FIG 3 - uploaded by Nuno M Neves
Content may be subject to copyright.
Hippocampal neurons incubated with human umbilical cord perivascular cells (HUCPVCs) conditioned media (CM). Similarly to what was observed for glial cells, CM induced an increase of metabolic viability (all values statistically signifi cant different from the control, one way ANOVA, n = 3, mean ± for SD, P < 0.05) was observed. MAP-2 cell counts normalized to the content of total protein/well revealed that there were drastic differences to the control (one way ANOVA, n = 3, mean ± SD, P < 0.05). Therefore it can be stated that HUCPVCs CM possess neuroregulatory molecules that increase cell viability, proliferation and survival/differentiation on primary cultures of hippocampal neurons.  

Hippocampal neurons incubated with human umbilical cord perivascular cells (HUCPVCs) conditioned media (CM). Similarly to what was observed for glial cells, CM induced an increase of metabolic viability (all values statistically signifi cant different from the control, one way ANOVA, n = 3, mean ± for SD, P < 0.05) was observed. MAP-2 cell counts normalized to the content of total protein/well revealed that there were drastic differences to the control (one way ANOVA, n = 3, mean ± SD, P < 0.05). Therefore it can be stated that HUCPVCs CM possess neuroregulatory molecules that increase cell viability, proliferation and survival/differentiation on primary cultures of hippocampal neurons.  

Source publication
Article
Full-text available
It has been recently reported that mesenchymal progenitor/stem cells isolated from the Wharton's Jelly (WJ) of umbilical cords (UC) ameliorate the condition of animals suffering from central nervous system (CNS)-related conditions. However, little is known on the mechanisms that regulate these actions. Therefore, the objective of the present work w...

Contexts in source publication

Context 1
... fact, the graph shown in Figure 1E indicates that cultures incubated with HUCPVCs CM had lower cell densities of microglial cells, namely those cultured with CM 24 h. Figures 3 and 4 show the results for the experiments per- formed with primary hippocampal cultures. Cell viability assays revealed that in the absence of supplements the control cultures disclosed lower levels of cell viability, up to 3-fold smaller (P < 0.05) when compared to the CM-incubated cultures (Fig. 3A). ...
Context 2
... cells, namely those cultured with CM 24 h. Figures 3 and 4 show the results for the experiments per- formed with primary hippocampal cultures. Cell viability assays revealed that in the absence of supplements the control cultures disclosed lower levels of cell viability, up to 3-fold smaller (P < 0.05) when compared to the CM-incubated cultures (Fig. 3A). Simultaneously, it was also possible to observe that CM 24 h (Fig. 3B) drastically increased cell pro- liferation in this culture system (P < 0.05), while CM 48 h and 72 h did not induce any changes in these parameters. MAP-2 immunostaining showed that no differences were observed between the different CM groups for neuronal ...
Context 3
... for the experiments per- formed with primary hippocampal cultures. Cell viability assays revealed that in the absence of supplements the control cultures disclosed lower levels of cell viability, up to 3-fold smaller (P < 0.05) when compared to the CM-incubated cultures (Fig. 3A). Simultaneously, it was also possible to observe that CM 24 h (Fig. 3B) drastically increased cell pro- liferation in this culture system (P < 0.05), while CM 48 h and 72 h did not induce any changes in these parameters. MAP-2 immunostaining showed that no differences were observed between the different CM groups for neuronal densities, but drastic differences were observed when compared to the con- trol ...
Context 4
... < 0.05), while CM 48 h and 72 h did not induce any changes in these parameters. MAP-2 immunostaining showed that no differences were observed between the different CM groups for neuronal densities, but drastic differences were observed when compared to the con- trol (P < 0.05), as the latter had almost undetected numbers of MAP-2-positive cells (Figs. 3C and ...
Context 5
... the experiments with hippocampal neurons similar results were obtained for cell viability and proliferation. As it can be concluded from Figures 3 and 4, the CM collected from HUCPVCs for all time points increased the overall cell metabolic viability when compared to control cultures. Moreover it was also possible to observe that CM 24 h had notorious effects on the up-regulation of cell proliferation (P < 0.05). ...

