More Information

Submitted: November 23, 2023 | Approved: December 04, 2023 | Published: December 05, 2023

How to cite this article: Al-Anazi KA, Al-Ansari RY. Update on the Use of Mesenchymal Stem Cells in the Treatment of Various Infectious Diseases Including COVID-19 Infection. J Stem Cell Ther Transplant. 2023; 7: 034-042.

DOI: 10.29328/journal.jsctt.1001033

Copyright License: © 2023 Al-Anazi KA, et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Keywords: Mesenchymal stem cells; Sepsis; Acute respiratory distress syndrome; Chagas disease; Mycobacterial tuberculosis; COVID-19 infection

 FullText PDF

Update on the Use of Mesenchymal Stem Cells in the Treatment of Various Infectious Diseases Including COVID-19 Infection

Khalid A Al-Anazi1* and Rehab Y Al-Ansari2

1Department of Hematology and Hematopoietic Stem Cell Transplantation, Oncology Center, King Fahad Specialist Hospital, P.O. Box: 15215, Dammam 31444, Saudi Arabia
2Consultant Hematologist, Section of Adult Hematology, Department of Internal Medicine, King Fahad Military Medical Complex, Dhahran 31932, Saudi Arabia

*Address for Correspondence: Dr. Khalid A Al-Anazi, Consultant Hemato-Oncologist, Department of Hematology and Hematopoietic Stem Cell Transplantation, Oncology Center, King Fahad Specialist Hospital, P.O. Box: 15215, Dammam 31444, Saudi Arabia, Email: kaa_alanazi@yahoo.com

Mesenchymal Stem Cells (MSCs) have antimicrobial, anti-inflammatory, immunomodulatory, and regenerative potentials. Additionally, utilization of MSCs in the clinical arena has been shown to be safe and well tolerated. Hence, this form of cellular therapy has gained particular attention in the treatment of several infectious disorders and their complications.

MSCs have been successfully used in the treatment of the following infections and their complications: bacterial infections including complicated sepsis; viral infections including Human Immunodeficiency Virus (HIV), hepatitis B and C viruses, and Coronavirus disease (COVID-19) complicated by acute respiratory distress syndrome; parasitic infections including schistosomiasis, malaria, and Chagas disease; and mycobacterial infections including tuberculosis. The use of MSCs derived from certain sources and Extracellular Vesicles (ECVs) derived from MSCs has improved their efficacy and reduced their side effects. However, the clinical application of MSCs in the treatment of several infectious diseases still faces real challenges that need to be resolved. The current status of MSCs and the controversies related to their utilization in various infections will be thoroughly discussed in this review.

MSCs are heterogeneous, non-hematopoietic, adult multipotent stromal progenitor cells that are capable of self-renewal and differentiation into multiple lineages and various cell types [1-3]. MSCs can be obtained from various body tissues and organs and they have characteristic features and surface markers on flow cytometry as shown in Table 1 [1-5]. MSCs have several antimicrobial, anti-inflammatory, and regenerative properties which make them suitable candidates for use in the treatment of various infections and their complications. These properties include: (1) inhibition of biofilm formation; (2) excessive recruitment of neutrophils to the sites of infection or inflammation; (3) reduction of tissue damage and release of paracrine factors that direct tissue regeneration and wound healing; (4) detection and elimination of invading pathogens and release of antimicrobial factors that are needed for clearance of pathogens and reducing their load as shown in preclinical models of sepsis, cystic fibrosis and Acute Respiratory Distress Syndrome (ARDS); (5) having the capacity to enhance antibacterial activity by interaction with the host innate immune system; (6) activation of phagocytosis and reprogramming of macrophages and neutrophils towards a more anti-inflammatory phenotype; (7) activation of the immune responses by induction of proinflammatory responses and downregulation of proinflammatory cytokines; (8) secretion of antimicrobial peptides, molecules, and proteins such as: interleukin (IL)-17, indoleamine 2,3 dioxygenase, β-defensins, lipocalin-2, and cathelicidin LL-37; (9) inhibition of inflammation and regulation of multiple inflammatory networks; and (10) changing the microenvironment at the site of infection and inhibition of T-cell proliferation at the site of tissue injury [2,3,5-7].

Table 1: Sources and characteristic features and surface markers of MSCs.
Sources of MSCs Characteristic features of MSCs Surface markers of MSCs
  1. Bone marrow
  2. Peripheral blood
  3. Umbilical cord (UC) blood
  4. Wharton's jelly of UC
  5. Placenta
  6. Amniotic fluid
  7. Menstrual blood
  8. Breast milk
  9. Adipose tissues
  10. Dental pulp
  11. Palatal tonsils
  12. Salivary glands
  13. Lung and liver tissues
     
  1. Differentiation into osteoblasts, adipocytes, and chondrocytes.
  2. Adherence to the plastic vessel under optimal culture conditions.
  3. Having characteristic surface markers on flow cytometry.
Characteristically positive:
CD 105
CD 73
CD 90 Characteristically negative:
CD 45
CD 34
CD 14
CD 11b
CD 19
CD 79a
HLA-DR
- MSCs: Mesenchymal Stem Cells
- HLA: Human Leukocyte Antigen

MSCs have demonstrated potent antimicrobial effects through direct and indirect mechanisms against the major classes of human pathogens including bacteria, viruses, fungi, and parasites across a wide range of infection models. The immunomodulatory and antibacterial activity of MSCs can be enhanced by various forms of preconditioning protocols [1,8]. Currently, more than 80 Clinical Trials (CTs) are investigating the use of MSCs in the treatment of various infections including sepsis, local bacterial infections, and viral infections such as COVID-19 [1,9]. The completed CTs on the use of MSCs in difficult-to-treat infectious diseases have demonstrated safety, tolerability, as well as potential efficacy as adjuvant therapy in complex or resistant infections [1]. The use of ECVs of MSCs is a promising cell-free treatment strategy that allows for solving problems associated with the safety of cell therapy and increasing its effectiveness. Hence, MSCs and their ECVs can be a promising tool for the treatment of various infectious diseases, particularly in combination with antimicrobial agents [2,4].

Factors that affect the outcome of mesenchymal stem cell therapies

Recent studies have suggested that factors such as age, gender, route of administration, infused stem cell dose, as well as biological sources of MSCs, have a significant impact on the outcome of MSC therapies [10-14]. Also, studies have shown that: (1) it is preferable to use MSCs obtained from Bone Marrows (BMs) of young healthy donors or derived from Umbilical Cords (UCs) directly after birth; and (2) administration of MSCs that are compatible with the biological gender of the recipient can avoid gender-specific immunological complications [12]. Although similar, MSCs derived from different sources possess distinct characteristics, advantages, and disadvantages, including their differentiation potential and proliferation capacity, which influence their clinical applicability [15]. However, the preferred source may vary according to the clinical indication. Examples are: MSCs derived from BM, adipose tissue, and dental tissues are preferable for use in oral and dental regeneration while MSCs obtained from UC and other perinatal tissues are preferable in COVID-19 infections [16-20].

Mesenchymal stem cells in bacterial infections

Chronic bacterial infections associated with biofilm formation are often difficult to treat without extended courses of antibiotic therapy [21]. The antimicrobial activity of MSCs relies on direct effects and indirect effects by secreting paracrine factors to inhibit bacterial growth [6]. In animal models, the combination of activated MSCs and antibiotic therapy may become a promising approach to treating infections caused by drug-resistant bacteria and accelerate the healing of infected tissues [22,23]. In patients with sepsis or pneumonia, administration of MSCs has been shown to inhibit bacterial growth and enhance survival by improving clearance of pathogenic bacteria [24,25]. So, MSCs should be considered as novel therapeutic strategies to control chronic infections and infections caused by multi-drug resistant organisms [6,21].

