1
|
Nishimura E, Pridal L, Glendorf T, Hansen BF, Hubálek F, Kjeldsen T, Kristensen NR, Lützen A, Lyby K, Madsen P, Pedersen TÅ, Ribel-Madsen R, Stidsen CE, Haahr H. Molecular and pharmacological characterization of insulin icodec: a new basal insulin analog designed for once-weekly dosing. BMJ Open Diabetes Res Care 2021; 9:9/1/e002301. [PMID: 34413118 PMCID: PMC8378355 DOI: 10.1136/bmjdrc-2021-002301] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/04/2021] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Insulin icodec is a novel, long-acting insulin analog designed to cover basal insulin requirements with once-weekly subcutaneous administration. Here we describe the molecular engineering and the biological and pharmacological properties of insulin icodec. RESEARCH DESIGN AND METHODS A number of in vitro assays measuring receptor binding, intracellular signaling as well as cellular metabolic and mitogenic responses were used to characterize the biological properties of insulin icodec. To evaluate the pharmacological properties of insulin icodec in individuals with type 2 diabetes, a randomized, double-blind, double-dummy, active-controlled, multiple-dose, dose escalation trial was conducted. RESULTS The long half-life of insulin icodec was achieved by introducing modifications to the insulin molecule aiming to obtain a safe, albumin-bound circulating depot of insulin icodec, providing protracted insulin action and clearance. Addition of a C20 fatty diacid-containing side chain imparts strong, reversible albumin binding, while three amino acid substitutions (A14E, B16H and B25H) provide molecular stability and contribute to attenuating insulin receptor (IR) binding and clearance, further prolonging the half-life. In vitro cell-based studies showed that insulin icodec activates the same dose-dependent IR-mediated signaling and metabolic responses as native human insulin (HI). The affinity of insulin icodec for the insulin-like growth factor-1 receptor was proportionately lower than its binding to the IR, and the in vitro mitogenic effect of insulin icodec in various human cells was low relative to HI. The clinical pharmacology trial in people with type 2 diabetes showed that insulin icodec was well tolerated and has pharmacokinetic/pharmacodynamic properties that are suited for once-weekly dosing, with a mean half-life of 196 hours and close to even distribution of glucose-lowering effect over the entire dosing interval of 1 week. CONCLUSIONS The molecular modifications introduced into insulin icodec provide a novel basal insulin with biological and pharmacokinetic/pharmacodynamic properties suitable for once-weekly dosing. TRIAL REGISTRATION NUMBER NCT02964104.
Collapse
Affiliation(s)
- Erica Nishimura
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | - Lone Pridal
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | - Tine Glendorf
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | - Bo Falk Hansen
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | | | - Thomas Kjeldsen
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | | | - Anne Lützen
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | | | - Peter Madsen
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | | | | | | | - Hanne Haahr
- Development, Novo Nordisk A/S, Søborg, Denmark
| |
Collapse
|
2
|
Rege NK, Liu M, Dhayalan B, Chen YS, Smith NA, Rahimi L, Sun J, Guo H, Yang Y, Haataja L, Phillips NFB, Whittaker J, Smith BJ, Arvan P, Ismail-Beigi F, Weiss MA. "Register-shift" insulin analogs uncover constraints of proteotoxicity in protein evolution. J Biol Chem 2020; 295:3080-3098. [PMID: 32005662 DOI: 10.1074/jbc.ra119.011389] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Globular protein sequences encode not only functional structures (the native state) but also protein foldability, i.e. a conformational search that is both efficient and robustly minimizes misfolding. Studies of mutations associated with toxic misfolding have yielded insights into molecular determinants of protein foldability. Of particular interest are residues that are conserved yet dispensable in the native state. Here, we exploited the mutant proinsulin syndrome (a major cause of permanent neonatal-onset diabetes mellitus) to investigate whether toxic misfolding poses an evolutionary constraint. Our experiments focused on an invariant aromatic motif (PheB24-PheB25-TyrB26) with complementary roles in native self-assembly and receptor binding. A novel class of mutations provided evidence that insulin can bind to the insulin receptor (IR) in two different modes, distinguished by a "register shift" in this motif, as visualized by molecular dynamics (MD) simulations. Register-shift variants are active but defective in cellular foldability and exquisitely susceptible to fibrillation in vitro Indeed, expression of the corresponding proinsulin variant induced endoplasmic reticulum stress, a general feature of the mutant proinsulin syndrome. Although not present among vertebrate insulin and insulin-like sequences, a prototypical variant ([GlyB24]insulin) was as potent as WT insulin in a rat model of diabetes. Although in MD simulations the shifted register of receptor engagement is compatible with the structure and allosteric reorganization of the IR-signaling complex, our results suggest that this binding mode is associated with toxic misfolding and so is disallowed in evolution. The implicit threat of proteotoxicity limits sequence variation among vertebrate insulins and insulin-like growth factors.
