The pancreatic islet as a signaling hub

https://doi.org/10.1016/j.jbior.2012.09.011Get rights and content

Abstract

Over the last two decades we have focused on beta cell signal transduction, bringing many new insights, especially in the context of insulin signal transduction, the role of inositol polyphosphates and the regulation of cytoplasmic free Ca2+ concentration. However, there has been a growing awareness that the beta cell, which is mandatory for insulin secretion, has a unique context within the micro-organ of the pancreatic Islet of Langerhans. In this environment the beta cell both mediates and receives paracrine regulation, critical for the control of blood glucose homeostasis. Failure of an appropriate beta cell function leads to the development of diabetes mellitus.

In our quest to understand the molecular events maintaining beta cell function we have faced two key challenges. Firstly, whilst there are many similarities between signal transduction in pancreatic islets between the much used rodent models and humans there are some notable differences. Critical distinctions between rodent and primate can be made in the structure of the islet, including the arrangement of the islet cells, the innervation pattern and the microcirculation. This means that important signaling interactions between islets cells, mediated through for example insulin, glucagon, GABA, glutamate and ATP, will have a unique human framework. The second challenge was to be able to take the discoveries we have made using in vitro systems and examine them in an in vivo context. Advances in in vivo imaging achieved by utilizing the anterior chamber of the eye as a transplantation site for pancreatic islets make it possible for non-invasive, longitudinal studies at single cell resolution in real time of islet cell physiology and pathology. Thus it is becoming possible to study the insulin secreting pancreatic beta cell within the framework of the unique micro-organ, the Islet of Langerhans, for the first time in a physiological context, i.e. when being innervated and connected to the blood supply.

Introduction

Over the last decade our laboratory has made significant progress in developing an understanding of signal transduction in the pancreatic beta cell. Areas of particular note include the regulation of cytoplasmic free Ca2+ concentration, [Ca2+]i, the function of inositol polyphosphates and insulin receptor signaling (Barker and Berggren, 2012; Leibiger et al., 2008; Yang and Berggren, 2005). These investigations, in turn, have lead to novel insights into important regulatory events in pancreatic beta cells including apoptosis, the secretion of insulin, and the regulation of key genes (Barker and Berggren, 2012; Leibiger et al., 2008; Yang and Berggren, 2005). The beta cell, however, does not operate in isolation, but is part of a micro-organ known as the Islet of Langerhans. Islets consist not only of beta cells but also of alpha, delta and PP cells, together with other cells like endothelial cells, that constitute the blood capillaries, or neurons that innervate the islets. This architecture immediately suggests the opportunity for paracrine interactions between the different islet cells and thus increases the complexity of the signaling processes. There are also important ramifications that have emerged from studies looking at beta cell–beta cell interactions (Jain and Lammert, 2009) and the importance of the interactions of the cells with the endothelial cells that constitute the blood vessels (Eberhard et al., 2010). Notwithstanding this complexity, we were faced with two significant further challenges.

The first challenge was the fact that ours and other studies have largely been based on rodent islets and not human islets, mostly due to lack of availability of human material. This became a serious issue when our investigations into human islets revealed radical differences between rodent and primate islet structure/function relationships (Cabrera et al., 2006, 2008; Jacques-Silva et al., 2010; Rodriguez-Diaz et al., 2011a, 2011b) and thus the possibility of fundamentally different signaling contexts. Ultimately, we and others wish to understand the context of the pancreatic beta cell and islet function and dysfunction from the perspective of an important human disease, namely diabetes. Thus information that has come purely from rodent models is likely to be at best preliminary, and at worst misleading, about how islet signal transduction may be disrupted in diabetes.

The second challenge we faced was the lack of an in vivo context. Virtually all experiments regarding islet cell signal transduction, and indeed islet cell biology in general, have been conducted in an in vitro environment. Unfortunately, it is quite likely that the in vivo environment is rather different from that found in vitro. This reflects the need to orchestrate a coordinated regulation of plasma glucose homeostasis in the whole organism. Clearly, systematic human in vivo experiments of the breadth and depth needed to resolve the expected molecular complexity of islet cell signal transduction are not possible and the in vivo context therefore needed to be in terms of an animal model.

Faced with these two challenges, we have first studied signal transduction in human islets and how it may differ from that of rodents. Secondly, we have established an in vivo system to examine both islet cell biology and ultimately, signal transduction. This novel in vivo platform is the application of the anterior chamber of the eye as non-invasive imaging platform suitable for real-time, longitudinal studies. The remainder of this review will detail how far we have progressed in addressing these two challenges and how we see this developing in the future.

