Elsevier

Current Opinion in Pharmacology

Volume 43, December 2018, Pages 34-39
Current Opinion in Pharmacology

Potential of mesenchymal stromal cells for improving islet transplantation outcomes

https://doi.org/10.1016/j.coph.2018.07.011Get rights and content

Highlights

  • Mesenchymal stromal cells (MSCs)/their secretome improve islet revascularisation.

  • MSC-derived secretory products reduce pathogenic host T cell responses.

  • MSCs and their secretory products improve donor islet cell functional survival.

  • Harnessing the MSC secretome has potential to improve transplantation efficiency.

Allogeneic islet transplantation as a therapy for Type 1 Diabetes (T1D) is restricted by the limited availability of donor islets, loss of functional islets during pre-transplantation culture in vitro and further extensive loss during the immediate post-transplantation period when islet function and survival is compromised by the hypoxic, inflammatory host environment. In the longer term pathogenic T cell responses drive autoimmunity and chronic allograft rejection. Experimental studies have demonstrated that mesenchymal stromal cells (MSCs) have significant potential to improve the outcomes of clinical islet transplantation. This review explores the potential for MSCs and their ‘secretome’ to influence donor islet cell function and survival, as well as the host niche. We discuss the possibility of harnessing the therapeutic benefits of MSCs in a cell-free strategy to offer a well-defined, cell-free approach to improve the outcomes of clinical islet transplantation.

Introduction

Type 1 diabetes mellitus (T1DM) is an autoimmune disorder in which insulin-secreting pancreatic β-cells are selectively destroyed. The consequent insulin deficiency results in dysregulation of metabolic control with hyperglycemia, hyperlipidemia and ketosis leading to a fatal outcome. People with T1DM have been treated by the administration of exogenous insulin since the first successful isolation of biologically active insulin in 1921. However, insulin therapy treats the symptoms of T1DM rather than offering a cure, and it has become clear over the decades that exogenous insulin often fails to maintain tight glycaemic control, and that the subsequent hyperglycemic excursions are responsible for the progressive development of a range of devastating side effects known as ‘secondary complications’ [1]. Replacement of the damaged β-cells offers the potential for restoring physiological glycemic control and thus avoiding the development of secondary complications. However, pancreas transplantation is surgically invasive and associated with significant co-morbidity [2]. An alternative is to transplant only the endocrine component of the pancreas — the islets of Langerhans-which comprise 2–3% of the total pancreas, and which can be isolated from the intact pancreas by collagenase digestion. Islet transplantation first became a viable therapeutic option with the publication of the landmark ‘Edmonton Protocol’ in 2000 [3], which stimulated human islet transplantation programmes around the world, such that over 1500 people with T1DM have now received islet grafts in 40 centres globally (https://citregistry.org/content/citr-9th-annual-report). Islet transplantation is a safe procedure with little or no co-morbidity [4••] and the clinical outcomes have improved year-on-year [4••,5]. Recent figures suggest that approximately 50% of graft recipients remain insulin independent at 5 years and more, making islet transplantation as clinically effective as whole pancreas transplantation [6]. However, the wider adoption of islet transplantation as a therapeutic option is currently limited by a shortage of tissue donors and much effort is currently directed at generating functionally-competent substitutes for primary human islets to expand the available pool of graft material.

Islet β-cells are metabolically active and use glucose metabolism and ATP generation to maintain appropriate rates of insulin secretion for the prevailing extracellular glucose concentrations. Islets deteriorate rapidly during and after their isolation [7], losing 20–50% of the β-cell mass within 24 hours of culture in vitro. The loss of β-cell function continues during the immediate post-transplantation period (24–72 hours) when up to 70% of the graft function is lost because of deleterious responses of the transplanted β-cells to the hypoxic, inflammatory, immunogenic host environment [8]. Strategies which improve the functional survival of islets both before and after transplantation will improve the outcome of individual grafts and enable the limited pool of donor islets to treat many more people with T1DM. One emerging strategy is the use of mesenchymal stromal cells (MSCs) to take advantage of their anti-inflammatory, immunoregulatory, angiogenic and regenerative properties.

Section snippets

Mesenchymal stromal cells and islet transplantation

MSCs are multipotent adult stromal progenitor cells located in the perivascular niche of most adult tissues, where they are involved in regeneration and repair in response to tissue ageing or damage. Importantly, MSCs can be isolated from their host tissues and maintained and expanded in vitro. There is no single specific marker for identifying MSCs, so the International Society for Cellular Therapy has specified three essential criteria to define cells as MSCs. First, cells must be

Influencing the host environment

Isolated islets are metabolically fragile because of the stresses (mechanical, enzymatic), loss of extracellular matrix (ECM) [22] and ischaemia associated with the isolation process. These fragile islets are transplanted into a hostile inflammatory environment leading to further β-cell death [23]. The clinically-preferred route for graft delivery via the hepatic portal vein has the advantages of being simple, minimally-invasive and targeting insulin to the liver, but intravascular delivery

