13
Mulpotent Potenal of Human Adult Mesenchymal Stem Cells Quratulain Yousaf *, Amna Tirmzi, Saman Ahsan and Amber Afroz Department of Biochemistry and Biotechnology, University of Gujrat, Pakistan *Corresponding author: Quratulain Yousaf, Department of Biochemistry and Biotechnology, University of Gujrat, Pakistan, Tel: +92-308-8042812; E-mail: [email protected] Received Date: June 27, 2018; Accepted Date: July 06, 2018; Published Date: July 09, 2018 Citaon: Yousaf Q, Tirmzi A, Ahsan S, Afroz A (2018) Mulpotent Potenal of Human Adult Mesenchymal Stem Cells. Biochem Mol Biol J Vol. 4: No.2:16. Abstract Aim of the study is to cite the recent research findings about mulpotent adult mesenchymal stem cells or stromal cells (MSCs) considering their origin, phenotypical and funconal characterizaon as well as being a bio medical ideal candidate for their effecve administraon in MSC-mediated clinical trials. MSCs were firstly reported by Friedenstein along with his coworkers within the bone marrow of an adult individual categorized as plasc adherent, colony forming fibroblasc cells with capability of reconstung a hematopoiec medium when graſted in vivo. Gradually with years, it has been revealed that these stromal cells with potenal to differenate in mulple mesenchymal mesodermal and non-mesodermal cell lines like adipocytes, osteoblasts and chondrocytes, are not limited to bone marrow cells but are also exclusive part of some other ssues and organs. Many evidences have provided an insight supporng their self-renewal ability combined with their mulpotency both in vivo and invitro and it has exploited their potenal in mulorgan engraſtment, ssue engineering therapies, immune regulatory properes along with capacity to aid as natural repair mechanism for ssues both invitro and in vivo. Though, despite this striking spectrum of invesgaon, many queries regarding their developmental origin, reproducible pluripotency and their in vivo funconal mechanism of modeling and remodeling of bones along with ssue mending are yet answerable and a lot of challenges are needed to deal with before administering them in broad clinical prosecuons. Keywords Stromal cells; Pluripotency; Reproducible; Mulorgan engraſtment; Striking spectrum; Prosecuons; Tissue mending Abbreviaons MSCs: Mesenchymal Stem Cell; BM: Bone Marrow; HSCs: Hematopoiec Stem cells; CFU-Fs: Colonized fibroblasc units; GVHD: Graſt Versus Host Disease; AT: Adipose Tissues; EPCs: Endothelial Progenitor Cells; BMP: Bone Morphogenic Protein; NEBL: Nebulee; LPL: Lipoprotein Lipase Genes; qRT PCR: Qualitave Real Time-PCR; Runx2: Runt-Related Transcripon Factor; IBMX: Isobutyl Methyl Xanthine; GNDF: Glial Cell Lines Derived Neurotrophic Growth Factors; HNF-4α: hepatocyte nuclear Factor; ZNF: Zinc Finger Protein; PDGFR: Platelet Derived Growth Factor Receptor; LVEF: Leſt Ventricular Ejecon Fracon; CNS: Central Nervous System; DCs: Dendric Cells; CM: Condioning Media; CARCs: CXCL Abundant Recular Cells Introducon Mesenchymal stromal cells stem cells (MSCs) have elucidated the global and scienfic aenon mainly due to their potenal to harbor the self-renewal and regenerave capability during repairing, liming and reconstung the respecve ssue components in bone regeneraon phenomenon [1]. Being characterized as heterogenous, plasc adherent, disnct stromal sub-populaon, MSCs having the lineage commitment capability is an acve field of invesgaon in the world of stem cell based innovaons [2]. Aſter HSCs discovery, research work of Friedenstein tesfied the presence of non- hematopoiec stromal cells with adipose and skeletal potenal observed during heterotopic transplantaon that can form recular cells, carlage, fat cells and bone [3]. With passing years, researchers have extended and confirmed these observaons by isolang them from bone marrow (BM) and by elaborang the passaging and induced differenaon procedures of MSCs under the invitro cultured condions into mulple mesodermal and non-mesodermal lineages [4,5]. Progressive researches in field of MSCs has revealed the MSCs niche component and it has explained that though bone marrow is direct source of isolaon of MSCs but it is also found in post-natal organs considering from the birth ssues to skeletal muscles and even dental pulp. However, the frequency and number of MSCs present in different ssues can be varied depending on the corresponding ssue type [6-8]. Significant novel methods of immunoselecon, enzymac and non-enzymac degradaon and analycal PCR has been performed for analyzing their phenotypic characteriscs, cells expansion, and differenaon in vitro as well the recognion of factors that served to play part during their in vivo Review iMedPub Journals www.imedpub.com DOI: 10.21767/2471-8084.100065 Biochemistry & Molecular Biology Journal ISSN 2471-8084 Vol.4 No.2:16 2018 © Copyright iMedPub | This article is available from: http://biochem-molbio.imedpub.com/inpress.php 1

Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

Multipotent Potential of Human Adult Mesenchymal Stem CellsQuratulain Yousaf *, Amna Tirmzi, Saman Ahsan and Amber Afroz

Department of Biochemistry and Biotechnology, University of Gujrat, Pakistan

*Corresponding author: Quratulain Yousaf, Department of Biochemistry and Biotechnology, University of Gujrat, Pakistan, Tel:+92-308-8042812; E-mail: [email protected]

Received Date: June 27, 2018; Accepted Date: July 06, 2018; Published Date: July 09, 2018

Citation: Yousaf Q, Tirmzi A, Ahsan S, Afroz A (2018) Multipotent Potential of Human Adult Mesenchymal Stem Cells. Biochem Mol Biol J Vol. 4:No.2:16.

Abstract

Aim of the study is to cite the recent research findingsabout multipotent adult mesenchymal stem cells orstromal cells (MSCs) considering their origin, phenotypicaland functional characterization as well as being a biomedical ideal candidate for their effective administrationin MSC-mediated clinical trials. MSCs were firstly reportedby Friedenstein along with his coworkers within the bonemarrow of an adult individual categorized as plasticadherent, colony forming fibroblastic cells with capabilityof reconstituting a hematopoietic medium when graftedin vivo. Gradually with years, it has been revealed thatthese stromal cells with potential to differentiate inmultiple mesenchymal mesodermal and non-mesodermalcell lines like adipocytes, osteoblasts and chondrocytes,are not limited to bone marrow cells but are also exclusivepart of some other tissues and organs. Many evidenceshave provided an insight supporting their self-renewalability combined with their multipotency both in vivo andinvitro and it has exploited their potential in multiorganengraftment, tissue engineering therapies, immuneregulatory properties along with capacity to aid as naturalrepair mechanism for tissues both invitro and in vivo.Though, despite this striking spectrum of investigation,many queries regarding their developmental origin,reproducible pluripotency and their in vivo functionalmechanism of modeling and remodeling of bones alongwith tissue mending are yet answerable and a lot ofchallenges are needed to deal with before administeringthem in broad clinical prosecutions.

KeywordsStromal cells; Pluripotency; Reproducible; Multiorgan

engraftment; Striking spectrum; Prosecutions; Tissue mending

AbbreviationsMSCs: Mesenchymal Stem Cell; BM: Bone Marrow; HSCs:

Hematopoietic Stem cells; CFU-Fs: Colonized fibroblastic units;GVHD: Graft Versus Host Disease; AT: Adipose Tissues; EPCs:Endothelial Progenitor Cells; BMP: Bone Morphogenic Protein;

NEBL: Nebulette; LPL: Lipoprotein Lipase Genes; qRT PCR:Qualitative Real Time-PCR; Runx2: Runt-Related TranscriptionFactor; IBMX: Isobutyl Methyl Xanthine; GNDF: Glial Cell LinesDerived Neurotrophic Growth Factors; HNF-4α: hepatocytenuclear Factor; ZNF: Zinc Finger Protein; PDGFR: PlateletDerived Growth Factor Receptor; LVEF: Left VentricularEjection Fraction; CNS: Central Nervous System; DCs: DendriticCells; CM: Conditioning Media; CARCs: CXCL AbundantReticular Cells

IntroductionMesenchymal stromal cells stem cells (MSCs) have

elucidated the global and scientific attention mainly due totheir potential to harbor the self-renewal and regenerativecapability during repairing, limiting and reconstituting therespective tissue components in bone regenerationphenomenon [1]. Being characterized as heterogenous, plasticadherent, distinct stromal sub-population, MSCs having thelineage commitment capability is an active field ofinvestigation in the world of stem cell based innovations [2].

After HSCs discovery, research work of Friedenstein testifiedthe presence of non- hematopoietic stromal cells with adiposeand skeletal potential observed during heterotopictransplantation that can form reticular cells, cartilage, fat cellsand bone [3]. With passing years, researchers have extendedand confirmed these observations by isolating them from bonemarrow (BM) and by elaborating the passaging and induceddifferentiation procedures of MSCs under the invitro culturedconditions into multiple mesodermal and non-mesodermallineages [4,5].

Progressive researches in field of MSCs has revealed theMSCs niche component and it has explained that though bonemarrow is direct source of isolation of MSCs but it is also foundin post-natal organs considering from the birth tissues toskeletal muscles and even dental pulp. However, the frequencyand number of MSCs present in different tissues can be varieddepending on the corresponding tissue type [6-8].

