Skip to main content

Bone marrow vs Wharton’s jelly mesenchymal stem cells in experimental sepsis: a comparative study

Abstract

Background

The use of mesenchymal stem cells (MSCs) is being extensively studied in clinical trials in the setting of various diseases including diabetes, stroke, and progressive multiple sclerosis. The unique immunomodulatory properties of MSCs also point them as a possible therapeutic tool during sepsis and septic shock, a devastating syndrome associated with 30–35% mortality. However, MSCs are not equal regarding their activity, depending on their tissue origin. Here, we aimed at comparing the in vivo properties of MSCs according to their tissue source (bone marrow (BM) versus Wharton’s jelly (WJ)) in a murine cecal ligation and puncture (CLP) model of sepsis that mimics a human peritonitis. We hypothesized that MSC properties may vary depending on their tissue source in the setting of sepsis.

Methods

CLP, adult, male, C57BL/6 mice were randomized in 3 groups receiving respectively 0.25 × 106 BM-MSCs, 0.25 × 106 WJ-MSCs, or 150 μL phosphate-buffered saline (PBS) intravenously 24 h after the CLP procedure.

Results

We observed that both types of MSCs regulated leukocyte trafficking and reduced organ dysfunction, while only WJ-MSCs were able to improve bacterial clearance and survival.

Conclusion

This study highlights the importance to determine the most appropriate source of MSCs for a given therapeutic indication and suggests a better profile for WJ-MSCs during sepsis.

Background

Mesenchymal stem cell (MSC) administration is being extensively studied in clinical trials in the setting of many different disorders such as graft versus host disease, cardiomyopathy, diabetes, stroke, bronchopulmonary dysplasia, progressive multiple sclerosis, or osteoarthritis. Indeed, MSCs are an attractive therapeutic candidate for several reasons: these cells display immunomodulatory, anti-inflammatory, antibacterial, and differentiation properties [1]. Their isolation and expansion are both easy and fast as compare to other stem cells like embryonic stem cells. They are devoid of MHC class II antigens and express only low levels of MHC class I antigens, allowing their use in an allogeneic setting due to their low immunogenicity [2]. Finally, several clinical trials reported no adverse events after MSC infusion, describing those cells as safe for clinical use [3].

Since their discovery in the bone marrow (BM) by Friedenstein’s team in 1976, MSCs have been found in the skeletal muscle, adipose tissue [4], dental pulp, trabecular bone synovial membrane, lungs [5], heart [6], synovial membrane, trabecular bone, periosteum [7, 8], and menstrual blood [9], as well as in different birth tissues, including the amniotic fluid and membrane [10], placenta [11], umbilical cord blood [12], and Wharton’s jelly (WJ) [13].

The International Society for Cellular Therapies defined MSCs as (i) CD34neg CD45neg HLADRneg CD90+ CD73+ CD105+ cells with (ii) plastic adherence (iii) and ability to differentiate into osteocytes, adipocytes, and chondrocytes. However, despite this consensual definition, MSCs remain a very heterogeneous cell population and large variations in their properties, partly related to their tissue source, have been described [14]. For example, Alcayaga et al. demonstrated a superior frequency of menstrual stem cell fibroblast colony-forming units as compared to bone marrow stem cells (BM-MSCs) [15]. Paneppucci et al. described better osteogenic differentiation when MSCs were derived from BM as compared to WJ [16]. Li et al. found that MSCs from birth tissues have stronger immunomodulatory properties than BM-derived cells [17]. Accordingly, it is essential to determine the most suitable source of MSCs to get the best-expected effect depending on the therapeutic indication considered.

Sepsis, defined as life-threatening organ dysfunction caused by a deregulated host response to infection, is a leading cause of admission to intensive care units and is associated with high mortality rates [18, 19]. Unfortunately, due to its complex physiopathology, there is still no specific treatment for this syndrome. Mei et al. [20] were the first to suggest that MSCs improve survival and decrease organ failure in a mouse model of endotoxemia, and subsequent studies showed that MSCs can increase bacterial clearance [21], modulate cytokine production [22,23,24,25], and improve renal, pulmonary, liver, cardiac, and muscular functions [21, 26,27,28,29]. Although promising, these studies used MSCs derived from adult tissues (BM and adipose tissue) exhibiting many drawbacks with regard to their potential for clinical applications: the number of adult MSC donors is limited, and adult MSCs remain difficult to produce. By contrast, fetal tissues, and particularly the umbilical cord, are much easier to obtain and MSCs are present in large numbers in these tissues and can be expanded [30].

Therefore, in this study, we compared the in vivo properties of MSCs according to their tissue source: BM versus WJ, which is an attractive source due to its abundance, during a cecal ligation and puncture model of sepsis that mimics a human peritonitis with immune deregulation, organ injury bacterial invasion [31]. We hypothesized that MSC properties may vary depending on their tissue source in the setting of sepsis.

Methods

MSC preparation

Umbilical cords were collected at Nancy Maternity Hospital from new mothers who had signed an informed consent form in compliance with the French national legislation regarding human sample collection, manipulation, and personal data protection. Umbilical cord removal was performed in parallel in a context of hematopoietic stem cell allograft. Briefly, after the removal of umbilical cord vessels, WJ was cut into small pieces and plated in a six-well plate with complete medium (minimal essential medium alpha (αMEM; Lonza, Walkersville, MD, USA) supplemented with 10% fetal bovine serum (FBS), 2 mM glutamine, 100 IU/mL penicillin, 100 μg/mL streptomycin, and 2.5 μg/mL amphotericin B). After 7 days, pieces were removed and culture continued until passage 3.

Bone marrow-MSCs were isolated from a sample of healthy human BM collected in a hematopoietic stem cell allograft context, after donors and patients’ informed consent in compliance with national legislation regarding human sample collection, manipulation, and personal data protection. Nuclear cells were seeded at 50000/cm2 in a complete medium. After 2 days, cultures were washed to eliminate non-adherent cells, the medium replaced, and cultures continued until passage 1.

MSC cultures were carried out at 37 °C in hypoxic conditions (5% of O2 and 5% of CO2).

All donors met the criteria for HSC allogeneic transplant: for example, no medical history against donation, negative serology for less than 1 month, and age lower than 50 years. To minimize the impact of donor variabilities, we used 6 donors from each MSC source.

At the end of culture, MSCs were washed with HBSS (Hanks balanced salt solution) and detached by trypsinization. One million MSCs were labeled with anti-CD90, CD73, CD44, CD105, CD34, CD45, CD11b, CD19, and HLA-DR mAbs (Stemflow hMSC Analysis kit, Becton Dickinson, Franklin Lakes, USA) for characterization; the remaining cells were frozen and stored in vapor phase nitrogen. For their use in our experimental sepsis model, MSCs were administered immediately after thawing to reflect the clinical setting.

Functional characterization of MSC

As a control of MSC characteristics, osteogenic differentiation was induced by seeding MSCs at a density of 3100 cells/cm2 and by culturing them for 28 days in an osteogenic induction medium (Lonza, USA). After 28 days, the samples were fixed in 4% paraformaldehyde and then embedded in paraffin before being stained with alizarin red. To induce adipocyte differentiation, 21,000 MSCs/cm2 were seeded onto a 24-well plate. When 100% confluence was reached, 3 cycles of induction/maintenance were performed. One cycle of induction/maintenance consisted in 3 day-culture in induction medium (Lonza, USA), followed by 1 to 3 days of culture in maintenance medium (Lonza). After 3 cycles of induction/maintenance, the cells were cultured for 7 days in complete maintenance medium (Lonza, USA) before being stained with oil red.

Cecal ligation and puncture polymicrobial sepsis model

Experiments were performed in compliance with the National Institute of Health guidelines on the use of laboratory animals and evaluated by our Institutional Animal Care and Use Committee (CELMEA-CE2A-66). Cecal ligation and puncture (CLP) was performed, as previously described [32]. Eight- to 10-week-old male C57BL/6 mice were anesthetized by isoflurane inhalation (4% isoflurane for induction; 1.5% isoflurane for maintenance). After laparotomy, the distal end of the cecum was ligated, a single perforation was performed with an 18-gauge needle, and a small amount of stool was taken out. The cecum was then replaced into the peritoneal cavity, and the abdominal incision was sutured in two layers with 4.0 nylon suture. Five hundred microliters of 0.9% NaCl was administered sub-cutaneously for fluid resuscitation. Mice were then immediately randomized in three groups: BM-MSC, WJ-MSC, and phosphate-buffered saline (PBS) by a person who did not perform surgery. Twenty-four hours after CLP procedure, 2.5 × 105 MSCs in 150 μl of PBS or 150 μl PBS alone were slowly administered into the retro-orbital sinus under sevoflurane anesthesia.

Inflammation studies

Forty-eight hours or 7 days after CLP procedure, animals were sacrificed by pentobarbital i.p. injection and blood and organs were harvested. Blood count was determined by a hemocytometer, and plasma concentrations of IL1β, IL-6, IL-10, IFNγ, and TNFα were measured by multiplex immunoassays (Bio-Plex Pro Mouse Th1 cytokine, Biorad, France) according to the manufacturer’s recommendations. The same protocol was carried out on healthy mice (H0).

Leukocyte trafficking was analyzed by flow cytometry as previously described [33]. The spleen and liver were crushed in HBSS and filtered on a 70-μm nylon filter. The bone marrow was extracted from the femur, after the bone has been clipped, by rapidly injecting 1 ml of PBS into the medullary cavity. The lungs were cut into fine pieces and incubated in a cocktail of collagenase I and DNase I at 37 °C for 45 min before being crushed and filtered. After washing, a cell count was performed by a hemocytometer with Trypan blue staining (BioRad). Cell suspensions were labeled with a combination of anti-CD4-PerCP, CD25-PE, CD11b-Vioblue, Ly6C-FITC, Ly6G-PE, FoxP3-APC, and CD45-PerCP mAbs (Miltenyi, France) after permeabilization according to the manufacturer’s recommendations. Data were acquired on a Gallios FACS analyzer (Beckman Coulter). The same protocol was carried out on healthy mice (H0).

