pcMSC Modulates Immune Dysregulation in Patients With COVID-19-Induced Refractory Acute Lung Injury

Mei-Chuan Chen, Kevin Shu-Leung Lai, Ko-Ling Chien, Sing Teck Teng, Yuh-Rong Lin, Wei Chao, Meng-Jung Lee, Po-Li Wei, Yen-Hua Huang, Han-Pin Kuo, Chih-Ming Weng, Chun-Liang Chou, Mei-Chuan Chen, Kevin Shu-Leung Lai, Ko-Ling Chien, Sing Teck Teng, Yuh-Rong Lin, Wei Chao, Meng-Jung Lee, Po-Li Wei, Yen-Hua Huang, Han-Pin Kuo, Chih-Ming Weng, Chun-Liang Chou

Abstract

Background and objectives: The novel coronavirus disease 2019 (COVID-19) has been a pandemic health issue in 30 January 2020. The mortality rate is as high as 50% in critically ill patients. Stem cell therapy is effective for those who are refractory to standard treatments. However, the immune responses that underlie stem cell therapy have not been well reported, particularly, in patients associated with moderate to severe acute respiratory distress syndrome (ARDS).

Methods: On Days 0 and 4, an intravenous infusion of 2 × 107 placenta-derived mesenchymal stem cells (pcMSCs) (MatriPlax) were administered to five severe COVID-19 patients refractory to current standard therapies. Peripheral blood inflammatory markers and immune profiles were determined by multi-parameter flow cytometry and studied at Days 0, 4, and 8. Clinical outcomes were also observed.

Results: None of the pc-MSC treated patients experienced 28-day mortality compared with the control group and showed a significant improvement in the PaO2/FiO2 ratio, Murray's lung injury scores, reduction in serum ferritin, lactate dehydrogenase (LDH), and C-reactive protein (CRP) levels. The cytokine profiles also showed a reduction in IL-1β, IFN-γ, IL-2, and IL-6, and an increase in IL-13 and IL-5 type 2 cytokines within 7 days of therapy. Lymphopenia was also significantly improved after 7 days of treatment. Immune cell profiles showed an increase in the proportions of CD4+ T cells (namely, CD4+ naïve T cells and CD4+ memory T cell subtypes), Treg cells, CD19+ B cells (namely, CD19+ naïve B cells, CD27+ switched B cell subtypes) and dendritic cells, and a significant decrease in the proportion of CD14+ monocytes (namely, CD16- classical and CD16+ non-classical subtypes), and plasma/plasmablast cells. No adverse effects were seen at the serial follow-up visits for 2 months after initial therapy.

Conclusion: pc-MSCs therapy suppressed hyper-inflammatory states of the innate immune response to COVID-19 infection by increasing Treg cells, decreasing monocytes and plasma/plasmablast cells, and promoting CD4+ T cells and CD19+ B cells toward adaptive immune responses in severely critically ill COVID-19 patients with moderate to severe ARDS, especially those who were refractory to current standard care and immunosuppressive therapies.

Keywords: COVID-19; Treg cells; immune response; mesenchymal stem cell; severe lung injury.

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 © 2022 Chen, Lai, Chien, Teng, Lin, Chao, Lee, Wei, Huang, Kuo, Weng and Chou.

