Mesenchymal Stromal Cells From Emphysematous Donors and Their Extracellular Vesicles Are Unable to Reverse Cardiorespiratory Dysfunction in Experimental Severe Emphysema

Mariana A Antunes, Cassia L Braga, Tainá B Oliveira, Jamil Z Kitoko, Ligia L Castro, Debora G Xisto, Mariana S Coelho, Nazareth Rocha, Rodrigo P Silva-Aguiar, Celso Caruso-Neves, Eduarda G Martins, Clara Fernandes Carvalho, Antônio Galina, Daniel J Weiss, José R Lapa E Silva, Miquéias Lopes-Pacheco, Fernanda F Cruz, Patricia R M Rocco, Mariana A Antunes, Cassia L Braga, Tainá B Oliveira, Jamil Z Kitoko, Ligia L Castro, Debora G Xisto, Mariana S Coelho, Nazareth Rocha, Rodrigo P Silva-Aguiar, Celso Caruso-Neves, Eduarda G Martins, Clara Fernandes Carvalho, Antônio Galina, Daniel J Weiss, José R Lapa E Silva, Miquéias Lopes-Pacheco, Fernanda F Cruz, Patricia R M Rocco

Abstract

Although bone marrow-derived mesenchymal stromal cells (BM-MSCs) from patients with chronic obstructive pulmonary disease (COPD) appear to be phenotypically and functionally similar to BM-MSCs from healthy sources in vitro, the impact of COPD on MSC metabolism and mitochondrial function has not been evaluated. In this study, we aimed to comparatively characterize MSCs from healthy and emphysematous donors (H-MSCs and E-MSCs) in vitro and to assess the therapeutic potential of these MSCs and their extracellular vesicles (H-EVs and E-EVs) in an in vivo model of severe emphysema. For this purpose, C57BL/6 mice received intratracheal porcine pancreatic elastase once weekly for 4 weeks to induce emphysema; control animals received saline under the same protocol. Twenty-four hours after the last instillation, animals received saline, H-MSCs, E-MSCs, H-EVs, or E-EVs intravenously. In vitro characterization demonstrated that E-MSCs present downregulation of anti-inflammatory (TSG-6, VEGF, TGF-β, and HGF) and anti-oxidant (CAT, SOD, Nrf2, and GSH) genes, and their EVs had larger median diameter and lower average concentration. Compared with H-MSC, E-MSC mitochondria also exhibited a higher respiration rate, were morphologically elongated, expressed less dynamin-related protein-1, and produced more superoxide. When co-cultured with alveolar macrophages, both H-MSCs and E-MSCs induced an increase in iNOS and arginase-1 levels, but only H-MSCs and their EVs were able to enhance IL-10 levels. In vivo, emphysematous mice treated with E-MSCs or E-EVs demonstrated no amelioration in cardiorespiratory dysfunction. On the other hand, H-EVs, but not H-MSCs, were able to reduce the neutrophil count, the mean linear intercept, and IL-1β and TGF-β levels in lung tissue, as well as reduce pulmonary arterial hypertension and increase the right ventricular area in a murine model of elastase-induced severe emphysema. In conclusion, E-MSCs and E-EVs were unable to reverse cardiorespiratory dysfunction, whereas H-EVs administration was associated with a reduction in cardiovascular and respiratory damage in experimental severe emphysema.

Keywords: COPD; cell therapy; extracellular vesicles; inflammation; macrophages; mesenchymal stromal cells; mitochondria.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Antunes, Braga, Oliveira, Kitoko, Castro, Xisto, Coelho, Rocha, Silva-Aguiar, Caruso-Neves, Martins, Carvalho, Galina, Weiss, Lapa e Silva, Lopes-Pacheco, Cruz and Rocco.

