Next Article in Journal
Optimization of Antibacterial Activity and Biosafety through Ultrashort Peptide/Cyclodextrin Inclusion Complexes
Next Article in Special Issue
Following Natural Autoantibodies: Further Immunoserological Evidence Regarding Their Silent Plasticity and Engagement in Immune Activation
Previous Article in Journal
Protective Role of Ethanol Extract of Cibotium barometz (Cibotium Rhizome) against Dexamethasone-Induced Muscle Atrophy in C2C12 Myotubes
Previous Article in Special Issue
The Local Anaesthetic Procaine Prodrugs ProcCluster® and Procaine Hydrochloride Impair SARS-CoV-2 Replication and Egress In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mesenchymal Stem Cells in the Treatment of COVID-19

1
Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
2
Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
3
School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
4
Department of Emergency Medicine, Tungs’ Taichung Metro Harbor Hospital, Taichung 43503, Taiwan
5
Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan
6
Department of Pediatrics, Taichung Veterans General Hospital, Taichung 43503, Taiwan
7
Laboratory of Epidemiology and Biostastics, Changhua Christian Hospital, Changhua 50006, Taiwan
8
Department of Emergency Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua 50544, Taiwan
9
Division of Cardiology, Department of Medicine, Taichung Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan
10
Department of Medicine, College of Medicine, Tzu Chi University, Hualien 97002, Taiwan
11
College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
12
Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(19), 14800; https://doi.org/10.3390/ijms241914800
Submission received: 28 August 2023 / Revised: 21 September 2023 / Accepted: 29 September 2023 / Published: 30 September 2023
(This article belongs to the Special Issue COVID-19 Pandemic: Therapeutic Strategies and Vaccines)

Abstract

:
Since the emergence of the coronavirus disease 2019 (COVID-19) pandemic, many lives have been tragically lost to severe infections. The COVID-19 impact extends beyond the respiratory system, affecting various organs and functions. In severe cases, it can progress to acute respiratory distress syndrome (ARDS) and multi-organ failure, often fueled by an excessive immune response known as a cytokine storm. Mesenchymal stem cells (MSCs) have considerable potential because they can mitigate inflammation, modulate immune responses, and promote tissue regeneration. Accumulating evidence underscores the efficacy and safety of MSCs in treating severe COVID-19 and ARDS. Nonetheless, critical aspects, such as optimal routes of MSC administration, appropriate dosage, treatment intervals, management of extrapulmonary complications, and potential pediatric applications, warrant further exploration. These research avenues hold promise for enriching our understanding and refining the application of MSCs in confronting the multifaceted challenges posed by COVID-19.

1. Introduction

Since 2020, the relentless grip of the coronavirus disease 2019 (COVID-19) has profoundly shaped people’s lives worldwide, causing extensive global devastation [1,2]. As of 13 September 2023, 770,563,467 cases of COVID-19 have been confirmed and countless lives lost [3,4]. The COVID-19 virus (SARS-CoV-2) primarily spreads through large respiratory droplets, airborne particles, and fecal–oral and surface contact [5,6]. These modes of transmission significantly contribute to the rapid and difficult-to-control spread of the virus, despite the implementation of various government response policies in many countries. Consequently, the global community has actively dedicated itself to identifying effective methods for preventing and treating the disease. COVID-19 vaccines have been developed to prevent the spread of the disease and end the pandemic [7]. Significant progress has also been made in developing antiviral drugs, which have proven instrumental in treating the disease [8,9]. However, their effectiveness against COVID-19 has declined with the emergence of new virus variants [10]. COVID-19 can be categorized into mild, moderate, severe, and critical [11]. In mild and moderate COVID-19 cases, supportive care, oxygen therapy, and antiviral treatment are important for treatment [12,13]. However, in patients with severe and critical COVID-19, hyperinflammation and a cytokine storm resulting from a dysregulated host innate immune response often occurs [14,15], leading to the potential development of acute respiratory distress syndrome, septic shock, multiple organ damage, and even death [16,17,18,19,20]. In addition to the traditional supportive care for viral diseases, various strategies have been proposed to address the dysregulated immune response and mitigate the inflammatory cascade of COVID-19.
Immunotherapy-related agents, such as glucocorticoids and anti-interleukin-6 (IL-6) receptor antibodies, have effectively modulated the immune response and reduced excessive inflammation in these patients [21]. These treatments have demonstrated promising results in reducing mortality among individuals affected by the disease [22,23,24,25]. However, it is important to consider the potential risk of secondary infections associated with these therapies [26,27].
Unlike immunosuppressants, mesenchymal stem cells (MSCs) possess a unique combination of properties, including the ability to reduce inflammation, immunomodulation, and regenerative capabilities [28]. COVID-19 primarily affects the respiratory tract and can potentially lead to the development of acute respiratory distress syndrome (ARDS), cardiac injury, acute kidney injury, and multi-organ failure, often triggered by a cytokine storm [29,30]. MSC-based therapy has been proposed as a promising option for managing COVID-19 due to its ability to significantly suppress cytokine storms, leading to improved clinical symptoms and higher survival rates [31]. In this review, our objective is to investigate clinical trials related to MSCs in the treatment of COVID-19, as published in the scientific literature. We aim to provide a comprehensive summary that covers clinical symptoms, biomarkers associated with the cytokine storm, and lung imaging within the context of MSC therapy for COVID-19. Additionally, we will include a summary of the pathology and organ damage aspects. Furthermore, we will discuss potential future directions in this field.

2. Pathology and Vital Organ Damage in COVID-19

2.1. Mechanisms of Entry and Exit of COVID-19

The pathogenesis of COVID-19 is complex. The coronavirus virion comprises four structural proteins: envelope (E), membrane (M), nucleocapsid (N), and spike (S) [32]. The viral S protein binds to the ACE2 receptor via the receptor-binding domain (RBD) of the S1 subunit, facilitating viral entry into various human host cells, including those in the vascular endothelium, lungs, heart, brain, kidneys, intestines, liver, and pharynx [33,34,35,36]. Type II alveolar epithelial cells are considered to be the cells most predisposed to viral infections [37]. Upon RBD-receptor interaction, the S protein undergoes proteolytic cleavage by enzymes such as furin, or cellular proteases such as transmembrane serine protease 2 (TMPRSS2) in both the S1 and S2 subunits [33,38,39,40,41]. This cleavage exposes the fusion peptide of the S2 protein, enabling the S2 subunit trimers to fuse directly with the host cell membrane [42]. This process leads to deposition of the virus-positive RNA genome into the host cell. Another entry route that the virus may use is through the endosome. This occurs when the target cell expresses insufficient TMPRSS2 or if the virus-ACE2 complex does not encounter TMPRSS2. In such cases, the virus-ACE2 complex is internalized via clathrin-mediated endocytosis into endolysosomes, where S2’ cleavage is performed by cathepsins. Cathepsins require an acidic environment for their activity [33,43]. Furthermore, various membrane proteins can act as ACE2 cofactors or alternative receptors, enabling SARS-CoV-2 entry into different cell types, even those that do not typically express ACE2 [44]. However, their specific roles in SARS-CoV-2 pathogenesis remain unclear [45].
After entering the host cells, the viral genome hijacks the host cell machinery to initiate replication and translation. The molecular machinery of the host cell reads viral genetic material, leading to the synthesis of viral proteins. Among these proteins are the envelope glycoproteins processed within the Golgi apparatus [33]. When virus-containing vesicles fuse with the plasma membrane, assembled viruses are released into the extracellular space. This process occurs outside the cell.

2.2. Cytokine Storm and Severity of COVID-19

SARS-CoV-2 primarily infects type 2 pneumocytes that express ACE2 receptors in the alveoli and trigger an immune response characterized by the production of inflammatory cytokines and a weak interferon (IFN) response [46]. After cytokine release, membrane-bound immune receptors and downstream signaling pathways play crucial roles in mediating the proinflammatory immune responses of pathogenic Th1 cells and intermediate CD14+CD16+ monocytes. This process promotes the infiltration of macrophages and neutrophils into lung tissue, contributing to the cytokine storm’s onset [47]. Proinflammatory cytokines, including granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-6, are secreted by pathogenic Th1 cells. GM-CSF then activates CD14+CD16+ monocytes, leading them to produce significant quantities of IL-6, tumor necrosis factor-alpha (TNF-α), and other cytokines [48]. Neutrophil extracellular traps, which are extracellular nets released by neutrophils, can potentially contribute to the release of cytokines [49]. Cytokine storms are primarily characterized by the overexpression of IL-6 and TNF-α, two proinflammatory cytokines that play a significant role in the exaggerated immune response observed in severe cases of the disease.
In severe cases of COVID-19, multiple studies have indicated higher levels of IL-2, IL-6, IL-7, IL-10, CXCL10(IP-10), CCL2 (MCP-1), TNF-α, CCL3(MIP-1α), and G-CSF when compared to patients with mild and moderate infections [14,21,50,51,52,53]. Furthermore, increased levels of proinflammatory cytokines have been observed in patients with COVID-19 [54]. Patients experiencing severe COVID-19 demonstrate significant and sustained decreases in lymphocyte counts, particularly CD4+ and CD8+ T cells, but show increases in neutrophil counts when compared to those with milder cases [50,55]. The loss of T cells may worsen specific pathological inflammatory responses during SARS-CoV-2 infection [56], whereas the restoration of T cells could potentially alleviate inflammatory responses during SARS-CoV-2 infection [51]. Thus, T-cell lymphopenia, and dynamic cytokine storm are associated with COVID-19 severity [57,58]. Therefore, these findings highlight the need for clinicians to identify patients at risk of developing severe COVID-19 as early as possible through monitoring the dynamic cytokine storms.

3. Organ Damage of COVID-19

The severity of COVID-19 encompasses a broad spectrum, ranging from asymptomatic cases to mild presentations characterized by upper respiratory symptoms, such as fever, cough, sneezing, fatigue, and sore throat. This progression can lead to moderate illness with pneumonia, and in severe cases, individuals may experience dyspnea and hypoxemia, resulting in blood oxygen levels falling below the critical threshold of 94% [59,60]. The impact of the virus extends beyond the lungs and can damage other organs [61,62,63]. The pulmonary and extrapulmonary manifestations of COVID-19 are presented in Table 1. In more critical scenarios, COVID-19 can cause a range of severe conditions, including acute respiratory distress syndrome, shock, encephalopathy, heart failure, acute kidney injury, coagulopathy, and multiple organ dysfunction [60,64]. Cytokine storms are widely recognized as significant contributors to the development of multi-organ failure in COVID-19 [65].

3.1. Lung Involvement in COVID-19

SARS-CoV-2 primarily targets the lungs, and its severity ranges from asymptomatic to respiratory failure or death [66]. Inhaled viral particles of COVID-19 are primarily deposited in the nasal mucosa, where the virus infects and replicates within the epithelial target cells [67]. Infection leads to alveolar injury and inflammation in the lung tissue [68]. Dendritic cells (DCs) and alveolar macrophages phagocytose virus-infected apoptotic epithelial cells, initiating T-cell responses that activate both innate and adaptive immune mechanisms [69].
One critical aspect of COVID-19 pathology is the cytokine storm, characterized by an excessive release of proinflammatory cytokines and chemokines like TNF-α, IL-1β, and IL-6 [29]. This immune response contributes to immunopathology, leading to lung damage and, in some cases, immune suppression characterized by reduced T-cell counts and increased vulnerability to bacterial infections [70,71].
Autopsy studies of patients with COVID-19 pneumonia revealed acute interstitial pneumonia and diffuse alveolar damage. The lung tissue showed macrophage infiltration, hyaline membrane formation, and alveolar wall edema. Microvascular involvement includes hyaline thrombosis, hemorrhage, vessel wall edema, and immune cell infiltration [72]. Lung-restricted vascular immunopathology, also known as “diffuse pulmonary intravascular coagulopathy” (PIC), is associated with COVID-19, particularly in severe cases accompanied by ARDS [73].
An interesting feature of COVID-19 pneumonia is “silent hypoxia,” wherein patients tolerate low oxygen levels without dyspnea [74,75]. This unique characteristic distinguishes it from typical ARDS and is attributed to preserved lung compliance and high air volume with hypocapnia [75,76]. The disease presents with two time-associated phenotypes. Early disease stages show an L-phenotype with high lung compliance and a low Va/Q ratio. The lung weight and recruitment were low. Progression to the H phenotype includes decreased compliance, edema, and shunting. Type L patients can improve or worsen, transitioning to type H owing to COVID-19 pneumonia and high-stress ventilation [77,78].

3.2. Cardiovascular System Involvement in COVID-19

COVID-19 patients with hypertension, diabetes, and cardiovascular disease experience worse outcomes [79,80]. Cardiovascular complications, such as myocardial injury, heart failure, and arrhythmias are associated with poor survival [81,82]. Obesity is also a predictor of adverse cardiovascular outcomes in COVID-19 [83]. Myocardial injury is a notable finding in COVID-19 patients and is believed to be facilitated by elevated ACE2 expression in the cardiac tissue, allowing for direct virus-induced damage [84,85]. Isolated cases of COVID-19-induced myocarditis have been reported, suggesting that direct myocardial injury caused by the virus is a possible mechanism [86,87]. The binding of SARS-CoV-2 to ACE2 may lead to downregulation of ACE2, potentially compromising heart function [88]. COVID-19 can induce a hyper-inflammatory state with increased levels of inflammatory cytokines, rendering atherosclerotic lesions more vulnerable to disruption and increasing the risk of acute coronary syndrome [89,90]. The direct effect of the virus on endothelial cells and the host inflammatory response can lead to endothelial dysfunction in various organs, including the heart [91]. Myocardial injury can result from a mismatch between oxygen supply and demand, known as type 2 myocardial infarction [92]. Hypotension in sepsis and cytokine storms reduces myocardial perfusion [93], whereas infection and fever increase myocardial cell demand [94,95].
Fulminant myocarditis may occur as a manifestation of COVID-19 and lead to left ventricular dysfunction or cardiogenic shock [96]. Heart failure and arrhythmias are more prevalent in patients with severe COVID-19, particularly critically ill patients [97,98]. In addition, Kawasaki-like syndrome, characterized by circulatory dysfunction and macrophage activation, has been reported in some COVID-19 patients [99], suggesting a potential predisposing mechanism associated with the virus’s cytokine storm.

