You are on page 1of 19

Biol. Rev. (2021), 96, pp. 357–375.

357
doi: 10.1111/brv.12659

New insights on the reparative cells in bone


regeneration and repair
Shuo Huang , Min Jin, Nan Su* and Lin Chen*
Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and
Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University),
10 Changjiang zhi Road, Yuzhong District, Chongqing, China

ABSTRACT

Bone possesses a remarkable repair capacity to regenerate completely without scar tissue formation. This unique char-
acteristic, expressed during bone development, maintenance and injury (fracture) healing, is performed by the reparative
cells including skeletal stem cells (SSCs) and their descendants. However, the identity and functional roles of SSCs remain
controversial due to technological difficulties and the heterogeneity and plasticity of SSCs. Moreover, for many years,
there has been a biased view that bone marrow is the main cell source for bone repair. Together, these limitations have
greatly hampered our understanding of these important cell populations and their potential applications in the treatment
of fractures and skeletal diseases. Here, we reanalyse and summarize current understanding of the reparative cells in bone
regeneration and repair and outline recent progress in this area, with a particular emphasis on the temporal and spatial
process of fracture healing, the sources of reparative cells, an updated definition of SSCs, and markers of skeletal stem/
progenitor cells contributing to the repair of craniofacial and long bones, as well as the debate between SSCs and peri-
cytes. Finally, we also discuss the existing problems, emerging novel technologies and future research directions in
this field.

Key words: skeletal stem cells, suture stem cells, mesenchymal stem cells, mesenchymal stromal cells, MSCs, SSCs, frac-
ture healing, bone repair, bone maintenance, craniofacial suture

CONTENTS

I.Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
II.Processes of fracture healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
III.Definition of skeletal stem cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361
IV. Sources of cells for long bone repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361
(1) Periosteum-, endosteum-, and bone marrow-derived cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .361
(2) Muscle-derived stem/progenitor cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .362
(3) Cells from the circulatory system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .362
V. Sources of cells for craniofacial bone repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363
VI. Sources of osteoblasts for bone maintenance and repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363
VII. Markers of skeletal stem/progenitor cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 364
(1) Prrx1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .364
(2) Ctsk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .364
(3) Gli1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .366
(4) Axin2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .367
(5) αSMA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .367
(6) Cxcl12 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .367
(7) Mx1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .368

* Authors for correspondence (Tel: +0086 23 68757551; E-mail: linchen70@163.com); (Tel: +0086 23 68757553; E-
mail: sunansyd@163.com)

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


358 Shuo Huang et al.

(8) Grem1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .368


(9) Sox9, Col2 and aggrecan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .368
VIII. The debate between pericytes and mscs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
IX. Future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 370
X. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371
. Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371
XI. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371

I. INTRODUCTION and their descendants in skeletal development, maintenance


and repair remain uncertain due to technological difficulties
Bone possesses an extraordinary regeneration capacity and the heterogeneity and plasticity of SSCs. Moreover, for
expressed during injury (fracture) repair, as well as during many years, there has been a biased view that bone marrow
bone development and continuous remodelling throughout is the main cell source for bone repair. Such uncertainties
adult life (Dimitriou et al., 2011). In the clinical setting, the greatly hamper our understanding of these important cell
most common form of bone regeneration is fracture healing. populations and their potential application in the treatment
Although most fractures can be cured through proper surgi- of fractures and skeletal diseases.
cal reduction and fixation, approximately 10–15% of frac-
ture patients in the United States experience delayed union
or non-union due to various physiological (e.g. age and
smoking) and pathological conditions (e.g. concomitant II. PROCESSES OF FRACTURE HEALING
infections, osteoporosis, diabetes, and osteogenesis imper-
fecta) (Einhorn, 1995; Gruber et al., 2006; Kline et al., 2009; Craniofacial bones are derived from mesodermal and cranial
Colnot, 2011; Abou-Khalil & Colnot, 2014). The success of neural crest cells, whereas the appendicular skeleton is
current therapies on such abnormal fracture healing is still derived from the lateral plate mesoderm and the axial skele-
very limited (Colnot, 2011; Abou-Khalil & Colnot, 2014; ton is derived from the paraxial mesoderm (Senarath-Yapa
Senarath-Yapa et al., 2014). Thus, understanding the cellular et al., 2014; Berendsen & Olsen, 2015). Fractures of craniofa-
and molecular mechanisms of bone regeneration and repair cial bones heal through IO (Zhao et al., 2015; Ono &
is important for the development of treatments and novel Kronenberg, 2016) (Fig. 2). In long bones, microfractures
therapeutic strategies. or mechanically stable fractures heal mainly through IO,
Numerous studies have indicated that the processes of whereas the majority of severe fractures are repaired through
bone repair are similar to those of bone development. The a combination of IO and EO, with the approach determined
two major categories of vertebral bones are the flat bones mainly by the mechanical stability of the fracture site. For
in the skulls and the long bones that comprise most bones example, after a complete transverse fracture of a long bone,
in the axial and appendicular skeleton (Dirckx, Van Hul, & rigid fixation using fixators such as bone plates will result in
Maes, 2013). All these bones are formed by two mechanisms: complete stability, and allow healing mainly through IO
long bones are formed following the formation and resorp- (Bahney et al., 2015). If the fracture is not rigidly reduced to
tion of cartilaginous templates through a process called enable some motion, such as fixation by an intramedullary
endochondral ossification (EO), and the majority of craniofa- pin, or is not subjected to fixation, it heals mainly through
cial bones develop through intramembranous ossification EO accompanied by IO in a location-specific manner
(IO), in which stem cells differentiate directly into osteoblasts (Bahney et al., 2015) (Fig. 1). Along the periosteal surface of
that lay down the mineralized matrix (Senarath-Yapa the distal ends of the fractured bone (Fig. 1), where there is
et al., 2014; Ono & Kronenberg, 2016). While bone injuries no motion, periosteal cells form bones via IO, with a large
are also repaired through EO and/or IO (Zhao et al., 2015; cartilaginous callus formed around the fracture gap (Fig. 1);
Ono & Kronenberg, 2016), most pathways involved in bone subsequently, bone tissue is formed through the more com-
development are activated in the repair processes (Vortkamp plex process of EO (Bahney et al., 2015).
et al., 1998; Ferguson et al., 1999; Gerstenfeld et al., 2003), The processes of fracture healing normally include
including homeotic genes in pathways that control bone three distinct but overlapping stages: the inflammatory stage,
development (Bais et al., 2009). the repair stage and the remodelling stage (DePalma
Bone regeneration and repair is mainly performed by the et al., 1972; Burchardt & Enneking, 1978) (Fig. 1). In the
reparative cells including skeletal stem cells (SSCs) and their inflammatory stage, upon injury, blood vessels, bone and
descendants in a definable spatiotemporal manner. SSCs the surrounding soft tissues are disrupted, and a haematoma
possess self-renewal capacity and can be activated rapidly develops at the fracture site within the first few hours to days
in response to the injury, differentiating into bone and carti- (Abou-Khalil & Colnot, 2014). The haematoma serves as an
lage (or bone only) within the bone callus that will go through important source of haematopoietic cells, neutrophils/
a remodelling process to be replaced by new bone tissue macrophages, and platelets that are involved in the initiation
(Figs 1, 2). However, the identity and functional roles of SSCs of the inflammatory response (Mountziaris & Mikos, 2008)

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 359

Fig 1. Repair processes of an unstable long bone fracture. Unstable long bone fractures are normally repaired through both
intramembranous ossification (IO) and endochondral ossification (EO). Repair initiates after the inflammatory reaction, and
includes a soft and hard callus formation stage. In the inflammatory stage, upon injury, blood vessels, bone and the surrounding
soft tissues are disrupted, and a haematoma develops at the fracture site within the first few hours to days. The haematoma serves
as an important source of neutrophils, macrophages, and platelets that are involved in the initiation of the inflammatory response.
The outer layer of the periosteum gradually reconstitutes to contain the haematoma, while cells from the inner cambial layer
initiate and start to proliferate (periosteal reaction). The initial stage of repair begins from the periphery of the stable and well-
vascularized cortex bone at the distal ends of the fracture site, where cells from the inner layer of the lifted periosteum continually
condense, proliferate and differentiate into woven bone, resulting in the formation of a bone collar via IO. In the soft callus
formation stage, around the fracture gap, the mechanical instability of the fracture ends together with hypoxia of the local
microenvironment due to the disrupted blood supply lead to the formation of a cartilaginous mass. Chondrocytes progressively
mature into hypertrophic chondrocytes (HCs). HCs secrete collagen X in the transition zone, where cartilage will be replaced by
bone tissue and express a variety of active molecules that promote mineralization and vascular invasion into the callus from the
distal ends of the fracture site and the reconstituted periosteum at the outer margin of the callus. Along with angiogenesis,
osteoblasts derived from osteoprogenitors secrete the osteoid at the edges of cartilage tissues, leading to subsequent mineralization.
In the hard callus formation stage, the entire cartilage is continuously replaced by woven bones, and a bony framework is
eventually constructed across the fracture gap.

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


360 Shuo Huang et al.

Fig 2. Maintenance and repair of craniofacial bone. Craniofacial bones are coated by the periosteum and dura/endosteum and
contain interspersed bone marrow. Sutures between craniofacial bones are composed of two osteogenic fronts with a strip of
mesenchyme between them. Specific postnatal suture stem cells (SuSCs) expressing glioma-associated oncogene 1 (Gli1), Axin2,
paired related homeobox 1 (Prrx1) and cathepsin K (Ctsk) reside within the suture mesenchyme and have been found to play a
predominant role in craniofacial bone maintenance and fracture repair in adult mice.

(Fig. 1). A variety of inflammatory cytokines, such as tumor the action of muscle contraction, cartilage in the callus is orga-
necrosis factor alpha (TNF-α), interleukin-1a (IL-1a), IL-1b, nized to act as a bidirectional growth plate, which in turn gen-
IL-6, and IL-18 (Abou-Khalil & Colnot, 2014), and growth erates force in the opposing direction that drives the natural
factors, such as platelet derived growth factor (PDGF), vascu- reduction of the fractured bone (Rot et al., 2014). In the
lar endothelial growth factor (VEGF), bone morphogenetic cartilaginous callus, chondrocytes progressively mature into
proteins (BMPs), fibroblast growth factors (FGFs), transform- hypertrophic chondrocytes (HCs). HCs secrete collagen X in
ing growth factor beta (TGFβ), and insulin like growth fac- the transition zone (Fig. 1), where cartilage will be replaced
tors (IGFs) (Ferguson et al., 1998; Gerstenfeld et al., 2003; by bone tissue, and express a variety of active molecules,
Schindeler et al., 2008), are released at the fracture site, such as VEGF (Gerber et al., 1999; Colnot & Helms, 2001;
resulting in the formation of vascular tissue and the recruit- Zelzer et al., 2004), placental growth factor (PIGF) (Maes
ment of inflammatory cells and reparative cells that are et al., 2006), BMP (Yu et al., 2010), matrix metalloproteinase
essential for bone regeneration and proper fracture healing 13 (MMP13) (Behonick et al., 2007) and alkaline phosphatase,
(Gerstenfeld et al., 2001; Kon et al., 2001; Cho, Gerstenfeld, & which promote mineralization as well as vascular invasion into
Einhorn, 2002; Cho et al., 2007; Yang et al., 2007; Einhorn & the callus from the distal ends of the fracture site and the recon-
Gerstenfeld, 2015). Thereafter, the outer layer of the perios- stituted periosteum at the outer margin of the callus (Kurdy,
teum is gradually reconstituted to contain the haematoma, Weiss, & Bate, 1996; Hausman, Schaffler, & Majeska, 2001;
while cells from the inner cambial layer initiate and start to Bahney et al., 2015). Furthermore, HCs can also act as a
proliferate (periosteal reaction) (Shapiro, 2008) (Fig. 1). secondary source of osteoblasts (Dy et al., 2012; Yang
In the repair stage, if the fracture heals through IO (includ- et al., 2014a; Zhou et al., 2014b), which will be discussed in
ing craniofacial fracture), skeletal stem/osteoprogenitor cells Section VI. In parallel with angiogenesis, osteoblasts derived
directly differentiate into osteoblasts and lay down the mineral- from osteoprogenitors secrete osteoid at the edges of cartilage
ized matrix. In fractures healed through a combination of IO tissues with subsequent mineralization. In the hard callus for-
and EO, the repair process includes the soft and hard callus for- mation stage, the entirety of the cartilage is continuously
mation stage. The initial stage of repair begins in the periphery replaced by woven bones, and a bony framework is eventually
of the stable and still well vascularized cortex bone at the distal constructed across the fracture gaps (Kalfas, 2001) (Fig. 1).
ends of the fracture site, where cells from the inner layer of the In the remodelling stage, the bony callus continues to be
lifted periosteum continually condense, proliferate and differ- resorbed by osteoclasts, and secondary compact bone is laid
entiate into woven bone, which results in the formation of a down by osteoblasts, which is termed coupled remodelling.
bone collar via IO (Bahney et al., 2015) (Fig. 1). In the soft callus During this stage, the marrow space is re-established and
formation stage, around the fracture gap, the mechanical insta- returns to its original structure as haematopoietic tissue.
bility of the fracture ends together with hypoxia of the local The enhanced vascular flow rate gradually returns to pre-
microenvironment due to a disrupted blood supply leads to injury levels, accompanied by extensive vascular remodelling
the formation of a cartilaginous mass (Fig. 1) that functions as (Melnyk et al., 2008; Holstein et al., 2013). Finally, the healing
a ‘mechanical jack’ (Rot et al., 2014; Bahney et al., 2015). Under bone is gradually restored to its original shape, structure, and

