Stem Cells: A Promising Therapeutic Target for COVID-19

Abstract

A large variety of therapeutic products have been developed as vaccines or treatments of COVID-19 infection. Meanwhile SARS-CoV-2 continues to mutate and spread. The severe COVID-19 patients have limited treatment options. Therefore, mesenchymal stem cells (MSCs) are proposed as another therapeutic target. They are known for their regenerative and immunomodulatory properties. They showed potent efficacy and safety abilities for the treatment of critical COVID-19 patients and intriguing benefits in reducing lungs damage, mortality, and cytokine storm in several clinical studies. Another promising treatment option is the use of dental pulp stem cells (DPSCs) to combat SARS-CoV-2, despite the fact that detailed therapeutic strategies and mechanisms are still lacking. In this review we shed light on the immune response against COVID-19 infection and the relevant knowledge considering the role of MSCs in innate and adaptive immune responses to better understand the immune-physiological mechanisms of MSCs treatment. Lastly, we summarize the latest progress in clinical and research studies suggesting potential MSCs/DPSCs as therapeutic targets in battling COVID-19.

Share and Cite:

Hardin, L. and Xiao, N. (2022) Stem Cells: A Promising Therapeutic Target for COVID-19. Stem Cell Discovery, 12, 1-27. doi: 10.4236/scd.2022.121001.

1. Introduction

Although the COVID-19 pandemic began over two years ago and the pathological mechanisms of SARS-CoV-2 have sufficiently been revealed, there is no current effective treatment [1] [2]. The virus provokes cytokine storm [3], in which hyperinflammation and severe multiorgan failure were observed through excessive cytokine release from uncontrolled immune activation [4]. Stem cells therapy is one of the numerous therapeutic avenues used to save people’s lives especially in severe COVID-19 cases [5]. MSCs are stromal cells that originated from different tissues such as bone marrow, fat, dental tissue, umbilical cord, placenta, and many other sources [6] [7] [8] [9]. MSCs possess immunomodulatory capabilities as well as their endowed regenerative ability and multipotency [8] [10]. After infecting the host cells, SARS-CoV-2 showed ability to overcome the host immune system [11]. MSC therapies were showed to be a fruitful avenue to treat COVID-19 infection [3] [12] [13]. Hence, several MSCs therapy clinical studies (clinical trials, patient case reports) have been reported. An impressing decrease of inflammatory cytokines was observed few days after administration of the MSCs, in addition to a decrease of lung damage and time to recovery [2] [3]. Moreover, a great survival rate and MSCs tolerance were reported [14] [15] [16] [17] [18]. DPSCs are MSCs isolated from the dental pulps. They share many similarities with bone marrow MSCs and present many unique proprieties such as the easy isolation, great proliferation and differentiation potentials, and immunomodulatory activity [19] [20] [21]. In addition, they are characterized by a low immunogenicity. All these properties make them great candidates for the treatment of COVID-19. DPSCs have been shown to be efficient as a potential therapy for severe COVID-19 and they have shown a great curative potential despite the lack of a full understanding of the mechanisms [22] [23]. DPSCs hold promising immunomodulatory potential that might enhance their medical use in infectious diseases and inflammation [24].

Standardizing MSC use and MSC derivatives protocols are fundamental [2]. In order to determine the cell-therapy products, cells, tissues, or products derived from cells and tissues are categorized as pharmaceutical methods by the US Food and Drug Administration (US FDA) [2] [25]. The umbilical cord blood is the sole stem cell product approved for use in patients who have blood production problems [2] [26].

Immuno-regulators agents seemed to play a key role in fighting COVID-19 infection. With more reports and ongoing research revealing the possible viral pathological pathways and virus-host interactions, mechanism of therapeutic strategies could be better elucidated, and alternative treatments are being investigated. Some studies suggest that the immunomodulatory potential of MSCs is particularly paracrine and it results from MSC-derived exosome which could be secreted as extracellular vesicles (EVs) through paracrine pathway [27] [28]. The exosomes contain a variety of cargos, including messenger ribonucleic acids (mRNAs), DNA, lipids, growth factors, carbohydrates, and miRNAs [29] [30] [31].

This paper discusses the plausible MSCs as well as DPSCs immuno-modulatory potentials to suppress COVID-19 infection. Also, we particularly review the recent progress in MSC therapy in clinical and research studies.

2. Immune Response against COVID-19

When SARS-CoV-2 invades the host cell, the immune system tries to eliminate the virus through the innate and acquired immunity [32] (Figure 1).

2.1. Innate Immune Response

SARS-CoV-2 cell entry is insured by many cellular players such as ACE2, TMP-RSS2, integrins, neuropilins 1, DC/L-SIGN, and C-type lectins mediated endocytosis [33] [34] [35] [36]. It is reported that viral mRNA is recognized by Toll-like receptor (TLR) which triggers transcriptional responses and cytoplasmic changes that generate the induction of the Nod-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome [32]. NLRP3 is an intracellular pattern recognition receptor (PRR) sensor that recognizes a large variety of microbial motifs, intracellular danger signals and extracellular stressors [37]. Activated NLRP3 then recruits adaptor and effector proteins to form inflammasome and provokes caspase 1-mediated secretion of the pro-inflammatory cytokines IL-1β and IL-18, and gasdermin [37] [38] [39], which further induces pyroptosis [40], inflammation [41] and coagulopathy [42].

The virus hijacks the immunity actors and triggers a cytokine storm [43] [44] [45] [46] [47]. These chemokines are chemotactic cytokines that play a major role in controlling the migration, the positioning of immune cells in tissues, and the selective recruitment of innate immune effectors [48]. Stimulated macrophages release tumor necrosis factor α (TNF-α) and interleukin-1β (IL-1β) which activates the innate immune system and triggers the acute inflammatory response immediately following infection. Interferon-γ(IFN-γ) is

Figure 1. Innate and adaptative immune response of MSCs to COVID-19.

essentially secreted by activated T cells and natural killer (NK) cells, and its principal roles are the stimulation of the macrophage population, the achievement of antigen presentation and the mobilization of the innate immune system and the antimicrobial immunity [49]. A high plasma level of interferon IFN-γwas noticed in COVID-19 patients [50]. Also, a significant increase in plasma levels of the inflammatory interleukins (IL-1β, IL-8, and TNF-α) and the anti-inflammatory interleukins (IL-6, IL-10) was reported [51] [52]. Besides, the monocyte chemoattractant peptide (MCP)-1, which is a key player in monocytes/macrophages recruitment [53], was found with high concentration in COVID-19 patients [54]. Similarly, granulocytes colony stimulating factor (GCSF), MIP)-1A and MIP-1B, and MIP were found to be abundant in plasma of COVID-19 which stimulates the “emergency production” of neutrophils that occurs in response to acute injury or infection [52].

SARS-CoV-2 showed to trigger cell pyroptosis, which is a programmed cell death with pro-inflammatory cytokines [55]. Indeed, the high secretion of cytokines recruit different immune cells to the site needed [56], which partially reveals the lymphopenia especially in severe cases of COVID19 [57]. It appears that SARS-CoV-2 acts by differentially suppressing some cells and activating other cells, lymphocytes were found to decrease in opposite neutrophils showed to increase [52] [58].

2.2. Adaptive Immunity

Upon stimulation by the pathogen, T helper 1 (Th1) and NK cells secrete IFN-γ, which induces macrophages, activates antigen processing, and coordinates both innate and adaptive immune response [49]. The number of CD8+ T and NK cells was found to decrease but the cells were particularly induced. Cell lytic granules (perforin and granzyme) were greatly detected in some immune cells [56], which can impair the viral infection but also disrupt the cellular mechanisms [59].

CD4+ T and B cells numbers were not correlated to COVID-19 severity [60], but in the study of Wang et al. the B cells number remarkably decreased in severe and critical cases [61]. The study of Bobcakova et al. reported that memory B cell frequency was significantly reduced in asymptomatic and critical cases [62]. COVID-19 patients showed a low count of lymphocytes especially for patients with severe disease [63], and the counts of T cells, NK cells, T helper cells, cytotoxic T cells (also known as CD8+ cells), and Treg cells also decreased, which yield a reduction of their capabilities to battle the virus infection [64] [65].

SARS-CoV-2 humoral immune response is mediated by antibodies that are targeting viral surface glycoproteins, mainly S and N. As the coronavirus S glycoprotein is surface-exposed and insures entry into host cells, it represent essentially the most targetable protein the neutralizing antibodies [66]. The study of Sterlin et al. showed that IgA antibodies were detected earlier than other antibodies in COVID-19 disease [67]. Moreover, serum IgA showed to be more efficient in neutralizing the virus than IgG. IgM and IgA antibodies showed to be detectable early during the first week of symptoms appearance, but IgG showed to be identifiable at least two weeks from the appearance of symptoms. IgM seropositivity level was reported to decline after 4 weeks, whereas IgG and IgA seropositivity persisted at detectable level for more than eight weeks [68].

Determining IgA and IgG subclasses helps to understand the infection mechanisms. The study of Dogan et al., reported that the IgG1 subclass was the most dominant in COVID-19 cases, but other subclasses were detectable such as IgA and IgG2-4 at lower rates [69].

3. MSCs as a Potential Therapy for COVID-19

MSCs were discovered for the first time in mouse bone marrow [70], and have since then been found in many organs [71]. They have been extensively tested and proven effective for many disorders, and they have showed an advantageous potential to treat multiple diseases [72] [73] [74] [75].

MSCs can intervene in injury sites through different mechanisms: they can 1) differentiateinto different cell types [76], 2) repair the damaged cells through cell fusion [77], 3) release paracrine bioactive molecules such as growth and differentiation factors, and chemokines [78], 4) transfer organelles (e.g., mitochondria) and/or molecules through tunneling nanotubes (TNTs) [79], and 5) mediate the transfer of proteins, lipids, DNAs, mRNAs, non-coding RNAs (ncRNAs), carbohydrates, and growth factors by exosomes [80].

Recently, MSCs curative potential has been reported in fighting COVID-19 disease [31] [81] [82]. It was reported that MSCs, cells source was not disclosed in the publication, lacked ACE2 expression [14]. MSCs did not express ACE2 and transmembrane serine protease 2 (TMPRSS2), which is mainly expressed by endothelial cells in respiratory and digestive tracks and facilitates entry and activation of SARS-CoV-2 in host cells [14]. A different study also did not find expression of ACE2 and TMPRSS2 in fetal and adult human MSCs [83]. These results suggest that MSCs could be resistant to COVID-19 [84], which is another expectation from MSC therapeutic potential in battling the virus. Although ACE2 and TMPRSS2 are expressed in oral epithelia cells [85], there is limited report about their expression in DPSCs. Our preliminary unpublished data indicates low expression of ACE2 and TMPRSS2 compared to lung epithelia cells. More experiments are warranted to confirm the observation.

MSCs showed anti-inflammatory properties and they demonstrated to reduce the cytokine storm via paracrine way, and their activities are controlled by chemokines [86] [87]. They are able to decrease inflammation through the release of anti-inflammatory factors or the inactivation of some immune cells [88].

3.1. Potential Role of MSCs in Immune Response

MSCs are endowed with high immune modulatory properties. They communicate with many immune cells of innate or acquired responses, including macrophages, dendritic cells (DCs), T cells, B cells, and NK [89]. The components of the secretome of stem and immune cells represent the most important key players to decode the biological pathways of any disorder [90] and possibly COVID-19 infection as well [91].

3.1.1. Antigen Presentation

MSCs showed to suppress the migration and maturity of DCs [92]. MSCs suppressed antigen processing and presentation of DCs, differentiation, and maturation of DCs [93] [94]. In addition, MSCs showed to suppress monocyte differentiation into DCs by decreasing the presentation molecules signature and the cytokines secretion. When MSCs are exposed to TNF-α and IFN-γ, they stimulate the over-expression of anti-inflammatory factors essentially the prostaglandin E2 (PGE2) and the programmed death ligand 1 (PDL1), which inhibit the antigen processing by DCs and expansion of cytotoxic, helper, and regulatory T cells and reduce the evolved immune cells [95]. DPSCs, possess stem cell properties and they showed to suppress macrophage functions via the TNF-α/IDO axis, this provided a physiologically pertinent evidence for their innate immunomodulatory activity [96]. MSCs are capable of immune-suppressing the activity and cytokine secretion of neutrophils and macrophages [97] [98], consequently this may reduce the cytokine storm.

3.1.2. Phagocytosis

MSCs showed to modulate the activity of macrophages [99] [100], which exert both pro and anti-inflammatory properties by a direct cell-to-cell contact or by soluble mediators [101]. Chen et al. showed that BM-MSCs conditioned medium can suppress phagocytosis [102]. While another study reported that MSCs are able to induce macrophages phagocytosis [79]. In addition, MSCs exhibited a great induction potential to polarize in the polarization of macrophages to an anti-inflammatory profile [101] [103] [104] [105].

Increased concentrations of IL-10 were ensured by IL-6 and hepatocyte growth factor (HGF) secreted by MSCs [106]. IL-10 consequently inhibits the differentiation of monocytes to DCs but switches it to macrophages [107]. Liu et al. found released bodies by MSCs able to create a tissue repair through the generation of an anti-inflammatory profile [108]. The anti-inflammatory environment can reduce the inflammation caused by SARS-CoV2.

Another mechanism was employed by the menstrual blood-derived stromal cells (MenSCs) to exacerbate infections is by decreasing the migration of immune cells to the damaged site and suppressing immune clearance [109], this could reduce leukopenia observed in COVID-19. A recent study showed that the co-culture of DPSCs and macrophages decrease pro-inflammatory and increase anti-inflammatory cytokines from macrophages [110].

3.1.3. Apoptosis

MSCs showed to inhibit apoptosis of immune cells. Fadhi et al. evaluated the impact of human bone marrow mesenchymal stem cells (BMMSCs) on human immortalized myelogenous leukemia cell line K562 by co-culture [111]. In addition, Gu et al. found that MSCs decrease neural apoptosis and improve learning-memory function in hypoxic–ischemic brain damage (HIBD) rats by inhibiting the TLR2/NFκB signaling pathway [112]. DPSC-EVs showed to reduce cytotoxicity through anti-apoptotic mechanism by upregulating endogenous Bcl-2, and decrease the expression of the pro-apoptotic regulator Bax in Aβ peptide-exposed human neuroblastoma cells [113]. Bax was reported to be expressed in overnight-cultured neutrophils. However, upon co-culture with BMMSCs, the neutrophils showed a significant decrease of Bax expression. MCL-1, a well-known mitochondrial antiapoptotic protein, was upregulated in neutrophils after co-culture with MSCs compared with neutrophils cultured in medium alone [95]. As SARS-CoV-2 has showed to trigger cell pyroptosis [40], [55], the use of MSCs may help to decrease it.

3.1.4. NK Cells Cytotoxicity

NK cells are granular lymphocytes of the innate immune system, implicated in the microbe-infected cells and the immune surveillance of tumors. When stimulated, NK cells release multiple chemokines such as IFN-γ, MIP-1α, MIP-1β, TNF-α, GM-CSF, and chemokines (IL-5, IL-8, IL-10, IL-13) that can regulate other innate and adaptive immune cells [114] [115]. MSCs are considered as powerful inhibitors of natural killer cell proliferation [116] [117]. The co-culture of NK cells with MSCs seemed to impede the cell signaling through cytokines release [118]. Many cytokines (prostaglandin E2 (PGE2), TGF-β1, and IL-6) were secreted by MSCs and showed to hinder the NK cells properties [118] [119]. Along the same line, Sotiropoulou et al. found that MSCs showed to reduce the NK cells activation and secretion promoted by IL-2, this interaction sometimes requires direct cell contact, whereas soluble factors exhibited to play an important role, such as TGF-β1 and PGE2 [120]. DPSCs reported to impede NK cells proliferation and to promote apoptosis of activated NK cells also the cytotoxic capacity of NK cells showed to be notably reduced [121]. Severe COVID-19 resulted in an increase in armed NK cells containing high levels of cytotoxic proteins such as perforin [122]. The NK suppression could reduce the cytokine secretion and consequently the cytokine storm.