Figures

FIGURE 1
FIGURE 1
Charaterization of mediators produced by MSCs from healthy and emphysematous donors. H-MSCs, bone marrow-derived mesenchymal stromal cells (BM-MSCs) obtained from healthy donors; E-MSCs, BM-MSCs obtained from emphysematous donors. Gene expression profile of tumor necrosis factor α-induced protein-6 (TSG-6), indoleamine 2,3-dioxygenase 1 (IDO-1), interleukin-1 receptor antagonist (IL-1RN), interleukin 10 (IL-10), vascular endothelial growth factor (VEGF), transforming growth factor β (TGF-β), and hepatocyte growth factor (HGF) in H-MSCs and E-MSCs. Data are expressed as means ± standard deviation of 5–6 samples in each group. *P < 0.05 vs. the H-MSC group.
FIGURE 2
FIGURE 2
Characterization of EVs secreted by MSCs from healthy and emphysematous donors. H-MSCs, bone marrow-derived mesenchymal stromal cells (BM-MSCs) obtained from healthy donor; E-MSCs, BM-MSCs obtained from emphysematous donors; H-EVs, extracellular vesicles derived from BM-MSCs obtained from healthy donors; E-EVs, extracellular vesicles derived from BM-MSCs obtained from emphysematous donors. (A) Representative scanning electron microscopy of H-MSCs and E-MSCs, illustrating EV release from the surface of both types of MSCs. (B–D) Characterization of H-EVs and E-EVs, as determined by total protein content (B) and the size (C), concentration (D), and distribution (E) of secreted particles, measured by the Bradford assay and nanoparticle tracking analysis, respectively. Data are expressed as medians (interquartile range) of 5–6 samples in each group. *P < 0.05 vs. the H-EV group.
FIGURE 3
FIGURE 3
Characterization of mitochondria of MSCs from healthy and emphysematous donors. (A) An OROBOROS system was used to measure basal (Routine), spare respiration capacity (SPC) and maximum (electron transfer system, ETS) mitochondrial respiration in H-MSCs and E-MSCs. (B) Citrate synthase was used as a quantitative enzyme marker for the presence of intact mitochondria in H-MSCs and E-MSCs. (C) Oxygraphy parameters were normalized by citrate synthase activity. (D) Mean fluorescence intensity detected by MitoSOX-based flow cytometry in H-MSCs and E-MSCs. Quantitative analysis of several antioxidant (E) and mitochondrial dynamics-related (F) genes in H-MSCs and E-MSCs by real-time RT-PCR. (G) Transmission electron microscopy to assess morphological features of mitochondria in H-MSCs and E-MSCs. Note mitochondrial elongation in E-MSCs (arrows). CAT, catalase; SOD2, superoxide dismutase 2; Nrf2, nuclear factor erythroid 2-related factor; GSH, glutathione; MFN1, mitofusin-1; MFN2, mitofusion-2; DNM1, dynamin-1. Data are expressed as means ± standard deviation of 5–6 samples in each group. *P < 0.05 vs. the H-MSC group.
FIGURE 4
FIGURE 4
Representative immunoblotting showing mitofusin-1 (MFN1), mitofusin-2 (MNF2), dynamin-related protein 1 (DRP1), and β-actin in MSCs from health (H) and emphysematous (E) donors. Data are expressed (right panel) as the ratio between protein expression and β-actin obtained by densitometry of 2 samples in each group.
FIGURE 5
FIGURE 5
Mitochondrial transfer from healthy or emphysematous MSCs and EVs to alveolar macrophages (MΦ). (A) Intensity of MitoRed fluorescence of healthy (H) or emphysematous (E) MSCs decreased after direct co-culture with MΦ (light blue and pink histograms, respectively). The extent of mitochondrial transfer to MΦ was measured by flow cytometry. Co-culture of MΦ with MitoRed-pretreated H-MSCs and E-MSCs resulted in increased MitoRed (APC+) mean fluorescent intensity of MΦ. (B) Scatter plots show MΦ and EV after 24 h in co-culture. The presence of CD45+ MΦ, demonstrating acquisition of MitoRed fluorescence (APC+), indicates that mitochondrial transfer occurs from H-EVs and E-EVs. Data are representative of three independent experiments. The histograms represented graphically in (A) are from a single representative experiment. *P < 0.05 vs. the MΦ + H-MSC.