3.3. Gastrointestinal Tract and Liver Involvement in COVID-19

Digestive symptoms in COVID-19 patients, including diarrhea, nausea, vomiting, and abdominal pain, are likely linked to the affinity of the virus for ACE2 receptors found abundantly in the GI tract, including the intestine, liver, and pancreas [100,101]. These symptoms can be the primary manifestations in COVID-19 patients, potentially leading to delayed diagnosis owing to their nonspecific nature [102,103]. SARS-CoV-2 shedding has been detected in the stool of COVID-19 patients, and as with most enteric viruses, it could be a potential fecal–oral transmission route [104].
Mild and transient liver injury, as well as severe liver damage, can occur during COVID-19 [105]. However, the mechanism of liver injury is not fully understood and can be caused by direct viral infection of hepatocytes, immune-related injury, or drug hepatotoxicity [106]. A systematic review and meta-analysis found that patients with severe COVID-19 have a higher risk of developing gastrointestinal symptoms and liver injury than those with non-severe disease [107].

3.4. Hematological Involvement in COVID-19

COVID-19 is a systemic infection that significantly affects the hematopoietic system and homeostasis. The cytokine storm is likely a key factor underlying the observed lymphopenia [108]. Lymphopenia is more frequent in moderate and severe COVID-19 cases, making it a valid marker of disease severity and mortality [108,109]. Neutrophilia has been identified as a predictor of poor outcomes in COVID-19 because severe disease is linked to an increased neutrophil-to-lymphocyte ratio and elevated expression of the neutrophil-related cytokines IL-8 and IL-6 in the serum [110,111]. COVID-19-induced severe lung inflammation may impair platelet production in the lungs because the lungs possess potential hematopoietic functions and serve as a primary site for terminal platelet production, accounting for approximately half of the total platelet production [110,112]. Thrombocytopenia is associated with an increased risk of severe disease and mortality in patients with COVID-19 [113,114]. Therefore, it should be considered a crucial clinical indicator of worsening illness during hospitalization [113]. Elevated D-dimer levels and coagulation abnormalities indicate blood hypercoagulability, particularly in severe COVID-19 cases [115]. Biomarkers such as high serum procalcitonin, C-reactive protein (CRP), and ferritin levels at presentation have emerged as poor prognostic factors associated with severe disease and poor survival [116]. COVID-19 is associated with a high risk of venous thromboembolism [117], and prompt thromboprophylaxis with low-molecular-weight heparin is recommended [118].

3.5. Neurological Involvement in COVID-19

SARS-CoV-2 can infiltrate the nervous system through two main routes: directly by utilizing the olfactory nerve pathway or by compromising the blood–brain barrier (BBB); and indirectly by triggering a vigorous immune response resulting in systemic inflammation or hypoxia [119]. COVID-19 can lead to neurological symptoms affecting the central nervous system (CNS), peripheral nervous system (PNS), and skeletal muscles in approximately one third of patients [120,121]. Common symptoms include dizziness, headache, taste impairment, anosmia, impaired consciousness, and nerve pain [122]. Severe cases of COVID-19 have a higher prevalence of neurological symptoms than milder cases [123]. Encephalopathy is a devastating CNS complication frequently encountered in older and critically ill patients. It arises from hypoxic/metabolic changes caused by cytokine storms and has severe consequences [124]. Moreover, patients with severe disease or individuals with vascular risk factors, such as hypertension, diabetes mellitus, or dyslipidemia, are at an increased risk of acute cerebrovascular diseases, primarily acute ischemic stroke [125]. Additionally, patients with stroke and concurrent COVID-19 were found to have a significantly higher risk of severe disability and death than those without COVID-19 [126].

3.6. Kidney Involvement in COVID-19

In COVID-19, kidney-related symptoms such as proteinuria and dipstick hematuria are not uncommon [127,128] and have been identified as significant predictors of severe or critically severe cases [127].
Acute kidney injury (AKI) affects approximately 30% of hospitalized COVID-19 patients and is one of the most frequent extrapulmonary complications [129]. Kidney damage induced by SARS-CoV-2 is believed to be multifactorial. ACE2, a receptor in various kidney cells, plays a significant role in COVID-19-related kidney abnormalities [130]. Other factors, including macrophage activation syndrome, cytokine storm, lymphopenia, endothelial dysfunction, organ interactions, hypercoagulability, rhabdomyolysis, and sepsis, can also contribute to AKI development [131]. The rate of AKI is higher in severe COVID-19 cases [132,133], and patients in the intensive care unit (ICU) show elevated levels of inflammatory cytokines, suggesting a potential association with cytokine release syndrome (CRS) leading to kidney injury [53].

3.7. Endocrine Involvement in COVID-19

Patients with diabetes mellitus or obesity have an increased risk of developing severe manifestations of COVID-19 [83,134]. The infection caused by SARS-CoV-2 can trigger a hyperglycemic state and potentially lead to ketoacidosis in both diabetic and non-diabetic patients, resulting in critical outcomes [135,136]. Notably, ACE2 expression has been identified in the endocrine pancreas [137], suggesting that SARS-CoV-2 directly binds to ACE2 on-cells, potentially contributing to insulin deficiency and hyperglycemia [138]. SARS-CoV-2-related factors, including increased cytokine levels, may adversely affect pancreatic β-cell function and induce apoptosis. This can result in diminished insulin production and the onset of ketosis [139]. Well-managed diabetic condition has been correlated with a significantly reduced risk of mortality and the need for invasive mechanical ventilation in individuals with COVID-19 [140].

4. MSCs in COVID-19 Treatment

4.1. Source and Immunomodulation of MSCs

MSCs, a type of versatile stem cells, can be sourced from various tissues of both adult and neonatal origin. These tissues include the bone marrow (BM), adipose tissue (AT), and peripheral blood [91] from adults, as well as neonatal birth-associated tissues, such as the umbilical cord (UC), cord blood (CB), and placenta (PL) [141]. Derived from the ethereal realm of fetal tissues, particularly the umbilical cord, MSCs have emerged as a true spectacle and have the advantage of rapid proliferation and enhanced immunomodulatory properties. This sets them apart from other sources, sidestepping any inconveniences that might arise while procuring MSCs from the bone marrow or adipose tissue [142].
MSCs possess differentiative and regenerative attributes and can secrete factors, such as hepatocyte growth factor, vascular endothelial growth factor, and keratinocyte growth factor. These molecules play crucial roles in regenerating type II alveolar epithelial cells [143]. Moreover, MSCs can be drawn to sites of inflammation through diverse chemokines and can regulate the activities of various immune cells, including NK cells, dendritic cells, B cells, T cells, neutrophils, and macrophages. This modulation occurs via direct and paracrine mechanisms. The major effectors of this process include indoleamine 2,3-dioxygenase, transforming growth factor β, human leukocyte antigen isoforms, and prostaglandin E2 [144]. Thus, MSCs possess immunomodulatory properties, can reduce immunity, and may offer a therapeutic option for patients with severe or critical COVID-19 to suppress overactivated inflammatory responses.

4.2. Effectiveness of MSC Treatment in Other Diseases

MSCs have demonstrated significant clinical efficacy in the treatment of various immune disorders. In animal studies, human umbilical cord-derived MSCs have shown promising results in alleviating cytokine storms and reducing lung damage in mice with LPS-induced acute lung injury (ALI) [145]. Furthermore, a review highlighted the positive impact of MSC therapy on survival rates, hematopoietic reconstitution, and the recovery of peripheral blood cells in animal models of aplastic anemia [146]. In clinical practice, MSCs have been successfully used to treat numerous immune disorders [147], including inflammatory bowel disease [148,149,150], systemic lupus erythematosus [151,152,153], graft-versus-host disease [154,155], or multiple sclerosis [156,157,158]. MSCs exert significant effects on tumor growth and treatment response, making them promising candidates for cancer treatment, either alone or in combination with other therapies [159,160]. MSCs have the potential to effectively treat severe COVID-19 and its complications.

4.3. Current Outcomes of Clinical Trials of MSCs in COVID-19 Treatment

Numerous clinical trials involving MSCs have demonstrated their effectiveness in treating COVID-19 and its associated complications. Currently, over 100 registered clinical trials have investigated the use of MSCs for COVID-19 treatment [161]. Most of these trials focused on severely ill COVID-19 patients. Nevertheless, a phase I clinical study has provided evidence of the safety of both high and low doses of DW-MSC infusion in patients with non-severe COVID-19 [162]. To substantiate the therapeutic efficacy of MSCs in these patients, large-scale randomized controlled trials could be imperative for definitive confirmation. Our review encompassed more than 20 clinical trials, the details of which are presented in Table 2. It is important to note that all the aforementioned clinical trials have demonstrated the safety of MSC treatment. The effectiveness of the MSC treatment in terms of clinical symptoms, biomarkers, and medical imaging in clinical trials is discussed below, with summary provided in Table 3.

4.3.1. Effectiveness of MSC Treatment in Clinical Symptoms

MSC therapy has shown promising outcomes in reducing clinical symptoms in patients with severe COVID-19. As stated by Shu et al. [163], individuals who underwent treatment with umbilical cord mesenchymal stem cells (UC-MSC) experienced enhancements in clinical symptoms, such as weakness, fatigue, shortness of breath, and improved oxygenation index as early as the third day of treatment. Prenatal MSCs derived from umbilical cord (UC-MSC) or placental (PL-MSC) tissues can be utilized to treat critically ill patients with COVID-19-induced ARDS, resulting in reduced dyspnea and increased SpO2 within 2–4 days after the initial infusion in 64% of patients [164]. A multicenter randomized, double-blind trial unveiled a significant increase in PaO2/FiO2 ratios in the UC-MSC group compared to the placebo group in COVID-19-associated ARDS [165]. In a clinical trial using human menstrual blood-derived mesenchymal stromal cells for the treatment of severe and critically ill COVID-19 patients, significant improvement in dyspnea after MSC infusion on days 1, 3, and 5 and significant improvements in SpO2 and PaO2 following MSC infusion were noted. Additionally, patients in the MSC group showed significantly lower mortality (7.69% in the experimental group vs. 33.33% in the control group; p = 0.048) [166]. Farkhad et al. demonstrated that mesenchymal stromal cell therapy could improve the SPO2/FIO2 ratio in COVID-19-induced ARDS patients [167]. Sadeghi et al. showed that in COVID-19-induced ARDS cases treated with placenta-derived decidual stromal cells, there was a noticeable improvement in oxygenation levels, with a median increase from 80.5% (range, 69–88) to 95% (range, 78–99) (p = 0.012) [168].
Lanzoni et al. [169] reported improved patient survival and a shorter time to recovery after administering two rounds of intravenous allogeneic UC-MSC to patients with ARDS. In a phase I/II study involving patients with severe COVID-19, survival rates were notably higher in the MSC group at both 28 and 60 days after BM-MSC treatment (both p values < 0.05) [170]. In Indonesia, a randomized controlled trial demonstrated a survival rate 2.5 times higher in the UC-MSC group than in the control group in critical COVID-19 cases [171]. Fathi-Kazerooni et al. demonstrated that the survival rate in severe COVID-19 patients treated with human Mesenchymal Stromal Cells was significantly higher than that of patients who received placebo treatment (p < 0.001) [172].
Following a 2-year monitoring period for severe COVID-19 patients subjected to UC-MSC treatment, a slightly smaller subset of individuals within the MSC-treated group exhibited a 6 min walking distance (6-MWD) falling below the lower boundary of the normal range in comparison to the placebo-administered group (OR = 0.19, 95% CI: 0.04–0.80, Fisher’s exact test, p = 0.015). Furthermore, at month 18, the MSC-treated group demonstrated a higher general health score on the Short Form 36 questionnaire than the placebo group (MSC group: 50.00, placebo group: 35.00) (95% CI: 0.00–20.00, Wilcoxon rank sum test, p = 0.018) [173].

4.3.2. Effectiveness of MSC Treatment in Biomarkers Related to Cytokine Storm

Many inflammatory biomarkers and cytokines are closely associated with severe and critical COVID-19. Notably, significant improvements in the levels of these biomarkers were observed after MSC treatment. Shu et al. [163] reported that CRP and IL-6 levels significantly decreased starting from day three, and the time required for lymphocyte counts to return to the normal range was notably shorter in patients with severe COVID-19 following UC-MSC infusion. In a study by Lanzoni et al. [169], significantly lower concentrations of GM-CSF, IFN-r, IL-5, IL-6, IL-7, TNF-a, and TNF-b were observed in COVID-19-related acute respiratory distress syndrome. Meng et al. [174] showed an improvement in the percentage of inspired oxygen (PaO2/FiO2) ratio and a declining trend in levels of inflammatory cytokines, including high levels of IL-6, IFN-γ, TNF-α, MCP-1, interferon-inducible cytokine IP-10 (IP-10), IL-22, interleukin 1 receptor type 1 (IL-1RA), IL-18, IL-8, and macrophage inflammatory protein 1-alpha (MIP-1), after three rounds of UC-MSC treatment. Fathi-Kazerooni et al. conducted a study that showed that in severe COVID-19 patients treated with human mesenchymal stromal cells, the levels of CRP on day five were notably lower than those in patients who underwent placebo treatment (p < 0.03). Additionally, within the treatment group of critical COVID-19 patients, significant reductions in CRP, LDH, D-dimer, and ferritin levels were observed after MSC treatment (all p < 0.05) [172]. Sadeghi et al. demonstrated that in cases of COVID-19-induced ARDS treated with placenta-derived decidua stromal cells, there were significant reductions in levels of IL-6, with a median decrease from 69.3 (range 35.0–253.4) to 11 (range 4.0–38.3) pg/mL, and CRP, with a median decrease from 69 (range 5–169) to 6 (range 2–31) mg/mL (p = 0.028) [168]. After aerosol inhalation of exosomes derived from human adipose-derived MSCs (haMSC-Exos) in COVID-19 patients, a pilot study reported an increase in lymphocyte counts and a decrease in CRP, LDH, and IL-6 levels [175].
In a case series study, considerable reductions in the serum levels of TNF-α, IL-8, and CRP were evident in all six critically ill COVID-19-induced ARDS survivors [164]. In a randomized clinical trial involving critically ill COVID-19 patients who underwent mesenchymal stromal cell infusion, the UC-MSC group exhibited reduced ferritin, IL-6, and MCP1-CCL2 levels on the fourteenth day. In the second month, decreases in reactive C-protein, D-dimer, and neutrophil levels were observed, along with increases in the numbers of TCD3, TCD4, and NK lymphocytes [176]. In a phase I/II clinical trial, patients with severe COVID-19 treated with BM-MSCs exhibited significantly lower D-dimer levels on day seven than control patients [170].
An Indonesian randomized controlled trial revealed that the infusion of UC-MSC led to a significant decrease in IL-6 levels in recovered COVID-19 patients with ARDS (p = 0.023) [171]. A randomized controlled trial showed that MSC infusion was associated with a decrease in inflammatory cytokines, such as IL-6 (p = 0.015), TNF-α (p = 0.034), IFN-γ (p = 0.024), and CRP (p = 0.041) in ARDS COVID-19 patient [177]. A prospective double-controlled trial revealed that after adding MSCs transplantation therapy to treat critical COVID-19 patients in the ICU, serum ferritin, fibrinogen, and CRP levels significantly decreased compared to conventional therapy alone [178]. In a successful phase 1 clinical trial with a control-placebo group, a noteworthy reduction (p < 0.05) was noted in biomarkers such as CRP, IL-6, IFN-γ, TNF-α, and IL-17A in COVID-19-induced ARDS patients. Conversely, there was a significant increase in the serum levels of TGF-B, IL-1B, and IL-10 [167]. In a phase 1 clinical trial, a generalized estimating equation analysis showed a significant decrease in ferritin levels (p = 0.008) after MSC treatment in COVID-19 patients [179].