Sepsis and septic shock are life-threatening disorders that are associated with high rates of morbidity and mortality [26,27]. Recently, due to the failure of conventional therapies, research has been focused on innovative treatments such as cellular therapies [26]. The antimicrobial, immunomodulatory, anti-inflammatory, anti-apoptotic, and regenerative properties of MSCs can protect against organ failure caused by sepsis and septic shock. Consequently, MSCs have been extensively utilized in preclinical as well as clinical trials in various infectious diseases [26,27]. However, the mechanisms involved in the way MSCs exert their beneficial effects to control inflammation and prolong survival in septic conditions remain unclear [28]. Based on their immunomodulatory and antimicrobial properties, adult MSCs represent an emerging therapeutic strategy to treat sepsis and sepsis-induced organ dysfunction or failure [29,30]. Adult MSCs have been shown to reduce mortality in experimental models of sepsis [7]. Three phase I CTs, that included small numbers of patients, on the use of MSCs in the treatment of patients having septic shock have shown safety and positive impact of MSC therapy on survival rates of patients during the early phase of sepsis. However, there is an urgent need to perform phase II as well as randomized phase III CTs to determine the role of MSC therapies in patients with septic shock [31-33]. Additionally, ECVs of MSCs have therapeutic effects that are similar to the parent MSCs and they can protect against sepsis-induced organ dysfunction [27]. Studies in animal models have shown that the use of ECVs obtained from BM-derived MSCs was associated with less organ damage in comparison to ECVs derived from MSCs obtained from other sources [34]. Additionally, ECVs derived from MSCs have shown a superior safety profile as well as the ability to be stored safely without loss of function compared to the parent cells. Hence, MSC-ECVs may be used as a novel alternative to MSC-based therapy in sepsis [27].

Mesenchymal stem cells and viral infections

The interplay between MSCs and viruses can be described as a double-edged sword as MSCs have been found to be highly susceptible to viral infections in vitro [35]. Upon encountering certain viruses, MSCs appear to produce deleterious effects and they act as viral transmitters and this may raise concerns about their therapeutic efficacy. On the other hand, MSCs provide beneficial effects such as allowing the proliferation and function of specific antiviral effector cells instead of suppressing them thus they may serve as an ideal tool to study viral pathogenesis and protect hosts against viral challenges by using their antimicrobial activity [35-37]. Additionally, MSCs express receptors that are used by HIV, hepatitis B virus (HBV), and herpes viruses for their interaction with target cells. Hence, MSCs are permissive for these viruses and could transmit them to recipients of allogeneic stem cell transplantation [38]. As the clinical efficacy of MSC infusion may be impaired by some destructive outcomes, MSCs should be screened for HIV, HBV, herpes, and other viruses prior to transplantation in order to prevent the evolution of viral-associated diseases and to ensure the safety of MSC therapy [35,38].

Upon infection, Lung Resident (LR)-MSCs mount an antiviral response and release a variety of immunomodulatory mediators which may have a biological impact on the pulmonary microenvironment [39]. During the early phase of viral infection, MSCs that are activated by viral antigens elicit strong immune responses by production of proinflammatory cytokines which enhance the antiviral properties of immune cells [38]. However, after the elimination of viral pathogens, MSCs produce immunoregulatory cytokines and trophic factors that support the prevention of overactivation of immune cells, as well as the repair and regeneration of injured tissues. Thus, MSCs orchestrate antiviral immune responses that crucially contribute to the elimination of infected cells [38].

MSCs display considerable promise in the treatment of severe viral pneumonia as they possess several relevant mechanisms of action and they have demonstrated safety profile in early phase studies [40]. IL-18, which is highly expressed in influenza-A H1N1-induced severe lung injury, is a promising cytokine that can prime UC-MSCs to improve the efficacy of precision therapy against viral-induced pneumonia such as COVID-19 [41]. Respiratory syncytial virus-mediated lung injury activates and stimulates the expansion of LR-MSCs which mount a transcriptional program related to the mechanisms of repair and regeneration [39]. Soluble mediators released from avian influenza A-H5N1 virus-infected alveolar epithelial cells impair alveolar fluid clearance and protein permeability by down-regulating alveolar sodium and chloride transporter proteins [42]. Studies have shown that (1) MSCs reduce the impairment of alveolar fluid clearance induced by influenza A H5N1 infection in vitro and prevent or reduce influenza A-H5N1-associated Acute Lung Injury (ALI) in vivo, (2) transplantation of IL-18 primed human UC-MSCs has significantly enhanced the inhibition of inflammation, viral load, fibrosis, and cell apoptosis in ALIs, (3) treatment with IL-18 primed human UC-MSCs has superior inhibitory effect on T-cell exudation and proinflammatory cytokine secretion in bronchoalveolar lavage fluid, (4) in animal models, BM-MSCs exert antiviral properties and they may have therapeutic potential in the treatment of influenza virus-induced pneumonia and ALI, and (5) MSCs have been shown to enhance antiviral immune responses and protect against lethal or generalized herpes viral infections [42-44]. Additionally, exosomes derived from human UC-MSCs have a broad antiviral role in the inhibition of viral replication and provide new insights into the development of antiviral agents by bioengineering these exosomes to target respiratory viruses [45].

Stem cell-based therapies represent innovative approaches to rebuild the damaged immune system and ultimately eliminate the virus from the body [37]. The potential of MSCs to treat viral infections is still in its infancy due to the limited number of studies on the use of these cells in the treatment of virus-associated diseases [46]. In HBV-related acute or chronic liver failure, transplantation of MSCs has been shown to: improve liver function, increase survival of patients, and decrease the incidence of severe infections [47,48]. Despite that MSCs appear to have the potential to contribute to the HIV-1 reservoir, the use of MSCs derived from adipose tissues and UC in 2 CTs performed in patients with acquired immunodeficiency syndrome has not been shown to be effective in recovering immunity or aiding immune reconstitution in an immune non-responder group of patients although safety of MSC therapy has been confirmed [36,49,50].

Ten systematic reviews and meta-analyses that included 129 CTs comprising 4391 patients on the use of MSCs in the treatment of COVID-19 infection and its complications have shown the following results: (1) MSCs can reduce the mortality rates and increase overall survival in patients with COVID-19 infection; (2) MSCs can reduce the severity of COVID-19 pneumonia; (3) MSCs can induce remission of symptoms related to COVID-19 infection in the vast majority of studies included; (4) MSCs can reduce the duration of hospitalization and the requirement for invasive mechanical ventilation; (5) MSCs can improve lung function and radiological appearances in patients with COVID-19 pneumonia; and (6) MSCs can reduce the levels of inflammatory markers such as C-reactive protein and interferon-gamma in the majority of patients with severe COVID-19 infection [17,51-59]. Additionally, the included CTs have shown the safety of MSC therapy in COVID-19 infection without an increase in adverse effects [17,51-54,56-59]. In the CTs included: the commonest source of MSCs was the UC, intravenous infusion was the most common route of administration of MSCs, and the number of injections given ranged between 1 and 3 injections with variable MSC doses [17,18,52,56,58-60].