Significant novel methods of immunoselection, enzymaticand non-enzymatic degradation and analytical PCR has beenperformed for analyzing their phenotypic characteristics, cellsexpansion, and differentiation in vitro as well the recognitionof factors that served to play part during their in vivo

Review

iMedPub Journalswww.imedpub.com

DOI: 10.21767/2471-8084.100065

Biochemistry & Molecular Biology Journal

ISSN 2471-8084Vol.4 No.2:16

2018

© Copyright iMedPub | This article is available from: http://biochem-molbio.imedpub.com/inpress.php 1

Page 2: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

transplantation [9,10]. Various evidences have suggested thatmany signals transducing networks and interacting pathwaysare involved in regulating the differentiation of MSCs. Due tosignificant clinical advancement of MSCs in field of medicine, itis considered to be imperative to expose pathways adapted byregulators with which MSCs undergone differentiation anddiscovery of micro RNA dependent lineage commitment hasprovided a new insight in understanding the mechanismsregulating their differentiation [11,12].

Operationally, all the findings about MSCs is virtuallydependent on their invitro characterization and a very littlehas been cited about their in vivo counter partner. Therefore,criteria for defining MSCs is actually on basis of their in vitroself-renewal and differentiation potential [13]. Lately, an efforthas been done in explaining their anatomical origin and it hasshown their homology with in vivo residing pericytic cellssurrounded by blood vessels [14]. Additionally, no defined invivo assays have been identified so far that can explain itsunique marker of stemness, yet, recent studies has suggestedthe in vivo role of MSCs in maintaining the hematopoieticenvironment and stability of the bone marrow compartment[15].

An important reason to investigate the origin andunderlying differentiation mechanism of MSCs is to enhancetheir therapeutic potential in preclinical and clinical trials.Scientists have made advancement for analysis of the healingbenefits of MSCs according to their biological functions andthey have concluded that certain growth factors, solublesecretory molecules and paracrine factors are involved thatengage the recipient cells during tissue repair process.Currently, main goal of scientific innovations in study of MSCsis there in vivo characterization as it can lead to isolation ofenriched pure stromal cells population that can be used fororganogenesis, tissue engraftment and immunomodulatoryeffects which will renovate the world of clinical cell mediatedtherapies [16].

Literature ReviewThis brief review provides a brief understanding about MSCs

spotted right from their original perception and conception toits therapeutic properties focusing their in vivo and in vitroregulatory mechanisms of differentiation into multiple lineagesand their organizing roles in the clinical biology.

Origin and Perception of Mesenchymal StemCells

Cohnheim, in 1867, was the first scientist who hypothesizedthat a bone marrow (BM) source of the fibroblasts exists andplayed its role during the wound healing process. Followed bythis observation, Maximow described a vital link among freshlyforming blood constituents and mesodermal cells during theprocess of embryogenesis. He postulated the role of stromalmarrow in subsiding the progression and maintenance ofblood with hematopoietic organs. These observations done byboth scientists presented the very first idea about the

existence of stromal cells population in BM that served to beinvolved in natural healing process and hematopoiesis [17,18].

In early 1960s, in vivo BM transplantation demonstrated therole of these precursors as formers of cellular tissues.Transplanting these stromal cells in subcutaneous layer or justbeneath the kidney capsule led strangely towards growth ofectopic marrow. Decomposing the origins of these ectopicmarrow cells, it exposed the bony trabeculae of donor,adipocytes, myeloid supportive stroma and host derivatives ofhematopoietic stem cells that formed the colony and undergomaturation inside the lumen [19,20]. Friedenstein et al. werefirst who verified that stromal cells possibly be secluded fromBM based on their ability to adhere differentially with tissueculture plastic technique, still used for MSCs isolation. Thesecells were firstly regarded as non-phagocytic and adherentcells having ability to form colony forming units that arefibroblastic in nature [21]. Experiments made with connectivetissues and dermal fibroblasts failed to retrieve the similarhistological demonstration as made by when stromal cellswere transplanted in kidney capsule or other tissues,representing it a marrow stroma specific phenomenon. Thesepivotal experiments proved that MSCs has similar function likelymphoid stromal cells and recommended MSCs to be thepredecessors of BM connective tissues. Lately in 1980s,Maureen Owen with Caplan demonstrated the work ofFriedenstein and proposed about the presence of adult stemcells, considered to be responsible for process ofmesengenesis. Further, Owen et al. characterized thesemarrow stroma cells and also illustrate their heterogenicity[22,23]. Meanwhile, Caplan and his colleagues theorized thatsubpopulation of stromal cells was developmentally linkedwith mesenchymal tissues which they had been analyzing inthe process of chick embryogenesis. Moreover, scientistsidentified the first MSCs expressed antigens which show areaction with monoclonal antibodies at SH2 (CD105) and SH3(CD73) subsequently. He formulated the name “mesenchymalstem cells” for describing that subtype of stromal cells whichwere found to be involved in mesengenesis [23-25].

Presence of a stromal precursor differentiating intomesodermal cell lines in BM was initially theorized innineteenth century [26]. Succeeding towards approaches usedfor isolation and culturing of MSCs, this field began its progressrapidly and many scientists began to discover their therapeuticpractices. Few years after the discovery of MSCs, humanefforts were inaugurated to assess the safety and effectivenessof MSCs mediated therapy. Primarily, autologous MSCs wereinvestigated to aid in tissue engraftment and retrieval ofhematopoiesis after ablation and marrow transplantationperformed in treatment of cancer [27,28]. Concurrently,researchers conducted a series of innovative experiments toinvestigate about therapeutic probabilities for allogeneic MSCstransplants in treatment of children suffering fromosteogenesis imperfecta which is a genetic disease of skeletaldysplasia [29]. Thereafter, further investigations were madefor treating patients suffering with Hurler syndrome andmetachromatic leukodystrophy with help of allogenic MSCs.The results of these investigations were very significant as itprovided evidence for safety of MSCs mediated therapies [30].

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

2 This article is available from: http://biochem-molbio.imedpub.com/inpress.php

Page 3: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

Lately, International Society for the Mesenchymal Tissueand Stem Cell Committee projected the minimal criteria fordefining human MSCs (hMSCs). Firstly, hMSCs must be plasticadherent under standard cultured environment and must formcolonized fibroblastic units (CFU-Fs). Secondly, hMSCs mustexhibit CD73, CD90 and CD105 markers expression andabsence of CD14, CD11b, CD19, CD34, CD45, CD79 and HLA-DRmolecules. Lastly, hMSCs must differentiate intochondroblasts, osteoblasts and adipocytes in vitro [31].

Currently, on basis of recent innovation made aboutpleotropic function of MSCs that favors to attenuate immunedysfunction as well as enhancing endogenous repairment,MSCs transplantation has been investigated for treatingmyriad disease. Presently, registered experimental sites arepresent for assessing MSCs therapy all over the world,signifying global curiosity for exploring MSCs as a potentialtherapy source. Most of the trials are funded by academicmedical centers for exploring novel features of MSCs in diverseareas as liver cirrhosis, spinal cord injury and critical limbischemia. Osiris Therapeutics, instituted in 1991, has played aninfluential role in past decade in promoting the research andexpansion of MSC based clinical therapies. Based onresearches made on immunological role of MSCs, it hasintroduced a clinical trial to explore the therapeutic efficacy ofMSCs in humans. It has pioneered the studies to investigatethe administration of MSCs as a therapy for type 1- diabetesmellitus, crohon’s disease, myocardial infraction, graft-versus-host diseases (GvHD) and chronic obstructive pulmonarydisorders (COPD). A good safety record for MSCs injection hasexpanded the MSCs mediated clinical trials in Phase II andPhase III and currently MSCs have been approved as expandedaccess to be used in pediatric refractive GvHD [32-34].However, molecular signature and their in vivo status ofproliferation remain unclear and is subjected to investigation,though the ex vivo cultured MSCs are being extensively used inseveral studies [35,36].

Many clinical trials entail the administration of MSCssystemically and assume that MSCs engraftment provide longterm support by either directly replenishing damaged tissue orinteracting with neighboring cells to favor the endogenousrepair. Currently, it is widely debated whether MSCsengraftment, proliferation, or differentiation is obligatory fortherapeutic benefit [37]. Many recent studies are nowpredicting the paracrine signaling as the primary mechanismof action followed by soluble factors secreted by MSCs and afew studies have even demonstrated that direct injection ofthe molecules secreted by MSCs can provide an improvedbenefit when transplanted to the whole cells [38].

MSCs Niche evidences from several tissuesBM derived MSCs, due to their ease in accessibility,

multipotent nature and expandability, hold significantpotential for their role in the regenerative medicines as well astissue engineering [39]. However, besides the BM, wellrecognized sources are also reported in body for MSCs.Probably, primary MSCs population can be isolated from fetalbirth tissues like Wharton’s jelly, umbilical cord matrix and

umbilical cord blood. It is worthy to note that umbilical cordblood matrix serves more good source of MSCs than umbilicalcord blood [40]. Wharton’s jelly mainly contain primitive MSCsthus upsurges their potential in therapeutic submissions [41].Additionally, amniotic fluid has also been revealed to containMSCs and it can be attained with amniocentesis or during thebirth time.

Other birth related tissues like amnion and placenta thoughcontain MSCs, however due to their quantity and regarding thefact that they hold a heterogenous hematopoietic stem cells(HSCs) population and endothelial progenitor cells (EPC), theirpotential therapeutics differs from the pure MSCs [42]. Withgrowth, the next possible young cell source is the developingdental pulp which is easy to collect among 8-10 of ages inchildren. There is an ambiguity if these cells serve to bepluripotent or multipotent, however, they are proposed to bea key source for cell banking in future and have potential toreplace usage of umbilical cords [43].

In adults, Adipose tissues (AT) are found to be direct sourceof MSCs, which is simply accessible and a well-definedmethodology is present that favors their isolation from source.It is assessed that round about 500 times, more in number, ATderived MSCs can be isolated more easily from fat tissuescomparative to the BM of same amount [44]. Moreover,mobilized peripheral blood also serve as a source of MSCs aswell, anyhow, conceded by several aspects related to thedonors like sex, age, feeding habit, administered drugs,daytime and health status. It has been used in cartilage repairand efficient results are observed [45].

