Review
Restoring normal islet mass and function in type 1 diabetes through regenerative medicine and tissue engineering

https://doi.org/10.1016/S2213-8587(21)00170-4Get rights and content

Summary

Type 1 diabetes is characterised by autoimmune-mediated destruction of pancreatic β-cell mass. With the advent of insulin therapy a century ago, type 1 diabetes changed from a progressive, fatal disease to one that requires lifelong complex self-management. Replacing the lost β-cell mass through transplantation has proven successful, but limited donor supply and need for lifelong immunosuppression restricts widespread use. In this Review, we highlight incremental advances over the past 20 years and remaining challenges in regenerative medicine approaches to restoring β-cell mass and function in type 1 diabetes. We begin by summarising the role of endocrine islets in glucose homoeostasis and how this is altered in disease. We then discuss the potential regenerative capacity of the remaining islet cells and the utility of stem cell-derived β-like cells to restore β-cell function. We conclude with tissue engineering approaches that might improve the engraftment, function, and survival of β-cell replacement therapies.

Introduction

The advent of insulin therapy delivered a medical miracle in the early 20th century, rescuing people with type 1 diabetes from an inexorably progressive and ultimately fatal wasting disease. Since then, numbers of people with type 1 diabetes have steadily increased1, 2 in parallel with a global epidemic of type 2 diabetes over the second half of the 20th century and beyond.3 In 2019, it was estimated that 463 million adults had been diagnosed with diabetes worldwide. This number includes up to 10% with type 1 diabetes, in addition to more than 1·1 million children and adolescents being dependent on insulin replacement therapy.3 Although people with type 2 diabetes and rarer forms of diabetes are not absolutely dependent on insulin replacement, it is now acknowledged that all forms of diabetes are characterised by a suboptimal pancreatic islet cell insulin response.4

The dawn of the 21st century heralded β-cell replacement as an alternative to self-administered insulin therapy through seminal success with deceased donor islet transplantation in Edmonton, Canada.5 As regenerative medicine begins to deliver unparalleled outcomes across the spectrum of disease, we review its potential for transforming insulin-deficient diabetes.

Unfortunately, as we observe the centenary of insulin treatment, diabetes remains a leading cause of blindness, renal failure, and lower limb amputation,6 with increased incidence of myocardial infarction and reduced life expectancy.7 This is despite incontrovertible evidence that chronic complications can be prevented by early correction of high blood glucose concentrations.8 Achieving this correction with subcutaneously injected or infused insulin is unavoidably associated with risk of hypoglycaemia. Every year, nearly 50% of people with type 1 diabetes of more than 15 years duration require assistance for severe hypoglycaemia, one of the greatest fears of dependence on exogenous insulin.9 Great strides have been made in enhancing insulin delivery and glucose testing over the last century, including successful combination in integrated closed-loop systems.10 In contrast to the physiological β cell, which continuously senses glucose intravascularly with second-to-second control of insulin secretion directly into the portal vein, the wearable bioartifical pancreas has to overcome delays in detecting blood glucose changes due to subcutaneous sensing and in effecting a change in circulating insulin concentrations due to subcutaneous insulin delivery. This process necessitates frequent substantial changes in insulin infusion rate, typically associated with an average of at least 30 minutes each day with glucose less than 4 mmol/L even using the fastest marketed short-acting insulin analogue.11 This approach does not thus provide curative therapy, nor completely removes the burden of glucose uncertainty and unremitting daily self-management.12

Transplantation of vascularised whole pancreas from a deceased donor has confirmed that β-cell replacement can immediately normalise blood glucose concentrations in type 1 diabetes and in carefully selected recipients with insulin-treated type 2 diabetes.13 Despite established success, applicability to more than a minority is limited by the complexity of the surgery with unavoidable risk of mortality and substantial morbidity, in addition to the limited availability of suitable donor organs.14