Several studies have shown that the use of MSCs and their secretome in the treatment of severe COVID-19 infections has the following beneficial effects: (1) suppression of viral replication, viral shedding, and virus-induced damage to lung epithelial cells; (2) enhancement of the generation of regulatory Tcells that are suppressed by COVID-19; (3) MSCs shift the phenotype of antigen presenting cells including dendritic cells, B-lymphocytes, and macrophages; (4) MSCs modulate the proliferation and activation of naïve and effector T-cells, natural killer cells, and mononuclear cells; (5) MSCs prevent the formation of neutrophil extracellular traps that may have deleterious effects in patients with COVID-19 pneumonia and ARDS; (6) MSCs can inhibit the cytokine storm induced by COVID-19; (7) the secretome of MSCs including ECVs and exosomes has antiviral, antibacterial, and even analgesic effects; (8) reduction in pulmonary oedema associated with ARDS in COVID-19; (9) entrapment of intravenously infused MSCs in the lungs which is an advantage in patients with COVID-19 patients having pneumonia and ARDS; (10) enhancement of tissue regeneration and promotion of endogenous repair and healing in ALI induced by COVID-19; and (11) safety and efficacy of MSCs and their products provided good manufacturing practice guidelines and quality control measures of the whole process from harvesting till delivery are strictly applied [2,61-65].

Long-term follow-up of 65 patients having severe COVID-19, included in a CT, who received MSCs showed safety, but the efficacy of MSC treatment was not significantly sustained through the end of the 2-year follow-up period [66]. However, another CT that included 100 patients with COVID-19 infection on the use of UC-MSC treatment showed that UC-MSC administration achieved a long-term benefit in the recovery of lung lesions and symptoms after 1 year of follow-up [67]. Also, a third CT that included 17 patients with severe COVID-19 who required invasive mechanical ventilation in the intensive care unit showed that UC-MSCs infusion was safe and could play an important role in the chronic phase with a reduction in post-acute sequelae reduction in critically ill COVID-19 patients [68]. Additionally, MSC secretome could offer a new therapeutic approach to treating COVID-19 fibrotic lungs through its anti-inflammatory and antifibrotic factors [69].

Mesenchymal stem cells in mycobacterial infections

Recent studies have demonstrated that: (1) MSCs are present in and around the tuberculous granulomas which contain Mycobacterium Tuberculosis (MTB) bacilli, (2) MTB uses MSCs as a niche to evade host protective immune surveillance mechanisms and establish dormancy, (3) MSCs have the ability to restrict the growth of MTB to a certain extent and may be involved in the development of TB, (4) MSCs maintain antibacterial, immunomodulatory, anti-inflammatory, and regenerative properties during the regulation of TB immune responses, (5) MSCs express a large number of ATP-Binding Cassette (ABC) efflux pumps so that dormant MTB residing in MSCs are exposed to a suboptimal dose of drugs, and (6) MSCs represent the fifth element in cell-mediated immunity that is capable of regulating immune responses during TB infection [70-75]. Hence, it is highly likely that MSCs play significant roles in orchestrating: dormancy and reactivation of MTB, evasion of host immune responses, as well as resistance to anti-TB drugs [70,74,75]. Additionally, MSCs can be used as an immune target or immunotherapy agent for the treatment of TB and may provide a screening model for the development of new drugs or vaccines for TB [71].

Transplantation of MSCs and their exosomes has been

used in the treatment of Multidrug-Resistant (MDR)-TB [73,76]. Three CTs; that included 135 patients; on the use of autologous MSCs to treat MDR-TB and extensively DR (XDR)-TB have shown the following results: (1) MSCs induced clinical and radiological improvements in 70% - 80% of patients; (2) MSC transplantation induced persistent remission and even cure in 53% - 56% of patients; and (3) the addition of autologous MSC transplantation to conventional anti-TB chemotherapy significantly enhanced the response rates in patients with MDR-TB and XDR-TB [77-79].

Mesenchymal stem cells in parasitic infections

Stem cells exert inhibitory effects on parasitic infections and they improve the functions of the tissues and organs involved [80,81]. Stem cell therapies are necessary in the treatment of parasitic infections because there are limited drug choices, and there is continuous emergence of drug resistance [80]. Stem cells including MSCs have been used in the treatment of various parasitic infections including leishmaniasis, trypanosomiasis, schistosomiasis, echinococcosis, malaria, and toxoplasmosis [80,81].

In malaria, treatment with BM-derived MSCs showed the following effects: (1) improved the clearance of parasitized red blood cells, (2) increased the regeneration of hepatocytes and Kupffer cells, (3) increased the number of astrocytes and oligodendrocytes in the brain, (4) increased survival, and (5) decreased inflammation and malaria pigment accumulation in kidneys, lungs, and spleens of experimental animals [80,82]. Additionally, BM-derived MSCs are promising in the treatment of cerebral malaria as animal studies have shown that the combination of MSCs and adjuvant antimalarial treatment: protects against vascular damage, improves depression, and increases survival [82,83]. In experimental schistosomiasis, treatment with BM-derived MSCs significantly reduced the granuloma size as well as the expression of the fibrosis factor, alpha-smooth muscle actin, in hepatic cells [83]. Treatment with MSCs showed a significant reduction in the cutaneous lesions caused by Leishmania major, while in toxoplasmosis, experimental therapy with BM-derived MSCs combined with spiramycin, pyrimethamine, and folinic acid showed a significant decrease in the number and size of tissue cysts in the brains of mice [83].

In Chagas disease, unfortunately, more success has been achieved in animal or preclinical studies than in human studies [84]. In the murine model of Chagas disease, co-transplantation of MSCs and skeletal myoblasts has been shown to be effective in decreasing ventricular dysfunction [85]. In the Chagas disease model, cardiac MSCs have demonstrated potential as they exerted a protective effect on chronic Chagasic cardiomyopathy through immunomodulation but they neither reduced fibrosis nor contributed to cardiomyocyte formation [86]. Repeated injections of granulocyte colony-stimulating factor (G-CSF), which mobilizes stem cells from the BM, decreased inflammation and fibrosis in the hearts of mice having Chagas disease [80]. Also, G-CSF overexpression in MSCs potentiated the in vivo immunosuppressive effects of MSCs in chronic Chagas disease models [87].

The rising role of extracellular vesicles and exosomes of mesenchymal stem cells in the treatment of infectious diseases

Recently, studies have demonstrated that MSC-derived ECVs are at least partially responsible for the paracrine effects of MSCs including the transfer of molecules such as proteins/peptides, mRNA, microRNA, and lipids with immunoregulatory properties to recipient cells, and may offer specific advantages over the parent MSCs due to lower immunogenicity, storage without losing function, and superior safety profile [88,89]. ECVs can achieve their valuable roles in the treatment of infectious diseases through different mechanisms including elimination of the pathogen, regulation of immunity, modulation of drug resistance, repair of tissue damage, production of antimicrobial substances, inhibition of pathogen multiplication, and activating phagocytic activity of macrophages [90,91]. ECVs of MSCs have been extensively explored in treating various infectious diseases including respiratory tract infections, urinary tract infections, wound infections, sepsis, and intestinal infections [90,91]. MSC-ECVs have been shown to mimic MSCs in alleviating sepsis and protecting against sepsis-induced organ dysfunction and hence they may serve as an alternative to whole-cell therapy [88,89]. Thus, ECVs play a key role in infectious pathogenesis and hold great promise for developing innovative treatments [90].

Recent studies have shown that MSCs-derived exosomes may improve some complications of COVID-19 infection such as cytokine storm, ARDS, and ALI by suppressing inflammatory responses and regeneration of damaged tissues, and also serve as biomarkers, nanocarriers, and vaccines for the treatment of SARS-CoV-2 virus [92,93]. Nebulization of MSC-derived exosomes has been shown to be safe and effective, and administration of exosomes at the beginning of treatment of COVID-19 pneumonia may be more beneficial [94].

