@article{mei_li_wang_zhu_huang_hu_popowski_cheng_2023, title={An inhaled bioadhesive hydrogel to shield non-human primates from SARS-CoV-2 infection}, volume={2}, ISSN={1476-1122 1476-4660}, url={http://dx.doi.org/10.1038/s41563-023-01475-7}, DOI={10.1038/s41563-023-01475-7}, abstractNote={The surge of fast-spreading SARS-CoV-2 mutated variants highlights the need for fast, broad-spectrum strategies to counteract viral infections. In this work, we report a physical barrier against SARS-CoV-2 infection based on an inhalable bioadhesive hydrogel, named spherical hydrogel inhalation for enhanced lung defence (SHIELD). Conveniently delivered via a dry powder inhaler, SHIELD particles form a dense hydrogel network that coats the airway, enhancing the diffusional barrier properties and restricting virus penetration. SHIELD's protective effect is first demonstrated in mice against two SARS-CoV-2 pseudo-viruses with different mutated spike proteins. Strikingly, in African green monkeys, a single SHIELD inhalation provides protection for up to 8 hours, efficiently reducing infection by the SARS-CoV-2 WA1 and B.1.617.2 (Delta) variants. Notably, SHIELD is made with food-grade materials and does not affect normal respiratory functions. This approach could offer additional protection to the population against SARS-CoV-2 and other respiratory pathogens.}, journal={Nature Materials}, publisher={Springer Science and Business Media LLC}, author={Mei, Xuan and Li, Junlang and Wang, Zhenzhen and Zhu, Dashuai and Huang, Ke and Hu, Shiqi and Popowski, Kristen D. and Cheng, Ke}, year={2023}, month={Feb} } @article{wang_hu_zhu_li_cheng_liu_2023, title={Comparison of extruded cell nanovesicles and exosomes in their molecular cargos and regenerative potentials}, volume={16}, ISSN={["1998-0000"]}, DOI={10.1007/s12274-023-5374-3}, abstractNote={Extracellular vesicles (EVs) generated from mesenchymal stem cells (MSCs) play an essential role in modulating cell—cell communication and tissue regeneration. The clinical translation of EVs is constrained by the poor yield of EVs. Extrusion has recently become an effective technique for producing a large scale of nanovesicles (NVs). In this study, we systematically compared MSC NVs (from extrusion) and EVs (from natural secretion). Proteomics and RNA sequencing data revealed that NVs resemble MSCs more closely than EVs. Additionally, microRNAs in NVs are related to cardiac repair, fibrosis repression, and angiogenesis. Lastly, intravenous delivery of MSC NVs improved heart repair and cardiac function in a mouse model of myocardial infarction.}, number={5}, journal={NANO RESEARCH}, author={Wang, Xianyun and Hu, Shiqi and Zhu, Dashuai and Li, Junlang and Cheng, Ke and Liu, Gang}, year={2023}, month={May}, pages={7248–7259} } @article{zhu_liu_huang_li_mei_li_cheng_2023, title={Intrapericardial long non-coding RNA–Tcf21 antisense RNA inducing demethylation administration promotes cardiac repair}, volume={44}, ISSN={0195-668X 1522-9645}, url={http://dx.doi.org/10.1093/eurheartj/ehad114}, DOI={10.1093/eurheartj/ehad114}, abstractNote={Abstract Aims Epicardium and epicardium-derived cells are critical players in myocardial fibrosis. Mesenchymal stem cell–derived extracellular vesicles (EVs) have been studied for cardiac repair to improve cardiac remodelling, but the actual mechanisms remain elusive. The aim of this study is to investigate the mechanisms of EV therapy for improving cardiac remodelling and develop a promising treatment addressing myocardial fibrosis. Methods and results Extracellular vesicles were intrapericardially injected for mice myocardial infarction treatment. RNA-seq, in vitro gain- and loss-of-function experiments, and in vivo studies were performed to identify targets that can be used for myocardial fibrosis treatment. Afterward, a lipid nanoparticle–based long non-coding RNA (lncRNA) therapy was prepared for mouse and porcine models of myocardial infarction treatment. Intrapericardial injection of EVs improved adverse myocardial remodelling in mouse models of myocardial infarction. Mechanistically, Tcf21 was identified as a potential target to improve cardiac remodelling. Loss of Tcf21 function in epicardium-derived cells caused increased myofibroblast differentiation, whereas forced Tcf21 overexpression suppressed transforming growth factor-β signalling and myofibroblast differentiation. LncRNA–Tcf21 antisense RNA inducing demethylation (TARID) that enriched in EVs was identified to up-regulate Tcf21 expression. Formulated lncRNA–TARID-laden lipid nanoparticles up-regulated Tcf21 expression in epicardium-derived cells and improved cardiac function and histology in mouse and porcine models of myocardial infarction. Conclusion This study identified Tcf21 as a critical target for improving cardiac fibrosis. Up-regulating Tcf21 by using lncRNA–TARID-laden lipid nanoparticles could be a promising way to treat myocardial fibrosis. This study established novel mechanisms underlying EV therapy for improving adverse remodelling and proposed a lncRNA therapy for cardiac fibrosis.}, number={19}, journal={European Heart Journal}, publisher={Oxford University Press (OUP)}, author={Zhu, Dashuai and Liu, Shuo and Huang, Ke and Li, Junlang and Mei, Xuan and Li, Zhenhua and Cheng, Ke}, year={2023}, month={Mar}, pages={1748–1760} } @article{popowski_moatti_scull_silkstone_lutz_lópez de juan abad_george_belcher_zhu_mei_et al._2022, title={Inhalable dry powder mRNA vaccines based on extracellular vesicles}, volume={5}, ISSN={2590-2385}, url={http://dx.doi.org/10.1016/j.matt.2022.06.012}, DOI={10.1016/j.matt.2022.06.012}, abstractNote={•Lung extracellular vesicles (Lung-Exos) can package mRNA and protein drugs•Lung-Exos are deliverable through nebulization and dry powder inhalation•Dry powder Lung-Exos are room-temperature stable up to 28 days•Drug-loaded Lung-Exos can serve as an inhalable vaccine to illicit immune responses Research in extracellular vesicles (EVs) is important to the field of translational medicine to develop therapeutics that are limited by poor cellular targeting and efficacy. The biological composition of EVs can be exploited as drug-delivery vehicles that may be engineered for cellular targeting or eliciting specific immune responses through their functions in membrane trafficking and cellular signaling. With the molecular composition of EVs varying depending on their parent-cell origin, the derivation of EVs can further refine nanomedicine by utilizing nanoparticles that are recognized by specific cellular microenvironments. EVs are found in almost all biological fluids, opening the application of EVs as tailored drug-delivery vesicles to a wide range of diseases. Respiratory diseases are a global burden, with millions of deaths attributed to pulmonary illnesses and dysfunctions. Therapeutics have been developed, but they present major limitations regarding pulmonary bioavailability and product stability. To circumvent such limitations, we developed room-temperature-stable inhalable lung-derived extracellular vesicles or exosomes (Lung-Exos) as mRNA and protein drug carriers. Compared with standard synthetic nanoparticle liposomes (Lipos), Lung-Exos exhibited superior distribution to the bronchioles and parenchyma and are deliverable to the lungs of rodents and nonhuman primates (NHPs) by dry powder inhalation. In a vaccine application, severe acute respiratory coronavirus 2 (SARS-CoV-2) spike (S) protein encoding mRNA-loaded Lung-Exos (S-Exos) elicited greater immunoglobulin G (IgG) and secretory IgA (SIgA) responses than its loaded liposome (S-Lipo) counterpart. Importantly, S-Exos remained functional at room-temperature storage for one month. Our results suggest that extracellular vesicles can serve as an inhaled mRNA drug-delivery system that is superior to synthetic liposomes. Respiratory diseases are a global burden, with millions of deaths attributed to pulmonary illnesses and dysfunctions. Therapeutics have been developed, but they present major limitations regarding pulmonary bioavailability and product stability. To circumvent such limitations, we developed room-temperature-stable inhalable lung-derived extracellular vesicles or exosomes (Lung-Exos) as mRNA and protein drug carriers. Compared with standard synthetic nanoparticle liposomes (Lipos), Lung-Exos exhibited superior distribution to the bronchioles and parenchyma and are deliverable to the lungs of rodents and nonhuman primates (NHPs) by dry powder inhalation. In a vaccine application, severe acute respiratory coronavirus 2 (SARS-CoV-2) spike (S) protein encoding mRNA-loaded Lung-Exos (S-Exos) elicited greater immunoglobulin G (IgG) and secretory IgA (SIgA) responses than its loaded liposome (S-Lipo) counterpart. Importantly, S-Exos remained functional at room-temperature storage for one month. Our results suggest that extracellular vesicles can serve as an inhaled mRNA drug-delivery system that is superior to synthetic liposomes. IntroductionRespiratory diseases are among the leading causes of morbidity and mortality worldwide,1Wisnivesky J. De-Torres J.P. The global burden of pulmonary diseases: most prevalent problems and opportunities for improvement.Ann. Global Health. 2019; 85https://doi.org/10.5334/aogh.2411Crossref Scopus (17) Google Scholar with coronavirus disease 2019 (COVID-19)2Wang C. Horby P.W. Hayden F.G. Gao G.F. A novel coronavirus outbreak of global health concern.Lancet. 2020; 395: 470-473https://doi.org/10.1016/S0140-6736(20)30185-9Abstract Full Text Full Text PDF PubMed Scopus (3967) Google Scholar remaining prevalent in the ongoing pandemic. A wide range of therapeutics have been developed and repurposed to treat respiratory diseases, including small-molecule drugs,3Li D. Hu J. Li D. Yang W. Yin S.-F. Qiu R. Reviews on biological activity, clinical trial and synthesis progress of small molecules for the treatment of COVID-19.Top. Curr. Chem. 2021; 379: 4https://doi.org/10.1007/s41061-020-00318-2Crossref Scopus (11) Google Scholar,4Panoutsopoulos A.A. Known drugs and small molecules in the battle for COVID-19 treatment.Genes. Dis. 2020; 7: 528-534https://doi.org/10.1016/j.gendis.2020.06.007Crossref PubMed Scopus (11) Google Scholar stem cells5Cruz F.F. Rocco P.R.M. The potential of mesenchymal stem cell therapy for chronic lung disease.Expet. Rev. Respir. Med. 2020; 14: 31-39https://doi.org/10.1080/17476348.2020.1679628Crossref PubMed Scopus (69) Google Scholar, 6Ji H.-L. Liu C. Zhao R.-Z. Stem cell therapy for COVID-19 and other respiratory diseases: global trends of clinical trials.World J. Stem Cell. 2020; 12: 471-480https://doi.org/10.4252/wjsc.v12.i6.471Crossref PubMed Google Scholar, 7Henry E. Cores J. Hensley M.T. Anthony S. Vandergriff A. de Andrade J.B.M. Allen T. Caranasos T.G. Lobo L.J. Cheng K. Adult lung spheroid cells contain progenitor cells and mediate regeneration in rodents with bleomycin-induced pulmonary fibrosis.Stem Cells Transl. Med. 2015; 4: 1265-1274https://doi.org/10.5966/sctm.2015-0062Crossref PubMed Scopus (36) Google Scholar, 8Cores J. Hensley M.T. Kinlaw K. Rikard S.M. Dinh P.-U. Paudel D. Tang J. Vandergriff A.C. Allen T.A. Li Y. et al.Safety and efficacy of allogeneic lung spheroid cells in a mismatched rat model of pulmonary fibrosis.Stem Cells Transl. Med. 2017; 6: 1905-1916https://doi.org/10.1002/sctm.16-0374Crossref PubMed Scopus (18) Google Scholar, 9Cores J. Dinh P.U.C. Hensley T. Adler K.B. Lobo L.J. Cheng K. A pre- investigational new drug study of lung spheroid cell therapy for treating pulmonary fibrosis.Stem Cells Transl. Med. 2020; 9: 786-798https://doi.org/10.1002/sctm.19-0167Crossref PubMed Scopus (5) Google Scholar and their derivatives,10Sengupta V. Sengupta S. Lazo A. Woods P. Nolan A. Bremer N. Exosomes derived from bone marrow mesenchymal stem cells as treatment for severe COVID-19.Stem Cell. Dev. 2020; 29: 747-754https://doi.org/10.1089/scd.2020.0080Crossref PubMed Scopus (294) Google Scholar, 11Maremanda K.P. Sundar I.K. Rahman I. Protective role of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in cigarette smoke-induced mitochondrial dysfunction in mice.Toxicol. Appl. Pharmacol. 2019; 385: 114788https://doi.org/10.1016/j.taap.2019.114788Crossref PubMed Scopus (41) Google Scholar, 12Mansouri N. Willis G.R. Fernandez-Gonzalez A. Reis M. Nassiri S. Mitsialis S.A. Kourembanas S. Mesenchymal stromal cell exosomes prevent and revert experimental pulmonary fibrosis through modulation of monocyte phenotypes.JCI Insight. 2019; 4: 128060https://doi.org/10.1172/jci.insight.128060Crossref PubMed Scopus (99) Google Scholar, 13Dinh P.-U.C. Paudel D. Brochu H. Popowski K.D. Gracieux M.C. Cores J. Huang K. Hensley M.T. Harrell E. Vandergriff A.C. et al.Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis.Nat. Commun. 2020; 11: 1064https://doi.org/10.1038/s41467-020-14344-7Crossref PubMed Scopus (127) Google Scholar and, notably, messenger RNA (mRNA)14Zhang N.-N. Li X.-F. Deng Y.-Q. Zhao H. Huang Y.-J. Yang G. Huang W.-J. Gao P. Zhou C. Zhang R.-R. et al.A thermostable mRNA vaccine against COVID-19.Cell. 2020; 182: 1271-1283.e16https://doi.org/10.1016/j.cell.2020.07.024Abstract Full Text Full Text PDF PubMed Scopus (284) Google Scholar, 15Corbett K.S. Edwards D.K. Leist S.R. Abiona O.M. Boyoglu-Barnum S. Gillespie R.A. Himansu S. Schäfer A. Ziwawo C.T. DiPiazza A.T. et al.SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness.Nature. 2020; 586: 567-571https://doi.org/10.1038/s41586-020-2622-0Crossref PubMed Scopus (572) Google Scholar, 16Polack F.P. Thomas S.J. Kitchin N. Absalon J. Gurtman A. Lockhart S. Perez J.L. Pérez Marc G. Moreira E.D. Zerbini C. et al.Safety and efficacy of the BNT162b2 mRNA covid-19 vaccine.N. Engl. J. Med. 2020; 383: 2603-2615https://doi.org/10.1056/NEJMoa2034577Crossref PubMed Scopus (6139) Google Scholar, 17Walsh E.E. Frenck R.W.J. Falsey A.R. Kitchin N. Absalon J. Gurtman A. Lockhart S. Neuzil K. Mulligan M.J. Bailey R. et al.Safety and immunogenicity of two RNA-based covid-19 vaccine candidates.N. Engl. J. Med. 2020; 383: 2439-2450https://doi.org/10.1056/NEJMoa2027906Crossref PubMed Scopus (1259) Google Scholar and protein vaccines.18Wilkinson T.M.A. Schembri S. Brightling C. Bakerly N.D. Lewis K. MacNee W. Rombo L. Hedner J. Allen M. Walker P.P. et al.Non-typeable Haemophilus influenzae protein vaccine in adults with COPD: a phase 2 clinical trial.Vaccine. 2019; 37: 6102-6111https://doi.org/10.1016/j.vaccine.2019.07.100Crossref PubMed Scopus (29) Google Scholar,19Yang J. Wang W. Chen Z. Lu S. Yang F. Bi Z. Bao L. Mo F. Li X. Huang Y. et al.A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunity.Nature. 2020; 586: 572-577https://doi.org/10.1038/s41586-020-2599-8Crossref PubMed Scopus (352) Google Scholar Currently, three intramuscular (IM) vaccines (Pfizer-BioNTech, Moderna, and Janssen) have been authorized for COVID-19 by the US Food and Drug Administration. However, formulating drugs and vaccines for inhaled delivery may improve their efficacy and bioavailability in the lung as well as improve patient compliance.Although inhalation delivery is attractive due to its local, noninvasive, and highly absorptive properties, drug formulation with optimized physiochemical parameters remains the key obstacle.20Beija M. Salvayre R. Lauth-de Viguerie N. Marty J.