Next Article in Journal
Intelligent Control with Artificial Neural Networks for Automated Insulin Delivery Systems
Next Article in Special Issue
Intraoperative Creation of Tissue-Engineered Grafts with Minimally Manipulated Cells: New Concept of Bone Tissue Engineering In Situ
Previous Article in Journal
Solid PRF Serves as Basis for Guided Open Wound Healing of the Ridge after Tooth Extraction by Accelerating the Wound Healing Time Course—A Prospective Parallel Arm Randomized Controlled Single Blind Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mesenchymal Stem Cells and MSCs-Derived Extracellular Vesicles in Infectious Diseases: From Basic Research to Clinical Practice

by
Natalia Yudintceva
1,2,*,
Natalia Mikhailova
1,
Viacheslav Fedorov
2,
Konstantin Samochernych
2,
Tatiana Vinogradova
3,
Alexandr Muraviov
3 and
Maxim Shevtsov
1,2,*
1
Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
2
Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
3
Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, St. Petersburg 191036, Russia
*
Authors to whom correspondence should be addressed.
Bioengineering 2022, 9(11), 662; https://doi.org/10.3390/bioengineering9110662
Submission received: 27 September 2022 / Revised: 30 October 2022 / Accepted: 4 November 2022 / Published: 8 November 2022
(This article belongs to the Special Issue Mesenchymal Stem Cells for Tissue Engineering and Modelling)

Abstract

:
Mesenchymal stem cells (MSCs) are attractive in various fields of regenerative medicine due to their therapeutic potential and complex unique properties. Basic stem cell research and the global COVID-19 pandemic have given impetus to the development of cell therapy for infectious diseases. The aim of this review was to systematize scientific data on the applications of mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (MSC-EVs) in the combined treatment of infectious diseases. Application of MSCs and MSC-EVs in the treatment of infectious diseases has immunomodulatory, anti-inflammatory, and antibacterial effects, and also promotes the restoration of the epithelium and stimulates tissue regeneration. The use of MSC-EVs is a promising cell-free treatment strategy that allows solving the problems associated with the safety of cell therapy and increasing its effectiveness. In this review, experimental data and clinical trials based on MSCs and MSC-EVs for the treatment of infectious diseases are presented. MSCs and MSC-EVs can be a promising tool for the treatment of various infectious diseases, particularly in combination with antiviral drugs. Employment of MSC-derived EVs represents a more promising strategy for cell-free treatment, demonstrating a high therapeutic potential in preclinical studies.

1. Introduction

Infectious diseases are a large group of diseases caused by the impact of various pathogenic or conditionally biological agents on the human body. Several types are distinguished depending on the origin of the pathogen: viral, bacterial, fungal, as well as infections caused by prions, protozoa, and parasites. There are many historic examples of the devastating consequences caused by infectious diseases (e.g., smallpox, plague, cholera, typhoid, influenza, etc.), which are called “plague diseases”. Despite the sanitary well-being and the achievements of modern medicine, it is naive to believe that humanity has defeated infectious diseases, and each of us is not at risk. Currently, epidemics of COVID-19, tuberculosis (TB), AIDS, malaria, measles, influenza, and other diseases are constantly active in the world. According to the World Health Organization (WHO), about 50% of the world’s population lives in conditions of constant threat of epidemics (www.who.int (accessed on 12 February 2019)).
There are several objective conditions for the development of infectious diseases: the active development of tourism [1,2,3], the increase in migration processes [4,5,6,7,8], returning and re-emerging diseases [9,10], as well as the likelihood of using pathogens of various infectious diseases as biological weapons [11,12,13]. Improved social and environmental conditions help with reducing the risk of contracting and spreading infectious diseases [14,15], but, paradoxically, as the living standards rise, the mortality from some of them also increases. For example, in the case of paralytic poliomyelitis or chicken pox, the severity of the infectious process complications (including pneumonia, acute neurological disorders, thrombocytopenia, chickenpox encephalitis with damage to the myelin sheaths of the brain and spinal cord, etc.) is directly correlated to the age of the patient [16,17]. The use of antibiotics and active immunization of the population have made it possible to defeat or take control of most of the infections, however, there are still many infectious diseases that cannot be treated (AIDS, a multidrug resistant form of TB, viral hepatitis C, prion infections, etc.), as well as leading to serious complications (COVID-19, influenza, etc.).
The active growth of basic stem cell research has given impetus to the development of translational medicine, which is grounded on the results obtained and promotes new treatments for various diseases. One of these areas is cell therapy, which is based on the use of cells and cellular secretome that can stimulate tissue regeneration, provide anti-inflammatory, immunomodulatory, and other therapeutic effects on the body [18,19]. Despite the fact that many biological mechanisms concerning the effect of MSCs on damaged tissues remain insufficiently studied, the possibility of using cell therapy for various diseases, including infectious diseases, both as a monotherapy or in combination with other agents, is currently being actively studied [20,21,22,23].
The global stem cell therapy market is projected to grow to USD 18.51 billion by 2026 at a compound annual growth rate of 9.8% (Figure S1). Such predictions are driven primarily by increased awareness of the therapeutic efficacy of stem cells, as well as the development of the infrastructure associated with obtaining and banking stem cells. The largest companies in this market are Anterogen Co., Ltd. (Seoul, Korea), Mesoblast Ltd. (Melbourne, Australia), Osiris Therapeutics Inc. (Columbia, MD, USA), AlloSource (Centennial, CO, USA), Cellular Engineering Technologies (Coralville, IA, USA), BIOTIME Inc. (Carlsbad, CA, USA), Astellas Pharma US Inc. (Northbrook, IL, USA), Vericel (Cambridge, MA, USA), RTI Surgical Inc. (Deerfield, IL, USA), and Takara Bio Company (Kusatsu, Tokyo).
At the moment, cell therapy is not yet widely used and distributed, which is associated with its high cost, as well as the personalized approach and the use of autologous cells. However, in recent years, there has been a significant demand for allogeneic cells due to a more affordable process for their cultivation and an increase in the commercialization of allogeneic therapy products. The most commonly used allogeneic stem cells in clinical research include: mesenchymal stem cells (MSCs) isolated from bone marrow (bone marrow-derived mesenchymal stem cells (BM-MSCs), adipose tissue (adipose tissue-derived mesenchymal stem cells (A-MSCs)), umbilical cord (umbilical cord blood-derived mesenchymal stem cells (UC-MSCs), and placenta (placenta-derived mesenchymal stem cells (P-MSCs). The data available support the safety of therapy with both autologous and allogeneic MSCs. Despite the fact that the evidence on the effectiveness of cell therapy is often preliminary, the great advantages of MSCs are still their weak immunogenic properties and the possibility of rapid application for the treatment of various diseases [24,25].

2. MSCs and MSC-Derived Extracellular Vesicles (MSC-EVs)

The concept of “adult” MSCs, first proposed by Kaplan, appeared in accordance with the concept of the cell origin in the embryonic mesoderm [26]. Despite that it does not strictly correspond to the biological definition of MSCs [27,28], this term is widely used by clinicians and scientists to this day [29]. Due to the use of various methods for obtaining and culturing stem cells, the discussion on the specific characteristics used to determine MSCs is rather conflicting. Since these cells can be isolated from almost any tissue, it has been suggested that MSCs from different sources may be sufficiently distinct to combine them into a single classification (Table 1).
BM-MSCs have a longer doubling time and age earlier compared to cells obtained from other sources [41,42]. Approximately 98–100% of cells remain viable when derived from adipose tissue (A-MSCs) compared to cell isolation from bone marrow [43]. A-MSCs secrete various cytokines and growth factors with anti-inflammatory, antiapoptotic, and immunomodulatory characteristics including vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and insulin-like growth factor (IGF), which are involved in angiogenesis and damaged tissues repair. Additionally, due to their immunomodulatory effects, A-MSCs are an excellent source for allogeneic transplants, since they do not express type II major histocompatibility complex (MHC class II) and the risk of transplant rejection is thus minimized [44]. A minimal risk of immune response was also observed with in vivo administration of allogeneic UC-MSCs. This property and ease of their preparation also make UC-MSCs suitable candidates for cell therapy [45]. P-MSCs express embryonic stem cell markers such as c-Kit, Oct-4, stage-specific embryonic antigen (SSEA-4), as well as markers that determine the sex of the donor Y-box 2. One of the main advantages when applying P-MSCs is their high proliferative properties and plasticity [46,47].
In 2006, the International Society for Cell Therapy (www.isctglobal.org) defined three minimum criteria for identifying MSCs: (1) adherence to plastic under standard culture conditions; (2) the ability to differentiate into osteogenic, adipocyte, and chondrogenic directions under appropriate cultivation conditions; and (3) phenotyping by the presence/absence of surface markers: ≥95% positive for CD105, CD73, and CD90; ≤5.2% negative for CD45, CD34, CD14/CD11b, CD79a/CD19, and HLA-DR [48]. These cell types (BM-MSCs, A-MSCs, UC-MSCs, and P-MSCs) share the minimal criteria defined by ISCT and have additional characteristics which are associated with their tissue specificity [49,50].
Due to their therapeutic potential and unique properties, MSCs are appealing for various fields of regenerative medicine [51,52], cancer therapy [53,54,55], and infectious diseases [20,56,57,58,59]. MSCs synthesize factors that can restore damaged tissues. It has recently been suggested that MSCs are able to modulate cellular autophagy in damaged tissues/organs. MSCs can affect the autophagy of immune cells involved in injury-induced inflammation, reducing their survival, proliferation, and level of inflammation. At the same time, MSCs promote survival, proliferation, and differentiation of endogenous adult or progenitor cells, thereby promoting tissue repair [60].
Initial preclinical data on the therapeutic efficacy of MSCs focused mainly on the regenerative and differentiating ability of cells. However, there is now increasing evidence that many, if not all, of the positive effects of MSCs are associated with the paracrine activity of cells and their secretome, which consists of EVs [61,62], soluble proteins, cytokines, chemokines, and growth factors [63,64,65], and not only with the integration of cells into the damaged area [66,67]. MSCs have been found to play an immunomodulatory role in numerous infection diseases through the production of soluble factors, and the transfer of EVs containing various molecules [68]. It has been established that MSC-EVs have the same immunomodulatory and anti-inflammatory and other effects as their parental cells and recapitulate a broad range of the therapeutic effects shown by MSC treatment. The functional differences between MSC and MSC-EVs are not significant [69]. However, there are different mechanisms underlying the interaction of various MSCs or MSC-EVs with immune cells. EVs derived from different types of MSCs have similar and unique characteristics (Table 2).
Currently, most clinical trials of MSC therapy for viral and bacterial infectious diseases have focused on patients who have not responded to traditional disease drug therapy as COVID-19, AIDS, and TB. However, the use of MSCs in therapeutic treatments still has many challenges. An increasing number of studies reveal that MSCs are highly heterogeneous with different multipotential properties, progenitors, and cell states. In addition, MSCs isolated from different sources exhibit distinct characteristics, known as tissue sources-associated heterogeneity [86,87,88]. Moreover, the intravenous administration of MSCs can lead to aggregation or clumping of cells in the vascular system and is associated with the risk of mutagenicity and oncogenicity [89,90].
The results of preclinical studies in vitro and in vivo show that MSC-EVs also exhibit significant therapeutic properties in many pathophysiological conditions of the body, restoring damaged organs and tissues [91,92,93] without the risks associated with direct cell engraftment (i.e., immunogenicity, tumorigenicity, and teratoma formation) [94,95]. The application of MSC-EVs in the treatment of diseases is a novel concept with particular advantages over the whole-cell therapy. MSC-EVs are well-tolerated and have low immu-nogenicity and also have a more stable membrane structure than MSCs. Another advantage of MSC-EVs over MSCs is the possibility of storing them for several weeks/months allowing their safe transportation and delayed therapeutic use [96]. These advantages of EVs provide broader prospects for disease treatment. However, the studying of the mechanism of EVs in the treatment of diseases is the primary connection to future clinical research.

2.1. Extracellular Vesicles (EVs)

EVs are heterogeneous vesicles surrounded by a lipid bilayer and secreted not only by MSCs, but also by all cell types. EVs mediate intercellular communication and are involved in many physiological and pathophysiological processes, including modulating immune responses, maintaining homeostasis, inflammation, angiogenesis, and others [97,98,99,100] (Figure 1).
Depending on the origin and size of EVs, they are divided into various subtypes: ectosomes, microvesicles, microparticles, exosomes, oncosomes, apoptotic bodies, etc. [101]. However, these EV subtypes are further characterized by different, often overlapping, definitions based primarily on vesicle biogenesis (cellular pathway, cellular or tissue identity, etc.) [102]. In order to avoid contradictions in definitions, the International Society for Extracellular Vesicles (ISEV) (www.isev.org (accessed on 3 May 2018)) proposed in 2018 to call a particle secreted by a cell an “extracellular vesicle” if its specific biogenetic origin cannot be demonstrated.
The regenerative potential of EVs is mainly explained by the regulation of apoptosis, cell proliferation, differentiation, angiogenesis, and inflammation [103]. The exact mechanisms underlying the therapeutic effects of EVs remain to be fully elucidated. However, several factors have proven to be promising contenders for the transfer of regenerative potential: microRNA (miRNA), messenger RNA (mRNA), and proteins. MSC-EVs of different origin are quite heterogeneous and have significant differences in the qualitative and quantitative composition of proteins, cytokines, nucleic acids, lipids, mRNA, microRNA, and other active components [104,105]. The paracrine action of EVs can be mediated through three mechanisms, including internalization, direct fusion, and ligand–receptor interaction with the target cell [106]. Using these pathways, EVs deliver various biomolecules and are involved in the inhibition and/or induction of signaling in target cells.

2.2. Application of Tissue Engineering Methods to Improve Therapeutic Effectiveness of MSCs and MSC-EVs

The rather low efficiency of cell therapy is one of the factors that significantly limits its use. The effectiveness of cell therapy is influenced by various factors: methods of administration, multiplicity, stability, efficiency of retention in the target tissue, heterogeneity, content of vesicles, etc. Positive effects of MSCs can be further enhanced in various ways, for example, by changing the method of cultivation (under conditions of hypoxia compared with normoxic conditions) [107,108], or the form of cultivation (3D or 2D culture) [109,110,111], as well as exposing cells to various influences (e.g., heat shock) [112], or genetic modifications [113]. It has been shown that pretreated MSCs demonstrate enhanced differentiation efficiency [114,115,116,117,118], improved paracrine functions [119], superior survival [120], and an enhanced ability of EVs to accumulate and remain in damaged tissue [121,122,123,124]. An additional approach that makes it possible to increase the efficiency of cell therapy is the use of various biomaterials [125,126], as well as the cultivation of cells in cell sheets [127,128,129].
Various methods can also be used to enhance the therapeutic efficacy of MSC-EVs: preconditioning of donor cells to increase the beneficial contents of EVs [130,131,132], using genetically modified cells [133] to change the composition of EVs [134,135], and dosing and multiple application [136]. However, each of these methods has its drawbacks. For example, the use of preconditioned media, unfortunately, does not give a high yield of MSC-EVs, which is a limiting factor for the use of cell-free therapy, while the genetic modification of cells and repeated administration of EVs can be potential risk factors for tumor growth.
The structural versatility of EVs provides an opportunity for surface modifications that can be performed using various methods, such as genetic engineering, chemical and physical methods, and microfluidic technologies [137]. The efficiency and scalability of methods for modifying EVs are critical in defining the scope for clinical application [138]. It is known that integrins are present on the surface membranes, and RGD peptides (Arg-Gly-Asp) have a high binding affinity for integrins [139,140]. RGD-modified EVs have been shown to exhibit increased targeting to blood vessels and represent a potential new therapeutic tool for angiogenesis therapy [141].
Another approach that contributes to the creation of a more stable therapeutic effect of EVs is the use of hydrogels. To date, there are several studies on the development of hydrogel scaffolds for loading EVs and evaluating the mechanisms of interaction between gels and EVs, which are still unclear [142,143]. It has been shown that a biocompatible self-assembling RGD hydrogel easily conjugates with EVs, and such constructs increase the therapeutic efficacy of MSC-derived vesicles for the treatment of kidneys [144], liver [145], and other organs [146]. Loading EVs with various drugs also enhances their therapeutic effect [147,148,149].
Research conducted to reveal the therapeutic potential of EVs, especially those that secrete MSCs, has proven to be significant for regenerative medicine. However, how EVs promote tissue regeneration and what drives their regenerative effect is still far from clear.

