Description

Our Journey: firing the neurons leading to MS

Multiple sclerosis (MS) is a predominant disease “disease-personality” in neuroscientific research and the clinical world. For over 50 years, it has been a constant challenge for researchers and clinicians alike to unravel its well-hidden mysteries and uncover crucial treatment aspects of its pathophysiology. An underlying “flirt'' between the immune and nervous system intensifies itself to the extent that it takes on pathological dimensions, leading to a self-destructing love. “As a medical student”, Alexandros explained, “I've had numerous opportunities to learn about MS, mostly regarding current knowledge and progress already made. Then the notion struck, one that was absolutely and thoroughly fascinating: why not upend the current state of affairs by tackling novel therapeutically important aspects of MS etiology and setting ourselves apart from previously tested and established treatments? And all based on synthetic biology!” At this moment, the core motivation was established. NeuroMuSceteer was beginning to take shape!

Being a chronic illness of the central nervous system, the dynamic and unpredictable nature of MS presents a compelling challenge for medical research and patient care. MS leads to a range of physical and cognitive impairments. Despite its prevalence, public stigma and misinformation are not uncommon, often due to the lack of awareness and understanding. Therefore, bridging this knowledge gap and providing valuable insights through a new therapeutic strategy could be not only beneficial to patients themselves, but also their caregivers, and the community as a whole. Overcoming such a challenge could foster hope and encouragement among those affected by the disease. By shedding light on their daily struggles and triumphs, it could be possible both to inspire empathy and action as well as to “humanize” the complex parameters of the disease and the life it entails.

Yet, following our brainstorming timeline, “NeuroMuSceteer” was the last project proposal submitted. Prior to that, several remarkable ideas had already been discussed: an innovative point-of-care-testing (POCT) system for the early detection of Candida Auris; 'Conan,' a radiation-resistant bacterium as a key material for a novel space uniform design; two environmentally-focused projects addressing water pollution—one proposing a synthetic biofilter for efficient heavy metal and nitrogen-based compound collection, and the other suggesting a magnetic-bacteria biofilter to capture microplastics in the ocean. We also considered the idea of constructing 3D-printed organoids using cell cultures. Ultimately, there was an overwhelming consensus on the project's theme, though each choice was influenced by different criteria and reasoning.

Picture of the team
Figure 1: The day we chose the project.

9 REASONS WHY we chose this project

  • Elias
  • Panagiotis
  • Maria
  • Maria Christina
  • Ioanna
  • Antonis
  • Anastasia
  • Eva
  • Louiza

Ultimately, MS selection was all but random. “NeuroMuSceteer aspires to make a tangible difference, not only medically and scientifically, but also socially”, states Elias, our team leader. It has been an original goal and a firm belief of all of us that this project approach could launch a new therapeutic approach for MS as well as others in the spectrum of neurodegenerative/ neuroimmunological diseases, and, through increased education and sensitivity, could help us make a step towards a more supportive and informed society.

Finally, the interest stemming from the challenge of "incurable" was the main advantage of this idea for Panagiotis. In mathematics, for instance, there have been unsolved problems since ancient times. Mathematicians' engagement with them may not have led to their solution, but it contributed to advances in science, new problems and discoveries. Similarly, our attempt to tackle the so-far unsolved problem of MS will certainly open up new horizons and cultivate further ground for research and study in this field. "Aim for the moon and, even if you miss, you will still be among the stars".

The excitement of leveraging established methods and knowledge to create something truly innovative has been the driving force for Mary. “As I see it, this project gives us the opportunity to stand on the shoulders of giants and make a contribution of our own, not just to the scientific community but to an important issue as well”.

Maria-Christina opted for this project “because of a significant person in my life. Through this project, I would be able to understand their condition and contribute through scientific research”.

Meanwhile, working on an MS project seemed “personally fulfilling” for Ioanna as well, as “it provides an opportunity to contribute to a cause that truly matters. Knowing that our efforts could potentially lead to breakthroughs that improve the lives of millions of people worldwide is incredibly motivating. It gives a deeper purpose to our research and reinforces our commitment to making a positive difference in the world”.

As for Antonis, researching an MS-related project seems to have captured his attention all along. “Being aware of the daily struggles and the repercussions of this disease for those who suffer from it highlighted to me the urgent need for innovative solutions.” He claims that through this project, the team has the opportunity to deploy advanced scientific and technological methodologies and develop a breakthrough solution that may enhance the quality of life for people around the globe. “I am truly excited to contribute to our collective work!”

