Critically ill patients often suffer from multiple organ failures involving lung, kidney, liver, or brain. Genomic, proteomic, and metabolomic approaches highlight common injury mechanisms leading to acute organ failure. This underlines the need to focus on therapeutic strategies affecting multiple injury pathways. The use of adult stem cells such as mesenchymal stem or stromal cells (MSC) may represent a promising new therapeutic approach as increasing evidence shows that MSC can exert protective effects following injury through the release of promitotic, antiapoptotic, antiinflammatory, and immunomodulatory soluble factors. Furthermore, they can mitigate metabolomic and oxidative stress imbalance. In this work, the authors review the biological capabilities of MSC and the results of clinical trials using MSC as therapy in acute organ injuries. Although preliminary results are encouraging, more studies concerning safety and efficacy of MSC therapy are needed to determine their optimal clinical use. (Anesthesiology 2014; 121:1099-121)

IN the intensive care unit (ICU), the care of patients with acute organ injuries leading to organ failure remains challenging. Organ failure was defined by the 1991 Consensus Conference of the American College of Chest Physicians and the Society of Critical Care Medicine as “the presence of altered organ functions in an acutely ill patient such that homeostasis cannot be maintained without intervention.”1  This disorder represents a dynamic continuum of change over time.2  Multiple organ dysfunction syndrome (MODS) can lead to a mortality rate of 60% after severe trauma, 40% in sepsis, 50% in pancreatitis, 30% in burn injury, and 30% in patients admitted postcardiac arrest.3  The higher the number of failed organs, the higher the mortality.4  In the context of single organ injury without MODS, acute kidney injury (AKI),5  acute respiratory distress syndrome (ARDS),6  and acute liver failure (ALF)7  are responsible for up to 60%, 40%, and 30% of mortality, respectively.

The underlying mechanisms leading to cell death in organ injury are diverse: the proinflammatory nuclear factor-kappa B pathway, endothelial activation with coagulation disorders, lipid mediators, microcirculatory dysfunction, and ischemia–reperfusion (I/R) injury including oxidative stress (OS)-, metabolomic disruption-, and proapoptotic-induced injuries. Aside from the diversity, many mechanisms are also dependent on the sequence in time of injury and/or are organ specific. For instance, nuclear factor-kappa B pathway can be either damaging in the acute phase of sepsis, and/or can be involved in the repair process during the resolution phase of injury. Similarly, the function of phagocytes is dual-faced. Although beneficial in sepsis by clearing pathogens, macrophages can also generate neuron damage through phagocytosis and apoptosis.

This complexity probably explains in part why treatment strategies geared toward a single pathway and/or during a specific time point have failed, highlighting the limited therapeutic strategies available to clinicians to target the multiorgan injuries which may result, aside from the treatment of the initial cause of injury. Clinical management currently focuses on supporting failed organs until they recover, a period where patients may be exposed to new iatrogenic complications.3  Consequently, innovative therapies are needed. Therapeutic use of adult stem cells may be one of them. Stem cells are undifferentiated precursor cells capable of self-renewal and multilineage differentiation. They are classified by their potency (pluripotent vs. multipotent) and origin (adult vs. embryonic). Adult stem cells include hematopoietic stem cells, mesenchymal stem cells (MSC), endothelial progenitor cells, and organ specific stem cells. Although originally the beneficial effect of adult stem cells was thought to be through engraftment and regeneration,8  subsequent studies demonstrated the main therapeutic effects were mediated primarily through the secretion of soluble factors.

In this review, we focused on the potential therapeutic use of human MSC for acute organ injury, specifically in ARDS, AKI, ALF, acute brain injury encompassing stroke and traumatic brain injury (TBI), sepsis, and MODS. To accomplish this goal, we searched PubMed for relevant studies published over the past 10 yr (2003–2013) and the proceedings of major relevant conferences, clinical trial databases, the reference lists of identified trials, and major reviews. In this work, we decided to use the term “organ failure” and “organ injury” to define respectively the altered functional outcomes and the tissue lesions leading to this alteration in the corresponding organ.

MSC are adult nonhematopoietic precursor cells derived from a variety of tissues such as the bone marrow, adipose tissue, and placenta. The definition of MSC by the International Society of Cellular Therapy in 2006 is based on three criteria: (1) MSC must be adherent to plastic under standard tissue culture conditions; (2) MSC must express certain cell surface markers such as CD73, CD90, and CD105, but must not express CD45, CD34, CD14, or CD11b; and (3) MSC must have the capacity to differentiate into mesenchymal lineages including osteoblasts, adipocytes, and chondroblasts under in vitro conditions.9 

Engraftment versus Paracrine Effects

Therapeutic properties of MSC were originally thought to derive from their engraftment in the organ of injury and regeneration. However, subsequent in vivo studies demonstrated limited replacement of damaged tissue by transdifferentiated stem cells (<5%). Thus, the role of paracrine soluble factors with its endocrine actions were studied as potential mechanisms mediating the therapeutic effects.10–13  Despite the transient presence of MSC in the injured organ, ranging from several hours to several days,14,15  MSC are able to exert complex paracrine and endocrine actions, through the secretion of growth factors and cytokines.12  Moreover, recent in vivo studies also underscore the new potential role of microvesicles, small (50 to 200 nm) anuclear membrane bound particles released from MSC as a paracrine vehicle to deliver messenger RNA (mRNA), micro RNA or proteins that may reprogram the injured cells or induce secretion of cytoprotective factors.16–21  All these effects have been demonstrated in multiple organ injury models: acute lung injury (ALI),22–24  AKI,14,15,25–27  ALF,28–30  and acute brain injury.31–33 

Mesenchymal Stem Cells Homing Capacity

The ability of stem cells to preferentially traffic to inflammatory sites is thought to play a crucial role in the success of cellular therapy for organ injury. Intravenous or intraarterial infusion of MSC often initially result in the entrapment of the administered cells in organ capillary beds, especially in the lung and liver.34  In non-injured states, intravenous MSC tend to migrate to the bone marrow.35,36  However, following injury, MSC preferentially home to the site of inflammation where they migrate across the inflamed endothelium and enter the injured tissue bed.37–41  MSC trafficking have been shown to be driven by different interactions between chemokines released from the injured tissue and chemokine receptors expressed by MSC. For instance, stromal cell-derived factor-1/CXCR4 pathway, which is up-regulated under ischemic or hypoxic conditions, can mediate the localization of injected MSC into the injured brain or kidneys.42–46  Interaction between CD44 expressed by MSC and hyaluronic acid in the injured tissue, expressed when the extra-cellular matrix is exposed,47,48  is another major pathway.38 

The multiple mechanisms involved in organ injury are diverse. Although organ injuries do not fit into a single common combination of pathways, we will highlight those impacted by MSC.

Acute Proinflammatory Pathway

In addition to “septic” inflammation, a severe inflammatory response can be triggered by non-infectious sources, such as danger associated molecular patterns.49,50  In the acute phase of organ injury, multiple cells express pattern recognition receptors that can recognize either pathogen or danger associated molecular patterns. Pattern recognition receptors sense endogenous and exogenous danger signals and induce proinflammatory cytokines and type I interferons49  (fig. 1). Monocytes-macrophages and polymorphonuclear neutrophils migrate quickly to sites of injury and secrete reactive oxygen species (ROS) and proinflammatory cytokines/chemokines. Antigen-presenting cells also migrate to the site of injury and internalize and process either pathogen or danger-associated molecular patterns and initiate the adaptive immune response. Adaptive immune cells such as natural killer cells, natural killer T cells, mast cells, T-lymphocytes, and B-lymphocytes then converge, participating in the pro or antiinflammatory response. T cells are essential players in the acute and intermediate inflammatory phase of organ injury, bridging together innate and adaptive immunity. CD4+ T helper (Th) cells lead to polarization of the immune response in multiple pathways (Th1, Th2, Th17, Th22, Th3, T-regulatory), and CD8+ T cells are dramatically involved in the cytotoxic response leading to the lysis of the targeted cells. Rather than a patchwork process, acute organ injury is a continuum of responses from innate to adaptive immune cells.

Ischemia–Reperfusion Pathways: Oxidative Stress Injury, Metabolomic Disorders, and Apoptosis

Oxidative stress is caused by increased production of reactive oxygen and nitrogen species or by depletion of protective antioxidants. Resulting oxidative products can damage DNA, promoting cell death/apoptosis and cause end-organ tissue damage. OS is present in many pathological situations, such as during reperfusion after ischemia or following toxic exposures. Whether through low regional blood flow or hypoxemia or both, ischemia is responsible for a dramatic shift in cell metabolism. The lack of oxygen to drive oxidative phosphorylation and other oxygen dependant metabolic reactions (aerobic glycolysis, fatty acid beta oxidation) results in inefficient anaerobic glycolysis as the major source of adenosine triphosphate (ATP) production and leads to ATP deficit.51–54  Proteomic profiling indicate that during ischemia, metabolic key enzymes are decreased.53  The resultant ATP-dependant metabolic reaction shutdown then produces deep imbalance in cellular homeostasis eventually leading to cell death.53,55–57  Furthermore, any reduction in organ perfusion in terms of oxygen delivery51  can lead to organ damage by generating I/R injuries.58  I/R injury is present in most clinical conditions leading to acute organ injury such as shock, hypoxemia, sepsis, cardiac arrest, trauma, burn injuries, or following certain surgeries (cardiac, aortic, and organ transplantation surgeries). Although ischemia-induced tissue hypoxia can lead to irreversible tissue injury if the period of ischemia is prolonged, much of the tissue damage occurs following restoration of perfusion.59,60  While reperfusion can induce mitochondria to generate ATP and restore cell metabolism in less damaged tissue, it can also paradoxically exacerbate ischemia-induced injury in severely ischemic cells leading to release ROS generated by damaged mitochondria and nicotinamide adenine dinucleotide phosphate oxidase.58,61–64  Proteomic profiling show that reperfusion can lead to proglycolytic enzyme depletion, proapoptotic proteome shift, and mitochondrial dysfunction inducing OS.65  These I/R-induced pathways can lead to cell death and organ failure (fig. 2).

Immunomodulatory Properties

MSC can modulate innate and adaptive immune cells, by enhancing antiinflammatory pathways in the injured organ milieu.66–68  This immunomodulation is mediated by cell-contact-dependant and independent mechanisms through the release of soluble factors such as tumor necrosis factor-stimulated gene 6,69  prostaglandin E2,70  interleukin (IL)-10,70,71  IL-1 receptor antagonist,72  transforming growth factor (TGF)-β,73  hepatocyte growth factor73  or indolamine 2,3-dioxygenase.67  Both decrease in proinflammatory mediators (IL-1β, tumor necrosis factor (TNF)-α, interferon-γ, IL-6), and increase in antiinflammatory cytokines (IL-10, basic fibroblast growth factor, TGF-α, TGF-β) have been also pointed out as a key factor in preventing cell damage in acute kidney15,26  and liver30,74–76  injury models. Similar findings have been reported in acute stroke77  and sepsis78,79  animal models (fig. 3A).

Human MSC promote repolarization of monocytes and/or macrophages from a type 1 (proinflammatory) to a type 2 (antiinflammatory) monocyte phenotype characterized by high levels of IL-10 secretion, increased phagocytosis and low levels of TNF-α and interferon-γ production and major histocompatibility class II expression.80–82  This ability of MSC to reprogram monocytes/macrophages has been demonstrated in vivo in different models of sepsis,70,83–85  endotoxin86  or live Escherichia coli bacteria-induced ALI,87,88  ischemia,89  and regenerative medicine.82,90  Often in these injury models, MSC reprogrammed type 2 monocytes produced large quantities of IL-10, which blocked polymorphonuclear neutrophil influx into the injured tissue and prevented further damage (fig. 3A). However, in a mouse model of TBI, intracerebral administered MSC modulated the inflammatory response through decreasing the phagocytic capability of microglia macrophages.91  In this specific context, the reduction of phagocytosis by macrophages was beneficial, leading to better outcomes. These findings revealed the complexity of the crosstalk between MSC and macrophages, which may be organ specific and influenced by the injury milieu.

MSC can interfere with dendritic cells differentiation, maturation, and function, skewing them toward a regulatory phenotype.92,93  Dendritic cells generated in the presence of MSC have decreased capacity to induce activation of T cells, and exhibit an altered cytokine production pattern with lower proinflammatory and higher antiinflammatory cytokines66,92  (fig. 3A).

MSC also modulate natural killer cells, which are involved in both the elimination of virus-infected and damaged cells and the secretion of an array of proinflammatory cytokines such as interferon-γ. Several studies clearly show that MSC, when co-cultured with natural killer cells, impair their cytotoxic activity, cytokine production, and granzyme B release94–96  (fig. 3A). However, other studies have shown that MSC could enhance their proinflammatory phenotype depending on the culture conditions. Thus, the complex interplay between MSC and natural killer cells could result either in a proinflammatory or an antiinflammatory phenotype depending on the type of the activation state of both cells and on the surrounding milieu.67 

In addition, MSC are able to suppress T cell activation and proliferation and decrease their response by shifting them from a T helper (Th)1 to a Th2 immune phenotype.72,73,97,98  MSC have been shown to (1) inhibit the differentiation of naive T cells into Th17 cells,99–101  (2) inhibit secretion of proinflammatory cytokines by differentiated Th17 cells, (3) promote induction of immunosuppressive FoxP3+ T-regulatory cells,100,102  and (4) drive reprogramming of Th17 cells into FoxP3+ T-regulatory cells100  (fig. 3B). MSC also potentially inhibit cytotoxic effect of antigen-primed cytotoxic T cells98  and induce T cell anergy.67,73,103  This T regulatory–skewed response has been also demonstrated in vivo. In an ALI model, Sun et al. showed that MSC could up-regulate T-regulatory cells, reducing some key Th1 cytokines (interferon-γ, TNF-α, macrophage inflammatory protein-2) and increasing Th2 cytokines (IL-10). Others have also demonstrated that MSC decreased proinflammatory cytokines/chemokines such as macrophage inflammatory protein-1, B-lymphocyte chemoattractant, and IL-12, with subsequent decrease in Th cells.104 

Overall, an emerging body of data demonstrates at multiple levels the impact of MSC upon key cells involved in the continuum between innate and adaptive immunity, modulating inflammation in acute organ injury.

Antimicrobial Properties

Studies using bacteria-induced acute organ injury models demonstrated that MSC could exert direct and indirect antimicrobial properties. In E. coli pneumonia in mice, we demonstrated that MSC secreted antibacterial proteins/peptides such as LL-37105  and lipocalin-2,87  leading to improved bacterial clearance. Other antibacterial mechanisms of MSC include tryptophan catabolism by indolamine 2,3-dioxygenase106  or increased pathogen phagocytosis which inhibit overall bacterial growth.79,107–109  Using different in vivo and ex vivo models of sepsis or pneumonia, MSC were found to increase phagocytosis of bacteria by macrophages by switching from a type 1 to type 2 monocyte phenotype.79,87,88,110  In a mouse model of Pseudomonas aeruginosa-induced peritonitis, Krasnodembskaya et al.110  demonstrated that MSC reduced the number of colony-forming units of P. aeruginosa in the blood by increasing the monocyte phagocytic potency. The authors highlighted two potential underlying mechanisms: (1) the up-regulation of phagocytosis receptor CD11b on monocytes and (2) the increase in CD163 and CD206-positive activated monocytes/macrophages in the spleen.110  In a cecal ligation and puncture mice model of sepsis, Nemeth et al. showed a decrease in blood bacteria counts in the MSC treated group. The authors speculated that this increase in blood bacteria clearance could be explained by IL-10-mediated neutrophil retention within the vascular compartment.70  Recently, toll like receptor 3-triggered human MSC were shown to promote polymorphonuclear neutrophil activity, viability and improve its respiratory burst, increasing ROS release which is bactericidal108  (fig. 4).

