Skip to main content
Erschienen in: Diabetologia 11/2016

Open Access 21.07.2016 | Short Communication

Sequential intravital imaging reveals in vivo dynamics of pancreatic tissue transplanted under the kidney capsule in mice

verfasst von: Léon van Gurp, Cindy J. M. Loomans, Pim P. van Krieken, Gitanjali Dharmadhikari, Erik Jansen, Femke C. A. S. Ringnalda, Evelyne Beerling, Jacco van Rheenen, Eelco J. P. de Koning

Erschienen in: Diabetologia | Ausgabe 11/2016

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Aims/hypothesis

Dynamic processes in pancreatic tissue are difficult to study. We aimed to develop an intravital imaging method to longitudinally examine engraftment, vascularisation, expansion and differentiation in mature islets or embryonic pancreases transplanted under the kidney capsule.

Methods

Isolated pancreatic islets from adult mice and murine embryonic day (E)12.5 pancreases containing fluorescent biomarkers were transplanted under the kidney capsule of immunodeficient recipient mice. Human islet cells were dispersed, transduced with a lentivirus expressing a fluorescent label and reaggregated before transplantation. Graft-containing kidneys were positioned subcutaneously and an imaging window was fitted into the skin on top of the kidney. Intravital imaging using multiphoton microscopy was performed for up to 2 weeks. Volumes of fluorescently labelled cells were determined as a measure of development and survival.

Results

Transplanted islets and embryonic pancreases showed good engraftment and remained viable. Engraftment and vascularisation could be longitudinally examined in murine and human islet cells. Murine islet beta cell volume was unchanged over time. Transplanted embryonic pancreases increased to up to 6.1 times of their original volume and beta cell volume increased 90 times during 2 weeks.

Conclusions/interpretation

This method allows for repeated intravital imaging of grafts containing various sources of pancreatic tissue transplanted under the kidney capsule. Using fluorescent markers, dynamic information concerning engraftment or differentiation can be visualised and measured.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00125-016-4049-6) contains peer-reviewed but unedited supplementary material, which is available to authorised users.
Abkürzungen
AIW
Abdominal imaging window
CAG-DsRed
Cytomegalovirus–actin–globin promoter red fluorescent protein
GFP
Green fluorescent protein
HIP-GFP
Human insulin promoter-green fluorescent protein
MIP-EGFP
Mouse insulin promoter-enhanced green fluorescent protein
NSG mice
NOD severe combined immunodeficiency gamma mice

Introduction

Islet cell development, dynamic processes involved in islet engraftment and changes in islet composition are difficult processes to study in vivo. To obtain longitudinal information in combination with functional data, new imaging methods are required that allow sequential measurements in individual animals.
Transplantation of pancreatic islets under the kidney capsule is considered the gold standard for the in vivo evaluation of graft insulin secretory capacity and survival in mice [1]. Isolated islets derived from humans and different animal species can be used for transplantation at this site [2]. After engraftment, there is good vascularisation of the transplanted islets, allowing the rapid release and action of insulin. Nephrectomy of the graft-containing kidney followed by determination of blood glucose values validates graft function [3]. By transplanting a cell pellet that remains compact after transplantation, grafts can easily be retrieved and histologically analysed.
In this study, we adapted our previously published method [4, 5] to perform intravital imaging on islets of Langerhans or embryonic pancreatic tissue transplanted under the kidney capsule using an abdominal imaging window. This method allows sequential measurements of survival, vascularisation, expansion and differentiation of these tissues for a period up to 2 weeks.

