Skip to main content
Erschienen in: Molecular and Cellular Pediatrics 1/2016

Open Access 01.12.2016 | Mini review

Mesenchymal stem/stromal cells—a key mediator for regeneration after perinatal morbidity?

Erschienen in: Molecular and Cellular Pediatrics | Ausgabe 1/2016

Abstract

Perinatal complications in both term- and preterm-born infants are a leading cause of neonatal morbidities and mortality. Infants face different challenges in the neonatal intensive care unit with long-term morbidities such as perinatal brain injury and bronchopulmonary dysplasia being particularly devastating. While advances in perinatal medicine have improved our understanding of the pathogenesis, effective therapies to prevent and/or reduce the severity of these disorders are still lacking. The potential of mesenchymal stem/stromal cell (MSC) therapy has emerged during the last two decades, and an increasing effort is conducted to address brain- and lung-related morbidities in neonates at risk. Various studies support the notion that MSCs have protective effects. MSCs are an easy source and may be readily available after birth in a clinical setting. MSCs’ mechanisms of action are diverse, including migration and homing, release of growth factors and immunomodulation, and the potential to replace injured cells. Here, we review the pathophysiology of perinatally acquired brain and lung injuries and focus on MSCs as potential candidates for therapeutic strategies summarizing preclinical and clinical evidence.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

All authors have made substantive intellectual contributions to this manuscript. MM and BK designed, drafted, and wrote the manuscript. TW, AS, AG, and DS revised and provided critical and important intellectual content. All authors have given final approval of the version to be published and agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Introduction

Despite advances in obstetrics and neonatology, both preterm- and term-born infants continue to face serious risks during pregnancy, parturition, and adaptation after birth. Perinatal medicine is therefore a major public health issue around the world. The clinical presentation after perinatal complications or preterm birth in an individual child is complex. This complexity results from multiple potential causal pathways, signs, and symptoms of injury. Typical pathologies in these newborns include perinatal brain injury and bronchopulmonary dysplasia (BPD), among others. New avenues to treat these morbidities have emerged among which stem cells being particularly promising. The body of knowledge of stem cell biology and their function in tissue regeneration and protection has broadened exponentially in the last two decades. This review will discuss mesenchymal stem/stromal cells (MSCs) as a therapeutic approach for both brain and lung diseases in infants at risk. Furthermore, we will highlight the preclinical and clinical data that have emerged on the role of MSCs in perinatal medicine.

Perinatally acquired organ injury

Perinatally acquired organ injury affects both term and preterm infants. Depending on the timing of injury and/or delivery, infants need to cope with different challenges. In the developing brain of a preterm infant, the spectrum of injury suggests that the underlying pathophysiology is not due to a single lesion but consists of white and gray matter disturbances [1]. Thus, a comprehensive multidimensional assessment of potential contributing factors such as maternal medical history, obstetric antecedents, intrapartum factors (including fetal heart rate monitoring results and issues related to the delivery itself), and placental pathology is recommended [2]. In the term-born infants, perinatal insults such as birth asphyxia or perinatal stroke affect 1 to 3 newborns out of 1000 [3, 4]. In contrast, in preterm-born infants morbidity and mortality strongly relate to the gestational age. While preterm birth before 37 weeks of gestation occurs in 5–8 % of all pregnancies, very low gestational age (VLGA) before 32 weeks of gestation occurs in about 1 % of singletons and 9 % of twin pregnancies [5]. Mortality of VLGA infants ranges between 7.3 and 21.4 % at 30 days and 9.0 and 22.7 % at 1 year [6]. Additionally, a large number of survivors suffer significant long-term disabilities including cerebral palsy (CP), epilepsy, increased hyperactivity, and developmental disorders [7]. For example, the risk to develop CP is 30 times higher in infants born before 33 weeks of gestation compared to term-born infants [8]. Moreover, injury in these infants is frequently exacerbated by fetal inflammation and preferentially affects cerebral white matter resulting in periventricular leukomalacia and germinal matrix hemorrhage [1]. Currently, the only intervention known to reduce the burden of perinatal brain injury in the term population is hypothermia. Several large clinical trials confirmed that hypothermia in infants with neonatal hypoxic-ischemic encephalopathy is associated with a significant reduction in death and disability [9]. However, 40–50 % of infants treated with hypothermia still die or develop significant neurological disability [10]. In the preterm population, therapeutic options are lacking as hypothermia is contra-productive. Antenatal magnesium sulfate prior to birth at less than 30 weeks of gestation reduces CP and combined CP and mortality rate at 2 years of age. However, randomized control trials do not demonstrate long-term neurological benefits [11, 12]. Approximately 25 % of VLGA infants will develop BPD with long-lasting consequences such as chronic respiratory impairment and neurodevelopmental delay [13]. Changes in clinical management reduced the incidence of BPD significantly with a shift from VLGA to extreme low gestational age newborns developing BPD. Not surprisingly, the current pathogenesis of BPD is based on immaturity with disordered alveolar and capillary development and represents a developmental disorder [14]. The plasticity of the developing lung after preterm birth is poorly understood. Long-term follow-up studies suggest an incomplete regeneration of the lung growth in survivors with BPD. Current treatment options for BPD include vitamin A and caffeine in addition to supportive therapies, but their results often remain unsatisfactory [15].
Together, therapeutic approaches to counteract the consequences of perinatally acquired injury are sparse, and new measures are desperately needed especially for preterm infants [1, 1517].

Pathophysiology of brain and lung injury

Experimental studies identified different phases of perinatally induced neuronal death. Primary neuronal death is related to depletion of tissue energy reserves with primary energy failure. The secondary and tertiary phases are related to excitotoxicity, mitochondrial dysfunction, and free radical accumulation leading to cell necrosis or apoptosis with impaired myelination and/or axonal function [18]. The secondary and tertiary phases may cause persistent inflammation and gliosis, sensitization to further injury, and impaired oligodendrocyte maturation and myelination [19]. Interestingly, inflammation is increasingly recognized as being a critical contributor to both normal development and injury outcome especially in the immature brain [20]. Maternal infection/inflammation is not only a major risk for preterm birth but is linked to systemic fetal inflammatory response which, in turn, may elicit injury in the fetus. Perinatal inflammation modulates vulnerability to and development of brain injury [21] and influences critical phases of myelination and cortical plasticity [20]. Several studies suggest that inflammation may play a critical role in autism and schizophrenia [22]. Together, brain development, myelination, vascularization, and apoptosis are strongly influenced by inflammatory responses in both physiologic and pathophysiologic conditions [20, 23]. Pivotal regulators of inflammatory responses in the brain are glial cells which orchestrate the release of pro- and anti-inflammatory cytokines [24, 25]. Pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α and interferon gamma (IFN-γ) are cytotoxic to oligodendrocytes [26]. However, glial cells may produce anti-inflammatory cytokines (e.g., IL-10), which suppress expansion of IL-1β and TNF-α and contribute to resolving inflammation and repair processes [25]. Further, astrogliotic scar formation installs a barrier around tissue lesions to restrict the crossing of inflammatory cells into surrounding healthy areas [24].
The regulation of the inflammatory responses in the newborn appears to be a link that may explain some of the common features of organ injury in preterm infants. Many studies identified characteristic inflammatory changes and altered growth factor signaling in BPD such as initial influx of neutrophils into the lung followed by increased numbers of macrophages [27]. The release of cytokines and disturbance of growth factor signaling such as transforming growth factor (TGF)-β results in increased apoptotic process [28]. Furthermore, several studies showed that dysmorphic capillaries and subsequent development of pulmonary hypertension are related to an altered pattern of angiogenic growth factors such as vascular endothelial growth factor (VEGF) and its receptors [29]. Finally, lung injury leads to remodeling or early alveolar epithelial dysfunction which in turn promotes lung inflammation [30].
Taken together, perinatal inflammation is a strong modulator of both physiologic and pathophysiologic development of the neonatal organs. The fetal inflammatory response to certain cues such as lipopolysaccharides (LPS) [31] can be detected not only in the brain and the lung but also in remote tissues not directly exposed to LPS such as the spleen, liver, and mediastinal lymph nodes [3234]. Protective strategies to counteract the cascades leading to injury should therefore not focus on one organ or system but rather treat perinatally acquired injury globally, in which the immune system plays a key role. MSCs possess a regenerative potential. They were shown to modulate innate and adaptive immune responses, to have antiapoptotic effects, to decrease inflammation, and to enhance tissue repair, mostly through the release of paracrine factors [35, 36].

Mesenchymal stem/stromal cells

Stem cells are broadly defined as cells with self-renewing and differentiation capacity. Although stem cells derived from embryonic tissue were identified first, the clinical use is limited due to ethical concerns and tumorigenic potential [37, 38]. Clinical and animal stem cell-based studies to prevent or repair perinatally acquired injury have emerged during the recent years with MSCs being particularly promising. These cells are considered somatic stem cells as they originate from stem cell niches such as bone marrow (BM), skin, adipose umbilical cord, and placental tissues [39]. MSCs can be isolated from placental membranes and tissues [40, 41], amniotic fluid [4244], umbilical cord blood [45, 46], and the umbilical cord connective tissues (Wharton’s jelly; WJ) [47, 48]. Although all of these cells are MSCs, specific considerations with respect to clinical use, time of application, application route, availability, and ethical aspects need to be made.
MSC-based therapies are an attractive strategy since the pathophysiology of perinatally acquired injury is heterogeneous and MSCs have the capacity to adapt to the microenvironment of injured organs. The strategy may be either or both replacement/restoration of lost tissue and/or protection/salvage of injured cells. In term infants at risk for hypoxic-ischemic injury or neonatal ischemic stroke, MSCs could exert a neuroprotective effect starting at the acute phase of injury. The timing and presentation of the injury are usually well defined. MSCs could provide trophic support and/or amelioration of the inflammatory responses, leading to repair or reduced cell death. However, the different cell types, transplantation routes, and the timing need to be accounted for. Given the gold standard therapy of hypothermia for this kind of injury, MSCs have to proof additive/synergistic effects in order to be considered [49]. In contrast, in the preterm population, the timing of the injury is often unclear and the pathophysiology is more complex. The clinical diagnosis of infants at risk is challenging as symptoms such as CP or BPD are diagnosed in early childhood years. Thus, the injury may be considered more chronic as extensive atrophy and gliosis of the white matter tract or dysfunction of lung architecture are present. MSCs could modulate not only the inflammatory response after delivery but also the degree and magnitude of the injured white matter and epithelial cells as well. However, many questions such as altered pattern of growth factors and intercellular matrix proteins that could affect proliferation or differentiation of the desired cell types need to be addressed first.