4.3.3. Effectiveness of MSC Treatment in Lung Image

Among COVID-19 patients, the most frequently observed computed tomography (CT) findings include ground-glass opacification, infiltration, consolidation, pneumonia, and emphysematous changes [185]. After MSC treatment in COVID-19 patients, many clinical trials have shown improved lung change. Shu et al. [163] reported shorter lung inflammation absorption on CT imaging in patients with severe COVID-19 in the UC-MSC group than in the control group. In a clinical trial involving a case series, lung CT revealed remarkable indications for recovery, including a reduction in ground-glass opacities or consolidations, among patients with COVID-19-induced ARDS [164]. Chest CT images showed complete fading of lung lesions within 2 weeks in moderate COVID-19 patients after UC-MSCs transfusion in the study by Meng et al. [174]. In a randomized, double-blind, placebo-controlled phase 2 trial, UC-MSCs led to a significant reduction in the proportions of solid component lesion volume compared to the placebo group in patients with COVID-19-induced ARDS (p = 0.043) [180]. A decrease in the extent of lung damage was observed in the fourth month after three rounds mesenchymal stromal cell infusion in critical COVID-19 patients in a randomized clinical trial [176]. Different degrees of resolution of pulmonary lesions after aerosol inhalation of haMSC-Exos were observed in patients with COVID-19 in a pilot study [175]. Xu et al. demonstrated a noteworthy distinction between experimental and control groups concerning the improvement rate of chest CT findings during the initial month following MSC infusion in individuals with severe and critical COVID-19 [166]. Fathi-Kazerooni et al. showed that in severe COVID-19 patients treated with human mesenchymal stromal cells, the percentage of pulmonary involvement exhibited a significant improvement in the group receiving secretome treatment (p < 0.0001) [172]. Sadeghi et al. demonstrated that all pulmonary infiltrates disappeared in patients with COVID-19-induced ARDS treated with placenta-derived decidual stromal cells [168].

5. Future Prospect of MSC Treatment in COVID-19 Patients

MSCs are administered via multiple routes. The intravenous delivery of MSCs could potentially lead to their entrapment in the pulmonary microcirculatory system, allowing for selective homing to injured lobes in the lungs as well as their distribution to extrapulmonary organs [62,186,187,188]. Other administration routes, including intratracheal, intranasal, subcutaneous, and pulmonary artery, have also been explored for delivering stem cells to treat COVID-19 [189]. These local delivery offers several advantages, including prolonged cell half-life, increased utilization efficiency, and reduced off-target effects on other organs [189]. Intratracheal administration of MSCs has been shown to increase cell concentrations in the lungs and demonstrated efficacy in preclinical models of respiratory diseases [190,191]. However, it is important to consider that this route of administration may also increase the risk of virus spread in the case of COVID-19 [192]. Therapeutic intravenous doses of MSC per treatment ranged from 5 × 105 cells/kg body weight to 2 × 108 cells/dose from different studies (Table 2). A clinical trial has demonstrated that the dose of MSCs for the treatment of COVID-19 can be as high as 8 × 108 cells/kg/dose (clinical trial number: NCT04366323). Given the dose-dependent nature of the immunosuppressive effects and the documented safety of various cell doses, a higher MSC dosage may be warranted for critically ill patients [142]. However, the implementation of higher dosages requires meticulous clinical trial evaluation to determine the potential risks and benefits. For COVID-19, two to three courses of MSC therapy may be appropriate, with additional courses if insufficient recovery is observed [164,169,174,180,193]. Previous studies have proposed a 3-day interval between each course of MSC treatment during the acute phase [169,174,180]. However, the optimal cell dose, treatment duration, and interval protocols for MSC administration in COVID-19 management remain unclear. It is essential to conduct thorough research and develop guidelines to address these gaps and uncertainties.
The efficacy of MSCs in treating COVID-19-related conditions, including ARDS and severe cases in adults, has been well documented. However, there remains a need for further investigation into the potential of MSCs to address extrapulmonary complications, such as cardiovascular dysfunction and kidney injury in COVID-19 patients. Additionally, exploring their application in treating pediatric cases is crucial to expand our understanding of their therapeutic utility, underscoring the potential of MSCs in managing bronchopulmonary dysplasia in newborns and suggesting that MSCs hold promise in reducing lung inflammation, enhancing pulmonary architecture, mitigating pulmonary fibrosis, and ultimately improving survival rates [194]. Another study demonstrated encouraging results in managing acute graft-versus-host disease in pediatric patients, illustrating the versatility of MSCs in pediatric care [195]. In a phase I/II randomized, placebo-controlled clinical trial, intravenous transplantation of MSCs significantly improved HbA1c and C-peptide levels in pediatric patients. Furthermore, this treatment modulates the balance between proinflammatory and anti-inflammatory cytokines, highlighting its potential therapeutic impact [196]. Future research efforts should prioritize the evaluation of the effects of MSC-based cell therapy in COVID-19 patients, particularly in terms of restoring organ functionality and reducing mortality rates within this subgroup. Additionally, there is a need to investigate the efficacy of MSC-based therapy in pediatric patients with severe COVID-19.
By pursuing these avenues, we can deepen our understanding of the diverse therapeutic capacities of MSCs in various aspects of COVID-19 management. This endeavor holds significant promise for advancing our ability to effectively address the multifaceted challenges posed by the disease.

6. Conclusions

MSCs demonstrated significant therapeutic potential owing to their ability to attenuate inflammation, modulate immune responses, and facilitate tissue regeneration. MSCs have the potential to effectively treat severe COVID-19 and its complications. In our review, we conducted extensive research by reviewing dozens of clinical studies related to MSC treatment for COVID-19. We categorized these studies into clinical symptoms, cytokine storm biomarkers, and lung imaging for discussion. Additionally, we explored the prospects of MSC treatment in this context. These clinical trials have generated evidence showcasing the effectiveness of MSCs in addressing various facets of severe COVID-19 and COVID-19-related ARDS. These trials have yielded positive outcomes by improving clinical symptoms, enhancing survival rates, optimizing oxygen saturation levels, influencing cytokine storm-related biomarkers, and positively affecting lung imaging results. In the future, there may be a need to delve deeper into several areas. Investigating optimal administration routes, determining appropriate cell dosages, defining treatment intervals for MSC therapy, understanding the impact of severe extrapulmonary damage treatment, and exploring the potential applications of MSCs in pediatric cases are important aspects that warrant further investigation. These research directions hold promise for refining our understanding and utilization of MSCs as a therapeutic approach against the multifaceted challenges posed by COVID-19.