Methods

Mouse pancreatic islets and embryonic pancreas isolation
All experiments on animals were carried out in accordance with the guidelines of the Animal Welfare Committee of the Royal Netherlands Academy of Arts and Sciences. Mature pancreatic islets and intact E12.5 embryonic pancreases were isolated from mouse insulin promoter–enhanced green fluorescent protein (MIP-EGFP) (Jackson Laboratory, Bar Harbor, ME, USA, stock no. 006864) and cytomegalovirus–actin–globin promoter red fluorescent protein (CAG-DsRed) (Jackson Laboratory, stock no. 005441) mice. See ESM for further details.
Human pancreatic islets and lentiviral transduction
Human pancreatic islets were isolated at the Leiden University Medical Centre using standard procedures [6]. Isolated human islets could only be used with research consent and when the number and/or quality of the islets were insufficient for clinical islet transplantation, according to national laws. Islets were transduced with a human insulin promoter–GFP (HIP-GFP) virus before transplantation. See ESM for further details.
Islet transplantation and positioning of an intra-abdominal imaging window
NOD severe combined immunodeficiency gamma (NSG) mice (Jackson Laboratory, stock no. 005557) were anaesthetised and pancreatic islets or embryonic pancreases were transplanted under the kidney capsule. The kidney was fixed subcutaneously and an imaging window was placed in the skin (ESM Fig. 1a–f), as published previously [4]. See ESM for further details.
Intravital imaging
Intravital imaging was performed as described previously [5]. If visualisation of the vasculature was required, mice received an intravenous tail vein injection of 2.5 mg 70 kDa dextran-Texas Red conjugate (Thermo Fisher, Waltham, MA, USA, D-1830) in 100 μl PBS directly before imaging. Images were acquired at resolutions between 0.6 × 0.6 × 1.0 and 4.8 × 4.8 × 5.0 μm (xyz). Multiphoton excitation was performed at 960 nm, and the emission was collected at 505–555 nm (EGFP), and 555–695 nm (DsRed, Texas Red).
Histological analysis
Graft-containing kidneys were either fixed for paraffin sectioning or for cryosectioning. Paraffin-embedded sections (4 μm) were cut at and stained with haematoxylin and eosin for the determination of graft size and capsule thickness. Cryosections (10 μm) were used for immunohistochemical staining for insulin and glucagon. See ESM for further details.
Microscopy data processing
See ESM for further details.
Statistical analysis
Data are presented as mean ± SEM. R (www.​R-project.​org) was used to perform statistical analyses. IPGTT data were analysed using two-way ANOVA. All other statistical analyses were performed using independent two-way Student’s t-tests. Data were considered significant if the p value was <0.05.

Results

Transplanted islets function normally in mice fitted with an abdominal imaging window
Mice recovered quickly after surgery and were fully active after 1 h. There was no impairment of movement and mice did not show behaviour indicating pain or discomfort. Body weight increased normally after surgery (ESM Fig. 2a). To test if islet graft functionality was affected by the procedure, an IPGTT was performed and human C-peptide concentrations were measured in streptozotocin-induced hyperglycaemic NSG mice 4 weeks after transplantation of 2000 human islet equivalents. Both mice with an abdominal imaging window (AIW, n = 5) and controls (CTRL, n = 5) showed a normal response to an IPGTT (ESM Fig. 2b). Human C-peptide concentrations were similar between the two groups 4 weeks after surgery (358 ± 157 pmol/l [AIW] and 384 ± 47 pmol/l [CTRL], p = 0.88; ESM Fig. 2c). Graft thickness (801 ± 86 μm [AIW] and 827 ± 51 μm [CTRL]) was unaffected (p = 0.81). Capsule thickness was not significantly increased (477 ± 64 μm [AIW] and 324 ± 34 μm [CTRL], p = 0.09; ESM Fig. 2d). The imaging window did not affect the proportion of beta cells (relative to the total alpha and beta cells) in the transplanted islets (ESM Fig. 2e–g).
Transplanted islets can be repeatedly monitored during in vivo engraftment
To demonstrate that longitudinal intravital imaging of tissue grafts under the kidney capsule can be performed, normoglycaemic NSG mice (n = 4) were transplanted with 10–25 mouse MIP-EGFP/CAG-DsRed islets and fitted with an AIW. Imaging was performed on day 1, day 4, day 8 and day 15 after surgery (Fig. 1a–d). In total, 29 separate islet-containing regions of interest were identified. In these areas, beta cell and total cell volume were measured by volumetric calculation of the DsRed and EGFP signal. DsRed volume at 15 days was 2.1 times that at day 1 (7.4 × 105 ± 1.1 × 105 μm3 at day 1 vs 15.3 × 105 ± 3.1 × 105 μm3 at day 15, p = 0.029), while EGFP volume did not significantly increase over this time (4.7 × 105 ± 0.7 × 105 μm3 at day 1 vs 6.9 × 105 ± 1.6 × 105 μm3 at day 15, p = 0.22) (Fig. 1e). The relative EGFP content decreased from 69.5 ± 6.5% on day 1 to 44.5 ± 5.0% on day 15 (p = 0.004) (Fig. 1f). To show that human islet cells can also be sequentially imaged, HIP-GFP-transduced human islet cell aggregates were transplanted under the kidney capsule. Approximately 50% of all transplanted cells expressed GFP after transduction prior to transplantation (ESM Fig. 3). Beta cells expressing GFP could be clearly visualised on days 1, 4, 8 and 15 after transplantation under the kidney capsule (Fig. 1g–j).
Visualisation of islet graft vasculature and blood flow
Blood vessels were visualised in MIP-EGFP islet grafts after injection of Texas Red-labelled dextrans into the tail vein. The first signs of vascularisation were recorded 3 days after transplantation, when blood vessels could be identified within and surrounding the islets (Fig. 1k, ESM Fig. 4).
Transplanted embryonic pancreas increases in size and forms islets within 2 weeks
To demonstrate that large tissue grafts can be visualised and to monitor pancreatic development in a quantitative fashion, we transplanted two to four E12.5 embryonic pancreases from pregnant MIP-EGFP/CAG-DsRed mice under the kidney capsule of NSG mice before an AIW was fitted (n = 7). One day after transplantation, 27.3 ± 3.6 EGFP-positive cells were present per embryonic pancreas (n = 18). These cells were usually single and dispersed throughout the embryonic pancreas (Fig. 2a). When multiple pancreases were transplanted, the grafts joined together over time (Fig. 2a–e). After 7 days, the first islet-like clusters were visible in the grafts (Fig. 2f, ESM movie). Mature islets were present 2 weeks after transplantation, as illustrated by rounded and separated EGFP-positive cell clusters during intravital imaging (Fig. 2g) and immunohistochemical staining for insulin and glucagon (Fig. 2h). DsRed volume at 14 days was 6.1 times that at day 1 (9.5 ± 2.1 × 107 μm3 at day 1 vs 5.8 ± 1.2 × 108 μm3 at day 14, p = 0.014), and EGFP volume was 90 times that at day 1 (2.3 ± 0.5 × 105 μm3 at day 1 vs 2.1 ± 0.7 × 107 μm3 day 14, p = 0.04) (Fig. 2i). After 2 weeks, the EGFP-positive cells represented 3.34 ± 0.78% of the DsRed-positive cells (Fig. 2j).
ESM movie
Live intravital imaging movie of E12.5 MIP-EGFP::CAG-DsRed embryonic, 7 days after transplantation under the kidney capsule. Functional blood vessels are visualized in red, and erythrocytes can be observed moving through them. MIP-EGFP (green), CAG-DsRed (red). MIP-EGFP: Enhanced green fluorescent protein (GFP) under the transcriptional regulation of the mouse insulin promoter (MIP). CAG-DsRed: Red fluorescent protein (DsRed) under the transcriptional regulation of the cytomegalovirus-actin-globin (CAG) promoter. (MP4 26101 kb)

Discussion

Here we describe a novel intravital imaging model to study mature islet cells and developing pancreatic tissue using an abdominal imaging window. Tissues transplanted under the kidney capsule, which is considered the gold standard site for studying islet grafts, can be sequentially imaged for at least 2 weeks. Using fluorescent markers, we longitudinally measured engraftment, expansion, differentiation and vascularisation in transplanted developing pancreatic tissue and mature islets.
Alternative intravital imaging methods have been used to study islet function, survival and vascularisation over time. While the anterior chamber of the eye model allows prolonged (up to 6 months) sequential intravital imaging of transplanted pancreatic islets [7], it is limited by the tissue volume that can be transplanted. This reduces options to transplant large amounts of tissue, or tissues with expansion capacity over time.
Our model allows the dynamic characterisation of the embryonic pancreas, which undergoes further development and expansion after transplantation under the kidney capsule [8]. Fetal pancreases are by nature highly proliferative [9]. The time course of growth and differentiation of embryonic pancreases transplanted under the kidney capsule is very similar to that observed in eutopic pancreatic development [10]. Sequential intravital imaging shows the process of islet neogenesis, as indicated by the appearance of rounded clusters of fluorescent cells. In the majority of embryonic pancreatic tissue transplants no acinar tissue was observed, as reported previously [8]. This is probably the reason why the percentage of beta cells in our grafts is relatively high compared with normal pancreatic development.
We were also able to dynamically assess transplanted islet grafts. Islets were connected to the host vasculature within 3 days, which is in line with previous findings [7]. Generally, placement of an imaging window did not affect survival or function of islets transplanted under the kidney capsule.
Important processes such as engraftment, vascularisation, expansion and differentiation can be studied using this imaging method. The technique can therefore be a valuable tool in beta cell replacement therapy using progenitor cells, islet inflammation and rejection. In conclusion, we have developed a novel method to dynamically image both mature islet cells and developing pancreatic tissue.

Funding

This work was supported by the stichting Diabetes Onderzoek Nederland (DON).

Duality of interest

The authors declare that there is no duality of interest associated with this manuscript.

Contribution statement

LvG, CJML, JvR and EJPdK contributed to the design of the study. LvG, PPvK, GD, EJ, FCASR and EB contributed to the acquisition of the data. LvG and EJPdK contributed to the drafting of the article. All authors contributed to revision of the article and gave final approval. LvG is responsible for the integrity of the work as a whole.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Sakata N, Tan A, Chan N et al (2009) Efficacy comparison between intraportal and subcapsular islet transplants in a murine diabetic model. Transplant Proc 41:346–349CrossRefPubMedPubMedCentral Sakata N, Tan A, Chan N et al (2009) Efficacy comparison between intraportal and subcapsular islet transplants in a murine diabetic model. Transplant Proc 41:346–349CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Loganathan G, Graham ML, Radosevich DM et al (2013) Factors affecting transplant outcomes in diabetic nude mice receiving human, porcine, and nonhuman primate islets: analysis of 335 transplantations. Transplantation 95:1439–1447CrossRefPubMedPubMedCentral Loganathan G, Graham ML, Radosevich DM et al (2013) Factors affecting transplant outcomes in diabetic nude mice receiving human, porcine, and nonhuman primate islets: analysis of 335 transplantations. Transplantation 95:1439–1447CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Gao J, Tian L, Weng G, O’Brien TD, Luo J, Guo Z (2011) Stimulating beta-cell replication and improving islet graft function by AR231453, A gpr119 agonist. Transplant Proc 43:3217–3220CrossRefPubMed Gao J, Tian L, Weng G, O’Brien TD, Luo J, Guo Z (2011) Stimulating beta-cell replication and improving islet graft function by AR231453, A gpr119 agonist. Transplant Proc 43:3217–3220CrossRefPubMed
4.
Zurück zum Zitat Ritsma L, Steller EJ, Beerling E et al (2012) Intravital microscopy through an abdominal imaging window reveals a pre-micrometastasis stage during liver metastasis. Sci Transl Med 4:158ra145CrossRefPubMed Ritsma L, Steller EJ, Beerling E et al (2012) Intravital microscopy through an abdominal imaging window reveals a pre-micrometastasis stage during liver metastasis. Sci Transl Med 4:158ra145CrossRefPubMed
5.
Zurück zum Zitat Ritsma L, Steller EJ, Ellenbroek SI, Kranenburg O, Borel Rinkes IH, van Rheenen J (2013) Surgical implantation of an abdominal imaging window for intravital microscopy. Nat Protoc 8:583–594CrossRefPubMed Ritsma L, Steller EJ, Ellenbroek SI, Kranenburg O, Borel Rinkes IH, van Rheenen J (2013) Surgical implantation of an abdominal imaging window for intravital microscopy. Nat Protoc 8:583–594CrossRefPubMed
6.
Zurück zum Zitat Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW (1988) Automated method for isolation of human pancreatic islets. Diabetes 37:413–420CrossRefPubMed Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW (1988) Automated method for isolation of human pancreatic islets. Diabetes 37:413–420CrossRefPubMed
8.
Zurück zum Zitat Brown J, Molnar IG, Clark W, Mullen Y (1974) Control of experimental diabetes mellitus in rats by transplantation of fetal pancreases. Science 184:1377–1379CrossRefPubMed Brown J, Molnar IG, Clark W, Mullen Y (1974) Control of experimental diabetes mellitus in rats by transplantation of fetal pancreases. Science 184:1377–1379CrossRefPubMed
9.
Zurück zum Zitat Castaing M, Peault B, Basmaciogullari A, Casal I, Czernichow P, Scharfmann R (2001) Blood glucose normalization upon transplantation of human embryonic pancreas into beta-cell-deficient SCID mice. Diabetologia 44:2066–2076CrossRefPubMed Castaing M, Peault B, Basmaciogullari A, Casal I, Czernichow P, Scharfmann R (2001) Blood glucose normalization upon transplantation of human embryonic pancreas into beta-cell-deficient SCID mice. Diabetologia 44:2066–2076CrossRefPubMed
10.
Zurück zum Zitat Mullen YS, Clark WR, Molnar IG, Brown J (1977) Complete reversal of experimental diabetes mellitus in rats by a single fetal pancreas. Science 195:68–70CrossRefPubMed Mullen YS, Clark WR, Molnar IG, Brown J (1977) Complete reversal of experimental diabetes mellitus in rats by a single fetal pancreas. Science 195:68–70CrossRefPubMed
Metadaten
Titel
Sequential intravital imaging reveals in vivo dynamics of pancreatic tissue transplanted under the kidney capsule in mice
verfasst von
Léon van Gurp
Cindy J. M. Loomans
Pim P. van Krieken
Gitanjali Dharmadhikari
Erik Jansen
Femke C. A. S. Ringnalda
Evelyne Beerling
Jacco van Rheenen
Eelco J. P. de Koning
Publikationsdatum
21.07.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 11/2016
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-016-4049-6

Weitere Artikel der Ausgabe 11/2016

Diabetologia 11/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.