Next Article in Journal
Genetic Parameter and Hyper-Parameter Estimation Underlie Nitrogen Use Efficiency in Bread Wheat
Previous Article in Journal
Transcriptional Profiling and Transposon Mutagenesis Study of the Endophyte Pantoea eucalypti FBS135 Adapting to Nitrogen Starvation
Previous Article in Special Issue
Tenascin-C in Tissue Repair after Myocardial Infarction in Humans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Matricellular Proteins in the Homeostasis, Regeneration, and Aging of Skin

by
Erna Raja
1,
Maria Thea Rane Dela Cruz Clarin
1,2 and
Hiromi Yanagisawa
1,*
1
Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan
2
Ph.D. Program in Humanics, School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba 305-8577, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(18), 14274; https://doi.org/10.3390/ijms241814274
Submission received: 31 August 2023 / Revised: 13 September 2023 / Accepted: 15 September 2023 / Published: 19 September 2023
(This article belongs to the Special Issue Matricellular Proteins in Human Diseases)

Abstract

:
Matricellular proteins are secreted extracellular proteins that bear no primary structural functions but play crucial roles in tissue remodeling during development, homeostasis, and aging. Despite their low expression after birth, matricellular proteins within skin compartments support the structural function of many extracellular matrix proteins, such as collagens. In this review, we summarize the function of matricellular proteins in skin stem cell niches that influence stem cells’ fate and self-renewal ability. In the epidermal stem cell niche, fibulin 7 promotes epidermal stem cells’ heterogeneity and fitness into old age, and the transforming growth factor-β—induced protein ig-h3 (TGFBI)—enhances epidermal stem cell growth and wound healing. In the hair follicle stem cell niche, matricellular proteins such as periostin, tenascin C, SPARC, fibulin 1, CCN2, and R-Spondin 2 and 3 modulate stem cell activity during the hair cycle and may stabilize arrector pili muscle attachment to the hair follicle during piloerections (goosebumps). In skin wound healing, matricellular proteins are upregulated, and their functions have been examined in various gain-and-loss-of-function studies. However, much remains unknown concerning whether these proteins modulate skin stem cell behavior, plasticity, or cell–cell communications during wound healing and aging, leaving a new avenue for future studies.

1. Introduction

Matricellular proteins are nonstructural, modular, extracellular proteins that exert their effects by binding to cell surface receptors, extracellular matrix (ECM) proteins, soluble signaling molecules, and proteases, thereby modulating cellular responses to changes in their microenvironment, particularly during tissue remodeling [1,2]. The term matricellular protein was first introduced by Paul Bornstein in the 1990s to describe thrombospondin 1 (TSP1/THBS1), secreted protein acidic rich in cysteine (SPARC), and tenascin C [1]. Since then, the list has substantially expanded and includes protein families of the cellular communication network (CCN) factors, the fasciclin family, tenascins, galectins, R-Spondins (RSPOs), fibulins, ecto-nucleotide pyrophosphatase/phosphodiesterases, small integrin-binding ligand N-linked glycoproteins, and olfactomedins [3]. Although matricellular proteins have no direct structural functions, they can bind to structural ECM proteins such as collagens [4,5]. However, some structural ECM proteins have nonstructural functions, such as fibrillin-1 [6] and fibronectin [7], which bind and regulate the bioavailability of growth factors in the tissue microenvironment. Generally, matricellular proteins are highly expressed during development, which decreases to low levels during homeostasis but is upregulated upon tissue injury and disease [4]. While some knockout animal models of matricellular proteins with developmental roles have exhibited embryonic or neonatal lethality [8,9,10,11], others have resulted in no apparent post-natal phenotypic changes, and their biological functions were later discovered during tissue damage or in other pathological contexts [12,13,14,15]. In this review, we summarize reports on matricellular proteins in adult skin homeostasis, regeneration, and aging. We focus on the interaction between matricellular proteins and tissue stem cells and how they modify the tissue stem cell environment.

2. Skin Architecture, ECM, and Cellular Components

The skin is the largest organ of the body and protects us against environmental insults. It shields the body from mechanical abrasion, pathological infections, dehydration, and fluctuations in body temperature, while the nerves in the skin also provide us with sensations of touch [16]. The skin needs to act as a resilient mechanical barrier, yet provide structural flexibility. The functional unit of skin consists of the stratified epidermis and dermis (including dermal adipose and skin appendages such as hair follicles, sweat, and sebaceous glands) as well as the panniculus carnosus (PC) muscle and the subcutaneous fascia (Figure 1). Notably, the human skin has a thicker epidermis and dermis compared with mouse skin, and the epidermis exhibits undulations forming the rete ridge and inter-ridge (also known as dermal papillae) structures that are absent in mouse skin (Figure 1).

2.1. Epidermis

The construction of the epidermis begins with the basal epidermal stem cells that undergo a continuous and balanced process of symmetric self-renewal and asymmetric division to produce progenitors and differentiated cells. These provide a constant supply of keratinocytes committed to terminal differentiation which form the suprabasal layers and the cornified skin barrier [61]. Through hemidesmosomes and integrin receptors, the basal epidermal stem cells are anchored to the basement membrane (BM), which connects the epidermis to the underlying dermis. The BM’s primary constituents are structural scaffolding matrix proteins such as collagens IV, VII, XVII, laminin 332, and laminin 511 [62]. Fibronectin is also present in the lamina lucida of the BM (the laminin-rich area) [63]. Matricellular proteins, such as fibulin 2 [17,64], fibulin 7 [18], SPARC [19,20], hemicentin 1 (fibulin 6) [22,23], THBS1 [24], and thrombospondin 5 (THBS5 or COMP) [65], are contained in the epidermal BM via interactions with structural proteins or the integrin receptors.
Functionally, fibulin 2 binds to laminin 332 to stabilize the epidermal BM in neonatal skin. The absence of fibulin 2 results in separation of the dermal–epidermal junction (DEJ) and skin blisters [17]. Fibulin 7 is also expressed in the epidermal BM and has been shown to bind to collagen IV in vitro [18], although its role in the BM arrangement remains unknown. SPARC was similarly shown to bind to collagen IV in vitro and induce collagen IV and VII expression in the epidermal BM in reconstructed human three-dimensional skin culture models [19,20,21]. Hemicentin 1 co-localizes with laminin α2 at the epidermal BM [22]. Hmcn1 null mice exhibit unevenly widened lamina lucida and lamina densa and compromised hemidesmosomes [22], which suggests its function in BM organization. Hemicentin 1 was further shown to compete with laminins for its binding site to the nidogen 2 proteoglycan during BM formation and maintenance [23]. In contrast to other matricellular proteins in the BM, THBS1 is believed to act as an endogenous angiogenesis inhibitor, forming a barrier between the nonvascular epidermis and the vascularized dermis [25,26]. Matricellular proteins at the BM/DEJ, such as fibulin 2 and SPARC, are produced by both keratinocytes and fibroblasts [17,66,67,68,69], with hemicentin 1 and COMP mainly being secreted by fibroblasts [22,70]. In contrast, a subset of basal keratinocytes expresses Thbs1, albeit at low abundance during homeostasis [71]
In addition to keratinocytes, the epidermis is home to resident immune cells and melanocytes, which provide pigmentation to the skin and hair [16]. Melanocytes are regenerated by melanocyte stem cells, which are neural-crest-derived cells. Melanocytes produce melanin and have long dendrites that can make physical connections with up to 40 keratinocytes. Melanin is transferred to keratinocytes via caveolae-dependent internalization (lipid-raft-mediated endocytosis) to protect their nuclei from ultraviolet (UV) radiation [72]. Melanocyte survival also depends on its attachment to the epidermal BM via binding between its discoidin domain receptor 1 (DDR1) and collagen IV [27]. During stress or UV radiation, keratinocytes secrete paracrine factors such as IL-1β that, in turn, induce melanocytes to secrete the matricellular protein CCN3. CCN3 promotes DDR1 binding to collagen IV and inhibits melanocyte proliferation [27,28].

2.2. Dermis

Underneath the epidermal BM are the papillary and reticular dermal compartments, which are distinct in their cellularization and ECM components (Figure 1) [73]. The upper dermis/papillary dermis is densely populated with papillary fibroblasts that reside within thin and loose networks of fibrillar collagens (I and III) and elastic fibers. They produce ECMs such as collagen VI [74], fibronectin, and the matricellular proteins tenascin C and fibulin 2 [66]. These ECM proteins interact with each other to maintain structural integrity and provide biological cues for the surrounding cells. Fibronectin is a glycoprotein required for the formation of microfibrils. It acts as a scaffold for the elastic fibers, modulates the balance of skin rigidity and elasticity, and supports cell attachment and migration along the ECM matrix, such as collagen fibers [75,76]. Fibronectin also possesses growth-factor-binding ability [35,36]. Collagen VI fibrils interact with both fibronectin and collagen I [77] and may promote the tensile strength of the skin [78]. While fibulin 2 binds to fibrillin 1 and regulates the homeostasis of elastic fibers [37,38], further maintenance of the dermal ECM may come from the binding of tenascin C to fibronectin, collagen, and periostin [29]. Tenascin C has been reported to control cell proliferation by acting as a constitutive ligand to activate the cell surface epidermal growth factor receptor [30,31] and by sequestration of soluble growth factors such as Wnt3a, TGFβ, and VEGF [29,32,33,34]. Similarly, periostin is expressed in the papillary dermis, interacts with fibronectin, collagen I, and tenascin C [79] and is reported to modulate collagen structure and stability [43]. Periostin also impacts keratinocyte proliferation via fibroblast paracrine secretion of IL-6 [44].
The softer papillary dermis serves as a cushion connecting the stiffer epidermal BM to the lower reticular dermis, a major part of the dermal compartment. Reticular dermis contains more sparsely distributed reticular fibroblasts, thick and highly organized collagen bundles that contribute to skin tensile strength [62,73]. Reticular fibroblasts produce ECMs such as collagen I, fibrillin 1, fibronectin, emilin 1, and the matricellular proteins THBS1, tenascin C, and fibulin 2 [66]. Fibrillin 1, elastin, emilin 1, fibronectin, and matricellular proteins such as fibulin 2, 4, and 5 are expressed in the dermis and crucial for the formation of elastic fibers, which provide the skin with elastic properties [39,40,41,42,80]. Aside from regulating the balance of dermal vascularization, THBS1 is physically associated with the collagen I KGHR motif, which is important for cross-linking. The loss of this interaction results in abnormal collagen I and human dermal fibroblast differentiation into myofibroblasts [45]. Similarly, Thbs2 knockout mice have disarranged collagen fibril sizes and patterns in the dermis, which correlate with reduced skin tensile strength [46]. Abnormal collagen fibrils have also been observed in SPARC null mice, which exhibit decreases in the amount of dermal collagen I, the fibril diameter, and the tensile strength [47].
Fibroblasts in the embryonic/neonatal mouse skin are proliferative. However, in the mature skin, fibroblasts cease to divide and produce more ECM in the dermis. This ECM (collagen) enrichment in the adult skin acts as negative feedback to further inhibit fibroblast proliferation, although this quiescent state is reversible and fibroblasts are activated again upon skin injury [81]. Resident dermal immune cells are also activated to produce various cytokines that modulate fibroblast differentiation and proliferation [81,82]. Dermal fibroblasts originate from a common multipotent mesenchymal cell progenitor (expressing Pdgfrα, Dlk1, and Lrig1) that give rise to papillary and reticular fibroblast progenitors [82].

2.3. Dermal Adipose Tissue

Reticular fibroblast progenitors can give rise to adipocyte precursors and mature, lipid-filled adipocytes [82,83], which constitute the dermal fat layer. Dermal adipose functions as energy storage, thermal insulation, and mechanical support. Furthermore, it harbors innate immune antimicrobial functions [84,85]. Mature adipocytes express the collagen IV BM protein, which is deposited pericellularly and is believed to act as a strong scaffold to protect cells from mechanical stress in this loose connective tissue [86,87]. Collagen IV expression around these adipocytes is increased in obese compared with lean human subcutaneous adipose tissue [88], possibly to counterbalance the physical changes associated with an increase in adipocyte cell size. Other ECMs associated with skin adipose are collagen I, III, V [89], and VI [90]. Proteoglycans such as versican, biglycan, and decorin are also expressed in adipose tissue in order to bind to collagens, support their scaffolding function, and counteract compressive forces. However, in cases of obesity, these proteoglycans are present in excessive amounts, resulting in ECM defects and the promotion of tissue inflammation [91,92,93]. Adipogenesis is influenced by the matricellular protein SPARC, as its loss of expression leads to increased dermal adipose tissue [48] and accelerated wound healing [49]. Similarly, the secreted protein coiled-coil domain containing 80 (CCDC80/URB/DRO1) is highly expressed by adipocytes to modulate adipogenesis, and its genetic deletion in mice promotes body fat deposition, including subcutaneous fat [50,51,52].

2.4. The Hair Follicle

The hair follicle structure spans from the hypodermis (dermal adipose) up to the epidermis. Its size changes according to the hair cycle, which consists of the telogen (resting), anagen (growing), and catagen (regression) phases [94]. Hair follicles are connected to the sebaceous glands, arrector pili muscles, and dermal papilla at the base of the follicle (Figure 1 and Figure 2). During homeostasis, the hair follicle is mainly regenerated from hair follicle stem cells in the bulge region, with coordination from signals originating from mesenchymal cells in the dermal papilla, which control the exit from telogen and the duration of anagen [95,96,97,98,99,100] (Figure 2). The dermal papilla cells are, in turn, replenished by a subset of dermal sheath cells, which have self-renewal ability, also referred to as hair follicle dermal stem cells [96]. Hair follicle BM proteins are expressed by both epithelial cells and fibroblasts, with distinct contributions [101]. They consist of core structural BM proteins such as laminins, collagens IV, VI, VII, XVII, and XVIII, netrins 1 and 4, fibronectin, nephronectin, and matricellular proteins such as periostin, tenascin C, fibulin 1, SPARC, SMOC1, spondin-1, and R-spondin 3 [75,101,102]. The function of fibulin 1 is likely related to the regulation of BM homeostasis through its binding to laminin and collagen IV [53,103]. Nephronectin is produced by hair follicle stem cells to form an adhesion point for the arrector pili muscle cells expressing α8β1 integrins. Arrector pili muscles and the sympathetic nervous system cooperate with hair follicles to achieve piloerections to keep warm air closer to the skin in response to cold temperatures or emotions [54]. It has been proposed that hair follicle stem cells expressing periostin, tenascin C, and SPARC resemble tendon-related gene functions, i.e., they are expressed in the tendonous part of muscle tissue, which strengthens the bone–muscle connection and allows for skeletal movement [55,56,57]. Similarly, their deposition in the hair bulge matrix may stabilize the connection between arrector pili muscles and hair follicles during piloerections [54].
Figure 2. The hair follicle in anagen (growth phase, left) and telogen (resting phase, right) and the associated matricellular proteins in the hair follicle stem cell niche (bulge region, green box) and dermal papilla niche (yellow box). The functions of these matricellular proteins are summarized in Table 1 and Table 2. Adapted from BioRender.com (accessed on 2 August 2023).
Figure 2. The hair follicle in anagen (growth phase, left) and telogen (resting phase, right) and the associated matricellular proteins in the hair follicle stem cell niche (bulge region, green box) and dermal papilla niche (yellow box). The functions of these matricellular proteins are summarized in Table 1 and Table 2. Adapted from BioRender.com (accessed on 2 August 2023).
Ijms 24 14274 g002

2.5. The Sebaceous Gland

Sebaceous glands are located in the upper and permanent area of the hair follicle (junctional zone) in hair-associated skin. However, they may also be present in the skin independently of hair follicles such as in the meibomian glands of the eyelid [104,105]. The peripheral (basal) epidermal cells of these glands are proliferative and contain sebaceous gland stem cells. The differentiated sebocytes reside in the inner layer of the gland. Through cell death and lysis, lipids/sebum are released onto the skin surface to promote barrier functions, water repulsion, and antimicrobial- and antioxidant activities (vitamin E) [105,106,107]. To our knowledge, documentation regarding the ECM of the sebaceous gland is limited. A recent report indicated expression and interaction of fibronectin with the sebaceous gland basal cells to regulate its differentiation [58]. Another report discussed the ECM in the meibomian glands such as collagen IV, laminin α2, and β1, which may serve as BM scaffolding proteins, and the matricellular protein tenascin C, which may modulate growth factor bioavailability around sebaceous gland stem cells [59].
Table 2. Matricellular proteins for maintenance of hair follicles and sebaceous gland stem cells.
Table 2. Matricellular proteins for maintenance of hair follicles and sebaceous gland stem cells.
ProcessMatricellular
Protein
FunctionReferences & Study Model
Hair follicle stem cells homeostasisTenascin Cpromotes WNT signaling and maintains hair follicle stem cells[32] (mouse)
Periostinpro-cell proliferation in the hair follicles post-wounding[108] (mouse)
CCN2maintenance of hair follicle stem cell quiescence[109] (mouse)
R-spondin 2activates hair follicle stem cells, cell proliferation of dermal papilla and dermal sheath[110] (human hair follicles & mouse), [111] (human scalp & mouse skin, [96] (mouse)
R-spondin 3cell proliferation of dermal papilla and dermal sheath[96] (mouse), [111] (human scalp & mouse skin)
Mindin (Spon2)gene expression is decreased in aging hair follicle stem cells, role unknown in the hair follicle[112] (mouse)
Thrombospondin 1gene expression is increased in aging hair follicle stem cells, hair follicle and vasculature regression in catagen[112] (mouse skin), [113] (mouse)
Sebaceous gland stem cellsFibronectin *maintenance of basal sebocytes[58] (mouse)
* Fibronectin is not classified as matricellular proteins but they bear non-structural functions too via binding to matricellular proteins or regulating growth factors’ bio-availability.