-D. Colloidal systems for drug delivery: from design to therapy.Trends Biotechnol. 2012; 30: 485-496https://doi.org/10.1016/j.tibtech.2012.04.008Abstract Full Text Full Text PDF PubMed Scopus (130) Google Scholar A possible solution is colloidal drug-delivery systems, which help overcome poor drug solubility and hydrophobicity by providing protection through nanoparticle encapsulation.21Sharma J. Kalra S. Sharma A. Rani S. Colloidal drug carriers.Internet J. Fam. Pract. 2009; 9Google Scholar Plus, nanoparticles retained in the lung prolong drug release in the highly vascularized pulmonary and bronchial circulations.22Paranjpe M. Müller-Goymann C. Nanoparticle-mediated pulmonary drug delivery: a review.Int. J. Mol. Sci. 2014; 15: 5852-5873https://doi.org/10.3390/ijms15045852Crossref PubMed Scopus (271) Google Scholar Lipid nanoparticles (LNPs) have been successful as drug-delivery vesicles for vaccines, most notably in the liposome-encapsulated COVID-19 mRNA IM vaccines.23Nanomedicine and the COVID-19 vaccines.Nat. Nanotechnol. 2020; 15: 963https://doi.org/10.1038/s41565-020-00820-0Crossref PubMed Scopus (76) Google Scholar However, major limitations such as deep-freezing storage and healthcare professional administration have led to the expansion of the LNP platform to alternative delivery routes such as self-administered inhalation. In addition, the lung has sophisticated pulmonary defense mechanisms and surfactants that protect it against inhaled particulates and microbes.22Paranjpe M. Müller-Goymann C. Nanoparticle-mediated pulmonary drug delivery: a review.Int. J. Mol. Sci. 2014; 15: 5852-5873https://doi.org/10.3390/ijms15045852Crossref PubMed Scopus (271) Google Scholar,24Radiom M. Sarkis M. Brookes O. Oikonomou E.K. Baeza-Squiban A. Berret J.-F. Pulmonary surfactant inhibition of nanoparticle uptake by alveolar epithelial cells.Sci. Rep. 2020; 10: 19436https://doi.org/10.1038/s41598-020-76332-7Crossref PubMed Scopus (10) Google Scholar Therefore, the formulation of nanoparticle drug-delivery systems must be optimized to overcome these inherent obstacles.Exosomes, biologically derived nanoparticles, may be naturally equipped to withstand pulmonary conditions. They are nanosized extracellular vesicles secreted by numerous cell types and found in almost all biological fluids.25Théry C. Zitvogel L. Amigorena S. Exosomes: composition, biogenesis and function.Nat. Rev. Immunol. 2002; 2: 569-579https://doi.org/10.1038/nri855Crossref PubMed Scopus (3631) Google Scholar Initially regarded as cellular debris, exosomes are now understood to have potent roles in autocrine and paracrine signaling.26Maas S.L.N. Breakefield X.O. Weaver A.M. Extracellular vesicles: unique intercellular delivery vehicles.Trends Cell Biol. 2017; 27: 172-188https://doi.org/10.1016/j.tcb.2016.11.003Abstract Full Text Full Text PDF PubMed Scopus (756) Google Scholar, 27Sung B.H. von Lersner A. Guerrero J. Krystofiak E.S. Inman D. Pelletier R. Zijlstra A. Ponik S.M. Weaver A.M. A live cell reporter of exosome secretion and uptake reveals pathfinding behavior of migrating cells.Nat. Commun. 2020; 11: 2092https://doi.org/10.1038/s41467-020-15747-2Crossref PubMed Scopus (71) Google Scholar, 28van Niel G. D’Angelo G. Raposo G. Shedding light on the cell biology of extracellular vesicles.Nat. Rev. Mol. Cell Biol. 2018; 19: 213-228https://doi.org/10.1038/nrm.2017.125Crossref PubMed Scopus (3018) Google Scholar Originating from the endosomal system and shedding from the plasma membrane, exosomes contain various cocktails of RNA, protein, and lipid cargo with unique parent-cell signatures.29Kao C.-Y. Papoutsakis E.T. Extracellular vesicles: exosomes, microparticles, their parts, and their targets to enable their biomanufacturing and clinical applications.Curr. Opin. Biotechnol. 2019; 60: 89-98https://doi.org/10.1016/J.COPBIO.2019.01.005Crossref PubMed Scopus (0) Google Scholar,30Popowski K. Lutz H. Hu S. George A. Dinh P.-U. Cheng K. Exosome therapeutics for lung regenerative medicine.J. Extracell. Vesicles. 2020; 9: 1785161https://doi.org/10.1080/20013078.2020.1785161Crossref PubMed Scopus (36) Google Scholar Lung-derived exosomes can be utilized as sophisticated drug-delivery systems that offer cargo components and membrane features tailored to the lung microenvironment.20Beija M. Salvayre R. Lauth-de Viguerie N. Marty J.-D. Colloidal systems for drug delivery: from design to therapy.Trends Biotechnol. 2012; 30: 485-496https://doi.org/10.1016/j.tibtech.2012.04.008Abstract Full Text Full Text PDF PubMed Scopus (130) Google Scholar,31Popowski K.D. Dinh P.-U.C. George A. Lutz H. Cheng K. Exosome therapeutics for COVID-19 and respiratory viruses.View. 2021; : 20200186https://doi.org/10.1002/VIW.20200186Crossref Google Scholar Our group has demonstrated the lung regenerative abilities of human lung spheroid cells (LSCs)8Cores J. Hensley M.T. Kinlaw K. Rikard S.M. Dinh P.-U. Paudel D. Tang J. Vandergriff A.C. Allen T.A. Li Y. et al.Safety and efficacy of allogeneic lung spheroid cells in a mismatched rat model of pulmonary fibrosis.Stem Cells Transl. Med. 2017; 6: 1905-1916https://doi.org/10.1002/sctm.16-0374Crossref PubMed Scopus (18) Google Scholar,9Cores J. Dinh P.U.C. Hensley T. Adler K.B. Lobo L.J. Cheng K. A pre- investigational new drug study of lung spheroid cell therapy for treating pulmonary fibrosis.Stem Cells Transl. Med. 2020; 9: 786-798https://doi.org/10.1002/sctm.19-0167Crossref PubMed Scopus (5) Google Scholar in rodent models of idiopathic pulmonary fibrosis (IPF), and their safety and efficacy are being tested in a human clinical trial (HALT-IPF, ClinicialTrials.gov: NCT04262167). LSCs and their secreted exosomes (lung-derived extracellular vesicles [Lung-Exos]) have regenerative abilities in IPF models13Dinh P.-U.C. Paudel D. Brochu H. Popowski K.D. Gracieux M.C. Cores J. Huang K. Hensley M.T. Harrell E. Vandergriff A.C. et al.Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis.Nat. Commun. 2020; 11: 1064https://doi.org/10.1038/s41467-020-14344-7Crossref PubMed Scopus (127) Google Scholar and protective abilities against COVID-19 as decoys.32Li Z. Wang Z. Dinh P.-U.C. Zhu D. Popowski K.D. Lutz H. Hu S. Lewis M.G. Cook A. Andersen H. et al.Cell-mimicking nanodecoys neutralize SARS-CoV-2 and mitigate lung injury in a non-human primate model of COVID-19.Nat. Nanotechnol. 2021; 16: 942-951https://doi.org/10.1038/s41565-021-00923-2Crossref PubMed Scopus (43) Google Scholar In both disease models, Lung-Exos maintained therapeutic efficacy through jet-nebulization administration, demonstrating the ability of Lung-Exos to function as an inhalable drug-delivery and vaccine vehicle. Additionally, exosomes can be synthetically supplemented to enhance cellular targeting and therapeutic efficacy.33Hutcheson J.D. Aikawa E. Extracellular vesicles in cardiovascular homeostasis and disease.Curr. Opin. Cardiol. 2018; 33: 290-297https://doi.org/10.1097/HCO.0000000000000510Crossref PubMed Scopus (29) Google Scholar, 34Li S.p. Lin Z.x. Jiang X. Yu X.y. Yu X. Exosomal cargo-loading and synthetic exosome-mimics as potential therapeutic tools.Acta Pharmacol. Sin. 2018; 39: 542-551https://doi.org/10.1038/aps.2017.178Crossref PubMed Scopus (170) Google Scholar, 35Lutz H. Popowski K.D. Dinh P.