For Anastasia, the choice was natural; “As a woman, I am particularly aware of how this condition affects mostly females.” Witnessing the struggles of those living with MS, she felt compelled to support a project dedicated to advancing research and improving treatments. The project’s focus on innovative solutions resonated deeply with her, making the casting of her vote for this project feel like a step towards a hopeful future for the MS community.

Equally motivated by the social impact and the scientific thrill, Eva shares her thoughts; “This project represents more than just an academic challenge; it embodies a commitment to making a real difference. By working on this, I feel that we can contribute to something truly meaningful and transformative”. Pushing the boundaries of science and technology and providing a real chance to make a lasting impact are considered by her as the amazing opportunity arising from the interdisciplinary nature of this research.

“Apart from the fact that MS is a very serious disease, I wanted to contribute to the effort of dealing with this medical issue, even though, as a student of Agriculture, it is not in my field of knowledge, thus highlighting the value of interdisciplinary and collaboration of different fields for the best possible result”, mentioned Louiza, unraveling the interdisciplinary plexus of our team.

MS Effects

Local Solutions for Local Problems: Thessaloniki in the spotlight

Multiple sclerosis is a particularly pressing issue in our region, Thessaloniki, Greece, due to higher prevalence rates than in other parts of the country. According to studies, northern Greece, including Thessaloniki, has one of the highest MS incidences in Europe, most likely due to genetic, environmental, and lifestyle factors unique to the region 1. This increased prevalence could be attributed to specific genetic markers found more frequently in northern Greek populations, as well as environmental triggers such as vitamin D deficiency 2.

Furthermore, the Panhellenic Reference and Excellence Center for MS Research and Treatment lies in Thessaloniki, fostering pioneering research. The Second Neurology Department of AHEPA University Hospital 3 has been the cornerstone of this center. Nevertheless, addressing MS in Thessaloniki requires not only medical interventions but also public health strategies aimed at improving awareness, reducing stigma, and ensuring equitable access to care and support services. For the experiments and the implementation of our project, we were hosted at the Gene and Cell Therapy Center of George Papanikolaou Hospital in Thessaloniki, a GMP-certified facility for developing advanced gene and cell therapies in Greece. It constitutes the first facility of its kind in our country and its focus on CAR-T cells lately was a great inspiration for our project 4.

The Problem: where neurology meets immunology

Multiple sclerosis is unquestionably the most common non-traumatic disease affecting young people. The global incidence of MS is increasing (more than 2 million people), as is the disease's socioeconomic burden 5. Although the etiology is unknown, it is widely assumed that intricate gene-environment interactions play a substantial role and that the disease is autoimmune in origin. The disease is thought to begin with the activation of CD4+ T cells that target central nervous system (CNS) autoantigens and their infiltration into the CNS, followed by cytokine and antibody storms against oligodendrocyte and neuron antigens, which eventually lead to their irreversible destruction. MS pathology includes plaque formation in the CNS after inflammation and demyelination 6. Depending on which area(s) of the CNS is damaged, this can lead to a wide range of clinical symptoms.

Neurons On MS

Meeting CAAR: a cell therapy approach tailored to autoimmunity

CAAR T cells could be a new therapeutic strategy for autoimmune diseases, targeting and eliminating autoreactive B cells responsible for producing pathogenic antibodies. CAAR T cells have an extracellular domain derived from the autoantigen involved in the autoimmune response, which is linked to intracellular signaling domains that activate T cells when they bind to autoreactive B-cell receptors 7. Recent research has shown the effectiveness of CAAR T cells targeting desmoglein in mouse models of pemphigus vulgaris, aquaporin-4 in neuromyelitis optica, autoimmune encephalitis, NMDAR encephalitis and potential in treating systemic lupus erythematosus 8 9. However, challenges such as off-target effects, potential cytokine release syndrome, and long-term safety concerns need thorough investigation. Additionally, the manufacturing complexity and costs associated with personalized cell therapies present significant hurdles for widespread clinical application 10.

CRISPR in the roster: making T regulatory cells

We sought to employ the CRISPR-Cas9 system to epigenetically induce expression of FOXP3, a transcription factor thought to be the defining characteristic of regulatory cells and necessary for their activation and survival, in order to lessen the potentially harmful side effects of our CAAR-T cells and to further suppress the autoimmune response. This would essentially turn our effector T cells into immunosuppressive Tregs, allowing us to retain immunological tolerance and improve therapy safety 11.