2.3. Mechanisms of Immunomodulatory Action of MSCs and MSC-EVs

MSCs are involved in innate and adaptive immunity and their immunomodulatory functions are manifested mainly when interacting with immune cells (T-cells, B-cells, natural killer (NK-cells), macrophages, monocytes, dendritic cells, and neutrophils) through the formation of intercellular contacts and implementation of paracrine activity [150]. By influencing the adaptive immune system, T-cells in particular, MSCs inhibit the differentiation of Th17, inducing the production of IL-10 and PGE2, as well as inhibiting IL-17, IL-22, and IFN-γ [151]. However, the mechanisms underlying the interactions between MSCs and Th17 lymphocytes have not yet been fully understood. In the innate immune system, MSCs interact with NK-cells, inhibit their proliferation with the help of IL-2, and induce cytotoxic activity, as well as the production of cytokines through the secretion of IDO and PGE2 [152].
The key role in the development of the immunomodulatory potential of MSCs is played by the interaction of cells with regulatory T-cells and monocytes. A-MSCs have been shown to regulate T-cell function by inducing suppressor T-cells and inhibiting the production of cytotoxic CD8+ T-cells, NK-cells, and proinflammatory cytokines including tumor necrosis factor-alpha (TFN-alpha), IFN-gamma, and IL-12. The secretion of A-MSCs of soluble factors such as IL-10, TGF-beta, and PGE2 renders cells immunosuppressive [44,153]. In this regard, A-MSCs have the strongest immunomodulatory effect compared to BM-MSCs and can become a better alternative for immunomodulatory therapy [154].
Through intercellular interactions, MSCs increase the survival of B-cells and promote their differentiation [155]. A-MSCs not only inhibit caspase-3-mediated B-cell apoptosis by up-regulating VEGF expression, but also inhibit proliferation by blocking the B-lymphocyte cell cycle in the G0/G1 phase by activating p38 protein kinase (MARK) [156]. In addition, MSCs prevent the death of neutrophils through an ICAM-1-dependent mechanism and exert a tissue protective effect [157]. Thus, the interaction of MSCs with immune cells contributes to a decrease in the inflammatory response, as well as the regeneration of damaged tissue.

3. Viral Infectious Diseases

Over the past decades, a huge number of experimental and clinical studies have been devoted to the use of cell therapy in the treatment of oncological, cardiovascular, neurodegenerative, and other diseases [158,159,160]. Basic stem cell research and the global COVID-19 pandemic have given rise to the development of cell therapy for infectious diseases, which currently stands at 121 registered clinical trials [161].

3.1. COVID-19

Coronavirus and other respiratory viruses are the leading cause of morbidity and mortality in acute lung injury (ALI) and acute respiratory distress syndrome. Although scientific advances have enabled rapid progress in understanding pathogenesis and developing therapeutic agents, stem cell therapy has recently found numerous applications in the treatment of viral infections.
Severe forms of the disease caused by COVID-19 are accompanied by increased activation of the immune system, which, in addition to antiviral protection, leads to a side effect—damage to lung tissue and other organs. To date, several studies have proposed the use of MSCs for the treatment of pneumonia caused by COVID-19 [162,163,164,165]. MSCs have been shown to reduce inflammation and suppress viral infection [166]. In the ALI mouse model, it was shown that, due to the anti-inflammatory effect, MSCs improve lung function, synthesizing the keratinocytes growth factor (KGF), VEGF, and HGF to restore damaged epithelial cells and lung tissues. IDO, TGF-ß, and granulocyte-macrophage colony-stimulating factor (G-CSF) act on macrophages, neutrophils, and T-cells (Figure 2). The main mechanism of action is probably to reduce the secretion of inflammatory factors [167].
Clinical intravenous administration of MSCs has shown an increase in the number of peripheral lymphocytes, hyperactivation of some types of T-cells as well as a decrease in the level of C-reactive protein [168]. A major factor in organ damage in severe COVID-19 cases is the cytokine storm. Due to their strong immunomodulatory ability, MSCs not only suppress the cytokine storm, but also promote the activation of the endogenous regenerative mechanism [169]. At the same time, MSC-EVs play an important role in the implementation of intercellular communication, since they are able to enter the bloodstream, pass through it for long distances and pass through histohematic barriers [170,171].
Several clinical studies have demonstrated the ability of MSC-EVs to reduce the level of inflammatory factors and increase immunity in various forms of COVID-19 (NCT04384445, USA; NCT04276987, China; NCT04491240, Russia). Two clinical trials are currently underway: one study group (NCT04276987) is investigating the efficacy of inhaled treatment of COVID-19 pneumonia using EVs derived from A-MSCs, and the second one (NCT04313647) is evaluating their safety and tolerability in healthy volunteers.
Due to their specific structure, various drugs can be introduced into MSC-EVs to use them as delivery systems [172] and one of the tools in the treatment of viral infection [173]. In addition, compared with other types of treatment, such as monoclonal antibody therapy, the economic costs of obtaining and using MSC-EV are significantly lower, which is important when using this method during a pandemic [28]. Ongoing clinical trials highlight the potential benefits of using both MSCs and MSC-EVs for the treatment of patients with COVID-19. However, further studies to evaluate and confirm their efficacy and safety are needed.

3.2. Flu

Due to the fact that infectious diseases of the respiratory organs caused by various viruses can occur like the common cold but also have severe acute respiratory syndromes [174], it is rather difficult to determine the specific agents involved in the infection [175]. Currently, influenza therapy mainly includes antibacterial and antiviral drugs.
Several studies on animal models infected with the influenza virus have shown a positive effect of the use of MSCs of various tissue origins [176,177,178,179,180]. Cocultivation of BM-MSCs with H5N1 virus-infected AECs inhibits their permeability under in vitro conditions. Possible mechanisms for this are related to the secretion of angiopoietin-1 (Ang1) and KGF by BM-MSCs [178]. In vivo experiments demonstrate that BM-MSCs have a significant anti-inflammatory effect by increasing the number of macrophages and releasing various cytokines and interleukins: IL-1 beta, IL-4, IL-6, IL-8, and IL-17 [180,181]. Similar anti-inflammatory effects have been shown using another model of lung injury caused by the H9N2 virus [179]. Intravenous injection of a suspension of BM-MSCs into virus-infected mice significantly attenuates virus-induced lung inflammation by reducing the levels of chemokines (GM-CSF, MCP-1, KC, MIP-1α, and MIG) and proinflammatory factors IL-1 alpha, IL-6, TNF-alpha, and IFN-gamma. Using an in vitro model of lung injury caused by the H5N1 virus, human UC-MSCs, through the secretion of Ang1 and HGF, had the same anti-inflammatory effect as BM-MSCs [182]. In one clinical study in patients with lung injury caused by the H7N9 influenza virus, the use of MSCs did not cause side effects and significantly increased their survival [183]. Despite the data indicating the therapeutic effect of MSCs in various preclinical models of lung injury, some studies have shown that the use of a suspension of MSCs with an antiviral drug was ineffective [184,185]. In addition, when using cell therapy, it is necessary to take into account the condition of the donor and recipient. It has been shown that when MSCs are administered to a patient with ongoing disease, cells can become infected with the influenza virus, and transplantation of BM-MSCs from influenza virus-infected donors, in turn, can also transmit the infection to recipients. Thus, when using cell therapy in the treatment of pulmonary influenza, it is imperative to take into account these factors and observe safety.

3.3. AIDS

The human immunodeficiency virus (HIV) is caused by a retrovirus of the lentivirus genus. It affects cells of the immune system that have CD4 receptors on their surface: T-helpers, monocytes, macrophages, Langerhans cells, dendritic cells, and microglial cells. As a result, the work of the immune system is inhibited and the syndrome of acquired immune deficiency (AIDS) develops, the patient’s body loses the ability to defend itself against infections and tumors. Despite the fact that the first case of AIDS was discovered almost 27 years ago, it is still not possible to effectively control the AIDS pandemic [186]. Of the 35 million people living with HIV infection, a fraction survives thanks to antiretroviral therapy, but in the absence of it, death occurs on average 9–11 years after infection. There are currently three known cases of a cure for the virus. In the medical literature, they appear under the names “Berlin”, “London”, and “Sao Paulo” patients [187,188].
Recently, a new strategy for the treatment of HIV and AIDS using stem cells, in particular BM-MSCs, has emerged [189]. According to the data, circulating replicative HIV remains the most serious threat to effective AIDS therapy. The main therapy strategy is aimed at reducing the number of replicating virus particles. As a result of its application, the destruction of HIV circulating in the blood occurs with the help of erythrocytes integrated with the CD4 receptor and chemokine receptors, which selectively bind circulating HIV particles [190,191,192,193].
One of the most interesting studies focused on the use of MSCs to increase antiviral immune activity and minimize the amount of virus. It has been shown that the administration of MSCs, even in the absence of antiviral drugs, can enhance the host’s antiviral response due to the restoration of lymphoid follicles and mucosal immunity, all of which become the target of the virus at an early stage [194]. The results of scientific and clinical studies provide an appropriate scientific basis for the future use of MSCs in the treatment of HIV and other infectious diseases. Researchers are still developing comprehensive and effective treatments for AIDS and related conditions.
Cell-based therapies initially were reserved to the most severely affected patients with viral infectious diseases (COVID-19, flu, and AIDS) and most clinical trials were also focused on them (Table 3).
The application of cell and vesicles therapy in most of the clinical trials resulted in symptomatic relief and treatment success. However, in order to ensure the widespread clinical implementation of MSC-based therapy, there are many challenges that need to be resolved (stages of the disease, clinical indicators, gender and age of patients, the source and age of MSCs, etc.).

4. Bacterial Infectious Diseases

4.1. Tuberculosis

Tuberculosis (TB) is one of the 10 leading causes of death worldwide. According to WHO data for 2021, over 9.9 million people worldwide became infected and about 1.3 million people died from TB [218]. The emergence of the COVID-19 pandemic has severely disrupted global TB prevention and control [219,220]. Nearly half a million people suffer from the rifampin-resistant TB strain, of which 78% are multidrug-resistant. In this regard, the actual direction is the search for fundamentally new approaches in the treatment of resistant TB, among which a certain place is occupied by MSC therapy.
Once in the lower respiratory tract, mycobacteria (Mycobacterium tuberculosis (µTb)) are mainly absorbed by macrophages. In this case, the resulting inflammatory reaction causes a large number of immune cells (monocytes, dendritic cells, neutrophils, and T-lymphocytes) to be attracted to the infected area, resulting in the formation of tuberculous granuloma (TG), which is a pathological sign of TB [221,222]. TG formation is a key event in preventing the spread of infection, and the period during which µTb are able to avoid the host’s immune response and remain dormant can be decades [223]. Numerous studies show that MSCs are involved in the formation and development of TG. It was found using CD29 as a marker that the cells are in a cluster with acid-resistant bacteria and are distributed in the TG area. In the pathogenesis of TB, MSCs, on the one hand, are able to inhibit the T-cell response through the synthesis of nitric oxide (NO) and, thereby, reduce the immune response, and on the other hand, NO itself can inhibit the growth of µTb and limit their proliferation within TG. Thus, it can be assumed that the formation of TG is associated precisely with this mechanism [224]. It has been shown that MSCs are able to regulate and limit the growth of µTb [225] using scavenger receptors for this [226,227]. Whether MSCs can influence the growth of µTb in any other way is still a question that still needs further study.
It has been shown that MSCs are natural host cells of latent µTb infection. In addition, recent research found that MSCs exist in the lungs and extrapulmonary tuberculosis granuloma. After infection with MSCs, the metabolic activity of µTb in cells becomes low, and, thus, they gradually acquire resistance to antituberculosis drugs [228,229]. Thus, along with immune cells, MSCs can not only provide a niche for dormant µTb but also limit their growth to a certain extent and participate in the emergence and development of TB.
The incidence of TB largely depends not on primary or secondary infection but on the reactivation of the dormant form of TB against the background of the emerging immunodeficiency [230]. In this regard, in recent years, therapy methods aimed at increasing infection control, reducing inflammation by modulating the immune response, and reducing tissue damage have become widespread [231]. Immunomodulatory properties and the ability to replace or repair damaged tissues make MSCs ideal candidates for the treatment of both pulmonary and extrapulmonary TB [232]. A number of studies have shown that the therapeutic potential of MSCs is associated with the antibacterial activity of cells directed against various pathogens (Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, Streptococcus pneumonia) through the secretion of antimicrobial peptides [233,234,235]. However, it is not yet known whether MSCs affect µTb growth in the same way.
MSCs and MSC-EVs have a wide range of immunomodulatory effects on various cells of the immune system: they promote the function of regulatory T cells (Treg and Th2) [236,237], inhibit the release of IFN-gamma, regulate the balance of Th1/Th2 [238], promote polarization of macrophages from M1 to M2 by expression of IDO and activation of CD39 and CD73/adenosine signaling pathways [237,238,239], and inhibit activation and promote B cell transformation [240,241,242]. In addition, MSCs are able to regulate the survival of the alveolar epithelium by secreting factors KGF and HGF that protect cells from apoptosis [243].
Previously, we showed that intravenous administration of MSCs results in accumulation and retention of MSCs in µTb-affected rabbit kidney tissue, due to which the cells are able to reduce the level of the inflammatory response and enhance the process of tissue repair [244]. A decrease in the level of expression and synthesis of hydroxyproline, collagen Types I and III leads to a decrease in fibrosis, restoration of damage, and prevention of pulmonary edema [245,246,247].
A number of studies have shown that, at low concentrations, MSCs can inhibit the activation of lymphocytes [248]. Thus, the ratio of the number of MSCs and immune cells can be a turning point for inhibiting or activating the immune response. Overall, the results obtained with MSCs in vivo are encouraging, but the safety and efficacy of MSCs in the treatment of TB remains to be confirmed.

4.2. Cholera

Vibrio cholerae (VCh) is the causative agent of cholera, which is commonly associated with a high infection rate, mortality, and a major public health problem in many parts of the world [249,250]. According to WHO data, each year there are from 1.3 to 4 million cases of cholera, and 100,000 to 130,000 deaths worldwide due to cholera per year. The emergence of multidrug-resistant VCh strains in developing countries is of great concern [251,252]. The high mortality rate and the lack of effective antimicrobials necessitate the development of new effective approaches for the treatment of drug-resistant strains. Various vaccines have been developed (Dukoral, Shanhol, and Euvichol), but none provide complete long-term protection and are not approved for use in children under 1 year of age. Inflammation caused by the interaction of Vibrio cholerae with epithelial cells is considered as the main cause of the spread of bacteria in the gastrointestinal tract and the progression of its consequences. One of the effective therapeutic approaches to treatment is to reduce the level of inflammatory cytokines caused by VCh infection.
MSCs exert their antibacterial properties through the synthesis of compounds such as antimicrobial peptides (hCAP18/LL-37), which control the growth and reproduction of bacteria. One study using a neonatal mouse model showed the immunomodulatory effect of a medium conditioned with MSCs supplemented with LPS (lipopolysaccharide necessary to protect the body from VCh) [253], a decrease in the level of the inflammatory response and the induction of the production of vibriocidal antibodies that protect against VCh. In addition, MSCs have been shown to be effective in the treatment of bacterial sepsis [254,255].
A-MSCs show dual effects on inflammatory response and epithelial barrier integrity by reduction of bacterial attachment and increasing bacterial internalization. On the one hand, A-MSCs reduce bacterial adhesion and colony formation by secreting various antimicrobial peptides (including IDO, and TIMP). A decrease in the rate of bacterial adhesion, in turn, leads to a decrease in the expression of chloratoxin and an increase in the secretion of IL-6, which has a positive effect on maintaining the integrity of the epithelial barrier. On the other hand, increased bacterial internalization by cells stimulates the inflammatory reactions. An increase in the level of expression of the proinflammatory genes TNF-alpha, IL-1beta, and IL-8 leads to an increase in the level of cytokines, induction of apoptosis, and degradation of the tight junction between epithelial cells. Thus, A-MSCs are able to exert different effects on the inflammatory response and the integrity of the epithelial barrier by reducing bacterial adhesion and enhancing bacterial internalization. The probable reason for this effect is the high level of MSC expression of matrix metalloproteinases and tissue inhibitor of proteinases (TIMP), as well as other antibacterial peptides [256]. Therefore, it is recommended that future studies focus on the protective effects of MSCs’ secretome.
It can be assumed that the reduction of bacterial internalization may also become an appropriate therapeutic approach to limit the inflammatory reactions caused by VCh, while it is more efficient to use MSC-EVs as a therapeutic agent instead of intact cells.
Currently, the evaluation of the effectiveness of cell therapy for bacterial infectious diseases is carried out mainly in vitro and in vivo conditions. There are few clinical studies on this topic (Table 4).