2.6. The Panniculus Carnosus Muscle

The PC muscle is a layer of skeletal muscle located directly under the dermal adipose tissue in some mammals to facilitate skin movement independently of deeper muscle mass. The PC muscle has been suggested to mediate skin twitching during irritation, facial skin movement for social expressions, shivering thermogenesis, and skin contraction to promote wound closure [114]. In humans, PC muscle remnants exist only in certain anatomical regions, such as the craniofacial muscle, the platysma muscle (ventral region of the neck), and the palmaris brevis in the hand [114]. The PC muscle is reported to be highly regenerative [115] and vascularized [116]. The fibulin 4 matricellular protein may regulate PC muscle homeostasis, as the Fbln4 E57K homozygous mutation (the equivalent of the human mutation causing cutis laxa) in mice exhibits a thinner dermis and PC muscle layer, possibly due to elastic fiber defects and abnormal collagen fibrils [60]. However, it is unknown whether this mutation compromises PC muscle function.

2.7. Subcutaneous Fascia

The subcutaneous fascia ECM is enriched in an aqueous matrix containing glycosaminoglycans such as hyaluronan to facilitate smooth gliding between the skin and muscle. Fibroblasts and fasciacytes contribute to ECM production in the fascia [117]. Fibroblasts secrete fibrous matrix components such as fibrillin, elastin, fibronectin, and collagens I and III, whereas hyaluronan is produced by fasciacytes [118]. Fasciacytes are round, fibroblast-like cells expressing vimentin [119]. Little is known about the role of matricellular proteins in subcutaneous fascia maintenance, although the loss of fibulin 3 compromises elastic fibers in the visceral fascia [120,121]. Interestingly, lineage tracing experiments have demonstrated that, during wound healing, fascia fibroblasts are mainly responsible for generating scar tissue and not dermal fibroblasts [122,123].

3. Matricellular Proteins in Stem Cell Regulation during Homeostasis, Injury Repair, and Chronological Skin Aging

During physiological wear and tear or tissue injury, adult skin stem cells maintain tissue functions by replenishing damaged cells. Activated stem cells proliferate to self-renew and generate committed progenitor cells, forming terminally differentiated cells with specified functions. Stem cell fate regulation is influenced by changes in its microenvironment [112,124,125]. Through mechanotransduction pathways involving crucial players such as integrin receptors binding to the ECM, Piezo1 calcium channel, and YAP/TAZ signaling, both stem cells and stromal cells sense changes in the biochemical and mechanical properties of their microenvironment and respond accordingly [75,124,126,127,128]. The ECM/matricellular proteins are components of the microenvironment and changes in their quality and quantity with aging compromise skin functions.
In aged skin, the epidermis is thinner and less resistant to shear stress, and the rete ridge structures are compromised [129]. Moreover, hair follicles are miniaturized or depleted [130], while dermal fibroblasts lose their ability to produce collagen, exhibit altered identity [131], and become more adipogenic [132]. Immunopathological changes also occur in the dermis, and overall wound healing ability is impaired [129]. These declining skin features are partly attributed to impaired stem cell functions due to stem cell depletion or changes in their fate/behaviors. Furthermore, as skin ages, proliferating stem cells are prone to accumulate DNA damage due to intrinsic replication stress and exposure to UV irradiation over time. ECM stiffness may determine cells’ response of repairing double-strand DNA breaks, which can lead to genetic instability. Low ECM stiffness weakens the double-strand DNA break repair mechanism and renders cells more sensitive to genotoxic stress [133]. In this section, we will discuss the reported roles of matricellular proteins in maintaining stem cell homeostasis, changes during injury-induced regeneration, and aged skin.

3.1. Epidermal Stem Cells

Epidermal stem cells (more specifically interfollicular epidermal stem cells) that form stratified skin epithelium comprise heterogeneous populations in mouse [134] and human skin [135,136] (Figure 3). They differ in their proliferative capacity (hence the names: slow- and fast-cycling stem cell populations) and in their molecular characteristics [18,134,136,137]. In the mouse tail skin model, these two populations differentiate to form distinct territories, with fast-cycling stem cells replenishing the scale region, whereas the slow-cycling stem cells maintain the interscale region [134]. Interestingly, the gene expressions of the slow- and fast-cycling stem cells in the mouse tail skin also show territorial expression patterns that reflect the two basal epidermal stem cell regions of human skin, namely the inter-ridge (slow-cycling region) and rete ridge (fast-cycling region) [135]. An early study using monkey palm skin demonstrated that tritiated thymidine-labelled cells are located at the tips of the rete ridges [136], where basal cells express fast-cycling stem cell marker genes [135]. This suggests that the epidermal stem cell heterogeneity model is conserved [138,139].
Several signaling pathways organize the territorial segregation of the slow- and fast-cycling stem cells (in the interscale and the scale of tail skin), including Wnt, Lrig1, and Edaradd [140,141]. ECM, such as collagen XVII, a hemidesmosome component expressed by epidermal stem cells, is required for proper scale patterning [142], possibly through positive regulation of Wnt signaling [143,144]. The distinctive characteristics between slow- and fast-cycling stem cells raise intriguing questions about their biological relevance. It has been suggested that such heterogeneity confers robustness to skin homeostasis during environmental challenges such as UV exposure [135]. Interestingly, these two stem cell populations become interchangeable during wound repair, although they return to their initial territories once the wound is healed [134]. It is unknown whether the induction of matricellular protein expression during injury repair modulates the behavior of the two stem cell types.

3.1.1. Keratinocyte Regulation during Injury-Induced Skin Regeneration

Skin wound healing is an intricate process involving the interactions of many ECMs, growth factors, and various cell types [3,145,146]. It begins with the injury site filling with blood clotting factors and chemokines to attract leukocytes, including neutrophils, to clean the wounded area. Macrophages then enter the area to phagocytize neutrophils and secrete growth factors to stimulate the activation of keratinocytes and fibroblasts, resulting in ECM and BM production, re-epithelialization, and granulation tissue formation accompanied by neo-vascularization and tissue remodeling [146]. Here, we focus on the effect of matricellular proteins during the re-epithelialization process (Table 3), where keratinocytes and multiple skin stem cells proliferate and migrate to close the wounded area.
Table 3. Matricellular proteins regulating wound healing (re-epithelialization), epidermal stem cells, and aging skin.
Table 3. Matricellular proteins regulating wound healing (re-epithelialization), epidermal stem cells, and aging skin.
ProcessMatricellular
Protein
FunctionReferences & Study Model
Wound
healing
(Re-epithelialization)
CCN1upregulated during skin repair, induces myofibroblast senescence to minimize fibrosis, enhances neutrophil efferocytosis by macrophages for resolution of inflammation[147,148] (summary), [149,150] (mouse)
promotes keratinocytes proliferation and induces EMT during skin expansion[151] (human primary keratinocytes & in vivo mouse), [152] (human & rat skin)
Thrombospondin 1spatiotemporal expression regulates wound healing processes[153,154] (mouse)
promotes keratinocyte migration and proliferation[71] (human keratinocytes culture & in vivo mouse), [155] (mouse)
Thrombospondin 2influences vascularization but not re-epithelialization[153,156] (mouse)
Thrombospondin 4accelerates wound healing through increased keratinocytes proliferation and fibroblasts migration[157] (human & mouse skin)
Thrombospondin 5suppresses keratinocyte activation[65] (human skin)
Periostinregulates re-epithelialization and wound contraction, downregulated in aging skin[108,158,159] (mouse), [43] (human skin)
R-spondin 1regulates skin differentiation and malignancy, promotes wound healing and re-epithelialization[160] (human & mouse), [161] (rat)
TGFBIBM component, promotes epidermal stem cell proliferation and re-epithelialization[162] (human & mouse)
AgingFibulin 7decreased in aged BM, maintains epidermal stem cell heterogeneity, binds to periostin and tenascin C and improves re-epithelialization in aging skin wound healing[18] (mouse)
Thrombospondin 1upregulated in epidermal stem cells and dermal fibroblasts during aging and may contribute to aging-associated vasculature changes[124,163] (mouse), [164] (human dermal fibroblasts)
Mindin (Spon2)Mindin promotes epidermal stem/progenitor cell expansion downstream of Snail, inhibition of Mindin and fibulin 5 rescues Snail-induced skin fibrosis[165,166,167] (mouse)
Fibulin 5 [168] (mouse)
CCN1promotes fibroblast senescence in skin dermis[149,169] (mouse)
CCN2decreased in aging dermis is associated with collagen loss[170] (human skin & dermal fibroblasts), [171] (human dermal fibroblasts)
The CCN1 matricellular protein is upregulated during skin repair [147] and was initially found to promote wound healing by inducing myofibroblast senescence to minimize fibrosis [149] and by enhancing neutrophil efferocytosis by macrophages to stimulate inflammatory resolution [150]. Later, CCN1 was found to support re-epithelialization by promoting keratinocyte proliferation and migration [151]. Intriguingly, CCN1 has also been shown to increase in the epidermis during skin expansion in humans and rats, inducing proliferation and the epithelial-to-mesenchymal transition (EMT) [152,172], a reversible and temporary process whereby epithelial cells gain mesenchymal properties that contribute to increasing cell migration and re-epithelialization.
Thbs1 knockout mice exhibit delayed wound healing characterized by persistent inflammation, and reduced macrophage infiltration and TGFβ1 expression [153]. However, constitutive overexpression of Thbs1 under the keratin 14 promoter has also resulted in delayed re-epithelialization and overall wound healing due to defective granulation tissue and the inhibition of wound angiogenesis [26,154]. These observations underscore the importance of spatiotemporal regulation of Thbs1 expression in wound healing. Moreover, a recent report using single-cell RNA sequencing indicated that basal keratinocytes at the migrating front highly express Thbs1 [71,155]. These findings were supported by immunostainings and in vitro studies describing THBS1′s role in promoting keratinocyte migration and proliferation [71]. In contrast, Thbs2 deletion results in accelerated wound healing in mice without changes in the re-epithelialization rate due to higher vascularization, although the newly formed epidermis in Thbs2 null mice is thicker and forms an unusual rete ridge structure similar to the human skin architecture [153,156]. THBS4 is also upregulated in inflamed human psoriatic skin, post-burn injuries, and mouse dorsal skin wound assays. The administration of recombinant THBS4 to wounded skin in mice has led to faster wound healing, possibly due to increased keratinocyte proliferation and fibroblast migration [157]. Furthermore, COMP/THBS5 co-localizes with laminin α1 and integrin β1 at the DEJ, where it was found to suppress keratinocyte activation (proliferation and migration) in an ex vivo wound healing model [65].
The fasciclin family of matricellular proteins (periostin and TGFβ-Induced Protein (TGFBI)) is known to promote wound healing partly due to increased re-epithelialization [3,44,162]. Periostin-deficient mice exhibited delayed wound healing with defective re-epithelialization [108] and myofibroblast activation that facilitates wound contraction [158,159]. Similar to the over-expression of Thbs1 [154], constitutive overexpression of periostin dampens the wound repair process by inhibiting neutrophil and macrophage infiltration [173]. The other member of the family, TGFBI, was identified as a BM component in the epidermis. It enhances wound healing by promoting epidermal stem cell proliferation and re-epithelialization [162].
In wound healing, epidermal stem cells are empowered with plasticity that allows them to temporarily convert to other lineages (a term known as lineage infidelity) during re-epithelialization [174,175,176,177]. For example, post-injury hair follicle stem cells are mobilized to take on the role of epidermal stem cells and regenerate the epidermis [174,175]. There is still much that is unknown concerning how matricellular proteins modulate epidermal stem cells as part of the wound healing process. Among them, the R-Spondin family is known to activate Wnt signaling, which is crucial for adult epidermal stem cell maintenance [178,179]. R-Spondin 1 has been shown to regulate skin differentiation, have a predisposition for squamous cell carcinoma [160], and its topical application in rat skin promotes wound healing and re-epithelialization [161]. Interestingly, hair follicle-derived epidermal stem cells are more responsive to R-Spondin 1 and WNT7a stimulation (a characteristic of hair follicle stem cell activation) compared with native epidermal stem cells, suggesting they retain the memory from their original niches [175]. Whether matricellular proteins directly influence epidermal stem cell plasticity during injury remains unanswered and an open subject to be explored.