Figures

Figure 1
Figure 1
Clinical characteristics and clinical outcomes of patient with COVID-19. (A) PaO2/FiO2 ratio at Day 0 (before treatment), Day 4 (after the first treatment with MSC), and Day 8 (after second treatment with MSC) in five treated patients. (B) Murray’s lung injury score was shown before and after treatment; timing of after was adjusted as one week after recruitment. (C) Serum proinflammatory markers, derritin, LDH, CRP, and D-dimer on Days 0, 4, and 8 after pcMSCs therapy of five treated patients. The P value was as indicated. Nonparametric Wilcoxon sign-rank test.
Figure 2
Figure 2
Clinical characteristics of COVID-19 patient with standard or MSC treatment. (A) Murray’s lung injury score of standard treatment and MSC treatment group was shown as before and after treatment; timing of after was adjusted as one week after recruitment. (B) Duration of viral clearance by detecting viral COVID molecular testing by PCR in control and MSC treated group. (C) Survival analysis of control and MSC treated group.
Figure 3
Figure 3
The proinflammatory markers in the peripheral blood. IL-10 and type 2 cytokines changes of 5 treated patients in Days 0, 4 and 8 after pcMSCs therapy. The P value was as indicated. Nonparametric Wilcoxon sign-rank test.
Figure 4
Figure 4
Changes in T cells and sub-populations after pcMSC treatment. (A) Total lymphocyte count changes after Days 0, 4, and 8 of treatment. (B) The proportions of CD4, CD8, NKT, and Treg cells before and after treatment, compared with normal healthy individuals. (C) Distribution of T cells subpopulations was shown in the heat map. (D) The absolute cell number of total CD4+ T cell, naïve CD4+ T cell, and CD4+ memory cell increased after treatment. (E) No change in absolute cell number of CD8+ T cell, CD8+ Naïve T cell, or CD8+ memory T cell. (F) The absolute cell number of Treg cells was significantly increased after Day 8 of treatment. The P value was as indicated. Nonparametric Wilcoxon sign-rank test.
Figure 5
Figure 5
Changes of B cell populations after pcMSC treatment. (A) High proportion of IgM+ switched memory B cells and CD27+CD38+ plasma/plasmablast cells in the peripheral blood, compared to healthy controls, before and after treatment. Treatment with pcMSCs increased the proportions of CD27+ switched activated B cell and decreased proportion of plasma/plasmablast cells. (B) The absolute number of B cells, CD19+ total B cell count, CD19+ Naïve B cells count, and CD27+ switched B cell were significantly increased after D4 and D8 of treatment. The absolute number of Plasma/Plasmablast cell was significantly reduced after Day 8 of treatment. The P value was as indicated. Nonparametric Wilcoxon sign-rank test.
Figure 6
Figure 6
The changes of monocyte and NK cell population after pcMSC treatment. (A) The proportion and (B) the absolute number of total monocytes, classical monocytes, and non-classical monocytes were reduced after treatment. (C) The proportions of dendritic cells, NKT cells, and subpopulations were not significantly changed after treatment, compared with corresponding healthy controls. (D) The absolute number of dendritic cells, but not NK cells, or subpopulations, increased after Day 8 of treatment. *P <0.05 and **P <0.01, nonparametric Wilcoxon sign-rank test.