T regulatory cells (Tregs) are essential for maintaining immune tolerance by suppressing autoreactive T cells through mechanisms like cytokine secretion (e.g., IL-10, TGF-β) and direct cell contact. In autoimmunity, impaired Treg function or numbers lead to the loss of immune tolerance, allowing self-reactive cells to attack healthy tissues.

CRISPR, an acronym for “Clustered Regularly Interspaced Short Palindromic Repeats”, functions as a precise gene-editing tool, leveraging a naturally occurring system in bacteria that serves as a defense mechanism against viruses. The CRISPR-Cas9 system, the most widely used variant, involves two key components: a guide RNA (gRNA) and the Cas9 enzyme. The gRNA is designed to match a specific DNA sequence within the genome, guiding the Cas9 enzyme to this exact location. Once bound to the target DNA, the Cas9 enzyme acts as molecular scissors, creating a double-strand break (DSB) in the DNA. The cell's natural repair mechanisms then kick in, and this is where genetic editing occurs 12. Researchers can manipulate the repair process to disable a gene, insert a new gene, or correct a mutation. Undoubdedtly, this powerful technology is revolutionizing various scientific fields 13, however, the CRISPR-Cas-9 mediated introduction of DSBs has been associated with chromosomal translocations, chromothripsis and activation of apoptotic pathways[KP1]. Recently, epigenome editing has evolved as a safer and equally efficient approach. The epigenome editing tools consist of a DNA binding domain, such as deactivated Cas9 nuclease (dCas9) coupled with an epigenetic modifier domain (epigenome editor, epi-editor). Once the guide RNA (gRNA) successfully identifies and binds to the target genomic locus, such as a gene promoter or enhancer, the epi-editor is recruited to that specific site[KP2] . A broad range of epigenetic modifiers has been employed to either activate or suppress gene expression. One of the most well-known epigenetic activators is the dCas9-VPR[KP3], a synthetic transcriptional activation complex designed to strongly activate gene expression. It consists of three powerful transcriptional activators VP64, p65, Rta. Together, these activators form VPR, which significantly boosts transcriptional activation, much stronger than single transcriptional activators, when recruited to target gene. Unlike traditional CRISPR-Cas9, which introduces double-strand breaks to edit genes, dCas9-VPR modulates gene expression without altering the DNA sequence, thus reducing the risk of harmful mutations 14.

miRNAs in MS: some steps after the first date

Incorporating a remyelinating mechanism to the project design is crucial to combat the disease as a whole. After regulation of the autoimmune response against myelin, the damage that has already occurred could be repaired, and the disease progression would not only be decelerated but reversed. Myelin sheath restoration in the CNS, is the result of a cascade of steps that include the activation of stem cells called “Oligodendrocyte Progenitor Cells” (OPCs), their migration to demyelinated areas, their proliferation and finally, their differentiation to Oligodendrocytes (OLs), the mature cells responsible for myelin formation 15.

In multiple sclerosis lesions, while remyelination occurs initially -which explains remission periods-, the gradual accumulation of myelin debris and immune system cells inhibits it, mainly by blocking the differentiation of OPCs to OLs 16.

It should be highlighted that remyelination is one of the most sought-after targets for Multiple Sclerosis therapy, with the majority of research focusing on impaired OPC differentiation as the main culprit of failure. However, no such treatment is yet available 17.

MicroRNAs (miRNAs) are small, non-coding RNA molecules, typically about 22 nucleotides long, that play crucial roles in regulating gene expression. They bind to complementary sequences on target messenger RNAs (mRNAs), usually resulting in translational repression or target degradation and gene silencing 18.

The remyelination process in particular, involves a number of miRNAs in every stage that orchestrate OPC activity and can largely influence its success 19. That’s why miRNA delivery has been investigated as a remyelinating candidate for MS management yielding promising results 20 21. Moreover, as expected, miRNA levels are altered in patients compared to their healthy counterparts, contributing to the physiological changes and allowing them to act as disease biomarkers 22 23.

Combining those two functions, it is possible to utilize miRNAs in a singular theranostic mechanism that recognises commonly upregulated miRNAs in patient precursor cells and is then activated, leading to the release of miRNAs that boost cell maturation and remyelination.

