Skip to main content
Erschienen in: Journal of Translational Medicine 1/2007

Open Access 01.12.2007 | Review

Pancreatic islet cell therapy for type I diabetes: understanding the effects of glucose stimulation on islets in order to produce better islets for transplantation

verfasst von: Jiaqiang Ren, Ping Jin, Ena Wang, Eric Liu, David M Harlan, Xin Li, David F Stroncek

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2007

Abstract

While insulin replacement remains the cornerstone treatment for type I diabetes mellitus (T1DM), the transplantation of pancreatic islets of Langerhans has the potential to become an important alternative. And yet, islet transplant therapy is limited by several factors, including far too few donor pancreases. Attempts to expand mature islets or to produce islets from stem cells are far from clinical application. The production and expansion of the insulin-producing cells within the islet (so called β cells), or even creating cells that secrete insulin under appropriate physiological control, has proven difficult. The difficulty is explained, in part, because insulin synthesis and release is complex, unique, and not entirely characterized. Understanding β-cell function at the molecular level will likely facilitate the development of techniques to manufacture β-cells from stem cells. We will review islet transplantation, as well as the mechanisms underlying insulin transcription, translation and glucose stimulated insulin release.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1479-5876-5-1) contains supplementary material, which is available to authorized users.
Abkürzungen
bHLH
basic helix-loop-helix proteins
eIF2
Eukaryotic initiation factor 2
Glut 2
glucose transporter 2
IEGs
Immediate-early response genes
KATP-channels
ATP-sensitive potassium channels
L-VDCC
L-type voltage-dependent Ca2+channel
PDX-1
pancreas-duodenum homeobox-1
PERK
PKR-like ER kinase
PI-3-K
phosphatidylinositol (PI) 3-kinase
RRP
readily releasable pool
UTR
untranslated regions
4E-BPs
eIF4E-binding proteins

Background

Insulin, the body's only blood glucose-lowering hormone, is exclusively produced by the β-cells of the pancreatic islets of Langerhans. Type I diabetes mellitus (T1DM), formerly known as insulin-dependent diabetes or juvenile diabetes, affects an estimated 1,000,000 Americans, and is thought to result from the destruction of β cells by autologous cytotoxic T cells. The discovery of insulin in the 1920's was a major advance in diabetes treatment – freeing patients from the then "state of the art" starvation diets that, at best, prolonged survival for a few weeks, months, or in very rare cases, a few years. Insulin therapy changed diabetes from a rapidly fatal disease to a chronic disease associated with significant secondary complications, such as renal failure, neuropathy, cardiovascular disease, and retinopathy. While aggressive insulin therapy that maintains glucose levels near the normal range reduces the risk of secondary complications, patients often find such control difficult to achieve and suffer an increased risk of hypoglycemia [1]. Nonetheless, T1DM treatment has significantly improved with affordable glucose monitoring instruments, new insulin formulations, and improved insulin delivery systems.

Transplantation

Many clinical investigators have attempted β-cell replacement therapy, either in the form of whole pancreas or islet transplantation. Whole pancreas transplantation was first performed in the 1960s, but was a relatively uncommon procedure until the 1980's. From December 16, 1966 to December 31, 2004, more than 23,000 pancreas transplants were reported to the International Pancreas Transplant Registry (IPTR), including > 17000 from the US and almost 6000 from outside the US. The annual number of US in 2004 was close to 1500 [2]. The procedure's increasing popularity can be ascribed to improvements in organ preservation, surgical technique, and immunosuppressive therapy.
Patients given a pancreas transplant, at experienced centers, currently can anticipate a greater than 85% likelihood that they will enjoy insulin independent euglycemia 1 year later, and about 50% will maintain that excellent metabolic outcome 5 years following the transplant. While most pancreas transplant recipients no longer require exogenous insulin for blood glucose control, the procedure has not been shown to decrease the severity or frequency of the secondary complications associated with diabetes. For instance, while pancreas transplant has been shown to reverse histologically defined diabetic nephropathy ten years after transplantation, only a minority of grafts survive that long, and the immunosuppression given to preserve the transplanted organ may be more nephrotoxic than the diabetes prompting the transplant [3]. In fact, whether pancreas transplant alone offers any survival advantage versus insulin therapy alone is debated, and transplant recipient survival may even be worse [4, 5].
While pancreas transplantation has yielded promising results, other investigators have worked to develop isolated islet transplantation. Islet transplantation is potentially appealing because the transplant technique does not require major surgery. Using animal models in the 1970s, islets were first isolated from the pancreas and effectively transplanted [6]. Preliminary human islet cell transplant reports appeared in the late 1970's [7, 8]. Islet transplantation was not used clinically to treat type I diabetes until 1989 [9].
Although successful procedure of living-donor transplantation of islets has been reported [10], in its current status, most islet transplants involve the isolation of islets from cadaveric (deceased) donor organs. The isolation process entails both enzymatic (using collagenase) and mechanical disruption of a cadaveric pancreas into small fragments. Islets are then purified from the remaining exocrine tissue by density gradient separation [11]. Once "purified" (and isolated islets should more accurately be termed "enriched" because exocrine pancreatic fragments are also nearly always present), the islets are infused into the recipient's portal vein where they lodge in the liver's portal vein tree. Islets have been introduced into the portal circulation through the umbilical vein, but percutaneous transhepatic angiographic infusion is now used most frequently. Isolated islets are usually infused within 48 hours (some centers infuse them within hours), and most recipients require islets from more than one cadaver. The islets isolated from one cadaver are typically given with each infusion, such that most islet recipients end-up requiring two or more islet infusions. While islet infusions are generally well tolerated, they can be complicated by (at least temporary) portal hypertension, thrombosis or hemorrhage.
Before 2000, the preceding 2 decades of islet transplantation by investigators world-wide, had achieved 1-year insulin independence rates of less than 12% [12]. However, in 2000 the group from the University of Alberta, Edmonton reported that their protocol for islet transplantation and immunosuppression significantly improved graft function [13]. The Edmonton protocol used a steroid-free immunosuppressive regimen and high quality islet cells from 2 or more donors. The quality of islet cells obtained from each cadaver was improved by limiting cold ischemia time to 8 hours prior to initiating the isolation procedure and using a standardized collagenase preparation. Further, the Edmonton group transplanted the isolated islets, as soon as possible following the isolation, via transhepatic angiography. To prevent islet allograft rejection, the Edmonton team avoided glucocorticoids by giving the recipient induction immunosuppression with the anti- IL-2 receptor antibody (daclizumab), then more chronic immunosuppression was initiated with sirolimus (rapamycin) and tacrolimus (FK-506) [14, 15]. Recently, an international, multicenter trial tested whether islet transplantation using the Edmonton protocol could be generalized. From more than 2000 with T1DM expressing an interest in the protocol, 36 patients were selected and given allogeneic islets. Sixteen of the 36 (44%) attained the primary end point of insulin independence (defined as a fasting blood glucose not to exceed 140 mg/dl more than 3x/week, and a 2 hour post-prandial blood glucose not to exceed 180 mg/dl more than 4x/week), 28% had partial graft function and 28% had complete graft loss. While many of the insulin independent subjects did not have normal blood glucose values by currently accepted criteria, the subjects who reached the primary end point had no severe hypoglycemia or severe hyperglycemia, and those with partial function had a marked benefit in glycemia control in contrast to their baseline status [16]. Unfortunately, and for reasons not well understood, only 14% of the subjects remained insulin independent 2 years after receiving an islet transplant. Further, the current immunosuppressive regimen is associated with heightened risk of infection, β cell toxicity, and (most importantly) nephropathy such that the protocol participants lost, on average about 5% of their kidney function each year.

Alternative sources of β Islets

While pancreas and islet cell transplants have the potential to treat many patients with diabetes, these transplants require the collection of pancreata from organ donors [13, 17], and donors are very limited. Optimistic estimates suggest that in a typical year and using current techniques, islets isolated form U.S. cadaveric pancreata could transplant at most 1,000 to 2,000 patients [14]. In order to treat more patients, the field needs an alternative source of cells capable of physiologically regulated insulin secretion. Transplanting cells generated from stem cells is one potential treatment alternative [18] and another is the expansion adult islet β cells, xenogeneic islet is considered another alternative source.
While a stem cell that could be isolated from the pancreas, expanded, and differentiated in vitro into mature beta-like cells may exist, such a cell has not yet been identified [19], and culture techniques have not been perfected. If such a cell could be isolated from the pancreas and expanded in vitro, it is possible that enough islets could be produced from a pancreas from a single donor for one or more successful transplants.
Another alternative is to produce β cells from stem cells. Several groups have attempted to differentiate embryonic stem cells into β cells, but none has yet been successful. Islet-like cells have been generated from embryonic stem cells, but the cells do not secrete insulin in a physiologically regulated fashion [20]. In vitro differentiation of embryonic stem cells is to mimic normal embryonic development, the exposure of stem cells to growth factors, extracellular matrix components, and cell-cell interactions may promote and streamline the differentiation process. The existing protocols for generating insulin-producing cells from embryonic stem cells can be divided into spontaneous differentiation and induced differentiation. The efficiency of the former protocol is too low to be of practical value [2123] and induced differentiation is the mainstream. Lumelsky modified a protocol used to generate neurons from mouse embryonic stem cells and obtained insulin-producing cells [24], their results were repeated by other groups [25, 26]. However, some reports showed that the cells can not produce insulin themselves, but rather absorb insulin from the culture medium while undergoing apoptosis [27]. Recently, D'Amour developed a five-stage protocol to differentiate human embryonic stem cells to endocrine hormone-expressing cells through a series of endodermal intermediates resembling those that occur during pancreatic development in vivo [28]. Despite the facts, the procedure of pancreas development is so complex and precise that many aspects are not well understood now. Fortunately, advances in high-throughout technology such as microarray technology will facilitate the discovery of regulating network of pancreatic development, which will provide more information to better direct differentiating stem cells.
Adult stem cells are another potential source of islets [18]. Some animal hematopoietic transplant models have suggested that adult pluripotential stem cells from bone marrow can be induced to transdifferentiate into β-like cells, but subsequent studies have suggested that the cells were the result of cell-to-cell fusion rather than transdifferentiation [29]. It may also be possible to generate insulin producing cells using gene therapy. Some studies have suggested that transfecting hepatocytes with the gene encoding PDX-1 resulted in an insulin producing cell, but those results have not been widely reproduced [3032].
Xenogeneic islets is another alternative; a couple of papers were recently published in Nature Medicine showing that porcine islets xenografts can succeed and has potential for clinical application [33, 34]. However, xenogeneic transplants take major immunosuppression which significantly outweighs the risk of diabetes and the results were not consistent, several of the grafts failed early.
While all of these potential therapies are promising, none is yet useful clinically. Furthermore, since β cell insulin release to control glucose levels is both very complex and precise, it is likely that any cultured, expanded, or otherwise manufactured cells will have to function with β cell-like precision. We suggest therefore that it will not be possible to create highly effective expanded, cultured or manufactured β cells until the mechanisms responsible for glucose-induced β cell insulin synthesis and release are completely understood and these functions can be duplicated.

Insulin transcription, translation, and release

The production of insulin and its release from islets is complex and tightly regulated. Glucose affects insulin at all levels, including transcription, translation and release.

Transcription

Insulin is encoded by the insulin gene located on chromosome 11p15.5 [35]. Insulin expression is restricted to the β cell and insulin transcription is controlled by the insulin promoter, and in particular a highly conserved 340 bp region located immediately upstream from the transcription initiation start site [36]. The insulin promoter is responsible for tissue specific and metabolic regulation of the insulin gene. The most critical transcription activation elements of the insulin promoter are the A3, C1 and E1 sites.
The most important insulin transcription factor is the homeodomain transcription factor pancreatic/duodenal homebox-1 (PDX-1). In the adult islet, PDX-1 is only expressed in the pancreatic β cells and plays an important role in glucose-stimulated insulin gene transcription. PDX-1 is also essential for maintenance of the β cell phenotype and pancreatic development [36, 37]. PDX-1 binds to the A3 box of the insulin promoter. Alone PDX-1 has little activity, but it becomes a potent factor when it interacts with the heterodimer of basic helix-loop-helix (bHLH) proteins which bind to the E1 box. These bHLH heterodimers are made up of a Class A bHLH protein which is expressed ubiquitously in many different tissues and a Class B bHLH protein that is specifically expressed in β cells. The members of the class A bHLH family in the heterodimer that binds to the E1 site are products of the E2A gene; E12, E47, and E2/5 and the class B bHLH protein BETA2/NeuroD. The heterodimer E47/BETA2 is found in β cells.
Another important insulin transcription factor is MafA which belongs to the Maf family of transcription factors. This family of transcription factors contains a basic motif followed by a leucine zipper. In addition, MafA also contains an acidic domain that acts as a transcription initiation domain [38]. MafA binds to the C1 site of the insulin promoter. MafA is a glucose-regulated and pancreatic β cell-specific transcriptional activator for the insulin gene [3941], which produces synergistic activation with PDX-1 or BETA2.
In addition to PDX1, E47/BETA2/NeuroD, and MafA, other factors influence insulin transcription. Histones and DNA-binding proteins of the high mobility group (HMG) increases the binding of PDX-1 and bHLH heterodimers to the A and E sites. One important HMG protein is HMG 1(Y) which binds to the A3/A4 site of the insulin promoter [37]. Other proteins that contribute to insulin transcription include members of the hepatic nuclear factors and PAX families [36] (Figure 1).