Author Contributions

Conceptualization, W.-Y.L., Y.-J.C. and T.-A.L.; writing—original draft, B.-C.G. and K.-H.W.; writing—review and editing, H.-P.W. and C.-Y.C.; supervision, H.-P.W. and M.-J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Muralidar, S.; Ambi, S.V.; Sekaran, S.; Krishnan, U.M. The emergence of COVID-19 as a global pandemic: Understanding the epidemiology, immune response and potential therapeutic targets of SARS-CoV-2. Biochimie 2020, 179, 85–100. [Google Scholar] [CrossRef] [PubMed]
  2. Clemente-Suárez, V.J.; Navarro-Jiménez, E.; Moreno-Luna, L.; Saavedra-Serrano, M.C.; Jimenez, M.; Simón, J.A.; Tornero-Aguilera, J.F. The impact of the COVID-19 pandemic on social, health, and economy. Sustainability 2021, 13, 6314. [Google Scholar] [CrossRef]
  3. WHO. Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int (accessed on 13 September 2023).
  4. Herby, J.; Jonung, L.; Hanke, S. A literature review and meta-analysis of the effects of lockdowns on covid-19 mortality-II. 2022. Stud. Appl. Econ. 2022, 200. [Google Scholar]
  5. Zhao, X.; Liu, S.; Yin, Y.; Zhang, T.; Chen, Q. Airborne transmission of COVID-19 virus in enclosed spaces: An overview of research methods. Indoor Air 2022, 32, e13056. [Google Scholar] [CrossRef] [PubMed]
  6. Yeo, C.; Kaushal, S.; Yeo, D. Enteric involvement of coronaviruses: Is faecal–oral transmission of SARS-CoV-2 possible? Lancet Gastroenterol. Hepatol. 2020, 5, 335–337. [Google Scholar] [CrossRef]
  7. Ndwandwe, D.; Wiysonge, C.S. COVID-19 vaccines. Curr. Opin. Immunol. 2021, 71, 111–116. [Google Scholar] [CrossRef] [PubMed]
  8. Burki, T.K. The role of antiviral treatment in the COVID-19 pandemic. Lancet Respir. Med. 2022, 10, e18. [Google Scholar] [CrossRef]
  9. Vegivinti, C.T.R.; Evanson, K.W.; Lyons, H.; Akosman, I.; Barrett, A.; Hardy, N.; Kane, B.; Keesari, P.R.; Pulakurthi, Y.S.; Sheffels, E. Efficacy of antiviral therapies for COVID-19: A systematic review of randomized controlled trials. BMC Infect. Dis. 2022, 22, 107. [Google Scholar] [CrossRef]
  10. Ferdinands, J.M.; Rao, S.; Dixon, B.E.; Mitchell, P.K.; DeSilva, M.B.; Irving, S.A.; Lewis, N.; Natarajan, K.; Stenehjem, E.; Grannis, S.J. Waning of vaccine effectiveness against moderate and severe COVID-19 among adults in the US from the VISION network: Test negative, case-control study. BMJ 2022, 379. [Google Scholar] [CrossRef]
  11. Feng, Y.; Ling, Y.; Bai, T.; Xie, Y.; Huang, J.; Li, J.; Xiong, W.; Yang, D.; Chen, R.; Lu, F. COVID-19 with different severities: A multicenter study of clinical features. Am. J. Respir. Crit. Care Med. 2020, 201, 1380–1388. [Google Scholar] [CrossRef]
  12. Richardson, S.; Hirsch, J.S.; Narasimhan, M.; Crawford, J.M.; McGinn, T.; Davidson, K.W.; Barnaby, D.P.; Becker, L.B.; Chelico, J.D.; Cohen, S.L. Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City area. JAMA 2020, 323, 2052–2059. [Google Scholar] [CrossRef] [PubMed]
  13. Gandhi, R.T.; Lynch, J.B.; Del Rio, C. Mild or moderate Covid-19. N. Engl. J. Med. 2020, 383, 1757–1766. [Google Scholar] [CrossRef] [PubMed]
  14. Gustine, J.N.; Jones, D. Immunopathology of Hyperinflammation in COVID-19. Am. J. Pathol. 2021, 191, 4–17. [Google Scholar] [CrossRef]
  15. Blot, M.; Bour, J.-B.; Quenot, J.P.; Bourredjem, A.; Nguyen, M.; Guy, J.; Monier, S.; Georges, M.; Large, A.; Dargent, A. The dysregulated innate immune response in severe COVID-19 pneumonia that could drive poorer outcome. J. Transl. Med. 2020, 18, 1–14. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan, China. JAMA 2020, 323, 1061–1069. [Google Scholar] [CrossRef]
  17. Mathew, D.; Giles, J.R.; Baxter, A.E.; Oldridge, D.A.; Greenplate, A.R.; Wu, J.E.; Alanio, C.; Kuri-Cervantes, L.; Pampena, M.B.; D’Andrea, K. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 2020, 369, eabc8511. [Google Scholar] [CrossRef]
  18. Kuri-Cervantes, L.; Pampena, M.B.; Meng, W.; Rosenfeld, A.M.; Ittner, C.A.; Weisman, A.R.; Agyekum, R.S.; Mathew, D.; Baxter, A.E.; Vella, L.A. Comprehensive mapping of immune perturbations associated with severe COVID-19. Sci. Immunol. 2020, 5, eabd7114. [Google Scholar] [CrossRef]
  19. Song, J.-W.; Zhang, C.; Fan, X.; Meng, F.-P.; Xu, Z.; Xia, P.; Cao, W.-J.; Yang, T.; Dai, X.-P.; Wang, S.-Y. Immunological and inflammatory profiles in mild and severe cases of COVID-19. Nat. Commun. 2020, 11, 3410. [Google Scholar] [CrossRef]
  20. Chen, Q.; Yu, B.; Yang, Y.; Huang, J.; Liang, Y.; Zhou, J.; Li, L.; Peng, X.; Cheng, B.; Lin, Y. Immunological and inflammatory profiles during acute and convalescent phases of severe/critically ill COVID-19 patients. Int. Immunopharmacol. 2021, 97, 107685. [Google Scholar] [CrossRef]
  21. Zanza, C.; Romenskaya, T.; Manetti, A.C.; Franceschi, F.; La Russa, R.; Bertozzi, G.; Maiese, A.; Savioli, G.; Volonnino, G.; Longhitano, Y. Cytokine storm in COVID-19: Immunopathogenesis and therapy. Medicina 2022, 58, 144. [Google Scholar] [CrossRef]
  22. Sterne, J.A.; Murthy, S.; Diaz, J.V.; Slutsky, A.S.; Villar, J.; Angus, D.C.; Annane, D.; Azevedo, L.C.P.; Berwanger, O.; Cavalcanti, A.B. Association between administration of systemic corticosteroids and mortality among critically ill patients with COVID-19: A meta-analysis. JAMA 2020, 324, 1330–1341. [Google Scholar] [PubMed]
  23. El-Saber Batiha, G.; Al-Gareeb, A.I.; Saad, H.M.; Al-Kuraishy, H.M. COVID-19 and corticosteroids: A narrative review. Inflammopharmacology 2022, 30, 1189–1205. [Google Scholar] [CrossRef] [PubMed]
  24. Potere, N.; Batticciotto, A.; Vecchié, A.; Porreca, E.; Cappelli, A.; Abbate, A.; Dentali, F.; Bonaventura, A. The role of IL-6 and IL-6 blockade in COVID-19. Expert Rev. Clin. Immunol. 2021, 17, 601–618. [Google Scholar] [CrossRef] [PubMed]
  25. Ghosn, L.; Assi, R.; Evrenoglou, T.; Buckley, B.S.; Henschke, N.; Probyn, K.; Riveros, C.; Davidson, M.; Graña, C.; Bonnet, H. Interleukin-6 blocking agents for treating COVID-19: A living systematic review. Cochrane Database Syst. Rev. 2023, 6, CD013881. [Google Scholar] [CrossRef]
  26. Obata, R.; Maeda, T.; Rizk, D.; Kuno, T. Increased secondary infection in COVID-19 patients treated with steroids in New York City. Jpn. J. Infect. Dis. 2021, 74, 307–315. [Google Scholar] [CrossRef]
  27. Elahi, R.; Karami, P.; Heidary, A.H.; Esmaeilzadeh, A. An updated overview of recent advances, challenges, and clinical considerations of IL-6 signaling blockade in severe coronavirus disease 2019 (COVID-19). Int. Immunopharmacol. 2022, 105, 108536. [Google Scholar] [CrossRef]
  28. Wu, X.; Jiang, J.; Gu, Z.; Zhang, J.; Chen, Y.; Liu, X. Mesenchymal stromal cell therapies: Immunomodulatory properties and clinical progress. Stem Cell Res. Ther. 2020, 11, 1–16. [Google Scholar] [CrossRef]
  29. Montazersaheb, S.; Hosseiniyan Khatibi, S.M.; Hejazi, M.S.; Tarhriz, V.; Farjami, A.; Ghasemian Sorbeni, F.; Farahzadi, R.; Ghasemnejad, T. COVID-19 infection: An overview on cytokine storm and related interventions. Virol. J. 2022, 19, 1–15. [Google Scholar] [CrossRef]
  30. Mustafa, M.I.; Abdelmoneim, A.H.; Mahmoud, E.M.; Makhawi, A.M. Cytokine Storm in COVID-19 Patients, Its Impact on Organs and Potential Treatment by QTY Code-Designed Detergent-Free Chemokine Receptors. Mediat. Inflamm. 2020, 2020, 8198963. [Google Scholar] [CrossRef]
  31. Fang, Y.; Lao, P.; Tang, L.; Chen, J.; Chen, Y.; Sun, L.; Tan, W.; Fan, X. Mechanisms of Potential Therapeutic Utilization of Mesenchymal Stem Cells in COVID-19 Treatment. Cell Transplant. 2023, 32, 9636897231184611. [Google Scholar] [CrossRef]
  32. Troyano-Hernáez, P.; Reinosa, R.; Holguín, Á. Evolution of SARS-CoV-2 Envelope, Membrane, Nucleocapsid, and Spike Structural Proteins from the Beginning of the Pandemic to September 2020: A Global and Regional Approach by Epidemiological Week. Viruses 2021, 13, 243. [Google Scholar] [CrossRef] [PubMed]
  33. Jackson, C.B.; Farzan, M.; Chen, B.; Choe, H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Biol. 2022, 23, 3–20. [Google Scholar] [CrossRef] [PubMed]
  34. Nitin, P.; Nandhakumar, R.; Vidhya, B.; Rajesh, S.; Sakunthala, A. COVID-19: Invasion, pathogenesis and possible cure—A review. J. Virol. Methods 2022, 300, 114434. [Google Scholar] [CrossRef] [PubMed]
  35. Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef]
  36. Jain, U. Effect of COVID-19 on the Organs. Cureus 2020, 12, e9540. [Google Scholar] [CrossRef]
  37. Carcaterra, M.; Caruso, C. Alveolar epithelial cell type II as main target of SARS-CoV-2 virus and COVID-19 development via NF-Kb pathway deregulation: A physio-pathological theory. Med. Hypotheses 2021, 146, 110412. [Google Scholar] [CrossRef]
  38. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
  39. Mercurio, I.; Tragni, V.; Busto, F.; De Grassi, A.; Pierri, C.L. Protein structure analysis of the interactions between SARS-CoV-2 spike protein and the human ACE2 receptor: From conformational changes to novel neutralizing antibodies. Cell. Mol. Life Sci. 2021, 78, 1501–1522. [Google Scholar] [CrossRef]
  40. Chen, L.; Qu, J.; Kalyani, F.S.; Zhang, Q.; Fan, L.; Fang, Y.; Li, Y.; Xiang, C. Mesenchymal stem cell-based treatments for COVID-19: Status and future perspectives for clinical applications. Cell. Mol. Life Sci. 2022, 79, 142. [Google Scholar] [CrossRef]
  41. Örd, M.; Faustova, I.; Loog, M. The sequence at Spike S1/S2 site enables cleavage by furin and phospho-regulation in SARS-CoV2 but not in SARS-CoV1 or MERS-CoV. Sci. Rep. 2020, 10, 16944. [Google Scholar] [CrossRef]
  42. Papa, G.; Mallery, D.L.; Albecka, A.; Welch, L.G.; Cattin-Ortolá, J.; Luptak, J.; Paul, D.; McMahon, H.T.; Goodfellow, I.G.; Carter, A.; et al. Furin cleavage of SARS-CoV-2 Spike promotes but is not essential for infection and cell-cell fusion. PLoS Pathog. 2021, 17, e1009246. [Google Scholar] [CrossRef]
  43. Mykytyn, A.Z.; Breugem, T.I.; Riesebosch, S.; Schipper, D.; van den Doel, P.B.; Rottier, R.J.; Lamers, M.M.; Haagmans, B.L. SARS-CoV-2 entry into human airway organoids is serine protease-mediated and facilitated by the multibasic cleavage site. Elife 2021, 10, e64508. [Google Scholar] [CrossRef] [PubMed]
  44. Gusev, E.; Sarapultsev, A.; Solomatina, L.; Chereshnev, V. SARS-CoV-2-specific immune response and the pathogenesis of COVID-19. Int. J. Mol. Sci. 2022, 23, 1716. [Google Scholar] [CrossRef] [PubMed]
  45. Beumer, J.; Geurts, M.H.; Lamers, M.M.; Puschhof, J.; Zhang, J.; van der Vaart, J.; Mykytyn, A.Z.; Breugem, T.I.; Riesebosch, S.; Schipper, D.; et al. A CRISPR/Cas9 genetically engineered organoid biobank reveals essential host factors for coronaviruses. Nat. Commun. 2021, 12, 5498. [Google Scholar] [CrossRef] [PubMed]
  46. Sa Ribero, M.; Jouvenet, N.; Dreux, M.; Nisole, S. Interplay between SARS-CoV-2 and the type I interferon response. PLoS Pathog. 2020, 16, e1008737. [Google Scholar] [CrossRef]
  47. Murdaca, G.; Paladin, F.; Tonacci, A.; Isola, S.; Allegra, A.; Gangemi, S. The Potential Role of Cytokine Storm Pathway in the Clinical Course of Viral Respiratory Pandemic. Biomedicines 2021, 9, 1688. [Google Scholar] [CrossRef]
  48. Zhou, Y.; Fu, B.; Zheng, X.; Wang, D.; Zhao, C.; Qi, Y.; Sun, R.; Tian, Z.; Xu, X.; Wei, H. Aberrant pathogenic GM-CSF+ T cells and inflammatory CD14+ CD16+ monocytes in severe pulmonary syndrome patients of a new coronavirus. bioRxiv 2002. [Google Scholar] [CrossRef]
  49. Zuo, Y.; Yalavarthi, S.; Shi, H.; Gockman, K.; Zuo, M.; Madison, J.A.; Blair, C.; Weber, A.; Barnes, B.J.; Egeblad, M. Neutrophil extracellular traps in COVID-19. JCI Insight 2020, 5. [Google Scholar] [CrossRef] [PubMed]
  50. Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Investig. 2020, 130, 2620–2629. [Google Scholar] [CrossRef]
  51. Hu, B.; Huang, S.; Yin, L. The cytokine storm and COVID-19. J. Med. Virol. 2021, 93, 250–256. [Google Scholar] [CrossRef]
  52. Liu, J.; Li, S.; Liu, J.; Liang, B.; Wang, X.; Wang, H.; Li, W.; Tong, Q.; Yi, J.; Zhao, L. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. eBioMedicine 2020, 55. [Google Scholar] [CrossRef]