FIGURE 6
FIGURE 6
Direct co-culture of MSCs and EVs from healthy or emphysematous donors modulates the alveolar macrophage (MΦ) profile. H-MSCs, 1 × 106 bone marrow-derived mesenchymal stromal cells (BM-MSCs) obtained from healthy donors; E-MSCs, 1 × 106 BM-MSCs obtained from emphysematous donors; H-EVs, extracellular vesicles derived from 1 × 106 BM-MSCs obtained from healthy donors; E-EVs, extracellular vesicles derived from 1 × 106 BM-MSCs obtained from emphysematous donors. Data are expressed as medians (interquartile range) of 5–6 samples in each group. *P < 0.05 vs. MΦ. **P < 0.05 vs. the H-MSC. ***P < 0.05 vs. the E-MSC. &P < 0.05 vs. the H-EV.
FIGURE 7
FIGURE 7
Levels of IL-1β (A), IL-10 (B), and TGF-β (C) in lung tissue. C, intratracheal instillation of 50 μL of saline; ELA, intratracheal instillation of 0.2 IU of pancreatic porcine elastase; SAL, intravenous injection of 50 μL of saline; H-MSCs, 1 × 106 bone marrow-derived mesenchymal stromal cells (BM-MSCs) obtained from healthy donors; E-MSCs, 1 × 106 BM-MSCs obtained from emphysematous donors; H-EVs, extracellular vesicles derived from 1 × 106 BM-MSCs obtained from healthy donors; E-EVs, extracellular vesicles derived from 1 × 106 BM-MSCs obtained from emphysematous donors. IL-1β, interleukin-1β; IL-10, interleukin-10; TGF-β, transforming growth factor-β. Data are expressed as means ± standard deviation of 7–10 animals/group. *P < 0.05 vs. the C group. #P < 0.05 vs. the ELA-SAL group. **P < 0.05 vs. the H-MSC group.
FIGURE 8
FIGURE 8
Echocardiographic analysis. Parasternal short axis view of both ventricles (upper panel) and pulmonary artery flow dynamics (lower panel). (A) Pulmonary artery acceleration time/pulmonary artery ejection time ratio (PAT/PET) and (B) right ventricle area. C, intratracheal instillation of 50 μL of saline; ELA, intratracheal instillation of 0.2 IU of pancreatic porcine elastase; SAL, intravenous injection of 50 μL of saline; H-MSCs, 1 × 106 bone marrow-derived mesenchymal stromal cells (BM-MSCs) obtained from healthy donors; E-MSCs, 1 × 106 BM-MSCs obtained from emphysematous donors; H-EVs: extracellular vesicles derived from 1 × 106 BM-MSCs obtained from healthy donors; E-EVs, extracellular vesicles derived from 1 × 106 BM-MSCs obtained from emphysematous donors. RV, right ventricle. Data are expressed as means ± standard deviation of 7–10 animals/group. *P < 0.05 vs. the C group. #P < 0.05 vs. the ELA-SAL group.
FIGURE 9
FIGURE 9
Effects of H-MSCs, E-MSCs, H-EVs or E-EVs treatments on the remodeling process. Left panels: representative photomicrographs of lung parenchyma stained with Masson’s trichome staining collagen fibers in airways, (A); walls of blood vessels in interlobular septa, (B) and Weigert’s resorcin fuchsin elastic fibers in alveolar septa, (C). Note that blood vessels are highlighted by red arrowheads, elastic fibers by black arrowheads, and hyperinflated areas by red asterisks. C: intratracheal instillation of 50 μL of saline; ELA: intratracheal instillation of 0.2 UI of pancreatic porcine elastase (PPE); SAL: intravenous injection of 50 μL of saline; H-MSCs: 1 × 106 bone marrow mesenchymal stromal cells obtained from healthy donor; E-MSCs: 1 × 106 bone marrow mesenchymal stromal cells obtained from emphysematous donor; H-EVs: extracellular vesicles derived from 1 × 106 bone marrow mesenchymal stromal cells obtained from healthy donor; E-EVs: extracellular vesicles derived from 1 × 106 bone marrow mesenchymal stromal cells obtained from emphysematous donor. Right panels: Data are expressed as mean + SD of 5–7 animals/group. *vs. C group (p < 0.05). #Vs. ELA-SAL group (p < 0.05).

