Skip to main content
Erschienen in: Diabetologia 12/2012

Open Access 01.12.2012 | Article

Inhibition of activin/nodal signalling is necessary for pancreatic differentiation of human pluripotent stem cells

verfasst von: C. H.-H. Cho, N. R.-F. Hannan, F. M. Docherty, H. M. Docherty, M. Joåo Lima, M. W. B. Trotter, K. Docherty, L. Vallier

Erschienen in: Diabetologia | Ausgabe 12/2012

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Aims/hypothesis

Human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hIPSCs) offer unique opportunities for regenerative medicine and for the study of mammalian development. However, developing methods to differentiate hESCs/hIPSCs into specific cell types following a natural pathway of development remains a major challenge.

Methods

We used defined culture media to identify signalling pathways controlling the differentiation of hESCs/hIPSCs into pancreatic or hepatic progenitors. This approach avoids the use of feeders, stroma cells or serum, all of which can interfere with experimental outcomes and could preclude future clinical applications.

Results

This study reveals, for the first time, that activin/TGF-β signalling blocks pancreatic specification induced by retinoic acid while promoting hepatic specification in combination with bone morphogenetic protein and fibroblast growth factor. Using this knowledge, we developed culture systems to differentiate human pluripotent stem cells into near homogenous population of pancreatic and hepatic progenitors displaying functional characteristics specific to their natural counterparts. Finally, functional experiments showed that activin/TGF-β signalling achieves this essential function by controlling the levels of transcription factors necessary for liver and pancreatic development, such as HEX and HLXB9.

Conclusion/interpretation

Our methods of differentiation provide an advantageous system to model early human endoderm development in vitro, and also represent an important step towards the generation of pancreatic and hepatic cells for clinical applications.
Begleitmaterial
ESM Fig. 1 Screening of defined culture conditions driving differentiation of DE cells toward the pancreatic lineages. (a-c) Basal media composition affects the capacity of DE cells to differentiate into PDX1 expressing cells. DE cells generated as described in Figure 1 were grown for 9 additional days in the presence of RA, SB, FGF10 and Noggin in diverse basal media (Advanced DMEM (Invitrogen); RPMI, CDM-PVA, CDM-BSA (CDM5)). (d-f) Effect of absence and increasing dose of RA (0.3 mM, 1.5 mM, 3 mM, 6 mM) on expression of pancreatic endoderm markers. (g-i) BMP inhibits differentiation of DE toward pancreatic endoderm. DE cells were grown for 6 days in the presence of RA, SB, FGF10 and BMP4 10 ng/ml (N0, B6) or RA, SB, FGF10 and Noggin (N6, B0). (j-l) FGF signalling is not necessary for differentiation of DE into pancreatic endoderm. DE cells were grown for 6 days in the presence of RA, SB, SU5402 (10 mM) and Noggin (6SU, 0F) or RA, SB, FGF10 and Noggin (0SU, 6F). (m-o) Increasing dose of FGF10 (5 ng/ml, 25 ng/ml, 50 ng/ml, and 100 ng/ml) does not affect the expression of pancreatic endoderm markers during DE differentiation. (p-af) Expression of pluripotency (POU5F1, NANOG, SOX2), primitive streak markers (Brachyury (T), MIXL1, EOMES), endoderm (SOX17, CXCR4, HEX), pancreatic endoderm markers (FOXA2, HNF6, SOX9) and endocrine markers (NGN3, PTF1a, SST, Glucagon (GLU)) during differentiation of hESCs into pancreatic endoderm using 4 step protocol described in Figure 1. (PDF 228 kb)
ESM Fig. 3 Differentiation of hIPSCs into pancreatic endoderm cells using culture conditions developed with hESCs. (a-d) Expression of pancreatic markers in hIPSCs differentiating into pancreatic endoderm using the 4 step protocol described in Figure 1. (hIPSC1 = A1ATD-hIPSCs, hIPSC2 = BBHX8, hIPSC3 = JRO1D). (e-i) Expression of pancreatic endoderm and Hormonal markers in hIPSCs grown respectively for 15 days and 18 days in culture conditions inductive for pancreatic specification. (j) FACS analyses showing the fraction of PDX1 expressing cells generated after 12 days of differentiation. (k-m) C-peptide secretion upon glucose stimulation in endocrine cells generated from hIPSCs. (PDF 512 kb)
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00125-012-2687-x) contains peer-reviewed but unedited supplementary material, which is available to authorised users.
C. H.-H. Cho and N. R.-F. Hannan contributed equally to this study.
Abkürzungen
BMP
Bone morphogenetic protein
CDM
Chemically defined medium
DE
Definitive endoderm
DF
Dorsal foregut
FGF
Fibroblast growth factor
hESC
Human embryonic stem cell
HEX
Homeobox protein
hIPSC
Human induced pluripotent stem cell
HLXB9
Homeobox HB9
hPSC
Human pluripotent stem cell
PE
Pancreatic endoderm
qPCR
Quantitative PCR
RA
Retinoic acid
SB
SB431542
SCID
Severe combined immunodeficiency
shRNA
Small hairpin RNA
VF
Ventral foregut