Collapse
Affiliation(s)
- Nischay K Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Ming Liu
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia; Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, Heping District, 300052 China
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Nicholas A Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Leili Rahimi
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jinhong Sun
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia
| | - Huan Guo
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia
| | - Yanwu Yang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Leena Haataja
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia
| | - Nelson F B Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jonathan Whittaker
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Brian J Smith
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Peter Arvan
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, Michigan 48105, Australia
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202.
| |
Collapse
|
3
|
Yunn NO, Kim J, Kim Y, Leibiger I, Berggren PO, Ryu SH. Mechanistic understanding of insulin receptor modulation: Implications for the development of anti-diabetic drugs. Pharmacol Ther 2018; 185:86-98. [DOI: 10.1016/j.pharmthera.2017.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
4
|
Glidden MD, Aldabbagh K, Phillips NB, Carr K, Chen YS, Whittaker J, Phillips M, Wickramasinghe NP, Rege N, Swain M, Peng Y, Yang Y, Lawrence MC, Yee VC, Ismail-Beigi F, Weiss MA. An ultra-stable single-chain insulin analog resists thermal inactivation and exhibits biological signaling duration equivalent to the native protein. J Biol Chem 2017; 293:47-68. [PMID: 29114035 DOI: 10.1074/jbc.m117.808626] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/12/2017] [Indexed: 12/12/2022] Open
Abstract
Thermal degradation of insulin complicates its delivery and use. Previous efforts to engineer ultra-stable analogs were confounded by prolonged cellular signaling in vivo, of unclear safety and complicating mealtime therapy. We therefore sought an ultra-stable analog whose potency and duration of action on intravenous bolus injection in diabetic rats are indistinguishable from wild-type (WT) insulin. Here, we describe the structure, function, and stability of such an analog, a 57-residue single-chain insulin (SCI) with multiple acidic substitutions. Cell-based studies revealed native-like signaling properties with negligible mitogenic activity. Its crystal structure, determined as a novel zinc-free hexamer at 2.8 Å, revealed a native insulin fold with incomplete or absent electron density in the C domain; complementary NMR studies are described in the accompanying article. The stability of the analog (ΔGU 5.0(±0.1) kcal/mol at 25 °C) was greater than that of WT insulin (3.3(±0.1) kcal/mol). On gentle agitation, the SCI retained full activity for >140 days at 45 °C and >48 h at 75 °C. These findings indicate that marked resistance to thermal inactivation in vitro is compatible with native duration of activity in vivo Further, whereas WT insulin forms large and heterogeneous aggregates above the standard 0.6 mm pharmaceutical strength, perturbing the pharmacokinetic properties of concentrated formulations, dynamic light scattering, and size-exclusion chromatography revealed only limited SCI self-assembly and aggregation in the concentration range 1-7 mm Such a combination of favorable biophysical and biological properties suggests that SCIs could provide a global therapeutic platform without a cold chain.
