Biomedical Exploitation of Exosomes Delivered in Hydrogels: Comparison
Please note this is a comparison between Version 2 by Sirius Huang and Version 1 by Yaxin Xie.

Hydrogels are three-dimensional macromolecular polymeric networks composed of hydrophilic polymer chains. Exosomes functioning in the delivery of functional cargos are currently an active research hotspot. The biological features of exosomes make them suitable as potential therapeutics for the diagnosis and treatment of several diseases.

  • composite hydrogel
  • exosome
  • biomedical engineering

1. Introduction

Hydrogels are three-dimensional macromolecular polymeric networks composed of hydrophilic polymer chains. They can generally be divided into three categories according to their origin: natural, synthetic, and hybrid. Hydrogels are degradable, with a high affinity for water, and can be fabricated under physiological conditions, resulting in excellent biocompatibility [1]. They can be formed chemically and/or physically upon initiation with crosslinking agents and produced with a certain viscosity and elasticity. The innovation of Wichterle and Lim pioneered a new approach to applying crosslinked hydroxyethyl methacrylate (HEMA) hydrogels as biomaterials in 1960 [2]. In the two decades following this discovery, Lim and Sun demonstrated calcium alginate hydrogels with applications in cell encapsulation [3]. It is not surprising that hydrogels, having mechanical and structural properties similar to those of many tissues and the extracellular matrix (ECM), have attracted great attention, and significant progress has been made in designing, synthesizing, and using these materials for many biological and biomedical applications [4].
Exosomes are small, single-membrane, secreted extracellular vesicles (EVs), enriched in certain proteins, nucleic acids, and lipids. Budding at both the plasma and endosomal membranes of all the mammalian cell types studied to date, they are produced to remodel the ECM and deliver signals and functional macromolecules to adjacent cells. Numerous surface molecules on exosomes enable them to be internalized via endocytosis by recipient cells, playing an important role in regulating cell–cell communication [5]. Therefore, the study of exosomes in the pathology of various diseases is an active area of research, and the exploration of therapeutic exosomes as delivery vesicles has offered new insights for clinical applications in recent years. However, the stability and retention of exosomes released in vivo are major hurdles, as they are rapidly cleared by the innate immune system or accumulate in the liver, spleen, and lungs via the blood circulation [6].

The biological features of exosomes make them suitable as potential therapeutics for the diagnosis and treatment of several diseases. There are generally three approaches to obtaining exosomes with therapeutic and diagnostic potential. (1) Naturally derived exosomes (e.g., MSC-Exos) have been verified to be therapeutic by themselves [108][7]. (2) Engineering exosomes by transferring molecules such as microRNAs has achieved targeted applications [109][8]. (3) Exosome mimetics have been exploited as promising biomaterials [67,110][9][10].

2. Tissue Repair

Of the many classes of biomaterials that have been used in tissue repair, hydrogels have been regarded as one of the most prominent and versatile for supporting most cellular behaviors and nutrient transport. Protected by them, cellular secretions can maintain their biological activity and undergo controlled release in pathological environments (Table 1).
Table 1.
 Advances in tissue regeneration via the hydrogel encapsulation of EVs.