Antioxidative Effect

Recent studies of organ injuries involving the heart,57  brain,111,112  kidneys,113–115  and liver116–119  demonstrated that MSC could exert an antioxidative effect leading to a decrease in the severity of organ injury.56  This antioxidative property has been best exemplified in sepsis-induced organ failure models. In this context, authors have shown that MSC can reduce neutrophil-mediated oxidative injury in lungs, liver, and kidneys.70,78  This effect was primarily mediated through secretion of soluble factors, which prevent ROS accumulation through enhanced scavenging and antioxidant up-regulation.57,120  Interestingly, many of these studies focused on the adoptive transfer of antioxidant effects from exosomes by stem cells.59,60,121  Similar to microvesicles, exosomes are bilipid membrane vesicles with a diameter less than 50 nm. They can carry a complex cargo of proteins, lipids, DNA, mRNA, or microRNA which could be delivered into targeted cells and impact multiple cellular pathways.16,122  MSC release a large quantity of exosomes in their environment upon diverse stimuli.120  Both in vitro and in vivo studies have shown that MSC derived exosomes can decrease OS-induced injury by reversing the depletion of key enzymes in ROS metabolism and the resultant accumulation of toxic products from the electron transport chain.59,60,65,121,123  For example, the transfer of peroxiredoxins and glutathione S-transferase by MSC derived exosomes into injured cells has been shown.65  In addition, Zhou et al.121  recently demonstrated that the antioxidant effect of exosomes derived from human umbilical cord MSC in a cisplatin-induced AKI model may involve the inhibition of the p38 mitogen-activated protein kinases-caspase 3 pathway (fig. 2).

Metabolomics

Any potential treatment aimed at reversing the metabolomic disorders in acute organ injury should ideally overcome ATP deficit, compensate the proteomic alteration and repair the mitochondrial electron transport chain. Several studies demonstrate some direct beneficial effects from MSC on metabolomics disorders. Beiral et al.124  demonstrated in a rat kidney I/R model that MSC could restore ATP synthesis. In addition, proteomic and genomic profiling of MSC-derived exosomes (Exocarta,125  Vesiclepedia126 ) showed that they contain key enzymes involved in the ATP-generating stage of glycolysis so that they could potentially restore proteomic alterations in injured tissue. Lai et al.60  showed in injured rat cardiomyoblast, that MSC-derived exosomes increased intracellular ATP levels by 75 and 55% after 15 and 30 min, respectively. In an ex vivo myocardial I/R injury model, MSC-derived exosomes increased ATP production in reperfused myocardium.59  And in a model of lipopolysaccharide-induced ALI, Islam et al.127  demonstrated that mitochondrial transfer through connexin-43 may be involved in the restoration of ATP levels (fig. 2).

Promitotic/Antiapoptotic Effects

Multiple groups have studied the underlying mechanisms of MSC antiapoptotic effects in various organ injury models. Two main mechanisms have been proposed. (1) MSC secretion of growth factors. In animal models of AKI,27,128–131  acute stroke,132–135  and traumatic brain injury,91,136,137  a wide array of secreted growth factors such as insulin growth factor-1,128,131,133,134  vascular endothelial growth factor,27,130,135  hepatocyte growth factor,129  brain-derived neutrophic factor,91,132,136,137  nerve growth factor,91,133,134,136,137  and neurotrophin-391,136,137  have been linked to the proregenerative effects mediated by MSC. (2) And increased expression of proregenerative/antiapoptotic genes and/or possibly mRNA transfer to injured cells by MSC or MSC derived microvesicles or exosomes. In ALF, MSC induced over-expression of genes involved in hepatocellular regeneration such as hepatocyte growth factor, epidermal growth factor, transforming growth factor-β, stem cell factor, and tissue metalloproteinase 3.74  In AKI, Bruno et al.18,19  showed that MSC released microvesicles could transfer mRNAs or microRNAs involved in cell proliferation to damaged renal cells.138  In a glycerol-induced AKI model in immunocompromised mice, MSC microvesicles had a proliferative effect in tubular epithelial cells.19  RNAse pretreated microvesicles lost their therapeutic potencies, suggesting a RNA-dependent effect. The underlying mechanisms were mainly attributed to a microvesicle induced up-regulation of antiapoptotic genes (Bcl-xL, Bcl2) and to a down-regulation of apoptotic genes (caspase-1, caspase-8, lymphotoxin-α) in tubular epithelial cells. A similar decrease in apoptotic genes expression (caspase-3 pathway) and up-regulation of phosphorylated protein kinase B prosurvival pathway leading to new neuron generation139,140  were found in TBI treated with MSC.136,141  Finally, the over expression of genes involved in the antiapoptotic pathways (such as growth hormone and insulin growth factor-1 signaling) also played a therapeutic role in a model of sepsis treated with MSC78,79  (fig. 5).

Ischemia–Reperfusion Injury

Several in vivo studies have pointed out the beneficial effects of MSC with respect to I/R of the heart,142  lungs71,104,143–145  brain,146  kidney,15,147,148  and gut.149,150  More specifically, studies focused in I/R-induced ALI model, showed some beneficial effects through a combination of immunomodulation,71,143,145  antioxidant71,143,145  or antiapoptotic143  properties. Others demonstrated that MSC could increase the activity of antioxidant enzymes in I/R.151  Interestingly in a gut I/R model, MSC reduced rat intestinal I/R injury by increasing the expression of the intestinal tight junction protein zona occludens-1 and reducing tight junction disruption by suppressing the action of TNF-α.150  The proteomic alteration in I/R injury65  can be supplemented by the cellular contents of MSC-derived exosomes.60,123  By replenishing depleted glycolytic enzymes, supplementing damaged cells with additional protein components of the cellular antioxidant system, and activating prosurvival phosphatidylinositide 3-kinases/phosphorylated protein kinase B pathway via cluster of differentiation 73, MSC exosomes can increase ATP level and decrease OS and cell death59  (fig. 2).

Given the diversity of mechanisms involved in the potential therapeutic effect of MSC in various organ injuries (fig. 6), we will review the current literature underlying the rationale for the use of MSC in ARDS, AKI, ALF, acute brain injury, and sepsis.

ARDS is major cause of acute respiratory failure in critically ill patients. Despite improvements in supportive care, mortality associated with ARDS remains high, up to 40%, depending on the etiology.152,153  Current treatments remain focused on supportive care such as lung protective ventilation, fluid conservative strategy, and prone positioning.154–156  No pharmacological therapies from preclinical models have yet been translated to effective clinical treatment options. Past studies showed that focusing on either antiinflammatory or antifibrotic pathways were too simplistic as a therapy. Pathophysiology of ARDS involves complex crosstalks between the immune system and the alveolocapillary barrier leading to an excess of proinflammatory Th1 polarized responses, increase in lung protein permeability and formation of pulmonary edema. Pulmonary edema results in impaired gas exchange and eventual hypoxemia.153 

Mesenchymal Stem Cells Lung-specific Mechanism of Action

Aside from their immunomodulatory, antibacterial, antioxidant, and anti-I/R injury properties, MSC can also display some lung specific functional effects.

Alveolar Fluid Clearance.

ARDS is characterized by impaired alveolar fluid clearance (i.e., inability to decrease pulmonary edema) induced by excessive inflammation in the injured alveolar milieu.157  Several studies have demonstrated that MSC secrete keratinocyte growth factor, which increases alveolar fluid clearance by up-regulating key epithelial sodium channel gene expression and Na-K-ATPase activity, or by increasing trafficking of epithelial sodium channel proteins to the apical membrane.158  These keratinocyte growth factor mediated effects were shown in animal models83,159,160  as well as in an ex vivo perfused human88,161  preparation. Most recently, we demonstrated that MSC-derived microvesicles could protect against lipopolysaccharide-induced ALI through delivery of the keratinocyte growth factor mRNA with subsequent expression of the protein in the injured alveolus.21 

Lung Permeability.

In ARDS, the injured lung capillary endothelium leads to protein leakage from the vascular bed into the alveolar space. This phenomenon aggravates the ability of the lung epithelium to reduce pulmonary edema. Recently, MSC have been shown to secrete angiopoietin-1, a soluble factor capable of reducing endothelial permeability through enhanced endothelial survival and vascular stabilization, through the preservation of cell adhesion molecules and cell junctions and the prevention of actin “stress fiber” formation.162  We and others have demonstrated that angiopoietin-1 secreted by human MSC was essential to prevent an increase in lung protein permeability.163–165 

Preclinical Acute Lung Injury Studies

A recent review reported the benefits of administering MSC in preclinical small animal lung injury models.6  More than half of experimental studies concerned intratracheal lipopolysaccharide-induced ALI in rodents and intratracheal administration of MSC, whereas the intravenous route of delivery of MSC was preferred in bleomycin-induced, I/R or ventilator-induced lung injury. The beneficial effects of MSC have also been reported in bacterial-induced ALI models,84,87,88,105  such as pneumonia, peritonitis, and sepsis from cecal ligation and puncture, highlighting the antibacterial properties of MSC. Gupta et al.87  found a survival advantage from syngeneic mouse MSC in an E. coli bacterial pneumonia-induced ALI model. Lee et al.88  also showed beneficial effects of MSC in E. coli bacterial-induced ALI in an ex vivo perfused human lung preparation. Although this model excluded other systemic organs, which may generate an inflammatory response, it replicated many of the injury patterns seen in patients with ARDS. Aside from bone marrow, other sources of MSC have been studied. Human umbilical cord-derived MSC is currently being investigated in clinical trials, due to their accessibility (from the placenta), lack of ethical concerns, and their faster population doubling time.84,102,166  Although promising, adipose derived human MSC104,145  require further studies to clarify their potential therapeutic effects in ALI.

In ALI models in rodents, the mean dose of MSC typically was 20 to 30 × 106 cells/kg, and the timing of administration was within 6 h following ALI. The maximum therapeutic effect of MSC was found 2 to 3 days following administration. One study using an ALI model in mice with a large dose of MSC (889 × 106 cells/kg) showed a delayed effect on day 28.167  However, no dose response study has been yet published. Thus, it is still unclear whether there is a therapeutic ceiling or if a second dose of MSC is needed, especially during the resolution phase of ALI. Aside from the role of paracrine soluble factors, the role of MSC microvesicles or exosomes has been recently studied. Lee et al.20  found that murine MSC-derived exosomes could prevent hypoxic pulmonary hypertension by reducing vascular remodeling, pulmonary influx of macrophages, and proinflammatory and proliferative mediators. More recently, we demonstrated that human MSC microvesicles can reduce the severity of E. coli endotoxin-induced ALI in mice through the transfer of keratinocyte growth factor mRNA to the injured lung epithelium.21  These recent findings shed first light on a new stem cell-free therapy in ALI, circumventing caveats of MSC use such as genetic instability and potential malignant transformation.

Clinical Trials

Despite these multiple encouraging preclinical studies, translation into human clinical trials remains limited. Currently, two phase I/II clinical trials are underway (table, Supplemental Digital Content 1, https://links.lww.com/ALN/B92, which lists the ongoing clinical trials). One Phase I/II study (NCT01775774) uses human bone marrow-derived MSC (BM-MSC) in ARDS patients. The aim of this multi-center, single group assignment study is to assess the safety and then the feasibility of using escalating intravenous doses (1 to 10 × 106 cells/kg) of allogeneic human BM-MSC in patients with moderate or severe ARDS. Another randomized, double blind, placebo-controlled trial (NCT01902082), targets not only safety but also efficacy outcomes, using allogeneic adipose-derived MSC. In both studies, inclusion criteria are similar, the intravenous route is used and MSC therapeutic doses vary from 1 to 10 × 106 cells/kg. Both trials are still recruiting.

AKI is a clinical syndrome characterized by rapid loss of excretory function leading to accumulation of products of nitrogen metabolism and metabolic acids, increased potassium and phosphate serum concentration and decreased urine output. Incidence varies from 5,000 cases per million people per year for non-dialysis-requiring AKI to 295 cases per million people per year for dialysis-requiring disease.168  In critically ill patients, the AKI prevalence reaches 40% at admission to the ICU if sepsis is present169  and 60% during ICU stay.170  No pharmacological therapies are available. Treatment is essentially supportive, including renal replacement therapy if needed. Mortality from AKI ranges from 44.7 to 53% in critically ill patients.171  Most patients who survive recover their renal function ad integrum after a few weeks. However, some remain in chronic renal failure requiring definitive renal replacement therapy.

Etiology of clinical AKI is often multifactorial involving diverse triggers such as hypovolemia, ischemia, I/R, sepsis, and toxic injuries. Most of the AKI seen in the ICU, occur within 72 h from a combination of pre renal and renal injuries.171  Most existing preclinical animal AKI models use ischemia induced by acute occlusion of the renal artery.172–174  Although not wholly clinically relevant, these ischemic AKI models do imitate several activated pathways involved in AKI, such as coagulation system activation,175  leukocyte infiltration,176  endothelium injury177  with over-expression of adhesion molecules,178  cytokines release,179  Toll-like receptors induction,180  intrarenal vasoconstriction pathway, and apoptosis.181  In addition, in septic and hepatorenal preclinical AKI models, triggered by a decrease in blood pressure secondary to a systemic or hepatosplanchnic vasodilation,182  the renal sympathetic system,183  the renin–angiotensin–aldosterone system,184  and the tubuloglomerular feedback system184  are all activated. Depending on the intensity and the period of time of their association, these different factors contribute to a continuum ranging from tubular injuries to apoptosis/necrosis to renal failure.171 

Preclinical Acute Kidney Injury Studies

MSC therapy is effective in reducing AKI in diverse experimental models including those induced by cisplatin,19,25,38,128,130,185–190  glycerol,19,38  and I/R injury.14,15,26,129,191–193  Systemic route of administration is widely used via intravenous or intraperitoneal injection, except for I/R model where MSC are infused intraarterial.14,15,26,129,191–193  Delivered doses range from 8 × 106  to 2 × 108  cells/kg.186,187 In cisplatin-induced AKI models, MSC prevented renal function impairment, improved renal function and preserved tubular integrity,25,128  leading to an increase in the survival rate of mice following cisplatin injection188–190  compared with saline control. Interestingly, Morigi et al.188,189  found that, in the cisplatin-AKI model, cord blood derived MSC were more effective than BM-MSC in terms of renal function improvement and survival, whereas MSC derived from human adipose tissue did not improve renal function.194  In addition, mice treated by human adipose tissue-derived MSC showed some tubular alterations such as casts, nuclear fragmentations and necrosis. However, because these histological alterations are similar to those observed in a cisplatin-induced AKI, these lesions could not be interpreted as being harmful effects of MSC.

In a lethal AKI model induced by cisplatin administration, Bruno et al.18  showed MSC microvesicles could enhance survival in immunocompromised mice. In this model, a single administration of microvesicles increased survival rate and ameliorated renal failure but did not prevent chronic tubular injury. However, multiple injections of microvesicles not only improved survival but also normalized histology and renal function at day 21.

Clinical Trials

Despite strong preclinical evidence of the therapeutic effect of MSC in AKI, only three Phase I/II clinical trials have been carried out12,195,196  (table, Supplemental Digital Content 1, https://links.lww.com/ALN/B92, which lists the ongoing clinical trials). One ongoing trial (NCT00733876)12,196  aims to investigate safety and efficacy of allogeneic MSC in preventing and treating AKI following on-pump coronary artery bypass surgery, using suprarenal aortic MSC infusion. Patients at high risk of postoperative acute renal failure patients are included. Preliminary data from the trial indicates that MSC infusion is safe and feasible. Moreover, MSC infusion prevented any postoperative renal failure (0% vs. 20% AKI incidence compared with case control) and reduced by 40% the length of hospital stay and readmission rates.12,196  A double-blind, placebo controlled, multicenter phase II trial is planned by the same investigators. Another clinical trial used allogeneic human BM-MSC in a multicenter, double-blind, placebo-controlled phase II study (NCT01602328) in patients with post cardiopulmonary bypass-induced AKI. Safety and also efficacy outcomes such as time to kidney recovery and dialysis were the primary aims. The third ongoing pilot study (NCT01275612)195  investigates the safety and the feasibility of systemic infusion of donor ex vivo-expanded MSC in cisplatin-induced acute renal failure in chemotherapy treated patients with solid organ cancer. Preliminary data from these clinical trials are pending.