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 361

mechanical strength (Einhorn & Gerstenfeld, 2015). on the known expression of key transcription factors pre-
Although these processes take place consecutively, they over- dicted the cell state hierarchy in vivo. Data from two of these
lap substantially and involve a continuum of changing cell papers supported the classic concept that MSCs/SSCs pos-
populations and signalling processes within the regenerating sess the ability to differentiate into osteoblasts, chondrocytes
tissue. and adipocytes via three hierarchical differentiation trajecto-
ries in vivo (Wolock et al., 2019; Baccin et al., 2020). Data from
the other three papers showed that MSCs/SSCs might only
maintain the production of osteogenic- and adipogenic-
III. DEFINITION OF SKELETAL STEM CELLS lineage cells through bilineage differentiation pathways dur-
ing the steady-state differentiation process in vivo (Tikhonova
In the past, the stem cells regulating bone development, main- et al., 2019; Matsushita et al., 2020; Zhong et al., 2020). These
tenance and repair were usually termed mesenchymal stem/ findings refine our understanding of the cellular architecture
stromal cells (MSCs). MSCs are defined as multipotent stem of bone marrow cells, however, the inherent differentiation
cells that have the potential to self-renew with maintained potential of SSCs is still controversial, and the definition of
properties and differentiate into a variety of specialized cell SSCs needs to be further refined.
types, such as osteoblasts, chondrocytes, adipocytes, myocytes
and neurons (Pittenger et al., 1999; Caplan & Bruder, 2001;
Sanchez-Ramos, 2002). Due to their heterogeneity, there is
no single specific marker that can be used to characterize IV. SOURCES OF CELLS FOR LONG BONE
MSCs, and a panel of markers has been outlined by the Inter- REPAIR
national Society for Cellular Therapy (ISCT) and widely
adopted by researchers. The ISCT defines MSCs as culture- During long bone fracture healing, reparative cells could be
expanded cells that adhere to tissue culture plastic, retain recruited to the fracture site from both local tissues and the
trilineage differentiation capability (bone, cartilage, and fat) circulatory system. Local tissues include the periosteum, end-
in vitro, express the surface markers cluster of differentiation osteum, bone marrow, and surrounding soft tissues in close
73 (CD73), CD90 and CD105, and lack expression of the proximity to the bone, such as tendon, fat and muscle
CD34, CD45, CD14, CD11b, CD79α/CD19 and human leu- (Colnot, 2011). Cells obtained from systemic recruitment
kocyte antigen-DR (HLA-DR) surface molecules (Dominici include: (i) endothelial cells derived from blood vessels; (ii)
et al., 2006). However, with the development of biotechnologies stem/progenitor cells that exist in or are mobilized to the cir-
such as in vivo lineage tracing and single-cell RNA sequencing culatory system after injury; (iii) pericytes; and (iv) monocyte/
(scRNA-seq), a number of new markers have been proposed macrophage-lineage cells. The first three types possess the
to redefine/purify subpopulations of MSCs. Currently, it is ability to differentiate/transdifferentiate into chondrocytes
thought that distinct subpopulations of MSCs may have differ- and osteoblasts (Sempuku et al., 1996; Pittenger et al., 1999;
ent/overlapping phenotypes and functions. Based on scRNA- Lee et al., 2000; Zheng et al., 2006; Bi et al., 2007; Liu, Schin-
seq data, mouse stem cells in bone tissues were redefined as deler, & Little, 2010; Wosczyna et al., 2012; Shah et al., 2013);
‘mouse skeletal stem cells’ (mSSCs) that possess the ability to thus, it is hard to distinguish which sources of cells predomi-
differentiate into osteoblasts, chondrocytes and stromal cells nantly contribute to the fracture callus.
and are positive for integrin alpha V but negative for CD45,
lymphocyte antigen-76 (Ly-76/Ter119), tyrosine-protein
kinase receptor TIE-2 (Tie2), CD90, Ly-51 (BP-1/6C3) and (1) Periosteum-, endosteum-, and bone marrow-
derived cells
CD105 (Chan et al., 2015; Gulati et al., 2018). Chan et al. (2018)
further redefined human skeletal stem cells (hSSCs) as multipo- The periosteum is a highly vascularized connective tissue lining
tent stem cells that can differentiate into bone, cartilage, and the outer surface of bone. It is a bilayer membrane comprising:
stroma but not fat. hSSCs are present in fetal and adult bone (i) the outer fibrous layer, which consists of fibroblasts, collagen,
tissues and can also be obtained from induced pluripotent elastic fibres and microvessels (Popowics, Zhu, & Herring, 2002;
stem cells or BMP2-treated human adipose stroma (Chan Allen, Hock, & Burr, 2004) that provide the attachment points
et al., 2018). hSSCs are positive for podoplanin (PDPN), for tendons, ligaments and muscles and is thought to serve
CD73, and CD164 but negative for CD31, CD45, Tie2, mainly a structural and mechanical role (Yiannakopoulos
CD235a, and CD146 (Chan et al., 2018). These new definitions et al., 2008); and (ii) the inner layer, which is also known as the
and classification methods for SSCs have been adopted and cambium layer and consists of smaller and more isodiametric
applied by many researchers (Debnath et al., 2018; Mizuhashi fibroblasts, sympathetic nerves, osteoblasts and periosteum-
et al., 2018; Ransom et al., 2018). derived SSCs (Ozaki et al., 2000; Allen et al., 2004; Zhang
Five recent papers analysed the transcriptional landscape et al., 2005; Colnot, 2009; Colnot, Zhang, & Knothe
of mouse non-haematopoietic bone marrow cells using Tate, 2012; Matthews et al., 2014; Yukata et al., 2014).
scRNA-seq technology and computationally identified many It is generally accepted that cells from the periosteum form
cell clusters corresponding to distinct cell types or stages of the fracture callus and play the predominant role in long
differentiation. Pseudotemporal cell trajectory analysis based bone fracture healing (Zhang et al., 2005; Xie et al., 2008;

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


362 Shuo Huang et al.

Colnot, 2009; Kawanami et al., 2009; Yu et al., 2010), while the initial fracture site and decreased with the repair process
cells from the periosteum, endosteum, and bone marrow (Liu et al., 2011). Therefore, muscle might only contribute to
might collaborate with each other to form the new cortex bone bone fracture healing as an alternative source of cells when
connecting the graft and the host cortex (Colnot, 2009). Evi- the periosteum is disrupted.
dence has shown that periosteum activation occurs very rap-
idly within 24–48 h in response to fracture, which closely
coincides with local cell proliferation (Iwasaki, Le, & (3) Cells from the circulatory system
Helms, 1997). Mechanical disruption of the periosteum delays
healing (Utvag, Grundnes, & Reikeraos, 1996; Guichet It was reported that endothelial cells are able to transdiffer-
et al., 1998; Ozaki et al., 2000). entiate into chondrocytes and osteoblasts during heterotopic
Although there is currently a lack of periosteum-specific ossification (Medici et al., 2010). Endothelial cells were found
cyclization recombination enzyme (Cre) recombinants for in increased numbers in the circulatory system following
lineage tracing, Colnot (2009) compared in detail the distinct fracture. However, a genetic lineage-tracing approach
contributions to fracture healing of the periosteum, endos- proved that endothelial cells, which were represented by
teum, and bone marrow using tibia unicortical bone graft Tie2-expressing cells, do not appear to transdifferentiate into
transplantation approaches. Live bone grafts from geneti- cartilage or bone within the fracture callus and make no
cally labelled mice (Rosa26) containing periosteum and/or direct contribution to the fracture callus (Lu, Marcucio, &
endosteum were transplanted into the fracture site of the Miclau, 2006; Wang et al., 2011b). Therefore, there might
wild-type host (Colnot, 2009). The results revealed that peri- be a fundamental difference between heterotopic ossification
osteal cells are the major contributors to the fracture callus, and fracture healing (Liu et al., 2011).
giving rise to osteoblasts and osteocytes in a stabilized frac- Blood vessels carry cells from various sources systemically
ture and to chondrocytes, osteoblasts and osteocytes in a to the fracture site. To evaluate the contribution of systemic
non-stabilized fracture. Bone marrow and endosteal cells recruitment of skeletal stem/progenitor cells during bone
always give rise to osteoblasts and osteocytes at the endosteal repair, Taguchi et al. (2005) systemically injected green fluo-
surface and within the bone marrow cavity (Colnot, 2009). rescent protein (GFP)-labelled bone marrow cells into wild-
Moreover, periosteal cells have the potential to differentiate type mice, and found that GFP-expressing cells in the frac-
into cartilage and bone in the environment of the bone mar- ture callus were not incorporated into the new bone as oste-
row cavity, while cells from the endosteum and bone marrow ocytes. Another study observed systemic mobilization and
mainly differentiate into bone but do not support cartilage recruitment of osteoprogenitors to the fracture site via the
formation in the environment of the periosteum during peripheral circulation (Shirley et al., 2005), but these cells
non-stabilized fracture healing (Colnot, 2009). These data do not seem to differentiate into osteocytes in new bone.
suggest that the distinct contributions to fracture healing of Our previous studies also showed that systemic transplanted
periosteum-, endosteum-, and bone marrow-derived cells allogeneic MSCs are able to migrate into the fracture site,
might not be related to extrinsic differences in their environ- but few were found to participate in new bone formation
ment but to intrinsic cellular differences among these tissues. (Huang et al., 2015a; Huang et al., 2015b). Therefore,
the exact contribution of systemically recruited skeletal
stem/progenitor cells remains to be determined.
(2) Muscle-derived stem/progenitor cells Pericytes are closely associated with blood vessels. They
Skeletal muscles and bone are closely linked and coordinated can either be activated locally or transported from distant
to enable locomotion and posture control (Shah et al., 2013). tissues to the injury site and might participate in bone repair
Previous studies have demonstrated that muscle-derived (da Silva Meirelles, Caplan, & Nardi, 2008; Grcevic
stem/progenitor cells are capable of forming bone tissues et al., 2012) (Fig. 1). However, the contribution of pericytes
(Liu et al., 2010; Wosczyna et al., 2012; Shah et al., 2013), is still controversial (see Section VIII).
and ectopic bone formation can be induced in muscle by Monocyte/macrophage-lineage cells are crucial cell
injection of BMPs (Tachi et al., 2011; Wosczyna sources for bone repair. It was reported that macrophages
et al., 2012). However, using myogenic differentiation can be recruited from the circulatory system to the fracture
(MyoD) conditional reporter mice (MyoD-Cre+:Z/AP+), site, where they undergo phenotypic and functional changes
Liu et al. (2011) revealed that the intact periosteum might to coordinate repair (Wynn & Vannella, 2016). Depletion of
act as a physical barrier to prevent the involvement of macrophages delays fracture healing (Vi et al., 2015; Alexan-
muscle-derived stem/progenitor cells in fracture healing. In der et al., 2017). Heterochronic parabiosis experiments
a mouse tibial defect model or closed tibial fracture model, revealed that macrophages from old mice slowed down the
which produces limited damage to the adjacent soft tissues healing process in young mice, while young macrophages
while leaving the periosteum largely intact, no or a minimal or their secretions [e.g. low-density lipoprotein receptor-
number of MyoD-lineage cells were found in the fracture related protein 1 (Lrp1)], could rejuvenate fracture repair
callus. In a tibial open fracture model, MyoD-lineage cells in old mice (Vi et al., 2018). Evidence has also shown that
participated in all fracture healing stages; however, the pro- osteoclast precursors could migrate to the bone injury site
portion of MyoD-positive cells was lower in areas away from through the blood circulation and begin to participate in

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 363

the remodelling of bone on day 7 following bone injury in These results reveal that sutural cells rather than periosteal
mice (Yahara et al., 2020). cells play the predominant role in craniofacial bone repair,
Thus, systemically circulating cells recruited to the bone which might be related to differences in the populations of
injury site are mainly macrophages and osteoclast precursors skeletal stem/progenitor cells. Even so, the craniofacial
in the monocyte/macrophage lineages; systemically periosteum is rich in blood vessels that not only deliver sig-
recruited skeletal stem/progenitor cells are few, and their nals, nutrients and oxygen but also transfer suture-derived
contribution remains to be determined. reparative cells to the injury site; thus, they might still play
important roles in craniofacial bone repair.

V. SOURCES OF CELLS FOR CRANIOFACIAL


BONE REPAIR VI. SOURCES OF OSTEOBLASTS FOR BONE
MAINTENANCE AND REPAIR
Craniofacial bones are covered by the periosteum and dura/
endosteum and contain interspersed bone marrow Normally, osteoblasts have three possible fates: (i) to become
(Robey, 2011; Yang, Zhang, & Gangolli, 2014b; Zhao osteocytes embedded in mineralized bone matrix; (ii) to
et al., 2015) (Fig. 2). The craniofacial periosteum was undergo programmed cell death; or (iii) to transform into
reported to contain stem cells and osteoprogenitors (Kanou quiescent bone lining cells (BLCs) that are capable of being
et al., 2005; Zhang et al., 2005; De Bari et al., 2006; Brey reactivated and contributing to mature osteoblasts during
et al., 2007) and therefore was thought to share the same turn- modelling/remodelling (Hock et al., 2001; Franz-Odendaal,
over and repair mechanisms as those of the long bone perios- Hall, & Witten, 2006) (Fig. 1). Genetic pulse-chase experi-
teum (Ochareon & Herring, 2011; Pagni et al., 2012). ments using Ocn-CreER;R26-YFP mice revealed that
However, recent evidence has shown that the craniofacial mouse mature osteoblasts might be short-lived non-
periosteum might have distinct skeletal progenitors and bio- replicative cells (Park et al., 2012). Mature osteoblasts labelled
logical characteristics compared to those of the long bone by osteocalcin (Ocn) turned over rapidly and were continu-
periosteum (Leucht et al., 2008; Lin et al., 2014; Wilk ously replaced by other cell populations in 6- to 8-week-old
et al., 2017). Sutures between craniofacial bones might play mice. The number of Ocn+ endosteal and periosteal osteo-
a predominant role in craniofacial bone development, main- blasts reached a peak 14 days after induction, followed by a
tenance and repair (Mao & Prockop, 2012; Zhao et al., 2015; significant decrease of 50% by day 30, with the number grad-
Maruyama et al., 2016; Wilk et al., 2017). ually decreasing to the basal level by day 60 in both femurs
Sutures are composed of two osteogenic fronts with a strip and calvaria. By contrast, bone-embedded Ocn-labelled
of mesenchyme between them (Fig. 2). Most sutures in mice osteocytes peaked in number at day 30 after induction and
remain patent throughout their lifetime, whereas human cra- then dropped over time (Park et al., 2012). These results
nial sutures normally fuse in adults between 20 and 30 years suggest that the half-life of mouse mature osteoblasts is
old, and facial sutures fuse after 50 years old (Badve only 30 days in vivo; thus they need to be continuously
et al., 2013; Zhao et al., 2015). Utilizing the bone transplanta- replenished during bone maintenance and repair.
tion method, Zhao et al. (2015) proved that sutures rather than Osterix (Osx) is expressed earlier than osteocalcin in the oste-
craniofacial periosteum and dura possess regeneration capac- oblast lineage, and is considered a marker of pre-osteoblasts
ity. They transplanted a piece of GFP-labelled calvarial bone (osteogenic precursors) (Rodda & McMahon, 2006). Osx-
containing the sagittal suture but without the periosteum and labelled cells were reported to be the origin of trabecular osteo-
dura into nude mice. After 1 month, the transplanted suture blasts and osteocytes in developing bones (Maes et al., 2010).
vigorously generated new GFP-labelled periosteum, dura Using a genetic pulse-chase method similar to that described
and bones in the transplant (Zhao et al., 2015), while transplan- above, Park et al. (2012) found that the average number of
tation of calvarial bone containing intact periosteum and dura Osx-labelled cells gradually increased over 30 days following
but without sutures failed to grow (Zhao et al., 2015). Further induction but then decreased to the basal level after 90 days
studies have shown that the midline of the suture mesenchyme in both femurs and calvaria, implying that Osx-labelled cells
contains a distinctive stem cell population known as suture were not durable and did not represent a self-renewing pool
stem cells (SuSCs) (Fig. 2), which possess long-term self- of osteoprogenitors.
renewing, colony-forming and multidifferentiation abilities BLCs are post-mitotic flat osteoblast-lineage cells covering
in vitro and can give rise to osteogenic fronts, periosteum, dura non-remodelling (quiescent) bone surfaces and are character-
and bone during natural bone turnover in vivo (Ouyang ized by their morphology and location without specific
et al., 2013; Zhao et al., 2015; Maruyama et al., 2016; Wilk markers (Miller et al., 1989; Matic et al., 2016) (Fig. 1). BLCs
et al., 2017). When a craniofacial injury occurs, SuSCs are rap- can be induced to proliferate and/or differentiate into mature
idly activated within 24 h and migrate into the injury site to osteoblasts by parathyroid hormone (PTH), FGF2, inhibition
proliferate and differentiate into osteocytes embedded in the of sclerostin or high doses of γ-radiation (Dobnig &
bone plate, directly contributing to craniofacial bone repair Turner, 1995; Kim et al., 2012; Turner et al., 2013; Delgado-
(Zhao et al., 2015; Maruyama et al., 2016) (Fig. 2). Calle & Bellido, 2017). BLCs display gene expression profiles

