Part.1 Background

Type I diabetes is an autoimmune disease due to the destruction of beta cells of islets that produce insulin and the absolute insufficiency of insulin secretion.

Figure 1.

Distribution of type I diabetes patients in the global population

Table 1.

Top 10 countries or territories for estimated number of incident (new) cases o type 1 diabetes in children and adolescents (0-19 years) per annum

Table 2.

Challenges with the diagnosis of adult-onsettype 1 diabetes

In China, the incidence rate of people aged 0-14 years is 1.93/100000, significantly higher than that of people of other ages2.(The incidence rate of people aged 15-29 years is 1.28/100000, while the incidence rate of the elderly is 0.69/100000.)

Figure 2.

Age distribution of type I diabetes patients in the global population

From the above chart, we can see that in 2022, there were 530000 new confirmed cases of T1D, of which 20.1000 were patients under the age of 20. Most patients with type I diabetes get sick in children or adolescence, and some adults will also be affected.

The risk factors of type I diabetes include heredity, toxin, immune system disorder and virus infection.
Specific HLA alleles are closely associated with TD.

Figure 3.

The risk factors of type I diabetes

Type I diabetes can cause a variety of complications, including acute ketoacidosis, chronic diabetes foot and other complications involving other systems and organs. In addition, studies have shown that adults with type 1 diabetes have an increased risk of other autoimmune diseases. About 30% of adults with type 1 diabetes have thyroid autoimmunity.

Why do we do this?
Present

At present, the treatment of T1D mainly includes drug therapy and transplantation therapy.
Insulin is the main medication for T1D treatment, but if patients want to successfully manage T1D, they need to inject multiple times a day to maintain blood sugar stability, or use automatic insulin delivery systems such as insulin pumps, and continuously monitor blood sugar changes.

The above methods require lifelong use and can lead to some side effects, such as pain at the injection site, subcutaneous fat hyperplasia, and hypoglycemia.
The transplantation of pancreas, islets, and stem cells is an ideal treatment for T1D, but the lack of immune response and donor resources severely limits the widespread use of this method.

If...

 We designed the Glycemic Stabilizer project to construct a Designer cell that can secrete insulin using synthetic biology methods. In this cell, high blood sugar promotes insulin synthesis and secretion, and once blood sugar drops to a certain level, it can prevent further insulin synthesis and prevent the occurrence of hypoglycemia.

Part.2 Introduction
Our system includes systems that promote insulin synthesis, including:

the GI-GAL4 and LOV-VP16 genes regulated by the GIP promoter (a glucose sensitive promoter that enhances GIP promoter activity)

the insulin genes regulated by the UAS promoter (which can be activated by GAL-VP6)

When the glucose concentration increases, GI-GAL4 and LOV-VP16 proteins are expressed, but when GI-GAL4 and LOV-VP16 proteins are present alone, they cannot activate glucose expression.

We designed a switch that senses glucose concentration.

When the glucose concentration reaches a pathological state, the switch opens and activates a blue light, catalyzing the covalent binding of GI-GAL4 and LOV-VP16 proteins, i.e. GI-GAL4: LOV-VP16, to assemble an active GAL4-VP16 transcription factor that binds to the UAS promoter, promoting insulin synthesis and secretion.

In this system, we utilized the promoter of GIP to control the expression of GI-GAL4 and LOV-VP16 proteins, in order to prevent blue light from mistakenly turning on and causing insulin synthesis at low blood sugar concentrations.

Additionally, considering that the GIP promoter cannot accurately sense changes in blood sugar, such as at 5mM, The GIP promoter can be activated, but at this point blood sugar is normal. Therefore, we have designed a blood sugar concentration sensing switch that can accurately sense pathological blood sugar levels to promote insulin secretion.

Figure 4.

