Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 698
Filtrer
1.
J Diabetes Res ; 2024: 5574968, 2024.
Article de Anglais | MEDLINE | ID: mdl-38800586

RÉSUMÉ

Islet transplantation (ITx) is an established and safe alternative to pancreas transplantation for type 1 diabetes mellitus (T1DM) patients. However, most ITx recipients lose insulin independence by 3 years after ITx due to early graft loss, such that multiple donors are required to achieve insulin independence. In the present study, we investigated whether skeletal myoblast cells could be beneficial for promoting angiogenesis and maintaining the differentiated phenotypes of islets. In vitro experiments showed that the myoblast cells secreted angiogenesis-related cytokines (vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and stromal-derived factor-1α (SDF-1α)), contributed to maintenance of differentiated islet phenotypes, and enhanced islet cell insulin secretion capacity. To verify these findings in vivo, we transplanted islets alone or with myoblast cells under the kidney capsule of streptozotocin-induced diabetic mice. Compared with islets alone, the group bearing islets with myoblast cells had a significantly lower average blood glucose level. Histological examination revealed that transplants with islets plus myoblast cells were associated with a significantly larger insulin-positive area and significantly higher number of CD31-positive microvessels compared to islets alone. Furthermore, islets cotransplanted with myoblast cells showed JAK-STAT signaling activation. Our results suggest two possible mechanisms underlying enhancement of islet graft function with myoblast cells cotransplantation: "indirect effects" mediated by angiogenesis and "direct effects" of myoblast cells on islets via the JAK-STAT cascade. Overall, these findings suggest that skeletal myoblast cells enhance the function of transplanted islets, implying clinical potential for a novel ITx procedure involving myoblast cells for patients with diabetes.


Sujet(s)
Diabète expérimental , Insuline , Transplantation d'ilots de Langerhans , Myoblastes squelettiques , Néovascularisation physiologique , Animaux , Transplantation d'ilots de Langerhans/méthodes , Diabète expérimental/métabolisme , Myoblastes squelettiques/transplantation , Myoblastes squelettiques/métabolisme , Souris , Mâle , Insuline/métabolisme , Facteur de croissance des hépatocytes/métabolisme , Souris de lignée C57BL , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Ilots pancréatiques/métabolisme , Ilots pancréatiques/vascularisation , Chimiokine CXCL12/métabolisme , Glycémie/métabolisme , Diabète de type 1/métabolisme , Diabète de type 1/physiopathologie , Diabète de type 1/chirurgie , Transduction du signal , Sécrétion d'insuline , Différenciation cellulaire
2.
Int J Biol Macromol ; 271(Pt 2): 132376, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38750865

RÉSUMÉ

Diabetes is a complex metabolic disease and islet transplantation is a promising approach for the treatment of diabetes. Unfortunately, the transplanted islets at the subcutaneous site are also affected by various adverse factors such as poor vascularization and hypoxia. In this study, we utilize biocompatible copolymers l-lactide and D,l-lactide to manufacture a biomaterial scaffold with a mesh-like structure via 3D printing technology, providing a material foundation for encapsulating pancreatic islet cells. The scaffold maintains the sustained release of vascular endothelial growth factor (VEGF) and a slow release of oxygen from calcium peroxide (CPO), thereby regulating the microenvironment for islet survival. This helps to improve insufficient subcutaneous vascularization and reduce islet death due to hypoxia post-transplantation. By pre-implanting VEGF-CPO scaffolds subcutaneously into diabetic rats, a sufficiently vascularized site is formed, thereby ensuring early survival of transplanted islets. In a word, the VEGF-CPO scaffold shows good biocompatibility both in vitro and in vivo, avoids the adverse effects on the implanted islets, and displays promising clinical transformation prospects.