3.1.2. Epidermal Stem Cell Aging

As we age, our skin becomes thinner and more fragile, and its wound healing ability decreases. This is partly due to the impaired epidermal regeneration process in aged skin, which suffers from imbalanced BM ECM and growth factor signaling, an increased inflammatory microenvironment, the loss of epidermal stem cell heterogeneity, and diminished stem cell potential [18,125,180,181,182,183,184,185]. During aging, epidermal stem cells proliferate less [186,187], Wnt signaling components are downregulated [112,188,189], and fast-cycling epidermal stem cells are gradually depleted, which leads to reduced tissue fitness and resilience. However, slow-cycling epidermal stem cells are maintained into old age [18]. These slow-cycling stem cells are enriched for collagen XVII expression (Figure 3) [135], enabling their firm attachment to the BM and a higher self-renewal potential. In contrast, low collagen XVII-expressing cells are outcompeted, delaminated, and differentiated in aged skin [181].
What changes occur in the BM during aging that promote the loss of epidermal stem cell heterogeneity? In young human skin, wavy, undulating structures increase the DEJ area and strengthen the connection between the epidermis and the underlying dermis to increase skin mechanical resistance [190]. In aged human skin, however, the epidermis appears flatter than in young skin [182,191]. Human epidermal stem cells residing on the different areas of the undulating BM structures are also characterized by unique mechanical properties that may contribute to their heterogeneity; cells in the tip area are softer (lower Young’s modulus) and express higher β1 integrin stem cell marker levels compared with cells at the base area [192]. Flattening of the DEJ in aging skin may affect epidermal stem cell properties and shift the population balance towards stiffer stem cell populations, similar to stem cells grown on a flat surface [192].
Although epidermal BM becomes stiffer and thicker during aging [125,193,194], the expression of multiple constituents of the BM structural ECM is decreased, including collagen IV, VII, and XVII and laminin-332 [181,194,195,196,197]. Aberrant matricellular protein expression results in matrix re-arrangement, collagen cross-linking, and dysregulated enzymatic activity, thus promoting tissue stiffness in tumorigenesis [4]. However, it is unclear whether aging also induces alterations to matricellular proteins at the BM to promote stiffness and epidermal stem cell deregulation. Here, we discuss the studies that address some aspects of this question (Table 3).
The abundance of the matricellular protein fibulin 7 (encoded by FBLN7) at the epidermal BM is decreased in aged skin, with a concomitant loss of epidermal stem cell heterogeneity [18]. Fibulin 7 belongs to the short fibulin family of proteins but has no elastogenic function like other family members, such as fibulin 4 and 5 [198]. Instead, fibulin 7 is essential in maintaining long-term, fast-cycling epidermal stem cell potential during chronological aging. Fibulin 7 loss of function in mice leads to early depletion of fast-cycling stem cells, as shown by lineage tracing experiments after a 1-year chase [18]. Although the molecular mechanism of fibulin 7 regulation of fast-cycling stem cells is not well-defined, fibulin 7 deletion results in higher fast-cycling stem cell proliferation at a young age, which may have contributed to earlier fast-cycling stem cell exhaustion in the aging Fbln7 null mice compared with wild type mice. Moreover, fibulin 7 loss of function is correlated with augmented stem cell differentiation and lower collagen XVII expression, a stem cell fitness marker [18,181].
Fibulin 7 can also form a direct interaction with collagen IV, which may support its role in the BM to influence stem cell fate [18,199,200]. Other fibulin 7-binding proteins include fibronectin [18,201], which regulates cell adhesion at the BM to keep basal keratinocytes in an undifferentiated state [202,203,204], as well as periostin and tenascin C, which are both matricellular proteins highly expressed during skin inflammation and wound healing [29,79]. Further characterization of fibulin 7 function in epidermal stem cells suggests that it suppresses aging-associated inflammatory responses and lineage misregulation. Finally, fibulin 7 has demonstrated an ability to ameliorate the re-epithelialization process during full-thickness wound healing in 1-year-old, middle-aged mice, likely due to the abovementioned roles and interactions [18].
Aging-induced epidermal stem cell differentiation and hemidesmosome instability are also linked to age-related dermal stiffening and vasculature atrophy, as the induction of dermal vasculature reverses this phenotype [124]. In this study, the authors focused on an increased calcium influx in the basal keratinocytes mediated by the mechanosensitive Piezo1 ion chanel and the secretion of the immune-responsive protein pentraxin 3 by aged dermal fibroblasts, which plays an anti-angiogenic role in this context. Likewise, aged epidermal stem cells and dermal fibroblasts upregulate the Thbs1 matricellular protein, inhibiting angiogenesis [124]. This is supported by earlier studies that also found THBS1 upregulation in secreted proteins from human dermal fibroblasts isolated from intrinsically aged skin compared with young donors [164] and found that cutaneous blood flow is recovered and induced in aged Thbs1 or CD47 (a receptor for THBS1) null mouse skin [163]. It is still unknown whether the THBS1-CD47 axis directly regulates epidermal stem cells. However, in lung endothelial cells, THBS1-CD47 signaling inhibits self-renewal ability in cultures [205], and BMP4-dependent THBS1 expression in lung endothelial cells triggers differentiation of bronchioalveolar stem cells, which are activated during lung injury repair [206,207]. Increased THBS1-CD47 signaling has also been observed in aging human lung vasculature, promoting endothelial cell senescence [207,208].
Aging skin is associated with an increased risk of tumorigenic growth. Snail is a transcription factor known to be a driver of the EMT, a process involved in the maintenance of cancer stem cells and metastasis [209]. When Snail is overexpressed in the mouse epidermis, epidermal stem/progenitor cell populations expand, and stemness is promoted [165,166,210]. In turn, Snail induces the transcription of Mindin (SPON2) matricellular protein, which binds to integrin αMβ2 (CD11b) in an autocrine manner and activates c-Src and STAT3. Loss of function in Mindin was further shown to rescue the effect of Snail overexpression on epidermal expansion and in squamous cell carcinoma xenograft models [165]. In addition, a higher SNAI1 expression was found in the epidermis of patients with systemic sclerosis, linking its function in skin fibrosis to the induction of inflammation and fibroblast-mediated collagen secretion [167,168]. Mindin and fibulin 5 inhibition can rescue Snail-induced skin fibrosis, making them potential therapeutic targets. As fibrosis and ECM stiffness are linked to tissue aging, it would be interesting to address the role of these proteins in the context of aging skin.

3.2. Hair Follicle Stem Cells

Hair follicle stem cells reside in the bulge region, which is enriched for matricellular proteins such as tenascin C, periostin, and Mindin/Spon2 [101] (Table 3). Tenascin C can interact with the Wnt3a ligand, thus facilitating Wnt signaling to maintain the hair follicle stem cell population and suppress aberrant differentiation [32]. Periostin positively regulates the proliferation of cells in the hair follicles after skin wounding and in cultured keratinocytes via activation of NFκB. The loss of periostin expression is further associated with decreased fibronectin expression around the hair follicle BM [108].
Matricellular proteins are also expressed in the dermal papilla to regulate hair follicle growth. For example, CCN2 maintains hair follicle stem cell quiescence, as its genetic deletion from the dermal papilla niche results in an increased number of hair follicles with a shorter telogen phase [109]. In contrast, the Wnt agonist R-Spondin 2, through intradermal administration, activates hair follicle stem cells and sustains the anagen phase, promoting hair growth [110]. Dermal papilla cells also express R-spondin 2 and R-spondin 3 to promote cell proliferation of hair follicle dermal stem cells that replenish the dermal sheath and hair follicle progenitors [111]. However, the inhibition of TGFβ signaling in skin fibroblasts induces dermal papilla niche reorganization, resulting in inefficient hair production and a redistribution of progenitor cells, although their proliferation and differentiation are generally preserved [211].
In aged hair follicle stem cells, Mindin/Spon2 gene expression is decreased while Thbs1 is increased [112]. The role of Mindin in hair follicle stem cells has not been reported, although it may be related to stemness, as in the case of epidermal stem cells [165]. Hair follicle stem cell activation is associated with the skin vasculature [212,213]. THBS1 is an angiogenesis inhibitor and has been shown to induce hair follicle shrinkage and vascular regression during the catagen phase of the hair cycle [113]. Thus, an increase in THBS1 expression may be linked to reduced hair density associated with aging. In line with the involution of vasculature in aged skin and dermal ECM changes [124], YAP/TAZ mechanosignaling is suppressed in aged dermal fibroblasts [127]. Conditional knockout of YAP in these fibroblasts mimics the effect of aging skin (i.e., a reduced number of hair follicles). In contrast, the expression of constitutively active YAP rescues skin from the aging phenotype (a restored number of hair follicles) [127]. Fibroblast deregulation could be further attributed to an increase in matricellular protein CCN1 in the aging dermis [169]. CCN1 augments fibroblast senescence, suppressing fibrotic genes such as Col1a1 [149], one of the hallmarks of aging skin [214].

3.3. Sebaceous Gland Stem Cells

Sebaceous gland stem cells are originated from hair follicle stem cells expressing Sox9 or Lrig1 [215,216]. Sebaceous gland stem cells and progenitor cells express the embigin cell surface receptor, which binds to fibronectin at the N-terminus domain for cell adhesion. Embigin expression is regulated by Wnt signaling, and its deletion drives basal cell detachment and differentiation, resulting in increased sebaceous glands areas and an increased number of sebocytes. Intriguingly, tissue stiffness is increased in the ECM of the basal progenitor cells upon loss of embigin [58]. Tissue stiffness has been reported in skin aging, such as increased BM stiffness in the hair follicle stem cells [125], and sebaceous glands tend to become larger during aging despite reduced sebum production [217]. It has been suggested that the increased number of differentiated sebocytes due to embigin deletion is potentiated with increased age [58], indicating that an aging-dependent factor may be further modulating the embigin–fibronectin interaction.
Several signaling pathways involving Wnt, TGFβ, Shh, and Notch have been reported for homeostasis regulation and aging of the sebaceous glands [218]. Matricellular proteins such as the CCN family can bind to TGFβ, Notch, and the Wnt co-receptor LRP6, thus affecting signaling outcomes [148,219,220,221], although their roles in sebaceous glands aging have not been reported. In line with this, CCN protein expressions are changed during aging, with CCN1 upregulated and CCN2/CTGF (connective tissue growth factor) downregulated in the dermis of aged human skin [169,170,171].

3.4. Cross-Communications and Lineage-Fate Plasticity Potentially Involving Matricellular Proteins in the Skin Network

Cross-communications between stem cells and the surrounding cells in the niche, such as nerve cells, muscle cells, fibroblasts, immune cells, lymphatics, or vasculature, sustain skin homeostasis. The importance of matricellular proteins in maintaining such cellular cross-talks is largely unknown. For example, interactions between the hair follicle stem cells with the sympathetic nervous system secreting the neurotransmitter noradrenaline control stem cell activity. This interaction requires the arrector pili muscle, which harbors the sympathetic innervations [222]. The attachment of the arrector pili muscle to the bulge region has been attributed to the nephronectin ECM; however, matricellular proteins such as periostin, fibulin 1, and tenascin C are also present in this region, possibly stabilizing the structure [54]. Likewise, the melanocyte stem cells in the hair follicle are modulated by the sympathetic innervation. Stress causes the rush of noradrenaline and hyperproliferation of melanocyte stem cells, followed by their differentiation, eventually depleting the melanocyte stem cell pool leading to greying hair [223]. The lymphatic vasculature also functions as a hair follicle stem cell niche [224,225]. The secretome of hair follicle stem cells alters with the hair cycle; in the resting phase, they secrete angiopoietin-like protein 7 to promote lymphatic fitness and stem cell quiescence, whereas during the growing phase, netrin 4 ECM and angiopoietin-like protein 4 are increased to induce lymphatic capillary remodeling and stem cell activation [225]. Similarly, the hair cycle and hair follicle stem cells are influenced by their neighboring blood vasculature [212]. Aging hair follicle stem cells upregulate Thbs1 expression [112], which is anti-angiogenic and may change the stem cell–vasculature interactions in aging skin. Moreover, there are defective interactions between epidermal cells and dendritic T-cells in aging skin wound healing, and overall cellular composition, transcriptomics, and cell–cell communication are compromised [180,226].
Much remains unknown regarding whether and how matricellular proteins are involved in skin stem cell regulation during wound healing and inflammation. Do matricellular proteins influence skin cell plasticity during injury-induced regeneration? Inflammation-induced skin stem cells and skin cell plasticity have been well reported [73,81,174,175,176,177,227,228,229,230,231,232]. Slow- and fast-cycling epidermal stem cells leave their unique territories and become interchangeable in wound repair [134]. Hair follicle stem cells contribute as epidermal stem cells in post-wounding epidermal regeneration. On the contrary, epidermal stem cells are described as participating in large wound-induced hair follicle neogenesis [174,176,177]. Interestingly, differentiated sebocytes expressing Gata6 undergo de-differentiation and become epidermal stem cells during injury repair [229], and a high degree of fibroblast heterogeneity has been observed in wound healing [83,233,234]. While the cause of such plasticity or heterogeneity is linked to the upregulation of stress-associated transcription factors [174,175,227], the role of the ECM or microenvironment is unclear. In aging skin, DNA damage leads to the proteolysis of collagen XVII in the BM of hair follicle stem cells, resulting in an epidermal fate and hair loss [130], although aging-induced lineage conversion and the role of the stem cell microenvironment is still an ongoing discussion [112]. Our findings of the loss of fibulin 7 in the aging epidermal BM and its association with the loss of fast-cycling epidermal stem cells further suggest that microenvironmental alterations affect stem cell fate [18]. Matricellular proteins can influence cell behaviors [3,146]; hence, future studies that address their roles in skin stem cell regulation and cell–cell interactions would be of great interest.

4. Concluding Remarks

While matricellular proteins are abundantly expressed during skin development or in an inflammatory context during injury repair or disease, a basal level of expression is maintained in various compartments of the skin, which is necessary to support overall skin functioning. This occurs through matricellular proteins interacting with structural ECMs, such as collagens and elastic fibers, and through regulation of growth factor bioavailability and cell–cell or cell–matrix adhesion, thereby orchestrating cellular responses and tissue functions (Table 1, Table 2 and Table 3). The importance of matricellular protein’s spatiotemporal expression has been demonstrated in studies where constitutive null and overexpression mice have yielded identical phenotypes, adding difficulties to studying the function of these proteins in vivo. Inducible tissue-specific over-expression or loss-of-function studies would facilitate efforts to further elucidate their roles in the skin, especially in stem cell regulation. Of note, even though human and mouse skin share some similarities, they do have notable differences [235] that could limit our interpretation from studies using rodent skin.
Given the known significance of the presence of matricellular proteins during tissue development or inflammation, where various stem cells are activated, it would be unsurprising if matricellular proteins modulate stem cell niche and function during organogenesis, as well as adult stem cells and their response to injury, as shown by studies using fibulin 7 and TGFBI (Table 3) [18,162]. Nevertheless, few of the matricellular proteins that have been reported to regulate the re-epithelialization process (Table 3) link their functions to epidermal stem cells. Finally, it remains to be explored whether matricellular protein dynamics in the stem cells’ niche affect epigenetic changes involved in tissue stem cell inflammatory memory and stem cell aging [125,175,228]. This holds promise for potential future applications in regenerative medicine.

Author Contributions

E.R. and H.Y. contributed to the conceptualization and editing of the manuscript. E.R. wrote the manuscript’s first draft. M.T.R.D.C.C. contributed to figures and tables construction. All authors have read and agreed to the published version of the manuscript.

Funding

E.R is supported by Grant-in-Aid for Scientific Research (C) (23K07737) and Lydia O’leary Memorial PIAS Dermatological Foundation/Elastin molecular research grant. M.C is supported by the Ph.D. Program in Humanics Special Fellowship (University of Tsukuba). H.Y.’s laboratory is supported in part by the Japan Agency for Medical Research and Development (AMED) (grant number JP21ek0109553h001).