Bacterial count

The blood and spleen were obtained 48 h after the CLP procedure. The blood and crushed spleen were plated in serial log dilutions on blood agar plates. After plating, tryptic soy agar plates were incubated at 37 °C aerobically for 24 h and anaerobically for 48 h and colony-forming units (CFUs) were counted. Results are expressed as CFUs per milliliter of blood or per gram of spleen.

Analysis of organ injuries

Organ dysfunction was determined by measurement of biochemical indicators of organ functions in plasma samples and by histology scores. Plasma concentrations of urea and creatinine (indicators of renal dysfunction), alanine aminotransferase (ALT) and alkaline phosphatase (ALKP) (indicators of liver dysfunction), and amylase and glycemia (indicators of pancreatic dysfunction) were analyzed by an automate (VetTest GHP, Idexx, Saint-Denis, France) after the animals’ sacrifice 2 or 7 days after the CLP procedure.

Forty-eight hours after CLP, the lungs, liver, kidneys, and spleen were fixed in 4% paraformaldehyde during 24 h at 4 °C, dehydrated through a series of ethanol concentrations, cleared with toluene, embedded in paraffin wax, and cut into 5-mm-thick sections with a Leica micro-tome (RM2135, Leica, France). For histological examination, each specimen was stained with hematoxylin-erythrosin-saffron, mounted on glass slides, and visualized on an optical microscope (DMD 108, Leica, France).

Histology scores were performed by an experienced pathologist blinded to the treatment administered. The scoring system was adapted from [34, 35] and ranged from 0 to 9 for the lungs, 0 to 5 for the kidneys, 0 to 4 for the liver, and 0 to 3 for the spleen.

Survival study

The same previously described CLP procedure was performed for the survival study. All mice received every 12 h 50 μg/g of imipenem (Braun, France) sub-cutaneously for the survival study in accordance with Alcayaga-Miranda’s work which described a cumulative effect of MSCs and antibiotics [15]. Animals were followed up to 7 days.

Statistics

The normal distributions of the data were tested (Kolmogorov-Smirnov test), and data are presented as means ± SD. Between-group differences were tested for significance by two-way ANOVA with Bonferroni correction or Kruskal-Wallis test when appropriate. Analyses were performed using GraphPad Prism software.

Results

All the MSCs used in this study presented the typical MSC phenotype CD14neg-CD34neg-HLA-DRneg-CD11bneg-CD19neg-CD73+-CD90+-CD105+-CD44+, differentiation capacities into osteocytes and adipocytes, and an adherence to plastic in accordance with the standards described by the International Society for Cellular Therapy.

MSCs impact leukocyte trafficking

Sepsis induced an early neutrophilia and a progressive monocytosis and lymphocytosis (Fig. 1). MSCs showed no effect on these disturbances, except for a reduced late (day 7) monocytosis with BM-MSCs. However, no significant difference was found between WJ-MSC and BM-MSC. Of note, sepsis was also associated with deep and rapid thrombocytopenia, on which MSCs had no effect.

Fig. 1
figure 1

Evolution of blood cell count. Blood cell analysis was performed before (H0) and 2 and 7 days after the induction of sepsis. Results are expressed as mean ± SD (n = 3–6 per group). Group comparisons were analyzed by two-way ANOVA with Bonferroni correction. §p < 0.05 BM-MSC versus PBS

We next investigated leukocyte trafficking in various organs (Fig. 2). Following CLP, neutrophils progressively populated the liver, spleen, lungs, and BM. This was partially prevented by MSCs regardless of their origin (Fig. 2a). Monocytes were also recruited by the lungs and the liver, though to a less extent in MSC-treated animals (Fig. 2b). In mouse, monocytes are classically categorized as “pro-inflammatory” Ly6Chigh and “anti-inflammatory” Ly6Clow. Mobilization of Ly6Chigh monocytes to the lung was reduced in mice receiving MSCs regardless of their origin while Ly6Clow was largely unaffected except in the liver (Fig. 2c, d). Likewise, we observed a significant reduction of Ly6Chigh monocytes in BM in the WJ-MSC group in comparison to the PBS group. However, no significant difference was found between WJ-MSC and BM-MSC. Regarding Treg-cells, known to favor resolution of inflammation, WJ-MSC-treated mice displayed higher numbers in their lungs, liver, and BM, than BM-MSC-treated mice though with no significant differences as compared to control animals (Fig. 2e). Altogether, these data show that MSCs are able to impact leukocyte trafficking, towards a more “anti-inflammatory” profile conferred after treatment by WJ-derived cells.