System Overview

The above system plays a more precise regulatory role in insulin synthesis, but, the half-life of GI-GAL4 and LOV-VP16 proteins reaches the hour level, and undigested proteins continue to promote insulin synthesis, thereby posing a risk of hypoglycemia.

To avoid this situation, we have designed a braking system.

The system consists of miRNA and miR-BS (miRNA binding site) located in the insulin expression vector 3 '- UTR.

When miRNA is expressed, it binds to miR-BS and inhibits insulin expression. In order to achieve the regulation of blood glucose levels in the system, we further constructed a sponge controlled by the GIP promoter. Under high blood glucose conditions, the sponge binds to miRNA, releasing the binding of miRNA to insulin expression.

However, when blood glucose drops to normal levels, the expression of the sponge decreases, MiRNA is liberated and binds to miR-BS, inhibiting insulin expression.

Now let's systematically review the operational process again.
Precise regulatory system:
preventing the initiation of insulin synthesis under physiological conditions, thereby making insulin synthesis regulation more precise.
When the blood glucose concentration increases, the glucose induced promoter is activated, the GI-GAL4 and LOV-VP16 proteins were synthesized.
At this point, if blood sugar continues to rise to pathological levels, our biosensor will emit blue light, insulin expression will be activated, and cells will secrete insulin.
Braking system:
If there is nothing to stop it, excessive secretion can occur, which is dangerous for patients.
To avoid such dangerous situations, we have designed a negative feedback system that can prevent insulin expression when blood sugar drops.

We can use materials such as microcapsules, hollow fiber membrane tubes, and sodium alginate gel as carriers to implant engineered cells subcutaneously and achieve the goal of controlling blood sugar.

Part.3 Glycemic stabilizer
Precise Control System Based on Biosensors
Figure 6. Physiological state - blue light not turned on, insulin not expressed
We utilize P-GIP, which may be activated at physiological blood glucose concentrations, leading to GI-GAL4 The LOV-VP16 protein was synthesized and the inhibitory effect of miRNA was relieved.
Figure 7. Pathological state - blue light on, insulin expression
When blood sugar rises and reaches a pathological state, The GAL4-VP16 transcription factor is synthesized, and insulin is synthesized and released.

However, due to the threshold for blue light activation being when blood glucose concentration exceeds the physiological state, blue light does not activate and cannot synthesize the GAL-VP16 promoter, resulting in the inability of insulin expression.

