Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

| |

Received: 21 August 2019    Revised: 14 November 2019    Accepted: 31 December 2019

DOI: 10.1111/exd.14070

FOCUS THEME ISSUE: REVIEW ARTICLE

Immune cell regulation of the hair cycle

Etienne C. E. Wang1  | Claire A. Higgins2

1
Skin Research Institute of Singapore (SRIS),
National Skin Centre, Singapore, Singapore Abstract
2
Department of Bioengineering, Imperial The ability to manipulate the mammalian hair cycle will lead to novel therapies and
College London, London, UK
strategies to combat all forms of alopecia. Thus, in addition to the epithelial-mes-
Correspondence enchymal interactions in the hair follicle, niche and microenvironmental signals that
Etienne Wang, National Skin Centre,
accompany the phases of growth, regression and rest need to be scrutinized. Immune
1 Mandalay Road, Singapore 308205,
Singapore. cells are well described in skin homeostasis and wound healing and have recently
Email: etienne@nsc.com.sg
been shown to play an important role in the mammalian hair cycle. In this review, we
will summarize our current knowledge of the role of immune cells in hair cycle control
and discuss their relevance to human hair cycling disorders. Increased attention to
this aspect of the hair cycle will provide new avenues to manipulate hair regeneration
in humans and provide better insight into developing better ex vivo models of hair
growth.

KEYWORDS

anagen, catagen, macrophages, mast cells, T cells, telogen

1 | I NTRO D U C TI O N are precisely co-ordinated. This co-ordination is partially controlled


by intrinsic properties of the HFSC, but is also reliant on the HF
Alopecia, or loss of hair, is caused by the disrupted integrity of the microenvironment, made up of surrounding cells and tissues that
hair follicle (HF), a sine qua non appendage of mammalian skin. The contribute to the HFSC niche.[4] These cell types include the dermal
main role of the hair follicle is to produce the keratinaceous hair papilla (DP),[5,6] arrector pili muscle,[7] adipose tissue,[8] blood ves-
shaft, whose maximum length in situ is dictated by the length of sels [9] and even lymphatics.[10] Historically, immune cells have been
the growth phase of the hair cycle, which varies across body sites described to be closely associated with hair follicles,[11] with char-
[1]
and between species. The hair follicle cycles due to complex, or- acteristic compositions at each stage of the hair cycle,[12] but their
chestrated fluxes of stem cells which arise from distinct populations influence on the HFSC niche has only recently been uncovered.
of keratinocytes of the HF, with the slowest cycling population in In this review, we have assembled the current knowledge on
the bulge region, responding to morphogenetic and inhibitory sig- immune cells in and around the HF. We detail the role of immune
nals coming from neighbouring cells.[2,3] Disorders of the hair follicle cells in various stages of HF cycling (in both mouse and humans) and
include classically immune-mediated alopecias like alopecia areata discuss immune cells in the context of common human diseases that
(AA) and cicatricial alopecias (eg. discoid lupus erythematosus, li- result from interference of the normal hair cycle.
chen planopilaris) or non–immune-mediated alopecias like androge-
netic alopecia and telogen effluvium. These various disorders occur
when the co-ordination of the hair cycle and functions of the hair 2 | I M M U N E C E LL S A N D TI S S U E
follicle stem cells (HFSCs) have become impaired. H O M EOS TA S I S
At steady state, after the HF has completed morphogenesis (a
process that begins in utero), hair follicles go through cyclic phases Specialized macrophages, known as microglia, in the central nerv-
of growth (anagen), regression (catagen) and relative rest (telogen), ous system have long been known to be responsible for the mainte-
whereby keratinocyte proliferation, differentiation and quiescence nance of synaptic health and optimal function of neurons.[13] More

|
322     © 2020 John Wiley & Sons A/S. wileyonlinelibrary.com/journal/exd Experimental Dermatology. 2020;29:322–333.
Published by John Wiley & Sons Ltd
WANG and HIGGINS |
      323

recently, immune cells, in addition to their nascent functions of an- sheath (ORS) and matrix,[31] and a loss in volume of the DP (caused
tigen surveillance and microbial defense, have started to emerge as by a loss of extracellular matrix and migration of cells out of the
significant contributors to tissue homeostasis in a variety of other DP[32]). The HF begins to regress and the bulb (consisting of the few
tissues. For example, another subset of macrophages have adapted remaining matrix keratinocytes and the DP) as drawn away from the
to facilitate propagation of membrane potential and contractility of subcutaneous adipose tissue towards the epidermis (Figure 1A).
[14]
cardiomyocytes in the heart. In the bone marrow, immune cells The decision for a terminal HF to enter catagen is likely a com-
such as macrophages, neutrophils and T regulatory (T reg) cells dou- bination of signals from the microenvironment, including bone
ble up as components of the hematopoietic stem cell niche,[15‒18] as morphogenetic protein (BMP) signalling from the underlying adi-
do specialized macrophages in the skeletal muscle.[19] pose tissue.[33] In human HFs in culture, transforming growth factor
In the interfollicular epidermis (IFE) of the skin, Langerhans cells (TGF)-β1 and TGF-β2 induce catagen and are likely produced by the
and dendritic epidermal T cells (DETCs) are found in close approx- DP.[34,35] Interferon (IFN)-γ, which is typically produced by lymphoid
imation with basal keratinocyte stem cells,[20] while DETCs have cells of the immune system, is also a potent inducer of catagen.[36]
been shown to support the IFE niche via insulin-like growth factor FGF-5 from perifollicular macrophages have also been implicated in
[21] [22]
(IGF)-1 and wound healing via fibroblast growth factor (FGF)-7. promoting catagen,[37] while FGF-5 mutations result in trichomegaly
Immune cells are recruited to the developing HF during em- of the eyelashes in humans[30,38] and the long-haired angora pheno-
bryogenesis and have been described to change dynamically with type in several mammalian species, including mice[39] and dogs.[40,41]
the post-natal hair cycle.[23] The HF epithelium is also densely popu- This knowledge has allowed researchers to disrupt the FGF-5 gene
[24‒26]
lated by DETCs in both mice and humans. Likewise, dermal mac- in cashmere goats, resulting in gene-edited animals with prolonged
rophages are found to be associated with almost every stage of the hair anagen that result in longer coats for the manufacture of clothing.[42]
[27,28]
cycle and may play multiple roles in the maintenance of the HF. Mast cells, which are the arbiters of allergic inflammation, also
play a role HF regression.[43] Mast cell degranulation is associated
with catagen HFs, and granule contents substance P and adrenocorti-
3 |  S P O NTA N EO U S H A I R C YC LI N G cotrophic hormone (ACTH) induce catagen in mouse HFs in vivo. Mast
cell-deficient mice also have delayed catagen entry, as do neurokinin-1
The hair cycle is an exquisitely orchestrated series of cellular in- knock-out mice that cannot respond to substance P secretion.[44]
teractions between the epidermal keratinocytes and mesenchymal Interestingly, the HF contains mast cell precursors in mice,[45] which
“control centre” in the DP. Evolutionarily, the hair cycle functions to can differentiate into mature mast cells under appropriate conditions
provide a protective coat of fur for mammals, which has the capacity and contribute to wound healing in the skin.[46] Mast cells are also as-
to regenerate easily when plucked, to moult and shed periodically, or sociated with HFs in human skin,[47] and substance P produced by mast
change colour in order to adapt to temperate climates with changing cells may contribute to HF regression by upregulating nerve growth
seasons. The spontaneous hair cycle is the rhythmic progression of factor (NGF) and its receptor p75NTR on HF ORS keratinocytes.[48]
anagen-catagen-telogen-anagen in an unperturbed animal and can Cellular debris (apoptotic keratinocytes and redundant extra-
be disrupted, induced and stimulated experimentally. cellular matrix, ECM) from the regressing HF are cleared by macro-
In rodents and other small mammals, the spontaneous hair cycle phages[49] and possibly neighbouring keratinocytes, which appear to
is tightly synchronized across their entire coat for the first few take on phagocytic abilities.[50] As the HF regresses, the DP reduces
months of life, and anagen can be observed spreading from the ante- in size and migrates upwards, leaving a “fibrous streamer” of degen-
rior to the posterior integument in waves. Inbred laboratory strains erating elastic fibres.[51] Macrophages are involved in the clearance
like the C57BL/6 mouse are an ideal model animal to observe hair of this streamer, as histiocytes and multinucleated giant cells are ob-
cycling because the phases of anagen, catagen and telogen are pre- served in association with it.[52] The control of DP size during the
dictable according to their age up to about 80-100 days after birth hair cycle is currently poorly understood and may be a combination
(postnatal day 80-100 or P80-P100), at which time they enter their of fluctuations in cell number and ECM components.[32,53] Whether
third, asynchronous anagen. The co-ordination of the synchronous immune cells are involved in this process is still unknown.
waves of anagen in these animal models is currently under investiga-
tion[29] and may involve juxtacrine signals from the DP, IFE, subder-
mal adipose tissue and very likely also the immune cells (summarized 3.2 | Telogen
in Table 1 and Figure 1).
At the end of catagen, the HF enters the telogen phase, whereby the
DP has reduced in size and is located subjacent to the base of the so-
3.1 | Catagen called “permanent portion” of the HF (Figure 1B). During this phase
of “rest,” the bulge and ORS keratinocytes are mitotically quiescent,
At the end of HF morphogenesis, the completed terminal HF enters and the hair shaft (“club hair”) remains anchored in the HF. This appar-
the first catagen, around P19 in a C57BL/6 mouse.[30] This process ent “quiescence” of the HF is the result of active inhibitory macro- and
involves apoptosis of the keratinocytes that form the outer root microenvironment signals from cell types comprising the HF niche,
|
324       WANG and HIGGINS