Efficient population of MSCs has been reported in menstrualblood and endometrium (Figure 1), that are found to beexhibiting stem cell like marker and phenotypes and are foundto show neuroprotection in a stroke condition [46,47]. Skinand fore skin are also source of MSCs and evidenced to bedifferentiated into multiple lineages. [48].

Figure 1 A flow representation of Mesenchymal StromalCells (MSCs). MSCs are reported to find in various tissuesources in the body and can be isolated from them. MSCsare found in endometrium, adipose tissues, cartilage, dentalpulp, bone marrow, neuronal cells, placental tissue and skinand foreskin.

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

© Copyright iMedPub 3

Page 4: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

Characterization and Potency Analysisof MSCs Invitro:

Prospective isolation techniques for MSCsApplication of MSCs in clinal trials require proper isolation,

culturing and the differentiation into subsequent lineage.Since time of its discovery, a density gradient centrifugationmethod was implied to separate the mononuclear cells andred blood cells in BM unless it was potentiated by percollgradient separation and ficoll gradient separation [49]. Recentinvestigation has suggested that percoll density separationmethod is best in term of providing adequate yield of MSCswhereas as ficoll protocol is reported to cause extensive graftloss and less productive yield of MSCs [50]. The latestinnovation involves invention of automated, closed systemthat supervise the processing of MSCs from its collection to itsapplication in clinical therapy. This automated system has beenapplied extensively on broad scale for AT derived MSCs [51].

Some studies, on clinical scale, adapted direct platingapproach to separate the plastic adherent cells from non-adherent ones. In presence of optimal conditions ficoll, percolland whole BM was directly plated and compared for the samesamples to check efficiency of separation methods. OptimizedSeeding densities are considered as the most crucial factorwhile isolating and expanding MSCs. After attainingconfluence, cells were replenished after detaching and wereplated once again. Results suggests that plating of wholemarrow directly is more advantageous in term of cell growth,CFU-Fs number along with minimal manipulation. Thechallenge is to establish a compromise with both cell qualityand expansion along with cost issues in clinical scale expansionof MSCs [52].

One of conventional method for isolation of hMSCs involvesenzymatic digestion method by using collagenase enzyme forisolation of human adipose derived MSCs. After enzymaticdigestion and the preliminary adherence phase, nonadherentcells are discarded and the remaining adherent ones areappeared as fibroblastic cells. Now it has been replaced byexplant derived MSCs isolation process. In this method, smallpieces of human AT derived MSCs were placed in a cultureflask with suitable medium elements via liposuction and iskept being incubated. Homogeneity of the isolated cellpopulation, high yield and less time consumption make itadvantageous over conventional enzymatic degradationmethod [9,53].

Immunoisolation is another method for isolation of MSCsbefore culturing on basis of cell surface markers. It is actuallythe positive selection technique performed by usingmonoclonal antibodies cross reacting against MSCs markers[54]. Moreover, immunodepletion is another negativeselection method where MSCs population is supplemented bywashing after being labeled with the antibodies, directedagainst mostly the hematopoietic markers. It is actuallypurifying method of MSCs from hematopoietic contamination.At early stages of passaging, MSCs can be isolated and purified

from hematopoietic cells by immumodepletion ofhematopoietic cells [55]. Recently, more particular and pureMSCs populations can be isolated utilizing a combinatorialmethod of immunoisolation along with immunodepletionbased on the surface markers. The novel technique forisolation of non-cultured MSCs evolved includes affinity-basedmethods, microscale technologies as well as biophysicalproperties-based methods demanding no labelling of cellsfollowed by suspending under fluid conditions [10].

Culturing analysis of MSCsMSCs after being successfully isolated from different

sources, are cultured using condition media namely Dulbeccomodified Eagle media (DMEM) and RPMI (Roswell ParkMemorial Institute medium) with minimum essential mediumalpha (MEM α) mentioned to be one of the best medium forculturing of MSCs in vitro [56]. Dexamethasone is consideredto be leading factor in culture medium for inducing osteogenicdifferentiation while bone morphogenetic protein BMP2provide supportive properties when added in culture mediaalong with vitamin D3 and fibroblast growth factor [57]. Oneof the major challenge during culturing is to obtain sufficientnumber of cells as it has been observed that starting freshtissue culture available is in limited amount and on sub-culturing, these cells showed a decline in potency along withtime consumption problems and genetic instabilities thatresults ion MSCs aging and senescence. Currently anadvancement has been made by establishing an evaluation ofa new cell bank strategy that has resulted in MSCs cell lineswith stable genotypic and immunophenotypic characters [58].

Differentiation of MSCs into multiple lineageshMSCs have the ability to differentiate by invitro high

capacity assays into all three i.e. endodermal, ectodermal andmesodermal lineages that confirms their self- renewal andmultipotent ability [59].

Mesodermal lineages: The invitro differentiation potentialof MSCs into mesodermal lineages evidenced by performingqRT PCR (qualitative real time-PCR), flowcytometry andcytochemical analysis has been evaluated [7,60]. MSCsdifferentiation into adipocytes is induced by providingappropriate media supplementations, which stimulates theactivation of transcription factors and genes responsible foradipogenesis. Cultured MSCs in growth medium supplementedwith growth factors were analyzed for adipogenesis byexpression of adipocytes specific genes, lipoprotein lipasegenes (LPL), adipocyte protein ap2 and peroxisome proliferatoractivated receptor PPAR γ [61,62]. Induction of MSCs towardsadipogenesis is a two-phase process. During the determinationphase, cells are committed towards the preadipocytes, aremorphologically similar to fibroblasts and eventually are notdistinguished from MSCs precursors. At terminal phase, thepre-adipocytes form the mature adipocytes along with lipiddroplets and also express the adipocytes particular proteins.Overall, adipogenesis is an sequential ordered process withmultiple signaling pathways [63].

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

4 This article is available from: http://biochem-molbio.imedpub.com/inpress.php

Page 5: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

Osteogenic induction of MSCs is characterized by mineralsaggregation and an increase in enzyme activity of alkalinephosphatase in final stages of differentiation. Thesemineralized aggregates were found to be positive for von kossastaining and Alizarin Red [64]. The progression of osteogenesisinitiates with presence of osteoprogenitors which at earlystages, differentiate towards pre-osteocytes and then latterlyinto mature osteoblasts. Factors that are involved in theinduction of osteogenesis includes activation of runt-relatedtranscription factor-2 (Runx2) along with bone morphogenicprotein BMP 9, and many other extracellular signalingmolecules [65,66]. In the bone formation process, osteoblastsfirstly synthesize the bone matrix and then allows the mineraldeposition and bone remodeling [67]. Many researchers havestated that a relationship exist between osteogenesis andadipogenesis. Several signaling cascades are demonstrated tobe the positive regulators of osteogenesis and negativeregulators of adipogenesis. Signaling pathways like NEL-likeprotein, Hedge Hog and Wnt pathways are well demonstratedfor anti adipogenic and pro-osteogenic inductions in MSCs[68-70].

hMSCs also have the potential to differentiate towardschondrocytes lineages. In final step, pre-chondrocytesdifferentiate towards the mature chondrocytes and show anexpression of chondrogenic transcription factors like Sox5,Sox6 and Sox9 [71]. Along with transforming growth factorTGF-β1, insulin like growth factor-I and BMP-2 also contributedtowards chondrogenic differentiation of MSCs. In hMSCs,growth factor TGF-β1 interacts to Wnt β-catenin dependentpathways and hinders the osteoblast differentiation andinduces the chondrogenesis. Apart from the growth factors,parathyroid hormone related peptides and triiodothyronine T3also influenced the induction of chondrogenesis [72,73].

Ectodermal lineages: On providing neural induction culturemedia along with growth factors cocktail, like retinoic acid,brain derived neurotropic factors, isobutyl methyl xanthine(IBMX) and Glial cell lines derived neurotrophic growth factors(GNDF), expanded MSCs transdifferentiate towards neuronalcells exhibiting particular neuron specific phenotypes. Theinduced neuronal cells were found to express Nestin, neuronalnuclear protein NeuN, dopamine transporter protein DAT andtyrosine hydroxylase protein TH, neuronal potential ofdifferentiation [74]. Normally an enhancer or Zest Homologtype 2 controls the stem cell proliferation, maintenance andeventually differentiation into multiple lineages involvingneurons. It is noted that changes observed in concentration ofintracellular calcium plays a key role in induced neuronaldifferentiation of MSCs [75]. Gangliosides, a class ofglycosphingolipids, when interact with epidermal growthfactor receptor is reported to enhance the osteoblastformation (Figure 2).

Whereas decrease in gangliosides biosynthesis contributetowards inhibiting the neuronal differentiation [76]. hMSCswhen co-transfected with certain growth factors revealed tomaintain the neuronal differentiation, effective in the recoveryof hypoxia ischemic brain damage. hMSCs originated frommesodermal line, have potential to transdifferentiate towards

the neuronal cells which has revolutionized the regenerativecellular therapy in treatment of neurological disorders [77].

Figure 2 A schematic representation of differentiation ofMesenchymal Stromal Cells (MSCs) is shown. MSCs inpresence of certain growth factors differentiate intomultiple lineages by adapting different pathways. Growthfactors used are EGF, epidermal growth factors; PDGF,platelet derived growth factor; FGF, Fibroblast growth factorand VEGF, vascular endothelial growth factors. Severalpathways mediated that regulation of differentiation aresummarized: LPL-lipoprotein lipase gene; ap-2, adipocyteprotein; PPAR, peroxisome proliferator activated receptor;BMP-9, Bone morphogenetic protein; TGF, tumor growthfactor; GDNF, Glial cell lines derived neurotropic factors;IBMX, isobutyl methyl xanthine.