MSCs: homing and migration

The approach of MSCs as a therapy for perinatal injury is based on several crucial properties of MSCs, including delivery of the cells “homing” to the site of injury. Migration and homing to the tissue of injury is influenced by multiple factors including age, passage, and number of cells; culture conditions; and delivery method [50]. The apparent migration and homing abilities of MSCs without tumorigenic potential were described by several groups and in different disease models [5154]. In BPD animal models, peripherally injected cells were detected in the hyperoxia-induced injured lung [55, 56]. Experimental studies identified chemokines as major molecules responsible for cell homing with chemokine receptors CXCR3, CXCR4, and CXCR6 being particularly important [5759]. Further secretion of factors such as stromal cell-derived factor-1α (SDF-1α), which is a CXCR4 ligand, promotes migration of MSC to the injury site [60]. Interestingly, the phenotype of MSCs is an important criterion as well. CD9 (high)-positive MSCs display improved engraftment compared to the CD9 (low)-positive population in a murine ischemic hind limb model [61]. This observation highlights the rather heterogeneous MSC population and the importance of proper MSC characterization and definition for future studies. Currently, MSC characterization is based on a set of minimal criteria [62], and they display a cell surface repertoire and gene expression pattern which differ among MSCs from various tissues of origin and culture conditions used [6366]. For example, MSCs derived from amniotic fluid express many cell surface markers characteristic for BM-derived MSCs including CD73, CD90, CD105, and major histocompatibility complex (MHC) class I [44]. The lack of MHC class II, CD40, CD80, and CD86 molecules suggests a low immunogenic phenotype of MSCs when compared to other stem cell sources [67]. In contrast, WJ-MSCs express cell surface markers CD29, CD44, CD73, CD90, CD105, CD146, and CD166 [68, 69] and are considered more primitive cell population relative to BM-derived MSCs [70]. As a result, WJ-MSCs differentiate more efficiently into neural progenitors compared to BM-derived MSC [71]. In addition, the underlying clinical condition may also affect the phenotype of MSCs. WJ-MSCs derived from umbilical cords collected after preeclamptic pregnancies seem to be more committed to neuroglial differentiation compared to cords from uncomplicated pregnancies [66].

MSCs: secretome and immunomodulation

While MSCs have a proven restorative capacity in response to injury cues, the question of potential protective mechanisms remains unclear. Most of the available data comes from adult neurodegenerative and lung diseases or in vitro studies. Studies identified the induction of cytokines, interleukins, and trophic factors predominately involved in neurogenesis, angiogenesis, hematopoiesis, and cardiovascular regeneration being crucial for the mostly paracrine effects [58, 72]. For example, WJ-MSCs’ secretome triggers neuronal survival and differentiation in vitro and in vivo [73, 74]. Secreted factors such as VEGF-A, angiopoietin-1, fibroblast growth factor (FGF)-I, hepatocyte growth factor (HGF), FGF-II, brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and platelet-derived growth factor (PDGF)-AB were identified [7579]. Importantly, MSCs’ secretome alters both adaptive and innate immune responses [55, 80]. MSCs inhibit autoreactive T cell responses in animal models of multiple sclerosis and hypoxic-ischemic brain injury [8183]. MSCs shift the alveolar macrophages from a M1 (pro-inflammatory) to a M2 (protective) phenotype ameliorating pulmonary injury in acute LPS-induced acute lung injury model [84]. Thus, the shifting from the M1 to the M2 states and promoting regulatory T cells are a function unique to MSCs [85]. Besides T cell modulation, MSCs inhibit B cell proliferation, neutrophil and monocyte function, and NK toxicity [8689]. Although these modulatory effects are partially understood, direct cell-to-cell contact and soluble factors are relevant [90]. Additionally, MSC effects expand beyond constitutive immune modulatory properties with the release of cytokines and growth factors such as VEGF, transforming growth factor beta-1 (TGF-β1), TNF-α, interleukin-1 (IL-1), interleukin-6 (IL-6), and IFN-γ [9194].

MSCs: regeneration/replacement of injured cells

MSCs’ multipotency and self-renewal properties make them valid candidates for providing both lung and brain cell regeneration/replacement. Although, this strategy for repair carries risks such as tumorigenic potential [37, 38], MSCs were successfully differentiated into various types of cells including cardiomyocytes, myocytes, and epidermal and endothelial cells [54, 9597]. Importantly, MSCs express neuroglial commitment and can be differentiated into lung cells as well [98102]. Not surprisingly, MSCs are currently tested in various animal models and clinical trials for lung and brain regeneration [103105]. Although this line of investigation is particularly intriguing, MSCs’ potential to replace injured cells is not proven and is a matter of constant debate [103, 104, 106]. For example, intravenously injected MSCs improve myocardial infarction without permanent replacement of injured cells [107]. In the lung, MSCs embolize causing endothelial damage and are cleared in a matter of hours [107]. Taken together, the MSCs’ low rate of in vivo engraftment and differentiation suggests that transplanted cells affect tissue injury and repair through paracrine factors. Whether the factors released by the MSCs or the cells themselves are more promising for the therapy of lung and brain injury in the newborn still remains an open question.

MSCs: extracellular vehicles

The translation from bench to bedside requires the most efficacious and safest approach. Thus, the question of cell-based versus cell-free therapy needs to be addressed. Given that the MSCs’ therapeutic potential has been shown to be largely triggered via paracrine effects and not differentiation, recent studies focus on extracellular vehicles (EV) [108]. These are all types of vehicles present in the extracellular space, including shedding vesicles, apoptotic bodies, and exosomes. Exosomes (40–100 nm in diameter) are secreted by cells in a regulated fashion, possess the ability to transfer proteins and functional genetic materials such as mRNA and microRNAs, and are involved in cell-to-cell signaling and regulation [80]. Not surprisingly, MSC-derived EV are contributing to tissue repair in brain injury including stroke and Alzheimer’s disease [109111]. The cell-free approach is very promising; however, it is still in its infancy. In fact, stem cells do not just secrete growth factors and/or cytokines but encourage the growth and even supplement (host) cells [112114]. Importantly, the stem cells’ potential of immunomodulation and protection after injury seems to depend on the bidirectional communication between the injured host cells and the graft via the exchange of specific information [115].

MSCs: clinical trials

As a result of the remarkable regenerative potential of MSCs, MSCs are ideal candidates for clinical cell therapy. MSCs are easily available, have a good safety profile and homing capacity, and importantly are relatively immunoprivileged, allowing allogeneic transplantation. Not surprisingly, MSCs have been tested in clinical trials in several neurodegenerative diseases such as stroke [116118], amyotrophic lateral sclerosis [119], multiple sclerosis [120, 121], and spinal cord injury [122]. Several clinical trials indicated no serious side effects or dose-limiting toxicity in acute respiratory distress syndrome [123] and chronic obstructive pulmonary disease [124, 125]. Also, safety and feasibility trials for CP [126128] and BPD [129, 130] were successful. A recent double-blind randomized control study used allogeneic umbilical cord blood, in combination with erythropoietin, in children diagnosed with CP showing motor and cognitive benefits [131]. In contrast to previous studies, a large safety study used autologous umbilical cord blood directly after birth for infants at risk for hypoxic-ischemic encephalopathy [132]. This approach differs significantly from previous studies as it aims mainly to prevent and not to replace affected cells. Cells were transplanted directly after birth and in combination with hypothermia. Authors concluded that the collection, preparation, and infusion of fresh autologous umbilical cord blood cells for use in infants with hypoxic-ischemic encephalopathy are feasible. The feasibility and safety of intra-tracheal infusion of umbilical cord-derived MSCs in BPD was reported as well [129]. MSCs reduced BPD severity and retinopathy of prematurity and improved inflammatory cytokine profile in tracheal aspirates in a phase 1 dose escalation study. Several other clinical trials for neonatal brain and lung injury are currently listed as in progress or completed, and more results should become available in the near future (ClinicalTrials.gov Identifiers: NCT01297205, NCT01828957, NCT02023788, NCT01832454, NCT01962233, NCT01988584, NCT01207869).