Modulating the host immune response

MSCs can influence cells of the innate and adaptive immune systems and their co-transplantation at extrahepatic sites creates an immunosuppressive niche [14]. For example, MSCs influence macrophage phenotype by inducing a shift from pro-inflammatory (M1) to anti-inflammatory (M2) macrophages (reviewed in [19]), which reduces graft neutrophil infiltration and inflammation during the immediate post-transplantation period. MSCs also have the potential to intercept the priming and amplification of

Improving revascularisation

Transplanted islets are avascular during the immediate post transplantation period exposing β-cells to a hypoxic microenvironment [25] which can lead to ischaemic cell death, so enhancing graft revascularisation should improve transplantation outcomes [10, 11, 12,21,27]. Numerous studies have demonstrated that co-transplantation of MSCs [11, 12, 13,21] or MAPCs [27] improves islet revascularisation via a range of pro-angiogenic factors [12,43]. For example, Cunha and colleagues [27] identified

Improving graft functional survival

Several experimental studies have shown that MSCs maintain functional β-cell mass during culture in vitro [46, 47, 48], with associated improvements in islet graft function in vivo [46,47], and it is now well established that MSCs have direct effects on β-cells to enhance their insulin secretory capacity [12,46, 47, 48, 49] and protect them from cytokine-induced apoptosis [50]. Direct cell–cell contact plays a role in the capacity of MSCs to promote insulin secretory function [46,47,51], with

Conclusion

There is convincing experimental evidence that MSCs improve the outcomes of experimental islet transplantation and therefore have the potential to improve human islet transplantation as a therapy for T1DM. It has become evident that MSCs secrete molecules which have beneficial effects on the islet graft and/or on the host environment. Defining a cocktail of MSC secretory factors which improves the functional survival of donor islets, enhances their revascularisation and reduces inflammatory and

Conflict of interest statement

Nothing declared.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

Our work on MSCs and islet function is supported by Diabetes UK (Grant number 15/0005146 to P.M.J. and C.L.R.) and a David Matthews Research Fellowship from the Diabetes Research and Wellness Foundation (C.L.R.).

References (60)

  • E.J. Jung et al.

    Bone marrow-derived mesenchymal stromal cells support rat pancreatic islet survival and insulin secretory function in vitro

    Cytotherapy

    (2011)
  • S.P. Ki-Soo et al.

    Trophic molecules derived from human mesenchymal stem cells enhance survival, function, and angiogenesis of isolated islets after transplantation

    Transplantation

    (2010)
  • S. Amisten et al.

    A comparative analysis of human and mouse islet G-protein coupled receptor expression

    Sci Rep

    (2017)
  • S.W. Schive et al.

    Human adipose-derived mesenchymal stem cells respond to short-term hypoxia by secreting factors beneficial for human islets in vitro and potentiate antidiabetic effect in vivo

    Cell Med

    (2017)
  • K.A. Adeshara et al.

    Diabetes and complications: cellular signaling pathways, current understanding and targeted therapies

    Curr Drug Targets

    (2016)
  • S. Dholakia et al.

    The rise and potential fall of pancreas transplantation

    Br Med Bull

    (2017)
  • A.M. Shapiro et al.

    Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen

    N Engl J Med

    (2000)
  • A.M. Shapiro et al.

    Clinical pancreatic islet transplantation

    Nat Rev Endocrinol

    (2017)
  • F.B. Barton et al.

    Improvement in outcomes of clinical islet transplantation: 1999–2010

    Diabetes Care

    (2012)
  • A.C. Gruessner

    2011 update on pancreas transplantation: comprehensive trend analysis of 25,000 cases followed up over the course of twenty-four years at the international pancreas transplant registry (IPTR)

    Rev Diabet Stud

    (2011)
  • T. Kin et al.

    Risk factors for islet loss during culture prior to transplantation

    Transpl Int

    (2008)
  • A. Citro et al.

    Anti-inflammatory strategies to enhance islet engraftment and survival

    Curr Diab Rep

    (2013)
  • M. Dominici et al.

    Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement

    Cytotherapy

    (2006)
  • D.J. Borg et al.

    Mesenchymal stromal cells improve transplanted islet survival and islet function in a syngeneic mouse model

    Diabetologia

    (2014)
  • T. Ito et al.

    Mesenchymal stem cell and islet co-transplantation promotes graft revascularization and function

    Transplantation

    (2010)
  • S. Jacobson et al.

    Co-transplantation of stromal cells interferes with the rejection of allogeneic islet grafts

    Ann N Y Acad Sci

    (2008)
  • N. Moufida Ben et al.

    Co-transplantation of autologous mscs delays islet allograft rejection and generates a local immunoprivileged site

    Acta Diabetol

    (2015)
  • J. Mohammadi Ayenehdeh et al.

    Immunomodulatory and protective effects of adipose tissue-derived mesenchymal stem cells in an allograft islet composite transplantation for experimental autoimmune type 1 diabetes

    Immunol Lett

    (2017)
  • K. Le Blanc et al.

    Mesenchymal stromal cells and the innate immune response

    Immunol Lett

    (2015)
  • C.L. Rackham et al.

    Co-transplantation of mesenchymal stem cells maintains islet organisation and morphology in mice

    Diabetologia

    (2011)
  • Cited by (14)

    View all citing articles on Scopus
    View full text