2.1. Bone and Cartilage Defects

Overwhelming evidence shows that the exogenous transport of miRNAs by exosomes can regulate osteogenic and angiogenic differentiation. An example of this is a study carried out by Mi et al., who created a cocktail therapy by transferring miR-26a-5p into endothelial cell-derived exosomes (EC-Exos) in an HA hydrogel. The EC-ExosmiR−26a−5p promoted osteogenic and osteoclast differentiation in mice with femoral fractures [109][8]. In another study, Hu et al. found that human umbilical cord MSC-derived small EVs (hUCMSC-sEVs) activated the PTEN/AKT signaling pathway by transferring miR-23a-3p when investigating the role and mechanism of cartilage regeneration [31][11]. Compared to increasing the specific miRNA in the target cells, the inhibition of miR-29a was verified to stimulate endogenous BMP/Smad signaling, which triggers subsequent osteogenic differentiation [67][9]. Therefore, the overexpression of miRNA can be an attractive method for improving the therapeutic effects. For example, miR-375 could be enriched in human adipose MSC (hASC)-derived exosomes by overexpressing the miRNA cargo in the parent cells [121][23].
Extensive research has shown that the essential properties of a bone and cartilage engineering scaffold are mechanical strength and a porous structure, to support the attachment and infiltration of osteogenic cells [122][24]. Hu et al. recently utilized an injectable and UV-crosslinked gelatin methacrylate (GelMA) to fabricate with nanoclay and achieved the sustained release of small EVs with the degradation of the hydrogel. The addition of laponite nanoclay significantly enhanced its ultimate strength for local administration in cartilage defects [31][11]. In addition to additives, 3D technology can also be applied to customize the shapes and sizes of porous scaffolds in accordance with bone defects. Fan et al. encapsulated umbilical MSC-derived exosomes (UMSC-Exos) in an HA hydrogel and combined them with 3D-printed nanohydroxyapatite/poly-ε-caprolactone (nHP) scaffolds [123][25]. Taken together, hydrogels can regulate extracellular matrix (ECM) formation, which provides a three-dimensional (3D) culture system for exosome secretion [89,124][26][27].
 

2.2. Wound Repair

As a complicated biological process, wound healing consists of inflammation, proliferation, and remodeling [125][28]. The conventional treatment of chronic wounds includes regular wound debridement for stimulating skin regeneration and the protection of the wound using a specific dressing [126][29]. Recent interventions inspired by cell therapy approaches involve exosomes derived from MSCs, plasma, and cancer cells, while stem cell-derived exosomes are being developed for tissue recovery [68,127,128][14][30][31]. In a diabetes-impaired wound model, a wound dressing biomaterial was applied by combining antioxidant polyurethane (PUAO) for attenuating oxidative stress and adipose-derived stem cell (ADSC) exosomes for tissue remodeling [128][31]. Similarly, immobilizing ADSC-derived exosomes in a composite hydrogel that includes poly-ε-L-lysine (EPL), a natural cationic polypeptide from Streptomyces albulus, can help to realize antibacterial activity and adhesive ability [129][32]. Another study explored the feasibility of a composite hydrogel formed from silk fibroin (SF) and silk sericin (SS) due to the excellent mechanical properties of SF, and the cell-adhesion and biocompatibility properties of SS. After encapsulating and delivering UMSC-Exos, SF–SS hydrogels promoted wound healing and angiogenesis [130][33]. Additionally, the delivery of platelet-rich plasma exosomes in a composite chitosan–silk hydrogel sponge was found to upregulate collagen synthesis and deposition, as well as angiogenesis, at the wound site in diabetic rat models [127][30]. In addition, exosomes were enriched in miR-21, miR-23a, miR-125b, and miR-14, which can be blocked to reduce scar formation when they are laden in hydrogels [131][34]. Chitosan hydrogels functionalized with exosomes from synovium MSCs transduced to overexpress miR-126 promoted healing and angiogenesis in skin wounds [132][35].

2.3. Cardiovascular Diseases

Ischemic myocardial infarction (MI) results from the severe blockage of blood arteries, which, in turn, interrupts nutrient supply. However, clinical treatments may lead to further myocardial ischemia/reperfusion injury [133][36]. New findings have triggered studies investigating the potential of utilizing MSC-derived EVs after MI to promote angiogenesis and restore cardiac function [117,134,135,136][19][37][38][39]. For example, Zou at al. elaborated an exo-anchoring conductive hydrogel enabling electrical conduction within the myocardial fibrotic area and promoting the synchronous contraction of the myocardium. In this study, an aniline tetramer (AT) was employed as a crosslinker, and the researchers endowed it with electroconductibility. The CP05 peptide was applied for its capability of binding to CD63 on the exosomal surface, to anchor and capture exosomes from human UC-MSCs [116][18]. Based on the intended application, hydrogels can be synthesized with different preparations. A notable application is to encapsulate EVs from induced pluripotent stem cells in a hydrogel patch and apply them directly onto the rat myocardium. The hydrogel patch enabled sustainable release, which protected the acutely injured heart against pathological hypertrophy [89][26].