Section snippets

The challenge of the disparate architecture between rodent and human islets

There has been a common misconception that there was little difference between the architecture of human and rodent islets, perpetuated by textbooks on the subject (e.g. Nussey and Whitehead, 2001). In reality, a careful inspection of the literature would have revealed significant differences between rodent and primate (including human) islets (Orci et al., 1976; Orci and Unger, 1975). This difference was an inconvenient truth to a field that relied heavily on rodent models to better define

Functional in vivo imaging of pancreatic islet cell physiology and pathology – the anterior chamber of the eye as a natural body window

The other challenge we faced was to establish techniques that allowed us to study pancreatic islet cell physiology/pathology in vivo. Although progress has been made in the development of non-invasive monitoring of beta cell mass by for example magnetic resonance imaging (MRI) or positron emission tomography (PET), these techniques are at present missing the resolution and specificity due to the lack of high affinity islet cell-selective probes. We have chosen the anterior chamber of the eye as

Conclusion

A dramatic increase in the incidence of type 2 diabetes worldwide and associated burden for the individual patient as well as for society require novel strategies in diagnosis and treatment. The data obtained from genome-wide association studies in large study samples of type 2 diabetes patients revealed target genes with potentially associated functions in pancreatic islet cells, thus pinpointing the importance of the pancreatic islet micro-organ for proper function and survival of the

Acknowledgments

This work was supported by grants from Karolinska Institutet, Novo Nordisk Foundation, The Swedish Research Council, The Swedish Diabetes Association, The Family Erling-Persson Foundation, Berth von Kantzow’s Foundation, Torsten and Ragnar Söderberg's Foundation, The Knut and Alice Wallenberg Foundation, VIBRANT (FP7-228933-2), Skandia Insurance Company, Ltd., Strategic Research Program in Diabetes at Karolinska Institutet, The Stichting af Jochnick Foundation and The Diabetes Research

References (38)

  • M.H. Abdulreda et al.

    High-resolution, noninvasive longitudinal live imaging of immune responses

    Proc Natl Acad Sci U S A

    (2011)
  • B. Ahren

    Autonomic regulation of islet hormone secretion–implications for health and disease

    Diabetologia

    (2000)
  • O. Cabrera et al.

    The unique cytoarchitecture of human pancreatic islets has implications for islet cell function

    Proc Natl Acad Sci U S A

    (2006)
  • J. Fernandez-Alvarez et al.

    P2 receptor agonists stimulate insulin release from human pancreatic islets

    Pancreas

    (2001)
  • Y. Guo et al.

    CHRM3 gene variation is associated with decreased acute insulin secretion and increased risk for early-onset type 2 diabetes in Pima Indians

    Diabetes

    (2006)
  • J.A. Healy et al.

    Cholinergic augmentation of insulin release requires ankyrin-B

    Sci Signal

    (2010)
  • M.C. Jacques-Silva et al.

    ATP-gated P2X3 receptors constitute a positive autocrine signal for insulin release in the human pancreatic beta cell

    Proc Natl Acad Sci U S A

    (2010)
  • R. Jain et al.

    Cell–cell interactions in the endocrine pancreas

    Diabetes Obes Metab

    (2009)
  • M. Kohler et al.

    On-line monitoring of apoptosis in insulin-secreting cells

    Diabetes

    (2003)
  • Cited by (25)

    • The eye as a novel imaging site in diabetes research

      2019, Pharmacology and Therapeutics
      Citation Excerpt :

      To meet this challenge, it is important to find ways to implement non-invasive, longitudinal experiments on pancreatic islets in live animals and humans at high-resolution. The body's tissues/organs including islets behave differently in vivo versus in vitro (Barker, Leibiger, & Berggren, 2013; Leibiger & Berggren, 2017; Weigert et al., 2010). However, in vivo and in situ visualization of islets is not practical with non-invasive optical approaches since the islets are deeply embedded in the pancreas and covered by the opaque exocrine pancreas, other tissues and organs as well as the abdominal wall.

    • Development of the human pancreas and its vasculature — An integrated review covering anatomical, embryological, histological, and molecular aspects

      2019, Annals of Anatomy
      Citation Excerpt :

      Zhou et al. (2001) suggested that an impaired pattern of insular drainage vessels could be the morphological basis of the pancreatic exocrine pathologic lesion observed in human diabetes. Understanding the developmental process of visceral vessels is essential for successful transplantation of pancreatic islets (Barker et al., 2013). The venous system develops simultaneously with the arterial system at the end of 3rd gestational week (Azizoglu et al., 2016).

    • Imaging endocrinology in animal models of endocrine disease

      2018, Best Practice and Research: Clinical Endocrinology and Metabolism
    • Chapter 4 - Pancreatic Physiology and Functional Assessment

      2016, Blumgart's Surgery of the Liver, Biliary Tract and Pancreas: Sixth Edition
    • Lipocalin-type prostaglandin D<inf>2</inf> synthase reduces glucagon secretion in alpha TC-1 clone 6 cells via the DP1 receptor

      2015, Biochemistry and Biophysics Reports
      Citation Excerpt :

      Lipocalin-type prostaglandin D2 synthase (l-PGDS) is the enzyme which plays an important role in the metabolism of arachidonic acid by converting PGH2 into PGD2. PGD2 gets converted into numerous prostaglandins including and 15-deoxy-D12, 14-PGJ2 (15d-PGJ2) [1,2]. PGD2 is primarily produced in mast cells and in smooth muscle cells with a synthetic phenotype [3] which has also been reported for its accumulation during atherosclerotic lesion formation, suggesting their strong involvement in the pathogenesis of cardiovascular injuries [4].

    View all citing articles on Scopus
    1

    These authors have made an equal contribution to this manuscript.

    View full text