Our Solution: “NeuroMuSceteer” genesis

Multiple sclerosis (MS) is a complex autoimmune disorder characterized by neuroinflammation and demyelination within the central nervous system (CNS). This project aims to address these two core aspects of MS pathophysiology through innovative therapeutic strategies.

Pillar 1: T-Cell Modification (CRISPR and CAAR)

Overview

The first part of our approach focuses on the modification of T-immune cells derived from MS patients. By leveraging CRISPR technology, we aim to convert these cells into regulatory T cells (T-regs), expressing the transcription factor Foxp3. This conversion is crucial, as T-regs play a vital role in maintaining immune homeostasis and preventing excessive inflammation.

Methodology

  1. Epigenetic CRISPR Modification: We utilize a lentiviral vector system to introduce a dCas9-VPR epigenetic tool into the T cells to effectively activate Foxp3 expression and switch conventional T cells into T regulatory cells (Tregs). This step is supported by studies demonstrating the efficacy of CRISPR in enhancing T-reg cell populations 24.
  2. CAAR Construction: Following T-reg conversion, we integrate a specific Chimeric Auto-Antibody Receptor (CAAR) construct into Treg cells, targeting key epitopes involved in MS pathogenesis, particularly Myelin Basic Protein (MBP) 25 and Myelin Oligodendrocyte Glycoprotein (MOG) 26. These epitopes are critical as they are often recognized by autoimmune B cells, leading to demyelination. The binding of these autoimmune B cells to modified CAAR T-regs can help modulate the autoimmune response by by-passing the detrimental binding of myelin proteins to auto-antibodies.
  3. Blood-Brain Barrier (BBB) Modifications: The passage of CAAR Treg cells across the BBB when they are administered intravenously, is expected based on their documented migratory capabilities. Recent studies have shown that engineered T cells can effectively cross the BBB, which in case of MS could be further facilitated by the active CNS inflammation leading to partial BBB disruption 27. Intrathecal administration should also be taken into consideration, provided that modified CAR-T cells are unable to sparkle inflammatory reactions in the CNS 28.
Project abstract
Figure 2: T-Cell modification.

Pillar 2: miRNAs Delivery

Overview

The second component of our solution involves the introduction of microRNAs (miRNAs) encapsulated in nanoparticles. This method is inspired by previous proposals, mainly iGEM Thessaloniki 2022 Project “Theriac” 29. As an administration route, the olfactory nerve is recommended for targeted delivery 30.

miRNA Selection and Function

We focus on a cluster of four miRNAs, strategically chosen for their roles in MS pathogenesis:

  • Identifier miRNAs: miR-27a and miR-125a-3p are upregulated in MS plaques, specifically in oligodendrocyte lineage cells. Their identification within these lesions underscores their potential as biomarkers.
  • Therapeutic miRNAs: miR-219 and miR-338 have been shown to promote remyelination and protect against myelin sheath damage. By delivering these miRNAs to targeted MS lesions, we aim to reverse the demyelination process 31.

Mechanism of Action

The proposed mechanism involves the activation of a series of reactions known as Hybridization Chain Reaction (HCR) upon the targeted release of the therapeutic miRNAs 28 at the identified MS plaque sites. This approach ensures localized remyelination while minimizing potential off-target effects, thereby enhancing safety and efficacy.

miRNAs
Figure 3: microRNA delivery.

In brief, our project proposes a dual-targeted approach to tackle the neuroinflammation and demyelination associated with MS. By modifying T cells to act as regulatory agents and employing nanoparticles for miRNA delivery, we aim to provide a comprehensive and effective therapeutic strategy for MS patients.