5. Conclusions

The therapeutic use of MSCs is not an unrealistic goal, as the cells offer a promising treatment option for a number of diseases. Using MSC-EVs instead of cells seems to be a promising strategy for cell-free treatment, as it allows to solve various problems associated with cell administration. Nevertheless, for clinical use, a preliminary assessment of the safety, efficacy, and long-term results of using both various types of MSCs, regardless of the source of their production, and MSC-EVs, first in animal models and then in preclinical trials, is necessary. At present, studies are being actively carried out on the selection and establishment of optimal therapeutic doses and the frequency of administration of cells and vesicles, optimal methods for their management, assessment of cellular and vesicular heterogeneity, etc. In addition, it is known that the properties of MSCs change significantly under inflammatory or anti-inflammatory stimuli, and therefore, it remains to be seen how variability affects the immunomodulatory effects induced by cells and to establish which subpopulations of cells or extracellular vesicles are the most therapeutically effective. Although clinical research on MSCs is still in its infancy, there is great hope that MSCs and MSC-EVs will become promising tools for future clinical applications in the treatment of infectious diseases.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/bioengineering9110662/s1, Figure S1: Growth of the global stem cell therapy market.

Author Contributions

Conceptualization, N.Y. and M.S.; writing—original draft preparation, N.Y., V.F. and A.M.; writing—review and editing, M.S, N.M. and T.V.; supervision, K.S. and M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Ministry of Science and Higher Education of the Russian Federation (Agreement No. 075-15-2022-301).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that there is no conflict of interest regarding the publication of this article.

