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










Publication year range
1.
Physiol Rep ; 8(13): e14444, 2020 07.
Article in English | MEDLINE | ID: mdl-32618430

ABSTRACT

Differences in pancreatic islet susceptibility during type 1 diabetes development may be explained by interislet variations. This study aimed to investigate if heterogeneities in vascular support and metabolic activity in rat and human islets may explain why some islets are attacked earlier than other islets. In rats, highly blood perfused islets were identified by injection of microspheres into the ascending aorta, whereas a combination of anterograde and retrograde injections of microspheres into pancreas was used to determine the islet vascular drainage system. Highly blood perfused islets had superior function and lower glucose threshold for insulin release when compared with other islets. These islets had a preferential direct venous drainage to the portal vein, whereas other islets mainly were incorporated into the exocrine capillary system. In BioBreeding rats, the hypothesis that islets with high islet blood perfusion was more prone to immune cell infiltration was investigated. Indeed, highly blood perfused islets were the first affected by the immune attack. In human subjects, differences in glucose threshold for insulin (C-peptide) secretion was evaluated in individuals recently diagnosed for type 1 diabetes and compared to control subjects. A preferential loss of capacity for insulin release in response to low glucose concentrations was observed at debut of type 1 diabetes. Our study indicates that highly blood perfused islets with direct venous drainage and lower glucose threshold for insulin release are of great importance for normal glucose homeostasis. At the same time, these highly metabolically active islets were the primary target of the immune system.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Insulin Secretion , Islets of Langerhans/immunology , Regional Blood Flow , Animals , Blood Glucose/metabolism , Cells, Cultured , Diabetes Mellitus, Type 1/physiopathology , Insulin/metabolism , Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , Male , Microspheres , Rats , Rats, Sprague-Dawley
2.
Cell Transplant ; 28(11): 1455-1460, 2019 11.
Article in English | MEDLINE | ID: mdl-31359771

ABSTRACT

Beta cell replacement is an exciting field where new beta cell sources and alternative sites are widely explored. The liver has been the implantation site of choice in the clinic since the advent of islet transplantation. However, in most cases, repeated islet transplantation is needed to achieve normoglycemia in diabetic recipients. This study aimed to investigate whether there are differences in islet survival and engraftment between a first and a second transplantation, performed 1 week apart, to the liver. C57BL/6 mice were accordingly transplanted twice with an initial infusion of syngeneic islets expressing green fluorescent protein (GFP). The second islet transplant was performed 1 week later and consisted of islets isolated from non-GFP C57BL/6-mice. Animals were sacrificed either 1 day or 1 month after the second transplantation. A control group received a saline infusion instead of GFP-expressing islets, 1 week later obtained a standard non-GFP islet transplant, and was subsequently sacrificed 1 month later. Islet engraftment in the liver was assessed by immunohistochemistry and serum was analyzed for angiogenic factors induced by the first islet transplantation. Almost 70% of islets found in the liver following repeated islet transplantation originated from the second transplantation. The vascular density in the transplanted non-GFP-expressing islets did not differ depending on whether their transplantation was preceded by a primary islet transplantation or saline administration only nor did angiogenic factors in serum prior to the transplantation of non-GFP islets differ between animals that had received a previous islet transplantation or a saline infusion. We conclude that first islet transplantation creates, by unknown mechanisms, favorable conditions for the survival of a second transplant to the liver.


Subject(s)
Graft Survival , Islets of Langerhans Transplantation/methods , Animals , Cells, Cultured , Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Transplantation, Homologous/methods
3.
Diabetologia ; 61(4): 896-905, 2018 04.
Article in English | MEDLINE | ID: mdl-29209740