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


364 Shuo Huang et al.

different from those of osteoblasts, such as increased levels of and periosteum-derived cells, and bone marrow cells were
macrophage colony-stimulating factor (M-CSF), receptor acti- usually used for cell identification in vitro; and (iv) the lack of
vator of nuclear factor-κ B ligand (RANKL), and MMP13, well-accepted cell surface markers for cell identification.
and share some of the surface markers of MSCs, such as Ly- Below, we summarize some populations of skeletal stem
6A/E (Sca1), CD51 and leptin receptor (Lepr) (Matic and/or progenitor cells involved in craniofacial and/or long
et al., 2016; Matthews et al., 2016). Matic et al. (2016) discov- bone repair with a focus on their specific markers, spatiotem-
ered by accident using dentin matrix protein 1 (Dmp1) poral distribution, stemness, and contributions to bone main-
reporter mice (Dmp1-CreERT;R26-tdTomato) that Dmp1 tenance and repair (summarized in Table 1).
could label BLCs, mature osteoblasts (Col2.3-GFP+) and oste-
ocytes. To remove mature osteoblasts from Dmp1+ cells, they
(1) Prrx1
crossed Dmp1-CreERT;R26-tdTomato mice with Col2.3Δtk
mice, after which Col2.3-GFP+ cells could be selectively elim- Paired related homeobox 1 (Prrx1; also known as MHox or
inated by 16 days of ganciclovir (GCV) treatment (Visnjic Prx1) is usually acknowledged as a marker of SSCs in both
et al., 2001). After ablation, osteoblasts were absent from the craniofacial and long bones. Prrx1 expression is restricted
bone surface, and calcein labelling was minimal; however, to the early limb bud mesenchyme during embryonic devel-
after 21 days of recovery, rapid bone formation appeared, opment in mice (Ono & Kronenberg, 2016). In adult mice,
and the number of Dmp1+ osteoblasts and osteocytes embed- Prrx1-expressing cells are localized immediately outside of
ded in the bone dramatically increased in both cortical and collagen type I alpha 1 (col1a1)-expressing osteoblasts in
trabecular bones, suggesting the replenishment by BLCs of the cambium layer of the long bone periosteum (Ouyang
mature osteoblasts (Matic et al., 2016). Therefore, BLCs might et al., 2013), and very few are localized in the endosteum
be an alternative to SSCs as a source of ‘predetermined’ oste- and bone marrow (Kawanami et al., 2009). Prrx1 is also
ogenic precursors contributing to bone formation in expressed in SuSCs (Ouyang et al., 2013; Wilk et al., 2017),
adult mice. tendon cells (Ouyang et al., 2013) and in the precursors of
HCs are generally considered to be a postmitotic cell type, white adipocytes (Sanchez-Gurmaches et al., 2015) but not
and they undergo programmed cell death through either apo- in skeletal muscles (Logan et al., 2002; Sanchez-Gurmaches
ptotic or autophagic mechanisms (Ahmed et al., 2007; Kroe- et al., 2015). Both calvarial suture- and periosteum-derived
mer et al., 2009; Tsang et al., 2014). However, it has recently Prrx1+ cells could give rise to chondrocytes and osteoblasts
been reported that HCs are able to re-enter the cell cycle in vitro (Kawanami et al., 2009; Ouyang et al., 2013; Wilk
and might undergo asymmetric cell division (Roach, Eren- et al., 2017). Mice with deletion of Prrx1 show limb shorten-
preisa, & Aigner, 1995; Roach & Erenpreisa, 1996), giving rise ing, craniofacial defects, and incompletely penetrant spina
to two daughter cells, one of which undergoes apoptosis, bifida (Martin & Olson, 2000). Prrx1 also inhibits adipogen-
whereas the other survives and subsequently transdifferenti- esis by activating TGFβ signalling (Sanchez-Gurmaches
ates into osteoblasts (Dy et al., 2012; Yang et al., 2014a; Zhou et al., 2015). During long bone fracture healing, which is
et al., 2014b) or dedifferentiates into progenitor-like cells mainly exerted through EO, Prrx1+ cells derived from the
(Song & Tuan, 2004; Bahney et al., 2014). Moreover, trans- periosteum could give rise to cartilage and bone in the frac-
planted cartilage grafts can participate in bone repair ture callus (Kawanami et al., 2009; Wang et al., 2017;
(Bahney et al., 2014), and genetically labelled HCs in donor Duchamp de Lageneste et al., 2018), while Prrx1+ cells in
graft cartilage can become osteoblasts and osteocytes (Yang the sutures contribute to calvarial bone defect healing
et al., 2014a; Tsang, Chan, & Cheah, 2015). Therefore, HCs through IO (Wilk et al., 2017).
might be an important source of osteoblasts during long bone
development and fracture healing through EO.
(2) Ctsk
In summary, mouse mature osteoblasts are considered to
be short-lived non-replicative cells, and they can be replen- Cathepsin K (Ctsk), a lysosomal cysteine protease, was origi-
ished by BLCs, HCs, and skeletal stem/progenitor/precur- nally recognized as a marker of mature osteoclasts both in vivo
sor cells during bone maintenance and repair. and in vitro (Lotinun et al., 2013; Bonnet et al., 2017; Panwar
et al., 2017). Ctsk plays important roles in bone resorption
and remodelling through the degradation of matrix proteins;
inactivation or deletion of Ctsk suppresses bone resorption
VII. MARKERS OF SKELETAL STEM/ (Lotinun et al., 2013; Bonnet et al., 2017; Panwar
PROGENITOR CELLS et al., 2017). Yang et al. (2013) identified a new type of Ctsk+
cells within Ranvier’s groove and termed them Ctsk+ chon-
The identities and functional roles of skeletal stem/ droid progenitors (CCPs). Conditional knockout of tyrosine
progenitor cells remain unclear or controversial due to: (i) phosphatase 2 (SHP2) in Ctsk+ cells resulted in multiple
the multiple sources of cells that intrinsically or extrinsically osteochondromas and enchondromas, implying that Ctsk+
(are induced to) possess trilineage differentiation capability; cells in Ranvier’s groove might possess the functional proper-
(ii) the heterogeneity of SSCs/MSCs; (iii) the fact that most ties of skeletal stem/progenitor cells (Yang et al., 2013). Most
previous studies did not distinguish between bone marrow- recently, Debnath et al. (2018) found that Ctsk not only labels

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 365

Table 1. Markers of skeletal stem/progenitor cells in adult mice

Target cells and their locations in Cell lineages involved in bone


Markers References
adult mice maintenance and repair
Long bones
Prrx1 SSCs in the periosteum, endosteum Periosteal and endosteal cells, Kawanami et al. (2009); Ouyang
and bone marrow, precursors of BMSCs, osteoblasts, osteocytes, et al. (2013); Logan et al. (2002);
white adipocytes, tendon cells, not chondrocytes (in fracture callus) Sanchez-Gurmaches, Hsiao, &
in skeletal muscles Guertin (2015); Wang, Zhang, &
Bikle (2017); Duchamp de
Lageneste et al. (2018)
Ctsk SSCs in the periosteum, endosteum, Periosteal and endosteal cells, Lotinun et al. (2013); Panwar
bone marrow and Ranvier’s BMSCs, osteoblasts, osteocytes, et al. (2017); Bonnet et al. (2017);
groove, osteoprogenitors, chondrocytes (in fracture callus), Yang et al. (2013); Debnath
osteoblasts, osteocytes#, mature osteoclasts et al. (2018)
osteoclasts
Gli1 SSCs in the periosteum, bone Periosteal cells, BMSCs, muscle cells, Shi et al. (2017); Hojo et al. (2012);
marrow* and Ranvier’s groove, adipocytes, osteoblasts, osteocytes, Zhao et al. (2015)
cells in the chondro-osseous chondrocytes
junction immediately beneath the
growth plate, chondroprogenitors*
in the upper layers of the growth
plate, cells in articular cartilage,
incisors and muscles
αSMA/Acta2 SSCs in the periosteum* and bone Periosteal* and endosteal* cells, Morikawa et al. (2002); Nehls,
marrow, periodontal and sutural BMSCs, muscle cells*, osteoblasts, Denzer, & Drenckhahn (1992);
cells, satellite cells, adipose stem osteocytes, chondrocytes (in Baryawno et al. (2019); Kalajzic
cells, vSMCs, pericytes fracture callus) et al. (2008); Grcevic et al. (2012);
Matthews et al. (2014), Matthews
et al. (2016); Roguljic et al. (2013)
Cxcl12 Pericytes, vSMCs and CAR cells in BMSCs, adipocytes in the bone Sugiyama et al. (2006); Matsushita
the bone marrow, adipogenic marrow, osteoblasts and osteocytes et al. (2020); Coutu et al. (2017);
precursors, not osteoblasts or in the trabecula during bone Zhong et al. (2020)
osteocytes maintenance, osteoblasts and
osteocytes in the callus and newly
formed cortical bone during
fracture healing
Mx1 SSCs in the periosteum and Periosteal and endosteal cells, Walkley et al. (2007); Park et al. (2012),
endosteum, mature osteoblasts, BMSCs, osteoblasts, osteocytes, Park et al. (2014)
osteoclasts but not chondrocytes
Mx1 + αSMA SSCs in the periosteum and calvarial Periosteal and sutural cells, Ortinau et al. (2019)
sutures, nearly undetectable in osteoblasts, osteocytes and
bone marrow chondrocytes* (in fracture callus)
Grem1 Periosteal cells*, OCR stem cells in Periosteal cells, reticular marrow Worthley et al. (2015); Mendez-Ferrer
bone marrow, not in the stromal cells, chondrocytes (in the et al. (2010); Zhou et al. (2014a);
perisinusoidal space growth plate and articular Debnath et al. (2018)
cartilage), diaphyseal osteoblasts,
osteocytes, but not adipocytes
Sox9, Col2, Skeletal stem/progenitor cells in the Periosteal cells, BMSCs, He et al. (2017); Shintaku et al. (2011);
aggrecan periosteum and bone marrow, cells chondrocytes, osteoblasts, Akiyama et al. (2005); Ono
in the growth plate and articular osteocytes et al. (2014); Debnath et al. (2018)
cartilage
Lepr Skeletal stem/progenitor cells in the BMSCs, adipocytes in the bone Zhou et al. (2014a); Baryawno
periosteum* and bone marrow, marrow, osteoblasts and osteocytes et al. (2019); Baccin et al. (2020)
CAR cells, adipogenic precursors, in the trabecula during bone
pericytes maintenance; osteoblasts,
osteocytes and chondrocytes in the
callus and osteocytes in newly
formed cortical bone during
fracture healing
Craniofacial bones
Gli1 SuSCs in the suture mesenchyme Periosteal, sutural and endosteal/ Zhao et al. (2015); Shi et al. (2017)
(after P21) dural cells, osteoblasts, osteocytes

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


366 Shuo Huang et al.

Table 1. (Cont.)

Target cells and their locations in Cell lineages involved in bone


Markers References
adult mice maintenance and repair
Axin2 SuSCs in the midline of almost all Periosteal* and sutural cells, Maruyama et al. (2016); Usami
calvarial sutures, osteoblasts, osteocytes, et al. (2019)
chondroprogenitors in the resting chondroprogenitors and
zone of the growth plate and the chondrocytes (in the growth plate)
outermost layer of the growth plate
facing Ranvier’s groove
Prrx1 SuSCs in the suture mesenchyme Sutural cells, osteoblasts, osteocytes Ouyang et al. (2013); Wilk et al. (2017)
Ctsk SuSCs in the suture mesenchyme; Periosteal, sutural and endosteal/ Debnath et al. (2018)
cells in the periosteum, dural cells, BMSCs, osteoblasts,
endosteum/dura and bone osteocytes, osteoclasts
marrow*

Prrx1, Paired related homeobox 1; Ctsk, Cathepsin K; Gli1, Glioma-associated oncogene 1; αSMA, Alpha smooth muscle actin; Cxcl12, C-
X-C motif chemokine 12; Mx1, Myxovirus resistance 1; Grem1, Gremlin 1; Sox9, Sex-determining region Y (SRY)-box 9; Col2, collagen
type II; Lepr, leptin receptor; SSCs, skeletal stem cells; BMSCs, bone marrow mesenchymal stem/stromal cells; vSMCs, vascular smooth
muscle cells; OCR, osteochondroreticular; CAR, Cxcl12-abundant reticular; SuSCs, suture stem cells.
*Expression
#
is not mentioned in the papers referenced but can be inferred from their figures or single-cell RNA sequencing data.
Rare osteocytes express Ctsk.

osteoblasts, osteoprogenitors and osteoclasts in calvarial domains of long bones: the periosteum, articular cartilage,
sutures, periosteum, endosteum and bone marrow but also upper layers of the growth plates, perichondrium flanking the
labels SSCs in the periosteum of long bones and sutures of growth plate, and the chondro-osseous junction immediately
the calvarium. Non-haematopoietic Ctsk+ periosteum/ beneath the growth plate; it is not expressed in mature osteo-
suture cells contain three main subpopulations: periosteal blasts, osteocytes and bone marrow (Shi et al., 2017). After
stem cells (PSCs: CD31− CD45− Ter119− 6C3− CD90− administrations of tamoxifen to Gli1-CreERT2;R26-tdTomato
CD49flow CD51low CD105− CD200+) and periosteal pro- mice at 1 month of age, lineage tracing of Gli1+ cells was carried
genitors 1 and 2 (PP1: CD31− CD45− Ter119− 6C3− out over the following 1, 3, and 9 months. tdTomato (Gli1+)
CD90− CD49flow CD51low CD105− CD200−; PP2: could label certain periosteal cells, osteoblasts and osteocytes
CD31− CD45− Ter119− 6C3− CD90− CD49flow CD51low of cortical bone and the remaining bone trabeculae as well as
CD105+ CD200variable) (Debnath et al., 2018). PSCs are at growth plate chondrocytes. It was noted that tdTomato also
the top of the Ctsk+ cell differentiation hierarchy and can labelled many adipocytes, as shown by the identification of
give rise to all the other Ctsk+ populations of periosteum/ perilipin- and Lepr-positive cells in bone marrow (Shi
suture cells, while PP1 and PP2 do not give rise to PSCs et al., 2017). Moreover, to investigate the contribution of Gli1+
(Debnath et al., 2018). In addition, PSCs are able to self- cells to long bone fracture healing, Gli1-CreERT2;
renew and differentiate into osteoblasts and chondrocytes R26-tdTomato mice were injected with tamoxifen when they
in vivo and in vitro and still retain these abilities even after serial were 4 weeks old, subjected to semistabilized fracture at
rounds of transplantation into the kidney capsule and mam- 10 weeks old, and killed after 10 days. Gli1-lineage cells were
mary fat pad (Debnath et al., 2018). PSCs can differentiate detected throughout the fracture callus, including in both
into bone and cartilage in the fracture callus and contribute bone and cartilage, indicating that Gli1 might be a marker of
to long bone fracture healing (Debnath et al., 2018). Notably, periosteal SSCs in long bones in postnatal mice.
multipotent PSCs also exist in the periosteal tissue of human Zhao et al. (2015) reported that Gli1 is expressed within the
femurs (Debnath et al., 2018). entire craniofacial periosteum, dura and suture mesenchyme
but not in the fontanelles and osteocytes on postnatal day
0 (P0), P7, and P14. After P21, Gli1 expression is gradually
(3) Gli1
restricted to the mesenchyme of most craniofacial sutures
Glioma-associated oncogene 1 (Gli1), a transcription factor and to the inner surface of some bone marrow spaces (Zhao
and an effector of the hedgehog pathway, identifies MSCs et al., 2015). Lineage-tracing results showed that when
in many tissues of adult mice, such as craniofacial bones, long Gli1-CreERT2;R26-tdTomato mice were induced with
bones and incisors (Hojo et al., 2012; Zhao et al., 2015; Shi tamoxifen at 1 month of age, 2 months later, tdTomato
et al., 2017). Utilizing Gli1-CreERT2;R26-tdTomato mice, labelled the entire suture mesenchyme, osteogenic fronts,
Shi et al. (2017) discovered that during embryonic long bone periosteum, dura and parts of the calvarial osteocytes that
development, Gli1 is predominantly expressed in the peri- were located either next to the suture or on the external sur-
chondrium, including the layers covering the joint surface, face of the craniofacial bones (Zhao et al., 2015). Gli1+ suture
Ranvier’s groove, and the bone shaft where osteoblasts first cells normally do not undergo active division; however, they
form. In postnatal mice, Gli1 is mainly expressed in five could be rapidly activated within 24 h after craniofacial bone