References

  1. Flaherty, G.T.; Hamer, D.H.; Chen, L.H. Travel in the Time of COVID: A Review of international travel health in a global pandemic. Curr. Infect. Dis. Rep. 2022, 24, 129–145. [Google Scholar] [CrossRef] [PubMed]
  2. Du, M.; Yuan, J.; Jing, W.; Liu, M.; Liu, J. The effect of international travel arrivals on the new HIV infections in 15-49 years aged group among 109 countries or territories from 2000 to 2018. Front. Public Health 2022, 10, 833551. [Google Scholar] [CrossRef] [PubMed]
  3. Bonato, F.; Ferreli, C.; Satta, R.; Rongioletti, F.; Atzori, L. Syphilis and the COVID-19 pandemic: Did the lockdown stop risky sexual behavior? Clin. Dermatol. 2021, 39, 710–713. [Google Scholar] [CrossRef] [PubMed]
  4. Abbas, M.; Aloudat, T.; Bartolomei, J.; Carballo, M.; Durieux-Paillard, S.; Gabus, L.; Jablonka, A.; Jackson, Y.; Kaojaroen, K.; Koch, D.; et al. Migrant and refugee populations: A public health and policy perspective on a continuing global crisis. Antimicrob. Resist. Infect. Control 2018, 7, 113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Dunn, R.N.; Husien, M.B. Spinal tuberculosis: Review of current management. Bone Jt. J. 2018, 1, 425–431. [Google Scholar] [CrossRef]
  6. Greenaway, C.; Castelli, F. Infectious diseases at different stages of migration: An expert review. J. Travel. Med. 2019, 26, taz007. [Google Scholar] [CrossRef]
  7. Seedat, F.; Hargreaves, S.; Nellums, L.B.; Ouyang, J.; Brown, M.; Friedland, J.S. How effective are approaches to migrant screening for infectious diseases in Europe? A systematic review. Lancet Infect. Dis. 2018, 18, e259–e271. [Google Scholar] [CrossRef]
  8. Castelli, F.; Sulis, G. Migration and infectious diseases. Clin. Microbiol. Infect. 2017, 23, 283–289. [Google Scholar] [CrossRef] [Green Version]
  9. Lycett, S.J.; Duchatel, F.; Digard, P. A brief history of bird flu. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2019, 74, 20180257. [Google Scholar] [CrossRef]
  10. Ashraf, N.; Kubat, R.C.; Poplin, V.; Adenis, A.A.; Denning, D.W.; Wright, L.; McCotter, O.; Schwartz, I.S.; Jackson, B.R.; Chiller, T.; et al. Redrawing the maps for endemic mycoses. Mycopathologia 2020, 185, 843–865. [Google Scholar] [CrossRef]
  11. Kotwal, A.; Yadav, A. Biothreat & One Health: Current scenario & way forward. Indian J. Med. Res. 2021, 153, 257–263. [Google Scholar] [CrossRef]
  12. Green, M.S.; LeDuc, J.; Cohen, D.; Franz, D.R. Confronting the threat of bioterrorism: Realities, challenges, and defensive strategies. Lancet Infect. Dis. 2019, 19, e2–e13. [Google Scholar] [CrossRef]
  13. Biselli, R.; Nisini, R.; Lista, F.; Autore, A.; Lastilla, M.; De Lorenzo, G.; Peragallo, M.S.; Stroffolini, T.; D’Amelio, R. A historical review of military medical strategies for fighting infectious diseases: From battlefields to global health. Biomedicines 2022, 10, 2050. [Google Scholar] [CrossRef]
  14. Cisse, G. Food-borne and water-borne diseases under climate change in low- and middle-income countries: Further efforts needed for reducing environmental health expo-sure risks. Acta Trop. 2019, 194, 181–188. [Google Scholar] [CrossRef]
  15. Kostyusheva, A.; Brezgin, S.; Babin, Y.; Vasilyeva, I.; Glebe, D.; Kostyushev, D.; Chulanov, V. CRISPR-Cas systems for diagnosing infectious diseases. Methods 2022, 203, 431–446. [Google Scholar] [CrossRef]
  16. Meiner, Z.; Marmor, A.; Jalagel, M.; Levine, H.; Shiri, S.; Schwartz, I. Risk factors for functional deterioration in a cohort with late effects of poliomyelitis: A ten-year follow-up study. NeuroRehabilitation 2021, 49, 491–499. [Google Scholar] [CrossRef]
  17. Pirrotta, P.; Tavares-Da-Silva, F.; Co, M.; Lecrenier, N.; Herve, C.; Stegmann, J.U. An analysis of spontaneously reported data of vesicular and bullous cutaneous eruptions occurring following vaccination with the adjuvanted recombinant zoster vaccine. Drug Saf. 2021, 44, 1341–1353. [Google Scholar] [CrossRef] [PubMed]
  18. Miceli, V.; Bulati, M.; Iannolo, G.; Zito, G.; Gallo, A.; Conaldi, G.P. Therapeutic properties of mesenchymal stromal/stem cells: The need of cell priming for cell-free therapies in regenerative medicine. Int. J. Mol. Sci. 2021, 22, 763. [Google Scholar] [CrossRef]
  19. Zakrzewski, W.; Dobrzyński, M.; Szymonowicz, M.; Rybak. Z. Stem cells: Past, present, and future. Stem Cell Res. Ther. 2019, 10, 68. [Google Scholar] [CrossRef]
  20. Galipeau, J.; Sensebe, L. Mesenchymal stromal cells: Clinical challenges and therapeutic opportunities. Cell Stem Cell 2018, 22, 824–833. [Google Scholar] [CrossRef]
  21. Squillaro, T.; Peluso, G.; Galderisi, U. Clinical trials with mesenchymal stem cells: An update. Cell Transplant. 2016, 25, 829–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Trounson, A.; McDonald, C. Stem cell therapies in clinical trials: Progress and challenges. Cell Stem Cell 2015, 17, 11–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Naji, A.; Rouas-Freiss, N.; Durrbach, A.; Carosella, E.D.; Sensébé, L.; Deschaseaux, F. Concise review: Combining human leukocyte antigen G and mesenchymal stem cells for immunosuppressant biotherapy. Stem Cells 2013, 31, 2296–2303. [Google Scholar] [CrossRef] [PubMed]
  24. Naji, A.; Suganuma, N.; Espagnolle, N.; Yagyu, K.; Baba, N.; Sensebe, L.; Deschaseaux, F. Rationale for determining the functional potency of mesenchymal stem cells in preventing regulated cell death for therapeutic use. Stem Cells Transl. Med. 2017, 6, 713–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Aleksandrushkina, N.A.; Danilova, N.V.; Grigorieva, O.A.; Mal’kov, P.G.; Popov, V.S.; Efimenko, A.Y.; Makarevich, P.I. Cell sheets of mesenchymal stromal cells effectively stimulate healing of deep soft tissue defects. Bull. Exp. Biol. Med. 2019, 167, 159–163. [Google Scholar] [CrossRef]
  26. Caplan, A.I. Mesenchymal stem cells. J. Orthop. Res. 1991, 9, 641–650. [Google Scholar] [CrossRef]
  27. Bianco, P. “Mesenchymal” stem cells. Annu. Rev. Cell Dev. Biol. 2014, 30, 677–704. [Google Scholar] [CrossRef]
  28. Caplan, A.I. Mesenchymal stem cells: Time to change the name! Stem Cells Transl. Med. 2017, 6, 1445–1451. [Google Scholar] [CrossRef] [Green Version]
  29. Bhartiya, D. The need to revisit the definition of mesenchymal and adult stem cells based on their functional attributes. Stem Cell Res. 2018, 9, 78. [Google Scholar] [CrossRef] [Green Version]
  30. Alvarez-Viejo, M.; Menendez-Menendez, Y.; Otero-Hernández, J. CD271 as a marker to identify mesenchymal stem cells from diverse sources be-fore culture. World J. Stem Cells 2015, 7, 470–476. [Google Scholar] [CrossRef]
  31. Lv, F.-J.; Tuan, R.S.; Cheung, K.M.C.; Leung, V.Y.L. Concise Review: The surface markers and identity of human mesenchymal stem cells. Stem Cells 2014, 32, 1408–1419. [Google Scholar] [CrossRef]
  32. Berebichez-Fridman, R.; Gomez-García, R.; Granados-Montiel, J.; Berebichez-Fastlicht, E.; Olivos-Meza, A.; Granados, J. The holy grail of orthopedic surgery: Mesenchymal stem cells—Their current uses and potential applications. Stem Cells Int. 2017, 2017, 2638305. [Google Scholar] [CrossRef] [Green Version]
  33. Hass, R.; Kasper, C.; Bohm, 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, 12. [Google Scholar] [CrossRef] [Green Version]
  34. Costela-Ruiz, V.J.; Melguizo-Rodriguez, L.; Bellotti, C.; Il-lescas-Montes, R.; Stanco, D.; Arciola, C.R.; Lucarelli, E. Different sources of mesenchymal stem cells for tissue regeneration: A guide to identifying the most favorable one in orthopedics and dentistry applications. Int. J. Mol. Sci. 2022, 23, 6356. [Google Scholar] [CrossRef]
  35. Mazini, L.; Rochette, L.; Amine, M.; Malka, G. Regenerative capacity of adipose-derived stem cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs). Int. J. Mol. Sci. 2019, 20, 2523. [Google Scholar] [CrossRef] [Green Version]
  36. Jin, H.J.; Bae, Y.K.; Kim, M.; Kwon, S.J.; Jeon, H.B.; Choi, S.J.; Kim, S.W.; Yang, Y.S.; Oh, W.; Chang, J.W. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int. J. Mol. Sci. 2013, 14, 17986–18001. [Google Scholar] [CrossRef] [Green Version]
  37. Arutyunyan, I.; Elchaninov, A.; Makarov, A.; Fatkhudinov, T. Umbilical cord as prospective source for mesenchymal stem cell-based therapy. Stem Cells Int. 2016, 2016, 6901286. [Google Scholar] [CrossRef] [Green Version]
  38. Selich, A.; Zimmermann, K.; Tenspolde, M.; Dittrich-Breiholz, O.; von Kaisenberg, C.; Schambach, A.; Rothe, M. Umbilical cord as a long-term source of activatable mesenchymal stromal cells for immunomodulation. Stem Cell Res. Ther. 2019, 1, 285. [Google Scholar] [CrossRef]
  39. Fukuchi, Y.; Nakajima, H.; Sugiyama, D.; Hirose, I.; Kitamura, T.; Tsuji, K. Human placenta-derived cells have mesenchymal stem/progenitor cell potential. Stem Cells 2004, 22, 649–658. [Google Scholar] [CrossRef] [Green Version]
  40. Thaweesapphithak, S.; Tantrawatpan, C.; Kheolamai, P.; Tantikanlayaporn, D.; Roytrakul, S.; Manochantr, S. Human serum enhances the proliferative capacity and immunomodulatory property of MSCs derived from human placenta and umbilical cord. Stem Cell Res. Ther. 2019, 10, 79. [Google Scholar] [CrossRef]
  41. Mastrolia, I.; Foppiani, E.M.; Murgia, A.; Candini, O.; Samarelli, A.V.; Grisendi, G.; Veronesi, E.; Horwitz, E.M.; Dominici, M. Challenges in clinical development of mesenchymal stromal/stem cells: Concise Review. Stem Cells Transl. Med. 2019, 8, 1135–1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Cheng, H.Y.; Ghetu, N.; Wallace, C.G.; Wei, F.C.; Liao, S.K. The impact of mesenchymal stem cell source on proliferation, differentiation, immunomodulation and therapeutic efficacy. J. Stem Cell Res. Ther. 2014, 4, 237. [Google Scholar] [CrossRef] [Green Version]
  43. Choudhery, M.S.; Badowski, M.; Muise, A.; Pierce, J.; Harris, D.T. Donor age negatively impacts adipose tissue-derived mesenchymal stem cell expansion and differentiation. J. Transl. Med. 2014, 12, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Cagliani, J.; Grande, D.; Molmenti, E.P.; Miller, E.J.; Rilo, H.L.R. Immunomodulation by mesenchymal stromal cells and their clinical applications. J. Stem Cell Regen. Biol. 2017, 3, 1–26. [Google Scholar] [CrossRef]
  45. Kim, J.H.; Chris, H.J.; Kim, H.R.; Hwang, Y.I. Comparison of immunological characteristics of mesenchymal stem cells from the periodontal ligament, umbilical cord, and adipose tissue. Stem Cells Int. 2018, 2018, 8429042. [Google Scholar] [CrossRef]
  46. In’t Anker, P.S.; Scherjon, S.A.; Kleijburg-van der Keur, C.; de Groot-Swings, G.M.; Claas, F.H.; Fibbe, W.E. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 2004, 22, 1338–1345. [Google Scholar] [CrossRef]
  47. Pipino, C.; Shangaris, P.; Resca, E.; Zia, S.; Deprest, J.; Sebire, N.J.; David, A.L.; Guillot, P.V.; De Coppiet, P. Placenta as a reservoir of stem cells: An underutilized resource? Br. Med. Bull. 2013, 105, 43–68. [Google Scholar] [CrossRef] [Green Version]
  48. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
  49. Mushahary, D.; Spittler, A.; Kasper, C.; Weber, V.; Charwat, V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry A 2018, 1, 19–31. [Google Scholar] [CrossRef] [Green Version]
  50. Nagamura-Inoue, T.; He, H. Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. World J. Stem Cells 2014, 6, 195–202. [Google Scholar] [CrossRef]
  51. Chen, Y.; Shao, J.Z.; Xiang, L.X.; Dong, X.J.; Zhang, G.R. Mesenchymal stem cells: A promising candidate in regenerative medicine. Int. J. Biochem. Cell Biol. 2008, 40, 815–820. [Google Scholar] [CrossRef]
  52. Nimiritsky, P.P.; Eremichev, R.Y.; Alexandrushkina, N.A.; Efimenko, A.Y.; Tkachuk, V.A.; Makarevich, P.I. Unveiling mesenchymal stromal cells’ organizing function in regeneration. Int. J. Mol. Sci. 2019, 20, 823. [Google Scholar] [CrossRef] [Green Version]
  53. Dai, L.-J.; Moniri, M.R.; Zeng, Z.-R.; Zhou, J.X.; Rayat, J.; Warnock, G.L. Potential implications of mesenchymal stem cells in cancer therapy. Cancer Lett. 2011, 305, 8–20. [Google Scholar] [CrossRef]
  54. Shah, K. Mesenchymal stem cells engineered for cancer therapy. Adv. Drug Deliv. Rev. 2012, 64, 739–748. [Google Scholar] [CrossRef] [Green Version]
  55. Yudintceva, N.; Lomert, E.; Mikhailova, N.; Tolkunova, E.; Agadzhanian, N.; Samochernych, K.; Multhoff, G.; Timin, G.; Ryzhov, V.; Deriglazov, V.; et al. Targeting brain tumors with mesenchymal stem cells in the experimental model of the orthotopic glioblastoma in rats. Biomedicines 2021, 9, 1592. [Google Scholar] [CrossRef]
  56. Shi, Y.; Wang, Y.; Li, Q.; Liu, K.; Hou, J.; Shao, C.; Wang, Y. Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases. Nat. Rev. Nephrol. 2018, 14, 493–507. [Google Scholar] [CrossRef]
  57. Jin, S.S.; He, D.Q.; Luo, D.; Wang, Y.; Yu, M.; Guan, B.; Fu, Y.; Li, Z.X.; Zhang, T.; Zhou, Y.H.; et al. A Biomimetic hierarchical nanointerface orchestrates macrophage polarization and mesenchymal stem cell recruitment to promote endogenous bone regeneration. ACS Nano 2019, 13, 6581–6595. [Google Scholar] [CrossRef]
  58. Zhang, Y.; Xu, J.; Liu, S.; Lim, M.; Zhao, S.; Cui, K.; Zhang, K.; Wang, L.; Ji, Q.; Han, Z.; et al. Embryonic stem cell-derived extracellular vesicles enhance the therapeutic effect of mesenchymal stem cells. Theranostics 2019, 9, 6976–6990. [Google Scholar] [CrossRef]
  59. Cao, X.; Duan, L.; Hou, H.; Liu, Y.; Chen, S.; Zhang, S.; Liu, Y.; Wang, C.; Qi, X.; Liu, N.; et al. IGF-1C hydrogel improves the therapeutic effects of MSCs on colitis in mice through PGE2-mediated M2macrophage polarization. Theranostics 2020, 10, 7697–7709. [Google Scholar] [CrossRef]
  60. Ceccariglia, S.; Cargnoni, A.; Silini, A.R.; Parolini, O. Autophagy: A potential key contributor to the therapeutic action of mesenchymal stem cells. Autophagy 2020, 1, 28–37. [Google Scholar] [CrossRef]
  61. Liu, Y.; Cui, J.; Wang, H.; Hezam, K.; Zhao, X.; Huang, H.; Chen, S.; Han, Z.; Han, Z.C.; Guo, Z.; et al. Enhanced therapeutic effects of MSC-derived extracellular vesicles with an injectable collagen matrix for experimental acute kidney injury treatment. Stem Cell Res. Ther. 2020, 11, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Tao, H.; Chen, X.; Cao, H.; Zheng, L.; Li, Q.; Zhang, K.; Han, Z.; Han, Z.C.; Guo, Z.; Li, Z.; et al. Mesenchymal stem cell-derived extracellular vesicles for corneal wound repair. Stem Cells Int. 2019, 2019, 5738510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Crivelli, B.; Chlapanidas, T.; Perteghella, S.; Lucarelli, E.; Pascucci, L.; Brini, A.T.; Ferrero, I.; Marazzi, M.; Pessina, A.; Torre, M.L. Mesenchymal stem/stromal cell extracellular vesicles: From active principle to next generation drug delivery system. J. Control. Release 2017, 262, 104–117. [Google Scholar] [CrossRef] [PubMed]
  64. Taghavi-Frahabadi, M.; Mahmoudi, M.; Soudi, S.; Hashemi, S.M. Hypothesis for the management and treatment of the COVID-19-induced acute respiratory distress syndrome and lung injury using mesenchymal stem cell—Derived exosomes. Med. Hypotheses 2020, 144, 109865. [Google Scholar] [CrossRef] [PubMed]
  65. Rahmati, S.; Shojaei, F.; Shojaeian, A.; Rezakhani, L.; Dehkordi, M.B. An overview of current knowledge in biological functions and potential theragnostic applications of exosomes. Chem. Phys. Lipids 2020, 226, 104836. [Google Scholar] [CrossRef]