Sujet(s)
Matériaux biocompatibles , Diabète expérimental , Transplantation d'ilots de Langerhans , Ilots pancréatiques , Impression tridimensionnelle , Structures d'échafaudage tissulaires , Facteur de croissance endothéliale vasculaire de type A , Animaux , Structures d'échafaudage tissulaires/composition chimique , Rats , Transplantation d'ilots de Langerhans/méthodes , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Diabète expérimental/thérapie , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/vascularisation , Ilots pancréatiques/métabolisme , Mâle , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Peroxydes
3.
Sci Rep ; 14(1): 10136, 2024 05 02.
Article de Anglais | MEDLINE | ID: mdl-38698049

RÉSUMÉ

Exocrine and endocrine pancreas are interconnected anatomically and functionally, with vasculature facilitating bidirectional communication. Our understanding of this network remains limited, largely due to two-dimensional histology and missing combination with three-dimensional imaging. In this study, a multiscale 3D-imaging process was used to analyze a porcine pancreas. Clinical computed tomography, digital volume tomography, micro-computed tomography and Synchrotron-based propagation-based imaging were applied consecutively. Fields of view correlated inversely with attainable resolution from a whole organism level down to capillary structures with a voxel edge length of 2.0 µm. Segmented vascular networks from 3D-imaging data were correlated with tissue sections stained by immunohistochemistry and revealed highly vascularized regions to be intra-islet capillaries of islets of Langerhans. Generated 3D-datasets allowed for three-dimensional qualitative and quantitative organ and vessel structure analysis. Beyond this study, the method shows potential for application across a wide range of patho-morphology analyses and might possibly provide microstructural blueprints for biotissue engineering.


Sujet(s)
Imagerie tridimensionnelle , Imagerie multimodale , Pancréas , Animaux , Imagerie tridimensionnelle/méthodes , Pancréas/imagerie diagnostique , Pancréas/vascularisation , Suidae , Imagerie multimodale/méthodes , Microtomographie aux rayons X/méthodes , Ilots pancréatiques/imagerie diagnostique , Ilots pancréatiques/vascularisation , Tomodensitométrie/méthodes
4.
Diabetes ; 73(7): 1127-1139, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38603470

RÉSUMÉ

Pluripotent stem cell-derived islets (SC-islets) have emerged as a new source for ß-cell replacement therapy. The function of human islet transplants is hampered by excessive cell death posttransplantation; contributing factors include inflammatory reactions, insufficient revascularization, and islet amyloid formation. However, there is a gap in knowledge of the engraftment process of SC-islets. In this experimental study, we investigated the engraftment capability of SC-islets at 3 months posttransplantation and observed that cell apoptosis rates were lower but vascular density was similar in SC-islets compared with human islets. Whereas the human islet transplant vascular structures were a mixture of remnant donor endothelium and ingrowing blood vessels, the SC-islets contained ingrowing blood vessels only. Oxygenation in the SC-islet grafts was twice as high as that in the corresponding grafts of human islets, suggesting better vascular functionality. Similar to the blood vessel ingrowth, reinnervation of the SC-islets was four- to fivefold higher than that of the human islets. Both SC-islets and human islets contained amyloid at 1 and 3 months posttransplantation. We conclude that the vascular and neural engraftment of SC-islets are superior to those of human islets, but grafts of both origins develop amyloid, with potential long-term consequences.


Sujet(s)
Transplantation d'ilots de Langerhans , Ilots pancréatiques , Humains , Transplantation d'ilots de Langerhans/méthodes , Ilots pancréatiques/vascularisation , Ilots pancréatiques/cytologie , Ilots pancréatiques/physiologie , Animaux , Souris , Apoptose/physiologie , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/physiologie , Survie du greffon/physiologie , Mâle
5.
Acta Diabetol ; 61(7): 897-907, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38530415

RÉSUMÉ

OBJECTIVE: To examine the association between islet autoantibodies (IAbs) and the retinal neurovascular changes in type 1 diabetes mellitus (T1DM) with no diabetic retinopathy (NDR). METHODS: This cross-sectional study measured the neural retinal structure and microvascular density of 118 NDR eyes using spectral-domain optical coherence tomography angiography. Retinal structure parameters included retinal thickness (RT), inner retinal thickness (iRT), retina never fibral layer thickness (RNFL thickness), ganglion cell complex thickness (GCC thickness), and loss volume of GCC. Microvascular parameters included vessel density of superficial capillary plexus (sVD), vessel density of deep capillary plexus, and vessel density of choroid capillary plexus. Comparison and correlation analyses of these OCTA parameters were made with various IAbs, including glutamic acid decarboxylase antibody (GADA), tyrosine phosphatase-related islet antigen 2 antibody (IA2A), and zinc transporter 8 antibody (ZnT8A). A general linear model was used to understand the association of IAbs with the retina parameters. RESULTS: The IAb positive (IAbs +) group, which included 85 patients, had thinner RT (235.20 ± 18.10 mm vs. 244.40 ± 19.90 mm at fovea, P = 0.021) and thinner iRT (120.10 ± 9.00 mm vs. 124.70 ± 6.90 mm at parafovea, P = 0.015), compared with the IAb negative (IAbs-) group comprising 33 patients. Furthermore, a more severe reduction of RT was demonstrated in the presence of multiple IAbs. Among the three IAbs, GADA was the most significant independent risk factor of all-round RT decrease (ß = -0.20 vs. -0.27 at fovea and parafovea, respectively, P < 0.05), while titers of IA2A negatively affect sVD in the parafovea (ß = -0.316, P = 0.003). CONCLUSIONS: IAbs are associated with neural retinal thinning and microcirculation reduction in T1DM patients before the clinical onset of diabetic retinopathy.


Sujet(s)
Autoanticorps , Diabète de type 1 , Rétinopathie diabétique , Microcirculation , Rétine , Humains , Autoanticorps/sang , Autoanticorps/immunologie , Diabète de type 1/immunologie , Diabète de type 1/anatomopathologie , Diabète de type 1/imagerie diagnostique , Mâle , Femelle , Études transversales , Adulte , Rétinopathie diabétique/immunologie , Rétinopathie diabétique/anatomopathologie , Rétinopathie diabétique/imagerie diagnostique , Rétine/imagerie diagnostique , Rétine/immunologie , Rétine/anatomopathologie , Adulte d'âge moyen , Tomographie par cohérence optique , Ilots pancréatiques/immunologie , Ilots pancréatiques/imagerie diagnostique , Ilots pancréatiques/anatomopathologie , Ilots pancréatiques/vascularisation , Vaisseaux rétiniens/imagerie diagnostique , Vaisseaux rétiniens/anatomopathologie , Jeune adulte
6.
J Histochem Cytochem ; 72(3): 131-148, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38454609

RÉSUMÉ

Diabetes is not only an endocrine but also a vascular disease. Vascular defects are usually seen as consequence of diabetes. However, at the level of the pancreatic islet, vascular alterations have been described before symptom onset. Importantly, the cellular and molecular mechanisms underlying these early vascular defects have not been identified, neither how these could impact the function of islet endocrine cells. In this review, we will discuss the possibility that dysfunction of the mural cells of the microvasculature-known as pericytes-underlies vascular defects observed in islets in pre-symptomatic stages. Pericytes are crucial for vascular homeostasis throughout the body, but their physiological and pathophysiological functions in islets have only recently started to be explored. A previous study had already raised interest in the "microvascular" approach to this disease. With our increased understanding of the crucial role of the islet microvasculature for glucose homeostasis, here we will revisit the vascular aspects of islet function and how their deregulation could contribute to diabetes pathogenesis, focusing in particular on type 1 diabetes (T1D).