References

  1. Dadouli K, Ntellas P, Anagnostopoulos L, Bonotis K, Dardiotis E, Hadjichristodoulou C. Geography of hospital admissions for multiple sclerosis in Greece. Mult Scler Relat Disord. 2021 Apr;49:102735. doi: 10.1016/j.msard.2021.102735. Epub 2021 Jan 5. PMID: 33444957.
  2. Hedström AK, Olsson T, Kockum I, Hillert J, Alfredsson L. Low sun exposure increases multiple sclerosis risk both directly and indirectly. J Neurol. 2020 Apr;267(4):1045-1052. doi: 10.1007/s00415-019-09677-3. Epub 2019 Dec 17. PMID: 31844981; PMCID: PMC7109160.
  3. https://www.med.auth.gr/en/content/2nd-department-neurology
  4. https://aimatologiko-pap.gr/brief-overview-hematology-department-of-g-papanicolaou-hospital/
  5. Dobson, R., & Giovannoni, G. (2019). Multiple sclerosis – a review. In European Journal of Neurology (Vol. 26, Issue 1, pp. 27–40). Blackwell Publishing Ltd. https://doi.org/10.1111/ene.13819
  6. Dighriri, I. M., Aldalbahi, A. A., Albeladi, F., Tahiri, A. A., Kinani, E. M., Almohsen, R. A., Alamoudi, N. H., Alanazi, A. A., Alkhamshi, S. J., Althomali, N. A., Alrubaiei, S. N., & Altowairqi, F. K. (2023). An Overview of the History, Pathophysiology, and Pharmacological Interventions of Multiple Sclerosis. Cureus. https://doi.org/10.7759/cureus.33242
  7. Christoph T. Ellebrecht et al.,Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease.Science353,179-184(2016).DOI:10.1126/science.aaf6756
  8. Reincke, S. M., von Wardenburg, N., Homeyer, M. A., Kornau, H. C., Spagni, G., Li, L. Y., Kreye, J., Sánchez-Sendín, E., Blumenau, S., Stappert, D., Radbruch, H., Hauser, A. E., Künkele, A., Edes, I., Schmitz, D., & Prüss, H. (2023). Chimeric autoantibody receptor T cells deplete NMDA receptor-specific B cells. Cell, 186(23), 5084-5097.e18. https://doi.org/10.1016/j.cell.2023.10.001
  9. Blache, U., Tretbar, S., Koehl, U., Mougiakakos, D., & Fricke, S. (2023). CAR T cells for treating autoimmune diseases. In RMD Open (Vol. 9, Issue 4). BMJ Publishing Group. https://doi.org/10.1136/rmdopen-2022-002907
  10. Műzes, G., & Sipos, F. (2023). CAR-Based Therapy for Autoimmune Diseases: A Novel Powerful Option. In Cells (Vol. 12, Issue 11). MDPI. https://doi.org/10.3390/cells12111534
  11. Walker, M. R., Kasprowicz, D. J., Gersuk, V. H., Bènard, A., van Landeghen, M., Buckner, J. H., & Ziegler, S. F. (2003). Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25- T cells. Journal of Clinical Investigation, 112(9), 1437–1443. https://doi.org/10.1172/JCI200319441
  12. Doudna J, Knott G. CRISPR–Cas guides the future of genetic engineering. Science (80−) 2018;361(6405):1–4.
  13. Wang JY, Doudna JA. CRISPR technology: A decade of genome editing is only the beginning. Science. 2023 Jan 20;379(6629):eadd8643. doi: 10.1126/science.add8643. Epub 2023 Jan 20. PMID: 36656942.
  14. Chavez, A., Scheiman, J., Vora, S., Pruitt, B. W., Tuttle, M., P R Iyer, E., Lin, S., Kiani, S., Guzman, C. D., Wiegand, D. J., Ter-Ovanesyan, D., Braff, J. L., Davidsohn, N., Housden, B. E., Perrimon, N., Weiss, R., Aach, J., Collins, J. J., & Church, G. M. (2015). Highly efficient Cas9-mediated transcriptional programming. Nature Methods, 12(4), 326–328. https://doi.org/10.1038/nmeth.3312
  15. Franklin, R. J. M., & Simons, M. (2022). CNS remyelination and inflammation: From basic mechanisms to therapeutic opportunities. In Neuron (Vol. 110, Issue 21, pp. 3549–3565). Cell Press. https://doi.org/10.1016/j.neuron.2022.09.023
  16. Hagemeier, K., Brück, W., & Kuhlmann, T. (2012). Multiple sclerosis - remyelination failure as a cause of disease progression. In Histology and Histopathology (Vol. 27, Issue 3, pp. 277–287). https://doi.org/10.14670/HH-27.277