Endodermal lineages: Relatively, it was assumed thathepatocytes could be derived only via endodermal source andthe progenitor cells. Fortunately, MSCs have been exposed tohold the ability of transdifferentiation into pancareocytes andhepatocytes upon induction with the growth supplementedmedia [78]. Human BM derived MSCs were transdifferentiatedtowards hepatocytes via two steps phases: differentiationphase followed by the maturation phase. Hepatocytesdifferentiated cells displayed the liver specific transcriptionfactors like nuclear factor 4 α (HNF-4α), albumin and α-fetoprotein. Amongst these factors, HNF-4α is considered tobe critical transcription factor for functional and morphologicaland differentiation to hepatocytes. When human derivedMSCs were treated with HNF-4α, it enhanced thedifferentiation potential of cells and show an increase liverspecific expression markers [79]. In another research, it wasshown that on extracellular decellularized cell depositedmatrix with protein mixture of fibronectin, laminin andcollagen along with protein kinases upregulates the activationof hepatic markers [80]. Human dental stem cells are alsofound to be differentiated towards hepatocytes by induction ofthe growth factors and particularly melatonin when

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

© Copyright iMedPub 5

Page 6: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

transplanted during treatment of liver cirrhosis [81]. Moreover,the paracrine factors enhance the differentiation potential andmaturation of human BM-MSCs towards the pancreaticlineages without any kind of genetic manipulation and thustransplantation is being used for treating diabetes. Up till now,hMSCs derived from amnion, dental, adipose, umbilical cord,placental tissues and Wharton jelly tissues have effectivelydifferentiated towards insulin forming β-cells. Theseinvestigations have shown that hMSCs can differentiatetowards the endodermal lineages which can renovate theconventional drug related therapies towards the promising cellbased clinical therapies [82,83].

Regulation of differentiation: The commitment as well asdifferentiation of MSCs from precursors towards specificmature cells is a temporally and tightly controlled method thatinclude the activation of several transcription factors, growthfactors, extracellular matrix component and cytokines.Globally, the gene expression profiling with help of DNAmicroarray technique is rendered as useful tool foridentification of genes for commitment and differentiation ofstem cells in presence of inductive microenvironments [84].Several genes are found to have role in differentiation of threelineages of MSCs. One of major intracellular transcriptionfactor include Zinc finger protein ZNF-145 that acts as aupstream regulator of SOX9, thus favoring osteoblastic andadipocytic differentiation of MSCs [85]. The in vitrodifferentiation analysis of MSCs under proper conditions haveprogressed in identification of several factors crucial to stemcell commitment process. These factors include secretedmolecules along with their receptors like transforming growthfactor TGF-β, extracellular matrix components includingproteoglycans collagen and actin, interleukin-1 β, nebulette(NEBL) and micro RNAs. Functional investigation of the genesis mandatory to govern how they are tangled in progressionand commitment of stem cells towards different stages ofdifferentiation [86-89]. An interesting factor observed duringinvitro differentiation is the switching phenomenon ofphenotypes among fully differentiated MSC-derivedadipocytes, osteoblast and chondrocytes when enthused bystimulus from extra cellular environment.

During this trans differentiation, fully differentiated MSCsundergoes dedifferentiation both in function and morphologyand followed by next stimulation, these dedifferentiated cellsare re-differentiated into new phenotype. On the whole, it isreasonably concluded that these pre-committed precursorcells along with fully differentiated ones sustain theirmultipotentiality and that their plasticity during thisphenotypic switching phenomenon can be well-maintainedunder well defined, suitable microenvironmental conditions,observed in a tissue regeneration and repair mechanism [90].

Recent advancement in understanding mechanism of MSCsdifferentiation involves the members of Wnt family which playtheir role in osteogenesis of MSCs. Wnts belong to a family ofsecreted cysteine enriched glycoproteins that have beenimplied for instructing the stem cell maintenance,propagation, and differentiation in embryonic developmentalstages. Canonical Wnt signaling mechanism works as stem cell

mitogen by their role in stabilizing intracellular β-cateninreceptor and activating the transcription complex, resulting instimulated expression of cell cycle controlling genes, like cyclinD1, Myc and Msx1. When MSCs are triggered towards Wnt3a,which is a prototypical Wnt signal, under defined growth andmedium environment, remarkable increase in cell proliferationalong with decrease in the apoptotic process and Wntsmitogenic role in HSCs was observed. Anyhow, overlyexpressed Wnt3a has led to inhibitory effect on osteogenicdifferentiation invitro due to β-catenin arbitrated downregulation of transcription complex. Contrarily, a member ofnon-canonical Wnt pathway, Wnt5a has been observed toencourage osteogenesis differentiation in vitro. As Wnt3afavors MSC proliferation in early osteogenesis, it is concludedthat canonical Wnt signaling pathway has role in the initialstages of osteogenic commitment by enhancingosteoprecursors in stem cell compartment. On the other hand,non-canonical pathway of Wnt regulates the proliferation ofthese osteoprecursors to from functional mature osteoblasts.Taken into account, Wnt signaling pathway activation leads toa key role in inducing MSCs differentiation. In humans, themutations in LRP5, a Wnt co-receptor, primed towards thedefective bone development. Regain of functional mutationindicates the higher density and mass of bones whereas loss offunction results in the complete loss of the bone mass as wellas strength, indicating positive feedback effect of Wntsignaling at embryonic osteogenesis stage [12,91].

Characterization of MSCs In-vivoCurrent studies have provided appreciated insight for

understanding the identity and physiology of bone marrowresident MSCs via novel markers to detect and purify the MSCsenriched cell populations and then assay them in vivo. Aplatelet derived growth factor receptor, PDGFR, indicates apopulation of non-hematopoietic cells residing during boneformation process and provides a novel understanding ofmechanism involve in MSCs differentiation to adipocytes,reticular cells and osteoblasts, in vivo when transplanted to anrecipient [92]. Recently, Nestin, neural stem cell marker, isreported to express in undifferentiated nervous system stemcells. For the first time, expression of the Nestin was checkedin progenitor endothelial cells derived from adherent cultureof bone marrow cells. Both, BM derived mesenchymal cellsand Nestin, can be serially transplanted and used forneovascularization [93] (Figure 3).

MSCs anatomical analysis In vivoResearches have influentially explained self-renewing as

well as differentiation potential of BM resident mesenchymalstem cells. BM-derived MSCs are being investigated for clinicaltrials and the innate functions of these cells in BM have beendeliberated to know more about the therapeutic role andactivity. The location of MSCs has been problematic to traceand even tough to observe vigorously because cavity is notpossible to probe easily in vivo and also there is no specificmarker for MSC identification in vivo. However, using organculture system, immunophenotypic analysis, CFU-Fs assays

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

6 This article is available from: http://biochem-molbio.imedpub.com/inpress.php

Page 7: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

vivo and invitro labeled assays, MSCs existence in perivascularlocations has been supported. This organ culture system hasalso preserved the original niche of MSCs [94].

Figure 3 The proposed model for BM- derived MSCsbiological roles in vivo. BM derived stromal cells undergoesdifferentiation into adipocytes, reticular cells as shown withthe bold arrow heads and osteoblasts where they serve toensures and favors hematopoietic supportive environmentfor their development and are assumed to have their rolesin native turnover of MSCs into the bone marrow.

Perivascular location has been confirmed by theobservations concluding that MSCs are apparently founded inmany post-natal organs where their frequency in given tissue isbalanced with density of micro-vasculature. MSCs secretecertain angiogenic and trophic factors that contributes inneovasculogenesis along with proliferation and stabilization ofendothelial layer depending upon the tissue from which theyare derive. The stromal counterparts differentiate and may bemigrated towards abluminal sinusoids of marrow and made athree dimensional network which embedded in capillary bed[95].

Acknowledging the capacity of MSCs in forming bone andtheir assembly into functional bone marrow stroma onheterotopic sites in vivo, a group of researchers recognized thepopulation of osteoprogenitors in the outmost connectivetissues layer surrounding BM microvessels. Adventitialreticular cells also known as pericytes projects into sinusoidallumens with their fibroblastic extensions and are supposed tobe in vivo counter parts of CFU-Fs. These pericytes and MSCsshare comparable surface and intracellular expressionalpatterns of proteins which confirms their ontological relation.Moreover, tissue engineering constructs juxtaposed themesenchymal stromal cells along with endothelial cells and itserves to form long vascularized structures with MSCsnaturally exhibiting the pericytic phenotype as well asfunction. This localization proposes that MSCs may beconfidentially involved in angiogenesis, wound healing processand in interacting with blood borne components along with

differentiation in multiple lineages into smooth muscle sandmyocytes as well [14]. It must be worth to mention thatpericytes relates to cells that lies adjacent to capillaries as wellas post capillary venules but MSCs are isolated from walls ofarteries and veins. Additionally, because pericytes illustratesthe wide tissue distribution along microvascular beds and havemany functions including phagocytosis, regulating vascularintegrity and vessel stabilization. Therefore, regardless of beingperivascular, all MSCs cannot be stated as pericytes and viceversa [96].

Fibrocytes represents to be circulating BM-derived cells,found in peripheral blood, are phenotypically similar tomonocyte hybrids and fibroblasts articulating the surfacemarker CD34 and CD35, presence of chemotactic receptorsCCR3, CXCR4, CCR5 and CCR7, surface molecules likehistocompatibility complex class II MHC-I, MHC-II as well asMSC markers collagen I, collagen 111 and pro-collagen.Moreover, Fibrocytes have also been reported in areas wherethere was some kind of inflammation, cancer or fibrosis asresult of any injury. In these areas, they were supposed tomature towards myofibroblast that reside in tissues. Actually,fibrocytes are found to be phenotypically and functionallysimilar to BM-MSCs, so recognizing unique characteristicsbetween these two types of cells may help in understandingthe MSCs and fibrocytes homology [97].

Role of MSCs in maintenance of hematopoiesisin vivo

As stated, MSCs were initially isolated from bone marrowcompartment. Subsequently then, bone marrow derived MSCshave been studied most extensively and are considered to bekey managers of bone marrow physiology. Blood cells areconstantly produced principally in our bone marrow duringadulthood process. MSCs have longer been considered toserve as in vivo progenitors of some non-hematopoieticconstituents of BM that control hematopoiesis, likeadipocytes, osteoblasts and reticular cells of fibroblasts.Therefore, MSCs are expected to contribute in maintaininghomeostasis within hematopoietic compartment in vivo viathe regulatory roles of their matured progeny [98].

HSCs are considered to localized in the confined sites withinbone marrow microenvironment. This microenvironment isconstituted by soluble factors, surrounding cells and extracellular matrix proteins that latterly contribute in promotingmaintenance of HSCs [99]. Osteoblasts have been assumed tosignificantly contribute towards stem cell niches and promotesregulation of HSCs homeostasis via direct interactions fromcell to cell. Though this osteoblast-HSC niche are stilldebatable, it is believed that osteoblasts serve as vitalcomponent in microenvironment of BM either directly or bycertain secreting factors by the involvement of signalingpathway and play a key role in developmental stages ofhematopoiesis [100]. Additionally, BM stroma also have MSCsderived adipocytes which act to negatively regulateprogression of hematopoietic precursors. Molecularmechanism is not known. However several matters concerningthe in vivo precise developmental phases of MSCs during

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

© Copyright iMedPub 7

Page 8: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

differentiation, the pathways leading to the lineagecommitment in vivo as well as the ratio of adipocytes andosteoblast production in hematopoietic environment areneeded to be considered [101]. Osteoblasts are considered tobe significant as niches of HSCs and positively interact andregulate HSCs activity in BM. On the other hand, adipocyteshave a role in negative regulation of HSCs activity. CARCsrepresents CXCL-12 abundant reticular cells that lie adjacent toHSCs as shown in the figure, which have potential todifferentiate into adipocytes and osteocytes due their poorcharacterization and are considered to be originate from BM-derived MSCs [3].

Therapeutic Effects of MSCs Relatedto the Biological Properties

MSCs have the beneficial effects in the medical applicationsand it requires the complete understanding of their biologicalproperties. There are the four properties of MSCs that play animportant role in the medical applications. Firstly, their abilityto move to the site of inflammation when given intravenouslyduring the tissue injury. Secondly, their ability to differentiateinto different and various cell types. Thirdly, their ability toproduce various bio active molecules that are responsible forcuring the wounded cells or inhibit the inflammations duringthe course of injury or any foreign particle invasion. Lastly, thelack of immunogenicity and their ability to perform variousimmunomodulatory functions [13].

Capacity to migrate, differentiate and engraftat site of injury

After the regular administration of MSCs, they migrate tothe sites of injury or inflammation and provide the localbeneficial and functional effect at the local sites or tissues.MSCs move selectively to the sites of the injury irrespective tothe tissue under diverse pathological condition and isinvestigated by many studies. Scientists observed that in themice that has been challenged to bleomycin, theadministration of MSCs result in lowering the inflammation ofthe lungs tissues during injury by moving to the lungs andacquire a phenotype like the epithelium. We also noticed thatin the mice, the transplanted MSCs could move to the muscletissues that has been injured. Migration of the cells totallydepend on the signals that is produced by the cells at theinjured sites and it consists of the immune cells or growthfactors such as chemokines. Migration of MSCs may also beunder the control of these signals and chemokines. It has beenseen by the various studies that the migration of MSCs is alsodepend on the growth factors such as receptors of TyrosineKinases Growth Factors, Platelets Derived Growth Factors(PDGF), Insulin Growth Factors 1 (IGF1) and several otherchemokines [102]. In a study, it has been noticed that theMSCs when injected into mice that has been injured afterirradiation, MSCs differentiate into functional cells of thelungs, these functional cells are the epithelial or endothelialcells. Other studies demonstrated that when MSCs is given tothe model of the animals that has been injured by exposure of

bleomycin, it moves to the lungs and differentiate intoepithelial cells or pneumocytes and sustain the phenotype ofall the lungs cells such as myofibroblasts, fibroblasts andepithelial cells. Additionally, MSCs could also have the abilityto differentiated into the cells of the ectoderm. For example,MSCs when inserted into the Central Nervous Systems (CNS) ofthe infant mice, it acquired the functional, structural orphenotypic uniqueness of the astrocytes or the neurons. Byobserving all the facts that MSCs have the ability todifferentiate into specialized cells/tissues giving us theopportunity to use the MSCs for the disease treatment [103].In a model of mouse of Ischemic/Reperfusion (I/R) of thekidney, when MSCs were transplanted at the initial stage ofinjury, they differentiated into the renal tubular epithelium.The donor cells that are differentiated replaced the emptyspace that is left over by the dead cells and fill the gap, andhence maintaining the structural features or the tissue and cellrepair processes. On the other hand, the clinical aspects ofMSCs give us the knowledge to repair the damaged cells byspecific differentiation procedures [104].

Releasing the multiple bio active moleculesClinical applications of MSCs showed that it could release

multiple bio active molecules that has the functional orbeneficial effects on the cellular dynamics. They include manygrowth factors such as cytokines and chemokines.Administration of these bio active molecules showed thebeneficial effect for tissue and cell repair. For example, in MSCsConditioning Medium (MSC CM), MSCs move toward theinjury sites of the liver and inhibit the liver cells death andregenerate the damaged cells and provide a new departmentto treat the hepatic failure or liver cirrhosis. Many bio activemolecules including the cytokines also takes part in improvingthe functions of the damaged heart by preventing theapoptosis of the cardiomyocytes and restoring the contractilecapacity of the heart and also induce the productive andcurative angiogenesis of the damaged or infracted heart. Manyof the observed proteins that are secreted as soluble factorsare recognized as growth factors such as cytokines and thechemokines. All these molecules have the well-recognized antiapoptotic or the regenerating properties. These properties canbe direct or indirect or both. Direct are in producing oractivating the signaling intracellularly or indirect by allowinganother cell in the small environment to release functionallyactive bio molecules [105] (Figure 4).

Immunomodulatory aspects of MSCsMSCs have the distinctive immunologic or

immunomodulatory properties that permit their perseverancein the xenogeneic environment. Emerging techniques or datacomplete or enhance the immunomodulatory functions of theMSCs but the mechanisms of their fundamental immunogeniceffects are not yet fully understood by the researchers. Directconnection between the cells or the release of solubleimmune suppressive factors or molecules may have a majorrole in the treatment. MSCs have the ability to communicatewith a huge no. of immune cells, that consist of T lymphocytes,

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

8 This article is available from: http://biochem-molbio.imedpub.com/inpress.php

Page 9: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

B lymphocytes, natural killers and the dendritic cells, theimmunomodulatory role of MSCs are mentioned in the Table1. The immunogenic effects of MSCs are also observed in avariety of animals of the immune diseases as in rat model oftransplantation of heart, a long-term Allograft Acceptance isproduced by the donor derived MSCs. The immunomodulatoryproperties of MSCs could also played an important role in theimmune disorder, graft versus host diseases (GVHD) treatment[106].

Table 1 shows the immuno-modulatory functions ofMesenchymal Stem Cells (MSCs) effecting different types ofimmune cells.

Figure 4 Therapeutic effects of stromal cells with referenceto their effects on immunomodulation and differentiation iscompiled. On basis of bioactive molecule secretion,migration and engraftment, these stromal cells serve toreduce inflammation, increase tissue repair and is used inregenerative medicinal therapies. NK, Natural killer; igG,immunoglobulin G.

Applications of MSCS in gene therapyThe multipotent potential of MSCs and the ability of self-

renewal have shown the promising effect in great number ofanimal transplantation studies and represented thewidespread applications in cell and gene therapy. MSCs thatwere extended in vitro were responsible for differentiationinto cells of the residing tissue, reestablishes its normalfunction and also renovate the damaged tissue due to traumaor any other disorders. They are not only responsible to repairthe tissues of mesenchymal lineages, such as cartilage, bone,cardiomyocytes, and articular cartilage at knee joints, but alsorepair the cells of other embryonic layers, such as neurons andepithelia in the skin, liver, lung, intestine, spleen, and kidney.These applications express the effectiveness and the plasticityof these adult stem cells in multiple tissue repair anddevelopmental procedures and in the cell therapy applications.It is also notable that MSCs proves to be a principle carrier indelivering the genes into the cell of interest or it does notbring any immune reactivity in the host due to the

transplantation. Various strategies have been examined andused to transfer the exogenous DNA into MSCs and make themmore useful in tissue regeneration therapies and applications.

Table 1 Immune-modulatory functions of Mesenchymal StemCells (MSCs) effecting different types of immune cells. MSCsare shown to affect activity of immune cells including naturalkillers NK, dendritic cells, DCs; B lymphocytes and Tlymphocytes.

Immune Cells Effects of MSCs on immune Cells

T lymphocytes • Repress the proliferation of T cells caused bynonspecific mutagenic signals

• Enhance the functions of T cells

• Adjust and modify the secretion of cytokines of newand Effector

• T cells

B lymphocytes • Slow down the propagation of B lymphocyte

• Affect the chemical characteristics of B cells

• Suppress the terminal B-cells differentiation

NK • Affect the physical appearance of Natural Killer cells

• Inhibit the propagation, secretion of cytokines, andcytotoxic effects

DCs • Induce segregation, maturation and the functions ofdendritic cells

• Inhibit the migration of dendritic cells (DCs) andmaturation of antigen presenting cells

• Stimulate the mature DCs into the regulatoryproduction of DCs

Viral transduction, mostly using the adenovirus mediatedgene transfer, can produce stable cell clones with high efficacyand the low cell mortality, making it an accepted option ingene therapy applications. On the other hand, the safetyconcerns related with viral transduction have provoked us tolook for another non-viral gene delivery strategies. Forinstance, the brittle bone disease known as osteogenesisimperfecta is treated by using the MSCs that regenerate thebones in the infected individuals. However, Conventionaltransfection methods, including calcium phosphateprecipitation method, lipofection, and electroporation indelivering the plasmid DNA into MSCs have revealed the littlesuccess and result in less than 1% efficiency of transfectionmethod and high cell mortality, so these methods are notuseful and appropriate for producing adequate amount oftransfected cells for the gene delivery and transplantation[107].

Discussion and ConclusionIt is not easy to isolate the hMSCs but it has been seen that

MSC also have the ability of expansion without losing itsparticular characteristics. On the other hand, besides themesodermal lineages, MSCs have the ability to differentiateinto the endodermal or ectodermal lineages. Moreover, it has

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

© Copyright iMedPub 9

Page 10: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

been observed that hMSCs also show the immunomodulatoryproperties by releasing the receptors relevant to immunesystem and the cytokines that help in the modification ofimmune system of the host. These distinctive properties ofMSCs make them unique from the other stem cells and can beused in the future for cell replacement therapies. Many clinicalor preclinical studies demonstrated the usefulness of hMSCs inthe treatment of the chronic diseases, such asneurodegenerative diseases, cardiovascular diseases orautoimmune diseases, but on the large experimental or clinicalscale there are many questions that must be answer before weuse the hMSC. Firstly, the issues of safety of MSCs must beresolved, because when we administered the MSCs for thelong-term culturing possible side effects can be seen and itleads to the growth of tumor or metastasis. The second issue isof quality control, additional tests have to be performedbefore the direct transplantation of MSCs in vivo, these testsinclude the oncogenic tests, endotoxins assays or the cellviability. An optimum dose has to be decided depending onthe disease severity. The third issue is of the clinical productionof the MSCs as cells are needed in large number for the clinicalutilization of MSCs, in this the in vitro expansion plays a vitalrole. By this it is concluded that the MSCs obtained from theadult tissues are the favorite choice for the scientists and thecomplete understanding of the MSCs is necessary to regulatethe role of MSCs in the clinical applications.

Current discoveries in the MSCs engineering made it aprinciple source in the regenerative medicine for the futurecell therapies. The clinical experiments of MSCs is now on theway to victory after doing large no. of preclinical experiments,but the complete understanding of mechanism of MSCs is onthe preliminary state. The main focus of the future research ofMSCs is to isolate the particular markers that is specific forMSCs and also involved the understanding of mechanism ofdifferentiation to revolutionized the therapy of cellregeneration. Special attention should be put on the geneticsafety to lower the risk of oncogenesis during cellpreparations. However, there should be a dynamic researchthat focus on the bio banking to develop the novel protocolwithout disturbing the properties of MSCs in the future on thelarge scale.

References1. Manfrini M (2012) Human mesenchymal stem cells as a model

of study for new biomaterials in bone tissue regeneration.Università degli studi di ferrara.

2. Phinney DG (2012) Functional heterogeneity of mesenchymalstem cells: Implications for cell therapy. J cell biochem 113:2806-2812.

3. Nombela-Arrieta C, Ritz J, Silberstein LE (2011) The elusivenature and function of mesenchymal stem cells. Nat Rev MolCell Biol 12: 126-131.

4. Mitrano TI, Grob MS,Carrión F, Nova-Lamperti E, Luz PA, et al.(2010) Culture and characterization of mesenchymal stem cellsfrom human gingival tissue. J periodontol 81: 917-925.

5. Vater C, Kasten P, Stiehler M (2011) Culture media for thedifferentiation of mesenchymal stromal cells. Acta biomaterialia7: 463-477.

6. Goh TKP, Zhang ZY, Chen AKL, Reuveny S, Choolani M, et al.(2013) Microcarrier culture for efficient expansion andosteogenic differentiation of human fetal mesenchymal stemcells. Biores open access 2: 84-97.

7. Al-Nbaheen M, Ali D, Bouslimi A, Al-Jassir F, Megges M, et al.(2013) Human stromal (mesenchymal) stem cells from bonemarrow, adipose tissue and skin exhibit differences in molecularphenotype and differentiation potential. Stem Cell Rev Rep 9:32-43.

8. Uezumi A, Fukada S, Yamamoto N, Ikemoto-Uezumi M, NakataniM, et al. (2014) Identification and characterization ofPDGFRalpha+ mesenchymal progenitors in human skeletalmuscle. Cell Death Dis 5: e1186.

9. Ghorbani A, Jalali SA, Varedi M (2014) Isolation of adipose tissuemesenchymal stem cells without tissue destruction: A non-enzymatic method. Tissue and Cell 46: 54-58.

10. Diogo MM, Da Silva CL, Cabral JM (2014) Separationtechnologies for stem cell bioprocessing. Stem Cells and CellTherapy (1st edn) 157-181.

11. Huang J, Zhao L, Xing L, Chen D (2010) MicroRNA-204 regulatesRunx2 protein expression and mesenchymal progenitor celldifferentiation. Stem cells 28: 357-364.

12. Visweswaran M, Pohl S, Arfuso F, Newsholme P, Dilley R, et al.(2015) Multi-lineage differentiation of mesenchymal stem cells-To Wnt, or not Wnt. Int J Biochem Cell Biol 68: 139-147.

13. Salem HK, Thiemermann C (2010) Mesenchymal stromal cells:Current understanding and clinical status. Stem cells 28:585-596.

14. Corselli M, Chen CW, Crisan M, Lazzari L, Péault B (2010)Perivascular ancestors of adult multipotent stem cells.Arterioscler Thromb Vasc Biol 30: 1104-1109.

15. Augello A, Kurth TB, De Bari C (2010) Mesenchymal stem cells: aperspective from in vitro cultures to in vivo migration andniches. Eur Cell Mater 20: e33.

16. Tamama K, Kawasaki H, Wells A (2010) Epidermal growth factor(EGF) treatment on multipotential stromal cells (MSCs). Possibleenhancement of therapeutic potential of MSC. Biomed Res Int.

17. Cohnheim J (1867) Uber Entzundung und Eiterung [Aboutinflammation and sepsis]. Pathol Anat Physiol Klin Med 40: 79.

18. Maximow AA (1924)Relation of blood cells to connective tissuesand endothelium. Physiol rev 4: 533-563.

19. Friedenstein A, Piatetzky-Shapiro I, Petrakova K (1966)Osteogenesis in transplants of bone marrow cells. Development16: 381-390.

20. Friedenstein A, Chailakhyan RK, Latsinik NV, AF Panasyuk, Keiliss-Borok IV, (1974) Stromal cells responsible for transferring themicroenvironment of the hemopoietic tissues: Cloning in vitroand retransplantation in vivo. Transplant 17: 331-340.

21. Friedenstein AJ, Gorskaja J, Kulagina N (1976) Fibroblastprecursors in normal and irradiated mouse hematopoieticorgans. Exp hematol4: 267-274.

22. Owen M (1988) Marrow stromal stem cells. J Cell Sci 10: 63-76.

23. Caplan AI (1991) Mesenchymal stem cells. J Orthop Res 9:641-650.

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

10 This article is available from: http://biochem-molbio.imedpub.com/inpress.php

Page 11: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

24. Haynesworth S, Goshima J, Goldberg V, Caplan A (1992)Characterization of cells with osteogenic potential from humanmarrow. Bone 13: 81-88.

25. Goshima J, Goldberg VM, Caplan AI (1991) The osteogenicpotential of culture-expanded rat marrow mesenchymal cellsassayed in vivo in calcium phosphate ceramic blocks. Clin OrthopRelat Res 262: 298-311.

26. Prockop DJ (1997) Marrow stromal cells as stem cells fornonhematopoietic tissues. Science 276: 71-74.

27. Lazarus H, Haynesworth S, Gerson S, Rosenthal N, Caplan A(1995) Ex-vivo expansion and subsequent infusion of humanbone marrow-derived stromal progenitor cells (mesenchymalprogenitor cells): Implications for therapeutic use. Bone marrowtransplant 16: 557-564.

28. Koc ON, Gerson SL, Cooper BW, Dyhouse SM, Haynesworth SE,et al. (2000) Rapid hematopoietic recovery after coinfusion ofautologous-blood stem cells and culture-expanded marrowmesenchymal stem cells in advanced breast cancer patientsreceiving high-dose chemotherapy. J Clin Oncol18: 307.

29. Horwitz EM, Gordon PL, Koo WKK, Marx JC, Neel MD, et al.(2002) Isolated allogeneic bone marrow-derived mesenchymalcells engraft and stimulate growth in children with osteogenesisimperfecta: Implications for cell therapy of bone. Proc Natl AcadSci USA 99: 8932-8937.

30. Koc O, Day J, Nieder M, Gerson S, Lazarus H, et al. (2002)Allogeneic mesenchymal stem cell infusion for treatment ofmetachromatic leukodystrophy (MLD) and Hurler syndrome(MPS-IH). Bone marrow transplant 30: 215.

31. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F,et al. (2006) Minimal criteria for defining multipotentmesenchymal stromal cells The International Society for CellularTherapy position statement. Cytotherapy 8: 315-317.

32. Parekkadan B, Van Poll D, Suganuma K, Carter EA, Berthiaume F,et al. (2007) Mesenchymal stem cell-derived molecules reversefulminant hepatic failure. PloS one 2: e941.

33. Van Poll D, Parekkadan B, Cho CH, Berthiaume F, Nahmias Y, etal. (2008) Mesenchymal stem cell–derived molecules directlymodulate hepatocellular death and regeneration in vitro and invivo. Hepatol47: 1634-1643.

34. Phinney DG, Prockop DJ (2007) Concise review: Mesenchymalstem/multipotent stromal cells: The state of transdifferentiationand modes of tissue repair-current views. Stem cells 25:2896-2902.

35. Kubo H, Shimizu M, Taya Y, Kawamoto T, Michida M, et al. (2009)Identification of mesenchymal stem cell (MSC)-transcriptionfactors by microarray and knockdown analyses, and signaturemolecule-marked MSC in bone marrow byimmunohistochemistry. Genes to cells, 14: 407-424.

36. Pricola KL, Kuhn NZ, Haleem-Smith H, Song Y, Tuan RS (2009)Interleukin-6 maintains bone marrow-derived mesenchymalstem cell stemness by an ERK1/2-dependent mechanism. Jcellbiochem108: 577-588.

37. Von Bahr L, Batsis I, Moll G, Hägg M, Szakos A, et al. (2012)Analysis of tissues following mesenchymal stromal cell therapyin humans indicates limited long-term engraftment and noectopic tissue formation. Stem Cells 30: 1575-1578.

38. Gnecchi M, Danieli P, Malpasso G, Ciuffreda MC (2016) Paracrinemechanisms of mesenchymal stem cells in tissue repair.Methods Mol Biol 14: 123-146.

39. Kagami H, Agata H, Tojo A (2011) Bone marrow stromal cells(bone marrow-derived multipotent mesenchymal stromal cells)for bone tissue engineering: basic science to clinical translation.Int J Biochem Cell Biol 43: 286-289.

40. Zeddou M, Briquet A, Relic B, Josse C, Malaise MG, et al. (2010)The umbilical cord matrix is a better source of mesenchymalstem cells (MSC) than the umbilical cord blood. Cell Biol Int 34:693-701.

41. Peng J, Wang Y, Zhang L, Zhao B, Zhao Z, et al. (2011) Humanumbilical cord Wharton's jelly-derived mesenchymal stem cellsdifferentiate into a Schwann-cell phenotype and promoteneurite outgrowth in vitro. Brain Res Bull 84: 235-243.

42. Murphy S, Atala A (2013) Amniotic fluid and placentalmembranes: Unexpected sources of highly multipotent cells.Semin Reprod Med 31: 062-068.

43. Karaöz E, Doğan BN, Aksoy A, Gacar G, Akyüz S, et al. (2010)Isolation and in vitro characterisation of dental pulp stem cellsfrom natal teeth. Histochem Cell Biol 133: 95-112.

44. Vishnubalaji R, Al-Nbaheen M, Kadalmani B, Aldahmash A,Ramesh T (2012) Comparative investigation of thedifferentiation capability of bone-marrow-and adipose-derivedmesenchymal stem cells by qualitative and quantitative analysis.Cell Tissue Res 347: 419-427.

45. Fu WL, Zhou CY, Yu JK (2014) A new source of mesenchymalstem cells for articular cartilage repair: MSCs derived frommobilized peripheral blood share similar biologicalcharacteristics in vitro and chondrogenesis in vivo as MSCs frombone marrow in a rabbit model. Am J Sports Med 42: 592-601.

46. Borlongan CV, Kaneko Y, Maki M, Yu SJ, Ali M, et al. (2010)Menstrual blood cells display stem cell–like phenotypic markersand exert neuroprotection following transplantation inexperimental stroke. Stem Cells Dev 19: 439-452.

47. Rossignoli F, Caselli A, Grisendi G, Piccinno S, Burns JS, et al.(2013) Isolation, characterization, and transduction ofendometrial decidual tissue multipotent mesenchymal stromal/stem cells from menstrual blood. Biomed Res Int 2013: 1-14.

48. Huang HI, Chen SK, Ling QD, Chien CC, Liu HT, et al. (2010)Multilineage differentiation potential of fibroblast-like stromalcells derived from human skin. Tissue Eng Part A 16: 1491-1501.

49. Pösel C, Möller K, Fröhlich W, Schulz I, Boltze J, et al. (2012)Density gradient centrifugation compromises bone marrowmononuclear cell yield. PloS one 7: e50293.

50. Bourzac C, Smith LC, Vincent P, Beauchamp G, Lavoie JP, et al.(2010) Isolation of equine bone marrow-derived mesenchymalstem cells: A comparison between three protocols. Equine Vet J42: 519-527.

51. Fraser JK, Hicok KC, Shanahan R, Zhu M, Miller S, et al. (2014)The Celution® system: Automated processing of adipose-derivedregenerative cells in a functionally closed system. Adv WoundCare 3: 38-45.

52. Mareschi K, Rustichelli D, Calabrese R, Gunetti M, Sanavio F, etal. (2012) Multipotent mesenchymal stromal stem cellexpansion by plating whole bone marrow at a low cellulardensity: A more advantageous method for clinical use. StemCells Int 2012: 1-10.

53. Yoon JH, Roh EY, Shin S, Jung N, Song EY, et al. (2013)Comparison of explant-derived and enzymatic digestion-derivedMSCs and the growth factors from Wharton’s jelly. Biomed ResInt 2013: 1-8.

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

© Copyright iMedPub 11

Page 12: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

54. Rozemuller H, Prins HJ, Naaijkens B, Staal J, Bühring HJ, et al.(2010) Prospective isolation of mesenchymal stem cells frommultiple mammalian species using cross-reacting anti-humanmonoclonal antibodies. Stem Cells and Development 19:1911-1921.

55. Fan G, Wen L, Li M, Li C, Luo B, et al. (2011) Isolation of mousemesenchymal stem cells with normal ploidy from bone marrowsby reducing oxidative stress in combination with extracellularmatrix. BMC Cell Bio 12: 30.

56. Watchrarat K, Korchunjit W, Buranasinsup S, Taylor J, RitruechaiP, et al. (2017) MEM α promotes cell proliferation andexpression of bone marrow derived equine mesenchymal stemcell gene markers but depresses differentiation gene markers. JEquine Vet Sci 50: 8-14.

57. Mostafa NZ, Fitzsimmons R, Major PW, Adesida A, Jomha N, etal. (2012) Osteogenic differentiation of human mesenchymalstem cells cultured with dexamethasone, vitamin D3, basicfibroblast growth factor, and bone morphogenetic protein-2.Connect Tissue Res 53: 117-131.

58. Oliver-Vila I, Coca MI, Grau-Vorster M, Pujals-Fonts N, CaminalM, et al. (2016) Evaluation of a cell-banking strategy for theproduction of clinical grade mesenchymal stromal cells fromWharton's jelly. Cytotherapy 18: 25-35.

59. Russell KC, Phinney DG, Lacey MR, Barrilleaux BL, MeyertholenKE, et al. (2010) In vitro high-capacity assay to quantify theclonal heterogeneity in trilineage potential of mesenchymalstem cells reveals a complex hierarchy of lineage commitment.Stem cells 28: 788-798.

60. Gharibi B, Hughes FJ (2012) Effects of medium supplements onproliferation, differentiation potential, and in vitro expansion ofmesenchymal stem cells. Stem Cells Transl Med 1: 771-782.

61. Menssen A, Häupl T, Sittinger M, Delorme B, Charbord P, et al.(2011) Differential gene expression profiling of human bonemarrow-derived mesenchymal stem cells during adipogenicdevelopment. BMC genomics 12: 461.

62. Muruganandan S, Parlee SD, Rourke JL, Ernst MC, Goralski KB, etal. (2011) Chemerin, a novel peroxisome proliferator-activatedreceptor γ (PPARγ) target gene that promotes mesenchymalstem cell adipogenesis. J Biol Chem 286: 23982-23995.

63. Muruganandan S, Roman A, Sinal C (2009) Adipocytedifferentiation of bone marrow-derived mesenchymal stemcells: Cross talk with the osteoblastogenic program. Cell Mol LifeSci 66: 236-253.

64. Wang W, Olson D, Cheng B, Guo X, Wang K (2012) Sanguisdraconis resin stimulates osteoblast alkaline phosphataseactivity and mineralization in MC3T3-E1 cells. J Ethnopharmacol142: 168-174.

65. Zhang Y, Su J, Yu J, Bu X, Ren T, et al. (2011) An essential role ofdiscoidin domain receptor 2 (DDR2) in osteoblast differentiationand chondrocyte maturation via modulation of Runx2activation. J Bone Miner Res 26: 604-617.

66. Xu D, Zhao Y, Wang J, He J, Weng Y, et al. (2012) p38 and ERK1/2are involved in BMP9-induced osteogenic differentiation ofC3H10T1/2 mesenchymal stem cells. BMB reports 45: 247-252.

67. Birmingham E, Niebur G, McHugh P (2012) Osteogenicdifferentiation of mesenchymal stem cells is regulated byosteocyte and osteoblast cells in a simplified bone niche. EurCell Mater 23: 13-27.

68. Wang J, Liao J, Zhang F, Song D, Lu M, et al. (2017) NEL-LikeMolecule-1 (Nell1) Is Regulated by bone morphogenetic protein9 (BMP9) and potentiates BMP9-induced osteogenicdifferentiation at the expense of adipogenesis in mesenchymalstem cells. Cell Physiol Biochem 41: 484-500.

69. Oliveira F, Bellesini L, Defino H, Herrero CS, Beloti M, et al.(2012) Hedgehog signaling and osteoblast gene expression areregulated by purmorphamine in human mesenchymal stemcells. J Cell Biochem 113: 204-208.

70. Cawthorn WP, Bree AJ, Yao Y, Du B, Hemati N, et al. (2012) Wnt6,Wnt10a and Wnt10b inhibit adipogenesis and stimulateosteoblastogenesis through a β-catenin-dependent mechanism.Bone 50: 477-489.

71. Liu CF, Lefebvre V (2015) The transcription factors SOX9 andSOX5/SOX6 cooperate genome-wide through super-enhancersto drive chondrogenesis. Nucleic Acids Res 43: 8183-8203.

72. Karl A (2013) Role of BMP and TGFß signalling in the terminaldifferentiation in the in vitro chondrogenesis of mesenchymalstem cells.

73. Statman LY (2012) Mechanosensitive β-Catenin signalingmodulates mesenchymal stem cell chondrogenesis, University ofCalifornia, San Francisco.

74. Chen L, He DM, Zhang Y (2009) The differentiation of humanplacenta-derived mesenchymal stem cells into dopaminergiccells in vitro. Cell Mol Biol Lett 14: 528-536.

75. Yu YL, Chou RH, Chen LT, Shyu WC, Hsieh SC, et al. (2011) EZH2regulates neuronal differentiation of mesenchymal stem cellsthrough PIP5K1C-dependent calcium signaling. J Biol Chem 286:9657-9667.

76. Yang HJ, Jung KY, Kwak DH, Lee SH, Ryu JS, et al. (2011) Inhibitionof ganglioside GD1a synthesis suppresses the differentiation ofhuman mesenchymal stem cells into osteoblasts. Dev GrowthDiffer 53: 323-332.

77. Andres RH, Horie N, Slikker W, Keren-Gill H, Zhan K, et al. (2011)Human neural stem cells enhance structural plasticity andaxonal transport in the ischaemic brain. Brain 134: 1777-1789.

78. Ayatollahi M, Soleimani M, Tabei SZ, Kabir Salmani M (2011)Hepatogenic differentiation of mesenchymal stem cells inducedby insulin like growth factor-I. World J Stem Cells 3: 113-121.

79. Bishi DK, Mathapati S, Venugopal JR, Guhathakurta S, CherianKM, et al. (2013) Trans-differentiation of human mesenchymalstem cells generates functional hepatospheres on poly (L-lacticacid)-co-poly (ε-caprolactone)/collagen nanofibrous scaffolds. JMater Chem B 1: 3972-3984.

80. He H, Liu X, Peng L, Gao Z, Ye Y, et al. (2013) Promotion ofhepatic differentiation of bone marrow mesenchymal stem cellson decellularized cell-deposited extracellular matrix. BioMedRes Int 2013: 406871.

81. Cho YA, Noh K, Jue SS, Lee SY, Kim EC (2015) Melatoninpromotes hepatic differentiation of human dental pulp stemcells: clinical implications for the prevention of liver fibrosis. JPineal Res 58: 127-135.

82. Prabakar KR, Domínguez-Bendala J, Molano RD, Pileggi AS,Ricordi VC (2012) Generation of Glucose-Responsive, Insulin-Producing Cells From Human Umbilical Cord Blood-DerivedMesenchymal Stem Cells. Cell Transplant 21: 1321-1339.

83. Cao X, Han ZB, Zhao H, Liu Q (2014) Transplantation ofmesenchymal stem cells recruits trophic macrophages to induce

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

12 This article is available from: http://biochem-molbio.imedpub.com/inpress.php

Page 13: Multipotent Potential of Human Adult Mesenchymal Stem Cells · lymphoid stromal cells and recommended MSCs to be the predecessors of BM connective tissues. Lately in 1980s, Maureen

pancreatic beta cell regeneration in diabetic mice. Int J BiochemCell Biol 53: 372-379.

84. Hsiao STF, Asgari A, Lokmic Z, Sinclair R, Dusting GJ (2011)Comparative analysis of paracrine factor expression in humanadult mesenchymal stem cells derived from bone marrow,adipose, and dermal tissue. Stem Cells Develop 21: 2189-2203.

85. Liu TM, Guo XM, Tan HS, Hui JH, Lim B (2011) Zinc-finger protein145, acting as an upstream regulator of SOX9, improves thedifferentiation potential of human mesenchymal stem cells forcartilage regeneration and repair. Arthritis Rheumatol 63:2711-2720.

86. Sonomoto K, Yamaoka K, Oshita K, Fukuyo S, Zhang X (2012)Interleukin-1β induces differentiation of human mesenchymalstem cells into osteoblasts via the Wnt-5a/receptor tyrosinekinase–like orphan receptor 2 pathway. Arthritis Rheumatol 64:3355-3363.

87. Aino M, Nishida E, Fujieda Y, Orimoto A, Mitani A, et al. (2014)Isolation and characterization of the human immatureosteoblast culture system from the alveolar bones of ageddonors for bone regeneration therapy. Expert Opin Biol Ther 14:1731-1744.

88. Ragni E, Montemurro T, Montelatici E, Lavazza C, Viganò M, etal. (2013) Differential microRNA signature of humanmesenchymal stem cells from different sources reveals an“environmental-niche memory” for bone marrow stem cells. ExpCell Res 319: 1562-1574.

89. Park JS, Chu JS, Tsou AD, Diop RZ, Wang TA, et al. (2011) Theeffect of matrix stiffness on the differentiation of mesenchymalstem cells in response to TGF-β. Biomaterials 32: 3921-3930.

90. Ullah M, Sittinger M, Ringe J (2014) Transdifferentiation ofadipogenically differentiated cells into osteogenically orchondrogenically differentiated cells: Phenotype switching viadedifferentiation. Int J Biochem Cell Biol 46: 124-137.

91. Baksh D, Boland GM, Tuan RS (2007) Cross-talk between Wntsignaling pathways in human mesenchymal stem cells leads tofunctional antagonism during osteogenic differentiation. J CellBiochem 101: 1109-1124.

92. Caplan AI, Correa D (2011) PDGF in bone formation andregeneration: new insights into a novel mechanism involvingMSCs. J Orthop Res 29: 1795-1803.

93. Suzuki S, Namiki J, Shibata S, Mastuzaki Y, Okano H (2010) Theneural stem/progenitor cell marker nestin is expressed inproliferative endothelial cells, but not in mature vasculature. JHistochem. Cytochem 58: 721-730.

94. Choi MY, Kim HI, Yang YI, Kim JT, Jang SH, et al. (2012) Theisolation and in situ identification of MSCs residing in looseconnective tissues using a niche-preserving organ culturesystem. Biomaterials 33: 4469-4479.

95. Hoch AI, Binder BY, Genetos DC, Leach JK (2012) Differentiation-dependent secretion of proangiogenic factors by mesenchymalstem cells. PloS one 7: e35579.

96. Corselli M, Chen CW, Sun B, Yap S, Rubin JP, (2011) The tunicaadventitia of human arteries and veins as a source ofmesenchymal stem cells. Stem Cells Dev 21: 1299-1308.

97. Fu Xy, Zhang Dw, Li Yd, Zhao Pw, Tang YQ (2015) Curcumintreatment suppresses CCR7 expression and the differentiationand migration of human circulating fibrocytes. Cell PhysiolBiochem 35: 489-498.

98. Pontikoglou C, Deschaseaux F, Sensebé L, Papadaki HA (2011)Bone marrow mesenchymal stem cells: biological properties andtheir role in hematopoiesis and hematopoietic stem celltransplantation. Stem Cell Rev 7: 569-589.

99. Nombela-Arrieta G, Pivarnik B, Winkel KJ, Canty B, Harley JE, etal. (2013) Quantitative imaging of hematopoietic stem andprogenitor cell localization and hypoxic status in the bonemarrow microenvironment. Nat Cell Biol 15: 533-543.

100. Rankin EB, Wu C, Khatri R, Wilson TL, Andersen R, et al. (2012)The HIF signaling pathway in osteoblasts directly modulateserythropoiesis through the production of EPO. Cell 149: 63-74.

101. Bethel M, Chitteti BR, Srour EF, Kacena MA (2013) The changingbalance between osteoblastogenesis and adipogenesis in agingand its impact on hematopoiesis. Curr Osteoporos Rep 11:99-106.

102. Burdon TJ, Paul A, Noiseux N, Prakash S, Shum-Tim D (2011)Bone marrow stem cell derived paracrine factors forregenerative medicine: current perspectives and therapeuticpotential. Bone Marrow Res 2011: 207326.

103. Bhaskar B, Mekala NK, Baadhe RR, Rao PS (2014) Role ofsignaling pathways in mesenchymal stem cell differentiation.Curr Stem Cell Res Ther 9: 508-512.

104. Zou X, Gu D, Xing X, Cheng Z, Gong D, et al. (2016) Humanmesenchymal stromal cell-derived extracellular vesicles alleviaterenal ischemic reperfusion injury and enhance angiogenesis inrats, Am J Transl Res 8: 4289.

105. Timmers L, Lim SK, Hoefer IE, Arslan F, Lai RC, et al. (2011)Human mesenchymal stem cell-conditioned medium improvescardiac function following myocardial infarction. Stem Cell Res 6:206-214.

106. De Witte SF, Franquesa M, Baan CC, Hoogduijn MJ (2015)Toward development of iMesenchymal stem cells forimmunomodulatory therapy. Front Immunol 6: 648.

107. Uchibori R, Tsukahara T, Ohmine K, Ozawa K (2014) Cancer genetherapy using mesenchymal stem cells. Int J Hematol Res 99:377-382.

Biochemistry & Molecular Biology Journal

ISSN 2471-8084 Vol.4 No.2:16

2018

© Copyright iMedPub 13