Introduction

Production of pancreatic beta cells is a major objective of regenerative medicine. An increased supply of these cells will enable the development of cell-based therapy against diabetes, which is currently limited by the lack of donated organs and difficulties with increasing insulin-secreting cells in vitro. Human pluripotent stem cells (hPSCs) of embryonic origin (human embryonic stem cells [hESCs]) [1] or generated from reprogrammed somatic cells (human induced pluripotent stem cells [hIPSCs]) [2] offer the prospect of bypassing these restrictions. These cells are capable of proliferating indefinitely in vitro while maintaining the capacity to differentiate into a broad number of cell types, including pancreatic progenitors [36]; however, robust protocols allowing for the production of homogenous populations of these cells in defined culture conditions have not yet been established. Current methods involve undefined animal products such as feeders, FBS and Matrigel and only allow for the generation of heterogeneous populations of cells, thus increasing the risk of teratoma formation after transplantation [7, 8]. They also appear to work efficiently on a limited number of hPSC lines [3], which hinders their use in a large number of laboratories.
Most of the culture systems currently used to direct the differentiation of hPSCs mimic normal development, since this approach could facilitate the generation of fully functional cell types. Consequently, the knowledge coming from studies on mice or other vertebrate animal models has been used to inform strategies driving hPSCs towards specific lineages. The pancreas and liver arise at approximately embryonic day 8.5–9.5 from adjacent regions of the developing primitive foregut under the influence of inductive signals secreted by the nearby mesoderm [9]. These signals probably command the production of transcription factors necessary for pancreatic specification, such as HLXB9, which marks the dorsal foregut (DF) prior to the formation of the pancreatic bud [10, 11], and PDX1, which marks regions of the foregut from which the ventral and dorsal pancreatic buds arise [12, 13]. The newly specified pancreatic endoderm quickly produces additional markers, including PTF1A, NKX6.1 and SOX9, and these progenitors give rise to both endocrine (islets of Langerhans) and exocrine (acinar and ductal cells) cells of the pancreas. Similar mechanisms control hepatic specification, although they involve a different set of transcription factors, such as homeobox protein (HEX), GATA-binding factor 6 (GATA6), Prospero homeobox protein 1 (PROX1) and hepatic nuclear factor 4α (HNF4A) [14], and signalling pathways, such as bone morphogenetic protein (BMP) and fibroblast growth factors (FGFs) [15]. Despite this broad knowledge, the molecular mechanisms enabling extracellular signalling pathways to orchestrate the transcriptional networks characterising pancreatic or hepatic progenitors remain to be elucidated. Especially in humans, hPSCs could present a unique advantage in completing this major task.
In the current study, we screened defined culture conditions to differentiate human definitive endoderm (DE) from multiple hPSC lines into a near homogenous population of pancreatic endoderm cells. Our analyses revealed that activin/TGF-β controls DE cell fate choice between the pancreatic and hepatic lineages by controlling the levels of key transcription factors. These observations facilitated the development of defined culture systems to differentiate DE cells into near homogenous populations of pancreatic and hepatic endoderm following a natural path of development. Therefore, the method described here not only provides a unique in vitro model of development for basic studies, but also represents a first step towards the production of pancreatic and hepatic cell types for therapeutic use.

Methods

hESC and hIPSC culture conditions
hESCs (H9 from WiCell, Maddison, WI, USA) and hIPSCs (BBHX8, A1ATD-1, JRO1D; University of Cambridge, Cambridge, UK) [16] were passaged weekly using collagenase IV and maintained in chemically defined medium (CDM) supplemented with activin A (10 ng/ml) and FGF2 (12 ng/ml), as described previously [17]. Differentiation was carried out as described in Fig. 1 and is explained in detail in the electronic supplementary material (ESM) Methods.
RT-qPCR, immunostaining, FACS analyses and HEX/HLXB9 knockdown
The methods used for RT-quantitative (q)PCR, immunostaining and FACS analyses have been described elsewhere [18, 19] and are outlined in the ESM Methods and ESM Table 1. All data are presented as average of three independent biological triplicates and error bars indicate SDs.
C-peptide release, cytochrome P450 activity, periodic acid Schiff and LDL-cholesterol uptake assays
C-peptide ELISA (Mercodia, Uppsala, Sweden), Cyp3A4 P450-Glo assays (Promega, Maddison, WI, USA), periodic acid Schiff staining (Sigma, St Louis, MO, USA) and 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine-labelled acetylated LDL (Dil-LDL) uptake (Cayman Chemicals, MA, USA) were measured in triplicate according to the manufacturers’ instructions.
Microarray profiling
RNA was hybridised to Illumina (San Diego, CA, USA) HumanHT-12 v4.0R1 Expression BeadChips using the manufacturer’s standard protocols. BeadChip probe sets that did not pass the Illumina signal detection statistic at a threshold of p < 0.01 in all sample replicates of at least one sample group were removed from further analysis. Array processing was performed using the BeadArray package of the Bioconductor suite of software (www.​bioconductor.​org) for the R statistical programming language (www.​r-project.​org). See ESM for more detailed methods.
Animal studies
Differentiated cells (5 × 106) were grafted under the kidney capsule of NOD/severe combined immunodeficiency (SCID) mice using a 24G catheter attached to a positive displacement pipette. Blood samples were removed from the tail at various time intervals for C-peptide analysis. Kidneys were harvested at the indicated time points, and a section containing the grafted cells was fixed in 4% paraformaldehyde, wax embedded and processed for immunohistochemistry. Antibody binding was visualised using 3,3′-diaminobenzidine.
Ethics approvals
Ethics approval was obtained for hPSCs (authorisation 09/H0306/73 and 08/H0311/201) and hESCs were imported under the guidelines of the UK Stem Cell Bank Steering Committee (authorisation SCSC10-44).
All animal experiments were conducted following UK regulations under the project licence PPL60/4242. Animals were sourced from Harlan laboratories (Indianapolis, IN, USA).