  53. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef]
  54. Darif, D.; Hammi, I.; Kihel, A.; El Idrissi Saik, I.; Guessous, F.; Akarid, K. The pro-inflammatory cytokines in COVID-19 pathogenesis: What goes wrong? Microb. Pathog. 2021, 153, 104799. [Google Scholar] [CrossRef] [PubMed]
  55. Henry, B.M.; Cheruiyot, I.; Vikse, J.; Mutua, V.; Kipkorir, V.; Benoit, J.; Plebani, M.; Bragazzi, N.; Lippi, G. Lymphopenia and neutrophilia at admission predicts severity and mortality in patients with COVID-19: A meta-analysis. Acta Biomedica: Atenei Parm. 2020, 91, e2020008. [Google Scholar]
  56. de Candia, P.; Prattichizzo, F.; Garavelli, S.; Matarese, G. T cells: Warriors of SARS-CoV-2 infection. Trends Immunol. 2021, 42, 18–30. [Google Scholar] [CrossRef]
  57. Zhang, S.; Asquith, B.; Szydlo, R.; Tregoning, J.S.; Pollock, K.M. Peripheral T cell lymphopenia in COVID-19: Potential mechanisms and impact. Immunother. Adv. 2021, 1, ltab015. [Google Scholar] [CrossRef] [PubMed]
  58. Moore, J.B.; June, C.H. Cytokine release syndrome in severe COVID-19. Science 2020, 368, 473–474. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, T.; Wu, D.; Chen, H.; Yan, W.; Yang, D.; Chen, G.; Ma, K.; Xu, D.; Yu, H.; Wang, H. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: Retrospective study. BMJ 2020, 368. [Google Scholar] [CrossRef]
  60. Carlotti, A.P.d.C.P.; Carvalho, W.B.d.; Johnston, C.; Rodriguez, I.S.; Delgado, A.F. COVID-19 diagnostic and management protocol for pediatric patients. Clinics 2020, 75. [Google Scholar] [CrossRef]
  61. Falzone, L.; Gattuso, G.; Tsatsakis, A.; Spandidos, D.A.; Libra, M. Current and innovative methods for the diagnosis of COVID-19 infection. Int. J. Mol. Med. 2021, 47, 1–23. [Google Scholar] [CrossRef]
  62. Gupta, A.; Madhavan, M.V.; Sehgal, K.; Nair, N.; Mahajan, S.; Sehrawat, T.S.; Bikdeli, B.; Ahluwalia, N.; Ausiello, J.C.; Wan, E.Y. Extrapulmonary manifestations of COVID-19. Nat. Med. 2020, 26, 1017–1032. [Google Scholar] [CrossRef] [PubMed]
  63. Chowdhury, M.R.; Mas-ud, M.A.; Ali, M.R.; Fatamatuzzohora, M.; Shimu, A.S.; Haq, M.A.; Islam, M.A.; Hossain, M.F.; Hosenuzzaman, M.; Islam, M.M. Harmful Effects of COVID-19 on Major Human Body Organs: A Review. J. Pure Appl. Microbiol. 2021, 15. [Google Scholar] [CrossRef]
  64. World Health Organization. Living Guidance for Clinical Management of COVID-19: Living Guidance; World Health Organization: Geneva, Switzerland, 23 November 2021. [Google Scholar]
  65. Zaim, S.; Chong, J.H.; Sankaranarayanan, V.; Harky, A. COVID-19 and multiorgan response. Curr. Probl. Cardiol. 2020, 45, 100618. [Google Scholar] [CrossRef] [PubMed]
  66. Wu, Z.; McGoogan, J.M. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: Summary of a report of 72,314 cases from the Chinese Center for Disease Control and Prevention. JAMA 2020, 323, 1239–1242. [Google Scholar] [CrossRef] [PubMed]
  67. Gengler, I.; Wang, J.C.; Speth, M.M.; Sedaghat, A.R. Sinonasal pathophysiology of SARS-CoV-2 and COVID-19: A systematic review of the current evidence. Laryngoscope Investig. Otolaryngol. 2020, 5, 354–359. [Google Scholar] [CrossRef]
  68. Erjefält, J.S.; Costa, N.d.S.X.; Jönsson, J.; Cozzolino, O.; Dantas, K.C.; Clausson, C.-M.; Siddhuraj, P.; Lindö, C.; Alyamani, M.; Lombardi, S.C.F.S. Diffuse alveolar damage patterns reflect the immunological and molecular heterogeneity in fatal COVID-19. EBioMedicine 2022, 83. [Google Scholar] [CrossRef]
  69. Channappanavar, R.; Zhao, J.; Perlman, S. T cell-mediated immune response to respiratory coronaviruses. Immunol. Res. 2014, 59, 118–128. [Google Scholar] [CrossRef]
  70. Xu, B.; Fan, C.Y.; Wang, A.L.; Zou, Y.L.; Yu, Y.H.; He, C.; Xia, W.G.; Zhang, J.X.; Miao, Q. Suppressed T cell-mediated immunity in patients with COVID-19: A clinical retrospective study in Wuhan, China. J. Infect. 2020, 81, e51–e60. [Google Scholar] [CrossRef]
  71. Zhou, G.; Chen, S.; Chen, Z. Advances in COVID-19: The virus, the pathogenesis, and evidence-based control and therapeutic strategies. Front. Med. 2020, 14, 117–125. [Google Scholar] [CrossRef]
  72. Doglioni, C.; Ravaglia, C.; Chilosi, M.; Rossi, G.; Dubini, A.; Pedica, F.; Piciucchi, S.; Vizzuso, A.; Stella, F.; Maitan, S.; et al. Covid-19 Interstitial Pneumonia: Histological and Immunohistochemical Features on Cryobiopsies. Respiration 2021, 100, 488–498. [Google Scholar] [CrossRef]
  73. McGonagle, D.; O’Donnell, J.S.; Sharif, K.; Emery, P.; Bridgewood, C. Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia. Lancet Rheumatol. 2020, 2, e437–e445. [Google Scholar] [CrossRef] [PubMed]
  74. Rahman, A.; Tabassum, T.; Araf, Y.; Al Nahid, A.; Ullah, M.A.; Hosen, M.J. Silent hypoxia in COVID-19: Pathomechanism and possible management strategy. Mol. Biol. Rep. 2021, 48, 3863–3869. [Google Scholar] [CrossRef] [PubMed]
  75. Dhont, S.; Derom, E.; Van Braeckel, E.; Depuydt, P.; Lambrecht, B.N. The pathophysiology of ‘happy’ hypoxemia in COVID-19. Respir. Res. 2020, 21, 198. [Google Scholar] [CrossRef] [PubMed]
  76. Ottestad, W.; Søvik, S. COVID-19 patients with respiratory failure: What can we learn from aviation medicine? Br. J. Anaesth. 2020, 125, e280–e281. [Google Scholar] [CrossRef]
  77. Gattinoni, L.; Chiumello, D.; Caironi, P.; Busana, M.; Romitti, F.; Brazzi, L.; Camporota, L. COVID-19 pneumonia: Different respiratory treatments for different phenotypes? Intensive Care Med. 2020, 46, 1099–1102. [Google Scholar] [CrossRef]
  78. Panwar, R.; Madotto, F.; Laffey, J.G.; van Haren, F.M. Compliance phenotypes in early acute respiratory distress syndrome before the COVID-19 pandemic. Am. J. Respir. Crit. Care Med. 2020, 202, 1244–1252. [Google Scholar] [CrossRef]
  79. Alkhemeiri, A.; Al Zaabi, S.; Lakshmanan, J.; El-Khatib, Z.; Awofeso, N. COVID-19 Case Management Outcomes Amongst Diabetes and Hypertensive Patients in the United Arab Emirates: A Prospective Study. Int. J. Environ. Res. Public. Health 2022, 19, 15967. [Google Scholar] [CrossRef]
  80. Guo, T.; Fan, Y.; Chen, M.; Wu, X.; Zhang, L.; He, T.; Wang, H.; Wan, J.; Wang, X.; Lu, Z. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19). JAMA Cardiol. 2020, 5, 811–818. [Google Scholar] [CrossRef]
  81. Davis, G.K.; Adlan, A.; Majewski, J.; Ibrahim, B. SARS-CoV-2 pandemic and the cardiovascular system: What the non-cardiologist needs to know. Clin. Med. 2020, 20, 262. [Google Scholar] [CrossRef]
  82. Giustino, G.; Pinney, S.P.; Lala, A.; Reddy, V.Y.; Johnston-Cox, H.A.; Mechanick, J.I.; Halperin, J.L.; Fuster, V. Coronavirus and cardiovascular disease, myocardial injury, and arrhythmia: JACC focus seminar. J. Am. Coll. Cardiol. 2020, 76, 2011–2023. [Google Scholar] [CrossRef]
  83. Sattar, N.; McInnes, I.B.; McMurray, J.J. Obesity is a risk factor for severe COVID-19 infection: Multiple potential mechanisms. Circulation 2020, 142, 4–6. [Google Scholar] [CrossRef] [PubMed]
  84. Sun, H.; Su, X.; Huang, L.; Mu, D.; Qu, Y. Research progress on the cardiac injury from ACE2 targeting in SARS-CoV-2 infection. Biomolecules 2021, 11, 196. [Google Scholar] [CrossRef] [PubMed]
  85. Magadum, A.; Kishore, R. Cardiovascular Manifestations of COVID-19 Infection. Cells 2020, 9, 2508. [Google Scholar] [CrossRef] [PubMed]
  86. Okor, I.; Bob-Manuel, T.; Price, J.; Sleem, A.; Amoran, O.; Kelly, J.; Ekerete, M.F.; Bamgbose, M.O.; Bolaji, O.A.; Krim, S.R. COVID-19 myocarditis: An emerging clinical conundrum. Curr. Probl. Cardiol. 2022, 47, 101268. [Google Scholar] [CrossRef]
  87. Mele, D.; Flamigni, F.; Rapezzi, C.; Ferrari, R. Myocarditis in COVID-19 patients: Current problems. Intern. Emerg. Med. 2021, 1–7. [Google Scholar] [CrossRef]
  88. Silhol, F.; Sarlon, G.; Deharo, J.-C.; Vaïsse, B. Downregulation of ACE2 induces overstimulation of the renin–angiotensin system in COVID-19: Should we block the renin–angiotensin system? Hypertens. Res. 2020, 43, 854–856. [Google Scholar] [CrossRef]
  89. Cancro, F.P.; Bellino, M.; Esposito, L.; Romei, S.; Centore, M.; D’Elia, D.; Cristiano, M.; Maglio, A.; Carrizzo, A.; Rasile, B. Acute coronary syndrome in patients with SARS-CoV-2 infection: Pathophysiology and translational perspectives. Transl. Med. @ UniSa 2022, 24, 1. [Google Scholar] [CrossRef]
  90. Schiavone, M.; Gobbi, C.; Biondi-Zoccai, G.; D’Ascenzo, F.; Palazzuoli, A.; Gasperetti, A.; Mitacchione, G.; Viecca, M.; Galli, M.; Fedele, F. Acute coronary syndromes and Covid-19: Exploring the uncertainties. J. Clin. Med. 2020, 9, 1683. [Google Scholar] [CrossRef]
  91. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
  92. Talanas, G.; Dossi, F.; Parodi, G. Type 2 myocardial infarction in patients with coronavirus disease 2019. J. Cardiovasc. Med. 2021, 22, 603–605. [Google Scholar] [CrossRef]
  93. Libby, P. The Heart in COVID-19: Primary Target or Secondary Bystander? JACC Basic Transl. Sci. 2020, 5, 537–542. [Google Scholar] [CrossRef] [PubMed]
  94. Musher, D.M.; Abers, M.S.; Corrales-Medina, V.F. Acute infection and myocardial infarction. N. Engl. J. Med. 2019, 380, 171–176. [Google Scholar] [CrossRef] [PubMed]
  95. Li, S.; Wang, J.; Yan, Y.; Zhang, Z.; Gong, W.; Nie, S. Clinical characterization and possible pathological mechanism of acute myocardial injury in COVID-19. Front. Cardiovasc. Med. 2022, 9, 862571. [Google Scholar] [CrossRef] [PubMed]
  96. Noone, S.; Flinspach, A.N.; Fichtlscherer, S.; Zacharowski, K.; Sonntagbauer, M.; Raimann, F.J. Severe COVID-19-associated myocarditis with cardiogenic shock–management with assist devices—A case report & review. BMC Anesthesiol. 2022, 22, 1–10. [Google Scholar]
  97. Italia, L.; Tomasoni, D.; Bisegna, S.; Pancaldi, E.; Stretti, L.; Adamo, M.; Metra, M. COVID-19 and heart failure: From epidemiology during the pandemic to myocardial injury, myocarditis, and heart failure sequelae. Front. Cardiovasc. Med. 2021, 8, 713560. [Google Scholar] [CrossRef]
  98. Karamchandani, K.; Quintili, A.; Landis, T.; Bose, S. Cardiac Arrhythmias in Critically Ill Patients With COVID-19: A Brief Review. J. Cardiothorac. Vasc. Anesth. 2021, 35, 3789–3796. [Google Scholar] [CrossRef]
  99. Viner, R.M.; Whittaker, E. Kawasaki-like disease: Emerging complication during the COVID-19 pandemic. Lancet 2020, 395, 1741–1743. [Google Scholar] [CrossRef]
  100. Zhang, J.; Garrett, S.; Sun, J. Gastrointestinal symptoms, pathophysiology, and treatment in COVID-19. Genes Dis. 2021, 8, 385–400. [Google Scholar] [CrossRef]
  101. Li, M.Y.; Li, L.; Zhang, Y.; Wang, X.S. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect. Dis. Poverty 2020, 9, 45. [Google Scholar] [CrossRef]
  102. Luo, S.; Zhang, X.; Xu, H. Don’t Overlook Digestive Symptoms in Patients With 2019 Novel Coronavirus Disease (COVID-19). Clin. Gastroenterol. Hepatol. 2020, 18, 1636–1637. [Google Scholar] [CrossRef]
  103. Pan, L.; Mu, M.; Yang, P.; Sun, Y.; Wang, R.; Yan, J.; Li, P.; Hu, B.; Wang, J.; Hu, C.; et al. Clinical Characteristics of COVID-19 Patients With Digestive Symptoms in Hubei, China: A Descriptive, Cross-Sectional, Multicenter Study. Am. J. Gastroenterol. 2020, 115, 766–773. [Google Scholar] [CrossRef] [PubMed]
  104. Khreefa, Z.; Barbier, M.T.; Koksal, A.R.; Love, G.; Del Valle, L. Pathogenesis and Mechanisms of SARS-CoV-2 Infection in the Intestine, Liver, and Pancreas. Cells 2023, 12, 262. [Google Scholar] [CrossRef]
  105. Wong, S.H.; Lui, R.N.; Sung, J.J. Covid-19 and the digestive system. J. Gastroenterol. Hepatol. 2020, 35, 744–748. [Google Scholar] [CrossRef] [PubMed]
  106. Zhang, C.; Shi, L.; Wang, F.-S. Liver injury in COVID-19: Management and challenges. Lancet Gastroenterol. Hepatol. 2020, 5, 428–430. [Google Scholar] [CrossRef]
  107. Mao, R.; Qiu, Y.; He, J.-S.; Tan, J.-Y.; Li, X.-H.; Liang, J.; Shen, J.; Zhu, L.-R.; Chen, Y.; Iacucci, M. Manifestations and prognosis of gastrointestinal and liver involvement in patients with COVID-19: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2020, 5, 667–678. [Google Scholar] [CrossRef] [PubMed]