References

    1. Abreu S. C., Lopes-Pacheco M., Weiss D. J., Rocco P. R. M. (2021). Mesenchymal stromal cell-derived extracellular vesciles in lung diseases: current status and perspectives. Front. Cell Dev. Biol. 9:600711. 10.3389/fcell.2021.600711
    1. Abreu S. C., Xisto D. G., de Oliveira T. B., Blanco N. G., de Castro L. L., Kitoko J. Z., et al. (2019). Serum from asthmatic mice potentiates the therapeutic effects of mesenchymal stromal cells in experimental allergic asthma. Stem Cells Transl. Med. 8 301–312. 10.1002/sctm.18-0056
    1. Antunes M. A., Abreu S. C., Cruz F. F., Teixeira A. C., Lopes-Pacheco M., Bandeira E., et al. (2014). Effects of diferente mesenchymal stromal cell sources and delivery routes in experimental emphysema. Respir. Res. 15:118. 10.1186/s12931-014-0118-x
    1. Antunes M. A., Lopes-Pacheco M., Rocco P. R. M. (2021). Oxidative stress-derived mitochondrial dysfunction in chronic obstructive pulmonary disease: a concise review. Oxid. Med. Cell. Longev. 2021:6644002. 10.115/2021/6644002
    1. Barnes P. J., Burney P. G. J., Silverman E. K., Celli B. R., Vestbo J., Wedzicha J. A., et al. (2015). Chronic obstructive pulmonary disease. Nat. Rev. Dis. Primers 1:15076. 10.1038/nrdp.2015.76
    1. Bates J. H., Ludwig M. S., Sly P. D., Brown K., Martin J. G., Fredberg J. J. (1988). Interrupter resistance elucidated by alveolar pressure measurement in open-chest normal dogs. J. Appl. Physiol. 65 408–414. 10.1152/jappl.1988.65.1.408
    1. Broekman W., Roelofs H., Zarcone M. C., Taube C., Stolk J., Hiemstra P. S. (2016). Functional characterisation of bone marrow-derived mesenchymal stromal cells from COPD patients. ERJ Open Res. 2 00045–2015. 10.1183/23120541.00045-2015
    1. Chang S.-H., Park C.-G. (2019). Allogeneic ADSCs induces CD8 T cell-mediated cytotoxicity and faster cell death after exposure to xenogeneic serum or proinflammatory cytokines. Exp. Mol. Med. 51 1–10. 10.1038/s12276-019-0231-5
    1. Chen H., Chomyn A., Chan D. C. (2005). Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J. Biol. Chem. 280 26185–26192. 10.1074/jbc.M503062200
    1. Collino F., Pomatto M., Bruno S., Lindoso S., Tapparo M., Sicheng W., et al. (2017). Exosome and microvesicle-enriched franctions isolated from mesenchymal stem cells by gradient separation showed different molecular signatures and functions on renal tubular epithelial cells. Stem Cell Rev. Rep. 13 226–243. 10.1007/s12015-016-9713-1
    1. de Carvalho L. R. P., Abreu S. C., de Castro L. L., Andrade da Silva L. H., Silva P. L., Vieira J. B., et al. (2021). Mitochondria-rich fraction isolated from mesenchymal stromal cells reduces lung and distal organ injury in experimental sepsis. Crit. Care Med. 10.1097/ccm.0000000000005056 [Epub Online ahead of print].
    1. Dyer D. P., Thomson J. M., Hermant A., Jowitt T. A., Handel T. M., Proudfoot A. E. I., et al. (2014). TSG-6 inhibits neutrophil migration via direct interaction with the chemokine CXCL8. J. Immunol. 192 2177–2185. 10.4049/jimmunol.1300194
    1. Eapen M. S., Hansbro P. M., McAlinden K., Kin R. Y., Ward C., Hackett T.-L., et al. (2017). Abnormal M1/M2 macrophage phenotype profiles in the small airway wall and lumen in smokers and chronic obstructive pulmonary disease (COPD). Sci. Rep. 7:13392. 10.1038/s41598-017-13888-x
    1. El-Gayar S., Thuring-Nahler H., Pfeilschifter J., Rollinghoff M., Bogdan C. (2003). Translational control of inducible nitric oxide synthase by IL-13 and arginine availability in inflammatory macrophages. J. Immunol. 171 4561–4568. 10.4049/jimmunol.171.9.4561
    1. Global Initiative for Chronic Obstructive Lung Diseases [GOLD] (2020). Global Strategy for Prevention, Diagnosis and Management of COPD. Available online at: (accessed December 30, 2020).
    1. Guan X.-J., Song L., Han F.-F., Cui Z.-L., Chen X., Gui X.-J., et al. (2013). Mesenchymal stem cells protect cigarette smoke-damaged lung and pulmonary function partly via VEGF-VEGF receptors. J. Cell Biochem. 114 323–335. 10.1002/jcb.24377
    1. Henriques I., Lopes-Pacheco M., Padilha G. A., Marques O. S., Magalhães R. F., Antunes M. A., et al. (2016). Moderate aerobic training improves cardiorespiratory parameters in elastase-induced emphysema. Front. Physiol. 7:329. 10.3389/fphys.2016.00329
    1. Hsia C. C. W., Hyde D. M., Ochs M., Weibel E. R. Ats/Ers Joint Taks Force on Quantitative Assessment of Lung Structure. (2010). An official research policy statement of the American Thoracic Society/European Respiratory Society: standards for quantitative assessment of lung structure. Am. J. Respir. Crit. Care Med. 181 394–418. 10.1164/rccm.200809-1522ST
    1. Huertas A., Testa U., Riccioni R., Petrucci E., Riti V., Savi D., et al. (2010). Bone marrow-derived progenitors are greatly reduced in patients with severe COPD and low-BMI. Respir. Physiol. Neurobiol. 170 23–31. 10.1016/j.resp.2009.10.003
    1. Islam M. N., Das S. R., Emin M. T., Wei M., Sun L., Westphalen K., et al. (2012). Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat. Med. 18 759–765. 10.1038/nm.2736
    1. Jackson M. V., Krasnodembskaya A. D. (2017). Analysis of mitochondrial transfer in direct co-cultures of human monocyte-derived macrophages (MDM) and mesenchymal stem cells (MSC). Bio Protoc. 7:e2255. 10.21769/BioProtoc.2255
    1. Jackson M. V., Morrison T. J., Doherty D. F., McAuley D. F., Matthay M. A., Kissenpfennig A., et al. (2016). Mitochondrial transfer via tunneling nanotubes is an important mechanism by which mesenchymal stem cells enhance macrophage phagocytosis in the in vitro and in vivo models of ARDS. Stem Cells 34 2210–2223. 10.1002/stem.2372
    1. Janssen W. J., Yunt Z. X., Muldrow A., Kearns M. T., Kloepfer A., Barthel L., et al. (2014). Circulating hematopoietic progenitor cells are decreased in COPD. COPD 11 277–289.
