TISSUE-SPECIFIC STEM CELLS

Size: px
Start display at page:

Download "TISSUE-SPECIFIC STEM CELLS"

Transcription

1 TISSUE-SPECIFIC STEM CELLS Concise Review: Wnt Signaling Pathways in Skin Development and Epidermal Stem Cells ANTHONY VELTRI, CHRISTOPHER LANG, WEN-HUI LIEN Key Words. Wnt signaling Skin development Hair follicle morphogenesis Epidermal stem cells de Duve Institute, Faculty of Medicine, Universite Catholique de Louvain, Brussels, Belgium Correspondence: Wen-Hui Lien, Ph.D., de Duve Institute, Universite catholique de Louvain, Avenue Hippocrate 74, Box B , 1200 Brussels, Belgium. Telephone: ; uclouvain.be Received August 22, 2017; accepted for publication September 23, 2017; first published online in STEM CELLS EXPRESS October 19, Available online without subscription through the open access option /stem.2723 ABSTRACT Mammalian skin and its appendages constitute the integumentary system forming a barrier between the organism and its environment. During development, skin epidermal cells divide rapidly and stratify into a multilayered epithelium, as well as invaginate downward in the underlying mesenchyme to form hair follicles (HFs). In postnatal skin, the interfollicular epidermal (IFE) cells continuously proliferate and differentiate while HFs undergo cycles of regeneration. Epidermal regeneration is fueled by epidermal stem cells (SCs) located in the basal layer of the IFE and the outer layer of the bulge in the HF. Epidermal development and SC behavior are mainly regulated by various extrinsic cues, among which Wnt-dependent signaling pathways play crucial roles. This review not only summarizes the current knowledge of Wnt signaling pathways in the regulation of skin development and governance of SCs during tissue homeostasis, but also discusses the potential crosstalk of Wnt signaling with other pathways involved in these processes. STEM CELLS 2018;36:22 35 SIGNIFICANCE STATEMENT This review article provides a concise overview on the current knowledge of Wnt signaling pathways in the regulation of skin development, hair follicle morphogenesis, and governance of epidermal stem cells during tissue homeostasis. The spatial and temporal interconnections of Wnt/ß-catenin and ß-catenin-independent Wnt signaling in the developmental and homeostatic processes of skin are also highlighted. Using epidermal development and regeneration as examples, this study calls attention to the potential crosstalk between Wnt signaling with other pathways that co-orchestrate these processes. INTRODUCTION Secreted Wnt proteins can stimulate multiple intracellular signaling pathways and act as growth factors that regulate diverse processes, including cell proliferation, differentiation, migration, and polarity [1 3]. Among Wntstimulated pathways, Wnt/b-catenin signaling is known as an important regulatory pathway that governs developmental processes and fate choices during tissue morphogenesis [4 6]. Deregulation of Wnt/b-catenin signaling has been linked to several human diseases and cancers [5, 7]. In addition to b-catenin-dependent Wnt signaling, Wnt-activated signaling pathways that do not depend on b-catenin are referred as non-canonical Wnt pathways and also play divergent roles in development and cancer [8, 9]. While the role of Wnt/b-catenin signaling has been extensively characterized in many different tissues, studies of non-canonical Wnt pathways, to date, have mainly stressed their ability to inhibit Wnt/b-catenin signaling. In this review, we focus on the cellular processes of skin development and homeostasis to point out the spatial and temporal interconnections of Wnt-dependent signaling pathways. Using epidermal regeneration as an example, we draw attention to the controversial question of whether Wnt/b-catenin signaling is essential for stem cell (SC) maintenance. Last, we discuss the potential crosstalk of Wnt signaling with other pathways that co-orchestrate skin development and SC activation. THE WNT SIGNALING PATHWAYS Wnt signaling is one of the major cues directing skin development and maintenance [1, 3]. To drive these cellular activities, Wnt ligandreceptor interactions initiate signaling cascades that can be divided into canonical (b-catenindependent) and non-canonical (b-cateninindependent) Wnt signaling. The detailed description of Wnt signaling pathways has STEM CELLS 2018;36: VC AlphaMed Press 2017