ABSTRACT

AIMS/HYPOTHESIS: Genetic studies show coupling of genes affecting beta cell function to type 1 diabetes, but hitherto no studies on whether beta cell dysfunction could precede insulitis and clinical onset of type 1 diabetes are available. METHODS: We used 40-day-old BioBreeding (BB) DRLyp/Lyp rats (a model of spontaneous autoimmune type 1 diabetes) and diabetes-resistant DRLyp/+ and DR+/+ littermates (controls) to investigate beta cell function in vivo, and insulin and glucagon secretion in vitro. Beta cell mass was assessed by optical projection tomography (OPT) and morphometry. Additionally, measurements of intra-islet blood flow were performed using microsphere injections. We also assessed immune cell infiltration, cytokine expression in islets (by immunohistochemistry and qPCR), as well as islet Glut2 expression and ATP/ADP ratio to determine effects on glucose uptake and metabolism in beta cells. RESULTS: DRLyp/Lyp rats were normoglycaemic and without traces of immune cell infiltrates. However, IVGTTs revealed a significant decrease in the acute insulin response to glucose compared with control rats (1685.3 ± 121.3 vs 633.3 ± 148.7; p < 0.0001). In agreement, insulin secretion was severely perturbed in isolated islets, and both first- and second-phase insulin release were lowered compared with control rats, while glucagon secretion was similar in both groups. Interestingly, after 5-7 days of culture of islets from DRLyp/Lyp rats in normal media, glucose-stimulated insulin secretion (GSIS) was improved; although, a significant decrease in GSIS was still evident compared with islets from control rats at this time (7393.9 ± 1593.7 vs 4416.8 ± 1230.5 pg islet-1 h-1; p < 0.0001). Compared with controls, OPT of whole pancreas from DRLyp/Lyp rats revealed significant reductions in medium (4.1 × 109 ± 9.5 × 107 vs 3.8 × 109 ± 5.8 × 107 µm3; p = 0.044) and small sized islets (1.6 × 109 ± 5.1 × 107 vs 1.4 × 109 ± 4.5 × 107 µm3; p = 0.035). Finally, we found lower intra-islet blood perfusion in vivo (113.1 ± 16.8 vs 76.9 ± 11.8 µl min-1 [g pancreas]-1; p = 0.023) and alterations in the beta cell ATP/ADP ratio in DRLyp/Lyp rats vs control rats. CONCLUSIONS/INTERPRETATION: The present study identifies a deterioration of beta cell function and mass, and intra-islet blood flow that precedes insulitis and diabetes development in animals prone to autoimmune type 1 diabetes. These underlying changes in islet function may be previously unrecognised factors of importance in type 1 diabetes development.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Insulin-Secreting Cells/cytology , Insulin/metabolism , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Animals , Blood Glucose/metabolism , Female , Genotype , Glucose/metabolism , Islets of Langerhans/metabolism , Langerhans Cells/metabolism , Male , Pancreas/metabolism , Perfusion , Rats , Rats, Inbred BB , Rats, Wistar
4.
Ups J Med Sci ; 121(2): 81-95, 2016 May.
Article in English | MEDLINE | ID: mdl-27124642

ABSTRACT

Pancreatic islets are richly vascularized, and islet blood vessels are uniquely adapted to maintain and support the internal milieu of the islets favoring normal endocrine function. Islet blood flow is normally very high compared with that to the exocrine pancreas and is autonomously regulated through complex interactions between the nervous system, metabolites from insulin secreting ß-cells, endothelium-derived mediators, and hormones. The islet blood flow is normally coupled to the needs for insulin release and is usually disturbed during glucose intolerance and overt diabetes. The present review provides a brief background on islet vascular function and especially focuses on available techniques to measure islet blood perfusion. The gold standard for islet blood flow measurements in experimental animals is the microsphere technique, and its advantages and disadvantages will be discussed. In humans there are still no methods to measure islet blood flow selectively, but new developments in radiological techniques hold great hopes for the future.


Subject(s)
Blood Flow Velocity , Islets of Langerhans/blood supply , Animals , Blood Glucose/metabolism , Blood Pressure , Capillaries/metabolism , Hemodynamics , Hormones/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Microspheres , Neurotransmitter Agents/metabolism , Pancreas/metabolism , Perfusion , Regional Blood Flow , Veins/metabolism
5.
Transplantation ; 99(10): 2077-82, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26426924

ABSTRACT

BACKGROUND: Muscle is a promising alternative site for islet transplantation that facilitates rapid restoration of islet vascularization. However, the development of fibrosis suggests massive cellular death after transplantation. This study tested the hypothesis that islet graft function is limited by hypoxia-related death early after intramuscular transplantation, but that this can be overcome by cotransplantation of an oxygen carrier, that is, polymerized bovine hemoglobin (PolyHb). METHODS: Two hundred islets were transplanted with or without different doses of PolyHb intramuscularly to nondiabetic C57BL/6 and diabetic C57BL/6 nu/nu mice. ß-cell hypoxia and apoptosis were evaluated by immunohistochemistry after injection of the biochemical marker pimonidazole or by staining for caspase-3, respectively. Blood glucose concentrations were monitored for 30 days after islet transplantation and animals were then subjected to an intravenous glucose tolerance test. RESULTS: Substantial hypoxia was observed in control islet grafts during the first 4 days after transplantation. Cotransplantation of PolyHb resulted in a dose-dependent reduction of ß-cell hypoxia, but ß-cell apoptosis was only reduced by cotransplantation of low-dose PolyHb (0.03 mg/g body weight) due to the inflammatory effects of higher PolyHb concentrations. Cotransplantation of low-dose PolyHb resulted in improved islet graft function 30 days after transplantation in diabetic mice, with a glucose tolerance comparable to transplantation of 50% more islets. CONCLUSION: We conclude that cotransplantation of islets with PolyHb can be used to effectively bridge the critical hypoxic phase immediately after transplantation, improve islet graft function, and reduce the number of islets needed for successful intramuscular transplantation.