Mechanical and enzymatic dissociation of a donor pancreas has enabled separation of the endocrine cells within the islets of Langerhans from their blood supply and from the digestive enzyme-producing majority of the organ.15 Islet transplantation is a minimally invasive procedure that was initially undertaken as an autologous procedure following total pancreatectomy for painful pancreatitis.15 Subsequently, allogeneic islets were infused into the hepatic portal vein with systemic immunosuppression in type 1 diabetes.5

Islet transplantation offers prevention of severe hypoglycaemia, stabilisation of blood glucose concentrations, and attainment of optimal HbA1c (<53 mmol/mol [7·0%]) and is established as standard-of-care for recurrent dangerous hypoglycaemia despite optimised conventional therapy in the integrated UK programme and in other countries around the world.16, 17, 18 However, sustainable insulin independence cannot be reproducibly achieved from a single transplantation procedure, and the requirement for lifelong systemic immunosuppression currently limits recipients to those with life-threatening complications.19

Portal vein infusion via a percutaneous transhepatic or mini-laparotomy approach is minimally invasive, only very rarely complicated by haemorrhage or thrombosis in experienced centres.16, 17, 18 Intravascularly transplanted cells are, however, subject to an instant blood-mediated inflammatory reaction.20 PET scanning strongly suggests that this thrombotic and innate immune response leads to a substantial loss of transplanted islet mass immediately following infusion.21 Moreover, β cells are uniquely sensitive to hypoxia during pancreas retrieval and preservation; during islet isolation, culture, and shipment for transplantation; and within the relatively low oxygen tension portal vein environment before revascularisation from the hepatic artery.22 Transient ex-vivo exposure of primary mouse islets to hypoxia has been shown to lead to upregulation of hypoxia-inducible genes and adaptation enabling β-cell survival but with impaired glucose-stimulated insulin secretion persisting after transplantation.23 Loss of fully mature end-differentiated phenotype (absence of β-cell urocortin-3 expression) several years after transplantation despite full vascularisation has been reported following clinical islet transplantation.24 Together these limitations have led to maximal engraftment of only approximately 25–40% of normal adult β-cell functional mass despite often repeated transplantations from sequential deceased donors.25 Although this level of functional mass can deliver short-term insulin independence, true physiological endocrine function has not yet been achieved.26

Human islets have been transplanted clinically without encapsulation into alternative sites including skeletal muscle and omentum.27 Although islets might become well vascularised, superior outcomes to portal transplantation have yet to be achieved, potentially due to irreversible adaptation during even short-term ischaemia and unpredictable inflammatory response.

In this Review we consider how the remaining challenges eluding a type 1 diabetes cure might be overcome through regenerative medicine approaches including stem cell-derived β cells, gene therapy, and tissue engineering. The highly vascularised endogenous islet niche and mechanisms for maintenance of β-cell mass and function will be considered as the context for intelligent recapitulation through: (1) generation of an unlimited source of physiologically functional islets; (2) vascularised engraftment of sufficient β-cell mass without tissue ischaemia; and (3) overcoming cell loss through innate and adaptive immune response.

Section snippets

The endogenous β-cell niche

In considering β-cell replacement strategies, it is important to understand physiological β-cell function in the context of its endogenous niche. β cells do not exist in isolation in the normal human pancreas, but in islet mini-organs. β cells respond to glucose stimulation above a threshold varying between species (approximately 7·8–8·3 mmol/L in mice; approximately 4·4–5·0 mmol/L in humans28) with closure of ATP-sensitive potassium leak channels triggering opening of voltage-gated calcium

Reversal of functional impairment

The honeymoon period—ie, the period following type 1 diabetes diagnosis with transient reduction in requirement for exogenous insulin after initial glucose stabilisation—supports the potential for β-cell functional recovery. This β-cell rest is insufficient to prevent declining endogenous insulin concentrations as type 1 diabetes progresses, however, although a case series of three individuals aged 36–40 years presenting with type 1 diabetes and confirmed underlying autoimmunity reported

Human pluripotent stem cell-derived pancreatic progenitor cells for the treatment of diabetes