  66. Lener, T.; Gimona, M.; Aigner, L.; Borger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; Del Portillo, H.A.; et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef] [Green Version]
  67. Jia, X.H.; Lu, H.; Li, C.; Feng, G.W.; Yao, X.P.; Mao, L.N.; Ke, T.Y.; Che, Y.Z.; Xu, Y.; Li, Z.J.; et al. Human embryonic stem cells-derived endothelial cell therapy facilitates kidney regeneration by stimulating renal resident stem cell proliferation in acute kidney injury. Chin. Sci. Bull. 2013, 58, 2820–2827. [Google Scholar] [CrossRef] [Green Version]
  68. Mardpour, S.; Hamidieh, A.A.; Taleahmad, S.; Sharifzad, F.; Taghikhani, A.; Baharvand, H. Interaction between mesenchymal stromal cell-derived extracellular vesicles and immune cells by distinct protein content. J. Cell Physiol. 2019, 234, 8249–8258. [Google Scholar] [CrossRef]
  69. Kim, H.; Lee, M.J.; Bae, E.H.; Ryu, J.S.; Kaur, G.; Kim, H.J.; Kim, J.Y.; Barreda, H.; Jung, S.Y.; Choi, J.M. Comprehensive Molecular Profiles of Functionally Effective MSC-Derived Extracellular Vesicles in Immunomodulation. Mol. Ther. 2020, 28, 1628–1644. [Google Scholar] [CrossRef]
  70. Franco da Cunha, F.; Andrade-Oliveira, V.; Candido de Almeida, D.; Borges da Silva, T.; Naffah de Souza Breda, C.; Costa Cruz, M. Extracellular vesicles isolated from mesenchymal stromal cells modulate CD4(+) T Lymphocytes toward a regulatory profile. Cells 2020, 9, 1059. [Google Scholar] [CrossRef]
  71. Shigemoto-Kuroda, T.; Oh, J.Y.; Kim, D.K.; Jeong, H.J.; Park, S.Y.; Lee, H.J.; Park, J.W.; Kim, T.W.; An, S.Y.; Prockop, D.J.; et al. MSC-derived extracellular vesicles attenuate immune responses in two autoimmune murine models: Type 1 diabetes and uveoretinitis. Stem Cell Rep. 2017, 8, 1214–1225. [Google Scholar] [CrossRef] [Green Version]
  72. Zhang, Z.; Huang, S.; Wu, S.; Qi, J.; Li, W.; Liu, S. Clearance of apoptotic cells by mesenchymal stem cells contributes to immunosuppression via PGE2. EBioMedicine 2019, 45, 341–350. [Google Scholar] [CrossRef] [Green Version]
  73. Wang, K.; Shi, Y.J.; Song, Z.L.; Wu, B.; Zhou, C.L.; Liu, W.; Gao, W. Regulatory effect of rat bone marrow mesenchymal stem cells on Treg/Th17 immune balance in vitro. Mol. Med. Rep. 2020, 21, 2123–2130. [Google Scholar] [CrossRef] [Green Version]
  74. Luo, S.; Ding, S.; Liao, J.; Zhang, P.; Liu, Y.; Zhao, M.; Zhao, M.; Lu, Q. Excessive miR-152-3p Results in Increased BAFF Expression in SLE B-Cells by Inhibiting the KLF5 Expression. Front. Immunol. 2019, 10, 1127. [Google Scholar] [CrossRef]
  75. Adamo, A.; Brandi, J.; Caligola, S.; Delfino, P.; Bazzoni, R.; Carusone, R.; Cecconi, D.; Giugno, R.; Manfredi, M.; Robotti, E.; et al. Extracellular vesicles mediate mesenchymal stromal cell-dependent regulation of B cell PI3K-AKT signaling pathway and actin cytoskeleton. Front. Immunol. 2019, 10, 446. [Google Scholar] [CrossRef]
  76. Yang, L.; Li, N.; Yang, D.; Chen, A.; Tang, J.; Jing, Y.; Kang, D.; Jiang, P.; Dai, X.; Luo, L.; et al. CCL2 Regulation of MST1-mTOR-STAT1 Signaling Axis Controls BCR Signaling and B-Cell Differentiation. Cell Death Differ. 2021, 28, 2616–2633. [Google Scholar] [CrossRef]
  77. Reis, M.; Mavin, E.; Nicholson, L.; Green, K.; Dickinson, A.M.; Wang, X.N. Mesenchymal stromal cell-derived extracellular vesicles attenuate dendritic cell maturation and function. Front. Immunol. 2018, 9, 2538. [Google Scholar] [CrossRef] [Green Version]
  78. Hu, C.D.; Kosaka, Y.; Marcus, P.; Rashedi, I.; Keating, A. Differential immunomodulatory effects of human bone marrow-derived mesenchymal stromal cells on natural killer cells. Stem Cells Dev. 2019, 28, 933–943. [Google Scholar] [CrossRef]
  79. Najar, M.; Fayyad-Kazan, M.; Meuleman, N.; Bron, D.; Fayyad-Kazan, H.; Lagneaux, L. Mesenchymal stromal cells of the bone marrow and natural killer cells: Cell interactions and cross modulation. J. Cell Commun. Signal. 2018, 12, 673–688. [Google Scholar] [CrossRef]
  80. Selleri, S.; Bifsha, P.; Civini, S.; Pacelli, C.; Dieng, M.M.; Lemieux, W.; Jin, P.; Bazin, R.; Patey, N.; Marincolet, F.M.; et al. Human Mesenchymal Stromal Cell-Secreted Lactate Induces M2-Macrophage Differentiation by Metabolic Reprogramming. Oncotarget 2016, 7, 30193–30210. [Google Scholar] [CrossRef]
  81. Hyvarinen, K.; Holopainen, M.; Skirdenko, V.; Ruhanen, H.; Lehenkari, P.; Korhonen, M. Mesenchymal stromal cells and their extracellular vesicles enhance the anti-inflammatory phenotype of regulatory macrophages by downregulating the production of Interleukin (IL)-23 and IL-22. Front. Immunol. 2018, 9, 771. [Google Scholar] [CrossRef] [PubMed]
  82. Lo Sicco, C.; Reverberi, D.; Balbi, C.; Ulivi, V.; Principi, E.; Pascucci, L. Mesenchymal stem cell-derived extracellular vesicles as mediators of anti-inflammatory effects: Endorsement of macrophage polarization. Stem Cells Transl. Med. 2017, 6, 1018–1028. [Google Scholar] [CrossRef] [PubMed]
  83. He, X.; Dong, Z.; Cao, Y.; Wang, H.; Liu, S.; Liao, L.; Jin, Y.; Yuan, L.; Li, B. MSC-derived exosome promotes M2 polarization and enhances cutaneous wound healing. Stem Cells Int. 2019, 2019, 7132708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Xin, D.; Li, T.; Chu, X.; Ke, H.; Yu, Z.; Cao, L.; Wang, Z. Mesenchymal stromal cell-derived extracellular vesicles modulate microglia/macrophage polarization and protect the brain against hypoxia-ischemic injury in neonatal mice by targeting delivery of miR-21a-5p. Acta Biomater. 2020, 113, 597–613. [Google Scholar] [CrossRef] [PubMed]
  85. Chen, X.; Wei, Q.; Sun, H.; Zhang, X.; Yang, C.; Tao, Y.; Nong, G. Exosomes derived from human umbilical cord mesenchymal stem cells regulate macrophage polarization to attenuate systemic lupus erythematosus-associated diffuse alveolar hemorrhage in mice. Int. J. Stem Cells 2021, 14, 331–340. [Google Scholar] [CrossRef]
  86. Costa, L.A.; Eiro, N.; Fraile, M.; Gonzalez, L.O.; Saa, J.; Garcia-Portabella, P.; Vega, B.; Schneider, J.; Vizoso, F.J. Functional heterogeneity of mesenchymal stem cells from natural niches to culture conditions: Implications for further clinical uses. Cell Mol. Life Sci. 2021, 78, 447–467. [Google Scholar] [CrossRef]
  87. Levy, O.; Kuai, R.; Siren, E.M.J.; Bhere, D.; Milton, Y.; Nissar, N.; De Biasio, M.; Heinelt, M.; Reeve, B.; Abdi, R.; et al. Shattering barriers toward clinically meaningful MSC therapies. Sci. Adv. 2020, 6, eaba6884. [Google Scholar] [CrossRef]
  88. Brown, C.; McKee, C.; Bakshi, S.; Walker, K.; Hakman, E.; Halassy, S.; Svinarich, D.; Dodds, R.; Govind, C.K.; Chaudhry, G.R. Mesenchymal stem cells: Cell therapy and regeneration potential. J. Tissue Eng. Regen. Med. 2019, 13, 1738–1755. [Google Scholar] [CrossRef]
  89. Tavakoli, S.; Ghaderi Jafarbeigloo, H.R.; Shariati, A.; Jahangiryan, A.; Jadidi, F.; Jadidi Kouhbanani, M.A.; Hassanzadeh, A.; Zamani, M.; Javidi, K.; Naimi, A. Mesenchymal stromal cells; a new horizon in regenerative medicine. J. Cell Physiol. 2020, 235, 9185–9210. [Google Scholar] [CrossRef]
  90. Tang, Y.; Zhou, Y.; Li, H.J. Advances in mesenchymal stem cell exosomes: A review. Stem Cell Res. Ther. 2021, 12, 71. [Google Scholar] [CrossRef]
  91. He, C.; Zheng, S.; Luo, Y.; Wang, B. Exosome Theranostics: Biology and Translational Medicine. Theranostics. 2018, 8, 237–255. [Google Scholar] [CrossRef]
  92. Wu, J.; Kuang, L.; Chen, C.; Yang, J.; Zeng, W.N.; Li, T.; Chen, H.; Huang, S.; Fu, Z.; Li, J.; et al. miR-100−5p-Abundant exosomes derived from infrapatellar fat pad MSCs protect articular cartilage and ameliorate gait abnormalities via inhibition of mTOR in osteoarthritis. Biomaterials 2019, 206, 87–100. [Google Scholar] [CrossRef]
  93. Zhang, S.; Teo, K.Y.W.; Chuah, S.J.; Lai, R.C.; Lim, S.; Toh, W.S. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials 2019, 200, 35–47. [Google Scholar] [CrossRef]
  94. Liao, Z.; Luo, R.; Li, G.; Song, Y.; Zhan, S.; Zhao, K.; Hua, W.; Zhang, Y.; Wu, X.; Yang, C. Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo. Theranostics 2019, 9, 4084–4100. [Google Scholar] [CrossRef]
  95. Mihajlovic, M.; Wever, K.E.; Van der Made, T.K.; de Vries, R.B.M.; Hilbrands, L.B.; Masereeuw, R. Are cell-based therapies for kidney disease safe? A systematic review of preclinical evidence. Pharmacol. Ther. 2019, 197, 191–211. [Google Scholar] [CrossRef]
  96. Lai, P.; Weng, J.; Guo, L.; Chen, X.; Du, X. Novel insights into MSC-EVs therapy for immune diseases. Biomark Res. 2019, 7, 6. [Google Scholar] [CrossRef] [Green Version]
  97. Gould, S.J.; Raposo, G. As we wait: Coping with an imperfect nomenclature for extracellular vesicles. J. Extracell. Vesicles 2013, 2, 2892. [Google Scholar] [CrossRef]
  98. Yuana, Y.; Sturk, A.; Nieuwland, R. Extracellular vesicles in physiological and pathological conditions. Blood Rev. 2013, 27, 31–39. [Google Scholar] [CrossRef] [Green Version]
  99. Ludwig, A.-K.; Giebel, B. Exosomes: Small vesicles participating in intercellular communication. Int. J. Biochem. Cell Boil. 2012, 44, 11–15. [Google Scholar] [CrossRef]
  100. Yanez-Mo, M.; Siljander, P.; Andreu, Z.; Zavec, A.B.; Borras, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef]
  101. Yates, A.G.; Pink, R.C.; Erdbrügger, U.; Siljander, P.R.; Dellar, E.R.; Pantazi, P.; Akbar, N.; Cooke, W.R.; Vatish, M.; Dias-Neto, E.; et al. In sickness and in health: The functional role of extracellular vesicles in physiology and pathology in vivo: Part I: Health and Normal Physiology: Part I: Health and Normal Physiology. J. Extracell. Vesicles 2022, 11, e12151. [Google Scholar] [CrossRef] [PubMed]
  102. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yates, A.G.; Pink, R.C.; Erdbrügger, U.; Siljander, P.R.; Dellar, E.R.; Pantazi, P.; Akbar, N.; Cooke, W.R.; Vatish, M.; Dias-Neto, E.; et al. In sickness and in health: The functional role of extracellular vesicles in physiology and pathology in vivo: Part II: Pathology: Part II: Pathology. J. Extracell. Vesicles 2022, 11, e12190. [Google Scholar] [CrossRef] [PubMed]
  104. Lobov, A.A.; Yudintceva, N.M.; Mittenberg, A.G.; Shabelnikov, S.V.; Mikhailova, N.A.; Malashicheva, A.B.; Khotin, M.G. Proteomic profiling of the human fetal multipotent mesenchymal stromal cells secretome. Molecules 2020, 25, 5283. [Google Scholar] [CrossRef] [PubMed]
  105. Rahbarghazi, R.; Jabbari, N.; Sani, N.A.; Asghari, R.; Salimi, L.; Kalashani, S.A. Tumor-derived extracellular vesicles: Reliable tools for Cancer diagnosis and clinical applications. Cell Commun. Signal. 2019, 17, 73. [Google Scholar] [CrossRef] [Green Version]
  106. Gonda, A.; Kabagwira, J.; Senthil, G.N.; Wall, N.R. Internalization of Exosomes through Receptor-Mediated Endocytosis. Mol. Cancer Res. 2019, 2, 337–347. [Google Scholar] [CrossRef] [Green Version]
  107. Cheng, H.; Chang, S.; Xu, R.; Chen, L.; Song, X.; Wu, J.; Qian, J.; Zou, Y.; Ma, J. Hypoxia-challenged MSC-derived exosomes deliver miR-210 to attenuate post-infarction cardiac apoptosis. Stem Cell Res. Ther. 2020, 11, 224. [Google Scholar] [CrossRef]
  108. Feng, J.; Wang, W. Hypoxia pretreatment and EPO-modification enhance the protective effects of MSC on neuron-like PC12 cells in a similar way. Biochem. Biophys. Res. Commun. 2017, 482, 232–238. [Google Scholar] [CrossRef]
  109. Carter, K.; Lee, H.J.; Na, K.S.; Fernandes-Cunha, G.M.; Blanco, I.J.; Djalilian, A.; Myung, D. Characterizing the impact of 2D and 3D culture conditions on the therapeutic effects of human mesenchymal stem cell secretome on corneal wound healing in vitro and ex vivo. Acta Biomater. 2019, 99, 247–257. [Google Scholar] [CrossRef]
  110. Fernandes-Cunha, G.M.; Na, K.S.; Putra, I.; Lee, H.; Hull, S.; Cheng, Y.C.; Blanco, I.J.; Eslani, M.; Djalilian, A.R.; Myung, D. Corneal wound healing effects of mesenchymal stem cell secretome delivered within a viscoelastic gel carrier. Stem Cells Transl. Med. 2019, 8, 478–489. [Google Scholar] [CrossRef]
  111. Kaminska, A.; Wedzinska, A.; Kot, M.; Sarnowska, A. Effect of long-term 3D spheroid culture on WJ-MSC. Cells 2021, 10, 719. [Google Scholar] [CrossRef]
  112. Kanada, M.; Ashammakhi, N. Discussion of the role of extracellular vesicles secreted from thermal stress-induced adipose-derived stem cells on bone regeneration. J. Craniofac. Surg. 2021, 32, 2251. [Google Scholar] [CrossRef]
  113. Jafari, D.; Shajari, S.; Jafari, R.; Mardi, N.; Gomari, H.; Ganji, F.; Moghadam, M.F.; Samadikuchaksaraei, A. Designer Exosomes: A new platform for biotechnology therapeutics. BioDrugs 2020, 34, 567–586. [Google Scholar] [CrossRef]
  114. Jiang, Y.; Zhang, P.; Zhang, X.; Lv, L.; Zhou, Y. Advances in mesenchymal stem cell transplantation for the treatment of osteoporosis. Cell Prolif. 2021, 54, e12956. [Google Scholar] [CrossRef]
  115. Xu, L.; Huang, S.; Hou, Y.; Liu, Y.; Ni, M.; Meng, F.; Wang, K.; Rui, Y.; Jiang, X.; Li, G. Sox11-modified mesenchymal stem cells (MSCs) accelerate bone fracture healing: Sox11 regulates differentiation and migration of MSCs. FASEB J. 2015, 29, 1143–1152. [Google Scholar] [CrossRef] [Green Version]
  116. Garcia-Sanchez, D.; Fernandez, D.; Rodríguez-Rey, J.C.; Perez-Campo, F.M. Enhancing survival, engraftment, and osteogenic potential of mesenchymal stem cells. World J. Stem Cells 2019, 11, 748–763. [Google Scholar] [CrossRef]
  117. Song, H.; Song, B.-W.; Cha, M.-J.; Choi, I.-G.; Hwang, K.-C. Modification of mesenchymal stem cells for cardiac regeneration. Expert Opin. Biol. Ther. 2010, 10, 309–319. [Google Scholar] [CrossRef]
  118. Ocansey, D.K.W.; Pei, B.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; Mao, F. Improved therapeutics of modified mesenchymal stem cells: An update. J. Transl. Med. 2020, 18, 42. [Google Scholar] [CrossRef] [Green Version]
  119. Wang, H.; Wang, X.; Qu, J.; Yue, Q.; Hu, Y.; Zhang, H. VEGF enhances the migration of MSCs in neural differentiation by regulating focal adhesion turnover. J. Cell Physiol. 2015, 230, 2728–2742. [Google Scholar] [CrossRef]
  120. De Becker, A.; Van Riet, I. Homing and migration of mesenchymal stromal cells: How to improve the efficacy of cell therapy? World J. Stem Cells 2016, 8, 73–87. [Google Scholar] [CrossRef]
  121. Nolta, J.A.; Galipeau, J.; Phinney, D.G. Improving mesenchymal stem/stromal cell potency and survival: Proceedings from the International Society of Cell Therapy (ISCT) MSC preconference held in May 2018, Palais des Congrès de Montréal, Organized by the ISCT MSC Scientific Committee. Cytotherapy 2020, 22, 123–126. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, L.; Xu, Y.; Zhao, J.; Zhang, Z.; Yang, R.; Xie, J.; Liu, X.; Qi, S. Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice. PLoS ONE 2014, 9, e96161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Van der Pol, E.; Boing, A.N.; Harrison, P.; Sturk, A.; Nieuwland, R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol. Rev. 2012, 64, 676–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Hu, L.; Wang, J.; Zhou, X.; Xiong, Z.; Zhao, J.; Yu, R.; Huang, F.; Zhang, H.; Chen, L. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 2016, 6, 32993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Borges, F.T.; Reis, L.A.; Schor, N. Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases. Braz. J. Med. Biol. Res. 2013, 46, 824–830. [Google Scholar] [CrossRef] [Green Version]
  126. Facklam, A.L.; Volpatti, L.R.; Anderson, D.G. Biomaterials for personalized cell therapy. Adv. Mater. 2020, 32, 1902005. [Google Scholar] [CrossRef]