Subject(s)
Hemoglobins/chemistry , Hypoxia/pathology , Insulin-Secreting Cells/cytology , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Muscles/pathology , Polymers/chemistry , Animals , Apoptosis , Blood Glucose/chemistry , Caspase 3/metabolism , Cattle , Cell Survival , Glucose Tolerance Test , Immunohistochemistry , Injections, Intramuscular , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Oxygen/chemistry
6.
Microvasc Res ; 97: 124-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25446368

ABSTRACT

The blood perfusion of pancreatic islets is regulated independently from that of the exocrine pancreas, and is of importance for multiple aspects of normal islet function, and probably also during impaired glucose tolerance. Single islet blood flow has been difficult to evaluate due to technical limitations. We therefore adapted a hydrogen gas washout technique using microelectrodes to allow such measurements. Platinum micro-electrodes monitored hydrogen gas clearance from individual endogenous and transplanted islets in the pancreas of male Lewis rats and in human and mouse islets implanted under the renal capsule of male athymic mice. Both in the rat endogenous pancreatic islets as well as in the intra-pancreatically transplanted islets, the vascular conductance and blood flow values displayed a highly heterogeneous distribution, varying by factors 6-10 within the same pancreas. The blood flow of human and mouse islet grafts transplanted in athymic mice was approximately 30% lower than that in the surrounding renal parenchyma. The present technique provides unique opportunities to study the islet vascular dysfunction seen after transplantation, but also allows for investigating the effects of genetic and environmental perturbations on islet blood flow at the single islet level in vivo.


Subject(s)
Hydrogen , Islets of Langerhans Transplantation , Islets of Langerhans/blood supply , Islets of Langerhans/surgery , Rheology/methods , Animals , Blood Flow Velocity , Equipment Design , Feasibility Studies , Gases , Heterografts , Humans , Hydrogen/blood , Laser-Doppler Flowmetry , Male , Mice, Inbred C57BL , Mice, Nude , Microelectrodes , Microspheres , Predictive Value of Tests , Rats, Inbred Lew , Regional Blood Flow , Rheology/instrumentation , Time Factors
7.
Acta Radiol ; 55(3): 328-34, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23963152

ABSTRACT

BACKGROUND: Previous studies have demonstrated that magnetic resonance imaging may be a method of choice to visualize transplanted pancreatic islets. However, contrast agents may interfere with microcirculation and affect graft function. PURPOSE: To evaluate the effects manganese-containing contrast media on regional blood flow and glucose tolerance. MATERIAL AND METHODS: Anesthetized rats were injected intravenously with MnCl2 (10 µM/kg body weight) or Mn-DPDP (Teslascan™; 5 µM/kg body weight). Blood flow measurements were made with a microsphere technique 10 min later. In separate animals vascular arteriolar reactivity in isolated, perfused islets was examined. Furthermore, an intraperitoneal glucose tolerance test was performed in separate rats. RESULTS: Glucose tolerance was unaffected by both agents. No changes in regional blood flow were seen after administration of Mn-DPDP, except for an increase in arterial liver blood flow. MnCl2 increased all blood flow values except that of the kidney. MnCl2, but not Mn-DPDP, caused a vasoconstriction in isolated rat islet arterioles but only at very high doses. CONCLUSION: Mn-DPDP administration does not affect glucose tolerance or regional blood flow, besides an increase in arterial hepatic blood flow, and may therefore be suitable for visualization of islets.


