14.03.2024 Views

Review: MSCs and Exosomes Production

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

www.pelobiotech.com<br />

<strong>MSCs</strong> <strong>and</strong> Extracellular Vesicle <strong>Production</strong>:<br />

A Gateway to Regenerative Medicine<br />

Mesenchymal stem / stromal cells (<strong>MSCs</strong>) have emerged as a promising tool in regenerative<br />

medicine due to their unique properties, including self-renewal <strong>and</strong> differentiation capability.<br />

One of the key mechanisms through which <strong>MSCs</strong> exert their therapeutic effects is via paracrine<br />

signalling, which includes the production <strong>and</strong> secretion of Extracellular Vesicle (EVs), often<br />

termed as exosomes or macrovesicles that play a crucial role in intercellular communication<br />

<strong>and</strong> tissue repair. This article provides an overview of <strong>MSCs</strong>, their role in EVs production, <strong>and</strong><br />

the potential applications of MSC-derived EVs in regenerative medicine. In the scientific<br />

community is a very active discussion regarding the appropriate nomenclature for<br />

mesenchymal stem cells (<strong>MSCs</strong>): Some experts suggest <strong>MSCs</strong> should be more accurately<br />

termed "stromal cells" to reflect their supportive role in tissue rather than their stem cell-like<br />

qualities 24-27 . So here we will incorporate both names <strong>and</strong> refer to these cells mesenchymal<br />

stem/stromal cells (<strong>MSCs</strong>) throughout this article.<br />

Underst<strong>and</strong>ing <strong>MSCs</strong><br />

<strong>MSCs</strong> non-hematopoietic, multipotent, adult stem / stromal cells that can be isolated from<br />

various biological sources such as bone marrow, adipose tissue, Wharton's jelly of the<br />

umbilical cord, brain, spleen, kidneys, liver, placenta, dental pulp, neurons, lungs, skin, <strong>and</strong><br />

Tel: +49 (0) 89 517 286 59 0<br />

Mail: info@pelobiotech.com


www.pelobiotech.com<br />

breast milk. <strong>MSCs</strong> have further been defined by the International Society for Cellular Therapy<br />

(ISCT) based on specific criteria outlined in a position statement from 2006. According to these<br />

criteria, <strong>MSCs</strong> must exhibit certain characteristics to be classified as such. These include the<br />

ability to adhere to plastic surfaces during in vitro culture, expression of specific surface<br />

markers such as CD105, CD73, <strong>and</strong> CD90 while lacking CD45, CD34, CD14, or CD11b,<br />

expression markers <strong>and</strong> the capacity to differentiate into osteoblasts, adipocytes, <strong>and</strong><br />

chondrocytes under specific in vitro conditions 22,23 . These criteria are crucial for the<br />

identification <strong>and</strong> st<strong>and</strong>ardization of <strong>MSCs</strong> across various tissue sources. Importantly, the<br />

tissue source of <strong>MSCs</strong> can influence their therapeutic potential, making it essential to<br />

underst<strong>and</strong> the differences between <strong>MSCs</strong> isolated from different tissues to predict their<br />

behaviour <strong>and</strong> widen their clinical use 1 .<br />

Extracellular Vesicles: Nature's Nanoscale Messengers<br />

EVs, nanoscale membrane-bound vesicles released by various cell types, including <strong>MSCs</strong>, play<br />

a vital role in an array of cellular functions, including intercellular communication, cell<br />

differentiation, <strong>and</strong> proliferation, angiogenesis, stress response, <strong>and</strong> immune signalling. The<br />

ability to carry out these different functions is because of the complexity of EVs. These vesicles<br />

carry <strong>and</strong> transfer functional cargo like proteins, messenger RNAs, microRNAs, cytokines,<br />

lipids, cell surface receptors, enzymes, <strong>and</strong> transcription factors from cells to the recipient cells.<br />

Their sizes range from 30 to 150 nanometres, originating from a specialized biogenesis<br />

pathway.<br />

The composition of EVs is contingent on the donor cell type <strong>and</strong> the physiological context of<br />

production. EVs interact with recipient cells through specific adhesion molecules <strong>and</strong> can be<br />

internalized via multiple pathways, dependent on the proximity of the target cells. The nucleic<br />

acid content in EVs is particularly influential in their functional capacity. They harbour distinct<br />

markers including tetraspanins (CD9, CD63, CD81), integrins, MHC molecules, HSP70,<br />