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 367

injury (Zhao et al., 2015). One month after induction with involved in the lateral growth of the growth plate (Usami
tamoxifen, tdTomato labelled the periosteum, dura and et al., 2019). In summary, these findings suggested that Axin2
many osteocytes in the repair region, indicating that Gli1+ might be an ideal marker of SuSCs and chondroprogenitors
cells contribute to injury repair in craniofacial bones (Zhao in the growth plate.
et al., 2015). Moreover, ablation of Gli1+ cells in adult
Gli1-CreERT2;DTA mice led to skull growth arrest, osteo-
(5) αSMA
porosis, craniosynostosis and impaired craniofacial bone
defect healing (Zhao et al., 2015). Overall, Gli1+ cells in post- Alpha smooth muscle actin (αSMA, also known as Acta2) was
natal mice contribute to maintenance and repair in both long reported to label 0.02–0.03% of bone marrow cells, approx-
bones and craniofacial bones. imately 1% of scraped periosteal cells (Grcevic et al., 2012),
SuSCs, satellite cells in muscles, and MSCs in adipose tissue
and periodontium, and these cells possess self-renewing and
(4) Axin2
dual/multiple-lineage differentiation potential (Kalajzic
Axin2 inhibits canonical wingless-type MMTV integration et al., 2008; Grcevic et al., 2012; Roguljic et al., 2013; Mat-
site (Wnt)/T cell specific transcription factor (TCF) signalling thews et al., 2014; Matthews et al., 2016). αSMA is not only
by enhancing the formation of the beta-catenin destruction expressed in pericytes localized to venules (Nehls
complex. By utilizing Axin2-GFP mice, Maruyama et al., 1992; Morikawa et al., 2002; Baryawno et al., 2019),
et al. (2016) found that Axin2 expression is restricted to the but also present in vascular smooth muscle cells (vSMCs) of
midline of almost all calvarial sutures, including the patent larger blood vessels (Kalajzic et al., 2008; Baryawno
posterior frontal suture, and is colocalized with slow-cycling et al., 2019). Grcevic et al. (2012) found that αSMA+ bone
cells that retained 5-ethynyl-20 -deoxyuridine (Edu). This marrow cells have the ability to differentiate into mature
highly specific pattern of expression was first detected at P9 osteoblasts, chondrocytes and adipocytes in vitro. However,
and was maintained in adult mice. By contrast, Gli1+ cells most of these cells express the haematopoietic lineage
in craniofacial bones might target a larger area within the markers CD11b, CD45 and Ter119; αSMA+ (CD11b−
sutures and be retained in the periosteum and dura at an CD45− Ter119−) bone marrow cells are mostly negative
early stage (Zhao et al., 2015; Maruyama et al., 2016). For for the mesenchymal lineage markers Sca1, CD51, CD90,
lineage-tracing analysis, Axin2-Cre-Dox;R26RLacZ mice CD106, CD140b and CD146 (Grcevic et al., 2012). By con-
were administered doxycycline for 3 days starting at P28 trast, the majority of αSMA+ periosteal cells are negative
and analysed after 1, 3, and 12 months. The results showed for CD11b, CD45, and Ter119, but αSMA+ (CD11b−
that Axin2+ labelled all patent sutures and calvarial bones CD45− Ter119−) periosteal cells express higher levels of
but was not detectable in the osteoblasts and osteocytes in Sca1, CD51, CD90, and CD140b than bone marrow cells
long bones, suggesting that there was very little contribution (Grcevic et al., 2012). Using αSMA-CreERT2;
of Axin2+ cells to the homeostasis of long bones (Maruyama R26-tdTomato mice for lineage tracing, results showed that
et al., 2016). Further analysis showed that Axin2+ suture cells during tibia fracture healing, αSMA+ cells are able to expand
possess long-term self-renewal, clonal expansion and differ- and differentiate towards chondrogenic and osteogenic line-
entiation abilities in vitro (Maruyama et al., 2016). Moreover, ages (Grcevic et al., 2012; Matthews et al., 2014; Wang
in response to calvarial injury, Axin2+ suture cells could et al., 2017) and contribute to new cortical bone formation
move into the damage site and then differentiate into osteo- 6 weeks following the fracture (Grcevic et al., 2012; Matthews
cytes embedded in the bone plate, directly contributing to et al., 2016), which indicates that αSMA may label periosteal
calvarial injury healing (Maruyama et al., 2016). By contrast, SSCs of long bones in postnatal mice.
there was a very limited contribution of Axin2+ cells to the
long bone fracture repair (Maruyama et al., 2016).
(6) Cxcl12
Recently, using Axin2-CreERT2;R26-ZsGreen mice,
Usami et al. (2019) identified two new Axin2+ cell populations C–X–C motif chemokine 12 (Cxcl12) is also known as stro-
as chondroprogenitors: one population in the resting zone of mal cell-derived factor 1 (SDF1). Cxcl12-abundant reticular
the central part of the growth plate in postnatal mice, and the (CAR) cells are the major producers of Cxcl12 in mouse bone
other localized to the outermost layer of the growth plate fac- marrow (Sugiyama et al., 2006). CAR cells can be found in
ing Ranvier’s groove (the perichondrium adjacent to the the periosteum and endosteum and are widely scattered in
growth plate). Lineage-tracing results showed that the first adult bone marrow surrounding the sinusoids but Cxcl12
population of Axin2+ cells contributed to the formation of are barely detected in osteoblasts and osteocytes (Tokoyoda
the central part of the growth plate columns in the growth et al., 2004; Sugiyama et al., 2006; Matsushita et al., 2020).
stage, and ablation of β-catenin in these Axin2+ cells strongly CAR cells usually colocalize with haematopoietic stem cells
inhibited their expansion and induced ectopic chondrogen- (HSCs) and play important roles in the maintenance of the
esis in the growth plate (Usami et al., 2019). The second pop- HSC pool (Sugiyama et al., 2006). Recently, using
ulation could give rise to the outer growth plate during the Cxcl12-CreERT;R26-tdTomato mice carrying either
neonatal stage, but their growth was paused when the growth Col12.3-GFP or Cxcl12-GFP reporters, Matsushita
plate stopped growing, and these cells were thought to be et al. (2020) identified a population of quiescent perisinusoidal

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


368 Shuo Huang et al.

Cxcl12-CreERT+ bone marrow MSCs (BMSCs). Although (8) Grem1


these cells displayed little colony-forming activity, they pos-
Gremlin 1 (Grem1), a secreted antagonist of BMP2, -4, and
sessed trilineage differentiation potential in vitro and could
-7 and a VEGFR2 agonist (Merino et al., 1999; Khokha
contribute to the formation of trabecular bone and fat in
et al., 2003; Canalis, Parker, & Zanotti, 2012), is critical for
the bone marrow cavity but did not participate in cortical
osteogenesis. Grem1 null mice displayed decreased mass
bone formation during bone maintenance in vivo
and body fat and a shortened femoral length (Canalis
(Matsushita et al., 2020). However, these cells partly contrib-
et al., 2012). They also exhibited developmental skeletal
uted to newly formed bone in the callus and cortical bone in
abnormalities with incomplete formation of the forelimbs,
both the cortical injury model and the tibial complete frac-
hindlimbs and metatarsal bones (Canalis et al., 2012). Worth-
ture model. Matsushita et al. (2020) suggested that these cells
ley et al. (2015) reported that Grem1 could label a population
might transform into osteoblast precursor cells in a manner
of osteochondroreticular (OCR) stem cells in the embryonic
mediated by canonical Wnt signalling in response to bone
and postnatal bone marrow. To trace Grem1 lineages
injury. We found that Lepr+ cells might display similar char-
in vivo, Grem1-CreERT;R26-tdTomato mice were orally
acteristics according to fate-mapping data from Lepr-cre;
administered tamoxifen at 6–8 weeks of age. After approxi-
R26-tdTomato;Col2.3-GFP mice (Zhou et al., 2014a).
mately 12 months of induction, Grem1+ cells differentiated
Therefore, Cxcl12 might be a marker of particular popula-
into columns of chondrocytes in the growth plate, articular
tions of skeletal stem/progenitor cells in some circumstances.
cartilage, periosteal cells, reticular marrow stromal cells,
diaphyseal osteoblasts, and osteocytes but not in adipocytes
in either the vertebral bodies or the femur (Worthley
(7) Mx1 et al., 2015). Grem1+ cells also have the ability to self-renew
Myxovirus resistance 1 (Mx1) was originally reported to be in vivo and in vitro and are tripotent, differentiating into bone,
expressed in bone marrow stromal cells, cranial stem/pro- cartilage, and reticular marrow stromal cells in both embry-
genitor cells, a fraction of osteocalcin (Ocn)-positive mature onic and postnatal mice (Worthley et al., 2015). Gene-
osteoblasts, haematopoietic-derived osteoclasts, and perios- expression microarray analysis of Lin−(CD31− CD45−
teal (inner cambial) and endosteal cells in growing and adult Ter119−)Grem1+ and Lin−Grem1− mesenchymal cells
mice (Walkley et al., 2007; Park et al., 2012; Park et al., 2014). revealed that Lin−Grem1+ cells were significantly more
Chondrocytes, adipocytes, skeletal muscle cells, fibroblasts, active in BMP signalling as well as extracellular matrix
osteocytes and blood vessels were not labelled by Mx1 (ECM)-receptor interactions, phosphatidylinositol 3-kinase
(Park et al., 2012). In vivo pulse-chase results showed that (PI3K)-serine/threonine kinase Akt (Akt) signalling, and
Mx1+ cells could differentiate into osteoblasts, osteocytes, focal adhesion pathways, which might be correlated with
bone marrow cells, and periosteal and endosteal cells but the osteochondral lineage potential of Lin−Grem1+ cells
not chondrocytes (Park et al., 2012). Mx1+ cells could also (Worthley et al., 2015). To investigate the contribution of
respond to tissue stress and migrate to fracture sites, supply- Grem1+ cells to fracture healing, adult Grem1-creERT;
ing new bone tissue during microfracture healing of the cal- R26-TdTomato;2.3ColGFP mice were administered tamox-
varium and metaphysis. Therefore, Mx1 was identified as a ifen with a 1-week washout period. Mice were killed at 7 days
marker of osteogenic progenitor cells. following unilateral femoral fracture. The results showed that
Recent findings show that Mx1 and αSMA in combination Grem1+ cells could generate osteoblasts and chondrocytes in
can selectively label postnatal periosteal stem cells (P-SSCs) the callus during fracture healing through EO (Worthley
and calvarial SuSCs but are nearly undetectable in bone et al., 2015). These data indicate that Grem1 might label
marrow (Ortinau et al., 2019). Mx1+aSMA+ periosteal cells SSCs in long bones.
possess SSC characteristics in vitro and are able to form colo-
nies with chondrogenic, osteogenic and adipogenic differen-
(9) Sox9, Col2 and aggrecan
tiation abilities at the single-cell level (Ortinau et al., 2019).
Mx1+αSMA+ P-SSCs/SuSCs could respond rapidly and Sex-determining region Y (SRY)-box 9 (Sox9), collagen type
migrate towards the injury site. Mx1+αSMA+ P-SSCs repo- II (Col2) and aggrecan (Agc) are markers of chondrocytes.
pulated the outer layer of the callus, generated new perios- However, recent lineage-tracing experiments using a
teum, and formed the majority of callus by supplying tamoxifen-inducible form of Cre (CreERT) system revealed
osteoblasts and chondrocytes following tibial transverse frac- that early postnatal cells at P3 labelled by Sox9-CreERT,
ture (Ortinau et al., 2019). Mx1+αSMA+ SuSCs at the top Col2-CreERT and Agc-CreERT progressively contributed
surface of the injury maintained their undifferentiated status to the formation of chondrocytes, osteoblasts, osteocytes
and formed a new periosteum, while Mx1+αSMA+ cells in and bone marrow stromal cells for at least 6 months in mice
the middle of the callus differentiated into Mx1+aSMA− (Ono et al., 2014). In addition, Col2-CreERT-labelled cells
osteoblasts and osteocytes embedded in newly formed bone could become adipocytes in the metaphyseal bone marrow
plates after 21 days during calvarial defect healing (Ortinau (Ono et al., 2014). Conditional gene manipulation using
et al., 2019). Therefore, Mx1 and αSMA in combination Col2-Cre affected both cartilage and bone (Ford-Hutchinson
might label SSCs in both long bones and craniofacial bones. et al., 2007; Wang et al., 2011a). Another study reported that