    1. Ji H., Chen M., Greening D. W., He W., Rai A., Zhang W., et al. (2014). Deep sequencing of RNA from three different extracellular vesicle (EV) subtypes released from the human LIM1863 colon cancer cell line uncovers distinct miRNA-enrichment signatures. PLoS One 9:e110314. 10.1371/journal.pone.0110314
    1. Jin E., Lee P. T., Jeon W. B., Li W.-J. (2016). Effects of elastin-like peptide on regulation of human stem cell behavior. Reg. Eng. Transl. Med. 2 85–97. 10.1007/s40883-016-0015-6
    1. Kauffman M. E., Kaufmann M. K., Traore K., Zhu H., Trush M. A., Jia Z., et al. (2016). MitoSOX-based flow cytometry for detecting mitochondrial ROS. React. Oxyg. Species 2 361–370. 10.20455/ros.2016.865
    1. Kennelly H., Mahon B. P., English K. (2016). Human mesenchymal stromal cells exert HGF dependent cytoprotective effects in a human relevant pre-clinical model of COPD. Sci. Rep. 6:38207. 10.1038/srep38207
    1. Kulkarni T., O’Reilly P., Antony V. B., Gaggar A., Thannickal V. J. (2016). Matrix remodeling in pulmonary fibrosis and emphysema. Am. J. Respir. Cell Mol. Biol. 54 751–760.
    1. Lang R. M., Bierig M., Devereux R. B., Flachskampf F. A., Foster E., Pellikka P. A., et al. (2005). Recommendations for chamber quantification: a report from the American Society of Echocardiography’s Guidelines and Standards Committee and the Chamber Quantification Writing Group, Developed in conjunction with the European Association of Echocardiography, a branch of the European Society of Cardiology. J. Am. Soc. Echocardiogr. 18 1440–1463. 10.1016/j.echo.2005.10.005
    1. Leboucher G. P., Tsai Y. C., Yang M., Shaw K. C., Zhou M., Veenstra T. D. (2012). Stress-induced phosphorylation and proteasomal degradation of mitofusion 2 facilitates mitochondrial fragmentation and apoptosis. Mol. Cell 47 547–557. 10.1016/j.molcel.2012.05.041
    1. Li Y., Fung J., Lin F. (2016). Local inhibition of complement improves mesenchymal stem cell viability and function after administration. Mol. Ther. 24 1665–1674. 10.1038/mt.2016.142
    1. Li Y., Lin F. (2012). Mesenchymal stem cells are injured by complement after their contact with serum. Blood 120 3436–3443. 10.1182/blood-2012-03-420612
    1. Lopes-Pacheco M., Robba C., Rocco P. R. M., Pelosi P. (2020). Current understanding of the therapeutic benefits of mesenchymal stem cells in acute respiratory distress syndrome. Cell Biol. Toxicol. 36 83–102. 10.1007/s10565-019-09493-5
    1. Martínez J., Tarallo D., Martínez-Palma L., Victoria S., Bresque M., Rodríguez-Bottero S., et al. (2019). Mitofusins modulate the increase in mitochondrial lenght, bioenergetics and secretory phenotype in therapy-induced senescent melanoma cells. Biochem. J. 476 2463–2486. 10.1042/bcj20190405
    1. Moravcikova E., Meyer E. M., Corselli M., Donnenberg V. S., Donnenberg A. D. (2018). Proteomic profiling of native unpassaged and culture-expanded mesenchymal stromal cells (MSC). Cytometry A 93 894–904. 10.1002/cyto.a.23574
    1. Morrison T. J., Jackson M. V., Cunningham E. K., Kissenpfennig A., McAuley D. F., O’Kane C. M., et al. (2017). Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellulatr vesicles mitochondrial transfer. Am. J. Respir. Crit. Care Med. 196 1275–1286. 10.1164/rccm.201701-0170OC