References

    1. Hu B, Guo H, Zhou P, Shi ZL. Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol (2021) 19(3):141–54. doi: 10.1038/s41579-020-00459-7
    1. Wu Z, McGoogan JM. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72314 Cases From the Chinese Center for Disease Control and Prevention. JAMA (2020) 323(13):1239–42. doi: 10.1001/jama.2020.2648
    1. Bhatraju PK, Ghassemieh BJ, Nichols M, Kim R, Jerome KR, Nalla AK, et al. . Covid-19 in Critically Ill Patients in the Seattle Region - Case Series. N Engl J Med (2020) 382(21):2012–22. doi: 10.1056/NEJMoa2004500
    1. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, et al. . Clinical Course and Outcomes of Critically Ill Patients With SARS-CoV-2 Pneumonia in Wuhan, China: A Single-Centered, Retrospective, Observational Study. Lancet Respir Med (2020) 8(5):475–81. doi: 10.1016/S2213-2600(20)30079-5
    1. Mathew D, Giles JR, Baxter AE, Oldridge DA, Greenplate AR, Wu JE, et al. . Deep Immune Profiling of COVID-19 Patients Reveals Distinct Immunotypes With Therapeutic Implications. Science (2020) 369(6508):eabc8511. doi: 10.1126/science.abc8511
    1. Yang L, Xie X, Tu Z, Fu J, Xu D, Zhou Y. The Signal Pathways and Treatment of Cytokine Storm in COVID-19. Signal Transduct Target Ther (2021) 6(1):255. doi: 10.1038/s41392-021-00679-0
    1. Group RC, Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, et al. . Dexamethasone in Hospitalized Patients With Covid-19. N Engl J Med (2021) 384(8):693–704. doi: 10.1056/NEJMoa2021436
    1. Group RC. Tocilizumab in Patients Admitted to Hospital With COVID-19 (RECOVERY): A Randomised, Controlled, Open-Label, Platform Trial. Lancet (2021) 397(10285):1637–45. doi: 10.1016/S0140-6736(21)00676-0
    1. Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V, et al. . Baricitinib Plus Remdesivir for Hospitalized Adults With Covid-19. N Engl J Med (2021) 384(9):795–807. doi: 10.1056/NEJMoa2031994
    1. Arentz M, Yim E, Klaff L, Lokhandwala S, Riedo FX, Chong M, et al. . Characteristics and Outcomes of 21 Critically Ill Patients With COVID-19 in Washington State. JAMA (2020) 323(16):1612–4. doi: 10.1001/jama.2020.4326
    1. Thompson M, Mei SHJ, Wolfe D, Champagne J, Fergusson D, Stewart DJ, et al. . Cell Therapy With Intravascular Administration of Mesenchymal Stromal Cells Continues to Appear Safe: An Updated Systematic Review and Meta-Analysis. EClinicalMedicine (2020) 19:100249. doi: 10.1016/j.eclinm.2019.100249
    1. Zhuang WZ, Lin YH, Su LJ, Wu MS, Jeng HY, Chang HC, et al. . Mesenchymal Stem/Stromal Cell-Based Therapy: Mechanism, Systemic Safety and Biodistribution for Precision Clinical Applications. J BioMed Sci (2021) 28(1):28. doi: 10.1186/s12929-021-00725-7
    1. de Witte SFH, Luk F, Sierra Parraga JM, Gargesha M, Merino A, Korevaar SS, et al. . Immunomodulation By Therapeutic Mesenchymal Stromal Cells (MSC) Is Triggered Through Phagocytosis of MSC By Monocytic Cells. Stem Cells (2018) 36(4):602–15. doi: 10.1002/stem.2779
    1. Ge W, Jiang J, Arp J, Liu W, Garcia B, Wang H. Regulatory T-Cell Generation and Kidney Allograft Tolerance Induced by Mesenchymal Stem Cells Associated With Indoleamine 2,3-Dioxygenase Expression. Transplantation (2010) 90(12):1312–20. doi: 10.1097/TP.0b013e3181fed001
    1. Luz-Crawford P, Djouad F, Toupet K, Bony C, Franquesa M, Hoogduijn MJ, et al. . Mesenchymal Stem Cell-Derived Interleukin 1 Receptor Antagonist Promotes Macrophage Polarization and Inhibits B Cell Differentiation. Stem Cells (2016) 34(2):483–92. doi: 10.1002/stem.2254
    1. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. . Bone Marrow Stromal Cells Attenuate Sepsis via Prostaglandin E(2)-Dependent Reprogramming of Host Macrophages to Increase Their Interleukin-10 Production. Nat Med (2009) 15(1):42–9. doi: 10.1038/nm.1905
    1. Vignali DA, Collison LW, Workman CJ. How Regulatory T Cells Work. Nat Rev Immunol (2008) 8(7):523–32. doi: 10.1038/nri2343