Insulin synthesis

Translating the insulin mRNA leads to preproinsulin production. Approximately 30 to 60 seconds after preproinsulin is synthesized in the ER, the pre portion is removed enzymatically and proinsulin is transported along the microtubule network system in transport vesicles to the cis part of the Golgi apparatus. The proinsulin was first packaged into clathrin-coated immature granules, where proinsulin is further converted to insulin and C-peptide [42]. The clathrin-coated granules then become mature granules, where the insulin crystals are formed; thus the insulin is stored in the mature secretory granules until it is either released by exocytosis or degraded by crinophagy [43].
The β cell has highly developed ER specialized for the synthesis of insulin. β Cells can control the rate of insulin production by regulating insulin synthesis in the ER in response to glucose stimulation. Eukaryotic initiation factor 2 (eIF2) is an important factor regulating protein biosynthesis. The eIF2 factor participates in the formation of translational ternary complex (eIF2-GTP·Met-tRNAi), recruiting charged initiator methionyl-tRNA to the 40S ribosomal subunit. The activity of the eIF2 complex is dependent on its state of phosphorylation. Pancreatic ER kinase (PERK), an important regulator of insulin translation in β cells, phosphorylates eIF2 complex and hence lowers insulin translation [44]. PERK activity is sensitive to glucose levels [45]. Therefore, PERK signaling is particularly important to normal β cell function.
The initiation complex eIF4F initiates the recruitment of the 40S ribosome to mRNA in insulin synthesis. Hypophosphorylated eIF4E-binding proteins (4E-BPs) inhibit eIF4F complex formation, but phosphorylation of 4E-BP1 leads to loss of this inhibition, therefore inducing an increase in mRNA translation [46, 47].

Insulin release

Nearly all insulin released by β cells is from insulin secretory granules. To release insulin, the granules must be recruited from the cytoplasm, translocated to the plasma membrane where they are docked, fuse with the plasma membrane, and release their contents into the extracellular space. A group of proteins known as the SNAp REceptors (SNARES) are important for directing the insulin vesicles to the plasma membrane. The actual docking of the vesicle with the plasma membrane involves the linking of the plasma membrane proteins syntaxin and synaptosomal-associated protein 25 (SNAP-25) with the vesicle protein vesicle-associated protein 2 (VAMP-2) or synaptobrevin-2. Syntaxin and SNAP-25 are known as t-SNARE and VAMP-2 is known as v-SNARE.
Insulin granule secretion in response to glucose stimulation exhibits two characteristic phases. This biphasic pattern consists of a rapidly initiated, but transient first phase of insulin release, and a sustained second phase [4850]. β Cells contain two pools of insulin containing secretory granules that have distinct release processes. A limited pool of granules (< 5%) is available for immediate release and is referred to as the "readily releasable pool" (RRP). However, most of the insulin granules (> 95%) belong to a reserve pool and must undergo mobilization before they can gain release competence [5153]. The release of RRP granules accounts for the first phase of insulin secretion. The end of the first phase marked the depletion of this pool. The subsequent granule re-supply or mobilization from a reserve pool of granules and release of these mobilized granules is responsible for the second-phase of insulin secretion [54] (Figure 2).

Effects of glucose on insulin transcription, translation and release

Glucose controls all aspects of insulin regulation. Glucose is the major physiologic regulator of insulin transcription, translational regulation of insulin biosynthesis, and insulin secretion. The response of β cells to glucose is complex. Glucose has both immediate and long term effects that are mediated on several levels including granule release, protein translation and mRNA transcription. β Cells have large stores of insulin in granules and immediately following glucose stimulation a small proportion of these granules are released. To renew these stores, insulin biosynthesis starts immediately and during this period after glucose stimulation glucose-induced insulin biosynthesis is regulated mainly at the translational level [55, 56]. During this period glucose metabolism may be coupled to the transcriptional activation of immediate-early response factors. During periods of prolonged glucose stimulation (> 12 h), glucose increases insulin biosynthesis by accelerating insulin gene transcription, as well as by increasing preproinsulin mRNA stability and protein translation.

Glucose control of insulin secretion

The mechanism by which glucose triggers insulin secretion has been well established. β-cells do not release insulin in response to glucose itself, but to glucose metabolism [57]. Glucose enters β-cells via the GLUT 2 transporter. Intracellular glucose is then metabolized to generate ATP. This results in an increase in the cytosolic ATP: ADP ratio which in turn results in the closure of KATP-channels, membrane depolarization, and initiation of electrical activity [58].
Intracellular Ca2+ signaling plays a critical role in the regulation glucose-mediated insulin secretion. The depolarization of the plasma membrane by the closure of the KATP channels allows the opening of the L-type voltage dependent Ca2+-channels (L-VDCC) which results in an increase in the influx of calcium and an increase in the intracellular Ca2+ concentration, an event that causes exocytosis of the insulin granules [52]. This ATP-sensitive K+ (KATP) insulin release pathway is also known as the triggering pathway.
SNARE proteins play a critical role in insulin granule secretion [59, 60]. The granule protein, v-SNARE, and the β cell plasma membrane protein, t-SNARE, bring insulin granules in close contact with the plasma membrane and plasma membrane Ca 2+-channels [61]. When glucose stimulation leads to the closure of KATP channels and the opening of the Ca 2+-channels and the RRP granules located just beneath the inner mouth of Ca 2+-channels are exposed to high levels of Ca2+ causing RPR granule release or exocytosis. The release of insulin RPR granules proceeds in an essentially all-or-none fashion depending on whether the Ca 2+-channels are open or not [53, 54, 62, 63].
Glucose also affects calcium influx via cyclin-dependent kinase 5 kinase (Cdk5). Cdk5 phosphorylates the α1C subunit of L-VDCC which decreases its activity by preventing the binding of L-VDCC to the SNARE proteins. The activity of CdK5 kinase is dependent on glucose. High glucose concentrations inhibit the Cdk5 kinase activity which in turn increases the inward whole-cell Ca2+ channel current and increases Ca2+influx, leading to enhanced insulin secretion [64].
Glucose not only triggers insulin secretion but also amplifies it. The amplifying effect of glucose on insulin release is known as the augmentative pathway. This pathway does not cause insulin secretion by itself, but it enhances Ca2+ mediated secretion. The augmentation pathway is independent of KATP-channel inhibition. In fact, changes in ADP concentration mediate the amplifying effect while ATP represents a permissive factor [65, 66].
Before insulin secretory granules can be released, they must be primed or acidified. Insulin granule priming is dependent on the simultaneous operation of a V-type H+-ATPase and ClC-3 Cl- channels. Cl- ion uptake determines the extent of granular acidification by providing a counter-ion required to allow continuous H+ pumping. The activity of the ClC-3 Cl- channels is inhibited by high concentration of ADP. As a result of glucose metabolism, the ADP level is decreased while ATP level is increased, leading to granular acidification. Acidification is a prerequisite for insulin secretion and only after acidification, can granules undergo exocytosis whenever Ca 2+ influx increases to exocytotic levels and thus insulin secretion is amplified [67, 68] (Figure 3).

Glucose control of insulin transcription

The β-cells contain a large pool of cytoplasmic insulin mRNA which makes up 10 to15% of all the β cells' total mRNA [69]. Even at low glucose concentrations large quantities of insulin mRNA are present, however, at low plasma glucose concentrations (< 3 mM) the insulin mRNA reservoirs are due to basal insulin gene transcription. At higher glucose concentrations insulin transcription increases. For example, the mouse insulin gene promoter is transcriptionally active in the absence of glucose, but the addition of glucose stimulates transcription about 3-fold after 10 minutes, and the stimulatory effect is most pronounced 30 minutes after the glucose stimulus, but declines thereafter [70]. However, the regulation of insulin mRNA production is complex. No one factor completely controls insulin gene expression and transcription continues even when components involved in the stimulus-dependent up-regulation of insulin gene transcription are blocked or even knocked-out.
In addition to increasing the rate of insulin mRNA transcription, glucose also prolongs the half-life of preproinsulin mRNA. The half-life of preproinsulin mRNA in cells incubated in 17 mM of glucose is 76.8 hours, however, the half-life of those islets incubated in 3.3 mM is 29.0 hours [71]. Other studies have found that in rat islets lowering glucose from 11 mM to 2 mM decreased insulin mRNA levels by 38%, 79% and 66% at 3, 6 and 12 hours respectively [72]. Another study found that the content of preproinsulin mRNA in islets cultured at 3.3 mM glucose was reduced to 10% of the control islets after 24 hours and remained at that level for up to 7 days of incubation [73]. This suggests that the lower glucose concentrations may increase the proinsulin mRNA degradation rate.
The proinsulin mRNA 5' and 3' untranslated regions (UTR) also act cooperatively to increase glucose-induced preproinsulin biosynthesis [74]. Elements within the 3' UTR stabilize the mRNA and those within the 5' UTR stimulate preproinsulin translation. In addition, insulin gene 5'-untranslated region alternative splicing has been found in isolated human pancreatic islets, and the alternatively sliced mRNA is translated twice as efficiently (in vitro) as the native proinsulin mRNA. Twenty-four hours of hyperglycemia increases the expression of the alternatively spliced form by more than 2-fold and 72 hours of hyperglycemia increases it by more than 10-fold, suggesting that prolonged exposure to high glucose concentrations may indirectly affect the insulin splicing process and lead to the preferential use of the cryptic splice site [75].
The effect of short-term glucose stimulation on preproinsulin transcription is controversial. Isolated rat islets exposed to 16.7 mM glucose for 1 hour and 3 hours did not alter proinsulin mRNA levels, however, longer incubations (6, 12, and 24 hours)increased proinsulin mRNA levels 1.7-, 2.2-, and 2.6-foldi [76]. Similarly, incubating rat islets for 1 hour in 16.7 mM glucose, compared to 2.8 mM glucose, resulted in similar preproinsulin mRNA levels [77]. However, another study of rat islets found that proinsulin biosynthetic rates increased within 1 hour of exposure to high glucose concentrations, with fifty percent of this increase attributed to preproinsulin gene transcription. Furthermore, these investigators found that the glucose stimulatory effect was short-lived; the transcriptional activity was maximal at 30 minutes but markedly decreased thereafter [78].
The apparently contradictory results may be due to the dynamics of newly synthesized proinsulin mRNA, which undergoes faster degradation than pre-existing proinsulin mRNA [78]. Perhaps during 1-hour glucose stimulation, newly synthesized preproinsulin mRNA is degraded so rapidly that changes in preproinsulin mRNA levels are not detectable. Another potential explanation is that although glucose-induced insulin transcription can start within minutes, the preproinsulin mRNA levels accumulate slowly against the high background of steady-state mRNA.
Many factors contribute to glucose-induced insulin transcription, including the transcription factors PDX-1 [79], MafA [39, 80, 81], and a heterodimer (E12/E47 and E2/5) and (BETA 2/NeuroD) [82, 83] with the main glucose-responsive elements on the insulin promoter, the A3 [84], C1 and E1 [85] boxes. Insulin gene transcription is regulated through the activation of these factors and in combination, these factors can exert strong synergistic effects [37, 8688]. The upstream signaling that regulates these factors begins with β cell glucose metabolism.
The most important effects of glucose on insulin translation appear to be mediated by PDX-1. Glucose effects PDX-1 function in many different ways including a shift in the cellular distribution of PDX-1 from the cytoplasm to the nucleus, increases in the potential of the PDX-1 activation domain, and increases PDX1 binding to A3 [8994]. The effects of glucose on PDX-1 are, in part, due to the phosphorylation of PDX-1 through the activation of phosphatidylinositol 3-kinase (PI3-K), while stress-activated protein kinase 2 (SAPK2/P38) may also be involved in this process [95], its function is controversial [96]. In fact, the PI3-K kinase pathway is considered a central regulator of PDX-1 and of glucose-induced insulin gene transcription [95, 96]. Since PI3-K is a key molecule in the insulin receptor pathway, the activation of insulin gene transcription has been suggested, in part, to be the result of a feed forward mechanism involving the binding of secreted insulin to its receptor on the β cell surface [9799].
Glucose and PDX-1 also modulate insulin transcription by influencing histones. When glucose levels are low, PDX-1 interacts with histone deacetylases Hdac-1 and Hdac-2, recruiting them to the insulin gene promoter where they deacetylate histone H4 and thereby down-regulate insulin gene expression [100].
Glucose also increases bHLH heterodimer binding to the E site. E47/BETA 2 is critical for glucose-induced insulin gene transcription [101103]. Stimulatory glucose concentrations can activate β cell ERK1/2, and ERK1/2 promotes BETA 2 and E47 heterodimerization and binding to E-box sites [104]. Blocking ERK1/2 activity using kinase-defective ERK2 resulted in a substantial reduction in preproinsulin mRNA content after 24 hours[104]. Both the basal activity and the glucose-induced stimulation of the A2-E1 region of the insulin promoter are critically dependent upon ERK1/2 [105].
The mechanism by which MafA is activated by glucose is distinct. While glucose induces post translation modifications of PDX-1 and the bHLH heterodimer, glucose increases MafA transcription. At a glucose concentration of 2.8 mM, MafA expression is undetectable in whole β cell lysates. In contrast, at concentration of 16.7 mM MafA protein is readily detected. Exposing islets to 16.7 mM glucose for 24 hours also significantly increases MafA mRNA expression [89]. (Figure 4).
Immediate-early response genes (IEGs) are also involved with glucose mediated insulin biosythesis. IEGs are transcription factors that activate expression of downstream target genes, thus generating distinct biological responses by inducing specific long-term gene expression programs. In INS-1 β-like cells, glucose induces a Ca2+-dependent transcriptional activation of several immediate early genes such as c-fos, c-jun, JunB, zif-268 and nur-77 genes. In particular, c-fosand JunB proteins might facilitate glucose/cAMP mediated insulin gene induction [106]. 15 minutes of glucose stimulation induced egr-1 mRNA and protein synthesis, with maximum levels achieved in 30 minutes, in both glucose-responsive cell lines as well as primary islets. Glucose stimulation also induced expression of c-fos mRNA and JunB mRNA [107]. Thus, glucose metabolism could be coupled to transcriptional activation of immediate-early response factors.
Prolonged glucose stimulation leads to an increase in insulin transcription, which is supplementary to the glucose-induced translational control of proinsulin biosynthesis [77, 108]. Under conditions of sustained secretory drive, stimulation of proinsulin gene transcription by glucose appears to be necessary for maintaining preproinsulin biosynthesis and hence conserving pancreatic insulin stores [108].