HSP90, Alix, TSG101, <strong>and</strong> GTPases. The lipid bilayer encapsulating EVs confers stability <strong>and</strong><br />

protection, facilitating their biological roles. <strong>MSCs</strong> release large amounts of EVs for cell-tocell<br />

communication, maintaining a dynamic <strong>and</strong> homeostatic microenvironment for tissue<br />

repair <strong>and</strong> regeneration 2,28 . Furthermore, EVs have been implicated in various physiological<br />

<strong>and</strong> pathological processes, including cardiovascular diseases <strong>and</strong> neurogenesis 3,29-31 .<br />

Tel: +49 (0) 89 517 286 59 0<br />

Mail: info@pelobiotech.com


www.pelobiotech.com<br />

MSC-Derived EVs: Key Agents in Regenerative Medicine<br />

Past research has demonstrated that, despite positive effects in various settings, <strong>MSCs</strong> were<br />

barely detected in affected tissues, resulting in the hypothesis that they mainly act via their<br />

secretome rather than in a direct cellular manner 15 . Using the examples of an acute kidney<br />

injury model <strong>and</strong> a myocardial infarction model that <strong>MSCs</strong> were found to exert their<br />

therapeutic effects EVs 33 .<br />

MSC-derived EVs have become key players in regenerative medicine, showcasing a broad<br />

range of therapeutic effects. Originating from <strong>MSCs</strong>, these EVs carry immunomodulatory,<br />

regenerative, <strong>and</strong> anti-inflammatory traits, making them highly effective in tissue repair,<br />

angiogenesis, inflammation control, <strong>and</strong> wound healing. They contribute significantly to<br />

critical cellular processes, including angiogenesis, fibrosis reduction, <strong>and</strong> remodelling of the<br />

extracellular matrix. They are particularly promising for treating a spectrum of conditions such<br />

as cardiovascular, renal, hepatic, pulmonary, <strong>and</strong> neurodegenerative diseases, <strong>and</strong> they also<br />

exhibit antimicrobial effects.<br />

Unlike <strong>MSCs</strong> themselves, which can pose challenges related to cell viability, potential for<br />

immune rejection, <strong>and</strong> the complexity of direct use in regenerative therapies, MSC-EVs offer<br />

a safer, more stable, <strong>and</strong> potentially more effective alternative. In contrast to cell therapies,<br />

EVs are not self-replicating, <strong>and</strong> they lack endogenous tumour formation potentials. EVs do<br />

not seem to sense environmental conditions, <strong>and</strong> thus, their biological activity can be predicted<br />

more reliably than that of cells. Preconditioning <strong>and</strong> engineering techniques have enhanced the<br />

efficacy of MSC-EVs, paving the way for improved outcomes in cell-free therapeutic<br />

interventions 8 . This evolution emphasizes the strategic advantage of utilizing MSC-EVs over<br />

direct MSC therapies, as they represent an innovative method for harnessing the full<br />

regenerative potential of mesenchymal stem cells, setting a new st<strong>and</strong>ard in medical treatments.<br />

Culture <strong>and</strong> Expansion of <strong>MSCs</strong> for Extracellular Vesicle<br />

<strong>Production</strong><br />

The origin, culture, <strong>and</strong> expansion of <strong>MSCs</strong> are crucial for EVs production. The choice of<br />

expansion method significantly impacts EVs yield <strong>and</strong> quality.<br />

The field of MSC research faces challenges due to the inherent tendency of primary <strong>MSCs</strong> to<br />

undergo senescence during culture expansion. This limitation has prompted researchers to<br />

explore the generation <strong>and</strong> characterization of immortalized MSC (iMSC) lines as a potential<br />

solution. IMSC lines, such as those created by inducing the expression of human telomerase<br />

reverse transcriptase (hTERT), have been investigated for their ability to offer a reliable <strong>and</strong><br />

scalable source of <strong>MSCs</strong> for EVs production 17 . Studies have indicated that iMSC lines could<br />

serve as a consistent resource for EVs production, which is crucial for various therapeutic<br />

applications. However, it is important to note that i<strong>MSCs</strong> may exhibit functional alterations<br />

compared to primary <strong>MSCs</strong> 18 .<br />

Tel: +49 (0) 89 517 286 59 0<br />

Mail: info@pelobiotech.com


www.pelobiotech.com<br />

This difference in functionality raises concerns about the suitability of i<strong>MSCs</strong> for certain<br />

applications <strong>and</strong> underscores the importance of further research to underst<strong>and</strong> the implications<br />

of iMSC behaviour <strong>and</strong> characteristics. Moreover, the source of <strong>MSCs</strong>, whether primary,<br />

induced pluripotent stem cell (iPSC)-derived, or immortalized, can influence EVs production.<br />