Conclusion and future perspectives

MSCs have antimicrobial, anti-inflammatory, immuno-modulatory, and regenerative potentials that have enabled them to be used in the treatment of several bacterial, viral, mycobacterial, and fungal infections and their complications. The utilization of MSCs and their secretome have shown not only success and efficacy but also safety in the treatment of infections caused by MTB and COVID-19 infection and its complications in particular. Unfortunately, in MSC therapy for Chagas disease, more success has been achieved in animal or preclinical studies than in human studies.

Bioengineering of MSCs, and the use of MSCs obtained from certain sources, as well as the utilization of ECVs derived from MSCs have improved their efficacy and reduced their side effects. However, despite the progress achieved, the clinical application of MSCs in the treatment of several infectious diseases still faces real challenges that need to be resolved.

  1. Shaw TD, Krasnodembskaya AD, Schroeder GN, Zumla A, Maeurer M, O'Kane CM. Mesenchymal Stromal Cells: An Antimicrobial and Host-Directed Therapy for Complex Infectious Diseases. Clin Microbiol Rev. 2021 Dec 15;34(4):e0006421. doi: 10.1128/CMR.00064-21. Epub 2021 Oct 6. PMID: 34612662; PMCID: PMC8510528.
  2. Al-Anazi KA, Al-Anazi WK, Al-Jasser AM. Update on COVID-19 infections and the promising role of mesenchymal stem cell therapies in their management. Electronic Book. Heighten Science Publications Inc (HSPI) Published: June 23, 2020. doi: https://dx.doi.org/10.29328/ebook1002
  3. Al-Anazi KA, Al-Anazi WK, Al-Jasser AM. The rising role of mesenchymal stem cells in the treatment of various infectious complications. In: Update on Mesenchymal and Induced Pluripotent Stem Cells. Book edited by: Khalid Ahmed Al-Anazi. Intech Open. 2020. doi: 10.5772/intechopen. 91475
  4. 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 (Basel). 2022 Nov 8;9(11):662. doi: 10.3390/bioengineering9110662. PMID: 36354573; PMCID: PMC9687734.
  5. Sharma A, Chakraborty A, Jaganathan BG. Review of the potential of mesenchymal stem cells for the treatment of infectious diseases. World J Stem Cells. 2021 Jun 26;13(6):568-593. doi: 10.4252/wjsc.v13.i6.568. PMID: 34249228; PMCID: PMC8246252.
  6. Battah B. Mesenchymal stem cells: Potential role against bacterial infection. J Biosci Med. 2022; 10 (3): 97-113. doi: 10.4236/jbm.2022.103011
  7. Lombardo E, van der Poll T, DelaRosa O, Dalemans W. Mesenchymal stem cells as a therapeutic tool to treat sepsis. World J Stem Cells. 2015 Mar 26;7(2):368-79. doi: 10.4252/wjsc.v7.i2.368. PMID: 25815121; PMCID: PMC4369493.
  8. Alcayaga-Miranda F, Cuenca J, Khoury M. Antimicrobial Activity of Mesenchymal Stem Cells: Current Status and New Perspectives of Antimicrobial Peptide-Based Therapies. Front Immunol. 2017 Mar 30; 8:339. doi: 10.3389/fimmu.2017.00339. PMID: 28424688; PMCID: PMC5371613.
  9. Galderisi U, Peluso G, Di Bernardo G. Clinical Trials Based on Mesenchymal Stromal Cells are Exponentially Increasing: Where are We in Recent Years? Stem Cell Rev Rep. 2022 Jan;18(1):23-36. doi: 10.1007/s12015-021-10231-w. Epub 2021 Aug 16. PMID: 34398443; PMCID: PMC8365566.
  10. Yang G, Fan X, Liu Y, Jie P, Mazhar M, Liu Y, Dechsupa N, Wang L. Immunomodulatory Mechanisms and Therapeutic Potential of Mesenchymal Stem Cells. Stem Cell Rev Rep. 2023 Jul;19(5):1214-1231. doi: 10.1007/s12015-023-10539-9. Epub 2023 Apr 14. PMID: 37058201; PMCID: PMC10103048.
  11. Zhuang WZ, Lin YH, Su LJ, Wu MS, Jeng HY, Chang HC, Huang YH, Ling TY. Mesenchymal stem/stromal cell-based therapy: mechanism, systemic safety and biodistribution for precision clinical applications. J Biomed Sci. 2021 Apr 14;28(1):28. doi: 10.1186/s12929-021-00725-7. PMID: 33849537; PMCID: PMC8043779.
  12. Carp DM, Liang Y. Universal or Personalized Mesenchymal Stem Cell Therapies: Impact of Age, Sex, and Biological Source. Cells. 2022 Jun 30;11(13):2077. doi: 10.3390/cells11132077. PMID: 35805161; PMCID: PMC9265811.
  13. Margiana R, Markov A, Zekiy AO, Hamza MU, Al-Dabbagh KA, Al-Zubaidi SH, Hameed NM, Ahmad I, Sivaraman R, Kzar HH, Al-Gazally ME, Mustafa YF, Siahmansouri H. Clinical application of mesenchymal stem cell in regenerative medicine: a narrative review. Stem Cell Res Ther. 2022 Jul 28;13(1):366. doi: 10.1186/s13287-022-03054-0. PMID: 35902958; PMCID: PMC9330677.
  14. Fraile M, Eiro N, Costa LA, Martín A, Vizoso FJ. Aging and Mesenchymal Stem Cells: Basic Concepts, Challenges and Strategies. Biology (Basel). 2022 Nov 18;11(11):1678. doi: 10.3390/biology11111678. PMID: 36421393; PMCID: PMC9687158.
  15. Berebichez-Fridman R, Montero-Olvera PR. Sources and Clinical Applications of Mesenchymal Stem Cells: State-of-the-art review. Sultan Qaboos Univ Med J. 2018 Aug;18(3):e264-e277. doi: 10.18295/squmj.2018.18.03.002. Epub 2018 Dec 19. PMID: 30607265; PMCID: PMC6307657.
  16. Costela-Ruiz VJ, Melguizo-Rodríguez L, Bellotti C, Illescas-Montes R, Stanco D, Arciola CR, 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 Jun 6;23(11):6356. doi: 10.3390/ijms23116356. PMID: 35683035; PMCID: PMC9181542.
  17. Yang CW, Chen RD, Zhu QR, Han SJ, Kuang MJ. Efficacy of umbilical cord mesenchymal stromal cells for COVID-19: A systematic review and meta-analysis. Front Immunol. 2022 Aug 29; 13:923286. doi: 10.3389/fimmu.2022.923286. PMID: 36105796; PMCID: PMC9467457.
  18. Couto PS, Al-Arawe N, Filgueiras IS, Fonseca DLM, Hinterseher I, Catar RA, Chinnadurai R, Bersenev A, Cabral-Marques O, Moll G, Verter F. Systematic review and meta-analysis of cell therapy for COVID-19: global clinical trial landscape, published safety/efficacy outcomes, cell product manufacturing and clinical delivery. Front Immunol. 2023 Jun 21; 14:1200180. doi: 10.3389/fimmu.2023.1200180. PMID: 37415976; PMCID: PMC10321603.