Glucose control of translation

After insulin is secreted in response to glucose stimulation, renewed insulin biosynthesis begins immediately to replenish the insulin stores. After short term (less than 2 hours) glucose stimulation, insulin biosynthesis is mainly regulated at the translational level [55, 109]. When primary rat islets are stimulated by high glucose for 1 hour, preproinsulin biosynthesis increases 4-5 fold without any change in the total preproinsulin mRNA level [77]. In another study, preproinsulin biosynthesis increased 25-fold when glucose increased from 1 to 10 mM [110].
The mechanism by which glucose regulates proinsulin synthesis in β-cells remains unclear. It was previously hypothesized that the insulin protein production increase resulted from increased translation initiation, elongation and signal recognition particle (SRP) release. Evidence has shown however that translation elongation occurs only at non-physiological glucose concentrations [56, 111, 112] and does not play a major role in glucose-stimulated insulin synthesis. Translation initiation is now thought to be the primary mechanism underlying glucose stimulated insulin synthesis.
Factors involved with glucose regulation of preproinsulin translation initiation are the translation initiation factors eIF4F, eIF2 and PERK [113, 114]. The initiation complex eIF4F initiates the recruitment of the 40 S ribosome prerequisite to mRNA in protein synthesis. Hypophosphorylated eIF4E-binding proteins (4E-BPs) inhibit eIF4F complex formation but phosphorylation of 4E-BP1 decreases this inhibition, thereby increasing mRNA translation [46, 47]. In primary islets and pancreatic β-cell lines, glucose increases 4E-BP1 phosphorylation [113], suggesting the eIF4F assembly importance in glucose-stimulated protein synthesis.
Eukaryotic initiation factor 2 (eIF2) is another important factor regulating protein biosynthesis, participating in translational ternary complex (eIF2-GTP·Met-tRNAi) formation. This complex assembles only if eIF2 is in its GTP-bound state. After the formation of the 80S ribosome, the GTP bound to eIF2 is then hydrolyzed to GDP. The recycling of the inactive eIF2·GDP complex to the active eIF2·GTP complex is catalyzed by the guanine nucleotide exchange factor, eIF2B. eIF2B activity is transiently up-regulated in glucose stimulated isolated islets [111], suggesting that eIF2B may be an important modulator of insulin synthesis. Other studies suggest a role for the eIF2 alpha subunit (eIF2α); phosphorylation of eIF2α inhibits ternary complex formation, and glucose dephosphorylateseIF2α thereby increasing translational ternary complex availability [114] (Figure 5).

Other genes and proteins affected by glucose

9Glucose also induces delayed long-term responses by inducing the expression of other genes involved in β-cell function such as glucose transporter 2 (GLUT-2), pyruvate kinase, acetyl-coenzyme A-carboxylase [115118]. The expression of these genes is necessary to meet the increased metabolic and secretory demands during extended or repeated periods of hyperglycemia.