ALF still remains a leading cause of death in 30% of the cases.7  Principal etiologies include acetaminophen-induced injury, idiosyncratic drug induced liver injury, viral hepatitis, autoimmune hepatitis, Budd-Chiari syndrome, and Wilson disease. Up to 15% of the etiology of ALF are indeterminate. Depending on the cause, spontaneous recovery may vary from 30 to 60%.7  However, supportive therapies in ALF are dramatically limited and liver transplantation remains the gold standard for treating end-stage liver failure.7,197 

In ALF, innate immunity with its resultant inflammatory cascade is activated. Uncontrolled hepatic inflammation with clinically high serum levels of proinflammatory cytokines such as IL-1, TNF-α, IL-6, and IL-8 have been reported198–200  with resultant hepatic cytotoxicity.201  Necrosis and/or apoptosis may also take an important part in the loss of hepatic function, overwhelming hepatocyte regeneration.197  I/R and OS injuries can also take place in different causes of ALF such as toxic, post hepatectomy, or post transplantation injury. The prognosis of ALF is directly linked to liver regeneration, which in 40% of the cases can overcome the hepatocyte destruction.

Preclinical Acute Liver Failure Studies

To circumvent organ donor shortage, replacing injured hepatocytes by stem cells initially appeared as the main aim of liver-oriented cell-based therapy. Although several studies showed that MSC can transdifferentiate towards a hepatocyte phenotype in vitro and in vivo,202  the beneficial effects of MSC are more complex, encompassing regenerative,203–207  immunoregulatory,206–208  and anti-OS injury117  pathways.

Most preclinical studies using MSC used mice and rats with carbon tetrachloride,30,76,209–212  thioacetamide,118 d-galactosamine,29,74,75,213,214  or I/R-induced liver injury.116,215,216  However, two studies used d-galactosamine induced fulminant hepatic failure in pigs.29,213  Therapeutic dose ranged from 2 to 10 × 106 cells/kg.217  Most of the studies used intravenous MSC administration, but others chose the intraportal route29,215  aiming at circumventing trapping in the pulmonary circulation.215  Overall, MSC decreased the severity of histological liver injury,74,75,210,213–215  improved liver function,75,210–212,215  and finally enhanced survival.29,74,75,210,213–215  In contrast, Boeykens et al.218  did not find any beneficial effects of intraportally administrated MSC in terms of improved liver recovery. However, the authors used MSC in a complex liver injury model, combining a partial hepatectomy in a previously steatotic liver which may not be applicable to ALF. Regardless all these promising findings, no clinical trial has been carried out in this field.

Stroke causes 15 million death worldwide every year.219  In the United States of America, it remains the leading cause of disability and the third leading cause of mortality behind cardiovascular disease and cancer.220  Currently, tissue plasminogen activator administration within 4.5 h of the onset of ischemia is the only validated treatment for ischemic stroke. Alternate or complementary therapeutics are urgently needed.

In acute stroke, reduction in the oxygen and glucose supplies lead to neuronal cell death through several mechanisms including intracellular calcium movement and energetic metabolism impairment.221–224  Secondary, restoration of the cerebral blood flow leads to I/R. As in the other organs, I/R injury in the brain triggers ROS production as well as proinflammatory pathways.225,226  Microglia cells secrete proinflammatory cytokines such as IL-6, TNF-α, and IL-1β.227,228  Taken together, all these mechanisms increase neuronal cell damage.

Preclinical Stroke Studies

Most preclinical animal studies using MSC have involved rodent, preferentially rats, in models of middle cerebral artery occlusion. Although some teams carried out a permanent occlusion model,77,134,229–232  most of the studies used a transient middle cerebral artery occlusion model ranging from 90 to 120 min of ischemic time. Three routes of MSC administration have been investigated: intracerebral,77,132,229,231–236  intracarotid,237,238  and intravenous.133–135,230,239–242 Time of treatment delivery after stroke varied from 2 h236  to 1 month.32  Both routes of MSC administration, intracerebral132,233–236  or intravenous,243  decreased infarct size and improved neurological outcomes in rats. Either intravenous or intracarotid administration of MSC also improved behavioral outcomes.244  However, it remains unclear which route, intracerebral243  or intravenous route,245  is more efficacious. The MSC doses range from 4 × 105  to 1.2 × 108  cells/kg depending on the model.77,240 A relation between cell dose and efficacy have been demonstrated with both neurological outcomes240  and neurotrophic factors secretion.77 

Clinical Trials

Based on the accumulation of these preclinical studies, clinical trials using MSC in stroke have increased dramatically. The number of clinical trials involving MSC in stroke (ischemic, hemorrhagic, acute, subacute, or chronic) rose from one completed phase I study in 2009246,247  to 22 phase I/II clinical trials.248  Bang et al.246  carried out the first phase I study for assessing feasibility and safety of intravenous administration of 108 autologous MSC in patients with severe neurological deficits due to subacute ischemic stroke. Five patients were included in the treatment group versus 25 in the control group. Although intravenous cell infusion appeared safe and feasible, the small sample size in the treated group and the non-blinded design of this study prevented any conclusions concerning the potential therapeutic benefits of MSC on neurological outcomes. Five years later, the same authors published a randomized placebo-controlled long-term follow-up study carried out on 52 subacute ischemic stroke patients.249  In this study, 16 patients were included in the intravenous MSC group. No difference was observed between groups concerning adverse events. More importantly, some of the neurological recovery scores were improved in the MSC group compared with the placebo group. Currently nine studies are underway to investigate the effect of intravenous or intraarterial administration of MSC in acute ischemic stroke patients (table, Supplemental Digital Content 1, https://links.lww.com/ALN/B92, which lists the ongoing clinical trials). All are phase I/II studies except one phase III. Four of the trials use autologous whereas five use allogeneic MSC. Time of MSC administration ranges from 1 day to 6 weeks after the onset of clinical signs of stroke. The therapeutic dose ranges from 1 to 2.5 × 106 cells/kg. Primary outcomes are safety, feasibility, tolerance, improvement of functional recovery assessed by neurological scores, and size of infarct. The maximum follow-up ranges from 1 month to 24 months after MSC administration. Despite the number of clinical trials, little data is yet available to demonstrate the potential therapeutic use of MSC in stroke management. Results of ongoing trials are expected soon, especially long-term safety data and the potential impact of MSC on neurological outcomes.

TBI remains a significant cause of morbidity, mortality, and disability among patients.250  After the initial trauma, multiple pathological pathways converge, generating secondary lesions and leading to increased neuronal cell death and brain damage. These different pathways include increased neurotransmitter release, ROS generation with OS injury, calcium-mediated signaling and increased apoptosis, mitochondrial dysfunction, and proinflammatory response.

Preclinical Traumatic Brain Injury Studies

MSC can both suppress these different injury mechanisms and also express neuronal and glial markers,251  although regeneration may not be a significant therapeutic mechanism. Most preclinical studies in TBI have used BM-MSC,39,136,137,139,252–255  except two studies which used peripheral blood-derived141  or umbilical cord-derived MSC.91  Rats were the most frequent small animal used,39,137,139,141,252–256  although, a few studies with TBI have been performed in mice.91  In these studies, MSC were typically given from 24 h to 7 days following TBI and, doses varied from 6 × 106  to 3.2 × 108  cells/kg depending on the administration route,91,254 which included intravenous39,136,137,139,140,252–255,257  or intracerebral.91,140,141  MSC route of administration in TBI remains controversial. Multiple studies demonstrated that in rat models of TBI, most of the MSC are initially trapped in the lungs, liver, and spleen, leaving a small portion of cells, ranging from 0.0005%256  to 1.4%,33  to cross the blood brain barrier to reach the cerebral parenchyma. Harting et al.256  showed that intravenous MSC treatment failed to improve any motor or cognitive outcomes in a rat TBI model. Although some studies highlighted the beneficial effects of intravenous MSC in TBI,39,136,137,140,253–255  most of studies were from the same experimental team. Interestingly, Mahmood et al.140  compared the intravenous with the intracerebral route of administration of MSC at doses of 3 × 106 and 7 × 106 cells/kg in a rat TBI model. They found differences in terms of localization of the induced neuronal cells proliferation but none regarding neurological functional recovery. Overall, the beneficial effects of MSC have been demonstrated in terms of functional neurologic improvements from 15 to 90 days after TBI.39,136,137,140,141,253–255,257  MSC are believed to migrate into the injured brain parenchyma91,141,255  with a high affinity for the periphery of the lesions,253  leading to a decrease in the contusion volume measured one month after the TBI.91  Possibly due to the small number of published preclinical animal studies and to the unresolved issue of optimal route of delivery, no clinical trial using MSC in TBI have been yet carried out.

Despite decades of clinical trials and improvement in antibiotic and supportive care, sepsis remains a challenging life-threatening disease in critically ill patients and the leading cause of morbidity and mortality in ICU patients.258  In the United States, sepsis is responsible for more than 200,000 patient deaths and utilizes US$17 billion per year.259,260  Sepsis results from a complex host-pathogen interaction leading to a dysregulation of the host response in terms of inflammation and coagulation. Proapoptotic pathways, metabolomic disorders, OS, and I/R injuries are also involved in patients treated for sepsis. Eventually, sepsis can evolve toward septic shock, MODS, and death. Currently, all clinical trials using therapeutics targeting a single specific pathway have failed to demonstrate any clinical benefits261–264  such as high dose corticoids265,266  or activated protein C.267  Consequently, immunomodulatory approach using a multifaceted therapy is required to overcome the inflammatory imbalance. MSC is an attractive approach due to its ability to home to injured sites, mitigate the proinflammatory cascade, modulate multiple immune cell types, promote cell survival, protect against OS injuries, and exhibit some antibacterial properties. In addition, another advantage of cell-based therapy in sepsis is that stem cells can potentially interact with their environment, so that they can adopt some dynamic phenotypes and secrete a variety array of soluble factors depending on the pathological context.268,269 

Preclinical Sepsis Studies

In this review, we have excluded studies using endotoxin-induced injury models of sepsis and focused only on preclinical studies using live bacteria. Although lipopolysaccharide represents one part of the multiple bacterial factors involved in the septic process, these models have obvious limitations.270  Thus, we considered the live bacteria models more clinically relevant. The therapeutic use of MSC has been used in three different sepsis models: cecal ligation and puncture,70,78,79 P. aeruginosa peritonitis,110  and E. coli pneumonia.86,88  The cecal ligation and puncture model is the only one that generates a polymicrobial sepsis, since the procedure exposes directly the peritoneum to the gut microbiome. Intravenous,70,79,110  intraperitoneal,78  and intratracheal87,88  route of MSC administration have been used. Dose of MSC ranged from 1 × 107  to 4 × 108  cells/kg.79,88 The main findings were that MSC were able to enhance bacteria clearance and attenuate septic organ injury in lungs, liver, and kidneys.70,78,79 

Although MSC have been extensively studied in heart I/R injury and used in clinical trials in patients with acute myocardial infarction,271  no data have been published concerning their potential therapeutic effects in sepsis-induced cardiac injury.272,273  And yet, half of patients with severe sepsis and septic shock present with reversible left ventricular systolic or diastolic dysfunction274  which is associated with increased mortality. Since the main pathways involved in this sepsis-related heart injury are those encountered in inflammation and I/R injuries, it seems to be important to study MSC in this context.

Beyond their ability of organ functional improvement in sepsis-induced injury, several studies showed a significant survival advantage in mice treated with MSC in peritonitis70,78,79  or pneumonia models.87  In addition, Lee et al.88  demonstrated a similar beneficial effect of MSC on macrophage phagocytosis and bacteria clearance in an E. coli bacterial-induced lung injury in an ex vivo human lung preparation. Even though most of these studies highlighted promising therapeutic properties of MSC within the early inflammatory phase of sepsis, it is still unknown whether they could be beneficial or harmful during the later antiinflammatory phases where immunity is impaired.275  However, what makes the therapeutic use of MSC unusual is that their phenotype can be skewed either towards a pro or antiinflammatory side depending on the surrounding milieu.268,269,276  The importance of this property of MSC needs to be studied, such as their use in the later phase of sepsis.

Possibly due to the heterogeneity of the animal septic models and the lack of data comparing MSC to the multiple therapeutics commonly used in sepsis, no clinical trial has been carried out yet.

As we described previously, the dose of MSC used in the preclinical small animal studies are extremely large and varies substantially (from 4 × 105 to 4 × 108 cells/kg). The optimal dose remains unknown in clinical trials although the typical dose in human is 5 to 10 × 106 cell/kg per dose. Additionally, the optimal route of delivery to generate the best therapeutic effect is still largely unknown between systemic and local administration. For example, the two clinical trials (NCT02097641, NCT01902082) in ARDS use intravenous administration whereas, in bronchopulmonary dysplasia in neonates, the only clinical trial (NCT01297205) uses intrabronchial administration.

Most injury models have shown benefits of MSC administration shortly after injury. Given that organ injury is a dynamic process over time, it is still unknown whether any beneficial effects might be found if MSC was given at a later phase such as during the resolution of injury; thus, it is unclear whether a second dose of MSC is needed for the resolution phase. Overall, the optimal dose, route, and time-sequence remain to be determined.

Even though organ failure is associated with poor outcome, it remains unclear whether organ failure or the initial underlying cause of injury or both is responsible for death. Organ failure has been even seen by others as an adaptive process of the organism in response to injury. Consequently, MSC should be considered as an adjuvant therapy; treating the initial cause of injury still remains the priority. For example, MSC should be considered an adjuvant therapy to ARDS caused by bacterial pneumonia, not supplanting antibiotics or other supportive therapies.

In addition, although MSC have antimicrobial properties in preclinical animal models, it is still worth questioning whether an immunosuppressive therapy such as MSC is appropriate during injury from an infectious etiology. For example, recent studies suggest that MSC fail to improve outcomes in acute phase of severe influenza.277  Whether this is a limitation to the murine model used needs to be studied further.

And finally, although a recent meta-analysis demonstrated no severe adverse outcome associated with MSC therapy,278  the potential of malignant transformation of MSC or the ability of MSC to enhance preexisting tumors still remains a serious clinical question, especially in light of the limitations of the tests available to detect cancer (i.e., computerized tomography scan).

The beneficial effects of cell-based therapy with MSC are apparent in multiple preclinical injury models involving all the organs in MODS. Attracted by signals from the injured and inflamed tissues, MSC appear to migrate to the site of damage and secrete an array of soluble factors and/or exosomes/microvesicles which suppress the injury. This review highlights the preclinical evidence which provided the underlying rationale for several phase I/II clinical trials in ARDS, AKI, and stroke. Based on promising preliminary results, further phase II and III trials are underway, the results of which are pending. However, no clinical studies are underway for ALF, TBI, sepsis, and MODS.

Some concerns still remain with MSC cell-based therapy which will need to be addressed in ongoing Phase I/II clinical trials such as the long term adverse effects of systemic immune suppression, potential for ectopic tissue formation, and MSC immunogenicity. Although very promising, the evidence is still unclear whether MSC cell-based therapy is superior to current therapies. We still await the results from the clinical trials.

Dr. Monsel was funded by the International Research Grant from the Société Française d’Anesthésie-Réanimation (Paris, France). Dr. Lee was supported by the National Heart, Lung, and Blood Institute (Bethesda, Maryland; grant no. HL-113022) and Hamilton Endowment Funds (University of California San Francisco Department of Anesthesiology, San Francisco, California).

The authors declare no competing interests.