    1. Harrell CR, Sadikot R, Pascual J, Fellabaum C, Jankovic MG, Jovicic N, et al. . Mesenchymal Stem Cell-Based Therapy of Inflammatory Lung Diseases: Current Understanding and Future Perspectives. Stem Cells Int (2019) 2019:4236973. doi: 10.1155/2019/4236973
    1. Singh S, Chakravarty T, Chen P, Akhmerov A, Falk J, Friedman O, et al. . Allogeneic Cardiosphere-Derived Cells (CAP-1002) in Critically Ill COVID-19 Patients: Compassionate-Use Case Series. Basic Res Cardiol (2020) 115(4):36. doi: 10.1007/s00395-020-0795-1
    1. Leng Z, Zhu R, Hou W, Feng Y, Yang Y, Han Q, et al. . Transplantation of ACE2(-) Mesenchymal Stem Cells Improves the Outcome of Patients With COVID-19 Pneumonia. Aging Dis (2020) 11(2):216–28. doi: 10.14336/AD.2020.0228
    1. Arabpour E, Khoshdel S, Tabatabaie N, Akhgarzad A, Zangiabadian M, Nasiri MJ. Stem Cells Therapy for COVID-19: A Systematic Review and Meta-Analysis. Front Med (Lausanne) (2021) 8:737590. doi: 10.3389/fmed.2021.737590
    1. Mahendiratta S, Bansal S, Sarma P, Kumar H, Choudhary G, Kumar S, et al. . Stem Cell Therapy in COVID-19: Pooled Evidence From SARS-CoV-2, SARS-CoV, MERS-CoV and ARDS: A Systematic Review. BioMed Pharmacother (2021) 137:111300. doi: 10.1016/j.biopha.2021.111300
    1. Zanirati G, Provenzi L, Libermann LL, Bizotto SC, Ghilardi IM, Marinowic DR, et al. . Stem Cell-Based Therapy for COVID-19 and ARDS: A Systematic Review. NPJ Regener Med (2021) 6(1):73. doi: 10.1038/s41536-021-00181-9
    1. Li Y, Li H, Cao Y, Wu F, Ma W, Wang Y, et al. . Placentaderived Mesenchymal Stem Cells Improve Airway Hyperresponsiveness and Inflammation in Asthmatic Rats by Modulating the Th17/Treg Balance. Mol Med Rep (2017) 16(6):8137–45. doi: 10.3892/mmr.2017.7605
    1. Li Y, Qu T, Tian L, Han T, Jin Y, Wang Y. Human Placenta Mesenchymal Stem Cells Suppress Airway Inflammation in Asthmatic Rats by Modulating Notch Signaling. Mol Med Rep (2018) 17(4):5336–43. doi: 10.3892/mmr.2018.8462
    1. Richardson S, Hirsch JS, Narasimhan M, Crawford JM, McGinn T, Davidson KW, et al. . Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized With COVID-19 in the New York City Area. JAMA (2020) 323(20):2052–9. doi: 10.1001/jama.2020.6775
    1. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. . Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA (2020) 323(11):1061–9. doi: 10.1001/jama.2020.1585
    1. Guo YR, Cao QD, Hong ZS, Tan YY, Chen SD, Jin HJ, et al. . The Origin, Transmission and Clinical Therapies on Coronavirus Disease 2019 (COVID-19) Outbreak - an Update on the Status. Mil Med Res (2020) 7(1):11. doi: 10.1186/s40779-020-00240-0
    1. Henry BM, Aggarwal G, Wong J, Benoit S, Vikse J, Plebani M, et al. . Lactate Dehydrogenase Levels Predict Coronavirus Disease 2019 (COVID-19) Severity and Mortality: A Pooled Analysis. Am J Emerg Med (2020) 38(9):1722–6. doi: 10.1016/j.ajem.2020.05.073
    1. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ, et al. . COVID-19: Consider Cytokine Storm Syndromes and Immunosuppression. Lancet (2020) 395(10229):1033–4. doi: 10.1016/S0140-6736(20)30628-0
    1. Ruddell RG, Hoang-Le D, Barwood JM, Rutherford PS, Piva TJ, Watters DJ, et al. . Ferritin Functions as a Proinflammatory Cytokine via Iron-Independent Protein Kinase C Zeta/Nuclear Factor kappaB-Regulated Signaling in Rat Hepatic Stellate Cells. Hepatology (2009) 49(3):887–900. doi: 10.1002/hep.22716
    1. Wu Y, Hoogduijn MJ, Baan CC, Korevaar SS, de Kuiper R, Yan L, et al. . Adipose Tissue-Derived Mesenchymal Stem Cells Have a Heterogenic Cytokine Secretion Profile. Stem Cells Int (2017) 2017:4960831. doi: 10.1155/2017/4960831
    1. Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal Stem Cells Inhibit Natural Killer-Cell Proliferation, Cytotoxicity, and Cytokine Production: Role of Indoleamine 2,3-Dioxygenase and Prostaglandin E2. Blood (2008) 111(3):1327–33. doi: 10.1182/blood-2007-02-074997