While iPSC derived <strong>MSCs</strong> have shown promise for specific applications, primary <strong>MSCs</strong> are<br />

still preferred in many cases due to their superior supportive capabilities in co-culture<br />

systems 19 . The choice of MSC source is a critical consideration in EVs production, as it can<br />

impact the quantity <strong>and</strong> quality of EVs generated for therapeutic purposes.<br />

The culture media used for MSC expansion can also influence EVs production <strong>and</strong><br />

functionality. The use of defined media has been suggested as advantageous for maintaining<br />

the desired characteristics of <strong>MSCs</strong> <strong>and</strong> their derived EVs 20 . Additionally, it may be necessary<br />

to add special lipid cocktails for high EVs production. The balance between high<br />

proliferation/expansion rates <strong>and</strong> EVs production is a critical consideration. While high<br />

proliferation rates are desirable for obtaining large quantities of <strong>MSCs</strong>, it may lead to<br />

competition for resources, such as lipids, which are essential for both proliferation <strong>and</strong> EVs<br />

biogenesis 21 . Studies have indicated that the efficiency of EVs production may inversely<br />

correlate with the developmental maturity of the MSC donor, further highlighting the<br />

importance of donor selection for optimal EVs yield 21 .<br />

Moreover, the choice of having serum in the cell culture, such as FBS, hPL, or AB serum, can<br />

introduce both variability in proliferation, <strong>and</strong> expansion. Utilizing defined media can help to<br />

overcome these batch-to-batch variabilities <strong>and</strong> ensure consistent functional EVs production 20 .<br />

In conclusion, the culture <strong>and</strong> expansion of <strong>MSCs</strong> for EVs production involve various factors<br />

that influence the quantity <strong>and</strong> quality of EVs. Careful consideration of expansion methods,<br />

culture media, cell source, proliferation rates, <strong>and</strong> serum choice is essential to optimize EVs<br />

production for therapeutic applications. Special lipid cocktails may be necessary to enhance<br />

EVs production efficiency <strong>and</strong> yield, further emphasizing the importance of optimizing culture<br />

conditions for successful EVs-based therapies.<br />

Isolation Techniques for MSC-Derived EVs<br />

Isolating EVs from <strong>MSCs</strong> is a noteworthy area of research, essential for obtaining pure EVs<br />

samples for applications ranging from therapeutic use, drug delivery, regenerative medicine,<br />

<strong>and</strong> tissue engineering. Various methods such as ultracentrifugation, differential<br />

ultracentrifugation, <strong>and</strong> tangential flow filtration are employed, each with its distinct<br />

advantages <strong>and</strong> limitations 4 .<br />

• Ultracentrifugation: Ultracentrifugation is one of the most traditional <strong>and</strong> widely used<br />

methods for Extracellular Vesicles isolation. This technique relies on the application of<br />

extremely high centrifugal forces, typically ranging from 100,000 to 200,000g to<br />

sediment EVs from MSC culture media or other biological fluids. The process involves<br />

multiple centrifugation steps at varying speeds <strong>and</strong> durations to progressively remove<br />

cells, cell debris, <strong>and</strong> larger vesicles, culminating in the sedimentation of EVs.<br />

o<br />

Advantages:<br />

Widely available: The equipment required for ultracentrifugation is available in<br />

most research laboratories.<br />

Tel: +49 (0) 89 517 286 59 0<br />

Mail: info@pelobiotech.com


www.pelobiotech.com<br />

Scalable: It can be adapted for large-volume preparations, making it suitable for<br />

both research <strong>and</strong> clinical applications.<br />

o<br />

Limitations:<br />

Time-consuming: The process is labor-intensive <strong>and</strong> requires several hours to<br />

complete.<br />

Potential for contamination: Co-isolation of protein aggregates or other vesicles<br />

of similar density can occur.<br />

Sample integrity: The high forces applied can potentially damage the EVs or<br />

alter their functional properties.<br />

• Differential Ultracentrifugation<br />

Differential ultracentrifugation refines the basic ultracentrifugation process by<br />

employing a series of centrifugation steps at gradually increasing speeds. This method<br />

allows for more precise separation of EVs from other components based on their size<br />