  108. Tavakolpour, S.; Rakhshandehroo, T.; Wei, E.X.; Rashidian, M. Lymphopenia during the COVID-19 infection: What it shows and what can be learned. Immunol. Lett. 2020, 225, 31–32. [Google Scholar] [CrossRef]
  109. Toori, K.U.; Qureshi, M.A.; Chaudhry, A. Lymphopenia: A useful predictor of COVID-19 disease severity and mortality. Pak. J. Med. Sci. 2021, 37, 1984–1988. [Google Scholar] [CrossRef] [PubMed]
  110. Liu, X.; Zhang, R.; He, G. Hematological findings in coronavirus disease 2019: Indications of progression of disease. Ann. Hematol. 2020, 99, 1421–1428. [Google Scholar] [CrossRef]
  111. McKenna, E.; Wubben, R.; Isaza-Correa, J.M.; Melo, A.M.; Mhaonaigh, A.U.; Conlon, N.; O’Donnell, J.S.; Ní Cheallaigh, C.; Hurley, T.; Stevenson, N.J. Neutrophils in COVID-19: Not innocent bystanders. Front. Immunol. 2022, 13, 864387. [Google Scholar] [CrossRef]
  112. Shan, C.; Yu, F.; Deng, X.; Ni, L.; Luo, X.; Li, J.; Cai, S.; Huang, M.; Wang, X. Biogenesis aberration: One of the mechanisms of thrombocytopenia in COVID-19. Front. Physiol. 2023, 14, 1100997. [Google Scholar] [CrossRef]
  113. Lippi, G.; Plebani, M.; Henry, B.M. Thrombocytopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: A meta-analysis. Clin. Chim. Acta 2020, 506, 145–148. [Google Scholar] [CrossRef] [PubMed]
  114. Yang, X.; Yang, Q.; Wang, Y.; Wu, Y.; Xu, J.; Yu, Y.; Shang, Y. Thrombocytopenia and its association with mortality in patients with COVID-19. J. Thromb. Haemost. 2020, 18, 1469–1472. [Google Scholar] [CrossRef] [PubMed]
  115. Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 844–847. [Google Scholar] [CrossRef] [PubMed]
  116. Huang, I.; Pranata, R.; Lim, M.A.; Oehadian, A.; Alisjahbana, B. C-reactive protein, procalcitonin, D-dimer, and ferritin in severe coronavirus disease-2019: A meta-analysis. Ther. Adv. Respir. Dis. 2020, 14, 1753466620937175. [Google Scholar] [CrossRef] [PubMed]
  117. Kollias, A.; Kyriakoulis, K.G.; Lagou, S.; Kontopantelis, E.; Stergiou, G.S.; Syrigos, K. Venous thromboembolism in COVID-19: A systematic review and meta-analysis. Vasc. Med. 2021, 26, 415–425. [Google Scholar] [CrossRef] [PubMed]
  118. Schulman, S.; Hu, Y.; Konstantinides, S. Venous thromboembolism in COVID-19. Thromb. Haemost. 2020, 120, 1642–1653. [Google Scholar] [CrossRef]
  119. Ousseiran, Z.H.; Fares, Y.; Chamoun, W.T. Neurological manifestations of COVID-19: A systematic review and detailed comprehension. Int. J. Neurosci. 2023, 133, 754–769. [Google Scholar] [CrossRef]
  120. Khatoon, F.; Prasad, K.; Kumar, V. COVID-19 associated nervous system manifestations. Sleep Med. 2022, 91, 231–236. [Google Scholar] [CrossRef]
  121. Mao, L.; Jin, H.; Wang, M.; Hu, Y.; Chen, S.; He, Q.; Chang, J.; Hong, C.; Zhou, Y.; Wang, D.; et al. Neurologic Manifestations of Hospitalized Patients With Coronavirus Disease 2019 in Wuhan, China. JAMA Neurol. 2020, 77, 683–690. [Google Scholar] [CrossRef]
  122. Chen, X.; Laurent, S.; Onur, O.A.; Kleineberg, N.N.; Fink, G.R.; Schweitzer, F.; Warnke, C. A systematic review of neurological symptoms and complications of COVID-19. J. Neurol. 2021, 268, 392–402. [Google Scholar] [CrossRef]
  123. Romagnolo, A.; Balestrino, R.; Imbalzano, G.; Ciccone, G.; Riccardini, F.; Artusi, C.A.; Bozzali, M.; Ferrero, B.; Montalenti, E.; Montanaro, E. Neurological comorbidity and severity of COVID-19. J. Neurol. 2021, 268, 762–769. [Google Scholar] [CrossRef] [PubMed]
  124. Garg, R.K.; Paliwal, V.K.; Gupta, A. Encephalopathy in patients with COVID-19: A review. J. Med. Virol. 2021, 93, 206–222. [Google Scholar] [CrossRef] [PubMed]
  125. Nannoni, S.; de Groot, R.; Bell, S.; Markus, H.S. Stroke in COVID-19: A systematic review and meta-analysis. Int. J. Stroke 2021, 16, 137–149. [Google Scholar] [CrossRef]
  126. Ntaios, G.; Michel, P.; Georgiopoulos, G.; Guo, Y.; Li, W.; Xiong, J.; Calleja, P.; Ostos, F.; González-Ortega, G.; Fuentes, B. Characteristics and outcomes in patients with COVID-19 and acute ischemic stroke: The global COVID-19 stroke registry. Stroke 2020, 51, e254–e258. [Google Scholar] [CrossRef] [PubMed]
  127. Hong, D.; Long, L.; Wang, A.Y.; Lei, Y.; Tang, Y.; Zhao, J.W.; Song, X.; He, Y.; Wen, E.; Zheng, L.; et al. Kidney manifestations of mild, moderate and severe coronavirus disease 2019: A retrospective cohort study. Clin. Kidney J. 2020, 13, 340–346. [Google Scholar] [CrossRef] [PubMed]
  128. Cheng, Y.; Luo, R.; Wang, K.; Zhang, M.; Wang, Z.; Dong, L.; Li, J.; Yao, Y.; Ge, S.; Xu, G. Kidney disease is associated with in-hospital death of patients with COVID-19. Kidney Int. 2020, 97, 829–838. [Google Scholar] [CrossRef] [PubMed]
  129. Adamczak, M.; Surma, S.; Więcek, A. Acute kidney injury in patients with COVID-19: Epidemiology, pathogenesis and treatment. Adv. Clin. Exp. Med. 2022, 31, 317–326. [Google Scholar] [CrossRef]
  130. Azinheira Nobrega Cruz, N.; Gonçalves de Oliveira, L.C.; Tedesco Silva Junior, H.; Osmar Medina Pestana, J.; Casarini, D.E. Angiotensin-Converting Enzyme 2 in the Pathogenesis of Renal Abnormalities Observed in COVID-19 Patients. Front. Physiol. 2021, 12, 700220. [Google Scholar] [CrossRef]
  131. Ahmadian, E.; Hosseiniyan Khatibi, S.M.; Razi Soofiyani, S.; Abediazar, S.; Shoja, M.M.; Ardalan, M.; Zununi Vahed, S. Covid-19 and kidney injury: Pathophysiology and molecular mechanisms. Rev. Med. Virol. 2021, 31, e2176. [Google Scholar] [CrossRef]
  132. Silver, S.A.; Beaubien-Souligny, W.; Shah, P.S.; Harel, S.; Blum, D.; Kishibe, T.; Meraz-Munoz, A.; Wald, R.; Harel, Z. The prevalence of acute kidney injury in patients hospitalized with COVID-19 infection: A systematic review and meta-analysis. Kidney Med. 2021, 3, 83–98.e1. [Google Scholar] [CrossRef]
  133. Chávez-Valencia, V.; Orizaga-de-la-Cruz, C.; Lagunas-Rangel, F.A. Acute Kidney Injury in COVID-19 Patients: Pathogenesis, Clinical Characteristics, Therapy, and Mortality. Diseases 2022, 10, 53. [Google Scholar] [CrossRef]
  134. Alberti, A.; Schuelter-Trevisol, F.; Iser, B.P.M.; Traebert, E.; Freiberger, V.; Ventura, L.; Rezin, G.T.; da Silva, B.B.; Meneghetti Dallacosta, F.; Grigollo, L.; et al. Obesity in people with diabetes in COVID-19 times: Important considerations and precautions to be taken. World J. Clin. Cases 2021, 9, 5358–5371. [Google Scholar] [CrossRef] [PubMed]
  135. De Sa-Ferreira, C.O.; da Costa, C.H.M.; Guimarães, J.C.W.; Sampaio, N.S.; Silva, L.d.M.L.; de Mascarenhas, L.P.; Rodrigues, N.G.; Dos Santos, T.L.; Campos, S.; Young, E.C. Diabetic ketoacidosis and COVID-19: What have we learned so far? Am. J. Physiol.-Endocrinol. Metab. 2022, 322, E44–E53. [Google Scholar] [CrossRef] [PubMed]
  136. Li, J.; Wang, X.; Chen, J.; Zuo, X.; Zhang, H.; Deng, A. COVID-19 infection may cause ketosis and ketoacidosis. Diabetes Obes. Metab. 2020, 22, 1935–1941. [Google Scholar] [CrossRef] [PubMed]
  137. El-Huneidi, W.; Hamad, M.; Taneera, J. Expression of SARS-CoV-2 receptor "ACE2" in human pancreatic β cells: To be or not to be! Islets 2021, 13, 106–114. [Google Scholar] [CrossRef] [PubMed]
  138. Yang, J.-K.; Lin, S.-S.; Ji, X.-J.; Guo, L.-M. Binding of SARS coronavirus to its receptor damages islets and causes acute diabetes. Acta Diabetol. 2010, 47, 193–199. [Google Scholar] [CrossRef]
  139. Eizirik, D.L.; Darville, M.I. beta-cell apoptosis and defense mechanisms: Lessons from type 1 diabetes. Diabetes 2001, 50, S64. [Google Scholar] [CrossRef]
  140. Bhatti, J.M.; Raza, S.A.; Shahid, M.O.; Akhtar, A.; Ahmed, T.; Das, B. Association between glycemic control and the outcome in hospitalized patients with COVID-19. Endocrine 2022, 77, 213–220. [Google Scholar] [CrossRef]
  141. Hass, R.; Kasper, C.; Böhm, S.; Jacobs, R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 2011, 9, 1–14. [Google Scholar] [CrossRef]
  142. Chan, C.-K.; Wu, K.-H.; Lee, Y.-S.; Hwang, S.-M.; Lee, M.-S.; Liao, S.-K.; Cheng, E.-H.; See, L.-C.; Tsai, C.-N.; Kuo, M.-L. The comparison of interleukin 6-associated immunosuppressive effects of human ESCs, fetal-Type MSCs, and adult-type MSCs. Transplantation 2012, 94, 132–138. [Google Scholar] [CrossRef]
  143. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [PubMed]
  144. Zhou, Y.; Yamamoto, Y.; Xiao, Z.; Ochiya, T. The immunomodulatory functions of mesenchymal stromal/stem cells mediated via paracrine activity. J. Clin. Med. 2019, 8, 1025. [Google Scholar] [CrossRef] [PubMed]
  145. Wu, K.-H.; Li, J.-P.; Chao, W.-R.; Lee, Y.-J.; Yang, S.-F.; Cheng, C.-C.; Chao, Y.-H. Immunomodulation via MyD88-NFκB signaling pathway from human umbilical cord-derived mesenchymal stem cells in acute lung injury. Int. J. Mol. Sci. 2022, 23, 5295. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, X.-A.; Li, J.-P.; Wu, K.-H.; Yang, S.-F.; Chao, Y.-H. Mesenchymal Stem Cells in Acquired Aplastic Anemia: The Spectrum from Basic to Clinical Utility. Int. J. Mol. Sci. 2023, 24, 4464. [Google Scholar] [PubMed]
  147. Jasim, S.A.; Yumashev, A.V.; Abdelbasset, W.K.; Margiana, R.; Markov, A.; Suksatan, W.; Pineda, B.; Thangavelu, L.; Ahmadi, S.H. Shining the light on clinical application of mesenchymal stem cell therapy in autoimmune diseases. Stem Cell Res. Ther. 2022, 13, 1–15. [Google Scholar]
  148. Zhang, H.-M.; Yuan, S.; Meng, H.; Hou, X.-T.; Li, J.; Xue, J.-C.; Li, Y.; Wang, Q.; Nan, J.-X.; Jin, X.-J. Stem cell-based therapies for inflammatory bowel disease. Int. J. Mol. Sci. 2022, 23, 8494. [Google Scholar] [CrossRef]
  149. Saadh, M.J.; Mikhailova, M.V.; Rasoolzadegan, S.; Falaki, M.; Akhavanfar, R.; Gonzáles, J.L.A.; Rigi, A.; Kiasari, B.A. Therapeutic potential of mesenchymal stem/stromal cells (MSCs)-based cell therapy for inflammatory bowel diseases (IBD) therapy. Eur. J. Med. Res. 2023, 28, 47. [Google Scholar]
  150. Huldani, H.; Margiana, R.; Ahmad, F.; Opulencia, M.J.C.; Ansari, M.J.; Bokov, D.O.; Abdullaeva, N.N.; Siahmansouri, H. Immunotherapy of inflammatory bowel disease (IBD) through mesenchymal stem cells. Int. Immunopharmacol. 2022, 107, 108698. [Google Scholar] [CrossRef]
  151. Zhou, T.; Li, H.-Y.; Liao, C.; Lin, W.; Lin, S. Clinical efficacy and safety of mesenchymal stem cells for systemic lupus erythematosus. Stem Cells Int. 2020, 2020. [Google Scholar] [CrossRef]
  152. Li, A.; Guo, F.; Pan, Q.; Chen, S.; Chen, J.; Liu, H.-f.; Pan, Q. Mesenchymal stem cell therapy: Hope for patients with systemic lupus erythematosus. Front. Immunol. 2021, 12, 728190. [Google Scholar]
  153. Radmanesh, F.; Mahmoudi, M.; Yazdanpanah, E.; Keyvani, V.; Kia, N.; Nikpoor, A.R.; Zafari, P.; Esmaeili, S.A. The immunomodulatory effects of mesenchymal stromal cell-based therapy in human and animal models of systemic lupus erythematosus. IUBMB Life 2020, 72, 2366–2381. [Google Scholar] [PubMed]
  154. Bader, P.; Kuçi, Z.; Bakhtiar, S.; Basu, O.; Bug, G.; Dennis, M.; Greil, J.; Barta, A.; Kállay, K.M.; Lang, P. Effective treatment of steroid and therapy-refractory acute graft-versus-host disease with a novel mesenchymal stromal cell product (MSC-FFM). Bone Marrow Transplant. 2018, 53, 852–862. [Google Scholar] [CrossRef] [PubMed]
  155. Kelly, K.; Rasko, J.E. Mesenchymal stromal cells for the treatment of graft versus host disease. Front. Immunol. 2021, 4457. [Google Scholar]
  156. Odinak, M.; Bisaga, G.; Novitskiĭ, A.; Tyrenko, V.; Fominykh, M.; Bilibina, A.; Krugliakov, P.; Polyntsev, D. Transplantation of mesenchymal stem cells in multiple sclerosis. Zhurnal Nevrol. I Psikhiatrii Im. SS Korsakova 2011, 111, 72–76. [Google Scholar] [CrossRef]
  157. Riordan, N.H.; Morales, I.; Fernández, G.; Allen, N.; Fearnot, N.E.; Leckrone, M.E.; Markovich, D.J.; Mansfield, D.; Avila, D.; Patel, A.N. Clinical feasibility of umbilical cord tissue-derived mesenchymal stem cells in the treatment of multiple sclerosis. J. Transl. Med. 2018, 16, 1–12. [Google Scholar] [CrossRef] [PubMed]
  158. Gugliandolo, A.; Bramanti, P.; Mazzon, E. Mesenchymal stem cells in multiple sclerosis: Recent evidence from pre-clinical to clinical studies. Int. J. Mol. Sci. 2020, 21, 8662. [Google Scholar] [CrossRef] [PubMed]
