Научная статья на тему 'BIOLOGICAL ACTIVITY OF MESENCHYMAL STEM CELLS SECRETOME AS A BASIS FOR CELL-FREE THERAPEUTIC APPROACH'

BIOLOGICAL ACTIVITY OF MESENCHYMAL STEM CELLS SECRETOME AS A BASIS FOR CELL-FREE THERAPEUTIC APPROACH Текст научной статьи по специальности «Биотехнологии в медицине»

CC BY
477
81
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
MESENCHYMAL STEM CELLS / PRECONDITIONING / SECRETOME / REGENERATION / IMMUNOMODULATION / THERAPEUTIC POTENTIAL

Аннотация научной статьи по биотехнологиям в медицине, автор научной работы — Pokrovskaya L.A., Zubareva E.V., Nadezhdin S.V., Lysenko A.S., Litovkina T.L.

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.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «BIOLOGICAL ACTIVITY OF MESENCHYMAL STEM CELLS SECRETOME AS A BASIS FOR CELL-FREE THERAPEUTIC APPROACH»

# Research Results in Pharmacology

Research Results in Pharmacology 6(1): 57-68 UDC: 576.5, 57.04, 615.324, 604.4 DOI 10.3897/rrpharmacology. 6.49413

3 Review Article

Biological activity of mesenchymal stem cells secretome as a basis for cell-free therapeutic approach

Liubov A. Pokrovskaya1, Ekaterina V. Zubareva2, Sergey V. Nadezhdin2,3, Anna S. Lysenko2, Tatyana L. Litovkina2

1 National Research Tomsk State University, 36 Lenin Ave., Tomsk 634050, Russia

2 Belgorod State National Research University, 85 Pobedy St., Belgorod 308015, Russia

3 Research Laboratory of Cellular, Assisted Reproductive and DNA Technologies, Belgorod State National Research University, 85 Pobedy St., Belgorod 308015, Russia

Corresponding author: Ekaterina V. Zubareva (zubareva@bsu.edu.ru)

Academic editor: Mikhail Korokin ♦ Received 14 December 2019 ♦ Accepted 20 January 2020 ♦ Published 30 March 2020

Citation: Pokrovskaya LA, Zubareva EV, Nadezhdin SV, Lysenko AS, Litovkina TL (2020) Biological activity of mesenchymal stem cells secretome as a basis for cell-free therapeutic approach. Research Results in Pharmacology 6(1): 57-68. https://doi. org/10.3897/rrpharmacology.6.49413

Abstract

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.

Keywords

mesenchymal stem cells; pre-conditioning; secretome; regeneration; immunomodulation; therapeutic potential.

Introduction

Mesenchymal stem (stromal) cells (MSCs) are self-renewing, culture expandable adult stem cells that have been shown to be a promising candidate for cell-based therapy (Ferreira et al. 2018). Using mesenchymal stem cells for regenerative purposes is possible due to their trophic, paracrine, and immunomodulatory properties (Stagg and Galipeau 2013; Del Papa et al. 2015; Mafia et al.

2015; Leavitt et al. 2016; Del Papa et al. 2019; Kuchar-zewski et al. 2019; L et al. 2019; Lombardi et al. 2019). Besides, MSCs also have anti-tumorigenic, anti-fibrotic, anti-apoptotic, anti-inflammatory, pro-angiogenic, neuroprotective, anti-bacterial and chemo-attractive effects (Maumus et al. 2013; Bartosh et al. 2016; L et al. 2019).

MSCs harvested from numerous anatomical locations, including the bone marrow, adipose tissue, Wharton's jelly of the umbilical cord, display similar immunopheno-

Copyright Pokrovskaya LA et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC-BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

typic profiles. However, there is a large body of evidence showing that, despite the similarity in their immunophe-notypes, MSCs secrete a complex set of multiple soluble biologically active molecules, the secretome, composition of which varies significantly, depending on the age of the host and niches where the cells reside (Baksh et al. 2004; Traktuev et al. 2008; Amos et al. 2010; Daquinag et al. 2011; Cheng et al. 2012; Daquinag et al. 2013; Ka-pur and Katz 2013; Kyurkchiev et al. 2014; Madrigal et al. 2014; Dubey et al. 2018; Ferreira et al. 2018; L et al. 2019; Lombardi et al. 2019; Meiliana et al. 2019). The MSCs secretome in general consists of such biologically active factors as chemokines and cytokines, cell adhesion molecules, lipid mediators, interleukins (ILs), growth factors (GFs), hormones, micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), messenger RNAs (mR-NAs), exosomes, as well as microvesicles (Kyurkchiev et al. 2014; Madrigal et al. 2014; Dubey et al. 2018; Ferreira et al. 2018; Lombardi et al. 2019; Meiliana et al. 2019). It is revealed that MSC secretion include in particular vascular endothelial growth factor (VEGF), insulin-like growth factor 1 (IGF-1), basic fibroblast growth factor (bFGF), transforming growth factor beta 1 (TGF-b1), nerve growth factor (NGF), placental growth factor (PGF), stromal-derived growth factor (SDF-1/CXCL12), monocyte chemo-attractant protein-1 (MCP-1/CCL2), IL-6, IL-8, IL-10 and IL-13 (Meiliana et al. 2019), bone morphogenetic proteins (BMP), CC chemokine ligand 5/ Regulated on activation, normal T cell expressed and secreted (CCL5/RANTES), epidermal growth factor (EGF), granulocyte colony-stimulating factor (G-CSF), granulo-cyte-macrophage colony-stimulating factor (GM-CSF), hepatocyte growth factor (HGF), inter-cellular adhesion molecules (ICAM), indoleamine-2,3-dioxygenase (IDO), leukemia inhibitory factor (LIF), matrix metalloproteas-es (MMP-1, MMP-2, MMP-3, MMP-7), platelet-derived growth factor (PDGF), metalloproteinase inhibitors (TIMP-1, TIMP-2) (Polacek et al. 2011; Osugi et al. 2012; Inukai et al. 2013; Kyurkchiev et al. 2014; Pereira et al. 2014; Ferreira et al. 2018; Linero and Chaparro 2014).

Herewith, mesenchymal stem cells of various origin, including bone-marrow-derived mesenchymal stem cells (BM-MSCs), adipose tissue-derived stem cells (ADSCs) or human uterine cervical stromal stem cells (hUCESCs), may be good candidates for obtaining products from secretome (Zhao et al. 2013; Vizoso et al. 2017). However different populations of MSCs should be used for specific purposes because the composition of the secretome depends on the stromal cells source. For instance, ADSCs have higher expression of mRNA, VEGF-D, IGF-1 and IL-8, while dermal-sheath- and dermal-papilla-derived cells secrete higher concentrations of CCL2 and leptin (Hsiao et al. 2012). It is known that placenta-derived MSCs are characterized with increased expression levels of HGF, bFGF, IL-6, IL-8, IL-1a and IL-1b, while in secretome obtained from bone marrow-derived MSCs the levels of VEGF-A, NGF and angiogenin are higher (Du et al. 2016; Meiliana et al. 2019).

There is evidence that the molecules produced by MSCs (secretomes), especially those packaged in extracellular vesicles (EVs), influence the tissue repair even better than the cells themselves (Lepperdinger et al. 2008; Madrigal et al. 2014; Dubey et al. 2018; Ferreira and Gomes 2018; Pelizzo et al. 2018; Campanella et al. 2019; Lombardi et al. 2019; Mitchell et al. 2019).

Therefore due to the paracrine activity, MSCs conditioned medium (CM) or purified MSC-derived extracellular vesicles having a plethora of bioactive factors are assumed to have the most prominent cell-free therapeutic impact and can serve as a better option in the field of regenerative medicine in the future (Maguire 2013; Zhou et al. 2013; Vizoso et al. 2017; Park et al. 2018; L et al. 2019; Lombardi et al. 2019).

This new frontier of research provides several key advantages over cell based applications: (a) the administration of proteins instead of whole cells as a new therapeutic option in regenerative medicine; (b) CM can be stored without any toxic cryopreservatives, such as dimethyl sulfoxide (DMSO), for a relatively long period; (c) preparation of CM is more economical as it can be mass-produced from the available MSC populations under current good manufacturing practice (cGMP) conditions; (d) evaluation of CM for safety and efficacy will be much simpler and analogous to conventional pharmaceutical agents (Bermudez et al. 2015; L et al. 2019).

Anti-inflammatory activity

It is well known that there are anti-inflammatory factors in the MSCs secretome, including tumor necrosis factor ß1 (TGFß1) (Zagoura et al. 2012), interleukin 13 (IL13) (Bermudez et al. 2016), interleukin 18 binding protein (IL18BP), ciliary neurotrophic factor (CNTF), neurot-rophin 3 (NT-3) factor, interleukin 10 (IL10), interleukin 12 p70 (IL12p70), interleukin-25 (IL-25), which is also known as interleukin-17E (IL17E), interleukin 27 (IL27), or interleukin 1 receptor antagonist (IL1RA). On the other hand, pro-inflammatory cytokines are also present in MSCs conditioned medium, for example, IL1b, interleukin 6 (IL6) (See et al. 2011; Cantinieaux et al. 2013), interleukin 8 (IL8) (Mirabella et al. 2011), and interleukin 9 (IL9) (Lee et al. 2011). Thus, the balance between the anti-and pro-inflammatory factors will determine the final effect of conditioned medium on the inflammatory process.