  19. Chu M, Wang H, Bian L, Huang J, Wu D, Zhang R, Fei F, Chen Y, Xia J. Nebulization Therapy with Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes for COVID-19 Pneumonia. Stem Cell Rev Rep. 2022 Aug;18(6):2152-2163. doi: 10.1007/s12015-022-10398-w. Epub 2022 Jun 4. PMID: 35665467; PMCID: PMC9166932.
  20. Dilogo IH, Aditianingsih D, Sugiarto A, Burhan E, Damayanti T, Sitompul PA, Mariana N, Antarianto RD, Liem IK, Kispa T, Mujadid F, Novialdi N, Luviah E, Kurniawati T, Lubis AMT, Rahmatika D. Umbilical cord mesenchymal stromal cells as critical COVID-19 adjuvant therapy: A randomized controlled trial. Stem Cells Transl Med. 2021 Sep;10(9):1279-1287. doi: 10.1002/sctm.21-0046. Epub 2021 Jun 8. PMID: 34102020; PMCID: PMC8242692.
  21. Johnson V, Webb T, Norman A, Coy J, Kurihara J, Regan D, Dow S. Activated Mesenchymal Stem Cells Interact with Antibiotics and Host Innate Immune Responses to Control Chronic Bacterial Infections. Sci Rep. 2017 Aug 29;7(1):9575. doi: 10.1038/s41598-017-08311-4. PMID: 28851894; PMCID: PMC5575141.
  22. Pezzanite LM, Chow L, Phillips J, Griffenhagen GM, Moore AR, Schaer TP, Engiles JB, Werpy N, Gilbertie J, Schnabel LV, Antczak D, Miller D, Dow S, Goodrich LR. TLR-activated mesenchymal stromal cell therapy and antibiotics to treat multi-drug resistant Staphylococcal septic arthritis in an equine model. Ann Transl Med. 2022 Nov;10(21):1157. doi: 10.21037/atm-22-1746. PMID: 36467344; PMCID: PMC9708491.
  23. Johnson V, Chow L, Harrison J, Soontararak S, Dow S. Activated Mesenchymal Stromal Cell Therapy for Treatment of Multi-Drug Resistant Bacterial Infections in Dogs. Front Vet Sci. 2022 Jun 23; 9:925701. doi: 10.3389/fvets.2022.925701. PMID: 35812842; PMCID: PMC9260693.
  24. Javaregownda PK, Yoon JW, Jang G. Roles of mesenchymal stem cells (MSCs) in bacterial diseases. J Biomed Res. 2013: 184-194.
  25. Yang H, Xu F, Zheng X, Yang S, Ren Z, Yang J. Human Umbilical Cord Mesenchymal Stem Cells Prevent Bacterial Biofilm Formation. Biomed Res Int. 2022 Mar 3; 2022:1530525. doi: 10.1155/2022/1530525. PMID: 35281594; PMCID: PMC8913149.
  26. Laroye C, Gibot S, Reppel L, Bensoussan D. Concise Review: Mesenchymal Stromal/Stem Cells: A New Treatment for Sepsis and Septic Shock? Stem Cells. 2017 Dec;35(12):2331-2339. doi: 10.1002/stem.2695. Epub 2017 Sep 16. PMID: 28856759.
  27. Cheng Y, Cao X, Qin L. Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Novel Cell-Free Therapy for Sepsis. Front Immunol. 2020 Apr 21; 11:647. doi: 10.3389/fimmu.2020.00647. PMID: 32373121; PMCID: PMC7186296.
  28. Daniel M, Bedoui Y, Vagner D, Raffray L, Ah-Pine F, Doray B, Gasque P. Pathophysiology of Sepsis and Genesis of Septic Shock: The Critical Role of Mesenchymal Stem Cells (MSCs). Int J Mol Sci. 2022 Aug 17;23(16):9274. doi: 10.3390/ijms23169274. PMID: 36012544; PMCID: PMC9409099.
  29. Horák J, Nalos L, Martínková V, Beneš J, Štengl M, Matějovič M. Mesenchymal Stem Cells in Sepsis and Associated Organ Dysfunction: A Promising Future or Blind Alley? Stem Cells Int. 2017; 2017:7304121. doi: 10.1155/2017/7304121. Epub 2017 Sep 14. PMID: 29098010; PMCID: PMC5618761.
  30. Matthay MA, Pati S, Lee JW. Concise Review: Mesenchymal Stem (Stromal) Cells: Biology and Preclinical Evidence for Therapeutic Potential for Organ Dysfunction Following Trauma or Sepsis. Stem Cells. 2017 Feb;35(2):316-324. doi: 10.1002/stem.2551. Epub 2017 Jan 19. PMID: 27888550.
  31. McIntyre LA, Stewart DJ, Mei SHJ, Courtman D, Watpool I, Granton J, Marshall J, Dos Santos C, Walley KR, Winston BW, Schlosser K, Fergusson DA; Canadian Critical Care Trials Group; Canadian Critical Care Translational Biology Group. Cellular Immunotherapy for Septic Shock. A Phase I Clinical Trial. Am J Respir Crit Care Med. 2018 Feb 1;197(3):337-347. doi: 10.1164/rccm.201705-1006OC. PMID: 28960096.
  32. Schlosser K, Wang JP, Dos Santos C, Walley KR, Marshall J, Fergusson DA, Winston BW, Granton J, Watpool I, Stewart DJ, McIntyre LA, Mei SHJ; Canadian Critical Care Trials Group and the Canadian Critical Care Translational Biology Group. Effects of Mesenchymal Stem Cell Treatment on Systemic Cytokine Levels in a Phase 1 Dose Escalation Safety Trial of Septic Shock Patients. Crit Care Med. 2019 Jul;47(7):918-925. doi: 10.1097/CCM.0000000000003657. PMID: 30720538; PMCID: PMC6629173.
  33. Alp E, Gonen ZB, Gundogan K, Esmaoglu A, Kaynar L, Cetin A, Karakukcu M, Cetin M, Kalin G, Doganay M. The Effect of Mesenchymal Stromal Cells on the Mortality of Patients with Sepsis and Septic Shock: A Promising Therapy. Emerg Med Int. 2022 Jun 24; 2022:9222379. doi: 10.1155/2022/9222379. PMID: 35784641; PMCID: PMC9249540.
  34. Blanco NG, Machado NM, Castro LL, Antunes MA, Takiya CM, Trugilho MRO, Silva LR, Paes Leme AF, Domingues RR, Pauletti BA, Miranda BT, Silva JD, Dos Santos CC, Silva PL, Rocco PRM, Cruz FF. Extracellular Vesicles from Different Sources of Mesenchymal Stromal Cells Have Distinct Effects on Lung and Distal Organs in Experimental Sepsis. Int J Mol Sci. 2023 May 4;24(9):8234. doi: 10.3390/ijms24098234. PMID: 37175936; PMCID: PMC10179270.
  35. Thanunchai M, Hongeng S, Thitithanyanont A. Mesenchymal Stromal Cells and Viral Infection. Stem Cells Int. 2015; 2015:860950. doi: 10.1155/2015/860950. Epub 2015 Jul 29. PMID: 26294919; PMCID: PMC4532961.
  36. Kallmeyer K, Ryder MA, Pepper MS. Mesenchymal Stromal Cells: A Possible Reservoir for HIV-1? Stem Cell Rev Rep. 2022 Apr;18(4):1253-1280. doi: 10.1007/s12015-021-10298-5. Epub 2022 Jan 1. PMID: 34973144; PMCID: PMC9033703.
  37. Kandula UR, Wake AD. Promising Stem Cell therapy in the Management of HIV and AIDS: A Narrative Review. Biologics. 2022 Jul 8; 16:89-105. doi: 10.2147/BTT.S368152. PMID: 35836496; PMCID: PMC9275675.