Results

Inhibition of activin and BMP signalling in the presence of retinoic acid induces PDX1 expression in hESC-derived endoderm cells grown in fully defined culture conditions
We recently established a defined culture system to differentiate hESCs and hIPSCs into near homogenous populations of DE cells [20]. This culture system relies on a CDM devoid of animal product including BSA, serum, complex extracellular matrix such as Matrigel (BD Biosciences, NJ, USA) or feeder cells, thereby avoiding the presence of unknown factors that might interfere with experimental outcomes. To further extend this protocol, we screened a broad number of combinations of growth factors and inhibitors of signalling pathways to identify defined culture conditions driving the differentiation of DE cells into pancreatic progenitors. These analyses revealed that a combination of retinoic acid (RA), FGF10, noggin (a BMP inhibitor) and SB431542 (SB; an activin/TGF-β receptor antagonist) was able to induce expression of the pancreatic markers PDX1, HNF6, PTF1A, SOX9 and HLXB9 (also known as MNX1), while inhibiting the expression of gut (CDX2) and liver (AFP) markers in hESC-derived DE cells (Fig. 2a–g). Importantly, this cocktail of factors only induced PDX1 in specific basal medium (advanced DMEM) while the presence of serum, Matrigel or feeders (ESM Fig. 1a–c and data not shown) inhibited pancreatic endoderm differentiation, confirming that DE differentiation can be influenced by a diversity of factors.
We then sought to validate and optimise the role of each of these additives. Absence of RA in the presence of noggin, FGF10 and SB inhibited the expression of pancreatic markers (Fig. 2a–c and ESM Fig. 1d–f), confirming that RA is necessary for the induction of pancreatic specification [21]. Absence of noggin or addition of BMP4 at any time during the differentiation process (Fig. 2a–g and ESM Fig. 1g–i) resulted in a significant decrease in the expression of pancreatic endoderm markers while inducing gut (CDX2) and liver (AFP) markers, thereby reinforcing previous studies showing that BMP signalling inhibits pancreatic specification to promote alternative cell fate [22]. Inhibition of FGF signalling using SU5402 (an FGF receptor antagonist) or increasing the dose of FGF10 did not affect the expression of pancreatic endoderm markers such as PDX1, SOX9 and HLXB9 (Fig. 2a–g, ESM Fig. 1j–o). However, the expression of the gut marker CDX2 [23] was augmented and the expression of PTF1A was strongly reduced in the absence of FGF signalling, suggesting that FGF10 might block the specification of PDX1-expressing cells towards duodenum [23, 24] during pancreatic bud specification. Furthermore, FGF inhibition caused significant cell death, implying that FGFs are also necessary for the proliferation and survival of pancreatic endoderm in vitro. More importantly, we observed that the addition of activin abolished the expression of pancreatic markers while inhibition of activin/TGF-β signalling by SB had the opposite effect (Fig. 2a–g), demonstrating for the first time that activin/TGF-β signalling inhibits pancreatic specification in vitro. Interestingly, the presence of SB was only required for the first 3 days of differentiation, indicating that activin/TGF-β signalling acted on the earliest steps of pancreatic specification preceding PDX1 expression. Together, these results show that RA acts as an inductive signal driving the differentiation of DE cells towards the pancreatic lineage, while TGF-β signalling pathways (i.e. activin and BMP) act as a potent inhibitor of this cell fate choice.
Inhibition of activin/TGF-β induces differentiation of endoderm into a near homogenous population of pancreatic endoderm following a native path of development
Based on the results described above, we established a four-step protocol to differentiate hESCs into pancreatic endoderm using defined culture media (Def-Panc, Fig. 1). During the first step (days 1–3), hESCs were grown in CDM supplemented with activin/BMP/FGF2/LY294002 (a phosphoinositide 3-kinase inhibitor) [20]. The resulting cells were positive for the expression of DE markers including SOX17, CXCR4, HEX (also known as HHEX), FOXA2 and EOMES, and simultaneously negative for the expression of the pluripotency markers POU5f1 (also known as OCT3/4), NANOG and SOX2 and the primitive streak markers T (brachyury) and MIXL1 (Fig. 2h–m and ESM Fig. 1p–af). The second step of the Def-Panc protocol involved growing DE cells in the presence of RA/noggin/FGF10/SB431542 for 3 days (days 4–6). The resulting cells expressed HNF1B FOXA2, HNF4A, RFX6 and HLXB9 (Fig. 2i–m and ESM Fig. 1p–z), all of which mark the foregut during early mammalian development.
Notably, the expression of HLXB9 and the absence of HEX expression suggested a dorsal identity for these foregut cells, while the absence of CDX2 excluded the presence of midgut or hindgut cells (Fig. 2k, ESM Fig. 1x and data not shown). In the third step of the protocol, DF cells were grown for three additional days in the presence of RA/noggin/FGF10/cyclopamine (days 7–9). The resulting cells expressed a combination of foregut markers (HNF1B, SOX2, FOXA2 and HLXB9) and pancreatic endoderm markers (SOX9, HNF6, PTF1A and PDX1) (Fig. 2i–l and ESM Fig. 1s–af). The expression of pancreatic endoderm markers was further reinforced in the fourth step of the protocol by the addition of FGF10 for 3 days (days 10–12). The resulting cells expressed NKX6.1, SOX9, HNF6, PTF1A, PDX1, HNF1B, SOX2 and FOXA2, while the expression of HLXB9 was strongly diminished (Fig. 2i–m and ESM Fig. 1a,b). FACS analyses performed at the end of step 1 showed that the DE-enriched cells were homogeneously positive for CXCR4 and after the fourth step of the protocol (day 12), 80% of cells expressed PDX1 (Fig. 2n,o). Immunostaining analyses also confirmed that PDX1 was coexpressed with SOX9, HNF6, HNF4, NKX6.1 and GATA4 (Fig. 2p–t) in approximately 80% of the cells generated. Together, these results indicate that the Def-Panc protocol drives differentiation of hESCs towards a near homogenous population of pancreatic endoderm cells following successive events of specifications reminiscent of those that occur during pancreatic development.
PDX1 endoderm generated in defined culture conditions can differentiate into insulin secreting cells in vitro and in vivo
To confirm the capacity of pancreatic endoderm cells to differentiate further towards the endocrine lineage, PDX1-expressing cells obtained at the end of stage 4 were grown for six additional days in culture conditions previously shown to stimulate endocrine cell differentiation [3]. qPCR analyses showed that PDX1 expression decreased after 3 days, while expression of NGN3 and hormonal markers (insulin, glucagon and somatostatin) progressively increased (Fig. 2i–m and ESM Fig. 1ac–af). By day 18, 11% of cells stained positive for C-peptide (Fig. 3a). Interestingly, these hESC-derived insulin-expressing cells were able to release C-peptide upon glucose stimulation, mimicking insulin release by pancreatic beta cells (Fig. 3c). Nonetheless, the expression of hormonal markers (insulin, somatostatin and glucagon, ESM Fig. 2a–k) was relatively low when compared with that of human adult islets cells, while the expression of markers specific to endocrine progenitors was maintained (NKX6.1, NGN3 and SOX9, ESM Fig. 2a–k). Furthermore, a fraction of C-peptide-expressing cells was also positive for glucagon or somatostatin (Fig. 3b).
Polyhormonal expression could mark islet cells of embryonic origin [25], thereby confirming that our in vitro culture conditions are not sufficient to generate fully functional endocrine cells. To overcome this limitation of the in vitro system, pancreatic endoderm cells obtained after 12 days of differentiation were injected under the kidney capsule of NOD/SCID mice to provide an environment known to favour their differentiation into endocrine cells [3]. A low level of human C-peptide was detected in the blood stream of three out of eight transplanted animals as soon as 12 weeks after transplantation (negative control = 0.021 ng/ml; mouse 1 = 0.1 ng/ml, mouse 2 = 0.43 ng/ml, mouse 3 = 0.1635 ng/ml) (Fig. 3d). In addition, histology analyses performed after 19 weeks of differentiation in vivo revealed the presence of islet-like clusters with cells containing glucagon and C-peptide (Fig. 3e–i). Together, these results demonstrate that pancreatic endoderm cells generated with the Def-Panc protocol have the capacity to differentiate further into endocrine cells and thus represent early pancreatic progenitors. Finally, similar results were obtained with three hIPSCs lines (ESM Fig. 3), indicating that the Def-Panc protocol could be successfully used to produce pancreatic progenitors from diverse hPSCs.
Activin/TGF-β drives differentiation of endoderm cells into hepatic progenitors, which can differentiate into fetal hepatocytes
During screening of the culture conditions described above, we noted that DE cells grown in the presence of activin acquired the appearance of fetal hepatocytes with large darkened cytoplasmic space and canaliculi-like structures (data not shown). Further analyses confirmed that DE cells grown in the presence of activin for 5 days expressed genes marking the ventral foregut (VF), the site of liver bud formation (HEX, SOX17, HNF4A, FOXA1, FOXA2 and TBX3 in Fig. 4a–h). Conversely, inhibition of activin by SB decreased the expression of HNF4A, SOX17, HEX and TBX3, while blocking known hepatic inducers such as FGF signalling also decreased the expression of liver bud genes such as HEX, SOX17 and TBX3 (Fig. 4i–l). Surprisingly, noggin only induced a moderate decrease in HNF4A expression, suggesting that BMP signalling might have a limited function in hepatic specification in vitro. Alternatively, unknown signalling pathways could activate the same programme of differentiation. Considered together, these results suggest that the combined effect of activin, BMP and FGF is necessary to fully promote hepatic specification of DE cells in vitro.
Based on this observation, we developed a three-step protocol to generate hepatocytes from hPSCs in defined culture conditions (Def-Hep, Fig. 1). The first step of the Def-Hep protocol consisted of differentiating hESCs into DE cells, as described above, while the second step involved promoting DE specification towards the hepatic lineage using first activin alone for 3 days and then activin combined with BMP4 and FGF10. In the third step of the Def-Hep protocol, hepatic endoderm cells were grown for 15 additional days in the presence of oncostatin M and hepatocyte growth factor, two growth factors known to control hepatoblast differentiation into hepatocytes. Accordingly, the cells generated with the Def-Hep protocol expressed hepatocyte markers such as ALB, AAT, αAPOF, TAT, TDO2, TTR, HNF4A and HEX (Fig. 5a–j). These observations were confirmed by immunostaining and FACS analyses, which showed homogenous coexpression of ALB, CK18, AAT and AFP (Fig. 5k–m). These cells also displayed functional characteristic of hepatocytes, such as: (i) albumin and α-1-antitrypsin secretion (Fig. 5n–o); (ii) CYP3A4 activity inducible by dexamethasone (Fig. 5p); (iii) cholesterol uptake; and (iv) glycogen storage (Fig. 5q–r). Together, these data demonstrate that activin drives DE specification towards VF-like cells and then hepatic endoderm, which has the capacity to differentiate into cells displaying characteristics of fetal hepatocytes.
HEX and HLXB9 knockdown during pancreatic and hepatic differentiation of hESCs blocks hepatic and pancreatic differentiation, respectively
We then decided to take advantage of the Def-Panc and Def-Hep culture systems to study the mechanisms by which activin can control the cell-fate choice between the pancreatic and hepatic lineages. For that, we performed gene-expression profiling experiments to identify genes that were up- or downregulated by the presence of activin during pancreatic specification. These analyses revealed that activin could activate or block the expression of a broad number of genes, including HEX and HLXB9 (Fig. 6a), that are known to be essential for foregut development. Thus, we hypothesised that activin could direct DE specification by controlling the expression of these transcription factors. To test this hypothesis, we knocked down HEX or HLXB9 levels in hESCs using stable expression of small hairpin (sh)RNA. The resulting hESC sublines (shHEX-hESCs and shHLXB9-hESCs) were then differentiated as described in Fig. 1. qPCR analyses showed that knockdown of HEX levels during VF differentiation (Fig. 6b) was systematically associated with downregulation of hepatic markers such as AFP and albumin (Fig. 6c–k). A similar decrease was not observed with DE cells derived from shHLXB9-hESCs or DE cells derived from hESCs stably overexpressing a non-targeting shRNA (shScramble-hESCs) (Fig. 6c, f and i, and data not shown). However, we also observed that reduced HEX expression increased cell death during VF differentiation (Fig. 6l). Therefore, HEX expression appears to be necessary for the survival and differentiation of VF-like cells towards the hepatic lineage in vitro. Finally, shHEX-hESCs were able to differentiate into pancreatic endoderm expressing successively HLXB9 and PDX1 (Fig. 6m,n).
Similar experiments performed with shHLXB9-hESCs showed that knockdown of HLXB9 levels during foregut differentiation strongly decreased the expression of pancreatic endoderm markers, including PDX1/SOX9 (Fig. 7a–g). Interestingly, a decrease in HLXB9 expression did not affect the expression of foregut makers such as HNF4A, FOXA2 and HNF1B (Fig. 7a–f), suggesting that HLXB9 is not required for DF specification while being necessary for its differentiation towards the pancreatic lineage. Importantly, HLXB9 is not expressed during hepatic differentiation (Fig. 7h), and thus DE cells generated from shHLXB9-hESCs were able to differentiate into VF-like cells and into hepatic endoderm when grown in the presence of activin (data not shown).
Collectively, these results recapitulate studies performed in the mouse embryo showing that absence of HEX disrupts hepatic bud development without affecting dorsal pancreatic specification, while HLXB9 is necessary for induction of PDX1 expression in the pancreas [26]. Therefore, they demonstrate the general application of our culture system in modelling DE development and studying early organogenesis of the pancreas and liver in vitro.

Discussion

Robust protocols allowing for the production of homogenous populations of liver and pancreatic progenitors from hPSCs under culture conditions compatible with clinical applications have not yet been established. Available methods often contain undefined animal products such as feeders or FBS. To address these challenges, we screened defined culture conditions to differentiate human DE into a near homogenous population of pancreatic and liver endoderm from multiple hPSC lines. The result of this screening shows that RA has an essential function in promoting pancreatic specification while BMP signalling blocks the expression of the pancreatic marker PDX1, reinforcing previous studies [21, 22]. However, our results concerning the function of FGF signalling contradict those of previous studies [27] by suggesting that FGF acts as a permissive rather than an inductive signal of pancreatic specification.
This apparent divergence might be explained by the absence in our culture conditions of feeders, serum and Matrigel, all of which contain unknown components that are likely to interfere with FGF signalling. In addition, we observed that inhibition of FGF signalling decreases cell survival of pancreatic progenitors, thus justifying the use of FGFs in our protocol. More importantly, our analyses also revealed that activin/TGF-β controls DE cell fate choice towards the pancreas lineage by inhibiting DF specification while promoting the hepatic lineage. Previous studies have shown that TGF-β signalling controls ventral pancreatic bud induction in the mouse embryo [15] and, thus, our data demonstrate for the first time that similar mechanisms could occur in the dorsal pancreas, confirming the interest of our culture system in modelling foregut development in vitro. Interestingly, a recent report has shown that inhibition of TGF-β signalling can block gut specification induced by WNT signalling [24] and also promote endocrine differentiation of PDX1-expressing progenitors [28, 29]. Thus, TGF-β can control different stages of pancreatic development. Further studies will be necessary to fully understand the successive functions of TGF-β pathways during primitive tube patterning and organogenesis.
Our results also show that activin/TGF-β signalling directs hepatic vs pancreatic specification by controlling transcription factors directing foregut differentiation, such as HEX and HLXB9. Interestingly, we recently identified HEX as a direct target of Smad2/3 using chromatin immunoprecipitation-sequencing analyses in differentiating DE cells [30], and thus activin/TGF-β signalling could ‘directly’ control hepatic specification by maintaining HEX expression during DE development. Our findings are in keeping with a recent demonstration that HEX plays a pivotal role in the induction of liver development from mouse embryonic stem-cell-derived endoderm [31], and the observation that transient overexpression of HEX in hESCs and hIPSCs enhances the formation of hepatoblasts [32]. However, our findings also suggest an essential function for HEX in the early stage of hepatic specification, which differs from the results obtained with mouse embryos showing that HEX is not necessary for liver bud induction [33]. Nevertheless, these experiments were performed either with whole embryos or with culture explant, mixing several tissues grown in culture media containing serum and unknown factors. Our culture system is based on defined culture conditions that might not contain all of the necessary factors to maintain hepatic endoderm in the absence of HEX. This hypothesis is in agreement with recent studies indicating that BMP and HEX display a synergistic effect on hepatic specification [31].
Similarly, activin/TGF-β signalling could inhibit pancreatic specification by blocking HLXB9 expression, which is necessary for the subsequent induction of PDX1 [10, 11]. This mechanism is likely to involve other factors since Smad2/3 was not detected on the HLXB9 promoter upon DE differentiation and thus cannot play a ‘direct’ repressive function. The importance of HLXB9 also indicates that cells generated in vitro have a DF origin, since HLXB9 is not necessary for ventral bud development [10, 11]. Nevertheless, further studies will be necessary to confirm this hypothesis and to identify additional culture conditions for generating ventral pancreatic cells.
Finally, these results have important practical significance since protocols currently available to generate pancreatic cells from hPSCs often rely on feeders, Matrigel and serum, all of which represent a potential source of TGF-β signalling with the capacity to compromise pancreatic specification. Moreover, recent studies have shown that endogenous levels of nodal expression might determine the capacity of specific hIPSC lines to differentiate into mesodermal derivatives [34]. Such differences in the endogenous level of nodal/TGF-β growth factors could affect the capacity of diverse hPSC lines to differentiate into pancreatic endoderm, and the inhibition of this signalling pathway with SB could bypass this limitation. Accordingly, we recently differentiated ten hIPSC lines into pancreatic endoderm using our four-step protocol, and observed that only those hIPSC lines that failed to differentiate into DE (two out of ten) also lacked the ability to differentiate into pancreatic cells (M. Brimpari, University of Cambridge, Cambridge, UK and L. Vallier, personal observations).
Another advantage of inhibiting TGF-β signalling during pancreatic specification resides in the possibility of eliminating contaminating pluripotent cells. Indeed, we and others have extensively demonstrated that inhibition of activin/nodal/TGF-β signalling induces differentiation of hPSCs [35]. Thus, inhibition of activin during DE specification could decrease contamination by undifferentiated cells. Accordingly, we have failed to observe teratoma formation in mice transplanted with pancreatic progenitors. Therefore, inhibiting activin signalling during pancreatic specification could allow for the generation of ‘safer’ pancreatic progenitor for potential cell-based therapy.
To conclude, our study could greatly facilitate the production of homogenous populations of pancreatic and liver cells in defined culture conditions for clinical applications. However, this culture system also provides a robust and efficient in vitro model of development to study human endoderm differentiation.