2.4. Spinal Cord Injury

Spinal cord injury (SCI) is among the most fatal diseases of the central nervous system, resulting in a temporary or permanent loss of sensation, movement, strength, and body functions [137][40]. To overcome the low cell survival resulting from the inhibitory environment at the lesion site, the local injection of exosomes protected by hydrogels is a promising therapeutic strategy. Li et al. improved the affinity of HA hydrogels and MSC-derived exosomes by a laminin modification, and successfully promoted spinal cord regeneration and the recovery of hindlimb motor function in vivo [119][21]. Surprisingly, plant (e.g., ginseng)-derived exosomes that can stimulate the neural differentiation of BMSCs have been demonstrated, and can be loaded in GelMA to fit the irregular shapes of injury defects [138][41]. The promotion of angiogenesis is beneficial for the regeneration of neuronal networks after SCI. Inspired by this, Luo et al. utilized a hybrid hydrogel system comprising GelMA, HA-NB, and a photoinitiator (LAP) to immobile exosomes from M2 macrophages. The hydrogel-mediated release system protected the exosomes from severe oxidative stress and inflammation [129][32].

2.5. Other Diseases

In addition to the aforementioned applications, exosomes have also played important roles in periodontal, endometrial, and corneal repairs. In the context of periodontitis, the incorporation of dental pulp stem cell-derived exosomes and chitosan hydrogels repolarized macrophages and accelerated periodontal regeneration [108][7]. The dynamic coordination of adipose stem cell-derived exosomes and PEG hydrogels via Ag+–S resulted in outstanding injectable, self-healing, and antibacterial properties for endometrial and fertility restoration [113][15]. To effectively promote the repair of corneal damage, exosomes derived from MSCs were loaded in thermosensitive chitosan-based hydrogels [95][42].

3. Immune Regulation

Commonly, the adaptive immune response is regulated by antigen-presenting cells (APCs), such as dendritic cells (DCs), B cells, and macrophages, directly interacting with T cells and natural killer (NK) cells through cell-surface proteins [90][43]. Exosomes produced by APCs play an important role in the regulation of immunity, mediating immune stimulation or suppression, and driving inflammatory, autoimmune, and infectious disease pathology [96][44]. Inspired by dendritic cell-derived exosomes (DEXs), which improve cardiac function by activating CD4+ T cells in the spleen and lymph nodes [139][45], Zhang et al. encapsulated DEXs in a simple alginate hydrogel and injected the DEX-Gel into the MI model. The DEXs significantly upregulated the infiltration of Treg cells and M2 macrophages, which resulted in better wound remodeling, and preserved systolic function after MI. Furthermore, the combined application of the hydrogel provides physical support to the infarcted area [140][46].
MSCs confer regenerative effects in different tissue injuries, while in some cases, MSCs have been confirmed to secrete immunosuppressive cytokines and other factors, resulting in anti-inflammatory effects from stem cells [141][47]. Notably, the analysis of MSC-derived EVs revealed that they also have immunosuppressive therapeutic effects [142][48]. To harness EVs’ immunosuppressive properties, Fuhrmann et al. innovatively incorporated enzyme-loaded vesicles from MSCs into PVA hydrogels and applied this bioactive material for enzyme prodrug therapy. Once vesicles are released into the desired site, the injected nontoxic prodrugs are converted to anti-inflammatory drugs by enzymes [143][49]. The polarization of M2 macrophages, which can inhibit inflammation and induce tissue regeneration, has recently drawn great attention [108,109,144][7][8][50]. A classic cue is osteoimmunology, in which exosomes overexpressing miR-181 from human bone marrow-derived MSCs (hBM-MSCs) combined with a hydrogel were verified to significantly enhance osseointegration [144][50].
Tumor-derived EVs have been revealed to suppress tumor-specific and non-specific immune responses [96][44]. Metastatic melanoma releases a high level of exosomes carrying PD-L1 on their surfaces, which help in the evasion of immune surveillance. Based on how tumor cells suppress the immune system, Su et al. isolated exosomes from melanoma cells overexpressing PD-L1 to decrease T cell proliferation in a wound-healing model. The application of the thermoresponsive Pluronic F-127 hydrogel ensured that exosomes were released in a sustained manner [68][14].

4. Pathogenesis Study

Along with mediating physiological intercellular communication, exosomes also spread pathogenetic cargoes in diseases. Identifying the proteins and RNAs of exosomes can provide therapeutic targets. However, exosomal behavior can be dictated by the environment [4]. Therefore, hydrogels providing certain mechanical, structural, and compositional cues in the extracellular microenvironment are adopted as a novel strategy to recapitulate numerous physiologically relevant cell behaviors [145][51].
Tumor-derived exosomes can assist tumor growth and promote metastasis. To demonstrate the role of exosomes in ECM stiffness-triggered breast cancer invasiveness, Patwardhan et al. fabricated stiffness-tunable polyacrylamide (PA) gels as ECM mimics. Interestingly, stiff ECM cultures fostered exosome secretion by a series of changes in cell morphology, adhesion, and protrusion dynamics, which resulted in the invasion of breast cancer cells [146][52]. Aberrant cell behaviors can be induced by in vitro 2D culture, and the heterogeneity of exosomal behaviors also depends on the culture conditions [147][53]. Therefore, Millan et al. created 3D-engineered microtissues using the polysaccharides alginate and chitosan for the study of prostate cancer-derived EVs. Proteomics and RNA sequencing comparing 2D- and 3D-cultured cells revealed significantly differential expression of EV biomarkers. Some proteins known to be drivers of prostate cancer progression that were not detectable in the 2D conditions were enriched in the 3D cultures [148][54].
 
Exosomes from different cells such as endothelial cells and smooth muscle cells can contribute to atherosclerosis and cardiovascular disease when circulating in the blood [149,150][55][56]. In atherosclerosis-prone areas, EVs from smooth muscle cells (SMCs) and valvular interstitial cells (VICs) can cause a phospholipidic imbalance and, consequently, vascular and valvular calcification. Three-dimensional collagen hydrogels were utilized to produce a cardiovascular calcification model with which to observe the aggregation and microcalcification at the EV level [91][57]. Moreover, lesion macrophages can deliver exosomes that regulate vascular SMCs during the progression of atherosclerosis. In a study investigating the potential role of exosomes from nicotine-treated macrophages, Zhu et al. incorporated the above exosomes with chitosan hydrogels to stimulate release at the abdominal aorta [151][58].

References

  1. Zhao, X.H.; Chen, X.Y.; Yuk, H.; Lin, S.T.; Liu, X.Y.; Parada, G. Soft Materials by Design: Unconventional Polymer Networks Give Extreme Properties. Chem. Rev. 2021, 121, 4309–4372.
  2. Wichterle, O.; Lim, D. Hydrophilic gels for biological USE. Nature 1960, 185, 117–118.
  3. Lim, F.; Sun, A.M. Microencapsulated islets as bioartificial endocrine pancreas. Science 1980, 210, 908–910.
  4. Rosales, A.M.; Anseth, K.S. The design of reversible hydrogels to capture extracellular matrix dynamics. Nat. Rev. Mater. 2016, 1, 15012.
  5. Pegtel, D.M.; Gould, S.J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514.
  6. Riau, A.K.; Ong, H.S.; Yam, G.H.F.; Mehta, J.S. Sustained Delivery System for Stem Cell-Derived Exosomes. Front. Pharmacol. 2019, 10, 1368.
  7. Shen, Z.; Kuang, S.; Zhang, Y.; Yang, M.; Qin, W.; Shi, X.; Lin, Z. Chitosan hydrogel incorporated with dental pulp stem cell-derived exosomes alleviates periodontitis in mice via a macrophage-dependent mechanism. Bioact. Mater. 2020, 5, 1113–1126.
  8. Mi, B.; Chen, L.; Xiong, Y.; Yang, Y.; Panayi, A.C.; Xue, H.; Hu, Y.; Yan, C.; Hu, L.; Xie, X.; et al. Osteoblast/Osteoclast and Immune Cocktail Therapy of an Exosome/Drug Delivery Multifunctional Hydrogel Accelerates Fracture Repair. Acs Nano 2022, 6, 771–782.
  9. Fan, J.; Lee, C.S.; Kim, S.; Chen, C.; Aghaloo, T.; Lee, M. Generation of Small RNA-Modulated Exosome Mimetics for Bone Regeneration. ACS Nano 2020, 14, 11973–11984.
  10. Xu, Z.; Tsai, H.-i.; Xiao, Y.; Wu, Y.; Su, D.; Yang, M.; Zha, H.; Yan, F.; Liu, X.; Cheng, F.; et al. Engineering Programmed Death Ligand-1/Cytotoxic T-Lymphocyte-Associated Antigen-4 Dual-Targeting Nanovesicles for Immunosuppressive Therapy in Transplantation. Acs Nano 2020, 14, 7959–7969.
  11. Hu, H.; Dong, L.; Bu, Z.; Shen, Y.; Luo, J.; Zhang, H.; Zhao, S.; Lv, F.; Liu, Z. miR-23a-3p-abundant small extracellular vesicles released from Gelma/nanoclay hydrogel for cartilage regeneration. J. Extracell. Vesicles 2020, 9, 1778883.
  12. Guan, P.; Liu, C.; Xie, D.; Mao, S.; Ji, Y.; Lin, Y.; Chen, Z.; Wang, Q.; Fan, L.; Sun, Y. Exosome-loaded extracellular matrix-mimic hydrogel with anti-inflammatory property Facilitates/promotes growth plate injury repair. Bioact. Mater. 2022, 10, 145–158.
  13. Zhang, Y.; Wang, X.; Chen, J.; Qian, D.; Gao, P.; Qin, T.; Jiang, T.; Yi, J.; Xu, T.; Huang, Y.; et al. Exosomes derived from platelet-rich plasma administration in site mediate cartilage protection in subtalar osteoarthritis. J Nanobiotechnol. 2022, 20, 56.
  14. Su, D.; Tsai, H.I.; Xu, Z.; Yan, F.; Wu, Y.; Xiao, Y.; Liu, X.; Wu, Y.; Parvanian, S.; Zhu, W.; et al. Exosomal PD-L1 functions as an immunosuppressant to promote wound healing. J Extracell Vesicles 2019, 9, 1709262.
  15. Lin, J.; Wang, Z.; Huang, J.; Tang, S.; Saiding, Q.; Zhu, Q.; Cui, W. Microenvironment-Protected Exosome-Hydrogel for Facilitating Endometrial Regeneration, Fertility Restoration, and Live Birth of Offspring. Small 2021, 17, e2007235.
  16. Wang, C.; Liang, C.; Wang, R.; Yao, X.; Guo, P.; Yuan, W.; Liu, Y.; Song, Y.; Li, Z.; Xie, X. The fabrication of a highly efficient self-healing hydrogel from natural biopolymers loaded with exosomes for the synergistic promotion of severe wound healing. Biomater. Sci. 2019, 8, 313–324.
  17. Zhu, D.; Li, Z.; Huang, K.; Caranasos, T.G.; Rossi, J.S.; Cheng, K. Minimally invasive delivery of therapeutic agents by hydrogel injection into the pericardial cavity for cardiac repair. Nat. Commun. 2021, 12, 1412.
  18. Zou, Y.; Li, L.; Li, Y.; Chen, S.; Xie, X.; Jin, X.; Wang, X.; Ma, C.; Fan, G.; Wang, W. Restoring Cardiac Functions after Myocardial Infarction-Ischemia/Reperfusion via an Exosome Anchoring Conductive Hydrogel. Acs Appl. Mater. Interfaces 2021, 13, 56892–56908.
  19. Waters, R.; Alam, P.; Pacelli, S.; Chakravarti, A.R.; Ahmed, R.P.H.; Paul, A. Stem cell-inspired secretome-rich injectable hydrogel to repair injured cardiac tissue. Acta Biomater. 2018, 69, 95–106.
  20. Liang, L.; Shen, Y.; Dong, Z.; Gu, X. Photoacoustic image-guided corpus cavernosum intratunical injection of adipose stem cell-derived exosomes loaded polydopamine thermosensitive hydrogel for erectile dysfunction treatment. Bioact. Mater. 2022, 9, 147–156.
  21. Li, L.; Zhang, Y.; Mu, J.; Chen, J.; Zhang, C.; Cao, H.; Gao, J. Transplantation of Human Mesenchymal Stem-Cell-Derived Exosomes Immobilized in an Adhesive Hydrogel for Effective Treatment of Spinal Cord Injury. Nano Lett. 2020, 20, 4298–4305.
  22. Yu, H.; Cheng, J.; Shi, W.; Ren, B.; Zhao, F.; Shi, Y.; Yang, P.; Duan, X.; Zhang, J.; Fu, X.; et al. Bone marrow mesenchymal stem cell-derived exosomes promote tendon regeneration by facilitating the proliferation and migration of endogenous tendon stem/progenitor cells. Acta Biomater. 2020, 106, 328–341.
  23. Chen, S.; Tang, Y.; Liu, Y.; Zhang, P.; Lv, L.; Zhang, X.; Jia, L.; Zhou, Y. Exosomes derived from miR-375-overexpressing human adipose mesenchymal stem cells promote bone regeneration. Cell Prolif. 2019, 52, e12669.
  24. De Witte, T.-M.; Fratila-Apachitei, L.E.; Zadpoor, A.A.; Peppas, N.A. Bone tissue engineering via growth factor delivery: From scaffolds to complex matrices. Regen. Biomater. 2018, 5, 197–211.
  25. Wu, D.; Qin, H.; Wang, Z.; Yu, M.; Liu, Z.; Peng, H.; Liang, L.; Zhang, C.; Wei, X. Bone Mesenchymal Stem Cell-Derived sEV-Encapsulated Thermosensitive Hydrogels Accelerate Osteogenesis and Angiogenesis by Release of Exosomal miR-21. Front. Bioeng. Biotechnol. 2021, 9, 829136.
  26. Liu, B.; Lee, B.W.; Nakanishi, K.; Villasante, A.; Williamson, R.; Metz, J.; Kim, J.; Kanai, M.; Bi, L.; Brown, K.; et al. Cardiac recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat. Biomed. Eng. 2018, 2, 293–303.
  27. Yu, W.; Li, S.; Guan, X.; Zhang, N.; Xie, X.; Zhang, K.; Bai, Y. Higher yield and enhanced therapeutic effects of exosomes derived from MSCs in hydrogel-assisted 3D culture system for bone regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2022, 112646.
  28. Martin, P. Wound healing-Aiming for perfect skin regeneration. Science 1997, 276, 75–81.
  29. Frykberg, R.G.; Banks, J. Challenges in the Treatment of Chronic Wounds. Adv. Wound Care 2015, 4, 560–582.
  30. Xu, N.; Wang, L.; Guan, J.; Tang, C.; He, N.; Zhang, W.; Fu, S. Wound healing effects of a Curcuma zedoaria polysaccharide with platelet-rich plasma exosomes assembled on chitosan/silk hydrogel sponge in a diabetic rat model. Int. J. Biol. Macromol. 2018, 117, 102–107.
  31. Shiekh, P.A.; Singh, A.; Kumar, A. Exosome laden oxygen releasing antioxidant and antibacterial cryogel wound dressing OxOBand alleviate diabetic and infectious wound healing. Biomaterials 2020, 249, 120020.
  32. Shi, Q.; Qian, Z.; Liu, D.; Sun, J.; Wang, X.; Liu, H.; Xu, J.; Guo, X. GMSC-Derived Exosomes Combined with a Chitosan/Silk Hydrogel Sponge Accelerates Wound Healing in a Diabetic Rat Skin Defect Model. Front. Physiol. 2017, 8, 904.
  33. Han, C.; Liu, F.; Zhang, Y.; Chen, W.; Luo, W.; Ding, F.; Lu, L.; Wu, C.; Li, Y. Human Umbilical Cord Mesenchymal Stem Cell Derived Exosomes Delivered Using Silk Fibroin and Sericin Composite Hydrogel Promote Wound Healing. Front. Cardiovasc. Med. 2021, 8, 713021.
  34. Tao, S.C.; Guo, S.C.; Li, M.; Ke, Q.F.; Guo, Y.P.; Zhang, C.Q. Chitosan Wound Dressings Incorporating Exosomes Derived from MicroRNA-126-Overexpressing Synovium Mesenchymal Stem Cells Provide Sustained Release of Exosomes and Heal Full-Thickness Skin Defects in a Diabetic Rat Model. Stem Cells Transl. Med. 2017, 6, 736–747.
  35. Fang, S.; Xu, C.; Zhang, Y.T.; Xue, C.Y.; Yang, C.; Bi, H.D.; Qian, X.J.; Wu, M.J.; Ji, K.H.; Zhao, Y.P.; et al. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomal MicroRNAs Suppress Myofibroblast Differentiation by Inhibiting the Transforming Growth Factor-beta/SMAD2 Pathway During Wound Healing. Stem Cells Transl. Med. 2016, 5, 1425–1439.
  36. Hashimoto, H.; Olson, E.N.; Bassel-Duby, R. Therapeutic approaches for cardiac regeneration and repair. Nat. Rev. Cardiol. 2018, 15, 585–600.
  37. Zhang, K.; Zhao, X.; Chen, X.; Wei, Y.; Du, W.; Wang, Y.; Liu, L.; Zhao, W.; Han, Z.; Kong, D.; et al. Enhanced Therapeutic Effects of Mesenchymal Stem Cell-Derived Exosomes with an Injectable Hydrogel for Hindlimb Ischemia Treatment. ACS Appl Mater Interfaces 2018, 10, 30081–30091.
  38. Monguio-Tortajada, M.; Prat-Vidal, C.; Moron-Font, M.; Clos-Sansalvador, M.; Calle, A.; Gastelurrutia, P.; Cserkoova, A.; Morancho, A.; Ramirez, M.A.; Rosell, A.; et al. Local administration of porcine immunomodulatory, chemotactic and angiogenic extracellular vesicles using engineered cardiac scaffolds for myocardial infarction. Bioact. Mater. 2021, 6, 3314–3327.
  39. Wang, Q.; Zhang, L.; Sun, Z.; Chi, B.; Zou, A.; Mao, L.; Xiong, X.; Jiang, J.; Sun, L.; Zhu, W.; et al. HIF-1alpha overexpression in mesenchymal stem cell-derived exosome-encapsulated arginine-glycine-aspartate (RGD) hydrogels boost therapeutic efficacy of cardiac repair after myocardial infarction. Mater. Today Bio 2021, 12, 100171.
  40. Ahuja, C.S.; Wilson, J.R.; Nori, S.; Kotter, M.R.N.; Druschel, C.; Curt, A.; Fehlings, M.G. Traumatic spinal cord injury. Nat. Rev. Dis. Primers 2017, 3, 17018.
  41. Xu, X.H.; Yuan, T.J.; Dad, H.A.; Shi, M.Y.; Huang, Y.Y.; Jiang, Z.H.; Peng, L.H. Plant Exosomes As Novel Nanoplatforms for MicroRNA Transfer Stimulate Neural Differentiation of Stem Cells In Vitro and In Vivo. Nano Lett. 2021, 21, 8151–8159.
  42. Tang, Q.; Lu, B.; He, J.; Chen, X.; Fu, Q.; Han, H.; Luo, C.; Yin, H.; Qin, Z.; Lyu, D.; et al. Exosomes-loaded thermosensitive hydrogels for corneal epithelium and stroma regeneration. Biomaterials 2022, 280, 121320.
  43. Cheng, L.; Hill, A.F. Therapeutically harnessing extracellular vesicles. Nat. Rev. Drug Discov. 2022, 21, 379–399.
  44. Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208.
  45. Liu, H.; Gao, W.; Yuan, J.; Wu, C.; Yao, K.; Zhang, L.; Ma, L.; Zhu, J.; Zou, Y.; Ge, J. Exosomes derived from dendritic cells improve cardiac function via activation of CD4(+) T lymphocytes after myocardial infarction. J. Mol. Cell. Cardiol. 2016, 91, 123–133.
  46. Zhang, Y.; Cai, Z.; Shen, Y.; Lu, Q.; Gao, W.; Zhong, X.; Yao, K.; Yuan, J.; Liu, H. Hydrogel-load exosomes derived from dendritic cells improve cardiac function via Treg cells and the polarization of macrophages following myocardial infarction. J Nanobiotechnol. 2021, 19, 271.
  47. van Koppen, A.; Joles, J.A.; van Balkom, B.W.M.; Lim, S.K.; de Kleijn, D.; Giles, R.H.; Verhaar, M.C. Human embryonic mesenchymal stem cell-derived conditioned medium rescues kidney function in rats with established chronic kidney disease. PLoS ONE 2012, 7, e38746.
  48. Cantaluppi, V.; Gatti, S.; Medica, D.; Figliolini, F.; Bruno, S.; Deregibus, M.C.; Sordi, A.; Biancone, L.; Tetta, C.; Camussi, G. Microvesicles derived from endothelial progenitor cells protect the kidney from ischemia-reperfusion injury by microRNA-dependent reprogramming of resident renal cells. Kidney Int. 2012, 82, 412–427.
  49. Fuhrmann, G.; Chandrawati, R.; Parmar, P.A.; Keane, T.J.; Maynard, S.A.; Bertazzo, S.; Stevens, M.M. Engineering Extracellular Vesicles with the Tools of Enzyme Prodrug Therapy. Adv. Mater. 2018, 30, 1706616.
  50. Liu, W.; Yu, M.; Chen, F.; Wang, L.; Ye, C.; Chen, Q.; Zhu, Q.; Xie, D.; Shao, M.; Yang, L. A novel delivery nanobiotechnology: Engineered miR-181b exosomes improved osteointegration by regulating macrophage polarization. J Nanobiotechnol. 2021, 19, 269.
  51. Hippler, M.; Lemma, E.D.; Bertels, S.; Blasco, E.; Barner-Kowollik, C.; Wegener, M.; Bastmeyer, M. 3D Scaffolds to Study Basic Cell Biology. Adv. Mater. 2019, 31, 1808110.
  52. Patwardhan, S.; Mahadik, P.; Shetty, O.; Sen, S. ECM stiffness-tuned exosomes drive breast cancer motility through thrombospondin-1. Biomaterials 2021, 279, 121185.
  53. Palviainen, M.; Saari, H.; Karkkainen, O.; Pekkinen, J.; Auriola, S.; Yliperttula, M.; Puhka, M.; Hanhineva, K.; Siljander, P.R.M. Metabolic signature of extracellular vesicles depends on the cell culture conditions. J. Extracell. Vesicles 2019, 8, 1596669.
  54. Millan, C.; Prause, L.; Vallmajo-Martin, Q.; Hensky, N.; Eberli, D. Extracellular Vesicles from 3D Engineered Microtissues Harbor Disease-Related Cargo Absent in EVs from 2D Cultures. Adv. Healthc. Mater. 2022, 11, e2002067.
  55. van Balkom, B.W.M.; de Jong, O.G.; Smits, M.; Brummelman, J.; den Ouden, K.; de Bree, P.M.; van Eijndhoven, M.A.J.; Pegtel, D.M.; Stoorvogel, W.; Wuerdinger, T.; et al. Endothelial cells require miR-214 to secrete exosomes that suppress senescence and induce angiogenesis in human and mouse endothelial cells. Blood 2013, 121, 3997–4006.
  56. Bobryshev, Y.V.; Killingsworth, M.C.; Orekhov, A.N. Increased Shedding of Microvesicles from Intimal Smooth Muscle Cells in Athero-Prone Areas of the Human Aorta: Implications for Understanding of the Predisease Stage. Pathobiology 2013, 80, 24–31.
  57. Rogers, M.A.; Buffolo, F.; Schlotter, F.; Atkins, S.K.; Lee, L.H.; Halu, A.; Blaser, M.C.; Tsolaki, E.; Higashi, H.; Luther, K.; et al. Annexin A1-dependent tethering promotes extracellular vesicle aggregation revealed with single-extracellular vesicle analysis. Sci. Adv. 2020, 6, eabb1244.
  58. Zhu, J.M.; Liu, B.; Wang, Z.Y.; Di, W.; Ni, H.E.; Zhang, L.L.; Wang, Y. Exosomes from nicotine-stimulated macrophages accelerate atherosclerosis through miR-21-3p/PTEN-mediated VSMC migration and proliferation. Theranostics 2019, 9, 6901–6919.
More