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 369

Sox9 was expressed in the primary spongiosa, periosteum periosteum and bone marrow, and were reported to contrib-
and endosteum of adult long bones (He et al., 2017). In a fem- ute to callus formation during bone repair (Diaz-Flores
oral fracture model, Sox9-CreERT;R26-tdTomato mice et al., 1992). In bone marrow, there are two types of pericytes
were given three consecutive doses of tamoxifen 2 weeks expressing two different surface markers, Lepr and nerve/
before fracture, and lineage-tracing results showed that at glial antigen 2 (NG2/Cspg4), which are generally associated
9 days postfracture, Sox9+-lineage cells differentiated into with sinusoids and arterioles, respectively (Kunisaki
large numbers of chondrocytes, osteoblasts and osteocytes et al., 2013; Coutu et al., 2017; Kunisaki, 2019; Liu, Li, &
in the callus (He et al., 2017). However, using the same Tan, 2019a). Both Lepr+ and NG2+ pericytes express stem
Sox9-CreERT mice but with different tamoxifen induction cell factor (SCF/Kitl), Cxcl12 and Nestin (Kunisaki
schedules for lineage tracing, Kuwahara et al. (2019) et al., 2013; Asada et al., 2017; Coutu et al., 2017), all of which
observed different results. When Sox9-CreERT; were reported as markers of bone marrow MSCs (Ding &
R26-tdTomato mice were injected with tamoxifen for three Morrison, 2013; Greenbaum et al., 2013; Kunisaki, 2019).
consecutive days starting 7 days before large-scale rib re- However, contradictory data hinted that pericytes might
section surgery, at 10 days post resection, only approxi- not or may only partly possess the characteristics of MSCs/
mately 22 ± 1.3% of tdTomato-labelled cells contributed SSCs in vivo. Zhou et al. (2014a) fate-mapped Lepr+ cells in vivo
to both cartilage and bone in the callus (Kuwahara using Lepr-Cre;R26-tdTomato;Col2.3-GFP mice and found
et al., 2019). However, when these mice were induced for that Lepr+ cells made little contribution to stromal cells and
4 consecutive days starting the day before the surgery, at bone in early postnatal bone marrow, but they increased in
10 days post resection, a much greater portion of the callus number over time and became the source of osteoblasts
was labeled by tdTomato (85.5 ± 2.8%), including the newly and adipocytes in adult bone marrow. However, as discussed
formed cartilage, bone and periosteum surrounding the cal- in Section VII-(6), both Cxcl12+ and Lepr+ cells only partly
lus (Kuwahara et al., 2019). Deletion of the obligate hedgehog contribute to cortical bone formation during bone repair,
co-receptor, Smoothened, in Sox9+ cells before surgery but do not participate in cortical bone maintenance. Utiliz-
resulted in a near-complete loss of rib bone regeneration abil- ing Nes-CreERT;R26-EGFP mice, Mendez-Ferrer
ity (Kuwahara et al., 2019). The latter authors speculated that et al. (2010) found that Nestin+ cells in bone marrow exhib-
Sox9-expressing periosteal cells (termed Sox9+ messenger ited spontaneous multilineage differentiation ability in cul-
cells) might play a major role in orchestrating large-scale ture and robust self-renewal ability after serial
bone regeneration by stimulating neighbouring cells to dif- transplantation in vivo. Lineage-tracing results showed that
ferentiate through an unknown signal into osteochondropro- Nestin+ cells did not give rise to bone-forming cells after a
genitors. We suggest that Sox9+ periosteal cells might 1-month chase but differentiated into chondrocytes in the
represent primed SSCs/transit-amplifying cells/osteochon- growth plate, osteoblasts, and osteocytes after an 8-month
droprogenitors. In summary, the above results imply that chase. However, Ono et al. (2014) reported that when Nes-
Col2, aggrecan and Sox9 might be considered markers of CreERT;R26-tdTomato mice received tamoxifen at P3,
skeletal stem/progenitor cells in long bones in some Nestin+ cells predominantly contributed to the formation of
circumstances. endothelial cells in bone marrow after a 1-month chase, only
a small number of Nestin+ cells became osteoblasts and oste-
ocytes in the cortical bone and very infrequently became
chondrocytes in the growth plate, but they never differenti-
VIII. THE DEBATE BETWEEN PERICYTES ated into adipocytes in bone marrow. Zhou et al. (2014a)
AND MSCS revealed that Nestin+ cells from different Nestin transgenic
mice displayed different characteristics in bone marrow.
Pericytes are perivascular multipotent cells that show hetero- Nestin+ cells from Nestin-GFP mice possessed colony-
geneity in different organs or even within the same tissue forming capacity in vitro, but Nestin+ cells from Nestin-
(Nirwane, Gautam, & Yao, 2017). Pericytes do not have a CreERT;R26-EYFP mice did not, and a lineage-tracing
precisely defined phenotype; instead, they are recognized experiment also confirmed that Nestin-CreERT lineages
by their phenotype, distribution and origin (Armulik, Gen- rarely contributed to the formation of osteoblasts and bone
ove, & Betsholtz, 2011). Pericytes reside on the abluminal in vivo after 9 months of tamoxifen induction (Zhou
surface of smaller calibre vessels of every vascularized con- et al., 2014a). Guimaraes-Camboa et al. (2017) found that
nective tissue, whereas vSMCs are found around larger ves- T-box transcription factor 18 (Tbx18) could selectively label
sels (Friedenstein et al., 1974). It is generally accepted that pericytes and vSMCs in multiple organs of adult mice.
pericytes and vSMCs belong to the same lineage and cate- Fluorescence-activated cell sorting (FACS)-purified
gory, and there are no specific markers to distinguish peri- Tbx18-expressing cells behaved as MSCs in vitro. However,
cytes from vSMCs (Armulik et al., 2011). Pericytes are lineage-tracing experiments using an inducible
believed to function as MSCs with the capacity to differenti- Tbx18-CreERT2 cell line indicated that pericytes and
ate into osteoblasts, chondrocytes, fibroblasts, adipocytes vSMCs remained identical in various pathological settings
(Nehls & Drenckhahn, 1993) and myogenic cells in vitro and during ageing but did not contribute significantly to
(Crisan et al., 2008). In addition, pericytes exist in the other cell lineages (Guimaraes-Camboa et al., 2017).

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


370 Shuo Huang et al.

Recently, two studies used scRNA-seq technology to ana- bone marrow cells, and this cluster was annotated as MSCs
lyse and compare MSCs and pericytes in periosteal, endos- (Wolock et al., 2019; Matsushita et al., 2020; Zhong
teal and bone marrow stromal cells of mouse limb bones, et al., 2020). It is still hard to distinguish which cell popula-
but resulted in almost diametrically opposed conclusions. tions are true MSCs/pericytes based on these opposing
Based on the ‘consensus’ that Lepr is a marker of adult bone views, however, these results indicate that MSCs and peri-
marrow MSCs (Zhou et al., 2014a), Baryawno et al. (2019) cytes might represent different cell types. Moreover, both
annotated one cell population as Lepr+ mesenchymal stromal populations are still heterogeneous according to their gene
cells (referred to as Lepr-MSCs) with adipogenic and osteo- expression profiles, and the homogeneous subsets need to
genic features and another distinct population of perivascular be further extracted and identified.
mesenchymal stromal cells and pericytes (referred to as peri-
cytes) according to their gene expression profiles. Lepr-MSCs
highly expressed Lepr and adiponectin (Adipoq), which is
highly expressed in preadipocytes (Lara-Castro et al., 2007), IX. FUTURE PROSPECTS
and the key HSC niche factors SCF, Cxcl12 and
angiopoietin-1 (Angpt1) (Baryawno et al., 2019). They also Current knowledge reveals the unique spatiotemporal distri-
expressed Grem1 and Runx2, which is the master regulator bution of SSCs/MSCs in the periosteum, bone marrow, and
transcription factor controlling the commitment of MSCs to suture mesenchyme as well as the growth plates and Ran-
osteolineages, as well as the classical MSC markers CD73, vier’s groove in the embryonic and postnatal period and
CD105 and CD106 (Vcam1), but they did not express the lineage-specific contributions of these cells during bone
CD90, Sca1, NG2, Nestin and αSMA (Baryawno development, maintenance and fracture repair. However,
et al., 2019). The annotated pericyte population expressed much remains to be confirmed due to inherent biases associ-
the classical pericyte markers NG2, Nestin, αSMA, myosin ated with experimental assays and conceptual controversy.
heavy chain 11 (Myh11), and melanoma cell adhesion mole- New technologies will help us to refine our understanding
cule (Mcam) (Armulik et al., 2011) but only expressed very and overcome the limitations of the technologies currently
low levels of Lepr, in contrast to Lepr-MSCs (Baryawno in use. The mineral composition, macro- and microarchitec-
et al., 2019). ture, remodelling capacity and biomechanics of bone in large
In the second paper, Baccin et al. (2020) annotated Ng2- animals such as sheep, pigs and monkeys are similar to those
and Nestin-expressing mesenchymal cells in Lin− CD45− of humans, and the availability of clustered regularly inter-
CD71− periosteal, endosteal and bone marrow stromal cells spaced short palindromic repeat (CRISPR)-mediated
of mouse limb bones as Ng2+ MSCs, which showed tran- genome editing in these large animals will provide us with
scriptomic similarity to previously described Ng2+Nestin+ better opportunities to study the cellular and molecular
MSCs [and were annotated as the ‘pericyte’ population by mechanisms of bone development, diseases, and injury/
Baryawno et al., 2019]. Moreover, they annotated another regeneration that are more similar to those in humans
two populations with similar overall transcriptomic profiles (Truong et al., 2019; Sparks et al., 2020). The dual-recombi-
as Adipo-CAR and Osteo-CAR cells that expressed adipo- nase-activated lineage-tracing platform (DeaLT) that com-
cyte and osteolineage genes, respectively (Baccin bines the D6 site-specific DNA recombinase-rox
et al., 2020). Most Adipo-CAR cells localized abundantly to recombination site (Dre-rox) and Cre recombinase-locus of
sinusoidal endothelia, and Osteo-CAR cells localized to crossover in P1 (Cre-loxP) recombination system could partly
non-vascular regions or covered arteriolar endothelia overcome the limitations of Cre-mediated lineage tracing,
(Baccin et al., 2020). Both CAR cell populations produced such as the specificity of Cre and off-target labelling
high levels of cytokines and growth factors involved in HSC (He et al., 2018; Li et al., 2018). The cellular barcoding tech-
maintenance and differentiation (Baccin et al., 2020). Inter- nique, in which individual cells are labelled with unique
estingly, the Adipo-CAR cells expressed Lepr and Adipoq nucleic acid sequences (termed barcodes), could be used for
at high levels and showed strong transcriptomic similarity fate mapping, lineage tracing and high-throughput screening
to Lepr-Cre cells (Baccin et al., 2020) [annotated as ‘Lepr- of millions of cells in parallel and thus is an efficient approach
MSCs’ in Baryawno et al., 2019]. Both Adipo-CAR cells for investigating heterogeneous populations of SSCs
and Ng2+ MSCs express NG2, Nestin, Cxcl12 and SCF; (Kebschull & Zador, 2018). The stochastic multicolour label-
however, Adipo-CAR cells express higher levels of Cxcl12 ling technique (Brainbow) that labels individual cells with
and SCF and lower levels of Nestin than Ng2+ MSCs but multiple fluorescent proteins (Livet et al., 2007; Cai
do not express Grem1 (Baccin et al., 2020). Furthermore, et al., 2013; Sakaguchi, Leiwe, & Imai, 2018) could also be
pseudo-time analysis results showed that Ng2+ MSCs are at used for investigating the heterogeneity of SSCs. If such a
the apex of a differentiation hierarchy with CAR cells, fibro- technique is combined with tissue-clearing techniques such
blasts, chondrocytes and osteoblasts downstream (Baccin as CUBIC (clear, unobstructed brain/body imaging cock-
et al., 2020). tails and computational analysis) (Susaki et al., 2015) and
It is interesting that three other studies used the scRNA- SHANEL (small micelle-mediated human organ efficient
seq technique to identify a similar cell cluster expressing high clearing and labeling) (Zhao et al., 2020), researchers could
levels of Lepr, Cxcl12 and many adipocyte markers in mouse comprehensively map cells in the whole body of mice or large

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 371

intact human organs at the cellular level. Intravital bone- Crombrugghe, B. (2005). Osteo-chondroprogenitor cells are derived from Sox9
expressing precursors. Proceedings of the National Academy of Sciences of the United States of
imaging technology using multiphoton microscopy is a novel America 102(41), 14665–14670.
and powerful tool that will enable us to visualize the spatio- Alexander, K. A., Raggatt, L. J., Millard, S., Batoon, L., Chiu-Ku Wu, A.,
temporal cellular behaviours of SSCs and their offspring in Chang, M. K., Hume, D. A. & Pettit, A. R. (2017). Resting and injury-induced
inflamed periosteum contain multiple macrophage subsets that are located at sites
their natural tissue microenvironment (Liu, Yuan, & of bone growth and regeneration. Immunology and Cell Biology 95(1), 7–16.
Zhang, 2019b). scRNA-seq will further deepen our aware- Allen, M. R., Hock, J. M. & Burr, D. B. (2004). Periosteum: biology, regulation, and
response to osteoporosis therapies. Bone 35(5), 1003–1012.
ness of distinct SSC populations and their niches, interrela- Armulik, A., Genove, G. & Betsholtz, C. (2011). Pericytes: developmental,
tionships and cellular and molecular mechanisms physiological, and pathological perspectives, problems, and promises. Developmental
(Baryawno et al., 2019; Tikhonova et al., 2019; Wolock Cell 21(2), 193–215.
Asada, N., Kunisaki, Y., Pierce, H., Wang, Z., Fernandez, N. F., Birbrair, A.,
et al., 2019; Baccin et al., 2020; Matsushita et al., 2020). The Ma’ayan, A. & Frenette, P. S. (2017). Differential cytokine contributions of
utilization of these new technologies in future studies will perivascular haematopoietic stem cell niches. Nature Cell Biology 19(3), 214–223.
help us increase our understanding of the mechanisms under- Baccin, C., Al-Sabah, J., Velten, L., Helbling, P. M., Grunschlager, F.,
Hernandez-Malmierca, P., Nombela-Arrieta, C., Steinmetz, L. M.,
lying the development, maintenance, degeneration, injury Trumpp, A. & Haas, S. (2020). Combined single-cell and spatial transcriptomics
and regeneration of the skeleton, leading to the development reveal the molecular, cellular and spatial bone marrow niche organization. Nature
of novel therapies to treat bone diseases and promote bone Cell Biology 22(1), 38–48.
Badve, C. A., K, M. M., Iyer, R. S., Ishak, G. E. & Khanna, P. C. (2013).
repair. Craniosynostosis: imaging review and primer on computed tomography. Pediatric
Radiology 43(6), 728–742.
Bahney, C. S., Hu, D. P., Taylor, A. J., Ferro, F., Britz, H. M.,
Hallgrimsson, B., Johnstone, B., Miclau, T. & Marcucio, R. S. (2014).
Stem cell-derived endochondral cartilage stimulates bone healing by tissue
X. CONCLUSIONS transformation. Journal of Bone and Mineral Research 29(5), 1269–1282.
Bahney, C. S., Hu, D. P., Miclau, T. 3rd & Marcucio, R. S. (2015). The
(1) We summarize in detail the temporal and spatial pro- multifaceted role of the vasculature in endochondral fracture repair. Frontiers in
Endocrinology 6, 4.
cesses of injury (fracture) repair in both craniofacial Bais, M., McLean, J., Sebastiani, P., Young, M., Wigner, N., Smith, T.,
and long bones. Kotton, D. N., Einhorn, T. A. & Gerstenfeld, L. C. (2009). Transcriptional
analysis of fracture healing and the induction of embryonic stem cell-related genes.
(2) We discuss the definition of MSCs and SSCs, as well as PLoS One 4(5), e5393.
their inherent differentiation potential. Baryawno, N., Przybylski, D., Kowalczyk, M. S., Kfoury, Y., Severe, N.,
(3) We describe newly discovered populations of skeletal Gustafsson, K., Kokkaliaris, K. D., Mercier, F., Tabaka, M., Hofree, M.,
Dionne, D., Papazian, A., Lee, D., Ashenberg, O., Subramanian, A. &
stem/progenitor cells involved in craniofacial and/or ET, A. L. (2019). A cellular taxonomy of the bone marrow stroma in homeostasis
long bone repair with a specific focus on their markers, and leukemia. Cell 177(7), 1915–1932.
spatiotemporal distribution, stemness, and contribu- Behonick, D. J., Xing, Z., Lieu, S., Buckley, J. M., Lotz, J. C., Marcucio, R. S.,
Werb, Z., Miclau, T. & Colnot, C. (2007). Role of matrix metalloproteinase
tions to bone maintenance and repair. 13 in both endochondral and intramembranous ossification during skeletal
(4) We compare the contributions of reparative cells from regeneration. PLoS One 2(11), e1150.
multiple sources to bone maintenance and repair, and Berendsen, A. D. & Olsen, B. R. (2015). Bone development. Bone 80, 14–18.
Bi, Y., Ehirchiou, D., Kilts, T. M., Inkson, C. A., Embree, M. C.,
also summarize the debate between the roles of Sonoyama, W., Li, L., Leet, A. I., Seo, B. M., Zhang, L., Shi, S. &
MSCs/SSCs and pericytes. Young, M. F. (2007). Identification of tendon stem/progenitor cells and the role
(5) We discuss the existing problems, emerging novel tech- of the extracellular matrix in their niche. Nature Medicine 13(10), 1219–1227.
Bonnet, N., Brun, J., Rousseau, J. C., Duong, L. T. & Ferrari, S. L. (2017).
nologies and future research directions in this field. Cathepsin K controls cortical bone formation by degrading Periostin. Journal of
Bone and Mineral Research 32(7), 1432–1441.
Brey, E. M., Cheng, M. H., Allori, A., Satterfield, W., Chang, D. W.,
Patrick, C. W. Jr. & Miller, M. J. (2007). Comparison of guided bone
formation from periosteum and muscle fascia. Plastic and Reconstructive Surgery 119(4),
ACKNOWLEDGEMENTS 1216–1222.
Burchardt, H. & Enneking, W. F. (1978). Transplantation of bone. Surgical Clinics of
North America 58(2), 403–427.
This work was supported by research funding from the Cai, D., Cohen, K. B., Luo, T., Lichtman, J. W. & Sanes, J. R. (2013). Improved
National Key Research and Development Program of tools for the Brainbow toolbox. Nature Methods 10(6), 540–547.
Canalis, E., Parker, K. & Zanotti, S. (2012). Gremlin1 is required for skeletal
China (2018YFA0800802), the National Natural Science development and postnatal skeletal homeostasis. Journal of Cellular Physiology 227(1),
Foundation of China (No. 81530071, No. 81772359, 269–277.
No. 81601906) and the Chongqing Postdoctoral Researchers Caplan, A. I. & Bruder, S. P. (2001). Mesenchymal stem cells: building blocks
for molecular medicine in the 21st century. Trends in Molecular Medicine 7(6),
Research Project (Xm2016084). 259–264.
Chan, C. K., Seo, E. Y., Chen, J. Y., Lo, D., McArdle, A., Sinha, R., Tevlin, R.,
Seita, J., Vincent-Tompkins, J., Wearda, T., Lu, W. J., Senarath-Yapa, K.,
Chung, M. T., Marecic, O., Tran, M. & ET, A. L. (2015). Identification and
XI. REFERENCES specification of the mouse skeletal stem cell. Cell 160(1–2), 285–298.
Chan, C. K. F., Gulati, G. S., Sinha, R., Tompkins, J. V., Lopez, M.,
Abou-Khalil, R. & Colnot, C. (2014). Cellular and molecular bases of skeletal Carter, A. C., Ransom, R. C., Reinisch, A., Wearda, T., Murphy, M.,
regeneration: what can we learn from genetic mouse models? Bone 64, 211–221. Brewer, R. E., Koepke, L. S., Marecic, O., Manjunath, A., Seo, E. Y. &
Ahmed, Y. A., Tatarczuch, L., Pagel, C. N., Davies, H. M., Mirams, M. & ET, A. L. (2018). Identification of the human skeletal stem cell. Cell 175(1),
Mackie, E. J. (2007). Physiological death of hypertrophic chondrocytes. 43–56.
Osteoarthritis and Cartilage 15(5), 575–586. Cho, T. J., Gerstenfeld, L. C. & Einhorn, T. A. (2002). Differential temporal
Akiyama, H., Kim, J. E., Nakashima, K., Balmes, G., Iwai, N., Deng, J. M., expression of members of the transforming growth factor beta superfamily during
Zhang, Z., Martin, J. F., Behringer, R. R., Nakamura, T. & de murine fracture healing. Journal of Bone and Mineral Research 17(3), 513–520.