Similar publications

Article
Full-text available
Simple Summary We review here what is currently known about the role of mesenchymal stromal-like cells, which complicate our understanding of the glioma microenvironment. We provide an overview of the major studies on these cells, highlighting their role in tumor progression and prognosis. Researchers and clinicians should consider these cells to b...

Citations

... Zhang et al. have also found that Dil dye can be used for retrograde labeling of brainstem neurons in vivo [27]. In addition, Salgado et al. [28] found that the secretome from Warton's Jelly-MSCs and HUCPVSs can enhance the survival, proliferation, and differentiation of hippocampal neurons by containing FGF and NGF [29]. Furthermore, axonal development is mediated by the secretomes formed from BDNF-induced MSCs [30]. ...
Article
Full-text available
Introduction Spinal cord injury (SCI) leads to significant destruction of nerve tissue, causing the degeneration of axons and the formation of cystic cavities. This study aimed to examine the characteristics of human umbilical cord-derived mesenchymal stem cells (HUCMSCs) cultured in a serum-free conditioned medium (CM) and assess their effectiveness in a well-established hemitransection SCI model. Materials and methods In this study, HUCMSCs cultured medium was collected and characterized by measuring IL-10 and identifying proteomics using mass spectroscopy. This collected serum-free CM was further used in the experiments to culture and characterize the HUMSCs. Later, neuronal cells derived from CM-enriched HUCMSC were tested sequentially using an injectable caffeic acid-bioconjugated gelatin (CBG), which was further transplanted in a hemitransection SCI model. In vitro, characterization of CM-enriched HUCMSCs and differentiated neuronal cells was performed using flow cytometry, immunofluorescence, electron microscopy, and post-transplant analysis using immunohistology analysis, qPCR, in vivo bioluminescence imaging, and behavioral analysis using an infrared actimeter. Results The cells that were cultured in the conditioned media produced a pro-inflammatory cytokine called IL-10. Upon examining the secretome of the conditioned media, the Kruppel-like family of KRAB and zinc-finger proteins (C2H2 and C4) were found to be activated. Transcriptome analysis also revealed an increased expression of ELK-1, HOXD8, OTX2, YY1, STAT1, ETV7, and PATZ1 in the conditioned media. Furthermore, the expression of Human Stem-101 confirmed proliferation during the first 3 weeks after transplantation, along with the migration of CBG-UCNSC cells within the transplanted area. The gene analysis showed increased expression of Nestin, NeuN, Calb-2, Msi1, and Msi2. The group that received CBG-UCNSC therapy showed a smooth recovery by the end of week 2, with most rats regaining their walking abilities similar to those before the spinal cord injury by week 5. Conclusions In conclusion, the CBG-UCNSC method effectively preserved the integrity of the transplanted neuronal-like cells and improved locomotor function. Thus, CM-enriched cells can potentially reduce biosafety risks associated with animal content, making them a promising option for clinical applications in treating spinal cord injuries. Graphical abstract
... Numerous reports have shown that CM contains antioxidants, growth factors, cytokines, and chemokines which can promote cell migration, angiogenesis, accelerate wound healing, and prevent cell aging (Sriramulu et al., 2018;Arutyunyan et al., 2016;Zhang et al., 2017). Studies have revealed the presence of transforming growth factor-β (TGF-β), insulin-like growth factor1 (IGF-1), vascular endothelial growth factor (VEGF), and brain-derived neurotrophic factor (BDNF) in the umbilical cord stem cell culture media (Salgado et al., 2010). These factors are responsible for the neurogenic functions and neuronal support to improve neuronal functions and reverse the synaptic plasticity in neurons of the hippocampus (Ruiz de Almodovar et al., 2009). ...