  127. Qazi, T.H.; Mooney, D.J.; Duda, G.N.; Geissler, S. Biomaterials that promote cell–cell interactions enhance the paracrine function of MSCs. Biomaterials 2017, 140, 103–114. [Google Scholar] [CrossRef]
  128. Dergilev, K.V.; Makarevich, P.I.; Tsokolaeva, Z.I.; Bodyrva, M.A.; Beloglazova, I.B.; Zubkova, E.S.; Menshikov, M.Y.; Parfyonova, Y.V. Comparison of cardiac stem cell sheets detached by Versen solution and from termoresponsive dishes revealssimilar properties of constructs. Tissue Cell 2017, 49, 64–71. [Google Scholar] [CrossRef]
  129. Kim, K.; Bou-Ghannam, S.; Kameishi, S.; Oka, M.; Grainger, D.W.; Okano, T. Allogeneic mesenchymal stem cell sheet therapy: A new frontier in drug delivery systems. J. Control. Release 2021, 330, 696–704. [Google Scholar] [CrossRef]
  130. Chang, D.; Fan, T.; Gao, S.; Jin, Y.; Zhang, M.; Ono, M. Application of mesenchymal stem cell sheet to treatment of ischemic heart disease. Stem Cell Res. Ther. 2021, 12, 384. [Google Scholar] [CrossRef]
  131. Du, W.; Zhang, K.Y.; Zhang, S.Q.; Wang, R.; Nie, Y.; Tao, H.Y.; Han, Z.B.; Liang, L.; Wang, D.; Liu, J.F.; et al. Enhanced proangiogenic potential of mesenchymal stem cell-derived exosomes stimulated by a nitric oxide releasing polymer. Biomaterials 2017, 133, 70–81. [Google Scholar] [CrossRef]
  132. Zhu, L.; Kalimuthu, S.; Oh, J.M.; Gangadaran, P.; Baek, S.H.; Jeong, S.Y.; Lee, S.-W.; Lee, J.; Ahn, B.-C. Enhancement of Antitumor Potency of Extracellular Vesicles Derived from Natural Killer Cells by IL-15 Priming. Biomaterials 2019, 190, 38–50. [Google Scholar] [CrossRef]
  133. Hu, C.; Li, L. Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J. Cell Mol. Med. 2018, 3, 1428–1442. [Google Scholar] [CrossRef] [Green Version]
  134. Phillips, M.I.; Tang, Y.L. Genetic modification of stem cells for transplantation. Adv. Drug Deliv. Rev. 2008, 60, 160–172. [Google Scholar] [CrossRef] [Green Version]
  135. Olson, S.D.; Kambal, A.; Pollock, K.; Mitchell, G.M.; Stewart, H.; Kalomoiris, S.; Cary, W.; Nacey, C.; Pepper, K.; Nolta, J.A. Examination of mesenchymal stem cell-mediated RNAi transfer to Huntington’s disease affected neuronal cells for reduction of Huntingtin. Mol. Cell. Neurosci. 2012, 49, 271–281. [Google Scholar] [CrossRef] [Green Version]
  136. Armstrong, J.P.; Holme, M.N.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS Nano 2017, 11, 69–83. [Google Scholar] [CrossRef] [Green Version]
  137. Gupta, D.; Zickler, A.M.; Andaloussi, S.E. Dosing extracellular vesicles. Adv. Drug Deliv. Rev. 2021, 178, 113961. [Google Scholar] [CrossRef]
  138. Villata, S.; Canta, M.; Cauda, V. EVs and Bioengineering: From cellular products to engineered nanomachines. Int. J. Mol. Sci. 2020, 21, 6048. [Google Scholar] [CrossRef]
  139. Mishra, A.; Singh, P.; Qayoom, I.; Prasad, A.; Kumar, A. Current strategies in tailoring methods for engineered exosomes and future avenues in biomedical applications. J. Mater. Chem. B 2021, 9, 6281–6309. [Google Scholar] [CrossRef]
  140. Wang, H.; Cui, J.; Zheng, Z.; Shi, Q.; Sun, T.; Liu, X.; Huang, Q.; Fukuda, T. Assembly of RGD-modified hydrogel micromodules into permeable three-dimensional hollow microtissues mimicking in vivo tissue structures. ACS Appl. Mater. Interfaces 2017, 9, 41669–41679. [Google Scholar] [CrossRef]
  141. Zheng, W.; Wang, Z.; Song, L.; Zhao, Q.; Zhang, J.; Li, D.; Wang, S.; Han, J.; Zheng, X.L.; Yang, Z.; et al. Endothelialization and patency of RGD-functionalized vascular grafts in a rabbit carotid artery model. Biomaterials 2012, 33, 2880–2891. [Google Scholar] [CrossRef] [PubMed]
  142. Wang, J.; Li, W.; Lu, Z.; Zhang, L.; Hu, Y.; Li, Q.; Du, W.; Feng, X.; Jia, H.; Liu, B.F. The use of RGD-engineered exosomes for enhanced targeting ability and synergistic therapy toward angiogenesis. Nanoscale 2017, 9, 15598–15605. [Google Scholar] [CrossRef] [PubMed]
  143. Han, C.; Zhou, J.; Liang, C.; Liu, B.; Pan, X.; Zhang, Y.; Wang, Y.; Yan, B.; Xie, W.; Liu, F.; et al. Human umbilical cord mesenchymal stem cell derived exosomes encapsulated in functional peptide hydrogels promote cardiac repair. Biomater. Sci. 2019, 7, 2920–2933. [Google Scholar] [CrossRef] [PubMed]
  144. Wang, C.; Wang, M.; Xu, T.; Zhang, X.; Lin, C.; Gao, W.; Xu, H.; Lei, B.; Mao, C. Engineering bioactive self-healing antibacterial exosomes hydrogel for promoting chronic diabetic wound healing and complete skin regeneration. Theranostics 2019, 9, 65–76. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, C.; Shang, Y.; Chen, X.; Midgley, A.C.; Wang, Z.; Zhu, D.; Wu, J.; Chen, P.; Wu, L.; Wang, X.; et al. Supramolecular nanofibers containing Arginine-Glycine-Aspartate (RGD) peptides boost therapeutic efficacy of extracellular vesicles in kidney repair. ACS Nano 2020, 14, 12133–12147. [Google Scholar] [CrossRef]
  146. Mardpour, S.; Ghanian, M.H.; Sadeghi-Abandansari, H.; Mardpour, S.; Nazari, A.; Shekari, F.; Baharvand, H. Hydrogel-mediated sustained systemic delivery of mesenchymal stem cell-derived extracellular vesicles improves hepatic regeneration in chronic liver failure. ACS Appl. Mater. Interfaces 2019, 11, 37421–37433. [Google Scholar] [CrossRef]
  147. Liu, X.; Yang, Y.; Li, Y.; Niu, X.; Zhao, B.; Wang, Y.; Bao, C.; Xie, Z.; Lin, Q.; Zhu, L. Integration of stem cell-derived exosomes with in situ hydrogel glue as a promising tissue patch for articular cartilage regeneration. Nanoscale 2017, 9, 4430–4438. [Google Scholar] [CrossRef]
  148. Aryani, A.; Denecke, B. Exosomes as a nanodelivery system: A key to the future of neuromedicine? Mol. Neurobiol. 2016, 53, 818–834. [Google Scholar] [CrossRef] [Green Version]
  149. Malekian, F.; Shamsian, A.; Kodam, S.P.; Ullah, M. Exosome engineering for efficient and targeted drug delivery: Current status and future perspective. J. Physiol. 2022, 16, 1–20. [Google Scholar] [CrossRef]
  150. Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.; Yuan, H. Engineering exosomes as refined biological nanoplatforms for drug delivery Acta Pharmacol. Sin. 2017, 38, 754–763. [Google Scholar] [CrossRef]
  151. 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] [Green Version]
  152. Ghannam, S.; Pene, J.; Moquet-Torcy, G.; Jorgensen, C.; Yssel, H. Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J. Immunol. 2010, 185, 302–312. [Google Scholar] [CrossRef] [Green Version]
  153. Spaggiari, G.M.; Capobianco, A.; Abdelrazik, H.; Becchetti, F.; Mingari, M.C.; Moretta, L. Mesenchymal stem cells inhibit natural killer–cell proliferation, cytotoxicity, and cytokine production: Role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 2008, 111, 1327–1333. [Google Scholar] [CrossRef]
  154. Liras, A. Future research and therapeutic applications of human stem cells: General, regulatory, and bioethical aspects. J. Transl. Med. 2010, 8, 131. [Google Scholar] [CrossRef] [Green Version]
  155. Melief, S.M.; Geutskens, S.B.; Fibbe, W.E.; Roelofs, H. Multipotent stromal cells skew monocytes towards an anti-inflammatory interleukin-10-producing phenotype by production of interleukin-6. Haematologica 2013, 98, 888–895. [Google Scholar] [CrossRef] [Green Version]
  156. Franquesa, M.; Mensah, F.K.; Huizinga, R.; Strini, T.; Boon, L.; Lombardo, E.; DelaRosa, O.; Laman, J.D.; Grinyó, J.M.; Weimar, W.; et al. Human adipose tissue-derived mesenchymal stem cells abrogate plasmablast formation and induce regulatory B cells independently of T helper cells. Stem Cells 2015, 33, 880–891. [Google Scholar] [CrossRef] [Green Version]
  157. Tabera, S.; Perez-Simón, J.A.; Diez-Campelo, M.; Sanchez-Abarca, L.I.; Blanco, B.; Lopez, A.; Benito, A.; Ocio, E.; Sanchez-Guijo, F.M.; Canizo, C.; et al. The effect of mesenchymal stem cells on the viability, proliferation and differentiation of B-lymphocytes. Haematologica 2008, 93, 1301–1309. [Google Scholar] [CrossRef]
  158. Jiang, D.; Muschhammer, J.; Qi, Y.; Kügler, A.; de Vries, J.C.; Saffarzadeh, M.; Sindrilaru, A.; Beken, S.V.; Wlaschek, M.; Kluth, M.A.; et al. Suppression of neutrophil-mediated tissue damage—A novel skill of mesenchymal stem cells. Stem Cells 2016, 3, 2393–2406. [Google Scholar] [CrossRef]
  159. Aly, R.M. Current state of stem cell-based therapies: An overview. Stem Cell Investig. 2020, 7, 8. [Google Scholar] [CrossRef]
  160. Chari, S.; Nguyen, A.; Saxe, J. Stem cells in the clinic. Cell Stem Cell 2018, 22, 781–782. [Google Scholar] [CrossRef]
  161. Volarevic, V.; Markovic, B.S.; Gazdic, M.; Volarevic, A.; Jovicic, N.; Arsenijevic, N.; Armstrong, L.; Djonov, V.; Lako, M.; Stojkovic, M. Ethical and safety issues of stem cell-based therapy. Int. J. Med. Sci. 2018, 15, 36–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Wang, L.T.; Liu, K.J.; Sytwu, H.K.; Yen, M.L.; Yen, B.L. Advances in mesenchymal stem cell therapy for immune and inflammatory diseases: Use of cell-free products and human pluripotent stem cell-derived mesenchymal stem cells. Stem Cells Transl. Med. 2021, 10, 1288–1303. [Google Scholar] [CrossRef] [PubMed]
  163. Metcalfe, S.M. Mesenchymal stem cells and management of COVID-19 pneumonia. Med. Drug Discov. 2020, 5, 100019. [Google Scholar] [CrossRef] [PubMed]
  164. Gentile, P.; Sterodimas, A. Adipose-derived stromal stem cells (ASCs) as a new regenerative immediate therapy combating coronavirus (COVID-19)-induced pneumonia. Expert Opin. Biol. Ther. 2020, 20, 711–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Leng, Z.; Zhu, R.; Hou, W.; Feng, Y.; Yang, Y.; Han, Q.; Zhao, R.C. Transplantation of ACE2-mesenchymal stem cells improves the outcome of patients with COVID-19 pneumonia. Aging Dis. 2020, 11, 216. [Google Scholar] [CrossRef] [Green Version]
  166. Golchin, A.; Seyedjafari, E.; Ardeshirylajimi, A. Mesenchymal stem cell therapy for COVID-19: Present or future. Stem Cell Rev. Rep. 2020, 16, 427–433. [Google Scholar] [CrossRef] [Green Version]
  167. Ji, F.; Li, L.; Li, Z.; Jin, Y.; Liu, W. Mesenchymal stem cells as a potential treatment for critically ill patients with coronavirus disease Stem Cells Transl. Med. 2020, 9, 813–814. [Google Scholar] [CrossRef] [Green Version]
  168. Zhao, L.; Hu, C.; Zhang, P.; Jiang, H.; Chen, J. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J. Cell Mol. Med. 2019, 23, 720–730. [Google Scholar] [CrossRef] [Green Version]
  169. Shu, L.; Niu, C.; Li, R.; Huang, T.; Wang, Y.; Huang, M.; Ji, N.; Zheng, Y.; Chen, X.; Shi, L.; et al. Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cells. Stem Cell Res. Ther. 2020, 11, 361. [Google Scholar] [CrossRef]
  170. Zhang, K.; Chen, S.; Sun, H.; Wang, L.; Li, H.; Zhao, J.; Zhang, C.; Li, N.; Guo, Z.; Han, Z.; et al. In vivo two-photon microscopy reveals the contribution of Sox9+ cell to kidney regeneration in a mouse model with extracellular vesicle treatment. J. Biol. Chem. 2020, 295, 12203–12213. [Google Scholar] [CrossRef]
  171. Eirin, A.; Zhu, X.Y.; Puranik, A.S.; Tang, H.; McGurren, K.A.; van Wijnen, A.J.; Lerman, A.; Lerman, L.O. Mesenchymal stem cell-derived extracellular vesicles attenuate kidney inflammation. Kidney Int. 2017, 92, 114–124. [Google Scholar] [CrossRef]
  172. Vader, P.; Mol, E.A.; Pasterkamp, G.; Schiffelers, R.M. Extracellular vesicles for drug delivery. Adv. Drug Deliv. Rev. 2016, 106, 148–156. [Google Scholar] [CrossRef]
  173. Gupta, P.S.; Krishnakumar, V.; Sharma, Y.; Dinda, A.K.; Mohanty, S. Mesenchymal stem cell derived exosomes: A nano platform for therapeutics and drug delivery in combating COVID-19. Stem Cell Rev. Rep. 2020, 1, 33–43. [Google Scholar] [CrossRef]
  174. Baker, S.C. Coronaviruses: From common colds to severe acute respiratory syndrome. Pediatr. Infect. Dis. J. 2004, 23, 1049–1050. [Google Scholar] [CrossRef]
  175. Sloots, T.P.; Whiley, D.M.; Lambert, S.B.; Nissena, M.D. Emerging respiratory agents: New viruses for old diseases? J. Clin. Virol. 2008, 42, 233–243. [Google Scholar] [CrossRef]
  176. Khatri, M.; Richardson, L.A.; Meulia, T. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model. Stem Cell Res. Ther. 2018, 9, 17. [Google Scholar] [CrossRef] [Green Version]
  177. Du, J.; Li, H.; Lian, J.; Zhu, X.; Qiao, L.; Lin, J. Stem cell therapy: A potential approach for treatment of influenza virus and coronavirus-induced acute lung injury. Stem Cell Res. Ther. 2020, 11, 192. [Google Scholar] [CrossRef]
  178. Chan, M.C.; Kuok, D.I.; Leung, C.Y.; Hui, K.P.; Valkenburg, S.A.; Lau, E.H.; Peiris, J.M. Human mesenchymal stromal cells reduce influenza A H5N1-associated acute lung injury in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2016, 113, 3621–3626. [Google Scholar] [CrossRef] [Green Version]
  179. Li, Y.; Xu, J.; Shi, W.; Chen, C.; Shao, Y.; Zhu, L.; Lu, W.; Hanet, X. Mesenchymal stromal cell treatment prevents H9N2 avian influenza virus-induced acute lung injury in mice. Stem Cell Res. Ther. 2016, 7, 159. [Google Scholar] [CrossRef] [Green Version]
  180. Loy, H.; Kuok, D.I.T.; Hui, K.P.Y.; Choi, M.H.L.; Yuen, W.; Nicholls, J.M.; Peiris, J.S.M.; Chan, M.C.W. Therapeutic implications of human umbilical cord mesenchymal stromal cells in attenuating influenza A (H5N1) virusassociated acute lung injury. J. Infect. Dis. 2019, 219, 186–196. [Google Scholar] [CrossRef]
  181. Song, N.; Scholtemeijer, M.; Shah, K. Mesenchymal stem cell immunomodulation: Mechanisms and therapeutic potential. Trends Pharmacol. Sci. 2020, 41, 653–664. [Google Scholar] [CrossRef] [PubMed]
  182. Suzdaltseva, Y.; Goryunov, K.; Silina, E.; Manturova, N.; Stupin, V.; Kiselev, S.L. Equilibrium among inflammatory factors determines human MSC-mediated immunosuppressive effect. Cells 2022, 11, 1210. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, J.; Hu, C.; Chen, L.; Tang, L.; Zhu, Y.; Xu, X.; Chen, L.; Gao, H.; Lu, X.; Yu, L.; et al. Clinical study of mesenchymal stem cell treatment for acute respiratory distress syndrome induced by epidemic influenza A (H7N9) infection: A hint for COVID-19 treatment. Engineering 2020, 6, 1153–1161. [Google Scholar] [CrossRef] [PubMed]
  184. Gotts, J.E.; Abbott, J.; Matthay, M.A. Influenza causes prolonged disruption of the alveolar-capillary barrier in mice unresponsive to mesenchymal stem cell therapy. Am. J. Physiol. Lung Cell Mol. Physiol. 2014, 307, L395–L406. [Google Scholar] [CrossRef] [PubMed]
  185. Darwish, I.; Banner, D.; Mubareka, S.; Kim, H.; Besla, R.; Kelvin, D.J.; Kain, K.C.; Liles, W.C. Mesenchymal stromal (stem) cell therapy fails to improve outcomes in experimental severe influenza. PLoS ONE 2013, 8, e71761. [Google Scholar] [CrossRef] [Green Version]
  186. Taylor, B.S.; Sobieszczyk, M.E.; McCutchan, F.E.; Hammer, S.M. The challenge of HIV-1 subtype diversity. N. Engl. J. Med. 2008, 358, 1590–1602. [Google Scholar] [CrossRef] [Green Version]
  187. Cohen, J. Has a second person with HIV been cured? Sci. Mag. 2019, 363, 1021. [Google Scholar] [CrossRef]
  188. Cohen, J. An intriguing—But far from proven—HIV cure in the ‘São Paulo Patient’. Sci. Mag. 2020. [Google Scholar] [CrossRef]
  189. Kitchen, S.G.; Zack, J.A. Stem cell-based approaches to treating HIV infection. Curr. Opin. HIV AIDS 2011, 6, 68–73. [Google Scholar] [CrossRef]
  190. Kandula, U.R.; Wake, A. Promising stem cell therapy in the management of HIV and AIDS: A narrative review. Biol. Targets Ther. 2022, 16, 89–105. [Google Scholar] [CrossRef]