Subject(s)
Chlorides/pharmacology , Contrast Media/pharmacology , Edetic Acid/analogs & derivatives , Hemodynamics/drug effects , Islets of Langerhans/blood supply , Islets of Langerhans/drug effects , Magnetic Resonance Imaging/methods , Manganese Compounds/pharmacology , Pyridoxal Phosphate/analogs & derivatives , Animals , Edetic Acid/pharmacology , Glucose Tolerance Test , Male , Microspheres , Pyridoxal Phosphate/pharmacology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects
8.
Am J Pathol ; 182(6): 2355-67, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23583653

ABSTRACT

Vascular therapeutic targeting requires thorough evaluation of the mechanisms activated in the specific context of each particular tumor type. We highlight structural, molecular, and functional microvascular aberrations contributing to development and maintenance of pancreatic neuroendocrine tumors (NETs), with special reference to multiple endocrine neoplasia 1 (MEN1) syndrome, using a Men1 mouse model. Tissue samples were analyzed by immunofluorescence to detect vessel density and pericyte distribution within the endocrine pancreas; expression of angiogenic factors was assessed by immunohistochemistry and quantitative real-time PCR in isolated islets and adenomas cultured under normoxic or hypoxic conditions. The increased vascular density of pancreatic NETs developed in Men1 mice was paralleled by an early and extensive redistribution of pericytes within endocrine tissue. These morphological alterations are supported by, and in some cases preceded by, fine-tuned variations in expression of several angiogenic regulators and are further potentiated by hypoxia. By combining two novel ex vivo and in vivo single-islet and tumor perfusion techniques, we demonstrated that both vascular reactivity and blood perfusion of tumor arterioles are significantly altered in response to glucose and L-nitro-arginine methyl ester. Our findings unravel multiple potential molecular and physiological targets differentially activated in the endocrine pancreas of Men1 mice and highlight the need for in-depth functional studies to fully understand the contribution of each component to development of pancreatic NETs in MEN1 syndrome.


Subject(s)
Islets of Langerhans/blood supply , Multiple Endocrine Neoplasia Type 1/blood supply , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood supply , Angiogenesis Inducing Agents/metabolism , Animals , Arterioles/physiology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Microvessels/pathology , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 1/pathology , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pericytes/pathology , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics
9.
J Exp Med ; 209(7): 1363-77, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22689825

ABSTRACT

Regulation of vascular endothelial (VE) growth factor (VEGF)-induced permeability is critical in physiological and pathological processes. We show that tyrosine phosphorylation of VEGF receptor 2 (VEGFR2) at Y951 facilitates binding of VEGFR2 to the Rous sarcoma (Src) homology 2-domain of T cell-specific adaptor (TSAd), which in turn regulates VEGF-induced activation of the c-Src tyrosine kinase and vascular permeability. c-Src was activated in vivo and in vitro in a VEGF/TSAd-dependent manner, and was regulated via increased phosphorylation at pY418 and reduced phosphorylation at pY527. Tsad silencing blocked VEGF-induced c-Src activation, but did not affect pathways involving phospholipase Cγ, extracellular regulated kinase, and endothelial nitric oxide. VEGF-induced rearrangement of VE-cadherin-positive junctions in endothelial cells isolated from mouse lungs, or in mouse cremaster vessels, was dependent on TSAd expression, and TSAd formed a complex with VE-cadherin, VEGFR2, and c-Src at endothelial junctions. Vessels in tsad(-/-) mice showed undisturbed flow and pressure, but impaired VEGF-induced permeability, as measured by extravasation of Evans blue, dextran, and microspheres in the skin and the trachea. Histamine-induced extravasation was not affected by TSAd deficiency. We conclude that TSAd is required for VEGF-induced, c-Src-mediated regulation of endothelial cell junctions and for vascular permeability.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Capillary Permeability/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/metabolism , Blotting, Western , Cadherins/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extravasation of Diagnostic and Therapeutic Materials/etiology , Female , Fluorescein-5-isothiocyanate/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microspheres , Phosphorylation/drug effects , Protein Binding , RNA Interference , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
10.
Xenotransplantation ; 19(3): 166-76, 2012.
Article in English | MEDLINE | ID: mdl-22702468

ABSTRACT

BACKGROUND: Nitrones such as 2-sulfo-phenyl-N-tert-butyl nitrone (S-PBN) are known to trap and stabilize free radicals and to reduce inflammation. Recently, S-PBN was shown to reduce infiltration of T lymphocytes and the expression of adhesion molecules on the endothelium in experimental traumatic brain injury. We hypothesized that S-PBN could reduce infiltration of T lymphocytes during cell-mediated xenograft rejection and thereby increase graft survival. The concordant mouse-to-rat heart transplantation model was used to test the hypothesis. In this model, grafts undergo acute humoral xenograft rejection (AHXR) almost invariably on day 3 and succumb to cell-mediated rejection on approximately day 8 if AHXR is inhibited by treatment with 15-deoxyspergualin (DSG). MATERIAL AND METHODS: Hearts from Naval Medical Research Institute (NMRI) mice were transplanted to the neck vessels of Lewis rats. Recipients were treated with S-PBN (n=9), DSG (n=9), S-PBN and DSG in combination (n=10) or left untreated (n=9) for survival studies. S-PBN was given daily intraperitoneally at a dose of 150 mg/kg body weight (BW) on day -1 to 30, and DSG was given daily intraperitoneally at a dose of 10 mg/kg BW on day -1 to 4 and 5 mg/kg BW on day 5 to 21. Nine additional recipients were given S-PBN only on days -1 and 0 in combination with continuous DSG treatment. Grafts were monitored until they stopped beating. Additional recipients were treated with S-PBN (n=5), DSG (n=5), S-PBN and DSG in combination (n=6) or left untreated (n=5) for morphological, immunohistochemical and flow cytometry analyses on days 2 and 6 after transplantation. RESULTS: S-PBN treatment in combination with DSG resulted in increased median graft survival compared to DSG treatment alone (14 vs. 7 days; P=0.019). Lower number of T lymphocytes on day 6 (P=0.019) was observed by ex vivo propagation and flow cytometry when combining S-PBN with DSG, whereas immunohistochemical analyses demonstrated a significant reduction in the number of infiltrated CD4+, but not TCR+, cells. S-PBN treatment alone had no impact on graft survival compared to untreated rats (3 vs. 3 days). No differences were seen in ICAM-1 and VCAM-1 expression or in morphology between the groups. CONCLUSION: The combination of S-PBN and DSG treatment increases xenograft survival. The main effect of S-PBN appears to be in direct connection with the transplantation. Because of its low toxicity, S-PBN could become useful in combination with other immunosuppressants to reduce cell-mediated xenograft rejection.


Subject(s)
Benzenesulfonates/pharmacology , Free Radical Scavengers/pharmacology , Graft Survival/drug effects , Guanidines/pharmacology , Heart Transplantation/methods , Transplantation, Heterologous/methods , Animals , Animals, Outbred Strains , Drug Therapy, Combination , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Survival/immunology , Immunosuppressive Agents/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Male , Mice , Rats , Rats, Inbred Lew , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Tissue and Organ Harvesting/methods , Vascular Cell Adhesion Molecule-1/metabolism
11.
Transpl Immunol ; 16(2): 81-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16860709

ABSTRACT

Objective and fast methods to diagnose rejection after organ transplantation are needed. In the present study, the ex vivo propagation technique was evaluated for its ability to detect rejection at two different time-points after experimental heart transplantation. Syngeneic and allogeneic heterotopic heart transplantations were performed using inbred rat strains. After 6 or 15 days, cardiac graft biopsies were put in culture and infiltrating cells isolated by the ex vivo propagation technique. The isolated cells were counted and phenotyped by flow cytometry. In parallel, graft sections were analysed with regard to morphology and the presence of infiltrating cells as determined by immunohistochemical stainings. On day 15 after transplantation, the number of cells possible to isolate through ex vivo propagation reflected the morphological changes of the graft, i.e. considerably more cells were obtained from allogeneic transplants undergoing rejection (1052 +/- 205) than from allogeneic grafts under cyclosporine protection (513 +/- 135; p < 0.05) or from syngeneic grafts (378 +/- 87; p < 0.01). Six days after transplantation the allogeneic grafts were strongly rejected with massive cellular infiltration, still there was no difference between allogeneic and syngeneic grafts as to the number of ex vivo propagated cells. However, the proportion of IL-2-receptor expressing T lymphocytes was increased (15.4 +/- 1.8% vs. 9.5 +/- 1.4%; p < 0.05) and the CD4/CD8 ratio reduced (1.0 +/- 0.1 vs. 2.8 +/- 0.2; p < 0.001) in the allogeneic group as compared with the syngeneic. We conclude that the ex vivo propagation technique can be used to distinguish rejection from non-rejection both early and later after transplantation, provided that not just cell counting but also phenotyping of the graft-infiltrating cells is performed.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Animals , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Graft Rejection/etiology , Graft Rejection/pathology , Heart Transplantation/adverse effects , Heart Transplantation/pathology , Male , Monitoring, Physiologic , Postoperative Period , Rats , Rats, Inbred WKY , Receptors, Interleukin-2/immunology , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...