Collapse
Affiliation(s)
- Michael D Glidden
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Khadijah Aldabbagh
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Nelson B Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Kelley Carr
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yen-Shan Chen
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jonathan Whittaker
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Manijeh Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | | | - Nischay Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Mamuni Swain
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yi Peng
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yanwu Yang
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael C Lawrence
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Vivien C Yee
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael A Weiss
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
5
|
Lundby A, Bolvig P, Hegelund AC, Hansen BF, Worm J, Lützen A, Billestrup N, Bonnesen C, Oleksiewicz MB. Surface-expressed insulin receptors as well as IGF-I receptors both contribute to the mitogenic effects of human insulin and its analogues. J Appl Toxicol 2014; 35:842-50. [DOI: 10.1002/jat.3082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/13/2014] [Accepted: 09/15/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Anders Lundby
- Department of Biomedical Sciences; University of Copenhagen; Blegdamsvej 3 2200 Copenhagen N Denmark
| | - Pernille Bolvig
- Diabetes Research Unit, Novo Nordisk A/S; Novo Nordisk Park 2760 Maaloev Denmark
| | | | - Bo F. Hansen
- Diabetes Research Unit, Novo Nordisk A/S; Novo Nordisk Park 2760 Maaloev Denmark
| | - Jesper Worm
- Diabetes Research Unit, Novo Nordisk A/S; Novo Nordisk Park 2760 Maaloev Denmark
| | - Anne Lützen
- Diabetes Research Unit, Novo Nordisk A/S; Novo Nordisk Park 2760 Maaloev Denmark
| | - Nils Billestrup
- Department of Biomedical Sciences; University of Copenhagen; Blegdamsvej 3 2200 Copenhagen N Denmark
| | - Christine Bonnesen
- Diabetes Research Unit, Novo Nordisk A/S; Novo Nordisk Park 2760 Maaloev Denmark
| | - Martin B. Oleksiewicz
- Centre for Biosecurity and Biopreparedness; Statens Serum Institute; Artillerivej 5 2300 Copenhagen S Denmark
| |
Collapse
|
6
|
Hvid H, Blouin MJ, Birman E, Damgaard J, Poulsen F, Fels JJ, Fledelius C, Hansen BF, Pollak M. Treatment with insulin analog X10 and IGF-1 increases growth of colon cancer allografts. PLoS One 2013; 8:e79710. [PMID: 24260289 PMCID: PMC3832545 DOI: 10.1371/journal.pone.0079710] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 09/24/2013] [Indexed: 12/28/2022] Open
Abstract
Obesity and type 2 diabetes are associated with an increased risk for development of certain forms of cancer, including colon cancer. The publication of highly controversial epidemiological studies in 2009 raised the possibility that use of the insulin analog glargine increases this risk further. However, it is not clear how mitogenic effects of insulin and insulin analogs measured in vitro correlate with tumor growth-promoting effects in vivo. The aim of this study was to examine possible growth-promoting effects of native human insulin, insulin X10 and IGF-1, which are considered positive controls in vitro, in a short-term animal model of an obesity- and diabetes-relevant cancer. We characterized insulin and IGF-1 receptor expression and the response to treatment with insulin, X10 and IGF-1 in the murine colon cancer cell line (MC38 cells) in vitro and in vivo. Furthermore, we examined pharmacokinetics and pharmacodynamics and monitored growth of MC38 cell allografts in mice with diet-induced obesity treated with human insulin, X10 and IGF-1. Treatment with X10 and IGF-1 significantly increased growth of MC38 cell allografts in mice with diet-induced obesity and we can therefore conclude that supra-pharmacological doses of the insulin analog X10, which is super-mitogenic in vitro and increased the incidence of mammary tumors in female rats in a 12-month toxicity study, also increase growth of tumor allografts in a short-term animal model.