<strong>and</strong> density.<br />

o<br />

o<br />

Advantages:<br />

Improved purity: By carefully adjusting the centrifugation parameters, it is<br />

possible to enhance the purity of the isolated EVs.<br />

Versatility: It can be used in conjunction with other purification steps to further<br />

increase the yield <strong>and</strong> purity of EVs.<br />

Limitations:<br />

Complexity: The process requires meticulous optimization of centrifugation<br />

speeds <strong>and</strong> times for each specific sample type.<br />

Sample loss: Each centrifugation step may lead to a loss of EVs yield.<br />

• Tangential Flow Filtration (TFF) 32<br />

Tangential flow filtration is a more modern approach that utilizes a crossflow<br />

mechanism, where the sample fluid flows tangentially across the surface of a membrane<br />

filter. This method effectively separates EVs based on their size, allowing them to pass<br />

through the membrane while larger particles are retained.<br />

o<br />

o<br />

Advantages:<br />

Efficiency: TFF can process large volumes of samples in a relatively short<br />

amount of time.<br />

Gentle on samples: The technique is less likely to damage EVs compared to<br />

ultracentrifugation.<br />

Scalability <strong>and</strong> reproducibility: TFF is easily scalable <strong>and</strong> offers high<br />

reproducibility, making it suitable for clinical applications.<br />

Limitations:<br />

Equipment cost: The initial investment for TFF systems can be high.<br />

Membrane maintenance: Over time, the membrane may become clogged with<br />

particles, requiring regular maintenance or replacement.<br />

The choice of an EVs isolation technique depends on various factors, including the source of<br />

<strong>MSCs</strong>, the volume of the sample, the desired purity <strong>and</strong> yield of EVs, <strong>and</strong> the available<br />

Tel: +49 (0) 89 517 286 59 0<br />

Mail: info@pelobiotech.com


www.pelobiotech.com<br />

resources. Each method has its trade-offs; thus, a combination of techniques should be used to<br />

achieve the best results. Continuous advancements in EVs isolation technologies are expected<br />

to enhance the efficiency, yield, <strong>and</strong> purity of EVs preparations.<br />

Enhancing EVs <strong>Production</strong> from <strong>MSCs</strong><br />

Optimizing the production of EVs from <strong>MSCs</strong> can significantly lead to more effective application<br />

possibilities by ensuring that enough potent, high-quality EVs are available for research <strong>and</strong> clinical<br />

therapy.<br />

Currently, several strategies are being developed to boost EVs production:<br />

• Culturing with Bioactive Glass (BG) Ion Products: Culturing <strong>MSCs</strong> with BG ion productenriched<br />

medium significantly increases Extracellular Vesicles production without altering<br />

their inherent characteristics 5 .<br />

• Use of Small Molecules: Identified specific small molecules capable of enhancing<br />

Extracellular Vesicles production in <strong>MSCs</strong>, with ongoing research exploring their effects on<br />

the EVs composition <strong>and</strong> regenerative capacity 6 .<br />

• Preconditioning <strong>and</strong> Engineering: Innovative strategies such as preconditioning <strong>MSCs</strong> <strong>and</strong><br />

engineering EVs are being investigated to amplify the therapeutic activity of MSC-EVs 7 .<br />

Navigating the Evolving L<strong>and</strong>scape of Engineered EVs Therapies:<br />

Opportunity <strong>and</strong> Challenges in Clinical Translation<br />

The l<strong>and</strong>scape of EV-based therapies growing exponentially, with over 150 clinical trials,<br />

spanning various domains such as respiratory disorders, infectious diseases, <strong>and</strong> oncology 9 .<br />

