Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Radiother Oncol ; 170: 118-121, 2022 05.
Article in English | MEDLINE | ID: mdl-35257850

ABSTRACT

Pre-operative 5-fraction breast radiotherapy followed by immediate breast-sparing surgery and sentinel node procedure was feasible in 14 patients with 15 clinical early-stage breast cancers. However wound problems occurred frequently and was documented in 5 of the 14 patients: 2 patients with a mastitis needing antibiotics, 2 patients developed a fistula with exudate needing antibiotics and local disinfection and 1 patient developed a fistula needing surgical reintervention. Other acute and late iatrogenic events were rather limited. Two patients had a pathological lymph node involvement, which underlines the importance to perform the sentinel node procedure before pre-operative radiotherapy.


Subject(s)
Breast Neoplasms , Sentinel Lymph Node , Anti-Bacterial Agents , Axilla/pathology , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Breast Neoplasms/surgery , Feasibility Studies , Female , Humans , Lymph Node Excision , Lymph Nodes/pathology , Sentinel Lymph Node/pathology , Sentinel Lymph Node Biopsy/methods
4.
Nat Commun ; 9(1): 5083, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504844

ABSTRACT

Whereas genomic aberrations in the SLIT-ROBO pathway are frequent in pancreatic ductal adenocarcinoma (PDAC), their function in the pancreas is unclear. Here we report that in pancreatitis and PDAC mouse models, epithelial Robo2 expression is lost while Robo1 expression becomes most prominent in the stroma. Cell cultures of mice with loss of epithelial Robo2 (Pdx1Cre;Robo2F/F) show increased activation of Robo1+ myofibroblasts and induction of TGF-ß and Wnt pathways. During pancreatitis, Pdx1Cre;Robo2F/F mice present enhanced myofibroblast activation, collagen crosslinking, T-cell infiltration and tumorigenic immune markers. The TGF-ß inhibitor galunisertib suppresses these effects. In PDAC patients, ROBO2 expression is overall low while ROBO1 is variably expressed in epithelium and high in stroma. ROBO2low;ROBO1high patients present the poorest survival. In conclusion, Robo2 acts non-autonomously as a stroma suppressor gene by restraining myofibroblast activation and T-cell infiltration. ROBO1/2 expression in PDAC patients may guide therapy with TGF-ß inhibitors or other stroma /immune modulating agents.


Subject(s)
Pancreas/metabolism , Pancreas/pathology , Receptors, Immunologic/metabolism , Transforming Growth Factor beta/metabolism , Animals , Blotting, Western , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cells, Cultured , Female , Flow Cytometry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , In Vitro Techniques , Male , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreatitis/genetics , Pancreatitis/metabolism , Receptors, Immunologic/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Trans-Activators/genetics , Trans-Activators/metabolism , Roundabout Proteins
5.
Diabetologia ; 61(8): 1804-1810, 2018 08.
Article in English | MEDLINE | ID: mdl-29789879

ABSTRACT

AIMS/HYPOTHESIS: The initial avascular period following islet transplantation seriously compromises graft function and survival. Enhancing graft revascularisation to improve engraftment has been attempted through virus-based delivery of angiogenic triggers, but risks associated with viral vectors have hampered clinical translation. In vitro transcribed mRNA transfection circumvents these risks and may be used for improving islet engraftment. METHODS: Mouse and human pancreatic islet cells were transfected with mRNA encoding the angiogenic growth factor vascular endothelial growth factor A (VEGF-A) before transplantation under the kidney capsule in mice. RESULTS: At day 7 post transplantation, revascularisation of grafts transfected with Vegf-A (also known as Vegfa) mRNA was significantly higher compared with non-transfected or Gfp mRNA-transfected controls in mouse islet grafts (2.11- and 1.87-fold, respectively) (vessel area/graft area, mean ± SEM: 0.118 ± 0.01 [n = 3] in Vegf-A mRNA transfected group (VEGF) vs 0.056 ± 0.01 [n = 3] in no RNA [p < 0.05] vs 0.063 ± 0.02 [n = 4] in Gfp mRNA transfected group (GFP) [p < 0.05]); EndoC-bH3 grafts (2.85- and 2.48-fold. respectively) (0.085 ± 0.02 [n = 4] in VEGF vs 0.030 ± 0.004 [n = 4] in no RNA [p < 0.05] vs 0.034 ± 0.01 [n = 5] in GFP [p < 0.05]); and human islet grafts (3.17- and 3.80-fold, respectively) (0.048 ± 0.013 [n = 3] in VEGF vs 0.015 ± 0.0051 [n = 4] in no RNA [p < 0.01] vs 0.013 ± 0.0046 [n = 4] in GFP [p < 0.01]). At day 30 post transplantation, human islet grafts maintained a vascularisation benefit (1.70- and 1.82-fold, respectively) (0.049 ± 0.0042 [n = 8] in VEGF vs 0.029 ± 0.0052 [n = 5] in no RNA [p < 0.05] vs 0.027 ± 0.0056 [n = 4] in GFP [p < 0.05]) and a higher beta cell volume (1.64- and 2.26-fold, respectively) (0.0292 ± 0.0032 µl [n = 7] in VEGF vs 0.0178 ± 0.0021 µl [n = 5] in no RNA [p < 0.01] vs 0.0129 ± 0.0012 µl [n = 4] in GFP [p < 0.001]). CONCLUSIONS/INTERPRETATION: Vegf-A mRNA transfection before transplantation provides a promising and safe strategy to improve engraftment of islets and other cell-based implants.