Negative Feedback System
Blood sugar decreases and returns to a physiological state, but the GAL4-VP16 transcription factor cannot be degraded in a short period of time, posing a risk of hypoglycemia.
To prevent this situation from happening, we added a miRNA-BS, which can be used when blood sugar drops, CircRNA is not synthesized and cannot exert its function, resulting in the binding of miRNA to miRNA-BS and inhibition of insulin synthesis.
Figure 8.
Physiological state - blood sugar drops, miRNA inhibits insulin synthesis
Application
  1. Desinger cells are embedded subcutaneously and achieve blood glucose regulation through controlled expression of insulin;
  2. We will design a wristband like device that can accurately sense glucose concentration and switch blue light to regulate Designer cells.
Significance
Except reliable methods, we also have...
The use of biosensors to control the blue light switch through changes in blood glucose concentration.
Concentration
We have a precise regulatory system that enables glucose concentration response to physiological and pathological changes, preventing insulin synthesis and release under physiological conditions.
Precision
To prevent hypoglycemia and hyperinsulinemia, the project has safety guarantees.
Safety
Reference
  1. Mobasseri, M. et al. Prevalence and incidence of type 1 diabetes in the world: a systematic review and meta-analysis. Health Promot Perspect 10, 98-115 (2020).
  2. Weng, J. et al. Incidence of type 1 diabetes in China, 2010-13: population based study. BMJ 360, j5295 (2018).
  3. Weng, J. et al. Incidence of type 1 diabetes in China, 2010-13: population based study. BMJ j5295 (2018) doi:10.1136/bmj.j5295.
  4. Rennert, O. M. & Francis, G. L. Update on the Genetics and Pathophysiology of Type I Diabetes Mellitus. Pediatr Ann 28, 570–575 (1999).
  5. MCGRAIL, C. & GAULTON, K. J. 138-OR: Characterization and Prediction of Genetic Risk of T1D in Individuals without HLA-DR3 or HLA-DR4. Diabetes 72, (2023).
  6. Wilson, V. An overview of complications associated with type 1 and type 2 diabetes. Nursing Standard 38, 77–82 (2023).
  7. Vondra, K., Bendlová, B., Sterzl, I., Vrbíková, J. & Zamrazil, V. [Diabetes mellitus in adult patients with type I diabetes shows immunological, functional and clinical differences depending on the presence of autoimmune thyroiditis]. Cas Lek Cesk 146, 267–72 (2007).
  8. Koike, Y. & Terauchi, Y. Are current SGLT2 Inhibitors efficacious treatment options for Type 1 diabetes? Expert Opin Pharmacother 22, 1079–1081 (2021).
  9. Calafiore, R. & Basta, G. Stem cells for the cell and molecular therapy of type 1 diabetes mellitus (T1D): the gap between dream and reality. Am J Stem Cells 4, 22–31 (2015).
  10. Boylan MO, Jepeal LI, Jarboe LA, Wolfe MM. Cell-specific expression of the glucose-dependent insulinotropic polypeptide gene in a mouse neuroendocrine tumor cell line. J Biol Chem. 1997 Jul 11;272(28):17438-43. doi: 10.1074/jbc.272.28.17438. PMID: 9211887.
  11. Yazawa M, Sadaghiani AM, Hsueh B, Dolmetsch RE. Induction of protein-protein interactions in live cells using light. Nat Biotechnol. 2009 Oct;27(10):941-5. doi: 10.1038/nbt.1569. Epub 2009 Oct 4. PMID: 19801976.
  12. Mansouri M, Hussherr MD, Strittmatter T, Buchmann P, Xue S, Camenisch G, Fussenegger M. Smart-watch-programmed green-light-operated percutaneous control of therapeutic transgenes. Nat Commun. 2021 Jun 7;12(1):3388. doi: 10.1038/s41467-021-23572-4. PMID: 34099676; PMCID: PMC8184832.
  13. Riddell MC, Gallen IW, Smart CE, Taplin CE, Adolfsson P, Lumb AN, Kowalski A, Rabasa-Lhoret R, McCrimmon RJ, Hume C, Annan F, Fournier PA, Graham C, Bode B, Galassetti P, Jones TW, Millán IS, Heise T, Peters AL, Petz A, Laffel LM. Exercise management in type 1 diabetes: a consensus statement. Lancet Diabetes Endocrinol. 2017 May;5(5):377-390. doi: 10.1016/S2213-8587(17)30014-1. Epub 2017 Jan 24. Erratum in: Lancet Diabetes Endocrinol. 2017 May;5(5):e3. PMID: 28126459.
  14. Nissim L, Wu MR, Pery E, Binder-Nissim A, Suzuki HI, Stupp D, Wehrspaun C, Tabach Y, Sharp PA, Lu TK. Synthetic RNA-Based Immunomodulatory Gene Circuits for Cancer Immunotherapy. Cell. 2017 Nov 16;171(5):1138-1150.e15. doi: 10.1016/j.cell.2017.09.049. Epub 2017 Oct 19. PMID: 29056342; PMCID: PMC5986174.
  15. Shaw JA, Delday MI, Hart AW, Docherty HM, Maltin CA, Docherty K. Secretion of bioactive human insulin following plasmid-mediated gene transfer to non-neuroendocrine cell lines, primary cultures and rat skeletal muscle in vivo. J Endocrinol. 2002 Mar;172(3):653-72. doi: 10.1677/joe.0.1720653. PMID: 11874714.