). Percent graft infiltration by T cells/macrophages was determined (immunohistochemistry), and rat IgG ELISAs were performed. There was >3 times more valve inflammation, >10 times more valve T-cell/macrophage infiltrate, and >3 times antibody rise in the G-F xenogeneic groups compared with the fresh syngeneic or the G-F syngeneic groups (P<0.05). There was >2 times more adventitial inflammation and T-cell/macrophage infiltrate in the xenogeneic groups (P<0.05). Steroid treatment decreased inflammation and antibody rise in the xenogeneic groups (P<0.05). Correlation analysis revealed media/adventitia inflammation (P=0.02) and percent macrophage (P=0.01) infiltration to be predictors of calcification. CONCLUSIONS: G-F xenografts have cellular/humoral rejection and calcify secondarily.


Subject(s)
Bioprosthesis , Calcinosis/etiology , Glutaral/pharmacology , Graft Rejection/etiology , Heart Valve Prosthesis , Prosthesis Failure , Animals , Calcinosis/pathology , Cell Movement , Glutaral/therapeutic use , Graft Rejection/immunology , Guinea Pigs , Inflammation/drug therapy , Macrophages/physiology , Rats , Steroids/pharmacology , Steroids/therapeutic use , T-Lymphocytes/physiology , Transplantation, Heterologous , Transplantation, Isogeneic
9.
Circulation ; 112(16): 2477-86, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-16230500

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia (BPD) and pulmonary emphysema, both significant global health problems, are characterized by a loss of alveoli. Vascular endothelial growth factor (VEGF) is a trophic factor required for endothelial cell survival and is abundantly expressed in the lung. METHODS AND RESULTS: We report that VEGF blockade decreases lung VEGF and VEGF receptor 2 (VEGFR-2) expression in newborn rats and impairs alveolar development, leading to alveolar simplification and loss of lung capillaries, mimicking BPD. In hyperoxia-induced BPD in newborn rats, air space enlargement and loss of lung capillaries are associated with decreased lung VEGF and VEGFR-2 expression. Postnatal intratracheal adenovirus-mediated VEGF gene therapy improves survival, promotes lung capillary formation, and preserves alveolar development in this model of irreversible lung injury. Combined VEGF and angiopoietin-1 gene transfer matures the new vasculature, reducing the vascular leakage seen in VEGF-induced capillaries. CONCLUSIONS: These findings underscore the importance of the vasculature in what is traditionally thought of as an airway disease and open new therapeutic avenues for lung diseases characterized by irreversible loss of alveoli through the modulation of angiogenic growth factors.


Subject(s)
Bronchopulmonary Dysplasia/therapy , Genetic Therapy , Hyperoxia/therapy , Lung/blood supply , Neovascularization, Physiologic , Pulmonary Alveoli/blood supply , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/therapeutic use , Adenoviridae , Angiography , Animals , Animals, Newborn , Disease Models, Animal , Endothelium, Vascular/physiology , Endothelium, Vascular/ultrastructure , Genetic Vectors , Humans , Infant, Newborn , Lung Injury , Pulmonary Artery , Rats
10.
Am J Respir Crit Care Med ; 172(6): 750-6, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15947285

ABSTRACT

RATIONALE: Bronchopulmonary dysplasia (BPD), the chronic lung disease of preterm infants, and pulmonary emphysema, both significant global health problems, are characterized by an arrest in alveolar growth/loss of alveoli structures. Mechanisms that inhibit distal lung growth are poorly understood, but recent studies suggest that impaired vascular endothelial growth factor signaling and reduced nitric oxide (NO) production decreases alveolar and vessel growth in the developing lung, features observed in experimental oxygen-induced BPD. NO exerts its biological activity by stimulating guanosine 3',5'-cyclic monophosphate (cGMP) production. OBJECTIVES: Because cGMP is inactivated by phosphodiesterase (PDE) enzymes, we hypothesized that the cGMP-specific PDE5 inhibitor sildenafil would promote angiogenesis and attenuate oxygen-induced lung injury in newborn rats. METHODS, MEASUREMENTS, AND MAIN RESULTS: In vitro, sildenafil (10(-4) M) increased endothelial capillary network formation of human pulmonary endothelial cells exposed to hyperoxia. In vivo, rat pups were randomly exposed from birth to normoxia, hyperoxia (95% O(2), BPD model), and hyperoxia+sildenafil (100 mg/kg/day subcutaneously). Rat pups exposed to hyperoxia showed fewer and enlarged air spaces as well as decreased capillary density, mimicking pathologic features seen in human BPD. These structural anomalies were associated with echographic (decreased pulmonary acceleration time) and structural (right ventricular hypertrophy and increased medial wall thickness) signs of pulmonary hypertension. Sildenafil preserved alveolar growth and lung angiogenesis, and decreased pulmonary vascular resistance, right ventricular hypertrophy and medial wall thickness. CONCLUSIONS: Our findings suggest a role for the NO/cGMP pathway during alveolar development. Sildenafil may have therapeutic potential in diseases associated with impaired alveolar structures.


Subject(s)
Hypertension, Pulmonary/physiopathology , Lung Diseases/physiopathology , Lung/pathology , Oxygen , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Pulmonary Alveoli/growth & development , Animals , Animals, Newborn , Cells, Cultured , Cyclic GMP/blood , Humans , Hypertension, Pulmonary/diagnostic imaging , Lung/blood supply , Lung/drug effects , Lung Diseases/chemically induced , Lung Diseases/pathology , Neovascularization, Physiologic/drug effects , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Purines , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Sulfones , Ultrasonography, Doppler
11.
Am J Physiol Endocrinol Metab ; 286(6): E1023-31, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14871885

ABSTRACT

Metformin, a drug widely used in the treatment of type 2 diabetes, has recently been shown to act on skeletal muscle and liver in part through the activation of AMP-activated protein kinase (AMPK). Whether metformin or the satiety factor leptin, which also stimulates AMPK in muscle, regulates this enzyme in pancreatic islets is unknown. We have recently shown that forced increases in AMPK activity inhibit insulin secretion from MIN6 cells (da Silva Xavier G, Leclerc I, Varadi A, Tsuboi T, Moule SK, and Rutter GA. Biochem J 371: 761-774, 2003). Here, we explore whether 1) glucose, metformin, or leptin regulates AMPK activity in isolated islets from rodent and human and 2) whether changes in AMPK activity modulate insulin secretion from human islets. Increases in glucose concentration from 0 to 3 and from 3 to 17 mM inhibited AMPK activity in primary islets from mouse, rat, and human, confirming previous findings in insulinoma cells. Incubation with metformin (0.2-1 mM) activated AMPK in both human islets and MIN6 beta-cells in parallel with an inhibition of insulin secretion, whereas leptin (10-100 nM) was without effect in MIN6 cells. These studies demonstrate that AMPK activity is subject to regulation by both glucose and metformin in pancreatic islets and clonal beta-cells. The inhibitory effects of metformin on insulin secretion may therefore need to be considered with respect to the use of this drug for the treatment of type 2 diabetes.


Subject(s)
Glucose/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Leptin/pharmacology , Metformin/pharmacology , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/enzymology , Mice , Mice, Inbred Strains , Multienzyme Complexes/genetics , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...