  38. Harrell CR, Popovska Jovicic B, Djonov V, Volarevic V. Molecular Mechanisms Responsible for Mesenchymal Stem Cell-Based Treatment of Viral Diseases. Pathogens. 2021 Apr 1;10(4):409. doi: 10.3390/pathogens10040409. PMID: 33915728; PMCID: PMC8066286.
  39. Brügger M, Démoulins T, Barut GT, Zumkehr B, Oliveira Esteves BI, Mehinagic K, Haas Q, Schögler A, Rameix-Welti MA, Eléouët JF, Moehrlen U, Marti TM, Schmid RA, Summerfield A, Posthaus H, Ruggli N, Hall SRR, Alves MP. Pulmonary mesenchymal stem cells are engaged in distinct steps of host response to respiratory syncytial virus infection. PLoS Pathog. 2021 Jul 28;17(7):e1009789. doi: 10.1371/journal.ppat.1009789. PMID: 34320038; PMCID: PMC8351988.
  40. Masterson CH, Ceccato A, Artigas A, Dos Santos C, Rocco PR, Rolandsson Enes S, Weiss DJ, McAuley D, Matthay MA, English K, Curley GF, Laffey JG. Mesenchymal stem/stromal cell-based therapies for severe viral pneumonia: therapeutic potential and challenges. Intensive Care Med Exp. 2021 Dec 31;9(1):61. doi: 10.1186/s40635-021-00424-5. PMID: 34970706; PMCID: PMC8718182.
  41. Liao Y, Fu Z, Huang Y, Wu S, Wang Z, Ye S, Zeng W, Zeng G, Li D, Yang Y, Pei K, Yang J, Hu Z, Liang X, Hu J, Liu M, Jin J, Cai C. Interleukin-18-primed human umbilical cord-mesenchymal stem cells achieve superior therapeutic efficacy for severe viral pneumonia via enhancing T-cell immunosuppression. Cell Death Dis. 2023 Jan 28;14(1):66. doi: 10.1038/s41419-023-05597-3. PMID: 36707501; PMCID: PMC9883134.
  42. Chan MC, Kuok DI, Leung CY, Hui KP, Valkenburg SA, Lau EH, Nicholls JM, Fang X, Guan Y, Lee JW, Chan RW, Webster RG, Matthay MA, Peiris JS. Human mesenchymal stromal cells reduce influenza A H5N1-associated acute lung injury in vitro and in vivo. Proc Natl Acad Sci U S A. 2016 Mar 29;113(13):3621-6. doi: 10.1073/pnas.1601911113. Epub 2016 Mar 14. PMID: 26976597; PMCID: PMC4822574.
  43. Jankauskaite L, Schmoldt C, Lohmeyer J, Herold S. LSC Abstract: Murine bone marrow-derived mesenchymal stem cells exert antiviral and barrier-protective properties in influenza virus infection. Eur Respir J. 2015; 46: OA3515. doi: 10.1183/13993003.congress-2015.OA3515
  44. Klimova RR, Momotyuk ЕD, Demidova NA, Yarigina EI, Kushch AA. [Mesenchymal stem cells enhance immune response and protect mice against lethal herpes viral infection.]. Vopr Virusol. 2018;63(6):261-267. Russian. doi: 10.18821/0507-4088-2018-63-6-261-267. PMID: 30641021.
  45. Oh SJ, Lee EN, Park JH, Lee JK, Cho GJ, Park IH, Shin OS. Anti-Viral Activities of Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Against Human Respiratory Viruses. Front Cell Infect Microbiol. 2022 Apr 21; 12:850744. doi: 10.3389/fcimb.2022.850744. PMID: 35558099; PMCID: PMC9085650.
  46. Sleem A, Saleh F. Mesenchymal stem cells in the fight against viruses: Face to face with the invisible enemy. Curr Res Transl Med. 2020 Aug;68(3):105-110. doi: 10.1016/j.retram.2020.04.003. Epub 2020 May 1. PMID: 32616467; PMCID: PMC7252154.
  47. Cheng F, Huang Z, Wei W, Li Z. Mesenchymal Stem Cell Transplantation For Hepatitis B Virus-Related Acute-on-Chronic Liver Failure: A Systematic Review and Meta-Analysis. Curr Stem Cell Res Ther. 2023;18(6):834-842. doi: 10.2174/1574888X18666221214123844. PMID: 36518036.
  48. Lin BL, Chen JF, Qiu WH, Wang KW, Xie DY, Chen XY, Liu QL, Peng L, Li JG, Mei YY, Weng WZ, Peng YW, Cao HJ, Xie JQ, Xie SB, Xiang AP, Gao ZL. Allogeneic bone marrow-derived mesenchymal stromal cells for hepatitis B virus-related acute-on-chronic liver failure: A randomized controlled trial. Hepatology. 2017 Jul;66(1):209-219. doi: 10.1002/hep.29189. Epub 2017 May 27. PMID: 28370357.
  49. Wang L, Zhang Z, Xu R, Wang X, Shu Z, Chen X, Wang S, Liu J, Li Y, Wang L, Zhang M, Yang W, Wang Y, Huang H, Tu B, Liang Z, Li L, Li J, Hou Y, Shi M, Wang FS. Human umbilical cord mesenchymal stem cell transfusion in immune non-responders with AIDS: a multicenter randomized controlled trial. Signal Transduct Target Ther. 2021 Jun 9;6(1):217. doi: 10.1038/s41392-021-00607-2. PMID: 34103473; PMCID: PMC8187429.
  50. Trujillo-Rodríguez M, Viciana P, Rivas-Jeremías I, Álvarez-Ríos AI, Ruiz-García A, Espinosa-Ibáñez O, Arias-Santiago S, Martínez-Atienza J, Mata R, Fernández-López O, Ruiz-Mateos E, Gutiérrez-Valencia A, López-Cortés LF. Mesenchymal stromal cells in human immunodeficiency virus-infected patients with discordant immune response: Early results of a phase I/II clinical trial. Stem Cells Transl Med. 2021 Apr;10(4):534-541. doi: 10.1002/sctm.20-0213. Epub 2020 Dec 2. PMID: 33264515; PMCID: PMC7980217.
  51. Wang J, Shi P, Chen D, Wang S, Wang P, Feng X, Zhang L. Research Status of the Safety and Efficacy of Mesenchymal Stem Cells in the Treatment of COVID-19-Related Pneumonia: A Systematic Review and Meta-Analysis. Stem Cells Dev. 2021 Oct 1;30(19):947-969. doi: 10.1089/scd.2021.0179. PMID: 34416823.
  52. Zhang Z, Shao S, Liu X, Tong Z. Effect and safety of mesenchymal stem cells for patients with COVID-19: Systematic review and meta-analysis with trial sequential analysis. J Med Virol. 2023 Apr;95(4):e28702. doi: 10.1002/jmv.28702. PMID: 36964933.
  53. Liu Q, Ma F, Zhong Y, Wang G, Hu L, Zhang Y, Xie J. Efficacy and safety of human umbilical cord-derived mesenchymal stem cells for COVID-19 pneumonia: a meta-analysis of randomized controlled trials. Stem Cell Res Ther. 2023 May 4;14(1):118. doi: 10.1186/s13287-023-03286-8. PMID: 37143167; PMCID: PMC10159228.
  54. Taufiq H, Shaik Fakiruddin K, Muzaffar U, Lim MN, Rusli S, Kamaluddin NR, Esa E, Abdullah S. Systematic review and meta-analysis of mesenchymal stromal/stem cells as strategical means for the treatment of COVID-19. Ther Adv Respir Dis. 2023 Jan-Dec; 17:17534666231158276. doi: 10.1177/17534666231158276. PMID: 37128999; PMCID: PMC10140776.
