Next Article in Journal
Diffuse Large B-Cell Lymphoma (DLBCL) in an Aged Raccoon (Procyon lotor)
Next Article in Special Issue
Antibiotic Recommendations for Treatment of Canine Stromal Corneal Ulcers
Previous Article in Journal
Oviduct Epithelial Cell-Derived Extracellular Vesicles Improve Porcine Trophoblast Outgrowth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Activated Cellular Therapy for Drug Resistant Infections: Rationale, Mechanisms, and Implications for Veterinary Medicine

1
Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
2
Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, The Ohio State University, Columbus, OH 43210, USA
3
Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Michigan State University, Lansing, MI 48824, USA
4
Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
*
Author to whom correspondence should be addressed.
Vet. Sci. 2022, 9(11), 610; https://doi.org/10.3390/vetsci9110610
Submission received: 29 September 2022 / Revised: 18 October 2022 / Accepted: 31 October 2022 / Published: 4 November 2022
(This article belongs to the Special Issue Antimicrobial Use in Companion Animals)

Abstract

:

Simple Summary

Mesenchymal stromal/stem cells have intrinsic antimicrobial properties, thus making them attractive as an alternative treatment strategy in chronic, drug-resistant bacterial infections. Recent evidence has suggested that these antimicrobial effects can be significantly enhanced by immune activation just prior to injection. This review examines the potential role for cellular therapies in treatment of drug resistant infections in veterinary medicine, drawing on insights across species and discussing the therapeutic potential of this approach overall in today’s veterinary patients.

Abstract

Antimicrobial resistance and biofilm formation both present challenges to treatment of bacterial infections with conventional antibiotic therapy and serve as the impetus for development of improved therapeutic approaches. Mesenchymal stromal cell (MSC) therapy exerts an antimicrobial effect as demonstrated in multiple acute bacterial infection models. This effect can be enhanced by pre-conditioning the MSC with Toll or Nod-like receptor stimulation, termed activated cellular therapy (ACT). The purpose of this review is to summarize the current literature on mechanisms of antimicrobial activity of MSC with emphasis on enhanced effects through receptor agonism, and data supporting use of ACT in treatment of bacterial infections in veterinary species including dogs, cats, and horses with implications for further treatment applications. This review will advance the field’s understanding of the use of activated antimicrobial cellular therapy to treat infection, including mechanisms of action and potential therapeutic applications.

1. Introduction

Selection of antibiotic resistant bacteria in both human and veterinary medicine necessitates novel therapeutic approaches for successful management. Chronic infections, particularly those involving biofilms and multi-drug resistant organisms, evade most attempts at effective treatment. Recent reports by the National Institutes of Health (NIH), National Institute of Allergy and Infectious Diseases (NIAID), Centers for Disease Control and Prevention (CDC), World Health Organization (WHO), and Natural Resources Defense Council (NRDC) reflect the magnitude of the problem in healthcare [1,2,3,4,5,6,7,8]. In 2013, the CDC reported that an estimated two million people developed antibiotic-resistant infections annually, with greater than 23,000 cases resulting in death [1]. Similarly, antimicrobial resistance has been extensively recently documented in veterinary medicine, and considered one of the most important issues threatening animal health worldwide [9]. Conventional approaches to treatment of bacterial infections (i.e., the development of new antibiotics), are not able to keep pace with the increasing incidence of multi-drug resistant infections [3].
Antimicrobial cellular therapy (ACT) represents a new approach to address the growing issue of chronic, drug-resistant infection. This approach employs living cells, mesenchymal stromal or ‘stem’ cells (MSC), to augment the activity of conventional antibiotic therapy. Recent work has focused on optimizing cellular therapeutic strategies to focus on use of ACT as an adjunctive therapy for multi-drug resistant (MDR) bacterial infections, including both acute and chronic cases, as will be discussed in this review. This work builds off the use of MSC for treatment of bacterial infections, previously reported in the lung or peritoneal cavity [10,11,12,13] and particularly in biofilms [14,15,16,17,18,19,20,21,22,23,24,25,26,27] and previous work by other groups demonstrating that pre-activation of MSC with inflammatory licensing agents enhances the antibacterial and immunomodulatory abilities of MSC which may enhance their effect in treatment of infection [16,17,24,25,26,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46]. Summary of the studies detailing the antimicrobial effects of mesenchymal stromal cell therapy in treatment of bacterial biofilms and that activation of MSC enhances their innate antibacterial and immunomodulatory effects are detailed in Table 1 and Table 2, respectively.
Several key features distinguish the current version of ACT from other forms of cellular therapy for treating infections. First, the use of allogeneic MSC that have been activated with toll or nod-like receptors prior to administration. Pre-activation takes advantage of receptors that are commonly present in inflammation and infection to enhance the migratory properties of MSC and activate host innate immune defenses against infection [16,17,19,24,25,26,28,29,30,32,33,34,36,37,38,39,41,44,45]. A second defining characteristic of this approach in ACT is the use of repeated cell infusions for optimal effect. In addition, both intravenous and local routes of delivery were explored [43]. Systemic administration ensures that activated MSC will reach sites of deep-seated infection via chemokine-mediated migration and interact fully with the host immune response to stimulate effective antibacterial immune responses. However, intra-articular administration in an equine model of septic arthritis demonstrated a beneficial effect in localized disease processes such as those isolated to synovial structures suggesting that route of administration may be tailored to the specific disease process [25]. Finally, the concurrent administration of conventional antibiotics with ACT enhances the effect in an additive or synergistic manner, which we will discuss further.
Evidence for the effectiveness of the ACT approach has been generated in both mouse models [17,24,36,45], pet dogs with spontaneous chronic, drug-resistant bacterial infections involving soft tissues and bones [26], and an induced case–control study modelling septic arthritis in horses [25]. Thus, there is compelling preclinical evidence that ACT may be an effective means of stimulating clearance of recalcitrant, drug-resistant infections. In this article, we will review the evidence supporting use of TLR agonism to improve cellular therapy in treatment of bacterial infections in murine, canine, and equine disease models and further discuss mechanisms of action by which ACT exerts an effect. Finally, we will discuss the implications of these studies in the clinical application of cellular therapy to manage patients with intractable MDR infections.

2. Principles of Cellular Therapy to Treat Bacterial Infection

2.1. Mechanisms of MSC Antimicrobial and Immunomodulatory Action

Direct antimicrobial activity of MSC from multiple species and tissue sources has been reported, primarily through secretion of antimicrobial peptides that potentiate the activity of conventional antibiotics by increasing drug permeability of bacterial cell walls [13,16,43,47,48,49,50,51,52,53,54,55,56,57]. In addition, while MSC themselves express low immunogenicity, MSC are immunologically active, suppressing inflammation associated with infection by both direct cell-to-cell contact and secreted factors [57,58,59,60,61,62,63] including immune suppressive cytokines (e.g., IL-10, TGF-ß), metabolites (e.g., IDO, PGE2, adenosine), and matrix factors (e.g., galectins) [19,57,62,64,65,66,67,68,69]. MSC secreted factors not only suppressed biofilm formation but further disrupted formed biofilms in vitro [23,70]. MSC embedded implants have previously been demonstrated to have enhanced bacterial clearance and be more resistant to biofilm formation [15]. As biofilms are a defining feature of chronic bacterial infections, including those involving bone, synovial structures, and implants [15,71,72,73], the biofilm dispersing properties displayed by MSC are key to their role in treatment of chronic infection. The rationale for and approach to ACT takes advantage of and optimizes these innate properties of MSC for enhanced treatment of MSC [31,42,57,74] (Figure 1).