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


372 Shuo Huang et al.

Cho, T. J., Kim, J. A., Chung, C. Y., Yoo, W. J., Gerstenfeld, L. C., Franz-Odendaal, T. A., Hall, B. K. & Witten, P. E. (2006). Buried alive: how
Einhorn, T. A. & Choi, I. H. (2007). Expression and role of interleukin-6 in osteoblasts become osteocytes. Developmental Dynamics 235(1), 176–190.
distraction osteogenesis. Calcified Tissue International 80(3), 192–200. Friedenstein, A. J., Chailakhyan, R. K., Latsinik, N. V., Panasyuk, A. F. &
Colnot, C. (2009). Skeletal cell fate decisions within periosteum and bone marrow Keiliss-Borok, I. V. (1974). Stromal cells responsible for transferring the
during bone regeneration (vol 24, pg 274, 2009). Journal of Bone and Mineral Research microenvironment of the hemopoietic tissues. Cloning in vitro and
24(4), 758–758. retransplantation in vivo. Transplantation 17(4), 331–340.
Colnot, C. (2011). Cell sources for bone tissue engineering: insights from basic Gerber, H. P., Vu, T. H., Ryan, A. M., Kowalski, J., Werb, Z. & Ferrara, N.
science. Tissue Engineering Part B-Reviews 17(6), 449–457. (1999). VEGF couples hypertrophic cartilage remodeling, ossification and
Colnot, C. I. & Helms, J. A. (2001). A molecular analysis of matrix remodeling and angiogenesis during endochondral bone formation. Nature Medicine 5(6), 623–628.
angiogenesis during long bone development. Mechanisms of Development 100(2), Gerstenfeld, L. C., Cho, T. J., Kon, T., Aizawa, T., Cruceta, J.,
245–250. Graves, B. D. & Einhorn, T. A. (2001). Impaired intramembranous bone
Colnot, C., Zhang, X. & Knothe Tate, M. L. (2012). Current insights on the formation during bone repair in the absence of tumor necrosis factor-alpha
regenerative potential of the periosteum: molecular, cellular, and endogenous signaling. Cells, Tissues, Organs 169(3), 285–294.
engineering approaches. Journal of Orthopaedic Research 30(12), 1869–1878. Gerstenfeld, L. C., Cullinane, D. M., Barnes, G. L., Graves, D. T. &
Coutu, D. L., Kokkaliaris, K. D., Kunz, L. & Schroeder, T. (2017). Three- Einhorn, T. A. (2003). Fracture healing as a post-natal developmental process:
dimensional map of nonhematopoietic bone and bone-marrow cells and molecular, spatial, and temporal aspects of its regulation. Journal of Cellular
molecules. Nature Biotechnology 35(12), 1202–1210. Biochemistry 88(5), 873–884.
Crisan, M., Yap, S., Casteilla, L., Chen, C. W., Corselli, M., Park, T. S., Grcevic, D., Pejda, S., Matthews, B. G., Repic, D., Wang, L., Li, H.,
Andriolo, G., Sun, B., Zheng, B., Zhang, L., Norotte, C., Teng, P. N., Kronenberg, M. S., Jiang, X., Maye, P., Adams, D. J., Rowe, D. W.,
Traas, J., Schugar, R., Deasy, B. M. & ET, A. L. (2008). A perivascular origin Aguila, H. L. & Kalajzic, I. (2012). In vivo fate mapping identifies
for mesenchymal stem cells in multiple human organs. Cell Stem Cell 3(3), 301–313. mesenchymal progenitor cells. Stem Cells 30(2), 187–196.
De Bari, C., Dell’Accio, F., Vanlauwe, J., Eyckmans, J., Khan, I. M., Greenbaum, A., Hsu, Y. M., Day, R. B., Schuettpelz, L. G.,
Archer, C. W., Jones, E. A., McGonagle, D., Mitsiadis, T. A., Christopher, M. J., Borgerding, J. N., Nagasawa, T. & Link, D. C. (2013).
Pitzalis, C. & Luyten, F. P. (2006). Mesenchymal multipotency of adult human CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-
periosteal cells demonstrated by single-cell lineage analysis. Arthritis and Rheumatism cell maintenance. Nature 495(7440), 227–230.
54(4), 1209–1221. Gruber, R., Koch, H., Doll, B. A., Tegtmeier, F., Einhorn, T. A. &
Debnath, S., Yallowitz, A. R., McCormick, J., Lalani, S., Zhang, T., Xu, R., Hollinger, J. O. (2006). Fracture healing in the elderly patient. Experimental
Li, N., Liu, Y., Yang, Y. S., Eiseman, M., Shim, J. H., Hameed, M., Gerontology 41(11), 1080–1093.
Healey, J. H., Bostrom, M. P., Landau, D. A. & ET, A. L. (2018). Discovery Guichet, J. M., Braillon, P., Bodenreider, O. & Lascombes, P. (1998).
of a periosteal stem cell mediating intramembranous bone formation. Nature 562 Periosteum and bone marrow in bone lengthening: a DEXA quantitative
(7725), 133–139. evaluation in rabbits. Acta Orthopaedica Scandinavica 69(5), 527–531.
Delgado-Calle, J. & Bellido, T. (2017). New insights into the local and systemic Guimaraes-Camboa, N., Cattaneo, P., Sun, Y., Moore-Morris, T., Gu, Y.,
functions of Sclerostin: regulation of quiescent bone lining cells and beige Dalton, N. D., Rockenstein, E., Masliah, E., Peterson, K. L.,
Adipogenesis in peripheral fat depots. Journal of Bone and Mineral Research 32(5), Stallcup, W. B., Chen, J. & Evans, S. M. (2017). Pericytes of multiple organs
889–891. do not behave as mesenchymal stem cells in vivo. Cell Stem Cell 20(3), 345–359.
DePalma, A. F., Rothman, R. H., Lewinnek, G. E. & Canale, S. T. (1972). Gulati, G. S., Murphy, M. P., Marecic, O., Lopez, M., Brewer, R. E.,
Anterior interbody fusion for severe cervical disc degeneration. Surgery, Gynecology Koepke, L. S., Manjunath, A., Ransom, R. C., Salhotra, A., Weissman, I. L.,
and Obstetrics 134(5), 755–758. Longaker, M. T. & Chan, C. K. F. (2018). Isolation and functional assessment of
Diaz-Flores, L., Gutierrez, R., Lopez-Alonso, A., Gonzalez, R. & Varela, H. mouse skeletal stem cell lineage. Nature Protocols 13(6), 1294–1309.
(1992). Pericytes as a supplementary source of osteoblasts in periosteal osteogenesis. Hausman, M. R., Schaffler, M. B. & Majeska, R. J. (2001). Prevention of fracture
Clinical Orthopaedics and Related Research 275, 280–286. healing in rats by an inhibitor of angiogenesis. Bone 29(6), 560–564.
Dimitriou, R., Jones, E., McGonagle, D. & Giannoudis, P. V. (2011). Bone He, X., Bougioukli, S., Ortega, B., Arevalo, E., Lieberman, J. R. &
regeneration: current concepts and future directions. BMC Medicine 9, 66. McMahon, A. P. (2017). Sox9 positive periosteal cells in fracture repair of the
Ding, L. & Morrison, S. J. (2013). Haematopoietic stem cells and early lymphoid adult mammalian long bone. Bone 103, 12–19.
progenitors occupy distinct bone marrow niches. Nature 495(7440), 231–235. He, L., Li, Y., Huang, X., Li, Y., Pu, W., Tian, X., Cai, D., Huang, H.,
Dirckx, N., Van Hul, M. & Maes, C. (2013). Osteoblast recruitment to sites of bone Lui, K. O. & Zhou, B. (2018). Genetic lineage tracing of resident stem cells by
formation in skeletal development, homeostasis, and regeneration. Birth Defects DeaLT. Nature Protocols 13(10), 2217–2246.
Research Part C-Embryo Today: Reviews 99(3), 170–191. Hock, J. M., Krishnan, V., Onyia, J. E., Bidwell, J. P., Milas, J. &
Dobnig, H. & Turner, R. T. (1995). Evidence that intermittent treatment with Stanislaus, D. (2001). Osteoblast apoptosis and bone turnover. Journal of Bone
parathyroid hormone increases bone formation in adult rats by activation of bone and Mineral Research 16(6), 975–984.
lining cells. Endocrinology 136(8), 3632–3638. Hojo, H., Ohba, S., Yano, F., Saito, T., Ikeda, T., Nakajima, K., Komiyama, Y.,
Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F., Nakagata, N., Suzuki, K., Takato, T., Kawaguchi, H. & Chung, U. I. (2012).
Krause, D., Deans, R., Keating, A., Prockop, D. & Horwitz, E. (2006). Gli1 protein participates in hedgehog-mediated specification of osteoblast lineage
Minimal criteria for defining multipotent mesenchymal stromal cells. The during endochondral ossification. Journal of Biological Chemistry 287(21),
International Society for Cellular Therapy position statement. Cytotherapy 8(4), 315–317. 17860–17869.
Duchamp de Lageneste, O., Julien, A., Abou-Khalil, R., Frangi, G., Holstein, J. H., Karabin-Kehl, B., Scheuer, C., Garcia, P., Histing, T.,
Carvalho, C., Cagnard, N., Cordier, C., Conway, S. J. & Colnot, C. Meier, C., Benninger, E., Menger, M. D. & Pohlemann, T. (2013).
(2018). Periosteum contains skeletal stem cells with high bone regenerative Endostatin inhibits callus remodeling during fracture healing in mice. Journal of
potential controlled by Periostin. Nature Communications 9(1), 773. Orthopaedic Research 31(10), 1579–1584.
Dy, P., Wang, W., Bhattaram, P., Wang, Q., Wang, L., Ballock, R. T. & Huang, S., Xu, L., Sun, Y., Zhang, Y. & Li, G. (2015a). The fate of systemically
Lefebvre, V. (2012). Sox9 directs hypertrophic maturation and blocks osteoblast administrated allogeneic mesenchymal stem cells in mouse femoral fracture
differentiation of growth plate chondrocytes. Developmental Cell 22(3), 597–609. healing. Stem Cell Research & Therapy 6, 206.
Einhorn, T. A. (1995). Enhancement of fracture-healing. The Journal of Bone & Joint Huang, S., Xu, L., Zhang, Y., Sun, Y. & Li, G. (2015b). Systemic and local
Surgery-American 77(6), 940–956. Administration of Allogeneic Bone Marrow-Derived Mesenchymal Stem Cells
Einhorn, T. A. & Gerstenfeld, L. C. (2015). Fracture healing: mechanisms and Promotes Fracture Healing in rats. Cell Transplantation 24(12), 2643–2655.
interventions. Nature Reviews Rheumatology 11(1), 45–54. Iwasaki, M., Le, A. X. & Helms, J. A. (1997). Expression of indian hedgehog, bone
Ferguson, C. M., Miclau, T., Hu, D., Alpern, E. & Helms, J. A. (1998). Common morphogenetic protein 6 and gli during skeletal morphogenesis. Mechanisms of
molecular pathways in skeletal morphogenesis and repair. Annals of the New York Development 69(1–2), 197–202.
Academy of Sciences 857, 33–42. Kalajzic, Z., Li, H., Wang, L. P., Jiang, X., Lamothe, K., Adams, D. J.,
Ferguson, C., Alpern, E., Miclau, T. & Helms, J. A. (1999). Does adult fracture Aguila, H. L., Rowe, D. W. & Kalajzic, I. (2008). Use of an alpha-smooth
repair recapitulate embryonic skeletal formation? Mechanisms of Development 87(1–2), muscle Actin GFP reporter to identify an osteoprogenitor population. Bone 43(3),
57–66. 501–510.
Ford-Hutchinson, A. F., Ali, Z., Lines, S. E., Hallgrimsson, B., Boyd, S. K. & Kalfas, I. H. (2001). Principles of bone healing. Neurosurgical Focus 10(4), E1.
Jirik, F. R. (2007). Inactivation of Pten in osteo-chondroprogenitor cells leads to Kanou, M., Ueno, T., Kagawa, T., Fujii, T., Sakata, Y., Ishida, N.,
epiphyseal growth plate abnormalities and skeletal overgrowth. Journal of Bone and Fukunaga, J. & Sugahara, T. (2005). Osteogenic potential of primed
Mineral Research 22(8), 1245–1259. periosteum graft in the rat calvarial model. Annals of Plastic Surgery 54(1), 71–78.