However, the numerous studies highlight MSCs anti-inflammatory effect. Yi and Song (2012) described that MSCs inhibited proinflammatory cytokines, such as interferon (IFN)-y and tumour necrosis factor a (TNFa), while increasing release of anti-inflammatory IL10. Legaki et al. (2016) showed that hUCESC-CM treatment significantly reduced mRNA expression of pro-inflammatory cytokines (IL6, IL8, TNFa and macrophage inflammatory protein-1 alpha (MIP-1a)), but increased mRNA expression of the anti-inflammatory cytokine (IL10). The similar results were obtained during experiments with MSC-CM from

amniotic fluid in a mice colitis model (Legaki et al. 2016). It was also found that hUCESC-CM reduced the infiltration of leucocytes in ocular tissues (Vishnubhatla et al. 2014).

Moreover, anti-inflammatory cytokines in MSC-CM can contribute to the beneficial effects seen in animal models of diabetes, acute colitis, inflammatory arthritis, etc. (Brini et al. 2017; Kay et al. 2017; Pouya et al. 2018). It was revealed that single intravenous injection of a conditioned medium derived from adipose tissue (hAT-CM) into streptozotocin-(STZ)-treated diabetic mice relieved the diabetic neuropathic pain by re-establishing the Th1/Th2 balance with a long-lasting relief of sensory hypersensitivity. In the experiments with the STZ-treated diabetic mice, it was demonstrated that the content of anti-inflammatory and immunomodulatory cytokines (IL-1p, IL-6 and TNF-a) in dorsal root ganglia, sciatic nerves and spinal cord restored to basal levels after 1 week of hAT-CM injection. The elevated level of IL-10 also confirmed realization of an anti-inflammatory mechanism (Brini et al. 2017). Pouya et al (2018) showed that an intraperitoneal injection of MSC-CM in C57BL/6 mice with colitis mediated a significant decrease in colon inflammation and an increase in colon weight and length, which led to the disease activity index and mortality rate reducing. Furthermore, the mesenteric lymph nodes and spleen of the mice infused with MSC-CM demonstrated increased levels of the anti-inflammatory cytokines IL-10 and TGF-p and reduced levels of the pro-inflammatory cytokine IL-17 confirming the antiinflammatory role of CM. Similarly, in the antigen-induced model of inflammatory arthritis, it was shown that an intra-articular injection of murine MSC-CM was effective in reducing disease severity and cartilage damage. The high levels of IL10 in CM were revealed, which correlated with an anti-inflammatory response (Kay et al. 2017; L et al. 2019).

In order to study whether the anti-inflammatory potential of ADSC secretome is higher than EV-enriched fraction of ADSCs secretome, the effect of both fractions was investigated on the TNF-a-induced nuclear translocation of the NF-kB subunit p65 in U251 cells. It is interesting to note that the level of nuclear NF-kB p65 was significantly increased by TNF-a treatment compared to control cells. The effect of the total secretome fraction on TNF-a stimulated cells was accompanied by a non-significant reduction of nuclear p65, whereas the influence of EV fraction led to a significantly reduced amount of p65 (Mitchell et al. 2019).

Anti-apoptotic activity

There are studies which illustrate that MSCs produce inhibitor proteins of apoptosis to restore local environment and prevent therefore cell death (Li et al. 2015). Tang et al. (2005) reported that MSCs decreased the pro-apoptotic factors expression (Bax and cleaved caspase-3) and stimulated at the same time the anti-apoptotic compounds levels (Bcl-2). It is noteworthy that MSCs treatment of hearts led to elevated level of pro-angiogenic factors expression, including basic fibroblast growth factor (bFGF), vascular

endothelial growth factor (VEGF), and stromal cell-derived factor-1 (SDF-1), which is also called chemokine (C-X-C motif) ligand 12 (CXCL12) (Tang et al. 2005).

There are also studies revealing a pro-apoptotic effect of hUCESC-CM on malignant cells. In accordance with these data, the effects of MSCs on normal and cancer cells are different. Along with an antiapoptotic effect of hUCESC-CM on normal cells (Bermudez et al. 2016), the apoptosis occurred in cancer cells under the influence of conditioned medium obtained from human uterine cervical stem cells in vitro and in vivo (Eiro et al. 2014; Vizoso et al. 2017).

Anti-fibrotic activity

An anti-fibrotic effect of stem cells conditioned medium is mediated by bioactive molecules in MSCs secretome which decrease accumulation of extracellular proteins and, therefore, lead to reduced scar formation. An et al. (2017) studied the influence of umbilical cord-derived mesenchymal cells (UCMSC) secretome on formation of fibrotic areas in mice with hepatic fibrosis. A decrease in the number of activated hepatic stellate cells (HSCs) expressing a- smooth muscle actin (a-SMA) was shown after an injection of the UCMSC-CM in the diseased mice, which was accompanied by reducing fibrotic areas. The researchers analysed the UCMSCs secretome using na-no-chip-LC/QTOF-MS and discovered the presence of milk fat globule EGF factor 8 (MFGE8), an anti-fibrotic protein known to down-regulate the expression of TGF-ßR1 (transforming growth factor ß type 1 receptor) at the mRNA and protein level, thereby decreasing the activation of human hepatic stellate cells (An et al. 2017).

Paracrine effect of MSCs in bone regeneration

It was revealed that secretome synthesized by different stem cells, including rat bone marrow-derived MSCs (rBM-MSCs), human adult mesenchymal stem cells (haMSCs) and human fetal mesenchymal stem cells (hfMSCs), promoted osteogenic differentiation of rBM-MSCs. It is quite important that the human MSC conditioned medium effects in the same way as rat MSCs secretome, or even better. Xu et al. (2016) described that the hfMSCs secretome is characterised as a conditioned medium with the strongest osteogenic induction ability compared to the conditioned medium obtained when cultivating rBM-MSC or haMSC. It was also revealed that hfMSC conditioned medium does not induce any significant immune response, which makes it different from the haMSC secretome. At the same time, conditioned media at different concentrations did not affect rBM-MSC viability or cell proliferation. Furthermore, hfMSCs secretome at the concentration of 100 ^g/^l could enhance mineralization during rBM-MSC osteogenic induction

via increasing of alkaline phosphatase (ALP) activity and formation of calcium nodules.

The expression levels of osteogenic marker genes, including runt-related transcription factor 2 (Runx2), osteocalcin (OCN), osteopontin (OPN), and osterix (Osx), were significantly upregulated on Days 3 and 10 after the hfMSCs secretome treatment. ALP is responsible for osteoblastic differentiation at an early stage; it hydrolyses pyrophosphyte and generates inorganic phosphate which promotes the process of mineralization. Runx2 produces bone matrix proteins and is essential for osteoblast differentiation. Growth and differentiation factors regulate expression levels of OPN and mediate bone formation and its remodeling. Osx acts downstream of Runx2, this factor takes part in the processes of osteoblast differentiation and bone tissue formation. It was shown that conditioned medium obtained from hfMSC stimulated differentiation of rBM-MSCs in osteogenic direction in vitro. Furthermore, application of the hfMSCs secretome locally into distraction osteogenesis gap in rats led to accelerated new bone formation and consolidation (Xu et al. 2016).

It is known that during the distraction osteogenesis procedure proliferation of bone progenitor cells increases and their recruitment to the target site occurs. The healing process is accompanied by the angiogenesis and bone tissue formation/mineralization. At the same time, the mechanisms of the hfMSCs conditioned media effect on bone tissue regeneration, including vascular network formation and remodelling, remain unknown. There is evidence that the MSCs secretome mediates the release of vascular endothelial growth factor (VEGF) at the site of tissue repair, being stimulated by hypoxia or normoxia. It is known that the expression of VEGF enhances bone tissue and blood vessels formation during distraction osteogenesis, and this biological active factor is required for osteogenic differentiation.

Moreover, osteogenic lineage commitment of MSCs is accompanied by the Osx and OCN osteoprogenitors markers expression. Treatment of the damaged area with the hfMSCs secretome upregulates the number of Osx-and OCN-positive osteoprogenitors in the distraction zone in comparison with the control group. Different signaling pathways may be involved in VEGF production following hfMSC conditioned medium influence, and further experiments are required to make the molecular mechanisms of these processes clear (Xu et al. 2016).

