@article{AshmwePosaRuehrnoessletal., author = {Ashmwe, Mohamed and Posa, Katja and R{\"u}hrn{\"o}ßl, Alexander and Heinzel, Johannes Christoph and Heimel, Patrick and Mock, Michael and Sch{\"a}dl, Barbara and Keibl, Claudia and Couillard-Despres, Sebastien and Redl, Heinz and Mittermayr, Rainer and Hercher, David}, title = {Effects of Extracorporeal Shockwave Therapy on Functional Recovery and Circulating miR-375 and miR-382-5p after Subacute and Chronic Spinal Cord Contusion Injury in Rats}, series = {Biomedicines}, volume = {2022}, journal = {Biomedicines}, number = {10(7)}, doi = {https://doi.org/10.3390/biomedicines10071630}, pages = {1630}, abstract = {Extracorporeal shockwave therapy (ESWT) can stimulate processes to promote regeneration, including cell proliferation and modulation of inflammation. Specific miRNA expression panels have been established to define correlations with regulatory targets within these pathways. This study aims to investigate the influence of low-energy ESWT-applied within the subacute and chronic phase of SCI (spinal cord injury) on recovery in a rat spinal cord contusion model. Outcomes were evaluated by gait analysis, µCT and histological analysis of spinal cords. A panel of serum-derived miRNAs after SCI and after ESWT was investigated to identify injury-, regeneration- and treatment-associated expression patterns. Rats receiving ESWT showed significant improvement in motor function in both a subacute and a chronic experimental setting. This effect was not reflected in changes in morphology, µCT-parameters or histological markers after ESWT. Expression analysis of various miRNAs, however, revealed changes after SCI and ESWT, with increased miR-375, indicating a neuroprotective effect, and decreased miR-382-5p potentially improving neuroplasticity via its regulatory involvement with BDNF. We were able to demonstrate a functional improvement of ESWT-treated animals after SCI in a subacute and chronic setting. Furthermore, the identification of miR-375 and miR-382-5p could potentially provide new targets for therapeutic intervention in future studies.}, subject = {Tissue Engineering}, language = {en} } @article{RomanelliBielerHeimeletal., author = {Romanelli, Pasquale and Bieler, Lara and Heimel, Patrick and Škokić, Siniša and Jakubecova, Dominika and Kreutzer, Christina and Zaunmair, Pia and Smolčić, Tomislav and Benedetti, Bruno and Rohde, Eva and Gimona, Mario and Hercher, David and Dobrivojević Radmilović, Marina and Couillard-Despres, Sebastien}, title = {Enhancing Functional Recovery Through Intralesional Application of Extracellular Vesicles in a Rat Model of Traumatic Spinal Cord Injury}, series = {Front Cell Neurosci}, volume = {15}, journal = {Front Cell Neurosci}, doi = {10.3389/fncel.2021.795008}, abstract = {Local inflammation plays a pivotal role in the process of secondary damage after spinal cord injury. We recently reported that acute intravenous application of extracellular vesicles (EVs) secreted by human umbilical cord mesenchymal stromal cells dampens the induction of inflammatory processes following traumatic spinal cord injury. However, systemic application of EVs is associated with delayed delivery to the site of injury and the necessity for high doses to reach therapeutic levels locally. To resolve these two constraints, we injected EVs directly at the lesion site acutely after spinal cord injury. We report here that intralesional application of EVs resulted in a more robust improvement of motor recovery, assessed with the BBB score and sub-score, as compared to the intravenous delivery. Moreover, the intralesional application was more potent in reducing inflammation and scarring after spinal cord injury than intravenous administration. Hence, the development of EV-based therapy for spinal cord injury should aim at an early application of vesicles close to the lesion.}, subject = {exosomes}, language = {en} }