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 373

Kawanami, A., Matsushita, T., Chan, Y. Y. & Murakami, S. (2009). Mice Lu, C., Marcucio, R. & Miclau, T. (2006). Assessing angiogenesis during fracture
expressing GFP and CreER in osteochondro progenitor cells in the periosteum. healing. The Iowa Orthopaedic Journal 26, 17–26.
Biochemical and Biophysical Research Communications 386(3), 477–482. Maes, C., Coenegrachts, L., Stockmans, I., Daci, E., Luttun, A., Petryk, A.,
Kebschull, J. M. & Zador, A. M. (2018). Cellular barcoding: lineage tracing, Gopalakrishnan, R., Moermans, K., Smets, N., Verfaillie, C. M.,
screening and beyond. Nature Methods 15(11), 871–879. Carmeliet, P., Bouillon, R. & Carmeliet, G. (2006). Placental growth factor
Khokha, M. K., Hsu, D., Brunet, L. J., Dionne, M. S. & Harland, R. M. (2003). mediates mesenchymal cell development, cartilage turnover, and bone remodeling
Gremlin is the BMP antagonist required for maintenance of Shh and Fgf signals during fracture repair. Journal of Clinical Investigation 116(5), 1230–1242.
during limb patterning. Nature Genetics 34(3), 303–307. Maes, C., Kobayashi, T., Selig, M. K., Torrekens, S., Roth, S. I., Mackem, S.,
Kim, S. W., Pajevic, P. D., Selig, M., Barry, K. J., Yang, J. Y., Shin, C. S., Carmeliet, G. & Kronenberg, H. M. (2010). Osteoblast precursors, but not
Baek, W. Y., Kim, J. E. & Kronenberg, H. M. (2012). Intermittent parathyroid mature osteoblasts, move into developing and fractured bones along with invading
hormone administration converts quiescent lining cells to active osteoblasts. Journal blood vessels. Developmental Cell 19(2), 329–344.
of Bone and Mineral Research 27(10), 2075–2084. Mao, J. J. & Prockop, D. J. (2012). Stem cells in the face: tooth regeneration and
Kline, A. J., Gruen, G. S., Pape, H. C., Tarkin, I. S., Irrgang, J. J. & beyond. Cell Stem Cell 11(3), 291–301.
Wukich, D. K. (2009). Early complications following the operative treatment of Martin, J. F. & Olson, E. N. (2000). Identification of a prx1 limb enhancer. Genesis 26
pilon fractures with and without diabetes. Foot & Ankle International 30(11), (4), 225–229.
1042–1047. Maruyama, T., Jeong, J., Sheu, T. J. & Hsu, W. (2016). Stem cells of the suture
Kon, T., Cho, T. J., Aizawa, T., Yamazaki, M., Nooh, N., Graves, D., mesenchyme in craniofacial bone development, repair and regeneration. Nature
Gerstenfeld, L. C. & Einhorn, T. A. (2001). Expression of osteoprotegerin, Communications 7, 10526.
receptor activator of NF-kappaB ligand (osteoprotegerin ligand) and related Matic, I., Matthews, B. G., Wang, X., Dyment, N. A., Worthley, D. L.,
proinflammatory cytokines during fracture healing. Journal of Bone and Mineral Rowe, D. W., Grcevic, D. & Kalajzic, I. (2016). Quiescent bone lining cells
Research 16(6), 1004–1014. are a major source of osteoblasts during adulthood. Stem Cells 34(12), 2930–2942.
Kroemer, G., Galluzzi, L., Vandenabeele, P., Abrams, J., Alnemri, E. S., Matsushita, Y., Nagata, M., Kozloff, K. M., Welch, J. D., Mizuhashi, K.,
Baehrecke, E. H., Blagosklonny, M. V., El-Deiry, W. S., Golstein, P., Tokavanich, N., Hallett, S. A., Link, D. C., Nagasawa, T., Ono, W. &
Green, D. R., Hengartner, M., Knight, R. A., Kumar, S., Lipton, S. A., Ono, N. (2020). A Wnt-mediated transformation of the bone marrow stromal cell
Malorni, W. & ET, A. L. (2009). Classification of cell death: recommendations identity orchestrates skeletal regeneration. Nature Communications 11(1), 332.
of the nomenclature committee on cell death 2009. Cell Death & Differentiation 16 Matthews, B. G., Grcevic, D., Wang, L., Hagiwara, Y., Roguljic, H.,
(1), 3–11. Joshi, P., Shin, D. G., Adams, D. J. & Kalajzic, I. (2014). Analysis of
Kunisaki, Y. (2019). Pericytes in bone marrow. Advances in Experimental Medicine and alphaSMA-labeled progenitor cell commitment identifies notch signaling as an
Biology 1122, 101–114. important pathway in fracture healing. Journal of Bone and Mineral Research 29(5),
Kunisaki, Y., Bruns, I., Scheiermann, C., Ahmed, J., Pinho, S., Zhang, D., 1283–1294.
Mizoguchi, T., Wei, Q., Lucas, D., Ito, K., Mar, J. C., Bergman, A. & Matthews, B. G., Torreggiani, E., Roeder, E., Matic, I., Grcevic, D. &
Frenette, P. S. (2013). Arteriolar niches maintain haematopoietic stem cell Kalajzic, I. (2016). Osteogenic potential of alpha smooth muscle Actin expressing
quiescence. Nature 502(7473), 637–643. muscle resident progenitor cells. Bone 84, 69–77.
Kurdy, N. M., Weiss, J. B. & Bate, A. (1996). Endothelial stimulating angiogenic Medici, D., Shore, E. M., Lounev, V. Y., Kaplan, F. S., Kalluri, R. &
factor in early fracture healing. Injury 27(2), 143–145. Olsen, B. R. (2010). Conversion of vascular endothelial cells into multipotent
Kuwahara, S. T., Serowoky, M. A., Vakhshori, V., Tripuraneni, N., stem-like cells. Nature Medicine 16(12), 1400–1406.
Hegde, N. V., Lieberman, J. R., Crump, J. G. & Mariani, F. V. (2019). Sox9+ Melnyk, M., Henke, T., Claes, L. & Augat, P. (2008). Revascularisation during
messenger cells orchestrate large-scale skeletal regeneration in the mammalian rib. fracture healing with soft tissue injury. Archives of Orthopaedic & Trauma Surgery 128
eLife 8, e40715. (10), 1159–1165.
Lara-Castro, C., Fu, Y., Chung, B. H. & Garvey, W. T. (2007). Adiponectin and Mendez-Ferrer, S., Michurina, T. V., Ferraro, F., Mazloom, A. R.,
the metabolic syndrome: mechanisms mediating risk for metabolic and Macarthur, B. D., Lira, S. A., Scadden, D. T., Ma’ayan, A.,
cardiovascular disease. Current Opinion in Lipidology 18(3), 263–270. Enikolopov, G. N. & Frenette, P. S. (2010). Mesenchymal and
Lee, J. Y., Qu-Petersen, Z., Cao, B., Kimura, S., Jankowski, R., Cummins, J., haematopoietic stem cells form a unique bone marrow niche. Nature 466(7308),
Usas, A., Gates, C., Robbins, P., Wernig, A. & Huard, J. (2000). Clonal 829–834.
isolation of muscle-derived cells capable of enhancing muscle regeneration and Merino, R., Rodriguez-Leon, J., Macias, D., Ganan, Y., Economides, A. N. &
bone healing. Journal of Cell Biology 150(5), 1085–1100. Hurle, J. M. (1999). The BMP antagonist gremlin regulates outgrowth,
Leucht, P., Kim, J. B., Amasha, R., James, A. W., Girod, S. & Helms, J. A. (2008). chondrogenesis and programmed cell death in the developing limb. Development
Embryonic origin and Hox status determine progenitor cell fate during adult bone 126(23), 5515–5522.
regeneration. Development 135(17), 2845–2854. Miller, S. C., de Saint-Georges, L., Bowman, B. M. & Jee, W. S. (1989). Bone
Li, Y., He, L., Huang, X., Bhaloo, S. I., Zhao, H., Zhang, S., Pu, W., Tian, X., lining cells: structure and function. Scanning Microscopy 3(3), 953–960.
Li, Y., Liu, Q., Yu, W., Zhang, L., Liu, X., Liu, K., Tang, J. & ET, A. L. (2018). Mizuhashi, K., Ono, W., Matsushita, Y., Sakagami, N., Takahashi, A.,
Genetic lineage tracing of nonmyocyte population by dual recombinases. Circulation Saunders, T. L., Nagasawa, T., Kronenberg, H. M. & Ono, N. (2018).
138(8), 793–805. Resting zone of the growth plate houses a unique class of skeletal stem cells. Nature
Lin, Z., Fateh, A., Salem, D. M. & Intini, G. (2014). Periosteum: biology and 563(7730), 254–258.
applications in craniofacial bone regeneration. Journal of Dental Research 93(2), Morikawa, S., Baluk, P., Kaidoh, T., Haskell, A., Jain, R. K. &
109–116. McDonald, D. M. (2002). Abnormalities in pericytes on blood vessels and
Liu, R., Schindeler, A. & Little, D. G. (2010). The potential role of muscle in bone endothelial sprouts in tumors. American Journal of Pathology 160(3), 985–1000.
repair. Journal of Musculoskeletal & Neuronal Interactions 10(1), 71–76. Mountziaris, P. M. & Mikos, A. G. (2008). Modulation of the inflammatory
Liu, R. J., Birke, O., Morse, A., Peacock, L., Mikulec, K., Little, D. G. & response for enhanced bone tissue regeneration. Tissue Engineering Part B-Reviews 14
Schindeler, A. (2011). Myogenic progenitors contribute to open but not closed (2), 179–186.
fracture repair. BMC Musculoskeletal Disorders 12(1), 1–9. Nehls, V. & Drenckhahn, D. (1993). The versatility of microvascular pericytes:
Liu, J. Q., Li, Q. W. & Tan, Z. (2019a). New insights on properties and spatial from mesenchyme to smooth muscle? Histochemistry 99(1), 1–12.
distributions of skeletal stem cells. Stem Cells International 2019, 9026729. Nehls, V., Denzer, K. & Drenckhahn, D. (1992). Pericyte involvement in capillary
Liu, Y., Yuan, Q. & Zhang, S. (2019b). Three-dimensional intravital imaging in bone sprouting during angiogenesis in situ. Cell & Tissue Research 270(3), 469–474.
research. Journal of Biophotonics 12(12), e201960075. Nirwane, A., Gautam, J. & Yao, Y. (2017). Isolation of type I and type II Pericytes
Livet, J., Weissman, T. A., Kang, H., Draft, R. W., Lu, J., Bennis, R. A., from mouse skeletal muscles. Journal of Visualized Experiments 123, e55904.
Sanes, J. R. & Lichtman, J. W. (2007). Transgenic strategies for combinatorial Ochareon, P. & Herring, S. W. (2011). Cell replication in craniofacial periosteum:
expression of fluorescent proteins in the nervous system. Nature 450(7166), 56–62. appositional vs. resorptive sites. Journal of Anatomy 218(3), 285–297.
Logan, M., Martin, J. F., Nagy, A., Lobe, C., Olson, E. N. & Tabin, C. J. (2002). Ono, N. & Kronenberg, H. M. (2016). Bone repair and stem cells. Current Opinion in
Expression of Cre recombinase in the developing mouse limb bud driven by a Prxl Genetics & Development 40, 103–107.
enhancer. Genesis 33(2), 77–80. Ono, N., Ono, W., Nagasawa, T. & Kronenberg, H. M. (2014). A subset of
Lotinun, S., Kiviranta, R., Matsubara, T., Alzate, J. A., Neff, L., Luth, A., chondrogenic cells provides early mesenchymal progenitors in growing bones.
Koskivirta, I., Kleuser, B., Vacher, J., Vuorio, E., Horne, W. C. & Nature Cell Biology 16(12), 1157–1167.
Baron, R. (2013). Osteoclast-specific cathepsin K deletion stimulates S1P- Ortinau, L. C., Wang, H., Lei, K., Deveza, L., Jeong, Y., Hara, Y., Grafe, I.,
dependent bone formation. Journal of Clinical Investigation 123(2), 666–681. Rosenfeld, S. B., Lee, D., Lee, B., Scadden, D. T. & Park, D. (2019).