Conclusions

Given the array of potential regenerative mechanisms of MSCs to protect neonatal brain and lungs, therapeutic applications can be envisioned in the near future. The mechanisms span anti-apoptotic/pro-mitotic capacities leading to neovascularization by the stimulation of angiogenesis and anti-inflammatory responses. Further, stimulation of neuro- and gliogenesis, synaptogenesis, neurite outgrowth, and also immunomodulation are crucial. Available data clearly demonstrate that MSC and secreted factors are beneficial to treat a variety of neurodegenerative and lung disorders. Although the data are very promising, critical questions need to be answered:
  • First, what are the appropriate age, passage, and dosage of the transplanted MSCs? Freshly isolated MSCs may be beneficial compared to cultured cells, but is this practicable in a clinical setting especially in the context of preterm infants [133]?
  • What is the best source and application route of MSCs? Banking of UCB and placental tissue offer an easily accessible and ethically responsible source of MSCs, and minimally invasive routes are currently tested [47, 66, 134136].
  • Finally, how does the transplantation of MSCs impact the standard care in the neonatal intensive care unit? The lack of effective interventions for many morbidities related to prematurity unlocks the potential of cell-based personalized treatments. Safe and effective clinical interventions are future perspectives bearing hope to improve the lifelong outcomes of the infants in our care. This has to be proven in long-term follow-up studies.
Open AccessThis 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.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