  159. Xuan, X.; Tian, C.; Zhao, M.; Sun, Y.; Huang, C. Mesenchymal stem cells in cancer progression and anticancer therapeutic resistance. Cancer Cell Int. 2021, 21, 1–16. [Google Scholar]
  160. Slama, Y.; Ah-Pine, F.; Khettab, M.; Arcambal, A.; Begue, M.; Dutheil, F.; Gasque, P. The Dual Role of Mesenchymal Stem Cells in Cancer Pathophysiology: Pro-Tumorigenic Effects versus Therapeutic Potential. Int. J. Mol. Sci. 2023, 24, 13511. [Google Scholar]
  161. Clinicaltrials.com. Available online: https://classic.clinicaltrials.gov/ct2/results?cond=COVID19&term=mesenchymal+stem+Cell+&cntry=&state=&city=&dist= (accessed on 8 August 2023).
  162. Karyana, M.; Djaharuddin, I.; Rif’ati, L.; Arif, M.; Choi, M.K.; Angginy, N.; Yoon, A.; Han, J.; Josh, F.; Arlinda, D. Safety of DW-MSC infusion in patients with low clinical risk COVID-19 infection: A randomized, double-blind, placebo-controlled trial. Stem Cell Res. Ther. 2022, 13, 134. [Google Scholar] [CrossRef]
  163. Shu, L.; Niu, C.; Li, R.; Huang, T.; Wang, Y.; Huang, M.; Ji, N.; Zheng, Y.; Chen, X.; Shi, L. Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cells. Stem Cell Res. Ther. 2020, 11, 1–11. [Google Scholar] [CrossRef]
  164. Hashemian, S.-M.R.; Aliannejad, R.; Zarrabi, M.; Soleimani, M.; Vosough, M.; Hosseini, S.-E.; Hossieni, H.; Keshel, S.H.; Naderpour, Z.; Hajizadeh-Saffar, E. Mesenchymal stem cells derived from perinatal tissues for treatment of critically ill COVID-19-induced ARDS patients: A case series. Stem Cell Res. Ther. 2021, 12, 1–12. [Google Scholar] [CrossRef] [PubMed]
  165. Monsel, A.; Hauw-Berlemont, C.; Mebarki, M.; Heming, N.; Mayaux, J.; Nguekap Tchoumba, O.; Diehl, J.-L.; Demoule, A.; Annane, D.; Marois, C. Treatment of COVID-19-associated ARDS with mesenchymal stromal cells: A multicenter randomized double-blind trial. Crit. Care 2022, 26, 48. [Google Scholar] [CrossRef] [PubMed]
  166. Xu, X.; Jiang, W.; Chen, L.; Xu, Z.; Zhang, Q.; Zhu, M.; Ye, P.; Li, H.; Yu, L.; Zhou, X. Evaluation of the safety and efficacy of using human menstrual blood-derived mesenchymal stromal cells in treating severe and critically ill COVID-19 patients: An exploratory clinical trial. Clin. Transl. Med. 2021, 11, e297. [Google Scholar] [CrossRef]
  167. Kaffash Farkhad, N.; Sedaghat, A.; Reihani, H.; Adhami Moghadam, A.; Bagheri Moghadam, A.; Khadem Ghaebi, N.; Khodadoust, M.A.; Ganjali, R.; Tafreshian, A.R.; Tavakol-Afshari, J. Mesenchymal stromal cell therapy for COVID-19-induced ARDS patients: A successful phase 1, control-placebo group, clinical trial. Stem Cell Res. Ther. 2022, 13, 283. [Google Scholar] [CrossRef]
  168. Sadeghi, B.; Roshandel, E.; Pirsalehi, A.; Kazemi, S.; Sankanian, G.; Majidi, M.; Salimi, M.; Aghdami, N.; Sadrosadat, H.; Samadi Kochaksaraei, S. Conquering the cytokine storm in COVID-19-induced ARDS using placenta-derived decidua stromal cells. J. Cell. Mol. Med. 2021, 25, 10554–10564. [Google Scholar] [CrossRef]
  169. Lanzoni, G.; Linetsky, E.; Correa, D.; Messinger Cayetano, S.; Alvarez, R.A.; Kouroupis, D.; Alvarez Gil, A.; Poggioli, R.; Ruiz, P.; Marttos, A.C. Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized controlled trial. Stem Cells Transl. Med. 2021, 10, 660–673. [Google Scholar] [CrossRef]
  170. Grégoire, C.; Layios, N.; Lambermont, B.; Lechanteur, C.; Briquet, A.; Bettonville, V.; Baudoux, E.; Thys, M.; Dardenne, N.; Misset, B. Bone marrow-derived mesenchymal stromal cell therapy in severe COVID-19: Preliminary results of a phase I/II clinical trial. Front. Immunol. 2022, 13, 932360. [Google Scholar] [CrossRef]
  171. Dilogo, I.H.; Aditianingsih, D.; Sugiarto, A.; Burhan, E.; Damayanti, T.; Sitompul, P.A.; Mariana, N.; Antarianto, R.D.; Liem, I.K.; Kispa, T. Umbilical cord mesenchymal stromal cells as critical COVID-19 adjuvant therapy: A randomized controlled trial. Stem Cells Transl. Med. 2021, 10, 1279–1287. [Google Scholar] [CrossRef]
  172. Fathi-Kazerooni, M.; Fattah-Ghazi, S.; Darzi, M.; Makarem, J.; Nasiri, R.; Salahshour, F.; Dehghan-Manshadi, S.A.; Kazemnejad, S. Safety and efficacy study of allogeneic human menstrual blood stromal cells secretome to treat severe COVID-19 patients: Clinical trial phase I & II. Stem Cell Res. Ther. 2022, 13, 96. [Google Scholar]
  173. Li, T.-T.; Zhang, B.; Fang, H.; Shi, M.; Yao, W.-Q.; Li, Y.; Zhang, C.; Song, J.; Huang, L.; Xu, Z. Human mesenchymal stem cell therapy in severe COVID-19 patients: 2-year follow-up results of a randomized, double-blind, placebo-controlled trial. EBioMedicine 2023, 92. [Google Scholar] [CrossRef] [PubMed]
  174. Meng, F.; Xu, R.; Wang, S.; Xu, Z.; Zhang, C.; Li, Y.; Yang, T.; Shi, L.; Fu, J.; Jiang, T. Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: A phase 1 clinical trial. Signal Transduct. Target. Ther. 2020, 5, 172. [Google Scholar] [CrossRef]
  175. Zhu, Y.-G.; Shi, M.-M.; Monsel, A.; Dai, C.-X.; Dong, X.; Shen, H.; Li, S.-K.; Chang, J.; Xu, C.-L.; Li, P. Nebulized exosomes derived from allogenic adipose tissue mesenchymal stromal cells in patients with severe COVID-19: A pilot study. Stem Cell Res. Ther. 2022, 13, 1–10. [Google Scholar] [CrossRef] [PubMed]
  176. Rebelatto, C.L.K.; Senegaglia, A.C.; Franck, C.L.; Daga, D.R.; Shigunov, P.; Stimamiglio, M.A.; Marsaro, D.B.; Schaidt, B.; Micosky, A.; de Azambuja, A.P. Safety and long-term improvement of mesenchymal stromal cell infusion in critically COVID-19 patients: A randomized clinical trial. Stem Cell Res. Ther. 2022, 13, 1–22. [Google Scholar] [CrossRef] [PubMed]
  177. Zarrabi, M.; Shahrbaf, M.A.; Nouri, M.; Shekari, F.; Hosseini, S.-E.; Hashemian, S.-M.R.; Aliannejad, R.; Jamaati, H.; Khavandgar, N.; Alemi, H. Allogenic mesenchymal stromal cells and their extracellular vesicles in COVID-19 induced ARDS: A randomized controlled trial. Stem Cell Res. Ther. 2023, 14, 169. [Google Scholar] [CrossRef] [PubMed]
  178. Adas, G.; Cukurova, Z.; Yasar, K.K.; Yilmaz, R.; Isiksacan, N.; Kasapoglu, P.; Yesilbag, Z.; Koyuncu, I.; Karaoz, E. The systematic effect of mesenchymal stem cell therapy in critical COVID-19 patients: A prospective double controlled trial. Cell Transplant. 2021, 30, 09636897211024942. [Google Scholar] [CrossRef]
  179. Saleh, M.; Vaezi, A.A.; Aliannejad, R.; Sohrabpour, A.A.; Kiaei, S.Z.F.; Shadnoush, M.; Siavashi, V.; Aghaghazvini, L.; Khoundabi, B.; Abdoli, S. Cell therapy in patients with COVID-19 using Wharton’s jelly mesenchymal stem cells: A phase 1 clinical trial. Stem Cell Res. Ther. 2021, 12, 1–13. [Google Scholar] [CrossRef]
  180. Shi, L.; Huang, H.; Lu, X.; Yan, X.; Jiang, X.; Xu, R.; Wang, S.; Zhang, C.; Yuan, X.; Xu, Z. Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: A randomized, double-blind, placebo-controlled phase 2 trial. Signal Transduct. Target. Ther. 2021, 6, 58. [Google Scholar] [CrossRef]
  181. Ye, Q.; Wang, H.; Xia, X.; Zhou, C.; Liu, Z.; Xia, Z.-e.; Zhang, Z.; Zhao, Y.; Yehenala, J.; Wang, S. 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 2020, 21, 1–4. [Google Scholar] [CrossRef]
  182. Aghayan, H.R.; Salimian, F.; Abedini, A.; Fattah Ghazi, S.; Yunesian, M.; Alavi-Moghadam, S.; Makarem, J.; Majidzadeh-A, K.; Hatamkhani, A.; Moghri, M. Human placenta-derived mesenchymal stem cells transplantation in patients with acute respiratory distress syndrome (ARDS) caused by COVID-19 (phase I clinical trial): Safety profile assessment. Stem Cell Res. Ther. 2022, 13, 1–12. [Google Scholar] [CrossRef]
  183. Chu, M.; Wang, H.; Bian, L.; Huang, J.; Wu, D.; Zhang, R.; Fei, F.; Chen, Y.; Xia, J. Nebulization therapy with umbilical cord mesenchymal stem cell-derived exosomes for COVID-19 pneumonia. Stem Cell Rev. Rep. 2022, 18, 2152–2163. [Google Scholar] [CrossRef]
  184. Bowdish, M.E.; Barkauskas, C.E.; Overbey, J.R.; Gottlieb, R.L.; Osman, K.; Duggal, A.; Marks, M.E.; Hupf, J.; Fernandes, E.; Leshnower, B.G. A randomized trial of mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome from COVID-19. Am. J. Respir. Crit. Care Med. 2023, 207, 261–270. [Google Scholar] [CrossRef] [PubMed]
  185. Al-Shudifat, A.E.; Al-Radaideh, A.; Hammad, S.; Hijjawi, N.; Abu-Baker, S.; Azab, M.; Tayyem, R. Association of Lung CT Findings in Coronavirus Disease 2019 (COVID-19) With Patients’ Age, Body Weight, Vital Signs, and Medical Regimen. Front. Med. 2022, 9, 912752. [Google Scholar] [CrossRef] [PubMed]
  186. Schrepfer, S.; Deuse, T.; Reichenspurner, H.; Fischbein, M.; Robbins, R.; Pelletier, M. Stem cell transplantation: The lung barrier. Transplant. Proc. 2007, 39, 573–576. [Google Scholar]
  187. Shi, L.; Wang, L.; Xu, R.; Zhang, C.; Xie, Y.; Liu, K.; Li, T.; Hu, W.; Zhen, C.; Wang, F.-S. Mesenchymal stem cell therapy for severe COVID-19. Signal Transduct. Target. Ther. 2021, 6, 339. [Google Scholar] [CrossRef] [PubMed]
  188. Paris, G.C.; Azevedo, A.A.; Ferreira, A.L.; Azevedo, Y.M.; Rainho, M.A.; Oliveira, G.P.; Silva, K.R.; Cortez, E.A.; Stumbo, A.C.; Carvalho, S.N. Therapeutic potential of mesenchymal stem cells in multiple organs affected by COVID-19. Life Sci. 2021, 278, 119510. [Google Scholar] [CrossRef]
  189. Ji, H.L.; Liu, C.; Zhao, R.Z. Stem cell therapy for COVID-19 and other respiratory diseases: Global trends of clinical trials. World J. Stem Cells 2020, 12, 471–480. [Google Scholar] [CrossRef]
  190. Khoury, M.; Cuenca, J.; Cruz, F.F.; Figueroa, F.E.; Rocco, P.R.; Weiss, D.J. Current status of cell-based therapies for respiratory virus infections: Applicability to COVID-19. Eur. Respir. J. 2020, 55. [Google Scholar] [CrossRef]
  191. Powell, S.B.; Silvestri, J.M. Safety of intratracheal administration of human umbilical cord blood derived mesenchymal stromal cells in extremely low birth weight preterm infants. J. Pediatr. 2019, 210, 209–213. [Google Scholar] [CrossRef]
  192. Wu, K.-H.; Chao, Y.-H.; Weng, T.-F.; Li, J.-P. Clinical Consideration for Mesenchymal Stem Cells in Treatment of COVID-19. Curr. Pharm. Des. 2022, 28, 2991–2994. [Google Scholar] [CrossRef]
  193. Guo, Z.; Chen, Y.; Luo, X.; He, X.; Zhang, Y.; Wang, J. Administration of umbilical cord mesenchymal stem cells in patients with severe COVID-19 pneumonia. Crit. Care 2020, 24, 1–3. [Google Scholar] [CrossRef]
  194. Omar, S.A.; Abdul-Hafez, A.; Ibrahim, S.; Pillai, N.; Abdulmageed, M.; Thiruvenkataramani, R.P.; Mohamed, T.; Madhukar, B.V.; Uhal, B.D. Stem-cell therapy for bronchopulmonary dysplasia (BPD) in newborns. Cells 2022, 11, 1275. [Google Scholar] [CrossRef]
  195. Ringdén, O.; Gustafsson, B.; Sadeghi, B. Mesenchymal stromal cells in pediatric hematopoietic cell transplantation a review and a pilot study in children treated with decidua stromal cells for acute graft-versus-host disease. Front. Immunol. 2020, 11, 567210. [Google Scholar] [CrossRef] [PubMed]
  196. Izadi, M.; Sadr Hashemi Nejad, A.; Moazenchi, M.; Masoumi, S.; Rabbani, A.; Kompani, F.; Hedayati Asl, A.A.; Abbasi Kakroodi, F.; Jaroughi, N.; Mohseni Meybodi, M.A.; et al. Mesenchymal stem cell transplantation in newly diagnosed type-1 diabetes patients: A phase I/II randomized placebo-controlled clinical trial. Stem Cell Res. Ther. 2022, 13, 264. [Google Scholar] [CrossRef] [PubMed]
Table 1. Systemic manifestations of COVID-19 infection.
Table 1. Systemic manifestations of COVID-19 infection.
Organ SystemManifestations of COVID-19 Infection
Pulmonary involvementPoor ventilation
Acute respiratory distress syndrome (ARDS)
Cardiovascular involvementCardiovascular disorder
Arrythmia
Myocarditis
Myocardial infarction
Acute coronary syndrome (ACS)
Acute cor pulmonale
Kawasaki-like syndrome
Gastrointestinal and hepatic involvementNausea and vomiting
Diarrhea
Abdominal pain
Anorexia
Elevated bilirubin
Elevated Aminotransferases
Renal involvementAcute kidney injury
Hematuria
Proteinuria
Hematological involvementNeutrophilia
Lymphopenia
Thrombocytopenia
Elevated procalcitonin, CRP and ferritin levels
Elevated d-dimer
Deep venous thrombosis
Pulmonary embolism
Catheter-related thrombosis
Neurological involvementHeadache
Dizziness
Anosmia
Ageusia
Nerve pain