-U.C. Cheng K. Advanced nanobiomedical approaches to combat coronavirus disease of 2019.Adv. Nanobiomed Res. 2021; : 2000063https://doi.org/10.1002/anbr.202000063Crossref PubMed Google Scholar For example, anti-inflammatory peptides36Kim G.Y. Lee Y. Ha J. Han S. Lee M. Engineering exosomes for pulmonary delivery of peptides and drugs to inflammatory lung cells by inhalation.J. Contr. Release. 2021; 330: 684-695https://doi.org/10.1016/j.jconrel.2020.12.053Crossref PubMed Scopus (21) Google Scholar can be linked onto the exosomal membrane, engineering an immune cell targeting nanomedicine. The combination of vesicle derivation and supplementation allows for a customizable nanoparticle delivery platform that can be utilized across many lung diseases.Although exosomes as inhaled therapeutics and RNA delivery vehicles have been demonstrated,13Dinh P.-U.C. Paudel D. Brochu H. Popowski K.D. Gracieux M.C. Cores J. Huang K. Hensley M.T. Harrell E. Vandergriff A.C. et al.Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis.Nat. Commun. 2020; 11: 1064https://doi.org/10.1038/s41467-020-14344-7Crossref PubMed Scopus (127) Google Scholar,37Zhang D. Lee H. Wang X. Rai A. Groot M. Jin Y. Exosome-Mediated small RNA delivery: a novel therapeutic approach for inflammatory lung responses.Mol. Ther. 2018; 26: 2119-2130https://doi.org/10.1016/j.ymthe.2018.06.007Abstract Full Text Full Text PDF PubMed Scopus (86) Google Scholar the distribution and retention of exosome particles in the lung have yet to be determined. Drug effectiveness depends on proper deposition of particles within the respirable fraction, requiring optimized nanoparticle formulation. In this study, we sought to elucidate the biodistribution of lung-derived exosomes upon nebulization, baselined to liposomes (Lipos) as a commercial standard. Furthermore, to provide a room-temperature-stable product, we formulated exosomes as a lyophilized dry powder to investigate their stability and inhaled biodistribution in the lung of both the mouse and African green monkey (AGM). The parent-cell signature of Lung-Exos may suggest that they are naturally optimized for the distribution and retention within the lung, which may allow them to bypass pulmonary clearance more efficiently than Lipos or exosomes derived from other cell types. Through this enhanced pulmonary bioavailability, we hypothesize that lung-derived exosomes elicit greater therapeutic responses for pulmonary diseases and serve as a customizable drug-delivery vehicle for room-temperature-stable inhaled mRNA therapeutics.ResultsExosome distribution in the bronchioles and parenchyma are superior to that of synthetic nanoparticlesRed fluorescent protein (RFP)-labeled lung-derived exosomes (RFP-Exos) and Lipos (RFP-Lipos) were fabricated to generate trackable nanoparticles for biodistribution analysis in the murine lung after inhalation treatment through three-dimensional (3D) imaging (Figure 1A ). The nanoparticles were characterized by transmission electron microscopy (TEM), confirming that the isolation of exosomes and Lipos did not disrupt vesicular membrane integrity (Figure 1B). RFP loading was verified by immunoblotting (Figure 1C). When co-cultured with lung parenchymal cells, RFP-Exo had a 6.7-fold increase in cellular uptake and RFP protein expression compared with cells cultured with RFP-Lipo (Figures 1D and 1E). Next, the biodistribution of nanoparticles in vivo were evaluated through light-sheet fluorescence microscopy (LSFM) (Figure 1F). Healthy mice received a single dose of RFP-Exos or RFP-Lipos via nebulization and were sacrificed after 24 h. LSFM imaging confirmed nanoparticle delivery to the conducting airways and the deep lung, with an accumulation of RFP-Exos in the upper pulmonary regions (Videos S1 and S2). Quantification of nanoparticle delivery to the whole lung demonstrated a 3.7-fold improvement in RFP-Exo retention and uptake compared with RFP-Lipo (Figure 1G). Segmentation of the lung into bronchial and parenchymal regions revealed 2.9- and 3.8-fold improvements in RFP-Exo retention and uptake, respectively, compared with RFP-Lipo (Figure 1H). Flow cytometry analysis in lung parenchymal cells (Figure 1I) and in the murine lung following nebulization (Figure 1J) confirmed greater cellular uptake of RFP-Exos than RFP-Lipos. The drug-loading capabilities of lung-derived exosomes (Lung-Exos and Lipos were expanded by loading GFP-encoding mRNA to evaluate nanoparticle mRNA uptake. Lung parenchymal cells that received GFP-Exos demonstrated more rapid internalization of exosomal mRNA than liposomal mRNA (Figure S1). These data confirm that our nanoparticle labeling system maintains nanoparticle integrity while delivering functional and translatable cargo after jet nebulization. In vitro and in vivo analyses suggest superior retention and cellular uptake of exosomes over Lipos in the lung. The native lung signature of lung-derived exosomes may enhance pulmonary bioavailability, resulting in an optimized nanoparticle vesicle for drug delivery for respiratory diseases.eyJraWQiOiI4ZjUxYWNhY2IzYjhiNjNlNzFlYmIzYWFmYTU5NmZmYyIsImFsZyI6IlJTMjU2In0.eyJzdWIiOiIyZDE5ZTI0ODk4NDRkYTFhYjJlNGUzMWEwMGE4OTEyYyIsImtpZCI6IjhmNTFhY2FjYjNiOGI2M2U3MWViYjNhYWZhNTk2ZmZjIiwiZXhwIjoxNjY5OTY2NjUwfQ.ZP_2__aVFlfG_3XKl06osGFI0GeEXb27ElTxZ-KGGM8IcSO676pqZKF6h65jz0cnnYuojIwvoPOnatMAd9qqR65EtMzNhGLYU-TdT2hEgjFIUL9AatnXvIyrTXEJt4Orap9iWn8eBBxr63jGbLdLvzqnA2A05saBmcwbPhbN3bVzqdWYJQCLOpzv6z8Yi-0i3xtJfwEmLHgdsvSLgQt8vnGKzY4IUA9jBXCdfho1Cp5PVg3eCXd0p9bQo-Df_q9s-Ghg1957V4C1OlRDoKdNdXo62FKjOcdLZPrCnVgzDNqmk_IPHTSkzr3rdFCG41UJVBXHBYkJMpXnhOvsDyhoXw Download .mp4 (30.4 MB) Help with .mp4 files Video S1. Biodistribution of nebulized RFP-Exos in mouse lungsLSFM imaging and 3D rendering and animation by Imaris confirms labeled exosome distribution throughout the lung. Tissue autofluorescence allows for morphological segmentation of bronchioles and parenchyma to quantify exosome distribution.eyJraWQiOiI4ZjUxYWNhY2IzYjhiNjNlNzFlYmIzYWFmYTU5NmZmYyIsImFsZyI6IlJTMjU2In0.eyJzdWIiOiJiNTYzZTFmZDNiYjJiOTQzMWQyMDAwODQzYzA2MDcxNCIsImtpZCI6IjhmNTFhY2FjYjNiOGI2M2U3MWViYjNhYWZhNTk2ZmZjIiwiZXhwIjoxNjY5OTY2NjUwfQ.rrwycH66nt21uB8nXcVXQaa02xcRy_2vJlbQsIigymt2QlJ24qCloQxenvyl4rqzmA9WIpCs6ZdvZ5yRY5FWlu-j2SfO4Sl8lcL2Xz3jrbu8PU-Ge_avejxOWdeaYEEEGIbmAWmXlVHWH2p6_LlFpfPB-j4E-KTapjf5CHzT4rGRom3mXroj6OiJkAkRl3ylnmxTdNaQDUbtDr2dJvIYzLCX_8TUNx9a7L4Rz-65ZWZD_ZXLADu_bgTFdnNOGodGmEY-9NZhPBqraOljjTPxzbe4TJJOttzGsmAySGpVDMWGFGcStxkuif_tgwujLF5wcLU1o-KQtk8eosKrcuRuxA Download .mp4 (38.39 MB) Help with .mp4 files Video S2. Biodistribution of nebulized RFP-Lipos in mouse lungsLSFM imaging and 3D rendering and animation by Imaris confirms labeled liposome distribution throughout the lung. Tissue autofluorescence allows for morphological segmentation of bronchioles and parenchyma to quantify liposome distribution.Lung-derived exosomes efficiently penetrate mucusDelivery of inhaled therapeutics must penetrate the lung’s protective mucus lining to provide pulmonary bioavailability. Lung-Exos were compared against human embryonic kidney (HEK)-derived exosomes (HEK-Exos) and Lipos to determine if nanoparticle derivation affected mucus penetrance. To test this, we used a model of the human airway at the air-liquid interface (Figure S2A), with human mucus-secreting bronchial epithelial cells lining the transwell membrane and human lung parenchymal cells lining the well (Figure S2B). Immunostaining confirmed the mucus lining in the transwell membrane and delivery of DiD-labeled nanoparticles (Figure S2C). Quantification of nanoparticle penetrance into the wells revealed the greatest uptake of Lung-Exos (Figure S2D), with the highest percentage of cellular uptake by lung parenchymal cells (Figure S2E) by 24 h. Likewise, Lung-Exos had the least entrapment by the mucus-lined membrane (Figure S2F) and the lowest percentage of cellular uptake by bronchial epithelial cells (Figure S2G). These data confirm mucus penetrance of the nanoparticles and suggest that Lung-Exos can most efficiently evade mucoadhesion, overcoming the lung’s natural defense mechanism and allowing for greater parenchymal bioavailability.Lung-derived exosomes are room-temperature stable and distributable in dry powder formulation in the murine lungRoom-temperature formulation of therapeutics circumvents major limitations in traditional IM vaccine delivery: deep-freezing storage, healthcare professional administration, and reduced patient compliance. Therefore, we reformulated our liquid nanoparticle suspensions into dry, lyophilized powder for dry powder inhalation (DPI) administration. We verified the efficacy and stability of room-temperature lyophilized Lung-Exos up to 28 days in the murine lung (Figure 2A ). To verify dry powder nanoparticle stability and shelf life, lyophilized nanoparticle cargo leakage was tested by an enzyme-linked immunosorbent assay (ELISA), where nanoparticles had less than 2.4% of total pg/mL cargo leakage at day 28 of room-temperature storage (Figures 2B and S3). Next, the morphology of nanoparticles was evaluated across their fresh and lyophilized formulations, as well as lyophilized powder reconstituted in water (reconstituted), to mimic rehydration of dry powder by saliva and mucus. TEM (Figures 2E and S4) and atomic force microscopy (AFM) verified that reformulation and rehydration did not affect nanoparticle membrane integrity (Figures 2C and S5) but did affect size distributions through clumping (Figure S6–S8). Lyophilization increased nanoparticle height and diameter (Figure 2D) but remained as small respiratory droplets upon reconstitution. Across all formulations, the nanoparticle diameters are approximately 10-fold larger, which may be explained by tip dilation38Wong C. West P.E. Olson K.S. Mecartney M.L. Starostina N. Tip dilation and AFM capabilities in the characterization of nanoparticles.JOM. 2007; 59: 12-16https://doi.org/10.1007/s11837-007-0003-xCrossref Scopus (40) Google Scholar that reports larger lateral dimensions than 2D analysis such as through TEM.39Eaton P. Quaresma P. Soares C. Neves C. de Almeida M.P. Pereira E. West P. A direct comparison of experimental methods to measure dimensions of synthetic nanoparticles.Ultramicroscopy. 2017; 182: 179-190https://doi.org/10.1016/j.ultramic.2017.07.001Crossref PubMed Scopus (164) Google Scholar Cross-section measurement curves demonstrate a restoration of membrane “smoothness” in reconstituted nanoparticles, mimicking fresh formulation (Figure S9). Next, we delivered the lyophilized Lung-Exos via DPI, where ex vivo images (Figure 2F) of mouse lungs who received fresh (fresh lyophilized) and 28-day-old (28-day lyophilized) dry powder Lung-Exos had no significant difference in exosomal mRNA and protein distribution (Figure 2G). mRNA activity showed greater variability at it 28-day-old state, but protein activity remained more stable.Figure 2Stability and distribution of lung-derived exosomes in dry powder formulation in the murine lungShow full caption(A) Schematic of mRNA and protein-loaded lung-derived exosome lyophilization, encapsulation, rodent DPI administration, and ex vivo histology. Created with BioRender.com.(B) Heatmaps of RFP leakage from Lung-Exos, HEK-Exos, and Lipos detected by ELISA; n = 2 per group.(C) Representative AFM height (I), amplitude (II), and phase (III) images of Lung-Exos; scale bar: 50 nm.(D) Quantification of the height and diameter of Lung-Exos, HEK-Exos, and Lipos from AFM images; n = 9 per group; data are represented as mean ± standard deviation.(E) TEM images of Lung-Exos at frozen (Frozen) or room (Lyophilized) temperatures; scale bar: 50 nm.(F) Ex vivo images of mouse lungs that received fresh lyophilized (0 days) and 28-day-old lyophilized Lung-Exos via dry powder inhalation after 24 h.(G) Quantification of the integrated density of GFP and RFP fluorescence in ex vivo mouse lungs 24 h after fresh (Fresh-Lyos) and 28-day-old (28-Day Lyos) dry powder inhalation; n = 3 per group; data are represented as mean ± standard deviation.(H) Quantification of the integrated density of GFP and RFP fluorescence in ex vivo mouse lungs 24 h after nebulization and fresh (Fresh-Lyos) dry powder inhalation; n = 3 per group; data are represented as mean ± standard deviation.(I) Quantification of the integrated density of GFP and RFP fluorescence in ex vivo mouse lungs 24 h after nebulization and 28-day-old (28-Day Lyos) dry powder inhalation; n = 3 per group; data are represented as mean ± standard deviation.View Large}, number={9}, journal={Matter}, publisher={Elsevier BV}, author={Popowski, Kristen D. and Moatti, Adele and Scull, Grant and Silkstone, Dylan and Lutz, Halle and López de Juan Abad, Blanca and George, Arianna and Belcher, Elizabeth and Zhu, Dashuai and Mei, Xuan and et al.}, year={2022}, month={Sep}, pages={2960–2974} } @article{zhu_liu_huang_wang_hu_li_li_cheng_2022, title={Intrapericardial Exosome Therapy Dampens Cardiac Injury via Activating Foxo3}, volume={131}, ISSN={["1524-4571"]}, url={https://doi.org/10.1161/CIRCRESAHA.122.321384}, DOI={10.1161/CIRCRESAHA.122.321384}, abstractNote={Mesenchymal stem cell (MSC)-derived exosomes are well recognized immunomodulating agents for cardiac repair, while the detailed mechanisms remain elusive. The Pericardial drainage pathway provides the heart with immunosurveillance and establishes a simplified model for studying the mechanisms underlying the immunomodulating effects of therapeutic exosomes.Myocardial infarction (MI) models with and without pericardiectomy (corresponding to Tomy MI and NonTomy MI) were established to study the functions of pericardial drainage pathway in immune activation of cardiac-draining mediastinal lymph node (MLN). Using the NonTomy MI model, MSC exosomes or vehicle PBS was intrapericardially injected for MI treatment. Via cell sorting and RNA-seq (RNA-sequencing) analysis, the differentially expressed genes were acquired for integrated pathway analysis to identify responsible mechanisms. Further, through functional knockdown/inhibition studies, application of cytokines and neutralizing antibodies, western blot, flow cytometry, and cytokine array, the molecular mechanisms were studied. In addition, the therapeutic efficacy of intrapericardially injected exosomes for MI treatment was evaluated through functional and histological analyses.We show that the pericardial draining pathway promoted immune activation in the MLN following MI. Intrapericardially injected exosomes accumulated in the MLN and induced regulatory T cell differentiation to promote cardiac repair. Mechanistically, uptake of exosomes by major histocompatibility complex (MHC)-II+ antigen-presenting cells (APCs) induced Foxo3 activation via the protein phosphatase (PP)-2A/p-Akt/forkhead box O3 (Foxo3) pathway. Foxo3 dominated APC cytokines (IL-10, IL-33, and IL-34) expression and built up a regulatory T cell (Treg)-inducing niche in the MLN. The differentiation of Tregs as well as their cardiac deployment were elevated, which contributed to cardiac inflammation resolution and cardiac repair.This study reveals a novel mechanism underlying the immunomodulation effects of MSC exosomes and provides a promising candidate (PP2A/p-Akt/Foxo3 signaling pathway) with a favorable delivery route (intrapericardial injection) for cardiac repair.