Human pluripotent stem cells (hPSCs) are capable of generating all cell types in vitro, if provided with the right environment. One source of hPSCs is human embryonic stem cells, which are derived from the blastocyst of a preimplantation embryo. Protocols to culture pluripotent human embryonic stem cells in the laboratory became available in 1998,123 but were limited by the need for embryonic donor tissue. In a series of seminal studies leading to the award of the Nobel Prize in Physiology or

Harnessing tissue engineering in β-cell replacement therapy

Systems capable of recreating a pancreatic niche for transplanted islets hold great promise for enhancing survival, engraftment, and long-term function. The following sections address some of the major challenges associated with transplantation, with a particular focus on access to nutrients and a supportive environment for survival and function, as well as modulating inflammation and adaptive cell responses to limit immune-mediated cell destruction.

Conclusions

Whether the approach is to maintain and restore β-cell function in situ or to replace β-cell mass through transplantation, truly transformative regenerative medicine for type 1 diabetes is dependent on fundamental understanding of human islets within the pancreas, from which we draw a number of conclusions.

Firstly, despite the presence of considerable functional reserve in individuals without diabetes for increasing insulin secretion several-fold in response to increasing demand (eg, during

Search strategy and selection criteria

We identified references for this Review through searches of PubMed for articles published between Jan 1, 2015, and May 1, 2021, by use of the terms “beta-cell differentiation” AND “stem cells”, “beta-cell transdifferentiation”, “beta-cell neogenesis”, “beta-cell dedifferentiation”, “human beta-cell proliferation”, “beta-cell heterogeneity”, “beta-cell transplantation”, “islet transplantation” AND “inflammation”, and “islet transplantation” AND “immunomodulation”. We reviewed articles published

Declaration of interests

LDS holds a licensed patent on nanoparticles for autoimmune tolerance, with patent applications pending on scaffolds for islet transplantation and local immune monitoring. MOH receives funding and consulting fees from Crinetics. JAMS has received hororaria for chairing academic meetings organised by Novo Nordisk and for participating in a Medtronic Scientific Advisory Board. NAJK declares no competing interests.

References (211)

  • JA Kellard et al.

    Reduced somatostatin signalling leads to hypersecretion of glucagon in mice fed a high-fat diet

    Mol Metab

    (2020)
  • M Makhmutova et al.

    Pancreatic β-cells communicate with vagal sensory neurons

    Gastroenterology

    (2021)
  • R Rodriguez-Diaz et al.

    Neural control of the endocrine pancreas

    Best Pract Res Clin Endocrinol Metab

    (2014)
  • N Damond et al.

    A map of human type 1 diabetes progression by imaging mass cytometry

    Cell Metab

    (2019)
  • J Rui et al.

    β cells that resist immunological attack develop during progression of autoimmune diabetes in NOD mice

    Cell Metab

    (2017)
  • RA Watkins et al.

    Proinsulin and heat shock protein 90 as biomarkers of beta-cell stress in the early period after onset of type 1 diabetes

    Transl Res

    (2016)
  • MEJ Lean et al.

    Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, cluster-randomised trial

    Lancet

    (2018)
  • B Bouillet et al.

    A low-carbohydrate high-fat diet initiated promptly after diagnosis provides clinical remission in three patients with type 1 diabetes

    Diabetes Metab

    (2020)
  • M von Herrath et al.

    Anti-interleukin-21 antibody and liraglutide for the preservation of β-cell function in adults with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled, phase 2 trial

    Lancet Diabetes Endocrinol

    (2021)
  • EJ Mayer-Davis et al.

    Incidence trends of type 1 and type 2 diabetes among youths, 2002–2012

    N Engl J Med

    (2017)
  • IDF Diabetes Atlas

    (2019)
  • E Cersosimo et al.

    Pathogenesis of type 2 diabetes mellitus. [Updated Feb 27, 2018]

  • AMJ Shapiro et al.

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

    N Engl J Med

    (2000)
  • SJ Livingstone et al.