Conclusion

Islet transplantation has the potential to benefit patients with type I diabetes, but it is a therapy limited by islet supply and other factors. While preliminary studies have found that stem cells can be induced to acquire a β-cell-like phenotype,, cells used for transplant therapy will need precisely replicate β-cell function. Characterizing the β-cell's unique molecular mechanisms underlying its glucose responsivity will allow investigators to better understand the critical elements stem cells must acquire and may allow investigators to better direct stem cell differentiation.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Literatur
1.
Zurück zum Zitat Steffes MW, Sibley S, Jackson M, Thomas W: Beta-cell function and the development of diabetes-related complications in the diabetes control and complications trial. Diabetes Care. 2003, 26 (3): 832-836.PubMed Steffes MW, Sibley S, Jackson M, Thomas W: Beta-cell function and the development of diabetes-related complications in the diabetes control and complications trial. Diabetes Care. 2003, 26 (3): 832-836.PubMed
2.
Zurück zum Zitat Gruessner AC, Sutherland DE: Pancreas transplant outcomes for United States (US) and non-US cases as reported to the United Network for Organ Sharing (UNOS) and the International Pancreas Transplant Registry (IPTR) as of June 2004. Clin Transplant. 2005, 19 (4): 433-455. 10.1111/j.1399-0012.2005.00378.x.PubMed Gruessner AC, Sutherland DE: Pancreas transplant outcomes for United States (US) and non-US cases as reported to the United Network for Organ Sharing (UNOS) and the International Pancreas Transplant Registry (IPTR) as of June 2004. Clin Transplant. 2005, 19 (4): 433-455. 10.1111/j.1399-0012.2005.00378.x.PubMed
3.
Zurück zum Zitat Coppelli A, Giannarelli R, Vistoli F, Del Prato S, Rizzo G, Mosca F, Boggi U, Marchetti P: The beneficial effects of pancreas transplant alone on diabetic nephropathy. Diabetes Care. 2005, 28 (6): 1366-1370.PubMed Coppelli A, Giannarelli R, Vistoli F, Del Prato S, Rizzo G, Mosca F, Boggi U, Marchetti P: The beneficial effects of pancreas transplant alone on diabetic nephropathy. Diabetes Care. 2005, 28 (6): 1366-1370.PubMed
4.
Zurück zum Zitat Venstrom JM, McBride MA, Rother KI, Hirshberg B, Orchard TJ, Harlan DM: Survival after pancreas transplantation in patients with diabetes and preserved kidney function. Jama. 2003, 290 (21): 2817-2823. 10.1001/jama.290.21.2817.PubMed Venstrom JM, McBride MA, Rother KI, Hirshberg B, Orchard TJ, Harlan DM: Survival after pancreas transplantation in patients with diabetes and preserved kidney function. Jama. 2003, 290 (21): 2817-2823. 10.1001/jama.290.21.2817.PubMed
5.
Zurück zum Zitat Gruessner RW, Sutherland DE, Gruessner AC: Mortality assessment for pancreas transplants. Am J Transplant. 2004, 4 (12): 2018-2026. 10.1111/j.1600-6143.2004.00667.x.PubMed Gruessner RW, Sutherland DE, Gruessner AC: Mortality assessment for pancreas transplants. Am J Transplant. 2004, 4 (12): 2018-2026. 10.1111/j.1600-6143.2004.00667.x.PubMed
6.
Zurück zum Zitat Ballinger WF, Lacy PE: Transplantation of intact pancreatic islets in rats. Surgery. 1972, 72 (2): 175-186.PubMed Ballinger WF, Lacy PE: Transplantation of intact pancreatic islets in rats. Surgery. 1972, 72 (2): 175-186.PubMed
7.
Zurück zum Zitat Najarian JS, Sutherland DE, Matas AJ, Steffes MW, Simmons RL, Goetz FC: Human islet transplantation: a preliminary report. Transplant Proc. 1977, 9 (1): 233-236.PubMed Najarian JS, Sutherland DE, Matas AJ, Steffes MW, Simmons RL, Goetz FC: Human islet transplantation: a preliminary report. Transplant Proc. 1977, 9 (1): 233-236.PubMed
8.
Zurück zum Zitat Largiader F, Kolb E, Binswanger U: A long-term functioning human pancreatic islet allotransplant. Transplantation. 1980, 29 (1): 76-77. 10.1097/00007890-198001000-00017.PubMed Largiader F, Kolb E, Binswanger U: A long-term functioning human pancreatic islet allotransplant. Transplantation. 1980, 29 (1): 76-77. 10.1097/00007890-198001000-00017.PubMed
9.
Zurück zum Zitat Scharp DW, Lacy PE, Santiago JV, McCullough CS, Weide LG, Falqui L, Marchetti P, Gingerich RL, Jaffe AS, Cryer PE: Insulin independence after islet transplantation into type I diabetic patient. Diabetes. 1990, 39 (4): 515-518.PubMed Scharp DW, Lacy PE, Santiago JV, McCullough CS, Weide LG, Falqui L, Marchetti P, Gingerich RL, Jaffe AS, Cryer PE: Insulin independence after islet transplantation into type I diabetic patient. Diabetes. 1990, 39 (4): 515-518.PubMed
10.
Zurück zum Zitat Matsumoto S, Okitsu T, Iwanaga Y, Noguchi H, Nagata H, Yonekawa Y, Yamada Y, Fukuda K, Tsukiyama K, Suzuki H, Kawasaki Y, Shimodaira M, Matsuoka K, Shibata T, Kasai Y, Maekawa T, Shapiro J, Tanaka K: Insulin independence after living-donor distal pancreatectomy and islet allotransplantation. Lancet. 2005, 365 (9471): 1642-1644. 10.1016/S0140-6736(05)66383-0.PubMed Matsumoto S, Okitsu T, Iwanaga Y, Noguchi H, Nagata H, Yonekawa Y, Yamada Y, Fukuda K, Tsukiyama K, Suzuki H, Kawasaki Y, Shimodaira M, Matsuoka K, Shibata T, Kasai Y, Maekawa T, Shapiro J, Tanaka K: Insulin independence after living-donor distal pancreatectomy and islet allotransplantation. Lancet. 2005, 365 (9471): 1642-1644. 10.1016/S0140-6736(05)66383-0.PubMed
11.
Zurück zum Zitat Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW: Automated method for isolation of human pancreatic islets. Diabetes. 1988, 37 (4): 413-420.PubMed Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW: Automated method for isolation of human pancreatic islets. Diabetes. 1988, 37 (4): 413-420.PubMed
12.
Zurück zum Zitat Bromberg JS, LeRoith D: Diabetes cure--is the glass half full?. N Engl J Med. 2006, 355 (13): 1372-1374. 10.1056/NEJMe068183.PubMed Bromberg JS, LeRoith D: Diabetes cure--is the glass half full?. N Engl J Med. 2006, 355 (13): 1372-1374. 10.1056/NEJMe068183.PubMed
13.
Zurück zum Zitat Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV: Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000, 343 (4): 230-238. 10.1056/NEJM200007273430401.PubMed Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV: Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000, 343 (4): 230-238. 10.1056/NEJM200007273430401.PubMed
14.
Zurück zum Zitat Rother KI, Harlan DM: Challenges facing islet transplantation for the treatment of type 1 diabetes mellitus. J Clin Invest. 2004, 114 (7): 877-883. 10.1172/JCI200423235.PubMedCentralPubMed Rother KI, Harlan DM: Challenges facing islet transplantation for the treatment of type 1 diabetes mellitus. J Clin Invest. 2004, 114 (7): 877-883. 10.1172/JCI200423235.PubMedCentralPubMed
15.
Zurück zum Zitat Naftanel MA, Harlan DM: Pancreatic islet transplantation. PLoS Med. 2004, 1 (3): e58; quiz e75-10.1371/journal.pmed.0010058.PubMed Naftanel MA, Harlan DM: Pancreatic islet transplantation. PLoS Med. 2004, 1 (3): e58; quiz e75-10.1371/journal.pmed.0010058.PubMed
16.
Zurück zum Zitat Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson RP, Secchi A, Brendel MD, Berney T, Brennan DC, Cagliero E, Alejandro R, Ryan EA, DiMercurio B, Morel P, Polonsky KS, Reems JA, Bretzel RG, Bertuzzi F, Froud T, Kandaswamy R, Sutherland DE, Eisenbarth G, Segal M, Preiksaitis J, Korbutt GS, Barton FB, Viviano L, Seyfert-Margolis V, Bluestone J, Lakey JR: International trial of the Edmonton protocol for islet transplantation. N Engl J Med. 2006, 355 (13): 1318-1330. 10.1056/NEJMoa061267.PubMed Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson RP, Secchi A, Brendel MD, Berney T, Brennan DC, Cagliero E, Alejandro R, Ryan EA, DiMercurio B, Morel P, Polonsky KS, Reems JA, Bretzel RG, Bertuzzi F, Froud T, Kandaswamy R, Sutherland DE, Eisenbarth G, Segal M, Preiksaitis J, Korbutt GS, Barton FB, Viviano L, Seyfert-Margolis V, Bluestone J, Lakey JR: International trial of the Edmonton protocol for islet transplantation. N Engl J Med. 2006, 355 (13): 1318-1330. 10.1056/NEJMoa061267.PubMed
17.
Zurück zum Zitat Hirshberg B, Rother KI, Digon BJ, Lee J, Gaglia JL, Hines K, Read EJ, Chang R, Wood BJ, Harlan DM: Benefits and risks of solitary islet transplantation for type 1 diabetes using steroid-sparing immunosuppression: the National Institutes of Health experience. Diabetes Care. 2003, 26 (12): 3288-3295.PubMed Hirshberg B, Rother KI, Digon BJ, Lee J, Gaglia JL, Hines K, Read EJ, Chang R, Wood BJ, Harlan DM: Benefits and risks of solitary islet transplantation for type 1 diabetes using steroid-sparing immunosuppression: the National Institutes of Health experience. Diabetes Care. 2003, 26 (12): 3288-3295.PubMed
18.
Zurück zum Zitat Hussain MA, Theise ND: Stem-cell therapy for diabetes mellitus. Lancet. 2004, 364 (9429): 203-205. 10.1016/S0140-6736(04)16635-X.PubMed Hussain MA, Theise ND: Stem-cell therapy for diabetes mellitus. Lancet. 2004, 364 (9429): 203-205. 10.1016/S0140-6736(04)16635-X.PubMed
19.
20.
Zurück zum Zitat Choi Y, Ta M, Atouf F, Lumelsky N: Adult pancreas generates multipotent stem cells and pancreatic and nonpancreatic progeny. Stem Cells. 2004, 22 (6): 1070-1084. 10.1634/stemcells.22-6-1070.PubMed Choi Y, Ta M, Atouf F, Lumelsky N: Adult pancreas generates multipotent stem cells and pancreatic and nonpancreatic progeny. Stem Cells. 2004, 22 (6): 1070-1084. 10.1634/stemcells.22-6-1070.PubMed
21.
Zurück zum Zitat Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki KL, Tzukerman M: Insulin production by human embryonic stem cells. Diabetes. 2001, 50 (8): 1691-1697.PubMed Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki KL, Tzukerman M: Insulin production by human embryonic stem cells. Diabetes. 2001, 50 (8): 1691-1697.PubMed
22.
Zurück zum Zitat Shiroi A, Yoshikawa M, Yokota H, Fukui H, Ishizaka S, Tatsumi K, Takahashi Y: Identification of insulin-producing cells derived from embryonic stem cells by zinc-chelating dithizone. Stem Cells. 2002, 20 (4): 284-292. 10.1634/stemcells.20-4-284.PubMed Shiroi A, Yoshikawa M, Yokota H, Fukui H, Ishizaka S, Tatsumi K, Takahashi Y: Identification of insulin-producing cells derived from embryonic stem cells by zinc-chelating dithizone. Stem Cells. 2002, 20 (4): 284-292. 10.1634/stemcells.20-4-284.PubMed
23.
Zurück zum Zitat Kahan BW, Jacobson LM, Hullett DA, Ochoada JM, Oberley TD, Lang KM, Odorico JS: Pancreatic precursors and differentiated islet cell types from murine embryonic stem cells: an in vitro model to study islet differentiation. Diabetes. 2003, 52 (8): 2016-2024.PubMed Kahan BW, Jacobson LM, Hullett DA, Ochoada JM, Oberley TD, Lang KM, Odorico JS: Pancreatic precursors and differentiated islet cell types from murine embryonic stem cells: an in vitro model to study islet differentiation. Diabetes. 2003, 52 (8): 2016-2024.PubMed
24.
Zurück zum Zitat Lumelsky N, Blondel O, Laeng P, Velasco I, Ravin R, McKay R: Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Science. 2001, 292 (5520): 1389-1394. 10.1126/science.1058866.PubMed Lumelsky N, Blondel O, Laeng P, Velasco I, Ravin R, McKay R: Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Science. 2001, 292 (5520): 1389-1394. 10.1126/science.1058866.PubMed
25.
Zurück zum Zitat Hori Y, Rulifson IC, Tsai BC, Heit JJ, Cahoy JD, Kim SK: Growth inhibitors promote differentiation of insulin-producing tissue from embryonic stem cells. Proc Natl Acad Sci U S A. 2002, 99 (25): 16105-16110. 10.1073/pnas.252618999.PubMedCentralPubMed Hori Y, Rulifson IC, Tsai BC, Heit JJ, Cahoy JD, Kim SK: Growth inhibitors promote differentiation of insulin-producing tissue from embryonic stem cells. Proc Natl Acad Sci U S A. 2002, 99 (25): 16105-16110. 10.1073/pnas.252618999.PubMedCentralPubMed
26.
Zurück zum Zitat Blyszczuk P, Czyz J, Kania G, Wagner M, Roll U, St-Onge L, Wobus AM: Expression of Pax4 in embryonic stem cells promotes differentiation of nestin-positive progenitor and insulin-producing cells. Proc Natl Acad Sci U S A. 2003, 100 (3): 998-1003. 10.1073/pnas.0237371100.PubMedCentralPubMed Blyszczuk P, Czyz J, Kania G, Wagner M, Roll U, St-Onge L, Wobus AM: Expression of Pax4 in embryonic stem cells promotes differentiation of nestin-positive progenitor and insulin-producing cells. Proc Natl Acad Sci U S A. 2003, 100 (3): 998-1003. 10.1073/pnas.0237371100.PubMedCentralPubMed
27.
Zurück zum Zitat Rajagopal J, Anderson WJ, Kume S, Martinez OI, Melton DA: Insulin staining of ES cell progeny from insulin uptake. Science. 2003, 299 (5605): 363-PubMed Rajagopal J, Anderson WJ, Kume S, Martinez OI, Melton DA: Insulin staining of ES cell progeny from insulin uptake. Science. 2003, 299 (5605): 363-PubMed
28.
Zurück zum Zitat D'Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK, Baetge EE: Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol. 2006, 24 (11): 1392-1401. 10.1038/nbt1259.PubMed D'Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK, Baetge EE: Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol. 2006, 24 (11): 1392-1401. 10.1038/nbt1259.PubMed
29.
Zurück zum Zitat Tang DQ, Cao LZ, Burkhardt BR, Xia CQ, Litherland SA, Atkinson MA, Yang LJ: In vivo and in vitro characterization of insulin-producing cells obtained from murine bone marrow. Diabetes. 2004, 53 (7): 1721-1732.PubMedCentralPubMed Tang DQ, Cao LZ, Burkhardt BR, Xia CQ, Litherland SA, Atkinson MA, Yang LJ: In vivo and in vitro characterization of insulin-producing cells obtained from murine bone marrow. Diabetes. 2004, 53 (7): 1721-1732.PubMedCentralPubMed
30.
Zurück zum Zitat Ferber S, Halkin A, Cohen H, Ber I, Einav Y, Goldberg I, Barshack I, Seijffers R, Kopolovic J, Kaiser N, Karasik A: Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat Med. 2000, 6 (5): 568-572. 10.1038/75050.PubMed Ferber S, Halkin A, Cohen H, Ber I, Einav Y, Goldberg I, Barshack I, Seijffers R, Kopolovic J, Kaiser N, Karasik A: Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat Med. 2000, 6 (5): 568-572. 10.1038/75050.PubMed
31.
Zurück zum Zitat Koizumi M, Doi R, Toyoda E, Tulachan SS, Kami K, Mori T, Ito D, Kawaguchi Y, Fujimoto K, Gittes GK, Imamura M: Hepatic regeneration and enforced PDX-1 expression accelerate transdifferentiation in liver. Surgery. 2004, 136 (2): 449-457. 10.1016/j.surg.2004.05.024.PubMed Koizumi M, Doi R, Toyoda E, Tulachan SS, Kami K, Mori T, Ito D, Kawaguchi Y, Fujimoto K, Gittes GK, Imamura M: Hepatic regeneration and enforced PDX-1 expression accelerate transdifferentiation in liver. Surgery. 2004, 136 (2): 449-457. 10.1016/j.surg.2004.05.024.PubMed
32.
Zurück zum Zitat Yamada S, Yamamoto Y, Nagasawa M, Hara A, Kodera T, Kojima I: In Vitro Transdifferentiation of Mature Hepatocytes into Insulin-Producing Cells. Endocr J. 2006 Yamada S, Yamamoto Y, Nagasawa M, Hara A, Kodera T, Kojima I: In Vitro Transdifferentiation of Mature Hepatocytes into Insulin-Producing Cells. Endocr J. 2006
33.
Zurück zum Zitat Cardona K, Korbutt GS, Milas Z, Lyon J, Cano J, Jiang W, Bello-Laborn H, Hacquoil B, Strobert E, Gangappa S, Weber CJ, Pearson TC, Rajotte RV, Larsen CP: Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways. Nat Med. 2006, 12 (3): 304-306. 10.1038/nm1375.PubMed Cardona K, Korbutt GS, Milas Z, Lyon J, Cano J, Jiang W, Bello-Laborn H, Hacquoil B, Strobert E, Gangappa S, Weber CJ, Pearson TC, Rajotte RV, Larsen CP: Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways. Nat Med. 2006, 12 (3): 304-306. 10.1038/nm1375.PubMed
34.
Zurück zum Zitat Hering BJ, Wijkstrom M, Graham ML, Hardstedt M, Aasheim TC, Jie T, Ansite JD, Nakano M, Cheng J, Li W, Moran K, Christians U, Finnegan C, Mills CD, Sutherland DE, Bansal-Pakala P, Murtaugh MP, Kirchhof N, Schuurman HJ: Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med. 2006, 12 (3): 301-303. 10.1038/nm1369.PubMed Hering BJ, Wijkstrom M, Graham ML, Hardstedt M, Aasheim TC, Jie T, Ansite JD, Nakano M, Cheng J, Li W, Moran K, Christians U, Finnegan C, Mills CD, Sutherland DE, Bansal-Pakala P, Murtaugh MP, Kirchhof N, Schuurman HJ: Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med. 2006, 12 (3): 301-303. 10.1038/nm1369.PubMed
35.
Zurück zum Zitat Ohneda K, Ee H, German M: Regulation of insulin gene transcription. Semin Cell Dev Biol. 2000, 11 (4): 227-233. 10.1006/scdb.2000.0171.PubMed Ohneda K, Ee H, German M: Regulation of insulin gene transcription. Semin Cell Dev Biol. 2000, 11 (4): 227-233. 10.1006/scdb.2000.0171.PubMed
36.
Zurück zum Zitat Poitout V, Hagman D, Stein R, Artner I, Robertson RP, Harmon JS: Regulation of the insulin gene by glucose and fatty acids. J Nutr. 2006, 136 (4): 873-876.PubMedCentralPubMed Poitout V, Hagman D, Stein R, Artner I, Robertson RP, Harmon JS: Regulation of the insulin gene by glucose and fatty acids. J Nutr. 2006, 136 (4): 873-876.PubMedCentralPubMed
37.
Zurück zum Zitat Ohneda K, Mirmira RG, Wang J, Johnson JD, German MS: The homeodomain of PDX-1 mediates multiple protein-protein interactions in the formation of a transcriptional activation complex on the insulin promoter. Mol Cell Biol. 2000, 20 (3): 900-911. 10.1128/MCB.20.3.900-911.2000.PubMedCentralPubMed Ohneda K, Mirmira RG, Wang J, Johnson JD, German MS: The homeodomain of PDX-1 mediates multiple protein-protein interactions in the formation of a transcriptional activation complex on the insulin promoter. Mol Cell Biol. 2000, 20 (3): 900-911. 10.1128/MCB.20.3.900-911.2000.PubMedCentralPubMed
38.
Zurück zum Zitat Zhang C, Moriguchi T, Kajihara M, Esaki R, Harada A, Shimohata H, Oishi H, Hamada M, Morito N, Hasegawa K, Kudo T, Engel JD, Yamamoto M, Takahashi S: MafA is a key regulator of glucose-stimulated insulin secretion. Mol Cell Biol. 2005, 25 (12): 4969-4976. 10.1128/MCB.25.12.4969-4976.2005.PubMedCentralPubMed Zhang C, Moriguchi T, Kajihara M, Esaki R, Harada A, Shimohata H, Oishi H, Hamada M, Morito N, Hasegawa K, Kudo T, Engel JD, Yamamoto M, Takahashi S: MafA is a key regulator of glucose-stimulated insulin secretion. Mol Cell Biol. 2005, 25 (12): 4969-4976. 10.1128/MCB.25.12.4969-4976.2005.PubMedCentralPubMed
39.
Zurück zum Zitat Kataoka K, Han SI, Shioda S, Hirai M, Nishizawa M, Handa H: MafA is a glucose-regulated and pancreatic beta-cell-specific transcriptional activator for the insulin gene. J Biol Chem. 2002, 277 (51): 49903-49910. 10.1074/jbc.M206796200.PubMed Kataoka K, Han SI, Shioda S, Hirai M, Nishizawa M, Handa H: MafA is a glucose-regulated and pancreatic beta-cell-specific transcriptional activator for the insulin gene. J Biol Chem. 2002, 277 (51): 49903-49910. 10.1074/jbc.M206796200.PubMed
40.
Zurück zum Zitat Kataoka K, Shioda S, Ando K, Sakagami K, Handa H, Yasuda K: Differentially expressed Maf family transcription factors, c-Maf and MafA, activate glucagon and insulin gene expression in pancreatic islet alpha- and beta-cells. J Mol Endocrinol. 2004, 32 (1): 9-20. 10.1677/jme.0.0320009.PubMed Kataoka K, Shioda S, Ando K, Sakagami K, Handa H, Yasuda K: Differentially expressed Maf family transcription factors, c-Maf and MafA, activate glucagon and insulin gene expression in pancreatic islet alpha- and beta-cells. J Mol Endocrinol. 2004, 32 (1): 9-20. 10.1677/jme.0.0320009.PubMed
41.
Zurück zum Zitat Matsuoka TA, Artner I, Henderson E, Means A, Sander M, Stein R: The MafA transcription factor appears to be responsible for tissue-specific expression of insulin. Proc Natl Acad Sci U S A. 2004, 101 (9): 2930-2933. 10.1073/pnas.0306233101.PubMedCentralPubMed Matsuoka TA, Artner I, Henderson E, Means A, Sander M, Stein R: The MafA transcription factor appears to be responsible for tissue-specific expression of insulin. Proc Natl Acad Sci U S A. 2004, 101 (9): 2930-2933. 10.1073/pnas.0306233101.PubMedCentralPubMed
42.
Zurück zum Zitat Orci L, Ravazzola M, Storch MJ, Anderson RG, Vassalli JD, Perrelet A: Proteolytic maturation of insulin is a post-Golgi event which occurs in acidifying clathrin-coated secretory vesicles. Cell. 1987, 49 (6): 865-868. 10.1016/0092-8674(87)90624-6.PubMed Orci L, Ravazzola M, Storch MJ, Anderson RG, Vassalli JD, Perrelet A: Proteolytic maturation of insulin is a post-Golgi event which occurs in acidifying clathrin-coated secretory vesicles. Cell. 1987, 49 (6): 865-868. 10.1016/0092-8674(87)90624-6.PubMed
43.
Zurück zum Zitat Orci L, Ravazzola M, Amherdt M, Yanaihara C, Yanaihara N, Halban P, Renold AE, Perrelet A: Insulin, not C-peptide (proinsulin), is present in crinophagic bodies of the pancreatic B-cell. J Cell Biol. 1984, 98 (1): 222-228. 10.1083/jcb.98.1.222.PubMed Orci L, Ravazzola M, Amherdt M, Yanaihara C, Yanaihara N, Halban P, Renold AE, Perrelet A: Insulin, not C-peptide (proinsulin), is present in crinophagic bodies of the pancreatic B-cell. J Cell Biol. 1984, 98 (1): 222-228. 10.1083/jcb.98.1.222.PubMed
44.
Zurück zum Zitat Wek RC, Jiang HY, Anthony TG: Coping with stress: eIF2 kinases and translational control. Biochem Soc Trans. 2006, 34 (Pt 1): 7-11.PubMed Wek RC, Jiang HY, Anthony TG: Coping with stress: eIF2 kinases and translational control. Biochem Soc Trans. 2006, 34 (Pt 1): 7-11.PubMed
45.
Zurück zum Zitat Harding HP, Ron D: Endoplasmic reticulum stress and the development of diabetes: a review. Diabetes. 2002, 51 Suppl 3: S455-61.PubMed Harding HP, Ron D: Endoplasmic reticulum stress and the development of diabetes: a review. Diabetes. 2002, 51 Suppl 3: S455-61.PubMed
46.
Zurück zum Zitat Haghighat A, Mader S, Pause A, Sonenberg N: Repression of cap-dependent translation by 4E-binding protein 1: competition with p220 for binding to eukaryotic initiation factor-4E. Embo J. 1995, 14 (22): 5701-5709.PubMedCentralPubMed Haghighat A, Mader S, Pause A, Sonenberg N: Repression of cap-dependent translation by 4E-binding protein 1: competition with p220 for binding to eukaryotic initiation factor-4E. Embo J. 1995, 14 (22): 5701-5709.PubMedCentralPubMed
47.
Zurück zum Zitat Pause A, Belsham GJ, Gingras AC, Donze O, Lin TA, Lawrence JC, Sonenberg N: Insulin-dependent stimulation of protein synthesis by phosphorylation of a regulator of 5'-cap function. Nature. 1994, 371 (6500): 762-767. 10.1038/371762a0.PubMed Pause A, Belsham GJ, Gingras AC, Donze O, Lin TA, Lawrence JC, Sonenberg N: Insulin-dependent stimulation of protein synthesis by phosphorylation of a regulator of 5'-cap function. Nature. 1994, 371 (6500): 762-767. 10.1038/371762a0.PubMed
48.
Zurück zum Zitat Curry DL, Bennett LL, Grodsky GM: Dynamics of insulin secretion by the perfused rat pancreas. Endocrinology. 1968, 83 (3): 572-584.PubMed Curry DL, Bennett LL, Grodsky GM: Dynamics of insulin secretion by the perfused rat pancreas. Endocrinology. 1968, 83 (3): 572-584.PubMed
49.
Zurück zum Zitat Henquin JC, Nenquin M, Stiernet P, Ahren B: In Vivo and In Vitro Glucose-Induced Biphasic Insulin Secretion in the Mouse: Pattern and Role of Cytoplasmic Ca2+ and Amplification Signals in {beta}-Cells. Diabetes. 2006, 55 (2): 441-451. 10.2337/diabetes.55.02.06.db05-1051.PubMed Henquin JC, Nenquin M, Stiernet P, Ahren B: In Vivo and In Vitro Glucose-Induced Biphasic Insulin Secretion in the Mouse: Pattern and Role of Cytoplasmic Ca2+ and Amplification Signals in {beta}-Cells. Diabetes. 2006, 55 (2): 441-451. 10.2337/diabetes.55.02.06.db05-1051.PubMed
50.
Zurück zum Zitat Fehse F, Trautmann M, Holst JJ, Halseth AE, Nanayakkara N, Nielsen LL, Fineman MS, Kim DD, Nauck MA: Exenatide augments first- and second-phase insulin secretion in response to intravenous glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab. 2005, 90 (11): 5991-5997. 10.1210/jc.2005-1093.PubMed Fehse F, Trautmann M, Holst JJ, Halseth AE, Nanayakkara N, Nielsen LL, Fineman MS, Kim DD, Nauck MA: Exenatide augments first- and second-phase insulin secretion in response to intravenous glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab. 2005, 90 (11): 5991-5997. 10.1210/jc.2005-1093.PubMed
51.
Zurück zum Zitat Kwan EP, Gaisano HY: Glucagon-like peptide 1 regulates sequential and compound exocytosis in pancreatic islet beta-cells. Diabetes. 2005, 54 (9): 2734-2743.PubMed Kwan EP, Gaisano HY: Glucagon-like peptide 1 regulates sequential and compound exocytosis in pancreatic islet beta-cells. Diabetes. 2005, 54 (9): 2734-2743.PubMed
52.
Zurück zum Zitat Bratanova-Tochkova TK, Cheng H, Daniel S, Gunawardana S, Liu YJ, Mulvaney-Musa J, Schermerhorn T, Straub SG, Yajima H, Sharp GW: Triggering and augmentation mechanisms, granule pools, and biphasic insulin secretion. Diabetes. 2002, 51 Suppl 1: S83-90.PubMed Bratanova-Tochkova TK, Cheng H, Daniel S, Gunawardana S, Liu YJ, Mulvaney-Musa J, Schermerhorn T, Straub SG, Yajima H, Sharp GW: Triggering and augmentation mechanisms, granule pools, and biphasic insulin secretion. Diabetes. 2002, 51 Suppl 1: S83-90.PubMed
53.
Zurück zum Zitat Rorsman P, Renstrom E: Insulin granule dynamics in pancreatic beta cells. Diabetologia. 2003, 46 (8): 1029-1045. 10.1007/s00125-003-1153-1.PubMed Rorsman P, Renstrom E: Insulin granule dynamics in pancreatic beta cells. Diabetologia. 2003, 46 (8): 1029-1045. 10.1007/s00125-003-1153-1.PubMed
54.
Zurück zum Zitat Daniel S, Noda M, Straub SG, Sharp GW: Identification of the docked granule pool responsible for the first phase of glucose-stimulated insulin secretion. Diabetes. 1999, 48 (9): 1686-1690.PubMed Daniel S, Noda M, Straub SG, Sharp GW: Identification of the docked granule pool responsible for the first phase of glucose-stimulated insulin secretion. Diabetes. 1999, 48 (9): 1686-1690.PubMed
55.
Zurück zum Zitat Itoh N, Okamoto H: Translational control of proinsulin synthesis by glucose. Nature. 1980, 283 (5742): 100-102. 10.1038/283100a0.PubMed Itoh N, Okamoto H: Translational control of proinsulin synthesis by glucose. Nature. 1980, 283 (5742): 100-102. 10.1038/283100a0.PubMed
56.
Zurück zum Zitat Welsh M, Scherberg N, Gilmore R, Steiner DF: Translational control of insulin biosynthesis. Evidence for regulation of elongation, initiation and signal-recognition-particle-mediated translational arrest by glucose. Biochem J. 1986, 235 (2): 459-467.PubMedCentralPubMed Welsh M, Scherberg N, Gilmore R, Steiner DF: Translational control of insulin biosynthesis. Evidence for regulation of elongation, initiation and signal-recognition-particle-mediated translational arrest by glucose. Biochem J. 1986, 235 (2): 459-467.PubMedCentralPubMed
57.
Zurück zum Zitat Maechler P, Carobbio S, Rubi B: In beta-cells, mitochondria integrate and generate metabolic signals controlling insulin secretion. Int J Biochem Cell Biol. 2006 Maechler P, Carobbio S, Rubi B: In beta-cells, mitochondria integrate and generate metabolic signals controlling insulin secretion. Int J Biochem Cell Biol. 2006
58.
Zurück zum Zitat MacDonald PE, Wheeler MB: Voltage-dependent K(+) channels in pancreatic beta cells: role, regulation and potential as therapeutic targets. Diabetologia. 2003, 46 (8): 1046-1062. 10.1007/s00125-003-1159-8.PubMed MacDonald PE, Wheeler MB: Voltage-dependent K(+) channels in pancreatic beta cells: role, regulation and potential as therapeutic targets. Diabetologia. 2003, 46 (8): 1046-1062. 10.1007/s00125-003-1159-8.PubMed
59.
Zurück zum Zitat Fukui K, Yang Q, Cao Y, Takahashi N, Hatakeyama H, Wang H, Wada J, Zhang Y, Marselli L, Nammo T, Yoneda K, Onishi M, Higashiyama S, Matsuzawa Y, Gonzalez FJ, Weir GC, Kasai H, Shimomura I, Miyagawa J, Wollheim CB, Yamagata K: The HNF-1 target collectrin controls insulin exocytosis by SNARE complex formation. Cell Metab. 2005, 2 (6): 373-384. 10.1016/j.cmet.2005.11.003.PubMed Fukui K, Yang Q, Cao Y, Takahashi N, Hatakeyama H, Wang H, Wada J, Zhang Y, Marselli L, Nammo T, Yoneda K, Onishi M, Higashiyama S, Matsuzawa Y, Gonzalez FJ, Weir GC, Kasai H, Shimomura I, Miyagawa J, Wollheim CB, Yamagata K: The HNF-1 target collectrin controls insulin exocytosis by SNARE complex formation. Cell Metab. 2005, 2 (6): 373-384. 10.1016/j.cmet.2005.11.003.PubMed
60.
Zurück zum Zitat Bruns D, Jahn R: Molecular determinants of exocytosis. Pflugers Arch. 2002, 443 (3): 333-338. 10.1007/s00424-001-0742-4.PubMed Bruns D, Jahn R: Molecular determinants of exocytosis. Pflugers Arch. 2002, 443 (3): 333-338. 10.1007/s00424-001-0742-4.PubMed
61.
Zurück zum Zitat Easom RA: Beta-granule transport and exocytosis. Semin Cell Dev Biol. 2000, 11 (4): 253-266. 10.1006/scdb.2000.0174.PubMed Easom RA: Beta-granule transport and exocytosis. Semin Cell Dev Biol. 2000, 11 (4): 253-266. 10.1006/scdb.2000.0174.PubMed
62.
Zurück zum Zitat Barg S, Ma X, Eliasson L, Galvanovskis J, Gopel SO, Obermuller S, Platzer J, Renstrom E, Trus M, Atlas D, Striessnig J, Rorsman P: Fast exocytosis with few Ca(2+) channels in insulin-secreting mouse pancreatic B cells. Biophys J. 2001, 81 (6): 3308-3323.PubMedCentralPubMed Barg S, Ma X, Eliasson L, Galvanovskis J, Gopel SO, Obermuller S, Platzer J, Renstrom E, Trus M, Atlas D, Striessnig J, Rorsman P: Fast exocytosis with few Ca(2+) channels in insulin-secreting mouse pancreatic B cells. Biophys J. 2001, 81 (6): 3308-3323.PubMedCentralPubMed
63.
Zurück zum Zitat Barg S, Eliasson L, Renstrom E, Rorsman P: A subset of 50 secretory granules in close contact with L-type Ca2+ channels accounts for first-phase insulin secretion in mouse beta-cells. Diabetes. 2002, 51 Suppl 1: S74-82.PubMed Barg S, Eliasson L, Renstrom E, Rorsman P: A subset of 50 secretory granules in close contact with L-type Ca2+ channels accounts for first-phase insulin secretion in mouse beta-cells. Diabetes. 2002, 51 Suppl 1: S74-82.PubMed
64.
Zurück zum Zitat Wei FY, Nagashima K, Ohshima T, Saheki Y, Lu YF, Matsushita M, Yamada Y, Mikoshiba K, Seino Y, Matsui H, Tomizawa K: Cdk5-dependent regulation of glucose-stimulated insulin secretion. Nat Med. 2005, 11 (10): 1104-1108. 10.1038/nm1299.PubMed Wei FY, Nagashima K, Ohshima T, Saheki Y, Lu YF, Matsushita M, Yamada Y, Mikoshiba K, Seino Y, Matsui H, Tomizawa K: Cdk5-dependent regulation of glucose-stimulated insulin secretion. Nat Med. 2005, 11 (10): 1104-1108. 10.1038/nm1299.PubMed
65.
Zurück zum Zitat Detimary P, Dejonghe S, Ling Z, Pipeleers D, Schuit F, Henquin JC: The changes in adenine nucleotides measured in glucose-stimulated rodent islets occur in beta cells but not in alpha cells and are also observed in human islets. J Biol Chem. 1998, 273 (51): 33905-33908. 10.1074/jbc.273.51.33905.PubMed Detimary P, Dejonghe S, Ling Z, Pipeleers D, Schuit F, Henquin JC: The changes in adenine nucleotides measured in glucose-stimulated rodent islets occur in beta cells but not in alpha cells and are also observed in human islets. J Biol Chem. 1998, 273 (51): 33905-33908. 10.1074/jbc.273.51.33905.PubMed
66.
Zurück zum Zitat Maechler P, Wollheim CB: Mitochondrial function in normal and diabetic beta-cells. Nature. 2001, 414 (6865): 807-812. 10.1038/414807a.PubMed Maechler P, Wollheim CB: Mitochondrial function in normal and diabetic beta-cells. Nature. 2001, 414 (6865): 807-812. 10.1038/414807a.PubMed
67.
Zurück zum Zitat Barg S, Huang P, Eliasson L, Nelson DJ, Obermuller S, Rorsman P, Thevenod F, Renstrom E: Priming of insulin granules for exocytosis by granular Cl(-) uptake and acidification. J Cell Sci. 2001, 114 (Pt 11): 2145-2154.PubMed Barg S, Huang P, Eliasson L, Nelson DJ, Obermuller S, Rorsman P, Thevenod F, Renstrom E: Priming of insulin granules for exocytosis by granular Cl(-) uptake and acidification. J Cell Sci. 2001, 114 (Pt 11): 2145-2154.PubMed
68.
Zurück zum Zitat Thevenod F: Ion channels in secretory granules of the pancreas and their role in exocytosis and release of secretory proteins. Am J Physiol Cell Physiol. 2002, 283 (3): C651-72.PubMed Thevenod F: Ion channels in secretory granules of the pancreas and their role in exocytosis and release of secretory proteins. Am J Physiol Cell Physiol. 2002, 283 (3): C651-72.PubMed
69.
Zurück zum Zitat Goodge KA, Hutton JC: Translational regulation of proinsulin biosynthesis and proinsulin conversion in the pancreatic beta-cell. Semin Cell Dev Biol. 2000, 11 (4): 235-242. 10.1006/scdb.2000.0172.PubMed Goodge KA, Hutton JC: Translational regulation of proinsulin biosynthesis and proinsulin conversion in the pancreatic beta-cell. Semin Cell Dev Biol. 2000, 11 (4): 235-242. 10.1006/scdb.2000.0172.PubMed
70.
Zurück zum Zitat Efrat S, Surana M, Fleischer N: Glucose induces insulin gene transcription in a murine pancreatic beta-cell line. J Biol Chem. 1991, 266 (17): 11141-11143.PubMed Efrat S, Surana M, Fleischer N: Glucose induces insulin gene transcription in a murine pancreatic beta-cell line. J Biol Chem. 1991, 266 (17): 11141-11143.PubMed
71.
Zurück zum Zitat Welsh M, Nielsen DA, MacKrell AJ, Steiner DF: Control of insulin gene expression in pancreatic beta-cells and in an insulin-producing cell line, RIN-5F cells. II. Regulation of insulin mRNA stability. J Biol Chem. 1985, 260 (25): 13590-13594.PubMed Welsh M, Nielsen DA, MacKrell AJ, Steiner DF: Control of insulin gene expression in pancreatic beta-cells and in an insulin-producing cell line, RIN-5F cells. II. Regulation of insulin mRNA stability. J Biol Chem. 1985, 260 (25): 13590-13594.PubMed
72.
Zurück zum Zitat Philippe J, Pacheco I, Meda P: Insulin gene transcription is decreased rapidly by lowering glucose concentrations in rat islet cells. Diabetes. 1994, 43 (4): 523-528.PubMed Philippe J, Pacheco I, Meda P: Insulin gene transcription is decreased rapidly by lowering glucose concentrations in rat islet cells. Diabetes. 1994, 43 (4): 523-528.PubMed
73.
Zurück zum Zitat Brunstedt J, Chan SJ: Direct effect of glucose on the preproinsulin mRNA level in isolated pancreatic islets. Biochem Biophys Res Commun. 1982, 106 (4): 1383-1389. 10.1016/0006-291X(82)91267-0.PubMed Brunstedt J, Chan SJ: Direct effect of glucose on the preproinsulin mRNA level in isolated pancreatic islets. Biochem Biophys Res Commun. 1982, 106 (4): 1383-1389. 10.1016/0006-291X(82)91267-0.PubMed
74.
Zurück zum Zitat Wicksteed B, Herbert TP, Alarcon C, Lingohr MK, Moss LG, Rhodes CJ: Cooperativity between the preproinsulin mRNA untranslated regions is necessary for glucose-stimulated translation. J Biol Chem. 2001, 276 (25): 22553-22558. 10.1074/jbc.M011214200.PubMed Wicksteed B, Herbert TP, Alarcon C, Lingohr MK, Moss LG, Rhodes CJ: Cooperativity between the preproinsulin mRNA untranslated regions is necessary for glucose-stimulated translation. J Biol Chem. 2001, 276 (25): 22553-22558. 10.1074/jbc.M011214200.PubMed
75.