Funding

This work was funded by Medical Research Council senior non-clinical fellowships (CH-HC, LV), the Cambridge Hospitals National Institute for Health Research Biomedical Research Center (LV), EU grant LivES (NR-FH), JDRF (KD, LV and CH-HC) and MOE Taiwan (CH-HC). Since contributing to this work, MWBT has become an employee of Celgene Research SLU (part of the Celgene Corporation)

Duality of interest

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

Contribution statement

CH-HC and NR-FH contributed equally to this study. All authors participated in concept and design of the study as well as collection, assembly and interpretation of data. CH-HC, NR-FH, KD and LV prepared the manuscript and all authors revised, edited and approved the final version of the manuscript.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

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.
ESM Fig. 1 Screening of defined culture conditions driving differentiation of DE cells toward the pancreatic lineages. (a-c) Basal media composition affects the capacity of DE cells to differentiate into PDX1 expressing cells. DE cells generated as described in Figure 1 were grown for 9 additional days in the presence of RA, SB, FGF10 and Noggin in diverse basal media (Advanced DMEM (Invitrogen); RPMI, CDM-PVA, CDM-BSA (CDM5)). (d-f) Effect of absence and increasing dose of RA (0.3 mM, 1.5 mM, 3 mM, 6 mM) on expression of pancreatic endoderm markers. (g-i) BMP inhibits differentiation of DE toward pancreatic endoderm. DE cells were grown for 6 days in the presence of RA, SB, FGF10 and BMP4 10 ng/ml (N0, B6) or RA, SB, FGF10 and Noggin (N6, B0). (j-l) FGF signalling is not necessary for differentiation of DE into pancreatic endoderm. DE cells were grown for 6 days in the presence of RA, SB, SU5402 (10 mM) and Noggin (6SU, 0F) or RA, SB, FGF10 and Noggin (0SU, 6F). (m-o) Increasing dose of FGF10 (5 ng/ml, 25 ng/ml, 50 ng/ml, and 100 ng/ml) does not affect the expression of pancreatic endoderm markers during DE differentiation. (p-af) Expression of pluripotency (POU5F1, NANOG, SOX2), primitive streak markers (Brachyury (T), MIXL1, EOMES), endoderm (SOX17, CXCR4, HEX), pancreatic endoderm markers (FOXA2, HNF6, SOX9) and endocrine markers (NGN3, PTF1a, SST, Glucagon (GLU)) during differentiation of hESCs into pancreatic endoderm using 4 step protocol described in Figure 1. (PDF 228 kb)
ESM Fig. 3 Differentiation of hIPSCs into pancreatic endoderm cells using culture conditions developed with hESCs. (a-d) Expression of pancreatic markers in hIPSCs differentiating into pancreatic endoderm using the 4 step protocol described in Figure 1. (hIPSC1 = A1ATD-hIPSCs, hIPSC2 = BBHX8, hIPSC3 = JRO1D). (e-i) Expression of pancreatic endoderm and Hormonal markers in hIPSCs grown respectively for 15 days and 18 days in culture conditions inductive for pancreatic specification. (j) FACS analyses showing the fraction of PDX1 expressing cells generated after 12 days of differentiation. (k-m) C-peptide secretion upon glucose stimulation in endocrine cells generated from hIPSCs. (PDF 512 kb)
Literatur
1.
Zurück zum Zitat Thomson JA, Itskovitz-Eldor J, Shapiro SS et al (1998) Embryonic stem cell lines derived from human blastocysts. Science 282:1145–1147PubMedCrossRef Thomson JA, Itskovitz-Eldor J, Shapiro SS et al (1998) Embryonic stem cell lines derived from human blastocysts. Science 282:1145–1147PubMedCrossRef
2.
Zurück zum Zitat Takahashi K, Tanabe K, Ohnuki M et al (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131:861–872PubMedCrossRef Takahashi K, Tanabe K, Ohnuki M et al (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131:861–872PubMedCrossRef
3.
Zurück zum Zitat D’Amour KA, Bang AG, Eliazer S et al (2006) Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 24:1392–1401PubMedCrossRef D’Amour KA, Bang AG, Eliazer S et al (2006) Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 24:1392–1401PubMedCrossRef
4.
Zurück zum Zitat Jiang W, Shi Y, Zhao D et al (2007) In vitro derivation of functional insulin-producing cells from human embryonic stem cells. Cell Res 17:333–344PubMedCrossRef Jiang W, Shi Y, Zhao D et al (2007) In vitro derivation of functional insulin-producing cells from human embryonic stem cells. Cell Res 17:333–344PubMedCrossRef
5.
Zurück zum Zitat Maehr R, Chen S, Snitow M et al (2009) Generation of pluripotent stem cells from patients with type 1 diabetes. Proc Natl Acad Sci USA 106:15768–15773PubMedCrossRef Maehr R, Chen S, Snitow M et al (2009) Generation of pluripotent stem cells from patients with type 1 diabetes. Proc Natl Acad Sci USA 106:15768–15773PubMedCrossRef
6.
Zurück zum Zitat Zhang D, Jiang W, Liu M et al (2009) Highly efficient differentiation of human ES cells and iPS cells into mature pancreatic insulin-producing cells. Cell Res 19:429–438PubMedCrossRef Zhang D, Jiang W, Liu M et al (2009) Highly efficient differentiation of human ES cells and iPS cells into mature pancreatic insulin-producing cells. Cell Res 19:429–438PubMedCrossRef
7.
Zurück zum Zitat Kroon E, Martinson LA, Kadoya K et al (2008) Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol 26:443–452PubMedCrossRef Kroon E, Martinson LA, Kadoya K et al (2008) Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol 26:443–452PubMedCrossRef