3.1.5. Cell Mediated Immune Response

Previous studies suggest that MSCs are not particularly capable to modulate the immune response, but they also induce the tolerance toward other transplanted cells [123] [124].

MSCs showed a spontaneous ability to suppress the expansion and function of T-cells (CD4+ and CD8+ T cell subsets) [125] [126] [127] [128] [129]. In inflammatory environment MSCs transform into suppressive cells to reduce the inflammation [130]. In the presence of DCs, MSCs lead the passage from pro-inflammatory Th1 to anti-inflammatory Th2 cells, consequently a secretion profiling modification toward anti-inflammation [131] [132] [133]. Moreover, this inhibition was dose dependent for both CD4+ and CD8+ T-cells. The 24 hours T-cells co-culture with MSCs showed to impact these T-cells activities [130]. MSCs showed to inhibit Th17 but not Treg differentiation [134]. MSCs induce an immunoregulation toward an anti-inflammatory response by promoting naive T cells into T cells with an anti-inflammatory Treg cells profile and by contributing to generate an immunosuppressive environment via the inhibition of proinflammatory T cells [134] [135] [136].

Moreover, MSCs can communicate with B cells and they affect the plasmablast differentiation also the expansion of reg B-reg cells, which possess an immune-suppressive activity facilitating the immunotolerance [103] [137]. MSCs regulate the proliferation in inflammatory environment, by inhibiting B cell expansion and differentiation by suppressing of transient receptor potential channels (TRP channels) [138], also secreting IL1RA inhibits B cell differentiation [139], and antibodies release of B cells without launching cell death [140].

DPSCs showed to activate compensatory pathways targeting to manage the inflammatory response by regulating pro-inflammatory cytokines in pre-activated T lymphocytes [24]. Another study confirmed that DPSCs can modulate the production of cytokines in vitro and suppress the proliferation of T and B cells [141], which could help to modulate SARS-CoV2 immune response by reducing the cytokine release and the cell migration to lungs and tissue damaging.

3.2. MSCs’s Clinical Use for COVID-19 Disease Treatment

3.2.1. MSCs-Cell Based Therapy

Since the identification of the new emerged SARS-CoV-2, more than 100 clinical trials have been submitted to ClinicalTrials.gov and the WHO Clinical Trials Registry Platform (WHO ICTRP) (https://clinicaltrials.gov) to investigate the potential treatment of MSCs and MSCs derived products. Most of these trials have not been yet published. Two clinical trials using dental pulp mesenchymal cells to treat COVID-19 (NCT04336254 and NCT04302519) were submitted to ClinicalTrails.gov, one of the studies is recruiting. However, most of published clinical interventions using intravenous or intramuscular MSCs treatment for both mild and critical types of COVID-19 cases showed improvement in clinical outcomes with some adverse reactions, and demonstrated that MSCs can play a major role in reducing the acute lung injury (ALI) and the respiratory distress syndrome (ARDS) [14] [15] [16] [81] [88] [142] (Table 1). Although the published MSCs therapies used stem cells from different sources (umbilical, placental, dental, menstrual, etc.), in different cells doses from one million to 5 × 107 cells given in one, two or three injections, different administration methods (intravenous and intramuscular) and also different infusion time, the MSCs treated patients showed promising pulmonary function improvement and decrease in deaths.

In all the studies, the cytokine storm started to be downregulated few days after MSCs administration. In the study of Leng et al. they noticed a cytokine storm’s alleviation in three to six days, but a significant improvement was observed in the pulmonary function and symptoms after 2 days from MSCs transplantation (one million cells) [14]. According to Shu et al., 2 × 106 of human

Table 1. Published MSCs clinical studies for COVID-19 treatment.

Abbreviations: CRP: C-reaction protein; CT: computed tomography; ICU: intensive care unit; MSCs: mesenchymal stem cells; PaO2/FiO2 ratio: ratio of arterial partial pressure of oxygen to fraction of inspired oxygen RR: respiration rate; SpO2: oxygen saturation. COVID-19 infection illness categories; Mild: showing various signs and symptoms of COVID-19 (e.g., fever, cough, sore throat, malaise, headache, muscle pain, nausea, vomiting, diarrhea, loss of taste and smell) but who do not have shortness of breath, dyspnea, or abnormal chest imaging; moderate: showing evidence of lower respiratory disease during clinical assessment or imaging and who have an oxygen saturation (SpO2) ≥ 94% on room air at sea level; severe: showing SpO2 < 94% on room air at sea level, a ratio PaO2/FiO2 < 300 mm Hg, respiratory frequency > 30 breaths/min, or lung infiltrates > 50%; critically ill: showing respiratory failure, septic shock, and/or multiple organ dysfunction [171].

umbilical cord mesenchymal stem cell (UCMSCs) per kilogram of weight intravenously administrated to 12 severe COVID-19 patients were able to reduce the CRP and IL-6 from day 3 of transplantation [88]. Multiple cytokines concentrations (IFN-γ, TNF-α, IL-18, IL-8, and MIP-1) noticeably decreased in less than two weeks. The administration of UCMSCs-treated subjects notably reduced the inflammatory cytokines at day 6. A remarkable patient survival rate and recovery time were noted. The use of UCMSC was safe according to this trial [17]. The transplantation of Wharton’s Jelly-derived MSCs to severe COVID-19 pneumonia patient in one dose improved breathing and decreased fever within two days of MSC administration [16].

Some adverse side effects have been reported like facial flushing and fever, allergic rash, liver and kidney failure, new cardia arrhythmia, and worsening hypoxemia. In the study of Lanzoni et al., the use of high dose of UCMSCs (108 cells) provoked one case of heart failure [17].

Interestingly, it was demonstrated that priming MSCs showed to be more efficient, Park et al. found that HGF-primed MSCs demonstrated a high regenerative potential and an elongated cells surviving [143]. In the submitted clinical trials, we noticed that no stimulating step was added to the therapeutic process. Several studies investigated the co-culture of dental pulp stem cells (DPSCs) and immune cells and their role in reducing the inflammation for COVID-19 patients and showed more anti-inflammatory benefits than DPSCs or immune cells cultured alone [23]. However, in resting conditions, MSCs do not have a high immunomodulatory activity, but when they are stimulated, they show their potential. This leads to the fact that the regulatory properties are not constitutively expressed by MSCs but are related to the process of priming [144].

Also, the preclinical study of Bustos et al. reported that BMMSCs treated with ARDS patients serum were more efficient to relieve pro-inflammatory cytokines secretion than unconditioned MSCs [145]. In another study, the activation of BMMSCs with same kind of serum led to a high secretion of IL-1RA and IL-10, reduced the release of inflammatory mediators, upregulated the TLR-4 and vascular endothelial growth factor (VEGF) genes, and launched an important immunoregulation through the prostaglandin E2 (PGE2) release [146].

There are obviously many limitations in the current studies. The small sample size to start with and the loss of treated subjects due to the severity of the COVID-19 during the studies. MSCs seem to be efficient and tolerable by patients in almost all the reported cases in COVID-19 therapy but long-term tracking and follow up information is pending. Future studies on larger pool of patients and long-term studies are required to evaluate the risks associated with MSC therapy, such mal-differentiation, immunosuppression, and instigation of malignant tumor growth [76].

Extracellular vesicles secretion represent another mechanism through it MSCs can regulate the immune response [147]. MSCs derived extracellular vesicles (MSC-EVs) contain heterogenous cargos such as proteins, lipids, DNAs, mRNAs, ncRNAs, carbohydrates, and growth factors [31] [148]. MSC-EVs are important for cell communication by acting as the delivers of the necessary factors of regeneration, antibacterial activity and immunomodulation [7] [149] [150] [151]. The most important advantages of using exosome-based therapy are 1) cell-free therapy without vascular obstruction, 2) low immunogenicity, and 3) ability to load content of miRNA in exosome [152] (Figure 2).

3.2.2. Promising Immunomodulatory and Regenerative Potential of DPSCs

Several clinical and research studies investigate the safety and efficacy of DPSCs in suppressing the cytokine storm and promoting injuries repairing in COVID-19 disease [22] [23] [153]. Likewise, accumulating evidence revealed the powerful properties of DPSCs. They are known for their immunomodulation and regenerative potential, they demonstrated to reduce inflammatory reactions in many auto-immune and inflammatory diseases [96] [154] [155] [156] and to induce tissues regeneration [110] [157] [158]. Moreover, DPSCs showed interesting regenerative and immunomodulatory properties when they are primed or co-cultured with other types of cells. TGF-β1 showed to induce DPSCs differentiation into functional pericyte-like cells [159]. The DPSCs also showed to secrete important amounts of prostaglandin E2, which is a key mediator for the anti-inflammatory activity and it contributes to the resolution of inflammation, and

Figure 2. Mechanisms of action of MSCs against COVID-19 infection. COVID-19 patient can develop serious cytokine storm and ARDS. Immune cells are activated, pro-inflammatory cytokines (IL-6, TNF-α, etc.) are over-released, lung cells are injured, and multiple organs fail. MSCs, through exosome, can induce an anti-inflammatory reaction and a cytokine storm reduction, a tissue regeneration, and an antimicrobial activity. Abbreviation: ARDS: acute respiratory distress syndrome; MSCs: mesenchymal stem cells.

they maintained their immunomodulatory activities even upon differentiation [160].

A study investigated the co-culture of DPSCs and immune cells and their role in reducing the inflammation for COVID-19 patients and showed more anti-inflammatory benefits than DPSCs or immune cells cultured separately [23]. The potent immunomodulatory functions of DPSCs due to their soluble factors and cytokines showed to play a crucial role in clinical cell therapy by T-lymphocyte function inhibition and upregulation of T cell regulatory (Tregs) stimulating immune tolerance [161] [162].

4. Conclusion

SARS-CoV-2 continues to grip the world with high infection rate, uncertain long haul symptoms in recovered patients, and an economic burden. Since the available vaccines do not protect from a re-infection with SARS-CoV-2, developing therapies to boost host immune systems to fight the virus infection and treat ALI and ARDS still seem to be of high priority. MSCs and especially DPSCs could be an interesting therapeutic candidate, since many phase-1 and 2 trials have shown promising results. However, consistent studies are still needed to reveal the whole MSCs therapeutic mechanism and to ensure their safety and efficacy.

Acknowledgements

This work was supported by Tobacco-Related Diseases Research Program Grant T30IP0917 from Regents of the University of California.

Funding Information

This work was supported by Tobacco-Related Diseases Research Program Grant T30IP0917 from Regents of the University of California.

Author Contributions

Conceptualization: Leyla Tahrani Hardin, Nan Xiao; Writing-original draft: Leyla Tahrani Hardin; Writing-review and editing: Leyla Tahrani Hardin, Nan Xiao; Validation: Nan Xiao; Supervision: Nan Xiao.

Conflict of Interest Statement

The authors declare that they have no conflicts of interest.

References

  1. 1. Spicer, A.J. and Jalkanen, S. (2021) Why Haven’t We Found an Effective Treatment for COVID-19? Frontiers in Immunology, 12, Article ID: 644850. https://doi.org/10.3389/fimmu.2021.644850

  2. 2. Shi, L., et al. (2021) Mesenchymal Stem Cell Therapy for Severe COVID-19. Signal Transduction and Targeted Therapy, 6, Article No. 339. https://doi.org/10.1038/s41392-021-00754-6

  3. 3. Yang, L., Xie, X., Tu, Z., Fu, J., Xu, D. and Zhou, Y. (2021) The Signal Pathways and Treatment of Cytokine Storm in COVID-19. Signal Transduction and Targeted Therapy, 6, Article No. 255. https://doi.org/10.1038/s41392-021-00679-0

  4. 4. Henderson, L.A., et al. (2020) On the Alert for Cytokine Storm: Immunopathology in COVID-19. Arthritis & Rheumatology, 72, 1059-1063. https://doi.org/10.1002/art.41285

  5. 5. Riedel, R.N., Pérez-Pérez, A., Sánchez-Margalet, V., Varone, C.L. and Maymó, J.L. (2021) Stem Cells and COVID-19: Are the Human Amniotic Cells a New Hope for Therapies against the SARS-CoV-2 Virus? Stem Cell Research & Therapy, 12, Article No. 155. https://doi.org/10.1186/s13287-021-02216-w

  6. 6. Dominici, M., et al. (2006) Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells. The International Society for Cellular Therapy Position Statement. Cytotherapy, 8, 315-317. https://doi.org/10.1080/14653240600855905

  7. 7. Durand, N., Mallea, J. and Zubair, A.C. (2020) Insights into the Use of Mesenchymal Stem Cells in COVID-19 Mediated Acute Respiratory Failure. npj Regenerative Medicine, 5, Article No. 17. https://doi.org/10.1038/s41536-020-00105-z

  8. 8. Gao, F., et al. (2016) Mesenchymal Stem Cells and Immunomodulation: Current Status and Future Prospects. Cell Death & Disease, 7, e2062. https://doi.org/10.1038/cddis.2015.327

  9. 9. Xu, R., Feng, Z. and Wang, F.-S. (2022) Mesenchymal Stem Cell Treatment for COVID-19. eBioMedicine, 77, Article ID: 103920. https://doi.org/10.1016/j.ebiom.2022.103920

  10. 10. Kean, T.J., Lin, P., Caplan, A.I. and Dennis, J.E. (2013) MSCs: Delivery Routes and Engraftment, Cell-Targeting Strategies, and Immune Modulation. Stem Cells International, 2013, e732742. https://doi.org/10.1155/2013/732742

  11. 11. Deng, Y., et al. (2020) Clinical Characteristics of Fatal and Recovered Cases of Coronavirus Disease 2019 in Wuhan, China: A Retrospective Study. Chinese Medical Journal, 133, 1261-1267. https://doi.org/10.1097/CM9.0000000000000824

  12. 12. Moll, G., Drzeniek, N., Kamhieh-Milz, J., Geissler, S., Volk, H.-D. and Reinke, P. (2020) MSC Therapies for COVID-19: Importance of Patient Coagulopathy, Thromboprophylaxis, Cell Product Quality and Mode of Delivery for Treatment Safety and Efficacy. Frontiers in Immunology, 11, Article 1091. https://doi.org/10.3389/fimmu.2020.01091

  13. 13. Yao, W., Shi, L., Zhang, Y., Dong, H. and Zhang, Y. (2022) Mesenchymal Stem/Stromal Cell Therapy for COVID-19 Pneumonia: Potential Mechanisms, Current Clinical Evidence, and Future Perspectives. Stem Cell Research & Therapy, 13, Article No. 124. https://doi.org/10.1186/s13287-022-02810-6

  14. 14. Leng, Z., et al. (2020) Transplantation of ACE2- Mesenchymal Stem Cells Improves the Outcome of Patients with COVID-19 Pneumonia. Aging and Disease, 11, 216-228.