Zurück zum Zitat Shalev A, Blair PJ, Hoffmann SC, Hirshberg B, Peculis BA, Harlan DM: A proinsulin gene splice variant with increased translation efficiency is expressed in human pancreatic islets. Endocrinology. 2002, 143 (7): 2541-2547. 10.1210/en.143.7.2541.PubMed Shalev A, Blair PJ, Hoffmann SC, Hirshberg B, Peculis BA, Harlan DM: A proinsulin gene splice variant with increased translation efficiency is expressed in human pancreatic islets. Endocrinology. 2002, 143 (7): 2541-2547. 10.1210/en.143.7.2541.PubMed
76.
Zurück zum Zitat Leibowitz G, Oprescu AI, Uckaya G, Gross DJ, Cerasi E, Kaiser N: Insulin does not mediate glucose stimulation of proinsulin biosynthesis. Diabetes. 2003, 52 (4): 998-1003.PubMed Leibowitz G, Oprescu AI, Uckaya G, Gross DJ, Cerasi E, Kaiser N: Insulin does not mediate glucose stimulation of proinsulin biosynthesis. Diabetes. 2003, 52 (4): 998-1003.PubMed
77.
Zurück zum Zitat Wicksteed B, Alarcon C, Briaud I, Lingohr MK, Rhodes CJ: Glucose-induced translational control of proinsulin biosynthesis is proportional to preproinsulin mRNA levels in islet beta-cells but not regulated via a positive feedback of secreted insulin. J Biol Chem. 2003, 278 (43): 42080-42090. 10.1074/jbc.M303509200.PubMed Wicksteed B, Alarcon C, Briaud I, Lingohr MK, Rhodes CJ: Glucose-induced translational control of proinsulin biosynthesis is proportional to preproinsulin mRNA levels in islet beta-cells but not regulated via a positive feedback of secreted insulin. J Biol Chem. 2003, 278 (43): 42080-42090. 10.1074/jbc.M303509200.PubMed
78.
Zurück zum Zitat Leibiger B, Moede T, Schwarz T, Brown GR, Kohler M, Leibiger IB, Berggren PO: Short-term regulation of insulin gene transcription by glucose. Proc Natl Acad Sci U S A. 1998, 95 (16): 9307-9312. 10.1073/pnas.95.16.9307.PubMedCentralPubMed Leibiger B, Moede T, Schwarz T, Brown GR, Kohler M, Leibiger IB, Berggren PO: Short-term regulation of insulin gene transcription by glucose. Proc Natl Acad Sci U S A. 1998, 95 (16): 9307-9312. 10.1073/pnas.95.16.9307.PubMedCentralPubMed
79.
Zurück zum Zitat Marshak S, Totary H, Cerasi E, Melloul D: Purification of the beta-cell glucose-sensitive factor that transactivates the insulin gene differentially in normal and transformed islet cells. Proc Natl Acad Sci U S A. 1996, 93 (26): 15057-15062. 10.1073/pnas.93.26.15057.PubMedCentralPubMed Marshak S, Totary H, Cerasi E, Melloul D: Purification of the beta-cell glucose-sensitive factor that transactivates the insulin gene differentially in normal and transformed islet cells. Proc Natl Acad Sci U S A. 1996, 93 (26): 15057-15062. 10.1073/pnas.93.26.15057.PubMedCentralPubMed
80.
Zurück zum Zitat Olbrot M, Rud J, Moss LG, Sharma A: Identification of beta-cell-specific insulin gene transcription factor RIPE3b1 as mammalian MafA. Proc Natl Acad Sci U S A. 2002, 99 (10): 6737-6742. 10.1073/pnas.102168499.PubMedCentralPubMed Olbrot M, Rud J, Moss LG, Sharma A: Identification of beta-cell-specific insulin gene transcription factor RIPE3b1 as mammalian MafA. Proc Natl Acad Sci U S A. 2002, 99 (10): 6737-6742. 10.1073/pnas.102168499.PubMedCentralPubMed
81.
Zurück zum Zitat Matsuoka TA, Zhao L, Artner I, Jarrett HW, Friedman D, Means A, Stein R: Members of the large Maf transcription family regulate insulin gene transcription in islet beta cells. Mol Cell Biol. 2003, 23 (17): 6049-6062. 10.1128/MCB.23.17.6049-6062.2003.PubMedCentralPubMed Matsuoka TA, Zhao L, Artner I, Jarrett HW, Friedman D, Means A, Stein R: Members of the large Maf transcription family regulate insulin gene transcription in islet beta cells. Mol Cell Biol. 2003, 23 (17): 6049-6062. 10.1128/MCB.23.17.6049-6062.2003.PubMedCentralPubMed
82.
Zurück zum Zitat Qiu Y, Sharma A, Stein R: p300 mediates transcriptional stimulation by the basic helix-loop-helix activators of the insulin gene. Mol Cell Biol. 1998, 18 (5): 2957-2964.PubMedCentralPubMed Qiu Y, Sharma A, Stein R: p300 mediates transcriptional stimulation by the basic helix-loop-helix activators of the insulin gene. Mol Cell Biol. 1998, 18 (5): 2957-2964.PubMedCentralPubMed
83.
Zurück zum Zitat Kaneto H, Sharma A, Suzuma K, Laybutt DR, Xu G, Bonner-Weir S, Weir GC: Induction of c-Myc expression suppresses insulin gene transcription by inhibiting NeuroD/BETA2-mediated transcriptional activation. J Biol Chem. 2002, 277 (15): 12998-13006. 10.1074/jbc.M111148200.PubMed Kaneto H, Sharma A, Suzuma K, Laybutt DR, Xu G, Bonner-Weir S, Weir GC: Induction of c-Myc expression suppresses insulin gene transcription by inhibiting NeuroD/BETA2-mediated transcriptional activation. J Biol Chem. 2002, 277 (15): 12998-13006. 10.1074/jbc.M111148200.PubMed
84.
Zurück zum Zitat Melloul D, Ben-Neriah Y, Cerasi E: Glucose modulates the binding of an islet-specific factor to a conserved sequence within the rat I and the human insulin promoters. Proc Natl Acad Sci U S A. 1993, 90 (9): 3865-3869. 10.1073/pnas.90.9.3865.PubMedCentralPubMed Melloul D, Ben-Neriah Y, Cerasi E: Glucose modulates the binding of an islet-specific factor to a conserved sequence within the rat I and the human insulin promoters. Proc Natl Acad Sci U S A. 1993, 90 (9): 3865-3869. 10.1073/pnas.90.9.3865.PubMedCentralPubMed
85.
Zurück zum Zitat Sharma A, Fusco-DeMane D, Henderson E, Efrat S, Stein R: The role of the insulin control element and RIPE3b1 activators in glucose-stimulated transcription of the insulin gene. Mol Endocrinol. 1995, 9 (11): 1468-1476. 10.1210/me.9.11.1468.PubMed Sharma A, Fusco-DeMane D, Henderson E, Efrat S, Stein R: The role of the insulin control element and RIPE3b1 activators in glucose-stimulated transcription of the insulin gene. Mol Endocrinol. 1995, 9 (11): 1468-1476. 10.1210/me.9.11.1468.PubMed
86.
Zurück zum Zitat Aramata S, Han SI, Yasuda K, Kataoka K: Synergistic activation of the insulin gene promoter by the beta-cell enriched transcription factors MafA, Beta2, and Pdx1. Biochim Biophys Acta. 2005, 1730 (1): 41-46.PubMed Aramata S, Han SI, Yasuda K, Kataoka K: Synergistic activation of the insulin gene promoter by the beta-cell enriched transcription factors MafA, Beta2, and Pdx1. Biochim Biophys Acta. 2005, 1730 (1): 41-46.PubMed
87.
Zurück zum Zitat Glick E, Leshkowitz D, Walker MD: Transcription factor BETA2 acts cooperatively with E2A and PDX1 to activate the insulin gene promoter. J Biol Chem. 2000, 275 (3): 2199-2204. 10.1074/jbc.275.3.2199.PubMed Glick E, Leshkowitz D, Walker MD: Transcription factor BETA2 acts cooperatively with E2A and PDX1 to activate the insulin gene promoter. J Biol Chem. 2000, 275 (3): 2199-2204. 10.1074/jbc.275.3.2199.PubMed
88.
Zurück zum Zitat Zhao L, Guo M, Matsuoka TA, Hagman DK, Parazzoli SD, Poitout V, Stein R: The islet beta cell-enriched MafA activator is a key regulator of insulin gene transcription. J Biol Chem. 2005, 280 (12): 11887-11894. 10.1074/jbc.M409475200.PubMed Zhao L, Guo M, Matsuoka TA, Hagman DK, Parazzoli SD, Poitout V, Stein R: The islet beta cell-enriched MafA activator is a key regulator of insulin gene transcription. J Biol Chem. 2005, 280 (12): 11887-11894. 10.1074/jbc.M409475200.PubMed
89.
Zurück zum Zitat Hagman DK, Hays LB, Parazzoli SD, Poitout V: Palmitate inhibits insulin gene expression by altering PDX-1 nuclear localization and reducing MafA expression in isolated rat islets of Langerhans. J Biol Chem. 2005, 280 (37): 32413-32418. 10.1074/jbc.M506000200.PubMedCentralPubMed Hagman DK, Hays LB, Parazzoli SD, Poitout V: Palmitate inhibits insulin gene expression by altering PDX-1 nuclear localization and reducing MafA expression in isolated rat islets of Langerhans. J Biol Chem. 2005, 280 (37): 32413-32418. 10.1074/jbc.M506000200.PubMedCentralPubMed
90.
Zurück zum Zitat Mosley AL, Corbett JA, Ozcan S: Glucose regulation of insulin gene expression requires the recruitment of p300 by the beta-cell-specific transcription factor Pdx-1. Mol Endocrinol. 2004, 18 (9): 2279-2290. 10.1210/me.2003-0463.PubMed Mosley AL, Corbett JA, Ozcan S: Glucose regulation of insulin gene expression requires the recruitment of p300 by the beta-cell-specific transcription factor Pdx-1. Mol Endocrinol. 2004, 18 (9): 2279-2290. 10.1210/me.2003-0463.PubMed
91.
Zurück zum Zitat Pedersen AA, Petersen HV, Videbaek N, Skak K, Michelsen BK: PDX-1 mediates glucose responsiveness of GAD(67), but not GAD(65), gene transcription in islets of Langerhans. Biochem Biophys Res Commun. 2002, 295 (2): 243-248. 10.1016/S0006-291X(02)00674-5.PubMed Pedersen AA, Petersen HV, Videbaek N, Skak K, Michelsen BK: PDX-1 mediates glucose responsiveness of GAD(67), but not GAD(65), gene transcription in islets of Langerhans. Biochem Biophys Res Commun. 2002, 295 (2): 243-248. 10.1016/S0006-291X(02)00674-5.PubMed
92.
Zurück zum Zitat Melloul D, Marshak S, Cerasi E: Regulation of insulin gene transcription. Diabetologia. 2002, 45 (3): 309-326. 10.1007/s00125-001-0728-y.PubMed Melloul D, Marshak S, Cerasi E: Regulation of insulin gene transcription. Diabetologia. 2002, 45 (3): 309-326. 10.1007/s00125-001-0728-y.PubMed
93.
Zurück zum Zitat Peshavaria M, Cissell MA, Henderson E, Petersen HV, Stein R: The PDX-1 activation domain provides specific functions necessary for transcriptional stimulation in pancreatic beta-cells. Mol Endocrinol. 2000, 14 (12): 1907-1917. 10.1210/me.14.12.1907.PubMed Peshavaria M, Cissell MA, Henderson E, Petersen HV, Stein R: The PDX-1 activation domain provides specific functions necessary for transcriptional stimulation in pancreatic beta-cells. Mol Endocrinol. 2000, 14 (12): 1907-1917. 10.1210/me.14.12.1907.PubMed
94.
Zurück zum Zitat Shushan EB, Cerasi E, Melloul D: Regulation of the insulin gene by glucose: stimulation of trans-activation potency of human PDX-1 N-terminal domain. DNA Cell Biol. 1999, 18 (6): 471-479. 10.1089/104454999315196.PubMed Shushan EB, Cerasi E, Melloul D: Regulation of the insulin gene by glucose: stimulation of trans-activation potency of human PDX-1 N-terminal domain. DNA Cell Biol. 1999, 18 (6): 471-479. 10.1089/104454999315196.PubMed
95.
Zurück zum Zitat Elrick LJ, Docherty K: Phosphorylation-dependent nucleocytoplasmic shuttling of pancreatic duodenal homeobox-1. Diabetes. 2001, 50 (10): 2244-2252.PubMed Elrick LJ, Docherty K: Phosphorylation-dependent nucleocytoplasmic shuttling of pancreatic duodenal homeobox-1. Diabetes. 2001, 50 (10): 2244-2252.PubMed
96.
Zurück zum Zitat Rafiq I, da Silva Xavier G, Hooper S, Rutter GA: Glucose-stimulated preproinsulin gene expression and nuclear trans-location of pancreatic duodenum homeobox-1 require activation of phosphatidylinositol 3-kinase but not p38 MAPK/SAPK2. J Biol Chem. 2000, 275 (21): 15977-15984. 10.1074/jbc.275.21.15977.PubMed Rafiq I, da Silva Xavier G, Hooper S, Rutter GA: Glucose-stimulated preproinsulin gene expression and nuclear trans-location of pancreatic duodenum homeobox-1 require activation of phosphatidylinositol 3-kinase but not p38 MAPK/SAPK2. J Biol Chem. 2000, 275 (21): 15977-15984. 10.1074/jbc.275.21.15977.PubMed
97.
Zurück zum Zitat Leibiger IB, Leibiger B, Moede T, Berggren PO: Exocytosis of insulin promotes insulin gene transcription via the insulin receptor/PI-3 kinase/p70 s6 kinase and CaM kinase pathways. Mol Cell. 1998, 1 (6): 933-938. 10.1016/S1097-2765(00)80093-3.PubMed Leibiger IB, Leibiger B, Moede T, Berggren PO: Exocytosis of insulin promotes insulin gene transcription via the insulin receptor/PI-3 kinase/p70 s6 kinase and CaM kinase pathways. Mol Cell. 1998, 1 (6): 933-938. 10.1016/S1097-2765(00)80093-3.PubMed
98.