2.2. Cellular Activation Techniques

The functional properties of MSCs can be modified through activation of Toll-like receptors (TLR), nucleotide-binding oligomerization domain (NOD-like receptors or NLRs), or RIG-I-like receptors (RLR) [75]. Toll-like receptors (TLRs) specifically have been recognized as regulators of stromal cell functions, including survival, differentiation, and growth [35], with thirteen different TLRs identified to date in mammalian species [35]. TLRs are expressed either on intracellular membranes of the endoplasmic reticulum, lysosomes, and endosomes (TLRs 3, 7, 8 and 9) or on the cell surface (TLRs 1, 2, 4, 5, and 6) [42]. MSC derived from multiple tissue sources and species express TLRs (e.g., TLR2, TLR3, TLR4, and TLR9), which play an important role in their regulatory effects in immune modulation and response to inflammation in infection [33,76], and signaling through TLR pathways is regulated at multiple levels from transcriptional to post-translational [42]. Furthermore, interactions between TLR pathways and micro-RNAs (miRNAs) dictate either suppression or activation of the TLR signaling and downstream responses in MSCs [42]. Differences in TLR stimuli used, culture conditions or MSC source have been shown to play a role in resultant action following MSC priming, leading to inconsistent findings reported with TLR activation of MSC [31]. MSCs activated with TLRs have been demonstrated to exhibit immunosuppressive properties through induction of indoleamine-2,3-dioxygenase-1 via protein kinase R and interferon-ß [29] and to recruit immune inflammatory cells, through upregulation of secretion of immunomodulatory cytokines (CCL5, IL1ß, IL-6, IL-8) [30]. In vivo injection of various ligands (NLR2, TLR3,4 and 5) further enhanced proliferation of MSCs, increased cloning efficiency, and affected cell differentiation [36].
Importantly, activation with different TLR ligands have resulted in differential effects [46]. For example, TLR4 activation was found to induce a pro-inflammatory phenotype in MSC, termed MSC1, whereas TLR3 activation resulted in an MSC2 phenotype with upregulation of more immunosuppressive pathways [77,78,79]. TLR3 but not TLR4 primed MSC enhanced their immune-suppressive activity again natural killer cells, through modulation of natural killer group 2D ligand major histocompatibility complex class I chain A and ULBP3 and DNAM-1 ligands, which was also found to be context dependent to the site of inflammation [34]. Ligation of TLR3 and TLR4 further inhibited MSCs’ ability to suppress T-cell proliferation by affecting Notch signaling pathways, which are transmembrane receptor proteins important in cell–cell communication, solidifying MSCs’ role in immunosuppression [28,37]. In addition, TLR4 activation can stimulate the release of cytokines, especially immunomodulatory chemokines such as MCP-1 and IL-8 that recruit monocytes and neutrophils, respectively [41]. Priming of equine MSC with both TLR3 and TLR4 increased expression of CXCL10, CCL2, and IL-6 and resulted in decreased T cell proliferation (TLR3 to a greater extent than TLR4) [39]. TLR3 agonist polyinosinic:polycytidylic acid (poly I:C) stimulation of MSC further regulated key innnate immune cells known to be important to anti-viral immunity in a time-dependent fashion where early activated MSC secrete type I interferon to enhance NK cell effector function and at later time points produce greater amounts of IL-6 and TGF-ß to induce senescence in NK cells and terminate inflammatory responses [38].
Furthermore, ligation of specific TLR agonists (eg., TLR2 versus TLR4 activation) can actually inhibit MSC migration, MSC-mediated immunosuppression, and reduce expansion of regulatory T cells, diminishing MSC potential effect in treating inflammatory disease [33]. In another study, inhibition of TLR4 resulted in reduced proliferation and osteogenic differentiation of adipose derived MSC. These findings indicate that TLR receptors also regulate cell differentiation pathways, which may be relevant in the setting of bacterial infections where multiple different TLR and NLR ligands are expressed.
In a study evaluating the effect of TLR activation of murine MSC in the treatment of pulmonary infection, activation with TLR 2, 4 and 9 resulted in significantly decreased production of pro-inflammatory cytokines IL-6 and TNF- α [17]. Finally, multiple aspects of culture techniques, including time of TLR agonist exposure, concentration of TLR agonist, and MSC concentration during cell activation have all been demonstrated to affect both the immunosuppressive and the antibacterial activity of MSC [24,44]. These studies provide some explanation for the previously conflicting reports regarding overall net effects of TLR stimulation, suggesting MSC polarization and ligand selection are important aspects to consider in application of TLR agonists to activation of MSC in clinical scenarios. Specifically, MSC polarization refers to the process by which MSCs may be polarized by downstream TLR signaling into two relatively homogeneous phenotypes previously classified as MSC1 and MSC2, providing both a mechanism by which to reduce heterogeneity in cellular populations and potentially improve efficacy of current cell-based therapies [77]. Taken together, these findings support the concept that MSCs’ immunomodulatory and antimicrobial function can be significantly upregulated just prior to injection by priming or ‘licensing’ with innate immune ligands such as TLR agonists, and that selection of these agonists can significantly impact the quality and the magnitude of the downstream pathways that are activated.
Activation of MSC with TLR ligands stimulates production of antimicrobial peptides, including lipocalin-2, hepcidin, and beta-defensin-2, and cathelicidin [11,32,48,51,80]. Stimulation of MSC with IFN- γ, as would typically be found in an inflammatory microenvironment as in bacterial infection, resulted in enhanced mRNA expression of TLR3 as well as IDO1, and increased secretion of immunomodulatory cytokines including IL-10 [81]. When Toll-like receptor (TLR) activation was compared to that of nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) ligand stimulation of MSC specifically to enhance antimicrobial properties and immunomodulation, activation with TLR3 ligand poly I:C increased bactericidal activity, suppressed biofilm formation, enhanced neutrophil bacterial phagocytosis and increased immunomodulatory cytokine secretion (MCP-1) by equine MSC compared to nonstimulated MSC and activation with other TLR and NLR agonists [24]. Of all ligands evaluated, MSCs treated with TLR3 ligand poly I:C, of all ligands evaluated, resulted in greater production of indoleamine 2,3-dioxygenase (IDO), a clinically relevant therapeutic factor, and attenuated pathology in a mouse model of dextran sodium sulfate (DSS) induced colitis [82]. In an additional in vivo mouse model of chronic wound infection, mice treated with TLR3 activated MSC demonstrated migration to the site of infection, which was mechanistically shown to be mediated in part by upregulation of CXCR4 expression [16]. For example, activated MSC migrated more efficiently to an SDF-1 stimulus in vitro, and to sites of wound infection in vivo. Thus, pre-activation with a TLR ligand such as pIC was demonstrated to augment MSC antimicrobial activity through a variety of indirect mechanisms and was moved forward in clinical studies in dogs with naturally occurring wounds and horses with septic arthritis involving multidrug resistant organisms.

2.3. Route of Administration, Dosing, and Number of Injections

Both systemic and local intraperitoneal or intrasynovial injection of MSC have resulted in successful treatment of infection in animal models [25,82,83] and supports previous studies demonstrating that priming of MSC induces population-normalizing effects that can standardize what would otherwise be heterogenous cell populations [83]. Doses of 2 × 106 cells/kg and up to 1 × 109 cell/kg, which have previously been reported as optimal for immunomodulation in humans and large animals [84], were injected intravenously in mice with chronic Staphylococcus aureus impregnated implant infections and dogs with chronic naturally occurring wounds [16]. Mechanistically, when administered systemically via intravenous administration, MSC have been shown to interact with host innate immune cells, principally neutrophils and monocytes, at multiple sites, including lungs, spleen, liver, and sites of infection [64,65,85]. For example, these effects resulted in enhanced bacterial phagocytosis, mediated by MSC-secreted cytokines such as interleukin-18 (IL-8) and stimulation of neutrophil extracellular trap (NET) formation, leading to enhanced bacterial killing and neutrophil survival [16,26,78]. Recruitment of monocytes to sites of inflammation, such as bacterial infection, is mediated by chemokine CCL2 (MCP-1) produced by MSC, which mobilizes release of inflammatory monocytes from bone marrow and recruitment to sites of high CCL2 production (i.e., infection) [34]. Once recruited to wound tissues, monocytes rapidly differentiate to macrophages; important to the mechanism of ACT, TLR-3 activated MSC induce differentiation of wound macrophages from an M1 (pro-inflammatory) to M2 (reparative) phenotype [16]. This response is consistent with the anti-inflammatory phenotype of TLR-3 activated MSC previously reported [77,78,79].
When ACT was further explored in a large animal model of septic arthritis, local administration was investigated to minimize the need for larger numbers of MSC when dose was extrapolated to increased body mass [25], with positive results in reduction of local and systemic inflammation, decreased bacterial burden within joints and improved pain scores [25]. Furthermore, in a mouse model of induced colitis, intraperitoneal but not intravenous injection of TLR3 activated MSC was found to attenuate disease severity [82]. In previous studies, local injection of MSC at sites of wound infection have not been appreciated to be as effective as systemic administration [16], indicating that further investigation and comparison of routes of administration is warranted and the optimal route for a particular clinical scenario may depend on a number of factors. These studies illustrate the pros and cons of different routes of administration depending on the size of the patient, cost considerations, and condition for and accessibility of the lesion for which MSC are being administered.
Multiple versus single administrations may further improve eradication of chronic infections, theoretically due to a cumulative impact on activation of host defenses [25]. In studies performed in pet dogs with chronic MDR infections, some animals received up to 10 MSC infusions via intravenous administration [16]. A potential concern with the use of repeated injections of allogeneic MSC is the potential for induction of harmful host adaptive immune reactions to infused MSC; however, no adverse events were seen in dogs or horses receiving multiple MSC administrations for chronic infections, which may reflect the high level of systemic and local inflammation already present in multidrug resistant infections [16,25]. Future studies may employ recently investigated techniques to reduce immunogenicity when injecting allogeneic MSC such as major histocompatibility (MHC) haplotyping and matching or TGFß2 stimulation to reduce immunogenicity to MSC-mismatched stromal cell donors [86,87]. (Table 1 and Table 2).

2.4. Combination of MSC with Antibiotics for Enhanced Bacterial Killing

Co-administration of antibiotics with activated MSC has been a key feature of ACT for optimal bactericidal effect. Based on our studies, all major classes of antibiotics including beta-lactam drugs (penicillins, cephalosporins, carbapenems), aminoglycosides, fluoroquinolones, glycopeptide (vancomycin), and cyclic lipopeptide (daptomycin) antibiotics exhibit synergistic or additive activity with MSC secreted factors in vitro [70]. In support of this concept, the most effective treatment protocol for mice with chronic biofilm infections was activated MSC in combination with antibiotics compared to antibiotics alone, or activated or non-activated MSC alone [16]. Furthermore, canine clinical studies with spontaneous MDR infections demonstrated that administration of antibiotics to which the infecting bacteria are resistant can still be combined effectively with activated MSC treatment.

3. Evidence for Antimicrobial Activity in Animal Models

3.1. Rodent Models of Infection

Multiple rodent studies have supported both the antimicrobial effects of MSC in treatment of infection at various sites (e.g., thoracic and peritoneal cavities, subcutaneous chronic implant) [17] as well as the benefits of priming of MSC in culture prior to administration [17]. Mice with Streptococcus pneumoniae pulmonary infection treated with MSC exhibited reduced myeloperoxidase activity in the lungs, decreased neutrophil number in bronchoalveolar lavage fluid and lower levels of pro-inflammatory cytokines as well as bacterial load in the lungs following treatment [17]. In this model, activation of the murine MSC with TLR agonists 2,4,9 or live S. pneumoniae bacteria resulted in reduced production of IL-6 and TNF- α [17]. Intraperitoneal administration of TLR3 polyI:C activated MSC further reduced disease severity in mice with DSS-induced colitis through enhanced immunosuppressive activity by stimulating MSCs to increase production of indoleamine 2,3-dioxygenase (IDO) [82]. MSC can also be combined with various substrates or polymers to increase immune modulation ability [88]. In an acute model of bacterial wound infection, Kudinov et al. demonstrated that the combination of proteins secreted from MSC along with chitosan gel was able to ameliorate the presence of microorganisms in the burn wound area [89].

3.2. Naturally Occurring Canine Model of Chronic Infection

Dogs represent a translational model for orthopedic implant infection in humans as they develop naturally occurring implant infections in similar body sites which involve similar bacterial pathogens and antibiotic resistance patterns as chronic infections in humans. As infections were naturally occurring, induction in laboratory species could be avoided. Therefore, using the dog as a realistic, translational chronic infection model, activated allogeneic MSC were administered repeatedly intravenously without negative side effects, and in many cases, resolved infections that had resisted prolonged treatment (i.e., weeks to months) with conventional antibiotics. The canine model also addresses key issues regarding the scalability of ACT for treatment of chronic infection, as dogs in these studies have been treated with comparable doses of activated MSC (typically 2 × 106 cells per kg body weight) that have also been used for systemic MSC infusion in humans [35,90]. Moreover, dogs as an outbred species also address the safety issue of repeated intravenous delivery of fully allogeneic MSC, as the donor source for MSC in all the dog studies reported by our group were adipose tissues of unrelated dogs [16]. Adverse events associated with multiple repeated infusions of activated canine allogeneic MSC over periods of up to six months were not observed, and clinical study animals have now been followed for at least two years with no subsequent adverse events noted.

3.3. Induced Equine Model of Septic Arthritis

The encouraging findings demonstrated with TLR activation of MSC in vitro and in murine and canine models of infection prompted further evaluation of ACT in a large animal (equine) model of septic arthritis. The equine preclinical model is a clinically and translationally relevant model for human infection for several reasons. Development of infectious arthritis as a naturally occurring disease process in horses is well-documented, their large joint volume allows for repeated collection of synovial fluid to analyze a larger number of outcome parameters and their cartilage thickness, joint volume and loading forces more closely replicates that of people than many other veterinary species [91,92,93,94,95,96]. In this work, multi-drug resistant Staphylococcal septic arthritis was treated with three intra-articular injections of TLR3-activated MSC and antibiotics or antibiotics alone. Horse pain scores, diagnostic imaging findings (ultrasound, magnetic resonance imaging), quantitative bacterial counts, systemic parameters of inflammation (neutrophil counts and acute phase marker serum amyloid A), and intra-synovial cytokine levels of pro-inflammatory cytokines interleukin-6 and interleukin-18 were improved in MSC + antibiotic treated horses and no adverse events were noted (Figure 2). These studies serve as strong evidence that the use of ACT has considerable promise as a new approach to management of chronic and/or multidrug resistant infections.

4. Discussion

Cellular therapy is emerging as a promising adjunctive therapy to combat the growing problem of drug-resistant bacterial infections and those involving biofilms, and investigation of strategies to improve potency of MSCs in an ongoing area of research [42]. While there remains an incomplete understanding of the underlying mechanisms of action of TLR agonism in ACT, as well as the demonstrated additive and synergistic effects with specific antibiotics, it is apparent from these studies that TLR-activated cellular therapy for treatment of infection is well-tolerated, effective, and can be readily implemented using allogeneic sources (i.e., bone marrow or adipose tissue derived MSC obtained from young, healthy, unrelated donors) and in a variety of chronic inflammatory disease states [74]. The site of infection also does not appear to be a limiting factor, as intravenous delivery of cells was sufficient to home to sites of infection in mice and dog models and intrasynovial injection was used to effectively treat localized infections in horses. Moreover, specific resistance patterns or bacterial strains do not seem to reduce the antimicrobial effect of MSC, as activity of ACT has been observed against a variety of different Gram-positive and -negative bacterial isolates, many displaying multiple antibiotic resistances and for which development of resistance is very different. Further characterization of the effect of TLRs in biological regulation of stromal cell function could improve MSC-based cellular immunotherapies in treatment of infection [74].
Despite promising pre-clinical studies, potential obstacles to clinical implementation of ACT still must be addressed. Regulatory pathways for approval of veterinary cellular therapies in the United States by the Food and Drug Administration (FDA) is a lengthy and expensive process, with none approved to date despite greater than ten years of development efforts. Furthermore, the primary target for the majority of cellular therapies is osteoarthritis, as the market for infections in veterinary medicine may not justify development costs. In addition, there is generally a lack of spontaneous animal models of chronic infection in which to evaluate activated cellular therapies and therefore to use for FDA approval. Finally, the use of cellular therapy specifically to treat chronic drug resistant infections was not reported until 2017 by Johnson et al., so therapy for this specific indication is relataively early in the development process. As a result, a more complete understanding of the mechanisms of action of cellular activation and optimal combinations with various antibiotics is indicated. Recent evidence suggests that long noncoding RNAs (lncRNAs) regulate a wide range of biological processes and are differentially expressed in TLR3 activated MSC, providing some framework for better understanding the molecular mechanisms by which TLR activation modulates MSCs’ functions [35]. Another potential issue is donor-to-donor MSC variability as MSCs from different genetic backgrounds have been shown to exhibit distinct antibacterial phenotypes [83], which at present has been addressed by using MSC derived from young, healthy donor animals and avoiding extensive MSC passaging. Hirakawa et al. recently demonstrated that CRISPR-based gene modulation could be used to engineer MSCs with enhanced antibacterial properties through upregulation of CD14, and further investigation of these methods is indicated [83]. The relative impact of the host immune status on response to ACT is also a potential treatment variable, which may limit improvement following ACT therapy in elderly or immunocompromised patients. The optimal number of ACT treatments has also not been established, nor is it clear which clinical parameters (i.e., biomarkers) are best suited to monitor treatment responses, or time frame at which to assess treatment impact as response may take weeks to months to manifest in the case of persistent, chronic bacterial infections. Finally, recent studies have begun to investigate the application of MSC derived exosomes as an acellular therapy capable of reparation [97], immunomodulation and drug-delivery, specifically in the context of treating sepsis, which may represent a promising future direction for anti-infective cellular therapies.

5. Conclusions

In summary, the use of activated cellular therapy to manage refractory or drug resistant bacterial infections is promising as an innovative option to augment antibiotic therapy. Further evaluation of mechanisms of action and investigation of ACT in randomized controlled clinical trials is indicated.

6. Patents

Provisional patents have been filed covering immune activated MSC technology described herein (S.D., L.P., L.C.).

Author Contributions

L.M.P., L.C., A.S., V.J. and S.W.D. contributed to conception and design and drafted the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded in part by the Charlies Shipley Family Foundation, Grayson Jockey Club Research Foundation, ACVS Zoetis Dual Training Grant, NIH/NCATS CCTSI CTSA 5TL1TR002533-02, NIH 5T32OD010437-19, Carolyn Quan and Porter Bennett, Gates Center for Regenerative Medicine (Anschutz Medical Campus, University of Colorado Denver).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank and acknowledge members of the Center for Immune and Regenerative Medicine at Colorado State University for their contributions to the antimicrobial cellular therapy work described herein.

Conflicts of Interest

The co-authors declare commercial affiliations with Validus Cellular Therapeutics (S.D. co-founder) and eQcell Inc. (L.P., S.D. advisory board and stock options).

References

  1. CDC. Antibiotic Resistance Threats in the United States. 2013. Available online: https://www.cdc.gov/drugresistance/threat-report-2013/pdf/ar-threats-2013-508.pdf (accessed on 4 April 2022).
  2. World Health Organization (WHO). Antimicrobial Resistance. 2018. Available online: https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance (accessed on 4 April 2022).
  3. Ventola, C.L. The Antibiotic Resistance Crisis: Part 1: Causes and threats. Pharm. Ther. 2015, 40, 277–283. [Google Scholar]
  4. Aslam, B.; Wang, W.; Arshad, M.I.; Khurshid, M.; Muzammil, S.; Nisar, M.A.; Alvi, R.F.; Aslam, M.A.; Qamar, M.U.; Salamat, M.K.F.; et al. Antibiotic resistance: A rundown of a global crisis. Infect. Drug Resist. 2018, 11, 1645–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Bin Zaman, S.; Hussain, M.A.; Nye, R.; Mehta, V.; Mamun, K.T.; Hossain, N. A Review on Antibiotic Resistance: Alarm Bells are Ringing. Cureus 2017, 9, e1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Cumper, C. The Antibiotic Resistance Crisis. 2018. Available online: http://eureka.criver.com/the-antibiotic-resistance-crisis/ (accessed on 8 April 2022).
  7. NRDC. Next FDA Commissioner Must Address Antibiotic Resistance Crisis. 2019. Available online: https://www.nrdc.org/media/2019/190305 (accessed on 8 April 2022).
  8. NIHMedline Plus. The End Antibiotics? 2018. Available online: https://medlineplus.gov/magazine/issues/winter18/articles/winter18pg8-11.html (accessed on 8 April 2022).
  9. Palma, E.; Tilocca, B.; Roncada, P. Antimicrobial Resistance in Veterinary Medicine: An Overview. Int. J. Mol. Sci. 2020, 21, 1914. [Google Scholar] [CrossRef] [Green Version]
  10. 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]
  11. Gupta, N.; Krasnodembskaya, A.; Kapetanaki, M.; Mouded, M.; Tan, X.; Serikov, V.; Matthay, M.A. Mesenchymal stem cells enhance survival and bacterial clearance in murine Escherichia coli pneumonia. Thorax 2012, 67, 533–539. [Google Scholar] [CrossRef] [Green Version]
  12. Krasnodembskaya, A.; Samarani, G.; Song, Y.; Zhuo, H.; Su, X.; Lee, J.-W.; Gupta, N.; Petrini, M.; Matthay, M.A. Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 302, L1003–L1013. [Google Scholar] [CrossRef] [Green Version]
  13. 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, 8, 339. [Google Scholar] [CrossRef] [Green Version]
  14. Yuan, Y.; Lin, S.; Guo, N.; Zhao, C.; Shen, S.; Bu, X.; Ye, H. Marrow mesenchymal stromal cells reduce methicillin-resistant Staphylococcus aureus infection in rat models. Cytotherapy 2014, 16, 56–63. [Google Scholar] [CrossRef]
  15. Criman, E.T.; Kurata, W.E.; Matsumoto, K.W.; Aubin, H.T.; Campbell, C.E.; Pierce, L.M. Bone Marrow–Derived Mesenchymal Stem Cells Enhance Bacterial Clearance and Preserve Bioprosthetic Integrity in a Model of Mesh Infection. Plast. Reconstr. Surg. Glob. Open 2016, 4, e751. [Google Scholar] [CrossRef]
  16. 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, 7, 9575. [Google Scholar] [CrossRef] [PubMed]
  17. Asami, T.; Ishii, M.; Namkoong, H.; Yagi, K.; Tasaka, S.; Asakura, T.; Suzuki, S.; Kamo, T.; Okamori, S.; Kamata, H. Anti-inflammatory roles of mesenchymal stromal cells during acute Streptococcus pneumoniae pulmonary infection in mice. Cytotherapy 2018, 20, 302–313. [Google Scholar] [CrossRef] [PubMed]
  18. Wood, C.R.; Al Dhahri, D.; Al-Delfi, I.; Pickles, N.A.; Sammons, R.L.; Worthington, T.; Wright, K.T.; Johnson, W.E.B. Human adipose tissue-derived mesenchymal stem/stromal cells adhere to and inhibit the growth of Staphylococcus aureus and Pseudomonas aeruginosa. J. Med. Microbiol. 2018, 67, 1789–1795. [Google Scholar] [CrossRef] [PubMed]
  19. Chow, L.; Johnson, V.; Coy, J.; Regan, D.; Dow, S. Mechanisms of Immune Suppression Utilized by Canine Adipose and Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Dev. 2017, 26, 374–389. [Google Scholar] [CrossRef] [Green Version]
  20. Bujňáková, D.; Čuvalová, A.; Čížek, M.; Humenik, F.; Salzet, M.; Čížková, D. Canine Bone Marrow Mesenchymal Stem Cell Conditioned Media Affect Bacterial Growth, Biofilm-Associated Staphylococcus aureus and AHL-Dependent Quorum Sensing. Microorganisms 2020, 8, 1478. [Google Scholar] [CrossRef]
  21. Bahroudi, M.; Bakhshi, B.; Soudi, S.; Najar-Peerayeh, S. Antibacterial and antibiofilm activity of bone marrow-derived human mesenchymal stem cells secretome against Vibrio cholerae. Microb. Pathog. 2020, 139, 103867. [Google Scholar] [CrossRef]
  22. Marx, C.; Gardner, S.; Harman, R.M.; Van de Walle, G.R. The mesenchymal stromal cell secretome impairs methicillin-resistant Staphylococcus aureus biofilms via cysteine protease activity in the equine model. Stem Cells Transl. Med. 2020, 9, 746–757. [Google Scholar] [CrossRef] [Green Version]
  23. Marx, C.; Gardner, S.; Harman, R.M.; Wagner, B.; Van de Walle, G.R. Mesenchymal Stromal Cell-Secreted CCL2 Promotes Antibacterial Defense Mechanisms Through Increased Antimicrobial Peptide Expression in Keratinocytes. Stem Cells Transl. Med. 2021, 10, 1666–1679. [Google Scholar] [CrossRef]
  24. Pezzanite, L.M.; Chow, L.; Johnson, V.; Griffenhagen, G.M.; Goodrich, L.; Dow, S. Toll-like receptor activation of equine mesenchymal stromal cells to enhance antibacterial activity and immunomodulatory cytokine secretion. Vet. Surg. 2021, 50, 858–871. [Google Scholar] [CrossRef]
  25. Pezzanite, L.M.; Chow, L.; Phillips, J.; Griffenhagen, G.M.; Moore, A.R.; Schaer, T.P.; Engiles, J.B.; Werpy, N.; Gilbertie, J.; Schnabel, L.V.; et al. TLR-activated mesenchymal stromal cell therapy and antibiotics to treat multi-drug resistant Staphyloccocal septic arthritis in an equine model. Ann. Trans. Med. 2022, in press. [Google Scholar]
  26. 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, 9, 925701. [Google Scholar] [CrossRef] [PubMed]
  27. 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, 2022, 1530525. [Google Scholar] [CrossRef] [PubMed]
  28. Liotta, F.; Angeli, R.; Cosmi, L.; Filì, L.; Manuelli, C.; Frosali, F.; Mazzinghi, B.; Maggi, L.; Pasini, A.; Lisi, V.; et al. Toll-Like Receptors 3 and 4 Are Expressed by Human Bone Marrow-Derived Mesenchymal Stem Cells and Can Inhibit Their T-Cell Modulatory Activity by Impairing Notch Signaling. Stem Cells 2008, 26, 279–289. [Google Scholar] [CrossRef] [PubMed]
  29. Opitz, C.A.; Litzenburger, U.M.; Lutz, C.; Lanz, T.V.; Tritschler, I.; Köppel, A.; Tolosa, E.; Hoberg, M.; Anderl, J.; Aicher, W.K.; et al. Toll-Like Receptor Engagement Enhances the Immunosuppressive Properties of Human Bone Marrow-Derived Mesenchymal Stem Cells by Inducing Indoleamine-2,3-dioxygenase-1 via Interferon-β and Protein Kinase R. Stem Cells 2009, 27, 909–919. [Google Scholar] [CrossRef]
  30. Romieu-Mourez, R.; Francois, M.; Boivin, M.-N.; Bouchentouf, M.; Spaner, D.E.; Galipeau, J. Cytokine Modulation of TLR Expression and Activation in Mesenchymal Stromal Cells Leads to a Proinflammatory Phenotype. J. Immunol. 2009, 182, 7963–7973. [Google Scholar] [CrossRef] [Green Version]
  31. DelaRosa, O.; Lombardo, E. Modulation of Adult Mesenchymal Stem Cells Activity by Toll-Like Receptors: Implications on Therapeutic Potential. Mediat. Inflamm. 2010, 2010, 865601. [Google Scholar] [CrossRef] [Green Version]
  32. Cassatella, M.A.; Mosna, F.; Micheletti, A.; Lisi, V.; Tamassia, N.; Cont, C.; Calzetti, F.; Pelletier, M.; Pizzolo, G.; Krampera, M. Toll-Like Receptor-3-Activated Human Mesenchymal Stromal Cells Significantly Prolong the Survival and Function of Neutrophils. Stem Cells 2011, 29, 1001–1011. [Google Scholar] [CrossRef] [Green Version]
  33. Lei, J.; Wang, Z.; Hui, D.; Yu, W.; Zhou, D.; Xia, W.; Chen, C.; Zhang, Q.; Wang, Z.; Zhang, Q.; et al. Ligation of TLR2 and TLR4 on murine bone marrow-derived mesenchymal stem cells triggers differential effects on their immunosuppressive activity. Cell. Immunol. 2011, 271, 147–156. [Google Scholar] [CrossRef]
  34. Giuliani, M.; Bennaceur-Griscelli, A.; Nanbakhsh, A.; Oudrhiri, N.; Chouaib, S.; Azzarone, B.; Durrbach, A.; Lataillade, J.-J. TLR Ligands Stimulation Protects MSC from NK Killing. Stem Cells 2014, 32, 290–300. [Google Scholar] [CrossRef]
  35. Wang, L.; Zhu, C.-Y.; Ma, D.-X.; Gu, Z.-Y.; Xu, C.-C.; Wang, F.-Y.; Chen, J.-G.; Liu, C.-J.; Guan, L.-X.; Gao, R.; et al. Efficacy and safety of mesenchymal stromal cells for the prophylaxis of chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation: A meta-analysis of randomized controlled trials. Ann. Hematol. 2018, 97, 1941–1950. [Google Scholar] [CrossRef]
  36. Gorskaya, Y.F.; Tukhvatulin, A.I.; Nesterenko, V.G. NNLR2 and TLR3, TLR4, TLR5 ligands, injected in vivo, improve after 1 h the efficiency of cloning and proliferative activity of bone marrow multipotent stromal cells and reduce the content of osteogenic multipotent stromal cells in CBA mice. Microbiol. Immunol. 2017, 163, 356–360. [Google Scholar] [CrossRef]
  37. Rashedi, I.; Gómez-Aristizábal, A.; Wang, X.-H.; Viswanathan, S.; Keating, A. TLR3 or TLR4 Activation Enhances Mesenchymal Stromal Cell-Mediated Treg Induction via Notch Signaling. Stem Cells 2017, 35, 265–275. [Google Scholar] [CrossRef] [PubMed]
  38. Petri, R.M.; Hackel, A.; Hahnel, K.; Dumitru, C.A.; Bruderek, K.; Flohe, S.B.; Paschen, A.; Lang, S.; Brandau, S. Activated Tissue-Resident Mesenchymal Stromal Cells Regulate Natural Killer Cell Immune and Tissue-Regenerative Function. Stem Cell Rep. 2017, 9, 985–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Cassano, J.M.; Schnabel, L.V.; Goodale, M.B.; Fortier, L.A. The immunomodulatory function of equine MSCs is enhanced by priming through an inflammatory microenvironment or TLR3 ligand. Vet. Immunol. Immunopathol. 2018, 195, 33–39. [Google Scholar] [CrossRef] [PubMed]
  40. Cassano, J.M.; Schnabel, L.V.; Goodale, M.B.; Fortier, L.A. Inflammatory licensed equine MSCs are chrondroprotective and exhibit enhanced immunomodulation in an inflammatory environment. Stem Cell Res. Ther. 2018, 9, 82. [Google Scholar] [CrossRef] [Green Version]
  41. Cortés-Araya, Y.; Amilon, K.; Rink, B.E.; Black, G.; Lisowski, Z.; Donadeu, F.X.; Esteves, C.L. Comparison of Antibacterial and Immunological Properties of Mesenchymal Stem/Stromal Cells from Equine Bone Marrow, Endometrium, and Adipose Tissue. Stem Cells Dev. 2018, 27, 1518–1525. [Google Scholar] [CrossRef]
  42. Abdi, J.; Rashedi, I.; Keating, A. Concise review: TLR pathway-miRNA interplay in mesenchymal stromal cells: Regulatory roles and therapeutic directions. Cells. 2018, 36, 1655–1662. [Google Scholar] [CrossRef] [Green Version]
  43. Olsen, A.; Johnson, V.; Webb, T.; Santangelo, K.S.; Dow, S.; Duerr, F.M. Evaluation of Intravenously Delivered Allogeneic Mesenchymal Stem Cells for Treatment of Elbow Osteoarthritis in Dogs: A Pilot Study. Vet. Comp. Orthop. Traumatol. 2019, 32, 173–181. [Google Scholar] [CrossRef]
  44. Kurte, M.; Vega-Letter, A.M.; Luz-Crawford, P.; Djouad, F.; Noël, D.; Khoury, M.; Carrión, F. Time-dependent LPS exposure commands MSC immunoplasticity through TLR4 activation leading to opposite therapeutic outcome in EAF. Stem Cell Res. Ther. 2020, 11, 416. [Google Scholar] [CrossRef]
  45. Aqdas, M.; Singh, S.; Amir, M.; Maurya, S.K.; Pahari, S.; Agrewala, J.N. Cumulative Signaling Through NOD-2 and TLR-4 Eliminates the Mycobacterium Tuberculosis Concealed Inside the Mesenchymal Stem Cells. Front. Cell. Infect. Microbiol. 2021, 11, 669168. [Google Scholar] [CrossRef]
  46. Aziz, S.G.-G.; Alipour, S.; Ranjbarvan, P.; Azari, A.; Babaei, G.; Golchin, A. Critical roles of TLRs on the polarization of mesenchymal stem cells for cell therapy of viral infections: A notice for COVID-19 treatment. Comp. Clin. Pathol. 2021, 30, 119–128. [Google Scholar] [CrossRef] [PubMed]
  47. Krasnodembskaya, A.; Song, Y.; Fang, X.; Gupta, N.; Serikov, V.; Lee, J.-W.; Matthay, M.A. Antibacterial Effect of Human Mesenchymal Stem Cells Is Mediated in Part from Secretion of the Antimicrobial Peptide LL-37. Stem Cells 2010, 28, 2229–2238. [Google Scholar] [CrossRef] [PubMed]
  48. Sung, D.K.; Chang, Y.S.; Sung, S.I.; Yoo, H.S.; Ahn, S.Y.; Park, W.S. Antibacterial effect of mesenchymal stem cells against Escherichia coli is mediated by secretion of beta- defensin- 2 via toll- like receptor 4 signalling. Cell. Microbiol. 2016, 18, 424–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Kang, H.-K.; Kim, C.; Seo, C.H.; Park, Y. The therapeutic applications of antimicrobial peptides (AMPs): A patent review. J. Microbiol. 2017, 55, 1–12. [Google Scholar] [CrossRef] [PubMed]
  50. Fabisiak, A.; Murawska, N.; Fichna, J. LL-37: Cathelicidin-related antimicrobial peptide with pleiotropic activity. Pharmacol. Rep. 2016, 68, 802–808. [Google Scholar] [CrossRef]
  51. Doss, M.; White, M.R.; Tecle, T.; Hartshorn, K.L. Human defensins and LL-37 in mucosal immunity. J. Leukoc. Biol. 2010, 87, 79–92. [Google Scholar] [CrossRef]
  52. Chakraborty, K.; Ghosh, S.; Koley, H.; Mukhopadhyay, A.K.; Ramamurthy, T.; Saha, D.R.; Mukhopadhyay, D.; Roychowdhury, S.; Hamabata, T.; Takeda, Y.; et al. Bacterial exotoxins downregulate cathelicidin (hCAP-18/LL-37) and human beta-defensin 1 (HBD-1) expression in the intestinal epithelial cells. Cell Microbiol. 2008, 10, 2520–2537. [Google Scholar] [CrossRef]
  53. Armitage, A.E.; Eddowes, L.A.; Gileadi, U.; Cole, S.; Spottiswoode, N.; Selvakumar, T.A.; Ho, L.-P.; Townsend, A.R.M.; Drakesmith, H. Hepcidin regulation by innate immune and infectious stimuli. Blood 2011, 118, 4129–4139. [Google Scholar] [CrossRef] [Green Version]
  54. Michels, K.; Nemeth, E.; Ganz, T.; Mehrad, B. Hepcidin and Host Defense against Infectious Diseases. PLoS Pathog. 2015, 11, e1004998. [Google Scholar] [CrossRef] [Green Version]
  55. Kaundal, U.; Bagai, U.; Rakha, A. Immunomodulatory plasticity of mesenchymal stem cells: A potential key to successful solid organ transplantation. J. Transl. Med. 2018, 16, 31. [Google Scholar] [CrossRef] [Green Version]
  56. 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] [PubMed] [Green Version]
  57. Russell, K.A.; Garbin, L.C.; Wong, J.M.; Koch, T.G. Mesenchymal Stromal Cells as Potential Antimicrobial for Veterinary Use—A Comprehensive Review. Front. Microbiol. 2020, 11, 606404. [Google Scholar] [CrossRef] [PubMed]
  58. Kyurkchiev, D.; Bochev, I.; Ivanova-Todorova, E.; Mourdjeva, M.; Oreshkova, T.; Belemezova, K.; Kyurkchiev, S. Secretion of immunoregulatory cytokines by mesenchymal stem cells. World J. Stem Cells 2014, 6, 552–570. [Google Scholar] [CrossRef] [PubMed]
  59. Gonzalez-Rey, E.; Gonzalez, M.A.; Varela, N.; O’Valle, F.; Hernandez-Cortes, P.; Rico, L.; Büscher, D.; Delgado, M. Human adipose-derived mesenchymal stem cells reduce inflammatory and T cell responses and induce regulatory T cells in vitro in rheumatoid arthritis. Ann. Rheum. Dis. 2010, 69, 241–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Davies Lindsay, C.; Heldring, N.; Kadri, N.; Le Blanc, K. Mesenchymal stromal cell secretion of programmed death-1 ligands regulates t cell mediated immunosuppression. Stem Cells 2016, 35, 766–776. [Google Scholar] [CrossRef]
  61. Kadle, R.L.; Abdou, S.A.; Villarreal-Ponce, A.; Soares, M.A.; Sultan, D.L.; David, J.A.; Massie, J.; Rifkin, W.J.; Rabbani, P.; Ceradini, D.J. Microenvironmental cues enhance mesenchymal stem cell-mediated immunomodulation and regulatory T-cell expansion. PLoS ONE 2018, 13, e0193178. [Google Scholar] [CrossRef]
  62. Zhou, K.; Guo, S.; Tong, S.; Sun, Q.; Li, F.; Zhang, X.; Qiao, Y.; Liang, G. Immunosuppression of human adipose-derived stem cells on t-cell subsets via the reduction of nf-kappab activation mediated by pd-l1/pd-1 and gal-9/tim-3 pathways. Stem Cells Dev. 2018, 27, 1191–1202. [Google Scholar] [CrossRef]
  63. Rozenberg, A.; Rezk, A.; Boivin, M.-N.; Darlington, P.J.; Nyirenda, M.; Li, R.; Jalili, F.; Winer, R.; Artsy, E.A.; Uccelli, A.; et al. Human Mesenchymal Stem Cells Impact Th17 and Th1 Responses Through a Prostaglandin E2 and Myeloid-Dependent Mechanism. Stem Cells Transl. Med. 2016, 5, 1506–1514. [Google Scholar] [CrossRef] [Green Version]
  64. Takeda, K.; Webb, T.L.; Ning, F.; Shiraishi, Y.; Regan, D.P.; Chow, L.; Smith, M.J.; Ashino, S.; Guth, A.M.; Hopkins, S.; et al. Mesenchymal Stem Cells Recruit CCR2+ Monocytes to Suppress Allergic Airway Inflammation. J. Immunol. 2018, 200, 1261–1269. [Google Scholar] [CrossRef] [Green Version]
  65. Brandau, S.; Jakob, M.; Bruderek, K.; Bootz, F.; Giebel, B.; Radtke, S.; Mauel, K.; Jäger, M.; Flohe, S.; Lang, S. Mesenchymal Stem Cells Augment the Anti-Bacterial Activity of Neutrophil Granulocytes. PLoS ONE 2014, 9, e106903. [Google Scholar] [CrossRef] [Green Version]
  66. de Witte, S.F.H.; Luk, F.; Parraga, J.M.S.; Gargesha, M.; Merino, A.; Korevaar, S.S.; Shankar, A.S.; O'Flynn, L.; Elliman, S.J.; Roy, D.; et al. Immunomodulation by therapeutic mesenchymal stromal cells (MSC) is triggered through phagocytosis of MSC by monocytic cells. Stem Cells 2018, 36, 602–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Salami, F.; Tavassoli, A.; Mehrzad, J.; Parham, A. Immunomodulatory effects of mesenchymal stem cells on leukocytes with emphasis on neutrophils. Immunobiology 2018, 223, 786–791. [Google Scholar] [CrossRef] [PubMed]
  68. Kim, S.-N.; Lee, H.-J.; Jeon, M.-S.; Yi, T.; Song, S.U. Galectin-9 is involved in immunosuppression mediated by human bone marrow-derived clonal mesenchymal stem cells. Immune Netw. 2015, 15, 241–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Yagi, H.; Soto-Gutierrez, A.; Parekkadan, B.; Kitagawa, Y.; Tompkins, R.G.; Kobayashi, N.; Yarmush, M.L. Mesenchymal Stem Cells: Mechanisms of Immunomodulation and Homing. Cell Transplant. 2010, 19, 667–679. [Google Scholar] [CrossRef] [Green Version]
  70. 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] [Green Version]
  71. Bjarnsholt, T. The role of bacterial biofilms in chronic infections. APMIS 2013, 121, 1–58. [Google Scholar] [CrossRef]
  72. Bjarnsholt, T.; Ciofu, O.; Molin, S.; Givskov, M.; Høiby, N. Applying insights from biofilm biology to drug development—Can a new approach be developed? Nat. Rev. Drug Discov. 2013, 12, 791–808. [Google Scholar] [CrossRef]
  73. Walton, K.D.; Lord, A.; Kendall, L.V.; Dow, S.W. Comparison of 3 real-time, quantitative murine models of staphylococcal biofilm infection by using in vivo bioluminescent imaging. Comp. Med. 2014, 64, 25–33. [Google Scholar]
  74. Shirjang, S.; Mansoori, B.; Solali, S.; Hagh, M.F.; Shamsasenjan, K. Toll-like receptors as a key regulator of mesenchymal stem cell function: An up-to-date review. Cell. Immunol. 2017, 315, 1–10. [Google Scholar] [CrossRef]
  75. Chen, X.; Zhang, Z.; Zhou, H.; Zhou, G. Characterization of mesenchymal stem cells under the stimulation of Toll-like receptor agonists. Dev. Growth Differ. 2014, 56, 233–244. [Google Scholar] [CrossRef]
  76. Saeedi, P.; Halabian, R.; Fooladi, A.A.I. Antimicrobial effects of mesenchymal stem cells primed by modified LPS on bacterial clearance in sepsis. J. Cell. Physiol. 2018, 234, 4970–4986. [Google Scholar] [CrossRef] [PubMed]
  77. Waterman, R.S.; Tomchuck, S.L.; Henkle, S.L.; Betancourt, A.M. A New Mesenchymal Stem Cell (MSC) Paradigm: Polarization into a Pro-Inflammatory MSC1 or an Immunosuppressive MSC2 Phenotype. PLoS ONE 2010, 5, e10088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Ward, C.L.; Sanchez, C.J., Jr.; Pollot, B.E.; Romano, D.R.; Hardy, S.K.; Becerra, S.C.; Rathbone, C.R.; Wenke, J.C. Soluble factors from biofilms of wound pathogens modulate human bone marrow-derived stromal cell differentiation, migration, angiogenesis, and cytokine secretion. BMC Microbiol. 2015, 15, 75. [Google Scholar] [CrossRef] [PubMed]
  79. Hsu, S.-C.; Wang, L.-T.; Yao, C.-L.; Lai, H.-Y.; Chan, K.-Y.; Liu, B.-S.; Chong, P.; Lee, O.K.-S.; Chen, H.-W. Mesenchymal stem cells promote neutrophil activation by inducing IL-17 production in CD4+ CD45RO+ T cells. Immunobiology 2013, 218, 90–95. [Google Scholar] [CrossRef]
  80. Pezzanite, L.; Chow, L.; Griffenhagen, G.; Dow, S.; Goodrich, L. Impact of Three Different Serum Sources on Functional Properties of Equine Mesenchymal Stromal Cells. Front. Vet. Sci. 2021, 8, 634064. [Google Scholar] [CrossRef]
  81. Deng, J.; Li, D.; Huang, X.; Li, W.; Zhao, F.; Gu, C.; Shen, L.; Cao, S.; Ren, Z.; Zuo, Z.; et al. Interferon-y enhances the immunosuppressive ability of canine bone marrow-derived mesenchymal stem cells by activating the TLR3-dependent IDO/kynurenine pathway. Mol. Biol. Rep. 2022, 49, 8337–8347. [Google Scholar] [CrossRef] [PubMed]
  82. Ryu, D.-B.; Lim, J.-Y.; Lee, S.-E.; Park, G.; Min, C.-K. Induction of Indoleamine 2,3-dioxygenase by Pre-treatment with Poly(I:C) May Enhance the Efficacy of MSC Treatment in DSS-induced Colitis. Immune Netw. 2016, 16, 358–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hirakawa, M.P.; Tjahjono, N.; Light, Y.K.; Celebi, A.N.; Celebi, N.N.; Chintalapudi, P.; Butler, K.S.; Branda, S.S.; Krishnakumar, R. Upregulation of CD14 in mesenchymal stromal cells accelerates lipopolysaccharide-induced response and enhances antibacterial properties. iScience 2022, 25, 103759. [Google Scholar] [CrossRef] [PubMed]
  84. Schallmoser, K.; Rohde, E.; Reinisch, A.; Bartmann, C.; Thaler, D.; Drexler, C.; Obenauf, A.; Lanzer, G.; Linkesch, W.; Strunk, D. Rapid Large-Scale Expansion of Functional Mesenchymal Stem Cells from Unmanipulated Bone Marrow Without Animal Serum. Tissue Eng. Part C Methods 2008, 14, 185–196. [Google Scholar] [CrossRef] [PubMed]
  85. Pedrazza, L.; Cunha, A.A.; Luft, C.; Nunes, N.K.; Schimitz, F.; Gassen, R.B.; Breda, R.V.; Donadio, M.V.F.; de Souza Wyse, A.T.; Pitrez, P.M.C.; et al. Mesenchymal stem cells improves survival in LPS-induced acute lung injury acting through inhibition of NETs formation. J. Cell. Physiol. 2017, 232, 3552–3564. [Google Scholar] [CrossRef] [PubMed]
  86. Berglund, A.K.; Long, J.M.; Robertson, J.B.; Schnabel, L.V. TGF-ß2 reduces the cell-mediated immunogenicity of equine MHC-mismatched bone marrow-derived mesenchymal stem cells without altering immunomodulatory properties. Front. Cell Dev. Biol. 2021, 9, 628382. [Google Scholar] [CrossRef] [PubMed]
  87. Rowland, A.L.; Miller, D.; Berglund, A.; Schnabel, L.V.; Levine, G.J.; Antczak, D.F.; Watts, A.E. Cross-matching of allogeneic mesenchymal stromal cells eliminates recipient immune targeting. Stem Cells Transl. Med. 2021, 10, 694–710. [Google Scholar] [CrossRef] [PubMed]
  88. Arifka, M.; Wilar, G.; Elamin, K.M.; Wathoni, N. Polymeric Hydrogels as Mesenchymal Stem Cell Secretome Delivery System in Biomedical Applications. Polymers 2022, 14, 1218. [Google Scholar] [CrossRef] [PubMed]
  89. Kudinov, V.A.; Artyushev, R.I.; Zurina, I.M.; Lapshin, R.D.; Snopova, L.B.; Mukhina, I.V.; Grinakovskaya, O.S.; Saburina, I.N. Anntimicrobial and regenerative effects of placental multipotent mesenchymal stromal cell secretome-based chitosan gel on infected burns in rats. Pharmceuticals 2021, 14, 1263. [Google Scholar] [CrossRef] [PubMed]
  90. Fisher, S.A.; Cutler, A.; Doree, C.; Brunskill, S.J.; Stanworth, S.J.; Navarrete, C.; Girdlestone, J. Mesenchymal stromal cells as treatment or prophylaxis for acute or chronic graft-versus-host disease in haematopoietic stem cell transplant (HSCT) recipients with a haematological condition. Cochrane Database Syst. Rev. 2019, 2019, CD009768. [Google Scholar] [CrossRef]
  91. Frisbie, D.D.; Cross, M.W.; McIlwraith, C.W. A comparative study of articular cartilage thickness in the stifle of animal species used in human pre-clinical studies compared to articular cartilage thickness in the human knee. Vet. Comp. Orthop. Traumatol. 2006, 19, 142–146. [Google Scholar] [CrossRef] [Green Version]
  92. McIlwraith, C.W.; Fortier, L.A.; Frisbie, D.D.; Nixon, A.J. Equine Models of Articular Cartilage Repair. Cartilage 2011, 2, 317–326. [Google Scholar] [CrossRef] [Green Version]
  93. McIlwraith, C.W.; Frisbie, D.D.; Kawcak, C.E. The horse as a model of naturally occurring osteoarthritis. Bone Jt. Res. 2012, 1, 297–309. [Google Scholar] [CrossRef]
  94. Chu, C.R.; Szczodry, M.; Bruno, S. Animal Models for Cartilage Regeneration and Repair. Tissue Eng. Part B Rev. 2010, 16, 105–115. [Google Scholar] [CrossRef]
  95. Reesink, H.; Watts, A.; Mohammed, H.; Jay, G.; Nixon, A. Lubricin/proteoglycan 4 increases in both experimental and naturally occurring equine osteoarthritis. Osteoarthr. Cartil. 2017, 25, 128–137. [Google Scholar] [CrossRef] [Green Version]
  96. Frisbie, D.D.; Kawcak, C.E.; Trotter, G.W.; Powers, B.E.; Walton, R.M.; McIlwraith, C.W. Effects of triamcinolone acetonide on an in vivo equine osteochondral fragment exercise model. Equine Vet. J. 1997, 29, 349–359. [Google Scholar] [CrossRef] [PubMed]
  97. Wu, X.; Jin, S.; Ding, C.; Wang, Y.; He, D.; Liu, Y. Mesenchymal Stem Cell-Derived Exosome Therapy of Microbial Diseases: From Bench to Bed. Front. Microbiol. 2022, 12, 804813. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Immune mechanisms for antimicrobial properties of MSC against biofilms. Direct antimicrobial activity of MSC via secreted factors including antimicrobial peptides and indirect immunomodulatory activity of MSC are illustrated. Directly, cationic antimicrobial peptides (e.g., cathelicidin, lipocalin-2, ß-defensin 2), induce damage to bacterial membranes or alter bacterial function either directly or indirectly. Indirectly, MSC activate host immune cells, modulate local inflammation and induce angiogenesis and fibrogenesis, targeting several different cell types including T cells, macrophages, neutrophils, and dendritic cells. This activity is primarily mediated by up-regulation or inhibition of immunomodulatory cytokines and chemokines that in turn augment the immune system either to a pro-inflammatory or an anti-inflammatory state.
Figure 1. Immune mechanisms for antimicrobial properties of MSC against biofilms. Direct antimicrobial activity of MSC via secreted factors including antimicrobial peptides and indirect immunomodulatory activity of MSC are illustrated. Directly, cationic antimicrobial peptides (e.g., cathelicidin, lipocalin-2, ß-defensin 2), induce damage to bacterial membranes or alter bacterial function either directly or indirectly. Indirectly, MSC activate host immune cells, modulate local inflammation and induce angiogenesis and fibrogenesis, targeting several different cell types including T cells, macrophages, neutrophils, and dendritic cells. This activity is primarily mediated by up-regulation or inhibition of immunomodulatory cytokines and chemokines that in turn augment the immune system either to a pro-inflammatory or an anti-inflammatory state.
Vetsci 09 00610 g001
Figure 2. Evaluation of TLR poly I:C activated bone marrow derived MSC therapy in an equine model of multi-drug resistant USA300 methicillin resistant Staphylococcus aureus [25]; original unpublished images presented with permission from the authors]. Representative images of horses at day 7 following intra-articular inoculation of the left tarsocrural joint treated with three intra-articular injections of (A) MSC and antibiotics, or (B) antibiotics alone. Quantitative bacterial cultures were significantly reduced in horses treated with (C) MSC and antibiotics versus (D) antibiotics alone. Synovial fluid parameters serum amyloid A, lactate, and inflammatory biomarkers IL-6 and IL-18 were significantly improved in horses treated with MSC and antibiotics (left) versus antibiotics alone (right) (D).
Figure 2. Evaluation of TLR poly I:C activated bone marrow derived MSC therapy in an equine model of multi-drug resistant USA300 methicillin resistant Staphylococcus aureus [25]; original unpublished images presented with permission from the authors]. Representative images of horses at day 7 following intra-articular inoculation of the left tarsocrural joint treated with three intra-articular injections of (A) MSC and antibiotics, or (B) antibiotics alone. Quantitative bacterial cultures were significantly reduced in horses treated with (C) MSC and antibiotics versus (D) antibiotics alone. Synovial fluid parameters serum amyloid A, lactate, and inflammatory biomarkers IL-6 and IL-18 were significantly improved in horses treated with MSC and antibiotics (left) versus antibiotics alone (right) (D).
Vetsci 09 00610 g002
Table 1. Summary of studies demonstrating efficacy of (MSC) and conditioned medium (MSC-CM) in treatment of bacterial biofilms.
Table 1. Summary of studies demonstrating efficacy of (MSC) and conditioned medium (MSC-CM) in treatment of bacterial biofilms.
InvestigatorReferenceSpeciesCulture Conditions or LesionCell SourceCell DoseProtocolRoute of AdministrationOutcome ParametersMain Findings
Yuan et al. (2014)[14]RatSubcutaneous infection MRSABone marrow2 × 107, 2 × 106,Dosed daily for 4 dosesIntravenousQuantitative culturesMSC reduced bacterial colonies.
or 2 × 105 cells/rat Immunoassays cytokinesMSC reduced cytokine expression (IL1, IL6, IL10, CCL5).
Criman et al. (2016)[15]RatSubcutaneous E.coliBone marrow7.5 × 105 MSC/meshMSC seeded meshesSeeded in meshesMicrobiologic mesh evaluationAugmentation of bioprosthetic materials with MSC enhanced
inoculated meshes vs non-seeded meshes Histologic mesh evaluationresistance to bacterial infection.
Johnson et al. (2017)[16]MurineStaphylococcus aureusAdipose1 × 106 cells/injectionTLR-3 poly I:C activated or notIntravenousIVIS luminescence imagingActivated MSC co-administered with antibiotics was most
implant infection model with or without antibiotics to determine bacterial burdeneffective to reduce bacterial bioburden.
Dosed every 3 days, 3 doses Wound tissue histology
CanineNaturally occurring woundsAdipose2 × 106 cells/kgTLR-3 poly I:C activated + antibioticsIntravenousQuantitative culturesRepeated MSC injection resulted in clearance of bacteria
Dosed every 2 weeks, 3 doses Clinical signsand wound healing.
Phone follow-up
Asami et al. (2018)[17]MurineStreptococcus pneumoniaeBone marrow1 × 106 cells/injectionOnce1 hour after bacterial inoculationIntravenousBacteria bronchoalveolar lavageMSC-CM modulates TNFα, IL-6, IL-10 after
pulmonary infection Myeloperoxidase activity assaystimulation with TLR2, TLR4, TLR9 ligands.
Bichinchoninic acid protein assayMSC-CM suppresses CXCL1, CXCL2 production
Histopathologic examinationafter stimulation with TLR2 and TLR9 ligands.
MSC IV decreased total cells, neutrophils, and
myeloperoxidase activity during pulmonary infection.
MSC IV decreased BALF cytokine levels TNFα, IL-6,
IFN-γ, CCL2, GM-CSF during pulmonary infection.
Wood et al. (2018)[18]HumanIn vitro Staphylococcus aureus,AdiposeN/A In vitroScanning electron microscopyMSC inhibited P. aeruginosa biofilm formation
Pseudomonas co-culture Colony forming unitsdue to bacterial adhesion, engulfment/phagocytosis
Biofilm assayand secretion of antibacterial factors.
Chow et al. (2019)[19]HumanStaphylococcus aureus
In vitro biofilm assayBone marrowN/ATLR and Nod-like receptor agonistsIn vitroLive/dead biofilms confocal microscopyMSC secreted factors disrupted MRSA biofilm formation.
Mouse mesh implant model 1 × 106 cells/injectionTLR-3 poly I:C activated with antibioticsIntravenousbacterial density via IVIS live imagingActivated MSC treatment decreases bacterial bioburden
dosed every 3 days for 4 doses in mouse chronic biofilm infection model.
Bujnakova et al. (2020)[20]CanineIn vitro biofilmBone marrowN/AIn vitro coculture S. aureus, E.coli biofilmsIn vitroDisc diffusion testMSC-CM inhibited biofilm formation and quorum sensing.
Staphylococcus aureus Spectrophotometric crystal violet assay
Escherichia coli Bioluminescence assay
Bahroudi et al. (2020)[21]HumanIn vitro Vibrio choleraeBone marrowN/AMSC secretome cocultureIn vitroPlate crystal violet assayMSC secretome prevented biofilm formation
co-culture with MSC secretome V. cholerae 1:8 to 1:128 of Vibrio cholerae in a dose-dependent manner.
Marx et al. (2020)[22]EquineIn vitro Pseudomonas,Peripheral bloodN/AIn vitro co-culture with PseudomonasIn vitroProtease arrayMSC secretome inhibits biofilm formation and mature
Staphylococcus biofilms and Staphylococcus biofilms Confocal microscopy biofilm compositionbiofilms of Pseudomonas and Staphylococcus spp.
Western blot analysisMSC secrete cysteine proteases that destabilize MRSA
biofilms increasing efficacy of antibiotics.
Marx et al. (2021)[23]EquineEx vivo equine skinPeripheral bloodN/AIn vitro co-culture MSC-CMIn vitro explantImmunofluorescence activityMSC decreased MRSA viability in mature biofilms.
biofilm explant model with MRSA and MSSA Biofilm live/dead stainingEquine MSCs secrete CCL2 that increased antimicrobial
peptide secretion by equine keratinocytes.
Pezzanite et al. (2021)[24]EquineIn vitro MRSA biofilm assaysBone marrowN/ATLR-3, TLR-4 NOD activated MSCIn vitro biofilmsBactericidal activityMSC stimulation TLR3 poly I:C suppressed biofilm formation
Neutrophil bacterial phagocytosisenhanced neutrophil phagocytosis
Cytokine analysisincreased MCP-1 secretion,
Antimicrobial peptide secretionenhanced antimicrobial peptide production.
Pezzanite et al. (2022)[25]EquineIn vivo MRSA septic arthritisBone marrow20 × 106 cells/jointTLR-3 poly I:C activated MSCIntra-articularClinical pain scoringActivated MSC therapy resulted in improved pain scores,
Quantitative bacterial culturesultrasound and MRI scoring, quantittative bacterial counts,
Complete blood countssystemic neutrophil and serum amyloid A,
Dosed every 3 days for 3 doses Cytokines synovial fluid, plasmasynovial fluid lactate and serum amyloid A
Imaging (radiographs, ultrasound, MRI)synovial fluid IL-6 and IL-18.
Macroscopic joint scoring
Histologic changes
Johnson et al. (2022)[26]CanineNaturally occurring chronicAdipose2 × 106 cells/kgTLR-3 poly I:C activated with antibioticsIntravenousQuantitative culturesRepeated delivery of activated allogeneic MSC resulted
multidrug resistant infections Dosed every 2 weeks for 3 doses Clinical signsin infection clearance and wound healing.
Phone follow-up
Yang et al. (2022)[27]HumanPseudomonas aeruginosaUmbilical cordN/AIn vitro co-culture, 8 MSC concentrationsIn vitro biofilmsTitration MSC concentrationAntibacterial peptides from MSC affected biofim formation
inoculated tracheal tubes Anti-biofilm experimentby downregulating polysaccharide biosynthesis
Bacterial motility assayprotein which correlated to MSC concentration.
DNA microarray experiment
Table 2. Summary of studies demonstrating evidence that activation of MSC enhances their innate antibacterial and immunomodulatory properties.
Table 2. Summary of studies demonstrating evidence that activation of MSC enhances their innate antibacterial and immunomodulatory properties.
InvestigatorReferenceSpeciesCulture Conditions or LesionCell SourceCell DoseProtocolRouteOutcome ParametersMain Findings
Liotta et al. (2008)[28]HumanIn vitro TLR activationBone marrowN/ATLR-3 poly I:C or TLR-4 LPS activationIn vitroFlow cytometric evaluationBM-MSCs expressed high levels TLR3 and 4 which induce nuclear factor k- activity, IL6, IL8, CXCL10
T-cell co-culture MSC differentiation assaysLigation TLR3 and TLR4 on MSCs inhibited ability of MSC to suppress T-cell proliferation without
T-cell proliferation assaysinfluencing immunophenotype or differentiation potential
ELISA cytokines/chemokines analysisTLR-triggering was related to impaired Notch receptor signaling in T cells
IDO activity measuresTLR3 and TLR4 expression on MSCs provide effective mechanisms to block immunosuppressive activities
Confocal microscopyand restore efficient T-cell response to infection such as viruses or Gram-negative bacteria
Quantitative analysis NFK- translocation
RNA extraction and rtPCR
Opitz et al. (2009)[29]HumanIn vitro co-cultureBone marrowN/AMSC T-cells in mixed leukocyte reactionsIn vitroKaryotype analysis of MSCTLR ligation activates innate and adaptive immune response pathways to protect against pathogens
MSC with T-cells TLR-3 poly I:C or TLR-4 LPS activation Flow cytometric analysis MSCTLR expressed on human bm-MSC enhanced immunosuppressive phenotype of MSC
Mixed leukocyte reactionsImmnunosuppression mediated by TLR was dependent on production of IDO1
Quantitative rt-PCRInduction of IDO1 by TLR involved autocrine interferon signaling loop which depended on protein kinase R
Liquid chromatography
Western blot analysis, siRNA
ELISA cell culture supernatants
Romieu-mourez et al. (2009)[30]HumanIn vitro activationBone marrowN/ATLR-3 poly I:C or TLR-4 LPS activationIn vitroFlow cytometric analysisHuman MSC and macrophages expressed TLR3 and TLR4 at comparable levels
cytokines, TLR agonists real-time RT-PCRTLR-mediated activation of MSC resulted in production inflammatory mediators IL-1, IL-6, IL-8/CXCL8, CCL5
Immunoblot analysisIFN priming combined with TLR activation increases immune responses induced by Ag-presenting MSC
Growth response to TNF-α, IFN-α, IFN-γTLR activation resulted in inflammatory site attracting innate immune cells
Immune effector infiltration analysis
Neutrophil chemotaxis assay
Cassatella et al. (2011)[32]HumanIn vitro activatedBone marrowN/ATLR-3 poly I:C or TLR-4 LPS activationIn vitroCytofluorometric analysisTLR-3 MSC activation enhanced anti-apoptosis of neutrophils more than TLR-4
MSC neutrophil coculture ELISA immunoassaysTLR-3 and TLR-4 activation enhanced respiratory burst ability and CD11b expression by PMN
Respiratory burst cytochrome C reductionTLR-3 activation effects mediated by IL-6, IFN- and GM-CSF
TLR-4 activation effects mediated by GM-CSF
Lei et al. (2011)[33]MurineIn vitro TLR activationBone marrowN/ATLR-2 or TLR-4 activationIn vitroMSC migrationTLR2 ligation (but not TLR4) inhibited MSC migration, MSC mediated immunosuppression on allo-MLR,
Allogeneic mixed lymphocyte reactionand reduced MSC mediated expansion of Treg cells
Induction Treg cellTLR2 activation induced lower CXCL10 mRNA and protein expressions
TLR2 and TLR4 had different effects on immunomodulatory capacity of MSC
Giuliani et al. (2014)[34]HumanIn vitro MSC NK cell cocultureBone marrowN/ATLR-3 or TLR-4 activationIn vitroFlow cytometry CD107 degranulationTLR primed MSC are more resistant than unprimed MSC to IL-2 activated NK-induced killing
Embryonic NK cell MSC coculture ELISA culture supernatantsTLR-primed MSC modulated naturall killer group 2D ligands MHC class I chain A, ULBP3, DNAM-1 ligands
Chromium release assayMSC adapt their immunobehavior in inflammatory context, decreasing susceptibility to NK killing
TLR3 but not TL4 primed MSC enhance suppressive functionns against NK cells
Johnson et al. (2017)[16]MurineStaphylococcus aureusAdipose1 × 106 cells/TLR-3 poly I:C activation +/- antibioticsIntravenousBacterial burden IVIS imagingActivated MSC co-administered with antibiotics was most effective to reduce bacterial bioburden
implant infection model /injectiondosed every 3 days for 3 doses Wound tissue histology
CanineNaturally occurring woundsAdipose2 × 106 cells/kgTLR-3 poly I:C activated with antibioticsIntravenousQuantitative culturesClearance of bacteria and wound healing following repeated IV injection
dosed every 2 weeks for 3 doses Clinical signs, Phone follow-up
Gorskaya et al. (2017)[36]MurineIntraperitoneal injectionBone marrowNLR/TLR ligandsNLR2 and TLR (LPS, flagellin, CpG, poly I:C)IntraperitonealEfficiency bone marrow MSC colony formationNLR, TLR and S. typhimurium antigenic complex increase efficiency of MSC cloning and content by 1 hr
NLR, TLR, S. typhimurium 10 µg/mouseand S. typhimurium antigenic complex
Rashedi et al. (2017)[37]HumanIn vitro activation TLR ligandsBone marrowN/ATLR-3, TLR-4 effect on MSC Treg inductionIn vitroMSC, CD4+ lymphocyte co-culture assaysTLR3/4 activation MSC enhanced Treg generation in CD4+ lymphocyte/MSC cultures
Gene and protein expression analysisTLR3/4 activation augmented Treg induction via Notch pathway
Flow cytometric analysis
Quantification cytokines culture medium
Petri et al. (2017)[38]HumanIn vitro coculture TLR-3Nasal mucosaN/ATLR-3 activated MSC effect on NK cellsIn vitroELISA immunoassaysEarly time points TLR3-activated MSC secrete type I interferon to enhance NK cell effector function
TLR-3 activated Flow cytometric analysisLater time points NK cell function limited by TGF- and IL-6
MSCs and NK cells Surface/intracellular stainingFeedback regulatory NK cells to MSCs promote survival, proliferation, pro-angiogenic properties
Cytotoxicity assays
Degranulation assays
NK cell proliferation assays
MSC invasion and proliferation assays
Cassano et al. (2018)[39]EquineIn vitro co-culture TLR ligandsBone marrowN/ATLR-3 or TLR-4 activationIn vitroT-cell proliferation via flow cytometryTLR3/4 priming increased MSC expression IL6, CCL2, CXCL10
MSC co-culture inflammatory macrophages Macrophage RNA gene expressionTLR3/4 priming or exposure to inflammatory macrophages enhanced immunomodulatory function
Suppression T-cell proliiferation assay demonstrated by decreased T-cell proliferation
Cortes-Araya et al. (2018)[41]EquineIn vitro comparison MSC tissue sourcesEndometriumN/ATLR-4 primed MSC versus unprimedIn vitroAntimicrobial peptide immunocytochemistryLipocalin-2 was expressed at higher levels in EM-MSC than AD or BMD
In vitro activation with TLR4 ligandAdipose Cytokine secretion via ELISATLR-4 stimulated lipocalin-2 production by all three cell types
Bone marrow Gene expression analysesTLR-4 induced expression IL-6, IL-8, MCP-1, chemokine ligand-5, TLR4 by all three cell types
Asami et al. (2018)[17]MurineIn vitro activation with TLR ligandsBone marrow1 × 106 cells1 injection 1 hour after bacterial inoculationIntravenousBacteria bronchoalveolar lavageMSC-CM modulates TNFα, IL-6, IL-10 after
Streptococcus pneumoniae /injection Myeloperoxidase activity assaystimulation with TLR2, TLR4, TLR9 ligands.
pulmonary infection Bichinchoninic acid protein assayMSC-CM suppresses CXCL1, CXCL2 production
Histopathologic examinationafter stimulation with TLR2 and TLR9 ligands.
MSC IV decreased total cells, neutrophils, and
myeloperoxidase activity during pulmonary infection.
MSC IV decreased BALF cytokine levels TNFα, IL-6,
IFN-γ, CCL2, GM-CSF during pulmonary infection.
Chow et al. (2019)[19]HumanIn vitro Staphylococcus aureus biofilm assayBone marrowN/AComparison TLR, NLR receptor agonistsIn vitroLive/dead biofilms via confocal microscopyMSC secreted factors disrupted MRSA biofilm formation
Mice with mesh implant biofilm animal model 1 × 106 cellsTLR-3 poly I:C activated with antibioticsIntravenousbacterial density by IVIS live imagingActivated MSC treatment decreases bacterial bioburden in mouse chronic biofilm infection model
/injectionDosed every 3 days for 4 doses
Kurte et al. (2020)[44]MurineIn vitro splenocyte and MSC and TcellBone marrowN/A In vitroQuantitative real-time PCRTime dependent LPS activation regulate IL6 and iNOS expression in MSCs.
and MSC co-cultures SubcutaneousFlow cytometryImmunosuppressive activity of MSCs on T cell proliferation depends on time dependent LPS activation.
Murine autoimmune encephalomyelitis (EAE) Immunosuppression assayLong exposure to LPS enhances MSC therapeutic potential in EAE.
Treg, Th17, Th1 differentiation assayTLR4 expression involved in immunosuppressive capacity of MSCs in vitro.
Thelper analysis in treated mouse lymph nodesTLR4 inhibition disrupts capacity of MSCs to inhibit Th1 and Th17 cells in vitro.
TLR4 deficiency reduces therapeutic effect of MSCs in EAE.
Aqdas et al. (2021)[45]MurineIn vitro co-culture MSC withBone marrowN/ATLR-4 or NOD-2 activated MSCIn vitroCytokine secretion ELISA (IL-6, IL-10, IL-12, TNF-α)TLR4/NOD-2 augmented pro-inflammatory cytokine secretion.
Mycobacterium tuberculosis (Mtb) RT-qPCR (IL-6, IL-12, IL-10, iNOS, TNF-α, TGF-)TLR4/NOD-2 co-localized Mtb in lysosomes.
Phenotypic charactization of MSC markersTLR4-NOD-2 induced autophagy.
Evaluation MSC differentiationTLR4-NOD-2 enhanced NF-κ activity via p38 MAPK.
Bacterial load determination post-infectionTLR4-NOD-2 reduced intracellular Mtb survival.
Bacterial tracking into autolysosomesTriggering TLR4-NOD-2 pathway may be future immunotherapy.
Pezzanite et al. (2021)[24]EquineIn vitro MRSA biofilm assaysBone marrowN/ATLR-3, TLR-4 and NOD activated MSCIn vitroBactericidal activityMSC stimulation with TLR3 poly I:C suppressed biofilm formation, enhanced neutrophil phagocytosis,
Neutrophil bacterial phagocytosisincreased MCP-1 secretion and enhanced antimicrobial peptide cathetlicidin production
Cytokine analysis
Antimicrobial peptide secretion
Johnson et al. (2022)[26]CanineNaturally occurring chronicAdipose2 × 106 cells/kgTLR-3 poly I:C activated with antibioticsIntravenousQuantitative culturesRepeated delivery of activated allogeneic MSC resulted in infection clearance and wound healing
multidrug resistant infections dosed every 2 weeks for 3 doses Clinical signs, Phone follow-up
Pezzanite et al. (2022)[25]EquineMRSA inoculated septic arthritisBone marrow20 × 106 cells/jointTLR-3 poly I:C activated MSCIntra-articularClinical pain scoringActivated MSC therapy resulted in improved pain scores, ultrasound and MRI scoring, quantitative
dosed every 3 days for 3 doses Quantitative bacterial culturesbacterial counts, systemic neutrophil and serum amyloid A, and synovial fluid lactate, serum
Complete blood counts
Cytokine analyses (blood, synovial fluid)
Imaging (radiographs, ultrasound, MRI)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pezzanite, L.M.; Chow, L.; Strumpf, A.; Johnson, V.; Dow, S.W. Immune Activated Cellular Therapy for Drug Resistant Infections: Rationale, Mechanisms, and Implications for Veterinary Medicine. Vet. Sci. 2022, 9, 610. https://doi.org/10.3390/vetsci9110610

AMA Style

Pezzanite LM, Chow L, Strumpf A, Johnson V, Dow SW. Immune Activated Cellular Therapy for Drug Resistant Infections: Rationale, Mechanisms, and Implications for Veterinary Medicine. Veterinary Sciences. 2022; 9(11):610. https://doi.org/10.3390/vetsci9110610

Chicago/Turabian Style

Pezzanite, Lynn M., Lyndah Chow, Alyssa Strumpf, Valerie Johnson, and Steven W. Dow. 2022. "Immune Activated Cellular Therapy for Drug Resistant Infections: Rationale, Mechanisms, and Implications for Veterinary Medicine" Veterinary Sciences 9, no. 11: 610. https://doi.org/10.3390/vetsci9110610

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