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


374 Shuo Huang et al.

Identification of functionally distinct Mx1+alphaSMA+ periosteal skeletal stem Shintaku, Y., Murakami, T., Yanagita, T., Kawanabe, N., Fukunaga, T.,
cells. Cell Stem Cell 25(6), 784–796. Matsuzaki, K., Uematsu, S., Yoshida, Y., Kamioka, H., Takano-
Ouyang, Z., Chen, Z., Ishikawa, M., Yue, X., Kawanami, A., Leahy, P., Yamamoto, T., Takada, K. & Yamashiro, T. (2011). Sox9 expression during
Greenfield, E. M. & Murakami, S. (2013). Prx1 and 3.2kb Col1a1 promoters fracture repair. Cells, Tissues, Organs 194(1), 38–48.
target distinct bone cell populations in transgenic mice. Bone 58, 136–145. Shirley, D., Marsh, D., Jordan, G., McQuaid, S. & Li, G. (2005). Systemic
Ozaki, A., Tsunoda, M., Kinoshita, S. & Saura, R. (2000). Role of fracture recruitment of osteoblastic cells in fracture healing. Journal of Orthopaedic Research 23
hematoma and periosteum during fracture healing in rats: interaction of fracture (5), 1013–1021.
hematoma and the periosteum in the initial step of the healing process. Journal of da Silva Meirelles, L., Caplan, A. I. & Nardi, N. B. (2008). In search of the
Orthopaedic Science 5(1), 64–70. in vivo identity of mesenchymal stem cells. Stem Cells 26(9), 2287–2299.
Pagni, G., Kaigler, D., Rasperini, G., Avila-Ortiz, G., Bartel, R. & Song, L. & Tuan, R. S. (2004). Transdifferentiation potential of human mesenchymal
Giannobile, W. V. (2012). Bone repair cells for craniofacial regeneration. stem cells derived from bone marrow. Faseb Journal Official Publication of the Federation of
Advanced Drug Delivery Reviews 64(12), 1310–1319. American Societies for Experimental Biology 18(9), 980–982.
Panwar, P., Xue, L., Soe, K., Srivastava, K., Law, S., Delaisse, J. M. & Sparks, D. S., Saifzadeh, S., Savi, F. M., Dlaska, C. E., Berner, A., Henkel, J.,
Bromme, D. (2017). An Ectosteric inhibitor of Cathepsin K inhibits bone Reichert, J. C., Wullschleger, M., Ren, J., Cipitria, A., McGovern, J. A.,
resorption in Ovariectomized mice. Journal of Bone and Mineral Research 32(12), Steck, R., Wagels, M., Woodruff, M. A., Schuetz, M. A. & ET, A. L.
2415–2430. (2020). A preclinical large-animal model for the assessment of critical-size load-
Park, D., Spencer, J. A., Koh, B. I., Kobayashi, T., Fujisaki, J., Clemens, T. L., bearing bone defect reconstruction. Nature Protocols 15(3), 877–924.
Lin, C. P., Kronenberg, H. M. & Scadden, D. T. (2012). Endogenous bone Sugiyama, T., Kohara, H., Noda, M. & Nagasawa, T. (2006). Maintenance of the
marrow MSCs are dynamic, fate-restricted participants in bone maintenance and hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone
regeneration. Cell Stem Cell 10(3), 259–272. marrow stromal cell niches. Immunity 25(6), 977–988.
Park, D., Spencer, J. A., Lin, C. P. & Scadden, D. T. (2014). Sequential in vivo Susaki, E. A., Tainaka, K., Perrin, D., Yukinaga, H., Kuno, A. & Ueda, H. R.
imaging of osteogenic stem/progenitor cells during fracture repair. Journal of (2015). Advanced CUBIC protocols for whole-brain and whole-body clearing and
Visualized Experiments 87, e51289. imaging. Nature Protocols 10(11), 1709–1727.
Pittenger, M. F., Mackay, A. M., Beck, S. C., Jaiswal, R. K., Douglas, R., Tachi, K., Takami, M., Sato, H., Mochizuki, A., Zhao, B., Miyamoto, Y.,
Mosca, J. D., Moorman, M. A., Simonetti, D. W., Craig, S. & Tsukasaki, H., Inoue, T., Shintani, S., Koike, T., Honda, Y., Suzuki, O.,
Marshak, D. R. (1999). Multilineage potential of adult human mesenchymal Baba, K. & Kamijo, R. (2011). Enhancement of bone morphogenetic protein-
stem cells. Science 284(5411), 143–147. 2-induced ectopic bone formation by transforming growth factor-beta1. Tissue
Popowics, T. E., Zhu, Z. & Herring, S. W. (2002). Mechanical properties of the Engineering Part A 17(5–6), 597–606.
periosteum in the pig, Sus scrofa. Archives of Oral Biology 47(10), 733–741. Tikhonova, A. N., Dolgalev, I., Hu, H., Sivaraj, K. K., Hoxha, E., Cuesta-
Ransom, R. C., Carter, A. C., Salhotra, A., Leavitt, T., Marecic, O., Dominguez, A., Pinho, S., Akhmetzyanova, I., Gao, J., Witkowski, M.,
Murphy, M. P., Lopez, M. L., Wei, Y., Marshall, C. D., Shen, E. Z., Guillamot, M., Gutkin, M. C., Zhang, Y., Marier, C., Diefenbach, C. &
Jones, R. E., Sharir, A., Klein, O. D., Chan, C. K. F., Wan, D. C. & ET, A. L. (2019). The bone marrow microenvironment at single-cell resolution.
ET, A. L. (2018). Mechanoresponsive stem cells acquire neural crest fate in jaw Nature 569(7755), 222–228.
regeneration. Nature 563(7732), 514–521. Tokoyoda, K., Egawa, T., Sugiyama, T., Choi, B. I. & Nagasawa, T. (2004).
Roach, H. I. & Erenpreisa, J. (1996). The phenotypic switch from chondrocytes to Cellular niches controlling B lymphocyte behavior within bone marrow during
bone-forming cells involves asymmetric cell division and apoptosis. Connective Tissue development. Immunity 20(6), 707–718.
Research 35(1–4), 85–91. Truong, V. A., Hsu, M. N., Kieu Nguyen, N. T., Lin, M. W., Shen, C. C.,
Roach, H. I., Erenpreisa, J. & Aigner, T. (1995). Osteogenic differentiation of Lin, C. Y. & Hu, Y. C. (2019). CRISPRai for simultaneous gene activation and
hypertrophic chondrocytes involves asymmetric cell divisions and apoptosis. Journal inhibition to promote stem cell chondrogenesis and calvarial bone regeneration.
of Cell Biology 131(2), 483–494. Nucleic Acids Research 47(13), e74.
Robey, P. G. (2011). Cell sources for bone regeneration: the good, the bad, and the Tsang, K. Y., Tsang, S. W., Chan, D. & Cheah, K. S. (2014). The chondrocytic
ugly (but promising). Tissue Engineering Part B-Reviews 17(6), 423–430. journey in endochondral bone growth and skeletal dysplasia. Birth Defects Research
Rodda, S. J. & McMahon, A. P. (2006). Distinct roles for hedgehog and canonical Part C Embryo Today Reviews 102(1), 52–73.
Wnt signaling in specification, differentiation and maintenance of osteoblast Tsang, K. Y., Chan, D. & Cheah, K. S. (2015). Fate of growth plate hypertrophic
progenitors. Development 133(16), 3231–3244. chondrocytes: death or lineage extension? Development Growth & Differentiation 57(2),
Roguljic, H., Matthews, B. G., Yang, W., Cvija, H., Mina, M. & Kalajzic, I. 179–192.
(2013). In vivo identification of periodontal progenitor cells. Journal of Dental Turner, R. T., Iwaniec, U. T., Wong, C. P., Lindenmaier, L. B., Wagner, L. A.,
Research 92(8), 709–715. Branscum, A. J., Menn, S. A., Taylor, J., Zhang, Y., Wu, H. & Sibonga, J. D.
Rot, C., Stern, T., Blecher, R., Friesem, B. & Zelzer, E. (2014). A mechanical (2013). Acute exposure to high dose gamma-radiation results in transient activation
Jack-like mechanism drives spontaneous fracture healing in neonatal mice. of bone lining cells. Bone 57(1), 164–173.
Developmental Cell 31(2), 159–170. Usami, Y., Gunawardena, A. T., Francois, N. B., Otsuru, S., Takano, H.,
Sakaguchi, R., Leiwe, M. N. & Imai, T. (2018). Bright multicolor labeling of Hirose, K., Matsuoka, M., Suzuki, A., Huang, J., Qin, L., Iwamoto, M.,
neuronal circuits with fluorescent proteins and chemical tags. eLife 7, e40350. Yang, W., Toyosawa, S. & Enomoto-Iwamoto, M. (2019). Possible
Sanchez-Gurmaches, J., Hsiao, W. Y. & Guertin, D. A. (2015). Highly selective contribution of Wnt-responsive Chondroprogenitors to the postnatal murine
in vivo labeling of subcutaneous white adipocyte precursors with Prx1-Cre. Stem growth plate. Journal of Bone and Mineral Research 34(5), 964–974.
Cell Reports 4(4), 541–550. Utvag, S. E., Grundnes, O. & Reikeraos, O. (1996). Effects of periosteal stripping
Sanchez-Ramos, J. R. (2002). Neural cells derived from adult bone marrow and on healing of segmental fractures in rats. Journal of Orthopaedic Trauma 10(4), 279–284.
umbilical cord blood. Journal of Neuroscience Research 69(6), 880–893. Vi, L., Baht, G. S., Whetstone, H., Ng, A., Wei, Q., Poon, R., Mylvaganam, S.,
Schindeler, A., McDonald, M. M., Bokko, P. & Little, D. G. (2008). Bone Grynpas, M. & Alman, B. A. (2015). Macrophages promote osteoblastic
remodeling during fracture repair: the cellular picture. Seminars in Cell & differentiation in-vivo: implications in fracture repair and bone homeostasis.
Developmental Biology 19(5), 459–466. Journal of Bone and Mineral Research 30(6), 1090–1102.
Sempuku, T., Ohgushi, H., Okumura, M. & Tamai, S. (1996). Osteogenic Vi, L., Baht, G. S., Soderblom, E. J., Whetstone, H., Wei, Q., Furman, B.,
potential of allogeneic rat marrow cells in porous hydroxyapatite ceramics: a Puviindran, V., Nadesan, P., Foster, M., Poon, R., White, J. P.,
histological study. Journal of Orthopaedic Research 14(6), 907–913. Yahara, Y., Ng, A., Barrientos, T., Grynpas, M. & ET, A. L. (2018).
Senarath-Yapa, K., McArdle, A., Renda, A., Longaker, M. T. & Quarto, N. Macrophage cells secrete factors including LRP1 that orchestrate the rejuvenation
(2014). Adipose-derived stem cells: a review of signaling networks governing cell of bone repair in mice. Nature Communications 9(1), 5191.
fate and regenerative potential in the context of craniofacial and long bone skeletal Visnjic, D., Kalajzic, I., Gronowicz, G., Aguila, H. L., Clark, S. H.,
repair. International Journal of Molecular Sciences 15(6), 9314–9330. Lichtler, A. C. & Rowe, D. W. (2001). Conditional ablation of the osteoblast lineage
Shah, K., Majeed, Z., Jonason, J. & O’Keefe, R. J. (2013). The role of muscle in in Col2.3deltatk transgenic mice. Journal of Bone and Mineral Research 16(12), 2222–2231.
bone repair: the cells, signals, and tissue responses to injury. Current Osteoporosis Vortkamp, A., Pathi, S., Peretti, G. M., Caruso, E. M., Zaleske, D. J. &
Reports 11(2), 130–135. Tabin, C. J. (1998). Recapitulation of signals regulating embryonic bone
Shapiro, F. (2008). Bone development and its relation to fracture repair. The role of formation during postnatal growth and in fracture repair. Mechanisms of Development
mesenchymal osteoblasts and surface osteoblasts. European Cells & Materials 15, 53–76. 71(1–2), 65–76.
Shi, Y., He, G., Lee, W. C., McKenzie, J. A., Silva, M. J. & Long, F. (2017). Gli1 Walkley, C. R., Shea, J. M., Sims, N. A., Purton, L. E. & Orkin, S. H. (2007). Rb
identifies osteogenic progenitors for bone formation and fracture repair. Nature regulates interactions between hematopoietic stem cells and their bone marrow
Communications 8(1), 2043. microenvironment. Cell 129(6), 1081–1095.

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society


Reparative cells in bone regeneration and repair 375

Wang, W., Nyman, J. S., Ono, K., Stevenson, D. A., Yang, X. & Elefteriou, F. Yang, L., Tsang, K. Y., Tang, H. C., Chan, D. & Cheah, K. S. E. (2014a).
(2011a). Mice lacking Nf1 in osteochondroprogenitor cells display skeletal dysplasia Hypertrophic chondrocytes can become osteoblasts and osteocytes in
similar to patients with neurofibromatosis type I. Human Molecular Genetics 20(20), endochondral bone formation. Proceedings of the National Academy of Sciences of the
3910–3924. United States of America 111(33), 12097–12102.
Wang, X. X., Allen, R. J. Jr., Tutela, J. P., Sailon, A., Allori, A. C., Yang, M., Zhang, H. & Gangolli, R. (2014b). Advances of mesenchymal stem cells
Davidson, E. H., Paek, G. K., Saadeh, P. B., McCarthy, J. G. & derived from bone marrow and dental tissue in craniofacial tissue engineering.
Warren, S. M. (2011b). Progenitor cell mobilization enhances bone healing by Current Stem Cell Research & Therapy 9(3), 150–161.
means of improved neovascularization and osteogenesis. Plastic and Reconstructive Yiannakopoulos, C. K., Kanellopoulos, A. D., Trovas, G. P., Dontas, I. A. &
Surgery 128(2), 395–405. Lyritis, G. P. (2008). The biomechanical capacity of the periosteum in intact long
Wang, T., Zhang, X. & Bikle, D. D. (2017). Osteogenic differentiation of periosteal bones. Archives of Orthopaedic & Trauma Surgery 128(1), 117–120.
cells during fracture healing. Journal of Cellular Physiology 232(5), 913–921. Yu, Y. Y., Lieu, S., Lu, C. & Colnot, C. (2010). Bone morphogenetic protein
Wilk, K., Yeh, S. A., Mortensen, L. J., Ghaffarigarakani, S., 2 stimulates endochondral ossification by regulating periosteal cell fate during
Lombardo, C. M., Bassir, S. H., Aldawood, Z. A., Lin, C. P. & Intini, G. bone repair. Bone 47(1), 65–73.
(2017). Postnatal Calvarial skeletal stem cells expressing PRX1 reside exclusively in Yukata, K., Xie, C., Li, T. F., Takahata, M., Hoak, D., Kondabolu, S., Zhang, X. P.,
the Calvarial sutures and are required for bone regeneration. Stem Cell Reports 8(4), Awad, H. A., Schwarz, E. M., Beck, C. A., Jonason, J. H. & O’Keefe, R. J. (2014).
933–946. Aging periosteal progenitor cells have reduced regenerative responsiveness to bone injury
Wolock, S. L., Krishnan, I., Tenen, D. E., Matkins, V., Camacho, V., and to the anabolic actions of PTH 1-34 treatment. Bone 62, 79–89.
Patel, S., Agarwal, P., Bhatia, R., Tenen, D. G., Klein, A. M. & Zelzer, E., Mamluk, R., Ferrara, N., Johnson, R. S., Schipani, E. &
Welner, R. S. (2019). Mapping distinct bone marrow niche populations and their Olsen, B. R. (2004). VEGFA is necessary for chondrocyte survival during bone
differentiation paths. Cell Reports 28(2), 302–311. development. Development 131(9), 2161–2171.
Worthley, D. L., Churchill, M., Compton, J. T., Tailor, Y., Rao, M., Si, Y., Zhang, X., Xie, C., Lin, A. S., Ito, H., Awad, H., Lieberman, J. R.,
Levin, D., Schwartz, M. G., Uygur, A., Hayakawa, Y., Gross, S., Rubery, P. T., Schwarz, E. M., O’Keefe, R. J. & Guldberg, R. E. (2005).
Renz, B. W., Setlik, W., Martinez, A. N., Chen, X. & ET, A. L. (2015). Periosteal progenitor cell fate in segmental cortical bone graft transplantations:
Gremlin 1 identifies a skeletal stem cell with bone, cartilage, and reticular stromal implications for functional tissue engineering. Journal of Bone and Mineral Research 20
potential. Cell 160(1–2), 269–284. (12), 2124–2137.
Wosczyna, M. N., Biswas, A. A., Cogswell, C. A. & Goldhamer, D. J. (2012). Zhao, H., Feng, J., Ho, T. V., Grimes, W., Urata, M. & Chai, Y. (2015). The
Multipotent progenitors resident in the skeletal muscle interstitium exhibit robust suture provides a niche for mesenchymal stem cells of craniofacial bones. Nature
BMP-dependent osteogenic activity and mediate heterotopic ossification. Journal of Cell Biology 17(4), 386–396.
Bone and Mineral Research 27(5), 1004–1017. Zhao, S., Todorov, M. I., Cai, R., Maskari, R. A., Steinke, H., Kemter, E.,
Wynn, T. A. & Vannella, K. M. (2016). Macrophages in tissue repair, regeneration, Mai, H., Rong, Z., Warmer, M., Stanic, K., Schoppe, O., Paetzold, J. C.,
and fibrosis. Immunity 44(3), 450–462. Gesierich, B., Wong, M. N., Huber, T. B. & ET, A. L. (2020). Cellular and
Xie, C., Ming, X., Wang, Q., Schwarz, E. M., Guldberg, R. E., O’Keefe, R. J. & molecular probing of intact human organs. Cell 180(4), 796–812.e19.
Zhang, X. (2008). COX-2 from the injury milieu is critical for the initiation of Zheng, B., Cao, B., Li, G. & Huard, J. (2006). Mouse adipose-derived stem cells
periosteal progenitor cell mediated bone healing. Bone 43(6), 1075–1083. undergo multilineage differentiation in vitro but primarily osteogenic and
Yahara, Y., Barrientos, T., Tang, Y. J., Puviindran, V., Nadesan, P., chondrogenic differentiation in vivo. Tissue Engineering 12(7), 1891–1901.
Zhang, H., Gibson, J. R., Gregory, S. G., Diao, Y., Xiang, Y., Qadri, Y. J., Zhong, L., Yao, L., Tower, R. J., Wei, Y., Miao, Z., Park, J., Shrestha, R.,
Souma, T., Shinohara, M. L. & Alman, B. A. (2020). Erythromyeloid Wang, L., Yu, W., Holdreith, N., Huang, X., Zhang, Y., Tong, W.,
progenitors give rise to a population of osteoclasts that contribute to bone Gong, Y., Ahn, J. & ET, A. L. (2020). Single cell transcriptomics identifies a
homeostasis and repair. Nature Cell Biology 22(1), 49–59. unique adipose lineage cell population that regulates bone marrow environment.
Yang, X., Ricciardi, B. F., Hernandez-Soria, A., Shi, Y., Pleshko eLife 9, e54695.
Camacho, N. & Bostrom, M. P. (2007). Callus mineralization and maturation Zhou, B. O., Yue, R., Murphy, M. M., Peyer, J. G. & Morrison, S. J. (2014a).
are delayed during fracture healing in interleukin-6 knockout mice. Bone 41(6), Leptin-receptor-expressing mesenchymal stromal cells represent the main source
928–936. of bone formed by adult bone marrow. Cell Stem Cell 15(2), 154–168.
Yang, W., Wang, J., Moore, D. C., Liang, H., Dooner, M., Wu, Q., Terek, R., Zhou, X., von der Mark, K., Henry, S., Norton, W., Adams, H. & de
Chen, Q., Ehrlich, M. G., Quesenberry, P. J. & Neel, B. G. (2013). Ptpn11 Crombrugghe, B. (2014b). Chondrocytes transdifferentiate into osteoblasts in
deletion in a novel progenitor causes metachondromatosis by inducing hedgehog endochondral bone during development, postnatal growth and fracture healing in
signalling. Nature 499(7459), 491–495. mice. PLoS Genetics 10(12), e1004820.

(Received 1 April 2020; revised 6 October 2020; accepted 7 October 2020; published online 13 October 2020 )

Biological Reviews 96 (2021) 357–375 © 2020 Cambridge Philosophical Society

You might also like