    Estimated life expectancy in a Scottish cohort with type 1 diabetes, 2008-2010

    JAMA

    (2015)
  • DM Nathan et al.

    The diabetes control and complications trial/epidemiology of diabetes interventions and complications study at 30 years: overview

    Diabetes Care

    (2014)
  • Risk of hypoglycaemia in types 1 and 2 diabetes: effects of treatment modalities and their duration

    Diabetologia

    (2007)
  • E Bekiari et al.

    Artificial pancreas treatment for outpatients with type 1 diabetes: systematic review and meta-analysis

    BMJ

    (2018)
  • CK Boughton et al.

    Hybrid closed-loop glucose control with faster insulin aspart compared with standard insulin aspart in adults with type 1 diabetes: a double-blind, multicentre, multinational, randomized, crossover study

    Diabetes Obes Metab

    (2021)
  • KD Barnard et al.

    Psychosocial aspects of closed- and open-loop insulin delivery: closing the loop in adults with type 1 diabetes in the home setting

    Diabet Med

    (2015)
  • AJS Flatt et al.

    β-cell and renal transplantation options for diabetes

    Diabet Med

    (2020)
  • AMJ Shapiro

    A historical perspective on experimental and clinical islet transplantation

  • MC Vantyghem et al.

    Primary graft function, metabolic control, and graft survival after islet transplantation

    Diabetes Care

    (2009)
  • BJ Hering et al.

    Phase 3 trial of transplantation of human islets in type 1 diabetes complicated by severe hypoglycemia

    Diabetes Care

    (2016)
  • FB Barton et al.

    Improvement in outcomes of clinical islet transplantation: 1999–2010

    Diabetes Care

    (2012)
  • MA Kanak et al.

    Inflammatory response in islet transplantation

    Int J Endocrinol

    (2014)
  • AR Pepper et al.

    Revascularization of transplanted pancreatic islets and role of the transplantation site

    Clin Dev Immunol

    (2013)
  • J Cantley et al.

    A preexistent hypoxic gene signature predicts impaired islet graft function and glucose homeostasis

    Cell Transplant

    (2013)
  • AJS Flatt et al.

    Pancreatic islet reserve in type 1 diabetes

    Ann N Y Acad Sci

    (2021)
  • MR Rickels et al.

    Improvement in β-cell secretory capacity after human islet transplantation according to the CIT07 protocol

    Diabetes

    (2013)
  • P Addison et al.

    Considerations for an alternative site of islet cell transplantation

    J Diabetes Sci Technol

    (2020)
  • M Brissova et al.

    Assessment of human pancreatic islet architecture and composition by laser scanning confocal microscopy

    J Histochem Cytochem

    (2005)
  • DJ Steiner et al.

    Pancreatic islet plasticity: interspecies comparison of islet architecture and composition

    Islets

    (2010)
  • O Cabrera et al.

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

    Proc Natl Acad Sci USA

    (2006)
  • GM Noguchi et al.

    Integrating the inputs that shape pancreatic islet hormone release

    Nat Metab

    (2019)
  • SM Hartig et al.

    Paracrine signaling in islet function and survival

    J Mol Med (Berl)

    (2020)
  • ME Capozzi et al.

    β Cell tone is defined by proglucagon peptides through cAMP signaling

    JCI Insight

    (2019)
  • L Zhu et al.

    Intra-islet glucagon signaling is critical for maintaining glucose homeostasis

    JCI Insight

    (2019)
  • MO Huising et al.

    Evidence for a neogenic niche at the periphery of pancreatic islets

    BioEssays

    (2018)
  • T van der Meulen et al.

    Urocortin3 mediates somatostatin-dependent negative feedback control of insulin secretion

    Nat Med

    (2015)
  • JT Yue et al.

    Amelioration of hypoglycemia via somatostatin receptor type 2 antagonism in recurrently hypoglycemic diabetic rats

    Diabetes

    (2013)
  • Cited by (0)

    View full text