Notably, MSC-EVs are particularly promising, offering a compelling alternative to traditional<br />

stem cell therapies. As we have talked earlier, MSC-EVs can replicate the therapeutic impacts<br />

of their source <strong>MSCs</strong>, with added benefits like reduced size, increased stability, <strong>and</strong> more<br />

versatile administration routes 10 . Various companies are at the forefront of advancing the<br />

therapeutic potential of EVs through the engineering of EVs membrane proteins. These<br />

developments have led to innovative treatments, such as the creation of inhalable COVID-19<br />

vaccines utilizing EVs derived from lung stem cells. The contributions from multiple<br />

companies have played a significant role in driving progress in this field. The exciting world<br />

of engineered Extracellular Vesicles therapy is on the brink of transforming how we approach<br />

healing, opening a whole new world of medical possibilities 11 . Despite the promise of MSC-<br />

EVs, challenges persist in translating these therapies from bench to bedside. Issues concerning<br />

safety, st<strong>and</strong>ardized isolation protocols, <strong>and</strong> EVs characterization require resolution 12 .<br />

Additionally, the heterogeneity of EVs populations, influenced by extracellular environmental<br />

factors, complicates their therapeutic application 13 . A deeper underst<strong>and</strong>ing of exosomal cargo<br />

<strong>and</strong> its disease-specific roles is imperative for the full realization of exosomal potential in<br />

clinical settings.<br />

Lastly, the scale-up of MSC-EVs production for clinical applications encounters significant<br />

difficulties, primarily due to the substantial volume required to treat a single patient. Traditional<br />

volume reduction methods, such as ultracentrifugation, are notably inefficient for this scale,<br />

Tel: +49 (0) 89 517 286 59 0<br />

Mail: info@pelobiotech.com


www.pelobiotech.com<br />

with the maximum processing volume per run capped at under 500 mL, starkly inadequate for<br />

the quantities needed for EV-based therapeutics. This limitation highlights a critical bottleneck<br />

in the transition from laboratory-scale research to clinical applications. Key challenges include<br />

maintaining the purity <strong>and</strong> functionality of EVs, ensuring consistent quality across batches,<br />

source of EVs, isolation methods, <strong>and</strong> biodistribution, which are crucial for the successful<br />

translation of MSC-EVs into clinical use.<br />

In summary, the synergy between <strong>MSCs</strong> <strong>and</strong> EVs is illuminating new frontiers in regenerative<br />

medicine. As we unravel the complexities of MSC-EVs, we edge closer to a new epoch of<br />

therapeutic interventions that are safer, more efficacious, <strong>and</strong> transformative. These diminutive<br />

vesicles, emerging from the intricacies of cellular communication, hold the potential to redefine<br />

medical treatments, offering renewed hope for patients worldwide.<br />

References<br />

1. Cuesta-Gomez, N., Graham, G. J., & Campbell, J. (2021). Chemokines <strong>and</strong> their receptors: predictors of the<br />

therapeutic potential of mesenchymal stromal cells. Journal of Translational Medicine, 19(1).<br />

https://doi.org/10.1186/s12967-021-02822-5<br />

2. Ti, D., Hao, H., Tong, C., Liu, J., Dong, L., Zheng, J., … & Han, W. (2015). Lps-preconditioned mesenchymal<br />

stromal cells modify macrophage polarization for resolution of chronic inflammation via Extracellular Vesiclesshuttled<br />

let-7b. Journal of Translational Medicine, 13(1). https://doi.org/10.1186/s12967-015-0642-6<br />

3. Tian, J., Popal, M. S., Zhao, Y., Liu, Y., Chen, K., & Liu, Y. (2019). Interplay between Extracellular Vesicless <strong>and</strong><br />

autophagy in cardiovascular diseases: novel promising target for diagnostic <strong>and</strong> therapeutic application. Aging <strong>and</strong><br />

Disease, 10(6), 1302. https://doi.org/10.14336/ad.2018.1020<br />

4. Helwa I, Cai J, Drewry MD, Zimmerman A, Dinkins MB, Khaled ML, Seremwe M, Dismuke WM, Bieberich E,<br />

Stamer WD, Hamrick MW, Liu Y. A Comparative Study of Serum Extracellular Vesicles Isolation Using<br />

Differential Ultracentrifugation <strong>and</strong> Three Commercial Reagents. PLoS One. 2017 Jan 23;12(1): e0170628. doi:<br />

10.1371/journal.pone.0170628.<br />

5. Zhi Wu, Dan He, Haiyan Li. Bioglass enhances the production of Extracellular Vesicless <strong>and</strong> improves their<br />

capability of promoting vascularization. Bioactive materials (2021) doi: 10.1016/j.bioactmat.2020.09.011.<br />