Acknowledgments

We apologize for any omissions due to the spatial constraints of the manuscript. A special thanks to Aiko Sada for her continuous sharing of insightful work and references in skin stem cell biology.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bornstein, P. Diversity of function is inherent in matricellular proteins: An appraisal of thrombospondin 1. J. Cell Biol. 1995, 130, 503–506. [Google Scholar] [CrossRef]
  2. Murphy-Ullrich, J.E.; Sage, E.H. Revisiting the matricellular concept. Matrix Biol. 2014, 37, 1–14. [Google Scholar] [CrossRef] [PubMed]
  3. Cardenas-Leon, C.G.; Maemets-Allas, K.; Klaas, M.; Lagus, H.; Kankuri, E.; Jaks, V. Matricellular proteins in cutaneous wound healing. Front. Cell Dev. Biol. 2022, 10, 1073320. [Google Scholar] [CrossRef] [PubMed]
  4. Gerarduzzi, C.; Hartmann, U.; Leask, A.; Drobetsky, E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res. 2020, 80, 2705–2717. [Google Scholar] [CrossRef]
  5. Bornstein, P. Matricellular proteins: An overview. J. Cell Commun. Signal. 2009, 3, 163–165. [Google Scholar] [CrossRef]
  6. Asano, K.; Cantalupo, A.; Sedes, L.; Ramirez, F. The Multiple Functions of Fibrillin-1 Microfibrils in Organismal Physiology. Int. J. Mol. Sci. 2022, 23, 1892. [Google Scholar] [CrossRef]
  7. Theocharis, A.D.; Manou, D.; Karamanos, N.K. The extracellular matrix as a multitasking player in disease. FEBS J. 2019, 286, 2830–2869. [Google Scholar] [CrossRef] [PubMed]
  8. Rose, C.D.; Pompili, D.; Henke, K.; Van Gennip, J.L.M.; Meyer-Miner, A.; Rana, R.; Gobron, S.; Harris, M.P.; Nitz, M.; Ciruna, B. SCO-Spondin Defects and Neuroinflammation Are Conserved Mechanisms Driving Spinal Deformity across Genetic Models of Idiopathic Scoliosis. Curr. Biol. 2020, 30, 2363–2373.e6. [Google Scholar] [CrossRef]
  9. Mo, F.E.; Muntean, A.G.; Chen, C.C.; Stolz, D.B.; Watkins, S.C.; Lau, L.F. CYR61 (CCN1) is essential for placental development and vascular integrity. Mol. Cell. Biol. 2002, 22, 8709–8720. [Google Scholar] [CrossRef]
  10. Barque, A.; Jan, K.; De La Fuente, E.; Nicholas, C.L.; Hynes, R.O.; Naba, A. Knockout of the gene encoding the extracellular matrix protein SNED1 results in early neonatal lethality and craniofacial malformations. Dev. Dyn. 2021, 250, 274–294. [Google Scholar] [CrossRef]
  11. Ivkovic, S.; Yoon, B.S.; Popoff, S.N.; Safadi, F.F.; Libuda, D.E.; Stephenson, R.C.; Daluiski, A.; Lyons, K.M. Connective tissue growth factor coordinates chondrogenesis and angiogenesis during skeletal development. Development 2003, 130, 2779–2791. [Google Scholar] [CrossRef]
  12. Saga, Y.; Yagi, T.; Ikawa, Y.; Sakakura, T.; Aizawa, S. Mice develop normally without tenascin. Genes Dev. 1992, 6, 1821–1831. [Google Scholar] [CrossRef]
  13. He, Y.W.; Li, H.; Zhang, J.; Hsu, C.L.; Lin, E.; Zhang, N.; Guo, J.; Forbush, K.A.; Bevan, M.J. The extracellular matrix protein mindin is a pattern-recognition molecule for microbial pathogens. Nat. Immunol. 2004, 5, 88–97. [Google Scholar] [CrossRef]
  14. Daubon, T.; Leon, C.; Clarke, K.; Andrique, L.; Salabert, L.; Darbo, E.; Pineau, R.; Guerit, S.; Maitre, M.; Dedieu, S.; et al. Deciphering the complex role of thrombospondin-1 in glioblastoma development. Nat. Commun. 2019, 10, 1146. [Google Scholar] [CrossRef] [PubMed]
  15. Bhattacharyya, S.; Wang, W.; Morales-Nebreda, L.; Feng, G.; Wu, M.; Zhou, X.; Lafyatis, R.; Lee, J.; Hinchcliff, M.; Feghali-Bostwick, C.; et al. Tenascin-C drives persistence of organ fibrosis. Nat. Commun. 2016, 7, 11703. [Google Scholar] [CrossRef] [PubMed]
  16. Hsu, Y.C.; Fuchs, E. Building and Maintaining the Skin. Cold Spring Harb. Perspect. Biol. 2022, 14, a040840. [Google Scholar] [CrossRef]
  17. Longmate, W.M.; Monichan, R.; Chu, M.L.; Tsuda, T.; Mahoney, M.G.; DiPersio, C.M. Reduced fibulin-2 contributes to loss of basement membrane integrity and skin blistering in mice lacking integrin alpha3beta1 in the epidermis. J. Investig. Dermatol. 2014, 134, 1609–1617. [Google Scholar] [CrossRef]
  18. Raja, E.; Changarathil, G.; Oinam, L.; Tsunezumi, J.; Ngo, Y.X.; Ishii, R.; Sasaki, T.; Imanaka-Yoshida, K.; Yanagisawa, H.; Sada, A. The extracellular matrix fibulin 7 maintains epidermal stem cell heterogeneity during skin aging. EMBO Rep. 2022, 23, e55478. [Google Scholar] [CrossRef]
  19. Nakamura, T.; Yoshida, H.; Ota, Y.; Endo, Y.; Sayo, T.; Hanai, U.; Imagawa, K.; Sasaki, M.; Takahashi, Y. SPARC promotes production of type IV and VII collagen and their skin basement membrane accumulation. J. Dermatol. Sci. 2022, 107, 109–112. [Google Scholar] [CrossRef]
  20. Sasaki, T.; Hohenester, E.; Gohring, W.; Timpl, R. Crystal structure and mapping by site-directed mutagenesis of the collagen-binding epitope of an activated form of BM-40/SPARC/osteonectin. EMBO J. 1998, 17, 1625–1634. [Google Scholar] [CrossRef] [PubMed]
  21. Bradshaw, A.D. The role of SPARC in extracellular matrix assembly. J. Cell Commun. Signal. 2009, 3, 239–246. [Google Scholar] [CrossRef] [PubMed]
  22. Welcker, D.; Stein, C.; Feitosa, N.M.; Armistead, J.; Zhang, J.L.; Lutke, S.; Kleinridders, A.; Bruning, J.C.; Eming, S.A.; Sengle, G.; et al. Hemicentin-1 is an essential extracellular matrix component of the dermal-epidermal and myotendinous junctions. Sci. Rep. 2021, 11, 17926. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, J.L.; Richetti, S.; Ramezani, T.; Welcker, D.; Lutke, S.; Pogoda, H.M.; Hatzold, J.; Zaucke, F.; Keene, D.R.; Bloch, W.; et al. Vertebrate extracellular matrix protein hemicentin-1 interacts physically and genetically with basement membrane protein nidogen-2. Matrix Biol. 2022, 112, 132–154. [Google Scholar] [CrossRef]
  24. Wight, T.N.; Raugi, G.J.; Mumby, S.M.; Bornstein, P. Light microscopic immunolocation of thrombospondin in human tissues. J. Histochem. Cytochem. 1985, 33, 295–302. [Google Scholar] [CrossRef] [PubMed]
  25. Detmar, M. The role of VEGF and thrombospondins in skin angiogenesis. J. Dermatol. Sci. 2000, 24 (Suppl. 1), S78–S84. [Google Scholar] [CrossRef] [PubMed]
  26. Streit, M.; Velasco, P.; Brown, L.F.; Skobe, M.; Richard, L.; Riccardi, L.; Lawler, J.; Detmar, M. Overexpression of thrombospondin-1 decreases angiogenesis and inhibits the growth of human cutaneous squamous cell carcinomas. Am. J. Pathol. 1999, 155, 441–452. [Google Scholar] [CrossRef]
  27. Fukunaga-Kalabis, M.; Martinez, G.; Liu, Z.J.; Kalabis, J.; Mrass, P.; Weninger, W.; Firth, S.M.; Planque, N.; Perbal, B.; Herlyn, M. CCN3 controls 3D spatial localization of melanocytes in the human skin through DDR1. J. Cell Biol. 2006, 175, 563–569. [Google Scholar] [CrossRef]
  28. Fukunaga-Kalabis, M.; Santiago-Walker, A.; Herlyn, M. Matricellular proteins produced by melanocytes and melanomas: In search for functions. Cancer Microenviron. 2008, 1, 93–102. [Google Scholar] [CrossRef]
  29. Midwood, K.S.; Chiquet, M.; Tucker, R.P.; Orend, G. Tenascin-C at a glance. J. Cell Sci. 2016, 129, 4321–4327. [Google Scholar] [CrossRef]
  30. Swindle, C.S.; Tran, K.T.; Johnson, T.D.; Banerjee, P.; Mayes, A.M.; Griffith, L.; Wells, A. Epidermal growth factor (EGF)-like repeats of human tenascin-C as ligands for EGF receptor. J. Cell Biol. 2001, 154, 459–468. [Google Scholar] [CrossRef]
  31. Schalkwijk, J.; Steijlen, P.M.; van Vlijmen-Willems, I.M.; Oosterling, B.; Mackie, E.J.; Verstraeten, A.A. Tenascin expression in human dermis is related to epidermal proliferation. Am. J. Pathol. 1991, 139, 1143–1150. [Google Scholar]
  32. Hendaoui, I.; Tucker, R.P.; Zingg, D.; Bichet, S.; Schittny, J.; Chiquet-Ehrismann, R. Tenascin-C is required for normal Wnt/beta-catenin signaling in the whisker follicle stem cell niche. Matrix Biol. 2014, 40, 46–53. [Google Scholar] [CrossRef] [PubMed]
  33. De Laporte, L.; Rice, J.J.; Tortelli, F.; Hubbell, J.A. Tenascin C promiscuously binds growth factors via its fifth fibronectin type III-like domain. PLoS ONE 2013, 8, e62076. [Google Scholar] [CrossRef] [PubMed]
  34. Martino, M.M.; Briquez, P.S.; Guc, E.; Tortelli, F.; Kilarski, W.W.; Metzger, S.; Rice, J.J.; Kuhn, G.A.; Muller, R.; Swartz, M.A.; et al. Growth factors engineered for super-affinity to the extracellular matrix enhance tissue healing. Science 2014, 343, 885–888. [Google Scholar] [CrossRef] [PubMed]
  35. Huang, J.; Heng, S.; Zhang, W.; Liu, Y.; Xia, T.; Ji, C.; Zhang, L.J. Dermal extracellular matrix molecules in skin development, homeostasis, wound regeneration and diseases. Semin. Cell Dev. Biol. 2022, 128, 137–144. [Google Scholar] [CrossRef]
  36. Martino, M.M.; Tortelli, F.; Mochizuki, M.; Traub, S.; Ben-David, D.; Kuhn, G.A.; Muller, R.; Livne, E.; Eming, S.A.; Hubbell, J.A. Engineering the growth factor microenvironment with fibronectin domains to promote wound and bone tissue healing. Sci. Transl. Med. 2011, 3, 100ra189. [Google Scholar] [CrossRef]
  37. Reinhardt, D.P.; Sasaki, T.; Dzamba, B.J.; Keene, D.R.; Chu, M.L.; Gohring, W.; Timpl, R.; Sakai, L.Y. Fibrillin-1 and fibulin-2 interact and are colocalized in some tissues. J. Biol. Chem. 1996, 271, 19489–19496. [Google Scholar] [CrossRef]
  38. Lemaire, R.; Korn, J.H.; Schiemann, W.P.; Lafyatis, R. Fibulin-2 and fibulin-5 alterations in tsk mice associated with disorganized hypodermal elastic fibers and skin tethering. J. Investig. Dermatol. 2004, 123, 1063–1069. [Google Scholar] [CrossRef]
  39. Zhang, X.; Alanazi, Y.F.; Jowitt, T.A.; Roseman, A.M.; Baldock, C. Elastic Fibre Proteins in Elastogenesis and Wound Healing. Int. J. Mol. Sci. 2022, 23, 4087. [Google Scholar] [CrossRef]
  40. Hucthagowder, V.; Sausgruber, N.; Kim, K.H.; Angle, B.; Marmorstein, L.Y.; Urban, Z. Fibulin-4: A novel gene for an autosomal recessive cutis laxa syndrome. Am. J. Hum. Genet. 2006, 78, 1075–1080. [Google Scholar] [CrossRef]
  41. Markova, D.; Zou, Y.Q.; Ringpfeil, F.; Sasaki, T.; Kostka, G.; Timpl, R.; Uitto, J.; Chu, M.L. Genetic heterogeneity of cutis laxa: A heterozygous tandem duplication within the fibulin-5 (FBLN5) gene. Am. J. Hum. Genet. 2003, 72, 998–1004. [Google Scholar] [CrossRef] [PubMed]
  42. Yanagisawa, H.; Davis, E.C.; Starcher, B.C.; Ouchi, T.; Yanagisawa, M.; Richardson, J.A.; Olson, E.N. Fibulin-5 is an elastin-binding protein essential for elastic fibre development in vivo. Nature 2002, 415, 168–171. [Google Scholar] [CrossRef] [PubMed]
  43. Egbert, M.; Ruetze, M.; Sattler, M.; Wenck, H.; Gallinat, S.; Lucius, R.; Weise, J.M. The matricellular protein periostin contributes to proper collagen function and is downregulated during skin aging. J. Dermatol. Sci. 2014, 73, 40–48. [Google Scholar] [CrossRef]
  44. Taniguchi, K.; Arima, K.; Masuoka, M.; Ohta, S.; Shiraishi, H.; Ontsuka, K.; Suzuki, S.; Inamitsu, M.; Yamamoto, K.I.; Simmons, O.; et al. Periostin controls keratinocyte proliferation and differentiation by interacting with the paracrine IL-1alpha/IL-6 loop. J. Investig. Dermatol. 2014, 134, 1295–1304. [Google Scholar] [CrossRef]
  45. Rosini, S.; Pugh, N.; Bonna, A.M.; Hulmes, D.J.S.; Farndale, R.W.; Adams, J.C. Thrombospondin-1 promotes matrix homeostasis by interacting with collagen and lysyl oxidase precursors and collagen cross-linking sites. Sci. Signal. 2018, 11, eaar2566. [Google Scholar] [CrossRef] [PubMed]
  46. Kyriakides, T.R.; Zhu, Y.H.; Smith, L.T.; Bain, S.D.; Yang, Z.; Lin, M.T.; Danielson, K.G.; Iozzo, R.V.; LaMarca, M.; McKinney, C.E.; et al. Mice that lack thrombospondin 2 display connective tissue abnormalities that are associated with disordered collagen fibrillogenesis, an increased vascular density, and a bleeding diathesis. J. Cell Biol. 1998, 140, 419–430. [Google Scholar] [CrossRef]
  47. Bradshaw, A.D.; Puolakkainen, P.; Dasgupta, J.; Davidson, J.M.; Wight, T.N.; Helene Sage, E. SPARC-null mice display abnormalities in the dermis characterized by decreased collagen fibril diameter and reduced tensile strength. J. Investig. Dermatol. 2003, 120, 949–955. [Google Scholar] [CrossRef]
  48. Bradshaw, A.D.; Graves, D.C.; Motamed, K.; Sage, E.H. SPARC-null mice exhibit increased adiposity without significant differences in overall body weight. Proc. Natl. Acad. Sci. USA 2003, 100, 6045–6050. [Google Scholar] [CrossRef]
  49. Bradshaw, A.D.; Reed, M.J.; Sage, E.H. SPARC-null mice exhibit accelerated cutaneous wound closure. J. Histochem. Cytochem. 2002, 50, 1–10. [Google Scholar] [CrossRef]
  50. Tremblay, F.; Revett, T.; Huard, C.; Zhang, Y.; Tobin, J.F.; Martinez, R.V.; Gimeno, R.E. Bidirectional modulation of adipogenesis by the secreted protein Ccdc80/DRO1/URB. J. Biol. Chem. 2009, 284, 8136–8147. [Google Scholar] [CrossRef]
  51. Okada, T.; Nishizawa, H.; Kurata, A.; Tamba, S.; Sonoda, M.; Yasui, A.; Kuroda, Y.; Hibuse, T.; Maeda, N.; Kihara, S.; et al. URB is abundantly expressed in adipose tissue and dysregulated in obesity. Biochem. Biophys. Res. Commun. 2008, 367, 370–376. [Google Scholar] [CrossRef] [PubMed]
  52. Grill, J.I.; Neumann, J.; Herbst, A.; Ofner, A.; Hiltwein, F.; Marschall, M.K.; Zierahn, H.; Wolf, E.; Schneider, M.R.; Kolligs, F.T. Loss of DRO1/CCDC80 results in obesity and promotes adipocyte differentiation. Mol. Cell. Endocrinol. 2017, 439, 286–296. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, H.Y.; Timpl, R.; Sasaki, T.; Chu, M.L.; Ekblom, P. Fibulin-1 and fibulin-2 expression during organogenesis in the developing mouse embryo. Dev. Dyn. 1996, 205, 348–364. [Google Scholar] [CrossRef]