    1. Oliveira H. G., Cruz F. F., Antunes M. A., Neto A. V. M., Oliveira G. A., Svartman F., et al. (2017). Combined bone marrow-derived mesenchymal stromal cell therapy and one-way endobronchial valve placement in patients with pulmonary emphysema: a phase I clinical trial. Stem Cells Transl. Med. 6 962–969. 10.1002/sctm.16-0315
    1. Oliveira M. V., Abreu S. C., Padilha G. A., Rocha N. N., Maia L. A., Takiya C. M., et al. (2016). Characterization of a mouse modelo f emphysema induced by multiple instillations of low-dose elastase. Front. Physiol. 7:457. 10.3389/fphys.2016.00457
    1. Padilha G. A., Henriques I., Lopes-Pacheco M., Abreu S. C., Oliveira M. V., Morales M. M., et al. (2015). Therapeutic effects of LASSBio-596 in an elastase-induced mouse model of emphysema. Front. Physiol. 6:267. 10.3389/fphys.2015.00267
    1. Poggio H. A., Antunes M. A., Rocha N. N., Kitoko Morales M. M., Olsen P. C., Lopes-Pacheco M., et al. (2018). Impact of one versus two doses of mesenchymal stromal cells on lung and cardiovascular repair in experimental emphysema. Stem Cell Res. Ther. 9:296. 10.1186/s13287-018-1043-6
    1. Pyakurel A., Savoia C., Hess D., Scorrano L. (2015). Extracellular regulated kinase phosphorypates mitofusion 1 to control mitochondrial morphology and apoptosis. Mol. Cell 58 244–254. 10.1016/j.molcel.2015.02.021
    1. Somaiah C., Kumar A., Mawrie D., Sharma A., Patil S. D., Bhattacharyya J., et al. (2015). Collagen promotes higher adhesin, survival and proliferation of mesenchymal stem cells. PLoS One 10:e0145068. 10.1371/journal.pone.0145068
    1. Stolk J., Broekman W., Mauad T., Zwaginga J. J., Roelofs H., Fibbe W. E., et al. (2016). A phase I for intravenous autologous mesenchymal stromal cell administration to patients with severe emphysema. QJM 109 331–336. 10.1093/qjmed/hcw001
    1. Taguchi N., Ishihara N., Jofuku A., Oka T., Mihara K. (2007). Mitotic phosphorylation of dynamin-related GTPase Drp1 participates in mitochondrial fission. J. Biol. Chem. 282 11521–11529. 10.1074/jbc.m607279200
    1. Than U. T. T., Guanzon D., Broadbent J. A., Parker T. J., Leavesley D. I. (2020). Deep sequencing microRNAs from extracellular membrane vesicles revealed the association of the vesicle cargo with cellular origin. Int. J. Mol. Sci. 21:1141. 10.3390/ijms21031141
    1. Toledo-Pons N., Noell G., Jahn A., Iglesias A., Duran M. A., Iglesias J., et al. (2018). Bone marrow characterization in COPD: a multi-level network analysis. Respir. Res. 19:118. 10.1186/s12931-018-0824-x
    1. Weiss D. J., Casaburi R., Flannery R., LeRoux-Williams M., Tashkin D. P. (2013). A placebo-controlled, randomized trial of mesenchymal stem cells in COPD. Chest 143 1590–1598. 10.1378/chest.12-2094
    1. Wong S. W., Lenzini S., Cooper M. H., Mooney D. J., Shin J.-W. (2020). Sof extracellular matrix enhances inflammatory activation of mesenchymal stromal cells to induce monocyte production and trafficking. Sci. Adv. 6:eaaw0158. 10.1126/sciadv.aaw0158
    1. Yoon Y.-S., Yoon D.-S., Lim I. K., Yoon S.-H., Chung H.-Y., Rojo M., et al. (2006). Formation of elongated giant mitochondria in DFO-induced cellular senescence: involvement of enhanced fusion process through modulation of Fis1. J. Cell. Physiol. 209 468–480. 10.1002/jcp.20753
    1. Youle R. J., van der Bliek A. (2012). Mitochondrial fission, fusion, and stress. Science 337 1062–1065. 10.1126/science.1219855

Source: PubMed

3
Sottoscrivi