  191. Khalid, K.; Padda, J.; Fernando, R.W.; Mehta, K.A.; Almanie, A.H.; Hennawi, H.A.; Padda, S.; Cooper, A.C.; Jean-Charles, G. Stem cell therapy and its significance in HIV infection. Cureus 2021, 13, e17507. [Google Scholar] [CrossRef]
  192. Allers, K.; Hutter, G.; Hofmann, J.; Loddenkemper, C.; Rieger, K.; Thiel, E.; Schneider, T. Evidence for the cure of HIV infection by CCR5Δ32/Δ32 stem cell transplantation. Blood 2011, 117, 2791–2799. [Google Scholar] [CrossRef]
  193. Fackler, O.T.; Murooka, T.T.; Imle, A.; Mempel, T.R. Adding new dimensions: Towards an integrative understanding of HIV-1 spread. Nat. Rev. Microbiol. 2014, 12, 563–574. [Google Scholar] [CrossRef]
  194. Weber, M.G.; Walters-Laird, C.J.; Kol, A.; Rocha, C.S.; Hirao, L.A.; Mende, A.; Balan, B.; Arredondo, J.; Elizaldi, S.R.; Iyer, S.S.; et al. Gut germinal center regeneration and enhanced antiviral immunity by mesenchymal stem/stromal cells in SIV infection. JCI Insight 2021, 6, e149033. [Google Scholar] [CrossRef]
  195. Clinicaltrials.gov. Bone Marrow-Derived Mesenchymal Stem Cell Treatment for Severe Patients with Coronavirus Disease 2019 (COVID-19) ClinicalTrials.gov Identifier: NCT04346368. Available online: https://clinicaltrials.gov/ct2/show/NCT04346368 (accessed on 15 April 2020).
  196. Clinicaltrials.gov. Treatment of Severe COVID-19 Pneumonia with Allogeneic Mesenchymal Stromal Cells (COVID_MSV) Clinical Trials.gov Identifier: NCT04361942. Available online: https://clinicaltrials.gov/ct2/show/NCT04361942 (accessed on 28 October 2021).
  197. Clinicaltrials.gov. Mesenchymal Stem Cell Therapy for SARS-CoV-2-Related Acute Respiratory Distress Syndrome. ClinicalTrials.gov Identifier: NCT04366063. Available online: https://clinicaltrials.gov/ct2/show/NCT04366063 (accessed on 28 April 2021).
  198. Clinicaltrials.gov. Cellular Immuno-Therapy for COVID-19 Acute Respiratory Distress Syndrome—Vanguard (CIRCA-19). ClinicalTrials.gov Identifier: NCT04400032. Available online: https://clinicaltrials.gov/ct2/show/NCT04400032 (accessed on 26 April 2021).
  199. Clinicaltrials.gov. Mesenchymal Stromal Cells for the Treatment of SARS-CoV-2 Induced Acute Respiratory Failure (COVID-19 Disease). ClinicalTrials.gov Identifier: NCT04345601. Available online: https://clinicaltrials.gov/ct2/show/NCT04345601 (accessed on 26 September 2022).
  200. Clinicaltrials.gov. A Pilot Clinical Study on Inhalation of Mesenchymal Stem Cells Exosomes Treating Severe Novel Coronavirus Pneumonia. ClinicalTrials.gov Identifier: NCT 04276987. Available online: https://clinicaltrials.gov/ct2/show/NCT04276987 (accessed on 7 September 2020).
  201. Clinicaltrials.gov. Evaluation of Safety and Efficiency of Method of Exosome Inhalation in SARS-CoV-2 Associated Pneumonia. (COVID-19EXO). ClinicalTrials.gov Identifier: NCT04491240. Available online: https://clinicaltrials.gov/ct2/show/NCT04491240 (accessed on 4 November 2020).
  202. Clinicaltrials.gov. Mesenchymal Stem Cells (MSCs) in Inflammation-Resolution Programs of Coronavirus Disease 2019 (COVID-19) Induced Acute Respiratory Distress Syndrome (ARDS). ClinicalTrials.gov Identifier: NCT04377334. Available online: https://clinicaltrials.gov/ct2/show/NCT04377334 (accessed on 27 January 2020).
  203. Clinicaltrials.gov. Safety and Efficacy of Mesenchymal Stem Cells in the Management of Severe COVID-19 Pneumonia (CELMA). ClinicalTrials.gov Identifier: NCT 04429763. Available online: https://clinicaltrials.gov/ct2/show/NCT04429763 (accessed on 12 June 2020).
  204. Clinicaltrials.gov. Therapy for Pneumonia Patients Infected by 2019 Novel Coronavirus ClinicalTrials.gov Identifier: NCT04293692. Available online: https://clinicaltrials.gov/ct2/show/NCT04293692 (accessed on 18 March 2020).
  205. Clinicaltrials.gov. Use of UC-MSCs for COVID-19 Patients. ClinicalTrials.gov Identifier: NCT04355728. Available online: https://clinicaltrials.gov/ct2/show/NCT04355728 (accessed on 6 December 2021).
  206. Clinicaltrials.gov. Study of Human Umbilical Cord Mesenchymal Stem Cells in the Treatment of Severe COVID-19. ClinicalTrials.gov Identifier: NCT04273646. Available online: https://clinicaltrials.gov/ct2/show/NCT04273646 (accessed on 14 April 2020).
  207. Clinicaltrials.gov. Clinical Research of Human Mesenchymal Stem Cells in the Treatment of COVID-19 Pneumonia. ClinicalTrials.gov Identifier: NCT04339660. Available online: https://clinicaltrials.gov/ct2/show/NCT04339660 (accessed on 9 April 2020).
  208. Clinicaltrials.gov. Autologous Adipose-Derived Stem Cells (AdMSCs) for COVID-19. ClinicalTrials.gov Identifier: NCT04428801. Available online: https://clinicaltrials.gov/ct2/show/NCT04428801 (accessed on 5 May 2022).
  209. Clinicaltrials.gov. Battle against COVID-19 Using Mesenchymal Stromal Cells. ClinicalTrials.gov Identifier: NCT 04348461. Available online: https://clinicaltrials.gov/ct2/show/NCT04348461 (accessed on 17 March 2021).
  210. Clinicaltrials.gov. Clinical Trial to Assess the Safety and Efficacy of Intravenous Administration of Allogeneic Adult Mesenchymal Stem Cells of Expanded Adipose Tissue in Patients with Severe Pneumonia Due to COVID-19. ClinicalTrials. gov Identifier: NCT04366323. Available online: https://clinicaltrials.gov/ct2/show/NCT04366323 (accessed on 6 April 2022).
  211. Clinicaltrials.gov. ASC Therapy for Patients with Severe Respiratory COVID-19. ClinicalTrials.gov Identifier: NCT 04341610. Available online: https://clinicaltrials.gov/ct2/show/NCT04341610 (accessed on 27 May 2020).
  212. Clinicaltrials.gov. Zofin (Organicell Flow) for Patients with COVID-19. ClinicalTrials.gov Identifier: NCT04384445. Available online: https://clinicaltrials.gov/ct2/show/NCT04384445 (accessed on 21 September 2022).
  213. Clinicaltrials.gov. Umbilical Cord Mesenchymal Stem Cells for Immune Reconstitution in HIV-Infected Patients. ClinicalTrials.gov Identifier: NCT01213186. Available online: https://clinicaltrials.gov/ct2/show/NCT01213186 (accessed on 29 May 2013).
  214. Clinicaltrials.gov. Treatment with MSC in HIV-Infected Patients with Controlled Viremia and Immunological Discordant Response. ClinicalTrials.gov Identifier: NCT02290041. Available online: https://clinicaltrials.gov/ct2/show/NCT02290041 (accessed on 4 May 2020).
  215. Clinicaltrials.gov. A Tolerance Clinical Study on Aerosol Inhalation of Mesenchymal Stem Cells Exosomes in Healthy Volunteers. ClinicalTrials.gov Identifier: NCT04313647. Available online: https://clinicaltrials.gov/ct2/show/NCT04313647 (accessed on 4 August 2021).
  216. Clinicaltrials.gov. Using Human Menstrual Blood Cells to Treat Acute Lung Injury Caused by H7N9 Bird Flu Virus Infection. ClinicalTrials.gov Identifier: NCT02095444. Available online: https://clinicaltrials.gov/ct2/show/NCT02095444 (accessed on 24 March 2014).
  217. Clinicaltrials.gov. Regenerative Medicine for COVID-19 and Flu-Elicited ARDS Using Lomecel-B (RECOVER). ClinicalTrials.gov Identifier: NCT04629105. Available online: https://clinicaltrials.gov/ct2/show/NCT04629105 (accessed on 26 September 2022).
  218. Eveni, J.; Filipo, K.; Garfin, A.M.C.; Geocaniga-Gaviola, D.M.; Huot, C.; Iavro, E.; Ismail, K.; Itogo, N.; Kako, H.; Kal, M.; et al. Global Tuberculosis Report 2021; World Health Organization: Geneva, Switzerland, 2021; pp. 1–57. [Google Scholar]
  219. Can Sarinoglu, R.; Sili, U.; Eryuksel, E.; Olgun Yildizeli, S.; Cimsit, C.; Karahasan Yagci, A. Tuberculosis and COVID-19: An overlapping situation during pandemic. J. Infect. Dev. Ctries 2020, 14, 721–725. [Google Scholar] [CrossRef]
  220. Lange, C.; Dheda, K.; Chesov, D.; Mandalakas, A.M.; Udwadia, Z.; Horsburgh, C.R.J. Management of drug-resistant tuberculosis. Lancet 2019, 394, 953–966. [Google Scholar] [CrossRef]
  221. Cohen, S.B.; Gern, B.H.; Delahaye, J.L.; Adams, K.N.; Plumlee, C.R.; Winkler, J.K.; Sherman, D.R.; Gerner, M.Y.; Urdahl, K.B. Alveolar macrophages provide an early mycobacterium tuberculosis niche and initiate dissemination. Cell Host Microbe 2018, 24, 439–446.e4. [Google Scholar] [CrossRef] [Green Version]
  222. Orme, I.M.; Basaraba, R.J. The Formation of the granuloma in tuberculosis infection. Semin. Immunol. 2014, 26, 601–609. [Google Scholar] [CrossRef]
  223. Sandor, M.; Weinstock, J.V.; Wynn, T.A. Granulomas in Schistosome and Mycobacterial Infections: A Model of Local Immune Responses. Trends Immunol. 2003, 24, 44–52. [Google Scholar] [CrossRef]
  224. Raghuvanshi, S.; Sharma, P.; Singh, S.; Van Kaer, L.; Das, G. Mycobacterium tuberculosis evades host immunity by recruiting mesenchymal stem cells. Proc. Natl. Acad. Sci. USA 2010, 107, 21653–21658. [Google Scholar] [CrossRef]
  225. Schwartz, Y.S.; Belogorodtsev, S.N.; Filimonov, P.N.; Cherednichenko, A.G.; Pustylnikov, S.V.; Krasnov, V.A. BCG infection in mice is promoted by naive mesenchymal stromal cells (MSC) and suppressed by Poly(A:U)-conditioned MSC. Tuberculosis 2016, 101, 130–136. [Google Scholar] [CrossRef] [PubMed]
  226. Khan, A.; Mann, L.; Papanna, R.; Lyu, M.A.; Singh, C.R.; Olson, S. Mesenchymal stem cells internalize mycobacterium tuberculosis through scavenger receptors and restrict bacterial growth through autophagy. Sci. Rep. 2017, 7, 15010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Alquraini, A.; Khoury, J.E. Scavenger receptors. Curr. Biol. 2020, 30, R790–R795. [Google Scholar] [CrossRef] [PubMed]
  228. Jain, N.; Kalam, H.; Singh, L.; Sharma, V.; Kedia, S.; Das, P.; Ahuja, V.; Kumar, D. Mesenchymal stem cells offer a drug-tolerant and immune-privileged niche to mycobacterium tuberculosis. Nat. Commun. 2020, 11, 3062. [Google Scholar] [CrossRef] [PubMed]
  229. Singh, V.K.; Mishra, A.; Bark, S.; Mani, A.; Subbian, S.; Hunter, R.L.; Jagannath, C.; Khan, A. Human mesenchymal stem cell based intracellular dormancy model of mycobacterium tuberculosis. Microbes Infect. 2020, 22, 423–431. [Google Scholar] [CrossRef]
  230. Shamputa, I.C.; Van Deun, A.; Salim, M.A.; Hossain, M.A.; Fissette, K.; de Rijk, P.; Rigouts, L.; Portaels, F. Endogenous reactivation and true treatment failure as causes of recurrent tuberculosis in a high incidence setting with a low HIV infection. Trop. Med. Int. Health 2007, 12, 700–708. [Google Scholar] [CrossRef]
  231. Tsenova, L.; Singhal, A. Effects of host-directed therapies on the pathology of tuberculosis. J. Pathol. 2020, 250, 636–646. [Google Scholar] [CrossRef]
  232. Yudintceva, N.M.; Bogolyubova, I.O.; Muraviov, A.N.; Sheykhov, M.G.; Vinogradova, T.I.; Sokolovich, E.G.; Samusenko, I.A.; Shevtsov, M.A. Application of the allogenic mesenchymal stem cells in the therapy of the bladder tuberculosis. J. Tissue Eng. Regen. Med. 2018, 12, e1580–e1593. [Google Scholar] [CrossRef]
  233. Harman, R.M.; Yang, S.; He, M.K.; Van de Walle, G.R. Antimicrobial peptides secreted by equine mesenchymal stromal cells inhibit the growth of bacteria commonly found in skin wounds. Stem Cell Res. Ther. 2017, 8, 157. [Google Scholar] [CrossRef] [Green Version]
  234. Chow, L.; Johnson, V.; Impastato, R.; Coy, J.; Strumpf, A.; Dow, S. Antibacterial activity of human mesenchymal stem cells mediated directly by constitutively secreted factors and indirectly by activation of innate immune effector cells. Stem Cells Transl. Med. 2020, 9, 235–249. [Google Scholar] [CrossRef]
  235. Sutton, M.T.; Fletcher, D.; Ghosh, S.K.; Weinberg, A.; van Heeckeren, R.; Kaur, S.; Sadeghi, Z.; Hijaz, A.; Reese, J.; Lazarus, H.M.; et al. Antimicrobial properties of mesenchymal stem cells: Therapeutic potential for cystic fibrosis infection, and treatment. Stem Cells Int. 2016, 2016, 5303048. [Google Scholar] [CrossRef] [Green Version]
  236. Colombo, M.; Raposo, G.; Thery, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Ann. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  237. Favaro, E.; Carpanetto, A.; Lamorte, S.; Fusco, A.; Caorsi, C.; Deregibus, M.C.; Bruno, S.; Amoroso, A.; Giovarelli, M.; Port, M.; et al. Human Mesenchymal Stem Cell-Derived Microvesicles Modulate T Cell Response to Islet Antigen Glutamic Acid Decarboxylase in Patients With Type 1 Diabetes. Diabetologia 2014, 57, 1664–1673. [Google Scholar] [CrossRef]
  238. Li, P.; Zhao, Y.; Ge, L. Therapeutic Effects of Human Gingiva-Derived Mesenchymal Stromal Cells on Murine Contact Hypersensitivity via Prostaglandin E2-EP3 Signaling. Stem Cell Res. Ther. 2016, 7, 103. [Google Scholar] [CrossRef] [Green Version]
  239. Huang, F.; Chen, M.; Chen, W.; Gu, J.; Yuan, J.; Xue, Y.; Dang, J.; Su, W.; Wang, J.; Zadeh, H.H.; et al. Human Gingiva-Derived Mesenchymal Stem Cells Inhibit Xeno-Graft-Versus-Host Disease via CD39-CD73-Adenosine and IDO Signals. Front. Immunol. 2017, 8, 68. [Google Scholar] [CrossRef] [Green Version]
  240. Zhang, X.; Huang, F.; Li, W.; Dang, J.L.; Yuan, J.; Wang, J.; Zeng, D.-L.; Sun, C.-X.; Liu, Y.-Y.; Ao, Q.; et al. Human Gingiva-Derived Mesenchymal Stem Cells Modulate Monocytes/Macrophages and Alleviate Atherosclerosis. Front. Immunol. 2018, 9, 878. [Google Scholar] [CrossRef]
  241. Ren, W.; Hou, J.; Yang, C.; Wang, H.; Wu, S.; Wu, Y.; Zhao, X.; Lu, C. Extracellular Vesicles Secreted by Hypoxia Pre-Challenged Mesenchymal Stem Cells Promote non-Small Cell Lung Cancer Cell Growth and Mobility as Well as Macrophage M2 Polarization via miR-21-5p Delivery. J. Exp. Clin. Cancer Res. 2019, 38, 62. [Google Scholar] [CrossRef] [Green Version]
  242. Peng, Y.; Chen, X.; Liu, Q.; Zhang, X.; Huang, K.; Liu, L.; Li, H.; Zhou, M.; Huang, F.; Fan, Z.; et al. Mesenchymal Stromal Cells Infusions Improve Refractory Chronic Graft Versus Host Disease Through an Increase of CD5+ Regulatory B Cells Producing Interleukin 10. Leukemia 2015, 29, 636–646. [Google Scholar] [CrossRef] [Green Version]
  243. Budoni, M.; Fierabracci, A.; Luciano, R.; Petrini, S.; Di Ciommo, V.; Muraca, M. The Immunosuppressive Effect of Mesenchymal Stromal Cells on B Lymphocytes is Mediated by Membrane Vesicles. Cell Transplant. 2013, 22, 369–379. [Google Scholar] [CrossRef] [Green Version]
  244. Bernard, O.; Jeny, F.; Uzunhan, Y.; Dondi, E.; Terfous, R.; Label, R.; Sutton, A.; Larghero, J.; Vanneaux, V.; Nunes, H.; et al. Mesenchymal Stem Cells Reduce Hypoxia-Induced Apoptosis in Alveolar Epithelial Cells by Modulating HIF and ROS Hypoxic Signaling. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 314, L360–L371. [Google Scholar] [CrossRef]
  245. Yudintceva, N.; Mikhailova, N.; Bobkov, D.; Yakovleva, L.; Nikolaev, B.; Krasavina, D.; Muraviov, A.; Vinogradova, T.; Yablonskiy, P.; Samusenko, I.; et al. Evaluation of the biodistribution of mesenchymal stem cells in a pre-clinical renal tuberculosis model by non-linear magnetic response measurements. Front. Phys. 2021, 9, 198. [Google Scholar] [CrossRef]
  246. Chen, S.; Cui, G.; Peng, C.; Lavin, M.F.; Sun, X.; Zhang, E. Transplantation of Adipose-Derived Mesenchymal Stem Cells Attenuates Pulmonary Fibrosis of Silicosis via Anti-Inflammatory and Anti-Apoptosis Effects in Rats. Stem Cell Res. Ther. 2018, 9, 110. [Google Scholar] [CrossRef] [PubMed]
  247. Zhu, Y.G.; Feng, X.M.; Abbott, J.; Fang, X.H.; Hao, Q.; Monsel, A.; Qu, J.; Matthay, M.A.; Lee, J.W. Human Mesenchymal Stem Cell Microvesicles for Treatment of Escherichia Coli Endotoxin-Induced Acute Lung Injury in Mice. Stem Cells 2014, 3, 116–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Li, X.; Wang, Y.; An, G.; Liang, D.; Zhu, Z.; Lian, X.; Niu, P.; Guo, C.; Tian, L. Bone Marrow Mesenchymal Stem Cells Attenuate Silica-Induced Pulmonary Fibrosis via Paracrine Mechanisms. Toxicol. Lett. 2017, 270, 96–107. [Google Scholar] [CrossRef] [PubMed]