Article
Full-text available
Introduction Alzheimer’s disease (AD) is accompanied by progressive cognitive disorders and memory loss. This study aims to determine the combined effects of conditioned medium of human umbilical cord mesenchymal stem cells (CM) and platelet-rich plasma (PRP) on AD model rats. Methods Forty-eight male Sprague Dawley rats were classified into 6 groups: Control, Sham, AD, and three treatment groups. AD was induced by streptozotocin(STZ; 3 mg/kg, intracerebroventricular (ICV)) and the treatment groups received injections of CM [(200 µl, intraperitoneally (i.p.), and/or PRP (100 µl, intravenously(i.v)] for 8 days. Behavioral tests (Morris water maze and novel objective recognition) were used to assess learning ability and memory. At the end of the behavioral tests, the rats were sacrificed and their brain was entirely removed, sectioned, and stained with cresyl violet. The hippocampus volume and number of neurons were evaluated by stereological techniques. Results In the AD group, the discrimination ratio, time spent in the target zone, volume of Cornu Ammonis1 (CA1) and Dentate Gyrus (DG), and the number of pyramidal and granular cells decreased significantly compared to the Sham group. The mentioned parameters increased in the CM and CM+PRP groups compared to the AD group (p < 0.01). PRP did not have any noticeable effect on the examined parameters. Conclusions CM may be beneficial in the treatment of AD as it led to better improvement in STZ-induced learning and memory impairments as well as the structure of the hippocampus.
... Moreover, it has been shown in vitro and in vivo that human MSCs are capable of secreting neuroregulatory and neuroprotective factors, i.e., BDNF, ciliary neurotrophic factor (CNTF), glial cell line-derived neurotrophic factor (GDNF), or NGF thus increasing neuron viability; these are factors that increase neurogenesis or stimulate neurons proliferation and differentiation: bone morphogenetic proteins (BMP) family, granulocyte colony-stimulating factor (G-CSF), stem cell factor (SCF), TGF-β, and many others (e.g., IGF-1, HGF, vascular endothelial growth factor (VEGF), and stromal cell-derived factor-(SDF-) 1) involved in neurogenesis stimulation, apoptosis inhibition, glial scar formation, immunomodulation, and angiogenesis [66,67]. Moreover, human MSC-conditioned media promotes survival of rat neurons and glial cells as well as show neuroprotective properties [68,69]. There are also reports indicating that MSC exosomes promote axonal growth [70], inhibit neuronal cell apoptosis, suppress glial scar formation, and improve functional recovery after spinal cord injury in rats [71]. ...
Article
Full-text available
Facial nerve palsy is a serious neurological condition that strongly affects patient everyday life. Standard treatments provide insufficient improvement and are burdened with the risk of severe complications, e.g., facial synkinesis. Mesenchymal stromal cell-based therapies are a novel and extensively developed field which offers new treatment approaches with promising results in regards to the nervous tissue regeneration. The potential of mesenchymal stromal cells (MSCs) to aid the regeneration of damaged nerves has been demonstrated in several preclinical models, as well as in several clinical trials. However, therapies based on cell transplantation are difficult to standardize in the manner similar to that of routine clinical practices. On the other hand, treatments based on mesenchymal stromal cell secretome harness the proregenerative features of mesenchymal stromal cells but relay on a product with measurable parameters that can be put through standardization procedures and deliver a fully controllable end-product. Utilization of mesenchymal stromal cell secretome allows the controlled dosage and standardization of the components to maximize the therapeutic potential and ensure safety of the end-product.
... Abbreviations: Transforming growth factor beta (TGF-β), natural killer cells (NK), dendritic cells (DCs), prostaglandin E2 (PGE2), peripheral blood mononuclear cells (PBMCs), vascular endothelial growth factor (VEGF), T helper 1 cells (Th1), T helper 17 cells (Th17), Indoleamine-2,3-dioxygenase (IDO), monocyte chemoattractant protein-1 (CCL2/MCP-1), interleukin-6 (IL-6), tumor necrosis factor-inducible gene 6 protein (TSG6), hepatocyte growth factor-1 (HGF-1), nerve growth factor (NGF), glial cellderived neurotrophic factor (GDNF), galectin-1 (Gal-1), galectin-9 (Gal-9) (Kim et al. 2009;Rafei et al. 2009;Gieseke et al. 2010 factor (NGF), basic fibroblast growth factor (bFGF), cil iary neurotrophic factor (CNTF), erythropoietin (EPO), neurotrophin (NT) 3, and NT4/5 have been shown to have neuroprotection and neuroregeneration effects on the central nervous system (CNS) (Salgado et al. 2010). Whereas neurotrophic factors can be classified accord ing to their receptors into three groups: neurotrophins (BDNF, NGF, NT3, and NT4/5), neurokines (CNTF and leukemia inhibitory factor (LIF)); and the transforming growth factor β family (TGFβ1, TGFβ2, TGFβ3; and glial cellderived neurotrophic factor (GDNF)) (Fornaro et al. 2020). ...
Article
Full-text available
Low back pain is a crucial public health problem that is commonly associated with intervertebral disc de‐ generation and has vast socio‐economic impact worldwide. Current treatments for disc degeneration are conservative, non‐surgical, or surgical interventions, and there is no current clinical therapy aimed at directly reversing the degeneration. Given the limited capacity of intervertebral disc (IVD) cells to self‐repair, treatment aiming to regenerate IVDs is a topic of interest and mesenchymal stem cells (MSCs) have been identified as having potential in this regeneration. Recent studies have revealed that the benefits of MSC therapy could result from the molecules the cells secrete and that play principal roles in regulating essential biologic processes, rather than from the implanted cells themselves. Therefore, the objective of this study is to review the potential use of the MSC secretome to regenerate IVDs. Current evidence shows that the secretome may regenerate IVDs by modulating the gene expressions of nucleus pulposus cells (upregulation of keratin 19 and downregulation of matrix metalloproteinase 12 and matrix Gla protein) and stimulating IVD progenitor cells to repair the degenerated disc.
... The broad spectrum of molecules secreted by MSC is known as conditioned medium or secretome. In humans, the MSC secretome consists of hundreds of biologically active molecules, including anti-inflammatory cytokines, trophic factors, and antioxidant molecules [70,71]. The paracrine potential of MSC can be boosted by in vitro preconditioning of these cells with environmental or pharmacological stimuli, enhancing their therapeutic efficacy. ...
Article
Full-text available
Perinatal Asphyxia (PA) is a leading cause of motor and neuropsychiatric disability associated with sustained oxidative stress, neuroinflammation, and cell death, affecting brain development. Based on a rat model of global PA, we investigated the neuroprotective effect of intranasally administered secretome, derived from human adipose mesenchymal stem cells (MSC-S), preconditioned with either deferoxamine (an hypoxia-mimetic) or TNF-α+IFN-γ (pro-inflammatory cytokines). PA was generated by immersing fetus-containing uterine horns in a water bath at 37 °C for 21 min. Thereafter, 16 μL of MSC-S (containing 6 μg of protein derived from 2 × 105 preconditioned-MSC), or vehicle, were intranasally administered 2 h after birth to asphyxia-exposed and control rats, evaluated at postnatal day (P) 7. Alternatively, pups received a dose of either preconditioned MSC-S or vehicle, both at 2 h and P7, and were evaluated at P14, P30, and P60. The preconditioned MSC-S treatment (i) reversed asphyxia-induced oxidative stress in the hippocampus (oxidized/reduced glutathione); (ii) increased antioxidative Nuclear Erythroid 2-Related Factor 2 (NRF2) translocation; (iii) increased NQO1 antioxidant protein; (iv) reduced neuroinflammation (decreasing nuclearNF-κB/p65 levels and microglial reactivity); (v) decreased cleaved-caspase-3 cell-death; (vi) improved righting reflex, negative geotaxis, cliff aversion, locomotor activity, anxiety, motor coordination, and recognition memory. Overall, the study demonstrates that intranasal administration of preconditioned MSC-S is a novel therapeutic strategy that prevents the long-term effects of perinatal asphyxia.
... Recent evidence suggests that their capability to exert a neuroprotective effect following CNS injury may be through paracrine effects. 6,[13][14][15][16][17] In ischemic rat models, intracerebral transplantation of HUCPVCs modulates the expression of granulocyte colony-stimulating factor, vascular endothelial growth factor, glial-derived neurotrophic factor, and stem cell-derived factor 1, as well as brain-derived neurotrophic factor (BNDF). 18,19 Furthermore, HUCPVC-derived conditioned media (CM) enhances survival of neuronal and glial cells in vitro. ...
Article
Full-text available
The pathophysiological events of secondary brain injury contribute to poor outcome after traumatic brain injury (TBI). The neuroprotective effects of mesenchymal cells have been extensively studied and evidence suggests that their effects are mostly mediated through paracrine effects. Human umbilical cord perivascular cells (HUCPVCs) are mesenchymal stem cells with potential therapeutic value in TBI. In this study, we assessed the effect of HUCPVC-conditioned media (CM) in an established adult zebrafish model of TBI induced by pulsed high-intensity focused ultrasound (pHIFU). This model demonstrates similarities to mammalian outcome after TBI. Administration of HUCPVC-CM 1 h postinjury (hpi) resulted in improved outcome after pHIFU-induced TBI. Western blot and immunohistochemistry results demonstrated that the HUCPVC-CM reduced (p < 0.05) reactive astrogliosis at 24 hpi. Moreover, at 24 hpi, the HUCPVC-CM treatment resulted in reduced apoptosis in HUCPVC-CM-treated zebrafish. Behavioral analysis demonstrated improvement in locomotor activity (p < 0.05) and anxiety (p < 0.05) at 6 and 24 hpi following HUCPVC-CM treatment. Overall, HUCPVC-CM treatment improved acute outcome measures in pHIFU-injured zebrafish. Collectively, the data demonstrate a cell-free treatment approach for traumatic brain injuries.
... It was discovered on experimental animal models that stem cells obtained from bone marrow (BM-MSCs) and adipose tissue (ASCs) improved the healing process after stroke (Wei et al. 2009;Honmou et al. 2012;Teixeira et al. 2014 Populations of WJ-MSCs and HUCPVCs are also identified as mesenchymal stem cells (Sarugaser et al. 2005;Weiss and Troyer 2006;Baksh et al. 2007;Sarugaser et al. 2009). The major effects of MSCs are supposed to be determined by their secretomes (Salgado et al. 2010;Carvalho et al. 2011;Ribeiro et al. 2012;Teixeira et al. 2013;Teixeira et al. 2014). Both neural stem cells (NSCs) and MSCs secrete a variety of growth factors (Salgado et al. 2015). ...
... To restore the central nervous system (CNS), the stem/progenitor cells from different sources could be used. For example, stem cells present in the Warton jelly of the umbilical cord, known as Wharton jelly stem cells (WJ-MSCs) and human umbilical cord perivascular cells (HUCPVCs), have a great potential in healing CNS injuries(Salgado et al. 2010; Datta et al. 2011;Taghizadeh et al. 2011). ...
Article
Full-text available
Mesenchymal stem (stromal) cells (MSCs) are self-renewing, cultured adult stem cells which secrete a complex set of multiple soluble biologically active molecules such as chemokines, and cytokines, cell adhesion molecules, lipid mediators, interleukins (IL), growth factors (GFs), hormones, micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), messenger RNAs (mRNAs), exosomes, as well as microvesicles, the secretome. MSCs of various origin, including adipose-derived stem cells (ASCs), bone marrow derived mesenchymal stem cells (BM-MSCs), human uterine cervical stem cells (hUCESCs), may be good candidates for obtaining secretome-derived products. Different population of MSCs can secret different factors which could have anti-inflammatory, anti-apoptotic, anti-fibrotic activities, a neuroprotective effect, could improve bone, muscle, liver regeneration and wound healing. Therefore, the paracrine activity of conditioned medium obtained when cultivating MSCs, due to a plethora of bioactive factors, was assumed to have the most prominent cell-free therapeutic impact and can serve as a better option in the field of regenerative medicine in future.
... Moreover, Salgado and coworkers [82] pointed out that the secretome from WJ-MSCs and HUCPVCs is also able to increase survival, proliferation, and differentiation of hippocampal neurons due to the presence of, for example, NGF and FGF [82,83]. Additionally, the BDNF present on these cell secretomes is mediating axonal growth [84]. ...
... Moreover, Salgado and coworkers [82] pointed out that the secretome from WJ-MSCs and HUCPVCs is also able to increase survival, proliferation, and differentiation of hippocampal neurons due to the presence of, for example, NGF and FGF [82,83]. Additionally, the BDNF present on these cell secretomes is mediating axonal growth [84]. ...
Article
Full-text available
Transplantation of stem cells, in particular mesenchymal stem cells (MSCs), stands as a promising therapy for trauma, stroke or neurodegenerative conditions such as spinal cord or traumatic brain injuries (SCI or TBI), ischemic stroke (IS), or Parkinson’s disease (PD). Over the last few years, cell transplantation-based approaches have started to focus on the use of cell byproducts, with a strong emphasis on cell secretome. Having this in mind, the present review discusses the current state of the art of secretome-based therapy applications in different central nervous system (CNS) pathologies. For this purpose, the following topics are discussed: (1) What are the main cell secretome sources, composition, and associated collection techniques; (2) Possible differences of the therapeutic potential of the protein and vesicular fraction of the secretome; and (3) Impact of the cell secretome on CNS-related problems such as SCI, TBI, IS, and PD. With this, we aim to clarify some of the main questions that currently exist in the field of secretome-based therapies and consequently gain new knowledge that may help in the clinical application of secretome in CNS disorders.
... In the last decade it has been extensively shown that the sole use of secretome of MSCs positively impacts neuronal and glial cell survival, proliferation and differentiation in vitro (Ribeiro et al., 2012;Salgado et al., 2010;Teixeira et al., 2015) as well as axonal growth (Assunção- Silva et al., 2018;Martins et al., 2017), disclosing simultaneously an immuno-modulatory character upon microglial cells (Kong et al., 2018;Ooi et al., 2015). Additionally, in vivo studies in pre-clinical animal models of spinal cord injury, amyotrophic lateral sclerosis and Parkinson's disease, revealed that the use of MSCs secretome led to functional and histological recovery (Lu et al., 2019;Nicaise et al., 2011;Teixeira et al., 2017). ...
... In fact, early data from our group have demonstrated that the secretome is an active modulator of hippocampal neurogenesis, inducing new-born neuron proliferation and differentiation. Furthermore, the secretome is also capable of increasing neuronal and glial survival in vitro (Salgado et al., 2010;Ribeiro et al., 2012), as well as improving astrocytic densities when applied in vivo into the DG . ...
... By using conditioned media from hUC-MSC Salgado et al. [43] showed that these cells can potentiate astrocyte and oligodendrocyte cell densities, without stimulating the proliferation of microglial cells, and suggested that hUC-MSC release factors to the extracellular milieu that have different direct impact on the densities of each type of glial cells, possibly due to the existence of variable sensitivity. Different therapeutic methods have been studied to minimize the damage in central nervous system injuries. ...
Article
Full-text available
Spinal cord injury (SCI) is a common pathological condition that leads to permanent or temporal loss of motor and autonomic functions. Kainic acid (KA), an agonist of kainate receptors, a type of ionotropic glutamate receptor, is widely used to induce experimental neurodegeneration models of CNS. Mesenchymal Stem Cells (MSC) therapy applied at the injured nervous tissue have emerged as a promising therapeutic treatment. Here we used a validated SCI experimental model in which an intraparenchymal injection of KA into the C5 segment of rat spinal cord induced an excitotoxic lesion. Three days later, experimental animals were treated with an intracerebroventricular injection of human umbilical cord (hUC) MSC whereas control group only received saline solution. Sensory and motor skills as well as neuronal and glial reaction of both groups were recorded. Differences in motor behavior, neuronal counting and glial responses were observed between hUC-MSC-treated and untreated rats. According to the obtained results, we suggest that hUC-MSC therapy delivered into the fourth ventricle using the intracerebroventricular via can exert a neuroprotective or neurorestorative effect on KA-injected animals.