    1. McKechnie JL, Blish CA. The Innate Immune System: Fighting on the Front Lines or Fanning the Flames of COVID-19? Cell Host Microbe (2020) 27(6):863–9. doi: 10.1016/j.chom.2020.05.009
    1. Zhou Y, Fu B, Zheng X, Wang D, Zhao C, Qi Y, et al. . Pathogenic T-Cells and Inflammatory Monocytes Incite Inflammatory Storms in Severe COVID-19 Patients. Natl Sci Rev (2020) 7(6):998–1002. doi: 10.1093/nsr/nwaa041
    1. Zhao Q, Meng M, Kumar R, Wu Y, Huang J, Deng Y, et al. . Lymphopenia Is Associated With Severe Coronavirus Disease 2019 (COVID-19) Infections: A Systemic Review and Meta-Analysis. Int J Infect Dis (2020) 96:131–5. doi: 10.1016/j.ijid.2020.04.086
    1. Lu C, Liu Y, Chen B, Yang H, Hu H, Liu Y, et al. . Prognostic Value of Lymphocyte Count in Severe COVID-19 Patients With Corticosteroid Treatment. Signal Transduct Target Ther (2021) 6(1):106. doi: 10.1038/s41392-021-00517-3
    1. Huang G, Kovalic AJ, Graber CJ. Prognostic Value of Leukocytosis and Lymphopenia for Coronavirus Disease Severity. Emerg Infect Dis (2020) 26(8):1839–41. doi: 10.3201/eid2608.201160
    1. Jagadeesh A, Prathyusha AMVN, Sheela GM, Bramhachari PV. “T Cells in Viral Infections: The Myriad Flavours of Antiviral Immunity”. in: Dynamics of Immune Activation in Viral Diseases. (2020) pp. 139–48. doi: 10.1007/978-981-15-1045-8_9
    1. Sun J, Zhuang Z, Zheng J, Li K, Wong RL, Liu D, et al. . Generation of a Broadly Useful Model for COVID-19 Pathogenesis, Vaccination, and Treatment. Cell (2020) 182(3):734–43 e5. doi: 10.1016/j.cell.2020.06.010
    1. Mahmoudi S, Rezaei M, Mansouri N, Marjani M, Mansouri D. Immunologic Features in Coronavirus Disease 2019: Functional Exhaustion of T Cells and Cytokine Storm. J Clin Immunol (2020) 40(7):974–6. doi: 10.1007/s10875-020-00824-4
    1. Aggarwal S, Pittenger MF. Human Mesenchymal Stem Cells Modulate Allogeneic Immune Cell Responses. Blood (2005) 105(4):1815–22. doi: 10.1182/blood-2004-04-1559
    1. Engela AU, Hoogduijn MJ, Boer K, Litjens NH, Betjes MG, Weimar W, et al. . Human Adipose-Tissue Derived Mesenchymal Stem Cells Induce Functional De-Novo Regulatory T Cells With Methylated FOXP3 Gene DNA. Clin Exp Immunol (2013) 173(2):343–54. doi: 10.1111/cei.12120
    1. Khosravi M, Karimi MH, Hossein Aghdaie M, Kalani M, Naserian S, Bidmeshkipour A. Mesenchymal Stem Cells can Induce Regulatory T Cells via Modulating miR-126a But Not miR-10a. Gene (2017) 627:327–36. doi: 10.1016/j.gene.2017.06.012
    1. Bai L, Lennon DP, Eaton V, Maier K, Caplan AI, Miller SD, et al. . Human Bone Marrow-Derived Mesenchymal Stem Cells Induce Th2-Polarized Immune Response and Promote Endogenous Repair in Animal Models of Multiple Sclerosis. Glia (2009) 57(11):1192–203. doi: 10.1002/glia.20841
    1. McMahan K, Yu J, Mercado NB, Loos C, Tostanoski LH, Chandrashekar A, et al. . Correlates of Protection Against SARS-CoV-2 in Rhesus Macaques. Nature (2021) 590(7847):630–4. doi: 10.1038/s41586-020-03041-6
    1. Wang F, Nie J, Wang H, Zhao Q, Xiong Y, Deng L, et al. . Characteristics of Peripheral Lymphocyte Subset Alteration in COVID-19 Pneumonia. J Infect Dis (2020) 221(11):1762–9. doi: 10.1093/infdis/jiaa150
    1. Deng Z, Zhang M, Zhu T, Zhili N, Liu Z, Xiang R, et al. . Dynamic Changes in Peripheral Blood Lymphocyte Subsets in Adult Patients With COVID-19. Int J Infect Dis (2020) 98:353–8. doi: 10.1016/j.ijid.2020.07.003
    1. Rydyznski Moderbacher C, Ramirez SI, Dan JM, Grifoni A, Hastie KM, Weiskopf D, et al. . Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations With Age and Disease Severity. Cell (2020) 183(4):996–1012.e19. doi: 10.1016/j.cell.2020.09.038
    1. Rodda LB, Netland J, Shehata L, Pruner KB, Morawski PA, Thouvenel CD, et al. . Functional SARS-CoV-2-Specific Immune Memory Persists After Mild COVID-19. Cell (2021) 184(1):169–83.e17. doi: 10.1016/j.cell.2020.11.029

Source: PubMed

3
Iratkozz fel