Neuroprotective effect and impact on neural/glial proliferation

There have been several studies in which adult stem cells, including mesenchymal stem cells, were used as a possible tool for central nervous system (CNS) regeneration (Lindvall and Kokaia 2010; Shihabuddin and Aubert 2010; Kan et al. 2011; Teixeira et al. 2014), which could be a promising therapeutic option (Pittenger et al. 1999; Zuk et al. 2002; Wang et al. 2004; Teixeira et al. 2014).

It was shown that the MSCs secretome mediated neuroprotective and neurotrophic effects (Caseiro et al. 2016; Luarte et al. 2016; Ratajczak et al. 2016) due to a number of neurotrophic factors (de Almeida et al. 2014; Mead et al. 2014). There are studies which demonstrate on nerve injury models that the MSC-based approach generated healing effects, including enhanced vascularization of the regenerating site, increased thickness of the myelin sheaths, modulation of the Wallerian degeneration stage, accelerated fibre regeneration, reduction of fibrotic scaring, and improved fibre organization (Caseiro et al. 2016).

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), demyelination (Constantin et al. 2009; Cristo-fanilli et al. 2011; Teixeira et al. 2014), Parkinson's disease (Cova et al. 2010; Erba et al. 2010), and spinal cord injury (Arboleda et al. 2011; Park et al. 2012). 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). 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). It was shown that the molecular content of the MSCs secretome depended on the culture duration and tissue sources of MSCs, and it influenced significantly the changes of primary cultures of hippocampal neurons and glial cells viability (Kim et al. 2013).

During the in vitro experiments, the ability of the HUCPVCs conditioned media to modulate the survival and viability of both neuronal and glial cells populations was shown (Salgado et al. 2015). It was demonstrated that application of HUCPVCs-CM to human telencephalon neural precursor cells (htNPCs) in vitro led to an increase of neuronal cell differentiation, which was characterised by higher densities of immature (DCX+ cells) and mature neurons (MAP-2+ cells). Moreover, an injection of HUCPVCs and their secretome into the dentate gyrus (DG) was accompanied by increasing the endogenous proliferation (BrdU+ cells) in a week. It was revealed that application of HUCPVCs led to an increased number of newborn neurons (DCX+ cells). And an injection of CM or HUCPVCs into the DG tissue promoted an elevated level of fibroblast growth factor-2 (FGF-2) and, to a lesser extent, of nerve growth factor (NGF). Thus, either transplantation of HUCPVCs or the application of their conditioned media potentiated enhanced neuronal viability and

differentiation in vitro and in vivo (Teixeira et al. 2014; Salgado et al. 2015).

The effect of intranasal application of CM derived from stem cells of human exfoliated deciduous teeth (SHEDs) in an animal model of Alzheimer's disease was studied. And the cell-free treatment was accompanied by an improvement of cognitive function and induced neuroregenerative effects, for example, an attenuated pro-inflammatory response induced by amyloid plaques, and anti-inflammatory M2-like microglia (Mita et al. 2015; Vizoso et al. 2017).

It was revealed that application of a secretome obtained from hypoxic-preconditioned MSCs promoted reduce of neuronal cell loss and apoptosis and production of VEGF which stimulated recovery processes in the organisms of traumatic brain injury-induced rats (Chang et al. 2013).

Angiogenesis and revascularization

Angiogenesis is a process of new vasculature sprouts formation from pre-existing blood vessels. This process normally occurs during wound healing. Numerous studies illustrated a particular impact of the MSCs secretome at the different stages of angiogenesis (Burlacu et al. 2013).

The effect of MSCs on the process of angiogenesis is studied in different spectrum of diseases, including impaired vessel growth during atherosclerosis and wound healing. Several studies demonstrated that the MSCs application led to stimulation of blood vessels formation in animal models of myocardial infarction, neurogenic bladder, peripheral artery disease, stress urinary incontinence, and cerebral ischemia/stroke (Hsieh et al. 2013; Liu et al. 2013; Sharma et al. 2013).

The MSCs secretome contains numerous biologically active molecules which act as angiogenic stimulators and inhibitors (Kinnaird et al. 2004; Di Santo et al. 2009; Boomsma et al. 2012; Ho et al. 2012). An extensive pro-teomic analysis of the conditioned media of mesenchy-mal stem cells stimulated with inflammatory cytokines revealed the presence in a secretome of tissue inhibitor of metalloproteinase-1 (TIMP-1) which is responsible for the MSCs antiangiogenic impacts (Zanotti et al. 2016). Moreover, some studies showed the dependence of the pro- and anti-angiogenic factors secretion on chemokines and hypoxic conditions. In particular, to describe the effect of bioactive molecules on MSCs secretion ability, it is important to note that TGFa increases the level of growth factors in the secretome (i.e., VEGF, hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), IL6 and IL8). And a conditioned medium from MSCs treated with TFG-a induces blood vessel growth in an in vivo assay (De Luca et al. 2011; Vizoso et al. 2017). The obtained data demonstrate the complicated set of bioactive molecules in the MSCs secretome which can be balanced under the different interventions to promote angiogenesis.

The ASCs secretome also has a proangiogenic effect, which was demonstrated in acute myocardial infarction models. Rehman et al. (2004) showed that the conditioned

media obtained from ASCs stimulated angiogenesis in the ischemic environment. The following bioactive molecules were detected in the secretome after the analysis: G-CSF, TGF-p, VEGF, HGF, and bFGF (Rehman et al. 2004; Ka-pur and Katz 2013). The impact of HGF on the formation of blood vessels was proved through restricting the production of this factor by ASCs, which resulted in a decreasing effect by ASCs on endothelial cell proliferation, migration and survival in the ischemic environment (Cai et al. 2007).

Cutaneous wound healing

A cutaneous wound healing is a fascinating process, which requires cell migration, proliferation, matrix protein synthesis, and tissue remodelling. In particular, kera-tinocytes are involved in the epithelialization and dermal repair, and endothelial cells promote angiogenesis (Kober et al. 2016). The migration and proliferation of fibroblasts are the key processes in a wound healing mechanism. In the early stage of wound repair, they move to the damaged region and promote blood vessels regeneration and granulation tissue formation. In the advanced trauma stage, fibroblasts mature into myofibroblasts which are responsible for wound closure process (Zhao et al. 2013).

It was revealed that the adipose stem cell-conditioned medium (ASC-CM) had a marked stimulating effect on cutaneous wound healing, via affecting the mechanism for this response by influencing effector cells (Lombardi et al. 2019).

An increased proliferation and migration activity of primary human dermal fibroblasts (HDFs) as well as type I collagen secretion was shown after ASCs secretome application (Kim et al. 2007). Kober et al. (2016) revealed a stimulatory effect of ASC-CM on ASCs, and the proliferation of keratinocytes after application of ASCs secretome was significantly reduced. Furthermore, addition of ASC-CM did not affect cell migration, which had been tested with in vitro scratch assays. Collawn et al. (2016) demonstrated on a 3D skin model that application of ASC-CM as well as ASCs promoted an acceleration of wound repair. Seo et al. (2017) showed that ASC-CM, similarly to ASCs, stimulated the proliferation, migration, and invasion properties of HUVECs. An addition of ASC-CM led to increasing HaCaT cell proliferation and migration, as well as vascular endothelial growth factor (VEGF) secretion. Cooper et al. (2018) demonstrated that application of ASC-CM stimulated HDF migration ability in vitro. Park et al. (2018) collected CM samples from cultured ASCs isolated from adipose tissues of breast cancer patients and added the secretome to HDFs, normal adult human primary epidermal keratinocytes (HEKa), or HUVECs cultures. It resulted in increased cell proliferation, migration, and invasion. Kim et al. (2018) showed that ASC-CM from 3D-cultures influenced more significantly on the proliferation of Ha-CaT cells than the one obtained from 2D-cultivated ASCs. Noverina et al. (2019) analyzed the growth factor profile in ASC-CM via immunosorbent assay (ELISA) method and revealed a higher concentration level of fibroblast growth

factor (FGF), which is involved in wound healing and regeneration. Stojanovic and Najman (2019) demonstrated the immunomodulatory and wound healing potential of the secretome collected from stem cells previously isolated from adipose tissue, or lipoma (Lombardi et al. 2019).

Some studies described the presence of growth factors in the MSCs secretome that promoted tissue regeneration with a special focus on proliferation (Lee et al. 2011; Zagoura et al. 2012; Turner et al. 2013; Bhang et al. 2014; Bermudez et al. 2015). The experimental myocardial infarction models showed that anti-fibrotic and angiogenic effects of the MSCs secretome inhibited scar formation process (Cargonini et al. 2012; Preda et al. 2014) and stimulated the synthesis and consequent secretion of bi-oactive molecules responsible for remodelling (Williams et al. 2013; Vizoso et al. 2017).

It is known that such components of the ASCs secretome as bFGF significantly stimulate migration and proliferation of functional cells in wound healing; however PDGF-AA and VEGF influence only fibroblasts migration (Zhao et al. 2013). It was revealed that EGF could accelerate reepithelialisation process via stimulating the keratino-cytes proliferation and migration in an acute wound. Zhao et al. (2013) demonstrated an increased proliferation of fibroblasts under the EGF influence. However, the mechanism of the EGF effect on cells migration is still unclear.