}, number={10}, journal={CIRCULATION RESEARCH}, author={Zhu, Dashuai and Liu, Shuo and Huang, Ke and Wang, Zhenzhen and Hu, Shiqi and Li, Junlang and Li, Zhenhua and Cheng, Ke}, year={2022}, month={Oct}, pages={E135–E150} } @article{li_lv_zhu_mei_huang_wang_li_zhang_hu_popowski_et al._2022, title={Intrapericardial hydrogel injection generates high cell retention and augments therapeutic effects of mesenchymal stem cells in myocardial infarction}, volume={427}, ISSN={["1873-3212"]}, url={https://doi.org/10.1016/j.cej.2021.131581}, DOI={10.1016/j.cej.2021.131581}, abstractNote={Although cell therapy has shown potential efficacy in the treatment of heart diseases, one challenge is low cellular retention rate and poor engraftment. We sought to perform a head-to-head comparison on cell retention and therapeutic benefits of intramyocardial (IM) injection and intrapericardial cavity (IPC) injection of adult stem cells in hydrogel. Mouse green fluorescent protein (GFP)-labeled mesenchymal stem cells (MSCs) were combined in extracellular matrix (ECM) hydrogel and injected into the pericardial cavity or the myocardium of the heart of C57BL/6 mice that had been subjected to a myocardial infarction. The IPC injection, as an alternative cell delivery route, led to better cardiac function in our mouse model with myocardial infarction, which was showed by echocardiographies in the short term (2 weeks) and the long term (6 weeks). This result was attributed to 10-fold higher engraftment of MSCs injected via IPC route (42.5 ± 7.4%) than that of MSCs injected intramyocardially (4.4 ± 1.3%). Immunohistochemistry data revealed better cellular proliferation, less apoptosis, and better vascular regeneration in the myocardium after IPC delivery of MSCs. CD63-RFP exosome labeling system showed that heart cells including cardiomyocytes absorbed MSC-exosomes at higher rates when MSCs were injected via IPC route, compared to the results from IM injections, indicating more extensive paracrine activity of MSCs after IPC injections. What is more, the feasibility and safety of IPC injection were demonstrated in a porcine model with minimally invasive procedure. Intrapericardial cavity injection gave a promising solution for the low retention issue of MSCs in the infarcted heart.}, journal={CHEMICAL ENGINEERING JOURNAL}, publisher={Elsevier BV}, author={Li, Junlang and Lv, Yongbo and Zhu, Dashuai and Mei, Xuan and Huang, Ke and Wang, Xianyun and Li, Zhenhua and Zhang, Sichen and Hu, Shiqi and Popowski, Kristen D. and et al.}, year={2022}, month={Jan} } @article{zhang_zhang_zhu_li_wang_li_mei_xu_cheng_zhong_2022, title={Nanoparticles functionalized with stem cell secretome and CXCR4-overexpressing endothelial membrane for targeted osteoporosis therapy}, volume={20}, ISSN={["1477-3155"]}, url={http://www.scopus.com/inward/record.url?eid=2-s2.0-85122993345&partnerID=MN8TOARS}, DOI={10.1186/s12951-021-01231-6}, abstractNote={Abstract Background Osteoporosis is a chronic condition affecting patients’ morbidity and mortality and represents a big socioeconomic burden. Because stem cells can proliferate and differentiate into bone-forming cells, stem cell therapy for osteoporosis has been widely studied. However, cells as a live drug face multiple challenges because of their instability during preservation and transportation. In addition, cell therapy has potential adverse effects such as embolism, tumorigenicity, and immunogenicity. Results Herein, we sought to use cell-mimicking and targeted therapeutic nanoparticles to replace stem cells. We fabricated nanoparticles (NPs) using polylactic-co-glycolic acid (PLGA) loaded with the secretome (Sec) from mesenchymal stem cells (MSCs) to form MSC-Sec NPs. Furthermore, we cloaked the nanoparticles with the membranes from C–X–C chemokine receptor type 4 (CXCR4)-expressing human microvascular endothelial cells (HMECs) to generate MSC-Sec/CXCR4 NP. CXCR4 can target the nanoparticles to the bone microenvironment under osteoporosis based on the CXCR4/SDF-1 axis. Conclusions In a rat model of osteoporosis, MSC-Sec/CXCR4 NP were found to accumulate in bone, and such treatment inhibited osteoclast differentiation while promoting osteogenic proliferation. In addition, our results showed that MSC-Sec/CXCR4 NPs reduce OVX-induced bone mass attenuation in OVX rats. Graphical Abstract}, number={1}, journal={JOURNAL OF NANOBIOTECHNOLOGY}, author={Zhang, Chi and Zhang, Wei and Zhu, Dashuai and Li, Zhenhua and Wang, Zhenzhen and Li, Junlang and Mei, Xuan and Xu, Wei and Cheng, Ke and Zhong, Biao}, year={2022}, month={Jan} } @article{yao_huang_zhu_chen_jiang_zhang_mi_xuan_hu_li_et al._2021, title={A Minimally Invasive Exosome Spray Repairs Heart after Myocardial Infarction}, volume={15}, ISSN={["1936-086X"]}, url={http://www.scopus.com/inward/record.url?eid=2-s2.0-85110404046&partnerID=MN8TOARS}, DOI={10.1021/acsnano.1c00628}, abstractNote={Myocardial infarction (MI) remains the most common cause of death worldwide. Many MI survivors will suffer from recurrent heart failure (HF), which has been recognized as a determinant of adverse prognosis. Despite the success of improved early survival after MI by primary percutaneous coronary intervention, HF after MI is becoming the major driver of late morbidity, mortality, and healthcare costs. The development of regenerative medicine has brought hope to MI treatment in the past decade. Mesenchymal stem cell (MSC)-derived exosomes have been established as an essential part of stem cell paracrine factors for heart regeneration. However, its regenerative power is hampered by low delivery efficiency to the heart. We designed, fabricated, and tested a minimally invasive exosome spray (EXOS) based on MSC exosomes and biomaterials. In a mouse model of acute myocardial infarction, EXOS improved cardiac function and reduced fibrosis, and promoted endogenous angiomyogenesis in the post-injury heart. We further tested the feasibility and safety of EXOS in a pig model. Our results indicate that EXOS is a promising strategy to deliver therapeutic exosomes for heart repair.}, number={7}, journal={ACS NANO}, publisher={American Chemical Society (ACS)}, author={Yao, Jialu and Huang, Ke and Zhu, Dashuai and Chen, Tan and Jiang, Yufeng and Zhang, Junyi and Mi, Lijie and Xuan, He and Hu, Shiqi and Li, Junlang and et al.