TA B L E 1   Immune cells implicated in mammalian hair cycle

  Murine hair cycle Human hair cycle


[43]
Catagen Mast cells accumulate and degranulate during catagen. Macrophages undergo apoptosis and shirt to an “M2-like”
Macrophages may induce catagen by producing FGF-5[37] and may clear phenotype.[98]
debris from regressing HF.[49] FGF-5 mutations in humans result in delayed catagen
(prolong anagen)—cellular source yet to be identified[38]
Telogen Macrophages numbers increase during mid-telogen,[56] and a subset No data at the time of publication.
of TREM2+ macrophages maintain HFSC quiescence by producing
OSM.[55]
Ablation of macrophages using clodronate liposomes[56] or genetic/
pharmacologic methods [55] leads to anagen.
Early Numbers of macrophages decrease, possibly by apoptosis and release Downregulation of MHC Class I molecules on
anagen of Wnt ligands that stimulate anagen.[56] keratinocytes.[12]
T regs cluster around HFSC during the telogen-anagen transition.[57]
Mid-late Adipocyte niche produces macrophage-stimulating protein that High density of perifollicular macrophages and mast cells.
anagen maintains anagen.[71] MHC Class I downregulation and low numbers of NK
cells.[12]

including the Krt6+ inner bulge,[54] arrector pili muscle,[7] subdermal The telogen-to-anagen transition is a clinically relevant phase of
adipocytes[33] and HF-associated immune cells.[55] In the period imme- the hair cycle, as understanding its control might allow us to replicate
diately following catagen, the HF is in a state of refractory telogen, in it in patients with various forms of recalcitrant alopecia. In mouse skin,
which the threshold for anagen re-entry is higher, due in part to the the density of perifollicular macrophages decreases at the end of telo-
increased concentrations of BMPs secreted by neighbouring adipo- gen,[55,56] suggesting a process that is coupled to anagen re-entry, per-
[33]
cytes. By mid-telogen, these inhibitory signals are reduced, and ana- haps via macrophage apoptosis and release of Wnt ligands.[56]
gen is more readily induced by experimental means such as plucking, The anagen HF interacts with the immune system in a way that
depilation and topical application of drugs or chemicals. promotes immune privilege and prevents autoimmune attack of the
Immune cell numbers wax and wane according to the phase of shifting antigens in the growing HF. MHC class I expression on ana-
the hair cycle, but both T regulatory (T reg) cells and macrophages gen keratinocytes is upregulated,[65] and a relative immunosuppres-
[56‒58]
are most abundant during mid-telogen, consistent with their sion is observed in the murine skin.[66]
roles in tissue homeostasis and remodelling. Systemic administra- As anagen progresses, the DP enlarges in size, and the HF
tion of clodronate liposomes during mid-telogen, which ablates begins to extend into the adipose tissue. The adipose layer also
phagocytic cells (including macrophages, but also other cell types), thickens during anagen, in a manner that appears to be synchro-
has been shown to induce early anagen re-entry in mice. Single-cell nized with the HF cycle.[67] Adipocytes provide a supportive niche
RNA sequencing of murine macrophages during telogen identified for the growing anagen HF by producing a myriad adipokines
a rare subset of TREM2+ macrophages (coined “trichophages”) like leptin[68] and adiponectin [69]
; and growth factors, including
[55]
that produce oncostatin M (OSM), a potent inhibitor of HFSC macrophage colony-stimulating factor (M-CSF).[70] While macro-
proliferation and differentiation.[59] OSM maintains HFSC quies- phage-stimulating protein (MSP) has a direct effect on the anagen
cence via the JAK-STAT5 pathway, and anagen can be induced by HF,[71] its influence on the immune milieu itself during anagen has
topical janus kinase (JAK) inhibitors in wild-type C57BL/6 mice not been reported.
in telogen.[60] More specific methods of inhibiting macrophages Human HFs cultured ex vivo was shown to be maintained in ana-
in the dermis during telogen (ie. with genetic, immunological and gen with insulin, IGF-1 and IGF-2, and withdrawal of these factors led
pharmacological means) also initiates the telogen-to-anagen tran- to catagen entry.[72] Whether DETCs are the source of IGF-1 in hu-
[55]
sition in C57BL/6 mice. mans, as in the mouse during induced anagen,[24] is currently unknown.
The involvement of immune cells during spontaneous anagen remains
largely unexplored, as most studies on this phase of the hair cycle em-
3.3 | Anagen ploy the induced anagen model in small laboratory mammals.

The beginning of the growth phase (“anagen”) starts with prolifera-


tion in the DP, which produces stimulatory signals such as Wnt10b,[61] 3.4 | Exogen/Kenogen
FGF-2, FGF-7[62] and BMP antagonists[33] to induce proliferation of the
matrix keratinocytes in the hair germ (HG). The proliferation of the HG There are two sub-phases of the cycle that are independent of the
sends a signal (like Sonic Hedgehog (Shh)[63]) to the HF bulge to acti- cycling phases (anagen-catagen-telogen) that capture the physical
vate the quiescent, long-lived HFSCs that stimulates proliferation of shedding of the club hair from the follicle (“exogen”)[73] and a short
cells that will populate the ORS of the new anagen HF[64] (Figure 1C). period whereby a telogen HF is without a hair shaft (“kenogen”).[74]
WANG and HIGGINS |
      325

F I G U R E 1   Schematic of the spontaneous mammalian hair cycle, with current knowledge in mouse and human hair cycle depicted in
embedded tables. A, The previous hair cycle, or HF morphogenesis, ends with catagen whereby the HF regresses. Keratinocyte apoptosis, and
shrinking of the DP, are accompanied by mast cell and macrophage infiltration of the HF in mice. Macrophages secrete FGF-5, which has been
shown to induce catagen in many mammalian species. FGF-5 mutations in humans result in delayed catagen and prolonged anagen, leading to
trichomegaly of the eyelashes. B, The telogen HF is mitotically quiescent, but immune cells like macrophages and T reg cells have been described
to be in proximity with these HFs in mice. Quiescence of HFSCs during telogen is controlled by many factors secreted by HF niche cells, which
have been shown to include macrophages. There are currently no data on the immune cell infiltrate associated with human telogen HFs. Grey
dotted box denotes the “permanent portion” of the HF which is present at every stage of the hair cycle, as opposed to the “cycling portion” that
grows and regresses with each cycle. C, Anagen entry is marked by proliferation in the HFSCs in the matrix and bulge. Macrophage apoptosis is
postulated to be involved in this process in both mice and humans. The involvement of immune cells in the spontaneous hair cycle in both mice
and humans is currently poorly studied as most experiments utilize induced anagen models in rodents and small animals. D, Physical shedding of
the club hair fibre is known as exogen. HF, hair follicle; HFSC, hair follicle stem cell; ORS, outer root sheath

The term exogen refers to the physical shedding of the club HF. Shedding of the club hair is an active proteolytic process,[75] and
fibre (Figure 1D). To ensure a constant coat of fur in small mam- whether immune cells are involved is unknown. Some intriguing data
mals, the club hair fibre and growing fibre can reside in the same in birds suggest that there may be a link with immune cells and skin
|
326       WANG and HIGGINS

appendage retention; the relative force required to remove feathers be “gently” removed. This process also synchronizes the hair cycle in
(and escape predators) is larger in bird species with a stronger an- mice and begins a phase of induced anagen, which is not observed
ti-parasitic T cell–mediated immune response.[76] after close shaving. Using this method, T regulatory (T reg) cells have
Kenogen is not usually observed on the coats of mammals and is been postulated to be recruited to HFs after depilation in order to
more of a phenomenon restricted to human scalp follicles. Kenogen stimulate HFSC proliferation and differentiation via Jagged1-Notch
is common when the club hair is shed without a new shaft being signalling.[57] However, exposure to harsh chemicals also results
produced, and the empty follicle is in kenogen. This state may be in a degree of inflammation in the interfollicular epidermis[84] and
related to the miniaturized, refractory HFs in androgenetic alopecia might induce cytokine/chemokine release characteristic of a wound
(AGA).[77] Interestingly, in humans, HFs in AGA are associated with response. In fact, models of induced skin inflammation with topical
[78]
deposits of immunoglobulins along the basement membrane, and imiquimod also promote the telogen-to-anagen transition in mice.[85]
[79]
a lymphocytic immune infiltrate, the significance and composition In the epidermis, there is an increase in DETCs following depila-
of which remains uncharacterized. tion,[24] in a manner reminiscent of the wound healing process.[22]
Application of topical compounds onto mouse telogen skin has
also been used to induce anagen entry. In addition to compounds
4 |  I N D U C E D H A I R C YC LI N G such as Shh and Wnt agonists which are known to kick start HFSC
activation and proliferation, topical ciclosporin has been used to in-
Experimental models of the hair cycle in laboratory animals are con- duce the hair cycle in mice. Intriguingly, ciclosporin is used in humans
ceptually distinct from the spontaneous hair cycle. These techniques as a steroid-sparing immunosuppressant for organ transplant and in-
are convenient for synchronizing the hair cycle over the entire animal flammatory diseases, with the known side effect of hypertrichosis.
in adulthood so that observations are easily recorded and studied Both topical ciclosporin application and depilation induced anagen in
(Figure 2). Plucking telogen hairs in a C57BL/6 mouse is known to in- mice that was accompanied by mast cell degranulation at the point
duce a reliable anagen, with a characteristic time course that can be of anagen entry.[86,87] Mast cell stimulators and inhibitors were also
[80]
perturbed and easily examined in the laboratory. Similar effects able to induce and inhibit anagen entry in mice, respectively, sug-
are seen with chemical depilation or within and around accidental or gesting that they may also play a role in induced anagen entry.
experimental wounds, all of which are associated with a significant Anagen has also been induced in murine skin with various tech-
[81]
inflammatory infiltrate. Wounding-induced anagen progression is niques including fractional carbon dioxide laser,[88] microneedling
abnormal in mice lacking DETCs, suggesting shared mechanisms in tis- (with and without application of other chemicals)[89] and mechan-
[82]
sue allostasis. Evolutionarily, this allows the mammal to regenerate ical stretch.[90] Chemokines, in particular CCL2, released by tissue
its coat of fur quickly and efficiently after environmental or predato- damage might recruit anti-inflammatory or “M2-like” macrophages,
rial insults. While many of the signals that coordinate induced anagen which are associated with tissue regeneration and homeostasis. Chu
are similar to those in spontaneous anagen, there may be subtle differ- et al show that these “alternatively activated” macrophages might in-
ences that must be considered when interpreting these studies. duce HF anagen by producing growth factors (IGF-1 and hepatocyte
Early clues that spontaneous and induced hair cycling are dis- growth factor, HGF).[90]
tinct processes come from observations made by Sano et al, in which Hair follicle neogenesis has been observed within larger wounds
epidermal-specific STAT3 knock-out mice had delayed spontaneous of rodents and some small mammals and represents a solution to re-
anagen entry, but normal induced anagen entry with plucking or generate a continuous coat of fur in scarred tissue, which normally
phorbol 12-myristate 13-acetate (PMA) application.[83] The thresh- remains hairless in humans. This process (also known as wound-in-
old for telogen hairs to enter anagen in small mammals and rodents duced HF neogenesis, WIHN) occurs in large wounds[91] and reca-
after injury is hence quite low, and compounds that stimulate anagen pitulates the process of HF morphogenesis.[92] WIHN is distinct from
experimentally may not be relevant to the spontaneous hair cycle or the anagen re-entry of the HFs in the wound periphery, although
to the hair cycle in humans. both appear to be dependent on TNF-induced AKT/β-catenin signal-
Plucking telogen hairs in mice appear to induce anagen of the ling via macrophages.[93] Within the murine scar, macrophages with
surrounding HFs via a diffusible signal and is contingent on the den- an “M2-like” phenotype producing FGF-2 and IGF-1 are necessary for
sity and number of plucked HFs. Chen et al showed that there was WIHN,[94] as are γδ-T cells in the murine dermis that are recruited and
threshold density of plucked HFs that stimulated anagen in their un- secrete FGF-9 to activate dermal fibroblasts to start the HF morpho-
plucked neighbours, and this signal was mediated by the release of genesis cascade.[95] Human skin is almost devoid of γδ-T cells, which is
chemokine (C-C motif) ligand 2 (CCL2) and subsequent recruitment consistent with the inability of human scars to regrow hair.
of inflammatory macrophages that stimulated anagen by secreting
tumor necrosis factor (TNF)-α.[58]
Depilatory creams such as Nair or Veet are commonly used to 5 | H A I R C YC LI N G I N H U M A N D I S E A S E
loosen HFs and induce a chemical depilation. The active ingredients
include salts of thioglycolic or thiolactic acid, which disrupts disul- Unlike mammals with a coat of fur which predominantly contains
phide bonds in the keratinaceous hair shafts, allowing the hairs to HFs in telogen, 90% of human scalp hair is in the growing anagen
WANG and HIGGINS |
      327

F I G U R E 2   Methods of inducing
anagen experimentally in laboratory
mammals (usually mice). Mice in the
telogen phase may be subjected to
plucking, chemical depilation or wounding.
The immune cells that have been shown
to be recruited after some, but not
all, of these procedures include mast
cells,[44] macrophages,[58,90] T reg cells,[57]
DETCs[24] and γδ T cells,[82] depending
on the extent of injury. Growth factors
and cytokines produced by immune cells
play a role in stimulating anagen re-entry.
Unlike spontaneous anagen, this new
anagen phase is not associated with a club
hair fibre. These mechanisms have not
been shown to be present in human hair-
bearing skin

stage, reflected in the need for frequent haircuts. The anagen phase cycle independently of neighbouring follicles, making microenviron-
of human HFs last between 2 and 9 years,[96] making studying the mental regulation of the cycle extremely difficult to assess.
human hair cycle extremely challenging. Most of the knowledge Recently, careful study of microdissected follicular units from
about the human hair cycle is inferred from observations and experi- hair transplant surgery has allowed us to validate the existence of
ments carried out in mammalian (mostly murine) fur. This has obvi- these pathways (in particular Wnt signalling) in human HFs at the
ous limitations, not least as human HFs grow in a mosaic pattern, and telogen-to-anagen transition.[97] Similar techniques show dynamic
|
328       WANG and HIGGINS

macrophage polarization associated with different stages of the hair stem cell quiescence and is downstream of prolactin (PRL) during
cycle, with a possible Wnt-active macrophage subset during the begin- pregnancy[111] or OSM (from perifollicular trichophages) during nor-
[98]
ning of anagen. However, this method is still largely descriptive and mal telogen.[55] Thus, future treatments for AA using JAK inhibitors
correlative. Human HFs xenotransplanted onto immunodeficient mice might address both the immune-mediated attack on the HF, and the
is a model that might allow observation of human HF cycling,[99] but this hair cycle itself, in order to stimulate regrowth in patients.[112]
system is also artificial and does not take into account the extra-follicu- The role of mast cells in the hair cycle (facilitating catagen and
lar macroenvironmental signals, particularly from immune cells. anagen in the mouse) is confounded by their increased presence
Disorders of hair cycling in humans can be disfiguring and cause around human HFs in lesional AA.[113] Mast cells in this context ap-
significant psychological and social sequelae. Most conventional pear to upregulate pro-inflammatory markers, make contacts with
treatments for hair loss disorders are unsatisfactory and usually CD8+ cytotoxic cells and may play a role in immune privilege col-
does not address the hair cycle proper, largely due to our lack of lapse and antigen presentation. As AA episodes have been associ-
knowledge of the human hair cycle. ated with stressful life events, mast cells (via substance P) may play a
significant role in the collapse of immune privilege.[48]

5.1 | Alopecia areata (AA)


5.2 | Androgenetic alopecia (AGA)
Pigmented, anagen HFs are the main target of this autoimmune-
mediated alopecia.[100] Although the target antigen remains elu- Androgenetic alopecia, also known as male- or female-pattern bald-
sive, it is postulated to be upregulated during the anagen phase ness, is the most common form of alopecia and affects up to 40%-
and may be melanocyte-associated,[101] or part of the growing 60% the general adult population.[114‒116] AGA is largely viewed as
outer root sheath (ORS).[102] The hair cycle and its constant flux a cosmetic issue, but has enough negative impact on self-image
of self-antigens make it susceptible to autoimmune attack, if not and psychological well-being to drive significant health-seeking
for the relative immune privilege of the HF.[103] Immune privilege behaviour and spending among patients. FDA-approved therapies
is mediated in part by downregulation of major histocompatibil- for AGA (topical minoxidil, oral finasteride and some low-level light
ity complex (MHC) and natural killer group 2 member D (NKG2D) therapy (LLLT) devices) have inconsistent efficacy, and potentially
[104,105]
ligands on the HF keratinocytes (Figure 3A). Aberrant up- unacceptable side effects, prompting patients to spend a collective
regulation of NKG2D ligands ULBP3/ULBP6 on keratinocytes leads US$3.5 billion a year on unproven baldness treatments, according
to recruitment and activation of NKG2D+ CD8+ cytotoxic T cells to statistics provided by the American Hair Loss Association.
that destroy the HF matrix and induce a premature catagen, lead- The pathognomonic features in AGA are the miniaturization of
ing to non-scarring alopecia. The HF enters telogen, eventually re- HFs, with reduction in the size of DP during each hair cycle, leading
enters the hair cycle and begins the next anagen phase. However, to thinner hair shafts[117] (Figure 3B). The number of follicles in a
if the autoimmune process is still present, the growing anagen HF follicular unit also decreases, and hair density declines in an andro-
is attacked as soon as it begins to produce the anagen-associated gen-dependent manner. This leads to the characteristic bitemporal
autoantigen, is arrested once more in mid-anagen and forced into recession and thinning over the vertex in men[118] and diffuse thinning
a premature catagen yet again. Repeated attacks and truncation with accentuation of the central parting in women.[119] Interestingly,
of the hair cycle ultimately leads to a miniaturized, exhausted and occipital scalp HFs are spared from this process in men; thus, they
dystrophic, empty HF in a state specific to AA known as nano- are ideal donor follicular units in hair transplant surgery. Adipocytes,
[106]
gen. Discovery of the role of cytotoxic NKG2D+ T cells in this which appear to support anagen in mice, are also under investiga-
process has led to the use of JAK inhibitors for the treatment of AA, tion for the treatment of AGA in the form of stem cells derived from
as IFN-γ and IL-15 (crucial cytokines for NKG2D+ T-cell survival) the stromal-vascular fraction enriched through centrifugation[120] or
signal via the JAK-STAT pathway.[107,108] Recently, another subset their conditioned media derived from ex vivo cell culture.[70]
of regulatory invariant NKT (iNKT) cells are postulated to sup- Histologically, perifollicular infundibular inflammation has been
press NKG2D+ T-cell activity to promote regrowth and remission described in AGA,[78,79] but the nature of this infiltration remains un-
in AA.[109] Impairment in T regulatory (T reg) cells have also been characterized. Whether immune cells like macrophages, mast cells or
described in both lesional AA[110] and the circulatory system of AA T regs contribute to the microenvironment of HFs and participate in
patients, a possible permissive factor for the pathogenesis of AA. the miniaturization process remains an open question. Further, the
Preclinical experiments with JAK inhibitors also showed that JAK basic nature of miniaturized HFs, how they relate to the normal hair
inhibition in normal C57BL/6 mice (without AA) was also able to in- cycle, and whether the miniaturization process can be reversed, is
duce a telogen-to-anagen transition without overt inflammation.[60] poorly understood.
Transcriptomic analysis also showed dynamic JAK-STAT signalling The discovery of WIHN has prompted investigation into novel
across the spontaneous hair cycle in C57BL/6 mice, suggesting JAK therapeutic modalities for AGA that aim to recreate the wound mi-
inhibitor-induced anagen might more closely resemble spontaneous croenvironment and assess whether inducing tissue injury might
anagen. Further, JAK-STAT5 signalling in HFSCs is associated with rejuvenate miniaturized HFs.[121] Platelet-rich plasma, harvested
WANG and HIGGINS |
      329

F I G U R E 3   Human diseases of the hair cycle and potential immune cell involvement. A, Alopecia areata is characterized by loss of immune
privilege of anagen HFs. Upregulation of “danger signals” such as MHC Class I and ULBP3/ULBP6 on ORS and matrix keratinocytes attracts
NKG2D + cytotoxic T cells that form the characteristic “swarm of bees” around the bulb region of the HF. B, Androgenetic alopecia (AGA)
results from progressive miniaturization of terminal HFs, with a poorly characterized immune infiltrate around the infundibulum. The hair
shaft becomes finer, and the DP size is reduced. Further work is required to elucidate the role of immune cells in AGA. C, Telogen effluvium
is a biological response to physiological or emotional stress that is also poorly characterized and has no effective and proven treatments. The
premature and usually synchronized telogen leads to shedding of club hairs en masse. Immune involvement in this condition is also pending
interrogation

autologously from the patient's venous blood, has been shown to 5.3 | Telogen effluvium (TE)
have some effect on increasing hair density and preventing hair
shaft miniaturization,[122,123] likely via the release of cytokines Episodes of physical or emotional stress have been linked to the dis-
and growth factors from activated platelets.[124] These factors are ruption of the hair cycle in humans.[132] This is exemplified by the
commonly found in healing wounds and include platelet-derived normally self-limiting condition of TE, commonly experienced around
growth factor (PDGF), vascular endothelial growth factor (VEGF), 3-6 months postpartum,[133] whereby the HFs that entered telogen en
HGF and IGF-1, and an array of pro- and anti-inflammatory cyto- masse during the period of stress undergo shedding or “teloptosis” at
kines.[125] Other techniques like microneedling[126] and fractional the same time a few weeks or months later.[134] Other stressful events
non-ablative laser treatment have also been attempted alone or in that may precipitate TE include crash or fad diets, surgeries or even
combination with drugs or growth factors,[127,128] in order to boost starting a new medication for other medical issues.[135] The sudden
[129]
hair regrowth in AGA patients, with modest results. Low-level hair loss may lead to significant despair and requires detailed history-
laser therapy (LLLT) using visible red light laser-emitting LED de- taking, examination and investigations to exclude other forms of acute
[130]
vices may hold promise in stimulating hair growth in AGA, and and diffuse non-scarring alopecias.[136] Scalp biopsies, if performed at
the mechanism, which is still under investigation, may involve all, may show more than 25% of HFs in telogen.[135] Treatment is usu-
modulation of the local immune system and vasculature to pro- ally supportive after confirmation of the diagnosis, and relief accompa-
mote a wound healing-like milieu.[131] nies the emergence of the next cycle of anagen hairs.
|
330       WANG and HIGGINS

The actual mechanism of hair cycle disruption in human HFs is new treatments, but actual mechanistic studies in humans are lack-
currently unknown. Stressful situations, including pregnancy, may be ing. Several new technologies and advances are now available for
associated with high levels of serum mediators such as cortisol and researchers to probe the human hair cycle.
prolactin, which have a suppressive effect on immune cells or HFSCs Single-cell transcriptomics to identify immune cell subsets in
themselves[137] (Figure 3C), leading to the premature termination of human skin will allow identification of rare populations from limited
anagen. Increased mast cells are found in their degranulated state near tissue (ie. 4mm punch biopsies). These populations may be easily
catagen HFs in TE,[138] and substance P produced by mast cells may overlooked in immunohistochemical or immunofluorescence stud-
play a role in the induction of stress-associated keratinocyte apopto- ies or bulk RNA sequencing. Sampling hair-bearing skin from other
sis.[139] This apoptosis in humans may be mediated by TNF-α, which body sites using this technology will allow us to uncover the dis-
are upregulated in mast cells upon exposure to substance P. The roles tinct immunophenotype associated with different hair cycle stages,
of extra-medullary PRL, mast cells, substance P and TNF signalling in potentially identifying new therapeutic targets to combat hair loss.
stress-associated hair cycling defects such as TE and AA have been Discovering how the immune macroenvironment can epigenetically
proposed,[140] but their precise mechanism is yet to be elucidated. regulate transitions through the follicle cycle will open up exciting
Metabolic stress may be another mechanism, as manipulating new avenues for research.
glycolytic and oxidative substrates in HFSCs seem to impact the Similar to single-gene hair disorders described in the last cen-
hair cycle in mice.[141,142] Unfortunately, the diagnosis of acute TE is tury, the new era of biological and immunotherapy in dermatology
normally made several months after the precipitating event, and the and other diseases will provide valuable insight into the biologi-
molecular and cellular fluxes at this time are frequently missed. A cal pathways relevant in human hair cycling. Clinical vigilance for
small subset of patients progress to chronic TE, whereby hair shed- side effects affecting the hair cycle in patients receiving these
ding persists beyond 6 months and represents a more lasting shift treatments will give us clues to signalling pathways underlying
in the hair cycle which can last for years.[143,144] Studying the local the human hair cycle, and the cells involved can be confirmed by
immune and metabolic changes in this group of patients may provide cross-referencing with single-cell transcriptomic or proteomic
insight into the mechanism of hair cycle control in humans. data. Just like prostaglandin analogues were serendipitously
found to induce eyelash trichomegaly in glaucoma patients,[145]
recent reports suggest that EGFR inhibitors also cause eyelash
6 |  CO N C LU S I O N A N D FU T U R E trichomegaly and Hh blockade with vismodegib results in mous-
D I R EC TI O N S tache trichomegaly.[146] Tocilizumab, an IL-6 monoclonal antibody,
is also postulated to affect the human hair cycle.[147] A systematic,
Current strategies to therapeutically manipulate the hair cycle are standardized way of tracking and analysing the human hair cycle is
focused on the activation of HFSC or DP cells and their precursors. required to monitor these effects.
Engineered skin constructs and organoids currently also only in- While we have gleaned much useful information in mammalian
clude these epithelial and mesenchymal components. So far, none hair cycling from our murine counterparts, it is time to start system-
of these models have been shown to be able to cycle spontaneously. atically and thoroughly studying the mechanisms that underpin the
Expanding our knowledge into the supportive cells of the HF niche, human hair cycle. Only with this concerted effort will novel treat-
in particular the immune cells, is essential for the recreation of a ments and platforms be developed for all forms of hair loss in our
functional HF ex vivo. patients.
The role of the auxiliary cells in HF cycling has gradually been
uncovered in the past decade. Not only do immune cells play an im- C O N FL I C T O F I N T E R E S T
portant role in HF homeostasis, we have described in this review The authors have declared no conflicting interests.
how perturbations in follicle cycling can be initiated by alterations
in the immune environment. Macrophage subsets appear to have AU T H O R C O N T R I B U T I O N
pleiotropic and context-dependent roles at various stages of the E.C.E Wang conceptualized, outlined and wrote the manuscript and
spontaneous and induced hair cycle in mice, as do T reg cells and made the figures. Both E.C.E Wang and C.A. Higgins contributed to
mast cells. It is important to understand the function of these cells the literature search and formulated the arguments and edited the
in human hair cycling in order to recapitulate the HF niche in future manuscript.
tissue engineering efforts.
Currently, most of our understanding of the immune infiltrate ORCID
during the human hair cycle is descriptive and confined to mostly Etienne C. E. Wang  https://orcid.org/0000-0002-9139-1865
anagen HFs,[12] which is the predominant phase found in human Claire A. Higgins  https://orcid.org/0000-0002-9742-5149
scalps. Ex vivo analysis of human HFs is limited to anagen and cat-
agen follicles,[98] and telogen hairs are difficult to dissect and main- REFERENCES
tain in culture. Functional studies done in mice and small mammals [1] K. S. Stenn, R. Paus, Physiol. Rev. 2001, 81(1), 449.
have been extrapolated to human HF biology in order to develop [2] P. Rompolas, V. Greco, Semin. Cell Dev. Biol. 2014, 25–26, 34.
WANG and HIGGINS |
      331

[3] V. Jaks, M. Kasper, R. Toftgard, Exp. Cell Res. 2010, 316(8), 1422. [35] K. Foitzik, T. Spexard, M. Nakamura, U. Halsner, R. Paus, J. Invest.
[4] Y. C. Hsu, L. Li, E. Fuchs, Nat. Med. 2014, 20, 847. Dermatol. 2005, 124(6), 1119.
[5] C. A. Jahoda, A. J. Reynolds, J. Invest. Dermatol. 1993, 101(1 Suppl), [36] T. Ito, N. Ito, M. Saathoff, A. Bettermann, M. Takigawa, R. Paus, Br.
33S. J. Dermatol. 2005, 152, 623.
[6] T Matsuzaki, K Yoshizato, Wound Repair Regen. 1998, 6, 524. [37] S. Suzuki, T. Kato, H. Takimoto, S. Masui, H. Oshima, K. Ozawa, S.
[7] H. Fujiwara, M. Ferreira, G. Donati, D. K. Marciano, J. M. Linton, Suzuki, T. Imamura, J. Invest. Dermatol. 1998, 111, 963.
Y. Sato, A. Hartner, K. Sekiguchi, L. F. Reichardt, F. M. Watt, Cell [38] C. A. Higgins, L. Petukhova, S. Harel, Y. Y. Ho, E. Drill, L. Shapiro,
2011, 144, 577. M. Wajid, A. M. Christiano, Proc. Natl. Acad. Sci. U. S. A. 2014,
[8] E. Festa, J. Fretz, R. Berry, B. Schmidt, M. Rodeheffer, M. Horowitz, 111(29), 10648.
V. Horsley, Cell 2011, 146, 761. [39] J. M. Hébert, T. Rosenquist, J. Götz, G. R. Martin, Cell 1994, 78(6),
[9] K. N. Li, P. Jain, C. H. He, F. C. Eun, S. Kang, T. Tumbar, Elife 2019, 1017.
8, e45977. [40] D. J. Housley, P. J. Venta, Anim. Genet. 2006, 37, 309.
[10] S.-Y. Yoon, L. C. Dieterich, S. Karaman, S. T. Proulx, S. B. Bachmann, [41] C. Dierks, S. Mömke, U. Philipp, O. Distl, Anim. Genet. 2013, 44,
C. Sciaroni, M. Detmar, PLoS One 2019, 14(7), e0220341. 425.
[11] R. Paus, T. Christoph, S. Muller-Rover, J. Investig. Dermatol. Symp. [42] G. Li, S. Zhou, C. Li, B. Cai, H. Yu, B. Ma, Y. Huang, Y. Ding, Y. Liu,
Proc. 1999, 4, 226. Q. Ding, C. He, J. Zhou, Y. Wang, G. Zhou, Y. Li, Y. Yan, J. Hua, B.
[12] T. Christoph, S. Müller-Röver, H. Audring, D. J. Tobin, B. Hermes, G. Petersen, Y. Jiang, T. Sonstegard, X. Huang, Y. Chen, X. Wang, FEBS
Cotsarelis, R. Rückert, R. Paus, Br. J. Dermatol. 2000, 142, 862. J. 2019, 286, 4675.
[13] V. H. Perry, S. Gordon, Trends Neurosci. 1988, 11, 273. [43] M. Maurer, E. Fischer, B. Handjiski, E. von Stebut, B. Algermissen,
[14] M. Hulsmans, S. Clauss, L. Xiao, A. D. Aguirre, K. R. King, A. Hanley, A. Bavandi, R. Paus, Lab. Invest. 1997, 77, 319.
W. J. Hucker, E. M. Wülfers, G. Seemann, G. Courties, Y. Iwamoto, [44] P. C. Arck, B. Handjiski, A. Kuhlmei, E. M. J. Peters, M. Knackstedt,
Y. Sun, A. J. Savol, H. B. Sager, K. J. Lavine, G. A. Fishbein, D. E. A. Peter, S. P. Hunt, B. F. Klapp, R. Paus, J. Mol. Med. 2005, 83,
Capen, N. Da Silva, L. Miquerol, H. Wakimoto, C. E. Seidman, J. G. 386.
Seidman, R. I. Sadreyev, K. Naxerova, R. N. Mitchell, D. Brown, P. [45] T. Kumamoto, D. Shalhevet, H. Matsue, M. E. Mummert, B. R.
Libby, R. Weissleder, F. K. Swirski, P. Kohl, C. Vinegoni, D. J. Milan, Ward, J. V. Jester, A. Takashima, Blood 2003, 102(5), 1654.
P. T. Ellinor, M. Nahrendorf, Cell 2017, 169, 510. [46] D. E. A. Komi, K. Khomtchouk, P. L. Santa Maria, Review of the
[15] A. Hirao, F. Arai, T. Suda, Cell Cycle 2004, 3(12), 1481. Contribution of Mast Cells in Wound Healing: Involved Molecular
[16] I. G. Winkler, N. A. Sims, A. R. Pettit, V. Barbier, B. Nowlan, F. and Cellular Mechanisms. Clinic Rev Allerg Immunol. 2019.https​://
Helwani, I. J. Poulton, N. van Rooijen, K. A. Alexander, L. J. Raggatt, doiorg.libpr​oxy1.nus.edu.sg/10.1007/s12016-019-08729-w
J.-P. Lévesque, Blood 2010, 116, 4815. [47] A. Weber, J. Knop, M. Maurer, Br. J. Dermatol. 2003, 148, 224.
[17] M. Edinger, P. Hoffmann, J. Ermann, K. Drago, C. G. Fathman, S. [48] E. M. J. Peters, S. Liotiri, E. Bodó, E. Hagen, T. Bíró, P. C. Arck, R.
Strober, R. S. Negrin, Nat. Med. 2003, 9(9), 1144. Paus, Am. J. Pathol. 2007, 171(6), 1872.
[18] S. J. Morrison, D. T. Scadden, Nature 2014, 505, 327. [49] P. F. Parakkal, J. Ultrastruct. Res. 1969, 29, 210.
[19] F. Merly, L. Lescaudron, T. Rouaud, F. Crossin, M. F. Gardahaut, [50] K. R. Mesa, P. Rompolas, G. Zito, P. Myung, T. Y. Sun, S. Brown,
Muscle Nerve 1999, 22, 724. D. G. Gonzalez, K. B. Blagoev, A. M. Haberman, V. Greco, Nature
[20] E. K. Parkinson, Semin. Cell Biol. 1992, 3, 435. 2015, 522, 94.
[21] L. L. Sharp, J. M. Jameson, G. Cauvi, W. L. Havran, Nat. Immunol. [51] M. G. Horenstein, J. S. Jacob, J. Cutan. Pathol. 2008, 35(12), 1115.
2005, 6(1), 73. [52] S. McCoy, A. Evans, C. James, Lasers Surg. Med. 1999, 24(2), 142.
[22] J. M. Jameson, Front. Biosci. 2004, 9, 2640. [53] W. Chi, E. Wu, B. A. Morgan, Development 2013, 140(8), 1676.
[23] R. Paus, C. van der Veen, S. Eichmüller, T. Kopp, E. Hagen, S. [54] Y. C. Hsu, H. A. Pasolli, E. Fuchs, Cell 2011, 144(1), 92.
Müller-Röver, U. Hofmann, J. Invest. Dermatol. 1998, 111(1), 7. [55] E. C. E. Wang, Z. Dai, A. W. Ferrante, C. G. Drake, A. M. Christiano,
[24] H. Hashizume, Y. Tokura, M. Takigawa, J. Dermatol. Sci. 1994, 8(2), Cell Stem Cell 2019, 24, 654.
119. [56] D. Castellana, R. Paus, M. Perez-Moreno, PLoS Biol. 2014, 12(12),
[25] K. Nagao, T. Kobayashi, K. Moro, M. Ohyama, T. Adachi, D. Y. e1002002.
Kitashima, S. Ueha, K. Horiuchi, H. Tanizaki, K. Kabashima, A. [57] N. Ali, B. Zirak, R. S. Rodriguez, M. L. Pauli, H.-A. Truong, K. Lai, R.
Kubo, Y.-H. Cho, B. E. Clausen, K. Matsushima, M. Suematsu, G. Ahn, K. Corbin, M. M. Lowe, T. C. Scharschmidt, K. Taravati, M. R.
C. Furtado, S. A. Lira, J. M. Farber, M. C. Udey, M. Amagai, Nat. Tan, R. R. Ricardo-Gonzalez, A. Nosbaum, M. Bertolini, W. Liao, F.
Immunol. 2012, 13, 744. O. Nestle, R. Paus, G. Cotsarelis, A. K. Abbas, M. D. Rosenblum,
[26] R. Paus, U. Hofmann, S. Eichmuller, B. M. Czarnetzki, Br. J. Cell 2017, 169(6), 1119.
Dermatol. 1994, 130, 281. [58] C.-C. Chen, L. Wang, M. V. Plikus, T. X. Jiang, P. J. Murray, R.
[27] S. Eichmüller, C. van der Veen, I. Moll, B. Hermes, U. Hofmann, S. Ramos, C. F. Guerrero-Juarez, M. W. Hughes, O. K. Lee, S. Shi, R.
Müller-Röver, R. Paus, J. Histochem. Cytochem. 1998, 46, 361. B. Widelitz, A. D. Lander, C. M. Chuong, Cell 2015, 161, 277.
[28] F. Muneeb, J. A. Hardman, R. Paus, Exp. Dermatol. 2019, 28, 425. [59] M. Yu, S. Kissling, P. Freyschmidt-Paul, R. Hoffmann, J. Shapiro, K.
[29] M. V. Plikus, C. M. Chuong, J. Invest. Dermatol. 2008, 128(5), 1071. J. McElwee, Exp. Dermatol. 2008, 17(1), 12.
[30] S. Müller-Röver, K. Foitzik, R. Paus, B. Handjiski, C. van der Veen, S. [60] S. Harel, C. A. Higgins, J. E. Cerise, Z. Dai, J. C. Chen, R. Clynes, A.
Eichmüller, I. A. McKay, K. S. Stenn, J. Invest. Dermatol. 2001, 117(1), 3. M. Christiano, Sci. Adv. 2015, 1(9), e1500973.
[31] G. Lindner, V. A. Botchkarev, N. V. Botchkareva, G. Ling, C. van der [61] Y.-H. Li, K. Zhang, J.-X. Ye, X.-H. Lian, T. Yang, Clin. Exp. Dermatol.
Veen, R. Paus, Am. J. Pathol. 1997, 151(6), 1601. 2011, 36, 534.
[32] K. Elliott, T. J. Stephenson, A. G. Messenger, J. Invest. Dermatol. [62] K. A. Hwang, Y. L. Hwang, M. H. Lee, N. R. Kim, S. S. Roh, Y. Lee, C.
1999, 113, 873. D. Kim, J. H. Lee, K. C. Choi, Int. J. Mol. Med. 2012, 29(2), 195.
[33] M. V. Plikus, J. A. Mayer, D. de la Cruz, R. E. Baker, P. K. Maini, R. [63] A. E. Oro, K. Higgins, Dev. Biol. 2003, 255, 238.
Maxson, C.-M. Chuong, Nature 2008, 451, 340. [64] V. Greco, T. Chen, M. Rendl, M. Schober, H. A. Pasolli, N. Stokes, J.
[34] K. Foitzik, G. Lindner, S. Mueller-Roever, M. Maurer, N. dela Cruz-Racelis, E. Fuchs, Cell Stem Cell 2009, 4(2), 155.
Botchkareva, V. Botchkarev, B. Handjiski, M. Metz, T. Hibino, T. [65] R. Ruckert, U. Hofmann, C. van der Veen, S. Bulfone-Paus, R. Paus,
Soma, G. P. Dotto, R. Paus, FASEB J. 2000, 14, 752. J. Invest. Dermatol. 1998, 111(1), 25.
|
332       WANG and HIGGINS

[66] U. Hofmann, Y. Tokura, R. Rückert, R. Paus, Cell. Immunol. 1998, [97] N. J. Hawkshaw, J. A. Hardman, M. Alam, F. Jimenez, R. Paus, Br. J.
184(1), 65. Dermatol. 2019. https​://doi.org/10.1111/bjd.18356​
[67] H. B. Chase, W. Montagna, J. D. Malone, Anat. Rec. 1953, 116(1), [98] J. A. Hardman, F. Muneeb, J. Pople, R. Bhogal, A. Shahmalak, R.
75. Paus, J. Invest. Dermatol. 2019, 139, 2543.
[68] C.-C. Yang, H.-M. Sheu, P.-L. Chung, C.-H. Chang, Y.-S. Tsai, M. W. [99] J. W. Oh, J. Kloepper, E. A. Langan, Y. Kim, J. Yeo, M. J. Kim, T.-C.
Hughes, T.-L. Tuan, L. L. H. Huang, Exp. Dermatol. 2015, 24(1), 57. Hsi, C. Rose, G. S. Yoon, S.-J. Lee, J. Seykora, J. C. Kim, Y. K. Sung,
[69] C. H. Won, H. G. Yoo, K. Y. Park, S. H. Shin, W. S. Park, P. J. Park, J. M. Kim, R. Paus, M. V. Plikus, J. Invest. Dermatol. 2016, 136(1),
H. Chung, O. S. Kwon, K. H. Kim, J. Invest. Dermatol. 2012, 132(12), 34.
2849. [100] A. G. Messenger, D. N. Slater, S. S. Bleehen, Br. J. Dermatol. 1986,
[70] B.-S. Park, W.-S. Kim, J.-S. Choi, H.-K. Kim, J.-H. Won, F. Ohkubo, 114, 337.
H. Fukuoka, Biomed. Res. 2010, 31(1), 27. [101] R. Paus, A. Slominski, B. M. Czarnetzki, Yale J. Biol. Med. 1993, 66,
[71] K. J. McElwee, A. Huth, S. Kissling, R. Hoffmann, J. Invest. Dermatol. 541.
2004, 123(1), 34. [102] E. H. C Wang, M. Yu, T. Breitkopf, N. Akhoundsadegh, X. Wang,
[72] M. P. Philpott, D. A. Sanders, T. Kealey, J. Invest. Dermatol. 1994, F. T. Shi, G. Leung, J. P. Dutz, J. Shapiro, K. J. McElwee, J. Invest.
102, 857. Dermatol. 2016, 136(8), 1617.
[73] Y. Milner, M. Kashgarian, J. Sudnik, M. Filippi, M. Kizoulis, K. Stenn, [103] R. Paus, S. Bulfone-Paus, M. Bertolini, J. Investig. Dermatol. Symp.
J, Invest. Dermatol. 2002, 119, 639. Proc. 2018, 19(1), S12.
[74] A. Rebora, M. Guarrera, Dermatology 2002, 205(2), 108. [104] L. Petukhova, M. Duvic, M. Hordinsky, D. Norris, V. Price, Y.
[75] C. A. Higgins, G. E. Westgate, C. A. Jahoda, J. Invest. Dermatol. Shimomura, H. Kim, P. Singh, A. Lee, W. V. Chen, K. C. Meyer, R.
2009, 129, 2100. Paus, C. A. B. Jahoda, C. I. Amos, P. K. Gregersen, A. M. Christiano,
[76] A. P. Møller, J. T. Nielsen, J. Erritzøe, Behav. Ecol. 2006, 17(6), 1046. Nature 2010, 466(7302), 113.
[77] M. Guarrera, A. Rebora, Skin Appendage Disord. 2019, 5(3), 152. [105] T. Ito, N. Ito, M. Saatoff, H. Hashizume, H. Fukamizu, B. J. Nickoloff,
[78] J. W. Young, E. T. Conte, M. L. Leavitt, M. A. Nafz, A. L. Schroeter, M. Takigawa, R. Paus, J. Invest. Dermatol. 2008, 128(5), 1196.
J. Am. Osteopath. Assoc. 1991, 91, 765. [106] S. J. Peckham, S. B. Sloan, D. M. Elston, J. Am. Acad. Dermatol.
[79] C. M. Magro, A. Rossi, J. Poe, S. Manhas-Bhutani, N. Sadick, J. 2011, 65, 615.
Drugs Dermatol. 2011, 10(12), 1404. [107] L. Xing, Z. Dai, A. Jabbari, J. E. Cerise, C. A. Higgins, W. Gong,
[80] E. Johnson, F. J. Ebling, J. Embryol. Exp. Morphol. 1964, 12, 465. A. de Jong, S. Harel, G. M. DeStefano, L. Rothman, P. Singh, L.
[81] W. Qin, U. Baran, R. Wang, Lasers Surg. Med. 2015, 47, 669. Petukhova, J. Mackay-Wiggan, A. M. Christiano, R. Clynes, Nat.
[82] J. E. Kloepper, K. Kawai, M. Bertolini, T. Kanekura, R. Paus, J. Med. 2014, 20(9), 1043.
Invest. Dermatol. 2013, 133(6), 1666. [108] A. Gilhar, A. Keren, R. Paus, J. Investig. Dermatol. Symp. Proc. 2013,
[83] S. Sano, M. Kira, S. Takagi, K. Yoshikawa, J. Takeda, S. Itami, Proc. 16(1), S37.
Natl. Acad. Sci. U. S. A. 2000, 97(25), 13824. [109] A. Ghraieb, A. Keren, A. Ginzburg, Y. Ullmann, A. G. Schrum, R.
[84] N. Amberg, M. Holcmann, G. Stulnig, E. Glitzner, M. Sibilia, J. Paus, A. Gilhar, J. Autoimmun. 2018, 91, 61.
Invest. Dermatol. 2017, 137, 528. [110] J. J. Speiser, D. Mondo, V. Mehta, S. A. Marcial, A. Kini, K. A.
[85] N. Amberg, M. Holcmann, G. Stulnig, M. Sibilia, J. Invest. Dermatol. Hutchens, J. Cutan. Pathol. 2019, 46, 653.
2016, 136(11), 2140. [111] J. Goldstein, S. Fletcher, E. Roth, C. Wu, A. Chun, V. Horsley, Genes
[86] R. Paus, M. Maurer, A. Slominski, B. M. Czarnetzki, Dev. Biol. 1994, Dev. 2014, 28, 983.
163(1), 230. [112] E. C. E. Wang, Z. Dai, A. M. Christiano, J. Allergy Clin. Immunol.
[87] V. A. Botchkarev, R. Paus, B. M. Czarnetzki, V. S. Kupriyanov, D. S. 2018, 141, 499.
Gordon, O. Johansson, Arch. Dermatol. Res. 1995, 287, 683. [113] M. Bertolini, F. Zilio, A. Rossi, P. Kleditzsch, V. E. Emelianov, A.
[88] J. M. Bae, H. M. Jung, B. Goo, Y. M. Park, Lasers Surg. Med. 2015, Gilhar, A. Keren, K. C. Meyer, E. Wang, W. Funk, K. McElwee, R.
47, 433. Paus, PLoS One 2014, 9(5), e94260.
[89] K. H. Jeong, H. J. Joo, J. E. Kim, Y. M. Park, H. Kang, Clin. Exp. [114] P. H. Tang, H. P. Chia, L. L. Cheong, D. Koh, Singapore Med. J. 2000,
Dermatol. 2015, 40, 894. 41, 202.
[90] S.-Y. Chu, C.-H. Chou, H.-D. Huang, M.-H. Yen, H.-C. Hong, P.-H. [115] J.-H. Paik, J.-B. Yoon, W.-Y. Sim, B.-S. Kim, N.-I. Kim, Br. J. Dermatol.
Chao, Y.-H. Wang, P.-Y. Chen, S.-X. Nian, Y.-R. Chen, L.-Y. Liou, Y.- 2001, 145(1), 95.
C. Liu, H.-M. Chen, F.-M. Lin, Y.-T. Chang, C.-C. Chen, O. K. Lee, [116] D. Pathomvanich, S. Pongratananukul, P. Thienthaworn, S.

Nat. Commun. 2019, 10(1), 1524. Manoshai, Dermatol. Surg. 2002, 28, 804.
[91] M. Ito, Z. Yang, T. Andl, C. Cui, N. Kim, S. E. Millar, G. Cotsarelis, [117] N. Pantelireis, C. A. Higgins, Exp. Dermatol. 2018, 27, 959.
Nature 2007, 447, 316. [118] R. Sinclair, Br. Med. J. 1998, 317, 865.
[92] X. Wang, T.-C. Hsi, C. F. Guerrero-Juarez, K. Pham, K. Cho, C. [119] E. Ludwig, Br. J. Dermatol. 1977, 97, 247.
D. McCusker, E. S. Monuki, K. W. Y. Cho, D. L. Gay, M. V. Plikus, [120] D. Perez-Meza, C. Ziering, M. Sforza, G. Krishnan, E. Ball, E.

Regeneration 2015, 2(4), 169. Daniels, Stem Cells Cloning 2017, 10, 1.
[93] X. Wang, H. Chen, R. Tian, Y. Zhang, M. S. Drutskaya, C. Wang, J. [121] S.-C. Chueh, S.-J. Lin, C.-C. Chen, M. Lei, L. M. Wang, R. Widelitz,
Ge, Z. Fan, D. Kong, X. Wang, T. Cai, Y. Zhou, J. Wang, J. Wang, M. W. Hughes, T.-X. Jiang, C. M. Chuong, Expert Opin. Biol. Ther.
S. Wang, Z. Qin, H. Jia, Y. Wu, J. Liu, S. A. Nedospasov, E. E. 2013, 13, 377.
Tredget, M. Lin, J. Liu, Y. Jiang, Y. Wu, Nat. Commun. 2017, 8, [122] P. Gentile, S. Garcovich, A. Bielli, M. G. Scioli, A. Orlandi, V. Cervelli,
14091. Stem Cells Transl. Med. 2015, 4(11), 1317.
[94] A. Kasuya, T. Ito, Y. Tokura, J. Dermatol. Sci. 2018, 91, 250. [123] A. K. Gupta, J. Cole, D. P. Deutsch, P. A. Everts, R. P. Niedbalski,
[95] D. Gay, O. Kwon, Z. Zhang, M. Spata, M. V. Plikus, P. D. Holler, R. Panchaprateep, F. Rinaldi, P. T. Rose, R. Sinclair, J. E. Vogel, R. J.
M. Ito, Z. Yang, E. Treffeisen, C. D. Kim, A. Nace, X. Zhang, S. Welter, M. D. Zufelt, C. J. Puig, Dermatol. Surg. 2019, 45(10), 1262.
Baratono, F. Wang, D. M. Ornitz, S. E. Millar, G. Cotsarelis, Nat. [124] Z. J. Li, H.-I. Choi, D.-K. Choi, K.-C. Sohn, M. Im, Y.-J. Seo, Y.-H. Lee,
Med. 2013, 19, 916. J.-H. Lee, Y. Lee, Dermatol. Surg. 2012, 38(7 Pt 1), 1040.
[96] M. Saitoh, M. Uzuka, M. Sakamoto, J. Invest. Dermatol. 1970, 54(1), [125] B. L. Eppley, J. E. Woodell, J. Higgins, Plast. Reconstr. Surg. 2004,
65. 114(6), 1502.
WANG and HIGGINS |
      333

[126] R. Dhurat, M. S. Sukesh, G. Avhad, A. Dandale, A. Pal, P. Pund, Int. [141] A. Flores, J. Schell, A. S. Krall, D. Jelinek, M. Miranda, M. Grigorian,
J. Trichol. 2013, 5(1), 6. D. Braas, A. C. White, J. L. Zhou, N. A. Graham, T. Graeber, P. Seth,
[127] Y. Huang, F. Zhuo, L. Li, Lasers Med. Sci. 2017, 32(8), 1711. D. Evseenko, H. A. Coller, J. Rutter, H. R. Christofk, W. E. Lowry,
[128] A. C. J. Bertin, A. Vilarinho, A. L. A. Junqueira, J. Cosmet. Laser Ther. Nat. Cell Biol. 2017, 19(9), 1017.
2018, 20, 391. [142] M. Miranda, H. Christofk, D. L. Jones, W. E. Lowry, J. Invest.

[129] R. J. Dabek, W. G. Austen Jr, B. Bojovic, Plast. Reconstr. Surg. Glob. Dermatol. 2018, 138, 968.
Open 2019, 7(4), e2157. [143] D. A. Whiting, Dermatol. Clin. 1996, 14, 723.
[130] K.-H. Liu, D. Liu, Y.-T. Chen, S.-Y. Chin, Lasers Med. Sci. 2019, 34(6), [144] M. J. Garcia-Hernandez, F. M. Camacho, Arch. Dermatol. 1999,
1063. 135(9), 1123.
[131] D. Hawkins, N. Houreld, H. Abrahamse, Ann. N. Y. Acad. Sci. 2005, [145] T. Yoshino, T. Fukuchi, T. Togano, M. Seki, H. Ikegaki, H. Abe, Jpn. J.
1056, 486. Ophthalmol. 2013, 57(2), 172.
[132] A. Rebora, Int. J. Dermatol. 1993, 32, 339. [146] J. Riera-Monroig, A. Combalia, A. Rodríguez, L. Alós, C. Mangas, J.
[133] A. Rebora, M. Guarrera, F. Drago, J. Eur. Acad. Dermatol. Venereol. Ferrando, C. Carrera, J. Eur. Acad. Dermatol. Venereol. 2020, 34(1),
2016, 30, 518. e11-e13.
[134] C. Pierard-Franchimont, G. E. Pierard, Dermatology 2001, 203(2), [147] C. Vidon, M. Bossert, A. Lohse-Walliser, M. Godfrin-Valnet, J.-C.
115. Balblanc, D. Wendling, Joint Bone Spine 2014, 81(1), 100.
[135] S. Harrison, R. Sinclair, Clin. Exp. Dermatol. 2002, 27, 389.
[136] S. Harrison, W. Bergfeld, Cleve. Clin. J. Med. 2009, 76, 361.
[137] P. C. Arck, A. Slominski, T. C. Theoharides, E. M. J. Peters, R. Paus,
J. Invest. Dermatol. 2006, 126(8), 1697.
How to cite this article: Wang ECE, Higgins CA. Immune cell
[138] S. A. Grace, A. M. Sutton, N. Abraham, E. S. Armbrecht, C. I. Vidal,
Int. J. Trichology 2017, 9(1), 25.
regulation of the hair cycle. Exp Dermatol. 2020;29:322–333.
[139] P. C. Arck, B. Handjiski, E. Hagen, R. Joachim, B. F. Klapp, R. Paus, https​://doi.org/10.1111/exd.14070​
FASEB J. 2001, 15(13), 2536.
[140] E. A. Langan, S. Vidali, N. Pigat, W. Funk, E. Lisztes, T. Bíró, V.
Goffin, C. E. M. Griffiths, R. Paus, PLoS One 2013, 8(4), e60819.

You might also like