1.
Bone
RC
,
Balk
RA
,
Cerra
FB
,
Dellinger
RP
,
Fein
AM
,
Knaus
WA
,
Schein
RM
,
Sibbald
WJ
:
Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine.
Chest
1992
;
101
:
1644
55
2.
Awad
SS
:
State-of-the-art therapy for severe sepsis and multisystem organ dysfunction.
Am J Surg
2003
;
186
(
5A
):
23S
30S; discussion 31S–34S
3.
Mongardon
N
,
Dyson
A
,
Singer
M
:
Is MOF an outcome parameter or a transient, adaptive state in critical illness?
Curr Opin Crit Care
2009
;
15
:
431
6
4.
Vincent
JL
,
Sakr
Y
,
Sprung
CL
,
Ranieri
VM
,
Reinhart
K
,
Gerlach
H
,
Moreno
R
,
Carlet
J
,
Le Gall
JR
,
Payen
D
;
Sepsis Occurrence in Acutely Ill Patients Investigators
:
Sepsis in European intensive care units: Results of the SOAP study.
Crit Care Med
2006
;
34
:
344
53
5.
Uchino
S
,
Kellum
JA
,
Bellomo
R
,
Doig
GS
,
Morimatsu
H
,
Morgera
S
,
Schetz
M
,
Tan
I
,
Bouman
C
,
Macedo
E
,
Gibney
N
,
Tolwani
A
,
Ronco
C
;
Beginning and Ending Supportive Therapy for the Kidney (BEST Kidney) Investigators
:
Acute renal failure in critically ill patients: A multinational, multicenter study.
JAMA
2005
;
294
:
813
8
6.
Zhu
YG
,
Hao
Q
,
Monsel
A
,
Feng
XM
,
Lee
JW
:
Adult stem cells for acute lung injury: Remaining questions and concerns.
Respirology
2013
;
18
:
744
56
7.
Lee
WM
:
Recent developments in acute liver failure.
Best Pract Res Clin Gastroenterol
2012
;
26
:
3
16
8.
Ferrari
G
,
Cusella-De Angelis
G
,
Coletta
M
,
Paolucci
E
,
Stornaiuolo
A
,
Cossu
G
,
Mavilio
F
:
Muscle regeneration by bone marrow-derived myogenic progenitors.
Science
1998
;
279
:
1528
30
9.
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
10.
Gnecchi
M
,
Zhang
Z
,
Ni
A
,
Dzau
VJ
:
Paracrine mechanisms in adult stem cell signaling and therapy.
Circ Res
2008
;
103
:
1204
19
11.
Phinney
DG
,
Prockop
DJ
:
Concise review: Mesenchymal stem/multipotent stromal cells: The state of transdifferentiation and modes of tissue repair—current views.
Stem Cells
2007
;
25
:
2896
902
12.
Tögel
FE
,
Westenfelder
C
:
Mesenchymal stem cells: A new therapeutic tool for AKI.
Nat Rev Nephrol
2010
;
6
:
179
83
13.
Wagers
AJ
,
Sherwood
RI
,
Christensen
JL
,
Weissman
IL
:
Little evidence for developmental plasticity of adult hematopoietic stem cells.
Science
2002
;
297
:
2256
9
14.
Lange
C
,
Tögel
F
,
Ittrich
H
,
Clayton
F
,
Nolte-Ernsting
C
,
Zander
AR
,
Westenfelder
C
:
Administered mesenchymal stem cells enhance recovery from ischemia/reperfusion-induced acute renal failure in rats.
Kidney Int
2005
;
68
:
1613
7
15.
Tögel
F
,
Hu
Z
,
Weiss
K
,
Isaac
J
,
Lange
C
,
Westenfelder
C
:
Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms.
Am J Physiol Renal Physiol
2005
;
289
:
F31
42
16.
Biancone
L
,
Bruno
S
,
Deregibus
MC
,
Tetta
C
,
Camussi
G
:
Therapeutic potential of mesenchymal stem cell-derived microvesicles.
Nephrol Dial Transplant
2012
;
27
:
3037
42
17.
Bruno
S
,
Bussolati
B
:
Therapeutic effects of mesenchymal stem cells on renal ischemia–reperfusion injury: A matter of genetic transfer?
Stem Cell Res Ther
2013
;
4
:
55
18.
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
19.
Bruno
S
,
Grange
C
,
Deregibus
MC
,
Calogero
RA
,
Saviozzi
S
,
Collino
F
,
Morando
L
,
Busca
A
,
Falda
M
,
Bussolati
B
,
Tetta
C
,
Camussi
G
:
Mesenchymal stem cell-derived microvesicles protect against acute tubular injury.
J Am Soc Nephrol
2009
;
20
:
1053
67
20.
Lee
C
,
Mitsialis
SA
,
Aslam
M
,
Vitali
SH
,
Vergadi
E
,
Konstantinou
G
,
Sdrimas
K
,
Fernandez-Gonzalez
A
,
Kourembanas
S
:
Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension.
Circulation
2012
;
126
:
2601
11
21.
Zhu
Y-G
,
Feng
X-M
,
Abbott
J
,
Fang
X-H
,
Hao
Q
,
Monsel
A
,
Qu
J-M
,
Matthay
MA
,
Lee
JW
:
Human mesenchymal stem cell microvesicles for treatment of E.coli endotoxin-induced acute lung injury in mice.
Stem Cells
2014
;
32
:
116
25
22.
Fischer
UM
,
Harting
MT
,
Jimenez
F
,
Monzon-Posadas
WO
,
Xue
H
,
Savitz
SI
,
Laine
GA
,
Cox
CS
Jr
:
Pulmonary passage is a major obstacle for intravenous stem cell delivery: The pulmonary first-pass effect.
Stem Cells Dev
2009
;
18
:
683
92
23.
Kotton
DN
,
Fabian
AJ
,
Mulligan
RC
:
Failure of bone marrow to reconstitute lung epithelium.
Am J Respir Cell Mol Biol
2005
;
33
:
328
34
24.
Loi
R
,
Beckett
T
,
Goncz
KK
,
Suratt
BT
,
Weiss
DJ
:
Limited restoration of cystic fibrosis lung epithelium in vivo with adult bone marrow-derived cells.
Am J Respir Crit Care Med
2006
;
173
:
171
9
25.
Morigi
M
,
Imberti
B
,
Zoja
C
,
Corna
D
,
Tomasoni
S
,
Abbate
M
,
Rottoli
D
,
Angioletti
S
,
Benigni
A
,
Perico
N
,
Alison
M
,
Remuzzi
G
:
Mesenchymal stem cells are renotropic, helping to repair the kidney and improve function in acute renal failure.
J Am Soc Nephrol
2004
;
15
:
1794
804
26.
Tögel
F
,
Weiss
K
,
Yang
Y
,
Hu
Z
,
Zhang
P
,
Westenfelder
C
:
Vasculotropic, paracrine actions of infused mesenchymal stem cells are important to the recovery from acute kidney injury.
Am J Physiol Renal Physiol
2007
;
292
:
F1626
35
27.
Tögel
F
,
Zhang
P
,
Hu
Z
,
Westenfelder
C
:
VEGF is a mediator of the renoprotective effects of multipotent marrow stromal cells in acute kidney injury.
J Cell Mol Med
2009
;
13
(
8B
):
2109
14
28.
Ezzat
T
,
Dhar
DK
,
Malago
M
,
Olde Damink
SW
:
Dynamic tracking of stem cells in an acute liver failure model.
World J Gastroenterol
2012
;
18
:
507
16
29.
Li
J
,
Zhang
L
,
Xin
J
,
Jiang
L
,
Li
J
,
Zhang
T
,
Jin
L
,
Li
J
,
Zhou
P
,
Hao
S
,
Cao
H
,
Li
L
:
Immediate intraportal transplantation of human bone marrow mesenchymal stem cells prevents death from fulminant hepatic failure in pigs.
Hepatology
2012
;
56
:
1044
52
30.
Zhang
S
,
Chen
L
,
Liu
T
,
Zhang
B
,
Xiang
D
,
Wang
Z
,
Wang
Y
:
Human umbilical cord matrix stem cells efficiently rescue acute liver failure through paracrine effects rather than hepatic differentiation.
Tissue Eng Part A
2012
;
18
:
1352
64
31.
Park
BN
,
Shim
W
,
Lee
G
,
Bang
OY
,
An
YS
,
Yoon
JK
,
Ahn
YH
:
Early distribution of intravenously injected mesenchymal stem cells in rats with acute brain trauma evaluated by (99m)Tc-HMPAO labeling.
Nucl Med Biol
2011
;
38
:
1175
82
32.
Shen
LH
,
Li
Y
,
Chen
J
,
Zacharek
A
,
Gao
Q
,
Kapke
A
,
Lu
M
,
Raginski
K
,
Vanguri
P
,
Smith
A
,
Chopp
M
:
Therapeutic benefit of bone marrow stromal cells administered 1 month after stroke.
J Cereb Blood Flow Metab
2007
;
27
:
6
13
33.
Yoon
JK
,
Park
BN
,
Shim
WY
,
Shin
JY
,
Lee
G
,
Ahn
YH
:
In vivo tracking of 111In-labeled bone marrow mesenchymal stem cells in acute brain trauma model.
Nucl Med Biol
2010
;
37
:
381
8
34.
Gao
J
,
Dennis
JE
,
Muzic
RF
,
Lundberg
M
,
Caplan
AI
:
The dynamic in vivo distribution of bone marrow-derived mesenchymal stem cells after infusion.
Cells Tissues Organs
2001
;
169
:
12
20
35.
Devine
SM
,
Bartholomew
AM
,
Mahmud
N
,
Nelson
M
,
Patil
S
,
Hardy
W
,
Sturgeon
C
,
Hewett
T
,
Chung
T
,
Stock
W
,
Sher
D
,
Weissman
S
,
Ferrer
K
,
Mosca
J
,
Deans
R
,
Moseley
A
,
Hoffman
R
:
Mesenchymal stem cells are capable of homing to the bone marrow of non-human primates following systemic infusion.
Exp Hematol
2001
;
29
:
244
55
36.
Wynn
RF
,
Hart
CA
,
Corradi-Perini
C
,
O’Neill
L
,
Evans
CA
,
Wraith
JE
,
Fairbairn
LJ
,
Bellantuono
I
:
A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow.
Blood
2004
;
104
:
2643
5
37.
Chapel
A
,
Bertho
JM
,
Bensidhoum
M
,
Fouillard
L
,
Young
RG
,
Frick
J
,
Demarquay
C
,
Cuvelier
F
,
Mathieu
E
,
Trompier
F
,
Dudoignon
N
,
Germain
C
,
Mazurier
C
,
Aigueperse
J
,
Borneman
J
,
Gorin
NC
,
Gourmelon
P
,
Thierry
D
:
Mesenchymal stem cells home to injured tissues when co-infused with hematopoietic cells to treat a radiation-induced multi-organ failure syndrome.
J Gene Med
2003
;
5
:
1028
38
38.
Herrera
MB
,
Bussolati
B
,
Bruno
S
,
Morando
L
,
Mauriello-Romanazzi
G
,
Sanavio
F
,
Stamenkovic
I
,
Biancone
L
,
Camussi
G
:
Exogenous mesenchymal stem cells localize to the kidney by means of CD44 following acute tubular injury.
Kidney Int
2007
;
72
:
430
41
39.
Mahmood
A
,
Lu
D
,
Lu
M
,
Chopp
M
:
Treatment of traumatic brain injury in adult rats with intravenous administration of human bone marrow stromal cells.
Neurosurgery
2003
;
53
:
697
702; discussion 702–3
40.
Orlic
D
,
Kajstura
J
,
Chimenti
S
,
Limana
F
,
Jakoniuk
I
,
Quaini
F
,
Nadal-Ginard
B
,
Bodine
DM
,
Leri
A
,
Anversa
P
:
Mobilized bone marrow cells repair the infarcted heart, improving function and survival.
Proc Natl Acad Sci U S A
2001
;
98
:
10344
9
41.
Ortiz
LA
,
Gambelli
F
,
McBride
C
,
Gaupp
D
,
Baddoo
M
,
Kaminski
N
,
Phinney
DG
:
Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects.
Proc Natl Acad Sci U S A
2003
;
100
:
8407
11
42.
Honczarenko
M
,
Le
Y
,
Swierkowski
M
,
Ghiran
I
,
Glodek
AM
,
Silberstein
LE
:
Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors.
Stem Cells
2006
;
24
:
1030
41
43.
Hung
SC
,
Pochampally
RR
,
Hsu
SC
,
Sanchez
C
,
Chen
SC
,
Spees
J
,
Prockop
DJ
:
Short-term exposure of multipotent stromal cells to low oxygen increases their expression of CX3CR1 and CXCR4 and their engraftment in vivo.
PLoS One
2007
;
2
:
e416
44.
Ji
JF
,
He
BP
,
Dheen
ST
,
Tay
SS
:
Interactions of chemokines and chemokine receptors mediate the migration of mesenchymal stem cells to the impaired site in the brain after hypoglossal nerve injury.
Stem Cells
2004
;
22
:
415
27
45.
Ponte
AL
,
Marais
E
,
Gallay
N
,
Langonné
A
,
Delorme
B
,
Hérault
O
,
Charbord
P
,
Domenech
J
:
The in vitro migration capacity of human bone marrow mesenchymal stem cells: Comparison of chemokine and growth factor chemotactic activities.
Stem Cells
2007
;
25
:
1737
45
46.
Tögel
F
,
Isaac
J
,
Hu
Z
,
Weiss
K
,
Westenfelder
C
:
Renal SDF-1 signals mobilization and homing of CXCR4-positive cells to the kidney after ischemic injury.
Kidney Int
2005
;
67
:
1772
84
47.
Göransson
V
,
Johnsson
C
,
Jacobson
A
,
Heldin
P
,
Hällgren
R
,
Hansell
P
:
Renal hyaluronan accumulation and hyaluronan synthase expression after ischaemia–reperfusion injury in the rat.
Nephrol Dial Transplant
2004
;
19
:
823
30
48.
Zhu
H
,
Mitsuhashi
N
,
Klein
A
,
Barsky
LW
,
Weinberg
K
,
Barr
ML
,
Demetriou
A
,
Wu
GD
:
The role of the hyaluronan receptor CD44 in mesenchymal stem cell migration in the extracellular matrix.
Stem Cells
2006
;
24
:
928
35
49.
Nace
G
,
Evankovich
J
,
Eid
R
,
Tsung
A
:
Dendritic cells and damage-associated molecular patterns: Endogenous danger signals linking innate and adaptive immunity.
J Innate Immun
2012
;
4
:
6
15
50.
Matzinger
P
:
Tolerance, danger, and the extended family.
Annu Rev Immunol
1994
;
12
:
991
1045
51.
Cain
SM
,
Curtis
SE
:
Experimental models of pathologic oxygen supply dependency.
Crit Care Med
1991
;
19
:
603
12
52.
Deitch
EA
:
Organ failure.
Ann Surg
2005
;
216
:
117
34
53.
Rosano
GM
,
Fini
M
,
Caminiti
G
,
Barbaro
G
:
Cardiac metabolism in myocardial ischemia.
Curr Pharm Des
2008
;
14
:
2551
62
54.
Townsend
MC
,
Hampton
WW
,
Haybron
DM
,
Schirmer
WJ
,
Fry
DE
:
Effective organ blood flow and bioenergy status in murine peritonitis.
Surgery
1986
;
100
:
205
13
55.
Gurusamy
N
,
Goswami
S
,
Malik
G
,
Das
DK
:
Oxidative injury induces selective rather than global inhibition of proteasomal activity.
J Mol Cell Cardiol
2008
;
44
:
419
28
56.
Mohammadzadeh
M
,
Halabian
R
,
Gharehbaghian
A
,
Amirizadeh
N
,
Jahanian-Najafabadi
A
,
Roushandeh
AM
,
Roudkenar
MH
:
Nrf-2 overexpression in mesenchymal stem cells reduces oxidative stress-induced apoptosis and cytotoxicity.
Cell Stress Chaperones
2012
;
17
:
553
65
57.
Yan
B
,
Singla
DK
:
Transplanted induced pluripotent stem cells mitigate oxidative stress and improve cardiac function through the Akt cell survival pathway in diabetic cardiomyopathy.
Mol Pharm
2013
;
10
:
3425
32
58.
Granger
DN
:
Role of xanthine oxidase and granulocytes in ischemia–reperfusion injury.
Am J Physiol
1988
;
255
(
6 Pt. 2
):
H1269
75
59.
Arslan
F
,
Lai
RC
,
Smeets
MB
,
Akeroyd
L
,
Choo
A
,
Aguor
EN
,
Timmers
L
,
van Rijen
HV
,
Doevendans
PA
,
Pasterkamp
G
,
Lim
SK
,
de Kleijn
DP
:
Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury.
Stem Cell Res
2013
;
10
:
301
12
60.
Lai
RC
,
Yeo
RW
,
Tan
KH
,
Lim
SK
:
Mesenchymal stem cell exosome ameliorates reperfusion injury through proteomic complementation.
Regen Med
2013
;
8
:
197
209
61.
Becker
LB
:
New concepts in reactive oxygen species and cardiovascular reperfusion physiology.
Cardiovasc Res
2004
;
61
:
461
70
62.
Li
C
,
Jackson
RM
:
Reactive species mechanisms of cellular hypoxia-reoxygenation injury.
Am J Physiol Cell Physiol
2002
;
282
:
C227
41
63.
Reilly
PM
,
Schiller
HJ
,
Bulkley
GB
:
Pharmacologic approach to tissue injury mediated by free radicals and other reactive oxygen metabolites.
Am J Surg
1991
;
161
:
488
503
64.
Berthier
S
,
Nguyen
MV
,
Baillet
A
,
Hograindleur
MA
,
Paclet
MH
,
Polack
B
,
Morel
F
:
Molecular interface of S100A8 with cytochrome b558 and NADPH oxidase activation.
PLoS One
2012
;
7
:
e40277
65.
Li
X
,
Arslan
F
,
Ren
Y
,
Adav
SS
,
Poh
KK
,
Sorokin
V
,
Lee
CN
,
de Kleijn
D
,
Lim
SK
,
Sze
SK
:
Metabolic adaptation to a disruption in oxygen supply during myocardial ischemia and reperfusion is underpinned by temporal and quantitative changes in the cardiac proteome.
J Proteome Res
2012
;
11
:
2331
46
66.
Aggarwal
S
,
Pittenger
MF
:
Human mesenchymal stem cells modulate allogeneic immune cell responses.
Blood
2005
;
105
:
1815
22
67.
Le Blanc
K
,
Mougiakakos
D
:
Multipotent mesenchymal stromal cells and the innate immune system.
Nat Rev Immunol
2012
;
12
:
383
96
68.
Nauta
AJ
,
Fibbe
WE
:
Immunomodulatory properties of mesenchymal stromal cells.
Blood
2007
;
110
:
3499
506
69.
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
70.
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
71.
Manning
E
,
Pham
S
,
Li
S
,
Vazquez-Padron
RI
,
Mathew
J
,
Ruiz
P
,
Salgar
SK
:
Interleukin-10 delivery via mesenchymal stem cells: A novel gene therapy approach to prevent lung ischemia–reperfusion injury.
Hum Gene Ther
2010
;
21
:
713
27
72.
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 U S A
2007
;
104
:
11002
7
73.
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
74.
van Poll
D
,
Parekkadan
B
,
Cho
CH
,
Berthiaume
F
,
Nahmias
Y
,
Tilles
AW
,
Yarmush
ML
:
Mesenchymal stem cell-derived molecules directly modulate hepatocellular death and regeneration in vitro and in vivo.
Hepatology
2008
;
47
:
1634
43
75.
Zheng
YB
,
Zhang
XH
,
Huang
ZL
,
Lin
CS
,
Lai
J
,
Gu
YR
,
Lin
BL
,
Xie
DY
,
Xie
SB
,
Peng
L
,
Gao
ZL
:
Amniotic-fluid-derived mesenchymal stem cells overexpressing interleukin-1 receptor antagonist improve fulminant hepatic failure.
PLoS One
2012
;
7
:
e41392
76.
Burra
P
,
Arcidiacono
D
,
Bizzaro
D
,
Chioato
T
,
Di Liddo
R
,
Banerjee
A
,
Cappon
A
,
Bo
P
,
Conconi
MT
,
Parnigotto
PP
,
Mirandola
S
,
Gringeri
E
,
Carraro
A
,
Cillo
U
,
Russo
FP
:
Systemic administration of a novel human umbilical cord mesenchymal stem cells population accelerates the resolution of acute liver injury.
BMC Gastroenterol
2012
;
12
:
88
77.
Borlongan
CV
,
Lind
JG
,
Dillon-Carter
O
,
Yu
G
,
Hadman
M
,
Cheng
C
,
Carroll
J
,
Hess
DC
:
Bone marrow grafts restore cerebral blood flow and blood brain barrier in stroke rats.
Brain Res
2004
;
1010
:
108
16
78.
Gonzalez-Rey
E
,
Anderson
P
,
González
MA
,
Rico
L
,
Büscher
D
,
Delgado
M
:
Human adult stem cells derived from adipose tissue protect against experimental colitis and sepsis.
Gut
2009
;
58
:
929
39
79.
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
80.
Kim
J
,
Hematti
P
:
Mesenchymal stem cell-educated macrophages: A novel type of alternatively activated macrophages.
Exp Hematol
2009
;
37
:
1445
53
81.
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
82.
Zhang
QZ
,
Su
WR
,
Shi
SH
,
Wilder-Smith
P
,
Xiang
AP
,
Wong
A
,
Nguyen
AL
,
Kwon
CW
,
Le
AD
:
Human gingiva-derived mesenchymal stem cells elicit polarization of m2 macrophages and enhance cutaneous wound healing.
Stem Cells
2010
;
28
:
1856
68
83.
Ionescu
L
,
Byrne
RN
,
van Haaften
T
,
Vadivel
A
,
Alphonse
RS
,
Rey-Parra
GJ
,
Weissmann
G
,
Hall
A
,
Eaton
F
,
Thébaud
B
:
Stem cell conditioned medium improves acute lung injury in mice: In vivo evidence for stem cell paracrine action.
Am J Physiol Lung Cell Mol Physiol
2012
;
303
:
L967
77
84.
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
85.
Liang
ZX
,
Sun
JP
,
Wang
P
,
Tian
Q
,
Yang
Z
,
Chen
LA
:
Bone marrow-derived mesenchymal stem cells protect rats from endotoxin-induced acute lung injury.
Chin Med J (Engl)
2011
;
124
:
2715
22
86.
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
87.
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
88.
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
89.
Ohtaki
H
,
Ylostalo
JH
,
Foraker
JE
,
Robinson
AP
,
Reger
RL
,
Shioda
S
,
Prockop
DJ
:
Stem/progenitor cells from bone marrow decrease neuronal death in global ischemia by modulation of inflammatory/immune responses.
Proc Natl Acad Sci U S A
2008
;
105
:
14638
43
90.
Chen
L
,
Tredget
EE
,
Wu
PY
,
Wu
Y
:
Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing.
PLoS One
2008
;
3
:
e1886
91.
Zanier
ER
,
Montinaro
M
,
Vigano
M
,
Villa
P
,
Fumagalli
S
,
Pischiutta
F
,
Longhi
L
,
Leoni
ML
,
Rebulla
P
,
Stocchetti
N
,
Lazzari
L
,
De Simoni
MG
:
Human umbilical cord blood mesenchymal stem cells protect mice brain after trauma.
Crit Care Med
2011
;
39
:
2501
10
92.
Jiang
XX
,
Zhang
Y
,
Liu
B
,
Zhang
SX
,
Wu
Y
,
Yu
XD
,
Mao
N
:
Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells.
Blood
2005
;
105
:
4120
6
93.
Nauta
AJ
,
Kruisselbrink
AB
,
Lurvink
E
,
Willemze
R
,
Fibbe
WE
:
Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocyte-derived dendritic cells.
J Immunol
2006
;
177
:
2080
7
94.
Poggi
A
,
Prevosto
C
,
Massaro
AM
,
Negrini
S
,
Urbani
S
,
Pierri
I
,
Saccardi
R
,
Gobbi
M
,
Zocchi
MR
:
Interaction between human NK cells and bone marrow stromal cells induces NK cell triggering: Role of NKp30 and NKG2D receptors.
J Immunol
2005
;
175
:
6352
60
95.
Rasmusson
I
,
Ringdén
O
,
Sundberg
B
,
Le Blanc
K
:
Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells.
Transplantation
2003
;
76
:
1208
13
96.
Spaggiari
GM
,
Capobianco
A
,
Becchetti
S
,
Mingari
MC
,
Moretta
L
:
Mesenchymal stem cell-natural killer cell interactions: Evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation.
Blood
2006
;
107
:
1484
90
97.
Le Blanc
K
,
Tammik
L
,
Sundberg
B
,
Haynesworth
SE
,
Ringdén
O
:
Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex.
Scand J Immunol
2003
;
57
:
11
20
98.
Potian
JA
,
Aviv
H
,
Ponzio
NM
,
Harrison
JS
,
Rameshwar
P
:
Veto-like activity of mesenchymal stem cells: Functional discrimination between cellular responses to alloantigens and recall antigens.
J Immunol
2003
;
171
:
3426
34
99.
Duffy
MM
,
Pindjakova
J
,
Hanley
SA
,
McCarthy
C
,
Weidhofer
GA
,
Sweeney
EM
,
English
K
,
Shaw
G
,
Murphy
JM
,
Barry
FP
,
Mahon
BP
,
Belton
O
,
Ceredig
R
,
Griffin
MD
:
Mesenchymal stem cell inhibition of T-helper 17 cell- differentiation is triggered by cell–cell contact and mediated by prostaglandin E2 via the EP4 receptor.
Eur J Immunol
2011
;
41
:
2840
51
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.
Tatara
R
,
Ozaki
K
,
Kikuchi
Y
,
Hatanaka
K
,
Oh
I
,
Meguro
A
,
Matsu
H
,
Sato
K
,
Ozawa
K
:
Mesenchymal stromal cells inhibit Th17 but not regulatory T-cell differentiation.
Cytotherapy
2011
;
13
:
686
94
102.
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
103.
Krampera
M
,
Glennie
S
,
Dyson
J
,
Scott
D
,
Laylor
R
,
Simpson
E
,
Dazzi
F
:
Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide.
Blood
2003
;
101
:
3722
9
104.
Chien
MH
,
Bien
MY
,
Ku
CC
,
Chang
YC
,
Pao
HY
,
Yang
YL
,
Hsiao
M
,
Chen
CL
,
Ho
JH
:
Systemic human orbital fat-derived stem/stromal cell transplantation ameliorates acute inflammation in lipopolysaccharide-induced acute lung injury.
Crit Care Med
2012
;
40
:
1245
53
105.
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
106.
Meisel
R
,
Brockers
S
,
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
:
648
54
107.
Brandau
S
,
Jakob
M
,
Hemeda
H
,
Bruderek
K
,
Janeschik
S
,
Bootz
F
,
Lang
S
:
Tissue-resident mesenchymal stem cells attract peripheral blood neutrophils and enhance their inflammatory activity in response to microbial challenge.
J Leukoc Biol
2010
;
88
:
1005
15
108.
Cassatella
MA
,
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
11
109.
Raffaghello
L
,
Bianchi
G
,
Bertolotto
M
,
Montecucco
F
,
Busca
A
,
Dallegri
F
,
Ottonello
L
,
Pistoia
V
:
Human mesenchymal stem cells inhibit neutrophil apoptosis: A model for neutrophil preservation in the bone marrow niche.
Stem Cells
2008
;
26
:
151
62
110.
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
111.
Qiu
J
,
Li
W
,
Feng
S
,
Wang
M
,
He
Z
:
Transplantation of bone marrow-derived endothelial progenitor cells attenuates cerebral ischemia and reperfusion injury by inhibiting neuronal apoptosis, oxidative stress and nuclear factor-κB expression.
Int J Mol Med
2013
;
31
:
91
8
112.
Whone
AL
,
Kemp
K
,
Sun
M
,
Wilkins
A
,
Scolding
NJ
:
Human bone marrow mesenchymal stem cells protect catecholaminergic and serotonergic neuronal perikarya and transporter function from oxidative stress by the secretion of glial-derived neurotrophic factor.
Brain Res
2012
;
1431
:
86
96
113.
Chen
YT
,
Sun
CK
,
Lin
YC
,
Chang
LT
,
Chen
YL
,
Tsai
TH
,
Chung
SY
,
Chua
S
,
Kao
YH
,
Yen
CH
,
Shao
PL
,
Chang
KC
,
Leu
S
,
Yip
HK
:
Adipose-derived mesenchymal stem cell protects kidneys against ischemia–reperfusion injury through suppressing oxidative stress and inflammatory reaction.
J Transl Med
2011
;
9
:
51
114.
Liu
H
,
McTaggart
SJ
,
Johnson
DW
,
Gobe
GC
:
Original article anti-oxidant pathways are stimulated by mesenchymal stromal cells in renal repair after ischemic injury.
Cytotherapy
2012
;
14
:
162
72
115.
Zhuo
W
,
Liao
L
,
Xu
T
,
Wu
W
,
Yang
S
,
Tan
J
:
Mesenchymal stem cells ameliorate ischemia–reperfusion-induced renal dysfunction by improving the antioxidant/oxidant balance in the ischemic kidney.
Urol Int
2011
;
86
:
191
6
116.
Jin
G
,
Qiu
G
,
Wu
D
,
Hu
Y
,
Qiao
P
,
Fan
C
,
Gao
F
:
Allogeneic bone marrow-derived mesenchymal stem cells attenuate hepatic ischemia–reperfusion injury by suppressing oxidative stress and inhibiting apoptosis in rats.
Int J Mol Med
2013
;
31
:
1395
401
117.
Nyamandi
VZ
,
Johnsen
VL
,
Hughey
CC
,
Hittel
DS
,
Khan
A
,
Newell
C
,
Shearer
J
:
Enhanced stem cell engraftment and modulation of hepatic reactive oxygen species production in diet-induced obesity.
Obesity (Silver Spring)
2014
;
22
:
721
9
118.
Quintanilha
LF
,
Takami
T
,
Hirose
Y
,
Fujisawa
K
,
Murata
Y
,
Yamamoto
N
,
Goldenberg
RC
,
Terai
S
,
Sakaida
I
:
Canine mesenchymal stem cells show antioxidant properties against thioacetamide-induced liver injury in vitro and in vivo.
Hepatol Res
2013
[Epub ahead of print]
119.
Sun
CK
,
Chang
CL
,
Lin
YC
,
Kao
YH
,
Chang
LT
,
Yen
CH
,
Shao
PL
,
Chen
CH
,
Leu
S
,
Yip
HK
:
Systemic administration of autologous adipose-derived mesenchymal stem cells alleviates hepatic ischemia–reperfusion injury in rats.
Crit Care Med
2012
;
40
:
1279
90
120.
Baglio
SR
,
Pegtel
DM
,
Baldini
N
:
Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy.
Front Physiol
2012
;
3
:
359
121.
Zhou
Y
,
Xu
H
,
Xu
W
,
Wang
B
,
Wu
H
,
Tao
Y
,
Zhang
B
,
Wang
M
,
Mao
F
,
Yan
Y
,
Gao
S
,
Gu
H
,
Zhu
W
,
Qian
H
:
Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro.
Stem Cell Res Ther
2013
;
4
:
34
122.
Thébaud
B
,
Stewart
DJ
:
Exosomes: Cell garbage can, therapeutic carrier, or trojan horse?
Circulation
2012
;
126
:
2553
5
123.
Lai
RC
,
Yeo
RW
,
Tan
SS
,
Zhang
B
,
Yin
Y
,
Sze
NSK
,
Choo
A
,
Lim
SK
:
Mesenchymal stem cell exosomes: The future MSC-based therapy?
in
Mesenchymal Stem Cell Therapy
. Edited by
Chase
LG
,
Vemuri
MC
.
New York
,
Humana Press
,
2013
, pp
pp 39
62
124.
Beiral
HJ
,
Rodrigues-Ferreira
C
,
Fernandes
AM
,
Gonsalez
SR
,
Mortari
NC
,
Takiya
CM
,
Sorenson
MM
,
Figueiredo-Freitas
C
,
Galina
A
,
Vieyra
A
:
The impact of stem cells on electron fluxes, proton translocation, and ATP synthesis in kidney mitochondria after ischemia/reperfusion.
Cell Transplant
2014
;
23
:
207
20
125.
Mathivanan
S
,
Simpson
RJ
:
ExoCarta: A compendium of exosomal proteins and RNA.
Proteomics
2009
;
9
:
4997
5000
126.
Kalra
H
,
Simpson
RJ
,
Ji
H
,
Aikawa
E
,
Altevogt
P
,
Askenase
P
,
Bond
VC
,
Borràs
FE
,
Breakefield
X
,
Budnik
V
,
Buzas
E
,
Camussi
G
,
Clayton
A
,
Cocucci
E
,
Falcon-Perez
JM
,
Gabrielsson
S
,
Gho
YS
,
Gupta
D
,
Harsha
HC
,
Hendrix
A
,
Hill
AF
,
Inal
JM
,
Jenster
G
,
Krämer-Albers
EM
,
Lim
SK
,
Llorente
A
,
Lötvall
J
,
Marcilla
A
,
Mincheva-Nilsson
L
,
Nazarenko
I
,
Nieuwland
R
,
Nolte-‘t Hoen
EN
,
Pandey
A
,
Patel
T
,
Piper
MG
,
Pluchino
S
,
Prasad
TS
,
Rajendran
L
,
Raposo
G
,
Record
M
,
Reid
GE
,
Sánchez-Madrid
F
,
Schiffelers
RM
,
Siljander
P
,
Stensballe
A
,
Stoorvogel
W
,
Taylor
D
,
Thery
C
,
Valadi
H
,
van Balkom
BW
,
Vázquez
J
,
Vidal
M
,
Wauben
MH
,
Yáñez-Mó
M
,
Zoeller
M
,
Mathivanan
S
:
Vesiclepedia: A compendium for extracellular vesicles with continuous community annotation.
PLoS Biol
2012
;
10
:
e1001450
127.
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
128.
Imberti
B
,
Morigi
M
,
Tomasoni
S
,
Rota
C
,
Corna
D
,
Longaretti
L
,
Rottoli
D
,
Valsecchi
F
,
Benigni
A
,
Wang
J
,
Abbate
M
,
Zoja
C
,
Remuzzi
G
:
Insulin-like growth factor-1 sustains stem cell mediated renal repair.
J Am Soc Nephrol
2007
;
18
:
2921
8
129.
Chen
Y
,
Qian
H
,
Zhu
W
,
Zhang
X
,
Yan
Y
,
Ye
S
,
Peng
X
,
Li
W
,
Xu
W
:
Hepatocyte growth factor modification promotes the amelioration effects of human umbilical cord mesenchymal stem cells on rat acute kidney injury.
Stem Cells Dev
2011
;
20
:
103
13
130.
Yuan
L
,
Wu
MJ
,
Sun
HY
,
Xiong
J
,
Zhang
Y
,
Liu
CY
,
Fu
LL
,
Liu
DM
,
Liu
HQ
,
Mei
CL
:
VEGF-modified human embryonic mesenchymal stem cell implantation enhances protection against cisplatin-induced acute kidney injury.
Am J Physiol Renal Physiol
2011
;
300
:
F207
18
131.
Tomasoni
S
,
Longaretti
L
,
Rota
C
,
Morigi
M
,
Conti
S
,
Gotti
E
,
Capelli
C
,
Introna
M
,
Remuzzi
G
,
Benigni
A
:
Transfer of growth factor receptor mRNA via exosomes unravels the regenerative effect of mesenchymal stem cells.
Stem Cells Dev
2013
;
22
:
772
80
132.
Kurozumi
K
,
Nakamura
K
,
Tamiya
T
,
Kawano
Y
,
Kobune
M
,
Hirai
S
,
Uchida
H
,
Sasaki
K
,
Ito
Y
,
Kato
K
,
Honmou
O
,
Houkin
K
,
Date
I
,
Hamada
H
:
BDNF gene-modified mesenchymal stem cells promote functional recovery and reduce infarct size in the rat middle cerebral artery occlusion model.
Mol Ther
2004
;
9
:
189
97
133.
Li
Y
,
Chen
J
,
Chen
XG
,
Wang
L
,
Gautam
SC
,
Xu
YX
,
Katakowski
M
,
Zhang
LJ
,
Lu
M
,
Janakiraman
N
,
Chopp
M
:
Human marrow stromal cell therapy for stroke in rat: Neurotrophins and functional recovery.
Neurology
2002
;
59
:
514
23
134.
Zhang
J
,
Li
Y
,
Chen
J
,
Yang
M
,
Katakowski
M
,
Lu
M
,
Chopp
M
:
Expression of insulin-like growth factor 1 and receptor in ischemic rats treated with human marrow stromal cells.
Brain Res
2004
;
1030
:
19
27
135.
Chen
J
,
Zhang
ZG
,
Li
Y
,
Wang
L
,
Xu
YX
,
Gautam
SC
,
Lu
M
,
Zhu
Z
,
Chopp
M
:
Intravenous administration of human bone marrow stromal cells induces angiogenesis in the ischemic boundary zone after stroke in rats.
Circ Res
2003
;
92
:
692
9
136.
Kim
HJ
,
Lee
JH
,
Kim
SH
:
Therapeutic effects of human mesenchymal stem cells on traumatic brain injury in rats: Secretion of neurotrophic factors and inhibition of apoptosis.
J Neurotrauma
2010
;
27
:
131
8
137.
Mahmood
A
,
Lu
D
,
Chopp
M
:
Intravenous administration of marrow stromal cells (MSCs) increases the expression of growth factors in rat brain after traumatic brain injury.
J Neurotrauma
2004
;
21
:
33
9
138.
Collino
F
,
Deregibus
MC
,
Bruno
S
,
Sterpone
L
,
Aghemo
G
,
Viltono
L
,
Tetta
C
,
Camussi
G
:
Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs.
PLoS One
2010
;
5
:
e11803
139.
Chang
CP
,
Chio
CC
,
Cheong
CU
,
Chao
CM
,
Cheng
BC
,
Lin
MT
:
Hypoxic preconditioning enhances the therapeutic potential of the secretome from cultured human mesenchymal stem cells in experimental traumatic brain injury.
Clin Sci (Lond)
2013
;
124
:
165
76
140.
Mahmood
A
,
Lu
D
,
Chopp
M
:
Marrow stromal cell transplantation after traumatic brain injury promotes cellular proliferation within the brain.
Neurosurgery
2004
;
55
:
1185
93
141.
Nichols
JE
,
Niles
JA
,
DeWitt
D
,
Prough
D
,
Parsley
M
,
Vega
S
,
Cantu
A
,
Lee
E
,
Cortiella
J
:
Neurogenic and neuro-protective potential of a novel subpopulation of peripheral blood-derived CD133+ ABCG2+CXCR4+ mesenchymal stem cells: Development of autologous cell-based therapeutics for traumatic brain injury.
Stem Cell Res Ther
2013
;
4
:
3
142.
Wang
M
,
Tsai
BM
,
Crisostomo
PR
,
Meldrum
DR
:
Pretreatment with adult progenitor cells improves recovery and decreases native myocardial proinflammatory signaling after ischemia.
Shock
2006
;
25
:
454
9
143.
Chen
S
,
Chen
L
,
Wu
X
,
Lin
J
,
Fang
J
,
Chen
X
,
Wei
S
,
Xu
J
,
Gao
Q
,
Kang
M
:
Ischemia postconditioning and mesenchymal stem cells engraftment synergistically attenuate ischemia reperfusion-induced lung injury in rats.
J Surg Res
2012
;
178
:
81
91
144.
Pati
S
,
Gerber
MH
,
Menge
TD
,
Wataha
KA
,
Zhao
Y
,
Baumgartner
JA
,
Zhao
J
,
Letourneau
PA
,
Huby
MP
,
Baer
LA
,
Salsbury
JR
,
Kozar
RA
,
Wade
CE
,
Walker
PA
,
Dash
PK
,
Cox
CS
Jr
,
Doursout
MF
,
Holcomb
JB
:
Bone marrow derived mesenchymal stem cells inhibit inflammation and preserve vascular endothelial integrity in the lungs after hemorrhagic shock.
PLoS One
2011
;
6
:
e25171
145.
Sun
CK
,
Yen
CH
,
Lin
YC
,
Tsai
TH
,
Chang
LT
,
Kao
YH
,
Chua
S
,
Fu
M
,
Ko
SF
,
Leu
S
,
Yip
HK
:
Autologous transplantation of adipose-derived mesenchymal stem cells markedly reduced acute ischemia–reperfusion lung injury in a rodent model.
J Transl Med
2011
;
9
:
118
146.
Dharmasaroja
P
:
Bone marrow-derived mesenchymal stem cells for the treatment of ischemic stroke.
J Clin Neurosci
2009
;
16
:
12
20
147.
Kale
S
,
Karihaloo
A
,
Clark
PR
,
Kashgarian
M
,
Krause
DS
,
Cantley
LG
:
Bone marrow stem cells contribute to repair of the ischemically injured renal tubule.
J Clin Invest
2003
;
112
:
42
9
148.
Semedo
P
,
Palasio
CG
,
Oliveira
CD
,
Feitoza
CQ
,
Gonçalves
GM
,
Cenedeze
MA
,
Wang
PM
,
Teixeira
VP
,
Reis
MA
,
Pacheco-Silva
A
,
Câmara
NO
:
Early modulation of inflammation by mesenchymal stem cell after acute kidney injury.
Int Immunopharmacol
2009
;
9
:
677
82
149.
Jiang
H
,
Qu
L
,
Li
Y
,
Gu
L
,
Shi
Y
,
Zhang
J
,
Zhu
W
,
Li
J
:
Bone marrow mesenchymal stem cells reduce intestinal ischemia/reperfusion injuries in rats.
J Surg Res
2011
;
168
:
127
34
150.
Shen
ZY
,
Zhang
J
,
Song
HL
,
Zheng
WP
:
Bone-marrow mesenchymal stem cells reduce rat intestinal ischemia–reperfusion injury, ZO-1 downregulation and tight junction disruption via a TNF-α-regulated mechanism.
World J Gastroenterol
2013
;
19
:
3583
95
151.
Zhuo
W
,
Liao
L
,
Fu
Y
,
Xu
T
,
Wu
W
,
Yang
S
,
Tan
J
:
Efficiency of endovenous versus arterial administration of mesenchymal stem cells for ischemia–reperfusion-induced renal dysfunction in rats.
Transplant Proc
2013
;
45
:
503
10
152.
Rubenfeld
GD
,
Caldwell
E
,
Peabody
E
,
Weaver
J
,
Martin
DP
,
Neff
M
,
Stern
EJ
,
Hudson
LD
:
Incidence and outcomes of acute lung injury.
N Engl J Med
2005
;
353
:
1685
93
153.
Ware
LB
,
Matthay
MA
:
The acute respiratory distress syndrome.
N Engl J Med
2000
;
342
:
1334
49
154.
Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. The Acute Respiratory Distress Syndrome Network.
N Engl J Med
2000
;
342
:
1301
8
155.
National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network
Wiedemann
HP
,
Wheeler
AP
,
Bernard
GR
,
Thompson
BT
,
Hayden
D
,
deBoisblanc
B
,
Connors
AF
,
Hite
RD
,
Harabin
AL
;
National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network
:
Comparison of two fluid-management strategies in acute lung injury.
N Engl J Med
2006
;
354
:
2564
75
156.
Guérin
C
,
Reignier
J
,
Richard
JC
,
Beuret
P
,
Gacouin
A
,
Boulain
T
,
Mercier
E
,
Badet
M
,
Mercat
A
,
Baudin
O
,
Clavel
M
,
Chatellier
D
,
Jaber
S
,
Rosselli
S
,
Mancebo
J
,
Sirodot
M
,
Hilbert
G
,
Bengler
C
,
Richecoeur
J
,
Gainnier
M
,
Bayle
F
,
Bourdin
G
,
Leray
V
,
Girard
R
,
Baboi
L
,
Ayzac
L
;
PROSEVA Study Group
:
Prone positioning in severe acute respiratory distress syndrome.
N Engl J Med
2013
;
368
:
2159
68
157.
Folkesson
HG
,
Matthay
MA
:
Alveolar epithelial ion and fluid transport: Recent progress.
Am J Respir Cell Mol Biol
2006
;
35
:
10
9
158.
Guery
BP
,
Mason
CM
,
Dobard
EP
,
Beaucaire
G
,
Summer
WR
,
Nelson
S
:
Keratinocyte growth factor increases transalveolar sodium reabsorption in normal and injured rat lungs.
Am J Respir Crit Care Med
1997
;
155
:
1777
84
159.
Aguilar
S
,
Scotton
CJ
,
McNulty
K
,
Nye
E
,
Stamp
G
,
Laurent
G
,
Bonnet
D
,
Janes
SM
:
Bone marrow stem cells expressing keratinocyte growth factor via an inducible lentivirus protects against bleomycin-induced pulmonary fibrosis.
PLoS One
2009
;
4
:
e8013
160.
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
161.
Lee
JW
,
Fang
X
,
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 U S A
2009
;
106
:
16357
62
162.
Gamble
JR
,
Drew
J
,
Trezise
L
,
Underwood
A
,
Parsons
M
,
Kasminkas
L
,
Rudge
J
,
Yancopoulos
G
,
Vadas
MA
:
Angiopoietin-1 is an antipermeability and anti-inflammatory agent in vitro and targets cell junctions.
Circ Res
2000
;
87
:
603
7
163.
Fang
X
,
Neyrinck
AP
,
Matthay
MA
,
Lee
JW
:
Allogeneic human mesenchymal stem cells restore epithelial protein permeability in cultured human alveolar type II cells by secretion of angiopoietin-1.
J Biol Chem
2010
;
285
:
26211
22
164.
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
165.
Xu
J
,
Qu
J
,
Cao
L
,
Sai
Y
,
Chen
C
,
He
L
,
Yu
L
:
Mesenchymal stem cell-based angiopoietin-1 gene therapy for acute lung injury induced by lipopolysaccharide in mice.
J Pathol
2008
;
214
:
472
81
166.
Li
J
,
Li
D
,
Liu
X
,
Tang
S
,
Wei
F
:
Human umbilical cord mesenchymal stem cells reduce systemic inflammation and attenuate LPS-induced acute lung injury in rats.
J Inflamm (Lond)
2012
;
9
:
33
167.
Prota
LF
,
Lassance
RM
,
Maron-Gutierrez
T
,
Castiglione
RC
,
Garcia
CS
,
Santana
MC
,
Souza-Menezes
J
,
Abreu
SC
,
Samoto
V
,
Santiago
MF
,
Capelozzi
VL
,
Takiya
CM
,
Rocco
PR
,
Morales
MM
:
Bone marrow mononuclear cell therapy led to alveolar–capillary membrane repair, improving lung mechanics in endotoxin-induced acute lung injury.
Cell Transplant
2010
;
19
:
965
71
168.
Hsu
CY
,
McCulloch
CE
,
Fan
D
,
Ordoñez
JD
,
Chertow
GM
,
Go
AS
:
Community-based incidence of acute renal failure.
Kidney Int
2007
;
72
:
208
12
169.
Bagshaw
SM
,
George
C
,
Bellomo
R
;
ANZICS Database Management Committee
:
Early acute kidney injury and sepsis: A multicentre evaluation.
Crit Care
2008
;
12
:
R47
170.
Hoste
EA
,
Clermont
G
,
Kersten
A
,
Venkataraman
R
,
Angus
DC
,
De Bacquer
D
,
Kellum
JA
:
RIFLE criteria for acute kidney injury are associated with hospital mortality in critically ill patients: A cohort analysis.
Crit Care
2006
;
10
:
R73
171.
Bellomo
R
,
Kellum
JA
,
Ronco
C
:
Acute kidney injury.
Lancet
2012
;
380
:
756
66
172.
Heyman
SN
,
Lieberthal
W
,
Rogiers
P
,
Bonventre
JV
:
Animal models of acute tubular necrosis.
Curr Opin Crit Care
2002
;
8
:
526
34
173.
Heyman
SN
,
Rosenberger
C
,
Rosen
S
:
Experimental ischemia–reperfusion: Biases and myths—the proximal vs. distal hypoxic tubular injury debate revisited.
Kidney Int
2010
;
77
:
9
16
174.
Singh
AP
,
Junemann
A
,
Muthuraman
A
,
Jaggi
AS
,
Singh
N
,
Grover
K
,
Dhawan
R
:
Animal models of acute renal failure.
Pharmacol Rep
2012
;
64
:
31
44
175.
Thuillier
R
,
Favreau
F
,
Celhay
O
,
Macchi
L
,
Milin
S
,
Hauet
T
:
Thrombin inhibition during kidney ischemia–reperfusion reduces chronic graft inflammation and tubular atrophy.
Transplantation
2010
;
90
:
612
21
176.
Versteilen
AMG
,
Blaauw
N
,
Di Maggio
F
,
Groeneveld
ABJ
,
Sipkema
P
,
Musters
RJP
,
Tangelder
GJ
:
Rho-Kinase inhibition reduces early microvascular leukocyte accumulation in the rat kidney following ischemia–reperfusion injury: Roles of nitric oxide and blood flow.
Nephron Exp Nephrol
2011
;
118
:
e79
86
177.
Kwon
O
,
Hong
SM
,
Ramesh
G
:
Diminished NO generation by injured endothelium and loss of macula densa nNOS may contribute to sustained acute kidney injury after ischemia–reperfusion.
Am J Physiol Renal Physiol
2009
;
296
:
F25
33
178.
Kato
N
,
Yuzawa
Y
,
Kosugi
T
,
Hobo
A
,
Sato
W
,
Miwa
Y
,
Sakamoto
K
,
Matsuo
S
,
Kadomatsu
K
:
The E-selectin ligand basigin/CD147 is responsible for neutrophil recruitment in renal ischemia/reperfusion.
J Am Soc Nephrol
2009
;
20
:
1565
76
179.
Thurman
JM
:
Triggers of inflammation after renal ischemia/reperfusion.
Clin Immunol
2007
;
123
:
7
13
180.
Pulskens
WP
,
Teske
GJ
,
Butter
LM
,
Roelofs
JJ
,
van der Poll
T
,
Florquin
S
,
Leemans
JC
:
Toll-like receptor-4 coordinates the innate immune response of the kidney to renal ischemia/reperfusion injury.
PLoS One
2008
;
3
:
e3596
181.
Saikumar
P
,
Venkatachalam
MA
:
Role of apoptosis in hypoxic/ischemic damage in the kidney.
Semin Nephrol
2003
;
23
:
511
21
182.
Moreau
R
,
Lebrec
D
:
Acute kidney injury: New concepts. Hepatorenal syndrome: The role of vasopressors.
Nephron Physiol
2008
;
109
:
73
9
183.
Schrier
RW
,
Wang
W
:
Acute renal failure and sepsis.
N Engl J Med
2004
;
351
:
159
69
184.
Loutzenhiser
R
,
Griffin
K
,
Williamson
G
,
Bidani
A
:
Renal autoregulation: New perspectives regarding the protective and regulatory roles of the underlying mechanisms.
Am J Physiol Regul Integr Comp Physiol
2006
;
290
:
R1153
67
185.
Bi
B
,
Schmitt
R
,
Israilova
M
,
Nishio
H
,
Cantley
LG
:
Stromal cells protect against acute tubular injury via an endocrine effect.
J Am Soc Nephrol
2007
;
18
:
2486
96
186.
Eliopoulos
N
,
Zhao
J
,
Bouchentouf
M
,
Forner
K
,
Birman
E
,
Yuan
S
,
Boivin
MN
,
Martineau
D
:
Human marrow-derived mesenchymal stromal cells decrease cisplatin renotoxicity in vitro and in vivo and enhance survival of mice post-intraperitoneal injection.
Am J Physiol Renal Physiol
2010
;
299
:
F1288
98
187.
Herrera
MB
,
Bussolati
B
,
Bruno
S
,
Fonsato
V
,
Romanazzi
GM
,
Camussi
G
:
Mesenchymal stem cells contribute to the renal repair of acute tubular epithelial injury.
Int J Mol Med
2004
;
14
:
1035
41
188.
Morigi
M
,
Introna
M
,
Imberti
B
,
Corna
D
,
Abbate
M
,
Rota
C
,
Rottoli
D
,
Benigni
A
,
Perico
N
,
Zoja
C
,
Rambaldi
A
,
Remuzzi
A
,
Remuzzi
G
:
Human bone marrow mesenchymal stem cells accelerate recovery of acute renal injury and prolong survival in mice.
Stem Cells
2008
;
26
:
2075
82
189.
Morigi
M
,
Rota
C
,
Montemurro
T
,
Montelatici
E
,
Lo Cicero
V
,
Imberti
B
,
Abbate
M
,
Zoja
C
,
Cassis
P
,
Longaretti
L
,
Rebulla
P
,
Introna
M
,
Capelli
C
,
Benigni
A
,
Remuzzi
G
,
Lazzari
L
:
Life-sparing effect of human cord blood-mesenchymal stem cells in experimental acute kidney injury.
Stem Cells
2010
;
28
:
513
22
190.
Rota
C
,
Imberti
B
,
Pozzobon
M
,
Piccoli
M
,
De Coppi
P
,
Atala
A
,
Gagliardini
E
,
Xinaris
C
,
Benedetti
V
,
Fabricio
AS
,
Squarcina
E
,
Abbate
M
,
Benigni
A
,
Remuzzi
G
,
Morigi
M
:
Human amniotic fluid stem cell preconditioning improves their regenerative potential.
Stem Cells Dev
2012
;
21
:
1911
23
191.
Cao
H
,
Qian
H
,
Xu
W
,
Zhu
W
,
Zhang
X
,
Chen
Y
,
Wang
M
,
Yan
Y
,
Xie
Y
:
Mesenchymal stem cells derived from human umbilical cord ameliorate ischemia/reperfusion-induced acute renal failure in rats.
Biotechnol Lett
2010
;
32
:
725
32
192.
Hagiwara
M
,
Shen
B
,
Chao
L
,
Chao
J
:
Kallikrein-modified mesenchymal stem cell implantation provides enhanced protection against acute ischemic kidney injury by inhibiting apoptosis and inflammation.
Hum Gene Ther
2008
;
19
:
807
19
193.
Tögel
F
,
Cohen
A
,
Zhang
P
,
Yang
Y
,
Hu
Z
,
Westenfelder
C
:
Autologous and allogeneic marrow stromal cells are safe and effective for the treatment of acute kidney injury.
Stem Cells Dev
2009
;
18
:
475
85
194.
Morigi
M
,
Benigni
A
:
Mesenchymal stem cells and kidney repair.
Nephrol Dial Transplant
2013
;
28
:
788
93
195.
Gaspari
F
,
Cravedi
P
,
Mandalà
M
,
Perico
N
,
de Leon
FR
,
Stucchi
N
,
Ferrari
S
,
Labianca
R
,
Remuzzi
G
,
Ruggenenti
P
:
Predicting cisplatin-induced acute kidney injury by urinary neutrophil gelatinase-associated lipocalin excretion: A pilot prospective case–control study.
Nephron Clin Pract
2010
;
115
:
c154
60
196.
Gooch
A
,
Doty
J
,
Flores
J
,
Swenson
L
,
Toegel
FE
,
Reiss
GR
,
Lange
C
,
Zander
AR
,
Hu
Z
,
Poole
S
,
Zhang
P
,
Westenfelder
C
:
Initial report on a phase I clinical trial: Prevention and treatment of post-operative Acute Kidney Injury with allogeneic Mesenchymal Stem Cells in patients who require on-pump cardiac surgery.
Cell Ther Transplant
2008
;
1
:
31
5
197.
Chung
RT
,
Stravitz
RT
,
Fontana
RJ
,
Schiodt
FV
,
Mehal
WZ
,
Reddy
KR
,
Lee
WM
:
Pathogenesis of liver injury in acute liver failure.
Gastroenterology
2012
;
143
:
e1
7
198.
Leifeld
L
,
Dumoulin
FL
,
Purr
I
,
Janberg
K
,
Trautwein
C
,
Wolff
M
,
Manns
MP
,
Sauerbruch
T
,
Spengler
U
:
Early up-regulation of chemokine expression in fulminant hepatic failure.
J Pathol
2003
;
199
:
335
44
199.
Sgroi
A
,
Gonelle-Gispert
C
,
Morel
P
,
Baertschiger
RM
,
Niclauss
N
,
Mentha
G
,
Majno
P
,
Serre-Beinier
V
,
Buhler
L
:
Interleukin-1 receptor antagonist modulates the early phase of liver regeneration after partial hepatectomy in mice.
PLoS One
2011
;
6
:
e25442
200.
Yumoto
E
,
Higashi
T
,
Nouso
K
,
Nakatsukasa
H
,
Fujiwara
K
,
Hanafusa
T
,
Yumoto
Y
,
Tanimoto
T
,
Kurimoto
M
,
Tanaka
N
,
Tsuji
T
:
Serum gamma-interferon-inducing factor (IL-18) and IL-10 levels in patients with acute hepatitis and fulminant hepatic failure.
J Gastroenterol Hepatol
2002
;
17
:
285
94
201.
Tron
K
,
Novosyadlyy
R
,
Dudas
J
,
Samoylenko
A
,
Kietzmann
T
,
Ramadori
G
:
Upregulation of heme oxygenase-1 gene by turpentine oil-induced localized inflammation: Involvement of interleukin-6.
Lab Invest
2005
;
85
:
376
87
202.
Sato
Y
,
Araki
H
,
Kato
J
,
Nakamura
K
,
Kawano
Y
,
Kobune
M
,
Sato
T
,
Miyanishi
K
,
Takayama
T
,
Takahashi
M
,
Takimoto
R
,
Iyama
S
,
Matsunaga
T
,
Ohtani
S
,
Matsuura
A
,
Hamada
H
,
Niitsu
Y
:
Human mesenchymal stem cells xenografted directly to rat liver are differentiated into human hepatocytes without fusion.
Blood
2005
;
106
:
756
63
203.
Vassilopoulos
G
,
Wang
PR
,
Russell
DW
:
Transplanted bone marrow regenerates liver by cell fusion.
Nature
2003
;
422
:
901
4
204.
Muraca
M
,
Ferraresso
C
,
Vilei
MT
,
Granato
A
,
Quarta
M
,
Cozzi
E
,
Rugge
M
,
Pauwelyn
KA
,
Caruso
M
,
Avital
I
,
Inderbitzin
D
,
Demetriou
AA
,
Forbes
SJ
,
Realdi
G
:
Liver repopulation with bone marrow derived cells improves the metabolic disorder in the Gunn rat.
Gut
2007
;
56
:
1725
35
205.
Malhi
H
,
Irani
AN
,
Gagandeep
S
,
Gupta
S
:
Isolation of human progenitor liver epithelial cells with extensive replication capacity and differentiation into mature hepatocytes.
J Cell Sci
2002
;
115
(
Pt. 13
):
2679
88
206.
Caplan
AI
,
Dennis
JE
:
Mesenchymal stem cells as trophic mediators.
J Cell Biochem
2006
;
98
:
1076
84
207.
Haynesworth
SE
,
Baber
MA
,
Caplan
AI
:
Cytokine expression by human marrow-derived mesenchymal progenitor cells in vitro: Effects of dexamethasone and IL-1 alpha.
J Cell Physiol
1996
;
166
:
585
92
208.
Le Blanc
K
:
Mesenchymal stromal cells: Tissue repair and immune modulation.
Cytotherapy
2006
;
8
:
559
61
209.
Cho
KA
,
Woo
SY
,
Seoh
JY
,
Han
HS
,
Ryu
KH
:
Mesenchymal stem cells restore CCl4-induced liver injury by an antioxidative process.
Cell Biol Int
2012
;
36
:
1267
74
210.
Gruttadauria
S
,
Grosso
G
,
Pagano
D
,
Biondi
A
,
Echeverri
GJ
,
Seria
E
,
Pietrosi
G
,
Liotta
R
,
Basile
F
,
Gridelli
B
:
Marrow-derived mesenchymal stem cells restore biochemical markers of acute liver injury in experimental model.
Transplant Proc
2013
;
45
:
480
6
211.
Jin
SZ
,
Liu
BR
,
Xu
J
,
Gao
FL
,
Hu
ZJ
,
Wang
XH
,
Pei
FH
,
Hong
Y
,
Hu
HY
,
Han
MZ
:
Ex vivo-expanded bone marrow stem cells home to the liver and ameliorate functional recovery in a mouse model of acute hepatic injury.
Hepatobiliary Pancreat Dis Int
2012
;
11
:
66
73
212.
Yukawa
H
,
Noguchi
H
,
Oishi
K
,
Takagi
S
,
Hamaguchi
M
,
Hamajima
N
,
Hayashi
S
:
Cell transplantation of adipose tissue-derived stem cells in combination with heparin attenuated acute liver failure in mice.
Cell Transplant
2009
;
18
:
611
8
213.
Cao
H
,
Yang
J
,
Yu
J
,
Pan
Q
,
Li
J
,
Zhou
P
,
Li
Y
,
Pan
X
,
Li
J
,
Wang
Y
,
Li
L
:
Therapeutic potential of transplanted placental mesenchymal stem cells in treating Chinese miniature pigs with acute liver failure.
BMC Med
2012
;
10
:
56
214.
Parekkadan
B
,
van Poll
D
,
Suganuma
K
,
Carter
EA
,
Berthiaume
F
,
Tilles
AW
,
Yarmush
ML
:
Mesenchymal stem cell-derived molecules reverse fulminant hepatic failure.
PLoS One
2007
;
2
:
e941
215.
Kanazawa
H
,
Fujimoto
Y
,
Teratani
T
,
Iwasaki
J
,
Kasahara
N
,
Negishi
K
,
Tsuruyama
T
,
Uemoto
S
,
Kobayashi
E
:
Bone marrow-derived mesenchymal stem cells ameliorate hepatic ischemia reperfusion injury in a rat model.
PLoS One
2011
;
6
:
e19195
216.
Pan
GZ
,
Yang
Y
,
Zhang
J
,
Liu
W
,
Wang
GY
,
Zhang
YC
,
Yang
Q
,
Zhai
FX
,
Tai
Y
,
Liu
JR
,
Zhang
Q
,
Chen
GH
:
Bone marrow mesenchymal stem cells ameliorate hepatic ischemia/reperfusion injuries via inactivation of the MEK/ERK signaling pathway in rats.
J Surg Res
2012
;
178
:
935
48
217.
Dahlke
MH
,
Hoogduijn
M
,
Eggenhofer
E
,
Popp
FC
,
Renner
P
,
Slowik
P
,
Rosenauer
A
,
Piso
P
,
Geissler
EK
,
Lange
C
,
Chabannes
D
,
Mazzanti
B
,
Bigenzahn
S
,
Bertolino
P
,
Kunter
U
,
Introna
M
,
Rambaldi
A
,
Capelli
C
,
Perico
N
,
Casiraghi
F
,
Noris
M
,
Gotti
E
,
Seifert
M
,
Saccardi
R
,
Verspaget
HW
,
van Hoek
B
,
Bartholomew
A
,
Wekerle
T
,
Volk
HD
,
Remuzzi
G
,
Deans
R
,
Lazarus
H
,
Schlitt
HJ
,
Baan
CC
;
MISOT Study Group
:
Toward MSC in solid organ transplantation: 2008 position paper of the MISOT study group.
Transplantation
2009
;
88
:
614
9
218.
Boeykens
N
,
Ponsaerts
P
,
Van der Linden
A
,
Berneman
Z
,
Ysebaert
D
,
De Greef
K
:
Injury-dependent retention of intraportally administered mesenchymal stromal cells following partial hepatectomy of steatotic liver does not lead to improved liver recovery.
PLoS One
2013
;
8
:
e69092
219.
Donnan
GA
,
Fisher
M
,
Macleod
M
,
Davis
SM
:
Stroke.
Lancet
2008
;
371
:
1612
23
220.
Kissela
B
,
Broderick
J
,
Woo
D
,
Kothari
R
,
Miller
R
,
Khoury
J
,
Brott
T
,
Pancioli
A
,
Jauch
E
,
Gebel
J
,
Shukla
R
,
Alwell
K
,
Tomsick
T
:
Greater Cincinnati/Northern Kentucky Stroke Study: Volume of first-ever ischemic stroke among blacks in a population-based study.
Stroke
2001
;
32
:
1285
90
221.
Abdelkarim
GE
,
Gertz
K
,
Harms
C
,
Katchanov
J
,
Dirnagl
U
,
Szabó
C
,
Endres
M
:
Protective effects of PJ34, a novel, potent inhibitor of poly(ADP-ribose) polymerase (PARP) in in vitro and in vivo models of stroke.
Int J Mol Med
2001
;
7
:
255
60
222.
Li
X
,
Klaus
JA
,
Zhang
J
,
Xu
Z
,
Kibler
KK
,
Andrabi
SA
,
Rao
K
,
Yang
ZJ
,
Dawson
TM
,
Dawson
VL
,
Koehler
RC
:
Contributions of poly(ADP-ribose) polymerase-1 and -2 to nuclear translocation of apoptosis-inducing factor and injury from focal cerebral ischemia.
J Neurochem
2010
;
113
:
1012
22
223.
Mattson
MP
,
Culmsee
C
,
Yu
ZF
:
Apoptotic and antiapoptotic mechanisms in stroke.
Cell Tissue Res
2000
;
301
:
173
87
224.
Sims
NR
,
Muyderman
H
:
Mitochondria, oxidative metabolism and cell death in stroke.
Biochim Biophys Acta
2010
;
1802
:
80
91
225.
Matsuo
Y
,
Onodera
H
,
Shiga
Y
,
Shozuhara
H
,
Ninomiya
M
,
Kihara
T
,
Tamatani
T
,
Miyasaka
M
,
Kogure
K
:
Role of cell adhesion molecules in brain injury after transient middle cerebral artery occlusion in the rat.
Brain Res
1994
;
656
:
344
52
226.
Zhang
RL
,
Chopp
M
,
Zhang
ZG
,
Phillips
ML
,
Rosenbloom
CL
,
Cruz
R
,
Manning
A
:
E-selectin in focal cerebral ischemia and reperfusion in the rat.
J Cereb Blood Flow Metab
1996
;
16
:
1126
36
227.
Koo
JW
,
Duman
RS
:
IL-1beta is an essential mediator of the antineurogenic and anhedonic effects of stress.
Proc Natl Acad Sci U S A
2008
;
105
:
751
6
228.
Tuttolomondo
A
,
Di Raimondo
D
,
di Sciacca
R
,
Pinto
A
,
Licata
G
:
Inflammatory cytokines in acute ischemic stroke.
Curr Pharm Des
2008
;
14
:
3574
89
229.
Lee
J
,
Kuroda
S
,
Shichinohe
H
,
Ikeda
J
,
Seki
T
,
Hida
K
,
Tada
M
,
Sawada
K
,
Iwasaki
Y
:
Migration and differentiation of nuclear fluorescence-labeled bone marrow stromal cells after transplantation into cerebral infarct and spinal cord injury in mice.
Neuropathology
2003
;
23
:
169
80
230.
Liu
H
,
Honmou
O
,
Harada
K
,
Nakamura
K
,
Houkin
K
,
Hamada
H
,
Kocsis
JD
:
Neuroprotection by PlGF gene-modified human mesenchymal stem cells after cerebral ischaemia.
Brain
2006
;
129
(
Pt. 10
):
2734
45
231.
Shichinohe
H
,
Kuroda
S
,
Yano
S
,
Ohnishi
T
,
Tamagami
H
,
Hida
K
,
Iwasaki
Y
:
Improved expression of gamma-aminobutyric acid receptor in mice with cerebral infarct and transplanted bone marrow stromal cells: An autoradiographic and histologic analysis.
J Nucl Med
2006
;
47
:
486
91
232.
Yano
S
,
Kuroda
S
,
Shichinohe
H
,
Hida
K
,
Iwasaki
Y
:
Do bone marrow stromal cells proliferate after transplantation into mice cerebral infarct?—A double labeling study.
Brain Res
2005
;
1065
:
60
7
233.
Chen
J
,
Li
Y
,
Wang
L
,
Lu
M
,
Zhang
X
,
Chopp
M
:
Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats.
J Neurol Sci
2001
;
189
:
49
57
234.
Ikeda
N
,
Nonoguchi
N
,
Zhao
MZ
,
Watanabe
T
,
Kajimoto
Y
,
Furutama
D
,
Kimura
F
,
Dezawa
M
,
Coffin
RS
,
Otsuki
Y
,
Kuroiwa
T
,
Miyatake
S
:
Bone marrow stromal cells that enhanced fibroblast growth factor-2 secretion by herpes simplex virus vector improve neurological outcome after transient focal cerebral ischemia in rats.
Stroke
2005
;
36
:
2725
30
235.
Li
Y
,
Chopp
M
,
Chen
J
,
Wang
L
,
Gautam
SC
,
Xu
YX
,
Zhang
Z
:
Intrastriatal transplantation of bone marrow nonhematopoietic cells improves functional recovery after stroke in adult mice.
J Cereb Blood Flow Metab
2000
;
20
:
1311
9
236.
Zhao
MZ
,
Nonoguchi
N
,
Ikeda
N
,
Watanabe
T
,
Furutama
D
,
Miyazawa
D
,
Funakoshi
H
,
Kajimoto
Y
,
Nakamura
T
,
Dezawa
M
,
Shibata
MA
,
Otsuki
Y
,
Coffin
RS
,
Liu
WD
,
Kuroiwa
T
,
Miyatake
S
:
Novel therapeutic strategy for stroke in rats by bone marrow stromal cells and ex vivo HGF gene transfer with HSV-1 vector.
J Cereb Blood Flow Metab
2006
;
26
:
1176
88
237.
Li
Y
,
Chen
J
,
Wang
L
,
Lu
M
,
Chopp
M
:
Treatment of stroke in rat with intracarotid administration of marrow stromal cells.
Neurology
2001
;
56
:
1666
72
238.
Shen
LH
,
Li
Y
,
Chen
J
,
Zhang
J
,
Vanguri
P
,
Borneman
J
,
Chopp
M
:
Intracarotid transplantation of bone marrow stromal cells increases axon-myelin remodeling after stroke.
Neuroscience
2006
;
137
:
393
9
239.
Chen
J
,
Li
Y
,
Katakowski
M
,
Chen
X
,
Wang
L
,
Lu
D
,
Lu
M
,
Gautam
SC
,
Chopp
M
:
Intravenous bone marrow stromal cell therapy reduces apoptosis and promotes endogenous cell proliferation after stroke in female rat.
J Neurosci Res
2003
;
73
:
778
86
240.
Chen
J
,
Li
Y
,
Wang
L
,
Zhang
Z
,
Lu
D
,
Lu
M
,
Chopp
M
:
Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats.
Stroke
2001
;
32
:
1005
11
241.
Cui
X
,
Chen
J
,
Zacharek
A
,
Li
Y
,
Roberts
C
,
Kapke
A
,
Savant-Bhonsale
S
,
Chopp
M
:
Nitric oxide donor upregulation of stromal cell-derived factor-1/chemokine (CXC motif) receptor 4 enhances bone marrow stromal cell migration into ischemic brain after stroke.
Stem Cells
2007
;
25
:
2777
85
242.
Li
Y
,
Chen
J
,
Zhang
CL
,
Wang
L
,
Lu
D
,
Katakowski
M
,
Gao
Q
,
Shen
LH
,
Zhang
J
,
Lu
M
,
Chopp
M
:
Gliosis and brain remodeling after treatment of stroke in rats with marrow stromal cells.
Glia
2005
;
49
:
407
17
243.
Wakabayashi
K
,
Nagai
A
,
Sheikh
AM
,
Shiota
Y
,
Narantuya
D
,
Watanabe
T
,
Masuda
J
,
Kobayashi
S
,
Kim
SU
,
Yamaguchi
S
:
Transplantation of human mesenchymal stem cells promotes functional improvement and increased expression of neurotrophic factors in a rat focal cerebral ischemia model.
J Neurosci Res
2010
;
88
:
1017
25
244.
Gutiérrez-Fernández
M
,
Rodríguez-Frutos
B
,
Alvarez-Grech
J
,
Vallejo-Cremades
MT
,
Expósito-Alcaide
M
,
Merino
J
,
Roda
JM
,
Díez-Tejedor
E
:
Functional recovery after hematic administration of allogenic mesenchymal stem cells in acute ischemic stroke in rats.
Neuroscience
2011
;
175
:
394
405
245.
Willing
AE
,
Lixian
J
,
Milliken
M
,
Poulos
S
,
Zigova
T
,
Song
S
,
Hart
C
,
Sanchez-Ramos
J
,
Sanberg
PR
:
Intravenous versus intrastriatal cord blood administration in a rodent model of stroke.
J Neurosci Res
2003
;
73
:
296
307
246.
Bang
OY
,
Lee
JS
,
Lee
PH
,
Lee
G
:
Autologous mesenchymal stem cell transplantation in stroke patients.
Ann Neurol
2005
;
57
:
874
82
247.
De Keyser
J
:
Autologous mesenchymal stem cell transplantation in stroke patients.
Ann Neurol
2005
;
58
:
653
4; author reply 654–5
248.
Smith
HK
,
Gavins
FN
:
The potential of stem cell therapy for stroke: Is PISCES the sign?
FASEB J
2012
;
26
:
2239
52
249.
Lee
JS
,
Hong
JM
,
Moon
GJ
,
Lee
PH
,
Ahn
YH
,
Bang
OY
;
STARTING collaborators
:
A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke.
Stem Cells
2010
;
28
:
1099
106
250.
Lingsma
HF
,
Roozenbeek
B
,
Steyerberg
EW
,
Murray
GD
,
Maas
AI
:
Early prognosis in traumatic brain injury: From prophecies to predictions.
Lancet Neurol
2010
;
9
:
543
54
251.
Muñoz-Elías
G
,
Woodbury
D
,
Black
IB
:
Marrow stromal cells, mitosis, and neuronal differentiation: Stem cell and precursor functions.
Stem Cells
2003
;
21
:
437
48
252.
Chopp
M
,
Mahmood
A
,
Lu
D
,
Li
Y
:
Editorial. Mesenchymal stem cell treatment of traumatic brain injury.
J Neurosurg
2009
;
110
:
1186
8
253.
Mahmood
A
,
Lu
D
,
Qu
C
,
Goussev
A
,
Chopp
M
:
Human marrow stromal cell treatment provides long-lasting benefit after traumatic brain injury in rats.
Neurosurgery
2005
;
57
:
1026
31; discussion 1026–31
254.
Mahmood
A
,
Lu
D
,
Qu
C
,
Goussev
A
,
Chopp
M
:
Long-term recovery after bone marrow stromal cell treatment of traumatic brain injury in rats.
J Neurosurg
2006
;
104
:
272
7
255.
Mahmood
A
,
Lu
D
,
Wang
L
,
Li
Y
,
Lu
M
,
Chopp
M
:
Treatment of traumatic brain injury in female rats with intravenous administration of bone marrow stromal cells.
Neurosurgery
2001
;
49
:
1196
203; discussion 1203–4
256.
Harting
MT
,
Jimenez
F
,
Xue
H
,
Fischer
UM
,
Baumgartner
J
,
Dash
PK
,
Cox
CS
:
Intravenous mesenchymal stem cell therapy for traumatic brain injury.
J Neurosurg
2009
;
110
:
1189
97
257.
Lu
D
,
Mahmood
A
,
Wang
L
,
Li
Y
,
Lu
M
,
Chopp
M
:
Adult bone marrow stromal cells administered intravenously to rats after traumatic brain injury migrate into brain and improve neurological outcome.
Neuroreport
2001
;
12
:
559
63
258.
Wannemuehler
TJ
,
Manukyan
MC
,
Brewster
BD
,
Rouch
J
,
Poynter
JA
,
Wang
Y
,
Meldrum
DR
:
Advances in mesenchymal stem cell research in sepsis.
J Surg Res
2012
;
173
:
113
26
259.
Angus
DC
,
Linde-Zwirble
WT
,
Lidicker
J
,
Clermont
G
,
Carcillo
J
,
Pinsky
MR
:
Epidemiology of severe sepsis in the United States: Analysis of incidence, outcome, and associated costs of care.
Crit Care Med
2001
;
29
:
1303
10
260.
Dombrovskiy
VY
,
Martin
AA
,
Sunderram
J
,
Paz
HL
:
Rapid increase in hospitalization and mortality rates for severe sepsis in the United States: A trend analysis from 1993 to 2003.
Crit Care Med
2007
;
35
:
1244
50
261.
Abraham
E
,
Wunderink
R
,
Silverman
H
,
Perl
TM
,
Nasraway
S
,
Levy
H
,
Bone
R
,
Wenzel
RP
,
Balk
R
,
Allred
R
:
Efficacy and safety of monoclonal antibody to human tumor necrosis factor alpha in patients with sepsis syndrome. A randomized, controlled, double-blind, multicenter clinical trial. TNF-alpha MAb Sepsis Study Group.
JAMA
1995
;
273
:
934
41
262.
Cohen
J
,
Carlet
J
:
INTERSEPT: An international, multicenter, placebo-controlled trial of monoclonal antibody to human tumor necrosis factor-alpha in patients with sepsis. International Sepsis Trial Study Group.
Crit Care Med
1996
;
24
:
1431
40
263.
Fisher
CJ
Jr
,
Dhainaut
JF
,
Opal
SM
,
Pribble
JP
,
Balk
RA
,
Slotman
GJ
,
Iberti
TJ
,
Rackow
EC
,
Shapiro
MJ
,
Greenman
RL
:
Recombinant human interleukin 1 receptor antagonist in the treatment of patients with sepsis syndrome. Results from a randomized, double-blind, placebo-controlled trial. Phase III rhIL-1ra Sepsis Syndrome Study Group.
JAMA
1994
;
271
:
1836
43
264.
Fisher
CJ
Jr
,
Slotman
GJ
,
Opal
SM
,
Pribble
JP
,
Bone
RC
,
Emmanuel
G
,
Ng
D
,
Bloedow
DC
,
Catalano
MA
;
IL-1RA Sepsis Syndrome Study Group
:
Initial evaluation of human recombinant interleukin-1 receptor antagonist in the treatment of sepsis syndrome: A randomized, open-label, placebo-controlled multicenter trial.
Crit Care Med
1994
;
22
:
12
21
265.
The Veterans Administration Systemic Sepsis Cooperative Study Group:
:
Effect of high-dose glucocorticoid therapy on mortality in patients with clinical signs of systemic sepsis.
N Engl J Med
1987
;
317
:
659
65
266.
Bone
RC
,
Fisher
CJ
Jr
,
Clemmer
TP
,
Slotman
GJ
,
Metz
CA
,
Balk
RA
:
A controlled clinical trial of high-dose methylprednisolone in the treatment of severe sepsis and septic shock.
N Engl J Med
1987
;
317
:
653
8
267.
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
268.
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
269.
Tomchuck
SL
,
Zwezdaryk
KJ
,
Coffelt
SB
,
Waterman
RS
,
Danka
ES
,
Scandurro
AB
:
Toll-like receptors on human mesenchymal stem cells drive their migration and immunomodulating responses.
Stem Cells
2008
;
26
:
99
107
270.
Rittirsch
D
,
Hoesel
LM
,
Ward
PA
:
The disconnect between animal models of sepsis and human sepsis.
J Leukoc Biol
2007
;
81
:
137
43
271.
de Jong
R
,
Houtgraaf
JH
,
Samiei
S
,
Boersma
E
,
Duckers
HJ
:
Intracoronary stem cell infusion after acute myocardial infarction: A meta-analysis and update on clinical trials.
Circ Cardiovasc Interv
2014
;
7
:
156
67
272.
Prabhu
SD
:
Cytokine-induced modulation of cardiac function.
Circ Res
2004
;
95
:
1140
53
273.
Romero-Bermejo
FJ
,
Ruiz-Bailen
M
,
Gil-Cebrian
J
,
Huertos-Ranchal
MJ
:
Sepsis-induced cardiomyopathy.
Curr Cardiol Rev
2011
;
7
:
163
83
274.
Bouhemad
B
,
Nicolas-Robin
A
,
Arbelot
C
,
Arthaud
M
,
Féger
F
,
Rouby
JJ
:
Isolated and reversible impairment of ventricular relaxation in patients with septic shock.
Crit Care Med
2008
;
36
:
766
74
275.
Hotchkiss
RS
,
Monneret
G
,
Payen
D
:
Immunosuppression in sepsis: A novel understanding of the disorder and a new therapeutic approach.
Lancet Infect Dis
2013
;
13
:
260
8
276.
van den Akker
F
,
de Jager
SC
,
Sluijter
JP
:
Mesenchymal stem cell therapy for cardiac inflammation: Immunomodulatory properties and the influence of toll-like receptors.
Mediators Inflamm
2013
;
2013
:
181020
277.
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
278.
Lalu
MM
,
McIntyre
L
,
Pugliese
C
,
Fergusson
D
,
Winston
BW
,
Marshall
JC
,
Granton
J
,
Stewart
DJ
;
Canadian Critical Care Trials Group
:
Safety of cell therapy with mesenchymal stromal cells (SafeCell): A systematic review and meta-analysis of clinical trials.
PLoS One
2012
;
7
:
e47559