Collapse
Affiliation(s)
- Henning Hvid
- Diabetes Research Unit, Novo Nordisk A/S, Maaloev, Denmark
- * E-mail:
| | - Marie-José Blouin
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Elena Birman
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | | | - Fritz Poulsen
- Diabetes Research Unit, Novo Nordisk A/S, Maaloev, Denmark
| | | | | | | | - Michael Pollak
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Knudsen L, Hansen BF, Jensen P, Pedersen TÅ, Vestergaard K, Schäffer L, Blagoev B, Oleksiewicz MB, Kiselyov VV, De Meyts P. Agonism and antagonism at the insulin receptor. PLoS One 2012; 7:e51972. [PMID: 23300584 PMCID: PMC3531387 DOI: 10.1371/journal.pone.0051972] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/14/2012] [Indexed: 11/29/2022] Open
Abstract
Insulin can trigger metabolic as well as mitogenic effects, the latter being pharmaceutically undesirable. An understanding of the structure/function relationships between insulin receptor (IR) binding and mitogenic/metabolic signalling would greatly facilitate the preclinical development of new insulin analogues. The occurrence of ligand agonism and antagonism is well described for G protein-coupled receptors (GPCRs) and other receptors but in general, with the exception of antibodies, not for receptor tyrosine kinases (RTKs). In the case of the IR, no natural ligand or insulin analogue has been shown to exhibit antagonistic properties, with the exception of a crosslinked insulin dimer (B29-B’29). However, synthetic monomeric or dimeric peptides targeting sites 1 or 2 of the IR were shown to be either agonists or antagonists. We found here that the S961 peptide, previously described to be an IR antagonist, exhibited partial agonistic effects in the 1–10 nM range, showing altogether a bell-shaped dose-response curve. Intriguingly, the agonistic effects of S961 were seen only on mitogenic endpoints (3H-thymidine incorporation), and not on metabolic endpoints (14C-glucose incorporation in adipocytes and muscle cells). The agonistic effects of S961 were observed in 3 independent cell lines, with complete concordance between mitogenicity (3H-thymidine incorporation) and phosphorylation of the IR and Akt. Together with the B29-B’29 crosslinked dimer, S961 is a rare example of a mixed agonist/antagonist for the human IR. A plausible mechanistic explanation based on the bivalent crosslinking model of IR activation is proposed.
Collapse
Affiliation(s)
- Louise Knudsen
- Receptor Systems Biology Laboratory, Hagedorn Research Institute, Gentofte, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Hvid H, Fendt SM, Blouin MJ, Birman E, Voisin G, Svendsen AM, Frank R, Vander Heiden MG, Stephanopoulos G, Hansen BF, Pollak M. Stimulation of MC38 tumor growth by insulin analog X10 involves the serine synthesis pathway. Endocr Relat Cancer 2012; 19:557-74. [PMID: 22685267 DOI: 10.1530/erc-12-0125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Recent evidence suggests that type II diabetes is associated with increased risk and/or aggressive behavior of several cancers, including those arising from the colon. Concerns have been raised that endogenous hyperinsulinemia and/or exogenous insulin and insulin analogs might stimulate proliferation of neoplastic cells. However, the mechanisms underlying possible growth-promoting effects of insulin and insulin analogs in cancer cells in vivo, such as changes in gene expression, are incompletely described. We observed that administration of the insulin analog X10 significantly increased tumor growth and proliferation in a murine colon cancer model (MC38 cell allografts). Insulin and X10 altered gene expression in MC38 tumors in a similar fashion, but X10 was more potent in terms of the number of genes influenced and the magnitude of changes in gene expression. Many of the affected genes were annotated to metabolism, nutrient uptake, and protein synthesis. Strikingly, expression of genes encoding enzymes in the serine synthesis pathway, recently shown to be critical for neoplastic proliferation, was increased following treatment with insulin and X10. Using stable isotopic tracers and mass spectrometry, we confirmed that insulin and X10 increased glucose contribution to serine synthesis in MC38 cells. The data demonstrate that the tumor growth-promoting effects of insulin and X10 are associated with changes in expression of genes involved in cellular energy metabolism and reveal previously unrecognized effects of insulin and X10 on serine synthesis.
Collapse
Affiliation(s)
- Henning Hvid
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Cote-Ste.-Catherine, Montreal, Quebec, Canada H3T 1E2.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Hvid H, Fels JJ, Kirk RK, Thorup I, Jensen HE, Hansen BF, Oleksiewicz MB. In Situ Phosphorylation of Akt and ERK1/2 in Rat Mammary Gland, Colon, and Liver Following Treatment with Human Insulin and IGF-1. Toxicol Pathol 2011; 39:623-40. [DOI: 10.1177/0192623311406936] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
High doses of insulin and the insulin analog AspB10 have been reported to increase mammary tumor incidence in female rats likely via receptor-mediated mechanisms, possibly involving enhanced IGF-1 receptor activation. However, insulin and IGF-1 receptor functionality and intracellular signaling in the rat mammary gland in vivo is essentially unexplored. The authors investigated the effect of a single subcutaneous dose of 600 nmol/kg human insulin or IGF-1 on Akt and ERK1/2 phosphorylation in rat liver, colon, and mammary gland. Rat tissues were examined by Western blotting and immunohistochemistry by phosphorylation-specific antibodies. Insulin as well as IGF-1 caused Akt phosphorylation in mammary epithelial cells, with myoepithelial and basal epithelial cells being most sensitive. IGF-1 caused stronger Akt phosphorylation than insulin in mammary gland epithelial cells. Phosphorylation of ERK1/2 was not influenced by insulin or IGF-1. Rather, in liver and mammary gland P-ERK1/2 appeared to correlate with estrous cycling, supporting that ERK1/2 has important physiological roles in these two organs. In short, these findings supported that the rat mammary gland epithelium expresses functional insulin and IGF-1 receptors and that phosphorylation of Akt as well as ERK1/2 may be of value in understanding the effects of exogenous insulin in the rat mammary gland and colon.