6. Wang J, Bonacquisti EE, Brown AD, Nguyen J. Boosting the Biogenesis <strong>and</strong> Secretion of Mesenchymal Stem Cell-<br />

Derived Extracellular Vesicless. Cells. 2020 Mar 9;9(3):660. doi: 10.3390/cells9030660.<br />

7. Chen S, Sun F, Qian H, Xu W, Jiang J. Preconditioning <strong>and</strong> Engineering Strategies for Improving the Efficacy of<br />

Mesenchymal Stem Cell-Derived Extracellular Vesicless in Cell-Free Therapy. Stem Cells Int. 2022 May 14;<br />

2022:1779346. doi: 10.1155/2022/1779346.<br />

8. Lee, J., Park, B., Kim, J., Choo, Y. W., Kim, H. Y., Yoon, J., … & Kim, B. S. (2020). Nanovesicles derived from<br />

iron oxide nanoparticles–incorporated mesenchymal stem cells for cardiac repair. Science Advances, 6(18).<br />

https://doi.org/10.1126/sciadv.aaz0952<br />

9. Mendt, M. C., Rezvani, K., & Shpall, E. J. (2019). Mesenchymal stem cell-derived Extracellular Vesicless for<br />

clinical use. Bone Marrow Transplantation, 54(S2), 789-792. https://doi.org/10.1038/s41409-019-0616-z<br />

10. Levy, O., Kuai, R., Siren, E. M. J., Bhere, D., Milton, Y., Nissar, N., … & Karp, J. M. (2020). Shattering barriers<br />

toward clinically meaningful <strong>MSCs</strong> therapies. Science Advances, 6(30). https://doi.org/10.1126/sciadv.aba6884<br />

11. Yin, K., Wang, S., & Zhao, R. C. (2019). Extracellular Vesicless from mesenchymal stem/stromal cells: a new<br />

therapeutic paradigm. Biomarker Research, 7(1). https://doi.org/10.1186/s40364-019-0159-x<br />

12. Lee, B., Kang, I. H., & Yu, K. (2021). Therapeutic features <strong>and</strong> updated clinical trials of mesenchymal stem cell<br />

(<strong>MSCs</strong>)-derived Extracellular Vesicless. Journal of Clinical Medicine, 10(4), 711.<br />

https://doi.org/10.3390/jcm10040711<br />

13. Ahmadi, M. <strong>and</strong> Rezaie, J. (2020). Ageing <strong>and</strong> mesenchymal stem cells derived Extracellular Vesicless: molecular<br />

insight <strong>and</strong> challenges. Cell Biochemistry <strong>and</strong> Function, 39(1), 60-66. https://doi.org/10.1002/cbf.3602<br />

14. Xu, H., Chen, L., Zhou, S., Li, Y., & Xiang, C. (2020). Multifunctional role of micrornas in mesenchymal stem cellderived<br />

Extracellular Vesicless in treatment of diseases. World Journal of Stem Cells, 12(11), 1276-1294.<br />

https://doi.org/10.4252/wjsc.v12.i11.1276<br />

15. Arnold I. Caplan, Diego Correa, The MSC: An Injury Drugstore. Volume 9, Issue 1, 8 July 2011, Pages 11-15.<br />

https://doi.org/10.1016/j.stem.2011.06.008<br />

16. BioRender. 2023. “Sources <strong>and</strong> Potential Applications of Mesenchymal Stromal Cells” https://www.biorender.com/.<br />

17. Burk, J., Holl<strong>and</strong>, H., Lauermann, A., May, T., Siedlaczek, P., Charwat, V., … & Kasper, C. (2019). Generation <strong>and</strong><br />

characterization of a functional human adipose‐derived multipotent mesenchymal stromal cell line. Biotechnology<br />

<strong>and</strong> Bioengineering, 116(6), 1417-1426. https://doi.org/10.1002/bit.26950<br />

Tel: +49 (0) 89 517 286 59 0<br />

Mail: info@pelobiotech.com


www.pelobiotech.com<br />

18. Piñeiro-Ramil, M., Sanjurjo-Rodríguez, C., Rodríguez-Fernández, S., Castro-Viñuelas, R., Hermida-Gómez, T.,<br />

Blanco, F., … & Díaz-Prado, S. (2021). Generation of mesenchymal cell lines derived from aged donors.<br />