  54. Fujiwara, H.; Ferreira, M.; Donati, G.; Marciano, D.K.; Linton, J.M.; Sato, Y.; Hartner, A.; Sekiguchi, K.; Reichardt, L.F.; Watt, F.M. The basement membrane of hair follicle stem cells is a muscle cell niche. Cell 2011, 144, 577–589. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, T.; Wagner, A.; Gehwolf, R.; Yan, W.; Passini, F.S.; Thien, C.; Weissenbacher, N.; Lin, Z.; Lehner, C.; Teng, H.; et al. Load-induced regulation of tendon homeostasis by SPARC, a genetic predisposition factor for tendon and ligament injuries. Sci. Transl. Med. 2021, 13, eabe5738. [Google Scholar] [CrossRef]
  56. Wang, Y.; Jin, S.; Luo, D.; He, D.; Shi, C.; Zhu, L.; Guan, B.; Li, Z.; Zhang, T.; Zhou, Y.; et al. Functional regeneration and repair of tendons using biomimetic scaffolds loaded with recombinant periostin. Nat. Commun. 2021, 12, 1293. [Google Scholar] [CrossRef]
  57. Jarvinen, T.A.; Jozsa, L.; Kannus, P.; Jarvinen, T.L.; Hurme, T.; Kvist, M.; Pelto-Huikko, M.; Kalimo, H.; Jarvinen, M. Mechanical loading regulates the expression of tenascin-C in the myotendinous junction and tendon but does not induce de novo synthesis in the skeletal muscle. J. Cell Sci. 2003, 116, 857–866. [Google Scholar] [CrossRef] [PubMed]
  58. Sipila, K.; Rognoni, E.; Jokinen, J.; Tewary, M.; Vietri Rudan, M.; Talvi, S.; Jokinen, V.; Dahlstrom, K.M.; Liakath-Ali, K.; Mobasseri, A.; et al. Embigin is a fibronectin receptor that affects sebaceous gland differentiation and metabolism. Dev. Cell 2022, 57, 1453–1465.e7. [Google Scholar] [CrossRef]
  59. Chen, D.; Chen, X.; Xie, H.T.; Hatton, M.P.; Liu, X.; Liu, Y. Expression of extracellular matrix components in the meibomian gland. Front. Med. 2022, 9, 981610. [Google Scholar] [CrossRef]
  60. Igoucheva, O.; Alexeev, V.; Halabi, C.M.; Adams, S.M.; Stoilov, I.; Sasaki, T.; Arita, M.; Donahue, A.; Mecham, R.P.; Birk, D.E.; et al. Fibulin-4 E57K Knock-in Mice Recapitulate Cutaneous, Vascular and Skeletal Defects of Recessive Cutis Laxa 1B with both Elastic Fiber and Collagen Fibril Abnormalities. J. Biol. Chem. 2015, 290, 21443–21459. [Google Scholar] [CrossRef]
  61. Lippens, S.; Denecker, G.; Ovaere, P.; Vandenabeele, P.; Declercq, W. Death penalty for keratinocytes: Apoptosis versus cornification. Cell Death Differ. 2005, 12 (Suppl. 2), 1497–1508. [Google Scholar] [CrossRef] [PubMed]
  62. Nystrom, A.; Bruckner-Tuderman, L. Matrix molecules and skin biology. Semin. Cell Dev. Biol. 2019, 89, 136–146. [Google Scholar] [CrossRef]
  63. Fleischmajer, R.; Timpl, R. Ultrastructural localization of fibronectin to different anatomic structures of human skin. J. Histochem. Cytochem. 1984, 32, 315–321. [Google Scholar] [CrossRef]
  64. Utani, A.; Nomizu, M.; Yamada, Y. Fibulin-2 binds to the short arms of laminin-5 and laminin-1 via conserved amino acid sequences. J. Biol. Chem. 1997, 272, 2814–2820. [Google Scholar] [CrossRef]
  65. Bozo, R.; Szel, E.; Danis, J.; Guban, B.; Bata-Csorgo, Z.; Szabo, K.; Kemeny, L.; Groma, G. Cartilage Oligomeric Matrix Protein Negatively Influences Keratinocyte Proliferation via alpha5beta1-Integrin: Potential Relevance of Altered Cartilage Oligomeric Matrix Protein Expression in Psoriasis. J. Investig. Dermatol. 2020, 140, 1733–1742.e7. [Google Scholar] [CrossRef] [PubMed]
  66. Ghetti, M.; Topouzi, H.; Theocharidis, G.; Papa, V.; Williams, G.; Bondioli, E.; Cenacchi, G.; Connelly, J.T.; Higgins, C.A. Subpopulations of dermal skin fibroblasts secrete distinct extracellular matrix: Implications for using skin substitutes in the clinic. Br. J. Dermatol. 2018, 179, 381–393. [Google Scholar] [CrossRef]
  67. Hunzelmann, N.; Hafner, M.; Anders, S.; Krieg, T.; Nischt, R. BM-40 (osteonectin, SPARC) is expressed both in the epidermal and in the dermal compartment of adult human skin. J. Investig. Dermatol. 1998, 110, 122–126. [Google Scholar] [CrossRef]
  68. Wrana, J.L.; Overall, C.M.; Sodek, J. Regulation of the expression of a secreted acidic protein rich in cysteine (SPARC) in human fibroblasts by transforming growth factor beta. Comparison of transcriptional and post-transcriptional control with fibronectin and type I collagen. Eur. J. Biochem. 1991, 197, 519–528. [Google Scholar] [CrossRef]
  69. Ford, R.; Wang, G.; Jannati, P.; Adler, D.; Racanelli, P.; Higgins, P.J.; Staiano-Coico, L. Modulation of SPARC expression during butyrate-induced terminal differentiation of cultured human keratinocytes: Regulation via a TGF-beta-dependent pathway. Exp. Cell Res. 1993, 206, 261–275. [Google Scholar] [CrossRef] [PubMed]
  70. Agarwal, P.; Zwolanek, D.; Keene, D.R.; Schulz, J.N.; Blumbach, K.; Heinegard, D.; Zaucke, F.; Paulsson, M.; Krieg, T.; Koch, M.; et al. Collagen XII and XIV, new partners of cartilage oligomeric matrix protein in the skin extracellular matrix suprastructure. J. Biol. Chem. 2012, 287, 22549–22559. [Google Scholar] [CrossRef]
  71. Siriwach, R.; Ngo, A.Q.; Higuchi, M.; Arima, K.; Sakamoto, S.; Watanabe, A.; Narumiya, S.; Thumkeo, D. Single-cell RNA sequencing identifies a migratory keratinocyte subpopulation expressing THBS1 in epidermal wound healing. iScience 2022, 25, 104130. [Google Scholar] [CrossRef] [PubMed]
  72. Domingues, L.; Hurbain, I.; Gilles-Marsens, F.; Sires-Campos, J.; Andre, N.; Dewulf, M.; Romao, M.; Viaris de Lesegno, C.; Mace, A.S.; Blouin, C.; et al. Coupling of melanocyte signaling and mechanics by caveolae is required for human skin pigmentation. Nat. Commun. 2020, 11, 2988. [Google Scholar] [CrossRef]
  73. Rognoni, E.; Watt, F.M. Skin Cell Heterogeneity in Development, Wound Healing, and Cancer. Trends Cell Biol. 2018, 28, 709–722. [Google Scholar] [CrossRef] [PubMed]
  74. Sabatelli, P.; Gara, S.K.; Grumati, P.; Urciuolo, A.; Gualandi, F.; Curci, R.; Squarzoni, S.; Zamparelli, A.; Martoni, E.; Merlini, L.; et al. Expression of the collagen VI alpha5 and alpha6 chains in normal human skin and in skin of patients with collagen VI-related myopathies. J. Investig. Dermatol. 2011, 131, 99–107. [Google Scholar] [CrossRef] [PubMed]
  75. Watt, F.M.; Fujiwara, H. Cell-extracellular matrix interactions in normal and diseased skin. Cold Spring Harb. Perspect. Biol. 2011, 3, a005124. [Google Scholar] [CrossRef] [PubMed]
  76. Pfisterer, K.; Shaw, L.E.; Symmank, D.; Weninger, W. The Extracellular Matrix in Skin Inflammation and Infection. Front. Cell Dev. Biol. 2021, 9, 682414. [Google Scholar] [CrossRef] [PubMed]
  77. Cescon, M.; Gattazzo, F.; Chen, P.; Bonaldo, P. Collagen VI at a glance. J. Cell Sci. 2015, 128, 3525–3531. [Google Scholar] [CrossRef]
  78. Lettmann, S.; Bloch, W.; Maass, T.; Niehoff, A.; Schulz, J.N.; Eckes, B.; Eming, S.A.; Bonaldo, P.; Paulsson, M.; Wagener, R. Col6a1 null mice as a model to study skin phenotypes in patients with collagen VI related myopathies: Expression of classical and novel collagen VI variants during wound healing. PLoS ONE 2014, 9, e105686. [Google Scholar] [CrossRef]
  79. Kuwatsuka, Y.; Murota, H. Involvement of Periostin in Skin Function and the Pathogenesis of Skin Diseases. Adv. Exp. Med. Biol. 2019, 1132, 89–98. [Google Scholar] [CrossRef]
  80. Nakamura, T.; Lozano, P.R.; Ikeda, Y.; Iwanaga, Y.; Hinek, A.; Minamisawa, S.; Cheng, C.F.; Kobuke, K.; Dalton, N.; Takada, Y.; et al. Fibulin-5/DANCE is essential for elastogenesis in vivo. Nature 2002, 415, 171–175. [Google Scholar] [CrossRef]
  81. Rognoni, E.; Pisco, A.O.; Hiratsuka, T.; Sipila, K.H.; Belmonte, J.M.; Mobasseri, S.A.; Philippeos, C.; Dilao, R.; Watt, F.M. Fibroblast state switching orchestrates dermal maturation and wound healing. Mol. Syst. Biol. 2018, 14, e8174. [Google Scholar] [CrossRef]
  82. Lynch, M.D.; Watt, F.M. Fibroblast heterogeneity: Implications for human disease. J. Clin. Investig. 2018, 128, 26–35. [Google Scholar] [CrossRef] [PubMed]
  83. Ganier, C.; Rognoni, E.; Goss, G.; Lynch, M.; Watt, F.M. Fibroblast Heterogeneity in Healthy and Wounded Skin. Cold Spring Harb. Perspect. Biol. 2022, 14, a041238. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, S.X.; Zhang, L.J.; Gallo, R.L. Dermal White Adipose Tissue: A Newly Recognized Layer of Skin Innate Defense. J. Investig. Dermatol. 2019, 139, 1002–1009. [Google Scholar] [CrossRef]
  85. Zhang, L.J.; Guerrero-Juarez, C.F.; Hata, T.; Bapat, S.P.; Ramos, R.; Plikus, M.V.; Gallo, R.L. Innate immunity. Dermal adipocytes protect against invasive Staphylococcus aureus skin infection. Science 2015, 347, 67–71. [Google Scholar] [CrossRef]
  86. Sillat, T.; Saat, R.; Pollanen, R.; Hukkanen, M.; Takagi, M.; Konttinen, Y.T. Basement membrane collagen type IV expression by human mesenchymal stem cells during adipogenic differentiation. J. Cell. Mol. Med. 2012, 16, 1485–1495. [Google Scholar] [CrossRef]
  87. Mariman, E.C.; Wang, P. Adipocyte extracellular matrix composition, dynamics and role in obesity. Cell. Mol. Life Sci. 2010, 67, 1277–1292. [Google Scholar] [CrossRef]
  88. Reggio, S.; Rouault, C.; Poitou, C.; Bichet, J.C.; Prifti, E.; Bouillot, J.L.; Rizkalla, S.; Lacasa, D.; Tordjman, J.; Clement, K. Increased Basement Membrane Components in Adipose Tissue during Obesity: Links with TGFbeta and Metabolic Phenotypes. J. Clin. Endocrinol. Metab. 2016, 101, 2578–2587. [Google Scholar] [CrossRef]
  89. Mori, S.; Kiuchi, S.; Ouchi, A.; Hase, T.; Murase, T. Characteristic expression of extracellular matrix in subcutaneous adipose tissue development and adipogenesis; comparison with visceral adipose tissue. Int. J. Biol. Sci. 2014, 10, 825–833. [Google Scholar] [CrossRef] [PubMed]
  90. McCulloch, L.J.; Rawling, T.J.; Sjoholm, K.; Franck, N.; Dankel, S.N.; Price, E.J.; Knight, B.; Liversedge, N.H.; Mellgren, G.; Nystrom, F.; et al. COL6A3 is regulated by leptin in human adipose tissue and reduced in obesity. Endocrinology 2015, 156, 134–146. [Google Scholar] [CrossRef] [PubMed]
  91. Bolton, K.; Segal, D.; McMillan, J.; Jowett, J.; Heilbronn, L.; Abberton, K.; Zimmet, P.; Chisholm, D.; Collier, G.; Walder, K. Decorin is a secreted protein associated with obesity and type 2 diabetes. Int. J. Obes. 2008, 32, 1113–1121. [Google Scholar] [CrossRef]
  92. Han, C.Y.; Kang, I.; Harten, I.A.; Gebe, J.A.; Chan, C.K.; Omer, M.; Alonge, K.M.; den Hartigh, L.J.; Gomes Kjerulf, D.; Goodspeed, L.; et al. Adipocyte-Derived Versican and Macrophage-Derived Biglycan Control Adipose Tissue Inflammation in Obesity. Cell Rep. 2020, 31, 107818. [Google Scholar] [CrossRef] [PubMed]
  93. Daquinag, A.C.; Gao, Z.; Fussell, C.; Sun, K.; Kolonin, M.G. Glycosaminoglycan Modification of Decorin Depends on MMP14 Activity and Regulates Collagen Assembly. Cells 2020, 9, 2646. [Google Scholar] [CrossRef]
  94. Joost, S.; Annusver, K.; Jacob, T.; Sun, X.; Dalessandri, T.; Sivan, U.; Sequeira, I.; Sandberg, R.; Kasper, M. The Molecular Anatomy of Mouse Skin during Hair Growth and Rest. Cell Stem Cell 2020, 26, 441–457.e7. [Google Scholar] [CrossRef] [PubMed]
  95. Blanpain, C.; Lowry, W.E.; Geoghegan, A.; Polak, L.; Fuchs, E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 2004, 118, 635–648. [Google Scholar] [CrossRef]
  96. Rahmani, W.; Abbasi, S.; Hagner, A.; Raharjo, E.; Kumar, R.; Hotta, A.; Magness, S.; Metzger, D.; Biernaskie, J. Hair follicle dermal stem cells regenerate the dermal sheath, repopulate the dermal papilla, and modulate hair type. Dev. Cell 2014, 31, 543–558. [Google Scholar] [CrossRef] [PubMed]
  97. Clavel, C.; Grisanti, L.; Zemla, R.; Rezza, A.; Barros, R.; Sennett, R.; Mazloom, A.R.; Chung, C.Y.; Cai, X.; Cai, C.L.; et al. Sox2 in the dermal papilla niche controls hair growth by fine-tuning BMP signaling in differentiating hair shaft progenitors. Dev. Cell 2012, 23, 981–994. [Google Scholar] [CrossRef] [PubMed]
  98. Ng, K.J.; Lim, J.; Tan, Y.N.; Quek, D.; Lim, Z.; Pantelireis, N.; Clavel, C. Sox2 in the dermal papilla regulates hair follicle pigmentation. Cell Rep. 2022, 40, 111100. [Google Scholar] [CrossRef]
  99. Greco, V.; Chen, T.; Rendl, M.; Schober, M.; Pasolli, H.A.; Stokes, N.; Dela Cruz-Racelis, J.; Fuchs, E. A two-step mechanism for stem cell activation during hair regeneration. Cell Stem Cell 2009, 4, 155–169. [Google Scholar] [CrossRef]
  100. Harshuk-Shabso, S.; Dressler, H.; Niehrs, C.; Aamar, E.; Enshell-Seijffers, D. Fgf and Wnt signaling interaction in the mesenchymal niche regulates the murine hair cycle clock. Nat. Commun. 2020, 11, 5114. [Google Scholar] [CrossRef]
  101. Tsutsui, K.; Machida, H.; Nakagawa, A.; Ahn, K.; Morita, R.; Sekiguchi, K.; Miner, J.H.; Fujiwara, H. Mapping the molecular and structural specialization of the skin basement membrane for inter-tissue interactions. Nat. Commun. 2021, 12, 2577. [Google Scholar] [CrossRef]
  102. Morris, R.J.; Liu, Y.; Marles, L.; Yang, Z.; Trempus, C.; Li, S.; Lin, J.S.; Sawicki, J.A.; Cotsarelis, G. Capturing and profiling adult hair follicle stem cells. Nat. Biotechnol. 2004, 22, 411–417. [Google Scholar] [CrossRef] [PubMed]