All authors have made substantive intellectual contributions to this manuscript. MM and BK designed, drafted, and wrote the manuscript. TW, AS, AG, and DS revised and provided critical and important intellectual content. All authors have given final approval of the version to be published and agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Literatur
2.
Zurück zum Zitat Executive summary: Neonatal encephalopathy and neurologic outcome, second edition. Report of the American College of Obstetricians and Gynecologists' Task Force on Neonatal Encephalopathy (2014). Obstet Gynecol 123 (4):896-901. doi:10.1097/01.AOG.0000445580.65983.d2 Executive summary: Neonatal encephalopathy and neurologic outcome, second edition. Report of the American College of Obstetricians and Gynecologists' Task Force on Neonatal Encephalopathy (2014). Obstet Gynecol 123 (4):896-901. doi:10.1097/01.AOG.0000445580.65983.d2
4.
Zurück zum Zitat Cowan FM, Mercuri E, Rutherford MA (2005) Perinatal stroke in term infants with neonatal encephalopathy. Neurology 64(3):579, Author reply 579PubMedCrossRef Cowan FM, Mercuri E, Rutherford MA (2005) Perinatal stroke in term infants with neonatal encephalopathy. Neurology 64(3):579, Author reply 579PubMedCrossRef
6.
Zurück zum Zitat Numerato D, Fattore G, Tediosi F, Zanini R, Peltola M, Banks H, Mihalicza P, Lehtonen L, Svereus S, Heijink R, Klitkou ST, Fletcher E, Heijden A, Lundberg F, Over E, Hakkinen U, Seppala TT (2015) Mortality and length of stay of very low birth weight and very preterm infants: a EuroHOPE study. PLoS ONE 10(6):e0131685, 10.1371/journal.pone.0131685PubMedCentralPubMedCrossRef Numerato D, Fattore G, Tediosi F, Zanini R, Peltola M, Banks H, Mihalicza P, Lehtonen L, Svereus S, Heijink R, Klitkou ST, Fletcher E, Heijden A, Lundberg F, Over E, Hakkinen U, Seppala TT (2015) Mortality and length of stay of very low birth weight and very preterm infants: a EuroHOPE study. PLoS ONE 10(6):e0131685, 10.1371/journal.pone.0131685PubMedCentralPubMedCrossRef
8.
Zurück zum Zitat MacLennan AH, Thompson SC, Gecz J (2015) Cerebral palsy: causes, pathways, and the role of genetic variants. Am J Obstet Gynecol. doi:10.1016/j.ajog.2015.05.034. MacLennan AH, Thompson SC, Gecz J (2015) Cerebral palsy: causes, pathways, and the role of genetic variants. Am J Obstet Gynecol. doi:10.1016/j.ajog.2015.05.034.
9.
Zurück zum Zitat Higgins RD, Raju T, Edwards AD, Azzopardi DV, Bose CL, Clark RH, Ferriero DM, Guillet R, Gunn AJ, Hagberg H, Hirtz D, Inder TE, Jacobs SE, Jenkins D, Juul S, Laptook AR, Lucey JF, Maze M, Palmer C, Papile L, Pfister RH, Robertson NJ, Rutherford M, Shankaran S, Silverstein FS, Soll RF, Thoresen M, Walsh WF (2011) Hypothermia and other treatment options for neonatal encephalopathy: an executive summary of the Eunice Kennedy Shriver NICHD workshop. J Pediatr 159(5):851–858. doi:10.1016/j.jpeds.2011.08.004, e851PubMedCentralPubMedCrossRef Higgins RD, Raju T, Edwards AD, Azzopardi DV, Bose CL, Clark RH, Ferriero DM, Guillet R, Gunn AJ, Hagberg H, Hirtz D, Inder TE, Jacobs SE, Jenkins D, Juul S, Laptook AR, Lucey JF, Maze M, Palmer C, Papile L, Pfister RH, Robertson NJ, Rutherford M, Shankaran S, Silverstein FS, Soll RF, Thoresen M, Walsh WF (2011) Hypothermia and other treatment options for neonatal encephalopathy: an executive summary of the Eunice Kennedy Shriver NICHD workshop. J Pediatr 159(5):851–858. doi:10.​1016/​j.​jpeds.​2011.​08.​004, e851PubMedCentralPubMedCrossRef
10.
Zurück zum Zitat Edwards AD, Brocklehurst P, Gunn AJ, Halliday H, Juszczak E, Levene M, Strohm B, Thoresen M, Whitelaw A, Azzopardi D (2010) Neurological outcomes at 18 months of age after moderate hypothermia for perinatal hypoxic ischaemic encephalopathy: synthesis and meta-analysis of trial data. BMJ 340:c363. doi:10.1136/bmj.c363 PubMedCentralPubMedCrossRef Edwards AD, Brocklehurst P, Gunn AJ, Halliday H, Juszczak E, Levene M, Strohm B, Thoresen M, Whitelaw A, Azzopardi D (2010) Neurological outcomes at 18 months of age after moderate hypothermia for perinatal hypoxic ischaemic encephalopathy: synthesis and meta-analysis of trial data. BMJ 340:c363. doi:10.​1136/​bmj.​c363 PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Doyle LW, Anderson PJ, Haslam R, Lee KJ, Crowther C, Australasian Collaborative Trial of Magnesium Sulphate Study G (2014) School-age outcomes of very preterm infants after antenatal treatment with magnesium sulfate vs placebo. JAMA 312(11):1105–1113. doi:10.1001/jama.2014.11189 CrossRef Doyle LW, Anderson PJ, Haslam R, Lee KJ, Crowther C, Australasian Collaborative Trial of Magnesium Sulphate Study G (2014) School-age outcomes of very preterm infants after antenatal treatment with magnesium sulfate vs placebo. JAMA 312(11):1105–1113. doi:10.​1001/​jama.​2014.​11189 CrossRef
15.
16.
Zurück zum Zitat Hassell KJ, Ezzati M, Alonso-Alconada D, Hausenloy DJ, Robertson NJ (2015) New horizons for newborn brain protection: enhancing endogenous neuroprotection. Arch Dis Child Fetal Neonatal Ed. doi:10.1136/archdischild-2014-306284. Hassell KJ, Ezzati M, Alonso-Alconada D, Hausenloy DJ, Robertson NJ (2015) New horizons for newborn brain protection: enhancing endogenous neuroprotection. Arch Dis Child Fetal Neonatal Ed. doi:10.1136/archdischild-2014-306284.
17.
Zurück zum Zitat Buonocore G, Turrisi G, Kramer BW, Balduini W, Perrone S (2012) New pharmacological approaches in infants with hypoxic-ischemic encephalopathy., Curr Pharm Des Buonocore G, Turrisi G, Kramer BW, Balduini W, Perrone S (2012) New pharmacological approaches in infants with hypoxic-ischemic encephalopathy., Curr Pharm Des
18.
Zurück zum Zitat Pleasure D, Soulika A, Singh SK, Gallo V, Bannerman P (2006) Inflammation in white matter: clinical and pathophysiological aspects. Ment Retard Dev Disabil Res Rev 12(2):141–146. doi:10.1002/mrdd.20100 PubMedCrossRef Pleasure D, Soulika A, Singh SK, Gallo V, Bannerman P (2006) Inflammation in white matter: clinical and pathophysiological aspects. Ment Retard Dev Disabil Res Rev 12(2):141–146. doi:10.​1002/​mrdd.​20100 PubMedCrossRef
19.
Zurück zum Zitat Favrais G, van de Looij Y, Fleiss B, Ramanantsoa N, Bonnin P, Stoltenburg-Didinger G, Lacaud A, Saliba E, Dammann O, Gallego J, Sizonenko S, Hagberg H, Lelievre V, Gressens P (2011) Systemic inflammation disrupts the developmental program of white matter. Ann Neurol 70(4):550–565. doi:10.1002/ana.22489 PubMedCrossRef Favrais G, van de Looij Y, Fleiss B, Ramanantsoa N, Bonnin P, Stoltenburg-Didinger G, Lacaud A, Saliba E, Dammann O, Gallego J, Sizonenko S, Hagberg H, Lelievre V, Gressens P (2011) Systemic inflammation disrupts the developmental program of white matter. Ann Neurol 70(4):550–565. doi:10.​1002/​ana.​22489 PubMedCrossRef
21.
Zurück zum Zitat Eklind S, Mallard C, Leverin AL, Gilland E, Blomgren K, Mattsby-Baltzer I, Hagberg H (2001) Bacterial endotoxin sensitizes the immature brain to hypoxic-ischaemic injury. Eur J Neurosci 13(6):1101–1106PubMedCrossRef Eklind S, Mallard C, Leverin AL, Gilland E, Blomgren K, Mattsby-Baltzer I, Hagberg H (2001) Bacterial endotoxin sensitizes the immature brain to hypoxic-ischaemic injury. Eur J Neurosci 13(6):1101–1106PubMedCrossRef
23.
Zurück zum Zitat Shrivastava K, Chertoff M, Llovera G, Recasens M, Acarin L (2012) Short and long-term analysis and comparison of neurodegeneration and inflammatory cell response in the ipsilateral and contralateral hemisphere of the neonatal mouse brain after hypoxia/ischemia. Neurol Res Int 2012:781512. doi:10.1155/2012/781512 PubMedCentralPubMed Shrivastava K, Chertoff M, Llovera G, Recasens M, Acarin L (2012) Short and long-term analysis and comparison of neurodegeneration and inflammatory cell response in the ipsilateral and contralateral hemisphere of the neonatal mouse brain after hypoxia/ischemia. Neurol Res Int 2012:781512. doi:10.​1155/​2012/​781512 PubMedCentralPubMed
26.
Zurück zum Zitat Buntinx M, Moreels M, Vandenabeele F, Lambrichts I, Raus J, Steels P, Stinissen P, Ameloot M (2004) Cytokine-induced cell death in human oligodendroglial cell lines: I. Synergistic effects of IFN-gamma and TNF-alpha on apoptosis. J Neurosci Res 76(6):834–845. doi:10.1002/jnr.20118 PubMedCrossRef Buntinx M, Moreels M, Vandenabeele F, Lambrichts I, Raus J, Steels P, Stinissen P, Ameloot M (2004) Cytokine-induced cell death in human oligodendroglial cell lines: I. Synergistic effects of IFN-gamma and TNF-alpha on apoptosis. J Neurosci Res 76(6):834–845. doi:10.​1002/​jnr.​20118 PubMedCrossRef
30.
Zurück zum Zitat Atochina-Vasserman EN, Bates SR, Zhang P, Abramova H, Zhang Z, Gonzales L, Tao JQ, Gochuico BR, Gahl W, Guo CJ, Gow AJ, Beers MF, Guttentag S (2011) Early alveolar epithelial dysfunction promotes lung inflammation in a mouse model of Hermansky-Pudlak syndrome. Am J Respir Crit Care Med 184(4):449–458. doi:10.1164/rccm.201011-1882OC PubMedCentralPubMedCrossRef Atochina-Vasserman EN, Bates SR, Zhang P, Abramova H, Zhang Z, Gonzales L, Tao JQ, Gochuico BR, Gahl W, Guo CJ, Gow AJ, Beers MF, Guttentag S (2011) Early alveolar epithelial dysfunction promotes lung inflammation in a mouse model of Hermansky-Pudlak syndrome. Am J Respir Crit Care Med 184(4):449–458. doi:10.​1164/​rccm.​201011-1882OC PubMedCentralPubMedCrossRef
31.
Zurück zum Zitat Martinez-Lopez DG, Funderburg NT, Cerissi A, Rifaie R, Aviles-Medina L, Llorens-Bonilla BJ, Sleasman J, Luciano AA (2014) Lipopolysaccharide and soluble CD14 in cord blood plasma are associated with prematurity and chorioamnionitis. Pediatr Res 75(1-1):67–74. doi:10.1038/pr.2013.182 PubMedCrossRef Martinez-Lopez DG, Funderburg NT, Cerissi A, Rifaie R, Aviles-Medina L, Llorens-Bonilla BJ, Sleasman J, Luciano AA (2014) Lipopolysaccharide and soluble CD14 in cord blood plasma are associated with prematurity and chorioamnionitis. Pediatr Res 75(1-1):67–74. doi:10.​1038/​pr.​2013.​182 PubMedCrossRef
33.
Zurück zum Zitat Strackx E, Sparnaaij MA, Vlassaks E, Jellema R, Kuypers E, Vles JS, Kramer BW, Gavilanes AW (2015) Lipopolysaccharide-induced chorioamnionitis causes acute inflammatory changes in the ovine central nervous system. CNS Neurol Disord Drug Targets 14(1):77–84PubMedCrossRef Strackx E, Sparnaaij MA, Vlassaks E, Jellema R, Kuypers E, Vles JS, Kramer BW, Gavilanes AW (2015) Lipopolysaccharide-induced chorioamnionitis causes acute inflammatory changes in the ovine central nervous system. CNS Neurol Disord Drug Targets 14(1):77–84PubMedCrossRef
34.
Zurück zum Zitat Kuypers E, Willems MG, Jellema RK, Kemp MW, Newnham JP, Delhaas T, Kallapur SG, Jobe AH, Wolfs TG, Kramer BW (2015) Responses of the spleen to intraamniotic lipopolysaccharide exposure in fetal sheep. Pediatr Res 77(1-1):29–35. doi:10.1038/pr.2014.152 PubMedCrossRef Kuypers E, Willems MG, Jellema RK, Kemp MW, Newnham JP, Delhaas T, Kallapur SG, Jobe AH, Wolfs TG, Kramer BW (2015) Responses of the spleen to intraamniotic lipopolysaccharide exposure in fetal sheep. Pediatr Res 77(1-1):29–35. doi:10.​1038/​pr.​2014.​152 PubMedCrossRef
39.
40.
Zurück zum Zitat In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GM, Claas FH, Fibbe WE, Kanhai HH (2004) Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 22(7):1338–1345. doi:10.1634/stemcells.2004-0058 PubMedCrossRef In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GM, Claas FH, Fibbe WE, Kanhai HH (2004) Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 22(7):1338–1345. doi:10.​1634/​stemcells.​2004-0058 PubMedCrossRef
41.
Zurück zum Zitat Igura K, Zhang X, Takahashi K, Mitsuru A, Yamaguchi S, Takashi TA (2004) Isolation and characterization of mesenchymal progenitor cells from chorionic villi of human placenta. Cytotherapy 6(6):543–553PubMedCrossRef Igura K, Zhang X, Takahashi K, Mitsuru A, Yamaguchi S, Takashi TA (2004) Isolation and characterization of mesenchymal progenitor cells from chorionic villi of human placenta. Cytotherapy 6(6):543–553PubMedCrossRef
42.
Zurück zum Zitat Stefanidis K, Loutradis D, Anastasiadou V, Bletsa R, Kiapekou E, Drakakis P, Beretsos P, Elenis E, Mesogitis S, Antsaklis A (2008) Oxytocin receptor- and Oct-4-expressing cells in human amniotic fluid. Gynecol Endocrinol 24(5):280–284. doi:10.1080/09513590801977167 PubMedCrossRef Stefanidis K, Loutradis D, Anastasiadou V, Bletsa R, Kiapekou E, Drakakis P, Beretsos P, Elenis E, Mesogitis S, Antsaklis A (2008) Oxytocin receptor- and Oct-4-expressing cells in human amniotic fluid. Gynecol Endocrinol 24(5):280–284. doi:10.​1080/​0951359080197716​7 PubMedCrossRef
43.
Zurück zum Zitat Prusa AR, Marton E, Rosner M, Bernaschek G, Hengstschlager M (2003) Oct-4-expressing cells in human amniotic fluid: a new source for stem cell research? Hum Reprod 18(7):1489–1493PubMedCrossRef Prusa AR, Marton E, Rosner M, Bernaschek G, Hengstschlager M (2003) Oct-4-expressing cells in human amniotic fluid: a new source for stem cell research? Hum Reprod 18(7):1489–1493PubMedCrossRef
44.
Zurück zum Zitat In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FH, Willemze R, Fibbe WE, Kanhai HH (2003) Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 102(4):1548–1549. doi:10.1182/blood-2003-04-1291 PubMedCrossRef In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FH, Willemze R, Fibbe WE, Kanhai HH (2003) Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 102(4):1548–1549. doi:10.​1182/​blood-2003-04-1291 PubMedCrossRef
45.
Zurück zum Zitat Wang JF, Wang LJ, Wu YF, Xiang Y, Xie CG, Jia BB, Harrington J, McNiece IK (2004) Mesenchymal stem/progenitor cells in human umbilical cord blood as support for ex vivo expansion of CD34(+) hematopoietic stem cells and for chondrogenic differentiation. Haematologica 89(7):837–844PubMed Wang JF, Wang LJ, Wu YF, Xiang Y, Xie CG, Jia BB, Harrington J, McNiece IK (2004) Mesenchymal stem/progenitor cells in human umbilical cord blood as support for ex vivo expansion of CD34(+) hematopoietic stem cells and for chondrogenic differentiation. Haematologica 89(7):837–844PubMed
46.
Zurück zum Zitat Erices A, Conget P, Minguell JJ (2000) Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 109(1):235–242PubMedCrossRef Erices A, Conget P, Minguell JJ (2000) Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 109(1):235–242PubMedCrossRef
47.
Zurück zum Zitat Ma L, Feng XY, Cui BL, Law F, Jiang XW, Yang LY, Xie QD, Huang TH (2005) Human umbilical cord Wharton's Jelly-derived mesenchymal stem cells differentiation into nerve-like cells. Chin Med J (Engl) 118(23):1987–1993 Ma L, Feng XY, Cui BL, Law F, Jiang XW, Yang LY, Xie QD, Huang TH (2005) Human umbilical cord Wharton's Jelly-derived mesenchymal stem cells differentiation into nerve-like cells. Chin Med J (Engl) 118(23):1987–1993
53.
Zurück zum Zitat Schoeberlein A, Mueller M, Reinhart U, Sager R, Messerli M, Surbek DV (2011) Homing of placenta-derived mesenchymal stem cells after perinatal intracerebral transplantation in a rat model. Am J Obstet Gynecol 205(3):277. doi:10.1016/j.ajog.2011.06.044, e271-276PubMedCrossRef Schoeberlein A, Mueller M, Reinhart U, Sager R, Messerli M, Surbek DV (2011) Homing of placenta-derived mesenchymal stem cells after perinatal intracerebral transplantation in a rat model. Am J Obstet Gynecol 205(3):277. doi:10.​1016/​j.​ajog.​2011.​06.​044, e271-276PubMedCrossRef
54.
Zurück zum Zitat De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L, Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ, Furth ME, Soker S, Atala A (2007) Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 25(1):100–106. doi:10.1038/nbt1274 PubMedCrossRef De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L, Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ, Furth ME, Soker S, Atala A (2007) Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 25(1):100–106. doi:10.​1038/​nbt1274 PubMedCrossRef
56.
Zurück zum Zitat Zhang X, Wang H, Shi Y, Peng W, Zhang S, Zhang W, Xu J, Mei Y, Feng Z (2012) Role of bone marrow-derived mesenchymal stem cells in the prevention of hyperoxia-induced lung injury in newborn mice. Cell Biol Int 36(6):589–594. doi:10.1042/CBI20110447 PubMedCrossRef Zhang X, Wang H, Shi Y, Peng W, Zhang S, Zhang W, Xu J, Mei Y, Feng Z (2012) Role of bone marrow-derived mesenchymal stem cells in the prevention of hyperoxia-induced lung injury in newborn mice. Cell Biol Int 36(6):589–594. doi:10.​1042/​CBI20110447 PubMedCrossRef
58.
Zurück zum Zitat Gao LR, Zhang NK, Ding QA, Chen HY, Hu X, Jiang S, Li TC, Chen Y, Wang ZG, Ye Y, Zhu ZM (2013) Common expression of stemness molecular markers and early cardiac transcription factors in human Wharton's jelly-derived mesenchymal stem cells and embryonic stem cells. Cell Transplant 22(10):1883–1900. doi:10.3727/096368912X662444 PubMedCrossRef Gao LR, Zhang NK, Ding QA, Chen HY, Hu X, Jiang S, Li TC, Chen Y, Wang ZG, Ye Y, Zhu ZM (2013) Common expression of stemness molecular markers and early cardiac transcription factors in human Wharton's jelly-derived mesenchymal stem cells and embryonic stem cells. Cell Transplant 22(10):1883–1900. doi:10.​3727/​096368912X662444​ PubMedCrossRef
60.
Zurück zum Zitat Yang DY, Sheu ML, Su HL, Cheng FC, Chen YJ, Chen CJ, Chiu WT, Yiin JJ, Sheehan J, Pan HC (2012) Dual regeneration of muscle and nerve by intravenous administration of human amniotic fluid-derived mesenchymal stem cells regulated by stromal cell-derived factor-1alpha in a sciatic nerve injury model. J Neurosurg 116(6):1357–1367. doi:10.3171/2012.2.JNS111360 PubMedCrossRef Yang DY, Sheu ML, Su HL, Cheng FC, Chen YJ, Chen CJ, Chiu WT, Yiin JJ, Sheehan J, Pan HC (2012) Dual regeneration of muscle and nerve by intravenous administration of human amniotic fluid-derived mesenchymal stem cells regulated by stromal cell-derived factor-1alpha in a sciatic nerve injury model. J Neurosurg 116(6):1357–1367. doi:10.​3171/​2012.​2.​JNS111360 PubMedCrossRef
61.
62.
Zurück zum Zitat Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8(4):315–317. doi:10.1080/14653240600855905 PubMedCrossRef Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8(4):315–317. doi:10.​1080/​1465324060085590​5 PubMedCrossRef
64.
Zurück zum Zitat Brooke G, Tong H, Levesque JP, Atkinson K (2008) Molecular trafficking mechanisms of multipotent mesenchymal stem cells derived from human bone marrow and placenta. Stem Cells Dev 17(5):929–940. doi:10.1089/scd.2007.0156 PubMedCrossRef Brooke G, Tong H, Levesque JP, Atkinson K (2008) Molecular trafficking mechanisms of multipotent mesenchymal stem cells derived from human bone marrow and placenta. Stem Cells Dev 17(5):929–940. doi:10.​1089/​scd.​2007.​0156 PubMedCrossRef
65.
Zurück zum Zitat Mariotti E, Mirabelli P, Abate G, Schiattarella M, Martinelli P, Fortunato G, Di Noto R, Del Vecchio L (2008) Comparative characteristics of mesenchymal stem cells from human bone marrow and placenta: CD10, CD49d, and CD56 make a difference. Stem Cells Dev 17(6):1039–1041. doi:10.1089/scd.2008.0212 PubMedCrossRef Mariotti E, Mirabelli P, Abate G, Schiattarella M, Martinelli P, Fortunato G, Di Noto R, Del Vecchio L (2008) Comparative characteristics of mesenchymal stem cells from human bone marrow and placenta: CD10, CD49d, and CD56 make a difference. Stem Cells Dev 17(6):1039–1041. doi:10.​1089/​scd.​2008.​0212 PubMedCrossRef
66.
Zurück zum Zitat Joerger-Messerli M, Bruhlmann E, Bessire A, Wagner A, Mueller M, Surbek DV, Schoeberlein A (2015) Preeclampsia enhances neuroglial marker expression in umbilical cord Wharton's jelly-derived mesenchymal stem cells. J Matern Fetal Neonatal Med 28(4):464–469. doi:10.3109/14767058.2014.921671 PubMedCrossRef Joerger-Messerli M, Bruhlmann E, Bessire A, Wagner A, Mueller M, Surbek DV, Schoeberlein A (2015) Preeclampsia enhances neuroglial marker expression in umbilical cord Wharton's jelly-derived mesenchymal stem cells. J Matern Fetal Neonatal Med 28(4):464–469. doi:10.​3109/​14767058.​2014.​921671 PubMedCrossRef
68.
Zurück zum Zitat Fong CY, Chak LL, Biswas A, Tan JH, Gauthaman K, Chan WK, Bongso A (2011) Human Wharton's jelly stem cells have unique transcriptome profiles compared to human embryonic stem cells and other mesenchymal stem cells. Stem Cell Rev 7(1):1–16. doi:10.1007/s12015-010-9166-x PubMedCrossRef Fong CY, Chak LL, Biswas A, Tan JH, Gauthaman K, Chan WK, Bongso A (2011) Human Wharton's jelly stem cells have unique transcriptome profiles compared to human embryonic stem cells and other mesenchymal stem cells. Stem Cell Rev 7(1):1–16. doi:10.​1007/​s12015-010-9166-x PubMedCrossRef
69.
Zurück zum Zitat Subramanian A, Fong CY, Biswas A, Bongso A (2015) Comparative characterization of cells from the various compartments of the human umbilical cord shows that the Wharton's jelly compartment provides the best source of clinically utilizable mesenchymal stem cells. PLoS ONE 10(6):e0127992. doi:10.1371/journal.pone.0127992 PubMedCentralPubMedCrossRef Subramanian A, Fong CY, Biswas A, Bongso A (2015) Comparative characterization of cells from the various compartments of the human umbilical cord shows that the Wharton's jelly compartment provides the best source of clinically utilizable mesenchymal stem cells. PLoS ONE 10(6):e0127992. doi:10.​1371/​journal.​pone.​0127992 PubMedCentralPubMedCrossRef
71.
Zurück zum Zitat Balasubramanian S, Thej C, Venugopal P, Priya N, Zakaria Z, Sundarraj S, Majumdar AS (2013) Higher propensity of Wharton's jelly derived mesenchymal stromal cells towards neuronal lineage in comparison to those derived from adipose and bone marrow. Cell Biol Int 37(5):507–515. doi:10.1002/cbin.10056 PubMedCrossRef Balasubramanian S, Thej C, Venugopal P, Priya N, Zakaria Z, Sundarraj S, Majumdar AS (2013) Higher propensity of Wharton's jelly derived mesenchymal stromal cells towards neuronal lineage in comparison to those derived from adipose and bone marrow. Cell Biol Int 37(5):507–515. doi:10.​1002/​cbin.​10056 PubMedCrossRef
73.
74.
Zurück zum Zitat Teixeira FG, Carvalho MM, Neves-Carvalho A, Panchalingam KM, Behie LA, Pinto L, Sousa N, Salgado AJ (2015) Secretome of mesenchymal progenitors from the umbilical cord acts as modulator of neural/glial proliferation and differentiation. Stem Cell Rev 11(2):288–297. doi:10.1007/s12015-014-9576-2 PubMedCrossRef Teixeira FG, Carvalho MM, Neves-Carvalho A, Panchalingam KM, Behie LA, Pinto L, Sousa N, Salgado AJ (2015) Secretome of mesenchymal progenitors from the umbilical cord acts as modulator of neural/glial proliferation and differentiation. Stem Cell Rev 11(2):288–297. doi:10.​1007/​s12015-014-9576-2 PubMedCrossRef
75.
Zurück zum Zitat Constantin G, Marconi S, Rossi B, Angiari S, Calderan L, Anghileri E, Gini B, Bach SD, Martinello M, Bifari F, Galie M, Turano E, Budui S, Sbarbati A, Krampera M, Bonetti B (2009) Adipose-derived mesenchymal stem cells ameliorate chronic experimental autoimmune encephalomyelitis. Stem Cells 27(10):2624–2635. doi:10.1002/stem.194 PubMedCrossRef Constantin G, Marconi S, Rossi B, Angiari S, Calderan L, Anghileri E, Gini B, Bach SD, Martinello M, Bifari F, Galie M, Turano E, Budui S, Sbarbati A, Krampera M, Bonetti B (2009) Adipose-derived mesenchymal stem cells ameliorate chronic experimental autoimmune encephalomyelitis. Stem Cells 27(10):2624–2635. doi:10.​1002/​stem.​194 PubMedCrossRef
76.
Zurück zum Zitat Bai L, Lennon DP, Caplan AI, DeChant A, Hecker J, Kranso J, Zaremba A, Miller RH (2012) Hepatocyte growth factor mediates mesenchymal stem cell-induced recovery in multiple sclerosis models. Nat Neurosci 15(6):862–870. doi:10.1038/nn.3109 PubMedCentralPubMedCrossRef Bai L, Lennon DP, Caplan AI, DeChant A, Hecker J, Kranso J, Zaremba A, Miller RH (2012) Hepatocyte growth factor mediates mesenchymal stem cell-induced recovery in multiple sclerosis models. Nat Neurosci 15(6):862–870. doi:10.​1038/​nn.​3109 PubMedCentralPubMedCrossRef
77.
Zurück zum Zitat Voulgari-Kokota A, Fairless R, Karamita M, Kyrargyri V, Tseveleki V, Evangelidou M, Delorme B, Charbord P, Diem R, Probert L (2012) Mesenchymal stem cells protect CNS neurons against glutamate excitotoxicity by inhibiting glutamate receptor expression and function. Exp Neurol 236(1):161–170. doi:10.1016/j.expneurol.2012.04.011 PubMedCrossRef Voulgari-Kokota A, Fairless R, Karamita M, Kyrargyri V, Tseveleki V, Evangelidou M, Delorme B, Charbord P, Diem R, Probert L (2012) Mesenchymal stem cells protect CNS neurons against glutamate excitotoxicity by inhibiting glutamate receptor expression and function. Exp Neurol 236(1):161–170. doi:10.​1016/​j.​expneurol.​2012.​04.​011 PubMedCrossRef
79.
Zurück zum Zitat Meng F, Meliton A, Moldobaeva N, Mutlu G, Kawasaki Y, Akiyama T, Birukova AA (2015) Asef mediates HGF protective effects against LPS-induced lung injury and endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 308(5):L452–L463. doi:10.1152/ajplung.00170.2014 PubMedCrossRef Meng F, Meliton A, Moldobaeva N, Mutlu G, Kawasaki Y, Akiyama T, Birukova AA (2015) Asef mediates HGF protective effects against LPS-induced lung injury and endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 308(5):L452–L463. doi:10.​1152/​ajplung.​00170.​2014 PubMedCrossRef
80.
Zurück zum Zitat Bruno S, Deregibus MC, Camussi G (2015) The secretome of mesenchymal stromal cells: role of extracellular vesicles in immunomodulation. Immunol Lett. doi:10.1016/j.imlet.2015.06.007 Bruno S, Deregibus MC, Camussi G (2015) The secretome of mesenchymal stromal cells: role of extracellular vesicles in immunomodulation. Immunol Lett. doi:10.1016/j.imlet.2015.06.007
81.
Zurück zum Zitat Gerdoni E, Gallo B, Casazza S, Musio S, Bonanni I, Pedemonte E, Mantegazza R, Frassoni F, Mancardi G, Pedotti R, Uccelli A (2007) Mesenchymal stem cells effectively modulate pathogenic immune response in experimental autoimmune encephalomyelitis. Ann Neurol 61(3):219–227. doi:10.1002/ana.21076 PubMedCrossRef Gerdoni E, Gallo B, Casazza S, Musio S, Bonanni I, Pedemonte E, Mantegazza R, Frassoni F, Mancardi G, Pedotti R, Uccelli A (2007) Mesenchymal stem cells effectively modulate pathogenic immune response in experimental autoimmune encephalomyelitis. Ann Neurol 61(3):219–227. doi:10.​1002/​ana.​21076 PubMedCrossRef
82.
Zurück zum Zitat Jellema RK, Wolfs TG, Lima Passos V, Zwanenburg A, Ophelders DR, Kuypers E, Hopman AH, Dudink J, Steinbusch HW, Andriessen P, Germeraad WT, Vanderlocht J, Kramer BW (2013) Mesenchymal stem cells induce T-cell tolerance and protect the preterm brain after global hypoxia-ischemia. PLoS ONE 8(8):e73031. doi:10.1371/journal.pone.0073031 PubMedCentralPubMedCrossRef Jellema RK, Wolfs TG, Lima Passos V, Zwanenburg A, Ophelders DR, Kuypers E, Hopman AH, Dudink J, Steinbusch HW, Andriessen P, Germeraad WT, Vanderlocht J, Kramer BW (2013) Mesenchymal stem cells induce T-cell tolerance and protect the preterm brain after global hypoxia-ischemia. PLoS ONE 8(8):e73031. doi:10.​1371/​journal.​pone.​0073031 PubMedCentralPubMedCrossRef
84.
Zurück zum Zitat Maron-Gutierrez T, Silva JD, Asensi KD, Bakker-Abreu I, Shan Y, Diaz BL, Goldenberg RC, Mei SH, Stewart DJ, Morales MM, Rocco PR, Dos Santos CC (2013) Effects of mesenchymal stem cell therapy on the time course of pulmonary remodeling depend on the etiology of lung injury in mice. Crit Care Med 41(11):e319–e333. doi:10.1097/CCM.0b013e31828a663e PubMedCrossRef Maron-Gutierrez T, Silva JD, Asensi KD, Bakker-Abreu I, Shan Y, Diaz BL, Goldenberg RC, Mei SH, Stewart DJ, Morales MM, Rocco PR, Dos Santos CC (2013) Effects of mesenchymal stem cell therapy on the time course of pulmonary remodeling depend on the etiology of lung injury in mice. Crit Care Med 41(11):e319–e333. doi:10.​1097/​CCM.​0b013e31828a663e​ PubMedCrossRef
85.
Zurück zum Zitat Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM (2002) Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 99(10):3838–3843PubMedCrossRef Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM (2002) Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 99(10):3838–3843PubMedCrossRef
87.
Zurück zum Zitat Raffaghello L, Bianchi G, Bertolotto M, Montecucco F, Busca A, Dallegri F, Ottonello L, Pistoia V (2008) Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells 26(1):151–162. doi:10.1634/stemcells.2007-0416 PubMedCrossRef Raffaghello L, Bianchi G, Bertolotto M, Montecucco F, Busca A, Dallegri F, Ottonello L, Pistoia V (2008) Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells 26(1):151–162. doi:10.​1634/​stemcells.​2007-0416 PubMedCrossRef
88.
Zurück zum Zitat Ivanova-Todorova E, Bochev I, Mourdjeva M, Dimitrov R, Bukarev D, Kyurkchiev S, Tivchev P, Altunkova I, Kyurkchiev DS (2009) Adipose tissue-derived mesenchymal stem cells are more potent suppressors of dendritic cells differentiation compared to bone marrow-derived mesenchymal stem cells. Immunol Lett 126(1-2):37–42. doi:10.1016/j.imlet.2009.07.010 PubMedCrossRef Ivanova-Todorova E, Bochev I, Mourdjeva M, Dimitrov R, Bukarev D, Kyurkchiev S, Tivchev P, Altunkova I, Kyurkchiev DS (2009) Adipose tissue-derived mesenchymal stem cells are more potent suppressors of dendritic cells differentiation compared to bone marrow-derived mesenchymal stem cells. Immunol Lett 126(1-2):37–42. doi:10.​1016/​j.​imlet.​2009.​07.​010 PubMedCrossRef
89.
Zurück zum Zitat Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L (2008) Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 111(3):1327–1333. doi:10.1182/blood-2007-02-074997 PubMedCrossRef Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L (2008) Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 111(3):1327–1333. doi:10.​1182/​blood-2007-02-074997 PubMedCrossRef
90.
92.
Zurück zum Zitat Chang YS, Ahn SY, Jeon HB, Sung DK, Kim ES, Sung SI, Yoo HS, Choi SJ, Oh WI, Park WS (2014) Critical role of vascular endothelial growth factor secreted by mesenchymal stem cells in hyperoxic lung injury. Am J Respir Cell Mol Biol 51(3):391–399. doi:10.1165/rcmb.2013-0385OC PubMedCrossRef Chang YS, Ahn SY, Jeon HB, Sung DK, Kim ES, Sung SI, Yoo HS, Choi SJ, Oh WI, Park WS (2014) Critical role of vascular endothelial growth factor secreted by mesenchymal stem cells in hyperoxic lung injury. Am J Respir Cell Mol Biol 51(3):391–399. doi:10.​1165/​rcmb.​2013-0385OC PubMedCrossRef
93.
Zurück zum Zitat Guan XJ, Song L, Han FF, Cui ZL, Chen X, Guo XJ, Xu WG (2013) Mesenchymal stem cells protect cigarette smoke-damaged lung and pulmonary function partly via VEGF-VEGF receptors. J Cell Biochem 114(2):323–335. doi:10.1002/jcb.24377 PubMedCrossRef Guan XJ, Song L, Han FF, Cui ZL, Chen X, Guo XJ, Xu WG (2013) Mesenchymal stem cells protect cigarette smoke-damaged lung and pulmonary function partly via VEGF-VEGF receptors. J Cell Biochem 114(2):323–335. doi:10.​1002/​jcb.​24377 PubMedCrossRef
95.
Zurück zum Zitat Nartprayut K, U-Pratya Y, Kheolamai P, Manochantr S, Chayosumrit M, Issaragrisil S, Supokawej A (2013) Cardiomyocyte differentiation of perinatally derived mesenchymal stem cells. Mol Med Rep 7(5):1465–1469. doi:10.3892/mmr.2013.1356 PubMed Nartprayut K, U-Pratya Y, Kheolamai P, Manochantr S, Chayosumrit M, Issaragrisil S, Supokawej A (2013) Cardiomyocyte differentiation of perinatally derived mesenchymal stem cells. Mol Med Rep 7(5):1465–1469. doi:10.​3892/​mmr.​2013.​1356 PubMed
97.
Zurück zum Zitat Wu KH, Zhou B, Lu SH, Feng B, Yang SG, Du WT, Gu DS, Han ZC, Liu YL (2007) In vitro and in vivo differentiation of human umbilical cord derived stem cells into endothelial cells. J Cell Biochem 100(3):608–616. doi:10.1002/jcb.21078 PubMedCrossRef Wu KH, Zhou B, Lu SH, Feng B, Yang SG, Du WT, Gu DS, Han ZC, Liu YL (2007) In vitro and in vivo differentiation of human umbilical cord derived stem cells into endothelial cells. J Cell Biochem 100(3):608–616. doi:10.​1002/​jcb.​21078 PubMedCrossRef
98.
Zurück zum Zitat Leite C, Silva NT, Mendes S, Ribeiro A, de Faria JP, Lourenco T, dos Santos F, Andrade PZ, Cardoso CM, Vieira M, Paiva A, da Silva CL, Cabral JM, Relvas JB, Graos M (2014) Differentiation of human umbilical cord matrix mesenchymal stem cells into neural-like progenitor cells and maturation into an oligodendroglial-like lineage. PLoS ONE 9(10):e111059. doi:10.1371/journal.pone.0111059 PubMedCentralPubMedCrossRef Leite C, Silva NT, Mendes S, Ribeiro A, de Faria JP, Lourenco T, dos Santos F, Andrade PZ, Cardoso CM, Vieira M, Paiva A, da Silva CL, Cabral JM, Relvas JB, Graos M (2014) Differentiation of human umbilical cord matrix mesenchymal stem cells into neural-like progenitor cells and maturation into an oligodendroglial-like lineage. PLoS ONE 9(10):e111059. doi:10.​1371/​journal.​pone.​0111059 PubMedCentralPubMedCrossRef
99.
Zurück zum Zitat Yan ZJ, Hu YQ, Zhang HT, Zhang P, Xiao ZY, Sun XL, Cai YQ, Hu CC, Xu RX (2013) Comparison of the neural differentiation potential of human mesenchymal stem cells from amniotic fluid and adult bone marrow. Cell Mol Neurobiol 33(4):465–475. doi:10.1007/s10571-013-9922-y PubMedCrossRef Yan ZJ, Hu YQ, Zhang HT, Zhang P, Xiao ZY, Sun XL, Cai YQ, Hu CC, Xu RX (2013) Comparison of the neural differentiation potential of human mesenchymal stem cells from amniotic fluid and adult bone marrow. Cell Mol Neurobiol 33(4):465–475. doi:10.​1007/​s10571-013-9922-y PubMedCrossRef
101.
Zurück zum Zitat Huang K, Kang X, Wang X, Wu S, Xiao J, Li Z, Wu X, Zhang W (2015) Conversion of bone marrow mesenchymal stem cells into type II alveolar epithelial cells reduces pulmonary fibrosis by decreasing oxidative stress in rats. Mol Med Rep 11(3):1685–1692. doi:10.3892/mmr.2014.2981 PubMedCentralPubMed Huang K, Kang X, Wang X, Wu S, Xiao J, Li Z, Wu X, Zhang W (2015) Conversion of bone marrow mesenchymal stem cells into type II alveolar epithelial cells reduces pulmonary fibrosis by decreasing oxidative stress in rats. Mol Med Rep 11(3):1685–1692. doi:10.​3892/​mmr.​2014.​2981 PubMedCentralPubMed
102.
105.
106.
107.
Zurück zum Zitat Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, Semprun-Prieto L, Delafontaine P, Prockop DJ (2009) Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell 5(1):54–63. doi:10.1016/j.stem.2009.05.003 PubMedCentralPubMedCrossRef Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, Semprun-Prieto L, Delafontaine P, Prockop DJ (2009) Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell 5(1):54–63. doi:10.​1016/​j.​stem.​2009.​05.​003 PubMedCentralPubMedCrossRef
109.
Zurück zum Zitat Xin H, Li Y, Buller B, Katakowski M, Zhang Y, Wang X, Shang X, Zhang ZG, Chopp M (2012) Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells 30(7):1556–1564. doi:10.1002/stem.1129 PubMedCentralPubMedCrossRef Xin H, Li Y, Buller B, Katakowski M, Zhang Y, Wang X, Shang X, Zhang ZG, Chopp M (2012) Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells 30(7):1556–1564. doi:10.​1002/​stem.​1129 PubMedCentralPubMedCrossRef
111.
Zurück zum Zitat Katsuda T, Tsuchiya R, Kosaka N, Yoshioka Y, Takagaki K, Oki K, Takeshita F, Sakai Y, Kuroda M, Ochiya T (2013) Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci Rep 3:1197. doi:10.1038/srep01197 PubMedCentralPubMedCrossRef Katsuda T, Tsuchiya R, Kosaka N, Yoshioka Y, Takagaki K, Oki K, Takeshita F, Sakai Y, Kuroda M, Ochiya T (2013) Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci Rep 3:1197. doi:10.​1038/​srep01197 PubMedCentralPubMedCrossRef
112.
Zurück zum Zitat Ribeiro CA, Fraga JS, Graos M, Neves NM, Reis RL, Gimble JM, Sousa N, Salgado AJ (2012) The secretome of stem cells isolated from the adipose tissue and Wharton jelly acts differently on central nervous system derived cell populations. Stem Cell Res Ther 3(3):18. doi:10.1186/scrt109 PubMedCentralPubMedCrossRef Ribeiro CA, Fraga JS, Graos M, Neves NM, Reis RL, Gimble JM, Sousa N, Salgado AJ (2012) The secretome of stem cells isolated from the adipose tissue and Wharton jelly acts differently on central nervous system derived cell populations. Stem Cell Res Ther 3(3):18. doi:10.​1186/​scrt109 PubMedCentralPubMedCrossRef
113.
Zurück zum Zitat Ribeiro CA, Salgado AJ, Fraga JS, Silva NA, Reis RL, Sousa N (2011) The secretome of bone marrow mesenchymal stem cells-conditioned media varies with time and drives a distinct effect on mature neurons and glial cells (primary cultures). J Tissue Eng Regen Med 5(8):668–672. doi:10.1002/term.365 PubMedCrossRef Ribeiro CA, Salgado AJ, Fraga JS, Silva NA, Reis RL, Sousa N (2011) The secretome of bone marrow mesenchymal stem cells-conditioned media varies with time and drives a distinct effect on mature neurons and glial cells (primary cultures). J Tissue Eng Regen Med 5(8):668–672. doi:10.​1002/​term.​365 PubMedCrossRef
117.
Zurück zum Zitat Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY, collaborators S (2010) A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells 28(6):1099–1106. doi:10.1002/stem.430 PubMedCrossRef Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY, collaborators S (2010) A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells 28(6):1099–1106. doi:10.​1002/​stem.​430 PubMedCrossRef
118.
Zurück zum Zitat Diez-Tejedor E, Gutierrez-Fernandez M, Martinez-Sanchez P, Rodriguez-Frutos B, Ruiz-Ares G, Lara ML, Gimeno BF (2014) Reparative therapy for acute ischemic stroke with allogeneic mesenchymal stem cells from adipose tissue: a safety assessment: a phase II randomized, double-blind, placebo-controlled, single-center, pilot clinical trial. J Stroke Cerebrovasc Dis 23(10):2694–2700. doi:10.1016/j.jstrokecerebrovasdis.2014.06.011 PubMedCrossRef Diez-Tejedor E, Gutierrez-Fernandez M, Martinez-Sanchez P, Rodriguez-Frutos B, Ruiz-Ares G, Lara ML, Gimeno BF (2014) Reparative therapy for acute ischemic stroke with allogeneic mesenchymal stem cells from adipose tissue: a safety assessment: a phase II randomized, double-blind, placebo-controlled, single-center, pilot clinical trial. J Stroke Cerebrovasc Dis 23(10):2694–2700. doi:10.​1016/​j.​jstrokecerebrova​sdis.​2014.​06.​011 PubMedCrossRef
119.
Zurück zum Zitat Mazzini L, Ferrero I, Luparello V, Rustichelli D, Gunetti M, Mareschi K, Testa L, Stecco A, Tarletti R, Miglioretti M, Fava E, Nasuelli N, Cisari C, Massara M, Vercelli R, Oggioni GD, Carriero A, Cantello R, Monaco F, Fagioli F (2010) Mesenchymal stem cell transplantation in amyotrophic lateral sclerosis: a phase I clinical trial. Exp Neurol 223(1):229–237. doi:10.1016/j.expneurol.2009.08.007 PubMedCrossRef Mazzini L, Ferrero I, Luparello V, Rustichelli D, Gunetti M, Mareschi K, Testa L, Stecco A, Tarletti R, Miglioretti M, Fava E, Nasuelli N, Cisari C, Massara M, Vercelli R, Oggioni GD, Carriero A, Cantello R, Monaco F, Fagioli F (2010) Mesenchymal stem cell transplantation in amyotrophic lateral sclerosis: a phase I clinical trial. Exp Neurol 223(1):229–237. doi:10.​1016/​j.​expneurol.​2009.​08.​007 PubMedCrossRef
120.
Zurück zum Zitat Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, Du MQ, Luan SL, Altmann DR, Thompson AJ, Compston A, Scott MA, Miller DH, Chandran S (2012) Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study. Lancet Neurol 11(2):150–156. doi:10.1016/S1474-4422(11)70305-2 PubMedCentralPubMedCrossRef Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, Du MQ, Luan SL, Altmann DR, Thompson AJ, Compston A, Scott MA, Miller DH, Chandran S (2012) Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study. Lancet Neurol 11(2):150–156. doi:10.​1016/​S1474-4422(11)70305-2 PubMedCentralPubMedCrossRef
121.
Zurück zum Zitat Llufriu S, Sepulveda M, Blanco Y, Marin P, Moreno B, Berenguer J, Gabilondo I, Martinez-Heras E, Sola-Valls N, Arnaiz JA, Andreu EJ, Fernandez B, Bullich S, Sanchez-Dalmau B, Graus F, Villoslada P, Saiz A (2014) Randomized placebo-controlled phase II trial of autologous mesenchymal stem cells in multiple sclerosis. PLoS ONE 9(12):e113936. doi:10.1371/journal.pone.0113936 PubMedCentralPubMedCrossRef Llufriu S, Sepulveda M, Blanco Y, Marin P, Moreno B, Berenguer J, Gabilondo I, Martinez-Heras E, Sola-Valls N, Arnaiz JA, Andreu EJ, Fernandez B, Bullich S, Sanchez-Dalmau B, Graus F, Villoslada P, Saiz A (2014) Randomized placebo-controlled phase II trial of autologous mesenchymal stem cells in multiple sclerosis. PLoS ONE 9(12):e113936. doi:10.​1371/​journal.​pone.​0113936 PubMedCentralPubMedCrossRef
122.
Zurück zum Zitat Pal R, Venkataramana NK, Bansal A, Balaraju S, Jan M, Chandra R, Dixit A, Rauthan A, Murgod U, Totey S (2009) Ex vivo-expanded autologous bone marrow-derived mesenchymal stromal cells in human spinal cord injury/paraplegia: a pilot clinical study. Cytotherapy 11(7):897–911. doi:10.3109/14653240903253857 PubMedCrossRef Pal R, Venkataramana NK, Bansal A, Balaraju S, Jan M, Chandra R, Dixit A, Rauthan A, Murgod U, Totey S (2009) Ex vivo-expanded autologous bone marrow-derived mesenchymal stromal cells in human spinal cord injury/paraplegia: a pilot clinical study. Cytotherapy 11(7):897–911. doi:10.​3109/​1465324090325385​7 PubMedCrossRef
123.
Zurück zum Zitat Zheng G, Huang L, Tong H, Shu Q, Hu Y, Ge M, Deng K, Zhang L, Zou B, Cheng B, Xu J (2014) Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study. Respir Res 15:39. doi:10.1186/1465-9921-15-39 PubMedCentralPubMedCrossRef Zheng G, Huang L, Tong H, Shu Q, Hu Y, Ge M, Deng K, Zhang L, Zou B, Cheng B, Xu J (2014) Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study. Respir Res 15:39. doi:10.​1186/​1465-9921-15-39 PubMedCentralPubMedCrossRef
125.
Zurück zum Zitat Ribeiro-Paes JT, Bilaqui A, Greco OT, Ruiz MA, Marcelino MY, Stessuk T, de Faria CA, Lago MR (2011) Unicentric study of cell therapy in chronic obstructive pulmonary disease/pulmonary emphysema. Int J Chron Obstruct Pulmon Dis 6:63–71. doi:10.2147/COPD.S15292 PubMedCentralPubMedCrossRef Ribeiro-Paes JT, Bilaqui A, Greco OT, Ruiz MA, Marcelino MY, Stessuk T, de Faria CA, Lago MR (2011) Unicentric study of cell therapy in chronic obstructive pulmonary disease/pulmonary emphysema. Int J Chron Obstruct Pulmon Dis 6:63–71. doi:10.​2147/​COPD.​S15292 PubMedCentralPubMedCrossRef
126.
Zurück zum Zitat Lee YH, Choi KV, Moon JH, Jun HJ, Kang HR, Oh SI, Kim HS, Um JS, Kim MJ, Choi YY, Lee YJ, Kim HJ, Lee JH, Son SM, Choi SJ, Oh W, Yang YS (2012) Safety and feasibility of countering neurological impairment by intravenous administration of autologous cord blood in cerebral palsy. J Transl Med 10:58. doi:10.1186/1479-5876-10-58 PubMedCentralPubMedCrossRef Lee YH, Choi KV, Moon JH, Jun HJ, Kang HR, Oh SI, Kim HS, Um JS, Kim MJ, Choi YY, Lee YJ, Kim HJ, Lee JH, Son SM, Choi SJ, Oh W, Yang YS (2012) Safety and feasibility of countering neurological impairment by intravenous administration of autologous cord blood in cerebral palsy. J Transl Med 10:58. doi:10.​1186/​1479-5876-10-58 PubMedCentralPubMedCrossRef
127.
Zurück zum Zitat Mancias-Guerra C, Marroquin-Escamilla AR, Gonzalez-Llano O, Villarreal-Martinez L, Jaime-Perez JC, Garcia-Rodriguez F, Valdes-Burnes SL, Rodriguez-Romo LN, Barrera-Morales DC, Sanchez-Hernandez JJ, Cantu-Rodriguez OG, Gutierrez-Aguirre CH, Gomez-De Leon A, Elizondo-Riojas G, Salazar-Riojas R, Gomez-Almaguer D (2014) Safety and tolerability of intrathecal delivery of autologous bone marrow nucleated cells in children with cerebral palsy: an open-label phase I trial. Cytotherapy 16(6):810–820. doi:10.1016/j.jcyt.2014.01.008 PubMedCrossRef Mancias-Guerra C, Marroquin-Escamilla AR, Gonzalez-Llano O, Villarreal-Martinez L, Jaime-Perez JC, Garcia-Rodriguez F, Valdes-Burnes SL, Rodriguez-Romo LN, Barrera-Morales DC, Sanchez-Hernandez JJ, Cantu-Rodriguez OG, Gutierrez-Aguirre CH, Gomez-De Leon A, Elizondo-Riojas G, Salazar-Riojas R, Gomez-Almaguer D (2014) Safety and tolerability of intrathecal delivery of autologous bone marrow nucleated cells in children with cerebral palsy: an open-label phase I trial. Cytotherapy 16(6):810–820. doi:10.​1016/​j.​jcyt.​2014.​01.​008 PubMedCrossRef
128.
Zurück zum Zitat Wang X, Cheng H, Hua R, Yang J, Dai G, Zhang Z, Wang R, Qin C, An Y (2013) Effects of bone marrow mesenchymal stromal cells on gross motor function measure scores of children with cerebral palsy: a preliminary clinical study. Cytotherapy 15(12):1549–1562. doi:10.1016/j.jcyt.2013.06.001 PubMedCrossRef Wang X, Cheng H, Hua R, Yang J, Dai G, Zhang Z, Wang R, Qin C, An Y (2013) Effects of bone marrow mesenchymal stromal cells on gross motor function measure scores of children with cerebral palsy: a preliminary clinical study. Cytotherapy 15(12):1549–1562. doi:10.​1016/​j.​jcyt.​2013.​06.​001 PubMedCrossRef
130.
131.
Zurück zum Zitat Min K, Song J, Kang JY, Ko J, Ryu JS, Kang MS, Jang SJ, Kim SH, Oh D, Kim MK, Kim SS, Kim M (2013) Umbilical cord blood therapy potentiated with erythropoietin for children with cerebral palsy: a double-blind, randomized, placebo-controlled trial. Stem Cells 31(3):581–591. doi:10.1002/stem.1304 PubMedCentralPubMedCrossRef Min K, Song J, Kang JY, Ko J, Ryu JS, Kang MS, Jang SJ, Kim SH, Oh D, Kim MK, Kim SS, Kim M (2013) Umbilical cord blood therapy potentiated with erythropoietin for children with cerebral palsy: a double-blind, randomized, placebo-controlled trial. Stem Cells 31(3):581–591. doi:10.​1002/​stem.​1304 PubMedCentralPubMedCrossRef
132.
Zurück zum Zitat Cotten CM, Murtha AP, Goldberg RN, Grotegut CA, Smith PB, Goldstein RF, Fisher KA, Gustafson KE, Waters-Pick B, Swamy GK, Rattray B, Tan S, Kurtzberg J (2014) Feasibility of autologous cord blood cells for infants with hypoxic-ischemic encephalopathy. J Pediatr 164(5):973–979. doi:10.1016/j.jpeds.2013.11.036, e971PubMedCentralPubMedCrossRef Cotten CM, Murtha AP, Goldberg RN, Grotegut CA, Smith PB, Goldstein RF, Fisher KA, Gustafson KE, Waters-Pick B, Swamy GK, Rattray B, Tan S, Kurtzberg J (2014) Feasibility of autologous cord blood cells for infants with hypoxic-ischemic encephalopathy. J Pediatr 164(5):973–979. doi:10.​1016/​j.​jpeds.​2013.​11.​036, e971PubMedCentralPubMedCrossRef
133.
Zurück zum Zitat Ploemacher WJCRaRE (2003) Primary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia 17(1):160–170CrossRef Ploemacher WJCRaRE (2003) Primary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia 17(1):160–170CrossRef
135.
Zurück zum Zitat Donega V, Nijboer CH, van Velthoven CT, Youssef SA, de Bruin A, van Bel F, Kavelaars A, Heijnen CJ (2015) Assessment of long-term safety and efficacy of intranasal mesenchymal stem cell treatment for neonatal brain injury in the mouse. Pediatric research. doi:10.1038/pr.2015.145 Donega V, Nijboer CH, van Velthoven CT, Youssef SA, de Bruin A, van Bel F, Kavelaars A, Heijnen CJ (2015) Assessment of long-term safety and efficacy of intranasal mesenchymal stem cell treatment for neonatal brain injury in the mouse. Pediatric research. doi:10.1038/pr.2015.145
136.
Zurück zum Zitat Liu L, Mao Q, Chu S, Mounayar M, Abdi R, Fodor W, Padbury JF, De Paepe ME (2014) Intranasal versus intraperitoneal delivery of human umbilical cord tissue-derived cultured mesenchymal stromal cells in a murine model of neonatal lung injury. Am J Pathol 184(12):3344–3358. doi:10.1016/j.ajpath.2014.08.010 PubMedCrossRef Liu L, Mao Q, Chu S, Mounayar M, Abdi R, Fodor W, Padbury JF, De Paepe ME (2014) Intranasal versus intraperitoneal delivery of human umbilical cord tissue-derived cultured mesenchymal stromal cells in a murine model of neonatal lung injury. Am J Pathol 184(12):3344–3358. doi:10.​1016/​j.​ajpath.​2014.​08.​010 PubMedCrossRef
Metadaten
Titel
Mesenchymal stem/stromal cells—a key mediator for regeneration after perinatal morbidity?
Publikationsdatum
01.12.2016
Erschienen in
Molecular and Cellular Pediatrics / Ausgabe 1/2016
Elektronische ISSN: 2194-7791
DOI
https://doi.org/10.1186/s40348-016-0034-x

Weitere Artikel der Ausgabe 1/2016

Molecular and Cellular Pediatrics 1/2016 Zur Ausgabe

Häufigste Gründe für Brustschmerzen bei Kindern

06.05.2024 Pädiatrische Diagnostik Nachrichten

Akute Brustschmerzen sind ein Alarmsymptom par exellence, schließlich sind manche Auslöser lebensbedrohlich. Auch Kinder klagen oft über Schmerzen in der Brust. Ein Studienteam ist den Ursachen nachgegangen.

Endlich: Zi zeigt, mit welchen PVS Praxen zufrieden sind

IT für Ärzte Nachrichten

Darauf haben viele Praxen gewartet: Das Zi hat eine Liste von Praxisverwaltungssystemen veröffentlicht, die von Nutzern positiv bewertet werden. Eine gute Grundlage für wechselwillige Ärzte und Psychotherapeuten.

Durch übermäßige Internetnutzung wird oft die Schule verpasst

Häufige Fehlzeiten in der Schule können durch physische und psychische Probleme verursacht werden. Wie in einer Studie aus Finnland nun belegt wird, führt auch die exzessive Nutzung des Internets gehäuft zu Abwesenheiten.

Kinder mit anhaltender Sinusitis profitieren häufig von Antibiotika

30.04.2024 Rhinitis und Sinusitis Nachrichten

Persistieren Sinusitisbeschwerden bei Kindern länger als zehn Tage, ist eine Antibiotikatherapie häufig gut wirksam: Ein Therapieversagen ist damit zu über 40% seltener zu beobachten als unter Placebo.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.