  15. 15. Meng, F., et al. (2020) Human Umbilical Cord-Derived Mesenchymal Stem Cell Therapy in Patients with COVID-19: A Phase 1 Clinical Trial. Signal Transduction and Targeted Therapy, 5, Article No. 172. https://doi.org/10.1038/s41392-020-00286-5

  16. 16. Zhang, Y., et al. (2020) Intravenous Infusion of Human Umbilical Cord Wharton’s Jelly-Derived Mesenchymal Stem Cells as a Potential Treatment for Patients with COVID-19 Pneumonia. Stem Cell Research & Therapy, 11, Article No. 207. https://doi.org/10.1186/s13287-020-01725-4

  17. 17. Lanzoni, G., et al. (2021) Umbilical Cord Mesenchymal Stem Cells for COVID-19 Acute Respiratory Distress Syndrome: A Double-Blind, Phase 1/2a, Randomized Controlled Trial. STEM CELLS Translational Medicine, 10, 660-673. https://doi.org/10.1002/sctm.20-0472

  18. 18. Adugna, D.G. (2021) Current Clinical Application of Mesenchymal Stem Cells in the Treatment of Severe COVID-19 Patients: Review. Stem Cells and Cloning: Advances and Applications, 14, 71-80. https://doi.org/10.2147/SCCAA.S333800

  19. 19. Iohara, K., Zheng, L., Ito, M., Tomokiyo, A., Matsushita, K. and Nakashima, M. (2006) Side Population Cells Isolated from Porcine Dental Pulp Tissue with Self-Renewal and Multipotency for Dentinogenesis, Chondrogenesis, Adipogenesis, and Neurogenesis. Stem Cells, 24, 2493-2503. https://doi.org/10.1634/stemcells.2006-0161

  20. 20. Andrukhov, O., Behm, C., Blufstein, A. and Rausch-Fan, X. (2019) Immunomodulatory Properties of Dental Tissue-Derived Mesenchymal Stem Cells: Implication in Disease and Tissue Regeneration. World Journal of Stem Cells, 11, 604-617. https://doi.org/10.4252/wjsc.v11.i9.604

  21. 21. Tsutsui, T.W. (2020) Dental Pulp Stem Cells: Advances to Applications. Stem Cells and Cloning: Advances and Applications, 13, 33-42. https://doi.org/10.2147/SCCAA.S166759

  22. 22. Ye, Q., et al. (2020) Safety and Efficacy Assessment of Allogeneic Human Dental Pulp Stem Cells to Treat Patients with Severe COVID-19: Structured Summary of a Study Protocol for a Randomized Controlled Trial (Phase I/II). Trials, 21, Article No. 520. https://doi.org/10.1186/s13063-020-04380-5

  23. 23. Croci, S., et al. (2021) Human Dental Pulp Stem Cells Modulate Cytokine Production in vitro by Peripheral Blood Mononuclear Cells from Coronavirus Disease 2019 Patients. Frontiers in Cell and Developmental Biology, 8, Article ID: 609204. https://doi.org/10.3389/fcell.2020.609204

  24. 24. Di Tinco, R., et al. (2021) Role of PD-L1 in Licensing immunoregulatory Function of Dental Pulp Mesenchymal Stem Cells. Stem Cell Research & Therapy, 12, Article No. 598. https://doi.org/10.1186/s13287-021-02664-4

  25. 25. Part 15 Hearing: Draft Guidances Relating to the Regulation of human Cells, Tissues, or Cellular or Tissue-Based Products. 277.

  26. 26. Center for Biologics Evaluation and Research, US Food and Drug Administration (2020) Consumer Alert on Regenerative Medicine Products Including Stem Cells and Exosomes. FDA, July. https://www.fda.gov/vaccines-blood-biologics/consumers-biologics/consumer-alert-regenerative-medicine-products-including-stem-cells-and-exosomes

  27. 27. Bartel, D.P. (2009) MicroRNAs: Target Recognition and Regulatory Functions. Cell, 136, 215-233. https://doi.org/10.1016/j.cell.2009.01.002

  28. 28. Bartel, D.P. (2018) Metazoan MicroRNAs. Cell, 173, 20-51. https://doi.org/10.1016/j.cell.2018.03.006

  29. 29. Bulut, Ö. and Gürsel, İ. (2020) Mesenchymal Stem Cell Derived Extracellular Vesicles: Promising Immunomodulators against Autoimmune, Autoinflammatory Disorders and SARS-CoV-2 Infection. Turkish Journal of Biology, 44, Article 14. https://doi.org/10.3906/biy-2002-79

  30. 30. Öztürk, S., Elçin, A.E. and Elçin, Y.M. (2020) Mesenchymal Stem Cells for Coronavirus (COVID-19)-Induced Pneumonia: Revisiting the Paracrine Hypothesis with New Hopes? Aging and Disease, 11, 477-479. https://doi.org/10.14336/AD.2020.0403

  31. 31. Schultz, I.C., Bertoni, A.P.S. and Wink, M.R. (2021) Mesenchymal Stem Cell-Derived Extracellular Vesicles Carrying miRNA as a Potential Multi Target Therapy to COVID-19: An in Silico Analysis. Stem Cell Reviews and Reports, 17, 341-356. https://doi.org/10.1007/s12015-021-10122-0

  32. 32. Brodin, P. (2021) Immune Determinants of COVID-19 Disease Presentation and Severity. Nature Medicine, 27, 28-33. https://doi.org/10.1038/s41591-020-01202-8

  33. 33. Liu, J., Lu, F., Chen, Y., Plow, E. and Qin, J. (2022) Integrin Mediates Cell Entry of the SARS-CoV-2 Virus Independent of Cellular Receptor ACE2. Journal of Biological Chemistry, 298, Article ID: 101710. https://doi.org/10.1016/j.jbc.2022.101710

  34. 34. Cantuti-Castelvetri, L., et al. (2020) Neuropilin-1 Facilitates SARS-CoV-2 Cell Entry and Infectivity. Science, 370, 856-860. https://doi.org/10.1126/science.abd2985

  35. 35. Hoffmann, M., Kleine-Weber, H., et al. (2020) SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell, 181, 271-280.E8. https://doi.org/10.1016/j.cell.2020.02.052

  36. 36. Jackson, C.B., Farzan, M., Chen, B. and Choe, H. (2022) Mechanisms of SARS-CoV-2 Entry into Cells. Nature Reviews Molecular Cell Biology, 23, 3-20. https://doi.org/10.1038/s41580-021-00418-x

  37. 37. Swanson, K.V., Deng, M. and Ting, J.P.-Y. (2019) The NLRP3 Inflammasome: Molecular Activation and Regulation to Therapeutics. Nature Reviews Immunology, 19, 477-489. https://doi.org/10.1038/s41577-019-0165-0

  38. 38. Man, S.M., Karki, R. and Kanneganti, T.-D. (2017) Molecular Mechanisms and Functions of Pyroptosis, Inflammatory Caspases and Inflammasomes in Infectious Diseases. Immunological Reviews, 277, 61-75. https://doi.org/10.1111/imr.12534

  39. 39. Garay-Sevilla, M.E., Rojas, A., Portero-Otin, M. and Uribarri, J. (2021) Dietary AGEs as Exogenous Boosters of Inflammation. Nutrients, 13, Article 2802. https://doi.org/10.3390/nu13082802

  40. 40. Ferreira, A.C., et al. (2021) SARS-CoV-2 Engages Inflammasome and Pyroptosis in Human Primary Monocytes. Cell Death Discovery, 7, Article No. 116. https://doi.org/10.1038/s41420-021-00477-1

  41. 41. Marchetti, C., Mould, K., Tengesdal, I.W., Janssen, W.J. and Dinarello, C.A. (2021) Targeting of the NLRP3 Inflammasome for Early COVID-19. bioRxiv. https://doi.org/10.1101/2021.02.24.432734

  42. 42. de Rivero Vaccari, J.C., Dietrich, W.D., Keane, R.W. and de Rivero Vaccari, J.P. (2020) The Inflammasome in Times of COVID-19. Frontiers in Immunology, 11, Article ID: 583373. https://doi.org/10.3389/fimmu.2020.583373

  43. 43. Cron, R.Q. (2021) COVID-19 Cytokine Storm: Targeting the Appropriate Cytokine. The Lancet Rheumatology, 3, e236-e237. https://doi.org/10.1016/S2665-9913(21)00011-4

  44. 44. Fajgenbaum D.C. and June, C.H. (2020) Cytokine Storm. New England Journal of Medicine, 383, 2255-2273. https://doi.org/10.1056/NEJMra2026131

  45. 45. Hojyo, S., et al. (2020) How COVID-19 Induces Cytokine Storm with High Mortality. Inflammation and Regeneration, 40, Article No. 37. https://doi.org/10.1186/s41232-020-00146-3

  46. 46. Lucas, C., et al. (2020) Longitudinal Analyses Reveal Immunological Misfiring in Severe COVID-19. Nature, 584, 463-469. https://doi.org/10.1038/s41586-020-2588-y

  47. 47. Morris, S.B., et al. (2020) Case Series of Multisystem Inflammatory Syndrome in Adults Associated with SARS-CoV-2 Infection—United Kingdom and United States, March-August 2020. Morbidity and Mortality Weekly Report (MMWR), 69, 1450-1456. https://doi.org/10.15585/mmwr.mm6940e1

  48. 48. Sokol, C.L. and Luster, A.D. (2015) The Chemokine System in Innate Immunity. Cold Spring Harbor Perspectives in Biology, 7, a016303. https://doi.org/10.1101/cshperspect.a016303

  49. 49. Tau, G. and Rothman, P. (1999) Biologic Functions of the IFN-Gamma Receptors. Allergy, 54, 1233-1251. https://doi.org/10.1034/j.1398-9995.1999.00099.x

  50. 50. Galani, I.-E., et al. (2021) Untuned Antiviral Immunity in COVID-19 Revealed by Temporal Type I/III Interferon Patterns and Flu Comparison. Nature Immunology, 22, 32-40. https://doi.org/10.1038/s41590-020-00840-x

  51. 51. Del Valle, D.M., et al. (2020) An Inflammatory Cytokine Signature Predicts COVID-19 Severity and Survival. Nature Medicine, 26, 1636-1643. https://doi.org/10.1038/s41591-020-1051-9

  52. 52. Huang, C., et al. (2020) Clinical Features of Patients Infected with 2019 Novel Coronavirus in Wuhan, China. The Lancet, 395, 497-506. https://doi.org/10.1016/S0140-6736(20)30183-5

  53. 53. Deshmane, S.L., Kremlev, S., Amini, S. and Sawaya, B.E. (2009) Monocyte Chemoattractant Protein-1 (MCP-1): An Overview. Journal of Interferon & Cytokine Research, 29, 313-326. https://doi.org/10.1089/jir.2008.0027

  54. 54. Jøntvedt Jørgensen, M., et al. (2020) Increased Interleukin-6 and Macrophage Chemoattractant Protein-1 Are Associated with Respiratory Failure in COVID-19. Scientific Reports, 10, Article No. 21697. https://doi.org/10.1038/s41598-020-78710-7

  55. 55. Hasan, A., Al-Ozairi, E., Al-Baqsumi, Z., Ahmad, R. and Al-Mulla, F. (2021) Cellular and Humoral Immune Responses in COVID-19 and Immunotherapeutic Approaches. ImmunoTargets and Therapy, 10, 63-85. https://doi.org/10.2147/ITT.S280706

  56. 56. Xu, Z., et al. (2020) Pathological Findings of COVID-19 Associated with Acute Respiratory Distress Syndrome. The Lancet Respiratory Medicine, 8, 420-422. https://doi.org/10.1016/S2213-2600(20)30076-X

  57. 57. Guan, W., et al. (2020) Clinical Characteristics of Coronavirus Disease 2019 in China. New England Journal of Medicine, 382, 1708-1720. https://doi.org/10.1056/NEJMoa2002032

  58. 58. Farshi, E., Kasmapur, B. and Arad, A. (2021) Investigation of Immune Cells on Elimination of Pulmonary-Infected COVID-19 and Important Role of Innate Immunity, Phagocytes. Reviews in Medical Virology, 31, e2158. https://doi.org/10.1002/rmv.2158

  59. 59. Kigi, D., et al. (1994) Cytotoxicity Mediated by T Cells and Natural Killer Cells Is Greatly Impaired in Perforin-Deficient Mice. Nature, 369, 31-37. https://doi.org/10.1038/369031a0

  60. 60. Jiang, Y., et al. (2020) COVID-19 Pneumonia: CD8+ T and NK Cells Are Decreased in Number but Compensatory Increased in Cytotoxic Potential. Clinical Immunology, 218, Article ID: 108516. https://doi.org/10.1016/j.clim.2020.108516

  61. 61. Wang, F., et al. (2020) The Laboratory Tests and Host Immunity of COVID-19 Patients with Different Severity of Illness. JCI Insight, 5, e137799. https://doi.org/10.1172/jci.insight.137799

  62. 62. Bobcakova, A., et al. (2021) Immune Profile in Patients with COVID-19: Lymphocytes Exhaustion Markers in Relationship to Clinical Outcome. Frontiers in Cellular and Infection Microbiology, 11, Article ID: 646688. https://doi.org/10.3389/fcimb.2021.646688

  63. 63. Zhao, Q., et al. (2020) Lymphopenia Is Associated with Severe Coronavirus Disease 2019 (COVID-19) Infections: A Systemic Review and Meta-Analysis. International Journal of Infectious Diseases, 96, 131-135. https://doi.org/10.1016/j.ijid.2020.04.086

  64. 64. Jiang, M., et al. (2020) T-Cell Subset Counts in Peripheral Blood Can Be Used as Discriminatory Biomarkers for Diagnosis and Severity Prediction of Coronavirus Disease 2019. The Journal of Infectious Diseases, 222, 198-202. https://doi.org/10.1093/infdis/jiaa252

  65. 65. Zheng, M., et al. (2020) Functional Exhaustion of Antiviral Lymphocytes in COVID-19 Patients. Cellular & Molecular Immunology, 17, 533-535. https://doi.org/10.1038/s41423-020-0402-2

  66. 66. Walls, A.C., Park, Y.-J., Tortorici, M.A., Wall, A., McGuire, A.T. and Veesler, D. (2020) Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell, 181, 281-292.e6. https://doi.org/10.1016/j.cell.2020.02.058

  67. 67. Sterlin, D., et al. (2021) IgA Dominates the Early Neutralizing Antibody Response to SARS-CoV-2. Science Translational Medicine, 13. https://doi.org/10.1126/scitranslmed.abd2223

  68. 68. Chansaenroj, J., et al. (2021) Detection of SARS-CoV-2-Specific Antibodies via Rapid Diagnostic Immunoassays in COVID-19 Patients. Virology Journal, 18, Article No. 52. https://doi.org/10.1186/s12985-021-01530-2

  69. 69. Dogan, M., et al. (2020) Novel SARS-CoV-2 Specific Antibody and Neutralization Assays Reveal Wide Range of Humoral Immune Response during COVID-19. medRxiv. https://doi.org/10.1101/2020.07.07.20148106

  70. 70. Evans, M.J. and Kaufman, M.H. (1981) Establishment in Culture of Pluripotential Cells from Mouse Embryos. Nature, 292, 154-156. https://doi.org/10.1038/292154a0

  71. 71. Dwivedi, S. and Sharma, P. (2018) Stem Cell Biology: A New Hope in Regenerations and Replenishments Therapy. Indian Journal of Clinical Biochemistry, 33, 369-371. https://doi.org/10.1007/s12291-018-0792-4

  72. 72. Farini, A., Sitzia, C., Erratico, S., Meregalli, M. and Torrente, Y. (2014) Clinical Applications of Mesenchymal Stem Cells in Chronic Diseases. Stem Cells International, 2014, Article ID: 306573. https://doi.org/10.1155/2014/306573

  73. 73. Joyce, N., Annett, G., Wirthlin, L., Olson, S., Bauer, G. and Nolta, J.A. (2010) Mesenchymal Stem Cells for the Treatment of Neurodegenerative Disease. Regenerative Medicine, 5, 933-946. https://doi.org/10.2217/rme.10.72

  74. 74. Wang, S., Qu, X. and Zhao, R.C. (2012) Clinical Applications of Mesenchymal Stem Cells. Journal of Hematology & Oncology, 5, Article No. 19. https://doi.org/10.1186/1756-8722-5-19

  75. 75. Wei, X., Yang, X., Han, Z., Qu, F., Shao, L. and Shi, Y. (2013) Mesenchymal Stem Cells: A New Trend for Cell Therapy. Acta Pharmacologica Sinica, 34, 747-754. https://doi.org/10.1038/aps.2013.50

  76. 76. Parekkadan, B. and Milwid, J.M. (2010) Mesenchymal Stem Cells as Therapeutics. Annual Review of Biomedical Engineering, 12, 87-117. https://doi.org/10.1146/annurev-bioeng-070909-105309

  77. 77. Kouris, N.A., et al. (2012) Directed Fusion of Mesenchymal Stem Cells with Cardiomyocytes via VSV-G Facilitates Stem Cell Programming. Stem Cells International, 2012, Article ID: 414038. https://doi.org/10.1155/2012/414038

  78. 78. Hofer, H.R. and Tuan, R.S. (2016) Secreted Trophic Factors of Mesenchymal Stem Cells Support Neurovascular and Musculoskeletal Therapies. Stem Cell Research & Therapy, 7, Article No. 131. https://doi.org/10.1186/s13287-016-0394-0

  79. 79. Jackson, M.V., 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. https://doi.org/10.1002/stem.2372

  80. 80. Robbins, P.D. and Morelli, A.E. (2014) Regulation of Immune Responses by Extracellular Vesicles. Nature Reviews Immunology, 14, 195-208. https://doi.org/10.1038/nri3622

  81. 81. Liang, B., et al. (2020) Clinical Remission of a Critically Ill COVID-19 Patient Treated by Human Umbilical Cord Mesenchymal Stem Cells. Medicine (Baltimore), 99, e21429. https://doi.org/10.1097/MD.0000000000021429

  82. 82. Sánchez-Guijo, F., et al. (2020) Adipose-Derived Mesenchymal Stromal Cells for the Treatment of Patients with Severe SARS-CoV-2 Pneumonia Requiring Mechanical Ventilation. A Proof of Concept Study. eClinicalMedicine, 25, Article ID: 100454. https://doi.org/10.1016/j.eclinm.2020.100454

  83. 83. Avanzini, M.A., et al. (2021) Human Mesenchymal Stromal Cells Do Not Express ACE2 and TMPRSS2 and Are Not Permissive to SARS-CoV-2 Infection. STEM CELLS Translational Medicine, 10, 636-642. https://doi.org/10.1002/sctm.20-0385

  84. 84. Schäfer, R., et al. (2021) Human Mesenchymal Stromal Cells Are Resistant to SARS-CoV-2 Infection under Steady-State, Inflammatory Conditions and in the Presence of SARS-CoV-2-Infected Cells. Stem Cell Reports, 16, 419-427. https://doi.org/10.1016/j.stemcr.2020.09.003

  85. 85. Sakaguchi, W., et al. (2020) Existence of SARS-CoV-2 Entry Molecules in the Oral Cavity. International Journal of Molecular Sciences, 21, Article 6000. https://doi.org/10.3390/ijms21176000

  86. 86. Durand, N., Russell, A. and Zubair, A.C. (2019) Effect of Comedications and Endotoxins on Mesenchymal Stem Cell Secretomes, Migratory and Immunomodulatory Capacity. Journal of Clinical Medicine, 8, Article 497. https://doi.org/10.3390/jcm8040497

  87. 87. Russell, A.L., Lefavor, R., Durand, N., Glover, L. and Zubair, A.C. (2018) Modifiers of Mesenchymal Stem Cell Quantity and Quality. Transfusion, 58, 1434-1440. https://doi.org/10.1111/trf.14597

  88. 88. Shu, L., et al. (2020) Treatment of Severe COVID-19 with Human Umbilical Cord Mesenchymal Stem Cells. Stem Cell Research & Therapy, 11, Article No. 361. https://doi.org/10.1186/s13287-020-01875-5

  89. 89. de Witte, S.F.H., et al. (2018) Immunomodulation BY Therapeutic Mesenchymal Stromal Cells (MSC) Is Triggered Through Phagocytosis of MSC By Monocytic Cells. Stem Cells, 36, 602-615. https://doi.org/10.1002/stem.2779

  90. 90. Hu, C. and Li, L. (2019) The Immunoregulation of Mesenchymal Stem Cells Plays a Critical Role in Improving the Prognosis of Liver Transplantation. Journal of Translational Medicine, 17, Article No. 412. https://doi.org/10.1186/s12967-019-02167-0

  91. 91. Golchin, A., Seyedjafari, E. and Ardeshirylajimi, A. (2020) Mesenchymal Stem Cell Therapy for COVID-19: Present or Future. Stem Cell Reviews and Reports, 16, 427-433. https://doi.org/10.1007/s12015-020-09973-w

  92. 92. Lu, Z., et al. (2019) Mesenchymal Stem Cells Induce Dendritic Cell Immune Tolerance via Paracrine Hepatocyte Growth Factor to Alleviate Acute Lung Injury. Stem Cell Research & Therapy, 10, Article No. 372. https://doi.org/10.1186/s13287-019-1488-2

  93. 93. Park, M., Kim, Y.-H., Ryu, J.-H., Woo, S.-Y. and Ryu, K.-H. (2015) Immune Suppressive Effects of Tonsil-Derived Mesenchymal Stem Cells on Mouse Bone-Marrow-Derived Dendritic Cells. Stem Cells International, 2015, Article ID: 106540. https://doi.org/10.1155/2015/106540

  94. 94. Spaggiari, G.M. and Moretta, L. (2013) Interactions between Mesenchymal Stem Cells and Dendritic Cells. In: Weyand, B., Dominici, M., Hass, R., Jacobs, R. and Kasper, C., Eds., Mesenchymal Stem Cells—Basics and Clinical Application II, Springer, Berlin, Heidelberg, 199-208. https://doi.org/10.1007/10_2012_154

  95. 95. Raffaghello, L., et al. (2008) Human Mesenchymal Stem Cells Inhibit Neutrophil Apoptosis: A Model for Neutrophil Preservation in the Bone Marrow Niche. Stem Cells, 26, 151-162. https://doi.org/10.1634/stemcells.2007-0416

  96. 96. Lee, S., Zhang, Q.Z., Karabucak, B. and Le, A.D. (2016) DPSCs from Inflamed Pulp Modulate Macrophage Function via the TNF-α/IDO Axis. Journal of Dental Research, 95, 1274-1281. https://doi.org/10.1177/0022034516657817

  97. 97. Zhang, Y., Chen, Y. and Meng, Z. (2020) Immunomodulation for Severe COVID-19 Pneumonia: The State of the Art. Frontiers in Immunology, 11, Article ID: 577442. https://doi.org/10.3389/fimmu.2020.577442

  98. 98. Loke, X.Y., Imran, S.A.M., Tye, G.J., Wan Kamarul Zaman, W.S. and Nordin, F. (2021) Immunomodulation and Regenerative Capacity of MSCs for Long-COVID. International Journal of Molecular Sciences, 22, Article ID: 12421. https://doi.org/10.3390/ijms222212421

  99. 99. Tsyb, A.F., et al. (2008) In Vitro Inhibitory Effect of Mesenchymal Stem Cells on Zymosan-Induced Production of Reactive Oxygen Species. Bulletin of Experimental Biology and Medicine, 146, 158-164. https://doi.org/10.1007/s10517-008-0238-8

  100. 100. Vasandan, A.B., Jahnavi, S., Shashank, C., Prasad, P., Kumar, A. and Prasanna, S.J. (2016) Human Mesenchymal Stem Cells Program Macrophage Plasticity by Altering Their Metabolic Status via a PGE 2-Dependent Mechanism. Scientific Reports, 6, Article No. 38308. https://doi.org/10.1038/srep38308

  101. 101. Le Blanc, K. and Davies, L.C. (2015) Mesenchymal Stromal Cells and the Innate Immune Response. Immunology Letters, 168, 140-146. https://doi.org/10.1016/j.imlet.2015.05.004

  102. 102. Chen, B., Ni, Y., Liu, J., Zhang, Y. and Yan, F. (2018) Bone Marrow-Derived Mesenchymal Stem Cells Exert Diverse Effects on Different Macrophage Subsets. Stem Cells International, 2018, Article ID: 8348121. https://doi.org/10.1155/2018/8348121

  103. 103. Fan, L., Hu, C., Chen, J., Cen, P., Wang, J. and Li, L. (2016) Interaction between Mesenchymal Stem Cells and B-Cells. International Journal of Molecular Sciences, 17, Article 650. https://doi.org/10.3390/ijms17050650

  104. 104. Jiang, X.-X., et al. (2005) Human Mesenchymal Stem Cells Inhibit Differentiation and Function of Monocyte-Derived Dendritic Cells. Blood, 105, 4120-4126. https://doi.org/10.1182/blood-2004-02-0586

  105. 105. Melief, S.M., et al. (2013) Multipotent Stromal Cells Induce Human Regulatory T Cells through a Novel Pathway Involving Skewing of Monocytes toward Anti-Inflammatory Macrophages. Stem Cells, 31, 1980-1991. https://doi.org/10.1002/stem.1432

  106. 106. Deng, Y., et al. (2016) Umbilical Cord-Derived Mesenchymal Stem Cells Instruct Monocytes towards an IL10-producing Phenotype by Secreting IL6 and HGF. Scientific Reports, 6, Article No. 37566. https://doi.org/10.1038/srep37566

  107. 107. Allavena, P., et al. (1998) IL-10 Prevents the Differentiation of Monocytes to Dendritic Cells but Promotes Their Maturation to Macrophages. European Journal of Immunology, 28, 359-369. https://doi.org/10.1002/(SICI)1521-4141(199801)28:01<359::AID-IMMU359>3.0.CO;2-4

  108. 108. Liu, J., et al. (2020) Apoptotic Bodies Derived from Mesenchymal Stem Cells Promote Cutaneous Wound Healing via Regulating the Functions of Macrophages. Stem Cell Research & Therapy, 11, Article No. 507. https://doi.org/10.1186/s13287-020-02014-w

  109. 109. Martínez-Aguilar, R., Romero-Pinedo, S., Ruiz-Magaña, M.J., Olivares, E.G., Ruiz-Ruiz, C. and Abadía-Molina, A.C. (2020) Menstrual Blood-Derived Stromal Cells Modulate Functional Properties of Mouse and Human Macrophages. Scientific Reports, 10, Article No. 21389. https://doi.org/10.1038/s41598-020-78423-x

  110. 110. Nguyen, H.-Q.-D., Kao, C.-Y., Chiang, C.-P., Hung, Y.-H. and Lo, C.-M. (2022) Investigating the Immunomodulatory Potential of Dental Pulp Stem Cell Cultured on Decellularized Bladder Hydrogel towards Macrophage Response in Vitro. Gels, 8, Article 187. https://doi.org/10.3390/gels8030187

  111. 111. Fathi, E., Farahzadi, R., Valipour, B. and Sanaat, Z. (2019) Cytokines Secreted from Bone Marrow Derived Mesenchymal Stem Cells Promote Apoptosis and Change Cell Cycle Distribution of K562 Cell Line as Clinical Agent in Cell Transplantation. PLOS ONE, 14, e0215678. https://doi.org/10.1371/journal.pone.0215678

  112. 112. Gu, Y., et al. (2015) Mesenchymal Stem Cells Suppress Neuronal Apoptosis and Decrease IL-10 Release via the TLR2/NFκB Pathway in Rats with Hypoxic-Ischemic Brain Damage. Molecular Brain, 8, Article No. 65. https://doi.org/10.1186/s13041-015-0157-3

  113. 113. Ahmed, N.E.-M.B., Murakami, M., Hirose, Y. and Nakashima, M. (2016) Therapeutic Potential of Dental Pulp Stem Cell Secretome for Alzheimer’s Disease Treatment: An in Vitro Study. Stem Cells International, 2016, Article ID: 8102478. https://doi.org/10.1155/2016/8102478

  114. 114. Ferlazzo, G. and Carrega, P. (2012) Natural Killer Cell Distribution and Trafficking in Human Tissues. Frontiers in Immunology, 3, Article 347. https://doi.org/10.3389/fimmu.2012.00347

  115. 115. Paul, S. and Lal, G. (2017) The Molecular Mechanism of Natural Killer Cells Function and Its Importance in Cancer Immunotherapy. Frontiers in Immunology, 8, Article 1124. https://doi.org/10.3389/fimmu.2017.01124

  116. 116. Giuliani, M., et al. (2011) Human Mesenchymal Stem Cells Derived from Induced Pluripotent Stem Cells Down-Regulate NK-Cell Cytolytic Machinery. Blood, 118, 3254-3262. https://doi.org/10.1182/blood-2010-12-325324

  117. 117. Reinders, M.E. and Hoogduijn, M.J. (2014) NK Cells and MSCs: Possible Implications for MSC Therapy in Renal Transplantation. Journal of Stem Cell Research & Therapy, 4, Article ID: 1000166.

  118. 118. Spaggiari, G.M., Capobianco, A., Abdelrazik, H., Becchetti, F., Mingari, M.C. and Moretta, L. (2008) Mesenchymal Stem Cells Inhibit Natural Killer-Cell Proliferation, Cytotoxicity, and Cytokine Production: Role of Indoleamine 2,3-Dioxygenase and Prostaglandin E2. Blood, 111, 1327-1333. https://doi.org/10.1182/blood-2007-02-074997

  119. 119. Cui, R., et al. (2016) Human Mesenchymal Stromal/Stem Cells Acquire Immunostimulatory Capacity upon Cross-Talk with Natural Killer Cells and Might Improve the NK Cell Function of Immunocompromised Patients. Stem Cell Research & Therapy, 7, Article No. 88. https://doi.org/10.1186/s13287-016-0353-9

  120. 120. Sotiropoulou, P.A., Perez, S.A., Gritzapis, A.D., Baxevanis, C.N. and Papamichail, M. (2006) Interactions between Human Mesenchymal Stem Cells and Natural Killer Cells. Stem Cells, 24, 74-85. https://doi.org/10.1634/stemcells.2004-0359

  121. 121. Yan, F., et al. (2019) Human Dental Pulp Stem Cells Regulate Allogeneic NK Cells’ Function via Induction of Anti-Inflammatory Purinergic Signalling in Activated NK Cells. Cell Proliferation, 52, e12595. https://doi.org/10.1111/cpr.12595

  122. 122. Maucourant, C., et al. (2020) Natural Killer Cell Immunotypes Related to COVID-19 Disease Severity. Science Immunology, 5, eabd6832. https://doi.org/10.1126/sciimmunol.abd6832

  123. 123. Bartholomew, A., et al. (2002) Mesenchymal Stem Cells Suppress Lymphocyte Proliferation in Vitro and Prolong Skin Graft Survival in Vivo. Experimental Hematology, 30, 42-48. https://doi.org/10.1016/S0301-472X(01)00769-X

  124. 124. Beyth, S., et al. (2005) Human Mesenchymal Stem Cells Alter Antigen-Presenting Cell Maturation and Induce T-Cell Unresponsiveness. Blood, 105, 2214-2219. https://doi.org/10.1182/blood-2004-07-2921

  125. 125. Haddad, R. and Saldanha-Araujo, F. (2014) Mechanisms of T-Cell Immunosuppression by Mesenchymal Stromal Cells: What Do We Know So Far? BioMed Research International, 2014, Article ID: 216806. https://doi.org/10.1155/2014/216806