8.
Zurück zum Zitat Kelly OG, Chan MY, Martinson LA et al (2011) Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat Biotechnol 29:750–756PubMedCrossRef Kelly OG, Chan MY, Martinson LA et al (2011) Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat Biotechnol 29:750–756PubMedCrossRef
9.
Zurück zum Zitat Zaret KS, Grompe M (2008) Generation and regeneration of cells of the liver and pancreas. Science 322:1490–1494PubMedCrossRef Zaret KS, Grompe M (2008) Generation and regeneration of cells of the liver and pancreas. Science 322:1490–1494PubMedCrossRef
10.
Zurück zum Zitat Harrison KA, Thaler J, Pfaff SL, Gu H, Kehrl JH (1999) Pancreas dorsal lobe agenesis and abnormal islets of Langerhans in Hlxb9-deficient mice. Nat Genet 23:71–75PubMed Harrison KA, Thaler J, Pfaff SL, Gu H, Kehrl JH (1999) Pancreas dorsal lobe agenesis and abnormal islets of Langerhans in Hlxb9-deficient mice. Nat Genet 23:71–75PubMed
11.
Zurück zum Zitat Li H, Arber S, Jessell TM, Edlund H (1999) Selective agenesis of the dorsal pancreas in mice lacking homeobox gene Hlxb9. Nat Genet 23:67–70PubMed Li H, Arber S, Jessell TM, Edlund H (1999) Selective agenesis of the dorsal pancreas in mice lacking homeobox gene Hlxb9. Nat Genet 23:67–70PubMed
12.
Zurück zum Zitat Jonsson J, Carlsson L, Edlund T, Edlund H (1994) Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371:606–609PubMedCrossRef Jonsson J, Carlsson L, Edlund T, Edlund H (1994) Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371:606–609PubMedCrossRef
13.
Zurück zum Zitat Offield MF, Jetton TL, Labosky PA et al (1996) PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 122:983–995PubMed Offield MF, Jetton TL, Labosky PA et al (1996) PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 122:983–995PubMed
14.
Zurück zum Zitat Sherwood RI, Chen TY, Melton DA (2009) Transcriptional dynamics of endodermal organ formation. Dev Dyn 238:29–42PubMedCrossRef Sherwood RI, Chen TY, Melton DA (2009) Transcriptional dynamics of endodermal organ formation. Dev Dyn 238:29–42PubMedCrossRef
15.
Zurück zum Zitat Wandzioch E, Zaret KS (2009) Dynamic signaling network for the specification of embryonic pancreas and liver progenitors. Science 324:1707–1710PubMedCrossRef Wandzioch E, Zaret KS (2009) Dynamic signaling network for the specification of embryonic pancreas and liver progenitors. Science 324:1707–1710PubMedCrossRef
16.
Zurück zum Zitat Rashid ST, Corbineau S, Hannan N et al (2010) Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J Clin Invest 120:3127–3136PubMedCrossRef Rashid ST, Corbineau S, Hannan N et al (2010) Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J Clin Invest 120:3127–3136PubMedCrossRef
17.
Zurück zum Zitat Brons IG, Smithers LE, Trotter MW et al (2007) Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature 448:191–195PubMedCrossRef Brons IG, Smithers LE, Trotter MW et al (2007) Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature 448:191–195PubMedCrossRef
18.
Zurück zum Zitat Touboul T, Hannan NR, Corbineau S et al (2010) Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development. Hepatology 51:1754–1765PubMedCrossRef Touboul T, Hannan NR, Corbineau S et al (2010) Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development. Hepatology 51:1754–1765PubMedCrossRef
19.
Zurück zum Zitat Vallier L, Rugg-Gunn PJ, Bouhon IA, Andersson FK, Sadler AJ, Pedersen RA (2004) Enhancing and diminishing gene function in human embryonic stem cells. Stem Cells 22:2–11PubMedCrossRef Vallier L, Rugg-Gunn PJ, Bouhon IA, Andersson FK, Sadler AJ, Pedersen RA (2004) Enhancing and diminishing gene function in human embryonic stem cells. Stem Cells 22:2–11PubMedCrossRef
20.
Zurück zum Zitat Teo AK, Arnold SJ, Trotter MW et al (2011) Pluripotency factors regulate definitive endoderm specification through eomesodermin. Genes Dev 25:238–250PubMedCrossRef Teo AK, Arnold SJ, Trotter MW et al (2011) Pluripotency factors regulate definitive endoderm specification through eomesodermin. Genes Dev 25:238–250PubMedCrossRef
21.
Zurück zum Zitat Mfopou JK, Chen B, Mateizel I, Sermon K, Bouwens L (2010) Noggin, retinoids, and fibroblast growth factor regulate hepatic or pancreatic fate of human embryonic stem cells. Gastroenterology 138:2233–2245. e14PubMedCrossRef Mfopou JK, Chen B, Mateizel I, Sermon K, Bouwens L (2010) Noggin, retinoids, and fibroblast growth factor regulate hepatic or pancreatic fate of human embryonic stem cells. Gastroenterology 138:2233–2245. e14PubMedCrossRef
22.
Zurück zum Zitat Cai J, Yu C, Liu Y et al (2010) Generation of homogeneous PDX1(+) pancreatic progenitors from human ES cell-derived endoderm cells. J Mol Cell Biol 2:50–60PubMedCrossRef Cai J, Yu C, Liu Y et al (2010) Generation of homogeneous PDX1(+) pancreatic progenitors from human ES cell-derived endoderm cells. J Mol Cell Biol 2:50–60PubMedCrossRef
23.
Zurück zum Zitat Wells JM, Melton DA (2000) Early mouse endoderm is patterned by soluble factors from adjacent germ layers. Development 127:1563–1572PubMed Wells JM, Melton DA (2000) Early mouse endoderm is patterned by soluble factors from adjacent germ layers. Development 127:1563–1572PubMed