  249. Poggi, A.; Zocchi, M.R. Immunomodulatory properties of mesenchymal stromal cells: Still unresolved “Yin and Yang”. Curr. Stem Cell Res. Ther. 2019, 14, 344–350. [Google Scholar] [CrossRef]
  250. Sack, D.A.; Sack, R.B.; Chaignat, C.L. Getting serious about cholera. N. Engl. J. Med. 2006, 355, 649–651. [Google Scholar] [CrossRef] [Green Version]
  251. Bishop, A.L.; Camilli, A. Vibrio cholerae: Lessons for mucosal vaccine design. Expert Rev. Vaccines 2011, 10, 79–94. [Google Scholar] [CrossRef] [Green Version]
  252. Bhattacharya, D.; Dey, S.; Roy, S.; Parande, M.V.; Telsang, M.; Seema, M.H.; Parande, A.V.; Mantur, B.G. Multidrug-resistant Vibrio cholerae O1 was responsible for a cholera outbreak in 2013 in Bagalkot, North Karnataka. Jpn. J. Infect. Dis. 2015, 68, 347–350. [Google Scholar] [CrossRef] [Green Version]
  253. Bhattacharya, D.; Sayi, D.S.; Thamizhmani, R.; Bhattacharjee, H.; Bharadwaj, A.P.; Roy, A.; Sugunan, A.P. Emergence of multidrug-resistant Vibrio cholerae O1 biotype El Tor in Port Blair, India. Am. J. Trop. Med. Hyg. 2012, 86, 1015–1017. [Google Scholar] [CrossRef] [Green Version]
  254. Chatterjee, S.N.; Chaudhuri, K. Lipopolysaccharides of Vibrio cholerae: III. Biological functions. Biochim. Biophys. Acta 2006, 1762, 1–16. [Google Scholar] [CrossRef]
  255. Saeedi, P.; Halabian, R.; Fooladi, A.I. Antimicrobial effects of mesenchymal stem cells primed by modified LPS on bacterial clearance in sepsis. J. Cell Physiol. 2019, 234, 4970–4986. [Google Scholar] [CrossRef]
  256. Moulazadeh, A.; Soudi, S.; Bakhshi, B. Immunomodulatory effects of adipose-derived mesenchymal stem cells on epithelial cells function in response to Vibrio cholera in a co-culture model. Iran. J. Allergy Asthma Immunol. 2021, 20, 550–562. [Google Scholar] [CrossRef]
  257. Clinicaltrials.gov. Effectivity of Local Implantation of the Mesenchymal Stem Cell on Vertebral Bone Defect Due to Mycobaterium Tuberculosis Infection (Clinical Trial) Clinicaltrials.gov, Identifier: NCT04493918. Available online: https://clinicaltrials.gov/ct2/show/NCT04493918 (accessed on 3 August 2020).
  258. Erokhin, V.V.; Vasil’eva, I.A.; Konopliannikov, A.G.; Chukanov, V.I.; Tsyb, A.F.; Bagdasarian, T.R.; Danilenko, A.A.; Lepekhina, L.A.; Kal’sina, S.S.; Semenkova, I.V.; et al. Systemic transplantation of autologous mesenchymal stem cells of the bone marrow in the treatment of patients with multidrug-resistant pulmonary tuberculosis. Probl. Tuberk. Bolezn. Legk. 2008, 10, 3–6. [Google Scholar]
  259. Skrahin, A.; Ahmed, R.K.; Ferrara, G.; Rane, L.; Poiret, T.; Isaikina, Y.; Skrahina, A.; Zumla, A.; Maeurer, M.J. Autologous mesenchymal stromal cell infusion as adjunct treatment in patients with multidrug and extensively drug-resistant tuberculosis: An open-label Phase 1 Safety Trial. Lancet Respir. Med. 2014, 2, 108–122. [Google Scholar] [CrossRef]
  260. Skrahin, A.; Jenkins, H.E.; Hurevich, H.; Solodovnikova, V.; Isaikina, Y.; Klimuk, D.; Rohava, Z.; Skrahina, A. Effectiveness of a Novel Cellular Therapy to Treat Multidrug-Resistant Tuberculosis. Int. J. Mycobacteriol. 2016, 5, 23. [Google Scholar] [CrossRef]
Figure 1. Molecules released by MSC-EVs. The MSC-EVs contain cytokines, growth factors, and other active molecules that maintain the modulation of the immune response, have anti-inflammatory, antiviral/antibacterial effects, and promote epithelial repair and tissue regeneration. MSCs, mesenchymal stem/stromal cells; EVs, extracellular vesicles; AECs; alveolar epithelial cells; macrophage; T-cell; B-cell; NK, natural killer cell.
Figure 1. Molecules released by MSC-EVs. The MSC-EVs contain cytokines, growth factors, and other active molecules that maintain the modulation of the immune response, have anti-inflammatory, antiviral/antibacterial effects, and promote epithelial repair and tissue regeneration. MSCs, mesenchymal stem/stromal cells; EVs, extracellular vesicles; AECs; alveolar epithelial cells; macrophage; T-cell; B-cell; NK, natural killer cell.
Bioengineering 09 00662 g001
Figure 2. MSC therapies for treatment of coronavirus-induced lung injury. COVID-19, coronavirus; MSCs, mesenchymal stem/stromal cells; EVs, extracellular vesicles; macrophage; neutrophil; T-cell; B-cell; dendritic cell; NK, natural killer cell.
Figure 2. MSC therapies for treatment of coronavirus-induced lung injury. COVID-19, coronavirus; MSCs, mesenchymal stem/stromal cells; EVs, extracellular vesicles; macrophage; neutrophil; T-cell; B-cell; dendritic cell; NK, natural killer cell.
Bioengineering 09 00662 g002
Table 1. Commonly used sources of MSCs.
Table 1. Commonly used sources of MSCs.
SourceAbbreviationProliferation
Rate
Doubling
Time
ImmunogenicityMSCs PhenotypeReferences
1Bone MarrowBM-MSCsLowest40 hMediumStro-1, CD271, SSEA-4, CD146[30,31,32,33,34,35,36]
2Adipose TissueA-MSCsHigher5 daysHighCD271, CD146[33,34,35,36]
3Umbilical CordUC-MSCsMedium30 hHighCD146[35,36,37,38]
4PlacentaP-MSCsHigh36 hHighc-Kit, Oct-4, SSEA-4, Y-box 2[39,40]
Table 2. Mechanisms of MSCs and MSC-EVs influence on immune cells.
Table 2. Mechanisms of MSCs and MSC-EVs influence on immune cells.
AbbreviationImmune CellMechanismEffectReference
1BM-MSC-EVsCD4 + T cellEVs-encapsulating miR-23a-3p and post-transcriptionally regulated TGF-beta receptor 2 in T cellssuppressive Th1 differentiation[70]
2BM-MSC-EVsT cellincreasing IL-10
and TGF-beta
promote T cells apoptosis and inhibit proliferation[71]
3UC-MSCsT cellthrough the COX2/PGE2/NF-kB signaling pathwayinhibiting T cell proliferation and DC
differentiation
[72]
4AD-MSCT cellthrough regulating TGF-beta and PGE2regulate the Th17/Treg balance[73]
5BM-MSCB cellinhibition of BAFF productionsuppress the excessive activation of B-cells[74]
6BM-MSC-EVsB celltargeting PI3K/AKT signaling pathwayinhibit activation of B cell[75]
7BM-MSCB cellincreased expression of CCL2 by CCL2-MST1-mTOR-STAT1 mediated metabolic signaling pathwayprevent inhibition differentiation, proliferation,
and antibody secretion of B-cell
[76]
8BM-MSC-EVsDCsexpression of anti-inflammatory factors (TGF-beta 1 and IL-10) and reduce the generation of proinflammatory cytokines (L-6 and IL-12p70)attenuate DCs maturation and function[77]
9G-MSCNK cellregulating IDO and PGE2inhibit the activity of NK cells[78]
10BM-MSCsNK cellinhibit IL-12 and IL-21suppression NK cell proliferation but increase IFN-gamma and IFN-alpha production[79]
11UC-MSCmacrophageregulating macrophage metabolic pathwaysaffect M1/M2 balance[80]
12BM-MSC-EVsmacrophagedown-regulating
IL-23 and IL-22
enhances the anti-inflammatory phenotype of macrophages, promoting
inflammation remission
[81]
13AD-MSCsmacrophagedown-regulating
IL-23 and IL-22
toward M2 phenotype polarization[82]
14BM-MSC-EVsmacrophagethrough miR-223/pKNOX1 pathwaypromoting macrophages differentiation toward M2[83]
15BM-MSC-EVsmacrophagethrough TLR4/NF-kB/PI3K/Akt signaling cascadetoward M2 phenotype polarization[84]
16UC-MSC-EVsmacrophageincreased the proportion of M2 macrophage polarizationattenuate DAH induced inflammatory responses and alveolar hemorrhage[85]
MSC, mesenchymal stem cells; EVs, extracellular vesicles; BM-MSC, bone marrow-derived mesenchymal stem cells; BM-MSC-EVs, bone marrow MSC-derived extracellular vesicles; UC-MSC, umbilical cord-derived mesenchymal stem cells; AD-MSC, adipose-derived MSC; G-MSC, gingiva-derived mesenchymal stem cells; DCs, dendritic cells; IL, interleukin; IDO, indoleamine 2,3-dioxygenase; PGE2, prostaglandin E2; Th1, T-helper 1; Treg, regulatory T; IFN, interferon; TGF-beta 1, transforming growth factor beta 1; DAH, diffuse alveolar hemorrhage; Tfh, T follicular helper; IL-10, interleukin 10; TLR4, toll-like receptor 4; Th17, T-helper 17.
Table 3. MSCs and MSC-EVs based clinical trials of the viral infection diseases.
Table 3. MSCs and MSC-EVs based clinical trials of the viral infection diseases.
Study TitleAbbreviationViral Infectious DiseasesStatusCountryDescriptionReference
1Bone Marrow-Derived Mesenchymal Stem Cell Treatment for Severe Patients With Coronavirus Disease 2019 (COVID-19)BM-MSCsCOVID-19Phase 2ChinaConventional treatment plus BM-MSCs (1 × 106 cells/kg body weight intravenously[195]
2Treatment of Severe COVID-19 Pneumonia with Allogeneic Mesenchymal Stromal Cells (COVID_MSV)BM-MSCsCOVID-19Phase 2SpainIV injection of 1 × 106 cells/kg diluted in 100 mL saline[196]
3Mesenchymal Stem Cell Therapy for SARS-CoV-2-related Acute Respiratory Distress SyndromeBM-MSCs/
BM-MSC-EVs
COVID-19Phase 3IranTwo doses of MSCs 1 × 108 at Day 0 and Day 2 plus conventional treatment[197]
4Cellular Immuno-Therapy for COVID-19 Acute Respiratory Distress Syndrome—Vanguard (CIRCA-19)BM-MSCsCOVID-19Phase 1CanadaIV administration[198]
5Mesenchymal Stromal Cells for
the Treatment of SARS-CoV-2 Induced Acute Respiratory Failure (COVID-19 Disease)
BM-MSCsCOVID-19Early
Phase 1
USA1 × 108 cells/kg body weight intravenously[199]
6A Pilot Clinical Study on Inhalation of Mesenchymal Stem Cells Exosomes Treating Severe Novel Coronavirus PneumoniaBM-MSC-EVsCOVID-19Phase 1China5 times aerosol inhalation of MSC-EVs (2 × 108/3 mL at Day 1, Day 2, Day 3, Day 4, Day 5)[200]
7Evaluation of Safety and Efficiency of Method of Exosome Inhalation in SARS-CoV-2 Associated Pneumonia. (COVID-19EXO)BM-MSC-EVsCOVID-19Phase 1
Phase 2
RussiaTwice a day during 10 days inhalation of 3 mL special solution contained 0.5–2 × 1010 of EVs.[201]
8Mesenchymal Stem Cells (MSCs) in
Inflammation-Resolution Programs of Coronavirus Disease 2019 (COVID-19) Induced Acute Respiratory Distress Syndrome (ARDS)
BM-MSCsCOVID-19Phase 2GermanyInfusion of allogeneic bone marrow-derived human mesenchymal stem (stromal) cells[202]
9Safety and Efficacy of Mesenchymal Stem Cells in the Management of Severe COVID-19 Pneumonia (CELMA)UC-MSCsCOVID-19Phase 2USA1 × 106 cells/kg body weight intravenously[203]
10Therapy for Pneumonia Patients Infected by 2019 Novel CoronavirusUC-MSCsCOVID-19With- drawnChina0.5 × 106/kg body weight suspended in 100 mL saline intravenously at Day 1, Day 3, Day 5, Day 7[204]
11Use of UC-MSCs for COVID-19 PatientsUC-MSCsCOVID-19Phase 2USAConventional treatment plus UC-MSCs (1 × 108/kg body weight intravenously[205]
12Study of Human Umbilical Cord Mesenchymal Stem Cells in the Treatment of Severe COVID-19UC-MSCsCOVID-19Not yet recruitingChina4 times of UC-MSCs (0.5 × 106 UC-MSCs cell/kg body weight intravenously at Day 1, Day 3, Day 5, Day 7)[206]
13Clinical Research of Human Mesenchymal Stem Cells in the Treatment of COVID-19 PneumoniaUC-MSCsCOVID-19Phase 2China1 × 106 UC-MSCs/kg suspended in 100 mL saline[207]
14Autologous Adipose-derived Stem Cells (AdMSCs) for COVID-19A-MSCsCOVID-19Phase 2USA3 doses of 2 × 106 cells through IV every 3 days[208]
15Battle Against COVID-19 Using
Mesenchymal Stromal Cells
A-MSCsCOVID-19Phase 2SpainTwo serial doses of 1.5 × 106 cells/kg[209]
16Clinical Trial to Assess the Safety and Efficacy of Intravenous Administration of Allogeneic Adult Mesenchymal Stem Cells of Expanded Adipose Tissue in Patients With Severe Pneumonia Due to COVID-19A-MSCsCOVID-19Phase 2SpainTwo doses of 8 × 106
A-MSCs
[210]
17ASC Therapy for Patients with Severe Respiratory COVID-19A-MSCsCOVID-19Phase 2Denmark1 × 108 cells/kg diluted in
100 mL saline
[211]
18Zofin (Organicell Flow) for Patients With COVID-19MSC-EVsCOVID-19Phase 1USAZofin with 1ml, containing 2–5 × 1011 EVs/mL in addition to the Standard care.[212]
19Umbilical Cord Mesenchymal Stem Cells for Immune Reconstitution in HIV-infected PatientsUC-MSCsHIV/AIDSPhase 2ChinaHigh and low doses of MSCs (at 0, 4, 12, 24, 36 and 48 week since the onset of treatment)[213]
20Treatment with MSC in HIV-infected Patients with Controlled Viremia and Immunological Discordant ResponseA-MSCsHIV/AIDSPhase 1
Phase 2
SpainIntravenous infusion of 4 doses of A-MSCs (1 × 106 cells/kg, weeks 0-4-8-20).[214]
21A Tolerance Clinical Study on Aerosol Inhalation of Mesenchymal Stem Cells Exosomes In Healthy VolunteersMSC-EVsHealthy VolunteersPhase 1ChinaAerosol inhalation of MSC-EVs[215]
22Using Human Menstrual Blood Cells to Treat Acute Lung Injury Caused by H7N9 Bird Flu Virus InfectionMSCsH7N9 Bird Flu Virus InfectionPhase 1
Phase 2
China1~10 × 107 cells/kg infusion frequency: 2 times a week, 2 weeks for infusion[216]
23Regenerative Medicine for COVID-19 and Flu-Elicited ARDS Using Lomecel-B (RECOVER)MSCsARDS
COVID-19
Phase 1USA1 × 108 cells/kg on Day 0[217]
MSC, mesenchymal stem cells; EVs, extracellular vesicles; MSC-EVs, MSC-derived extracellular vesicles; BM-MSC, bone marrow-derived mesenchymal stem cells; AD-MSC, adipose-derived MSC; UC-MSC, umbilical cord-derived mesenchymal stem cells.
Table 4. MSC- and MSC-EV-based clinical trials of the bacterial infection diseases.
Table 4. MSC- and MSC-EV-based clinical trials of the bacterial infection diseases.
Study TitleAbbreviationBacterial DiseasesStatusCountryDescriptionReference
1Effectivity of Local Implantation of the Mesenchymal Stem Cell on Vertebral Bone Defect Due to Mycobaterium Tuberculosis Infection (Clinical Trial)MSCsExtrapulmonary tuberculosisPhase 2Indonesia3 × 107 cells/kg diluted in 2 mL 0.9% NaCl intravenously[257]
2Systemic Transplantation of Autologous Mesenchymal Stem Cells of the Bone Marrow in the Treatment of Patients With Multidrug-Resistant Pulmonary TuberculosisMSCsTuberculosis; multidrug resistant, extensive drug resistantCompletedRussiaNot stated[258]
3Autologous Mesenchymal Stromal Cell Infusion as Adjunct Treatment in Patients With Multidrug and Extensively Drug-Resistant Tuberculosis: An Open-Label Phase 1 Safety Trial.BM-MSCsTuberculosis; multidrug resistant, extensive drug resistantPhase 1Belarus1 × 107 cells/kg diluted in saline[259]
4Effectiveness of a Novel Cellular Therapy to Treat Multidrug-Resistant Tuberculosis.BM-MSCsTuberculosis; multidrug resistant, extensive drug resistantPhase 1Belarus1 × 107 cells/kg diluted in saline[260]
MSC, mesenchymal stem cells; BM-MSC, bone marrow-derived mesenchymal stem cells.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yudintceva, N.; Mikhailova, N.; Fedorov, V.; Samochernych, K.; Vinogradova, T.; Muraviov, A.; Shevtsov, M. Mesenchymal Stem Cells and MSCs-Derived Extracellular Vesicles in Infectious Diseases: From Basic Research to Clinical Practice. Bioengineering 2022, 9, 662. https://doi.org/10.3390/bioengineering9110662

AMA Style

Yudintceva N, Mikhailova N, Fedorov V, Samochernych K, Vinogradova T, Muraviov A, Shevtsov M. Mesenchymal Stem Cells and MSCs-Derived Extracellular Vesicles in Infectious Diseases: From Basic Research to Clinical Practice. Bioengineering. 2022; 9(11):662. https://doi.org/10.3390/bioengineering9110662

Chicago/Turabian Style

Yudintceva, Natalia, Natalia Mikhailova, Viacheslav Fedorov, Konstantin Samochernych, Tatiana Vinogradova, Alexandr Muraviov, and Maxim Shevtsov. 2022. "Mesenchymal Stem Cells and MSCs-Derived Extracellular Vesicles in Infectious Diseases: From Basic Research to Clinical Practice" Bioengineering 9, no. 11: 662. https://doi.org/10.3390/bioengineering9110662

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