Subject(s)
Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Neovascularization, Physiologic , RNA, Messenger/genetics , Transfection , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Survival , Humans , Insulin/metabolism , Insulin-Secreting Cells/transplantation , Islets of Langerhans Transplantation , Mice
6.
Physiol Rev ; 98(3): 1143-1167, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29717931

ABSTRACT

Diabetes mellitus results from disturbed glucose homeostasis due to an absolute (type 1) or relative (type 2) deficiency of insulin, a peptide hormone almost exclusively produced by the beta cells of the endocrine pancreas in a tightly regulated manner. Current therapy only delays disease progression through insulin injection and/or oral medications that increase insulin secretion or sensitivity, decrease hepatic glucose production, or promote glucosuria. These drugs have turned diabetes into a chronic disease as they do not solve the underlying beta cell defects or entirely prevent the long-term complications of hyperglycemia. Beta cell replacement through islet transplantation is a more physiological therapeutic alternative but is severely hampered by donor shortage and immune rejection. A curative strategy should combine newer approaches to immunomodulation with beta cell replacement. Success of this approach depends on the development of practical methods for generating beta cells, either in vitro or in situ through beta cell replication or beta cell differentiation. This review provides an overview of human beta cell generation.


Subject(s)
Cell Culture Techniques , Insulin-Secreting Cells/physiology , Regeneration , Animals , Homeostasis , Humans , Insulin-Secreting Cells/transplantation
7.
PLoS One ; 13(2): e0191249, 2018.
Article in English | MEDLINE | ID: mdl-29408875

ABSTRACT

Pancreas injury by partial duct ligation (PDL) activates beta cell differentiation and proliferation in adult mouse pancreas but remains controversial regarding the anticipated increase in beta cell volume. Several reports unable to show beta cell volume augmentation in PDL pancreas used automated digital image analysis software. We hypothesized that fully automatic beta cell morphometry without manual micrograph artifact remediation introduces bias and therefore might be responsible for reported discrepancies and controversy. However, our present results prove that standard digital image processing with automatic thresholding is sufficiently robust albeit less sensitive and less adequate to demonstrate a significant increase in beta cell volume in PDL versus Sham-operated pancreas. We therefore conclude that other confounding factors such as quality of surgery, selection of samples based on relative abundance of the transcription factor Neurogenin 3 (Ngn3) and tissue processing give rise to inter-laboratory inconsistencies in beta cell volume quantification in PDL pancreas.


Subject(s)
Automation , Islets of Langerhans/pathology , Animals
8.
Diabetologia ; 61(1): 168-181, 2018 01.
Article in English | MEDLINE | ID: mdl-28852792

ABSTRACT

AIMS/HYPOTHESIS: Identification of a pancreatic neuro-insular network in mice suggests that a similar integration of islets and nerves may be present in the human pancreas. To characterise the neuro-insular network and the intra-pancreatic ganglia in a clinically related setting, we examined human pancreases in health and with fatty infiltration via 3-dimensional (3D) histology and compared the human pancreatic microenvironment with its counterpart in mice. METHODS: Human pancreatic specimens from individuals with normal BMI, high BMI (≥ 25) and type 2 diabetes were used to investigate the neuro-insular network. Transparent specimens were prepared by tissue clearing for transmitted light and deep-tissue fluorescence imaging to simultaneously visualise infiltrated adipocytes, islets and neurovascular networks. RESULTS: High-definition images of human islets reveal that both the sympathetic and parasympathetic nerves enter the islet core and reside in the immediate microenvironment of islet cells. Around the islets, the neuro-insular network is visualised with 3D histology to identify the intra-pancreatic ganglia (peri-lobular and intra-parenchymal ganglia) and the islet-ganglionic association. In humans, but not in mice, pancreatic fatty infiltration (BMI dependent) features adipocytes infiltrating into the parenchyma and accumulating in the peri-lobular space, in which the peri-lobular ganglia also reside. We identified the formation of adipose-ganglionic complexes in the peri-lobular space and enlargement of ganglia around adipocytes. In the specimen from the individual with type 2 diabetes, an increase in the number of nerve projections from the intra-parenchymal ganglia is associated with severe fatty infiltration. CONCLUSIONS/INTERPRETATION: We present new perspectives of human pancreas and islet innervation via 3D histology. Our results strongly suggest that fatty infiltration in the human pancreas creates a neurotrophic microenvironment and promotes remodelling of pancreatic innervation.


Subject(s)
Pancreas/metabolism , Adipocytes/metabolism , Animals , Body Mass Index , Diabetes Mellitus, Type 2/metabolism , Humans , Islets of Langerhans/metabolism , Mice , Obesity/metabolism , Sympathetic Nervous System/metabolism
9.
Diabetes ; 66(6): 1703-1712, 2017 06.
Article in English | MEDLINE | ID: mdl-28246293

ABSTRACT

Insulin production by the pancreatic ß-cell is required for normal glucose homeostasis. While key transcription factors that bind to the insulin promoter are known, relatively little is known about the upstream regulators of insulin transcription. Using a whole-genome RNA interference screen, we uncovered 26 novel regulators of insulin transcription that regulate diverse processes including oxidative phosphorylation, vesicle traffic, and the unfolded protein response (UPR). We focused on Spry2-a gene implicated in human type 2 diabetes by genome-wide association studies but without a clear connection to glucose homeostasis. We showed that Spry2 is a novel UPR target and its upregulation is dependent on PERK. Knockdown of Spry2 resulted in reduced expression of Serca2, reduced endoplasmic reticulum calcium levels, and induction of the UPR. Spry2 deletion in the adult mouse ß-cell caused hyperglycemia and hypoinsulinemia. Our study greatly expands the compendium of insulin promoter regulators and demonstrates a novel ß-cell link between Spry2 and human diabetes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation/genetics , Insulin-Secreting Cells/metabolism , Insulin/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Unfolded Protein Response/genetics , Animals , Annexin A5/metabolism , Blotting, Western , Calcium/metabolism , Cell Line , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum/metabolism , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin/metabolism , Mice , Protein Serine-Threonine Kinases , RNA Interference , Real-Time Polymerase Chain Reaction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , eIF-2 Kinase/metabolism
10.
Diabetologia ; 59(9): 1948-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27318836

ABSTRACT

AIMS/HYPOTHESIS: When the beta cell mass or function declines beyond a critical point, hyperglycaemia arises. Little is known about the potential pathways involved in beta cell rescue. As two cytokines, epidermal growth factor (EGF) and ciliary neurotrophic factor (CNTF), restored a functional beta cell mass in mice with long-term hyperglycaemia by reprogramming acinar cells that transiently expressed neurogenin 3 (NGN3), the current study assesses the effect of these cytokines on the functional beta cell mass after an acute chemical toxic insult. METHODS: Glycaemia and insulin levels, pro-endocrine gene expression and beta cell origin, as well as the role of signal transducer and activator of transcription 3 (STAT3) signalling, were assessed in EGF+CNTF-treated mice following acute hyperglycaemia. RESULTS: The mice were hyperglycaemic 1 day following i.v. injection of the beta cell toxin alloxan, when the two cytokines were applied. One week later, 68.6 ± 4.6% of the mice had responded to the cytokine treatment and increased their insulin(+) cell number to 30% that of normoglycaemic control mice, resulting in restoration of euglycaemia. Although insulin(-) NGN3(+) cells appeared following acute EGF+CNTF treatment, genetic lineage tracing showed that the majority of the insulin(+) cells originated from pre-existing beta cells. Beta cell rescue by EGF+CNTF depends on glycaemia rather than on STAT3-induced NGN3 expression in acinar cells. CONCLUSIONS/INTERPRETATION: In adult mice, EGF+CNTF allows the rescue of beta cells in distress when treatment is given shortly after the diabetogenic insult. The rescued beta cells restore a functional beta cell mass able to control normal blood glucose levels. These findings may provide new insights into compensatory pathways activated early after beta cell loss.


Subject(s)
Ciliary Neurotrophic Factor/therapeutic use , Epidermal Growth Factor/therapeutic use , Hyperglycemia/drug therapy , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Alloxan/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Glucose/drug effects , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Signal Transduction/drug effects
11.
Diabetologia ; 59(9): 1834-7, 2016 09.
Article in English | MEDLINE | ID: mdl-27053238

ABSTRACT

The generation of beta(-like) cells to compensate for their absolute or relative shortage in type 1 and type 2 diabetes is an obvious therapeutic strategy. Patients first received grafts of donor islet cells over 25 years ago, but this procedure has not become routine in clinical practice because of a donor cell shortage and (auto)immune problems. Transplantation of differentiated embryonic and induced pluripotent stem cells may overcome some but not all the current limitations. Reprogramming exocrine cells towards functional beta(-like) cells would offer an alternative abundant and autologous source of beta(-like) cells. This review focuses on work by our research group towards achieving such a source of cells. It summarises a presentation given at the 'Can we make a better beta cell?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Amin Ardestani and Kathrin Maedler, DOI: 10.1007/s00125-016-3892-9 , and by Heiko Lickert and colleagues, DOI: 10.1007/s00125-016-3949-9 ) and a commentary by the Session Chair, Shanta Persaud (DOI: 10.1007/s00125-016-3870-2 ).


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pancreas/cytology , Animals , Cell Differentiation/physiology , Humans , Macrophages/metabolism , Transcription Factors/metabolism
12.
J Vis Exp ; (102): e52765, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26273954

ABSTRACT

Expansion of pancreatic beta cells in vivo or ex vivo, or generation of beta cells by differentiation from an embryonic or adult stem cell, can provide new expandable sources of beta cells to alleviate the donor scarcity in human islet transplantation as therapy for diabetes. Although recent advances have been made towards this aim, mechanisms that regulate beta cell expansion and differentiation from a stem/progenitor cell remain to be characterized. Here, we describe a protocol for an injury model in the adult mouse pancreas that can function as a tool to study mechanisms of tissue remodeling and beta cell proliferation and differentiation. Partial duct ligation (PDL) is an experimentally induced injury of the rodent pancreas involving surgical ligation of the main pancreatic duct resulting in an obstruction of drainage of exocrine products out of the tail region of the pancreas. The inflicted damage induces acinar atrophy, immune cell infiltration and severe tissue remodeling. We have previously reported the activation of Neurogenin (Ngn) 3 expressing endogenous progenitor-like cells and an increase in beta cell proliferation after PDL. Therefore, PDL provides a basis to study signals involved in beta cell dynamics and the properties of an endocrine progenitor in adult pancreas. Since, it still remains largely unclear, which factors and pathways contribute to beta cell neogenesis and proliferation in PDL, a standardized protocol for PDL will allow for comparison across laboratories.


Subject(s)
Cellular Reprogramming/physiology , Insulin-Secreting Cells/cytology , Pancreas/injuries , Pancreatic Ducts/surgery , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Humans , Intraoperative Complications/pathology , Ligation/methods , Male , Mice , Mice, Inbred BALB C , Pancreas/cytology
14.
EJNMMI Res ; 4: 32, 2014.
Article in English | MEDLINE | ID: mdl-25024930