  55. Chen L, Xu Q, Sheng F, Wang B. Efficacy and Safety of Mesenchymal Stem Cells for COVID-19 Infection: A Meta-Analysis and Systematic Review. Discov Med. 2023 Apr 1;35(175):201-207. doi: 10.24976/Discov.Med.202335175.21. PMID: 37105930.
  56. Kirkham AM, Monaghan M, Bailey AJM, Shorr R, Lalu MM, Fergusson DA, Allan DS. Mesenchymal stem/stromal cell-based therapies for COVID-19: First iteration of a living systematic review and meta-analysis: MSCs and COVID-19. Cytotherapy. 2022 Jun;24(6):639-649. doi: 10.1016/j.jcyt.2021.12.001. Epub 2022 Jan 31. PMID: 35219584; PMCID: PMC8802614.
  57. Yao W, Dong H, Qi J, Zhang Y, Shi L. Safety and efficacy of mesenchymal stem cells in severe/critical patients with COVID-19: A systematic review and meta-analysis. EClinicalMedicine. 2022 Jul 9;51:101545. doi: 10.1016/j.eclinm.2022.101545. PMID: 35844767; PMCID: PMC9270852.
  58. Javed A, Karki S, Sami Z, Khan Z, Shree A, Sah BK, Ghosh S, Saxena S. Association between Mesenchymal Stem Cells and COVID-19 Therapy: Systematic Review and Current Trends. Biomed Res Int. 2022 Jun 22; 2022:9346939. doi: 10.1155/2022/9346939. PMID: 35782071; PMCID: PMC9242780.
  59. Cao JX, You J, Wu LH, Luo K, Wang ZX. Clinical efficacy analysis of mesenchymal stem cell therapy in patients with COVID-19: A systematic review. World J Clin Cases. 2022 Sep 26;10(27):9714-9726. doi: 10.12998/wjcc.v10.i27.9714. PMID: 36186213; PMCID: PMC9516915.
  60. Nandina RQ, Eriani K, Asrizal CW, Azhar Al. Mesenchymal stem cell as a successful therapy for COVID-19 patient: Systematic review. Biosaintifica. 2022; 14 (1): 36-47. doi: 10.15294/biosaintifika.v14i1. 32367
  61. Bari E, Ferrarotti I, Saracino L, Perteghella S, Torre ML, Corsico AG. Mesenchymal Stromal Cell Secretome for Severe COVID-19 Infections: Premises for the Therapeutic Use. Cells. 2020 Apr 9;9(4):924. doi: 10.3390/cells9040924. PMID: 32283815; PMCID: PMC7226831.
  62. Rogers CJ, Harman RJ, Bunnell BA, Schreiber MA, Xiang C, Wang FS, Santidrian AF, Minev BR. Rationale for the clinical use of adipose-derived mesenchymal stem cells for COVID-19 patients. J Transl Med. 2020 May 18;18(1):203. doi: 10.1186/s12967-020-02380-2. PMID: 32423449; PMCID: PMC7232924.
  63. Liu S, Peng D, Qiu H, Yang K, Fu Z, Zou L. Mesenchymal stem cells as a potential therapy for COVID-19. Stem Cell Res Ther. 2020 May 4;11(1):169. doi: 10.1186/s13287-020-01678-8. PMID: 32366290; PMCID: PMC7197031.
  64. Zumla A, Wang FS, Ippolito G, Petrosillo N, Agrati C, Azhar EI, Chang C, El-Kafrawy SA, Osman M, Zitvogel L, Galle PR, Locatelli F, Gorman E, Cordon-Cardo C, O'Kane C, McAuley D, Maeurer M. Reducing mortality and morbidity in patients with severe COVID-19 disease by advancing ongoing trials of Mesenchymal Stromal (stem) Cell (MSC) therapy - Achieving global consensus and visibility for cellular host-directed therapies. Int J Infect Dis. 2020 Jul; 96:431-439. doi: 10.1016/j.ijid.2020.05.040. Epub 2020 May 17. PMID: 32425638; PMCID: PMC7231497.
  65. Rajarshi K, Chatterjee A, Ray S. Combating COVID-19 with mesenchymal stem cell therapy. Biotechnol Rep (Amst). 2020 May 14;26:e00467. doi: 10.1016/j.btre.2020.e00467. PMID: 32420049; PMCID: PMC7224671.
  66. Li TT, Zhang B, Fang H, Shi M, Yao WQ, Li Y, Zhang C, Song J, Huang L, Xu Z, Yuan X, Fu JL, Zhen C, Zhang Y, Wang ZR, Zhang ZY, Yuan MQ, Dong T, Bai R, Zhao L, Cai J, Dong J, Zhang J, Xie WF, Li Y, Shi L, Wang FS. Human mesenchymal stem cell therapy in severe COVID-19 patients: 2-year follow-up results of a randomized, double-blind, placebo-controlled trial. EBioMedicine. 2023 Jun; 92:104600. doi: 10.1016/j.ebiom.2023.104600. Epub 2023 May 5. PMID: 37149930; PMCID: PMC10161678.
  67. Shi L, Yuan X, Yao W, Wang S, Zhang C, Zhang B, Song J, Huang L, Xu Z, Fu JL, Li Y, Xu R, Li TT, Dong J, Cai J, Li G, Xie Y, Shi M, Li Y, Zhang Y, Xie WF, Wang FS. Human mesenchymal stem cells treatment for severe COVID-19: 1-year follow-up results of a randomized, double-blind, placebo-controlled trial. EBioMedicine. 2022 Jan; 75:103789. doi: 10.1016/j.ebiom.2021.103789. Epub 2021 Dec 25. PMID: 34963099; PMCID: PMC8709782.
  68. Rebelatto CLK, Senegaglia AC, Franck CL, Daga DR, Shigunov P, Stimamiglio MA, Marsaro DB, Schaidt B, Micosky A, de Azambuja AP, Leitão CA, Petterle RR, Jamur VR, Vaz IM, Mallmann AP, Carraro Junior H, Ditzel E, Brofman PRS, Correa A. Safety and long-term improvement of mesenchymal stromal cell infusion in critically COVID-19 patients: a randomized clinical trial. Stem Cell Res Ther. 2022 Mar 21;13(1):122. doi: 10.1186/s13287-022-02796-1. PMID: 35313959; PMCID: PMC8935270.
  69. Bari E, Ferrarotti I, Saracino L, Perteghella S, Torre ML, Richeldi L, Corsico AG. Mesenchymal Stromal Cell Secretome for Post-COVID-19 Pulmonary Fibrosis: A New Therapy to Treat the Long-Term Lung Sequelae? Cells. 2021 May 14;10(5):1203. doi: 10.3390/cells10051203. PMID: 34068958; PMCID: PMC8155949.
  70. Devi A, Pahuja I, Singh SP, Verma A, Bhattacharya D, Bhaskar A, Dwivedi VP, Das G. Revisiting the role of mesenchymal stem cells in tuberculosis and other infectious diseases. Cell Mol Immunol. 2023 Jun;20(6):600-612. doi: 10.1038/s41423-023-01028-7. Epub 2023 May 12. PMID: 37173422; PMCID: PMC10176304.
  71. Zhang X, Xie Q, Ye Z, Li Y, Che Z, Huang M, Zeng J. Mesenchymal Stem Cells and Tuberculosis: Clinical Challenges and Opportunities. Front Immunol. 2021 Jul 22; 12:695278. doi: 10.3389/fimmu.2021.695278. PMID: 34367155; PMCID: PMC8340780.
  72. 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 Jun 16;11(1):3062. doi: 10.1038/s41467-020-16877-3. PMID: 32546788; PMCID: PMC7297998.
  73. Khan A, Hunter RL, Jagannath C. Emerging role of mesenchymal stem cells during tuberculosis: The fifth element in cell mediated immunity. Tuberculosis (Edinb). 2016 Dec; 101S:S45-S52. doi: 10.1016/j.tube.2016.09.019. Epub 2016 Sep 28. PMID: 27743705.