  126. 126. Laing, A.G., Fanelli, G., Ramirez-Valdez, A., Lechler, R.I., Lombardi, G. and Sharpe, P.T. (2019) Mesenchymal Stem Cells Inhibit T-Cell Function through Conserved Induction of Cellular Stress. PLOS ONE, 14, e0213170. https://doi.org/10.1371/journal.pone.0213170

  127. 127. Najar, M., et al. (2009) Mesenchymal Stromal Cells Promote or Suppress the Proliferation of T Lymphocytes from Cord Blood and Peripheral Blood: The Importance of Low Cell Ratio and Role of Interleukin-6. Cytotherapy, 11, 570-583. https://doi.org/10.1080/14653240903079377

  128. 128. Martin-Rufino, J.D., Espinosa-Lara, N., Osugui, L. and Sanchez-Guijo, F. (2019) Targeting the Immune System with Mesenchymal Stromal Cell-Derived Extracellular Vesicles: What Is the Cargo’s Mechanism of Action? Frontiers in Bioengineering and Biotechnology, 7, Article 308. https://doi.org/10.3389/fbioe.2019.00308

  129. 129. Wang, L., et al. (2021) Regulation of Inflammatory Cytokine Storms by Mesenchymal Stem Cells. Frontiers in Immunology, 12, Article ID: 726909. https://doi.org/10.3389/fimmu.2021.726909

  130. 130. van Megen, K.M., van’t Wout, E.-J.T., Lages Motta, J., Dekker, B., Nikolic, T. and Roep, B.O. (2019) Activated Mesenchymal Stromal Cells Process and Present Antigens Regulating Adaptive Immunity. Frontiers in Immunology, 10, Article 694. https://doi.org/10.3389/fimmu.2019.00694

  131. 131. Kyurkchiev, D., et al. (2014) Secretion of Immunoregulatory Cytokines by Mesenchymal Stem Cells. World Journal of Stem Cells, 6, 552-570. https://doi.org/10.4252/wjsc.v6.i5.552

  132. 132. Moll, G., Hoogduijn, M.J. and Ankrum, J.A. (2020) Safety, Efficacy and Mechanisms of Action of Mesenchymal Stem Cell Therapies. Frontiers in Immunology, 11, Article 243. https://doi.org/10.3389/978-2-88963-600-6

  133. 133. Terraza-Aguirre, C., et al. (2020) Mechanisms behind the Immunoregulatory Dialogue between Mesenchymal Stem Cells and Th17 Cells. Cells, 9, Article 1660. https://doi.org/10.3390/cells9071660

  134. 134. Tatara, R., et al. (2011) Mesenchymal Stromal Cells Inhibit Th17 but Not Regulatory T-Cell Differentiation. Cytotherapy, 13, 686-694. https://doi.org/10.3109/14653249.2010.542456

  135. 135. Favaro, E., et al. (2016) Human Mesenchymal Stem Cells and Derived Extracellular Vesicles Induce Regulatory Dendritic Cells in Type 1 Diabetic Patients. Diabetologia, 59, 325-333. https://doi.org/10.1007/s00125-015-3808-0

  136. 136. Chen, Q.-H., et al. (2020) Mesenchymal Stem Cells Regulate the Th17/Treg Cell Balance Partly through Hepatocyte Growth Factor in Vitro. Stem Cell Research & Therapy, 11, Article No. 91. https://doi.org/10.1186/s13287-020-01612-y

  137. 137. de Cássia Noronha, N., et al. (2019) Priming Approaches to Improve the Efficacy of Mesenchymal Stromal Cell-Based Therapies. Stem Cell Research & Therapy, 10, Article No. 131. https://doi.org/10.1186/s13287-019-1224-y

  138. 138. Luk, F., et al. (2017) Inflammatory Conditions Dictate the Effect of Mesenchymal Stem or Stromal Cells on B Cell Function. Frontiers in Immunology, 8, Article 1042. https://doi.org/10.3389/fimmu.2017.01042

  139. 139. Luz-Crawford, P., et al. (2016) Mesenchymal Stem Cell-Derived Interleukin 1 Receptor Antagonist Promotes Macrophage Polarization and Inhibits B Cell Differentiation. Stem Cells, 34, 483-492. https://doi.org/10.1002/stem.2254

  140. 140. Corcione, A., et al. (2006) Human Mesenchymal Stem Cells Modulate B-Cell Functions. Blood, 107, 367-372. https://doi.org/10.1182/blood-2005-07-2657

  141. 141. Ding, G., Niu, J. and Liu, Y. (2015) Dental Pulp Stem Cells Suppress the Proliferation of Lymphocytes via Transforming Growth Factor-β1. Human Cell, 28, 81-90. https://doi.org/10.1007/s13577-014-0106-y

  142. 142. Feng, Y., et al. (2020) Safety and Feasibility of Umbilical Cord Mesenchymal Stem Cells in Patients with COVID-19 Pneumonia: A Pilot Study. Cell Proliferation, 53, e12947. https://doi.org/10.1111/cpr.12947

  143. 143. Park, B.-W., et al. (2020) In Vivo Priming of Human Mesenchymal Stem Cells with Hepatocyte Growth Factor-Engineered Mesenchymal Stem Cells Promotes Therapeutic Potential for Cardiac Repair. Science Advances, 6, eaay6994. https://doi.org/10.1126/sciadv.aay6994

  144. 144. Raza, S.S., Seth, P. and Khan, M.A. (2022) ‘Primed’ Mesenchymal Stem Cells: A Potential Novel Therapeutic for COVID19 Patients. Stem Cell Reviews and Reports, 17, 153-162. https://doi.org/10.1007/s12015-020-09999-0

  145. 145. Bustos, M.L., et al. (2013) Activation of Human Mesenchymal Stem Cells Impacts Their Therapeutic Abilities in Lung Injury by Increasing Interleukin (IL)-10 and IL-1RN Levels. STEM CELLS Translational Medicine, 2, 884-895. https://doi.org/10.5966/sctm.2013-0033

  146. 146. Xu, A.L., et al. (2019) Mesenchymal Stem Cells Reconditioned in Their Own Serum Exhibit Augmented Therapeutic Properties in the Setting of Acute Respiratory Distress Syndrome. STEM CELLS Translational Medicine, 8, 1092-1106. https://doi.org/10.1002/sctm.18-0236

  147. 147. Giunti, D., et al. (2021) Role of miRNAs Shuttled by Mesenchymal Stem Cell-Derived Small Extracellular Vesicles in Modulating Neuroinflammation. Scientific Reports, 11, Article No. 1740. https://doi.org/10.1038/s41598-021-81039-4

  148. 148. Öztürk, S., Elçin, A.E., Koca, A. and Elçin, Y.M. (2020) Therapeutic Applications of Stem Cells and Extracellular Vesicles in Emergency Care: Futuristic Perspectives. Stem Cell Reviews and Reports, 17, 390-410. https://doi.org/10.1007/s12015-020-10029-2

  149. 149. Askenase, P.W. (2020) COVID-19 Therapy with Mesenchymal Stromal Cells (MSC) and Convalescent Plasma Must Consider Exosome Involvement: Do the Exosomes in Convalescent Plasma Antagonize the Weak Immune Antibodies? Journal of Extracellular Vesicles, 10, e12004. https://doi.org/10.1002/jev2.12004

  150. 150. Jayaramayya, K., et al. (2020) Immunomodulatory Effect of Mesenchymal Stem Cells and Mesenchymal Stem-Cell-Derived Exosomes for COVID-19 Treatment. BMB Reports, 53, 400-412. https://doi.org/10.5483/BMBRep.2020.53.8.121

  151. 151. Wiklander, O.P.B., Brennan, M.á., Lötvall, J., Breakefield, X.O. and El Andaloussi, S. (2019) Advances in Therapeutic Applications of Extracellular Vesicles. Science Translational Medicine, 11, aav852. https://doi.org/10.1126/scitranslmed.aav8521

  152. 152. Chung, M.-J., et al. (2020) Mesenchymal Stem Cell and MicroRNA Therapy of Musculoskeletal Diseases. International Journal of Stem Cells, 14, 150-167. https://doi.org/10.15283/ijsc20167

  153. 153. Zayed, M. and Iohara, K. (2020) Immunomodulation and Regeneration Properties of Dental Pulp Stem Cells: A Potential Therapy to Treat Coronavirus Disease 2019. Cell Transplantation, 29. https://doi.org/10.1177/0963689720952089

  154. 154. Cui, S.J., et al. (2020) DPSCs Attenuate Experimental Progressive TMJ Arthritis by Inhibiting the STAT1 Pathway. Journal of Dental Research, 99, 446-455. https://doi.org/10.1177/0022034520901710

  155. 155. Matsumura-Kawashima, M., Ogata, K., Moriyama, M., Murakami, Y., Kawado, T. and Nakamura, S. (2021) Secreted Factors from Dental Pulp Stem Cells Improve Sjögren’s Syndrome via Regulatory T Cell-Mediated Immunosuppression. Stem Cell Research & Therapy, 12, Article No. 182. https://doi.org/10.1186/s13287-021-02236-6

  156. 156. Ogata, K., Matsumura-Kawashima, M., Moriyama, M., Kawado, T. and Nakamura, S. (2021) Dental Pulp-Derived Stem Cell-Conditioned Media Attenuates Secondary Sjögren’s Syndrome via Suppression of Inflammatory Cytokines in the Submandibular Glands. Regenerative Therapy, 16, 73-80. https://doi.org/10.1016/j.reth.2021.01.006

  157. 157. Fujii, Y., et al. (2018) Bone Regeneration by Human Dental Pulp Stem Cells Using a Helioxanthin Derivative and Cell-Sheet Technology. Stem Cell Research & Therapy, 9, Article No. 24. https://doi.org/10.1186/s13287-018-0783-7

  158. 158. Lam, C., et al. (2021) Human Dental Pulp Stem Cells (DPSCs) Therapy in Rescuing Photoreceptors and Establishing a Sodium Iodate-Induced Retinal Degeneration Rat Model. Tissue Engineering and Regenerative Medicine, 18, 143-154. https://doi.org/10.1007/s13770-020-00312-1

  159. 159. Zhang, Y., et al. (2021) DPSCs Treated by TGF-β1 Regulate Angiogenic Sprouting of Three-Dimensionally Co-Cultured HUVECs and DPSCs through VEGF-Ang-Tie2 Signaling. Stem Cell Research & Therapy, 12, Article No. 281. https://doi.org/10.1186/s13287-021-02349-y

  160. 160. Hossein-Khannazer, N., Hashemi, S.M., Namaki, S., Ghanbarian, H., Sattari, M. and Khojasteh, A. (2019) Study of the Immunomodulatory Effects of Osteogenic Differentiated Human Dental Pulp Stem Cells. Life Sciences, 216, 111-118. https://doi.org/10.1016/j.lfs.2018.11.040

  161. 161. Pierdomenico, L., et al. (2005) Multipotent Mesenchymal Stem Cells with Immunosuppressive Activity Can Be Easily Isolated from Dental Pulp. Transplantation, 80, 836-842. https://doi.org/10.1097/01.tp.0000173794.72151.88

  162. 162. Nakashima, M., Iohara, K. and Murakami, M. (2013) Dental Pulp Stem Cells and Regeneration. Endodontic Topics, 28, 38-50. https://doi.org/10.1111/etp.12027

  163. 163. Monsel, A., et al. (2022) Treatment of COVID-19-Associated ARDS with Mesenchymal Stromal Cells: A Multicenter Randomized Double-Blind Trial. Critical Care, 26, Article No. 48. https://doi.org/10.1186/s13054-022-03930-4

  164. 164. Peng, H., et al. (2020) A Synergistic Role of Convalescent Plasma and Mesenchymal Stem Cells in the Treatment of Severely Ill COVID-19 Patients: A Clinical Case Report. Stem Cell Research & Therapy, 11, Article No. 291. https://doi.org/10.1186/s13287-020-01802-8

  165. 165. Guo, Z., Chen, Y., Luo, X., He, X., Zhang, Y. and Wang, J. (2020) Administration of Umbilical Cord Mesenchymal Stem Cells in Patients with Severe COVID-19 Pneumonia. Critical Care, 24, Article No. 420. https://doi.org/10.1186/s13054-020-03142-8

  166. 166. Shi, L., et al. (2020) Treatment with Human Umbilical Cord-Derived Mesenchymal stem Cells for COVID-19 Patients with Lung Damage: A Randomised, Double-Blind, Placebo-Controlled Phase 2 Trial. medRxiv. https://doi.org/10.1101/2020.10.15.20213553

  167. 167. Hashemian, S.-M.R., et al. (2021) Mesenchymal Stem Cells Derived from Perinatal Tissues for Treatment of Critically Ill COVID-19-Induced ARDS Patients: A Case Series. Stem Cell Research & Therapy, 12, Article No. 91. https://doi.org/10.1186/s13287-021-02165-4

  168. 168. Häberle, H., et al. (2021) Mesenchymal Stem Cell Therapy for Severe COVID-19 ARDS. Journal of Intensive Care Medicine, 36, 681-688. https://doi.org/10.1177/0885066621997365

  169. 169. Tao, J., et al. (2020) Umbilical Cord Blood-Derived Mesenchymal Stem Cells in Treating a Critically Ill COVID-19 Patient. The Journal of Infection in Developing Countries, 14, 1138-1145. https://doi.org/10.3855/jidc.13081

  170. 170. Tang, L., et al. (2020) Clinical Study Using Mesenchymal Stem Cells for the Treatment of Patients with Severe COVID-19. Frontiers in Medicine, 14, 664-673. https://doi.org/10.1007/s11684-020-0810-9

  171. 171. COVID-19 Treatment Guidelines. Clinical Spectrum of SARS-CoV-2 Infection. https://www.covid19treatmentguidelines.nih.gov/overview/clinical-spectrum/