ABSTRACT

BACKGROUND: To combine the sensitivity of bioluminescent imaging (BLI) with the 3D and quantitative properties of pinhole single-photon emission computed tomography (SPECT)/micro-computed tomography (CT) (phSPECT/micro-CT), we generated stable cell lines that express a yellow-fluorescent protein (YFP) and Gaussia luciferase (GLuc) fusion protein (YFP/GLuc). For in vivo phSPECT detection of this YFP/GLuc protein, a nanobody, targeted against yellow and green fluorescent proteins (anti-YFP-Nb), was site specifically labelled with (99m)Tc. METHODS: Human embryonic kidney cells (HEK293T) were cultured and passaged every 3 days. 10E5 cells were transduced with YFP/GLuc-containing vector: both membrane-targeted (MT-YFP/GLuc) and non-targeted (YFP/GLuc) fusion proteins were developed. These vectors were compared against a SKOV-3 cell line stably expressing green fluorescent-firefly luciferase (GFP/Fluc) and HEK293T cells expressing red fluorescent protein in combination with a Gaussia luciferase (Red/GLuc). Transduction efficiencies were scored by fluorescence microscopy, and transduced cells were enriched by fluorescence-activated cell sorting (FACS). GLuc and FLuc functionality was tested in vitro by list-mode BLI. Subsequently, cells were transplanted subcutaneously in athymic (nu/nu) mice (MT-YFP/GLuc: n = 4, YFP/GLuc: n = 6, GFP/FLuc: n = 6, Red/GLuc: n = 4). Labelling efficiency of anti-YFP-Nb was measured using instant thin layer chromatography. One week after transplantation, (99m)Tc-labelled anti-YFP-Nb was injected intravenously and pinhole (ph) SPECT/micro-CT was performed, followed by in vivo BLI. RESULTS: Cells showed high levels of fluorescence after transduction. The cells containing the MT-YFP/GLuc were positive on fluorescence microscopy, with the fluorescent signal confined to the cell membrane. After cell sorting, transduced cells were assayed by BLI and showed a significantly higher light output both in vitro and in vivo compared with non-transduced HEK293T cells. The anti-YFP-Nb labelling efficiency was 98%, and subsequent phSPECT/micro-CT demonstrated visible cell binding and significantly higher transplant-to-muscle ratio for both the MT-YFP/GLuc and YFP/GLuc transplanted cells, compared with the GFP/FLuc and Red/GLuc group. CONCLUSION: This study provides a proof of principle for a nanobody-based cell tracking method, using a YFP/GLuc fusion protein and anti-YFP-Nb in a model of subcutaneously transplanted transduced HEK293T cells.

15.
Diabetologia ; 57(7): 1420-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24759958

ABSTRACT

AIMS/HYPOTHESIS: IL-6 was recently shown to control alpha cell expansion. As beta cells expand following partial pancreatic-duct ligation (PDL) in adult mice, we investigated whether PDL also causes alpha cells to expand and whether IL-6 signalling is involved. As alpha cells can reprogramme to beta cells in a number of beta cell (re)generation models, we examined whether this phenomenon also exists in PDL pancreas. METHODS: Total alpha cell volume, alpha cell size and total glucagon content were evaluated in equivalent portions of PDL- and sham-operated mouse pancreases. Proliferation of glucagon(+) cells was assessed by expression of the proliferation marker Ki67. Inter-conversions between alpha and beta cells were monitored in transgenic mice with conditional cell-type-specific labelling. The role of IL-6 in regulating alpha cell proliferation was evaluated by in situ delivery of an IL-6-inactivating antibody. RESULTS: In response to PDL surgery, alpha cell volume in the ligated tissue was increased threefold, glucagon content fivefold and alpha cell size by 10%. Activation of alpha cell proliferation in PDL pancreas required IL-6 signalling. A minor fraction of alpha cells derived from beta cells, whereas no evidence for alpha to beta cell conversion was obtained. CONCLUSIONS/INTERPRETATION: In PDL-injured adult mouse pancreas, new alpha cells are generated mainly by IL-6-dependent self-duplication and seldom by reprogramming of beta cells.