Myalgia
Increased depression
Cerebral hemorrhage
Stroke
Acute necrotizing encephalopathy
Guillain barre syndrome
Endocrine involvementHyperglycemia
Diabetic ketoacidosis
Dermatological involvementPetechiae
Urticaria
Vesicles
Table 2. Clinical trial on MSC infusion for patients with COVID-19.
Table 2. Clinical trial on MSC infusion for patients with COVID-19.
NoStudy TitleTrial ID NOPhaseIndicationsSource of MSCsRoute and Time of
Administration
Dose of MSCsNumber of PatientsCountryReference
1Safety of DW-MSC infusion in patients with low clinical risk COVID-19 infection: a randomized, double-blind, placebo-controlled trialNCT04535856Phase 1Low clinical risk COVID-19Allogenic UC-MSCIntravenous infusions, 1 roundHigh dose: 1 × 108 cells/round or
Low dose: 5 × 107 cells/round
9Indonesia[162]
2Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cellsChiCTR2000031494Phase 1Severe/critical COVID-19Human umbilical cordIntravenous administration
1 round
2 × 106 cells/kg41China[163]
3Mesenchymal stem cells derived from perinatal tissues for treatment of critically ill COVID-19-induced ARDS patients: a case seriesIRCT20200217046526N2Phase 1ARDS in COVID-19Allogeneic umbilical cord/placentaIntravenous infusions, 3 round (at days 0, 2, and 4)2 × 108 cells/round11Iran[164]
4Treatment of COVID-19-associated ARDS with mesenchymal stromal cells: a multicenter randomized double-blind trialNCT04333368Phase 2ARDS in COVID-19Umbilical cordIntravenous infusions, 3 round (at days 1, 3 ± 1, and 5 ± 1)1 × 106 cells/kg/round47French[165]
5Evaluation of the safety and efficacy of using human menstrual blood-derived mesenchymal stromal cells in treating severe and critically ill COVID-19 patients: An exploratory clinical trialChiCTR2000029606Phase 1Severe and critical COVID-19Allogenic menstrual bloodIntravenous infusions, 3 round (1, 3, 7)Total 9 × 107 cells44China[166]
6Mesenchymal stromal cell therapy for COVID-19-induced ARDS patients: a successful phase 1, control-placebo group, clinical trialIRCT20160809029275N1Phase 1ARDS in COVID-19Allogenic Umbilical cordIntravenous infusions, 3 round (1, 3, 5)1 × 106 cells/kg/round20Iran[167]
7Conquering the cytokine storm in COVID-19-induced ARDS using placenta-derived decidua stromal cellsIRCT2017010531786N1Phase 1/2ARDS in COVID-19PlacentaIntravenous infusions, 1 or 2 rounds1 × 106 cells/kg/round10Iran[168]
8Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized controlled trialNCT04355728
phase 1/2aARDS in COVID-19Allogeneic umbilical cordIntravenous infusions, 2 round (at days 0 and 3)10 ± 2 × 107 cells/round24USA[169]
9Bone marrow-derived mesenchymal stromal cell therapy in severe COVID-19: preliminary results of a phase I/II clinical trialNCT04445454Phase 1/2Severe COVID-19Bone marrowIntravenous infusions, 3 round (1, 4 (±1), 7 (±1))1.5–3 × 106 cells/kg/round32Belgium[170]
10Umbilical cord mesenchymal stromal cells as critical COVID-19 adjuvant therapy: A randomized controlled trialNCT04457609Phase 1ARDS in COVID-19Allogenic umbilical cordIntravenous infusions, 1 round1 × 106 cells/kg/round40Indonesia[171]
11Safety and efficacy study of allogeneic human menstrual blood stromal cell secretome to treat severe COVID-19 patients: clinical trial phase I & IIIRCT20180619040147N6Phase 1/2Severe COVID-19Allogeneic human menstrual bloodIntravenous infusions, 5 round (for 5 consecutive days)5 mL MenSCs-derived secretome diluted in 100 mL
of normalsaline
30Iran[172]
12Human mesenchymal stem cell therapy in severe COVID-19 patients: 2-year follow-up results of a randomized, double- blind, placebo-controlled trialNCT04288102Phase 2Severe COVID-19Allogenic umbilical cordIntravenous infusions, 3 round (at days 0, 3, and 6)4 × 107 cells/round100China[173]
13Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: a phase 1 clinical trialNCT04252118
Phase 1Moderate and severe COVID-19Allogeneic umbilical cordIntravenous infusions, 3 round (at days 0, 3, and 6)3 × 107 cells/round18China[174]
14Nebulized exosomes derived from allogenic adipose tissue mesenchymal stromal cells in patients with severe COVID-19: a pilot studyNCT04276987.Phase 2aSevere COVID-19Allogeneic adipose tissueInhalation 5 round (at days 1–5)2 × 108 particles/round (total 2 × 109)7China[175]
15Safety and long-term improvement of mesenchymal stromal cell infusion in critically
COVID-19 patients: a randomized clinical trial
U1111-1254-9819Phase 1/2Critical COVID-19Allogeneic umbilical cordIntravenous infusions, 3 round (at days 1, 3, and 5)5 × 105 cells/kg/round17Brazil[176]
16Allogenic mesenchymal stromal cells and their extracellular vesicles in COVID-19 induced ARDS: a randomized controlled trialIRCT20200217046526N2Phase 2ARDS in COVID-19Allogenic perinatal tissue1st round: intravenous infusions
2nd round:
intravenous infusion or inhalation
IV: 1 × 108 cells/round
Inhalation: 2 × 108 cells/round
43Iran[177]
17The systematic effect of mesenchymal stem cell therapy in critical COVID-19 patients: a prospective double controlled trialNCT04392778Phase 1/2Critical COVID-19Umbilical cordIntravenous infusions, 3 round (at days 0, 3, and 6)3 × 106 cells/kg/round30Turkey[178]
18Cell therapy in patients with COVID-19 using Wharton’s jelly mesenchymal stem cells: a phase 1 clinical trialIRCT20190717044241N2Phase 1Severe COVID-19Umbilical cordIntravenous infusions, 3 round (at days 0, 3, and 6)1.5 × 108 cells/round5Iran[179]
19Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: a randomized, double-blind, placebo-controlled phase 2 trialNCT04288102Phase 2Severe COVID-19Umbilical cordIntravenous infusions, 3 round (at days 0, 3, and 6)4 × 107 cells/round100China[180]
20Safety 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)NCT04336254Phase 1/2Severe COVID-19Allogeneic dental pulpIntravenous infusions, 3 round (at days 1, 4, and 7)3 × 107 cells/round20China[181]
21Human placenta-derived mesenchymal stem cells transplantation in patients with acute respiratory distress syndrome (ARDS) caused by COVID-19 (phase I clinical trial): safety profile assessmentIRCT20200621047859N4.Phase 1ARDS in COVID-19Allogenic placentaIntravenous infusions, 1 round1 × 106 cells/kg/round20Iran[182]
22Nebulization therapy with umbilical cord mesenchymal stem cell-derived exosomes for COVID-19 pneumoniaChiCTR2000030261Phase 1Pneumonia in COVID-19Umbilical cordNebulization, 1 round1 × 106 cells/kg/round7China[183]
23A randomized trial of mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome from COVID-19NCT04371393Phase 3ARDS in COVID-19Allogenic bone marrowIntravenous infusions, 2 round (two infusions
during the first week, with the second infusion
4 days after the first infusion (±1 day)
2 × 106 cells/kg/round222USA[184]
Table 3. Effectiveness of MSC treatment in clinical trial for patients with COVID-19.
Table 3. Effectiveness of MSC treatment in clinical trial for patients with COVID-19.
Study TitleIndicationsEffectiveness of MSC TreatmentReference
Clinical SymptomsCytokine Storm BiomarkersLung Image (Chest CT)
Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cellsSevere/critical COVID-19Improvement of weakness, fatigue, shortness of breath, and oxygenation index as early as the third day↓ CRP and IL-6Shorter lung inflammation absorption[163]
Mesenchymal stem cells derived from perinatal tissues for treatment of critically ill COVID-19-induced ARDS patients: a case seriesARDS in COVID-19Reduced dyspnea and increased SpO2 within 2–4 days↓ TNF-α, IL-8, and CRPReduction in ground-glass opacities or consolidation[164]
Treatment of COVID-19-associated ARDS with mesenchymal stromal cells: a multicenter randomized double-blind trialARDS in COVID-19Significant increase in PaO2/FiO2 ratios[165]
Evaluation of the safety and efficacy of using human menstrual blood-derived mesenchymal stromal cells in treating severe and critically ill COVID-19 patients: an exploratory clinical trialSevere and critical COVID-19Significant improvement in dyspnea on days 1, 3, and 5 and significant improvements in SpO2 and PaO2
Significantly lower mortality
Lung clearly[166]
Mesenchymal stromal cell therapy for COVID-19-induced ARDS patients: a successful phase 1, control-placebo group, clinical trialARDS in COVID-19Improve the SPO2/FIO2 ratio↓ CRP, IL-6, IFN-γ, TNF-α, and IL-17A
↑ TGF-B, IL-1B, and IL-10
[167]
Conquering the cytokine storm in COVID-19-induced ARDS using placenta-derived decidua stromal cellsARDS in COVID-19Improvement in oxygenation levels, with a median increase from 80.5% to 95%↓ IL-6 and CRPPulmonary infiltrates disappeared[168]
Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized controlled trialARDS in COVID-19Improved patient survival and a shorter time to recovery↓ GM-CSF, IFN-r, IL-5, IL-6, IL-7, TNF-a, and TNF-b[169]
Bone marrow-derived mesenchymal stromal cell therapy in severe COVID-19: preliminary results of a phase I/II clinical trialSevere COVID-19Higher survival rate in the MSC group at both 28 and 60 days↓ D-dimer[170]
Umbilical cord mesenchymal stromal cells as critical COVID-19 adjuvant therapy: A randomized controlled trialARDS in COVID-19Survival rate 2.5 times higher in the UC-MSC group than in the control group↓ IL-6[171]
Safety and efficacy study of allogeneic human menstrual blood stromal cells secretome to treat severe COVID-19 patients: clinical trial phase I & IISevere COVID-19Higher survival rate↓ CRP, LDH, D-dimer, and ferritin levelsImprovement of lung involvement[172]
Human mesenchymal stem cell therapy in severe COVID-19 patients: 2-year follow-up results of a randomized, double- blind, placebo-controlled trialSevere COVID-19Higher general health score on the Short Form 36 questionnaire[173]
Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: a phase 1 clinical trialModerate and severe COVID-19↓ IL-6, IFN-γ, TNF-α, MCP-1, IP-10, IL-22, IL-1RA, IL-18, IL-8, and MIP-1Complete fading of lung lesions within 2 weeks[174]
Nebulized exosomes derived from allogenic adipose tissue mesenchymal stromal cells in patients with severe COVID-19: a pilot studySevere COVID-19↓ CRP, LDH, and IL-6Resolution of pulmonary lesions[175]
Safety and long-term improvement of mesenchymal stromal cell infusion in critically COVID-19 patients: a randomized clinical trialCritical COVID-19↓ Ferritin, IL-6, and MCP1-CCL2, CRP, D-dimer, and neutrophil levels
↑ TCD3, TCD4, and NK lymphocytes
Decrease in the extent of lung damage was observed in the fourth month[176]
Allogenic mesenchymal stromal cells and their extracellular vesicles in COVID-19 induced ARDS: a randomized controlled trialARDS in COVID-19↓ IL-6, TNF-α, IFN-γ, and CRP[177]
The systematic effect of mesenchymal stem cell therapy in critical COVID-19 patients: a prospective double controlled trialCritical COVID-19↓ Ferritin, fibrinogen, and CRP[178]
Cell therapy in patients with COVID-19 using Wharton’s jelly mesenchymal stem cells: a phase 1 clinical trialSevere COVID-19↓ Ferritin[179]
Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: a randomized, double-blind, placebo-controlled phase 2 trialSevere COVID-19Significant reduction in the proportions of solid component lesion volume[180]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guo, B.-C.; Wu, K.-H.; Chen, C.-Y.; Lin, W.-Y.; Chang, Y.-J.; Lee, T.-A.; Lin, M.-J.; Wu, H.-P. Mesenchymal Stem Cells in the Treatment of COVID-19. Int. J. Mol. Sci. 2023, 24, 14800. https://doi.org/10.3390/ijms241914800

AMA Style

Guo B-C, Wu K-H, Chen C-Y, Lin W-Y, Chang Y-J, Lee T-A, Lin M-J, Wu H-P. Mesenchymal Stem Cells in the Treatment of COVID-19. International Journal of Molecular Sciences. 2023; 24(19):14800. https://doi.org/10.3390/ijms241914800

Chicago/Turabian Style

Guo, Bei-Cyuan, Kang-Hsi Wu, Chun-Yu Chen, Wen-Ya Lin, Yu-Jun Chang, Tai-An Lee, Mao-Jen Lin, and Han-Ping Wu. 2023. "Mesenchymal Stem Cells in the Treatment of COVID-19" International Journal of Molecular Sciences 24, no. 19: 14800. https://doi.org/10.3390/ijms241914800

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop