Sepsis is a life-threatening syndrome resulting in shock and organ dysfunction stemming from a microbial infection. Sepsis has a mortality of 40% and is implicated in half of all in-hospital deaths. The host immune response to microbial infection is critical, with early-phase sepsis characterized by a hyperinflammatory immune response, whereas the later phase of sepsis is often complicated by suppression. Sepsis has no treatment, and management remains supportive.

Stem cells constitute exciting potential therapeutic agents for sepsis. In this review, we examine the rationale for stem cells in sepsis, focusing on mesenchymal stem/stromal cells, which currently demonstrate the greatest therapeutic promise. We examine the preclinical evidence base and evaluate potential mechanisms of action of these cells that are important in the setting of sepsis. We discuss early-phase clinical trials and critically appraise translational barriers to the use of mesenchymal stem/stromal cells in patients with sepsis.

Sepsis is a life-threatening syndrome resulting in shock and multiple organ dysfunction as a consequence of microbial infection.1  Both pathogen and host factors influence the clinical presentation, severity, and ultimately patient outcome, including the nature and virulence of the microbial pathogen, which drives tissue invasion and toxin production, and the health status and comorbidities of the patient, which influence the host response. Patients frequently present with fever, shock, and dysfunction of one or more organs, including the lungs (acute respiratory distress syndrome), kidneys (acute kidney injury), brain (confusion, delirium, or coma), liver, and cardiovascular system (shock or myocardial dysfunction).

Infections of the lungs, abdominal cavity, urinary tract, and soft tissue constitute the most common sources of sepsis.2 Escherichia coli, Klebsiella spp, and Pseudomonas aeruginosa constitute the dominant Gram-negative pathogens, whereas Staphylococcus aureus and Streptococcus pneumoniae are the most common Gram-positive pathogens isolated.3  Recently, a global study of 14,000 critically ill patients found that 62% of isolates were Gram-negative, whereas 47% were Gram-positive and 19% were fungal.4  Fungal infections are an increasing source of severe sepsis, although in one third of cases the causative organism is not determined.

Sepsis exerts a significant socioeconomic impact and is now the leading cause of critical illness globally.5,6  In 2011, sepsis was responsible for more than $20 billion (5.2%) of hospital costs7  and a quarter of a million estimated deaths in the United States annually.8  The reported incidence of sepsis is increasing,9,10  perhaps because of changing patient demographics, with advanced age, more comorbidities, impaired immunity, and increasing clinician diagnosis and recognition of sepsis all playing a role.11  Sepsis has an overall mortality of 40%8,12  and may cause half of all in-hospital deaths in the United States.13  Furthermore, long-term follow-up studies demonstrate that sepsis survivors continue to have a higher mortality in the 5 yr after sepsis.14  In addition, survivors of sepsis endure long-term psychologic, cognitive, and physical impairments.15 

The host immune response, specifically the loss of immune homeostasis induced by the pathogen, is of critical importance to the initiation, evolution, and outcome from sepsis (fig. 1).16  Patients in the early phases (hours to days) of sepsis present with fever, shock, and multiorgan failure, as well as evidence of a hyperinflammatory innate immune response. Pathogen-associated molecular patterns, which originate from microorganisms, are specific molecular signatures recognized as foreign to the host, and they bind to pattern recognition receptors expressed on innate immune cells and initiate and drive this initial hyperinflammatory phase.17  Pattern recognition receptor activation generates diverse proinflammatory molecule expression including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-2, IL-6, IL-8, and interferon (IFN)-γ, as well as antiinflammatory cytokines such as IL-10.18  This process is further driven by the release of damage-associated molecular patterns from injured tissues and cells.17  This exuberant production of proinflammatory cytokines and other soluble mediators, coupled with the demonstration that injection of these mediators into animals could recapitulate some of the effects seen in sepsis, led to the concept of the “cytokine storm” as being responsible for early sepsis-related multiple organ failure and death.19  Key advances have occurred in the management of patients in the early phase of sepsis, with earlier recognition facilitating prompt broad-spectrum antimicrobial therapy,20  aggressive source control,8  and goal-directed resuscitation, all contributing to improved outcome and a reduction in mortality.

Fig. 1.

The adaptive and innate host immune responses to sepsis. Patients in the early phases of sepsis present with fever, shock, and multiorgan failure, as well as evidence of a hyperinflammatory innate immune response. Patients surviving this phase may recover or progress to a later phase of sepsis resulting in a more immunosuppressed profile, characterized by functional deficits of the immune system and by superinfection and poor outcome. Adapted and reprinted by permission from Macmillan Publishers Ltd: Hotchkiss et al.,16  Nature Review Immunology, copyright 2013.

Fig. 1.

The adaptive and innate host immune responses to sepsis. Patients in the early phases of sepsis present with fever, shock, and multiorgan failure, as well as evidence of a hyperinflammatory innate immune response. Patients surviving this phase may recover or progress to a later phase of sepsis resulting in a more immunosuppressed profile, characterized by functional deficits of the immune system and by superinfection and poor outcome. Adapted and reprinted by permission from Macmillan Publishers Ltd: Hotchkiss et al.,16  Nature Review Immunology, copyright 2013.

Close modal

Patients surviving the early phases of sepsis regain immune homeostasis, clear their infection, and recover, or they transition into a protracted immunosuppressive phase where sepsis persists16,21,22  (fig. 1). The proportion of patients who enter this later phase of sepsis is increasing, in part due to the advances in early supportive care reducing early deaths, as well as changes in the patient population, which is now older8  and has more comorbidities that render them immunosuppressed. Seventy percent of sepsis deaths now occur in this phase,16  which is characterized by opportunistic pathogen superinfections,23  latent viral reactivation,24  and evidence for profound immunosuppression.21,25 

Although fewer patients die in the early hyperinflammatory phase of sepsis, the increasing numbers of people experiencing severe sepsis, coupled by the failure to improve outcomes from the later phases of sepsis, means that the mortality burden of sepsis continues to increase.5,6,8,12,13  There are no therapies that directly modify the pathophysiology and injury mechanisms underlying sepsis. The focus of research over the last four decades has been on suppressing the early proinflammatory response to sepsis.19  To date, there have been more than 40 unsuccessful clinical trials of agents that reduce pathogen recognition and/or block proinflammatory cytokines and/or inflammation-signaling pathways.26,27 

A number of important insights have emerged from these efforts to find a therapy for sepsis. First, the traditional paradigm of sepsis as a hyperinflammatory disorder that led to the testing of interventions to suppress the immune response is likely an oversimplification, as discussed above. Second, given the complexity of the host response to sepsis, inhibition of a single mediator, however important to the injury process, is unlikely to be effective. Third, the timing of therapeutic interventions may be important. Although steroids can attenuate the early inflammatory response, these drugs have been demonstrated to worsen later immune suppression and increase mortality.28  In contrast, encouraging results have been reported from early-phase studies of immune stimulation strategies to reverse specific immune defects in late sepsis, such as administration of granulocyte macrophage colony stimulating factor29  and interferon-β1a,30  suggesting that the optimal therapeutic approach may vary considerably depending on the stage of sepsis. Fourth, sepsis is a heterogeneous disease, and identification of sepsis subphenotypes or endotypes, as has been demonstrated recently for acute respiratory distress syndrome (ARDS),31  may allow for focusing of therapeutic interventions on specific sepsis subpopulations more likely to benefit.

An additional concern is the ongoing emergence of pathogens resistant to multiple antimicrobial therapies. Taken together with the failure to date of drug therapy trials, these concerns suggest a need to consider alternative therapeutic approaches, aimed at attenuating the proinflammatory response while enhancing host immune function and tissue reparative capacity. Stem cells constitute an emerging therapeutic candidate that might meet these requirements and consequently are emerging as potential therapeutic agents for sepsis.

Stem cells (regardless of age of donor or source tissue) are undifferentiated cells with the capacity to self-renew and/or generate more than one differentiated functional daughter cell type. There is a hierarchy of “stemness,” from pluripotent cells to multipotent cells and to progenitor cells, where the capacity to differentiate into different cell lineages is progressively reduced. Hematopoietic stem cells used for treatment of blood disorders, for example, are pluripotent cells that generate platelets, erythrocytes, and a wide variety of leukocyte types. Another important classification is in relation to their tissue source, that is, whether they are derived from embryonic or adult tissues and, in the latter case, which specific tissues they originate from. The cell type for which there is the most interest as a therapy for sepsis at present is mesenchymal stem/stromal cells (MSCs). These cells have multiple potential advantages, including their convenient isolation from multiple adult tissues and relatively easy culture expansion, which make them strong therapeutic candidates in patients with sepsis. There are exciting preclinical data supporting their use, and early-phase clinical trials are in progress. Consequently, we focus on MSCs in this review.

The therapeutic potential of MSCs for sepsis is supported by several factors. First, MSCs are relatively immune privileged (low expression of cell-surface human leukocyte antigen class I and II molecules), they do not induce a classical cytotoxic T cell (rejection) response, and they can therefore be used as an allogeneic therapy without the need for immunosuppression. Second, they modulate diverse aspects of the host immune response. Although trials of agents that directly inhibit aspects of the immune response to sepsis have been unsuccessful, MSCs exert a more complex profile of immune effects. Importantly, MSCs may reprogram the immune system to reduce host tissue damage while preserving the immune response to microorganisms. Third, MSCs may enhance tissue repair and restoration after sepsis,32–34  restoring endothelial barrier function, mediated partly by secretion of factors that enhance resolution of tissue injury. Fourth, sepsis and septic shock frequently progress to dysfunction and failure of multiple organs. MSCs may decrease injury and/or restore function in diverse organs, including the liver, kidneys, heart, and lungs. Fifth, MSCs may directly enhance host bactericidal capacity by increasing macrophage bacterial phagocytosis and killing35  and increasing secretion of antimicrobial peptides.36  Sixth, MSCs are well studied in clinical trials, with a growing safety record in patients. Concerns regarding the potential for long-term effects are mitigated by the fact that they disappear from the tissues within days of administration, although their effects often outlast their residence time in the tissues.

Pulmonary Sepsis

Initial studies demonstrated the potential for MSC therapy to decrease injury after pulmonary endotoxin instillation in murine models.37  This, and subsequent studies in this model, provided important mechanistic insights, elucidating the role of MSC-secreted mediators, including keratinocyte growth factor,38  TNF-α–induced protein-6, and lipoxin A439  in modulating the immune response to endotoxin (reducing TNF-α and macrophage inflammatory protein-2 and increasing IL-10 concentrations) and in promoting injury resolution and repair. The MSC secretome and MSC-derived microvesicles,40  as well as embryonic stem cell–derived MSCs,41  also effectively attenuated endotoxin-induced injury.40  Mitochondrial transfer from MSCs to the pulmonary epithelium appears important in reducing the severity of injury caused by pulmonary endotoxin instillation42  (fig. 2).

Fig. 2.

Factors secreted from mesenchymal stem/stromal cells (MSCs) of importance for reducing severity of pneumonia and systemic sepsis. Exo = exosomes; GM-CSF = granulocyte-macrophage colony stimulating factor; KGF = keratinocyte growth factor; LXA4 = lipoxin A4; Mf = macrophages; MIP2 = macrophage inflammatory protein 2; Mt = mitochondria; Mv = microvesicles; PGE2 = prostaglandin E2; RvD1 = resolving D1; TNF = tumor necrosis factor.

Fig. 2.

Factors secreted from mesenchymal stem/stromal cells (MSCs) of importance for reducing severity of pneumonia and systemic sepsis. Exo = exosomes; GM-CSF = granulocyte-macrophage colony stimulating factor; KGF = keratinocyte growth factor; LXA4 = lipoxin A4; Mf = macrophages; MIP2 = macrophage inflammatory protein 2; Mt = mitochondria; Mv = microvesicles; PGE2 = prostaglandin E2; RvD1 = resolving D1; TNF = tumor necrosis factor.

Close modal

MSCs also demonstrate efficacy in relevant preclinical models of bacterial pneumonia (table 1) induced by Gram-negative organisms such as Escherichia coli,43–45 Klebsiella pneumonia,46  and Pseudomonas aeuroginosa.47,48  In the early phases of lung sepsis, several groups including ours have shown that human MSCs derived from bone marrow and umbilical cord tissues reduce E. coli lung injury, decreasing lung bacterial load, enhancing lung function, and reducing mortality in rodent, murine, and ovine models.44,49–53  The potential for MSCs to attenuate pneumonia induced by Gram-positive organisms, including Staphylococcus aureus and Streptococcus pneumoniae, has also been demonstrated.48  MSCs exert antimicrobial effects against methicillin-resistant S. aureus in a rodent model of pouch infection54  and in an infected wound model55  and can directly inhibit the growth of S aureus.56  Microvesicles secreted by MSCs are also effective in attenuating bacterial pneumonia in mice via mechanisms including enhanced macrophage phagocytosis, mediated in part through the expression of keratinocyte growth factor and cyclooxygenase-2 messenger RNA (mRNA) in the injured alveoli.53 

Table 1.

Selected Preclinical Studies Examining Mechanisms of Action of MSCs in Pneumonia Sepsis

Selected Preclinical Studies Examining Mechanisms of Action of MSCs in Pneumonia Sepsis
Selected Preclinical Studies Examining Mechanisms of Action of MSCs in Pneumonia Sepsis

Systemic Sepsis

MSCs demonstrate efficacy in several preclinical systemic sepsis models (table 2). MSCs from the bone marrow,41  adipose tissue,57  and macrophages cocultured with adipose tissue MSCs58  decreased systemic endotoxemia-induced lung injury, attenuated renal cell apoptosis, and decreased multiorgan injury in rodents. Bone marrow MSCs significantly reduced cytokine and chemokine (IL-1β, -6, -10, Chemokine [C-C motif] ligand-5 [CCL-5], and TNF-α) concentrations and improved survival after cecal ligation and puncture in mice, a key preclinical model of abdominal polymicrobial sepsis.35,59  MSCs maintained their efficacy when administered 6 h after cecal ligation and puncture-induced polymicrobial sepsis in mice.35  Bone marrow MSCs attenuated murine sepsis–induced kidney injury by decreasing the proinflammatory response and enhancing macrophage phagocytosis, with reductions in renal mRNA levels of IL-6, IL-17, TNF-α, IFN-γ, Chemokine (C-X-C motif) ligand (CXCL)-1, CXCL-2, CXCL-5, CCL-2 and CCL-3.60,61  The effect was also seen in methicillin-resistant S. aureus systemic infection, with reduced bacterial load and expression of cytokines and chemokines.54  In a genome-wide microarray analysis of septic animals, MSCs decreased transcription of proinflammatory genes while increasing transcription of genes relating to tissue repair and endothelial integrity and maintaining transcriptional pathways responsible for cellular bioenergetics62  (fig. 3). MSCs can also reduce the cytokine response induced by Staphylococcal enterotoxin B in mice, although it did not increase survival in this model.63 

Table 2.

Selected Preclinical Studies Examining Mechanisms of Action of MSCs in Systemic Sepsis

Selected Preclinical Studies Examining Mechanisms of Action of MSCs in Systemic Sepsis
Selected Preclinical Studies Examining Mechanisms of Action of MSCs in Systemic Sepsis
Fig. 3.

Organ-wide mesenchymal stem/stromal cell (MSC) effect on immune cells. MSCs exert beneficial effects on organ systems through their interaction with cells of the innate and adaptive immune systems, subsequent controlled tissue inflammation, and vascular integrity. Enhanced endothelial repair, through mitochondrial transfer, allows for tissue inflammatory milieu gatekeeping. M2-like macrophages enhance complement activation, increase microbe phagocytosis and clearance, and increase targeted neutrophil recruitment to injured tissue via CXCL2. Neutrophil extracellular traps release enhanced phagocytosis along with MSC-secreted antimicrobial peptides. The targeted control of microbial invasion is further influenced by inflammasome/caspase-1 deactivation, downregulation of GRK2 and PI3K, reduced secretion of TNF-α and iNOS, and an increased regulatory T-lymphocyte presence. ECM = extracellular matrix; GRK2 = G protein–coupled receptor kinase 2; IL-10 = interleukin-10; iNOS = inducible nitric oxide synthase; KGF = keratinocyte growth factor; MSC = mesenchymal stem cell; NETS = neutrophil extracellular traps; NK = natural killer cell; PGE2 = prostaglandin-E2; PI3K = phosphatidylinositol-4,5-bisphosphate 3-kinase; TEM = transendothelial migration; Treg = regulatory T cell.

Fig. 3.

Organ-wide mesenchymal stem/stromal cell (MSC) effect on immune cells. MSCs exert beneficial effects on organ systems through their interaction with cells of the innate and adaptive immune systems, subsequent controlled tissue inflammation, and vascular integrity. Enhanced endothelial repair, through mitochondrial transfer, allows for tissue inflammatory milieu gatekeeping. M2-like macrophages enhance complement activation, increase microbe phagocytosis and clearance, and increase targeted neutrophil recruitment to injured tissue via CXCL2. Neutrophil extracellular traps release enhanced phagocytosis along with MSC-secreted antimicrobial peptides. The targeted control of microbial invasion is further influenced by inflammasome/caspase-1 deactivation, downregulation of GRK2 and PI3K, reduced secretion of TNF-α and iNOS, and an increased regulatory T-lymphocyte presence. ECM = extracellular matrix; GRK2 = G protein–coupled receptor kinase 2; IL-10 = interleukin-10; iNOS = inducible nitric oxide synthase; KGF = keratinocyte growth factor; MSC = mesenchymal stem cell; NETS = neutrophil extracellular traps; NK = natural killer cell; PGE2 = prostaglandin-E2; PI3K = phosphatidylinositol-4,5-bisphosphate 3-kinase; TEM = transendothelial migration; Treg = regulatory T cell.

Close modal

Viral Infection

In vitro studies demonstrate that human MSCs exhibit antiviral effects, such as inhibition of virus-specific CD8+ T-cell proliferation,64  which is mediated through indoleamine 2,3-dioxygenase secretion.64,65  MSCs did not attenuate moderate66  or severe67  H1N1 (PR8 strain) influenza-induced lung injury in mice. In the study by Gotts et al.,66  MSCs modestly reduced viral load but failed to reduce disruption of the alveolar–capillary barrier in mouse lungs and severity of lung injury. In contrast, MSC therapy attenuated H9N2 avian influenza virus–induced acute lung inflammation and injury in mice68 via reduction in TNF-α, IFN-γ, IL-1α, and IL-6, as well as an increase in IL-10. This suggests that the efficacy of MSCs for influenza may be strain dependent, although additional studies are needed to further understand these issues.

MSCs exert multiple modulatory effects on diverse aspects of the immune response that are of direct relevance to their therapeutic potential for sepsis (fig. 3). In genome transcriptional studies in murine systemic sepsis models, MSC therapy has been demonstrated to modulate transcription of up to 13% of the murine genome, with immune response–related effects including the following: (1) down-regulation of toll-like receptor, nuclear factor-κB, and IL-6 signaling pathways; (2) up-regulation of nuclear factor of activated T cell–related genes; (3) up-regulation of genes involved in antigen presentation, phagocytosis, bacterial killing, complement, and coagulation regulation including platelet activation; and (4) enhancement of genes involved in cell-to-cell interactions and in the regulation of endothelial integrity.35,62 

Effects on Humoral Immune Response

Cytokines released from immune cells play key roles in the regulation of the host immune response. These intercellular messengers are the source of soluble regulatory signals that govern inflammatory responses to pathogens and tissue injury. Multiple studies demonstrate that MSCs decrease the proinflammatory cytokine response (TNF-α, IFN-γ, and IL-1α, -1β, -6, -12, and -17)35,37,57,60,69–79  while increasing concentrations of the antiinflammatory agents, including IL-1 receptor antagonist IL-10, cyclooxygenase-2, and prostaglandin E2.59,70,72,74,80,81 

Effects on the Inflammasome

The inflammasome was first described by Martinon et al.82  in 2002 and is a key component of the innate immune system. It is a multiprotein oligomer of pattern recognition receptors and sensors that regulate the activation of caspase-1 and a subsequent inflammatory response to infectious microbes and molecules derived from the host innate immune system. Several variants exist, and much interest centers on their role in health and in a host of inflammatory disorders. These structures may be on the membrane surface of myeloid cells, for example, toll-like receptors and C-type lectin receptors, or inside the cytoplasm, for example, nucleotide-binding oligomerization domain-like receptors MSCs appear to regulate inflammatory function. Miao et al.81  showed that MSCs can regulate the NLRP3 inflammasome in Kupffer cells via secretion of prostaglandin E2, leading to increased Kupffer cell production of IL-10. This reduced inflammasome activation and reduces the inflammatory response and ensuing organ dysfunction.81 

Effects on Neutrophil Response

Pathogen-associated molecular patterns, released from pathogens in infected tissue, bind to pattern recognition receptors, initiating a cascade of events and generating chemotactic (e.g., CXCL-2) and haplotactic gradients, which recruit activated neutrophils to the affected area. Neutrophils then attempt eradication of the offending microorganism via phagocytosis, the release of neutrophil extracellular traps, and the release of antimicrobial peptides.83  Neutrophil extracellular traps are structures released from neutrophils comprising a core of chromatin DNA and histones, surrounded by specific antimicrobial proteins (lactoferrin, cathepsin G, defensins, LL-37, and bacterial permeability increasing protein), proteases (neutrophil elastase, proteinase-3, and gelatinase), and reactive oxygen species–generating enzymes (myeloperoxidase).83  Neutrophil extracellular traps are extremely efficient in pathogen trapping, killing, and prevention of pathogen dissemination. This neutrophil response is generally advantageous and central to effective source control and pathogen eradication. However, when uncontrolled, such as in severe sepsis, activated neutrophils can migrate from inflamed tissues to other, noninfected tissue and organ systems (termed reverse migration), causing widespread host injury and organ dysfunction, potentially culminating in multiorgan dysfunction syndrome.84,85  In severe ongoing sepsis, infected tissues may have inadequate or dysfunctional neutrophils that are insufficient for source control due to neutrophil C-X-C motif chemokine receptor-2 downregulation, whereas an abundance of activated neutrophils contribute to injury in distant, healthy tissue due to neutrophil C-C motif chemokine receptor upregulation.84,86,87 

Multiple preclinical sepsis animal models demonstrate the potential for MSC therapy to alter neutrophil function to reduce host injury while maintaining bactericidal function.59,61  MSCs reduce neutrophil infiltration into the lung, liver, gut, and kidney, reducing injury and improving organ function in preclinical sepsis models.35,59–61,88,89  MSCs also enhance neutrophil-mediated phagocytosis, making them more effective in the clearance of bacteria.61  Neutrophil depletion, using anti-Ly6G antibody, totally abolished the protective effect of MSCs in systemic sepsis,61  highlighting the pivotal MSC–neutrophil interaction to the resolution of sepsis.

Effects on Monocyte/Macrophage Response

Macrophages are present in almost all tissues, where they coordinate developmental, metabolic, and immunologic functions and thus contribute to the maintenance of homeostasis.90  Macrophage dysfunction plays a key role in the pathogenesis of multiple diseases,91  including sepsis, and therefore these cells represent attractive therapeutic targets.

Much work has focused on the potential for MSCs to modulate macrophage function and phenotype.49,92  Macrophages, on stimulation, become activated into one of two phenotypes, namely classically activated M1 macrophages that were considered proinflammatory and play a key role in phagocytosis and killing of pathogens, and alternately activated M2 macrophages, with a more prorepair/resolution phenotype, that contribute to clearance of dead/injured host cells and tissue repair. A key effect of MSCs on macrophages may be their ability to favor development into an M2-like phenotype,44,49  with improved phagocytic activity and capacity for resolution of inflammation and injury repair.93  In a murine systemic sepsis model, MSCs secreted prostaglandin E2, which reprogrammed macrophages to the M2-like phenotype. Prostaglandin E2 increased macrophage production of IL-10, which reduced neutrophil transendothelial migration and neutrophil-induced organ damage and increased intravascular neutrophil and monocyte numbers, improving organ function and reducing pathogen load.35,49,59–61,70,72,73,88,89  MSCs can increase intravascular monocyte phagocytic potential via complement activation, increasing C5a levels, with subsequent CD11B up-regulation, both crucial for effective pathogen clearance.49,94  They also have an ability to enhance macrophage phagocytosis via several mechanisms, including secreted factors such as keratinocyte growth factor95  and mitochondrial transfer (from MSC to macrophage), either via direct cell–cell contact (via tunneling nanotubules) or indirectly (via exosomes).96  MSCs attenuate lipopolysaccharide-induced macrophage apoptosis via inhibition of the Wnt/β-catenin pathway.97 

Alteration of M1 macrophages to the M2 phenotype has been demonstrated to be important to injury resolution.98  More recently, emerging data have observed a wider spectrum of macrophage phenotypes.91  It appears that macrophages are activated to a spectrum of phenotypes depending on macrophage origin, current tissue of residence, and whether exposed previously to the same insult,90  and activation patterns display an element of temporal and spatial plasticity. Consequently, the effects of MSCs on macrophage phenotype may vary considerably based on these factors.

Effects on Adaptive Immune Response

The impact of MSCs on the T-cell response during sepsis has received limited attention. If anything, the well-described suppressive actions of MSCs on T-cell effector pathways in, for example, transplant studies, have been considered a potential concern in sepsis.99  Specifically, MSCs inhibit effector T-cell activation and can increase regulatory T-cell numbers,71,100  while suppressing proliferation of CD4+ T-helper cells, CD8+ cytotoxic T lymphocytes, and natural killer cells.99,101–103  These effects may be direct or may occur indirectly via effects on dendritic cells and/or other antigen-presenting cells.99 

The potential for MSCs to modulate regulatory T-cell function is of particular interest in the setting of sepsis and deserves additional attention. Regulatory T cells are a subpopulation of T cells that modulate the immune system, maintaining self-antigen tolerance and preventing autoimmune disease. They are classically considered to constitute a double-edged sword in infection, limiting inflammation and host tissue injury potentially at the price of reduced bacterial clearance.104  Regulatory T cells appear to have a role in suppressing the hyperinflammatory response to sepsis71 via suppression of the activation of autoreactive T effector cells.105  In contrast, in mice subjected to cecal ligation and puncture-induced sepsis, adoptive transfer of activated regulatory T cells enhanced bacterial clearance and increased animal survival,106  suggesting that the presence of regulatory T cells is essential to bacterial clearance and sepsis resolution.107  MSCs have been demonstrated to induce regulatory T-cell populations in multiple inflammatory models108–110  and during sepsis,71  potentially providing a mechanism by which MSCs may enhance sepsis resolution. MSCs may also modulate the activity of natural killer cells, alter dendritic cell differentiation,111  and regulate B-cell function via mechanisms that are not well understood.

Although much of the early enthusiasm for stem cells as a therapy derived from the concept that these cells could (trans-)differentiate to replace injured cells, this has been clearly demonstrated not to be a core mechanism by which MSCs exert their effects. Instead, MSCs work by multiple mechanisms, some of which require contact between the MSC and the target cells, while others are mediated via secreted products, both mediators and cell products, such as microvesicles and exosomes (figs. 2 and 3).

Cell Contact-dependent Effects

Although MSC engraftment is not required for efficacy in preclinical sepsis models, it seems that migration of MSCs to the site of injury and their retention there, at least for a short period, is required for efficacy after intrapulmonary endotoxin instillation.112  In murine endotoxemic sepsis, Xu et al.113  found that MSCs reduced lung inflammation and injury via a direct cell-to-cell contact-dependent mechanism. MSCs can bind to alveolar epithelial cells at connexin-43–positive gap junctions and transfer cellular products, including mitochondria, to increase cellular ATP levels, reducing epithelial cell dysfunction and mortality.42 

MSC Secretome

MSCs secrete multiple antimicrobial peptides, such as lipocalin-2,43  β-defensin-2,114  and LL-37.36,44  Other immunomodulatory mediators in the MSC secretome include prostaglandin E2,59  transforming growth factor-β,115  indoleamine 2,3-dioxygenase,116  IL-1 receptor antagonist,117  TNF-α–induced protein-6,79  and IL-1037 . MSC attenuation of cecal ligation and puncture sepsis is mediated in part via prostaglandin E2 secretion, which altered the host macrophage phenotype to an M2-like state.59  Endotoxin-induced stimulation of the toll-like receptor 4 expressed by the MSCs increases MCS production of prostaglandin E2 and cyclooxygenase 2.

MSC-derived Extracellular Vesicles

MSCs also release subcellular particles, termed extracellular vesicles, which incorporate cellular components, including mitochondria42  and gene products (i.e., mRNA and microRNAs).53  Two types of extracellular vesicles exist, namely microvesicles, which are in the 50- to 1000-nm range, and exosomes, which are in the 40- to 100-nm range. Microvesicles from MSCs decrease lung53  and kidney injury.118  These microvesicles decreased pulmonary edema, reduced the alveolar influx of neutrophils, and decreased alveolar macrophage inflammatory protein-2 concentrations after endotoxin-induced acute lung injury in mice,40  mainly through keratinocyte growth factor mRNA transferred to the injured alveolar epithelium. MSC-derived microvesicles decreased murine E. coli–induced severe pneumonia.53  MSC-derived exosomes exerted cardioprotective effects in polymicrobial sepsis through miR-223 transfer to cardiomyocytes and to macrophages, reducing the inflammatory response and enhancing survival of recipient cells.119  More recently, human-induced pluripotent stem cell–derived MSC exosomes had significant hepatoprotective effects in a hepatocellular injury model secondary to a combination of inflammatory response suppression, oxidative stress amelioration, and reduced apoptosis.120 

MSCs are activated by inflammatory mediators (including IFN-γ, IL-1β, and TNF-α) released from stimulated immune cells, potentially enhancing MSC function in sepsis.101  MSCs can also be modulated by toll-like receptor activators,121  which can polarize MSCs in vitro toward either a proinflammatory (MSC1) or antiinflammatory (MSC2) phenotype, depending on the specific receptor activator ligand.122  Activation of umbilical cord–derived MSCs with poly (I:C), a toll-like receptor-3 ligand, increased their efficacy in murine cecal ligation and puncture-induced systemic sepsis via inhibition of microRNA-143, which increased MSC expression of cyclooxygenase-2, leading to increased prostaglandin E2 production and enhanced MSC effects on macrophage function.89 

Overexpression of potentially therapeutic proteins is another strategy used to enhance MSC efficacy. MSCs overexpressing angiopoietin 1 were more effective than naive MSCs in reducing endotoxin-induced alveolar inflammation and lung permeability.123  Several gene overexpression strategies, using genes such as angiotensin-converting enzyme 2,124  fibroblast growth factor 2,125  and keratinocyte growth factor,126  have been demonstrated to enhance MSC efficacy in attenuating endotoxin-induced lung injury. MSCs transduced with E-prostanoid 2 receptor demonstrate enhanced homing to the injured lung, decreasing lung inflammation and reducing permeability.127  MSCs that overexpress the orphan receptor tyrosine kinase ROR2 further improved MSC-mediated protection against epithelial impairment in ARDS.128  Although these studies demonstrate useful proof-of-concept, additional studies in live bacterial models of pulmonary and systemic sepsis, with greater characterization of the effects on the immune response, would greatly enhance the translational potential of these approaches.

Despite multiple clinical trials of MSCs in diverse disease conditions, the evidence for therapeutic efficacy remains scant. An open-label phase I dose escalation trial for early septic shock, led by Drs McIntyre and Stewart at the University of Ottawa (Ottawa, Ontario, Canada), is due to publish preliminary results at the time of writing (table 3). The Cellular Immunotherapy for Septic Shock trial is a safety and dose escalation study of freshly cultured (i.e., not cryopreserved) allogeneic bone marrow MSCs for patients fulfilling clinical criteria for septic shock within 24 h of intensive care unit admission. MSC doses of 0.3- to 3.0-million cells per kilogram were used in the study. The investigators also enrolled a historical cohort that met study eligibility criteria to examine the adverse advents risk (NCT02421484). This key safety study will inform the design of larger-scale phase II septic shock trials that will determine the efficacy of MSCs for sepsis.

Table 3.

Selected Clinical Studies Examining Effects of MSCs of Relevance to Sepsis

Selected Clinical Studies Examining Effects of MSCs of Relevance to Sepsis
Selected Clinical Studies Examining Effects of MSCs of Relevance to Sepsis

A pilot study for a randomized, interventional trial, assessing the effect of MSCs on organ failure during septic shock, is due to commence enrollment in France (NCT02883803). A clinical trial of bone marrow MSCs recently concluded in Russia, which assessed neutropenic patients with septic shock (NCT01849237), demonstrated potentially promising results, although mortality was high in both groups.129  Of relevance to sepsis, a phase I trial of MSCs in ARDS has been published,39  a phase II trial has recently completed in the United States (NCT02097641), and a second is in progress in the Republic of Korea (NCT02112500).

However, despite multiple clinical studies of MSCs for diverse disease processes, there are no large-scale clinical trials demonstrating efficacy of MSC therapy. The study of MSC therapy for graft-versus-host disease, an immunologic condition with parallels to sepsis, is instructive. MSC therapy has been investigated for the prevention130  and treatment131  of graft-versus-host disease for more than 15 yr, and it is licensed for clinical use in certain countries, yet the clinical efficacy and mechanisms of action remain unclear. In acute graft-versus-host disease, ambiguity arose after failure of a phase III trial in the United States in 2009 (NCT00366145) to reach its clinical endpoint.132  This unexpected result contradicted European literature, with several smaller positive phase II trials emerging contemporaneously.133–136  Much discussion has focused on potential dissimilarities between large-scale, industrial-produced MSCs (used in a U.S. phase III trial) and smaller-scale MSC production in academic centers used in phase II trials, as a potential explanation for the contrasting trial results.137  Issues such as MSC donor variation, cell expansion techniques, immunogenicity of transfused products, and cryopreservation techniques are all challenges to large-scale MSC production. Currently, a number of graft-versus-host disease trials using MSCs have either recently completed (NCT01222039) or are currently enrolling (NCT01765634 and NCT01765660) to address these issues.

Other clinical trials that have not proven clinical efficacy include a trial of autologous MSCs in patients with myocardial ischemia138  and a trial of allogeneic MSCs in patients with chronic obstructive airways disease.139  These trials highlight potential translational challenges that lie ahead with regard to MSC therapy for sepsis.

Considerable barriers and knowledge gaps exist that significantly impede the clinical translation of MSCs for patients with sepsis. These issues will need to be better understood to enhance the likelihood of successful clinical efficacy studies. These challenges can be divided into those that relate MSCs as a therapy and that relate to sepsis as a disease target.

Heterogeneity of MSC Populations

There is no single marker or characteristic that identifies a cell as an MSC. The International Society for Cell and Gene Therapy (Vancouver, British Columbia, Canada) first defined MSCs for cellular therapy in 2006 based on the presence of three specific criteria, namely: (1) adherence to plastic; (2) the expression of, or lack thereof, certain surface molecules; and (3) their capacity for differentiation.140  Even if MSCs are sorted by consensus positive and negative surface markers, the resulting population is functionally heterogeneous.141  Consequently, MSC preparations produced by different cell-manufacturing facilities may differ in subtle but important ways, meaning that effects seen with one particular MSC product may not be seen with another, increasing the risk of therapeutic heterogeneity and failure of clinical translation. A more specific and robust approach to defining an MSC is imperative to reduce this heterogeneity.142,143 

Clonal derivation of MSCs has been described and may yield a more homogenous MSC population for subsequent expansion.144  Another approach to decrease MSC heterogeneity may be to generate them from homogenous populations of induced pluripotent stem cells.145  Proof of concept for this approach has been demonstrated, with induced pluripotent stem cell–derived MSCs demonstrating comparable efficacy to bone marrow–derived cells in preclinical models of corneal injury.146 

MSC Quality Assurance Issues Challenges

An advantage of MSCs as a therapy is their ease of passage and culture in vitro. However, this brings challenges, including the risk that repeated passaging can alter the MSC phenotype, ultimately resulting in reduced therapeutic efficacy.147  Repeated in vitro cellular passage can result in chromosomal damage, telomere shortening, and even malignant transformation.148  MSCs that have undergone cryopreservation and storage (generally used in clinical studies) may be less effective than fresh MSCs (used generally in preclinical studies), and this may explain inconsistencies in results between clinical and preclinical studies.149  Optimizing cryopreservation strategies for MSCs that maintain cell viability, potency, and efficacy is an important translational challenge. The criteria for the selection of donors for the establishment of master cell banks, which are then used to manufacture the MSC batches for clinical use and release criteria for the release of cell batches for clinical use are specified by regulatory agencies such as the U.S. Food and Drug Administration. Compliance with these criteria is a key issue in ensuring that cells of the highest quality are used in the clinical setting.

Optimal MSC Tissue Source

MSCs can be isolated from many tissues and organs. The bone marrow remains the standard tissue source of MSCs, and most preclinical and early-phase clinical sepsis studies use bone marrow MSCs. Other, potentially more plentiful sources of MSCs, including the umbilical cord and adipose tissues, are receiving increasing attention as potentially more feasible sources of cells for clinical use. Umbilical cords have the additional advantage of being a plentiful source; they are a waste biologic product and donor heterogeneity is reduced. Menstrual-derived MSCs, when combined with antibiotic therapy, synergistically improved the survival rate in mouse cecal ligation and puncture-induced sepsis, enhancing bacterial clearance and reducing organ injury.150 

Interestingly, with regard to sepsis, there appears to be differential immunomodulatory effects of MSCs derived from differing tissues. MSCs derived from the Wharton’s jelly of the umbilical cord attenuated increases in proinflammatory cytokines IL-1α, IL-6, and IFN-γ but did not modulate the response of antiinflammatory cytokines IL-4 and IL-10 in rats with cecal ligation and puncture-induced polymicrobial sepsis.73  Mouse adipose tissue MSCs protected mice from P. aeruginosa pulmonary infection by reducing lung bacterial load, neutrophil, and macrophage inflammatory protein-2 levels.47  Adipose tissue MSCs also enhanced the phagocytic and bactericidal abilities of mouse bone marrow–derived macrophages in vitro by inhibiting prostaglandin E2 signaling.47  Interestingly, it was observed that when prostaglandin E2 was administered to adipose tissue MSCs, their protective effects were negated.47  This contrasts with those effects observed with mouse bone marrow MSCs. Previous studies have suggested that bone marrow MSCs release prostaglandin E2, which enhances phagocytic ability and bacterial clearance by macrophages and stimulates them to release antiinflammatory IL-10.59,151  These differential immunomodulatory effects of MSCs, depending on their tissue source, may be important to consider when determining the optimal MSC for clinical testing.

Mechanisms of Action Relevant to Sepsis

The roles and relevance of the different mechanisms of action of MSCs in sepsis remain incompletely understood. Specifically, we need to better understand which MSC mechanisms of action are most relevant to sepsis and to develop strategies to enhance these effects. The most relevant MSC effects will likely differ based on the etiology, source, and phase of sepsis, highlighting the need to better characterize the biology of sepsis. Multiple secreted products, including prostaglandin E2,59  keratinocyte growth factor,53  and LL-37,36  exert therapeutic effects in preclinical sepsis models. Other potentially important effects are cell contact dependent, such as alteration of macrophage phenotype and phagocytic capacity. MSC-derived microvesicles play an important role via mechanisms involving transfer of mitochondria and nucleic acids.53  The injury microenvironment may further modulate MSC behavior.152  Evaluating the relative importance in sepsis of these diverse mechanisms of action will be important for maximizing the therapeutic efficacy of MSCs for sepsis.

MSC Dose and Timing

The majority of preclinical studies have used intravenous cell delivery. Local or regional cell delivery, for example, intraperitoneal for abdominal sepsis or intrapulmonary for pneumonia, is a feasible alternative option that may minimize systemic effects. However, the best dose and dosing regimen for MSCs in patients with sepsis are not known. Extrapolation from preclinical studies or from human studies for other conditions may be of limited relevance to patients with sepsis. The Cellular Immunotherapy for Septic Shock phase I study in sepsis is testing the safety of doses up to 3 million cells per kilogram. The optimal dose of MSCs may differ substantially in different disease states. The effect of factors such as the stage of illness, type of MSCs, route of cell delivery, viability and purity of MSCs, and condition of the patient, are all poorly understood. The timing of MSC therapy is also relevant, with preclinical studies to date generally focused on early MSC delivery. Characterization of MSC efficacy in later-phase sepsis, which is characterized by immune suppression, is a priority. The safety of repeated doses remains to be determined, with evidence to suggest that repeated administration does elicit an immune response.153 

MSC Safety Concerns

Although there is considerable experience in administering MSCs to patients, sepsis presents a number of safety concerns. Infusional toxicity is a concern during intravenous administration due to the risk of MSC clumping into microemboli that could obstruct the pulmonary circulation. Encouragingly, no infusional toxicities were seen in patients with ARDS in a recent phase I dose escalation clinical study.39  In the longer term, MSCs could potentially enhance tumorigenesis either by direct malignant transformation of MSCs or indirectly by facilitating growth of other tumor cells. Reassuringly, increased tumorigenesis has not been reported in the more than 6,000 patients who have received MSCs in clinical trials to date.154 

Population Heterogeneity

Sepsis is not a disease but rather a syndrome defined by a set of consensus clinical criteria that lump together patients who vary considerably in terms of their underlying biology, the source and nature of the inciting agent(s) and the host response, and a varying severity of illness. Some patients fulfilling clinical criteria for sepsis will not have a pathogen as the underlying inciting agent. The sepsis diagnostic criteria are useful in enabling rapid identification and early resuscitation and organ support of severely ill patients with sepsis. In addition, patients in different phases of sepsis may respond very differently to a therapeutic intervention. Consequently, this heterogeneity constitutes an impediment in identifying effective therapeutic strategies, especially where these strategies may have potentially harmful as well as beneficial effects.155  This heterogeneity of treatment effect may explain some negative trials in sepsis to date, whereby a treatment may have benefit in a particular patient subset, for example, severe sepsis with organ failure, but be ineffective or even harmful to patients with less severe sepsis.

It is very unlikely that MSC therapy will be useful in all patients with sepsis. Identification of patient subgroups within the population with sepsis that are more likely to respond to MSC therapy, and testing MSCs in these patient groups, will be necessary. In this regard, the identification of subphenotypes or endotypes within the sepsis population, as has been done in patients with ARDS by Calfee et al.,31  would be a key advance. A related approach, termed theranostics, involves identifying biomarkers of therapeutic responsiveness. Man et al.156  used this approach to identify potential subgroups of patients in the trial of Drotrecogin Alfa (Activated) in Adults with Septic Shock (PROWESS-SHOCK) that may have benefited from activated protein-C therapy.157  Similarly, Wong et al.158  identified a pediatric septic shock subgroup that had a higher mortality from corticosteroid administration. Based on our current elucidation of the biologic effects of MSCs, therapy might be more likely to be effective in patients with a hyperinflammatory phenotype.

Preclinical studies have demonstrated the therapeutic potential of MSCs for sepsis. The mechanisms of action of MSCs are increasingly well characterized and include modulation of the immune response, reduction of host injury from the proinflammatory response while augmenting bacterial clearance by indirect and direct mechanisms of action, and enhanced resolution of inflammation and enhanced tissue repair after injury. Although we await evidence of MSC benefit in patients with sepsis, phase I to II studies are underway, and initial reports are encouraging. However, significant hurdles still exist, both in terms of MSCs as a therapy and sepsis as a therapeutic target, which need to be overcome if the therapeutic potential of MSCs is to be realized. Addressing these ongoing knowledge gaps will help us to fully harness the therapeutic promise of MSCs for our patients with sepsis.

Supported by an operating grant from the Canadian Institutes for Health Research (Ottawa, Ontario, Canada), by the Science Foundation Ireland (16/FRL/3845; Dublin, Ireland), and by the Keenan Research Center for Biomedical Sciences at St. Michael’s Hospital (Toronto, Ontario, Canada).

The authors declare no competing interests.

1.
Gotts
JE
,
Matthay
MA
:
Sepsis: Pathophysiology and clinical management.
BMJ
2016
;
353
:
i1585
2.
Angus
DC
,
van der Poll
T
:
Severe sepsis and septic shock.
N Engl J Med
2013
;
369
:
840
51
3.
van der Poll
T
,
Opal
SM
:
Host-pathogen interactions in sepsis.
Lancet Infect Dis
2008
;
8
:
32
43
4.
Vincent
JL
,
Rello
J
,
Marshall
J
,
Silva
E
,
Anzueto
A
,
Martin
CD
,
Moreno
R
,
Lipman
J
,
Gomersall
C
,
Sakr
Y
,
Reinhart
K
;
EPIC II Group of Investigators
:
International study of the prevalence and outcomes of infection in intensive care units.
JAMA
2009
;
302
:
2323
9
5.
Vincent
JL
,
Marshall
JC
,
Namendys-Silva
SA
,
François
B
,
Martin-Loeches
I
,
Lipman
J
,
Reinhart
K
,
Antonelli
M
,
Pickkers
P
,
Njimi
H
,
Jimenez
E
,
Sakr
Y
;
ICON Investigators
:
Assessment of the worldwide burden of critical illness: The intensive care over nations (ICON) audit.
Lancet Respir Med
2014
;
2
:
380
6
6.
Fleischmann
C
,
Scherag
A
,
Adhikari
NK
,
Hartog
CS
,
Tsaganos
T
,
Schlattmann
P
,
Angus
DC
,
Reinhart
K
;
International Forum of Acute Care Trialists
:
Assessment of global incidence and mortality of hospital-treated sepsis: Current estimates and limitations.
Am J Respir Crit Care Med
2016
;
193
:
259
72
7.
Torio
CM
,
Andrews
RM
:
National inpatient hospital costs: The most expensive conditions by payer, 2011. Statistical Brief #160. Healthcare Cost and Utilization Project (HCUP) Statistical Briefs
.
Rockville, Agency for Healthcare Research and Quality
,
2013
8.
Martin
GS
,
Mannino
DM
,
Eaton
S
,
Moss
M
:
The epidemiology of sepsis in the United States from 1979 through 2000.
N Engl J Med
2003
;
348
:
1546
54
9.
Iwashyna
TJ
,
Cooke
CR
,
Wunsch
H
,
Kahn
JM
:
Population burden of long-term survivorship after severe sepsis in older Americans.
J Am Geriatr Soc
2012
;
60
:
1070
7
10.
Gaieski
DF
,
Edwards
JM
,
Kallan
MJ
,
Carr
BG
:
Benchmarking the incidence and mortality of severe sepsis in the United States.
Crit Care Med
2013
;
41
:
1167
74
11.
Dellinger
RP
,
Levy
MM
,
Rhodes
A
,
Annane
D
,
Gerlach
H
,
Opal
SM
,
Sevransky
JE
,
Sprung
CL
,
Douglas
IS
,
Jaeschke
R
,
Osborn
TM
,
Nunnally
ME
,
Townsend
SR
,
Reinhart
K
,
Kleinpell
RM
,
Angus
DC
,
Deutschman
CS
,
Machado
FR
,
Rubenfeld
GD
,
Webb
SA
,
Beale
RJ
,
Vincent
JL
,
Moreno
R
;
Surviving Sepsis Campaign Guidelines Committee including the Pediatric Subgroup
:
Surviving sepsis campaign: International guidelines for management of severe sepsis and septic shock: 2012.
Crit Care Med
2013
;
41
:
580
637
12.
Kaukonen
KM
,
Bailey
M
,
Suzuki
S
,
Pilcher
D
,
Bellomo
R
:
Mortality related to severe sepsis and septic shock among critically ill patients in Australia and New Zealand, 2000-2012.
JAMA
2014
;
311
:
1308
16
13.
Liu
V
,
Escobar
GJ
,
Greene
JD
,
Soule
J
,
Whippy
A
,
Angus
DC
,
Iwashyna
TJ
:
Hospital deaths in patients with sepsis from 2 independent cohorts.
JAMA
2014
;
312
:
90
2
14.
Beck
MK
,
Jensen
AB
,
Nielsen
AB
,
Perner
A
,
Moseley
PL
,
Brunak
S
:
Diagnosis trajectories of prior multi-morbidity predict sepsis mortality.
Sci Rep
2016
;
6
:
36624
15.
Iwashyna
TJ
,
Ely
EW
,
Smith
DM
,
Langa
KM
:
Long-term cognitive impairment and functional disability among survivors of severe sepsis.
JAMA
2010
;
304
:
1787
94
16.
Hotchkiss
RS
,
Monneret
G
,
Payen
D
:
Sepsis-induced immunosuppression: From cellular dysfunctions to immunotherapy.
Nat Rev Immunol
2013
;
13
:
862
74
17.
Pisetsky
DS
:
The origin and properties of extracellular DNA: From PAMP to DAMP.
Clin Immunol
2012
;
144
:
32
40
18.
Boomer
JS
,
Green
JM
,
Hotchkiss
RS
:
The changing immune system in sepsis: Is individualized immuno-modulatory therapy the answer?
Virulence
2014
;
5
:
45
56
19.
Hotchkiss
RS
,
Karl
IE
:
The pathophysiology and treatment of sepsis.
N Engl J Med
2003
;
348
:
138
50
20.
Kumar
A
,
Kethireddy
S
:
Emerging concepts in optimizing antimicrobial therapy of septic shock: Speed is life but a hammer helps too.
Crit Care
2013
;
17
:
104
21.
Boomer
JS
,
To
K
,
Chang
KC
,
Takasu
O
,
Osborne
DF
,
Walton
AH
,
Bricker
TL
,
Jarman
SD
2nd
,
Kreisel
D
,
Krupnick
AS
,
Srivastava
A
,
Swanson
PE
,
Green
JM
,
Hotchkiss
RS
:
Immunosuppression in patients who die of sepsis and multiple organ failure.
JAMA
2011
;
306
:
2594
605
22.
Hall
MW
,
Knatz
NL
,
Vetterly
C
,
Tomarello
S
,
Wewers
MD
,
Volk
HD
,
Carcillo
JA
:
Immunoparalysis and nosocomial infection in children with multiple organ dysfunction syndrome.
Intensive Care Med
2011
;
37
:
525
32
23.
Otto
GP
,
Sossdorf
M
,
Claus
RA
,
Rödel
J
,
Menge
K
,
Reinhart
K
,
Bauer
M
,
Riedemann
NC
:
The late phase of sepsis is characterized by an increased microbiological burden and death rate.
Crit Care
2011
;
15
:
R183
24.
Walton
AH
,
Muenzer
JT
,
Rasche
D
,
Boomer
JS
,
Sato
B
,
Brownstein
BH
,
Pachot
A
,
Brooks
TL
,
Deych
E
,
Shannon
WD
,
Green
JM
,
Storch
GA
,
Hotchkiss
RS
:
Reactivation of multiple viruses in patients with sepsis.
PLoS One
2014
;
9
:
e98819
25.
van Vught
LA
,
Klein Klouwenberg
PM
,
Spitoni
C
,
Scicluna
BP
,
Wiewel
MA
,
Horn
J
,
Schultz
MJ
,
Nürnberg
P
,
Bonten
MJ
,
Cremer
OL
,
van der Poll
T
;
MARS Consortium
:
Incidence, risk factors, and attributable mortality of secondary infections in the intensive care unit after admission for sepsis.
JAMA
2016
;
315
:
1469
79
26.
Angus
DC
:
The search for effective therapy for sepsis: Back to the drawing board?
JAMA
2011
;
306
:
2614
5
27.
Cohen
J
,
Opal
S
,
Calandra
T
:
Sepsis studies need new direction.
Lancet Infect Dis
2012
;
12
:
503
5
28.
Cronin
L
,
Cook
DJ
,
Carlet
J
,
Heyland
DK
,
King
D
,
Lansang
MA
,
Fisher
CJ
Jr
:
Corticosteroid treatment for sepsis: A critical appraisal and meta-analysis of the literature.
Crit Care Med
1995
;
23
:
1430
9
29.
Meisel
C
,
Schefold
JC
,
Pschowski
R
,
Baumann
T
,
Hetzger
K
,
Gregor
J
,
Weber-Carstens
S
,
Hasper
D
,
Keh
D
,
Zuckermann
H
,
Reinke
P
,
Volk
HD
:
Granulocyte-macrophage colony-stimulating factor to reverse sepsis-associated immunosuppression: A double-blind, randomized, placebo-controlled multicenter trial.
Am J Respir Crit Care Med
2009
;
180
:
640
8
30.
Bellingan
G
,
Maksimow
M
,
Howell
DC
,
Stotz
M
,
Beale
R
,
Beatty
M
,
Walsh
T
,
Binning
A
,
Davidson
A
,
Kuper
M
,
Shah
S
,
Cooper
J
,
Waris
M
,
Yegutkin
GG
,
Jalkanen
J
,
Salmi
M
,
Piippo
I
,
Jalkanen
M
,
Montgomery
H
,
Jalkanen
S
:
The effect of intravenous interferon-beta-1a (FP-1201) on lung CD73 expression and on acute respiratory distress syndrome mortality: an open-label study.
Lancet Respir Med
2014
;
2
:
98
107
31.
Calfee
CS
,
Delucchi
K
,
Parsons
PE
,
Thompson
BT
,
Ware
LB
,
Matthay
MA
;
NHLBI ARDS Network
:
Subphenotypes in acute respiratory distress syndrome: Latent class analysis of data from two randomised controlled trials.
Lancet Respir Med
2014
;
2
:
611
20
32.
Curley
GF
,
Hayes
M
,
Ansari
B
,
Shaw
G
,
Ryan
A
,
Barry
F
,
O’Brien
T
,
O’Toole
D
,
Laffey
JG
:
Mesenchymal stem cells enhance recovery and repair following ventilator-induced lung injury in the rat.
Thorax
2012
;
67
:
496
501
33.
Curley
GF
,
Ansari
B
,
Hayes
M
,
Devaney
J
,
Masterson
C
,
Ryan
A
,
Barry
F
,
O’Brien
T
,
Toole
DO
,
Laffey
JG
:
Effects of intratracheal mesenchymal stromal cell therapy during recovery and resolution after ventilator-induced lung injury.
Anesthesiology
2013
;
118
:
924
32
34.
Hayes
M
,
Masterson
C
,
Devaney
J
,
Barry
F
,
Elliman
S
,
O’Brien
T
,
O’Toole
D
,
Curley
GF
,
Laffey
JG
:
Therapeutic efficacy of human mesenchymal stromal cells in the repair of established ventilator-induced lung injury in the rat.
Anesthesiology
2015
;
122
:
363
73
35.
Mei
SH
,
Haitsma
JJ
,
Dos Santos
CC
,
Deng
Y
,
Lai
PF
,
Slutsky
AS
,
Liles
WC
,
Stewart
DJ
:
Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis.
Am J Respir Crit Care Med
2010
;
182
:
1047
57
36.
Krasnodembskaya
A
,
Song
Y
,
Fang
X
,
Gupta
N
,
Serikov
V
,
Lee
JW
,
Matthay
MA
:
Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37.
Stem Cells
2010
;
28
:
2229
38
37.
Gupta
N
,
Su
X
,
Popov
B
,
Lee
JW
,
Serikov
V
,
Matthay
MA
:
Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice.
J Immunol
2007
;
179
:
1855
63
38.
Lee
JW FX
,
Gupta
N
,
Serikov
V
,
Matthay
MA
:
Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung.
Proc Natl Acad Sci USA
2009
;
106
:
6
39.
Wilson
JG
,
Liu
KD
,
Zhuo
H
,
Caballero
L
,
McMillan
M
,
Fang
X
,
Cosgrove
K
,
Vojnik
R
,
Calfee
CS
,
Lee
JW
,
Rogers
AJ
,
Levitt
J
,
Wiener-Kronish
J
,
Bajwa
EK
,
Leavitt
A
,
McKenna
D
,
Thompson
BT
,
Matthay
MA
:
Mesenchymal stem (stromal) cells for treatment of ARDS: A phase 1 clinical trial.
Lancet Respir Med
2015
;
3
:
24
32
40.
Zhu
YG FX
,
Abbott
J
,
Fang
XH
,
Hao
Q
,
Monsel
A
,
Qu
JM
,
Matthay
MA
,
Lee
JW
:
Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice.
Stem Cells
2014
;
32
:
10
41.
Hao
Q ZY
,
Monsel
A
,
Gennai
S
,
Lee
T
,
Xu
F
,
Lee
JW
:
Study of bone marrow and embryonic stem cell-derived human mesenchymal stem cells for treatment of escherichia coli endotoxin-induced acute lung injury in mice.
Stem Cells Transl Med
2015
;
4
:
9
42.
Islam
MN
,
Das
SR
,
Emin
MT
,
Wei
M
,
Sun
L
,
Westphalen
K
,
Rowlands
DJ
,
Quadri
SK
,
Bhattacharya
S
,
Bhattacharya
J
:
Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury.
Nat Med
2012
;
18
:
759
65
43.
Gupta
N
,
Krasnodembskaya
A
,
Kapetanaki
M
,
Mouded
M
,
Tan
X
,
Serikov
V
,
Matthay
MA
:
Mesenchymal stem cells enhance survival and bacterial clearance in murine Escherichia coli pneumonia.
Thorax
2012
;
67
:
533
9
44.
Devaney
J
,
Horie
S
,
Masterson
C
,
Elliman
S
,
Barry
F
,
O’Brien
T
,
Curley
GF
,
O’Toole
D
,
Laffey
JG
:
Human mesenchymal stromal cells decrease the severity of acute lung injury induced by E. coli in the rat.
Thorax
2015
;
70
:
625
35
45.
Shalaby
SM
,
El-Shal
AS
,
Abd-Allah
SH
,
Selim
AO
,
Selim
SA
,
Gouda
ZA
,
Abd El Motteleb
DM
,
Zanfaly
HE
,
El-Assar
HM
,
Abdelazim
S
:
Mesenchymal stromal cell injection protects against oxidative stress in Escherichia coli-induced acute lung injury in mice.
Cytotherapy
2014
;
16
:
764
75
46.
Hackstein
H
,
Lippitsch
A
,
Krug
P
,
Schevtschenko
I
,
Kranz
S
,
Hecker
M
,
Dietert
K
,
Gruber
AD
,
Bein
G
,
Brendel
C
,
Baal
N
:
Prospectively defined murine mesenchymal stem cells inhibit Klebsiella pneumoniae-induced acute lung injury and improve pneumonia survival.
Respir Res
2015
;
16
:
123
47.
Mao
YX
,
Xu
JF
,
Seeley
EJ
,
Tang
XD
,
Xu
LL
,
Zhu
YG
,
Song
YL
,
Qu
JM
:
Adipose tissue-derived mesenchymal stem cells attenuate pulmonary infection caused by pseudomonas aeruginosa via inhibiting overproduction of prostaglandin E2.
Stem Cells
2015
;
33
:
2331
42
48.
Sutton
MT
,
Fletcher
D
,
Ghosh
SK
,
Weinberg
A
,
van Heeckeren
R
,
Kaur
S
,
Sadeghi
Z
,
Hijaz
A
,
Reese
J
,
Lazarus
HM
,
Lennon
DP
,
Caplan
AI
,
Bonfield
TL
:
Antimicrobial properties of mesenchymal stem cells: Therapeutic potential for cystic fibrosis infection, and treatment.
Stem Cells Int
2016
;
2016
:
5303048
49.
Krasnodembskaya
A
,
Samarani
G
,
Song
Y
,
Zhuo
H
,
Su
X
,
Lee
JW
,
Gupta
N
,
Petrini
M
,
Matthay
MA
:
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
13
50.
Curley
GF
,
M.
J
,
Dixon
S
,
Hogan
G
,
Masterson
C
,
O’Toole
D
,
Devaney
J
,
Laffey
JG
:
Cryopreserved, xeno-free human umbilical cord mesenchymal stromal cells reduce lung injury severity and bacterial burden in rodent E. coli induced acute respiratory distress syndrome.
Crit Care Med
2017
;
45
:
e202
12
51.
Kim
ES
,
Chang
YS
,
Choi
SJ
,
Kim
JK
,
Yoo
HS
,
Ahn
SY
,
Sung
DK
,
Kim
SY
,
Park
YR
,
Park
WS
:
Intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells attenuates Escherichia coli-induced acute lung injury in mice.
Respir Res
2011
;
12
:
108
52.
Asmussen
S
,
Ito
H
,
Traber
DL
,
Lee
JW
,
Cox
RA
,
Hawkins
HK
,
McAuley
DF
,
McKenna
DH
,
Traber
LD
,
Zhuo
H
,
Wilson
J
,
Herndon
DN
,
Prough
DS
,
Liu
KD
,
Matthay
MA
,
Enkhbaatar
P
:
Human mesenchymal stem cells reduce the severity of acute lung injury in a sheep model of bacterial pneumonia.
Thorax
2014
;
69
:
819
25
53.
Monsel
A
,
Zhu
YG
,
Gennai
S
,
Hao
Q
,
Hu
S
,
Rouby
JJ
,
Rosenzwajg
M
,
Matthay
MA
,
Lee
JW
:
Therapeutic effects of human mesenchymal stem cell-derived microvesicles in severe pneumonia in mice.
Am J Respir Crit Care Med
2015
;
192
:
324
36
54.
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
55.
Mot
YY
,
Othman
I
,
Sharifah
SH
:
Synergistic antibacterial effect of co-administering adipose-derived mesenchymal stromal cells and Ophiophagus hannah L-amino acid oxidase in a mouse model of methicillin-resistant Staphylococcus aureus-infected wounds.
Stem Cell Res Ther
2017
;
8
:
5
56.
Guerra
AD
,
Cantu
DA
,
Vecchi
JT
,
Rose
WE
,
Hematti
P
,
Kao
WJ
:
Mesenchymal stromal/stem cell and minocycline-loaded hydrogels inhibit the growth of staphylococcus aureus that evades immunomodulation of blood-derived leukocytes.
AAPS J
2015
;
17
:
620
30
57.
Shin
S
,
Kim
Y
,
Jeong
S
,
Hong
S
,
Kim
I
,
Lee
W
,
Choi
S
:
The therapeutic effect of human adult stem cells derived from adipose tissue in endotoxemic rat model.
Int J Med Sci
2013
;
10
:
11
58.
Hu
Y QC
,
Zheng
G
,
Lai
D
,
Tao
H
,
Zhang
Y
,
Qiu
G
,
Ge
M
,
Huang
L
,
Chen
L
,
Cheng
B
,
Shu
Q
,
Xu
J
:
Mesenchymal stem cell-educated macrophages ameliorate LPS-induced systemic response.
Mediators Inflamm
2016
;
2016
:
13
59.
Németh
K
,
Leelahavanichkul
A
,
Yuen
PS
,
Mayer
B
,
Parmelee
A
,
Doi
K
,
Robey
PG
,
Leelahavanichkul
K
,
Koller
BH
,
Brown
JM
,
Hu
X
,
Jelinek
I
,
Star
RA
,
Mezey
E
:
Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production.
Nat Med
2009
;
15
:
42
9
60.
Luo
CJ
,
Zhang
FJ
,
Zhang
L
,
Geng
YQ
,
Li
QG
,
Hong
Q
,
Fu
B
,
Zhu
F
,
Cui
SY
,
Feng
Z
,
Sun
XF
,
Chen
XM
:
Mesenchymal stem cells ameliorate sepsis-associated acute kidney injury in mice.
Shock
2014
;
41
:
123
9
61.
Hall
SR
,
Tsoyi
K
,
Ith
B
,
Padera
RF
Jr
,
Lederer
JA
,
Wang
Z
,
Liu
X
,
Perrella
MA
:
Mesenchymal stromal cells improve survival during sepsis in the absence of heme oxygenase-1: The importance of neutrophils.
Stem Cells
2013
;
31
:
397
407
62.
dos Santos
CC
,
Murthy
S
,
Hu
P
,
Shan
Y
,
Haitsma
JJ
,
Mei
SH
,
Stewart
DJ
,
Liles
WC
:
Network analysis of transcriptional responses induced by mesenchymal stem cell treatment of experimental sepsis.
Am J Pathol
2012
;
181
:
1681
92
63.
Kim
H
,
Darwish
I
,
Monroy
MF
,
Prockop
DJ
,
Liles
WC
,
Kain
KC
:
Mesenchymal stromal (stem) cells suppress pro-inflammatory cytokine production but fail to improve survival in experimental staphylococcal toxic shock syndrome.
BMC Immunol
2014
;
15
:
1
64.
Hong
J HA
,
Wang
L
,
Schmitt
A
,
Wuchter
P
,
Tabarkiewicz
J
,
Kleist
C
,
Bieback
K
,
Ho
AD
,
Schmitt
M
:
Indoleamine 2,3-dioxygenase mediates inhibition of virus- specific CD8(+) T cell proliferation by human mesenchymal stromal cells.
Cytotherapy
2016
;
18
:
9
65.
Meisel
R BS
,
Heseler
K
,
Degistirici
O
,
Bülle
H
,
Woite
C
,
Stuhlsatz
S
,
Schwippert
W
,
Jäger
M
,
Sorg
R
,
Henschler
R
,
Seissler
J
,
Dilloo
D
,
Däubener
W
:
Human but not murine multipotent mesenchymal stromal cells exhibit broad-spectrum antimicrobial effector function mediated by indoleamine 2,3-dioxygenase.
Leukemia
2011
;
25
:
7
66.
Gotts
JE
,
Abbott
J
,
Matthay
MA
:
Influenza causes prolonged disruption of the alveolar-capillary barrier in mice unresponsive to mesenchymal stem cell therapy.
Am J Physiol Lung Cell Mol Physiol
2014
;
307
:
L395
406
67.
Darwish
I
,
Banner
D
,
Mubareka
S
,
Kim
H
,
Besla
R
,
Kelvin
DJ
,
Kain
KC
,
Liles
WC
:
Mesenchymal stromal (stem) cell therapy fails to improve outcomes in experimental severe influenza.
PLoS One
2013
;
8
:
e71761
68.
Li
Y
,
Xu
J
,
Shi
W
,
Chen
C
,
Shao
Y
,
Zhu
L
,
Lu
W
,
Han
X
:
Mesenchymal stromal cell treatment prevents H9N2 avian influenza virus-induced acute lung injury in mice.
Stem Cell Res Ther
2016
;
7
:
159
69.
Fan
H
,
Wong
D
,
Ashton
SH
,
Borg
KT
,
Halushka
PV
,
Cook
JA
:
Beneficial effect of a CXCR4 agonist in murine models of systemic inflammation.
Inflammation
2012
;
35
:
130
7
70.
Weil
BR
,
Herrmann
JL
,
Abarbanell
AM
,
Manukyan
MC
,
Poynter
JA
,
Meldrum
DR
:
Intravenous infusion of mesenchymal stem cells is associated with improved myocardial function during endotoxemia.
Shock
2011
;
36
:
235
41
71.
Chao
YH
,
Wu
HP
,
Wu
KH
,
Tsai
YG
,
Peng
CT
,
Lin
KC
,
Chao
WR
,
Lee
MS
,
Fu
YC
:
An increase in CD3+CD4+CD25+ regulatory T cells after administration of umbilical cord-derived mesenchymal stem cells during sepsis.
PLoS One
2014
;
9
:
e110338
72.
Zullo
JA
,
Nadel
EP
,
Rabadi
MM
,
Baskind
MJ
,
Rajdev
MA
,
Demaree
CM
,
Vasko
R
,
Chugh
SS
,
Lamba
R
,
Goligorsky
MS
,
Ratliff
BB
:
The secretome of hydrogel-coembedded endothelial progenitor cells and mesenchymal stem cells instructs macrophage polarization in endotoxemia.
Stem Cells Transl Med
2015
;
4
:
852
61
73.
Cóndor
JM
,
Rodrigues
CE
,
Sousa Moreira
Rd
,
Canale
D
,
Volpini
RA
,
Shimizu
MH
,
Camara
NO
,
Noronha
Ide L
,
Andrade
L
:
Treatment with human Wharton’s jelly-derived mesenchymal stem cells attenuates sepsis-induced kidney injury, liver injury, and endothelial dysfunction.
Stem Cells Transl Med
2016
;
5
:
1048
57
74.
Li
N
,
Hu
DH
,
Wang
YJ
,
Hu
XL
,
Zhang
Y
,
Li
XQ
,
Shi
JH
,
Bai
XZ
,
Cai
WX
:
Effects of adipose-derived stem cells on renal injury in burn mice with sepsis [in Chinese].
Zhonghua Shao Shang Za Zhi
2013
;
29
:
249
54
75.
Weil
BR
,
Manukyan
MC
,
Herrmann
JL
,
Wang
Y
,
Abarbanell
AM
,
Poynter
JA
,
Meldrum
DR
:
Mesenchymal stem cells attenuate myocardial functional depression and reduce systemic and myocardial inflammation during endotoxemia.
Surgery
2010
;
148
:
444
52
76.
Sung
PH
,
Chang
CL
,
Tsai
TH
,
Chang
LT
,
Leu
S
,
Chen
YL
,
Yang
CC
,
Chua
S
,
Yeh
KH
,
Chai
HT
,
Chang
HW
,
Chen
HH
,
Yip
HK
:
Apoptotic adipose-derived mesenchymal stem cell therapy protects against lung and kidney injury in sepsis syndrome caused by cecal ligation puncture in rats.
Stem Cell Res Ther
2013
;
4
:
155
77.
Chen
HH
,
Chang
CL
,
Lin
KC
,
Sung
PH
,
Chai
HT
,
Zhen
YY
,
Chen
YC
,
Wu
YC
,
Leu
S
,
Tsai
TH
,
Chen
CH
,
Chang
HW
,
Yip
HK
:
Melatonin augments apoptotic adipose-derived mesenchymal stem cell treatment against sepsis-induced acute lung injury.
Am J Transl Res
2014
;
6
:
439
58
78.
Ou
H
,
Zhao
S
,
Peng
Y
,
Xiao
X
,
Wang
Q
,
Liu
H
,
Xiao
X
,
Yang
M
:
Comparison of bone marrow tissue- and adipose tissue-derived mesenchymal stem cells in the treatment of sepsis in a murine model of lipopolysaccharide-induced sepsis.
Mol Med Rep
2016
;
14
:
3862
70
79.
Danchuk
S
,
Ylostalo
JH
,
Hossain
F
,
Sorge
R
,
Ramsey
A
,
Bonvillain
RW
,
Lasky
JA
,
Bunnell
BA
,
Welsh
DA
,
Prockop
DJ
,
Sullivan
DE
:
Human multipotent stromal cells attenuate lipopolysaccharide-induced acute lung injury in mice via secretion of tumor necrosis factor-α-induced protein 6.
Stem Cell Res Ther
2011
;
2
:
27
80.
Letourneau
PA
,
Menge
TD
,
Wataha
KA
,
Wade
CE
,
S Cox
C
Jr
,
Holcomb
JB
,
Pati
S
:
Human bone marrow derived mesenchymal stem cells regulate leukocyte-endothelial interactions and activation of transcription factor NF-kappa B.
J Tissue Sci Eng
2011
;
suppl 3
:
001
81.
Miao
CM
,
Jiang
XW
,
He
K
,
Li
PZ
,
Liu
ZJ
,
Cao
D
,
Ou
ZB
,
Gong
JP
,
Liu
CA
,
Cheng
Y
:
Bone marrow stromal cells attenuate LPS-induced mouse acute liver injury via the prostaglandin E 2-dependent repression of the NLRP3 inflammasome in Kupffer cells.
Immunol Lett
2016
;
179
:
102
13
82.
Martinon
F
,
Burns
K
,
Tschopp
J
:
The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta.
Mol Cell
2002
;
10
:
417
26
83.
Kolaczkowska
E
,
Kubes
P
:
Neutrophil recruitment and function in health and inflammation.
Nat Rev Immunol
2013
;
13
:
159
75
84.
Sônego
F
,
Castanheira
FV
,
Ferreira
RG
,
Kanashiro
A
,
Leite
CA
,
Nascimento
DC
,
Colón
DF
,
Borges
Vde F
,
Alves-Filho
JC
,
Cunha
FQ
:
Paradoxical roles of the neutrophil in sepsis: Protective and deleterious.
Front Immunol
2016
;
7
:
155
85.
Nourshargh
S
,
Renshaw
SA
,
Imhof
BA
:
Reverse migration of neutrophils: Where, when, how, and why?
Trends Immunol
2016
;
37
:
273
86
86.
McAvoy
EF
,
McDonald
B
,
Parsons
SA
,
Wong
CH
,
Landmann
R
,
Kubes
P
:
The role of CD14 in neutrophil recruitment within the liver microcirculation during endotoxemia.
J Immunol
2011
;
186
:
2592
601
87.
Wei
J
,
Wei
C
,
Wang
M
,
Qiu
X
,
Li
Y
,
Yuan
Y
,
Jin
C
,
Leng
L
,
Wang
J
,
Yang
X
,
He
F
:
The GTPase-activating protein GIT2 protects against colitis by negatively regulating Toll-like receptor signaling.
2014; 111:8883
8
88.
Elman
JS
,
Li
M
,
Wang
F
,
Gimble
JM
,
Parekkadan
B
:
A comparison of adipose and bone marrow-derived mesenchymal stromal cell secreted factors in the treatment of systemic inflammation.
J Inflamm (Lond)
2014
;
11
:
1
89.
Zhao
X
,
Liu
D
,
Gong
W
,
Zhao
G
,
Liu
L
,
Yang
L
,
Hou
Y
:
The toll-like receptor 3 ligand, poly(I:C), improves immunosuppressive function and therapeutic effect of mesenchymal stem cells on sepsis via inhibiting MiR-143.
Stem Cells
2014
;
32
:
521
33
90.
Glass
CK
,
Natoli
G
:
Molecular control of activation and priming in macrophages.
Nat Immunol
2016
;
17
:
26
33
91.
Ginhoux
F
,
Schultze
JL
,
Murray
PJ
,
Ochando
J
,
Biswas
SK
:
New insights into the multidimensional concept of macrophage ontogeny, activation and function.
Nat Immunol
2016
;
17
:
34
40
92.
Luz-Crawford
P
,
Jorgensen
C
,
Djouad
F
:
Mesenchymal stem cells direct the immunological fate of macrophages.
Results Probl Cell Differ
2017
;
62
:
61
72
93.
Kim
J
,
Hematti
P
:
Mesenchymal stem cell-educated macrophages: A novel type of alternatively activated macrophages.
Exp Hematol
2009
;
37
:
1445
53
94.
Brekke
OL
,
Christiansen
D
,
Fure
H
,
Fung
M
,
Mollnes
TE
:
The role of complement C3 opsonization, C5a receptor, and CD14 in E. coli-induced up-regulation of granulocyte and monocyte CD11b/CD18 (CR3), phagocytosis, and oxidative burst in human whole blood.
J Leukoc Biol
2007
;
81
:
1404
13
95.
Lee
JW
,
Krasnodembskaya
A
,
McKenna
DH
,
Song
Y
,
Abbott
J
,
Matthay
MA
:
Therapeutic effects of human mesenchymal stem cells in ex vivo human lungs injured with live bacteria.
Am J Respir Crit Care Med
2013
;
187
:
751
60
96.
Jackson
MV
,
Morrison
TJ
,
Doherty
DF
,
McAuley
DF
,
Matthay
MA
,
Kissenpfennig
A
,
O’Kane
CM
,
Krasnodembskaya
AD
:
Mitochondrial transfer via tunneling nanotubes is an important mechanism by which mesenchymal stem cells enhance macrophage phagocytosis in the in vitro and in vivo models of ARDS.
Stem Cells
2016
;
34
:
2210
23
97.
Li
B
,
Zhang
H
,
Zeng
M
,
He
W
,
Li
M
,
Huang
X
,
Deng
DY
,
Wu
J
:
Bone marrow mesenchymal stem cells protect alveolar macrophages from lipopolysaccharide-induced apoptosis partially by inhibiting the Wnt/β-catenin pathway.
Cell Biol Int
2015
;
39
:
192
200
98.
Dal-Secco
D
,
Wang
J
,
Zeng
Z
,
Kolaczkowska
E
,
Wong
CH
,
Petri
B
,
Ransohoff
RM
,
Charo
IF
,
Jenne
CN
,
Kubes
P
:
A dynamic spectrum of monocytes arising from the in situ reprogramming of CCR2+ monocytes at a site of sterile injury.
J Exp Med
2015
;
212
:
447
56
99.
Duffy
MM
,
Ritter
T
,
Ceredig
R
,
Griffin
MD
:
Mesenchymal stem cell effects on T-cell effector pathways.
Stem Cell Res Ther
2011
;
2
:
34
100.
Ghannam
S
,
Pène
J
,
Moquet-Torcy
G
,
Torcy-Moquet
G
,
Jorgensen
C
,
Yssel
H
:
Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype.
J Immunol
2010
;
185
:
302
12
101.
Krampera
M
,
Cosmi
L
,
Angeli
R
,
Pasini
A
,
Liotta
F
,
Andreini
A
,
Santarlasci
V
,
Mazzinghi
B
,
Pizzolo
G
,
Vinante
F
,
Romagnani
P
,
Maggi
E
,
Romagnani
S
,
Annunziato
F
:
Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells.
Stem Cells
2006
;
24
:
386
98
102.
DelaRosa
O
,
Sánchez-Correa
B
,
Morgado
S
,
Ramírez
C
,
del Río
B
,
Menta
R
,
Lombardo
E
,
Tarazona
R
,
Casado
JG
:
Human adipose-derived stem cells impair natural killer cell function and exhibit low susceptibility to natural killer-mediated lysis.
Stem Cells Dev
2012
;
21
:
1333
43
103.
Di Nicola
M
,
Carlo-Stella
C
,
Magni
M
,
Milanesi
M
,
Longoni
PD
,
Matteucci
P
,
Grisanti
S
,
Gianni
AM
:
Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli.
Blood
2002
;
99
:
3838
43
104.
Belkaid
Y
,
Tarbell
K
:
Regulatory T cells in the control of host-microorganism interactions.
Annu Rev Immunol
2009
;
27
:
551
89
105.
Sun
J
,
Han
ZB
,
Liao
W
,
Yang
SG
,
Yang
Z
,
Yu
J
,
Meng
L
,
Wu
R
,
Han
ZC
:
Intrapulmonary delivery of human umbilical cord mesenchymal stem cells attenuates acute lung injury by expanding CD4+CD25+ Forkhead Boxp3 (FOXP3)+ regulatory T cells and balancing anti- and pro-inflammatory factors.
Cell Physiol Biochem
2011
;
27
:
587
96
106.
Heuer
JG
,
Zhang
T
,
Zhao
J
,
Ding
C
,
Cramer
M
,
Justen
KL
,
Vonderfecht
SL
,
Na
S
:
Adoptive transfer of in vitro-stimulated CD4+CD25+ regulatory T cells increases bacterial clearance and improves survival in polymicrobial sepsis.
J Immunol
2005
;
174
:
7141
6
107.
Kühlhorn
F
,
Rath
M
,
Schmoeckel
K
,
Cziupka
K
,
Nguyen
HH
,
Hildebrandt
P
,
Hünig
T
,
Sparwasser
T
,
Huehn
J
,
Pötschke
C
,
Bröker
BM
:
Foxp3+ regulatory T cells are required for recovery from severe sepsis.
PLoS One
2013
;
8
:
e65109
108.
Kavanagh
H
,
Mahon
BP
:
Allogeneic mesenchymal stem cells prevent allergic airway inflammation by inducing murine regulatory T cells.
Allergy
2011
;
66
:
523
31
109.
Engela
AU
,
Hoogduijn
MJ
,
Boer
K
,
Litjens
NH
,
Betjes
MG
,
Weimar
W
,
Baan
CC
:
Human adipose-tissue derived mesenchymal stem cells induce functional de-novo regulatory T cells with methylated FOXP3 gene DNA.
Clin Exp Immunol
2013
;
173
:
343
54
110.
English
K
,
Ryan
JM
,
Tobin
L
,
Murphy
MJ
,
Barry
FP
,
Mahon
BP
:
Cell contact, prostaglandin E(2) and transforming growth factor beta 1 play non-redundant roles in human mesenchymal stem cell induction of CD4+CD25(High) forkhead box P3+ regulatory T cells.
Clin Exp Immunol
2009
;
156
:
149
60
111.
Corcione
A
,
Benvenuto
F
,
Ferretti
E
,
Giunti
D
,
Cappiello
V
,
Cazzanti
F
,
Risso
M
,
Gualandi
F
,
Mancardi
GL
,
Pistoia
V
,
Uccelli
A
:
Human mesenchymal stem cells modulate B-cell functions.
Blood
2006
;
107
:
367
72
112.
Liu
L HH
,
Liu
A
,
Xu
J
,
Han
J
,
Chen
Q
,
Hu
S
,
Xu
X
,
Huang
Y
,
Guo
F
,
Yang
Y
,
Qiu
H
:
Therapeutic effects of bone marrow-derived mesenchymal stem cells in models of pulmonary and extrapulmonary acute lung injury.
Cell Transplant
2015
;
24
:
14
113.
Xu
G
,
Zhang
L
,
Ren
G
,
Yuan
Z
,
Zhang
Y
,
Zhao
RC
,
Shi
Y
:
Immunosuppressive properties of cloned bone marrow mesenchymal stem cells.
Cell Res
2007
;
17
:
240
8
114.
Sung
DK
,
Chang
YS
,
Sung
SI
,
Yoo
HS
,
Ahn
SY
,
Park
WS
:
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
36
115.
Nemeth
K
,
Keane-Myers
A
,
Brown
JM
,
Metcalfe
DD
,
Gorham
JD
,
Gorham
JD
,
Bundoc
VG
,
Bundoc
VG
,
Hodges
MG
,
Jelinek
I
,
Madala
S
,
Karpati
S
,
Mezey
E
:
Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma.
Proc Natl Acad Sci USA
2010
;
107
:
5652
7
116.
De Miguel
MP
,
Fuentes-Julián
S
,
Blázquez-Martínez
A
,
Pascual
CY
,
Aller
MA
,
Arias
J
,
Arnalich-Montiel
F
:
Immunosuppressive properties of mesenchymal stem cells: advances and applications.
Curr Mol Med
2012
;
12
:
574
91
117.
Ortiz
LA
,
Dutreil
M
,
Fattman
C
,
Pandey
AC
,
Torres
G
,
Go
K
,
Phinney
DG
:
Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury.
Proc Natl Acad Sci USA
2007
;
104
:
11002
7
118.
Bruno
S
,
Grange
C
,
Collino
F
,
Deregibus
MC
,
Cantaluppi
V
,
Biancone
L
,
Tetta
C
,
Camussi
G
:
Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury.
PLoS One
2012
;
7
:
e33115
119.
Wang
X GH
,
Qin
D
,
Yang
L
,
Huang
W
,
Essandoh
K
,
Wang
Y
,
Caldwell
CC
,
Peng
T
,
Zingarelli
B
,
Fan
GC
:
Exosomal miR-223 contributes to mesenchymal stem cell-elicited cardioprotection in polymicrobial sepsis.
Sci Rep
2015
;
5
:
16
120.
Nong
K
,
Wang
W
,
Niu
X
,
Hu
B
,
Ma
C
,
Bai
Y
,
Wu
B
,
Wang
Y
,
Ai
K
:
Hepatoprotective effect of exosomes from human-induced pluripotent stem cell-derived mesenchymal stromal cells against hepatic ischemia-reperfusion injury in rats.
Cytotherapy
2016
;
18
:
1548
59
121.
Delarosa
O
,
Dalemans
W
,
Lombardo
E
:
Toll-like receptors as modulators of mesenchymal stem cells.
Front Immunol
2012
;
3
:
182
122.
Waterman
RS
,
Tomchuck
SL
,
Henkle
SL
,
Betancourt
AM
:
A new mesenchymal stem cell (MSC) paradigm: Polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype.
PLoS One
2010
;
5
:
e10088
123.
Mei
SH
,
McCarter
SD
,
Deng
Y
,
Parker
CH
,
Liles
WC
,
Stewart
DJ
:
Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1.
PLoS Med
2007
;
4
:
e269
124.
He
H
,
Liu
L
,
Chen
Q
,
Liu
A
,
Cai
S
,
Yang
Y
,
Lu
X
,
Qiu
H
:
Mesenchymal stem cells overexpressing angiotensin-converting enzyme 2 rescue lipopolysaccharide-induced lung injury.
Cell Transplant
2015
;
24
:
1699
715
125.
Zhao
YF LY
,
Xiong
W
,
Ding
W
,
Li
YR
,
Zhao
W
,
Zeng
HZ
,
Gao
HC
,
Wu
XL
:
Mesenchymal stem cell-based FGF2 gene therapy for acute lung injury induced by lipopolysaccharide in mice.
Eur Rev Med Pharmacol Sci
2015
;
19
:
9
126.
Chen
J LC
,
Gao
X
,
Li
C
,
Liang
Z
,
Yu
L
,
Li
Y
,
Xiao
X
,
Chen
L
:
Keratinocyte growth factor gene delivery via mesenchymal stem cells protects against lipopolysaccharide-induced acute lung injury in mice.
PLoS One
2013
;
8
:
11
127.
Han
J LX
,
Zou
L
,
Xu
X
,
Qiu
H
:
E-Prostanoid 2 receptor overexpression promotes mesenchymal stem cell attenuated lung injury.
Hum Gene Ther
2016
;
27
:
10
128.
Cai
SX LA
,
Chen
S
,
He
HL
,
Chen
QH
,
Xu
JY
,
Pan
C
,
Yang
Y
,
Guo
FM
,
Huang
YZ
,
Liu
L
,
Qiu
HB
:
The orphan receptor tyrosine kinase ROR2 facilitates MSCs to repair lung injury in ARDS animal model.
Cell Transplant
2016
;
25
:
1561
74
129.
Galstian
GM
,
Parovichnikova
EN
,
Makarova
PM
,
Kuzmina
LA
,
Troitskaya
VV
,
Gemdzhian
E
,
Drize
NI
,
Savchenko
VG
:
The results of the Russian clinical trial of mesenchymal stromal cells (MSCs) in severe neutropenic patients (pts) with septic shock (SS) (RUMCESS trial).
Blood
2015
;
126
:
2220
130.
Lee
ST
,
Jang
JH
,
Cheong
JW
,
Kim
JS
,
Maemg
HY
,
Hahn
JS
,
Ko
YW
,
Min
YH
:
Treatment of high-risk acute myelogenous leukaemia by myeloablative chemoradiotherapy followed by co-infusion of T cell-depleted haematopoietic stem cells and culture-expanded marrow mesenchymal stem cells from a related donor with one fully mismatched human leucocyte antigen haplotype.
Br J Haematol
2002
;
118
:
1128
31
131.
Le Blanc
K
,
Rasmusson
I
,
Sundberg
B
,
Götherström
C
,
Hassan
M
,
Uzunel
M
,
Ringdén
O
:
Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells.
Lancet
2004
;
363
:
1439
41
132.
Martin
PJ
,
Uberti
JP
,
Soiffer
RJ
,
Klingemann
H
,
Waller
EK
,
Daly
AS
,
Herrmann
RP
,
Kebriaei
P
:
Prochymal improves response rates in patients with steroid-refractory acute graft versus host disease (SR-GVHD) involving the liver and gut: Results of a randomized, placebo-controlled, multicenter phase III trial in GVHD.
Biology of Blood and Marrow Transplantation
2010
;
16
:
S169
S170
133.
Le Blanc
K
,
Mougiakakos
D
:
Multipotent mesenchymal stromal cells and the innate immune system.
Nat Rev Immunol
2012
;
12
:
383
96
134.
Bernardo
ME
,
Ball
LM
,
Cometa
AM
,
Roelofs
H
,
Zecca
M
,
Avanzini
MA
,
Bertaina
A
,
Vinti
L
,
Lankester
A
,
Maccario
R
,
Ringden
O
,
Le Blanc
K
,
Egeler
RM
,
Fibbe
WE
,
Locatelli
F
:
Co-infusion of ex vivo-expanded, parental MSCs prevents life-threatening acute GVHD, but does not reduce the risk of graft failure in pediatric patients undergoing allogeneic umbilical cord blood transplantation.
Bone Marrow Transplant
2011
;
46
:
200
7
135.
Ringdén
O
,
Uzunel
M
,
Rasmusson
I
,
Remberger
M
,
Sundberg
B
,
Lönnies
H
,
Marschall
HU
,
Dlugosz
A
,
Szakos
A
,
Hassan
Z
,
Omazic
B
,
Aschan
J
,
Barkholt
L
,
Le Blanc
K
:
Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease.
Transplantation
2006
;
81
:
1390
7
136.
Kebriaei
P
,
Isola
L
,
Bahceci
E
,
Holland
K
,
Rowley
S
,
McGuirk
J
,
Devetten
M
,
Jansen
J
,
Herzig
R
,
Schuster
M
,
Monroy
R
,
Uberti
J
:
Adult human mesenchymal stem cells added to corticosteroid therapy for the treatment of acute graft-versus-host disease.
Biol Blood Marrow Transplant
2009
;
15
:
804
11
137.
Galipeau
J
:
The mesenchymal stromal cells dilemma–does a negative phase III trial of random donor mesenchymal stromal cells in steroid-resistant graft-versus-host disease represent a death knell or a bump in the road?
Cytotherapy
2013
;
15
:
2
8
138.
Nasseri
BA
,
Ebell
W
,
Dandel
M
,
Kukucka
M
,
Gebker
R
,
Doltra
A
,
Knosalla
C
,
Choi
YH
,
Hetzer
R
,
Stamm
C
:
Autologous CD133+ bone marrow cells and bypass grafting for regeneration of ischaemic myocardium: The Cardio133 trial.
Eur Heart J
2014
;
35
:
1263
74
139.
Weiss
DJ
,
Casaburi
R
,
Flannery
R
,
LeRoux-Williams
M
,
Tashkin
DP
:
A placebo-controlled, randomized trial of mesenchymal stem cells in COPD.
Chest
2013
;
143
:
1590
8
140.
Dominici
M
,
Le Blanc
K
,
Mueller
I
,
Slaper-Cortenbach
I
,
Marini
F
,
Krause
D
,
Deans
R
,
Keating
A
,
Prockop
DJ
,
Horwitz
E
:
Minimal criteria for defining multipotent mesenchymal stromal cells: The International Society for Cellular Therapy position statement.
Cytotherapy
2006
;
8
:
315
7
141.
Lee
RH
,
Hsu
SC
,
Munoz
J
,
Jung
JS
,
Lee
NR
,
Pochampally
R
,
Prockop
DJ
:
A subset of human rapidly self-renewing marrow stromal cells preferentially engraft in mice.
Blood
2006
;
107
:
2153
61
142.
Reger
RL
,
Prockop
DJ
:
Should publications on mesenchymal stem/progenitor cells include in-process data on the preparation of the cells?
Stem Cells Transl Med
2014
;
3
:
632
5
143.
Viswanathan
S
,
Keating
A
,
Deans
R
,
Hematti
P
,
Prockop
D
,
Stroncek
DF
,
Stacey
G
,
Weiss
DJ
,
Mason
C
,
Rao
MS
:
Soliciting strategies for developing cell-based reference materials to advance mesenchymal stromal cell research and clinical translation.
Stem Cells Dev
2014
;
23
:
1157
67
144.
Halleux
C
,
Sottile
V
,
Gasser
JA
,
Seuwen
K
:
Multi-lineage potential of human mesenchymal stem cells following clonal expansion.
J Musculoskelet Neuronal Interact
2001
;
2
:
71
6
145.
Takahashi
K
,
Yamanaka
S
:
A decade of transcription factor-mediated reprogramming to pluripotency.
Nat Rev Mol Cell Biol
2016
;
17
:
183
93
146.
Yun
YI
,
Park
SY
,
Lee
HJ
,
Ko
JH
,
Kim
MK
,
Wee
WR
,
Reger
RL
,
Gregory
CA
,
Choi
H
,
Fulcher
SF
,
Prockop
DJ
,
Oh
JY
:
Comparison of the anti-inflammatory effects of induced pluripotent stem cell-derived and bone marrow-derived mesenchymal stromal cells in a murine model of corneal injury.
Cytotherapy
2017
;
19
:
28
35
147.
Sarugaser
R
,
Hanoun
L
,
Keating
A
,
Stanford
WL
,
Davies
JE
:
Human mesenchymal stem cells self-renew and differentiate according to a deterministic hierarchy.
PLoS One
2009
;
4
:
e6498
148.
Rubio
D
,
Garcia
S
,
Paz
MF
,
De la Cueva
T
,
Lopez-Fernandez
LA
,
Lloyd
AC
,
Garcia-Castro
J
,
Bernad
A
:
Molecular characterization of spontaneous mesenchymal stem cell transformation.
PLoS One
2008
;
3
:
e1398
149.
François
M
,
Copland
IB
,
Yuan
S
,
Romieu-Mourez
R
,
Waller
EK
,
Galipeau
J
:
Cryopreserved mesenchymal stromal cells display impaired immunosuppressive properties as a result of heat-shock response and impaired interferon-γ licensing.
Cytotherapy
2012
;
14
:
147
52
150.
Alcayaga-Miranda
F
,
Cuenca
J
,
Martin
A
,
Contreras
L
,
Figueroa
FE
,
Khoury
M
:
Combination therapy of menstrual derived mesenchymal stem cells and antibiotics ameliorates survival in sepsis.
Stem Cell Res Ther
2015
;
6
:
199
151.
Maggini
J
,
Mirkin
G
,
Bognanni
I
,
Holmberg
J
,
Piazzón
IM
,
Nepomnaschy
I
,
Costa
H
,
Cañones
C
,
Raiden
S
,
Vermeulen
M
,
Geffner
JR
:
Mouse bone marrow-derived mesenchymal stromal cells turn activated macrophages into a regulatory-like profile.
PLoS One
2010
;
5
:
e9252
152.
Gregory
CA
,
Ylostalo
J
,
Prockop
DJ
:
Adult bone marrow stem/progenitor cells (MSCs) are preconditioned by microenvironmental “niches” in culture: A two-stage hypothesis for regulation of MSC fate.
Sci STKE
2005
;
2005
:
pe37
153.
Eliopoulos
N
,
Stagg
J
,
Lejeune
L
,
Pommey
S
,
Galipeau
J
:
Allogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice.
Blood
2005
;
106
:
4057
65
154.
National Institutes of Health. ClinicalTrials.gov
.
Available at: https://clinicaltrials.gov. Accessed July 19, 2017
.
155.
Prescott
HC
,
Calfee
CS
,
Thompson
BT
,
Angus
DC
,
Liu
VX
:
Toward smarter lumping and smarter splitting: Rethinking strategies for sepsis and acute respiratory distress syndrome clinical trial design.
Am J Respir Crit Care Med
2016
;
194
:
147
55
156.
Man
M
,
Close
SL
,
Shaw
AD
,
Bernard
GR
,
Douglas
IS
,
Kaner
RJ
,
Payen
D
,
Vincent
JL
,
Fossceco
S
,
Janes
JM
,
Leishman
AG
,
O’Brien
L
,
Williams
MD
,
Garcia
JG
:
Beyond single-marker analyses: Mining whole genome scans for insights into treatment responses in severe sepsis.
Pharmacogenomics J
2013
;
13
:
218
26
157.
Ranieri
VM
,
Thompson
BT
,
Barie
PS
,
Dhainaut
JF
,
Douglas
IS
,
Finfer
S
,
Gårdlund
B
,
Marshall
JC
,
Rhodes
A
,
Artigas
A
,
Payen
D
,
Tenhunen
J
,
Al-Khalidi
HR
,
Thompson
V
,
Janes
J
,
Macias
WL
,
Vangerow
B
,
Williams
MD
;
PROWESS-SHOCK Study Group
:
Drotrecogin alfa (activated) in adults with septic shock.
N Engl J Med
2012
;
366
:
2055
64
158.
Wong
HR
,
Cvijanovich
N
,
Lin
R
,
Allen
GL
,
Thomas
NJ
,
Willson
DF
,
Freishtat
RJ
,
Anas
N
,
Meyer
K
,
Checchia
PA
,
Monaco
M
,
Odom
K
,
Shanley
TP
:
Identification of pediatric septic shock subclasses based on genome-wide expression profiling.
BMC Med
2009
;
7
:
34