Subject(s)
Cell Proliferation/physiology , Glucagon-Secreting Cells/cytology , Interleukin-6/metabolism , Pancreatic Ducts/cytology , Animals , Cell Size , Glucagon-Secreting Cells/metabolism , Ligation , Mice , Pancreatic Ducts/metabolism
16.
Nat Biotechnol ; 32(1): 76-83, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24240391

ABSTRACT

Reprogramming of pancreatic exocrine cells into cells resembling beta cells may provide a strategy for treating diabetes. Here we show that transient administration of epidermal growth factor and ciliary neurotrophic factor to adult mice with chronic hyperglycemia efficiently stimulates the conversion of terminally differentiated acinar cells to beta-like cells. Newly generated beta-like cells are epigenetically reprogrammed, functional and glucose responsive, and they reinstate normal glycemic control for up to 248 d. The regenerative process depends on Stat3 signaling and requires a threshold number of Neurogenin 3 (Ngn3)-expressing acinar cells. In contrast to previous work demonstrating in vivo conversion of acinar cells to beta-like cells by viral delivery of exogenous transcription factors, our approach achieves acinar-to-beta-cell reprogramming through transient cytokine exposure rather than genetic modification.


Subject(s)
Ciliary Neurotrophic Factor/administration & dosage , Diabetes Mellitus/drug therapy , Epidermal Growth Factor/administration & dosage , Insulin-Secreting Cells/drug effects , Acinar Cells/drug effects , Acinar Cells/pathology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Ciliary Neurotrophic Factor/genetics , Diabetes Mellitus/genetics , Epidermal Growth Factor/genetics , Hyperglycemia/drug therapy , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred NOD/genetics , Signal Transduction
17.
Diabetes ; 62(12): 4165-73, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23974922

ABSTRACT

It is generally accepted that vascularization and oxygenation of pancreatic islets are essential for the maintenance of an optimal ß-cell mass and function and that signaling by vascular endothelial growth factor (VEGF) is crucial for pancreas development, insulin gene expression/secretion, and (compensatory) ß-cell proliferation. A novel mouse model was designed to allow conditional production of human sFlt1 by ß-cells in order to trap VEGF and study the effect of time-dependent inhibition of VEGF signaling on adult ß-cell fate and metabolism. Secretion of sFlt1 by adult ß-cells resulted in a rapid regression of blood vessels and hypoxia within the islets. Besides blunted insulin release, ß-cells displayed a remarkable capacity for coping with these presumed unfavorable conditions: even after prolonged periods of blood vessel ablation, basal and stimulated blood glucose levels were only slightly increased, while ß-cell proliferation and mass remained unaffected. Moreover, ablation of blood vessels did not prevent ß-cell generation after severe pancreas injury by partial pancreatic duct ligation or partial pancreatectomy. Our data thus argue against a major role of blood vessels to preserve adult ß-cell generation and function, restricting their importance to facilitating rapid and adequate insulin delivery.


Subject(s)
Hypoxia/physiopathology , Insulin-Secreting Cells/physiology , Ischemia/physiopathology , Islets of Langerhans/blood supply , Neovascularization, Pathologic/physiopathology , Animals , Hypoxia/metabolism , Insulin/metabolism , Ischemia/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/physiopathology , Mice , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
18.
BMC Biotechnol ; 12: 74, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23088534

ABSTRACT

BACKGROUND: Effective gene transfer to the pancreas or to pancreatic cells has remained elusive although it is essential for studies of genetic lineage tracing and modulation of gene expression. Different transduction methods and viral vectors were tested in vitro and in vivo, in rat and mouse pancreas. RESULTS: For in vitro transfection/transduction of rat exocrine cells lipofection reagents, adenoviral vectors, and Mokola- and VSV-G pseudotyped lentiviral vectors were used. For in vivo transduction of mouse and rat pancreas adenoviral vectors and VSV-G lentiviral vectors were injected into the parenchymal tissue. Both lipofection of rat exocrine cell cultures and transduction with Mokola pseudotyped lentiviral vectors were inefficient and resulted in less than 4% EGFP expressing cells. Adenoviral transduction was highly efficient but its usefulness for gene delivery to rat exocrine cells in vitro was hampered by a drastic increase in cell death. In vitro transduction of rat exocrine cells was most optimal with VSV-G pseudotyped lentiviral vectors, with stable transgene expression, no significant effect on cell survival and about 40% transduced cells. In vivo, pancreatic cells could not be transduced by intra-parenchymal administration of lentiviral vectors in mouse and rat pancreas. However, a high efficiency could be obtained by adenoviral vectors, resulting in transient transduction of mainly exocrine acinar cells. Injection in immune-deficient animals diminished leukocyte infiltration and prolonged transgene expression. CONCLUSIONS: In summary, our study remarkably demonstrates that transduction of pancreatic exocrine cells requires lentiviral vectors in vitro but adenoviral vectors in vivo.