Hu et al. (2013) showed that wound healing was mediated via application of the ASCs secretome which stimulated migration of vascular endothelial cells 4 hours later, fibro-blasts 12 hours later and then keratinocytes 24 hours later.

Park et al. (2018) reported that elevated levels of EGF, bFGF and HGF in conditioned media promoted wound healing.

And finally, it was revealed that increasing prolifera-tive and migratory characteristics of different dermis cellular components, including dermal fibroblasts, keratino-cytes, and endothelial cells, in vitro occurred by activating PI3K/Akt and FAK-ERK1/2 signaling (Park et al. 2017).

Muscle regeneration

Tissue regeneration and homeostasis are mediated by cell proliferation, migration and stem cell differentiation processes. Mitchell et al. (2019) showed that application of the total secretome mouse myoblast cell line C2C12 for 48 hours resulted in increased cell proliferation in comparison with the control group. Moreover, the total secretome stimulated the differentiation of C2C12 cells into myofibers (Mitchell et al. 2019).

Mitchell et al. (2019) studied the effect of the total ASCs secretome and its EV fraction on tissue repair in a mouse model of acute cardiotoxin-induced muscle injury. It was revealed that the total ASCs secretome stimulated the process of tissue regeneration, which was confirmed by a significant decrease in the activity of lysosomes in the group of animals treated with the total ASC-CM (Mitchell et al. 2019). Studying the cross-sectional area of new-

ly formed muscle fibres during the regeneration process revealed that the application of EV fraction had a greater effect than the total ASCs secretome (Mitchell et al. 2019).

Liver regeneration

Lee et al. (2014) demonstrated that liver regeneration in partially hepatectomised models occurred after systemic infusion of the ASCs secretome. It was shown that application of ASCs-CM increased mRNA expression of Lgr5 (a Wnt target gene), which was an indicator of actively dividing stem cells. Expression of Lgr5 occurred in small cells located near bile ducts as a result of a liver cell injury (Huch et al. 2013; Lee et al. 2014). During the repair phase, those cells were able to generate significant numbers of hepatocytes and biliary duct cells and, thus, could be considered a class of liver progenitor cells. Higher expression of Lgr5 in small cells located near bile ducts is an indicator of liver regeneration (Huch et al. 2013; Lee et al. 2014).

Lee et al. (2014) showed an increased expression of p-Akt, p-Erk1/2, which are the downstream effector components of HGF signaling pathways, and p-STAT3, which is responsible for cell cycle progression from G1 to S phase, after application of the ASC-secretome (Lee et al. 2014), which indicated that the ASC-secretome promoted liver regeneration.

Conclusion

The MSC-conditioned medium, or secretome, contains a plethora of cytokines and a wide array of bioactive factors, such as chemokines, cell adhesion molecules, lipid mediators, IL, growth factors, hormones, exosomes, mi-crovesicles, etc., which are secreted by MSCs (Lee et al. 2019). These factors have been considered as protagonists to participate in tissue repair and regeneration through their paracrine actions that mediate cell-to-cell signalling (Madrigal et al. 2014).

It is critical for the success of such a cell-free therapy to identify, analyze and elucidate the mechanism of action of each component of the secretome.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The research was carried out with the financial support of the Ministry of Science and Higher Education of the Russian Federation, Agreement № 14.575.21.0164, identifier RFMEFI57517X0164.

References

■ Amos PJ, Kapur SK, Stapor PC, Shang H, Bekiranov S, Khurgel M, Rodeheaver GT, Peirce SM, Katz AJ (2010) Human adipose-derived stromal cells accelerate diabetic wound healing: impact of cell formulation and delivery. Tissue Engineering Part A 16: 1595-1606. https://doi.org/10.1089/ten.tea.2009.0616 [PubMed] [PMC]

■ An SY, Jang YJ, Lim HJ, Han J, Lee J, Lee G, Park JY, Park SY, Kim JH, Do BR, Han C, Park HK, Kim OH, Song MJ, Kim SJ, Kim JH (2017) Milk fat Globule-EGF factor 8, secreted by mesenchymal stem cells, protects against liver fibrosis in mice. Gastroenterology 152: 1174-1186. https://doi.org/10.1053/j.gastro.2016.12.003 [PubMed]

■ Arboleda D, Forostyak S, Jendelova P, Marekova D, Amemori T, Pivonkova H, asinova K, Sykova E (2011) Transplantation of predif-ferentiated adipose-derived stromal cells for the treatment of spinal cord injury. Cellular and Molecular Neurobiology 31(7): 1113-1122. https://doi.org/10.1007/s10571-011-9712-3 [PubMed]

■ Baksh D, Song L, Tuan RS (2004) Adult mesenchymal stem cells: characterization, differentiation, and application in cell and gene therapy. Journal of Cellular and Molecular Medicine 8(3): 301-316. https:// doi.org/10.1111/j.1582-4934.2004.tb00320.x [PubMed] [PMC]

■ Baksh D, Yao R, Tuan RS (2007) Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells 25(6): 1384-1392. https://doi.org/10.1634/stemcells.2006-0709

■ Bartosh TJ, Ullah M, Zeitouni S, Beaver J, Prockop DJ (2016) Cancer cells enter dormancy after cannibalizing mesenchymal stem/stro-mal cells (MSCs). Proceedings of the National Academy of Sciences 113(42): E6447-E6456. https://doi.org/10.1073/pnas.1612290113 [PubMed] [PMC]

■ Bermudez MA, Sendon-Lago J, Eiro N, Trevino M, Gonzalez F, Ye-bra-Pimentel E, Giraldez MJ, Macia M, Lamelas ML, Saa J, Vizoso F, Perez-Fernandez R (2015) Corneal epithelial wound healing and bactericidal effect of conditioned medium from human uterine cervical stem cells. Investigative Ophthalmology & Visual Science 56(2): 983-992. https://doi.org/10.1167/iovs.14-15859 [PubMed]

■ Bermudez MA, Sendon-Lago J, Seoane S, Eiro N, Gonzalez F, Saa J, Vizoso F, Perez-Fernandez R (2016) Anti-inflammatory effect of conditioned medium from human uterine cervical stem cells in uveitis. Experimental Eye Research 149: 84-92. https://doi. org/10.1016/j.exer.2016.06.022 [PubMed]

■ Bhang SH, Lee S, Shin JY, Lee TJ, Jang HK, Kim BS (2014) Efficacious and clinically relevant conditioned medium of human adipose-derived stem cells for therapeutic angiogenesis. Molecular Therapy 22(4): 862-872. https://doi.org/10.1038/mt.2013.301 [PubMed] [PMC]

■ Boomsma RA, Geenen DL (2012) Mesenchymal stem cells secrete multiple cytokines that promote angiogenesis and have contrasting effects on chemotaxis and apoptosis. PLoS One 7(4): e35685. https://doi.org/10.1371/journal.pone.0035685 [PubMed] [PMC]

■ Brini AT, Amodeo G, Ferreira LM (2017) Therapeutic effect of human adipose-derived stem cells and their secretome in experimental diabetic pain. Scientific Reports 7(1): 9904. https://doi.org/10.1038/ s41598-017-09487-5 [PubMed] [PMC]

■ Burlacu A, Grigorescu G, Rosca AM, Preda MB, Simionescu M (2013) Factors secreted by mesenchymal stem cells and endothe-lial progenitor cells have complementary effects on angiogenesis

in vitro. Stem Cells and Development 22(4): 643-653. https://doi. org/10.1089/scd.2012.0273 [PubMed] [PMC]

■ Cai L, Johnstone BH, Cook TG, Liang Z, Traktuev D, Cornetta K, Ingram DA, Rosen ED, March KL (2007) Suppression of hepatocyte growth factor production impairs the ability of adipose-derived stem cells to promote ischemic tissue revascularization. Stem Cells 25: 3234-3243. https://doi.org/10.1634/stemcells.2007-0388

■ Campanella C, Caruso Bavisotto C, Logozzi M, Marino Gam-mazza A, Mizzoni D, Cappello F, Fais S (2019) On the choice of the extracellular vesicles for therapeutic purposes. International Journal of Molecular Sciences 20(2): 236. https://doi.org/10.3390/ ijms20020236 [PubMed] [PMC]

■ Cantinieaux D, Quertainmont R, Blacher S, Rossi L, Wanet T, Noel A, Brook G, Schoenen J, Franzen R (2013) Conditioned medium from bone marrow-derived mesenchymal stem cells improves recovery after spinal cord injury in rats: an original strategy to avoid cell transplantation. PLoS One 8(8): e69515. https://doi.org/10.1371/ journal.pone.0069515 [PubMed] [PMC]

■ Cargnoni A, Ressel L, Rossi D, Poli A, Arienti D, Lombardi G, Parolini O (2012) Conditioned medium from amniotic mesenchymal tissue cells reduces progression of bleomycin-induced lung fibrosis. Cytotherapy 14(2): 153-161. https://doi.org/10.3109/14653249.201 1.613930 [PubMed] [PMC]

■ Carvalho MM, Teixeira FG, Reis RL, Sousa N, Salgado AJ (2011) Mesenchymal stem cells in the umbilical cord: phenotypic characterization, secretome and applications in central nervous system regenerative medicine. Current Stem Cell Research & Therapy 6(3): 221-228. https://doi.org/10.2174/157488811796575332 [PubMed]

■ Caseiro AR, Pereira T, Ivanova G, Luis AL, Mauricio AC (2016) Neuromuscular regeneration: perspective on the application of mesenchymal stem cells and their secretion products. Stem Cells International 2016: 1-16. https://doi.org/10.1155/2016/9756973 [PubMed] [PMC]

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

■ Cheng NC, Wang S, Young TH (2012) The influence of spheroid formation of human adipose-derived stem cells on chitosan films on sternness and differentiation capabilities. Biomaterials 33(6): 17481758. https://doi.org/10.1016/j.biomaterials.2011.11.049 [PubMed]

■ Chang CP, Chio CC, Cheong CU, Chao CM, Cheng BC, Lin MT (2013) Hypoxic preconditioning enhances the therapeutic potential of the secretome from cultured human mesenchymal stem cells in experimental traumatic brain injury. Clinical Science 124(3): 165176. https://doi.org/10.1042/CS20120226 [PubMed]

■ Collawn SS, Mobley JA, Banerjee NS, Chow LT (2016) Conditioned media from adipose-derived stromal cells accelerates healing in 3-di-mensional skin cultures. Annals of Plastic Surgery 76(4): 446-452. https://doi.org/10.1097/SAP.0000000000000754 [PubMed]

■ 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. https:// doi.org/10.1002/stem.194

■ Cooper DR, Wang C, Patel R, Trujillo A, Patel NA, Prather J, Gould LJ, Wu MH (2018) Human adipose-derived stem cell conditioned media and exosomes containing MALAT1 promote human dermal fibroblast migration and ischemic wound healing. Advances in

Wound Care (New Rochelle) 7(9): 299-308. https://doi.org/10.1089/ wound.2017.0775 [PubMed] [PMC]

■ Cova L, Armentero MT, Zennaro E, Calzarossa C, Bossolasco P, Busca G, Lambertenghi Deliliers G, Polli E, Nappi G, Silani V, Blan-dini F (2010) Multiple neurogenic and neurorescue effects of human mesenchymal stem cell after transplantation in an experimental model of Parkinson's disease. Brain Research 1311: 12-27. https:// doi.org/10.1016/j.brainres.2009.11.041 [PubMed]

■ Cristofanilli M, Harris VK, Zigelbaum A, Goossens AM, Lu A, Rosenthal H, Sadiq SA (2011) Mesenchymal stem cells enhance the engraftment and myelinating ability of allogeneic oligodendrocyte progenitors in dysmyelinated mice. Stem Cells and Development 20(12): 2065-2076. https://doi.org/10.1089/scd.2010.0547 [PubMed]

■ Daquinag AC, Zhang Y, Amaya-Manzanares F, Simmons PJ, Kolonin MG (2011) An isoform of decorin is a resistin receptor on the surface of adipose progenitor cells. Cell Stem Cell 9(1): 74-86. https://doi.org/10.1016/j.stem.2011.05.017 [PubMed]

■ Daquinag AC, Souza GR, Kolonin MG (2013) Adipose tissue engineering in three dimensional levitation tissue culture system based on magnetic nanoparticles. Tissue Engineering Part C: Methods 19(5): 336-344. https://doi.org/10.1089/ten.tec.2012.0198 [PubMed] [PMC]

■ Datta I, Mishra S, Mohanty L, Pulikkot S, Joshi PG (2011) Neuronal plasticity of human Wharton's jelly mesenchymal stromal cells to the dopaminergic cell type compared with human bone marrow mes-enchymal stromal cells. Cytotherapy 13(8): 918-932. https://doi.org /10.3109/14653249.2011.579957 [PubMed]

■ de Almeida JF, Chen P, Henry MA, Diogenes A (2014) Stem cells of the apical papilla regulate trigeminal neurite outgrowth and targeting through a BDNF-dependent mechanism. Tissue Engineering Part A. 20(23-24): 3089-3100. https://doi.org/10.1089/ten.tea.2013.0347 [PubMed] [PMC]

■ Del Papa N, Caviggioli F, Sambataro D, Zaccara E, Vinci V, Di Luca G, Parafioriti A, Armiraglio E, Maglione W, Polosa R, Klinger F, Klinger M (2015) Autologous fat grafting in the treatment of fibrotic perioral changes in patients with systemic sclerosis. Cell Transplant 24(1): 63-72. https://doi.org/10.3727/096368914X674062 [PubMed]

■ Del Papa N, Di Luca G, Andracco R, Zaccara E, Maglione W, Pigna-taro F, Minniti A, Vitali C (2019) Regional grafting of autologous adipose tissue is effective in inducing prompt healing of indolent digital ulcers in patients with systemic sclerosis: results of a monocentric randomized controlled study. Arthritis Research and Therapy 21(1): 7. https://doi.org/10.1186/s13075-018-1792-8 [PubMed] [PMC]

■ De Luca A, Gallo M, Aldinucci D, Ribatti D, Lamura L, D'Alessio A, De Filippi R, Pinto A, Normanno N (2011) Role of the EGFR ligand/receptor system in the secretion of angiogenic factors in mes-enchymal stem cells. Journal of Cellular Physiology 226(8): 21312138. https://doi.org/10.1002/jcp.22548 [PubMed]

■ Di Santo S, Yang Z, Wyler von Ballmoos M, Voelzmann J, Diehm N, Baumgartner I, Kalka C (2009) Novel cell-free strategy for therapeutic angiogenesis: in vitro generated conditioned medium can replace progenitor cell transplantation. PLoS One 4(5): e5643. https://doi. org/10.1371/journal.pone.0005643 [PubMed] [PMC]

■ Du WJ, Chi Y, Yang ZX, Li ZJ, Cui JJ, Song BQ, Li X, Yang SG, Han ZB, Han ZC (2016) Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Research and Therapy 7(1): 163. https://doi.org/10.1186/s13287-016-0418-9 [PubMed] [PMC]

■ Dubey NK, Mishra VK, Dubey R, Deng YH, Tsai FC, Deng WP (2018) Revisiting the advances in isolation, characterization and secretome of adipose-derived stromal/stem cells. International Journal of Molecular Sciences 19(8): 2200. https://doi.org/10.3390/ ijms19082200 [PubMed] [PMC]

■ Eiro N, Sendon-Lago J, Seoane S, Bermüdez MA, Lamelas ML, Garcia-Caballero T, Schneider J, Perez-Fernandez R, Vizoso FJ (2014) Potential therapeutic effect of the secretome from human uterine cervical stem cells against both cancer and stromal cells compared with adipose tissue stem cells. Oncotarget 5(21): 10692-10708. https:// doi.org/10.18632/oncotarget.2530 [PubMed] [PMC]

■ Erba P, Terenghi G, Kingham PJ (2010) Neural differentiation and therapeutic potential of adipose tissue derived stem cells. Current Stem Cell Research & Therapy 5(2): 153-160. https://doi. org/10.2174/157488810791268645 [PubMed]

■ Ferreira ADF, Gomes DA (2018) Stem cell extracellular vesicles in skin repair. Bioengineering 6(1): 4. https://doi.org/10.3390/bioengi-neering6010004 [PubMed] [PMC]

■ Ferreira JR, Teixeira GQ, Santos SG, Barbosa MA, Almeida-Porada G, Gonjalves RM (2018) Mesenchymal stromal cell secretome: influencing therapeutic potential by cellular pre-conditioning. Frontiers in Immunology 9: 2837. https://doi.org/10.3389/fimmu.2018.02837 [PubMed] [PMC]

■ Ho JC, Lai WH, Li MF, Au KW, Yip MC, Wong NL, Ng ES, Lam FF, Siu CW, Tse HF (2012) Reversal of endothelial progenitor cell dysfunction in patients with type 2 diabetes using a conditioned medium of human embryonic stem cell-derived endothelial cells. Diabetes/Metabolism Research and Reviews 28(5): 462-473. https:// doi.org/10.1002/dmrr.2304 [PubMed]

■ Honmou O, Onodera R, Sasaki M, Waxman SG, Kocsis JD (2012) Mesenchymal stem cells: therapeutic outlook for stroke. Trends in Molecular Medicine 18(5): 292-297. https://doi.org/10.1016/j. molmed.2012.02.003 [PubMed]

■ Hsiao ST-F, Asgari A, Lokmic Z, Sinclair R, Dusting GJ, Lim SY, Dilley RJ (2012) Comparative analysis of paracrine factor expression in human adult mesenchymal stem cells derived from bone marrow, adipose,and dermal tissue. Stem Cells and Development 21(12): 2189-2203. https://doi.org/10.1089/scd.2011.0674 [PubMed] [PMC]

■ Hsieh JY, Wang HW, Chang SJ, Liao KH, Lee IH, Lin WS, Wu CH, Lin WY, Cheng SM (2013) Mesenchymal stem cells from human umbilical cord express preferentially secreted factors related to neuroprotection, neurogenesis, and angiogenesis. PLoS One 8(8): e72604. https://doi.org/10.1371/journal.pone.0072604 [PubMed] [PMC]

■ Hu L, Zhao J, Liu J, Gong N, Chen L (2013) Effects of adipose stem cell-conditioned medium on the migration of vascular endothelial cells, fibroblasts and keratinocytes. Experimental and Therapeutic Medicine 5: 701-706. https://doi.org/10.3892/etm.2013.887

■ Huch M, Dorrell C, Boj SF, van Es JH, Li VS, van de Wetering M, Sato T, Hamer K, Sasaki N, Finegold MJ, Haft A, Vries RG, Grompe M, Clevers H (2013) In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 494: 247-250. https://doi.org/10.1038/nature11826 [PubMed] [PMC]

■ Inukai T, Katagiri W, Yoshimi R, Osugi M, Kawai T, Hibi H, Ueda M (2013) Novel application of stem cell-derived factors for periodontal regeneration. Biochemical and Biophysical Research Communications 430(2): 763-768. https://doi.org/10.1016/j.bbrc.2012.11.074 [PubMed]

■ Kan I, Barhum Y, Melamed E, Offen D (2011). Mesenchymal stem cells stimulate endogenous neurogenesis in the subventricular

zone of adult mice. Stem Cell Reviews 7(2): 404-412. https://doi. org/10.1007/s12015-010-9190-x [PubMed]

■ Kapur SK, Katz AJ (2013) Review of the adipose derived stem cell secretome. Biochimie 95(12): 2222-2228. https://doi.org/10.1016/j. biochi.2013.06.001 [PubMed]

■ Kay AG, Long G, Tyler G, Stefan A, Broadfoot SJ, Piccinini AM, Middleton J, Kehoe O (2017) Mesenchymal stem cell-conditioned medium reduces disease severity and immune responses in inflammatory arthritis. Scientific Reports 7(1): 18019. https://doi. org/10.1038/s41598-017-18144-w [PubMed] [PMC]

■ Kim WS, Park BS, Sung JH, Yang JM, Park SB, Kwak SJ, Park JS (2007) Wound healing effect of adipose-derived stem cells: a critical role of secretory factors on human dermal fibroblasts. Journal of Dermatological Science 48(1): 15-24. https://doi.org/10.1016/j. jdermsci.2007.05.018 [PubMed]

■ Kim MH, Wu WH, Choi JH, Kim J, Jun JH, Ko Y, Lee JH (2018) Galectin-1 from conditioned medium of three-dimensional culture of adipose-derived stem cells accelerates migration and proliferation of human keratinocytes and fibroblasts. Wound Repair and Regeneration 26: S9-S18. https://doi.org/10.1111/wrr.12579 [PubMed]

■ Kinnaird T, Stabile E, Burnett MS, Shou M, Lee CW, Barr S, Fuchs S, Epstein SE (2004) Local delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation 109(12): 1543-1549. https://doi.org/10.1161/01. CIR.0000124062.31102.57 [PubMed]

■ Kober J, Gugerell A, Schmid M, Zeyda M, Buchberger E, Nickl S, Hacker S, Ankersmit HJ, Keck M (2016) Wound healing effect of conditioned media obtained from adipose tissue on human skin cells: a comparative in vitro study. Annals of Plastic Surgery 77(2): 156163. https://doi.org/10.1097/SAP.0000000000000358 [PubMed]

■ Kucharzewski M, Rojczyk E, Wilemska-Kucharzewska K, Wilk R, Hudecki J, Los MJ (2019) Novel trends in application of stem cells in skin wound healing. European Journal of Pharmacology 843: 307-315. https://doi.org/10.1016Zj.ejphar.2018.12.012 [PubMed]

■ Kyurkchiev D, Bochev I, Ivanova-Todorova E, Mourdjeva M, Ore-shkova T, Belemezova K, Kyurkchiev S (2014) Secretion of immu-noregulatory cytokines by mesenchymal stem cells. World Journal of Stem Cells 6(5): 552-570. https://doi.org/10.4252/wjsc.v6.i5.552 [PubMed] [PMC]

■ L PK, Kandoi S, Misra R, S V, K R, Verma RS (2019) The mesenchy-mal stem cell secretome: A new paradigm towards cell-free therapeutic mode in regenerative medicine. Cytokine and Growth Factor Reviews 46: 1-9. https://doi.org/10.1016/j.cytogfr.2019.04.002 [PubMed]

■ Leavitt T, Hu MS, Marshall CD, Barnes LA, Longaker MT, Lorenz HP (2016) Stem cells and chronic wound healing: State of the art. Chronic Wound Care Management and Research 3: 7-27. https://doi. org/10.2147/CWCMR.S84369

■ Lee MJ, Kim J, Lee KI, Shin JM, Chae JI, Chung HM (2011) Enhancement of wound healing by secretory factors of endothelial precursor cells derived from human embryonic stem cells. Cytotherapy 13(2): 165-178. https://doi.org/10.3109/14653249.2010.512632 [PubMed]

■ Lee SK, Lee SC, Kim S-J (2014) A novel cell-free strategy for promoting mouse liver regeneration: utilization of a conditioned medium from adipose-derived stem cells. Hepatology International 9(2): 310-320. https://doi.org/10.1007/s12072-014-9599-4 [PubMed]

■ Legaki E, Roubelakis MG, Theodoropoulos GE, Lazaris A, Kollia A, Karamanolis G, Marinos E, Gazouli M (2016) Therapeutic potential of secreted molecules derived from human amniotic fluid mesenchy-

mal stem/stroma cells in a mice model of colitis. Stem Cell Reviews and Reports 12(5): 604-612. https://doi.org/10.1007/s12015-016-9677-1 [PubMed]

■ Lepperdinger G, Brunauer R, Jamnig A, Laschober G, Kassem M (2008) Controversial issue: is it safe to employ mesenchymal stem cells in cell-based therapies? Experimental Gerontology 43(11): 1018-1023. https://doi.org/10.1016/j.exger.2008.07.004 [PubMed]

■ Li B, Zhang H, Zeng M, He W, Li M, Huang X, Deng DY, Wu J (2015) Bone marrow mesenchymal stem cells protect alveolar macrophages from lipopolysaccharide-induced apoptosis partially by inhibiting the Wnt/ß-catenin pathway. Cell Biology International 39(2): 192-200. https://doi.org/10.1002/cbin.10359 [PubMed]

■ Lindvall O, Kokaia Z (2010) Stem cells in human neurodegener-ative disorders-time for clinical translation? The Journal of Clinical Investigation 120(1): 29-40. https://doi.org/10.1172/JCI40543 [PubMed] [PMC]

■ Linero I, Chaparro O (2014) Paracrine effect of mesenchymal stem cells derived from human adipose tissue in bone regeneration. PLoS One 9(9): e107001. https://doi.org/10.1371/journal.pone.0107001

■ Liu G, Pareta RA, Wu R, Shi Y, Zhou X, Liu H, Deng C, Sun X, Atala A, Opara EC, Zhang Y (2013) Skeletal myogenic differentiation of urine-derived stem cells and angiogenesis using microbeads loaded with growth factors. Biomaterials 34(4): 1311-1326. https:// doi.org/10.1016/j.biomaterials.2012.10.038 [PubMed] [PMC]

■ Lombardi F, Palumbo P, Augello FR, Cifone MG, Cinque B, Giuliani M (2019) Secretome of adipose tissue-derived stem cells (ASCs) as a novel trend in chronic non-healing wounds: an overview of experimental in vitro and in vivo studies and methodological variables. International Jornal of Molecular Sciences 20(15): 3721. https://doi. org/10.3390/ijms20153721 [PubMed] [PMC]

■ Luarte A, Bätiz LF, Wyneken U, Lafourcade C (2016) Potential therapies by stem cell-derived exosomes in CNS diseases: focusing on the neurogenic niche. Stem Cells International 2016: 1-16. https:// doi.org/10.1155/2016/5736059 [PubMed] [PMC]

■ Madrigal M, Rao KS, Riordan NH (2014) A review of therapeutic effects of mesenchymal stem cell secretions and induction of secretory modification by different culture methods. Journal of Translational Medicine 12(1): 260. https://doi.org/10.1186/s12967-014-0260-8 [PubMed] [PMC]

■ Maguire G (2013) Stem cell therapy without the cells. Communicative and Integrative Biology 6(6): e26631. https://doi.org/10.4161/ cib.26631 [PubMed] [PMC]

■ Marfia G, Navone SE, Di Vito C, Ughi N, Tabano S, Miozzo M, Tremolada C, Bolla G, Crotti C, Ingegnoli F, Rampini P, Riboni L, Gualtierotti R, Campanella R (2015) Mesenchymal stem cells: potential for therapy and treatment of chronic non-healing skin wounds. Organogenesis 11(4): 183-206. https://doi.org/10.1080/15 476278.2015.1126018 [PubMed] [PMC]

■ Maumus M, Jorgensen C, Noel D (2013) Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: role of secretome and exosomes. Biochimie 95(12): 2229-2234. https://doi. org/10.1016/j.biochi.2013.04.017 [PubMed]

■ Mead B, Logan A, Berry M, Leadbeater W, Scheven BA (2014) Paracrine-mediated neuroprotection and neuritogenesis of axoto-mised retinal ganglion cells by human dental pulp stem cells: comparison with human bone marrow and adipose-derived mesenchymal stem cells. PLoS One 9(10): e109305. https://doi.org/10.1371/jour-nal.pone.0109305 [PubMed] [PMC]

■ Meiliana A, Dewi NM, Wijaya A (2019) Mesenchymal stem cell secretome: cell-free therapeutic strategy in regenerative medicine. The Indonesian Biomedical Journal 11(2): 113-124. https://doi. org/10.18585/inabj.v11i2.839

■ Mirabella T, Cilli M, Carlone S, Cancedda R, Gentili C (2011) Am-niotic liquid derived stem cells as reservoir of secreted angiogen-ic factors capable of stimulating neo-arteriogenesis in an ischemic model. Biomaterials 32(15): 3689-3699. https://doi.org/10.1016/j. biomaterials.2011.01.071 [PubMed]

■ Mita T, Furukawa-Hibi Y, Takeuchi H, Hattori H, Yamada K, Hibi H, Ueda M, Yamamoto A (2015) Conditioned medium from the stem cells of human dental pulp improves cognitive function in a mouse model of Alzheimer's disease. Behavioral Brain Research 293: 189197. https://doi.org/10.1016/j.bbr.2015.07.043 [PubMed]

■ Mitchell R, Mellows B, Sheard J, Antonioli M, Kretz O, Chambers D, Zeuner MT, Tomkins JE, Denecke B, Musante L, Joch B, De-bacq-Chainiaux F, Holthofer H, Ray S, Huber TB, Dengjel J, De Coppi P, Widera D, Patel K (2019) Secretome of adipose-derived mesenchymal stem cells promotes skeletal muscle regeneration through synergistic action of extracellular vesicle cargo and soluble proteins. Stem Cell Research and Therapy 10(1): 116. https://doi. org/10.1186/s13287-019-1213-1 [PubMed] [PMC]

■ Noverina R, Widowati W, Ayuningtyas W, Kurniawan D, Afifah E, Laksmitawati DR, Rinendyaputri R, Rilianawati R, Faried A, Bach-tiar I, Wirakusumah FF (2019) Growth factors profile in conditioned medium human adipose tissue-derived mesenchymal stem cells (cm-hatmscs). Clinical Nutrition Experimental 24: 34-44. https:// doi.org/10.1016/j.yclnex.2019.01.002

■ Osugi M, Katagiri W, Yoshimi R, Inukai T, Hibi H, Ueda M (2012) Conditioned media from mesenchymal stem cells enhanced bone regeneration in rat calvarial bone defects. Tissue Engineering Part A 18(13-14): 1479-1489. https://doi.org/10.1089/ten.tea.2011.0325 [PubMed] [PMC]

■ Park JH, Kim DY, Sung IY, Choi GH, Jeon MH, Kim KK, Jeon SR (2012) Long-term results of spinal cord injury therapy using mesenchymal stem cells derived from bone marrow in humans. Neurosurgery 70(5): 1238-1247. https://doi.org/10.1227/ NEU.0b013e31824387f9 [PubMed]

■ Park SR, Kim JW, Jun HS, Roh JY, Lee HY, Hong IS (2018) Stem cell secretome and its effect on cellular mechanisms relevant to wound healing. Molecular Therapy 26(2): 606-617. https://doi. org/10.1016/j.ymthe.2017.09.023 [PubMed] [PMC]

■ Pelizzo G, Avanzini MA, Icaro Cornaglia A, De Silvestri A, Mantelli M, Travaglino P, Croce S, Romano P, Avolio L, Iacob G, Dominici M, Calcaterra V (2018) Extracellular vesicles derived from mesen-chymal cells: perspective treatment for cutaneous wound healing in pediatrics. Regenerative Medicine 13(4): 385-394. https://doi. org/10.2217/rme-2018-0001 [PubMed]

■ Pereira T, Ivanova G, Caseiro AR, Barbosa P, Bartolo PJ, Santos JD, et al. (2014) MSCs conditioned media and umbilical cord blood plasma metabolomics and composition. PLoS ONE 9(11): e113769. https://doi.org/10.1371/journal.pone.0113769 [PubMed] [PMC]

■ Pittenger MF, Mackay AM, Beck SC, Jaiswai RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR (1999) Multilineage potential of adult human mesenchymal stem cells. Science 284(5411): 143-147. https://doi.org/10.1126/sci-ence.284.5411.143 [PubMed]

■ Polacek M, Bruun JA, Elvenes J, Figenschau Y, Martinez I (2011) The secretory profiles of cultured human articular chondrocytes and mesenchymal stem cells: implications for autologous cell transplantation strategies. Cell Transplantation 20(9): 1381-1393. https://doi. org/10.3727/096368910X550215 [PubMed]

■ Pouya S, Heidari M, Baghaei K, Asadzadeh Aghdaei H, Moradi A, Namaki S, Zali MR, Hashemi SM (2018) Study the effects of mesenchymal stem cell conditioned medium injection in mouse model of acute colitis. Internatinal Immunopharmacology 54: 86-94. https:// doi.org/10.1016/j.intimp.2017.11.001 [PubMed]

■ Preda MB, Renningen T, Burlacu A, Simionescu M, Moskaug J0, Valen G (2014) Remote transplantation of mesenchymal stem cells protects the heart against ischemia-reperfusion injury. Stem Cells 32(8): 2123-2134. https://doi.org/10.1002/stem.1687 [PubMed]

■ Ratajczak J, Bronckaers A, Dillen Y, Gervois P, Vangansewinkel T, Driesen RB, Wolfs E, Lambrichts I, Hilkens P (2016) The neurovas-cular properties of dental stem cells and their importance in dental tissue engineering. Stem Cells International 2016: 1-17. https://doi. org/10.1155/2016/9762871 [PubMed]

■ Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, Pell CL, Johnstone BH, Considine RV, March KL (2004) Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation 109(10): 1292-1298. https://doi. org/10.1161/01.CIR.0000121425.42966.F1 [PubMed]

■ 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 Research and Therapy 3(3): 18. https://doi.org/10.1186/scrt109 [PubMed]

■ Salgado AJ, Fraga JS, Mesquita AR, Neves NM, Reis RL, Sousa N (2010) Role of human umbilical cord mesenchymal progenitors conditioned media in neuronal/glial cell densities, viability, and proliferation. Stem Cells and Development 19(7): 1067-1074. https:// doi.org/10.1089/scd.2009.0279 [PubMed]

■ Salgado AJ, Sousa JC, Costa BM, Pires AO, Mateus-Pinheiro A, Teixeira FG, Pinto L, Sousa N (2015) Mesenchymal stem cells secretome as a modulator of the neurogenic niche: basic insights and therapeutic opportunities. Frontiers in Cellular Neuroscience 9: 249. https://doi.org/10.3389/fncel.2015.00249 [PubMed]

■ Sarugaser R, Lickorish D, Baksh D, Hosseini MM, Davies JE (2005) Human umbilical cord perivascular (HUCPV) cells: a source of mesenchymal progenitors. Stem Cells 23(2): 220-229. https://doi. org/10.1634/stemcells.2004-0166 [PubMed]

■ Sarugaser R, Ennis J, Stanford WL, Davies JE (2009) Isolation, propagation, and characterization of human umbilical cord perivascular cells (HUCPVCs). Stem Cells in Regenerative Medicines: Methods and Protocols 482: 269-279. https://doi.org/10.1007/978-1-59745-060-7_17 [PubMed]

■ See F, Seki T, Psaltis PJ, Sondermeijer HP, Gronthos S, Zannet-tino AC, Govaert KM, Schuster MD, Kurlansky PA, Kelly DJ, Krum H, Itescu S (2011) Therapeutic effects of human STRO-3-se-lected mesenchymal precursor cells and their soluble factors in experimental myocardial ischemia. Journal of Cellular and Molecular Medicine 15(10): 2117-2129. https://doi.org/10.1111/j.1582-4934.2010.01241.x [PubMed]

■ Seo E, Lim JS, Jun JB, Choi W, Hong IS, Jun HS (2017) Exendin-4 in combination with adipose-derived stem cells promotes angiogen-

esis and improves diabetic wound healing. Journal of Translation-al Medicine 15(1): 35. https://doi.org/10.1186/s12967-017-1145-4 [PubMed]

■ Sharma AK, Bury MI, Fuller NJ, Marks AJ, Kollhoff DM, Rao MV, Hota PV, Matoka DJ, Edassery SL, Thaker H, Sarwark JF, Janicki JA, Ameer GA, Cheng EY (2013) Cotransplantation with specific populations of spina bifida bone marrow stem/progenitor cells enhances urinary bladder regeneration. Proceedings of the National Academy of Sciences 110(10): 4003-4008. https://doi.org/10.1073/ pnas.1220764110 [PubMed]

■ Shihabuddin LS, Aubert I (2010) Stem cell transplantation for neuromet-abolic and neurodegenerative diseases. Neuropharmacology 58(6): 845-854. https://doi.org/10.1016/j.neuropharm.2009.12.015 [PubMed]

■ Stagg J, Galipeau J (2013) Mechanisms of immune modulation by mesenchymal stromal cells and clinical translation. Current Molecular Medicines 13(5): 856-867. https://doi. org/10.2174/1566524011313050016 [PubMed]

■ Stojanovic S, Najman S (2019) The effect of conditioned media of stem cells derived from lipoma and adipose tissue on macrophages' response and wound healing in indirect co-culture system in vitro. International Journal of Molecular Sciences 20(7): E1671. https:// doi.org/10.3390/ijms20071671 [PubMed]

■ Taghizadeh RR, Cetrulo KJ, Cetrulo CL (2011). Wharton's Jelly stem cells: future clinical applications. Placenta 32(Suppl 4): S311-S315. https://doi.org/10.1016/j.placenta.2011.06.010 [PubMed]

■ Tang YL, Zhao Q, Qin X, Shen L, Cheng L, Ge J, Phillips MI (2005) Paracrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarction. The Annals of Thoracic Surgery 80(1): 229237. https://doi.org/10.1016/j.athoracsur.2005.02.072 [PubMed]

■ Teixeira FG, Carvalho MM, Sousa N, Salgado AJ (2013) Mesen-chymal stem cells secretome: a new paradigm for central nervous system regeneration? Cellular and Molecular Life Sciences 70(20): 3871-3882. https://doi.org/10.1007/s00018-013-1290-8 [PubMed]

■ Teixeira FG, Carvalho MM, Neves-Carvalho A, Panchalingam KM, Behie LA, Pinto L, Sousa N, Salgado AJ (2014). Secretome of mes-enchymal progenitors from the umbilical cord acts as modulator of neural/glial proliferation and differentiation. Stem Cell Reviews and Reports 11(2): 288-297. https://doi.org/10.1007/s12015-014-9576-2

■ Traktuev DO, Merfeld-Clauss S, Li J, Kolonin M, Arap W, Pasqualini R, Johnstone BH, March KL (2008) A population of multipotent CD34-positive adipose stromal cells share pericyte and mesenchy-mal surface markers, reside in a periendothelial location, and stabilize endothelial networks. Circulation Research 102(1): 77-85. https://doi.org/10.1161/CIRCRESAHA.107.159475 [PubMed]

■ Turner JE, Morrison PJ, Wilhelm C, Wilson M, Ahlfors H, Renauld JC, Panzer U, Helmby H, Stockinger B (2013) IL-9-mediated survival of type 2 innate lymphoid cells promotes damage control in helminth-induced lung inflammation. Journal of Experimental Medicine 210(13): 2951-2965. https://doi.org/10.1084/jem.20130071 [PubMed]

■ Vishnubhatla I, Corteling R, Stevanato L, Hicks C, Sinden J (2014) The development of stem cell-derived exosomes as a cell-free regenerative medicine. Journal of Circulation Biomarkers 3: 3. https://doi. org/10.5772/58597

■ Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R (2017) Mesenchymal stem cell secretome: toward cell-free therapeutic

strategies in regenerative medicine. International Jurnal of Molecular Sciences 18(9): 1852. https://doi.org/10.3390/ijms18091852 [PubMed] [PMC]

■ Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, et al. (2004) Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord. Stem Cells 22(7): 1330-1337. https://doi. org/10.1634/stemcells.2004-0013 [PubMed]

■ Wei X, Zhao L, Zhong J, Gu H, Feng D, Johnstone BH, March KL, Farlow MR, Du Y (2009) Adipose stromal cells-secreted neu-roprotective media against neuronal apoptosis. Neuroscience Letters 462(1): 76-79. https://doi.org/10.1016/j.neulet.2009.06.054 [PubMed]

■ Weiss ML, Troyer DL (2006) Stem cells in the umbilical cord. Stem Cell Reviews 2(2): 155-162. https://doi.org/10.1007/s12015-006-0022-y [PubMed]

■ Williams AR, Suncion VY, McCall F, Guerra D, Mather J, Zambrano JP, Heldman AW, Hare JM (2013) Durable scar size reduction due to allogeneic mesenchymal stem cell therapy regulates whole-chamber remodeling. Journal of American Heart Association 2(3): e000140. https://doi.org/10.1161/JAHA.113.000140 [PubMed]

■ Xu J, Wang B, Sun Y, Wu T, Liu Y, Zhang J, Yukwai Lee W, Pan X, Chai Y, Li G (2016) Human fetal mesenchymal stem cell secretome enhances bone consolidation in distraction osteogenesis. Stem Cell Research and Therapy 7(1): 134. https://doi.org/10.1186/s13287-016-0392-2 [PMC]

■ Yi T, Song SU (2012) Immunomodulatory properties of mesenchy-mal stem cells and their therapeutic applications. Archives of Phar-macal Research 35(2): 213-221. https://doi.org/10.1007/s12272-012-0202-z [PubMed]

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

■ Zagoura DS, Roubelakis MG, Bitsika V, Trohatou O, Pappa KI, Kapelouzou A, Antsaklis A, Anagnou NP (2012) Therapeutic potential of a distinct population of human amniotic fluid mesen-chymal stem cells and their secreted molecules in mice with acute hepatic failure. Gut 61(6): 894-906. https://doi.org/10.1136/gut-jnl-2011-300908 [PubMed]

■ Zanotti L, Angioni R, Cali B, Soldani C, Ploia C, Moalli F, Garge-sha M, D'Amico G, Elliman S, Tedeschi G, Maffioli E, Negri A, Zacchigna S, Sarukhan A, Stein JV, Viola A (2016) Mouse mesenchymal stem cells inhibit high endothelial cell activation and lymphocyte homing to lymph nodes by releasing TIMP-1. Leukemia 30(5): 1143-1154. https://doi.org/10.1038/leu.2016.33 [PubMed]

■ Zhao J, Hu L, Liu J, Gong N, Chen L (2013) The effects of cyto-kines in adipose stem cell-conditioned medium on the migration and proliferation of skin fibroblasts in vitro. Biomedical Research International 2013: 1-11. https://doi.org/10.1155/2013/578479 [PubMed]

■ Zhou BR, Xu Y, Guo SL, Xu Y, Wang Y, Zhu F, Permatasari F, Wu D, Yin ZQ, Luo D (2013) The effect of conditioned media of adipose-derived stem cells on wound healing after ablative fractional carbon dioxide laser resurfacing. Biomedical Research International 2013: 1-9. https://doi.org/10.1155/2013/519126 [PubMed]

■ Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH (2002) Human adipose tissue is a source of multipotent stem cells. Molecular Biology of the Cell 13(12): 4279-4295. https://doi.org/10.1091/mbc.e02-02-0105 [PubMed]

Author contributions

■ Liubov A. Pokrovskaya, Junior Researcher of Laboratory of Polymers and Composite Materials, e-mail: pokrovskayal@ect-center.com, ORCID ID http://orcid.org/0000-0003-4764-0665. The author made substantial contributions to the conceptualization of the article and later participated in drafting the article.

■ Ekaterina V. Zubareva, PhD in Biology, Associate Professor of the Department of Biology, e-mail: zubareva@ bsu.edu.ru, ORCID ID http://orcid.org/0000-0002-6480-7810. The author made substantial contributions to the conceptualization of the article and later participated in drafting the article, prepared the final version of the paper.

■ Sergey V. Nadezhdin, PhD in Biology, Associate Professor, Head of the Scientific Research Laboratory of Cellular, Assisted Reproductive and DNA Technologies, e-mail: nadezhdin@bsu.edu.ru, ORCID ID http://orcid.org/0000-0001-8444-6330. The author made substantial contributions to the conceptualization of the article, participated in drafting the article, and gave the final approval of the version to be submitted.

■ Anna S. Lysenko, undergraduate student, e-mail: 1313833@bsu.edu.ru. The author participated in drafting the article.

■ Tatyana L. Litovkina, undergraduate student, e-mail: 1197105@bsu.edu.ru. The author participated in drafting the article.

i Надоели баннеры? Вы всегда можете отключить рекламу.