Collapse
Affiliation(s)
- Henning Hvid
- Department of Veterinary Disease Biology, Faculty of Life Sciences, University of Copenhagen, Denmark
- Pathology, Novo Nordisk A/S, Copenhagen, Denmark
| | | | - Rikke K. Kirk
- Histology and Delivery, Novo Nordisk A/S, Copenhagen, Denmark
| | - Inger Thorup
- Pathology, Novo Nordisk A/S, Copenhagen, Denmark
| | - Henrik E. Jensen
- Department of Veterinary Disease Biology, Faculty of Life Sciences, University of Copenhagen, Denmark
| | - Bo F. Hansen
- Insulin Biology, Novo Nordisk A/S, Copenhagen, Denmark
| | | |
Collapse
|
10
|
Andreas K, Zehbe R, Kazubek M, Grzeschik K, Sternberg N, Bäumler H, Schubert H, Sittinger M, Ringe J. Biodegradable insulin-loaded PLGA microspheres fabricated by three different emulsification techniques: investigation for cartilage tissue engineering. Acta Biomater 2011; 7:1485-95. [PMID: 21168535 DOI: 10.1016/j.actbio.2010.12.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 11/17/2010] [Accepted: 12/13/2010] [Indexed: 01/02/2023]
Abstract
Growth, differentiation and migration factors facilitate the engineering of tissues but need to be administered with defined gradients over a prolonged period of time. In this study insulin as a growth factor for cartilage tissue engineering and a biodegradable PLGA delivery device were used. The aim was to investigate comparatively three different microencapsulation techniques, solid-in-oil-in-water (s/o/w), water-in-oil-in-water (w/o/w) and oil-in-oil-in-water (o/o/w), for the fabrication of insulin-loaded PLGA microspheres with regard to protein loading efficiency, release and degradation kinetics, biological activity of the released protein and phagocytosis of the microspheres. Insulin-loaded PLGA microspheres prepared by all three emulsification techniques had smooth and spherical surfaces with a negative zeta potential. The preparation technique did not affect particle degradation nor induce phagocytosis by human leukocytes. The delivery of structurally intact and biologically active insulin from the microspheres was shown using circular dichroism spectroscopy and a MCF7 cell-based proliferation assay. However, the insulin loading efficiency (w/o/w about 80%, s/o/w 60%, and o/o/w 25%) and the insulin release kinetics were influenced by the microencapsulation technique. The results demonstrate that the w/o/w microspheres are most appropriate, providing a high encapsulation efficiency and low initial burst release, and thus these were finally used for cartilage tissue engineering. Insulin released from w/o/w PLGA microspheres stimulated the formation of cartilage considerably in chondrocyte high density pellet cultures, as determined by increased secretion of proteoglycans and collagen type II. Our results should encourage further studies applying protein-loaded PLGA microspheres in combination with cell transplants or cell-free in situ tissue engineering implants to regenerate cartilage.
Collapse
Affiliation(s)
- Kristin Andreas
- Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Oleksiewicz MB, Bonnesen C, Hegelund AC, Lundby A, Holm GMN, Jensen MB, Krabbe JS. Comparison of intracellular signalling by insulin and the hypermitogenic AspB10 analogue in MCF-7 breast adenocarcinoma cells. J Appl Toxicol 2010; 31:329-41. [DOI: 10.1002/jat.1590] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 08/16/2010] [Accepted: 08/16/2010] [Indexed: 12/17/2022]
|