International Journal of Molecular Sciences, 22(19), 10667. https://doi.org/10.3390/ijms221910667<br />

19. Vasko, T., Frobel, J., Lubberich, R., Goecke, T., & Wagner, W. (2016). Ipsc-derived mesenchymal stromal cells are<br />

less supportive than primary mscs for co-culture of hematopoietic progenitor cells. Journal of Hematology &<br />

Oncology, 9(1). https://doi.org/10.1186/s13045-016-0273-2<br />

20. Wang, J., Bonacquisti, E., Brown, A., & Nguyen, J. (2020). Boosting the biogenesis <strong>and</strong> secretion of mesenchymal<br />

stem cell-derived Extracellular Vesicless. Cells, 9(3), 660. https://doi.org/10.3390/cells9030660<br />

21. Chen, T., Yeo, R., Arslan, F., Yin, Y., Tan, S., Lai, R., … & Lim, S. (2013). Efficiency of Extracellular Vesicles<br />

production correlates inversely with the developmental maturity of msc donor. Journal of Stem Cell Research &<br />

Therapy, 3(3). https://doi.org/10.4172/2157-7633.1000145<br />

22. Dominici, M., Blanc, K. L., Mueller, I., Slaper‐Cortenbach, I., Marini, F. C., Krause, D. S., … & Horwitz, E. M.<br />

(2006). Minimal criteria for defining multipotent mesenchymal stromal cells. the international society for cellular<br />

therapy position statement. Cytotherapy, 8(4), 315-317. https://doi.org/10.1080/14653240600855905<br />

23. Murray, I. R., Chahla, J., Safran, M. R., Krych, A. J., Saris, D. B., Caplan, A. I., … & Nakamura, N. (2019).<br />

International expert consensus on a cell therapy communication tool: doses. Journal of Bone <strong>and</strong> Joint Surgery,<br />

101(10), 904-911. https://doi.org/10.2106/jbjs.18.00915<br />

24. Caplan AI. Mesenchymal Stem Cells: Time to Change the Name! Stem Cells Transl Med. 2017 Jun;6(6):1445-1451.<br />

doi: 10.1002/sctm.17-0051. Epub 2017 Apr 28. PMID: 28452204; PMCID: PMC5689741.<br />

25. Lindner U, Kramer J, Rohwedel J, Schlenke P. Mesenchymal Stem or Stromal Cells: Toward a Better Underst<strong>and</strong>ing<br />

of Their Biology? Transfus Med Hemother. 2010 Apr;37(2):75-83. doi: 10.1159/000290897. Epub 2010 Mar 15.<br />

PMID: 20737049; PMCID: PMC2914415.<br />

26. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj,<br />

Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for<br />

Cellular Therapy position statement. Cytotherapy. 2006;8(4):315-7. doi: 10.1080/14653240600855905. PMID:<br />

16923606.<br />

27. Elahi FM, Farwell DG, Nolta JA, Anderson JD. Preclinical translation of Extracellular Vesicless derived from<br />

mesenchymal stem/stromal cells. Stem Cells. 2020 Jan;38(1):15-21. doi: 10.1002/stem.3061. Epub 2019 Oct 1.<br />

PMID: 31381842; PMCID: PMC7004029.<br />

28. Yáñez-Mó M, Silj<strong>and</strong>er PR, Andreu Z, Zavec AB, Borràs FE, Buzas EI, Buzas K, Casal E, Cappello F, Carvalho J,<br />

Colás E, Cordeiro-da Silva A, Fais S, Falcon-Perez JM, Ghobrial IM, Giebel B, Gimona M, Graner M, Gursel I,<br />

Gursel M, Heegaard NH, Hendrix A, Kierulf P, Kokubun K, Kosanovic M, Kralj-Iglic V, Krämer-Albers EM,<br />

Laitinen S, Lässer C, Lener T, Ligeti E, Linē A, Lipps G, Llorente A, Lötvall J, Manček-Keber M, Marcilla A,<br />

Mittelbrunn M, Nazarenko I, Nolte-'t Hoen EN, Nyman TA, O'Driscoll L, Olivan M, Oliveira C, Pállinger É, Del<br />

Portillo HA, Reventós J, Rigau M, Rohde E, Sammar M, Sánchez-Madrid F, Santarém N, Schallmoser K, Ostenfeld<br />