  103. Timpl, R.; Brown, J.C. The laminins. Matrix Biol. 1994, 14, 275–281. [Google Scholar] [CrossRef]
  104. Niemann, C.; Horsley, V. Development and homeostasis of the sebaceous gland. Semin. Cell Dev. Biol. 2012, 23, 928–936. [Google Scholar] [CrossRef] [PubMed]
  105. Geueke, A.; Niemann, C. Stem and progenitor cells in sebaceous gland development, homeostasis and pathologies. Exp. Dermatol. 2021, 30, 588–597. [Google Scholar] [CrossRef]
  106. Kobayashi, T.; Voisin, B.; Kim, D.Y.; Kennedy, E.A.; Jo, J.H.; Shih, H.Y.; Truong, A.; Doebel, T.; Sakamoto, K.; Cui, C.Y.; et al. Homeostatic Control of Sebaceous Glands by Innate Lymphoid Cells Regulates Commensal Bacteria Equilibrium. Cell 2019, 176, 982–997.e16. [Google Scholar] [CrossRef] [PubMed]
  107. Thiele, J.J.; Weber, S.U.; Packer, L. Sebaceous gland secretion is a major physiologic route of vitamin E delivery to skin. J. Investig. Dermatol. 1999, 113, 1006–1010. [Google Scholar] [CrossRef]
  108. Nishiyama, T.; Kii, I.; Kashima, T.G.; Kikuchi, Y.; Ohazama, A.; Shimazaki, M.; Fukayama, M.; Kudo, A. Delayed re-epithelialization in periostin-deficient mice during cutaneous wound healing. PLoS ONE 2011, 6, e18410. [Google Scholar] [CrossRef]
  109. Liu, S.; Leask, A. CCN2 modulates hair follicle cycling in mice. Mol. Biol. Cell 2013, 24, 3939–3944. [Google Scholar] [CrossRef]
  110. Smith, A.A.; Li, J.; Liu, B.; Hunter, D.; Pyles, M.; Gillette, M.; Dhamdhere, G.R.; Abo, A.; Oro, A.; Helms, J.A. Activating Hair Follicle Stem Cells via R-spondin2 to Stimulate Hair Growth. J. Investig. Dermatol. 2016, 136, 1549–1558. [Google Scholar] [CrossRef]
  111. Hagner, A.; Shin, W.; Sinha, S.; Alpaugh, W.; Workentine, M.; Abbasi, S.; Rahmani, W.; Agabalyan, N.; Sharma, N.; Sparks, H.; et al. Transcriptional Profiling of the Adult Hair Follicle Mesenchyme Reveals R-spondin as a Novel Regulator of Dermal Progenitor Function. iScience 2020, 23, 101019. [Google Scholar] [CrossRef] [PubMed]
  112. Ge, Y.; Miao, Y.; Gur-Cohen, S.; Gomez, N.; Yang, H.; Nikolova, M.; Polak, L.; Hu, Y.; Verma, A.; Elemento, O.; et al. The aging skin microenvironment dictates stem cell behavior. Proc. Natl. Acad. Sci. USA 2020, 117, 5339–5350. [Google Scholar] [CrossRef]
  113. Yano, K.; Brown, L.F.; Lawler, J.; Miyakawa, T.; Detmar, M. Thrombospondin-1 plays a critical role in the induction of hair follicle involution and vascular regression during the catagen phase. J. Investig. Dermatol. 2003, 120, 14–19. [Google Scholar] [CrossRef]
  114. Naldaiz-Gastesi, N.; Bahri, O.A.; Lopez de Munain, A.; McCullagh, K.J.A.; Izeta, A. The panniculus carnosus muscle: An evolutionary enigma at the intersection of distinct research fields. J. Anat. 2018, 233, 275–288. [Google Scholar] [CrossRef] [PubMed]
  115. Naldaiz-Gastesi, N.; Goicoechea, M.; Alonso-Martin, S.; Aiastui, A.; Lopez-Mayorga, M.; Garcia-Belda, P.; Lacalle, J.; San Jose, C.; Arauzo-Bravo, M.J.; Trouilh, L.; et al. Identification and Characterization of the Dermal Panniculus Carnosus Muscle Stem Cells. Stem Cell Rep. 2016, 7, 411–424. [Google Scholar] [CrossRef] [PubMed]
  116. Machado, M.J.; Watson, M.G.; Devlin, A.H.; Chaplain, M.A.; McDougall, S.R.; Mitchell, C.A. Dynamics of angiogenesis during wound healing: A coupled in vivo and in silico study. Microcirculation 2011, 18, 183–197. [Google Scholar] [CrossRef]
  117. Pratt, R.L. Hyaluronan and the Fascial Frontier. Int. J. Mol. Sci. 2021, 22, 6845. [Google Scholar] [CrossRef]
  118. Fede, C.; Pirri, C.; Fan, C.; Petrelli, L.; Guidolin, D.; De Caro, R.; Stecco, C. A Closer Look at the Cellular and Molecular Components of the Deep/Muscular Fasciae. Int. J. Mol. Sci. 2021, 22, 1411. [Google Scholar] [CrossRef]
  119. Stecco, C.; Fede, C.; Macchi, V.; Porzionato, A.; Petrelli, L.; Biz, C.; Stern, R.; De Caro, R. The fasciacytes: A new cell devoted to fascial gliding regulation. Clin. Anat. 2018, 31, 667–676. [Google Scholar] [CrossRef]
  120. McLaughlin, P.J.; Bakall, B.; Choi, J.; Liu, Z.; Sasaki, T.; Davis, E.C.; Marmorstein, A.D.; Marmorstein, L.Y. Lack of fibulin-3 causes early aging and herniation, but not macular degeneration in mice. Hum. Mol. Genet. 2007, 16, 3059–3070. [Google Scholar] [CrossRef]
  121. Driver, S.G.W.; Jackson, M.R.; Richter, K.; Tomlinson, P.; Brockway, B.; Halliday, B.J.; Markie, D.M.; Robertson, S.P.; Wade, E.M. Biallelic variants in EFEMP1 in a man with a pronounced connective tissue phenotype. Eur. J. Hum. Genet. 2020, 28, 445–452. [Google Scholar] [CrossRef]
  122. Correa-Gallegos, D.; Jiang, D.; Christ, S.; Ramesh, P.; Ye, H.; Wannemacher, J.; Kalgudde Gopal, S.; Yu, Q.; Aichler, M.; Walch, A.; et al. Patch repair of deep wounds by mobilized fascia. Nature 2019, 576, 287–292. [Google Scholar] [CrossRef]
  123. Jiang, D.; Christ, S.; Correa-Gallegos, D.; Ramesh, P.; Kalgudde Gopal, S.; Wannemacher, J.; Mayr, C.H.; Lupperger, V.; Yu, Q.; Ye, H.; et al. Injury triggers fascia fibroblast collective cell migration to drive scar formation through N-cadherin. Nat. Commun. 2020, 11, 5653. [Google Scholar] [CrossRef] [PubMed]
  124. Ichijo, R.; Maki, K.; Kabata, M.; Murata, T.; Nagasaka, A.; Ishihara, S.; Haga, H.; Honda, T.; Adachi, T.; Yamamoto, T.; et al. Vasculature atrophy causes a stiffened microenvironment that augments epidermal stem cell differentiation in aged skin. Nat. Aging 2022, 2, 592–600. [Google Scholar] [CrossRef]
  125. Koester, J.; Miroshnikova, Y.A.; Ghatak, S.; Chacon-Martinez, C.A.; Morgner, J.; Li, X.; Atanassov, I.; Altmuller, J.; Birk, D.E.; Koch, M.; et al. Niche stiffening compromises hair follicle stem cell potential during ageing by reducing bivalent promoter accessibility. Nat. Cell Biol. 2021, 23, 771–781. [Google Scholar] [CrossRef] [PubMed]
  126. Kechagia, J.Z.; Ivaska, J.; Roca-Cusachs, P. Integrins as biomechanical sensors of the microenvironment. Nat. Rev. Mol. Cell. Biol. 2019, 20, 457–473. [Google Scholar] [CrossRef]
  127. Sladitschek-Martens, H.L.; Guarnieri, A.; Brumana, G.; Zanconato, F.; Battilana, G.; Xiccato, R.L.; Panciera, T.; Forcato, M.; Bicciato, S.; Guzzardo, V.; et al. YAP/TAZ activity in stromal cells prevents ageing by controlling cGAS-STING. Nature 2022, 607, 790–798. [Google Scholar] [CrossRef] [PubMed]
  128. Totaro, A.; Castellan, M.; Battilana, G.; Zanconato, F.; Azzolin, L.; Giulitti, S.; Cordenonsi, M.; Piccolo, S. YAP/TAZ link cell mechanics to Notch signalling to control epidermal stem cell fate. Nat. Commun. 2017, 8, 15206. [Google Scholar] [CrossRef] [PubMed]
  129. Russell-Goldman, E.; Murphy, G.F. The Pathobiology of Skin Aging: New Insights into an Old Dilemma. Am. J. Pathol. 2020, 190, 1356–1369. [Google Scholar] [CrossRef]
  130. Matsumura, H.; Mohri, Y.; Binh, N.T.; Morinaga, H.; Fukuda, M.; Ito, M.; Kurata, S.; Hoeijmakers, J.; Nishimura, E.K. Hair follicle aging is driven by transepidermal elimination of stem cells via COL17A1 proteolysis. Science 2016, 351, aad4395. [Google Scholar] [CrossRef]
  131. Sole-Boldo, L.; Raddatz, G.; Schutz, S.; Mallm, J.P.; Rippe, K.; Lonsdorf, A.S.; Rodriguez-Paredes, M.; Lyko, F. Single-cell transcriptomes of the human skin reveal age-related loss of fibroblast priming. Commun. Biol. 2020, 3, 188. [Google Scholar] [CrossRef] [PubMed]
  132. Salzer, M.C.; Lafzi, A.; Berenguer-Llergo, A.; Youssif, C.; Castellanos, A.; Solanas, G.; Peixoto, F.O.; Stephan-Otto Attolini, C.; Prats, N.; Aguilera, M.; et al. Identity Noise and Adipogenic Traits Characterize Dermal Fibroblast Aging. Cell 2018, 175, 1575–1590.e22. [Google Scholar] [CrossRef] [PubMed]
  133. Deng, M.; Lin, J.; Nowsheen, S.; Liu, T.; Zhao, Y.; Villalta, P.W.; Sicard, D.; Tschumperlin, D.J.; Lee, S.; Kim, J.; et al. Extracellular matrix stiffness determines DNA repair efficiency and cellular sensitivity to genotoxic agents. Sci. Adv. 2020, 6, abb2630. [Google Scholar] [CrossRef]
  134. Sada, A.; Jacob, F.; Leung, E.; Wang, S.; White, B.S.; Shalloway, D.; Tumbar, T. Defining the cellular lineage hierarchy in the interfollicular epidermis of adult skin. Nat. Cell Biol. 2016, 18, 619–631. [Google Scholar] [CrossRef]
  135. Ghuwalewala, S.; Lee, S.A.; Jiang, K.; Baidya, J.; Chovatiya, G.; Kaur, P.; Shalloway, D.; Tumbar, T. Binary organization of epidermal basal domains highlights robustness to environmental exposure. EMBO J. 2022, 41, e110488. [Google Scholar] [CrossRef] [PubMed]
  136. Lavker, R.M.; Sun, T.T. Heterogeneity in epidermal basal keratinocytes: Morphological and functional correlations. Science 1982, 215, 1239–1241. [Google Scholar] [CrossRef] [PubMed]
  137. Koren, E.; Feldman, A.; Yusupova, M.; Kadosh, A.; Sedov, E.; Ankawa, R.; Yosefzon, Y.; Nasser, W.; Gerstberger, S.; Kimel, L.B.; et al. Thy1 marks a distinct population of slow-cycling stem cells in the mouse epidermis. Nat. Commun. 2022, 13, 4628. [Google Scholar] [CrossRef]
  138. Negri, V.A.; Watt, F.M. Understanding Human Epidermal Stem Cells at Single-Cell Resolution. J. Investig. Dermatol. 2022, 142, 2061–2067. [Google Scholar] [CrossRef]
  139. Wang, S.; Drummond, M.L.; Guerrero-Juarez, C.F.; Tarapore, E.; MacLean, A.L.; Stabell, A.R.; Wu, S.C.; Gutierrez, G.; That, B.T.; Benavente, C.A.; et al. Single cell transcriptomics of human epidermis identifies basal stem cell transition states. Nat. Commun. 2020, 11, 4239. [Google Scholar] [CrossRef]
  140. Gomez, C.; Chua, W.; Miremadi, A.; Quist, S.; Headon, D.J.; Watt, F.M. The interfollicular epidermis of adult mouse tail comprises two distinct cell lineages that are differentially regulated by Wnt, Edaradd, and Lrig1. Stem Cell Rep. 2013, 1, 19–27. [Google Scholar] [CrossRef]
  141. Baess, S.C.; Burkhart, A.K.; Cappello, S.; Graband, A.; Sere, K.; Zenke, M.; Niemann, C.; Iden, S. Lrig1- and Wnt-dependent niches dictate segregation of resident immune cells and melanocytes in murine tail epidermis. Development 2022, 149, dev200154. [Google Scholar] [CrossRef] [PubMed]
  142. Wang, Y.; Kitahata, H.; Kosumi, H.; Watanabe, M.; Fujimura, Y.; Takashima, S.; Osada, S.I.; Hirose, T.; Nishie, W.; Nagayama, M.; et al. Collagen XVII deficiency alters epidermal patterning. Lab. Investig. 2022, 102, 581–588. [Google Scholar] [CrossRef]
  143. Natsuga, K.; Watanabe, M.; Nishie, W.; Shimizu, H. Life before and beyond blistering: The role of collagen XVII in epidermal physiology. Exp. Dermatol. 2019, 28, 1135–1141. [Google Scholar] [CrossRef] [PubMed]
  144. Watanabe, M.; Natsuga, K.; Nishie, W.; Kobayashi, Y.; Donati, G.; Suzuki, S.; Fujimura, Y.; Tsukiyama, T.; Ujiie, H.; Shinkuma, S.; et al. Type XVII collagen coordinates proliferation in the interfollicular epidermis. Elife 2017, 6, e26635. [Google Scholar] [CrossRef] [PubMed]
  145. Dekoninck, S.; Blanpain, C. Stem cell dynamics, migration and plasticity during wound healing. Nat. Cell Biol. 2019, 21, 18–24. [Google Scholar] [CrossRef]
  146. Rousselle, P.; Montmasson, M.; Garnier, C. Extracellular matrix contribution to skin wound re-epithelialization. Matrix Biol. 2019, 75–76, 12–26. [Google Scholar] [CrossRef]
  147. Kim, K.H.; Won, J.H.; Cheng, N.; Lau, L.F. The matricellular protein CCN1 in tissue injury repair. J. Cell Commun. Signal. 2018, 12, 273–279. [Google Scholar] [CrossRef]
  148. Jun, J.I.; Lau, L.F. Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nat. Rev. Drug Discov. 2011, 10, 945–963. [Google Scholar] [CrossRef] [PubMed]
  149. Jun, J.I.; Lau, L.F. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 2010, 12, 676–685. [Google Scholar] [CrossRef]
  150. Jun, J.I.; Kim, K.H.; Lau, L.F. The matricellular protein CCN1 mediates neutrophil efferocytosis in cutaneous wound healing. Nat. Commun. 2015, 6, 7386. [Google Scholar] [CrossRef]
  151. Du, H.; Zhou, Y.; Suo, Y.; Liang, X.; Chai, B.; Duan, R.; Huang, X.; Li, Q. CCN1 accelerates re-epithelialization by promoting keratinocyte migration and proliferation during cutaneous wound healing. Biochem. Biophys. Res. Commun. 2018, 505, 966–972. [Google Scholar] [CrossRef]
  152. Zhou, Y.; Li, H.; Liang, X.; Du, H.; Suo, Y.; Chen, H.; Liu, W.; Duan, R.; Huang, X.; Li, Q. The CCN1 (CYR61) protein promotes skin growth by enhancing epithelial-mesenchymal transition during skin expansion. J. Cell. Mol. Med. 2020, 24, 1460–1473. [Google Scholar] [CrossRef] [PubMed]
  153. Agah, A.; Kyriakides, T.R.; Lawler, J.; Bornstein, P. The lack of thrombospondin-1 (TSP1) dictates the course of wound healing in double-TSP1/TSP2-null mice. Am. J. Pathol. 2002, 161, 831–839. [Google Scholar] [CrossRef] [PubMed]
  154. Streit, M.; Velasco, P.; Riccardi, L.; Spencer, L.; Brown, L.F.; Janes, L.; Lange-Asschenfeldt, B.; Yano, K.; Hawighorst, T.; Iruela-Arispe, L.; et al. Thrombospondin-1 suppresses wound healing and granulation tissue formation in the skin of transgenic mice. EMBO J. 2000, 19, 3272–3282. [Google Scholar] [CrossRef]
  155. Haensel, D.; Jin, S.; Sun, P.; Cinco, R.; Dragan, M.; Nguyen, Q.; Cang, Z.; Gong, Y.; Vu, R.; MacLean, A.L.; et al. Defining Epidermal Basal Cell States during Skin Homeostasis and Wound Healing Using Single-Cell Transcriptomics. Cell Rep. 2020, 30, 3932–3947.e6. [Google Scholar] [CrossRef]
  156. Kyriakides, T.R.; Tam, J.W.; Bornstein, P. Accelerated wound healing in mice with a disruption of the thrombospondin 2 gene. J. Investig. Dermatol. 1999, 113, 782–787. [Google Scholar] [CrossRef]
  157. Klaas, M.; Maemets-Allas, K.; Heinmae, E.; Lagus, H.; Cardenas-Leon, C.G.; Arak, T.; Eller, M.; Kingo, K.; Kankuri, E.; Jaks, V. Thrombospondin-4 Is a Soluble Dermal Inflammatory Signal That Selectively Promotes Fibroblast Migration and Keratinocyte Proliferation for Skin Regeneration and Wound Healing. Front. Cell Dev. Biol. 2021, 9, 745637. [Google Scholar] [CrossRef]
  158. Nikoloudaki, G.; Creber, K.; Hamilton, D.W. Wound healing and fibrosis: A contrasting role for periostin in skin and the oral mucosa. Am. J. Physiol. Cell Physiol. 2020, 318, C1065–C1077. [Google Scholar] [CrossRef] [PubMed]
  159. Elliott, C.G.; Wang, J.; Guo, X.; Xu, S.W.; Eastwood, M.; Guan, J.; Leask, A.; Conway, S.J.; Hamilton, D.W. Periostin modulates myofibroblast differentiation during full-thickness cutaneous wound repair. J. Cell Sci. 2012, 125, 121–132. [Google Scholar] [CrossRef]
  160. Parma, P.; Radi, O.; Vidal, V.; Chaboissier, M.C.; Dellambra, E.; Valentini, S.; Guerra, L.; Schedl, A.; Camerino, G. R-spondin1 is essential in sex determination, skin differentiation and malignancy. Nat. Genet. 2006, 38, 1304–1309. [Google Scholar] [CrossRef]
  161. Kuppan, P.; Vasanthan, K.S.; Sundaramurthi, D.; Krishnan, U.M.; Sethuraman, S. Development of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) fibers for skin tissue engineering: Effects of topography, mechanical, and chemical stimuli. Biomacromolecules 2011, 12, 3156–3165. [Google Scholar] [CrossRef]
  162. Li, J.; Ma, J.; Zhang, Q.; Gong, H.; Gao, D.; Wang, Y.; Li, B.; Li, X.; Zheng, H.; Wu, Z.; et al. Spatially resolved proteomic map shows that extracellular matrix regulates epidermal growth. Nat. Commun. 2022, 13, 4012. [Google Scholar] [CrossRef] [PubMed]
  163. Rogers, N.M.; Roberts, D.D.; Isenberg, J.S. Age-associated induction of cell membrane CD47 limits basal and temperature-induced changes in cutaneous blood flow. Ann. Surg. 2013, 258, 184–191. [Google Scholar] [CrossRef] [PubMed]
  164. Waldera Lupa, D.M.; Kalfalah, F.; Safferling, K.; Boukamp, P.; Poschmann, G.; Volpi, E.; Gotz-Rosch, C.; Bernerd, F.; Haag, L.; Huebenthal, U.; et al. Characterization of Skin Aging-Associated Secreted Proteins (SAASP) Produced by Dermal Fibroblasts Isolated from Intrinsically Aged Human Skin. J. Investig. Dermatol. 2015, 135, 1954–1968. [Google Scholar] [CrossRef]
  165. Badarinath, K.; Dam, B.; Kataria, S.; Zirmire, R.K.; Dey, R.; Kansagara, G.; Ajnabi, J.; Hegde, A.; Singh, R.; Masudi, T.; et al. Snail maintains the stem/progenitor state of skin epithelial cells and carcinomas through the autocrine effect of matricellular protein Mindin. Cell Rep. 2022, 40, 111390. [Google Scholar] [CrossRef]
  166. De Craene, B.; Denecker, G.; Vermassen, P.; Taminau, J.; Mauch, C.; Derore, A.; Jonkers, J.; Fuchs, E.; Berx, G. Epidermal Snail expression drives skin cancer initiation and progression through enhanced cytoprotection, epidermal stem/progenitor cell expansion and enhanced metastatic potential. Cell Death Differ. 2014, 21, 310–320. [Google Scholar] [CrossRef]
  167. Rana, I.; Kataria, S.; Tan, T.L.; Hajam, E.Y.; Kashyap, D.K.; Saha, D.; Ajnabi, J.; Paul, S.; Jayappa, S.; Ananthan, A.; et al. Mindin (SPON2) Is Essential for Cutaneous Fibrogenesis in a Mouse Model of Systemic Sclerosis. J. Investig. Dermatol. 2022, 14, 699–710.e10. [Google Scholar] [CrossRef]
  168. Nakasaki, M.; Hwang, Y.; Xie, Y.; Kataria, S.; Gund, R.; Hajam, E.Y.; Samuel, R.; George, R.; Danda, D.; Mj, P.; et al. The matrix protein Fibulin-5 is at the interface of tissue stiffness and inflammation in fibrosis. Nat. Commun. 2015, 6, 8574. [Google Scholar] [CrossRef]
  169. Quan, T.; Xiang, Y.; Liu, Y.; Qin, Z.; Yang, Y.; Bou-Gharios, G.; Voorhees, J.J.; Dlugosz, A.A.; Fisher, G.J. Dermal Fibroblast CCN1 Expression in Mice Recapitulates Human Skin Dermal Aging. J. Investig. Dermatol. 2021, 141, 1007–1016. [Google Scholar] [CrossRef]
  170. Quan, T.; Shao, Y.; He, T.; Voorhees, J.J.; Fisher, G.J. Reduced expression of connective tissue growth factor (CTGF/CCN2) mediates collagen loss in chronologically aged human skin. J. Investig. Dermatol. 2010, 130, 415–424. [Google Scholar] [CrossRef] [PubMed]
  171. Qin, Z.; He, T.; Guo, C.; Quan, T. Age-Related Downregulation of CCN2 Is Regulated by Cell Size in a YAP/TAZ-Dependent Manner in Human Dermal Fibroblasts: Impact on Dermal Aging. JID Innov. 2022, 2, 100111. [Google Scholar] [CrossRef]
  172. Chai, J.; Norng, M.; Modak, C.; Reavis, K.M.; Mouazzen, W.; Pham, J. CCN1 induces a reversible epithelial-mesenchymal transition in gastric epithelial cells. Lab. Investig. 2010, 90, 1140–1151. [Google Scholar] [CrossRef]
  173. Nunomura, S.; Nanri, Y.; Ogawa, M.; Arima, K.; Mitamura, Y.; Yoshihara, T.; Hasuwa, H.; Conway, S.J.; Izuhara, K. Constitutive overexpression of periostin delays wound healing in mouse skin. Wound Repair. Regen. 2018, 26, 6–15. [Google Scholar] [CrossRef] [PubMed]
  174. Ge, Y.; Gomez, N.C.; Adam, R.C.; Nikolova, M.; Yang, H.; Verma, A.; Lu, C.P.; Polak, L.; Yuan, S.; Elemento, O.; et al. Stem Cell Lineage Infidelity Drives Wound Repair and Cancer. Cell 2017, 169, 636–650.e14. [Google Scholar] [CrossRef] [PubMed]
  175. Gonzales, K.A.U.; Polak, L.; Matos, I.; Tierney, M.T.; Gola, A.; Wong, E.; Infarinato, N.R.; Nikolova, M.; Luo, S.; Liu, S.; et al. Stem cells expand potency and alter tissue fitness by accumulating diverse epigenetic memories. Science 2021, 374, eabh2444. [Google Scholar] [CrossRef] [PubMed]
  176. Ito, M.; Liu, Y.; Yang, Z.; Nguyen, J.; Liang, F.; Morris, R.J.; Cotsarelis, G. Stem cells in the hair follicle bulge contribute to wound repair but not to homeostasis of the epidermis. Nat. Med. 2005, 11, 1351–1354. [Google Scholar] [CrossRef]
  177. Ito, M.; Yang, Z.; Andl, T.; Cui, C.; Kim, N.; Millar, S.E.; Cotsarelis, G. Wnt-dependent de novo hair follicle regeneration in adult mouse skin after wounding. Nature 2007, 447, 316–320. [Google Scholar] [CrossRef]
  178. Ter Steege, E.J.; Bakker, E.R.M. The role of R-spondin proteins in cancer biology. Oncogene 2021, 40, 6469–6478. [Google Scholar] [CrossRef]
  179. de Lau, W.B.; Snel, B.; Clevers, H.C. The R-spondin protein family. Genome Biol. 2012, 13, 242. [Google Scholar] [CrossRef]
  180. Keyes, B.E.; Liu, S.; Asare, A.; Naik, S.; Levorse, J.; Polak, L.; Lu, C.P.; Nikolova, M.; Pasolli, H.A.; Fuchs, E. Impaired Epidermal to Dendritic T Cell Signaling Slows Wound Repair in Aged Skin. Cell 2016, 167, 1323–1338.e14. [Google Scholar] [CrossRef]
  181. Liu, N.; Matsumura, H.; Kato, T.; Ichinose, S.; Takada, A.; Namiki, T.; Asakawa, K.; Morinaga, H.; Mohri, Y.; De Arcangelis, A.; et al. Stem cell competition orchestrates skin homeostasis and ageing. Nature 2019, 568, 344–350. [Google Scholar] [CrossRef] [PubMed]
  182. Roig-Rosello, E.; Rousselle, P. The Human Epidermal Basement Membrane: A Shaped and Cell Instructive Platform That Aging Slowly Alters. Biomolecules 2020, 10, 1607. [Google Scholar] [CrossRef] [PubMed]
  183. Doles, J.; Storer, M.; Cozzuto, L.; Roma, G.; Keyes, W.M. Age-associated inflammation inhibits epidermal stem cell function. Genes Dev. 2012, 26, 2144–2153. [Google Scholar] [CrossRef]
  184. Rube, C.E.; Baumert, C.; Schuler, N.; Isermann, A.; Schmal, Z.; Glanemann, M.; Mann, C.; Scherthan, H. Human skin aging is associated with increased expression of the histone variant H2A.J in the epidermis. NPJ Aging Mech. Dis. 2021, 7, 7. [Google Scholar] [CrossRef]
  185. Sola, P.; Mereu, E.; Bonjoch, J.; Casado-Pelaez, M.; Prats, N.; Aguilera, M.; Reina, O.; Blanco, E.; Esteller, M.; Di Croce, L.; et al. Targeting lymphoid-derived IL-17 signaling to delay skin aging. Nat. Aging 2023, 3, 688–704. [Google Scholar] [CrossRef]
  186. Changarathil, G.; Ramirez, K.; Isoda, H.; Sada, A.; Yanagisawa, H. Wild-type and SAMP8 mice show age-dependent changes in distinct stem cell compartments of the interfollicular epidermis. PLoS ONE 2019, 14, e0215908. [Google Scholar] [CrossRef] [PubMed]
  187. Giangreco, A.; Qin, M.; Pintar, J.E.; Watt, F.M. Epidermal stem cells are retained in vivo throughout skin aging. Aging Cell 2008, 7, 250–259. [Google Scholar] [CrossRef] [PubMed]
  188. Makrantonaki, E.; Brink, T.C.; Zampeli, V.; Elewa, R.M.; Mlody, B.; Hossini, A.M.; Hermes, B.; Krause, U.; Knolle, J.; Abdallah, M.; et al. Identification of biomarkers of human skin ageing in both genders. Wnt signalling—A label of skin ageing? PLoS ONE 2012, 7, e50393. [Google Scholar] [CrossRef]
  189. Kaur, A.; Webster, M.R.; Weeraratna, A.T. In the Wnt-er of life: Wnt signalling in melanoma and ageing. Br. J. Cancer 2016, 115, 1273–1279. [Google Scholar] [CrossRef]
  190. Langton, A.K.; Graham, H.K.; McConnell, J.C.; Sherratt, M.J.; Griffiths, C.E.M.; Watson, R.E.B. Organization of the dermal matrix impacts the biomechanical properties of skin. Br. J. Dermatol. 2017, 177, 818–827. [Google Scholar] [CrossRef]
  191. Giangreco, A.; Goldie, S.J.; Failla, V.; Saintigny, G.; Watt, F.M. Human skin aging is associated with reduced expression of the stem cell markers beta1 integrin and MCSP. J. Investig. Dermatol. 2010, 130, 604–608. [Google Scholar] [CrossRef] [PubMed]
  192. Mobasseri, S.A.; Zijl, S.; Salameti, V.; Walko, G.; Stannard, A.; Garcia-Manyes, S.; Watt, F.M. Patterning of human epidermal stem cells on undulating elastomer substrates reflects differences in cell stiffness. Acta Biomater. 2019, 87, 256–264. [Google Scholar] [CrossRef]
  193. Halfter, W.; Oertle, P.; Monnier, C.A.; Camenzind, L.; Reyes-Lua, M.; Hu, H.; Candiello, J.; Labilloy, A.; Balasubramani, M.; Henrich, P.B.; et al. New concepts in basement membrane biology. FEBS J. 2015, 282, 4466–4479. [Google Scholar] [CrossRef] [PubMed]
  194. Vazquez, F.; Palacios, S.; Aleman, N.; Guerrero, F. Changes of the basement membrane and type IV collagen in human skin during aging. Maturitas 1996, 25, 209–215. [Google Scholar] [CrossRef]
  195. Feru, J.; Delobbe, E.; Ramont, L.; Brassart, B.; Terryn, C.; Dupont-Deshorgue, A.; Garbar, C.; Monboisse, J.C.; Maquart, F.X.; Brassart-Pasco, S. Aging decreases collagen IV expression in vivo in the dermo-epidermal junction and in vitro in dermal fibroblasts: Possible involvement of TGF-beta1. Eur. J. Dermatol. 2016, 26, 350–360. [Google Scholar] [CrossRef]
  196. Tohgasaki, T.; Nishizawa, S.; Kondo, S.; Ishiwatari, S.; Sakurai, T. Long Hanging Structure of Collagen VII Connects the Elastic Fibers and the Basement Membrane in Young Skin Tissue. J. Histochem. Cytochem. 2022, 70, 751–757. [Google Scholar] [CrossRef] [PubMed]
  197. Yamada, T.; Hasegawa, S.; Miyachi, K.; Date, Y.; Inoue, Y.; Yagami, A.; Arima, M.; Iwata, Y.; Yamamoto, N.; Nakata, S.; et al. Laminin-332 regulates differentiation of human interfollicular epidermal stem cells. Mech. Ageing Dev. 2018, 171, 37–46. [Google Scholar] [CrossRef]
  198. Yanagisawa, H.; Davis, E.C. Unraveling the mechanism of elastic fiber assembly: The roles of short fibulins. Int. J. Biochem. Cell Biol. 2010, 42, 1084–1093. [Google Scholar] [CrossRef]
  199. Strachan, L.R.; Scalapino, K.J.; Lawrence, H.J.; Ghadially, R. Rapid adhesion to collagen isolates murine keratinocytes with limited long-term repopulating ability in vivo despite high clonogenicity in vitro. Stem Cells 2008, 26, 235–243. [Google Scholar] [CrossRef] [PubMed]
  200. Jones, P.H.; Watt, F.M. Separation of human epidermal stem cells from transit amplifying cells on the basis of differences in integrin function and expression. Cell 1993, 73, 713–724. [Google Scholar] [CrossRef]
  201. de Vega, S.; Iwamoto, T.; Nakamura, T.; Hozumi, K.; McKnight, D.A.; Fisher, L.W.; Fukumoto, S.; Yamada, Y. TM14 is a new member of the fibulin family (fibulin-7) that interacts with extracellular matrix molecules and is active for cell binding. J. Biol. Chem. 2007, 282, 30878–30888. [Google Scholar] [CrossRef]
  202. Adams, J.C.; Watt, F.M. Changes in keratinocyte adhesion during terminal differentiation: Reduction in fibronectin binding precedes alpha 5 beta 1 integrin loss from the cell surface. Cell 1990, 63, 425–435. [Google Scholar] [CrossRef]
  203. Adams, J.C.; Watt, F.M. Fibronectin inhibits the terminal differentiation of human keratinocytes. Nature 1989, 340, 307–309. [Google Scholar] [CrossRef] [PubMed]
  204. Nicholson, L.J.; Watt, F.M. Decreased expression of fibronectin and the alpha 5 beta 1 integrin during terminal differentiation of human keratinocytes. J. Cell Sci. 1991, 98 Pt 2, 225–232. [Google Scholar] [CrossRef]
  205. Kaur, S.; Soto-Pantoja, D.R.; Stein, E.V.; Liu, C.Y.; Elkahloun, A.G.; Pendrak, M.L.; Nicolae, A.; Singh, S.P.; Nie, Z.Q.; Levens, D.; et al. Thrombospondin-1 Signaling through CD47 Inhibits Self-renewal by Regulating c-Myc and Other Stem Cell Transcription Factors. Sci. Rep. 2013, 3, 1673. [Google Scholar] [CrossRef] [PubMed]
  206. Lee, J.H.; Bhang, D.H.; Beede, A.; Huang, T.L.; Stripp, B.R.; Bloch, K.D.; Wagers, A.J.; Tseng, Y.H.; Ryeom, S.; Kim, C.F. Lung Stem Cell Differentiation in Mice Directed by Endothelial Cells via a BMP4-NFATc1-Thrombospondin-1 Axis. Cell 2014, 156, 440–455. [Google Scholar] [CrossRef] [PubMed]
  207. Isenberg, J.S.; Roberts, D.D. Thrombospondin-1 in maladaptive aging responses: A concept whose time has come. Am. J. Physiol.-Cell Physiol. 2020, 319, C45–C63. [Google Scholar] [CrossRef]
  208. Meijles, D.N.; Sahoo, S.; Al Ghouleh, I.; Amaral, J.H.; Bienes-Martinez, R.; Knupp, H.E.; Attaran, S.; Sembrat, J.C.; Nouraie, S.M.; Rojas, M.M.; et al. The matricellular protein TSP1 promotes human and mouse endothelial cell senescence through CD47 and Nox1. Sci. Signal. 2017, 10, eaaj1784. [Google Scholar] [CrossRef]
  209. Wang, H.; Unternaehrer, J.J. Epithelial-mesenchymal Transition and Cancer Stem Cells: At the Crossroads of Differentiation and Dedifferentiation. Dev. Dyn. 2019, 248, 10–20. [Google Scholar] [CrossRef]
  210. Du, F.; Nakamura, Y.; Tan, T.L.; Lee, P.; Lee, R.; Yu, B.; Jamora, C. Expression of snail in epidermal keratinocytes promotes cutaneous inflammation and hyperplasia conducive to tumor formation. Cancer Res. 2010, 70, 10080–10089. [Google Scholar] [CrossRef]
  211. Wei, H.Y.; Du, S.S.; Parksong, J.; Pasolli, H.A.; Matte-Martone, C.; Regot, S.; Gonzalez, L.E.; Xin, T.C.; Greco, V. Organ function is preserved despite reorganization of niche architecture in the hair follicle. Cell Stem Cell 2023, 30, 962–972.e6. [Google Scholar] [CrossRef]
  212. Li, K.N.; Jain, P.; He, C.H.; Eun, F.C.; Kang, S.; Tumbar, T. Skin vasculature and hair follicle cross-talking associated with stem cell activation and tissue homeostasis. Elife 2019, 8, e45977. [Google Scholar] [CrossRef] [PubMed]
  213. Xiao, Y.; Woo, W.M.; Nagao, K.; Li, W.; Terunuma, A.; Mukouyama, Y.S.; Oro, A.E.; Vogel, J.C.; Brownell, I. Perivascular hair follicle stem cells associate with a venule annulus. J. Investig. Dermatol. 2013, 133, 2324–2331. [Google Scholar] [CrossRef]
  214. Ewald, C.Y. The Matrisome during Aging and Longevity: A Systems-Level Approach toward Defining Matreotypes Promoting Healthy Aging. Gerontology 2020, 66, 266–274. [Google Scholar] [CrossRef]
  215. Frances, D.; Niemann, C. Stem cell dynamics in sebaceous gland morphogenesis in mouse skin. Dev. Biol. 2012, 363, 138–146. [Google Scholar] [CrossRef]
  216. Nowak, J.A.; Polak, L.; Pasolli, H.A.; Fuchs, E. Hair follicle stem cells are specified and function in early skin morphogenesis. Cell Stem Cell 2008, 3, 33–43. [Google Scholar] [CrossRef] [PubMed]
  217. Zouboulis, C.C.; Boschnakow, A. Chronological ageing and photoageing of the human sebaceous gland. Clin. Exp. Dermatol. 2001, 26, 600–607. [Google Scholar] [CrossRef] [PubMed]
  218. Hou, X.; Wei, Z.; Zouboulis, C.C.; Ju, Q. Aging in the sebaceous gland. Front. Cell Dev. Biol. 2022, 10, 909694. [Google Scholar] [CrossRef]
  219. Abreu, J.G.; Ketpura, N.I.; Reversade, B.; De Robertis, E.M. Connective-tissue growth factor (CTGF) modulates cell signalling by BMP and TGF-beta. Nat. Cell Biol. 2002, 4, 599–604. [Google Scholar] [CrossRef]
  220. Sakamoto, K.; Yamaguchi, S.; Ando, R.; Miyawaki, A.; Kabasawa, Y.; Takagi, M.; Li, C.L.; Perbal, B.; Katsube, K. The nephroblastoma overexpressed gene (NOV/ccn3) protein associates with Notch1 extracellular domain and inhibits myoblast differentiation via Notch signaling pathway. J. Biol. Chem. 2002, 277, 29399–29405. [Google Scholar] [CrossRef]
  221. Mercurio, S.; Latinkic, B.; Itasaki, N.; Krumlauf, R.; Smith, J.C. Connective-tissue growth factor modulates WNT signalling and interacts with the WNT receptor complex. Development 2004, 131, 2137–2147. [Google Scholar] [CrossRef] [PubMed]
  222. Shwartz, Y.; Gonzalez-Celeiro, M.; Chen, C.L.; Pasolli, H.A.; Sheu, S.H.; Fan, S.M.; Shamsi, F.; Assaad, S.; Lin, E.T.; Zhang, B.; et al. Cell Types Promoting Goosebumps Form a Niche to Regulate Hair Follicle Stem Cells. Cell 2020, 182, 578–593.e19. [Google Scholar] [CrossRef]
  223. Zhang, B.; Ma, S.; Rachmin, I.; He, M.; Baral, P.; Choi, S.; Goncalves, W.A.; Shwartz, Y.; Fast, E.M.; Su, Y.; et al. Hyperactivation of sympathetic nerves drives depletion of melanocyte stem cells. Nature 2020, 577, 676–681. [Google Scholar] [CrossRef] [PubMed]
  224. Pena-Jimenez, D.; Fontenete, S.; Megias, D.; Fustero-Torre, C.; Grana-Castro, O.; Castellana, D.; Loewe, R.; Perez-Moreno, M. Lymphatic vessels interact dynamically with the hair follicle stem cell niche during skin regeneration in vivo. EMBO J. 2019, 38, e101688. [Google Scholar] [CrossRef]
  225. Gur-Cohen, S.; Yang, H.; Baksh, S.C.; Miao, Y.; Levorse, J.; Kataru, R.P.; Liu, X.; de la Cruz-Racelis, J.; Mehrara, B.J.; Fuchs, E. Stem cell-driven lymphatic remodeling coordinates tissue regeneration. Science 2019, 366, 1218–1225. [Google Scholar] [CrossRef] [PubMed]
  226. Vu, R.; Jin, S.; Sun, P.; Haensel, D.; Nguyen, Q.H.; Dragan, M.; Kessenbrock, K.; Nie, Q.; Dai, X. Wound healing in aged skin exhibits systems-level alterations in cellular composition and cell-cell communication. Cell Rep. 2022, 40, 111155. [Google Scholar] [CrossRef] [PubMed]
  227. Adam, R.C.; Yang, H.; Ge, Y.; Infarinato, N.R.; Gur-Cohen, S.; Miao, Y.; Wang, P.; Zhao, Y.; Lu, C.P.; Kim, J.E.; et al. NFI transcription factors provide chromatin access to maintain stem cell identity while preventing unintended lineage fate choices. Nat. Cell Biol. 2020, 22, 640–650. [Google Scholar] [CrossRef]
  228. Naik, S.; Larsen, S.B.; Gomez, N.C.; Alaverdyan, K.; Sendoel, A.; Yuan, S.; Polak, L.; Kulukian, A.; Chai, S.; Fuchs, E. Inflammatory memory sensitizes skin epithelial stem cells to tissue damage. Nature 2017, 550, 475–480. [Google Scholar] [CrossRef]
  229. Donati, G.; Rognoni, E.; Hiratsuka, T.; Liakath-Ali, K.; Hoste, E.; Kar, G.; Kayikci, M.; Russell, R.; Kretzschmar, K.; Mulder, K.W.; et al. Wounding induces dedifferentiation of epidermal Gata6+ cells and acquisition of stem cell properties. Nat. Cell Biol. 2017, 19, 603–613. [Google Scholar] [CrossRef]
  230. Donati, G.; Watt, F.M. Stem cell heterogeneity and plasticity in epithelia. Cell Stem Cell 2015, 16, 465–476. [Google Scholar] [CrossRef]
  231. Fujimura, Y.; Watanabe, M.; Ohno, K.; Kobayashi, Y.; Takashima, S.; Nakamura, H.; Kosumi, H.; Wang, Y.; Mai, Y.; Lauria, A.; et al. Hair follicle stem cell progeny heal blisters while pausing skin development. EMBO Rep. 2021, 22, e50882. [Google Scholar] [CrossRef]
  232. Oak, A.S.W.; Cotsarelis, G. Wound-Induced Hair Neogenesis: A Portal to the Development of New Therapies for Hair Loss and Wound Regeneration. Cold Spring Harb. Perspect. Biol. 2023, 15, a041239. [Google Scholar] [CrossRef] [PubMed]
  233. Guerrero-Juarez, C.F.; Dedhia, P.H.; Jin, S.; Ruiz-Vega, R.; Ma, D.; Liu, Y.; Yamaga, K.; Shestova, O.; Gay, D.L.; Yang, Z.; et al. Single-cell analysis reveals fibroblast heterogeneity and myeloid-derived adipocyte progenitors in murine skin wounds. Nat. Commun. 2019, 10, 650. [Google Scholar] [CrossRef] [PubMed]
  234. Mahmoudi, S.; Mancini, E.; Xu, L.; Moore, A.; Jahanbani, F.; Hebestreit, K.; Srinivasan, R.; Li, X.; Devarajan, K.; Prelot, L.; et al. Heterogeneity in old fibroblasts is linked to variability in reprogramming and wound healing. Nature 2019, 574, 553–558. [Google Scholar] [CrossRef]
  235. Zomer, H.D.; Trentin, A.G. Skin wound healing in humans and mice: Challenges in translational research. J. Dermatol. Sci. 2018, 90, 3–12. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic presentation of mouse and human dorsal skin. Abbreviations: DP, dermal papilla; DS, dermal sheath; pc, panniculus carnosus muscle; s.c. fat, subcutaneous fat; APM, arrector pili muscle; GAGs, glycosaminoglycans. The sweat gland is not shown in the image. Matricellular protein constituents and their roles are summarized in Table 1. Created with BioRender.com (accessed on 13 February 2023).
Figure 1. Schematic presentation of mouse and human dorsal skin. Abbreviations: DP, dermal papilla; DS, dermal sheath; pc, panniculus carnosus muscle; s.c. fat, subcutaneous fat; APM, arrector pili muscle; GAGs, glycosaminoglycans. The sweat gland is not shown in the image. Matricellular protein constituents and their roles are summarized in Table 1. Created with BioRender.com (accessed on 13 February 2023).
Ijms 24 14274 g001
Figure 3. Epidermal stem cell heterogeneity in human and mouse skin. Stem cells are illustrated in dark or light green (dark green for slow-cycling stem cells and light green for fast-cycling stem cells) with some of their respective gene expression markers in boxed regions [134,135]. Fibulin 7 matricellular protein is localized in the basement membrane (BM) to support epidermal stem cell heterogeneity during skin aging. Other matricellular proteins regulating the BM, epidermal stem cells homeostasis, and the re-epithelialization process in injury-induced regeneration are summarized in Table 1 and Table 3. Created with BioRender.com (accessed on 22 July 2023).
Figure 3. Epidermal stem cell heterogeneity in human and mouse skin. Stem cells are illustrated in dark or light green (dark green for slow-cycling stem cells and light green for fast-cycling stem cells) with some of their respective gene expression markers in boxed regions [134,135]. Fibulin 7 matricellular protein is localized in the basement membrane (BM) to support epidermal stem cell heterogeneity during skin aging. Other matricellular proteins regulating the BM, epidermal stem cells homeostasis, and the re-epithelialization process in injury-induced regeneration are summarized in Table 1 and Table 3. Created with BioRender.com (accessed on 22 July 2023).
Ijms 24 14274 g003
Table 1. Matricellular proteins in different compartments of the skin.
Table 1. Matricellular proteins in different compartments of the skin.
Skin
Compartment
Matricellular
Protein
FunctionReferences & Study Model
EpidermisFibulin 2binds to laminin 332 for BM Stability[17] (mouse)
Fibulin 7BM localization, binds to collagen IV in vitro[18] (mouse)
SPARCbinds to collagen IV in vitro and induces expression of collagen IV and VII[19] (human skin & 3D culture), [20] (molecular structure), [21] (summary of works in mouse)
Hemicentin 1
(Fibulin 6)
BM stability[22] (mouse), [23] (mouse & zebrafish)
Thrombospondin 1inhibits angiogenesis[24] (human skin), [25] (summary of works in mouse), [26] (mouse xenograft)
CCN3promotes DDR1 binding to collagen IV and inhibits melanocytes proliferation in UV-mediated stress[27] (human skin reconstructs), [28] (summary of works in human skin & cell culture)
DermisTenascin Cgrowth factors sequestration,
regulates cell proliferation
[29] (summary), [30] (mouse NR6 cells), [31] (human skin), [32] (mouse), [33,34] (molecular interactions)
Fibronectin *growth factor sequestration,
supports cell attachment and migration
[35] (summary), [36] (mouse)
Fibrillin 1 *supports elastic fiber formation and homeostasis [37] (human & bovine tissue)
Fibulin 2[38] (mouse), [37] (human & bovine tissue)
[39] (summary in human & mouse), [40,41] (human genetic mutations), [42] (mouse)
Fibulin 4
Fibulin 5
Periostinmodulates collagen structure and stability, regulates keratinocyte proliferation [43] (human skin), [44] (mouse skin 3D culture)
Thrombospondin 1dermal vascularization balance,
interacts with collagen I
[25] (summary), [26] (mouse xenograft), [45] (in vivo mouse & human dermal fibroblasts)
Thrombospondin 2collagen structural arrangement and abundance[46] (mouse)
SPARC[47] (mouse)
Dermal adipose tissueSPARCwound healing, modulates adipogenesis[48,49] (mouse)
CCDC80/URB/DRO1modulates adipogenesis[50] (mouse tissue and 3T3-L1 cells), [51] (human & mouse tissue), [52] (mouse)
Hair follicleFibulin 1BM homeostasis[53] (mouse), [54] [summary]
Periostintendon-related genes, may provide stability for arrector pili muscle and hair follicle connection[54,55] (mouse), [56,57] (rat)
Tenascin C
SPARC
Sebaceous glandsFibronectin*regulates cell differentiation[58] (mouse)
Tenascin Cgrowth factor sequestration[59] (human & mouse eyelids)
Panniculus
carnosus muscle
Fibulin 4 Panniculus carnosus muscle homeostasis[60] (mouse)
* Fibrillin 1 and Fibronectin are not classified as matricellular proteins, but they bear non-structural functions too via binding to matricellular proteins or regulating growth factors bio-availability.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Raja, E.; Clarin, M.T.R.D.C.; Yanagisawa, H. Matricellular Proteins in the Homeostasis, Regeneration, and Aging of Skin. Int. J. Mol. Sci. 2023, 24, 14274. https://doi.org/10.3390/ijms241814274

AMA Style

Raja E, Clarin MTRDC, Yanagisawa H. Matricellular Proteins in the Homeostasis, Regeneration, and Aging of Skin. International Journal of Molecular Sciences. 2023; 24(18):14274. https://doi.org/10.3390/ijms241814274

Chicago/Turabian Style

Raja, Erna, Maria Thea Rane Dela Cruz Clarin, and Hiromi Yanagisawa. 2023. "Matricellular Proteins in the Homeostasis, Regeneration, and Aging of Skin" International Journal of Molecular Sciences 24, no. 18: 14274. https://doi.org/10.3390/ijms241814274

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop