gingiva as a source of stem cells with therapeutic potential

71
1 1 Gingiva as a Source of Stem Cells with Therapeutic Potential Fournier BPJ 1,2,3 , Larjava H 1 , Häkkinen L 1 . 1 Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada; 2 Paris Diderot University, Dental School, Rotschild Hospital, AP-HP, Paris, 3 UMRS872, Team 5, Molecular Oral Physiopathology, CRC Les Cordeliers, Paris, 75006, INSERM UMRS872, Pierre et Marie Curie University, Paris Descartes University, France. Dr. Benjamin Fournier, Paris Diderot University, UMRS872, Team 5, Molecular Oral Physiopathology, 15 Rue de l'Ecole de Médecine, Paris 75006, France; Email: [email protected]; Tel: 331-4427-5587; Fax: 331-4427-5591. Dr. Hannu Larjava, University of British Columbia, Faculty of Dentistry, Department of Oral Biological and Medical Sciences, 2199 Wesbrook Mall, Vancouver, BC, V6T 1Z3 Canada; Email: [email protected]; Tel: 604-822-6822; Fax: 604-822-3562 Dr. Lari Häkkinen, University of British Columbia, Faculty of Dentistry, Department of Oral Biological and Medical Sciences, 2199 Wesbrook Mall, Vancouver, BC, V6T 1Z3 Canada; Email: [email protected]; Tel: 604-822-0096; Fax: 604-822-3562 Running Title: Therapeutic Potential of Gingival Stem Cells Corresponding Author: Dr. Lari Häkkinen, University of British Columbia, Faculty of Dentistry, Department of Oral Biological and Medical Sciences, 2199 Wesbrook Mall, Vancouver, BC, V6T 1Z3 Canada; Email: [email protected]; Tel: 604-822-0096. Page 1 of 72 Stem Cells and Development Gingiva as a Source of Stem Cells with Therapeutic Potential (doi: 10.1089/scd.2013.0015) This article has been peer-reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.

Upload: lari

Post on 10-Dec-2016

214 views

Category:

Documents


2 download

TRANSCRIPT

1

1

Gingiva as a Source of Stem Cells with Therapeutic Potential

Fournier BPJ1,2,3

, Larjava H1, Häkkinen L

1.

1Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British

Columbia, Vancouver, Canada; 2

Paris Diderot University, Dental School, Rotschild Hospital,

AP-HP, Paris, 3UMRS872, Team 5, Molecular Oral Physiopathology, CRC Les Cordeliers, Paris,

75006, INSERM UMRS872, Pierre et Marie Curie University, Paris Descartes University,

France.

Dr. Benjamin Fournier, Paris Diderot University, UMRS872, Team 5, Molecular Oral

Physiopathology, 15 Rue de l'Ecole de Médecine, Paris 75006, France; Email:

[email protected]; Tel: 331-4427-5587; Fax: 331-4427-5591.

Dr. Hannu Larjava, University of British Columbia, Faculty of Dentistry, Department of Oral

Biological and Medical Sciences, 2199 Wesbrook Mall, Vancouver, BC, V6T 1Z3 Canada;

Email: [email protected]; Tel: 604-822-6822; Fax: 604-822-3562

Dr. Lari Häkkinen, University of British Columbia, Faculty of Dentistry, Department of Oral

Biological and Medical Sciences, 2199 Wesbrook Mall, Vancouver, BC, V6T 1Z3 Canada;

Email: [email protected]; Tel: 604-822-0096; Fax: 604-822-3562

Running Title: Therapeutic Potential of Gingival Stem Cells

Corresponding Author: Dr. Lari Häkkinen, University of British Columbia, Faculty of

Dentistry, Department of Oral Biological and Medical Sciences, 2199 Wesbrook Mall,

Vancouver, BC, V6T 1Z3 Canada; Email: [email protected]; Tel: 604-822-0096.

Page 1 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

2

2

Abstract

Postnatal connective tissues contain phenotypically heterogeneous cells populations that include

distinct fibroblast subpopulations, pericytes, myofibroblasts, fibrocytes and tissue-specific

mesenchymal stem cells (MSCs). These cells play key roles in tissue development, maintenance

and repair and contribute to various pathologies. Depending on the origin of tissue, connective

tissue cells, including MSCs, have different phenotypes. Understanding the identity and specific

functions of these distinct tissue-specific cell populations may allow researchers to develop better

treatment modalities for tissue regeneration and find novel approaches to prevent pathological

conditions. Interestingly, MSCs from adult oral mucosal gingiva possess distinct characteristics,

including neural crest origin, multipotent differentiation capacity, fetal-like phenotype and potent

immunomodulatory properties. These characteristics and an easy, relatively noninvasive access to

gingival tissue and fast tissue regeneration after tissue biopsy make gingiva an attractive target

for cell isolation for therapeutic purposes aiming to promote tissue regeneration and fast, scar-

free wound healing. The purpose of this review is to discuss the identity, phenotypical

heterogeneity and function of gingival MSCs and summarize what is currently known about their

properties, role in scar-free healing and their future therapeutic potential.

Page 2 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

3

3

Introduction

Accumulating evidence has shown that the cells that reside in postnatal connective tissue are not

all alike but compose different phenotypic subpopulations with distinct properties and functions.

Among connective tissue cells and within fibroblast cultures established from connective tissues,

several tissue-specific mesenchymal stem cell (MSC) populations have been identified that

represent a distinct cell type from fibroblasts and other connective tissue cells (Figure 1) (1-4).

Depending on the origin of tissue, MSCs have different phenotypes (4-9). Understanding the

identity and specific functions of MSCs from various tissues may allow researches to develop

better treatment modalities for tissue regeneration and find novel approaches to prevent

pathological conditions. Interestingly, human oral mucosal cells have a distinct neural crest origin,

high regeneration potential, share some traits with fetal cells and show potent immunoregulatory

properties suggesting that they may possess specific therapeutic potential. The purpose of this

review is to discuss the identity, phenotype and function of MSCs from oral mucosal gingiva and

specifically summarize what is currently known about their therapeutic potential.

Strategies to isolate and identify MSCs from oral tissues

Practically all postnatal connective tissues, including bone marrow, skin, oral mucosa, dental and

periodontal tissues and various parenchymal tissues, contain stem or progenitor cell populations

(5). Nomenclature to describe these cells has been variable in the literature, but most commonly

they have been termed mesenchymal stromal cells (MSCs) or mesenchymal stem cells (also

MSCs) (5). Recently, a term connective tissue stem cells (CTSs) was also introduced to

specifically describe corresponding postnatal orofacial cells (8). For simplicity, we will use the

term MSCs to describe these cells in this review.

Page 3 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

4

4

By definition, MSCs are described as plastic-adherent fibroblast-like cells that are clonogenic and

have the capacity for self-renewal, i.e. making copies of themselves (daughter cells) that have the

same potential as the parent cell (Figure 1). Furthermore, they express a certain set of cell surface

markers, are multipotent and when appropriately stimulated differentiate into mature type of cells

that make up different connective tissues (2-4). These may include ability for osteo-, chondro-

and adipodifferentiation (2-5, 9). In addition, some MSCs may have a capacity for differentiation

into endothelial-like, muscle (myodifferentiation) or neuronal-like cells (neuronal and glial

differentiation). Conclusive evidence of functional neuronal and glial differentiation is still,

however, lacking in vivo (5, 10).

Adult MSCs can be propagated from tissue biopsies of connective tissue with identical protocols

as has been used to culture fibroblasts as they are both plastic adherent and grow in the same cell

culture medium (Figure 1). Therefore, MSCs are likely present in every standard fibroblast

culture although they appear to encompass only a small subpopulation, often representing less

than 1% of the total cell population. MSCs have also a similar morphology with fibroblasts and

share most of the molecular markers that have been traditionally used to identify fibroblasts

(Figure 1 and Table 1). Therefore, there has been some uncertainty about the relationship and

identity of these two cell types (5, 11, 12). The situation has been complicated further by

discoveries that in general connective tissue cells between different individuals and sexes, in

different connective tissues within the same individual or within the given connective tissue are

not alike but consist of subpopulations of adherent, fibroblast-like cells that show considerable

phenotypic heterogeneity (Figure 2) and share similar growth requirements in vitro (12-14).

Page 4 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

5

5

These fibroblast-like cells can include phenotypically distinct fibroblast subsets, pericytes,

fibrocytes, myofibroblasts and MSCs (Figures 2) (6, 11, 12). In any case, currently used

characterization of MSCs rely on properties present in cultured cells as identification of these

cells in their natural tissue niche is challenging largely due to lack of specific markers.

There are basically two strategies to isolate MSCs from adult tissues for cell culture that have

also been widely used to isolate MSCs from oral tissues, including gingiva. In the first approach,

cells are grown from a tissue biopsy as for establishing cultures of plastic-adherent fibroblast. To

this end, cells are either allowed to grow out of a connective tissue biopsy placed onto a plastic

cell culture dish (explant culture method) or they are first enzymatically released from the

connective tissue before seeding into the culture plate (enzymatic digestion method). From these

cultures, MSC colonies can be isolated or enriched by the limited dilution or colony-forming

unit-fibroblastic cells (CFU-F) techniques or by separating them based on expression of MSC

signature molecules using fluorescence-activated cell sorting (FACS) or other separation

techniques (4, 5). In the limited dilution or CFU-F techniques, cells are seeded in a very low

density to a culture dish allowing colonies to form from a single cell. In the enzymatic digestion

method, cells released from tissue can also be directly seeded at low density to isolate stem cells

without a preceding cell culture step. The largest colonies that develop are then considered

candidate MSC populations as they represent cells with the most growth (i.e. self-renewal)

potential. The colony-forming efficiency varies depending on origin of the cells but remains

usually below 10%, although colony CFU-F efficiency of up to about 23 % has been described

for gingival cells (15). However, not all of these colonies comply with the MSC criteria and,

therefore, only a proportion of the colonies contain in fact MSCs. To verify MSC characteristics

Page 5 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

6

6

of the colonies, presence or absence of certain key MSC molecular markers are screened and

their differentiation potential to various mesenchymal lineages is analyzed (6). When compared

to the parental cultures (i.e. mixed cultures containing mostly fibroblasts), the limited dilution-

derived or CFU-F colonies usually have an increased capacity for multipotent differentiation.

Experiments employing repeated enrichments from existing colonies have provided further

evidence that these populations can contain cells that maintain self-renewal and differentiation

potential over several repeated selections (4, 5). However, it has turned out that not all colonies

(i.e. putative stem cell populations) are alike, thus potentially reflecting phenotypic heterogeneity

in the cells of the parental culture or tissue. Furthermore, cells within the given clonal colony

have different characteristics possible reflecting phenotypic modulation, differentiation of the

cells during the colony formation and/or asymmetric cell division (5, 16). Therefore, cell

populations derived from limited dilution or CFU-F methods are phenotypically heterogeneous

and appear to enrich MSCs, although at a variable extent. However, with the current methods it is

not possible to identify which cells in these populations represent the true stem cells. In fact, it is

unclear whether the stem cell properties are characteristic of a specific “stem cell” per se or if

they are a more general characteristic of the given cell colony containing different cell

subpopulations that include subsets of fibroblasts or other fibroblast-like cells and MSCs. It is

possible that the communication between fibroblast subpopulations and MSCs is important for

the MSC phenotype and this can only be achieved in mixed cell populations. Therefore, the niche

created by the nearby fibroblasts, whether in culture or in tissue, maybe critical to determine the

MSC characteristics, fate and functions (17, 18). This niche may be distinct in different tissues

and cells cultures derived from them ultimately leading to a distinct MSC phenotype.

Page 6 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

7

7

In the second approach that have been used to isolate MSCs from oral and other tissues, MSCs

are sorted from the parental fibroblast cultures or from a cell population enzymatically released

directly from the connective tissue biopsy based on a panel of preselected cell surface markers.

This prospective isolation procedure relies on the utility of the selected markers to accurately

identify MSCs (2-5, 9). The challenge is that most of the markers that have been used are usually

present also in other connective tissue cells, including fibroblasts, pericytes, fibrocytes and

myofibroblasts (Table 1) (5, 9, 19). To at least partially overcome this problem, several different

markers should be analyzed in a single cell at the same time, which is technically challenging.

Furthermore, the presence or absence of the surface markers do not necessarily correlate with the

phenotype of these cells in vivo (9).

Recently, another approach was developed to identify the so-called skin-derived precursor cells

(SKPs) from human and mouse skin. In this approach, cells released from skin connective tissue

by enzymatic digestion are grown in suspension in a defined serum-free medium containing EGF

and FGF-2 that supports neural stem cell growth (20, 21). Unlike in the two approaches described

above where MSCs are grown in the presence of serum as plastic adherent cells, this technique

results to selective clustering of cells to form floating neurospheres. Cells in these neurospheres

express nestin, a neural crest cell marker, and vimentin and fibronectin, fibroblast markers. Cells

from the neurospheres can be subcultured and induced to differentiate into neuronal, glial, and

mesenchymal phenotypes that can include muscle cells, osteoblasts, chondrocytes and adipocytes

(20, 22, 23). The best-characterized origin for SKPs in adult tissues is neural crest-derived dermal

papilla and dermal sheath cells of the hair follicles of facial skin, but they also appear to have a

non-neural crest, extra-follicular niche because they can also be isolated from neonatal foreskin

Page 7 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

8

8

that is exclusively of mesodermal origin (21, 24, 25). It remains to be confirmed, however,

whether the SKP-like cells isolated from foreskin are, in fact, of non-neural crest origin and

identical to neural crest-derived SKPs (24). In any case, lineage-tracing experiments in

genetically engineered mice have also supported findings that SKPs can derive from both neural

crest and non-neural crest origins (24, 26). In the oral cavity, SKP-like cells have been isolated

from the nerve endings in rat hard palatal rugae, human and rat periodontal ligament, dental pulp

and buccal mucosa. Similar to SKPs, they express neural crest markers, form neurospheres and

differentiate efficiently into neuronal-like and mesenchymal phenotypes in culture (27-32). To

date, no information about the presence or absence of similar cells in oral mucosal gingiva has

been reported.

Properties of oral mucosal and dental MSC

Oral mucosal and dental cells originate from the neural crest and have distinct properties

Lineage-tracing and other experiments suggest that most oral mucosal connective tissues and

tooth-associated tissues originate embryonically from the cranial neural crest (24). In contrast, in

many other connective tissues, including trunk skin, cells derive from the mesoderm. In

craniofacial skin, however, the dermal papilla cells of hair follicles are also from the neural crest

while stromal cells originate from the mesoderm (27, 33-36). Likely due to the distinct

developmental origin, cells present in the oral mucosal connective tissue have a distinct gene

expression profile compared to skin cells (36, 37). Therefore, it is possible that the embryonic

origin and patterning may underlie some of the phenotypic differences between oral mucosal and

skin cells contributing to the different functional outcomes, including wound-healing responses

(see below) (Table 2). However, whether dermal papilla cells from craniofacial skin and oral

Page 8 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

9

9

mucosal cells, both likely derived from the neural crest, share similar cell phenotype remains to

be shown.

Oral mucosal cells have also specific functional properties. For instance, heterotopic

recombination studies have shown that oral mucosal connective tissues or cells differently

regulate epithelial phenotype as compared to corresponding skin connective tissue or cells (37-

39). As compared to skin, tissue response to injury is also distinct in oral mucosa (see below). In

addition, resulting from the different developmental origin and distinct epithelial-mesenchymal

interactions during organogenesis, oral mucosa harbors salivary glands and teeth while skin has

hair follicles and sweat glands (40). Hair follicles in skin and teeth in oral cavity are formed by

mechanisms involving similar epithelial-mesenchymal interactions. Therefore, it may not be

surprising that while hair follicles in skin are important niches for adult tissue-specific MSCs also

tissues associated with teeth, including gingiva, periodontal ligament and dental apical papilla,

pulp and follicle contain such stem cells (41, 42). In general, MSCs from oral tissues have similar

multipotent differentiation potential as MSCs isolated from other connective tissues, including

skin and bone marrow (41). However, they also have an increased capacity for neurogenic-like

differentiation (27) and may produce tissues specific for teeth and periodontal tissues when

placed in an extra-oral niche (43, 44). Thus, possibly due to their neural crest origin, oral cells

appear preprogrammed for neurogenic and dental and periodontal tissue formation. Interestingly,

cultured vibrissae dermal papilla-derived MSCs have also neurogenic potential and can undergo

odontogenic differentiation when placed in the microenvironment of a developing tooth (44, 45).

Therefore, also other cells may have similar differentiation capacity when exposed to an

appropriate niche. Similar to oral cells, vibrissae dermal papilla-derived MSCs derive from the

Page 9 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

10

10

neural crest (24, 27). It remains to be shown, however, whether the dual neurogenic-like and

odontogenic differentiation property is restricted only to cells that have the neural crest origin.

Characteristics of oral mucosal and dental MSCs

Using the above mentioned culture methods, MSC-like cells have been isolated in the oral cavity

from periodontal ligament (PDLSCs), periodontal granulation tissue, dental pulp (DPSCs,

DPPSCs and SHEDs), dental follicle (DFPCs), apical papilla of a developing tooth (SCAPs),

buccal oral mucosal lamina propria (OMLP-PCs) and gingiva (GMSCs) (Table 3) (46-76). In

general, these cells express various neural crest markers and are, like many neural crest-derived

stem cells, highly multipotent (44) and may possess capacity for both mesenchymal

(cementogenic, odontogenic, osteogenic, chondrogenic, adipogenic) and neural-like (neuronal

and glial) differentiation in vitro (Table 3) (8, 27, 44, 48, 49, 55). As mentioned above, dental-

derived MSCs can also maintain their dental-specific differentiation potential when placed to a

heterotopic tissue niche in vivo. For instance, when PDLSCs, DPSCs or SHEDs were mixed with

a hydroxyapatite carrier and placed subcutaneously in an immunocompromised mouse, they

formed periodontal ligament and tooth root cementum-like (PDLSCs) or dentin and dental pulp-

like (DPSCs and SHEDs) tissue (43, 50, 52). In addition, in their natural niche (i.e. in a tooth

extraction socket) in minipigs in vitro-generated and appropriately induced constructs of

allogenic or autogenic dental pulp MSCs surrounded by sheets of dental follicle MSCs generated

a functional mineralized, dentin-like tooth root and a periodontal ligament (73). On the other

hand, the non-dental oral mucosal lamina propria progenitor cells (OMLP-PCs) have an excellent

neurogenic potential, in addition to osteogenic and chondrogenic capacity and are strongly

immunosuppressive. This last property depends on a mechanism that is at least in part distinct

Page 10 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

11

11

from other adult MSCs being HLA II-independent and mediated by release of

immunosuppressive mediators by the cells (56) mimicking fetal-derived MSCs (77). This is

interesting keeping in mind that human gingival cells also share properties with fetal skin cells

(see below). The more primitive or fetal-like phenotype of oral cells is further supported by

findings showing that DPPSCs cultured from the pulp of human third molars share characteristics

with pluripotent embryonic and induced pluripotent stem cells (iPSCs), including similar gene

expression profile, ability to form embryonic bodies and differentiation into meso-, endo- and

ectoderm-like layers in vitro and teratoma formation in vivo (78).

Gingiva – a distinct tissue of oral mucosa with a fetal-like fast and scarless wound healing

response

While dental tissues are a promising source of MSCs with neural crest properties, their isolation

requires usually an invasive procedure involving tooth extraction. Therefore, oral mucosal soft

tissues may provide a more attractive and practical source of such MSCs. Although in general

oral tissues share similar developmental origin (27) there are several important structural and

functional differences even between various anatomical locations in the oral mucosa. For

example, while oral cavity is covered with a stratified squamous epithelium, in certain locations

the epithelium is non-keratinized (the so-called oral lining mucosa found in cheeks, vestibules

and floor of the mouth) while in others it is keratinized (the so-called masticatory mucosa of

gingiva and hard palate) or is composed of a specialized gustatory epithelium having areas of

both keratinized and non-keratinized epithelium and specialized taste buds (surface of the tongue)

(79, 80). The oral lining and masticatory mucosa cover approximately 60% and 25% of the total

oral mucosa surface area, respectively (81). Recombination experiments using separated

Page 11 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

12

12

epithelial and connective tissues or fibroblasts and epithelial cells from keratinized and non-

keratinized parts of the mucosa have shown that the epithelial phenotypes are determined by

signals from the underlying connective tissue (82-84). Thus, the connective tissue cells and niche

produced by these cells are functionally distinct also in different areas of the oral mucosa.

Accordingly, the structure of the oral mucosal connective tissue also shows regional variations.

The lining mucosa contains an elastic, loosely organized connective tissue with a submucosa

while the masticatory mucosa of gingiva and hard palate contain a dense connective tissue that

directly attaches to the underlying bone (80).

Among oral mucosal tissues, gingiva has further specific features. It is easily accessible for tissue

biopsy and subsequent wound healing is fast and complete with very little morbidity.

Anatomically, gingiva surrounds the teeth and is directly attached to the underlying bone

(alveolar bone) and to the teeth (Figure 3). Its primary function is to provide a dynamic seal

between the teeth and oral mucosa and participate in immune defense (85). Functionally, gingiva

is highly dynamic characterized by one of the fastest tissue turnover rates in body (80, 86).

Unlike in lining oral mucosa or most parts of the skin, the epithelium covering the gingiva forms

long rete ridges that protrude into the connective tissue. The keratinized gingival epithelium

shows also distinct expression of certain cytokeratins (CK), including CK6/CK16, that are not

normally present in skin or oral lining mucosal epithelium (87-89), but that are strongly induced

in these tissues during wound healing (90).

As implied above, gingival tissues and cells seem to be primed for fast tissue regeneration that

leads to a functional advantage over other adult tissues. For instance, wound healing in the

Page 12 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

13

13

keratinized masticatory mucosa of palate and gingiva results to a significantly reduced clinical

and histological scar formation as compared to similar skin wounds (91, 92). In addition, wound-

healing speed is accelerated in both gingiva and other parts of the oral mucosa compared to skin

(91-96). The fast wound healing in oral mucosa is associated with a mild and short-lived

inflammatory response, including a reduced recruitment of neutrophils, mast cells, macrophages

and T-cells as compared to skin wounds (91-93, 96). Furthermore, the level of

immunomodulatory and anti-fibrotic TGF-3 relative to pro-fibrotic TGF-1 is elevated (97, 98).

Although presence of saliva containing several factors that promote wound closure may in part

contribute to the fast and scarless wound healing response also other mechanisms are likely

involved (99). For instance, mechanical signals transmitted from the ECM to the cells are

powerful modulators of cell function (100). Direct adhesion of gingiva to the underlying bone

and tooth surface, low abundance of elastin and organization of collagen into densely packed

collagen fibers (so-called supra-alveolar fiber apparatus) that are thicker than in the oral lining

mucosa (101-104) results to significantly higher tensile strength and stiffness of the gingiva

compared to the oral lining mucosa (105). Interestingly, mechanosignaling and increased ECM

stiffness promotes cell migration (100, 106) and regulates MSC function and fate (107)

potentially promoting wound healing in gingiva. Both the mild inflammatory response and high

relative expression of TGF-3 in oral mucosal wounds are similar to fetal skin where wounds

close fast and do not form scars (93, 108, 109). Furthermore, distinct composition of gingival

ECM and phenotype of gingival fibroblasts, resembling fetal skin cells (13, 93), may provide a

significant functional advantage in wound healing (see below). Whether the preferential wound

healing response in gingiva depends specifically on increased abundance and/or distinct function

of MSC has not been studied in detail. However, several key features of gingival cells in general

Page 13 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

14

14

may contribute to this property.

Distinct phenotype of gingival cells

Developmental origin

Based on the neural crest origin of other oral and dental tissues (27, 33, 36), it has also been

assumed that gingival connective tissue cells have the same origin, but experimental evidence has

been provided only recently. For instance, subpopulations of human oral mucosal fibroblasts

have been reported to express neural crest markers nestin, III-tubulin and GFAP in culture (74).

Most recently, a study utilizing neural-crest specific Wnt-1 reporter mice showed that most cells

in the mouse gingiva in vivo are of neural crest origin but that a smaller proportion of cells are

likely of mesodermal origin. When gingival cells from these mice were propagated in culture as

plastic-adherent cells, about 90% of MSCs generated from these cultures were positive for the

neural crest reporter gene while the rest were from mesoderm (75). Therefore, it appears that

gingival MSCs are heterogeneous and the specific developmental programs may result to

different properties of the MSC subpopulations.

Extracellular matrix niche

In addition to developmental origin, the local tissue niche that the cells reside in determines their

phenotype and function (110). There is some evidence that gingival fibroblasts and likely also

MSCs secrete and interact with a specific ECM niche. For instance, there are quantitative and

qualitative differences between molecules that compose the fibrillar and non-fibrillar ECM,

including type III collagen, sulphated glycosaminoglycans, heparan, chondroitin, dermatan and

keratan sulphate, hyaluronan, and tenascin-C in adult gingiva and skin (92, 111-113). These

Page 14 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

15

15

differences in the ECM composition are likely due to a distinct phenotype of gingival cells as

cultured gingival fibroblasts produce larger dermatan sulphate proteoglycans and higher levels of

hyaluronan than dermal fibroblasts (93, 114). Furthermore, while elastin is abundantly expressed

by skin fibroblasts, gingival connective tissue shows elastin accumulation only in association

with blood vessels and gingival fibroblasts express very low levels of this molecule in vitro (101-

103). In spite of this, many of the microfibrillar components of elastic fiber system, including

fibulin-5 and fibrillin-1 and -2, are expressed in the gingiva (115-117). Interestingly, while skin

elastic fibers show age related changes, in gingiva they remain morphologically relatively stable

(118). A survey based on analysis of 44 genes by RT-PCR in cultured fibroblasts showed that,

although gingival and skin cells expressed comparable levels of major ECM proteins, including

type I collagen and fibronectin, the expression of certain ECM proteins, such as periostin,

osteopontin and chains for type III and V collagen, was significantly lower in gingival cells. In

addition, expression of several cell adhesion-related molecules and integrin-family ECM

receptors for collagens, laminin, fibronectin and teanscin-C was significantly different from skin

fibroblasts (119-121). Furthermore, gingival cells showed reduced adhesion and spreading on

collagen and fibronectin as compared to skin cells, suggesting that the repertoire and function of

ECM receptors in gingival cells is distinct (120).

The cellular niche contains also growth factors that are bound and stored in the ECM in an

inactive form, including TGF-1, vascular-endothelial growth factor (VEGF) and fibroblast

growth factor-2 (FGF-2) (122). Very little is known about the abundance of these molecules in

gingival MSC niche in vivo or in vitro. However, gingival fibroblasts produce distinct amounts of

ECM molecules, including various proteoglycans, glycosaminoglycans, fibrillin-1 and tenascin-C,

Page 15 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

16

16

that bind growth factors. Furthermore, gingival cells show differences to skin cells with respect to

levels of various growth factors produced (see below). Thus, gingival cells produce a specific

ECM niche that likely harbors distinct levels of growth factors.

In addition to ECM composition, mechanical signals mediated from the cellular

microenvironment to the cells are important modulators of cell phenotype and differentiation fate.

For instance, cells sense the stiffness of the ECM via integrin receptors that impacts their gene

expression and directs their lineage specifications and differentiations (123). Interestingly, elastic

modulus measurements from pig tissues indicate that stiffness in gingiva is higher (elastic

modulus of about 20 kPa) as compared to oral lining mucosa (about 5-10 kPa) or skin (about 5-

10 kPa) (124-125).

Taken together, the gingival ECM niche (composition, organization and mechanical properties)

and cell interactions with it appear distinct and may determine and maintain gingival MSC

phenotype and function (Table 2).

Cell heterogeneity within gingiva

As mentioned above, cells within a given connective tissue in vivo and in vitro, including gingiva,

are phenotypically heterogeneous. In gingiva, part of this may be explained by heterogeneous

neural crest and mesodermal origin of the cells (75), but other mechanisms may also be involved.

Structurally, gingival tissue is not uniform further suggesting that functionally distinct fibroblast

population and possibly MSCs also exist in different anatomical locations. For instance, the

molecular composition of the connective tissue in the interdental papilla (part of the gingiva that

Page 16 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

17

17

resides between teeth) and in the attached gingiva (facial or oral side of the gingiva) is different

(126). Other studies have shown that fibroblasts isolated from the connective tissue papilla of

gingiva (region of connective tissue residing between long epithelia rete ridges typical for gingiva)

show higher saturation density, smaller cell size and higher production of the migration

stimulating factor (MSF) as compared to the cells isolated from the deeper reticular connective

tissue (13). MSF is found only in fetal but not in adult skin fibroblasts (127, 128). While both

adult human gingiva and fetal skin are characterized by scarless wound healing the importance of

MSF in this context and specific molecular characteristics of MSF has not been studied in detail

(93).

In addition to phenotypic differences between different anatomical locations, gingival cells show

functional heterogeneity between individuals (129) and within the given cell culture originating

from the same biopsy (Figure 2). For instance, early studies by Hassell and Stanek (130)

demonstrated that distinct subpopulations of gingival fibroblasts can be isolated from the same

biopsy, in this case from the interdental papilla. These cell subpopulations showed differences in

doubling times, cell size and collagen and glycosaminoglycan synthesis. These properties were

stable during cell culture indicating that they were an inherent property of the cells and not

affected by the cell culture (130). Other studies have shown further phenotypic heterogeneity in

cell morphology, total protein synthesis, production of different collagens (type I, III, V) and

fibronectin, proliferation, response to cytokines, growth factors, C1q component of complement

and PGE2 and in enzymatic activities (131-140). Furthermore, gingival cells can be separated

into different subpopulations based on the expression of stem cell factor (SCF) and its receptor c-

Kit (CD117) (141), although gingival cells have also been reported to be negative for CD117 (49).

Page 17 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

18

18

Sorting of a gingival cell population based on the expression of the high affinity receptor for C1q

yields a subpopulation with short doubling time and an increased protein synthesis, particularly

type III and type V collagen. These properties are stable over various passages in culture and

reminiscent of cells that are activated for a wound healing response (134). Further studies showed

that these cell subpopulations had also a different property to interact with either the C1q

collagen-like or globular domains resulting to differential activation of intracellular signaling

pathways (140). Whether any of the above cell subpopulations present in the cultures or tissues

represent MSCs or if they result from distinct mesodermal or neural crest origin has not been

explored in detail.

Gingival cell heterogeneity and phenotype may also have importance for gingival pathology.

Drug-induced gingival fibromatosis affects about 10 to 50 % of individuals taking systemic

medications, including certain Ca2+

-channel blockers, anticonvulsants and immunosuppressants,

and results to an expansive enlargement of the gingival tissue due to excessive accumulation of

ECM produced by gingival fibroblasts. This condition appears to be limited to the gingiva

suggesting that only gingival cells are susceptible to these drugs (142). Interestingly, cultured

gingival fibroblasts are heterogeneous in their response to these drugs and only some

subpopulations are activated to accumulate excessive ECM. In addition, fibroblasts isolated from

the gingival overgrowth tissue maintain their high ECM producing phenotype in culture,

suggesting that use of the medication has selectively favored the expansion of the responding cell

population in vivo (143-145). The role of MSCs in the pathogenesis of gingival overgrowth has

not been addressed. However, MSCs can be isolated from gingival overgrowth tissue and have

comparable expression of cell surface markers, self-renewal, osteo-, adipo- and chondrogenic

Page 18 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

19

19

differentiation capacity and immunomodulatory properties as MSCs from normal gingiva (146).

Gingival multipotent MSCs

The fetal-like cell phenotype with potency to support tissue regeneration suggests that gingiva

may contain an abundant source of MSC that promote tissue regeneration. Consequently, cells

that comply with the classical MSC definitions have been isolated from human gingiva from

established gingival fibroblast explant cultures (49) as well as from cells released from the tissue

by enzymatic digestion (58, 59, 146, 147). In general, gingival stem cells express cell surface

markers typical to MSCs (Table 1), show self-renewal capacity and variable colony forming

efficiency of about 3-23% in culture. Comparisons of gingival MSCs with corresponding cells

from other sources have shown variable results. In one study, gingival MSCs were similar to

MSCs found in bone marrow (59) while in another study they expressed different cell surface

markers and differentiation capabilities (49). A more consistent finding seems to be that gingival

MSC proliferate faster in culture and maintain their characteristics, karyotype and telomerase

activity better after higher passages than bone marrow MSCs (59, 60, 146).

Perivascular tissue – a niche for gingival MSCs?

In general, the tissue niche for MSCs has remained undefined but it seems likely that this niche is

different in various tissues. Maybe because of this difference, MSCs from various origins also

possess distinct phenotypes (5). The tissue location of the gingival MSCs is elusive.

Immunostaining of cells expressing the classical MSC/embryonic stem cell markers STRO-1,

OCT-4 and SSEA-4 have indicated presence of a low number of single or double

immunopositive cells in subepithelial human gingival connective tissue (49, 59, 146). In addition,

Page 19 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

20

20

undefined chord-like structures in the subepithelial gingival connective tissue show positive

staining for p75NTR, a neural crest cell marker, along with positive staining for the embryonic

stem cell markers OCT4 and SOX-2 (74). However, as the cell surface markers used to identify

MSCs are not specific for these cells the exact tissue niche for putative MSCs still remains to be

clarified.

Certain populations of MSCs have been located previously in perivascular tissue in many tissues.

Therefore, it is likely that abundant vascular network may also provide such a MSC niche in

gingiva. In addition to the tooth and hair follicle-related niches described above, earlier studies

had suggested that some of the progenitor cells involved in wound healing and tissue

regeneration in various tissues, including skin, periodontal ligament and gingiva, reside at the

perivascular location (100, 148-151). This has been subsequently confirmed in several reports

showing that perivascular cells resembling “undifferentiated fibroblasts” can migrate into the

wound and produce collagen to repair the wound (99, 152-154). More recent findings from

immunolocalization and cell culture studies have also provided evidence that some cells that

reside in perivascular tissues have characteristics of MSCs (150). Association with vasculature

would provide an ideal tissue niche for MSCs as blood vessels form a vast network expanding

throughout the entire body allowing fast access for progenitor cells into various tissue sites and

into the circulation. Perivascular location can harbor various cells, including circulating cells

passing through the endothelium into the connective tissue, fibroblasts, vascular smooth muscle

cells and pericytes. In particular pericytes, which surround endothelial cells in capillaries and

microvessels throughout the body, possesses MSC properties. These cells can be identified by

expression of NG2, CD146, -SMA and PDGFRβ (151). Cultured pericytes share, however,

Page 20 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

21

21

several key properties with fibroblasts, including similar cell morphology, expression of several

of the cell markers (Table 1) and like fibroblasts they can be propagated as plastic adherent cells

with standard fibroblast growth medium. Pericytes have also similar properties with MSCs as

they express many of the same surface markers (Table 1) and are able to differentiate into

myofibers, adipocytes, chondroblasts and osteoblasts (151, 155, 156). Thus, pericytes maybe

present in fibroblast and/or MSC cultures. Whether pericytes can be considered as MSCs and

whether some of the MSC properties of fibroblast cultures or MSCs purified from them can be

attributed to pericytes needs further experimental clarification. In any case, perivascular tissue

appears to provide a distinct niche for MSCs or MSC-like cells throughout the body, including

periodontal tissues (155).

Therapeutic potential of gingival MSCs

Widely accepted key properties of MSCs to promote tissue regeneration include multipotent

differentiation, immunomodulation and stimulation of vasculogenesis and epithelialization (157).

While differentiation of MSCs into cells of the target tissue may occur in vivo, studies have

indicated that only very low numbers survive and engraft in the tissue for long term. Therefore, it

is likely that paracrine factors released by these cells play a key role in their potency to promote

regeneration by the host cells (69). In the following paragraphs we will review the regenerative

potential of gingival cells based on evidence about the above properties.

Differentiation capacity

Similar to other MSCs, gingival MSCs selected by CFU-F or limited dilution techniques also

display the classical mesodermal tri-lineage osteo-, adipo- and chondrogenic differentiation in

Page 21 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

22

22

vitro (15, 49, 59, 146). In addition, differentiation of gingival MSCs into endodermal-like and

neural-like cells has been described but these findings need further verification (Table 3) (59, 74,

75). Interestingly, when the two gingival MSC populations derived from the neural crest and

mesoderm were compared, the neural crest-derived cells showed a better potential for

chondrogenic differentiation and expressed higher levels of neural/neural-crest markers –tubulin,

nestin and neurofilament M under neurogenic differentiation conditions in vitro (75). When

compared to periodontal ligament MSCs, gingival cells show significantly reduced doubling time

and better cloning efficiency, but periodontal ligament MSCs appear to have better differentiation

potential (15).

Studies of differentiation of gingival MSCs in vivo have also shown promising but variable

results. In general, when gingival MSCs are implanted subcutaneously, without prior incubation

in any differentiation-inducing medium, they produce connective tissue reminiscent of gingiva

(158). When cultured in an osteogenic induction medium and incorporated into a

hydroxyapatite/tricalcium phosphate carrier, fibrin gel, or other carriers and then transplanted

subcutaneously into immunocompromised mice, gingival MSCs have been reported to either

produce (15) or not to produce (49, 59) mineralized tissue. When gingival MSCs grown inside a

collagen carrier were treated as above and transplanted into critical size bone defects created in

rat mandibles or calvaria they regenerated the bone defects (Table 3) (58). In a recent study,

STRO-1+ immunoselected gingival MSCs in deproteinized bovine cancelleous bone or collagen

carrier was applied into experimental periodontal defects in mini pigs. Results showed that MSCs

containing scaffolds induced regeneration of periodontal tissues including bone, cementum and

periodontal ligament (159. However, recent findings have shown that inflammatory environment

Page 22 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

23

23

in vivo or treatment with inflammatory cytokines in vitro may suppress the differentiation

capacity of gingival MSCs (15, 158, 160).

Interestingly, similar to MSC, also unselected gingival fibroblast cultures can be induced to

differentiate into osteo-, adipo- and chondrogenic lineages as well as to myofibroblasts in vitro

(60, 72, 74). In addition, differentiation into an endodermal and neurogenic pathway has been

described but needs further confirmation (74) (Table 3). It is somewhat surprising that non-

selected gingival fibroblast cultures may have similar properties as MSCs. There is some

evidence, however, that unselected gingival fibroblast cultures may loose their differentiation

potential during successive passaging in culture faster than gingival MSCs generated by CFU-F

method (49). This preliminary finding needs further verification but implies that the proportion of

MSCs in the fibroblast cultures decreases over time. Therefore, in addition to gingival MSCs,

non-selected gingival fibroblast cultures may possess potential for therapeutic applications, but

they may need to be used as primary cultures or in their early passages. Interestingly, in a recent

study, human gingival or lining mucosal cell cultures were established using a fibroblast explant

culture technique (76). Although no clonal selection was performed, about 90% of the cells

expressed typical MSC markers and about 40-70 % pluripotency genes Oct4, Sox2 and Nanog. In

addition, when subjected to appropriate induction media cultures showed osteogenic, adipogenic

and chondrogenic differentiation in vitro. Cells were then cultured in fibrin gels and transplanted

between skin and calvaria in immunocompromized mice which resulted to mineralized tissue

formation that contained proteins typical to bone, cementum and dentin. Thus, cultures generated

from gingiva and oral lining mucosa using typical fibroblast culture protocols maybe enriched

with MSC-like cells providing an attractive and practical source of cells for therapeutic

Page 23 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

24

24

applications.

Immunomodulation

In the oral cavity, gingiva protects underlying tissues from effects of constant build-up of

microbial biofilm at the tooth-gingiva interface. Therefore, gingival cells are adapted to the first

line of defense against the biofilm and interact with microbes, microbial-derived molecules and

immune cells. Gingival stromal cells communicate with immune cells directly by establishing

cell-cell contacts or indirectly by secreting and responding to inflammatory mediators from

immune cells (157, 161). Like other MSCs, gingival MSCs have potent immunosuppressive

properties that may contribute to their regenerative and other functions (75, 162). For instance,

when injected into circulation they suppress development of allogeneic rejection, contact

hypersensitivity, experimental colitis and collagen-induced arthritis in mouse models (75, 146,

162, 163). Cell culture findings have indicated that similar to bone marrow MSCs, gingival cells

can suppress T-cell proliferation and activation, and modulate function of innate immune cells,

including dendritic cells, macrophages and mast cells. Important for tissue regeneration, gingival

cells are able to induce repolarization of inflammatory M1 macrophages to anti-inflammatory,

reparative M2 macrophages (75, 146, 164, 165). Interestingly, the neural crest-derived gingival

MSC population appears to have better immunosuppressive properties than the mesodermal-

derived subpopulation as the former were more potent to induce apoptosis of activated T-cell in

vitro and to ameliorate experimental inflammatory colitis in a mouse model in vivo (75). These

anti-inflammatory and immunomodulatory functions can likely be attributed to cellular cross-talk

of gingival cells with immune cells and release of soluble factors, including indoleamine 2.3-

dioxygenase (IDO), IL-10 and PGE2, which is similar to MSCs from different tissues (162, 165).

Page 24 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

25

25

In addition to studies about immunomodulatory effects of gingival MSC, there is vast literature

about immunoregulation by non-selected gingival fibroblast populations that may also be

relevant in general to the utility of gingival cells in therapy. Most of this research has focused on

immunomodulatory effects of gingival cells on immune defense against acute or chronic

microbial infection and have provided evidence that gingival cells in general, like gingival MSC,

have immunosuppressive properties in vitro as they reduce peripheral blood monocyte

proliferation (72, 166, 167). In gingival fibroblasts, also similar to MSCs, IFN-stimulates type

II HLA expression but the cells are not able to stimulate alloreactive T-cells (3, 168-170).

Subsequent studies have shown that IFN- induces secretion of IDO in gingival fibroblasts

similar to MSCs and that this is at least in part responsible for suppressing T cell proliferation

(171, 172). In this context it is interesting to note that transplanted gingival fibroblasts have a

better potential than skin fibroblasts to down regulate inflammation in vivo (173, 174).

Furthermore, oral mucosal wounds show a milder and shorter inflammatory reaction in response

to wounding as compared to skin wounds (91, 92, 96). This may depends, at least partially, on

immunomodulatory functions of gingival cells as it is well established that MSCs and fibroblasts

take part in regulation of inflammation during wound healing (100). This is further supported by

findings from a mouse model where systemic infusion of gingival cells resulted to accelerated

skin wound repair with rapid re-epithelialization and angiogenesis. This effect was associated

with a local inflammatory response at the wound site (164). However, it was not completely clear

whether these effects were achieved with clonally selected MSC populations or by non-selected

parental cultures (i.e. composed mostly of fibroblasts).

Page 25 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

26

26

Stimulation of angiogenesis and epithelialization

Role of gingival cells in promoting angiogenesis has not been investigated extensively. However,

autologous gingival connective tissue grafts and free gingival grafts (composed of both gingival

epithelium and connective tissue) have been used extensively in periodontal therapy and they

revascularize readily in vivo as determined by histological methods (175-177). In addition,

gingival cell cultures are able to produce pro-angiogenic factors, including VEGF, CXCL12, IL8,

MCP-1 and CXCR1 and promote endothelial cell growth in vitro (178-180). However, whether

these properties depend on fibroblasts or MSCs present in the mixed populations of cells in these

experiments needs further clarification. In addition, whether these cells have comparable ability

to promote also vasculogenesis in regenerative therapy as has been reported for other MSCs (16)

remains to be shown.

As discussed above, gingival connective tissue or cells define the epithelial phenotype in the oral

cavity. This has been evidenced by grafting procedures where gingival connective tissue induces

formation of a keratinized epithelium that corresponds to the one that is present in the normal

gingiva (82). In addition, gingival connective tissue cells appear to have a distinct property to

regulate epithelialization also in extra-oral sites. For instance, bioengineered tracheal constructs

containing autologous gingival fibroblasts induced a functional tracheal epithelial regeneration in

a rat model (181, 182). This property was similar to adipose-derived MSCs but differed from skin

or nasal fibroblasts that were not able to induce an appropriate morphological and functional

epithelial phenotype (182). Whether these properties depend on the fibroblasts and/or MSCs

needs further clarification.

Page 26 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

27

27

Use of gingival cells in future cell therapy

Use of gingival cells to promote tissue regeneration

While stem cell therapies are still largely at an experimental stage, particularly skin fibroblasts in

various applications, including living skin tissue grafts or manufactured skin substitutes, have

been used to treat dermal deficiencies and to promote skin wound healing for decades. In general,

these modalities have improved treatment outcomes and benefited the patients by improving their

quality of life, but they have not been able to consistently regenerate the skin architecture and

function (183-186). However, animal studies have indicated that use of MSCs from various

sources, including bone marrow, adipose tissue or skin, have a better potential in skin and

possibly also in other tissues (187). Most recent findings have indicated that novel technologies,

including injection of allogeneic fibroblasts or MSCs or use of iPSCs generated from skin cells,

may also be beneficial to treat certain conditions (188, 189).

As discussed above, gingival cell cultures possess abundantly MSCs with distinct capacity for

multilineage differentiation and that promote re-epithelialization, support angiogenesis and

modulate inflammation. To what extent this property depends on presence of distinct MSC or

other cell subpopulations in the gingival tissues or cell cultures, developmental origin, specific

phenotype of the cells and tissue niche that they create or other factors remains to be shown. In

any case, due to their unique properties discussed above and easy access, gingival cells could

have certain advantages over skin cells and be considered for therapeutic applications also in

various areas of the body other than the oral cavity. Particularly, these would include conditions

that require connective tissue regeneration, healing without scarring or mucosal type epithelial

keratinization.

Page 27 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

28

28

Information about utility of gingival MSCs for tissue regeneration or clinical use of gingival cells

in general in tissue engineering in other anatomical locations than the oral cavity is currently

scarce. However, full thickness oral soft tissue autografts that contain, among other cells, viable

fibroblasts and likely MSCs, have been successfully used for decades. In particular, human

palatal tissue autografts are routinely used as free gingival (containing both gingival epithelium

and connective tissue) or connective tissue grafts to augment gingival tissue deficiencies. Both

graft types induce epithelial differentiation typical to the donor tissue (i.e. keratinized stratified

squamous epithelium) (82, 83). In addition, gingival fibroblasts have been recently applied in

vascular therapy in an animal model of a carotid aneurysm (173). The results showed that,

remarkably, grafting gingival fibroblasts into the arterial walls caused reduction of the lesions

while those treated with medium control or dermal fibroblasts continued to grow. Gingival

fibroblasts also supported the elastin repair in the arterial wall (173). Potential use of gingival

cells to improve wound-healing outcome in situations that have been traditionally treated with

skin cell-derived grafts, including burn wound and other skin trauma, is another attractive future

application. Wound healing of large skin defects, even with the best current therapeutic

modalities, usually results to excessive scar formation (190). Scar-free wound healing in gingiva

suggests that grafts of gingival cells maybe used to reduce scar-forming response in these

situations. Furthermore, ability of gingival cells to promote fast wound healing maybe utilized to

treat chronic non-healing skin wounds, a major clinical problem (191).

Gingiva as a source for induced pluripotent stem cells

Page 28 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

29

29

A novel therapeutic applications for gingival cells could be their use to generate iPSCs. iPSCs

were initially produced from non-pluripotent somatic cells by forced expression of four genes,

Oct4, Sox2, Klf4 and c-Myc by using viral vectors (192). More recently it was shown that iPSCs

can also be generated using corresponding recombinant proteins or by combined expression of

Oct4, Sox2 and Klf4 only or by a combination of Oct4, Sox2, Nanog and Lin28 (193, 194).

iPSCs share key properties, including gene expression, chromatin methylation, embryonic body

and teratoma formation, viable chimera formation after injection into blastocyst and ecto-, meso-

and endodermal differentiation potency, with embryonic stem cells (192). Interestingly, it appears

that iPSC-like cells can also be generated from human skin fibroblasts by overexpression of a

single gene encoding an ECM protein fibromodulin (195), but the mechanisms involved and

potential therapeutic potential of this finding remains to be shown and confirmed.

While skin fibroblasts from various animals and humans have been the most common source to

generate iPSCs, theoretically, they can be established from any somatic cell type. Accordingly,

they have also been produced from various mesenchymal, epithelial and blood-derived cells from

different animals or humans, but in general the generation efficiency is low (often less than 1%)

and varies greatly depending on the cell type (193, 196). Recently, iPSCs were produced from

cells of oral origin, namely from human dental pulp cells, buccal mucosa and periodontal

ligament fibroblasts (197-200). Recently, iPSCs were also produced and characterized from

human and mouse gingival fibroblasts (200, 201). Interestingly, generation of iPSCs was seven

times more efficient from mouse gingival fibroblasts as compared to fibroblasts obtained from

tail skin (201). Generation of patient-specific and disease-specific gingiva-derived iPSCs would

provide tools for disease modeling and drug screening in conditions related to oral cavity. These

Page 29 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

30

30

could include syndromes involving oral or dental tissues, conditions involving congenital dental

and/or oral malformations, hereditary gingival overgrowth or certain forms of periodontal disease

(202). Although promising, iPSC-based therapies are still in their initial stages of development

and several technical issues, including further improving efficacy of iPSC generation,

understanding the properties of iPSCs and safety concerns, such as tendency of iPSCs to form

tumors and adverse effects of the viral vectors, need to be addressed before iPSCs from human

gingiva or from any other source can be considered for human therapy (193, 194).

Due to their neural-crest origin, gingival cells may be suitable for the so-called direct somatic cell

fate reprogramming (induced transdifferentiation) where one differentiated cell type is converted

to another distinct cell type without a reversion to iPSCs or use of oncogenes. Recently, this

technique has been used to generate functional induced neurons (iNs) and induced neural stem

cells (iNSCs) from various cells (203), but there is no experimental evidence about the efficiency

of gingival cells in this application.

Conclusions and future directions

Connective tissue cells form a phenotypically heterogeneous group of cells that play key roles in

tissue development, maintenance and repair and contribute to various pathologies. These cells

can be relatively easily isolated and expanded in cell culture. Depending on tissue origin,

connective tissue cell cultures contain cells, most notably MSCs, that possess distinct ability for

multipotent differentiation and immunomodulation and promote vasculogenesis/angiogenesis and

epithelialization (Table 4). Identification of distinct phenotypic properties of these cells from

different tissues may provide novel opportunities to selectively use different phenotypic traits for

Page 30 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

31

31

various tissue regeneration modalities. Adult oral mucosal connective tissue cells, and in

particular MSCs, can be easily harvested with little morbidity and possess distinct characteristics,

including neural crest origin, multipotent differentiation capacity, fetal-like phenotype and potent

immunomodulatory properties, that may be utilized to promote tissue regeneration and fast, scar-

free wound healing. Future studies should be focused on understanding the phenotypic

differences among connective tissue cell populations and MSCs, to understand the specific

properties of neural crest-derived cells, to develop better molecular markers to identify and enrich

appropriate cell populations and in detail characterize their properties important for tissue

regeneration applications in vivo. In particular, studies systematically comparing gingival cells to

corresponding cells from other tissues and sources should be conducted to further verify the

distinct potential of gingival cells in vitro and in vivo. Furthermore, developing efficient cell

delivery modalities that accurately target the cells to the area of interest and provide an

appropriate cellular niche important for their engraftment and function need further development.

Finally, assessing putative risks and adverse effects of cell-based therapies will be critical.

Acknowledgements: We thank Dr. L. Loison-Robert for technical assistance. Supported by

Canadian Institutes of Health Research.

Author Disclosure Statement: Drs. B. Fournier, H. Larjava and L. Häkkinen have no competing

financial interests.

Page 31 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

32

32

References

1. Robert L. (1992). The fibroblast, definition of its phenotype by its "programme" of

biosynthesis of the extracellular matrix. Pathol Biol (Paris) 40:851-858.

2. Keating A. (2012). Mesenchymal stromal cells: new directions. Cell Stem Cell 14:709-

716.

3. Dominici M, K Le Blanc, I Mueller, I Slaper-Cortenbach, F Marini, D Krause, R Deans,

A Keating, D Prockop and E Horwitz. (2006). Minimal criteria for defining multipotent

mesenchymal stromal cells. The International Society for Cellular Therapy position statement.

Cytotherapy 8:315-317.

4. Bianco P, X Cao, PS Frenette, JJ Mao, PG Robey, PJ Simmons and CY Wang. (2013).

The meaning, the sense and the significance: translating the science of mesenchymal stem cells

into medicine. Nat Med 19:35-42.

5. Augello A, TB Kurth and C De Bari. (2010). Mesenchymal stem cells: a perspective from

in vitro cultures to in vivo migration and niches. Eur Cell Mater 20:121-133.

6. Pevsner-Fischer M, S Levin and D Zipori. (2011). The origins of mesenchymal stromal

cell heterogeneity. Stem Cell Rev 7:560-568.

7. Bieback K, P Wuchter, D Besser, W Franke, M Becker, M Ott, M Pacher, N Ma, C

Stamm, H Kluter, A Muller and AD Ho. (2012). Mesenchymal stromal cells (MSCs): science and

f(r)iction. J Mol Med 90:773-782.

8. Mao JJ and DJ Prockop. (2012). Stem cells in the face: tooth regeneration and beyond.

Cell Stem Cell 11:291-301.

9. Hematti P. (2012). Mesenchymal stromal cells and fibroblasts: a case of mistaken identity?

Page 32 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

33

33

Cytotherapy 14:516-521.

10. Chen L, DM He and Y Zhang. (2009). The differentiation of human placenta-derived

mesenchymal stem cells into dopaminergic cells in vitro. Cell Mol Biol Lett 14:528-536.

11. Sorrell JM and AI Caplan. (2004). Fibroblast heterogeneity: more than skin deep. J Cell

Sci 117:667-675.

12. Haniffa MA, MP Collin, CD Buckley and F Dazzi. (2009). Mesenchymal stem cells: the

fibroblasts' new clothes? Haematologica 94:258-263.

13. Irwin CR, M Picardo, I Ellis, P Sloan, A Grey, M McGurk and SL Schor. (1994). Inter-

and intra-site heterogeneity in the expression of fetal-like phenotypic characteristics by gingival

fibroblasts: potential significance for wound healing. J Cell Sci 107 (Pt 5):1333-1346.

14. Phipps RP, MA Borrello and TM Blieden. (1997). Fibroblast heterogeneity in the

periodontium and other tissues. J Periodontal Res 32:159-165.

15. Yang H, LN Gao, Y An, CH Hu, F Jin, J Zhou, Y Jin Y and FM Chen. (2013).

Comparison of mesenchymal stem cells derived from gingival tissue and periodontal ligament in

different incubation conditions. Biomaterials 34(29):7033-7047.

16. Phinney DG and DJ Prockop. (2007). Concise review: mesenchymal stem/multipotent

stromal cells: the state of transdifferentiation and modes of tissue repair--current views. Stem

Cells 25:2896-2902.

17. Kuhn NZ and RS Tuan. (2010). Regulation of stemness and stem cell niche of

mesenchymal stem cells: implications in tumorigenesis and metastasis. J Cell Physiol 222:268-

277.

18. Lu W, MO Casanueva, AP Mahowald, M Kato, D Lauterbach and EL Ferguson. (2012).

Niche-associated activation of rac promotes the asymmetric division of Drosophila female

Page 33 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

34

34

germline stem cells. PLoS Biol 10:e1001357.

19. Alt E, Y Yan, S Gehmert, YH Song, A Altman, S Gehmert, D Vykoukal and X Bai.

(2011). Fibroblasts share mesenchymal phenotypes with stem cells, but lack their differentiation

and colony-forming potential. Biol Cell 103:197-208.

20. Toma JG, M Akhavan, KJ Fernandes, F Barnabe-Heider, A Sadikot, DR Kaplan and FD

Miller. (2001). Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat

Cell Biol 3:778-784.

21. Toma JG, IA McKenzie, D Bagli and FD Miller. (2005). Isolation and characterization of

multipotent skin-derived precursors from human skin. Stem Cells 23:727-737.

22. Hunt DP, C Jahoda and S Chandran. (2009). Multipotent skin-derived precursors: from

biology to clinical translation. Curr Opin Biotechnol 20:522-530.

23. Kang HK, SK Min, SY Jung, K Jung, H Jang da, OB Kim, GS Chun, ZH Lee and BM

Min. (2011). The potential of mouse skin-derived precursors to differentiate into mesenchymal

and neural lineages and their application to osteogenic induction in vivo. Int J Mol Med 28:1001-

1011.

24. Achilleos A and PA Trainor. (2012). Neural crest stem cells: discovery, properties and

potential for therapy. Cell Res 22:288-304.

25. Uchugonova A, J Duong, N Zhang, K Konig and RM Hoffman. (2011). The bulge area is

the origin of nestin-expressing pluripotent stem cells of the hair follicle. J Cell Biochem

112:2046-2050.

26. Jinno H, O Morozova, KL Jones, JA Biernaskie, M Paris, R Hosokawa, MA Rudnicki, Y

Chai, F Rossi, MA Marra and FD Miller. (2010). Convergent genesis of an adult neural crest-like

dermal stem cell from distinct developmental origins. Stem Cells 28:2027-2040.

Page 34 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

35

35

27. Kaltschmidt B, C Kaltschmidt and D Widera. (2011). Adult Craniofacial Stem Cells:

Sources and Relation to the Neural Crest. Stem Cell Rev 8:658-671.

28. Arnold WH, S Becher, A Dannan, D Widera, T Dittmar, M Jacob, HG Mannherz, T

Dittmar, B Kaltschmidt, C Kaltschmidt and WD Grimm. (2010). Morphological characterization

of periodontium-derived human stem cells. Ann Anat 192:215-219.

29. Sasaki R, S Aoki, M Yamato, H Uchiyama, K Wada, T Okano and H Ogiuchi. (2008).

Neurosphere generation from dental pulp of adult rat incisor. Eur J Neurosci 27:538-548.

30. Techawattanawisal W, K Nakahama, M Komaki, M Abe, Y Takagi and I Morita. (2007).

Isolation of multipotent stem cells from adult rat periodontal ligament by neurosphere-forming

culture system. Biochem Biophys Res Commun 357:917-923.

31. Widera D, WD Grimm, JM Moebius, I Mikenberg, C Piechaczek, G Gassmann, NA

Wolff, F Thevenod, C Kaltschmidt and B Kaltschmidt. (2007). Highly efficient neural

differentiation of human somatic stem cells, isolated by minimally invasive periodontal surgery.

Stem Cells Dev 16:447-460.

32. Widera D, C Zander, M Heidbreder, Y Kasperek, T Noll, O Seitz, B Saldamli, H Sudhoff,

R Sader, C Kaltschmidt and B Kaltschmidt. (2009). Adult palatum as a novel source of neural

crest-related stem cells. Stem Cells 27:1899-1910.

33. Dupin E and L Sommer. (2012). Neural crest progenitors and stem cells: from early

development to adulthood. Dev Biol 366:83-95.

34. Fernandes KJ, JG Toma and FD Miller. (2008). Multipotent skin-derived precursors:

adult neural crest-related precursors with therapeutic potential. Philos Trans R Soc Lond B Biol

Sci 363:185-198.

35. Shakhova O and L Sommer. Neural crest-derived stem cells. (2010). StemBook,

Page 35 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

36

36

Cambridge, MA, Harvard Stem Cell Institute, p. 2008.

36. Chai Y, X Jiang, Y Ito, P Bringas, Jr., J Han, DH Rowitch, P Soriano, AP McMahon and

HM Sucov. (2000). Fate of the mammalian cranial neural crest during tooth and mandibular

morphogenesis. Development 127:1671-1679.

37. Rinn JL, JK Wang, H Liu, K Montgomery, M van de Rijn and HY Chang. (2008). A

systems biology approach to anatomic diversity of skin. J Invest Dermatol 128:776-782.

38. Hill MW and IC Mackenzie. (1989). The influence of subepithelial connective tissues on

epithelial proliferation in the adult mouse. Cell Tissue Res 255:179-182.

39. Okazaki M, K Yoshimura, Y Suzuki and K Harii. (2003). Effects of subepithelial

fibroblasts on epithelial differentiation in human skin and oral mucosa: heterotypically

recombined organotypic culture model. Plast Reconstr Surg 112:784-792.

40. Mikkola ML and SE Millar. (2006). The mammary bud as a skin appendage: unique and

shared aspects of development. J Mammary Gland Biol Neoplasia 11:187-203.

41. Huang GT, S Gronthos and S Shi. (2009). Mesenchymal stem cells derived from dental

tissues vs. those from other sources: their biology and role in regenerative medicine. J Dent Res

88:792-806.

42. Yang CC and G Cotsarelis. (2010). Review of hair follicle dermal cells. J Dermatol Sci

57:2-11.

43. Seo BM, M Miura, S Gronthos, PM Bartold, S Batouli, J Brahim, M Young, PG Robey,

CY Wang and S Shi. (2004). Investigation of multipotent postnatal stem cells from human

periodontal ligament. Lancet 364:149-155.

44. Hynes K, D Menicanin, S Gronthos and PM Bartold. (2012). Clinical utility of stem cells

for periodontal regeneration. Periodontol 2000 59:203-227.

Page 36 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

37

37

45. Wu G, ZH Deng, XJ Fan, ZF Ma, YJ Sun, DD Ma, JJ Wu, JN Shi and Y Jin. (2009).

Odontogenic potential of mesenchymal cells from hair follicle dermal papilla. Stem Cells Dev

18:583-589.

46. Atari M, J Caballe-Serrano, C Gil-Recio, C Giner-Delgado, E Martinez-Sarra, DA

Garcia-Fernandez, M Barajas, F Hernandez-Alfaro, E Ferres-Padro and L Giner-Tarrida. (2012).

The enhancement of osteogenesis through the use of dental pulp pluripotent stem cells in 3D.

Bone 50:930-941.

47. Davies LC, M Locke, RD Webb, JT Roberts, M Langley, DW Thomas, CW Archer and P

Stephens. (2010). A multipotent neural crest-derived progenitor cell population is resident within

the oral mucosa lamina propria. Stem Cells Dev 19:819-830.

48. Estrela C, AH Alencar, GT Kitten, EF Vencio and E Gava. (2011). Mesenchymal stem

cells in the dental tissues: perspectives for tissue regeneration. Braz Dent J 22:91-98.

49. Fournier BP, FC Ferre, L Couty, JJ Lataillade, M Gourven, A Naveau, B Coulomb, A

Lafont and B Gogly. (2010). Multipotent Progenitor Cells in Gingival Connective Tissue. Tissue

Eng Part A 16:2891-2899.

50. Gronthos S, M Mankani, J Brahim, PG Robey and S Shi. (2000). Postnatal human dental

pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci (USA) 97:13625-13630.

51. Hung TY, HC Lin, YJ Chan and K Yuan. (2012). Isolating stromal stem cells from

periodontal granulation tissues. Clin Oral Investig 16:1171-1180.

52. Miura M, S Gronthos, M Zhao, B Lu, LW Fisher, PG Robey and S Shi. (2003). SHED:

stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci (USA) 100:5807-5812.

53. Morsczeck C, W Gotz, J Schierholz, F Zeilhofer, U Kuhn, C Mohl, C Sippel and KH

Hoffmann. (2005). Isolation of precursor cells (PCs) from human dental follicle of wisdom teeth.

Page 37 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

38

38

Matrix Biol 24:155-165.

54. Sonoyama W, Y Liu, D Fang, T Yamaza, BM Seo, C Zhang, H Liu, S Gronthos, CY

Wang, S Wang and S Shi. (2006). Mesenchymal stem cell-mediated functional tooth regeneration

in swine. PLoS One 1:e79.

55. Hsu SH, GS Huang and F Feng. (2012). Isolation of the multipotent MSC subpopulation

from human gingival fibroblasts by culturing on chitosan membranes. Biomaterials 33:2642-

2655.

56. Davies LC, H Lonnies, M Locke, B Sundberg, K Rosendahl, C Gotherstrom, K Le Blanc

and P Stephens. (2012). Oral mucosal progenitor cells are potently immunosuppressive in a dose-

independent manner. Stem Cells Dev 21:1478-1487.

57. Strioga M, S Viswanathan, A Darinskas, O Slaby and J Michalek. (2012). Same or not the

same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and

stromal cells. Stem Cells Dev 21:2724-2752.

58. Wang F, M Yu, X Yan, Y Wen, Q Zeng, W Yue, P Yang and X Pei. (2011). Gingiva-

derived mesenchymal stem cell-mediated therapeutic approach for bone tissue regeneration. Stem

Cells Dev 20:2093-2102.

59. Zhang Q, S Shi, Y Liu, J Uyanne, Y Shi, S Shi and AD Le. (2009). Mesenchymal stem

cells derived from human gingiva are capable of immunomodulatory functions and ameliorate

inflammation-related tissue destruction in experimental colitis. J Immunol 183:7787-7798.

60. Tomar GB, RK Srivastava, N Gupta, AP Barhanpurkar, ST Pote, HM Jhaveri, GC Mishra

and MR Wani. (2010). Human gingiva-derived mesenchymal stem cells are superior to bone

marrow-derived mesenchymal stem cells for cell therapy in regenerative medicine. Biochem

Biophys Res Commun 393:377-383.

Page 38 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

39

39

61. Dezawa M, M Hoshino, Y Nabeshima and C Ide. (2005). Marrow stromal cells:

implications in health and disease in the nervous system. Curr Neuropharmacol 5:723-732.

62. Arthur A, G Rychkov, S Shi, SA Koblar and S Gronthos. (2008). Adult human dental

pulp stem cells differentiate toward functionally active neurons under appropriate environmental

cues. Stem Cells 26:1787-1795.

63. Ferro F, R Spelat, F D'Aurizio, E Puppato, M Pandolfi, AP Beltrami, D Cesselli, G Falini,

CA Beltrami and F Curcio. (2012). Dental pulp stem cells differentiation reveals new insights in

Oct4A dynamics. PLoS One 7:e41774.

64. Sakai K, A Yamamoto, K Matsubara, S Nakamura, M Naruse, M Yamagata, K Sakamoto,

R Tauchi, N Wakao, S Imagama, H Hibi, K Kadomatsu, N Ishiguro and M Ueda. (2012). Human

dental pulp-derived stem cells promote locomotor recovery after complete transection of the rat

spinal cord by multiple neuro-regenerative mechanisms. J Clin Invest122:80-90.

65. Zhang W, XF Walboomers, TH Van Kuppevelt, WF Daamen, PA Van Damme, Z Bian

and JA Jansen. (2008). In vivo evaluation of human dental pulp stem cells differentiated towards

multiple lineages. J Tissue Eng Regen Med 2:117-125.

66. Sakai VT, Z Zhang, Z Dong, KG Neiva, MA Machado, S Shi, CF Santos and JE Nor.

(2010). SHED differentiate into functional odontoblasts and endothelium. J Dent Res 89:791-796.

67. Seo BM, W Sonoyama, T Yamaza, C Coppe, T Kikuiri, K Akiyama, JS Lee and S Shi.

(2008). SHED repair critical-size calvarial defects in mice. Oral Dis 14:428-434.

68. Sonoyama W, Y Liu, T Yamaza, RS Tuan, S Wang, S Shi and GT Huang. (2008).

Characterization of the apical papilla and its residing stem cells from human immature permanent

teeth: a pilot study. J Endod 34:166-171.

69. d'Aquino R, V Tirino, V Desiderio, M Studer, GC De Angelis, L Laino, A De Rosa, D Di

Page 39 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

40

40

Nucci, S Martino, F Paino, M Sampaolesi and G Papaccio. (2011). Human neural crest-derived

postnatal cells exhibit remarkable embryonic attributes either in vitro or in vivo. Eur Cell Mater

21:304-316.

70. Yokoi T, M Saito, T Kiyono, S Iseki, K Kosaka, E Nishida, T Tsubakimoto, H Harada, K

Eto, T Noguchi and T Teranaka. (2007). Establishment of immortalized dental follicle cells for

generating periodontal ligament in vivo. Cell Tissue Res 327:301-311.

71. Dong R, X Liu, M Fan, L Yang, L Peng and L Zhang. (2010). Isolation and

differentiation of nestin positive cells from rat oral mucosal lamina propria. Differentiation 79:9-

14.

72. Mitrano TI, MS Grob, F Carrion, E Nova-Lamperti, PA Luz, FS Fierro, A Quintero, A

Chaparro and A Sanz. (2010). Culture and characterization of mesenchymal stem cells from

human gingival tissue. J Periodontol 81:917-925.

73. Wei F, T Song, G Ding, J Xu, Y Liu, D Liu, Z Fan, C Zhang, S Shi and S Wang. (2013).

Functional tooth restoration by allogeneic mesenchymal stem cell-based bio-root regeneration in

swine. Stem Cells Dev 22:1752-1762.

74. Marynka-Kalmani K, S Treves, M Yafee, H Rachima, Y Gafni, MA Cohen and S Pitaru.

(2010). The lamina propria of adult human oral mucosa harbors a novel stem cell population.

Stem Cells 28:984-995.

75. Xu X, C Chen, K Akiyama, Y Chai, AD Le, Z Wang Z and S Shi. (2013). Gingivae

Contain Neural-crest- and Mesoderm-derived Mesenchymal Stem Cells. J Dent Res Jul 18. [Epub

ahead of print]

76. Treves-Manusevitz S, L Hoz , H Rachima , G Montoya , E Tzur , A Vardimon , AS

Narayanan , S Amar , H Arzate and S Pitaru. (2013) Stem cells of the lamina propria of human

Page 40 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

41

41

oral mucosa and gingiva develop into mineralized tissues in vivo. J Clin Periodontol 40:73-81.

77. Gotherstrom C, O Ringden, C Tammik, E Zetterberg, M Westgren and K Le Blanc.

(2004). Immunologic properties of human fetal mesenchymal stem cells. Am J Obstet Gynecol

190:239-245.

78. Atari M, C Gil-Recio, M Fabregat, D Garcia-Fernandez, M Barajas, MA Carrasco, HS

Jung, FH Alfaro, N Casals, F Prosper, E Ferres-Padro and L Giner. (2012). Dental pulp of the

third molar: a new source of pluripotent-like stem cells. J Cell Sci 125:3343-3356.

79. Squier CA. (1991). The permeability of oral mucosa. Crit Rev Oral Biol Med 2:13-32.

80. Winning TA and GC Townsend. (2000). Oral mucosal embryology and histology. Clin

Dermatol 18:499-511.

81. Collins LM and C Dawes. (1987). The surface area of the adult human mouth and

thickness of the salivary film covering the teeth and oral mucosa. J Dent Res 66:1300-1302.

82. Karring T, NP Lang and H Loe. (1975). The role of gingival connective tissue in

determining epithelial differentiation. J Periodont Res 10:1-11.

83. Karring T, E Ostergaard and H Loe. (1971). Conservation of tissue specificity after

heterotopic transplantation of gingiva and alveolar mucosa. J Periodont Res 6:282-293.

84. Schweizer J, H Winter, MW Hill and IC Mackenzie. (1984). The keratin polypeptide

patterns in heterotypically recombined epithelia of skin and mucosa of adult mouse.

Differentiation 26:144-153.

85. Schroeder H. Oral Structural Biology. (1991). Thieme Medical Publisher, New York,

USA.

86. Page RC and WF Ammons. (1974). Collagen turnover in the gingiva and other mature

connective tissues of the marmoset Saguinus oedipus. Arch Oral Biol 19:651-658.

Page 41 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

42

42

87. Ouhayoun JP, F Gosselin, N Forest, S Winter and WW Franke. (1985). Cytokeratin

patterns of human oral epithelia: differences in cytokeratin synthesis in gingival epithelium and

the adjacent alveolar mucosa. Differentiation 30:123-129.

88. Ermich T, J Schulz and D Schumann. (1988). Pattern of oral cytokeratins: I. SDS-

electrophoretic analysis of the frequency of cytoskeletal keratins in the normal human mucosa of

mouth. Biomed Biochim Acta 47:737-742.

89. Presland RB and BA Dale. (2000). Epithelial structural proteins of the skin and oral

cavity: function in health and disease. Crit Rev Oral Biol Med 11:383-408.

90. Paladini RD, K Takahashi, NS Bravo and PA Coulombe. (1996). Onset of re-

epithelialization after skin injury correlates with a reorganization of keratin filaments in wound

edge keratinocytes: defining a potential role for keratin 16. J Cell Biol 132:381-397.

91. Mak K, A Manji, C Gallant-Behm, C Wiebe, DA Hart, H Larjava and L Hakkinen. (2009).

Scarless healing of oral mucosa is characterized by faster resolution of inflammation and control

of myofibroblast action compared to skin wounds in the red Duroc pig model. J Dermatol Sci

56:168-180.

92. Wong JW, C Gallant-Behm, C Wiebe, K Mak, DA Hart, H Larjava and L Hakkinen.

(2009). Wound healing in oral mucosa results in reduced scar formation as compared with skin:

evidence from the red Duroc pig model and humans. Wound Repair Regen 17:717-729.

93. Schor SL, I Ellis, CR Irwin, J Banyard, K Seneviratne, C Dolman, AD Gilbert and DM

Chisholm. (1996). Subpopulations of fetal-like gingival fibroblasts: characterisation and potential

significance for wound healing and the progression of periodontal disease. Oral Dis 2:155-166.

94. Sciubba JJ, JP Waterhouse and J Meyer. (1978). A fine structural comparison of the

healing of incisional wounds of mucosa and skin. J Oral Pathol 7:214-227.

Page 42 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

43

43

95. Szpaderska AM, CG Walsh, MJ Steinberg and LA DiPietro. (2005). Distinct patterns of

angiogenesis in oral and skin wounds. J Dent Res 84:309-314.

96. Szpaderska AM, JD Zuckerman and LA DiPietro. (2003). Differential injury responses in

oral mucosal and cutaneous wounds. J Dent Res 82:621-626.

97. Schrementi ME, AM Ferreira, C Zender and LA DiPietro. (2008). Site-specific

production of TGF-beta in oral mucosal and cutaneous wounds. Wound Repair Regen 16:80-86.

98. Eslami A, CL Gallant-Behm, DA Hart, C Wiebe, D Honardoust, H Gardner, L Hakkinen

and HS Larjava. (2009). Expression of integrin alphavbeta6 and TGF-beta in scarless vs scar-

forming wound healing. J Histochem Cytochem 57:543-557.

99. Hakkinen L, VJ Uitto and H Larjava. (2000). Cell biology of gingival wound healing.

Periodontol 2000 24:127-152.

100. Häkkinen L, H Larjava and L Koivisto. (2012). Granulation Tissue Formation and

Remodeling. In: Oral Wound Healing: Cell Biology and Clinical Management. H Larjava, ed.,

John Wiley & Sons, Inc., Ames, pp. 125-173.

101. Chavrier C. (1990). The elastic system fibres in healthy human gingiva. Arch Oral Biol

35 Suppl:223S-225S.

102. Hsieh PC, YT Jin, CW Chang, CC Huang, SC Liao and K Yuan. (2010). Elastin in oral

connective tissue modulates the keratinization of overlying epithelium. J Clin Periodontol

37:705-711.

103. Kielty CM, MJ Sherratt and CA Shuttleworth. (2002). Elastic fibres. J Cell Sci 115:2817-

2828.

104. Lozdan J. (1970). Studies on the mucogingival junction. Dent Pract Dent Rec 20:379-384.

105. Goktas S, JJ Dmytryk and PS McFetridge. (2011). Biomechanical behavior of oral soft

Page 43 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

44

44

tissues. J Periodontol 82:1178-1186.

106. Lee-Thedieck C, N Rauch, R Fiammengo, G Klein and JP Spatz. (2012). Impact of

substrate elasticity on human hematopoietic stem and progenitor cell adhesion and motility. J

Cell Sci 125:3765-3775.

107. Tschumperlin DJ, F Liu and AM Tager. (2013). Biomechanical regulation of

mesenchymal cell function. Curr Opin Rheumatol 25:92-100.

108. Bielefeld KA, S Amini-Nik and BA Alman. (2012). Cutaneous wound healing: recruiting

developmental pathways for regeneration. Cell Mol Life Sci Oct 4. [Epub ahead of print]

109. Namazi MR, MK Fallahzadeh and RA Schwartz. (2011). Strategies for prevention of

scars: what can we learn from fetal skin? Int J Dermatol 50:85-93.

110. Brizzi MF, G Tarone and P Defilippi. (2012). Extracellular matrix, integrins, and growth

factors as tailors of the stem cell niche. Curr Opin Cell Biol 24:645-651.

111. Bronson RE, JG Argenta, EP Siebert and CN Bertolami. (1988). Distinctive fibroblastic

subpopulations in skin and oral mucosa demonstrated by differences in glycosaminoglycan

content. In Vitro Cell Dev Biol 24:1121-1126.

112. Narayanan AS, RC Page and DF Meyers. (1980). Characterization of collagens of

diseased human gingiva. Biochemistry 19:5037-5043.

113. Pedlar J. (1984). Biochemistry of glycosaminoglycans in the skin and oral mucosa of the

rat. Arch Oral Biol 29:591-597.

114. Larjava H, J Heino, T Krusius, E Vuorio and M Tammi. (1988). The small dermatan

sulphate proteoglycans synthesized by fibroblasts derived from skin, synovium and gingiva show

tissue-related heterogeneity. Biochem J 256:35-40.

115. Tsuruga E, K Irie, Y Sakakura and T Yajima. (2002). Tropoelastin expression by

Page 44 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

45

45

periodontal fibroblasts. J Dent Res 81:198-202.

116. Tsuruga E, K Irie, Y Sakakura and T Yajima. (2002). Expression of fibrillins and

tropoelastin by human gingival and periodontal ligament fibroblasts in vitro. J Periodont Res

37:23-28.

117. Tsuruga E, K Irie and T Yajima. (2002). Gene expression and accumulation of fibrillin-1,

fibrillin-2, and tropoelastin in cultured periodontal fibroblasts. J Dent Res 81:771-775.

118. Gogly B, G Godeau, S Gilbert, JM Legrand, C Kut, B Pellat and M Goldberg. (1997).

Morphometric analysis of collagen and elastic fibers in normal skin and gingiva in relation to age.

Clin Oral Investig 1:147-152.

119. Guo F, DE Carter and A Leask. (2011). Mechanical tension increases CCN2/CTGF

expression and proliferation in gingival fibroblasts via a TGFbeta-dependent mechanism. PLoS

One 6:e19756.

120. Guo F, DE Carter, A Mukhopadhyay and A Leask. (2011). Gingival fibroblasts display

reduced adhesion and spreading on extracellular matrix: a possible basis for scarless tissue repair?

PLoS One 6:e27097.

121. Ebisawa K, R Kato, M Okada, T Sugimura, MA Latif, Y Hori, Y Narita, M Ueda, H

Honda and H Kagami. (2011). Gingival and dermal fibroblasts: their similarities and differences

revealed from gene expression. J Biosci Bioeng 111:255-258.

122. Macri L, D Silverstein and RA Clark. (2007). Growth factor binding to the pericellular

matrix and its importance in tissue engineering. Adv Drug Deliv Rev 59:1366-81.

123. Reilly GC and AJ Engler. (2010). Intrinsic extracellular matrix properties regulate stem

cell differentiation. J Biomech 43:55-62.

124. Goktas S, JJ Dmytryk and PS McFetridge. (2011). Biomechanical behavior of oral soft

Page 45 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

46

46

tissues. J Periodontol 82:1178-86.

125. Balestrini JL, S Chaudhry, V Sarrazy, A Koehler and B Hinz. (2012). The mechanical

memory of lung myofibroblasts. Integr Biol (Camb) 4:410-21.

126. Csiszar A, C Wiebe, H Larjava and L Hakkinen. (2007). Distinctive molecular

composition of human gingival interdental papilla. J Periodontol 78:304-314.

127. Grey AM, AM Schor, G Rushton, I Ellis and SL Schor. (1989). Purification of the

migration stimulating factor produced by fetal and breast cancer patient fibroblasts. Proc Natl

Acad Sci (USA) 86:2438-2442.

128. Schor SL, AM Schor, AM Grey and G Rushton. (1988). Foetal and cancer patient

fibroblasts produce an autocrine migration-stimulating factor not made by normal adult cells. J

Cell Sci 90 ( Pt 3):391-399.

129. Cockey GH, JA Boughman, EL Harris and TM Hassell. (1989). Genetic control of

variation in human gingival fibroblast proliferation rate. In Vitro Cell Dev Biol 25:255-258.

130. Hassell TM and EJ Stanek, 3rd. (1983). Evidence that healthy human gingiva contains

functionally heterogeneous fibroblast subpopulations. Arch Oral Biol 28:617-625.

131. Hakkinen L and H Larjava. (1992). Characterization of fibroblast clones from periodontal

granulation tissue in vitro. J Dent Res 71:1901-1907.

132. Ko SD, RC Page and AS Narayanan. (1977). Fibroblast heterogeneity and prostaglandin

regulation of subpopulations. Proc Natl Acad Sci (USA) 74:3429-3432.

133. Connor NS, JE Aubin and J Sodek. (1983). Independent expression of type I collagen and

fibronectin by normal fibroblast-like cells. J Cell Sci 63:233-244.

134. Bordin S, RC Page and AS Narayanan. (1984). Heterogeneity of normal human diploid

fibroblasts: isolation and characterization of one phenotype. Science 223:171-173.

Page 46 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

47

47

135. Hassell TM, DV Provenza and RA Foster. (1986). Synthetic activities of mass cultures

and clones of human gingival fibroblasts. Experientia 42:66-69.

136. Hou LT and JA Yaeger. (1993). Cloning and characterization of human gingival and

periodontal ligament fibroblasts. J Periodontol 64:1209-1218.

137. Fries KM, T Blieden, RJ Looney, GD Sempowski, MR Silvera, RA Willis and RP Phipps.

(1994). Evidence of fibroblast heterogeneity and the role of fibroblast subpopulations in fibrosis.

Clin Immunol Immunopathol 72:283-292.

138. Sempowski GD, MA Borrello, TM Blieden, RK Barth and RP Phipps. (1995). Fibroblast

heterogeneity in the healing wound. Wound Repair Regen 3:120-131.

139. Lekic PC, N Pender and CA McCulloch. (1997). Is fibroblast heterogeneity relevant to

the health, diseases, and treatments of periodontal tissues? Crit Rev Oral Biol Med 8:253-268.

140. Bordin S, LG Costa and X Tan. (1998). Fibroblast heterogeneity of signal transduction

mechanisms to complement-C1q. Analyses of calcium mobilization, inositol phosphate

accumulation, and protein kinases-C redistribution. J Periodontol 69:642-649.

141. Gagari E, MK Rand, L Tayari, H Vastardis, P Sharma, PV Hauschka and PD Damoulis.

(2006). Expression of stem cell factor and its receptor, c-kit, in human oral mesenchymal cells.

Eur J Oral Sci 114:409-415.

142. Trackman PC and A Kantarci. (2004). Connective tissue metabolism and gingival

overgrowth. Crit Rev Oral Biol Med 15:165-175.

143. Hassell TM and GH Gilbert. (1983). Phenytoin sensitivity of fibroblasts as the basis for

susceptibility to gingival enlargement. Am J Pathol 112:218-223.

144. Hassell TM, E Romberg, S Sobhani, L Lesko and R Douglas. (1988). Lymphocyte-

mediated effects of cyclosporine on human fibroblasts. Transplant Proc 20:993-1002.

Page 47 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

48

48

145. Stabellini G, C Calastrini, G Mariani, M Gioia, E Caramelli, N Gagliano, C Dellavia, C

Moscheni, L Vizzotto and EM Calvi. (2004). Extracellular glycosaminoglycan changes in healthy

and overgrown gingiva fibroblasts after cyclosporin A and cytokine treatments. J Oral Pathol

Med 33:346-353.

146. Tang L, N Li, H Xie and Y Jin. (2011). Characterization of mesenchymal stem cells from

human normal and hyperplastic gingiva. J Cell Physiol 226:832-842.

147. Mensing N, H Gasse, N Hambruch, JD Haeger, C Pfarrer and C Staszyk. (2011). Isolation

and characterization of multipotent mesenchymal stromal cells from the gingiva and the

periodontal ligament of the horse. BMC Vet Res 7:42.

148. Ross R and G Odland. (1968). Human wound repair. II. Inflammatory cells, epithelial-

mesenchymal interrelations, and fibrogenesis. J Cell Biol 39:152-168.

149. Helpap B, H Breining, S Cappel, KW Sturm and S Lymberopoulos. (1976). Wound

healing of the brain of rats after cryonecrosis. Autoradiographic investigations with 3H-

thymidine. Virchows Arch B Cell Pathol 22:151-161.

150. Crisan M, M Corselli, CW Chen and B Peault. (2011). Multilineage stem cells in the adult:

a perivascular legacy? Organogenesis 7:101-104.

151. Dulmovits BM and IM Herman. (2012). Microvascular remodeling and wound healing: a

role for pericytes. Int J Biochem Cell Biol 44:1800-1812.

152. Diaz-Flores L, R Gutierrez, JF Madrid, H Varela, F Valladares, E Acosta, P Martin-

Vasallo and L Diaz-Flores, Jr. (2009). Pericytes. Morphofunction, interactions and pathology in a

quiescent and activated mesenchymal cell niche. Histol Histopathol 24:909-969.

153. Lau K, R Paus, S Tiede, P Day and A Bayat. (2009). Exploring the role of stem cells in

cutaneous wound healing. Exp Dermatol 18:921-933.

Page 48 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

49

49

154. Feng J, A Mantesso, C De Bari, A Nishiyama and PT Sharpe. (2011). Dual origin of

mesenchymal stem cells contributing to organ growth and repair. Proc Natl Acad Sci (USA)

108:6503-6508.

155. Crisan M, S Yap, L Casteilla, CW Chen, M Corselli, TS Park, G Andriolo, B Sun, B

Zheng, L Zhang, C Norotte, PN Teng, J Traas, R Schugar, BM Deasy, S Badylak, HJ Buhring, JP

Giacobino, L Lazzari, J Huard and B Peault. (2008). A perivascular origin for mesenchymal stem

cells in multiple human organs. Cell Stem Cell 3:301-313.

156. Crisan M, B Deasy, M Gavina, B Zheng, J Huard, L Lazzari and B Peault. (2008).

Purification and long-term culture of multipotent progenitor cells affiliated with the walls of

human blood vessels: myoendothelial cells and pericytes. Methods Cell Biol 86:295-309.

157. Murakami S and H Okada. (1997). Lymphocyte-fibroblast interactions. Crit Rev Oral

Biol Med 8:40-50.

158. Ge S, KM Mrozik, D Menicanin, S Gronthos and PM Bartold. (2012). Isolation and

characterization of mesenchymal stem cell-like cells from healthy and inflamed gingival tissue:

potential use for clinical therapy. Regen Med 7(6):819-832.

159. Fawzy El-Sayed KM, S Paris , ST Becker , M Neuschl , W De Buhr , S Sälzer , A Wulff ,

M Elrefai , MS Darhous , M El-Masry , J Wiltfang and CE Dörfer . (2012). Periodontal

regeneration employing gingival margin-derived stem/progenitor cells: an animal study. J Clin

Periodontol 39:861-70.

160. Li N, N Liu, J Zhou, L Tang, B Ding, Y Duan and Y Jin. (2013). Inflammatory

environment induces gingival tissue-specific mesenchymal stem cells to differentiate towards a

pro-fibrotic phenotype. Biol Cell 105(6):261-275.

161. Takashiba S, K Naruishi and Y Murayama. (2003). Perspective of cytokine regulation for

Page 49 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

50

50

periodontal treatment: fibroblast biology. J Periodontol 74:103-110.

162. Zhang QZ, AL Nguyen, WH Yu and AD Le. (2012). Human oral mucosa and gingiva: a

unique reservoir for mesenchymal stem cells. J Dent Res 91:1011-8.

163. Chen M, W Su, X Lin, Z Guo J Wang, Q Zhang, D Brand, B Ryffel, J Huang, Z Liu, X

He, AD Le and SG Zheng. (2013). Adoptive transfer of human gingiva-derived mesenchymal

stem cells ameliorates collagen-induced arthritis VIA suppressing Th1 and Th17 and enhancing

regulatory T cell differentiation. Arthritis Rheum 65:1181-1193.

164. Zhang QZ, WR Su, SH Shi, P Wilder-Smith, AP Xiang, A Wong, AL Nguyen, CW Kwon

and AD Le. (2010). Human gingiva-derived mesenchymal stem cells elicit polarization of m2

macrophages and enhance cutaneous wound healing. Stem Cells 28:1856-68.

165. Su WR, Zhang QZ, Shi SH, Nguyen AL and Le AD. (2011). Human gingiva-derived

mesenchymal stromal cells attenuate contact hypersensitivity via prostaglandin E2-dependent

mechanisms. Stem Cells 29:1849-1860.

166. Cappellesso-Fleury S, B Puissant-Lubrano, PA Apoil, M Titeux, P Winterton, L Casteilla,

P Bourin and A Blancher. (2010). Human fibroblasts share immunosuppressive properties with

bone marrow mesenchymal stem cells. J Clin Immunol 30:607-619.

167. Wada N, D Menicanin, S Shi, PM Bartold and S Gronthos. (2009). Immunomodulatory

properties of human periodontal ligament stem cells. J Cell Physiol 219:667-676.

168. Shimabukuro Y, S Murakami and H Okada. (1992). Interferon-gamma-dependent

immunosuppressive effects of human gingival fibroblasts. Immunology 76:344-347.

169. Shimabukuro Y, S Murakami and H Okada. (1996). Antigen-presenting-cell function of

interferon gamma-treated human gingival fibroblasts. J Periodont Res 31:217-228.

170. Zou Z, Y Zhang, L Hao, F Wang, D Liu, Y Su and H Sun. (2010). More insight into

Page 50 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

51

51

mesenchymal stem cells and their effects inside the body. Expert Opin Biol Ther 10:215-230.

171. Mahanonda R, N Sa-Ard-Iam, P Montreekachon, A Pimkhaokham, K Yongvanichit, MM

Fukuda and S Pichyangkul. (2007). IL-8 and IDO expression by human gingival fibroblasts via

TLRs. J Immunol 178:1151-1157.

172. Nisapakultorn K, J Makrudthong, N Sa-Ard-Iam, P Rerkyen, R Mahanonda and O

Takikawa. (2009). Indoleamine 2,3-dioxygenase expression and regulation in chronic

periodontitis. J Periodontol 80:114-121.

173. Durand E, B Fournier, L Couty, M Lemitre, P Achouh, P Julia, L Trinquart, JN Fabiani, S

Seguier, B Gogly, B Coulomb and A Lafont. (2012). Endoluminal gingival fibroblast transfer

reduces the size of rabbit carotid aneurisms via elastin repair. Arterioscler Thromb Vasc Biol

32:1892-1901.

174. Gogly B, A Naveau, B Fournier, N Reinald, E Durand, C Brasselet, B Coulomb and A

Lafont. (2007). Preservation of rabbit aorta elastin from degradation by gingival fibroblasts in an

ex vivo model. Arterioscler Thromb Vasc Biol 27:1984-1990.

175. Guiha R, S el Khodeiry, L Mota and R Caffesse. (2001). Histological evaluation of

healing and revascularization of the subepithelial connective tissue graft. J Periodontol 72:470-

478.

176. Janson WA, MP Ruben, GM Kramer, AA Bloom and H Turner. (1969). Development of

the blood supply to split-thickness free ginival autografts. J Periodontol 40:707-716.

177. Oliver RC, H Loe and T Karring. (1968). Microscopic evaluation of the healing and

revascularization of free gingival grafts. J Periodont Res 3:84-95.

178. Suthin K, K Matsushita, M Machigashira, S Tatsuyama, T Imamura, M Torii and Y Izumi.

(2003). Enhanced expression of vascular endothelial growth factor by periodontal pathogens in

Page 51 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

52

52

gingival fibroblasts. J Periodont Res 38:90-96.

179. Hosokawa Y, I Hosokawa, K Ozaki, H Nakae and T Matsuo. (2005). Increase of CCL20

expression by human gingival fibroblasts upon stimulation with cytokines and bacterial

endotoxin. Clin Exp Immunol 142:285-291.

180. Okada Y, M Meguro, H Ohyama, S Yoshizawa, K Takeuchi-Hatanaka, N Kato, S

Matsushita, S Takashiba and F Nishimura. (2009). Human leukocyte histocompatibility antigen

class II-induced cytokines from human gingival fibroblasts promote proliferation of human

umbilical vein endothelial cells: potential association with enhanced angiogenesis in chronic

periodontal inflammation. J Periodont Res 44:103-109.

181. Kobayashi K, T Suzuki, Y Nomoto, Y Tada, M Miyake, A Hazama, I Wada, T Nakamura

and K Omori. (2010). A tissue-engineered trachea derived from a framed collagen scaffold,

gingival fibroblasts and adipose-derived stem cells. Biomaterials 31:4855-4863.

182. Kobayashi K, T Suzuki, Y Nomoto, Y Tada, M Miyake, A Hazama, T Nakamura and K

Omori. (2007). Potential of heterotopic fibroblasts as autologous transplanted cells for tracheal

epithelial regeneration. Tissue Eng 13:2175-2184.

183. Nolte SV, W Xu, HO Rennekampff and HP Rodemann. (2008). Diversity of fibroblasts--a

review on implications for skin tissue engineering. Cells Tissues Organs 187:165-176.

184. Felder JM, 3rd, E Hechenbleikner, M Jordan and J Jeng. (2012). Increasing the options

for management of large and complex chronic wounds with a scalable, closed-system dressing

for maggot therapy. J Burn Care Res 33:e169-175.

185. Metcalfe AD and MW Ferguson. (2007). Bioengineering skin using mechanisms of

regeneration and repair. Biomaterials 28:5100-5113.

186. Wong T, JA McGrath and H Navsaria. (2007). The role of fibroblasts in tissue

Page 52 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

53

53

engineering and regeneration. Br J Dermatol 156:1149-1155.

187. Jackson WM, LJ Nesti and RS Tuan. (2012). Mesenchymal stem cell therapy for

attenuation of scar formation during wound healing. Stem Cell Res Ther 3:20.

188. Nagy N, N Almaani, A Tanaka, JE Lai-Cheong, T Techanukul, JE Mellerio and JA

McGrath. (2011). HB-EGF induces COL7A1 expression in keratinocytes and fibroblasts:

possible mechanism underlying allogeneic fibroblast therapy in recessive dystrophic

epidermolysis Bullosa. J Invest Dermatol 131:1771-1774.

189. Tolar J, L Xia, MJ Riddle, CJ Lees, CR Eide, RT McElmurry, M Titeux, MJ Osborn, TC

Lund, A Hovnanian, JE Wagner and BR Blazar. (2011). Induced pluripotent stem cells from

individuals with recessive dystrophic epidermolysis bullosa. J Invest Dermatol 131:848-856.

190. Tziotzios C, C Profyris and J Sterling. (2012). Cutaneous scarring: Pathophysiology,

molecular mechanisms, and scar reduction therapeutics Part II. Strategies to reduce scar

formation after dermatologic procedures. J Am Acad Dermatol 66(1):13-24.

191. Greer N, N Foman J Dorrian, P Fitzgerald, R MacDonald, I Rutks I, and T Wilt.

Advanced Wound Care Therapies for Non-Healing Diabetic, Venous, and Arterial Ulcers: A

Systematic Review. (2012). Department of Veterans Affairs, Washington, DC, USA.

192. Takahashi K and S Yamanaka. (2006). Induction of pluripotent stem cells from mouse

embryonic and adult fibroblast cultures by defined factors. Cell 126:663-676.

193. Chun YS, K Byun and B Lee. (2011). Induced pluripotent stem cells and personalized

medicine: current progress and future perspectives. Anat Cell Biol 44:245-255.

194. Drews K, J Jozefczuk, A Prigione and J Adjaye. (2012). Human induced pluripotent stem

cells--from mechanisms to clinical applications. J Mol Med 90:735-745.

195. Zheng Z, J Jian, X Zhang, JN Zara, W Yin, M Chiang, Y Liu, J Wang, S Pang, K Ting

Page 53 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

54

54

and C Soo. (2012). Reprogramming of human fibroblasts into multipotent cells with a single

ECM proteoglycan, fibromodulin. Biomaterials 33:5821-5831.

196. Streckfuss-Bomeke K, F Wolf, A Azizian, M Stauske, M Tiburcy, S Wagner, D Hubscher,

R Dressel, S Chen, J Jende, G Wulf, V Lorenz, MP Schon, LS Maier, WH Zimmermann, G

Hasenfuss and K Guan. (2012). Comparative study of human-induced pluripotent stem cells

derived from bone marrow cells, hair keratinocytes, and skin fibroblasts. Eur Heart J. Jul 12.

[Epub ahead of print]

197. Miyoshi K, D Tsuji, K Kudoh, K Satomura, T Muto, K Itoh and T Noma. (2010).

Generation of human induced pluripotent stem cells from oral mucosa. J Biosci Bioeng 110:345-

350.

198. Tamaoki N, K Takahashi, T Tanaka, T Ichisaka, H Aoki, T Takeda-Kawaguchi, K Iida, T

Kunisada, T Shibata, S Yamanaka and K Tezuka. (2 010). Dental pulp cells for induced

pluripotent stem cell banking. J Dent Res 89:773-778.

199. Yan X, H Qin, C Qu, RS Tuan, S Shi and GT Huang. (2010). iPS cells reprogrammed

from human mesenchymal-like stem/progenitor cells of dental tissue origin. Stem Cells Dev

19:469-480.

200. Wada N, B Wang, NH Lin, AL Laslett, S Gronthos and PM Bartold. (2011). Induced

pluripotent stem cell lines derived from human gingival fibroblasts and periodontal ligament

fibroblasts. J Periodont Res 46:438-447.

201. Egusa H, K Okita, H Kayashima, G Yu, S Fukuyasu, M Saeki, T Matsumoto, S

Yamanaka and H Yatani. (2010). Gingival fibroblasts as a promising source of induced

pluripotent stem cells. PLoS One 5:e12743.

202. Srijaya TC, PJ Pradeep, RB Zain, S Musa, NH Abu Kasim and V Govindasamy. (2012).

Page 54 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

55

55

The promise of human induced pluripotent stem cells in dental research. Stem Cells Int

2012:423868.

203. Abdullah AI, A Pollock and T Sun. (2012). The path from skin to brain: generation of

functional neurons from fibroblasts. Mol Neurobiol 45:586-595.

Page 55 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

56

56

Figure legends

Page 56 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

57

57

Page 57 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

58

58

Figure 1. Comparison of fibroblasts and MSCs

A. Representative phase contrast microscope images of human gingival fibroblasts (GFBL) and

gingival stem cells (GSC) established from the fibroblast culture by CFU-F method. Images show

cells cultured at low and high density at passage 2. Morphologically, GFBLs and GSCs are

indistinguishable from each other. B. Key criteria for defining fibroblasts and mesenchymal stem

cells (MSCs). See 1-4 for references.

Page 58 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

59

59

Figure 2. Heterogeneity of connective tissue cells.

Plastic-adherent connective tissue cells form a group of cells that are phenotypically

heterogeneous between individuals, between different tissues within an individual and within a

given tissue. The phenotypic heterogeneity may result from presence of phenotypically different

fibroblast subpopulations, MSCs, myofibroblasts, pericytes and fibrocytes within a given cell

population. The different subsets of the cells share several morphological, molecular and

functional characteristics complicating their identification. However, relative proportions and

Page 59 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

60

60

distinct functions of these cell subsets may play important roles in tissue development,

maintenance, regeneration, repair and many disease processes.

Page 60 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

61

61

Figure 3. Clinical and histological features of gingiva within oral mucosa.

Page 61 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

62

62

A. Gingiva (G) represents the area of oral mucosa that surrounds teeth, is covered by a

keratinized epithelium and is demarcated from the non-keratinized oral lining mucosa (LM) by

the mucogingival junction (dotted line). B. Oral mucosa in the palate is covered by a similar

keratinized mucosa as gingiva. There is no apparent border between gingiva (G) that is closest to

the teeth and the palatal tissue proper. Characteristic features in the palate are the so-called rugae

(RG) that form through similar developmental program as other ectodermal appendages,

including teeth and hair (338). They can also contain so-called palatal neural crest-derived stem

cells (222). C. Representative histological section of human gingiva stained with hematoxylin

and eosin. Typical to the gingiva, epithelium forms long projections (rete pegs or rete ridges) that

extend deep into the connective tissue. D. Representative human gingival tissue section stained

with a monoclonal antibody that recognizes tenascin-C. Typically, gingiva displays strong

accumulation of tenascin-C at the subepithelial connective tissue. E. Collagen producing cells

populate gingival connective tissue. Representative image of immunostaining of type I

procollagen in human gingival tissue section. Type I collagen is most abundantly produced by

fibroblasts but can also be synthesized by other fibroblast-like cells. F. Immunostaining of CD90

(Thy-1) in human gingiva showing high density of immunopositive cells. In connective tissue,

CD90 can typically be expressed by fibroblasts, MSC, pericytes, myofibroblasts and fibrocytes. E:

epithelium; CT: connective tissue.

Page 62 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

63

63

Table 1. Comparison of cell line markers among fibroblasts (FBLs), gingival stem cells

(GSCs), bone marrow stem/stromal cells (MSCs), pericytes, myofibroblasts (MyoFBLs) and

fibrocytes. For references, see the text.

Markers/cell types FBLs GSCs MSCs Pericytes MyoFBLs Fibrocytes

Cell surface molecules

CD10 - Variable + +

CD11b - - +

CD13 + + + +

CD14 - - - -

CD29 + + + + + +

CD31 - - - + (low)

CD34 - - - - - +

CD40 - -

CD43 - +

CD44 + + + + + +

CD45 - - - - - +

CD56 - -

CD73 + + + + +

CD80 - -

CD86 - -

CD90 + + + + + -

CD105 + + + + + +

CD106 + (low) + (high) - -

CD108 + +

CD109 + +

CD117 +/- - - -

CD133 - - -

CD144 -

CD146 + (low) +(low) + (high) + (high)

Page 63 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

64

64

CD164 + (low) + + +

CD166 Variable + + -

CD200 - - - (low)

CD271 Variable - + +

CD318 + +

CD324 - - - -

CD326 - -

CD340 + +

CD344 - -

CD349 + +

CCR2 Variable

CRCX4 +

HLA I + +

HLA II - - - -

HLA-ABC + +

HLA-DR - - - +

NG2 + +

PDGF-Rβ + +

PPARg -

STRO-1 + (low) + + -

STRO-3 + (low) + (high)

SSEA-4 + (low) + + +

3G5 - + +

Cytoskeletal and cytoplasmic proteins and enzymes

-SMA Variable - + + + Variable

Fibronexus +

FSP-1 (S100A4) + +

LSP-1 - +

MyoD -

Vimentin + (high) + + variable variable + (low)

Prolyl-4-hydroxylase + + + +

Alkaline phosphatase + - + +

Secreted molecules

Page 64 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

65

65

Bone sialoprotein (BSP) -

FGF-2 (bFGF) + +

Fibronectin + + + -

Collagen I + (high) + + + + + (low)

Collagen II - - - -

Collagen III + (high) + + + + (low)

MMP-9 - +

Neurofilament -

Osteocalcin (OC) - -

Osteonectin (SPARC) + +

Page 65 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

66

66

Table 2. Comparison of key phenotypic differences of gingival and skin connective tissue

cells. See text for references.

Adult Gingival Cells Adult Skin Cells

Originate mostly from neural crest Originate mostly from mesoderm

Different gene expression profile and site-specific HOX gene expression

Quantitative and qualitative differences in production of ECM molecules

Differences in integrin-family ECM receptors

Different interactions with ECM proteins

Different production and response to inflammatory mediators

Fast remodeling of a fibrin lattice Slower remodeling of a fibrin lattice

Fast invasion into a collagen matrix Slower invasion into a collagen matrix

Fetal-like phenotype that associates with fast

wound healing without scar formation

Phenotype associates with slower wound

healing and scar formation

Page 66 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

67

67

Table 3. Comparison of human bone marrow (MSCs) and various oral stem cells.

Origin Isolation technique Positive

markers

Negative

markers

Differentiation

potential

Reference

MSCs Human

bone

marrow

Adhesion properties

and

CFU-F

STRO-1

SSEA-4

CD29

CD44

CD73

CD90

CD105

CD146

VCAM1C

Coll1

Coll3

ON

FGF2

CD11b

CD14

CD34

CD45

HLA-DR

Myod

NF

Coll2

OC

BSP

OP

PPARg

In vitro: osteo-, adipo-,

and chondrogenic,

hepatocyte, skeletal

muscle, myocardial,

tendon, smooth muscle

and endothelial cell,

neuronal and glial

differentiation.

In vivo: Osteo- and

chondrogenic,

hepatocyte, muscle and

endothelial cell

differentiation.

Clinical trials for

treatment in humans:

Osteogenesis

Imperfecta, urinary

incontinence (smooth

muscle), erectile

dysfunction

(endothelium), cartilage

regeneration,

myocardial infarction,

liver regeneration.

(49, 50, 57,

61)

DPSCs

Dental

pulp stem

cells

Human

tooth dental

pulp

Enzymatic digestion

and CFU-F

SSEA-4

STRO-1

CD10

CD29

CD44

CD49a

CD14

CD34

CD45

MyoD

NF

Coll2

In vitro: Osteo- and

adipogenic, hepatocyte,

myocyte and neuronal

differentiation.

In vivo: dentine-pulp

complex formation,

50, 62, 65,

73)

Page 67 of 72 Page 68 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

68

68

CD73

CD90

CD105

CD117

CD146

VCAM1

Coll1

Coll3

OC

ON

FGF2

BSP

OP

PPARg

osteogenic and

adipogenic

differentiation,

oligodendrocyte and

neuronal differentiation.

No chondrogenic

differentiation.

SHEDs

Stem cell

from

human

exfoliated

deciduous

tooth

Human

exfoliated

deciduous

tooth pulp

Enzymatic digestion STRO-1

CD146

ALP

MEPE

FGF2

VEGFR1

Endostatin

In vitro: adipo-,

dentino-, osteogenic and

neuronal differentiation.

In vivo: bone formation,

odontoblasts and

endothelial

differentiation.

(52, 66, 67)

SCAPs

Stem cells

from apical

papilla

Human

apical

papilla of

immature

permanent

teeth

Enzymatic digestion

and CFU-F

STRO-1

ALP

CD24

CD29

CD73

CD90

CD105

CD106

CD146

CD166

CD18

CD34

CD45

CD150

In vitro: osteo-, adipo-,

dentogenic and neuronal

differentiation.

In vivo: tooth root and

periodontal ligament

formation.

(54, 68)

DFSCs

Dental

follicle

stem cell

Human

dental

follicle of

impacted

3rd

molars

Enzymatic digestion

and CFU-F or

SSEA4 sorting

SSEA-4

OCT4

CD90

CD56

Tuj1

TRAI-60

CD133 In vitro: osteo-, adipo-,

myo-, cementogenic and

neuronal differentiation.

In vivo: periodontal

ligament, skeletal and

adipose tissue

(53, 69, 70,

73)

Page 69 of 72 St

em C

ells

and

Dev

elop

men

tG

ingi

va a

s a

Sour

ce o

f St

em C

ells

with

The

rape

utic

Pot

entia

l (do

i: 10

.108

9/sc

d.20

13.0

015)

Thi

s ar

ticle

has

bee

n pe

er-r

evie

wed

and

acc

epte

d fo

r pu

blic

atio

n, b

ut h

as y

et to

und

ergo

cop

yedi

ting

and

proo

f co

rrec

tion.

The

fin

al p

ublis

hed

vers

ion

may

dif

fer

from

this

pro

of.

69

69

TRAI-80

Flk1

SMA

p75

formation.

PDLSCs

Periodontal

ligament

stem cells

Human

tooth

periodontal

ligament

Enzymatic digestion

and CFU-F and/or

STRO-1/CD146

sorting

STRO-1

SSEA-4

CD105

CD146

CD166

CD31

CD34

CD45

CD117

In vitro: osteo-, adipo-,

chondro- and

dentinogenic

differentiation.

In vivo: periodontal

repair with periodontal

ligament and cementum-

like tissue formation.

(15, 43)

OMLPCs

Oral

mucosal

lamina

propria

cells

Human and

rat buccal

mucosa

Enzymatic digestion

and differential

adhesion on

fibronectin and CFU-

F (human) or nestin

immunoselection

(rat)

CD29

CD44

CD73

CD90

CD105

CD166

Oct4

CD34

CD45

In vitro: osteo-, adipo-,

chondro-, neuronal and

glial differentiation.

In vivo: bone like tissue

(47, 56, 71,

74, 76)

GSCs

Gingival

stem cells

Human

gingiva

Explant culture

followed by CFU-F

or enzymatic

digestion and CFU-F

STRO-1

SSEA-4

CD29

CD44

CD73

CD90

CD105

CD146

CD34

CD45

CD117

CD200

HLA-DR

In vitro: osteo-, adipo-,

chondrogenic and

endodermal and

neuronal-like

differentiation.

In vivo: collagen rich

connective tissue and

bone formation.

(15, 49, 55,

58, 59, 60,

72, 75)

Page 70 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

70

70

Table 4. Special characteristics of human gingiva and gingival fibroblasts and MSCs that

can be beneficial for their use in various tissue regeneration modalities.

Gingiva in vivo

Easy access and low morbidity after tissue biopsy

Mild and short inflammatory response to wounding

Fast wound re-epithelialization and closure

Scarless wound healing response

Distinct composition of ECM

Distinct regulation of epithelial phenotype by connective tissue

Majority of the connective tissue cells originate from the neural crest

Gingival cells

Neural crest origin

Fetal-like phenotype

Distinct phenotypical properties

Potent immunomodulatory properties

Produce factors that promote vasculogenesis

Produce factors that promote epithelialization

Fibroblast cultures contain MSCs

Possess capacity for multipotent differentiation into mesodermal lineages and neural-like

differentiation

Efficient source to produce iPSCs

Page 71 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.

71

71

Page 72 of 72

Stem

Cel

ls a

nd D

evel

opm

ent

Gin

giva

as

a So

urce

of

Stem

Cel

ls w

ith T

hera

peut

ic P

oten

tial (

doi:

10.1

089/

scd.

2013

.001

5)T

his

artic

le h

as b

een

peer

-rev

iew

ed a

nd a

ccep

ted

for

publ

icat

ion,

but

has

yet

to u

nder

go c

opye

ditin

g an

d pr

oof

corr

ectio

n. T

he f

inal

pub

lishe

d ve

rsio

n m

ay d

iffe

r fr

om th

is p

roof

.