  17. Cunniffe, N., & Coles, A. (2021). Promoting remyelination in multiple sclerosis. In Journal of Neurology (Vol. 268, Issue 1, pp. 30–44). Springer Science and Business Media Deutschland GmbH. https://doi.org/10.1007/s00415-019-09421-x
  18. O'Brien, J., Hayder, H., Zayed, Y., & Peng, C. (2018). Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Frontiers in endocrinology, 9, 402. https://doi.org/10.3389/fendo.2018.00402
  19. Maciak, K., Dziedzic, A., & Saluk, J. (2023). Remyelination in multiple sclerosis from the miRNA perspective. In Frontiers in Molecular Neuroscience (Vol. 16). Frontiers Media S.A. https://doi.org/10.3389/fnmol.2023.1199313
  20. Gao, Y., Han, D., & Feng, J. (2021). MicroRNA in multiple sclerosis. In Clinica Chimica Acta (Vol.516 ,pp. 92–99). Elsevier B.V. https://doi.org/10.1016/j.cca.2021.01.020
  21. Trideva Sastri, K., Vishal Gupta, N., Kannan, A., Balamuralidhara, V., & Ramkishan, A. (2022). Potential nanocarrier-mediated miRNA-based therapy approaches for multiple sclerosis. In Drug Discovery Today (Vol. 27, Issue 11). Elsevier Ltd. https://doi.org/10.1016/j.drudis.2022.103357
  22. Cipriano, G. L., Schepici, G., Mazzon, E., & Anchesi, I. (2024). Multiple Sclerosis: Roles of miRNA, lcnRNA, and circRNA and Their Implications in Cellular Pathways. In International Journal of Molecular Sciences (Vol. 25, Issue 4). Multidisciplinary Digital Publishing Institute (MDPI). https://doi.org/10.3390/ijms25042255
  23. Gandhi, R. (2015). MiRNA in multiple sclerosis: Search for novel biomarkers. In Multiple Sclerosis (Vol. 21, Issue 9, pp. 1095–1103). SAGE Publications Ltd. https://doi.org/10.1177/1352458515578771
  24. Ferreira LMR, Muller YD, Bluestone JA, Tang Q. Next-generation regulatory T cell therapy. Nat Rev Drug Discov. 2019 Oct;18(10):749-769. doi: 10.1038/s41573-019-0041-4. Epub 2019 Sep 20. PMID: 31541224; PMCID: PMC7773144.
  25. Martinsen V, Kursula P. Multiple sclerosis and myelin basic protein: insights into protein disorder and disease. Amino Acids. 2022 Jan;54(1):99-109. doi: 10.1007/s00726-021-03111-7. Epub 2021 Dec 10. PMID: 34889995; PMCID: PMC8810476.
  26. Androutsou ME, Tapeinou A, Vlamis-Gardikas A, Tselios T. Myelin Oligodendrocyte Glycoprotein and Multiple Sclerosis. Med Chem. 2018 Feb 6;14(2):120-128. doi: 10.2174/1573406413666170906123204. PMID: 28875859.
  27. Aydin S, Pareja J, Schallenberg VM, Klopstein A, Gruber T, Page N, Bouillet E, Blanchard N, Liblau R, Körbelin J, Schwaninger M, Johnson AJ, Schenk M, Deutsch U, Merkler D, Engelhardt B. Antigen recognition detains CD8+ T cells at the blood-brain barrier and contributes to its breakdown. Nat Commun. 2023 May 30;14(1):3106. doi: 10.1038/s41467-023-38703-2. PMID: 37253744; PMCID: PMC10229608.
  28. Del Baldo G, Del Bufalo F, Pinacchio C, Carai A, Quintarelli C, De Angelis B, Merli P, Cacchione A, Locatelli F, Mastronuzzi A. The peculiar challenge of bringing CAR-T cells into the brain: Perspectives in the clinical application to the treatment of pediatric central nervous system tumors. Front Immunol. 2023 Mar 21;14:1142597. doi: 10.3389/fimmu.2023.1142597. PMID: 37025994; PMCID: PMC10072260.
  29. https://2022.igem.wiki/thessaloniki/
  30. Formica, M. L., Real, D. A., Picchio, M. L., Catlin, E., Donnelly, R. F., & Paredes, A. J. (2022). On a highway to the brain: A review on nose-to-brain drug delivery using nanoparticles. In Applied Materials Today (Vol. 29). Elsevier Ltd. https://doi.org/10.1016/j.apmt.2022.101631
  31. Gao, Y., Han, D., & Feng, J. (2021). MicroRNA in multiple sclerosis. In Clinica Chimica Acta (Vol. 516, pp. 92–99). Elsevier B.V. https://doi.org/10.1016/j.cca.2021.01.020