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] Spicer, A.J. and Jalkanen, S. (2021) Why Haven’t We Found an Effective Treatment for COVID-19? Frontiers in Immunology, 12, Article ID: 644850.
https://doi.org/10.3389/fimmu.2021.644850
[2] Shi, L., et al. (2021) Mesenchymal Stem Cell Therapy for Severe COVID-19. Signal Transduction and Targeted Therapy, 6, Article No. 339.
https://doi.org/10.1038/s41392-021-00754-6
[3] Yang, L., Xie, X., Tu, Z., Fu, J., Xu, D. and Zhou, Y. (2021) The Signal Pathways and Treatment of Cytokine Storm in COVID-19. Signal Transduction and Targeted Therapy, 6, Article No. 255.
https://doi.org/10.1038/s41392-021-00679-0
[4] Henderson, L.A., et al. (2020) On the Alert for Cytokine Storm: Immunopathology in COVID-19. Arthritis & Rheumatology, 72, 1059-1063.
https://doi.org/10.1002/art.41285
[5] Riedel, R.N., Pérez-Pérez, A., Sánchez-Margalet, V., Varone, C.L. and Maymó, J.L. (2021) Stem Cells and COVID-19: Are the Human Amniotic Cells a New Hope for Therapies against the SARS-CoV-2 Virus? Stem Cell Research & Therapy, 12, Article No. 155.
https://doi.org/10.1186/s13287-021-02216-w
[6] Dominici, M., et al. (2006) Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells. The International Society for Cellular Therapy Position Statement. Cytotherapy, 8, 315-317.
https://doi.org/10.1080/14653240600855905
[7] Durand, N., Mallea, J. and Zubair, A.C. (2020) Insights into the Use of Mesenchymal Stem Cells in COVID-19 Mediated Acute Respiratory Failure. npj Regenerative Medicine, 5, Article No. 17.
https://doi.org/10.1038/s41536-020-00105-z
[8] Gao, F., et al. (2016) Mesenchymal Stem Cells and Immunomodulation: Current Status and Future Prospects. Cell Death & Disease, 7, e2062.
https://doi.org/10.1038/cddis.2015.327
[9] Xu, R., Feng, Z. and Wang, F.-S. (2022) Mesenchymal Stem Cell Treatment for COVID-19. eBioMedicine, 77, Article ID: 103920.
https://doi.org/10.1016/j.ebiom.2022.103920
[10] Kean, T.J., Lin, P., Caplan, A.I. and Dennis, J.E. (2013) MSCs: Delivery Routes and Engraftment, Cell-Targeting Strategies, and Immune Modulation. Stem Cells International, 2013, e732742.
https://doi.org/10.1155/2013/732742
[11] Deng, Y., et al. (2020) Clinical Characteristics of Fatal and Recovered Cases of Coronavirus Disease 2019 in Wuhan, China: A Retrospective Study. Chinese Medical Journal, 133, 1261-1267.
https://doi.org/10.1097/CM9.0000000000000824
[12] Moll, G., Drzeniek, N., Kamhieh-Milz, J., Geissler, S., Volk, H.-D. and Reinke, P. (2020) MSC Therapies for COVID-19: Importance of Patient Coagulopathy, Thromboprophylaxis, Cell Product Quality and Mode of Delivery for Treatment Safety and Efficacy. Frontiers in Immunology, 11, Article 1091.
https://doi.org/10.3389/fimmu.2020.01091
[13] Yao, W., Shi, L., Zhang, Y., Dong, H. and Zhang, Y. (2022) Mesenchymal Stem/Stromal Cell Therapy for COVID-19 Pneumonia: Potential Mechanisms, Current Clinical Evidence, and Future Perspectives. Stem Cell Research & Therapy, 13, Article No. 124.
https://doi.org/10.1186/s13287-022-02810-6
[14] Leng, Z., et al. (2020) Transplantation of ACE2- Mesenchymal Stem Cells Improves the Outcome of Patients with COVID-19 Pneumonia. Aging and Disease, 11, 216-228.
[15] Meng, F., et al. (2020) Human Umbilical Cord-Derived Mesenchymal Stem Cell Therapy in Patients with COVID-19: A Phase 1 Clinical Trial. Signal Transduction and Targeted Therapy, 5, Article No. 172.
https://doi.org/10.1038/s41392-020-00286-5
[16] Zhang, Y., et al. (2020) Intravenous Infusion of Human Umbilical Cord Wharton’s Jelly-Derived Mesenchymal Stem Cells as a Potential Treatment for Patients with COVID-19 Pneumonia. Stem Cell Research & Therapy, 11, Article No. 207.
https://doi.org/10.1186/s13287-020-01725-4
[17] Lanzoni, G., et al. (2021) Umbilical Cord Mesenchymal Stem Cells for COVID-19 Acute Respiratory Distress Syndrome: A Double-Blind, Phase 1/2a, Randomized Controlled Trial. STEM CELLS Translational Medicine, 10, 660-673.
https://doi.org/10.1002/sctm.20-0472
[18] Adugna, D.G. (2021) Current Clinical Application of Mesenchymal Stem Cells in the Treatment of Severe COVID-19 Patients: Review. Stem Cells and Cloning: Advances and Applications, 14, 71-80.
https://doi.org/10.2147/SCCAA.S333800
[19] Iohara, K., Zheng, L., Ito, M., Tomokiyo, A., Matsushita, K. and Nakashima, M. (2006) Side Population Cells Isolated from Porcine Dental Pulp Tissue with Self-Renewal and Multipotency for Dentinogenesis, Chondrogenesis, Adipogenesis, and Neurogenesis. Stem Cells, 24, 2493-2503.
https://doi.org/10.1634/stemcells.2006-0161
[20] Andrukhov, O., Behm, C., Blufstein, A. and Rausch-Fan, X. (2019) Immunomodulatory Properties of Dental Tissue-Derived Mesenchymal Stem Cells: Implication in Disease and Tissue Regeneration. World Journal of Stem Cells, 11, 604-617.
https://doi.org/10.4252/wjsc.v11.i9.604
[21] Tsutsui, T.W. (2020) Dental Pulp Stem Cells: Advances to Applications. Stem Cells and Cloning: Advances and Applications, 13, 33-42.
https://doi.org/10.2147/SCCAA.S166759
[22] Ye, Q., et al. (2020) Safety and Efficacy Assessment of Allogeneic Human Dental Pulp Stem Cells to Treat Patients with Severe COVID-19: Structured Summary of a Study Protocol for a Randomized Controlled Trial (Phase I/II). Trials, 21, Article No. 520.
https://doi.org/10.1186/s13063-020-04380-5
[23] Croci, S., et al. (2021) Human Dental Pulp Stem Cells Modulate Cytokine Production in vitro by Peripheral Blood Mononuclear Cells from Coronavirus Disease 2019 Patients. Frontiers in Cell and Developmental Biology, 8, Article ID: 609204.
https://doi.org/10.3389/fcell.2020.609204
[24] Di Tinco, R., et al. (2021) Role of PD-L1 in Licensing immunoregulatory Function of Dental Pulp Mesenchymal Stem Cells. Stem Cell Research & Therapy, 12, Article No. 598.
https://doi.org/10.1186/s13287-021-02664-4
[25] Part 15 Hearing: Draft Guidances Relating to the Regulation of human Cells, Tissues, or Cellular or Tissue-Based Products. 277.
[26] Center for Biologics Evaluation and Research, US Food and Drug Administration (2020) Consumer Alert on Regenerative Medicine Products Including Stem Cells and Exosomes. FDA, July.
https://www.fda.gov/vaccines-blood-biologics/consumers-biologics/consumer-alert-regenerative-medicine-products-including-stem-cells-and-exosomes
[27] Bartel, D.P. (2009) MicroRNAs: Target Recognition and Regulatory Functions. Cell, 136, 215-233.
https://doi.org/10.1016/j.cell.2009.01.002
[28] Bartel, D.P. (2018) Metazoan MicroRNAs. Cell, 173, 20-51.
https://doi.org/10.1016/j.cell.2018.03.006
[29] Bulut, Ö. and Gürsel, İ. (2020) Mesenchymal Stem Cell Derived Extracellular Vesicles: Promising Immunomodulators against Autoimmune, Autoinflammatory Disorders and SARS-CoV-2 Infection. Turkish Journal of Biology, 44, Article 14.
https://doi.org/10.3906/biy-2002-79
[30] Öztürk, S., Elçin, A.E. and Elçin, Y.M. (2020) Mesenchymal Stem Cells for Coronavirus (COVID-19)-Induced Pneumonia: Revisiting the Paracrine Hypothesis with New Hopes? Aging and Disease, 11, 477-479.
https://doi.org/10.14336/AD.2020.0403
[31] Schultz, I.C., Bertoni, A.P.S. and Wink, M.R. (2021) Mesenchymal Stem Cell-Derived Extracellular Vesicles Carrying miRNA as a Potential Multi Target Therapy to COVID-19: An in Silico Analysis. Stem Cell Reviews and Reports, 17, 341-356.
https://doi.org/10.1007/s12015-021-10122-0
[32] Brodin, P. (2021) Immune Determinants of COVID-19 Disease Presentation and Severity. Nature Medicine, 27, 28-33.
https://doi.org/10.1038/s41591-020-01202-8
[33] Liu, J., Lu, F., Chen, Y., Plow, E. and Qin, J. (2022) Integrin Mediates Cell Entry of the SARS-CoV-2 Virus Independent of Cellular Receptor ACE2. Journal of Biological Chemistry, 298, Article ID: 101710.
https://doi.org/10.1016/j.jbc.2022.101710
[34] Cantuti-Castelvetri, L., et al. (2020) Neuropilin-1 Facilitates SARS-CoV-2 Cell Entry and Infectivity. Science, 370, 856-860.
https://doi.org/10.1126/science.abd2985
[35] Hoffmann, M., Kleine-Weber, H., et al. (2020) SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell, 181, 271-280.E8.
https://doi.org/10.1016/j.cell.2020.02.052
[36] Jackson, C.B., Farzan, M., Chen, B. and Choe, H. (2022) Mechanisms of SARS-CoV-2 Entry into Cells. Nature Reviews Molecular Cell Biology, 23, 3-20.
https://doi.org/10.1038/s41580-021-00418-x
[37] Swanson, K.V., Deng, M. and Ting, J.P.-Y. (2019) The NLRP3 Inflammasome: Molecular Activation and Regulation to Therapeutics. Nature Reviews Immunology, 19, 477-489.
https://doi.org/10.1038/s41577-019-0165-0
[38] Man, S.M., Karki, R. and Kanneganti, T.-D. (2017) Molecular Mechanisms and Functions of Pyroptosis, Inflammatory Caspases and Inflammasomes in Infectious Diseases. Immunological Reviews, 277, 61-75.
https://doi.org/10.1111/imr.12534
[39] Garay-Sevilla, M.E., Rojas, A., Portero-Otin, M. and Uribarri, J. (2021) Dietary AGEs as Exogenous Boosters of Inflammation. Nutrients, 13, Article 2802.
https://doi.org/10.3390/nu13082802
[40] Ferreira, A.C., et al. (2021) SARS-CoV-2 Engages Inflammasome and Pyroptosis in Human Primary Monocytes. Cell Death Discovery, 7, Article No. 116.
https://doi.org/10.1038/s41420-021-00477-1
[41] Marchetti, C., Mould, K., Tengesdal, I.W., Janssen, W.J. and Dinarello, C.A. (2021) Targeting of the NLRP3 Inflammasome for Early COVID-19. bioRxiv.
https://doi.org/10.1101/2021.02.24.432734
[42] de Rivero Vaccari, J.C., Dietrich, W.D., Keane, R.W. and de Rivero Vaccari, J.P. (2020) The Inflammasome in Times of COVID-19. Frontiers in Immunology, 11, Article ID: 583373.
https://doi.org/10.3389/fimmu.2020.583373
[43] Cron, R.Q. (2021) COVID-19 Cytokine Storm: Targeting the Appropriate Cytokine. The Lancet Rheumatology, 3, e236-e237.
https://doi.org/10.1016/S2665-9913(21)00011-4
[44] Fajgenbaum D.C. and June, C.H. (2020) Cytokine Storm. New England Journal of Medicine, 383, 2255-2273.
https://doi.org/10.1056/NEJMra2026131
[45] Hojyo, S., et al. (2020) How COVID-19 Induces Cytokine Storm with High Mortality. Inflammation and Regeneration, 40, Article No. 37.
https://doi.org/10.1186/s41232-020-00146-3
[46] Lucas, C., et al. (2020) Longitudinal Analyses Reveal Immunological Misfiring in Severe COVID-19. Nature, 584, 463-469.
https://doi.org/10.1038/s41586-020-2588-y
[47] Morris, S.B., et al. (2020) Case Series of Multisystem Inflammatory Syndrome in Adults Associated with SARS-CoV-2 Infection—United Kingdom and United States, March-August 2020. Morbidity and Mortality Weekly Report (MMWR), 69, 1450-1456.
https://doi.org/10.15585/mmwr.mm6940e1
[48] Sokol, C.L. and Luster, A.D. (2015) The Chemokine System in Innate Immunity. Cold Spring Harbor Perspectives in Biology, 7, a016303.
https://doi.org/10.1101/cshperspect.a016303
[49] Tau, G. and Rothman, P. (1999) Biologic Functions of the IFN-Gamma Receptors. Allergy, 54, 1233-1251.
https://doi.org/10.1034/j.1398-9995.1999.00099.x
[50] Galani, I.-E., et al. (2021) Untuned Antiviral Immunity in COVID-19 Revealed by Temporal Type I/III Interferon Patterns and Flu Comparison. Nature Immunology, 22, 32-40.
https://doi.org/10.1038/s41590-020-00840-x
[51] Del Valle, D.M., et al. (2020) An Inflammatory Cytokine Signature Predicts COVID-19 Severity and Survival. Nature Medicine, 26, 1636-1643.
https://doi.org/10.1038/s41591-020-1051-9
[52] Huang, C., et al. (2020) Clinical Features of Patients Infected with 2019 Novel Coronavirus in Wuhan, China. The Lancet, 395, 497-506.
https://doi.org/10.1016/S0140-6736(20)30183-5
[53] Deshmane, S.L., Kremlev, S., Amini, S. and Sawaya, B.E. (2009) Monocyte Chemoattractant Protein-1 (MCP-1): An Overview. Journal of Interferon & Cytokine Research, 29, 313-326.
https://doi.org/10.1089/jir.2008.0027
[54] Jøntvedt Jørgensen, M., et al. (2020) Increased Interleukin-6 and Macrophage Chemoattractant Protein-1 Are Associated with Respiratory Failure in COVID-19. Scientific Reports, 10, Article No. 21697.
https://doi.org/10.1038/s41598-020-78710-7
[55] Hasan, A., Al-Ozairi, E., Al-Baqsumi, Z., Ahmad, R. and Al-Mulla, F. (2021) Cellular and Humoral Immune Responses in COVID-19 and Immunotherapeutic Approaches. ImmunoTargets and Therapy, 10, 63-85.
https://doi.org/10.2147/ITT.S280706
[56] Xu, Z., et al. (2020) Pathological Findings of COVID-19 Associated with Acute Respiratory Distress Syndrome. The Lancet Respiratory Medicine, 8, 420-422.
https://doi.org/10.1016/S2213-2600(20)30076-X
[57] Guan, W., et al. (2020) Clinical Characteristics of Coronavirus Disease 2019 in China. New England Journal of Medicine, 382, 1708-1720.
https://doi.org/10.1056/NEJMoa2002032
[58] Farshi, E., Kasmapur, B. and Arad, A. (2021) Investigation of Immune Cells on Elimination of Pulmonary-Infected COVID-19 and Important Role of Innate Immunity, Phagocytes. Reviews in Medical Virology, 31, e2158.
https://doi.org/10.1002/rmv.2158
[59] Kigi, D., et al. (1994) Cytotoxicity Mediated by T Cells and Natural Killer Cells Is Greatly Impaired in Perforin-Deficient Mice. Nature, 369, 31-37.
https://doi.org/10.1038/369031a0
[60] Jiang, Y., et al. (2020) COVID-19 Pneumonia: CD8+ T and NK Cells Are Decreased in Number but Compensatory Increased in Cytotoxic Potential. Clinical Immunology, 218, Article ID: 108516.