Zurück zum Zitat Xu GG, Gao ZY, Borge PD, Jegier PA, Young RA, Wolf BA: Insulin regulation of beta-cell function involves a feedback loop on SERCA gene expression, Ca(2+) homeostasis, and insulin expression and secretion. Biochemistry. 2000, 39 (48): 14912-14919. 10.1021/bi001260w.PubMed Xu GG, Gao ZY, Borge PD, Jegier PA, Young RA, Wolf BA: Insulin regulation of beta-cell function involves a feedback loop on SERCA gene expression, Ca(2+) homeostasis, and insulin expression and secretion. Biochemistry. 2000, 39 (48): 14912-14919. 10.1021/bi001260w.PubMed
99.
Zurück zum Zitat Leibiger B, Wahlander K, Berggren PO, Leibiger IB: Glucose-stimulated insulin biosynthesis depends on insulin-stimulated insulin gene transcription. J Biol Chem. 2000, 275 (39): 30153-30156. 10.1074/jbc.M005216200.PubMed Leibiger B, Wahlander K, Berggren PO, Leibiger IB: Glucose-stimulated insulin biosynthesis depends on insulin-stimulated insulin gene transcription. J Biol Chem. 2000, 275 (39): 30153-30156. 10.1074/jbc.M005216200.PubMed
100.
Zurück zum Zitat Mosley AL, Ozcan S: The pancreatic duodenal homeobox-1 protein (Pdx-1) interacts with histone deacetylases Hdac-1 and Hdac-2 on low levels of glucose. J Biol Chem. 2004, 279 (52): 54241-54247. 10.1074/jbc.M410379200.PubMed Mosley AL, Ozcan S: The pancreatic duodenal homeobox-1 protein (Pdx-1) interacts with histone deacetylases Hdac-1 and Hdac-2 on low levels of glucose. J Biol Chem. 2004, 279 (52): 54241-54247. 10.1074/jbc.M410379200.PubMed
101.
Zurück zum Zitat Naya FJ, Stellrecht CM, Tsai MJ: Tissue-specific regulation of the insulin gene by a novel basic helix-loop-helix transcription factor. Genes Dev. 1995, 9 (8): 1009-1019.PubMed Naya FJ, Stellrecht CM, Tsai MJ: Tissue-specific regulation of the insulin gene by a novel basic helix-loop-helix transcription factor. Genes Dev. 1995, 9 (8): 1009-1019.PubMed
102.
Zurück zum Zitat Odagiri H, Wang J, German MS: Function of the human insulin promoter in primary cultured islet cells. J Biol Chem. 1996, 271 (4): 1909-1915. 10.1074/jbc.271.4.1909.PubMed Odagiri H, Wang J, German MS: Function of the human insulin promoter in primary cultured islet cells. J Biol Chem. 1996, 271 (4): 1909-1915. 10.1074/jbc.271.4.1909.PubMed
103.
Zurück zum Zitat Amemiya-Kudo M, Oka J, Ide T, Matsuzaka T, Sone H, Yoshikawa T, Yahagi N, Ishibashi S, Osuga J, Yamada N, Murase T, Shimano H: Sterol regulatory element-binding proteins activate insulin gene promoter directly and indirectly through synergy with BETA2/E47. J Biol Chem. 2005, 280 (41): 34577-34589. 10.1074/jbc.M506718200.PubMed Amemiya-Kudo M, Oka J, Ide T, Matsuzaka T, Sone H, Yoshikawa T, Yahagi N, Ishibashi S, Osuga J, Yamada N, Murase T, Shimano H: Sterol regulatory element-binding proteins activate insulin gene promoter directly and indirectly through synergy with BETA2/E47. J Biol Chem. 2005, 280 (41): 34577-34589. 10.1074/jbc.M506718200.PubMed
104.
Zurück zum Zitat Khoo S, Griffen SC, Xia Y, Baer RJ, German MS, Cobb MH: Regulation of insulin gene transcription by ERK1 and ERK2 in pancreatic beta cells. J Biol Chem. 2003, 278 (35): 32969-32977. 10.1074/jbc.M301198200.PubMed Khoo S, Griffen SC, Xia Y, Baer RJ, German MS, Cobb MH: Regulation of insulin gene transcription by ERK1 and ERK2 in pancreatic beta cells. J Biol Chem. 2003, 278 (35): 32969-32977. 10.1074/jbc.M301198200.PubMed
105.
Zurück zum Zitat Lawrence MC, McGlynn K, Park BH, Cobb MH: ERK1/2-dependent activation of transcription factors required for acute and chronic effects of glucose on the insulin gene promoter. J Biol Chem. 2005, 280 (29): 26751-26759. 10.1074/jbc.M503158200.PubMed Lawrence MC, McGlynn K, Park BH, Cobb MH: ERK1/2-dependent activation of transcription factors required for acute and chronic effects of glucose on the insulin gene promoter. J Biol Chem. 2005, 280 (29): 26751-26759. 10.1074/jbc.M503158200.PubMed
106.
Zurück zum Zitat Susini S, Roche E, Prentki M, Schlegel W: Glucose and glucoincretin peptides synergize to induce c-fos, c-jun, junB, zif-268, and nur-77 gene expression in pancreatic beta(INS-1) cells. Faseb J. 1998, 12 (12): 1173-1182.PubMed Susini S, Roche E, Prentki M, Schlegel W: Glucose and glucoincretin peptides synergize to induce c-fos, c-jun, junB, zif-268, and nur-77 gene expression in pancreatic beta(INS-1) cells. Faseb J. 1998, 12 (12): 1173-1182.PubMed
107.
Zurück zum Zitat Josefsen K, Sorensen LR, Buschard K, Birkenbach M: Glucose induces early growth response gene (Egr-1) expression in pancreatic beta cells. Diabetologia. 1999, 42 (2): 195-203. 10.1007/s001250051139.PubMed Josefsen K, Sorensen LR, Buschard K, Birkenbach M: Glucose induces early growth response gene (Egr-1) expression in pancreatic beta cells. Diabetologia. 1999, 42 (2): 195-203. 10.1007/s001250051139.PubMed
108.
Zurück zum Zitat Leibowitz G, Uckaya G, Oprescu AI, Cerasi E, Gross DJ, Kaiser N: Glucose-regulated proinsulin gene expression is required for adequate insulin production during chronic glucose exposure. Endocrinology. 2002, 143 (9): 3214-3220. 10.1210/en.2002-220174.PubMed Leibowitz G, Uckaya G, Oprescu AI, Cerasi E, Gross DJ, Kaiser N: Glucose-regulated proinsulin gene expression is required for adequate insulin production during chronic glucose exposure. Endocrinology. 2002, 143 (9): 3214-3220. 10.1210/en.2002-220174.PubMed
109.
Zurück zum Zitat Leibiger B, Moede T, Uhles S, Berggren PO, Leibiger IB: Short-term regulation of insulin gene transcription. Biochem Soc Trans. 2002, 30 (2): 312-317. 10.1042/BST0300312.PubMed Leibiger B, Moede T, Uhles S, Berggren PO, Leibiger IB: Short-term regulation of insulin gene transcription. Biochem Soc Trans. 2002, 30 (2): 312-317. 10.1042/BST0300312.PubMed
110.
Zurück zum Zitat Schuit FC, In't Veld PA, Pipeleers DG: Glucose stimulates proinsulin biosynthesis by a dose-dependent recruitment of pancreatic beta cells. Proc Natl Acad Sci U S A. 1988, 85 (11): 3865-3869. 10.1073/pnas.85.11.3865.PubMedCentralPubMed Schuit FC, In't Veld PA, Pipeleers DG: Glucose stimulates proinsulin biosynthesis by a dose-dependent recruitment of pancreatic beta cells. Proc Natl Acad Sci U S A. 1988, 85 (11): 3865-3869. 10.1073/pnas.85.11.3865.PubMedCentralPubMed
111.
Zurück zum Zitat Gilligan M, Welsh GI, Flynn A, Bujalska I, Diggle TA, Denton RM, Proud CG, Docherty K: Glucose stimulates the activity of the guanine nucleotide-exchange factor eIF-2B in isolated rat islets of Langerhans. J Biol Chem. 1996, 271 (4): 2121-2125. 10.1074/jbc.271.4.2121.PubMed Gilligan M, Welsh GI, Flynn A, Bujalska I, Diggle TA, Denton RM, Proud CG, Docherty K: Glucose stimulates the activity of the guanine nucleotide-exchange factor eIF-2B in isolated rat islets of Langerhans. J Biol Chem. 1996, 271 (4): 2121-2125. 10.1074/jbc.271.4.2121.PubMed
112.
Zurück zum Zitat Yan L, Nairn AC, Palfrey HC, Brady MJ: Glucose regulates EF-2 phosphorylation and protein translation by a protein phosphatase-2A-dependent mechanism in INS-1-derived 832/13 cells. J Biol Chem. 2003, 278 (20): 18177-18183. 10.1074/jbc.M301116200.PubMed Yan L, Nairn AC, Palfrey HC, Brady MJ: Glucose regulates EF-2 phosphorylation and protein translation by a protein phosphatase-2A-dependent mechanism in INS-1-derived 832/13 cells. J Biol Chem. 2003, 278 (20): 18177-18183. 10.1074/jbc.M301116200.PubMed
113.
Zurück zum Zitat Xu G, Marshall CA, Lin TA, Kwon G, Munivenkatappa RB, Hill JR, Lawrence JC, McDaniel ML: Insulin mediates glucose-stimulated phosphorylation of PHAS-I by pancreatic beta cells. An insulin-receptor mechanism for autoregulation of protein synthesis by translation. J Biol Chem. 1998, 273 (8): 4485-4491. 10.1074/jbc.273.8.4485.PubMed Xu G, Marshall CA, Lin TA, Kwon G, Munivenkatappa RB, Hill JR, Lawrence JC, McDaniel ML: Insulin mediates glucose-stimulated phosphorylation of PHAS-I by pancreatic beta cells. An insulin-receptor mechanism for autoregulation of protein synthesis by translation. J Biol Chem. 1998, 273 (8): 4485-4491. 10.1074/jbc.273.8.4485.PubMed
114.
Zurück zum Zitat Gomez E, Powell ML, Greenman IC, Herbert TP: Glucose-stimulated protein synthesis in pancreatic beta-cells parallels an increase in the availability of the translational ternary complex (eIF2-GTP.Met-tRNAi) and the dephosphorylation of eIF2 alpha. J Biol Chem. 2004, 279 (52): 53937-53946. 10.1074/jbc.M408682200.PubMed Gomez E, Powell ML, Greenman IC, Herbert TP: Glucose-stimulated protein synthesis in pancreatic beta-cells parallels an increase in the availability of the translational ternary complex (eIF2-GTP.Met-tRNAi) and the dephosphorylation of eIF2 alpha. J Biol Chem. 2004, 279 (52): 53937-53946. 10.1074/jbc.M408682200.PubMed
115.
Zurück zum Zitat Nielsen DA, Welsh M, Casadaban MJ, Steiner DF: Control of insulin gene expression in pancreatic beta-cells and in an insulin-producing cell line, RIN-5F cells. I. Effects of glucose and cyclic AMP on the transcription of insulin mRNA. J Biol Chem. 1985, 260 (25): 13585-13589.PubMed Nielsen DA, Welsh M, Casadaban MJ, Steiner DF: Control of insulin gene expression in pancreatic beta-cells and in an insulin-producing cell line, RIN-5F cells. I. Effects of glucose and cyclic AMP on the transcription of insulin mRNA. J Biol Chem. 1985, 260 (25): 13585-13589.PubMed
116.
Zurück zum Zitat Yasuda K, Yamada Y, Inagaki N, Yano H, Okamoto Y, Tsuji K, Fukumoto H, Imura H, Seino S, Seino Y: Expression of GLUT1 and GLUT2 glucose transporter isoforms in rat islets of Langerhans and their regulation by glucose. Diabetes. 1992, 41 (1): 76-81.PubMed Yasuda K, Yamada Y, Inagaki N, Yano H, Okamoto Y, Tsuji K, Fukumoto H, Imura H, Seino S, Seino Y: Expression of GLUT1 and GLUT2 glucose transporter isoforms in rat islets of Langerhans and their regulation by glucose. Diabetes. 1992, 41 (1): 76-81.PubMed
117.
Zurück zum Zitat Brun T, Roche E, Kim KH, Prentki M: Glucose regulates acetyl-CoA carboxylase gene expression in a pancreatic beta-cell line (INS-1). J Biol Chem. 1993, 268 (25): 18905-18911.PubMed Brun T, Roche E, Kim KH, Prentki M: Glucose regulates acetyl-CoA carboxylase gene expression in a pancreatic beta-cell line (INS-1). J Biol Chem. 1993, 268 (25): 18905-18911.PubMed
118.
Zurück zum Zitat Marie S, Diaz-Guerra MJ, Miquerol L, Kahn A, Iynedjian PB: The pyruvate kinase gene as a model for studies of glucose-dependent regulation of gene expression in the endocrine pancreatic beta-cell type. J Biol Chem. 1993, 268 (32): 23881-23890.PubMed Marie S, Diaz-Guerra MJ, Miquerol L, Kahn A, Iynedjian PB: The pyruvate kinase gene as a model for studies of glucose-dependent regulation of gene expression in the endocrine pancreatic beta-cell type. J Biol Chem. 1993, 268 (32): 23881-23890.PubMed
119.
Zurück zum Zitat Choi Y, Ta M, Atouf F, Lumelsky N: Adult pancreas generates multipotent stem cells and pancreatic and nonpancreatic progeny. Stem Cells. 2004, 22: 1070-1084. 10.1634/stemcells.22-6-1070.PubMed Choi Y, Ta M, Atouf F, Lumelsky N: Adult pancreas generates multipotent stem cells and pancreatic and nonpancreatic progeny. Stem Cells. 2004, 22: 1070-1084. 10.1634/stemcells.22-6-1070.PubMed
Metadaten
Titel
Pancreatic islet cell therapy for type I diabetes: understanding the effects of glucose stimulation on islets in order to produce better islets for transplantation
verfasst von
Jiaqiang Ren
Ping Jin
Ena Wang
Eric Liu
David M Harlan
Xin Li
David F Stroncek
Publikationsdatum
01.12.2007
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2007
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/1479-5876-5-1

Weitere Artikel der Ausgabe 1/2007

Journal of Translational Medicine 1/2007 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.