}, year={2021}, month={Jul}, pages={11099–11111} } @article{mei_zhu_li_huang_hu_li_abad_cheng_2021, title={A fluid-powered refillable origami heart pouch for minimally invasive delivery of cell therapies in rats and pigs}, volume={2}, ISSN={["2666-6340"]}, url={https://doi.org/10.1016/j.medj.2021.10.001}, DOI={10.1016/j.medj.2021.10.001}, abstractNote={BackgroundCardiac repair after heart injury remains a big challenge, and current drug delivery to the heart is suboptimal. Repeated dosing of therapeutics is difficult due to the invasive nature of such procedures.MethodsWe developed a fluid-driven heart pouch with a memory-shaped microfabricated lattice structure inspired by origami. The pore size of the membrane is around 0.4 μm, which allows for the penetration of growth factors and exosomes.FindingsWe tested the pouch’s ability to deliver mesenchymal stem cells (MSCs) in a rodent model of acute myocardial infarction and demonstrated the feasibility of minimally invasive delivery in a swine model. The pouch’s semi-permeable membrane successfully protected delivered cells from their surroundings, maintaining their viability while releasing growth factors and exosomes to the infarcted site for cardiac repair.ConclusionsIn summary, we developed a fluid-driven heart pouch with a memory-shaped microfabricated lattice structure inspired by origami. The origami structure allowed minimally invasive delivery of the pouch to the heart and can be refilled with the therapeutic of choice.FundingThis work was supported by grants from the NIH ( HL123920 , HL137093 , HL144002 , HL146153 , HL147357 , and HL149940 to K.C.) and the American Heart Association ( 18TPA34230092 and 19EIA34660286 to K.C.).}, number={11}, journal={MED}, publisher={Elsevier BV}, author={Mei, Xuan and Zhu, Dashuai and Li, Junlang and Huang, Ke and Hu, Shiqi and Li, Zhenhua and Abad, Blanca Lopez de Juan and Cheng, Ke}, year={2021}, month={Nov}, pages={1253-+} } @article{zhang_zhu_li_huang_hu_lutz_xie_mei_li_neal-perry_et al._2021, title={A stem cell-derived ovarian regenerative patch restores ovarian function and rescues fertility in rats with primary ovarian insufficiency}, volume={11}, ISSN={["1838-7640"]}, url={http://www.scopus.com/inward/record.url?eid=2-s2.0-85114771196&partnerID=MN8TOARS}, DOI={10.7150/thno.61690}, abstractNote={Rationale: Primary ovarian insufficiency (POI) normally occurs before age 40 and is associated with infertility. Hormone replacement therapy is often prescribed to treat vasomotor symptom, but it cannot restore ovarian function or fertility. Stem cell therapy has been studied for the treatment of POI. However, the application of live stem cells has suffered from drawbacks, such as low cell retention/engraftment rate, risks for tumorigenicity and immunogenicity, and lack of off-the-shelf feasibility. Methods: We developed a therapeutic ovarian regenerative patch (ORP) that composed of clinically relevant hydrolysable scaffolds and synthetic mesenchymal stem cells (synMSCs), which are microparticles encapsulating the secretome from MSCs. The therapeutic potency of ORP was tested in rats with cisplatin induced POI injury. Results:In vitro studies revealed that ORP stimulated proliferation of ovarian somatic cells (OSCs) and inhibited apoptosis under injury stress. In a rat model of POI, implantation of ORP rescued fertility by restoring sexual hormone secretion, estrus cycle duration, and follicle development. Conclusion: ORP represents a cell-free, off-the-shelf, and clinically feasible treatment for POI.}, number={18}, journal={THERANOSTICS}, author={Zhang, Sichen and Zhu, Dashuai and Li, Zhenhua and Huang, Ke and Hu, Shiqi and Lutz, Halle and Xie, Mengjie and Mei, Xuan and Li, Junlang and Neal-Perry, Genevieve and et al.}, year={2021}, pages={8894–8908} } @article{xie_li_zhang_zhu_mei_wang_cheng_li_wang_cheng_2021, title={A trifunctional contraceptive gel enhances the safety and quality of sexual intercourse}, volume={6}, ISSN={["2452-199X"]}, url={https://doi.org/10.1016/j.bioactmat.2020.11.031}, DOI={10.1016/j.bioactmat.2020.11.031}, abstractNote={Current contraceptive methods come with a number of drawbacks, including low efficacy, in the case of commercial contraceptive gels, and a reduction in the quality of sexual intercourse, in the case of condoms. Adding pharmacologically-active agents to contraceptive gels holds the potential to improve sexual experience, and hardbor safety and hygiene. In this study, we fabricated a carbomer-based contraceptive gel consisting of three agents: tenofovir, gossypol acetate, and nitroglycerin (TGN), with pH adjusted to 4.5 (to be compatible with the vagina). In vitro, the gossypol component of the contraceptive gel proved to be an effective spermicide. When the concentration of gossypol acetate was 10 mg/ml, the spermicidal ability reached 100% after 30 s. In addition, tenofovir in the gel significantly inhibited lentiviral transfection efficiency in cell-containing media. In 6 pairs of rats, the gel successfully prevented all females from conceiving after successful mating. Moreover, increased sexual frequency and enhanced erection, which were promoted by the nitroglycerin in the components, were observed in male rats that had the gel applied to their penises. This novel TGN contraceptive gel yielded a higher contraceptive success rate than that of the commercial contraceptive gel (Contragel®). In addition, it has the added benefits to prevent sexually transmitted diseases and improve male libido and erectile function during sexual intercourse. Combining three FDA-approved and marketed agents together, our trifunctional TGN gel has a great potential for further translation and commercialization.}, number={6}, journal={BIOACTIVE MATERIALS}, publisher={Elsevier BV}, author={Xie, Mengjie and Li, Junlang and Zhang, Sichen and Zhu, Dashuai and Mei, Xuan and Wang, Zhenzhen and Cheng, Xiao and Li, Zhenhua and Wang, Shaowei and Cheng, Ke}, year={2021}, month={Jun}, pages={1777–1788} } @misc{zhang_zhu_mei_li_li_xie_xie_wang_cheng_2021, title={Advances in biomaterials and regenerative medicine for primary ovarian insufficiency therapy}, volume={6}, ISSN={["2452-199X"]}, url={https://doi.org/10.1016/j.bioactmat.2020.12.008}, DOI={10.1016/j.bioactmat.2020.12.008}, abstractNote={Primary ovarian insufficiency (POI) is an ovarian dysfunction that affects more than 1 % of women and is characterized by hormone imbalances that afflict women before the age of 40. The typical perimenopausal symptoms result from abnormal levels of sex hormones, especially estrogen. The most prevalent treatment is hormone replacement therapy (HRT), which can relieve symptoms and improve quality of life. However, HRT cannot restore ovarian functions, including secretion, ovulation, and fertility. Recently, as part of a developing field of regenerative medicine, stem cell therapy has been proposed for the treatment of POI. Thus, we recapitulate the literature focusing on the use of stem cells and biomaterials for POI treatment, and sum up the underlying mechanisms of action. A thorough understanding of the work already done can aid in the development of guidelines for future translational applications and clinical trials that aim to cure POI by using regenerative medicine and biomedical engineering strategies.