Fig. 2
figure 2

MSCs regulate leukocyte trafficking. Flow cytometric quantification of neutrophils (a), monocytes (b), monocytes Ly6Chigh (c), monocytes Ly6Clow (c), and T regulatory lymphocytes (e) in the spleen, lung, liver, and bone marrow at different time points. Results are expressed as mean ± SD (n = 4–6 per group). *p < 0.05 WJ-MSC versus PBS; **p < 0.01 WJ-MSC versus PBS; ***p < 0.001 WJ-MSC versus PBS; §p < 0.05 BM-MSC versus PBS; §§p < 0.01 BM-MSC versus PBS; §§§p < 0.001 BM-MSC versus PBS; #p < 0.05 BM-MSC versus WJ-MSC; ##p < 0.01 BM-MSC versus WJ-MSC

MSCs do not dampen systemic inflammation but reduce organ injury

As expected, sepsis induced an acute increase in both inflammatory (TNFα, IL1β, IL-6) and anti-inflammatory (IL-10) plasma cytokine concentrations (Fig. 3). Surprisingly, we observed no effect of MSCs on cytokine levels. Moderate kidney and liver dysfunctions develop after CLP (Fig. 4a). These early disturbances were partly prevented by MSCs regardless of their source. Histology studies of the lungs, spleens, kidneys, and livers revealed no significant differences between groups (Fig. 4b).

Fig. 3
figure 3

MSCs have no effect on plasma cytokine concentrations. Plasma concentrations of IL1β, IL-6, IL-10, IFNγ, and TNFα were determined by multiplex assay at baseline (H0) and 2 and 7 days after CLP. Results are expressed as mean ± SD(n = 5–10 per group). Group comparisons were analyzed by two-way ANOVA with Bonferroni correction

Fig. 4
figure 4

Effects of MSCs on organ dysfunction. Plasma concentrations of blood urea, creatinine, ALT, ALKP, amylase, and glycemia were measured at baseline and 2 or 7 days after CLP procedure (n = 3–7 per group) (a). The lungs, livers, kidneys, and spleens were harvested 48 h after CLP, and a pathologist blinded to the treatment group performed histology scoring (n = 4 per group) (b). Group comparisons were analyzed by two-way ANOVA with Bonferroni correction. *p < 0.05 WJ-MSC versus PBS; §p < 0.05 BM-MSC versus PBS; §§p < 0.01 BM-MSC versus PBS

MSCs improve bacterial clearance

We observed an important reduction of blood and spleen bacterial load as compared to controls after the administration of WJ-MSCs (Fig. 5). Although the trend was similar for BM-MSCs, it did not reach significance. Of note, 100% of control animals were bacteremic compared to 71% and 75% in the WJ- and BM-MSC groups respectively (Fig. 5b).

Fig. 5
figure 5

WJ-MSCs enhance bacterial clearance. Spleen (a) and blood (b) bacterial CFU were assessed 48 h after the induction of sepsis. Results are expressed as median (n = 7–12 per group). *p < 0.05 WJ-MSCs versus PBS. Group comparisons were analyzed by Kruskal-Wallis test

MSCs improve survival

Finally, we investigated the effects of MSCs on survival (Fig. 6). In the control group (antibiotics and fluid resuscitation), lethality reached 36% at day 7, but only 13% and 17% in the WJ- and BM-MSC-treated animals respectively. Significance was only obtained for the WJ-MSC group (Wilcoxon test, p = 0.04). No significant difference was found between WJ-MSC and BM-MSC groups.

Fig. 6
figure 6

WJ-MSCs improve survival. Kaplan-Meier estimate of survival after CLP (n = 18–48 per group). Survival curves were compared using the log-rank test. Group comparisons were analyzed by Wilcoxon test *p < 0.05 WJ-MSCs versus PBS

Discussion

Although the use of MSCs as a therapeutic tool has generated a great enthusiasm, important questions remain especially regarding their optimal tissue source that may depend on the target disorder. Sepsis is one of the diseases under scrutiny for MSC administration. This syndrome being a vital emergency, allogenic MSC banks will be necessary and the optimal MSC source must be determined. We aimed here to address whether in vivo MSC properties during sepsis could change according to their adult or fetal origin.

Indeed, we observed slightly different effects of MSCs depending on their tissue source. First, WJ-MSCs seem to be more potent than BM-MSCs to modulate leukocyte trafficking, conferring a more “anti-inflammatory” environment in organs with a lower neutrophil infiltration. Previous in vitro studies suggested that MSCs extracted from birth tissues have more immunomodulatory capacities than cells derived from adult tissues. Mareschi et al. studied the proliferative capacity of regulatory T cells during co-culture of mononuclear cells and MSCs from the birth tissue or bone marrow [36] and noted an increase in Treg cell proliferation with MSCs from birth tissue. Similarly, Barcia et al. observed that WJ-MSCs seemed to be less immunogenic and more immunosuppressive than BM-MSCs [37].

Although we demonstrated an important impact of MSCs on cellular traffic, no effects were noted on plasma cytokine concentrations. Our results contrast with previous findings demonstrating the MSC ability to increase anti-inflammatory cytokines and decrease pro-inflammatory cytokines [22,23,24, 38]. But it is crucial to underline the fact that we here administered MSCs as a treatment and not as a prophylactic agent: most if not all previous studies on MSCs and sepsis used cells at the same time (or even before) or just after the onset of sepsis. Here, we chose to give cells only 24 h after surgery (and thus analyzed cytokines at 48 h), to better mimic what could happen in the clinical setting. Therefore, our time window could have precluded from observing an MSC effect on cytokines, as their concentration peak at 24 h as observed in the Liu et al. study [39]. However, in a previous work in pigs, we also were unable to demonstrate an impact of an early WJ-MSC administration on plasma cytokines [29]. In a recent clinical trial investigating the effect of adipose tissue-derived MSCs after an intravenous injection of lipopolysaccharide (LPS) into healthy subjects, these cells did not decrease TNFα or IL6 release and only the higher MSC dose (4 × 106/kg) increased plasma level of IL10 [40]. It is also important to note that in these studies MSCs have been cryopreserved. The impact of freezing is unclear [41,42,43,44], and therefore, an effect on MSC anti-inflammatory properties cannot be excluded.

The antibacterial properties of MSCs seem also affected by their origin. Indeed, even if no significant difference was found between WJ-MSC and BM-MSC, we observed a significant decrease in bacterial clearance only with WJ-MSCs in comparison to the PBS group. Differences in terms of antimicrobial peptide secretion have been previously described. For example, Alcayaga et al. showed that BM-MSCs secrete LL-37, hepcidin, and β defensins, whereas MSCs derived from the menstrual liquid secrete only hepcidin, and MSCs from the umbilical cord produce only β defensins [45]. Obviously, in vitro experiments cannot reflect the complex environment of sepsis and therefore the behavior of in vivo MSCs. Moreover, antibacterial properties of MSCs are not just the consequence of antimicrobial peptide production as they can also increase neutrophil and monocyte phagocytic index [46, 47], a mechanism complex to reproduce in vitro.

Although the better profile of MSCs regarding their immunomodulatory and antibacterial properties should have been translated into a dampening of organ dysfunction, this was oddly not observed. An explanation could stem from the fact that our model was not severe enough to drive severe organ failure, as suggested by modest biochemistry and histologic disorders in the sacrificed mice 2 days after the CLP procedure.

Finally, only WJ-MSCs were able to improve survival in comparison to the PBS group, though it is important to note that no difference was observed between WJ-MSC and BM-MSC groups.

In summary, we observed very slight differences between MSC capacities depending on tissue source such as their impact on regulatory T cells but none justifying the use of a source over the other in terms of potency. Currently, three phase 1 clinical trials have reported the safety of using MSCs during septic shock: two using MSCs derived from adult tissues (bone marrow and adipose tissue) and the third using MSCs from the umbilical cord [40, 48, 49]. These studies demonstrate that intravenous infusion of allogeneic MSCs in a septic context was safe. None adverse effect was observed even at high doses, regardless of their origin. However, to treat a syndrome with a high prevalence such as sepsis, the accessibility to the source of MSC is essential and the number of adult MSC donors is limited. In this context, WJ is a more advantageous tissue source of MSCs: donations of the umbilical cord are devoid of risk and abundant, the amount of MSCs in an umbilical cord is important, and their expansion is fast and quite easy.

This study presents some limitations. First, we used MSCs at different passages. BM-MSCs were cryopreserved at passage 1 while WJ-MSC at passage 3. Indeed, the BM-MSC senescence is earlier than the WJ-MSC senescence [50]. Although unlikely, whether this could have negatively impacted WJ-MSC properties is unknown. Second, we could have taken into account differences in terms of BM and WJ donor characteristics which have been found to influence MSC functions such as age, gender, weight, smoking status, diabetes, or obstetric factors [51,52,53,54,55,56]. Third, we tested a single dose of MSCs. However, even if the efficiency of higher amounts of cells is unknown, the dose of 0.25 × 106 cells per animal corresponds to a high dose in humans (around 10 × 106 cells/kg).

Conclusion

We here showed that the effects of MSC administration in an in vivo model of murine sepsis marginally depend on their source, with an at least as potent profile conferred by WJ-MSCs.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Abbreviations

ALAT:

Alanine aminotransferase

ALKP:

Alkaline phosphatase

BM:

Bone marrow

BM-MSCs:

Mesenchymal stem cells derived from the bone marrow

CLP:

Cecal ligation and puncture

LPS:

Lipopolysaccharide

Ly6Chigh :

Inflammatory monocytes

Ly6Clow :

Anti-inflammatory monocytes

MHC:

Major histocompatibility complex

MSCs:

Mesenchymal stem cells

PBS:

Phosphate-buffered saline

Tregs:

Regulatory T cells

WJ-MSCs:

Mesenchymal stem cells derived from Wharton jelly

References

  1. Laroye C, Gibot S, Reppel L, Bensoussan D. Concise review: mesenchymal stromal/stem cells: a new treatment for sepsis and septic shock?: MSCs in the treatment for sepsis and septic shock. Stem Cells. 2017;35:2331–9.

    Article  Google Scholar 

  2. de Girolamo L, Lucarelli E, Alessandri G, Antonietta Avanzini M, Ester Bernardo M, Biagi E, et al. Mesenchymal stem/stromal cells: a new “cells as drugs” paradigm. Efficacy and critical aspects in cell therapy. Curr Pharm Des. 2013;19:2459–73.

    Article  Google Scholar 

  3. Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall JC, et al. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 2012;7:e47559 Beltrami AP, editor.

    Article  CAS  Google Scholar 

  4. Fraser JK, Wulur I, Alfonso Z, Hedrick MH. Fat tissue: an underappreciated source of stem cells for biotechnology. Trends Biotechnol. 2006;24:150–4.

    Article  CAS  Google Scholar 

  5. Griffiths MJ, Bonnet D, Janes SM. Stem cells of the alveolar epithelium. Lancet. 2005;366:249–60.

    Article  Google Scholar 

  6. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763–76.

    Article  CAS  Google Scholar 

  7. Chen Y, Shao J-Z, Xiang L-X, Dong X-J, Zhang G-R. Mesenchymal stem cells: a promising candidate in regenerative medicine. Int J Biochem Cell Biol. 2008;40:815–20.

    Article  CAS  Google Scholar 

  8. Sancricca C. Mesenchymal stem cells: molecular characteristics and clinical applications. World J Stem Cells. 2010;2:67.

    Article  Google Scholar 

  9. Rossignoli F, Caselli A, Grisendi G, Piccinno S, Burns JS, Murgia A, et al. Isolation, characterization, and transduction of endometrial decidual tissue multipotent mesenchymal stromal/stem cells from menstrual blood. Biomed Res Int. 2013;2013:1–14.

    Article  Google Scholar 

  10. Wolbank S, Griensven M, Grillari-Voglauer R, Peterbauer-Scherb A. Alternative sources of adult stem cells: human amniotic membrane. In: Kasper C, Griensven M, Pörtner R, editors. Bioreact Syst Tissue Eng II [Internet]. Berlin, Heidelberg: Springer Berlin Heidelberg; 2010. p. 1–27. [cited 2016 Mar 31]. Available from: http://link.springer.com/10.1007/10_2010_71.

    Google Scholar 

  11. In ’t Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GMJS, Claas FHJ, Fibbe WE, et al. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004;22:1338–45.

    Article  Google Scholar 

  12. Bieback K, Kern S, Klüter H, Eichler H. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells. 2004;22:625–34.

    Article  Google Scholar 

  13. Wang H-S, Hung S-C, Peng S-T, Huang C-C, Wei H-M, Guo Y-J, et al. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells. 2004;22:1330–7.

    Article  Google Scholar 

  14. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, Krause DS, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7.

    Article  CAS  Google Scholar 

  15. Alcayaga-Miranda F, Cuenca J, Martin A, Contreras L, Figueroa FE, Khoury M. Combination therapy of menstrual derived mesenchymal stem cells and antibiotics ameliorates survival in sepsis. Stem Cell Res Ther. 2015;6:199.

    Article  Google Scholar 

  16. Panepucci RA, Siufi JLC, Silva WA, Proto-Siquiera R, Neder L, Orellana M, et al. Comparison of gene expression of umbilical cord vein and bone marrow-derived mesenchymal stem cells. Stem Cells. 2004;22:1263–78.

    Article  CAS  Google Scholar 

  17. Li X, Bai J, Ji X, Li R, Xuan Y, Wang Y. Comprehensive characterization of four different populations of human mesenchymal stem cells as regards their immune properties, proliferation and differentiation. Int J Mol Med. 2014;34:695–704.

    Article  Google Scholar 

  18. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 2016;315:801.

    Article  CAS  Google Scholar 

  19. Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX, Deutschman CS, et al. Developing a new definition and assessing new clinical criteria for septic shock: for the third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 2016;315:775.

    Article  CAS  Google Scholar 

  20. 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.

    Article  Google Scholar 

  21. Mei SHJ, Haitsma JJ, Dos Santos CC, Deng Y, Lai PFH, Slutsky AS, et al. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am J Respir Crit Care Med. 2010;182:1047–57.

    Article  CAS  Google Scholar 

  22. 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.

    Article  CAS  Google Scholar 

  23. Pedrazza L, Lunardelli A, Luft C, Cruz CU, de Mesquita FC, Bitencourt S, et al. Mesenchymal stem cells decrease splenocytes apoptosis in a sepsis experimental model. Inflamm Res Off J Eur Histamine Res Soc Al. 2014;63:719–28.

    CAS  Google Scholar 

  24. Kim H, Darwish I, Monroy M-F, Prockop DJ, Liles WC, Kain KC. Mesenchymal stromal (stem) cells suppress pro-inflammatory cytokine production but fail to improve survival in experimental staphylococcal toxic shock syndrome. BMC Immunol. 2014;15:1.

    Article  Google Scholar 

  25. Németh K, Leelahavanichkul A, Yuen PST, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E2–dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15:42–9.

    Article  Google Scholar 

  26. Yagi H, Soto-Gutierrez A, Kitagawa Y, Tilles AW, Tompkins RG, Yarmush ML. Bone marrow mesenchymal stromal cells attenuate organ injury induced by LPS and burn. Cell Transplant. 2010;19:823–30.

    Article  Google Scholar 

  27. Rojas M, Xu J, Woods CR, Mora AL, Spears W, Roman J, et al. Bone marrow–derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol. 2005;33:145–52.

    Article  CAS  Google Scholar 

  28. Rocheteau P, Chatre L, Briand D, Mebarki M, Jouvion G, Bardon J, et al. Sepsis induces long-term metabolic and mitochondrial muscle stem cell dysfunction amenable by mesenchymal stem cell therapy. Nat Commun. 2015;6:10145.

    Article  CAS  Google Scholar 

  29. Laroye C, Lemarié J, Boufenzer A, Labroca P, Cunat L, Alauzet C, et al. Clinical-grade mesenchymal stem cells derived from umbilical cord improve septic shock in pigs. Intensive Care Med Exp. 2018;6:24.

    Article  Google Scholar 

  30. Jin H, Bae Y, Kim M, Kwon S-J, Jeon H, Choi S, et al. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int J Mol Sci. 2013;14:17986–8001.

    Article  Google Scholar 

  31. Dejager L, Pinheiro I, Dejonckheere E, Libert C. Cecal ligation and puncture: the gold standard model for polymicrobial sepsis? Trends Microbiol. 2011;19:198–208.

    Article  CAS  Google Scholar 

  32. Gibot S. A soluble form of the triggering receptor expressed on myeloid cells-1 modulates the inflammatory response in murine Sepsis. J Exp Med. 2004;200:1419–26.

    Article  CAS  Google Scholar 

  33. Jolly L, Carrasco K, Derive M, Lemarié J, Boufenzer A, Gibot S. Targeted endothelial gene deletion of triggering receptor expressed on myeloid cells-1 protects mice during septic shock. Cardiovasc Res. 2018;114:907–18.

    Article  CAS  Google Scholar 

  34. Hall SRR, Tsoyi K, Ith B, Padera RF, Lederer JA, Wang Z, et al. Mesenchymal stromal cells improve survival during sepsis in the absence of heme oxygenase-1: the importance of neutrophils. Stem Cells. 2013;31:397–407.

    Article  CAS  Google Scholar 

  35. Chen H-H, Chang C-L, Lin K-C, Sung P-H, Chai H-T, Zhen Y-Y, et al. Melatonin augments apoptotic adipose-derived mesenchymal stem cell treatment against sepsis-induced acute lung injury. Am J Transl Res. 2014;6:439.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Mareschi K, Castiglia S, Sanavio F, Rustichelli D, Muraro M, Defedele D, et al. Immunoregulatory effects on T lymphocytes by human mesenchymal stromal cells isolated from bone marrow, amniotic fluid, and placenta. Exp Hematol. 2016;44:138–150.e1.

    Article  CAS  Google Scholar 

  37. Bárcia RN, Santos JM, Filipe M, Teixeira M, Martins JP, Almeida J, et al. What makes umbilical cord tissue-derived mesenchymal stromal cells superior immunomodulators when compared to bone marrow derived mesenchymal stromal cells? Stem Cells Int. 2015;2015:1–14.

    Article  Google Scholar 

  38. Chao Y-H, Wu H-P, Wu K-H, Tsai Y-G, Peng C-T, Lin K-C, et al. An increase in CD3+CD4+CD25+ regulatory T cells after administration of umbilical cord-derived mesenchymal stem cells during sepsis. PLoS One. 2014;9:e110338 Zimmer J, editor.

    Article  Google Scholar 

  39. Liu W, Gao Y, Li H, Wang H, Ye M, Jiang G, et al. Intravenous transplantation of mesenchymal stromal cells has therapeutic effects in a sepsis mouse model through inhibition of septic natural killer cells. Int J Biochem Cell Biol. 2016;79:93–103.

    Article  CAS  Google Scholar 

  40. Perlee D, van Vught LA, Scicluna BP, Maag A, Lutter R, Kemper EM, et al. Intravenous infusion of human adipose mesenchymal stem cells modifies the host response to lipopolysaccharide in humans: a randomized, single-blind, parallel group, placebo controlled trial: mesenchymal stem cells in human endotoxemia. Stem Cells. 2018;36:1778–88.

    Article  CAS  Google Scholar 

  41. Moll G, Alm JJ, Davies LC, von Bahr L, Heldring N, Stenbeck-Funke L, et al. Do cryopreserved mesenchymal stromal cells display impaired immunomodulatory and therapeutic properties?: therapeutic efficacy of fresh versus thawed MSCs. Stem Cells. 2014;32:2430–42.

    Article  CAS  Google Scholar 

  42. François M, Copland IB, Yuan S, Romieu-Mourez R, Waller EK, Galipeau J. Cryopreserved mesenchymal stromal cells display impaired immunosuppressive properties as a result of heat-shock response and impaired interferon-γ licensing. Cytotherapy. 2012;14:147–52.

    Article  Google Scholar 

  43. Luetzkendorf J, Nerger K, Hering J, Moegel A, Hoffmann K, Hoefers C, et al. Cryopreservation does not alter main characteristics of good manufacturing process–grade human multipotent mesenchymal stromal cells including immunomodulating potential and lack of malignant transformation. Cytotherapy. 2015;17:186–98.

    Article  Google Scholar 

  44. Bárcia RN, Santos JM, Teixeira M, Filipe M, Pereira ARS, Ministro A, et al. Umbilical cord tissue–derived mesenchymal stromal cells maintain immunomodulatory and angiogenic potencies after cryopreservation and subsequent thawing. Cytotherapy. 2017;19:360–70.

    Article  Google Scholar 

  45. Alcayaga-Miranda F, Cuenca J, Khoury M. Antimicrobial activity of mesenchymal stem cells: current status and new perspectives of antimicrobial peptide-based therapies. Front Immunol. 2017;8:339.

    Article  Google Scholar 

  46. Jackson MV, Morrison TJ, Doherty DF, McAuley DF, Matthay MA, Kissenpfennig A, et al. Mitochondrial transfer via tunneling nanotubes (TNT) is an important mechanism by which mesenchymal stem cells enhance macrophage phagocytosis in the in vitro and in vivo models of ARDS: mitochondrial transfer from MSC to macrophages. Stem Cells. 2016;34:2210–23.

    Article  CAS  Google Scholar 

  47. Krasnodembskaya A, Samarani G, Song Y, Zhuo H, Su X, Lee J-W, et al. Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. AJP Lung Cell Mol Physiol. 2012;302:L1003–13.

    Article  CAS  Google Scholar 

  48. McIntyre LA, Stewart DJ, Mei SHJ, Courtman D, Watpool I, Granton J, et al. Cellular immunotherapy for septic shock. A phase I clinical trial. Am J Respir Crit Care Med. 2018;197:337–47.

    Article  CAS  Google Scholar 

  49. He X. Umbilical cord-derived mesenchymal stem (stromal) cells for treatment of severe sepsis: a phase 1 clinical trial. Transl Res. 2018;199:10.

    Article  Google Scholar 

  50. Batsali AK, Pontikoglou C, Koutroulakis D, Pavlaki KI, Damianaki A, Mavroudi I, et al. Differential expression of cell cycle and WNT pathway-related genes accounts for differences in the growth and differentiation potential of Wharton’s jelly and bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther. 2017;8:102.

    Article  Google Scholar 

  51. Avercenc-Léger L, Guerci P, Virion J-M, Cauchois G, Hupont S, Rahouadj R, et al. Umbilical cord-derived mesenchymal stromal cells: predictive obstetric factors for cell proliferation and chondrogenic differentiation. Stem Cell Res Ther. 2017;8:161.

    Article  Google Scholar 

  52. Sammour I, Somashekar S, Huang J, Batlahally S, Breton M, Valasaki K, et al. The effect of gender on mesenchymal stem cell (MSC) efficacy in neonatal hyperoxia-induced lung injury. PLoS One. 2016;11:e0164269 Kirchmair R, editor.

    Article  Google Scholar 

  53. Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Donor age negatively impacts adipose tissue-derived mesenchymal stem cell expansion and differentiation. J Transl Med. 2014;12:8.

    Article  Google Scholar 

  54. Greenberg JM, Carballosa CM, Cheung HS. Concise review: the deleterious effects of cigarette smoking and nicotine usage and mesenchymal stem cell function and implications for cell-based therapies: cigarette smoking, nicotine, and MSC function. Stem Cells Transl Med. 2017;6:1815–21.

    Article  Google Scholar 

  55. Boyle KE, Patinkin ZW, Shapiro ALB, Baker PR, Dabelea D, Friedman JE. Mesenchymal stem cells from infants born to obese mothers exhibit greater potential for adipogenesis: the healthy start BabyBUMP project. Diabetes. 2016;65:647–59.

    Article  CAS  Google Scholar 

  56. Pierdomenico L, Lanuti P, Lachmann R, Grifone G, Cianci E, Gialò L, et al. Diabetes mellitus during pregnancy interferes with the biological characteristics of Wharton’s jelly mesenchymal stem cells. Open Tissue Eng Regen Med J. 2011;4 [cited 2017 Aug 2]. Available from: https://benthamopen.com/ABSTRACT/TOTERMJ-4-103.

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank Natalia Da Isla, Naceur Cherif, Gishlaine Cauchois, and Anne-Laure Leblanc for the technical assistance.

Funding

This work was supported by INSERM U1116, IMoPA UMR 7365, and a grant from BPI France and La Région Lorraine.

Author information

Authors and Affiliations

Authors

Contributions

CL performed the experiments, analyzed the data, and wrote the manuscript. AB, LJ, CA, and LC, CY, JLM performed the experiments and analyzed the data. DB and LR analyzed the data. SG designed the study and analyzed the data. All authors approved the final version of the manuscript.

Corresponding author

Correspondence to Caroline Laroye.

Ethics declarations

Ethics approval and consent to participate

This study was carried out in accordance with the recommendations of the French national legislation regarding human sample collection, manipulation, and personal data protection with written informed consent from all subjects. All subjects gave written informed consent in accordance with the Declaration of Helsinki. The protocol was approved by the Nancy Hospital ethics committee and French ministry of research (No. DC-2014-2114).

This study was carried out in accordance with the recommendations of the National Institute of Health guidelines on the Use of Laboratory Animals. The protocol was approved by the University Animal Care Committee (Comité d’Éthique Lorrain en Matière d’Expérimentation Animale (CELMEA - CE2A-66)).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Laroye, C., Boufenzer, A., Jolly, L. et al. Bone marrow vs Wharton’s jelly mesenchymal stem cells in experimental sepsis: a comparative study. Stem Cell Res Ther 10, 192 (2019). https://doi.org/10.1186/s13287-019-1295-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-019-1295-9

Keywords