  74. Pathak L, Das B. Initiation of Post-Primary Tuberculosis of the Lungs: Exploring the Secret Role of Bone Marrow Derived Stem Cells. Front Immunol. 2021 Jan 21; 11:594572. doi: 10.3389/fimmu.2020.594572. PMID: 33584661; PMCID: PMC7873989.
  75. 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 U S A. 2010 Dec 14;107(50):21653-8. doi: 10.1073/pnas.1007967107. Epub 2010 Dec 6. PMID: 21135221; PMCID: PMC3003090.
  76. Liu M, Wang Z, Ren S, Zhao H. Exosomes derived from mycobacterium tuberculosis-infected MSCs induce a pro-inflammatory response of macrophages. Aging (Albany NY). 2021 Apr 19;13(8):11595-11609. doi: 10.18632/aging.202854. Epub 2021 Apr 19. PMID: 33872217; PMCID: PMC8109131.
  77.  Erokhin VV, Vasil'eva IA, Konopliannikov AG, Chukanov VI, Tsyb AF, Bagdasarian TR, Danilenko AA, Lepekhina LA, Kal'sina SSh, Semenkova IV, Agaeva EV. [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. Russian. PMID: 19086127.
  78. Skrahin A, Ahmed RK, Ferrara G, Rane L, Poiret T, Isaikina Y, Skrahina A, Zumla A, Maeurer MJ. 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 Feb;2(2):108-22. doi: 10.1016/S2213-2600(13)70234-0. Epub 2014 Jan 9. PMID: 24503266.
  79. Skrahin AE, Jenkins HE, Hurevich H, Solodovnokova V, Isaikina Y, Klimuk D, Rohava Z, Skrahina A. Potential role of autologous mesenchymal stromal cells in the treatment of multidrug and extensively drug-resistant tuberculosis. Eur Respir J. 2016; 48: PA1919. doi: 10.1183/13993003. congress-2016.PA1919
  80. Elsawey AM, Etewa SE, Aboulfotouh N. Stem cell therapy research for the treatment of parasitic infections. Parasitol United J. 2022; 15 (3): 232-237. doi: 10.21608/puj.2022.156355.1185
  81. Aryamand S, Khademvatan S, Diba K, Manafpour N, Abbasi E. Stem cell therapy in the treatment of parasitic diseases. Iran J Med Microbiol. 2017; 11 (3): 01-09.
  82. Wang W, Qian H, Cao J. Stem cell therapy: a novel treatment option for cerebral malaria? Stem Cell Res Ther. 2015 Aug 8;6(1):141. doi: 10.1186/s13287-015-0138-6. PMID: 26253514; PMCID: PMC4529732.
  83. Lima MN, Oliveira HA, Fagundes PM, Estato V, Silva AYO, Freitas RJRX, Passos BABR, Oliveira KS, Batista CN, Vallochi AL, Rocco PRM, Castro-Faria-Neto HC, Maron-Gutierrez T. Mesenchymal stromal cells protect against vascular damage and depression-like behavior in mice surviving cerebral malaria. Stem Cell Res Ther. 2020 Aug 26;11(1):367. doi: 10.1186/s13287-020-01874-6. PMID: 32843073; PMCID: PMC7448996.
  84. de Carvalho KA, Abdelwahid E, Ferreira RJ, Irioda AC, Guarita-Souza LC. Preclinical stem cell therapy in Chagas Disease: Perspectives for future research. World J Transplant. 2013 Dec 24;3(4):119-26. doi: 10.5500/wjt.v3.i4.119. PMID: 24392316; PMCID: PMC3879521.
  85. Guarita-Souza LC, Carvalho KA, Woitowicz V, Rebelatto C, Senegaglia A, Hansen P, Miyague N, Francisco JC, Olandoski M, Faria-Neto JR, Brofman P. Simultaneous autologous transplantation of cocultured mesenchymal stem cells and skeletal myoblasts improves ventricular function in a murine model of Chagas disease. Circulation. 2006 Jul 4;114(1 Suppl):I120-4. doi: 10.1161/CIRCULATIONAHA.105.000646. PMID: 16820560.
  86. Silva DN, de Freitas Souza BS, Azevedo CM, Vasconcelos JF, Carvalho RH, Soares MB, Dos Santos RR. Intramyocardial transplantation of cardiac mesenchymal stem cells reduces myocarditis in a model of chronic Chagas disease cardiomyopathy. Stem Cell Res Ther. 2014 Jul 1;5(4):81. doi: 10.1186/scrt470. PMID: 24984860; PMCID: PMC4229984.
  87. Silva DN, Souza BSF, Vasconcelos JF, Azevedo CM, Valim CXR, Paredes BD, Rocha VPC, Carvalho GB, Daltro PS, Macambira SG, Nonaka CKV, Ribeiro-Dos-Santos R, Soares MBP. Granulocyte-Colony Stimulating Factor-Overexpressing Mesenchymal Stem Cells Exhibit Enhanced Immunomodulatory Actions Through the Recruitment of Suppressor Cells in Experimental Chagas Disease Cardiomyopathy. Front Immunol. 2018 Jun 25; 9:1449. doi: 10.3389/fimmu.2018.01449. PMID: 30013550; PMCID: PMC6036245.
  88. Zheng G, Huang R, Qiu G, Ge M, Wang J, Shu Q, Xu J. Mesenchymal stromal cell-derived extracellular vesicles: regenerative and immunomodulatory effects and potential applications in sepsis. Cell Tissue Res. 2018 Oct;374(1):1-15. doi: 10.1007/s00441-018-2871-5. Epub 2018 Jun 28. PMID: 29955951.
  89. Cheng Y, Cao X, Qin L. Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Novel Cell-Free Therapy for Sepsis. Front Immunol. 2020 Apr 21; 11:647. doi: 10.3389/fimmu.2020.00647. PMID: 32373121; PMCID: PMC7186296.
  90. Manzoor T, Saleem A, Farooq N, Dar LA, Nazir J, Saleem S, Ismail S, Gugjoo MB, Shiekh PA, Ahmad SM. Extracellular vesicles derived from mesenchymal stem cells - a novel therapeutic tool in infectious diseases. Inflamm Regen. 2023 Feb 28;43(1):17. doi: 10.1186/s41232-023-00266-6. PMID: 36849892; PMCID: PMC9970864.
  91. You J, Fu Z, Zou L. Mechanism and Potential of Extracellular Vesicles Derived from Mesenchymal Stem Cells for the Treatment of Infectious Diseases. Front Microbiol. 2021 Oct 26; 12:761338. doi: 10.3389/fmicb.2021.761338. PMID: 34764947; PMCID: PMC8576143.
  92. Yousefi Dehbidi M, Goodarzi N, Azhdari MH, Doroudian M. Mesenchymal stem cells and their derived exosomes to combat Covid-19. Rev Med Virol. 2022 Mar;32(2):e2281. doi: 10.1002/rmv.2281. Epub 2021 Aug 7. PMID: 34363275; PMCID: PMC8420536.
  93. Akbari A, Rezaie J. Potential therapeutic application of mesenchymal stem cell-derived exosomes in SARS-CoV-2 pneumonia. Stem Cell Res Ther. 2020 Aug 14;11(1):356. doi: 10.1186/s13287-020-01866-6. PMID: 32795359; PMCID: PMC7427273.
  94. Chu M, Wang H, Bian L, Huang J, Wu D, Zhang R, Fei F, Chen Y, Xia J. Nebulization Therapy with Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes for COVID-19 Pneumonia. Stem Cell Rev Rep. 2022 Aug;18(6):2152-2163. doi: 10.1007/s12015-022-10398-w. Epub 2022 Jun 4. PMID: 35665467; PMCID: PMC9166932.