}, number={7}, journal={BIOACTIVE MATERIALS}, publisher={Elsevier BV}, author={Zhang, Sichen and Zhu, Dashuai and Mei, Xuan and Li, Zhenhua and Li, Junlang and Xie, Mengjie and Xie, Halle Jiang Williams and Wang, Shaowei and Cheng, Ke}, year={2021}, month={Jul}, pages={1957–1972} } @misc{li_hu_zhu_huang_mei_abad_cheng_2021, title={All Roads Lead to Rome (the Heart): Cell Retention and Outcomes From Various Delivery Routes of Cell Therapy Products to the Heart}, volume={10}, ISSN={["2047-9980"]}, url={https://doi.org/10.1161/JAHA.120.020402}, DOI={10.1161/JAHA.120.020402}, abstractNote={Abstract In the past decades, numerous preclinical studies and several clinical trials have evidenced the feasibility of cell transplantation in treating heart diseases. Over the years, different delivery routes of cell therapy have emerged and broadened the width of the field. However, a common hurdle is shared by all current delivery routes: low cell retention. A myriad of studies confirm that cell retention plays a crucial role in the success of cell‐mediated cardiac repair. It is important for any delivery route to maintain donor cells in the recipient heart for enough time to not only proliferate by themselves, but also to send paracrine signals to surrounding damaged heart cells and repair them. In this review, we first undertake an in‐depth study of primary theories of cell loss, including low efficiency in cell injection, “washout” effects, and cell death, and then organize the literature from the past decade that focuses on cell transplantation to the heart using various cell delivery routes, including intracoronary injection, systemic intravenous injection, retrograde coronary venous injection, and intramyocardial injection. In addition to a recapitulation of these approaches, we also clearly evaluate their strengths and weaknesses. Furthermore, we conduct comparative research on the cell retention rate and functional outcomes of these delivery routes. Finally, we extend our discussion to state‐of‐the‐art bioengineering techniques that enhance cell retention, as well as alternative delivery routes, such as intrapericardial delivery. A combination of these novel strategies and more accurate assessment methods will help to address the hurdle of low cell retention and boost the efficacy of cell transplantation to the heart.}, number={8}, journal={JOURNAL OF THE AMERICAN HEART ASSOCIATION}, publisher={Ovid Technologies (Wolters Kluwer Health)}, author={Li, Junlang and Hu, Shiqi and Zhu, Dashuai and Huang, Ke and Mei, Xuan and Abad, Blanca Lopez de Juan and Cheng, Ke}, year={2021}, month={Apr} } @article{wang_hu_li_zhu_wang_cores_cheng_liu_huang_2021, title={Extruded Mesenchymal Stem Cell Nanovesicles Are Equally Potent to Natural Extracellular Vesicles in Cardiac Repair}, volume={13}, ISSN={["1944-8252"]}, url={http://www.scopus.com/inward/record.url?eid=2-s2.0-85119954105&partnerID=MN8TOARS}, DOI={10.1021/acsami.1c08044}, abstractNote={Mesenchymal stem cells (MSCs) repair injured tissues mainly through their paracrine actions. One of the important paracrine components of MSC secretomes is the extracellular vesicle (EV). The therapeutic potential of MSC-EVs has been established in various cardiac injury preclinical models. However, the large-scale production of EVs remains a challenge. We sought to develop a scale-up friendly method to generate a large number of therapeutic nanovesicles from MSCs by extrusion. Those extruded nanovesicles (NVs) are miniature versions of MSCs in terms of surface marker expression. The yield of NVs is 20-fold more than that of EVs. In vitro, cell-based assays demonstrated the myocardial protective effects and therapeutic potential of NVs. Intramyocardial delivery of NVs in the injured heart after ischemia-reperfusion led to a reduction in scar sizes and preservation of cardiac functions. Such therapeutic benefits are similar to those injected with natural EVs from the same MSC parental cells. In addition, NV therapy promoted angiogenesis and proliferation of cardiomyocytes in the post-injury heart. In summary, extrusion is a highly efficient method to generate a large quantity of therapeutic NVs that can potentially replace extracellular vesicles in regenerative medicine applications.}, number={47}, journal={ACS APPLIED MATERIALS & INTERFACES}, publisher={American Chemical Society (ACS)}, author={Wang, Xianyun and Hu, Shiqi and Li, Junlang and Zhu, Dashuai and Wang, Zhenzhen and Cores, Jhon and Cheng, Ke and Liu, Gang and Huang, Ke}, year={2021}, month={Dec}, pages={55767–55779} } @article{hu_wang_li_zhu_cores_wang_li_mei_cheng_su_et al._2021, title={Platelet membrane and stem cell exosome hybrids enhance cellular uptake and targeting to heart injury}, volume={39}, ISSN={["1878-044X"]}, url={http://www.scopus.com/inward/record.url?eid=2-s2.0-85107566346&partnerID=MN8TOARS}, DOI={10.1016/j.nantod.2021.101210}, abstractNote={• Platelet membrane modification enhanced the accumulation of exosomes in injured tissues. • Platelet membrane substantially enhanced macropinocytosis-mediated cellular internalization of exosomes. • Platelet membrane hybrid improved the pharmacokinetic of stem cell derived exosomes. • This is a simple, fast, and translatable approach to modify therapeutic exosomes to make them disease targetable. We used platelet membrane to modify exosomes and rely on the natural “injury finding” ability of platelets to target the hybrid exosomes to vascular injury under myocardial infarction. Interestingly, we found that platelet membrane improved cellular binding and internalization of exosomes through enhanced macropinocytosis-mediated cellular internalization by endothelial cells and cardiomyocytes, but not by macrophages. Such modified exosomes showed improved targeting and functional benefits in vitro, and in a mouse myocardial infarction model in vivo, as compared to unmodified naive exosomes. Exosomes from mesenchymal stem cells have been widely studied as therapeutics to treat myocardial infarction. However, exosomes injected for therapeutic purposes face a number of challenges, including competition from endogenous exosomes, and the internalization/clearance by the mononuclear phagocyte system. There is also a lack of targeting. In this study, we hybridized stem cell-derived exosomes with platelet membranes to enhance their ability to target the injured heart and to reduce uptake by macrophages. Furthermore, we found that hybridization with platelet membranes induces macropinocytosis, enhancing the cellular uptake of exosomes by endothelial cells and cardiomyocytes drastically. In vivo studies showed the cardiac targeting ability of hybrid exosomes in a mouse model of myocardial infarction injury. Lastly, we determined cardiac functions and performed immunohistochemistry to confirm an enahnced therapeutic potency of platelet membrane modified exosomes as compared to non-modified exosomes. Our studies provide proof-of-concept data and a universal approach to enhance the binding and accumulation of exosomes in injured tissues.}, journal={NANO TODAY}, author={Hu, Shiqi and Wang, Xianyun and Li, Zhenhua and Zhu, Dashuai and Cores, Jhon and Wang, Zhenzhen and Li, Junlang and Mei, Xuan and Cheng, Xiao and Su, Teng and et al.}, year={2021}, month={Aug} } @misc{li_hu_cheng_2020, title={Engineering better stem cell therapies for treating heart diseases}, volume={8}, ISSN={["2305-5847"]}, DOI={10.21037/atm.2020.03.44}, abstractNote={: For decades, stem cells and their byproducts have shown efficacy in repairing tissues and organs in numerous pre-clinical studies and some clinical trials, providing hope for possible cures for many important diseases. However, the translation of stem cell therapy for heart diseases from bench to bed is still hampered by several limitations. The therapeutic benefits of stem cells are mediated by a combo of mechanisms. In this review, we will provide a brief summary of stem cell therapies for ischemic heart disease. Basically, we will talk about these barriers for the clinical application of stem cell-based therapies, the investigation of mechanisms behind stem-cell based cardiac regeneration and also, what bioengineers can do and have been doing on the translational stage of stem cell therapies for heart repair.}, number={8}, journal={ANNALS OF TRANSLATIONAL MEDICINE}, author={Li, Junlang and Hu, Shiqi and Cheng, Ke}, year={2020}, month={Apr} }