24.
Zurück zum Zitat Spence JR, Mayhew CN, Rankin SA et al (2011) Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470:105–109PubMedCrossRef Spence JR, Mayhew CN, Rankin SA et al (2011) Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470:105–109PubMedCrossRef
25.
Zurück zum Zitat Polak M, Bouchareb-Banaei L, Scharfmann R, Czernichow P (2000) Early pattern of differentiation in the human pancreas. Diabetes 49:225–232PubMedCrossRef Polak M, Bouchareb-Banaei L, Scharfmann R, Czernichow P (2000) Early pattern of differentiation in the human pancreas. Diabetes 49:225–232PubMedCrossRef
26.
Zurück zum Zitat Habener JF, Kemp DM, Thomas MK (2005) Minireview: transcriptional regulation in pancreatic development. Endocrinology 146:1025–1034PubMedCrossRef Habener JF, Kemp DM, Thomas MK (2005) Minireview: transcriptional regulation in pancreatic development. Endocrinology 146:1025–1034PubMedCrossRef
27.
Zurück zum Zitat Ameri J, Stahlberg A, Pedersen J et al (2010) FGF2 specifies hESC-derived definitive endoderm into foregut/midgut cell lineages in a concentration-dependent manner. Stem Cells 28:45–56PubMed Ameri J, Stahlberg A, Pedersen J et al (2010) FGF2 specifies hESC-derived definitive endoderm into foregut/midgut cell lineages in a concentration-dependent manner. Stem Cells 28:45–56PubMed
28.
Zurück zum Zitat Nostro MC, Sarangi F, Ogawa S et al (2011) Stage-specific signaling through TGF{beta} family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development 138:861–871PubMedCrossRef Nostro MC, Sarangi F, Ogawa S et al (2011) Stage-specific signaling through TGF{beta} family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development 138:861–871PubMedCrossRef
29.
Zurück zum Zitat Kunisada Y, Tsubooka-Yamazoe N, Shoji M, Hosoya M (2012) Small molecules induce efficient differentiation into insulin-producing cells from human induced pluripotent stem cells. Stem Cell Res 8:274–284PubMedCrossRef Kunisada Y, Tsubooka-Yamazoe N, Shoji M, Hosoya M (2012) Small molecules induce efficient differentiation into insulin-producing cells from human induced pluripotent stem cells. Stem Cell Res 8:274–284PubMedCrossRef
30.
Zurück zum Zitat Brown S, Teo A, Pauklin S et al (2011) Activin/nodal signaling controls divergent transcriptional networks in human embryonic stem cells and in endoderm progenitors. Stem Cells 29:1176–1185PubMedCrossRef Brown S, Teo A, Pauklin S et al (2011) Activin/nodal signaling controls divergent transcriptional networks in human embryonic stem cells and in endoderm progenitors. Stem Cells 29:1176–1185PubMedCrossRef
31.
Zurück zum Zitat Kubo A, Kim YH, Irion S et al (2009) The homeobox gene Hex regulates hepatocyte differentiation from embryonic stem cell-derived endoderm. Hepatology 51:633–641CrossRef Kubo A, Kim YH, Irion S et al (2009) The homeobox gene Hex regulates hepatocyte differentiation from embryonic stem cell-derived endoderm. Hepatology 51:633–641CrossRef
32.
Zurück zum Zitat Inamura M, Kawabata K, Takayama K et al (2011) Efficient generation of hepatoblasts from human ES cells and iPS cells by transient overexpression of homeobox gene HEX. Mol Ther 19:400–407PubMedCrossRef Inamura M, Kawabata K, Takayama K et al (2011) Efficient generation of hepatoblasts from human ES cells and iPS cells by transient overexpression of homeobox gene HEX. Mol Ther 19:400–407PubMedCrossRef
33.
Zurück zum Zitat Bort R, Martinez-Barbera JP, Beddington RS, Zaret KS (2004) Hex homeobox gene-dependent tissue positioning is required for organogenesis of the ventral pancreas. Development 131:797–806PubMedCrossRef Bort R, Martinez-Barbera JP, Beddington RS, Zaret KS (2004) Hex homeobox gene-dependent tissue positioning is required for organogenesis of the ventral pancreas. Development 131:797–806PubMedCrossRef
34.
Zurück zum Zitat Ramos-Mejia V, Melen GJ, Sanchez L et al (2010) Nodal/activin signaling predicts human pluripotent stem cell lines prone to differentiate towards the hematopoietic lineage. Mol Ther 18:2173–2181PubMedCrossRef Ramos-Mejia V, Melen GJ, Sanchez L et al (2010) Nodal/activin signaling predicts human pluripotent stem cell lines prone to differentiate towards the hematopoietic lineage. Mol Ther 18:2173–2181PubMedCrossRef
35.
Zurück zum Zitat Vallier L, Mendjan S, Brown S et al (2009) Activin/nodal signalling maintains pluripotency by controlling Nanog expression. Development 136:1339–1349PubMedCrossRef Vallier L, Mendjan S, Brown S et al (2009) Activin/nodal signalling maintains pluripotency by controlling Nanog expression. Development 136:1339–1349PubMedCrossRef
Metadaten
Titel
Inhibition of activin/nodal signalling is necessary for pancreatic differentiation of human pluripotent stem cells
verfasst von
C. H.-H. Cho
N. R.-F. Hannan
F. M. Docherty
H. M. Docherty
M. Joåo Lima
M. W. B. Trotter
K. Docherty
L. Vallier
Publikationsdatum
01.12.2012
Verlag
Springer-Verlag
Erschienen in
Diabetologia / Ausgabe 12/2012
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-012-2687-x

Weitere Artikel der Ausgabe 12/2012

Diabetologia 12/2012 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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