MS, Stoorvogel W, Stukelj R, Van der Grein SG, Vasconcelos MH, Wauben MH, De Wever O. Biological properties<br />

of extracellular vesicles <strong>and</strong> their physiological functions. J Extracell Vesicles. 2015 May 14;4:27066. doi:<br />

10.3402/jev.v4.27066. PMID: 25979354; PMCID: PMC4433489.<br />

29. Lener T, Gimona M, Aigner L, Börger V, Buzas E, Camussi G, Chaput N, Chatterjee D, Court FA, Del Portillo HA,<br />

O'Driscoll L, Fais S, Falcon-Perez JM, Felderhoff-Mueser U, Fraile L, Gho YS, Görgens A, Gupta RC, Hendrix A,<br />

Hermann DM, Hill AF, Hochberg F, Horn PA, de Kleijn D, Kordelas L, Kramer BW, Krämer-Albers EM, Laner-<br />

Plamberger S, Laitinen S, Leonardi T, Lorenowicz MJ, Lim SK, Lötvall J, Maguire CA, Marcilla A, Nazarenko I,<br />

Ochiya T, Patel T, Pedersen S, Pocsfalvi G, Pluchino S, Quesenberry P, Reischl IG, Rivera FJ, Sanzenbacher R,<br />

Schallmoser K, Slaper-Cortenbach I, Strunk D, Tonn T, Vader P, van Balkom BW, Wauben M, Andaloussi SE,<br />

Théry C, Rohde E, Giebel B. Applying extracellular vesicles based therapeutics in clinical trials - an ISEV position<br />

paper. J Extracell Vesicles. 2015 Dec 31;4:30087. doi: 10.3402/jev.v4.30087. PMID: 26725829; PMCID:<br />

PMC4698466.<br />

30. Agnes T. Reiner, Kenneth W. Witwer, Bas W.M. van Balkom, Joel de Beer, Chaya Brodie, R<strong>and</strong>olph L. Corteling,<br />

Susanne Gabrielsson, Mario Gimona, Ahmed G. Ibrahim, Dominique de Kleijn, Charles P. Lai, Jan Lötvall,<br />

Hern<strong>and</strong>o A. del Portillo, Ilona G. Reischl, Milad Riazifar, Carlos Salomon, Hidetoshi Tahara, Wei Seong Toh,<br />

Marca H.M. Wauben, Vicky K. Yang, Yijun Yang, Ronne Wee Yeh Yeo, Hang Yin, Bernd Giebel, Eva Rohde, Sai<br />

Kiang Lim, Concise <strong>Review</strong>: Developing Best-Practice Models for the Therapeutic Use of Extracellular Vesicles,<br />

Stem Cells Translational Medicine, Volume 6, Issue 8, August 2017, Pages 1730–1739,<br />

https://doi.org/10.1002/sctm.17-0055<br />

31. Dirk M. Hermann, Thorsten R. Doeppner, <strong>and</strong> Bernd Giebel, "Extracellular Vesicles as Diagnostic Tool in Transient<br />

Ischemic Attack <strong>and</strong> Ischemic Stroke," Stroke, vol. 52, no. 9, pp. 3348–3350, Aug. 2021.<br />

https://doi.org/10.1161/STROKEAHA.121.036150<br />

32. Börger, V., Dittrich, R., Staubach, S., Zumegen, S., Horn, P., & Giebel, B. (2019). Tangential flow filtration, a<br />

potential method for the scaled preparation of extracellular vesicles. Cytotherapy, 21(5), S89.<br />

https://doi.org/10.1016/j.jcyt.2019.03.431&#8203<br />

33. Zhang K, Chen S, Sun H, Wang L, Li H, Zhao J, Zhang C, Li N, Guo Z, Han Z, Han ZC, Zheng G, Chen X, Li Z.<br />

In vivo two-photon microscopy reveals the contribution of Sox9+ cell to kidney regeneration in a mouse model with<br />

extracellular vesicle treatment. J Biol Chem. 2020 Aug 21;295(34):12203-12213. doi: 10.1074/jbc.RA120.012732.<br />

Epub 2020 Jul 8. PMID: 32641493; PMCID: PMC7443503.<br />

Tel: +49 (0) 89 517 286 59 0<br />

Mail: info@pelobiotech.com

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!