https://doi.org/10.1016/j.clim.2020.108516
[61] Wang, F., et al. (2020) The Laboratory Tests and Host Immunity of COVID-19 Patients with Different Severity of Illness. JCI Insight, 5, e137799.
https://doi.org/10.1172/jci.insight.137799
[62] Bobcakova, A., et al. (2021) Immune Profile in Patients with COVID-19: Lymphocytes Exhaustion Markers in Relationship to Clinical Outcome. Frontiers in Cellular and Infection Microbiology, 11, Article ID: 646688.
https://doi.org/10.3389/fcimb.2021.646688
[63] Zhao, Q., et al. (2020) Lymphopenia Is Associated with Severe Coronavirus Disease 2019 (COVID-19) Infections: A Systemic Review and Meta-Analysis. International Journal of Infectious Diseases, 96, 131-135.
https://doi.org/10.1016/j.ijid.2020.04.086
[64] Jiang, M., et al. (2020) T-Cell Subset Counts in Peripheral Blood Can Be Used as Discriminatory Biomarkers for Diagnosis and Severity Prediction of Coronavirus Disease 2019. The Journal of Infectious Diseases, 222, 198-202.
https://doi.org/10.1093/infdis/jiaa252
[65] Zheng, M., et al. (2020) Functional Exhaustion of Antiviral Lymphocytes in COVID-19 Patients. Cellular & Molecular Immunology, 17, 533-535.
https://doi.org/10.1038/s41423-020-0402-2
[66] Walls, A.C., Park, Y.-J., Tortorici, M.A., Wall, A., McGuire, A.T. and Veesler, D. (2020) Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell, 181, 281-292.e6.
https://doi.org/10.1016/j.cell.2020.02.058
[67] Sterlin, D., et al. (2021) IgA Dominates the Early Neutralizing Antibody Response to SARS-CoV-2. Science Translational Medicine, 13.
https://doi.org/10.1126/scitranslmed.abd2223
[68] Chansaenroj, J., et al. (2021) Detection of SARS-CoV-2-Specific Antibodies via Rapid Diagnostic Immunoassays in COVID-19 Patients. Virology Journal, 18, Article No. 52.
https://doi.org/10.1186/s12985-021-01530-2
[69] Dogan, M., et al. (2020) Novel SARS-CoV-2 Specific Antibody and Neutralization Assays Reveal Wide Range of Humoral Immune Response during COVID-19. medRxiv.
https://doi.org/10.1101/2020.07.07.20148106
[70] Evans, M.J. and Kaufman, M.H. (1981) Establishment in Culture of Pluripotential Cells from Mouse Embryos. Nature, 292, 154-156.
https://doi.org/10.1038/292154a0
[71] Dwivedi, S. and Sharma, P. (2018) Stem Cell Biology: A New Hope in Regenerations and Replenishments Therapy. Indian Journal of Clinical Biochemistry, 33, 369-371.
https://doi.org/10.1007/s12291-018-0792-4
[72] Farini, A., Sitzia, C., Erratico, S., Meregalli, M. and Torrente, Y. (2014) Clinical Applications of Mesenchymal Stem Cells in Chronic Diseases. Stem Cells International, 2014, Article ID: 306573.
https://doi.org/10.1155/2014/306573
[73] Joyce, N., Annett, G., Wirthlin, L., Olson, S., Bauer, G. and Nolta, J.A. (2010) Mesenchymal Stem Cells for the Treatment of Neurodegenerative Disease. Regenerative Medicine, 5, 933-946.
https://doi.org/10.2217/rme.10.72
[74] Wang, S., Qu, X. and Zhao, R.C. (2012) Clinical Applications of Mesenchymal Stem Cells. Journal of Hematology & Oncology, 5, Article No. 19.
https://doi.org/10.1186/1756-8722-5-19
[75] Wei, X., Yang, X., Han, Z., Qu, F., Shao, L. and Shi, Y. (2013) Mesenchymal Stem Cells: A New Trend for Cell Therapy. Acta Pharmacologica Sinica, 34, 747-754.
https://doi.org/10.1038/aps.2013.50
[76] Parekkadan, B. and Milwid, J.M. (2010) Mesenchymal Stem Cells as Therapeutics. Annual Review of Biomedical Engineering, 12, 87-117.
https://doi.org/10.1146/annurev-bioeng-070909-105309
[77] Kouris, N.A., et al. (2012) Directed Fusion of Mesenchymal Stem Cells with Cardiomyocytes via VSV-G Facilitates Stem Cell Programming. Stem Cells International, 2012, Article ID: 414038.
https://doi.org/10.1155/2012/414038
[78] Hofer, H.R. and Tuan, R.S. (2016) Secreted Trophic Factors of Mesenchymal Stem Cells Support Neurovascular and Musculoskeletal Therapies. Stem Cell Research & Therapy, 7, Article No. 131.
https://doi.org/10.1186/s13287-016-0394-0
[79] Jackson, M.V., 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.
https://doi.org/10.1002/stem.2372
[80] Robbins, P.D. and Morelli, A.E. (2014) Regulation of Immune Responses by Extracellular Vesicles. Nature Reviews Immunology, 14, 195-208.
https://doi.org/10.1038/nri3622
[81] Liang, B., et al. (2020) Clinical Remission of a Critically Ill COVID-19 Patient Treated by Human Umbilical Cord Mesenchymal Stem Cells. Medicine (Baltimore), 99, e21429.
https://doi.org/10.1097/MD.0000000000021429
[82] Sánchez-Guijo, F., et al. (2020) Adipose-Derived Mesenchymal Stromal Cells for the Treatment of Patients with Severe SARS-CoV-2 Pneumonia Requiring Mechanical Ventilation. A Proof of Concept Study. eClinicalMedicine, 25, Article ID: 100454.
https://doi.org/10.1016/j.eclinm.2020.100454
[83] Avanzini, M.A., et al. (2021) Human Mesenchymal Stromal Cells Do Not Express ACE2 and TMPRSS2 and Are Not Permissive to SARS-CoV-2 Infection. STEM CELLS Translational Medicine, 10, 636-642.
https://doi.org/10.1002/sctm.20-0385
[84] Schäfer, R., et al. (2021) Human Mesenchymal Stromal Cells Are Resistant to SARS-CoV-2 Infection under Steady-State, Inflammatory Conditions and in the Presence of SARS-CoV-2-Infected Cells. Stem Cell Reports, 16, 419-427.
https://doi.org/10.1016/j.stemcr.2020.09.003
[85] Sakaguchi, W., et al. (2020) Existence of SARS-CoV-2 Entry Molecules in the Oral Cavity. International Journal of Molecular Sciences, 21, Article 6000.
https://doi.org/10.3390/ijms21176000
[86] Durand, N., Russell, A. and Zubair, A.C. (2019) Effect of Comedications and Endotoxins on Mesenchymal Stem Cell Secretomes, Migratory and Immunomodulatory Capacity. Journal of Clinical Medicine, 8, Article 497.
https://doi.org/10.3390/jcm8040497
[87] Russell, A.L., Lefavor, R., Durand, N., Glover, L. and Zubair, A.C. (2018) Modifiers of Mesenchymal Stem Cell Quantity and Quality. Transfusion, 58, 1434-1440.
https://doi.org/10.1111/trf.14597
[88] Shu, L., et al. (2020) Treatment of Severe COVID-19 with Human Umbilical Cord Mesenchymal Stem Cells. Stem Cell Research & Therapy, 11, Article No. 361.
https://doi.org/10.1186/s13287-020-01875-5
[89] de Witte, S.F.H., et al. (2018) Immunomodulation BY Therapeutic Mesenchymal Stromal Cells (MSC) Is Triggered Through Phagocytosis of MSC By Monocytic Cells. Stem Cells, 36, 602-615.
https://doi.org/10.1002/stem.2779
[90] Hu, C. and Li, L. (2019) The Immunoregulation of Mesenchymal Stem Cells Plays a Critical Role in Improving the Prognosis of Liver Transplantation. Journal of Translational Medicine, 17, Article No. 412.
https://doi.org/10.1186/s12967-019-02167-0
[91] Golchin, A., Seyedjafari, E. and Ardeshirylajimi, A. (2020) Mesenchymal Stem Cell Therapy for COVID-19: Present or Future. Stem Cell Reviews and Reports, 16, 427-433.
https://doi.org/10.1007/s12015-020-09973-w
[92] Lu, Z., et al. (2019) Mesenchymal Stem Cells Induce Dendritic Cell Immune Tolerance via Paracrine Hepatocyte Growth Factor to Alleviate Acute Lung Injury. Stem Cell Research & Therapy, 10, Article No. 372.
https://doi.org/10.1186/s13287-019-1488-2
[93] Park, M., Kim, Y.-H., Ryu, J.-H., Woo, S.-Y. and Ryu, K.-H. (2015) Immune Suppressive Effects of Tonsil-Derived Mesenchymal Stem Cells on Mouse Bone-Marrow-Derived Dendritic Cells. Stem Cells International, 2015, Article ID: 106540.
https://doi.org/10.1155/2015/106540
[94] Spaggiari, G.M. and Moretta, L. (2013) Interactions between Mesenchymal Stem Cells and Dendritic Cells. In: Weyand, B., Dominici, M., Hass, R., Jacobs, R. and Kasper, C., Eds., Mesenchymal Stem Cells—Basics and Clinical Application II, Springer, Berlin, Heidelberg, 199-208.
https://doi.org/10.1007/10_2012_154
[95] Raffaghello, L., et al. (2008) Human Mesenchymal Stem Cells Inhibit Neutrophil Apoptosis: A Model for Neutrophil Preservation in the Bone Marrow Niche. Stem Cells, 26, 151-162.
https://doi.org/10.1634/stemcells.2007-0416
[96] Lee, S., Zhang, Q.Z., Karabucak, B. and Le, A.D. (2016) DPSCs from Inflamed Pulp Modulate Macrophage Function via the TNF-α/IDO Axis. Journal of Dental Research, 95, 1274-1281.
https://doi.org/10.1177/0022034516657817
[97] Zhang, Y., Chen, Y. and Meng, Z. (2020) Immunomodulation for Severe COVID-19 Pneumonia: The State of the Art. Frontiers in Immunology, 11, Article ID: 577442.
https://doi.org/10.3389/fimmu.2020.577442
[98] Loke, X.Y., Imran, S.A.M., Tye, G.J., Wan Kamarul Zaman, W.S. and Nordin, F. (2021) Immunomodulation and Regenerative Capacity of MSCs for Long-COVID. International Journal of Molecular Sciences, 22, Article ID: 12421.
https://doi.org/10.3390/ijms222212421
[99] Tsyb, A.F., et al. (2008) In Vitro Inhibitory Effect of Mesenchymal Stem Cells on Zymosan-Induced Production of Reactive Oxygen Species. Bulletin of Experimental Biology and Medicine, 146, 158-164.
https://doi.org/10.1007/s10517-008-0238-8
[100] Vasandan, A.B., Jahnavi, S., Shashank, C., Prasad, P., Kumar, A. and Prasanna, S.J. (2016) Human Mesenchymal Stem Cells Program Macrophage Plasticity by Altering Their Metabolic Status via a PGE 2-Dependent Mechanism. Scientific Reports, 6, Article No. 38308.
https://doi.org/10.1038/srep38308
[101] Le Blanc, K. and Davies, L.C. (2015) Mesenchymal Stromal Cells and the Innate Immune Response. Immunology Letters, 168, 140-146.
https://doi.org/10.1016/j.imlet.2015.05.004
[102] Chen, B., Ni, Y., Liu, J., Zhang, Y. and Yan, F. (2018) Bone Marrow-Derived Mesenchymal Stem Cells Exert Diverse Effects on Different Macrophage Subsets. Stem Cells International, 2018, Article ID: 8348121.
https://doi.org/10.1155/2018/8348121
[103] Fan, L., Hu, C., Chen, J., Cen, P., Wang, J. and Li, L. (2016) Interaction between Mesenchymal Stem Cells and B-Cells. International Journal of Molecular Sciences, 17, Article 650.
https://doi.org/10.3390/ijms17050650
[104] Jiang, X.-X., et al. (2005) Human Mesenchymal Stem Cells Inhibit Differentiation and Function of Monocyte-Derived Dendritic Cells. Blood, 105, 4120-4126.
https://doi.org/10.1182/blood-2004-02-0586
[105] Melief, S.M., et al. (2013) Multipotent Stromal Cells Induce Human Regulatory T Cells through a Novel Pathway Involving Skewing of Monocytes toward Anti-Inflammatory Macrophages. Stem Cells, 31, 1980-1991.
https://doi.org/10.1002/stem.1432
[106] Deng, Y., et al. (2016) Umbilical Cord-Derived Mesenchymal Stem Cells Instruct Monocytes towards an IL10-producing Phenotype by Secreting IL6 and HGF. Scientific Reports, 6, Article No. 37566.
https://doi.org/10.1038/srep37566
[107] Allavena, P., et al. (1998) IL-10 Prevents the Differentiation of Monocytes to Dendritic Cells but Promotes Their Maturation to Macrophages. European Journal of Immunology, 28, 359-369.
https://doi.org/10.1002/(SICI)1521-4141(199801)28:01<359::AID-IMMU359>3.0.CO;2-4
[108] Liu, J., et al. (2020) Apoptotic Bodies Derived from Mesenchymal Stem Cells Promote Cutaneous Wound Healing via Regulating the Functions of Macrophages. Stem Cell Research & Therapy, 11, Article No. 507.
https://doi.org/10.1186/s13287-020-02014-w
[109] Martínez-Aguilar, R., Romero-Pinedo, S., Ruiz-Magaña, M.J., Olivares, E.G., Ruiz-Ruiz, C. and Abadía-Molina, A.C. (2020) Menstrual Blood-Derived Stromal Cells Modulate Functional Properties of Mouse and Human Macrophages. Scientific Reports, 10, Article No. 21389.
https://doi.org/10.1038/s41598-020-78423-x
[110] Nguyen, H.-Q.-D., Kao, C.-Y., Chiang, C.-P., Hung, Y.-H. and Lo, C.-M. (2022) Investigating the Immunomodulatory Potential of Dental Pulp Stem Cell Cultured on Decellularized Bladder Hydrogel towards Macrophage Response in Vitro. Gels, 8, Article 187.
https://doi.org/10.3390/gels8030187
[111] Fathi, E., Farahzadi, R., Valipour, B. and Sanaat, Z. (2019) Cytokines Secreted from Bone Marrow Derived Mesenchymal Stem Cells Promote Apoptosis and Change Cell Cycle Distribution of K562 Cell Line as Clinical Agent in Cell Transplantation. PLOS ONE, 14, e0215678.
https://doi.org/10.1371/journal.pone.0215678
[112] Gu, Y., et al. (2015) Mesenchymal Stem Cells Suppress Neuronal Apoptosis and Decrease IL-10 Release via the TLR2/NFκB Pathway in Rats with Hypoxic-Ischemic Brain Damage. Molecular Brain, 8, Article No. 65.
https://doi.org/10.1186/s13041-015-0157-3
[113] Ahmed, N.E.-M.B., Murakami, M., Hirose, Y. and Nakashima, M. (2016) Therapeutic Potential of Dental Pulp Stem Cell Secretome for Alzheimer’s Disease Treatment: An in Vitro Study. Stem Cells International, 2016, Article ID: 8102478.
https://doi.org/10.1155/2016/8102478
[114] Ferlazzo, G. and Carrega, P. (2012) Natural Killer Cell Distribution and Trafficking in Human Tissues. Frontiers in Immunology, 3, Article 347.
https://doi.org/10.3389/fimmu.2012.00347
[115] Paul, S. and Lal, G. (2017) The Molecular Mechanism of Natural Killer Cells Function and Its Importance in Cancer Immunotherapy. Frontiers in Immunology, 8, Article 1124.
https://doi.org/10.3389/fimmu.2017.01124
[116] Giuliani, M., et al. (2011) Human Mesenchymal Stem Cells Derived from Induced Pluripotent Stem Cells Down-Regulate NK-Cell Cytolytic Machinery. Blood, 118, 3254-3262.
https://doi.org/10.1182/blood-2010-12-325324
[117] Reinders, M.E. and Hoogduijn, M.J. (2014) NK Cells and MSCs: Possible Implications for MSC Therapy in Renal Transplantation. Journal of Stem Cell Research & Therapy, 4, Article ID: 1000166.
[118] Spaggiari, G.M., Capobianco, A., Abdelrazik, H., Becchetti, F., Mingari, M.C. and Moretta, L. (2008) Mesenchymal Stem Cells Inhibit Natural Killer-Cell Proliferation, Cytotoxicity, and Cytokine Production: Role of Indoleamine 2,3-Dioxygenase and Prostaglandin E2. Blood, 111, 1327-1333.
https://doi.org/10.1182/blood-2007-02-074997
[119] Cui, R., et al. (2016) Human Mesenchymal Stromal/Stem Cells Acquire Immunostimulatory Capacity upon Cross-Talk with Natural Killer Cells and Might Improve the NK Cell Function of Immunocompromised Patients. Stem Cell Research & Therapy, 7, Article No. 88.
https://doi.org/10.1186/s13287-016-0353-9
[120] Sotiropoulou, P.A., Perez, S.A., Gritzapis, A.D., Baxevanis, C.N. and Papamichail, M. (2006) Interactions between Human Mesenchymal Stem Cells and Natural Killer Cells. Stem Cells, 24, 74-85.
https://doi.org/10.1634/stemcells.2004-0359
[121] Yan, F., et al. (2019) Human Dental Pulp Stem Cells Regulate Allogeneic NK Cells’ Function via Induction of Anti-Inflammatory Purinergic Signalling in Activated NK Cells. Cell Proliferation, 52, e12595.
https://doi.org/10.1111/cpr.12595
[122] Maucourant, C., et al. (2020) Natural Killer Cell Immunotypes Related to COVID-19 Disease Severity. Science Immunology, 5, eabd6832.
https://doi.org/10.1126/sciimmunol.abd6832
[123] Bartholomew, A., et al. (2002) Mesenchymal Stem Cells Suppress Lymphocyte Proliferation in Vitro and Prolong Skin Graft Survival in Vivo. Experimental Hematology, 30, 42-48.
https://doi.org/10.1016/S0301-472X(01)00769-X
[124] Beyth, S., et al. (2005) Human Mesenchymal Stem Cells Alter Antigen-Presenting Cell Maturation and Induce T-Cell Unresponsiveness. Blood, 105, 2214-2219.
https://doi.org/10.1182/blood-2004-07-2921
[125] Haddad, R. and Saldanha-Araujo, F. (2014) Mechanisms of T-Cell Immunosuppression by Mesenchymal Stromal Cells: What Do We Know So Far? BioMed Research International, 2014, Article ID: 216806.
https://doi.org/10.1155/2014/216806
[126] Laing, A.G., Fanelli, G., Ramirez-Valdez, A., Lechler, R.I., Lombardi, G. and Sharpe, P.T. (2019) Mesenchymal Stem Cells Inhibit T-Cell Function through Conserved Induction of Cellular Stress. PLOS ONE, 14, e0213170.
https://doi.org/10.1371/journal.pone.0213170
[127] Najar, M., et al. (2009) Mesenchymal Stromal Cells Promote or Suppress the Proliferation of T Lymphocytes from Cord Blood and Peripheral Blood: The Importance of Low Cell Ratio and Role of Interleukin-6. Cytotherapy, 11, 570-583.
https://doi.org/10.1080/14653240903079377
[128] Martin-Rufino, J.D., Espinosa-Lara, N., Osugui, L. and Sanchez-Guijo, F. (2019) Targeting the Immune System with Mesenchymal Stromal Cell-Derived Extracellular Vesicles: What Is the Cargo’s Mechanism of Action? Frontiers in Bioengineering and Biotechnology, 7, Article 308.
https://doi.org/10.3389/fbioe.2019.00308
[129] Wang, L., et al. (2021) Regulation of Inflammatory Cytokine Storms by Mesenchymal Stem Cells. Frontiers in Immunology, 12, Article ID: 726909.
https://doi.org/10.3389/fimmu.2021.726909
[130] van Megen, K.M., van’t Wout, E.-J.T., Lages Motta, J., Dekker, B., Nikolic, T. and Roep, B.O. (2019) Activated Mesenchymal Stromal Cells Process and Present Antigens Regulating Adaptive Immunity. Frontiers in Immunology, 10, Article 694.
https://doi.org/10.3389/fimmu.2019.00694
[131] Kyurkchiev, D., et al. (2014) Secretion of Immunoregulatory Cytokines by Mesenchymal Stem Cells. World Journal of Stem Cells, 6, 552-570.
https://doi.org/10.4252/wjsc.v6.i5.552
[132] Moll, G., Hoogduijn, M.J. and Ankrum, J.A. (2020) Safety, Efficacy and Mechanisms of Action of Mesenchymal Stem Cell Therapies. Frontiers in Immunology, 11, Article 243.
https://doi.org/10.3389/978-2-88963-600-6
[133] Terraza-Aguirre, C., et al. (2020) Mechanisms behind the Immunoregulatory Dialogue between Mesenchymal Stem Cells and Th17 Cells. Cells, 9, Article 1660.
https://doi.org/10.3390/cells9071660
[134] Tatara, R., et al. (2011) Mesenchymal Stromal Cells Inhibit Th17 but Not Regulatory T-Cell Differentiation. Cytotherapy, 13, 686-694.
https://doi.org/10.3109/14653249.2010.542456
[135] Favaro, E., et al. (2016) Human Mesenchymal Stem Cells and Derived Extracellular Vesicles Induce Regulatory Dendritic Cells in Type 1 Diabetic Patients. Diabetologia, 59, 325-333.
https://doi.org/10.1007/s00125-015-3808-0
[136] Chen, Q.-H., et al. (2020) Mesenchymal Stem Cells Regulate the Th17/Treg Cell Balance Partly through Hepatocyte Growth Factor in Vitro. Stem Cell Research & Therapy, 11, Article No. 91.
https://doi.org/10.1186/s13287-020-01612-y
[137] de Cássia Noronha, N., et al. (2019) Priming Approaches to Improve the Efficacy of Mesenchymal Stromal Cell-Based Therapies. Stem Cell Research & Therapy, 10, Article No. 131.
https://doi.org/10.1186/s13287-019-1224-y
[138] Luk, F., et al. (2017) Inflammatory Conditions Dictate the Effect of Mesenchymal Stem or Stromal Cells on B Cell Function. Frontiers in Immunology, 8, Article 1042.
https://doi.org/10.3389/fimmu.2017.01042
[139] Luz-Crawford, P., et al. (2016) Mesenchymal Stem Cell-Derived Interleukin 1 Receptor Antagonist Promotes Macrophage Polarization and Inhibits B Cell Differentiation. Stem Cells, 34, 483-492.
https://doi.org/10.1002/stem.2254
[140] Corcione, A., et al. (2006) Human Mesenchymal Stem Cells Modulate B-Cell Functions. Blood, 107, 367-372.
https://doi.org/10.1182/blood-2005-07-2657
[141] Ding, G., Niu, J. and Liu, Y. (2015) Dental Pulp Stem Cells Suppress the Proliferation of Lymphocytes via Transforming Growth Factor-β1. Human Cell, 28, 81-90.
https://doi.org/10.1007/s13577-014-0106-y
[142] Feng, Y., et al. (2020) Safety and Feasibility of Umbilical Cord Mesenchymal Stem Cells in Patients with COVID-19 Pneumonia: A Pilot Study. Cell Proliferation, 53, e12947.
https://doi.org/10.1111/cpr.12947
[143] Park, B.-W., et al. (2020) In Vivo Priming of Human Mesenchymal Stem Cells with Hepatocyte Growth Factor-Engineered Mesenchymal Stem Cells Promotes Therapeutic Potential for Cardiac Repair. Science Advances, 6, eaay6994.
https://doi.org/10.1126/sciadv.aay6994
[144] Raza, S.S., Seth, P. and Khan, M.A. (2022) ‘Primed’ Mesenchymal Stem Cells: A Potential Novel Therapeutic for COVID19 Patients. Stem Cell Reviews and Reports, 17, 153-162.
https://doi.org/10.1007/s12015-020-09999-0
[145] Bustos, M.L., et al. (2013) Activation of Human Mesenchymal Stem Cells Impacts Their Therapeutic Abilities in Lung Injury by Increasing Interleukin (IL)-10 and IL-1RN Levels. STEM CELLS Translational Medicine, 2, 884-895.
https://doi.org/10.5966/sctm.2013-0033
[146] Xu, A.L., et al. (2019) Mesenchymal Stem Cells Reconditioned in Their Own Serum Exhibit Augmented Therapeutic Properties in the Setting of Acute Respiratory Distress Syndrome. STEM CELLS Translational Medicine, 8, 1092-1106.
https://doi.org/10.1002/sctm.18-0236
[147] Giunti, D., et al. (2021) Role of miRNAs Shuttled by Mesenchymal Stem Cell-Derived Small Extracellular Vesicles in Modulating Neuroinflammation. Scientific Reports, 11, Article No. 1740.
https://doi.org/10.1038/s41598-021-81039-4
[148] Öztürk, S., Elçin, A.E., Koca, A. and Elçin, Y.M. (2020) Therapeutic Applications of Stem Cells and Extracellular Vesicles in Emergency Care: Futuristic Perspectives. Stem Cell Reviews and Reports, 17, 390-410.
https://doi.org/10.1007/s12015-020-10029-2
[149] Askenase, P.W. (2020) COVID-19 Therapy with Mesenchymal Stromal Cells (MSC) and Convalescent Plasma Must Consider Exosome Involvement: Do the Exosomes in Convalescent Plasma Antagonize the Weak Immune Antibodies? Journal of Extracellular Vesicles, 10, e12004.
https://doi.org/10.1002/jev2.12004
[150] Jayaramayya, K., et al. (2020) Immunomodulatory Effect of Mesenchymal Stem Cells and Mesenchymal Stem-Cell-Derived Exosomes for COVID-19 Treatment. BMB Reports, 53, 400-412.
https://doi.org/10.5483/BMBRep.2020.53.8.121
[151] Wiklander, O.P.B., Brennan, M.á., Lötvall, J., Breakefield, X.O. and El Andaloussi, S. (2019) Advances in Therapeutic Applications of Extracellular Vesicles. Science Translational Medicine, 11, aav852.
https://doi.org/10.1126/scitranslmed.aav8521
[152] Chung, M.-J., et al. (2020) Mesenchymal Stem Cell and MicroRNA Therapy of Musculoskeletal Diseases. International Journal of Stem Cells, 14, 150-167.
https://doi.org/10.15283/ijsc20167
[153] Zayed, M. and Iohara, K. (2020) Immunomodulation and Regeneration Properties of Dental Pulp Stem Cells: A Potential Therapy to Treat Coronavirus Disease 2019. Cell Transplantation, 29.
https://doi.org/10.1177/0963689720952089
[154] Cui, S.J., et al. (2020) DPSCs Attenuate Experimental Progressive TMJ Arthritis by Inhibiting the STAT1 Pathway. Journal of Dental Research, 99, 446-455.
https://doi.org/10.1177/0022034520901710
[155] Matsumura-Kawashima, M., Ogata, K., Moriyama, M., Murakami, Y., Kawado, T. and Nakamura, S. (2021) Secreted Factors from Dental Pulp Stem Cells Improve Sjögren’s Syndrome via Regulatory T Cell-Mediated Immunosuppression. Stem Cell Research & Therapy, 12, Article No. 182.
https://doi.org/10.1186/s13287-021-02236-6
[156] Ogata, K., Matsumura-Kawashima, M., Moriyama, M., Kawado, T. and Nakamura, S. (2021) Dental Pulp-Derived Stem Cell-Conditioned Media Attenuates Secondary Sjögren’s Syndrome via Suppression of Inflammatory Cytokines in the Submandibular Glands. Regenerative Therapy, 16, 73-80.
https://doi.org/10.1016/j.reth.2021.01.006
[157] Fujii, Y., et al. (2018) Bone Regeneration by Human Dental Pulp Stem Cells Using a Helioxanthin Derivative and Cell-Sheet Technology. Stem Cell Research & Therapy, 9, Article No. 24.
https://doi.org/10.1186/s13287-018-0783-7
[158] Lam, C., et al. (2021) Human Dental Pulp Stem Cells (DPSCs) Therapy in Rescuing Photoreceptors and Establishing a Sodium Iodate-Induced Retinal Degeneration Rat Model. Tissue Engineering and Regenerative Medicine, 18, 143-154.
https://doi.org/10.1007/s13770-020-00312-1
[159] Zhang, Y., et al. (2021) DPSCs Treated by TGF-β1 Regulate Angiogenic Sprouting of Three-Dimensionally Co-Cultured HUVECs and DPSCs through VEGF-Ang-Tie2 Signaling. Stem Cell Research & Therapy, 12, Article No. 281.
https://doi.org/10.1186/s13287-021-02349-y
[160] Hossein-Khannazer, N., Hashemi, S.M., Namaki, S., Ghanbarian, H., Sattari, M. and Khojasteh, A. (2019) Study of the Immunomodulatory Effects of Osteogenic Differentiated Human Dental Pulp Stem Cells. Life Sciences, 216, 111-118.
https://doi.org/10.1016/j.lfs.2018.11.040
[161] Pierdomenico, L., et al. (2005) Multipotent Mesenchymal Stem Cells with Immunosuppressive Activity Can Be Easily Isolated from Dental Pulp. Transplantation, 80, 836-842.
https://doi.org/10.1097/01.tp.0000173794.72151.88
[162] Nakashima, M., Iohara, K. and Murakami, M. (2013) Dental Pulp Stem Cells and Regeneration. Endodontic Topics, 28, 38-50.
https://doi.org/10.1111/etp.12027
[163] Monsel, A., et al. (2022) Treatment of COVID-19-Associated ARDS with Mesenchymal Stromal Cells: A Multicenter Randomized Double-Blind Trial. Critical Care, 26, Article No. 48.
https://doi.org/10.1186/s13054-022-03930-4
[164] Peng, H., et al. (2020) A Synergistic Role of Convalescent Plasma and Mesenchymal Stem Cells in the Treatment of Severely Ill COVID-19 Patients: A Clinical Case Report. Stem Cell Research & Therapy, 11, Article No. 291.
https://doi.org/10.1186/s13287-020-01802-8
[165] Guo, Z., Chen, Y., Luo, X., He, X., Zhang, Y. and Wang, J. (2020) Administration of Umbilical Cord Mesenchymal Stem Cells in Patients with Severe COVID-19 Pneumonia. Critical Care, 24, Article No. 420.
https://doi.org/10.1186/s13054-020-03142-8
[166] Shi, L., et al. (2020) Treatment with Human Umbilical Cord-Derived Mesenchymal stem Cells for COVID-19 Patients with Lung Damage: A Randomised, Double-Blind, Placebo-Controlled Phase 2 Trial. medRxiv.
https://doi.org/10.1101/2020.10.15.20213553
[167] Hashemian, S.-M.R., et al. (2021) Mesenchymal Stem Cells Derived from Perinatal Tissues for Treatment of Critically Ill COVID-19-Induced ARDS Patients: A Case Series. Stem Cell Research & Therapy, 12, Article No. 91.
https://doi.org/10.1186/s13287-021-02165-4
[168] Häberle, H., et al. (2021) Mesenchymal Stem Cell Therapy for Severe COVID-19 ARDS. Journal of Intensive Care Medicine, 36, 681-688.
https://doi.org/10.1177/0885066621997365
[169] Tao, J., et al. (2020) Umbilical Cord Blood-Derived Mesenchymal Stem Cells in Treating a Critically Ill COVID-19 Patient. The Journal of Infection in Developing Countries, 14, 1138-1145.
https://doi.org/10.3855/jidc.13081
[170] Tang, L., et al. (2020) Clinical Study Using Mesenchymal Stem Cells for the Treatment of Patients with Severe COVID-19. Frontiers in Medicine, 14, 664-673.
https://doi.org/10.1007/s11684-020-0810-9
[171] COVID-19 Treatment Guidelines. Clinical Spectrum of SARS-CoV-2 Infection.
https://www.covid19treatmentguidelines.nih.gov/overview/clinical-spectrum/

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.