Sujet(s)
Diabète de type 1 , Ilots pancréatiques , Humains , Péricytes , Ilots pancréatiques/vascularisation , Diabète de type 1/anatomopathologie , Microvaisseaux/anatomopathologie
7.
Nat Commun ; 15(1): 1452, 2024 Feb 16.
Article de Anglais | MEDLINE | ID: mdl-38365780

RÉSUMÉ

The development of vascular networks in microfluidic chips is crucial for the long-term culture of three-dimensional cell aggregates such as spheroids, organoids, tumoroids, or tissue explants. Despite rapid advancement in microvascular network systems and organoid technologies, vascularizing organoids-on-chips remains a challenge in tissue engineering. Most existing microfluidic devices poorly reflect the complexity of in vivo flows and require complex technical set-ups. Considering these constraints, we develop a platform to establish and monitor the formation of endothelial networks around mesenchymal and pancreatic islet spheroids, as well as blood vessel organoids generated from pluripotent stem cells, cultured for up to 30 days on-chip. We show that these networks establish functional connections with the endothelium-rich spheroids and vascular organoids, as they successfully provide intravascular perfusion to these structures. We find that organoid growth, maturation, and function are enhanced when cultured on-chip using our vascularization method. This microphysiological system represents a viable organ-on-chip model to vascularize diverse biological 3D tissues and sets the stage to establish organoid perfusions using advanced microfluidics.


Sujet(s)
Ilots pancréatiques , Microfluidique , Organoïdes , Ingénierie tissulaire/méthodes , Endothélium , Ilots pancréatiques/vascularisation
8.
Diabetes ; 73(4): 533-541, 2024 04 01.
Article de Anglais | MEDLINE | ID: mdl-38215069

RÉSUMÉ

For many years, it has been taught in medical textbooks that the endocrine and exocrine parts of the pancreas have separate blood supplies that do not mix. Therefore, they have been studied by different scientific communities, and patients with pancreatic disorders are treated by physicians in different medical disciplines, where endocrine and exocrine function are the focus of endocrinologists and gastroenterologists, respectively. The conventional model that every islet in each pancreatic lobule receives a dedicated arterial blood supply was first proposed in 1932, and it has been inherited to date. Recently, in vivo intravital recording of red blood cell flow in mouse islets as well as in situ structural analysis of 3D pancreatic vasculature from hundreds of islets provided evidence for preferentially integrated pancreatic blood flow in six mammalian species. The majority of islets have no association with the arteriole, and there is bidirectional blood exchange between the two segments. Such vascularization may allow an entire downstream region of islets and acinar cells to be simultaneously exposed to a topologically and temporally specific plasma content, which could underlie an adaptive sensory function as well as common pathogeneses of both portions of the organ in pancreatic diseases, including diabetes.


Sujet(s)
Diabète , Ilots pancréatiques , Pancréas exocrine , Souris , Animaux , Humains , Ilots pancréatiques/vascularisation , Pancréas/physiologie , Cellules acineuses , Mammifères
9.
Transplantation ; 108(5): 1115-1126, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38192025

RÉSUMÉ

BACKGROUND: The utilization of islet-like cells derived from pluripotent stem cells may resolve the scarcity of islet transplantation donors. The subcutaneous space is a promising transplantation site because of its capacity for graft observation and removal, thereby ensuring safety. To guarantee subcutaneous islet transplantation, physicians should ensure ample blood supply. Numerous methodologies, including prevascularization, have been investigated to augment blood flow, but the optimal approach remains undetermined. METHODS: From C57BL/6 mice, 500 syngeneic islets were transplanted into the prevascularized subcutaneous site of recipient mice by implanting agarose rods with basic fibroblast growth factor at 1 and 2 wk. Before transplantation, the blood glucose levels, cell infiltration, and cytokine levels at the transplant site were evaluated. Furthermore, we examined the impact of the extracellular matrix capsule on graft function and the inflammatory response. RESULTS: Compared with the 1-wk group, the 2-wk group exhibited improved glycemic control, indicating that longer prevascularization enhanced transplant success. Flow cytometry analysis detected immune cells, such as neutrophils and macrophages, in the extracellular matrix capsules, whereas cytometric bead array analysis indicated the release of inflammatory and proinflammatory cytokines. Treatment with antitumor necrosis factor and anti-interleukin-6R antibodies in the 1-wk group improved graft survival, similar to the 2-wk group. CONCLUSIONS: In early prevascularization before subcutaneous transplantation, neutrophil and macrophage accumulation prevented early engraftment owing to inflammatory cytokine production.


Sujet(s)
Glycémie , Cytokines , Survie du greffon , Transplantation d'ilots de Langerhans , Souris de lignée C57BL , Transplantation d'ilots de Langerhans/méthodes , Transplantation d'ilots de Langerhans/immunologie , Animaux , Glycémie/métabolisme , Cytokines/métabolisme , Souris , Mâle , Facteurs temps , Diabète expérimental/immunologie , Diabète expérimental/chirurgie , Tissu sous-cutané/vascularisation , Tissu sous-cutané/immunologie , Matrice extracellulaire/métabolisme , Ilots pancréatiques/immunologie , Ilots pancréatiques/vascularisation , Néovascularisation physiologique
10.
Microvasc Res ; 151: 104617, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37918522

RÉSUMÉ

Type 1 diabetes mellitus (T1DM) is predominantly managed using insulin replacement therapy, however, pancreatic microcirculatory disturbances play a critical role in T1DM pathogenesis, necessitating alternative therapies. This study aimed to investigate the protective effects of glycine supplementation on pancreatic microcirculation in T1DM. Streptozotocin-induced T1DM and glycine-supplemented mice (n = 6 per group) were used alongside control mice. Pancreatic microcirculatory profiles were determined using a laser Doppler blood perfusion monitoring system and wavelet transform spectral analysis. The T1DM group exhibited disorganized pancreatic microcirculatory oscillation. Glycine supplementation significantly restored regular biorhythmic contraction and relaxation, improving blood distribution patterns. Further-more, glycine reversed the lower amplitudes of endothelial oscillators in T1DM mice. Ultrastructural deterioration of islet microvascular endothelial cells (IMECs) and islet microvascular pericytes, including membrane and organelle damage, collagenous fiber proliferation, and reduced edema, was substantially reversed by glycine supplementation. Additionally, glycine supplementation inhibited the production of IL-6, TNF-α, IFN-γ, pro-MMP-9, and VEGF-A in T1DM, with no significant changes in energetic metabolism observed in glycine-supplemented IMECs. A statistically significant decrease in MDA levels accompanied by an increase in SOD levels was also observed with glycine supplementation. Notably, negative correlations emerged between inflammatory cytokines and microhemodynamic profiles. These findings suggest that glycine supplementation may offer a promising therapeutic approach for protecting against pancreatic microcirculatory dysfunction in T1DM.


Sujet(s)
Diabète de type 1 , Ilots pancréatiques , Souris , Animaux , Microcirculation , Cellules endothéliales , Ilots pancréatiques/vascularisation , Ilots pancréatiques/métabolisme , Compléments alimentaires
11.
Cell Rep ; 42(8): 112913, 2023 08 29.
Article de Anglais | MEDLINE | ID: mdl-37531253

RÉSUMÉ

Pancreatic islets are endocrine organs that depend on their microvasculature to function. Along with endothelial cells, pericytes comprise the islet microvascular network. These mural cells are crucial for microvascular stability and function, but it is not known if/how they are affected during the development of type 1 diabetes (T1D). Here, we investigate islet pericyte density, phenotype, and function using living pancreas slices from donors without diabetes, donors with a single T1D-associated autoantibody (GADA+), and recent onset T1D cases. Our data show that islet pericyte and capillary responses to vasoactive stimuli are impaired early on in T1D. Microvascular dysfunction is associated with a switch in the phenotype of islet pericytes toward myofibroblasts. Using publicly available RNA sequencing (RNA-seq) data, we further found that transcriptional alterations related to endothelin-1 signaling and vascular and extracellular matrix (ECM) remodeling are hallmarks of single autoantibody (Aab)+ donor pancreata. Our data show that microvascular dysfunction is present at early stages of islet autoimmunity.


Sujet(s)
Diabète de type 1 , Ilots pancréatiques , Humains , Diabète de type 1/anatomopathologie , Péricytes/anatomopathologie , Cellules endothéliales/anatomopathologie , Ilots pancréatiques/vascularisation , Autoanticorps
12.
Diabetes ; 72(6): 758-768, 2023 06 01.
Article de Anglais | MEDLINE | ID: mdl-36929171

RÉSUMÉ

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.


Sujet(s)
Diabète de type 1 , Transplantation d'ilots de Langerhans , Ilots pancréatiques , Suidae , Humains , Animaux , Souris , Transplantation d'ilots de Langerhans/méthodes , Survie du greffon , Ilots pancréatiques/vascularisation , Diabète de type 1/chirurgie , Collagène
13.
Adv Biol Regul ; 87: 100919, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36266190

RÉSUMÉ

Pancreatic islets are micro-organs composed of a mixture of endocrine and non-endocrine cells, where the former secrete hormones and peptides necessary for metabolic homeostasis. Through vasculature and innervation the cells within the islets are in communication with the rest of the body, while they interact with each other through juxtacrine, paracrine and autocrine signals, resulting in fine-tuned sensing and response to stimuli. In this context, cellular protrusion in islet cells, such as primary cilia and filopodia, have gained attention as potential signaling hubs. During the last decade, several pieces of evidence have shown how the primary cilium is required for islet vascularization, function and homeostasis. These findings have been possible thanks to the development of ciliary/basal body specific knockout models and technological advances in microscopy, which allow longitudinal monitoring of engrafted islets transplanted in the anterior chamber of the eye in living animals. Using this technique in combination with optogenetics, new potential paracrine interactions have been suggested. For example, reshaping and active movement of filopodia-like protrusions of δ-cells were visualized in vivo, suggesting a continuous cell remodeling to increase intercellular contacts. In this review, we discuss these recent discoveries regarding primary cilia and filopodia and their role in islet homeostasis and intercellular islet communication.


Sujet(s)
Ilots pancréatiques , Pseudopodes , Animaux , Cils vibratiles , Ilots pancréatiques/vascularisation , Ilots pancréatiques/métabolisme , Communication cellulaire , Transduction du signal
14.
Front Endocrinol (Lausanne) ; 13: 992540, 2022.
Article de Anglais | MEDLINE | ID: mdl-36277698

RÉSUMÉ

Human islet transplantations into rodent models are an essential tool to aid in the development and testing of islet and cellular-based therapies for diabetes prevention and treatment. Through the ability to evaluate human islets in an in vivo setting, these studies allow for experimental approaches to answer questions surrounding normal and disease pathophysiology that cannot be answered using other in vitro and in vivo techniques alone. Intravital microscopy enables imaging of tissues in living organisms with dynamic temporal resolution and can be employed to measure biological processes in transplanted human islets revealing how experimental variables can influence engraftment, and transplant survival and function. A key consideration in experimental design for transplant imaging is the surgical placement site, which is guided by the presence of vasculature to aid in functional engraftment of the islets and promote their survival. Here, we review transplantation sites and mouse models used to study beta cell biology in vivo using intravital microscopy and we highlight fundamental observations made possible using this methodology.


Sujet(s)
Cellules à insuline , Transplantation d'ilots de Langerhans , Ilots pancréatiques , Souris , Animaux , Humains , Transplantation d'ilots de Langerhans/méthodes , Ilots pancréatiques/imagerie diagnostique , Ilots pancréatiques/vascularisation , Survie du greffon , Microscopie intravitale , Modèles animaux de maladie humaine
15.
Transpl Int ; 35: 10555, 2022.
Article de Anglais | MEDLINE | ID: mdl-36090775

RÉSUMÉ

Intrahepatic islet transplantation is a promising ß-cell replacement strategy for the treatment of type 1 diabetes. Instant blood-mediated inflammatory reactions, acute inflammatory storm, and graft revascularization delay limit islet engraftment in the peri-transplant phase, hampering the success rate of the procedure. Growing evidence has demonstrated that islet engraftment efficiency may take advantage of several bioengineering approaches aimed to recreate both vascular and endocrine compartments either ex vivo or in vivo. To this end, endocrine pancreas bioengineering is an emerging field in ß-cell replacement, which might provide endocrine cells with all the building blocks (vascularization, ECM composition, or micro/macro-architecture) useful for their successful engraftment and function in vivo. Studies on reshaping either the endocrine cellular composition or the islet microenvironment have been largely performed, focusing on a single building block element, without, however, grasping that their synergistic effect is indispensable for correct endocrine function. Herein, the review focuses on the minimum building blocks that an ideal vascularized endocrine scaffold should have to resemble the endocrine niche architecture, composition, and function to foster functional connections between the vascular and endocrine compartments. Additionally, this review highlights the possibility of designing bioengineered scaffolds integrating alternative endocrine sources to overcome donor organ shortages and the possibility of combining novel immune-preserving strategies for long-term graft function.


Sujet(s)
Insulines , Transplantation d'ilots de Langerhans , Ilots pancréatiques , Bioingénierie , Matrice extracellulaire , Humains , Ilots pancréatiques/vascularisation , Transplantation d'ilots de Langerhans/méthodes
17.
Diabetes ; 71(8): 1679-1693, 2022 08 01.
Article de Anglais | MEDLINE | ID: mdl-35587617

RÉSUMÉ

The pancreatic islet depends on blood supply to efficiently sense plasma glucose levels and deliver insulin and glucagon into the circulation. Long believed to be passive conduits of nutrients and hormones, islet capillaries were recently found to be densely covered with contractile pericytes with the capacity to locally control blood flow. Here, we determined the contribution of pericyte regulation of islet blood flow to plasma insulin and glucagon levels and glycemia. Selective optogenetic activation of pericytes in intraocular islet grafts contracted capillaries and diminished blood flow. In awake mice, acute light-induced stimulation of islet pericytes decreased insulin and increased glucagon plasma levels, producing hyperglycemic effects. Interestingly, pericytes are the targets of sympathetic nerves in the islet, suggesting that sympathetic control of hormone secretion may occur in part by modulating pericyte activity and blood flow. Indeed, in vivo activation of pericytes with the sympathetic agonist phenylephrine decreased blood flow in mouse islet grafts, lowered plasma insulin levels, and increased glycemia. We further show that islet pericytes and blood vessels in living human pancreas slices responded to sympathetic input. Our findings indicate that pericytes mediate vascular responses in the islet that are required for adequate hormone secretion and glucose homeostasis. Vascular and neuronal alterations that are commonly seen in the islets of people with diabetes may impair regulation of islet blood flow and thus precipitate islet dysfunction.


Sujet(s)
Glucagon , Ilots pancréatiques , Animaux , Glycémie , Glucose/pharmacologie , Homéostasie , Humains , Insuline , Ilots pancréatiques/vascularisation , Souris , Péricytes
18.
Wiad Lek ; 75(1 pt 2): 187-190, 2022.
Article de Anglais | MEDLINE | ID: mdl-35182120

RÉSUMÉ

OBJECTIVE: The aim: The purpose of the work was to study the peculiarities of blood supply the pancreatic islet of the 24-month-old rat, and its restructuring during the initial periods of experimental diabetes mellitus. PATIENTS AND METHODS: Materials and methods: The work was performed on 20 white outbred rats - males weighing 340-420g. 24 months of age, kept in standard vivarium conditions in compliance with all accepted ethical rules. Experimental streptozotocin diabetes mellitus was simulated in 16 animals. The material was taken on the 14th and 28th day of the experiment. RESULTS: Results: Reorganization of the endocrine part of the pancreas in the early stages of experimental diabetes is characterized by a decrease in the number and area of pancreatic islets, a decrease in the diameter of the lumen of arterioles, precapillaries, postcapillaries compared to the control group of animals by 7% and 5%. The diameter of the capillaries decreases by 16% and reaches 3.8 ± 0.62 µm2, and the diameter of the venules increases by 12%. In some blood vessels there are phenomena of desolation and edema of perivascular connective tissue, which is manifested by a decrease in optical density and stratification of collagen fibers. CONCLUSION: Conclusions: Thus, the reorganization of the circulatory system of the endocrine part of the pancreas of 24-month-old rats in the early stages of experimental diabetes mellitus is characterized by a decrease in the number and area of pancreatic islets.


Sujet(s)
Diabète expérimental , Ilots pancréatiques , Animaux , Vaisseaux capillaires , Ilots pancréatiques/vascularisation , Mâle , Pancréas , Rats , Streptozocine
19.
Bioengineered ; 13(2): 4385-4396, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35139776

RÉSUMÉ

As one of the most frequently prescribed antidiabetic drugs, metformin can lower glucose levels, improve insulin resistance manage body weight. However, the effect of metformin on islet microcirculation remains unclear. In the present study, to explore the effect of metformin on islet endothelial cells and investigated the underlying mechanism, we assessed the effects of metformin on islet endothelial cell survival, proliferation, oxidative stress and apoptosis. Our results suggest that metformin stimulates the proliferation of pancreatic islet endothelial cells and inhibits the apoptosis and oxidative stress caused by high glucose levels. By activating farnesoid X receptor (FXR), metformin increases the expression of vascular endothelial growth factor-A (VEGF-A) and endothelial nitric oxide synthase (eNOS), improves the production of nitric oxide (NO) and decreases the production of ROS. After the inhibition of FXR or VEGF-A, all of the effects disappeared. Thus, metformin appears to regulate islet microvascular endothelial cell (IMEC) proliferation, apoptosis and oxidative stress by activating the FXR/VEGF-A/eNOS pathway. These findings provide a new mechanism underlying the islet-protective effect of metformin.


Sujet(s)
Glucose/effets indésirables , Ilots pancréatiques , Metformine/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/cytologie , Ilots pancréatiques/vascularisation , Ilots pancréatiques/cytologie , Ilots pancréatiques/effets des médicaments et des substances chimiques , Souris , Microvaisseaux/cytologie , Stress oxydatif/effets des médicaments et des substances chimiques
20.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166339, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35017029

RÉSUMÉ

The pancreatic islet vasculature is of fundamental importance to the ß-cell response to obesity-associated insulin resistance. To explore islet vascular alterations in the pathogenesis of type 2 diabetes, we evaluated two insulin resistance models: ob/ob mice, which sustain large ß-cell mass and hyperinsulinemia, and db/db mice, which progress to diabetes due to secondary ß-cell compensation failure for insulin secretion. Time-dependent changes in islet vasculature and blood flow were investigated using tomato lectin staining and in vivo live imaging. Marked islet capillary dilation was observed in ob/ob mice, but this adaptive change was blunted in db/db mice. Islet blood flow volume was augmented in ob/ob mice, whereas it was reduced in db/db mice. The protein concentrations of total and phosphorylated endothelial nitric oxide synthase (eNOS) at Ser1177 were increased in ob/ob islets, while they were diminished in db/db mice, indicating decreased eNOS activity. This was accompanied by an increased retention of advanced glycation end-products in db/db blood vessels. Amelioration of diabetes by Elovl6 deficiency involved a restoration of capillary dilation, blood flow, and eNOS phosphorylation in db/db islets. Our findings suggest that the disability of islet capillary dilation due to endothelial dysfunction impairs local islet blood flow, which may play a role in the loss of ß-cell function and further exacerbate type 2 diabetes.


Sujet(s)
Vaisseaux sanguins/métabolisme , Ilots pancréatiques/physiologie , Animaux , Vitesse du flux sanguin , Diabète de type 2/anatomopathologie , Modèles animaux de maladie humaine , Fatty acid elongases/déficit , Fatty acid elongases/génétique , Femelle , Produits terminaux de glycation avancée/métabolisme , Insuline/métabolisme , Insulinorésistance , Ilots pancréatiques/anatomie et histologie , Ilots pancréatiques/vascularisation , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Souris obèse , Nitric oxide synthase type III/métabolisme , Phosphorylation
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...