Subject(s)
Acinar Cells/virology , Gene Transfer Techniques , Pancreas/cytology , Acinar Cells/cytology , Adenoviridae/genetics , Adenoviridae/physiology , Animals , Cells, Cultured , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/physiology , Humans , Lentivirus/genetics , Lentivirus/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreas/virology , Rats , Rats, Wistar , Transduction, Genetic , Transgenes
19.
Cell Metab ; 16(4): 449-61, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23000401

ABSTRACT

Reactive oxygen species (ROS) contribute to target-cell damage in inflammatory and iron-overload diseases. Little is known about iron transport regulation during inflammatory attack. Through a combination of in vitro and in vivo studies, we show that the proinflammatory cytokine IL-1ß induces divalent metal transporter 1 (DMT1) expression correlating with increased ß cell iron content and ROS production. Iron chelation and siRNA and genetic knockdown of DMT1 expression reduce cytokine-induced ROS formation and cell death. Glucose-stimulated insulin secretion in the absence of cytokines in Dmt1 knockout islets is defective, highlighting a physiological role of iron and ROS in the regulation of insulin secretion. Dmt1 knockout mice are protected against multiple low-dose streptozotocin and high-fat diet-induced glucose intolerance, models of type 1 and type 2 diabetes, respectively. Thus, ß cells become prone to ROS-mediated inflammatory damage via aberrant cellular iron metabolism, a finding with potential general cellular implications.


Subject(s)
Apoptosis/drug effects , Cation Transport Proteins/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-1beta/pharmacology , Iron/metabolism , Reactive Oxygen Species/metabolism , Animals , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Diabetes Mellitus, Experimental , Diet, High-Fat , Glucose Intolerance , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Mice , Mice, Knockout , Models, Biological , RNA Interference , RNA, Small Interfering/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
20.
Contrast Media Mol Imaging ; 5(4): 201-7, 2010.
Article in English | MEDLINE | ID: mdl-20665903

ABSTRACT

INTRODUCTION: Micro-CT provides non-invasive anatomic evaluation of small animals. Serial micro-CT measurements are, however, hampered by the severity of ionizing radiation doses cumulating over the total period of follow-up. The dose levels may be sufficient to influence experimental outcomes such as animal survival or tumor growth. AIM: This study was designed to evaluate the radiation dose of micro-CT and to optimize the scanning protocol for longitudinal micro-CT scans. METHODS AND MATERIALS: Normal C57Bl/6 mice were euthanized. Radiation exposure was measured using individually calibrated lithium fluoride thermoluminescent dosimeters (TLDs). Thirteen TLDs were placed in the mice at the thyroid, lungs, liver, stomach, colon, bladder and near the spleen. Micro-CT (SkyScan 1178) was performed using two digital X-ray cameras which scanned over 180 degrees at a resolution of 83 microm, a rotation step of 1.08 degrees , 50 kV, 615 microA and 121 s image acquisition time. The TLDs were removed after each scan. CTDI(100) was measured with a 100 mm ionization chamber, centrally positioned in a 2.7 cm diameter water phantom, and rotation steps were increased to reduce both scan time and radiation dose. RESULTS: Internal TLD analysis demonstrated median organ dose of 5.5 +/- 0.6 mGy per mA s, confirmed by CTDI(100) with result of 6.6 mGy per mA s. A rotation step of 2.16 resulted in qualitatively accurate images. At a resolution of 83 microm the scan time is reduced to 63 s with an estimated dose of 2.9 mGy per mA s. At 166 microm resolution, the scan time is limited to 27 s, with a concordant dose of 1.2 mGy per mA s. CONCLUSIONS: The radiation dose of a standard micro-CT scan is relatively high and could influence the experimental outcome. We believe that the presented adaptation of the scan protocol allows for accurate imaging without the risk of interfering with the experimental outcome of the study.


Subject(s)
Radiographic Image Interpretation, Computer-Assisted/methods , X-Ray Microtomography/methods , Animals , Male , Mice , Mice, Inbred C57BL , Radiation Dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...