2 Veltri, Lang, Lien 23 Figure 1. Wnt signaling pathways. This schematic diagram displays the processes of Wnt secretion, canonical and non-canonical Wnt signaling pathways. Synthesized Wnt protein is palmitoylated by Porcupine in ER, and transferred into the Golgi, where Wnt is complexed to Wls for secretion. Wls-bound Wnt protein is then escorted from Golgi to the plasma membrane and released into the extracellular environment where it can bind to target cells. In the absence of Wnt, free cytoplasmic b-catenin is targeted and phosphorylated for its degradation by a destructive complex, composed of the core proteins Axin, CK1a, APC, and GSK3b. The subsequent b-catenin degradation is mediated by b-trcp E3 ubiquitin ligase. Once Wnt ligands bind to receptor Fz and co-receptor LRP5/6, Dvl is recruited, leading to the inhibition of the degradation complex. Stabilized b-catenin is accumulated in the cytoplasm and then enters into the nucleus where it acts as a transcriptional co-activator for LEF/TCF transcription factors to activate Wnt-responsive genes. b-catenin-independent non-canonical Wnt pathways can be categorized into Wnt/Ca 21 and Wnt/PCP pathways. Binding of Wnt isoforms to either Fz or other tyrosine kinase-like receptors, for example, Ror2, can trigger multiple signaling cascades. In Wnt/Ca 21 signaling, Wnt-receptor interaction initiates intracellular calcium release, thereby activating CaMKII, CaN, or PKC. Among these effectors, CaMKII triggers TAK1-NLK cascade, which suppresses transcriptional activity of Wnt/b-catenin signaling. In parallel, activated CaN activates NFAT family proteins for transcriptional regulation. Wnt/PCP pathways involve activation of small GTPases Rho, Rac, and Cdc42, and their downstream JNK signaling that regulate cytoskeleton rearrangement and PCP. The mechanism underlying b-catenin-independent Wnt signaling may be dependent on the cellular context. Abbreviations: AP-1, activating protein-1; APC, adenomatous polyposis coli; CaMKII, calmodulin-dependent protein kinase II; CaN, calcineurin; CK1a, casein kinase 1a; DKK, Dickkopf proteins; Dvl, Dishevelled; ER, endoplasmic reticulum; Fz, Frizzled; GSK3b, glycogen synthase kinase 3b; JNK, c-jun N-terminal kinase; LEF/TCF, lymphoid-enhancing factor/t-cell factor; LRP5/6, lipoprotein receptor-related protein 5/6; NFAT, nuclear factor of activated T-cells; NLK, nemo-like kinase; PCP, planar cell polarity; PKC, protein kinase C; ROCK, Rho-associated kinase; ROR2, receptor tyrosine kinase-like orphan receptor 2; RYK, receptor-like tyrosine kinase; SFRP, secreted frizzled-related proteins; TAK1, TGF-b activated kinase 1; b-trcp, b-transducing repeat-containing protein; WIF, Wnt inhibitory factor; Wls, Wntless. been extensively covered elsewhere [3, 4, 10, 11], and we only briefly summarize those pathways below. Wnt signaling is activated when a Wnt ligand binds to receptor/co-receptor. Wnt proteins are encoded by a family of genes highly conserved across the animal kingdom, and so far 19 Wnt genes have been identified in mouse and human genomes [3]. Post-translational modifications of the Wnt proteins, including glycosylation and palmitoylation, are critical for the secretion and the binding to receptors [12, 13]. Some synthesized Wnt proteins are glycosylated and palmitoylated in the endoplasmic reticulum by the actions of porcupine [13, 14], and transferred into the Golgi, where they are complexed to Wntless (Wls), a transmembrane protein that is essential for Wnt secretion [15, 16]. Wls binds to Wnt proteins and escorts them from the Golgi to the plasma membrane; then released Wnt proteins bind to target cells in an autocrine or paracrine fashion (Fig. 1). Secreted Wnt ligands interact with receptor complexes consisting of the Frizzled (Fz) family receptors and/or co-receptors, such as low density lipoprotein receptor-related protein (LRP)-5/6, receptor tyrosine kinaselike orphan receptor 2 (ROR2), or receptor-like tyrosine kinase (RYK), to activate diverse signaling pathways [17 21]. The interaction of Wnt proteins with their cognate-receptors can be blocked by a number of secreted soluble regulators, including Dickkopf proteins (Dkk), secreted frizzled-related proteins (SFRP), or Wnt inhibitory factor (WIF) [22 25]. Furthermore, receptor-ligand interactions are regulated by modulation of receptor abundance via R-spondins and leucine-rich repeat-containing G-protein coupled receptor proteins [10, 26]. Among the Wnt-induced pathways, the b-catenindependent canonical Wnt signaling pathway appears to be the most conserved in both vertebrates and invertebrates [27]. The central output of the canonical Wnt (referred as Wnt/b-catenin) pathway is the stabilization of b-catenin, a cytoplasmic/nuclear protein that plays a dual role in adherens junctions and transcriptional regulation [11, 28] (Fig. 1). In the absence of Wnt stimulation, free cytoplasmic b-catenin proteins are phosphorylated by a destructive complex, consisting of Axin, casein kinase 1a (CK1a), adenomatous polyposis coli (APC), and glycogen synthase kinase 3b (GSK3b). Phosphorylated b-catenin is then ubiquitinated by a b-transducing repeat-containing protein (b-trcp) for proteasome-dependent degradation [29, 30]. Once the Wnt ligand binds to the receptor/co-receptor complex, the Dishevelled (Dvl) protein is recruited, leading to the inhibition of the degradation VC AlphaMed Press 2017

3 24 Wnt Signaling in Skin Development and Stem Cells complex, thereby stabilizing b-catenin [31, 32]. Stabilized b- catenin accumulates in the cytoplasm and then enters the nucleus where it acts as transcriptional co-activator for the transcription factor family of lymphoid-enhancing factor/t-cell factor (LEF/TCF) proteins [1, 28, 33]. The b-catenin and LEF/ TCF interaction engages transcriptional regulators and histone modifiers, which in turn mediates a plethora of developmental and homeostatic processes. The b-catenin-independent Wnt signaling can be triggered when the Wnt ligand binds to Fz receptor and the coreceptors, ROR2 or RYK. These non-canonical Wnt signaling cascades can be categorized into Wnt/Calcium (Ca 21 ) and Wnt/Planar cell polarity (PCP) signaling pathways [34 37] (Fig. 1). In Wnt/Ca 21 signaling, the Wnt ligand-receptor interaction leads to release of intracellular calcium, which in turn acts as a secondary messenger to activate calmodulin-dependent protein kinase II (CaMKII), calcineurin (CaN), or protein kinase C (PKC) [8, 38]. Among these activated effectors, CaMKII further triggers phosphorylation of TGF-b activated kinase 1 (TAK1), which in turn elevates the activity of nemo-like kinase (NLK), leading to the LEF1 b-catenin/dna dissociation, thereby inhibiting transcriptional activity of Wnt/b-catenin signaling [39]. In parallel, activated CaN induces nuclear translocation of its downstream effectors, nuclear factor of activated T-cells (NFAT) family proteins, which act as transcriptional regulators [40]. In parallel, NFAT transcriptional activity can also be reinforced by mitogen-activated protein kinase p38 upon Wnt stimulation [41]. In addition to the Ca 21 -mediated signaling cascades, induction of the non-canonical Wnt signaling also activates Rho family small GTPases, including Cdc42, Rac, and RhoA, via the recruitment to the receptor/co-receptor/dvl complex [42, 43] (Fig. 1). Activated Rac and Cdc42 initiate downstream c- Jun N-terminal kinase (JNK) signaling, which leads to transcriptional activation of the activating protein-1 (AP-1) complex, and orchestrates PCP by regulating actin cytoskeleton organization and cell migration [44 46]. Other than Rac and Cdc42, RhoA activation downstream of non-canonical Wnt signaling requires binding of Daam1 to Dvl. The activation of RhoA switches on the activity of the downstream effector, Rhoassociated kinase (ROCK), which leads to cytoskeletal rearrangement, thereby regulating PCP and cell migration [47 49]. There are several different Wnt proteins and known receptors in vertebrates, and their expression is spatially and temporally regulated during development. The responses to a given stimulus depend not only on which Wnt is present, but also on which cognate receptor is expressed on the cell. It is likely that one Wnt protein activates a combination of multiple signaling cascades that might act independently or collaboratively. Adding more layers of complexity, cofactors, secreted antagonists and co-receptors of Wnt signaling modulate both canonical and non-canonical actions. It is known that non-canonical Wnt pathways have the ability to inhibit b-catenin-dependent signaling; however, the mechanism underlying this inhibition remains unclear. Proposed explanations include the competition of Wnt ligands for the binding to Fz receptors [37], downregulation of b-catenin by the E3 ubiquitin ligase Siah2 [50], or the inhibition of Wnt/b-catenin transcriptional activity via TAK1-NLK-mediated phosphorylation of TCFs [51, 52]. VC AlphaMed Press 2017 WNT SIGNALING IN EPIDERMAL STRATIFICATION AND HF MORPHOGENESIS Mammalian skin is constituted of three primary layers, the epidermis, the dermis, and the hypodermis. The epidermis and its derivative appendages, such as hair follicles (HFs), sebaceous glands, and sweat glands, function together as a physical barrier that protects the organism from environmental stresses, such as dehydration, irradiation, and pathogenic infection. The underlying dermis contains blood vessels, which nourish the avascular epidermis, and protein fibers that enhance skin mechanical strength. This dermis lays on the hypodermis which is an adipose tissue providing thermal insulation and energy resource. Wnt signaling pathways have been shown to play crucial roles in epidermal development, HF morphogenesis, and regeneration, which will be discussed in depth below. The Stratification of the Developing Epidermis At the early stages of skin development, dynamic signaling crosstalk occurs between the embryonic epidermis and the dermis. The inter-tissue communication instructs basement membrane formation, stratification of the epidermis, and HF induction [53]. During embryogenesis, the neuroectoderm layer of the embryo gives rise to the nervous system and the skin epithelium. The specification of the ectodermal cells into the epidermal fate is driven by Wnt signaling, which inhibits the response of ectoderm to fibroblast growth factors (FGFs). In the absence of FGF signaling, ectodermal cells express bone morphogenetic proteins (BMPs) and start to acquire an epidermal fate [54 56]. Once ectodermal cells commit to an epidermal fate, they differentiate into keratin (K)-expressing cells, namely keratinocytes, and form a basal layer of embryonic epidermis (Fig. 2). Keratinocytes in the newly formed embryonic basal layer replace the expression of K8/K18 with K5/K14 [57]. At the initial step of epidermal stratification, the epidermal basal cells give rise to a transient layer of endodermis-like cells, called periderm, which protects epidermal basal cells from constant exposure to the amniotic fluid [58]. The next layer of the epidermis generated between the basal layer and the periderm is called intermediate layer, the development of which is associated with asymmetric cell division of the epidermal basal cells [59]. The intermediate layer cells initially undergo proliferation and then mature into spinous cells expressing K1/K10. The resulting spinous cells subsequently further mature into Involucrin-positive granular cells, which terminally differentiate into Filaggrin- and Loricrin- expressing cornified cells [60 62]. The cornified cells finally form the cornified envelop and fulfill the barrier function of the skin. When the stratification program is completed, the epidermis is composed of an inner layer of basal cells with proliferative potential and suprabasal layers of differentiated cells. At a molecular level, the epidermal stratification process is orchestrated by several transcriptional regulators and signaling pathways (reviewed in [61, 63]). Although Wnt signaling has been mainly implicated in HF induction during skin development, it has also been recently shown to regulate epidermal stratification. Using a genetic mouse model disrupting Wnt production in the developing epidermal basal cells, the authors found that epidermal Wnt production triggers Wnt- STEM CELLS

4 Veltri, Lang, Lien 25 Figure 2. Epidermal stratification and hair follicle (HF) formation. During early embryogenesis the surface of the embryo is covered by a single ectodermal layer, called surface ectoderm, which adheres to an underlying BM. Upon Wnt stimulation, ectodermal cells are specified into epidermal fate and form the first keratin-expressing BL. At the initial step of epidermal stratification, the epidermal basal cells give rise to a transient layer periderm, which protects basal cells from constant exposure to the amniotic fluid, and then generate intermediate layer via asymmetric cell division. The intermediate layer cells undergo proliferation and then mature into SL, which subsequently develops into GL. The granular layer cells in the last step of stratification terminally differentiate into CL to form cornified envelop and acquire the barrier function. In parallel with epidermal stratification, upon Wnt signal transmission between epidermis and dermis, HFs start forming from the developing epidermis. Before HFs are induced, dermal fibroblasts uniformly receive a widespread Wnt signal from epidermis. In response, dermal fibroblasts then produced the first dermal Wnt signal to induce aggregation of epidermal basal cells, leading to the formation of hair placode. After the initial induction, the developing placode produces Wnt ligands to induce underlying fibroblasts to form a dermal condensate; at the same time, the placode continuously grows and invaginates into a HG. The epidermal cells continue to penetrate the forming dermis and form an elongated column, called HP; meanwhile, the dermal condensate becomes a spherical DP enclosed by the lower end of HP. Once HG grows into HP, precortical matrix, a differentiated epidermal layer, becomes visible. As soon as HF down-growth reaches the subcutis, the HF starts forming SG at the upper part of HF and DP becomes thinner and gets fully enclosed by the matrix of the HF. Abbreviations: BL, basal layer; BM, basement membrane; CL, cornified layer; DP, dermal papilla; GL, granular layer; HG, hair germ; HP, hair peg; SG, sebaceous gland; SL, spinous layer. VC AlphaMed Press 2017

5 26 Wnt Signaling in Skin Development and Stem Cells dependent activation of a BMP-FGF signaling cascade in the underlying mesenchyme, and that this mesenchymal activation is essential for feedback regulations in the epidermis to modulate the spinous layer formation [64]. Furthermore, in primary human keratinocytes, Wnt5a acts as an autocrine stimulus to promote extracellular calcium-induced keratinocyte differentiation by coupling with Wnt/b-catenin pathway [65]. Although the underlying molecular networks are not fully understood, extracellular calcium-induced keratinocyte differentiation could be in part regulated by Wnt. HF Formation and Maturation HF morphogenesis includes three main stages: hair placode formation, HF organogenesis, and cytodifferentiation [66, 67]. The sequential events during morphogenesis are tightly regulated by signals transmitted between the dermis and the epidermis. Among those signals, Wnt signaling pathway is considered to be the master regulator. Before any visible hair placode formation, dermal fibroblasts uniformly receive a widespread epidermis-derived Wnt signal. In response, dermal fibroblasts then produce the first dermal Wnt signal that induces aggregation of epidermal basal cells at regularly spaced intervals, leading to the formation of hair placodes (Fig. 2). Notably, the pattern of the HF induction is dependent on a competition between slowly diffusible Wnt ligands and faster diffusing Wnt inhibitors [66, 68]. After the initial induction, the developing placode produces Wnt ligands to induce underlying fibroblasts to form a dermal condensate [69, 70]. Concomitantly, the placode continuously grows, invaginates into the underlying dermis, and then joins the dermal condensation to form the first structure of the HF organogenesis, the primary hair germ (HG). The epidermal cells continue to penetrate the forming dermis and generate a multi-layered and elongated column, called the hair peg. Meanwhile, the dermal condensate becomes a spherical dermal papilla (DP). The hair peg thickens at the lower end to form a hair bulb and half of which encloses the elongated DP. When the HG grows into a hair peg, differentiated epidermal layers that will give rise to the hair shaft become visible. As soon as HF down-growth reaches the subcutis, the program of cytodifferentiation initiates. At this point, the DP first becomes thinner and gets totally enclosed; then the sebaceous gland starts forming at the upper part of the HF. Finally, the fully formed hair shaft protrudes from the skin surface and the HF reaches its maximal length [66]. Wnt/b-catenin signaling is first upregulated uniformly in the upper dermis and then focally in both the hair placode and the underlying dermal condensate, where Lef1 is expressed [69, 71, 72]. When Wnt/b-catenin signaling is turned off either by overexpressing Dkk1 in the skin, by conditional ablation of b-catenin, or by transgenic expression of a truncated form of Lef1, HF formation is blocked [73 75]. Consistently, overexpression of a stable form of b-catenin or Lef1 induces de novo HF formation [72, 76, 77]. These findings suggest that the levels of Wnt/b-catenin signaling in the developing epidermis determine the specificity toward HF lineage. Further genetic investigations revealing Wnt signaling effects on the development of skin epidermis and HFs are summarized in Table 1. The development of DP occurring at early HF morphogenetic stages is also dependent on Wnt, and more precisely on VC AlphaMed Press 2017 Wnt5a [78]. Typically, Wnt5a is expressed in the developing dermis and highly enriched in the dermal condensate. Given that the dermal Wnt5a expression is completely abolished in sonic hedgehog (Shh) mutant skin, the dermal Wnt5a is considered as a target of Shh signaling activated by Wnt/b-catenin signaling in the hair placode. Although Wnt5a deletion in the skin does not lead to detectable defects in HF morphogenesis, Wnt5a-deficient DP cells lose HF-inductive ability in postnatal skin [79], underscoring the role of Wnt5a as an essential dermal signal. In embryonic murine skin, PCP is established within the entire epidermal plane in an anterior to posterior direction [70]. Over-activation of Wnt/b-catenin signaling in the developing epidermis can disrupt the polarity of HF orientation, and this is likely caused by perturbing the asymmetrical expression of Shh in the developing HF [72, 76]. Non-canonical Wnt pathways can also direct PCP via a Wnt-Fz-Dvl circuitry to activate Rho-ROCK and/or Rac-dependent signaling cascades. A Wnt receptor and PCP protein, Fz6, is asymmetrically localized in the embryonic epidermis and controls HF orientation in mice [80 82]. Although it remains unclear whether Wnt morphogens are needed in the establishment of hair patterning, Wnt/PCP signaling, known to regulate cytoskeletal rearrangement and cell migration, likely serves as a spatial signal to modulate HF orientation during morphogenesis. WNT SIGNALING IN SKIN EPIDERMAL STEM CELLS Throughout life the skin epidermis is regularly renewed. Skin epidermal SCs, capable of self-renewal and differentiation, provide unlimited sources of cells to maintain tissue homeostasis, as well as to regenerate HFs and repair the epidermis after injury. Skin epidermal SCs are located in the basal layer of the interfollicular epidermis, and also in the bulge region of the HF [83]. While adult interfollicular epidermal (IFE) basal cells sustain continual proliferation, the HFs undergo cycles of degeneration and regeneration. Both processes require the activation of SCs, which is largely modulated by signaling cues from their microenvironment. Among these signals, Wntdependent signaling plays crucial roles in the maintenance, activation, and fate determination of the SC populations. Stem Cells in the Interfollicular Epidermis The postnatal epidermis is continuously regenerated by the proliferative basal cells of the IFE. This IFE pool gives rise to progenies that will differentiate into suprabasal cells while migrating upward. During this process, the epidermal cell number remains constant as the number of newly generated cells exactly compensates for the number of cells that differentiate or die [53]. Two models, the hierarchical and the stochastic model, have been proposed to explain how SCs in the basal layer act to replenish the IFE [83]. The hierarchical model suggests that a slow-cycling SC located in each epidermal proliferative unit of the IFE generates short-lived transitamplifying cells (TACs), which then give rise to differentiated cells. The stochastic model, first proposed by Jones group, was that the progenitors in the basal layer of the IFE have equal potential to generate daughter cells which remain as progenitors or differentiate into suprabasal cells [84]. STEM CELLS

6 Veltri, Lang, Lien 27 Table 1. Summary of mouse models for Wnt signaling pathways in skin epidermis and hair follicles Target gene Target tissues Mutation Induction Phenotypes (stages) References b-catenin (loss-of-function) IFE and HFs K14-Cre; E9.5 - Placode formation is impaired (E15.5) Ctnnb1 fl/fl - Hair loss and cyst formation (P20) - Hyperproliferative epidermis (P40) [73] K14-CreER; P49 - Loss of SC marker - CD34 (P59) Ctnnb1 fl/fl - Fail to generate HF and adapt to sebocyte fate in cysts (P59) [131] K5-rtTA; TetO-Cre; Ctnnb1 fl/fl P Decreased cell proliferation and increased apoptosis of HF matrix (P8) - Precocious catagen entry (P14) - HF degeneration and loss of HFSCs (P60) [91] P HFs fail to enter anagen (P25) P plucking at P54 HFSCs K15-CrePR; P Ctnnb1 fl/fl plucking at P54 - The shg fails to proliferate (P57; 3DPP) - The shg fails to proliferate (P59; 5DPP) - HFs fail to enter anagen (P68; 14DPP) [91] P HFs fail to enter anagen (P95) - HFSCs remain expressing SC makers - CD34, SOX9, TCF4 (P200) [96] P plucking at P58 - HFSCs undergo proliferation (P60; 2DPP), but fail to differentiate into HFs (P65; 7DPP) - HFSCs differentiate into sebocytes (P81; 23 DPP) - Loss of bulge/hfscs, but with enlarged sebaceous glands (P610, 560DPP) Non-hairy IFE (footpad) Axin2-CreER; P21 - Decreased cell proliferation and increased proportion of K101 differentiated Ctnnb1 fl/fl cells in non-hairy IFE (P31) [92] K5-rtTA; TetO-Cre; Ctnnb1 fl/fl Dermal papilla Cor-Cre; P Decreased cell proliferation of non-hairy IFE (P71) [91] Ctnnb1 fl/- - Premature catagen (P16) P3/P7 - Reduced proliferation of HF matrix cells (P10) - Hair shortening and thinning (P60) - Impaired HF regeneration after depilation (P82; 12DPP) [132] Dermal fibroblasts En1-Cre; E Reduced proliferation of dermal fibroblasts (E14.5) Ctnnb1 fl/d - Failure in placode initiation and dermal condensate formation (E16.5) [69] b-catenin (gain-of-function) IFE and HFs K14-DN87bcat E9.5 - E De novo HF formation from IFE and the upper part of HFs (>P18) - Trichofolliculoma formation and Pilomatricoma development (Old homozygous mice) [76] - Precocious HFSC activation and hair cycle entry (P55 heterozygous mice) [131] K14-DNDCbcat E9.5 - E Formation of epidermal and sebaceous cysts from HFs (P24-P28) - Loss of TOPGAL activation in the bulge (P22) - De novo hair-like invagination from IFE (P28) [133] K14- S33Ybcat-ER P Precocious activation and anagen entry (P53) - Hyperplastic anagen HFs (P64) [134] K14-DNbcat -ER P Precocious activation and anagen entry (P45) - Abnormal HFs containing cysts (P53) - De novo HF formation from IFE (P50) [77] VC AlphaMed Press 2017

7 28 Wnt Signaling in Skin Development and Stem Cells Table 1. Continued Target gene Target tissues Mutation Induction Phenotypes (stages) References - Trichofolliculoma formation (P70), but tumors regress after induction stops K14-Cre; E9.5 - Precocious HF induction (E13.5) Ctnnb1 fl(ex3)/1 - Disrupted HF morphogenesis (E17) - Impaired hair shaft production in grafted skin (21DPG) [135] HFSCs K19-CreER; P20 - Induction of new hair growths from existing HFs and this induction is independent Ctnnb1 fl(ex3)/1 of DP signals (P32-P39) [136] Dermal fibroblasts HoxB6CreER; E Increased proliferation of dermal fibroblasts (E14.5) Ctnnb1 fl(ex3)/1 - Increased hair placode size and number (E16.5) - Accelerated differentiation of HF (E17.5) LEF1 (loss-of-function) IFE and HFs LEF1 -/- Constitutive - Arrest in HF development (P3) - Reduction in HF number (P3) - Lack of body hairs and whiskers (P16) [69] [137] K14-DNLef1 E9.5 - Suppression in hair differentiation and shg cells adapt sebocyte fate (>P21) [89] K14-DNLef1 E9.5 - Hair loss (P42) - Formation of epithelial cysts (P105) - Development of skin tumors (>P90) LEF1 (gain-of-function) IFE and HFs K14-Lef1 E9.5 - Irregularly spaced and misoriented HFs (P28) [72] TCF (loss-of-function) IFE and HFs K14-Cre; Tcf3 fl/fl E9.5 - No apparent epidermal defects [138] [74] K14-mycDNTcf4 E9.5 - Epidermal differentiation defects and impaired barrier function (P0) K14-Cre; Tcf3 fl/fl ; Tcf4 -/- E9.5 - Initiated HFs could not grow down (P0) - No hair formation in grafted skin (17DPG) - Failure of long-term IFE maintenance (60DPG) HFSCs K15CrePR; Tcf3 fl/fl ; Tcf4 -/ DPG - HFSC precocious activation (58DPG) - Precocious entry of hair cycle (65DPG) TCF (gain-of-function) IFE and HFs K14-Tcf3 E9.5 - Impaired epidermal barrier function (E18.5-P0) - Epidermal differentiation defects (P0) [96] [89] K14rtTA; TRE-mycTcf3 P1-5 - Repression of epidermal differentiation and sebaceous gland development (P5) - Suppression of HF development (d27, 60DPG) [139] P HFSCs fail to get activated (P23) [96] Wls (loss-of-function) IFE and HFs K14-Cre; Wls fl/fl E9.5 - Impaired hair placode induction (E14.5) - Loss of HF formation (E18.5, P7) [115] - Hyperproliferation of IFE basal cells (P16 28) - Epidermal differentiation and skin barrier defects (P16) [93] K14-CreER; Wls fl/fl P Impaired hair cycle entry (P37) - Hyperplasia of IFE and sebaceous glands (P47) - Defects in HFSC proliferation, but normal for HFSC maintenance (P91) - Failure of depilation-induced HF regeneration (15DPP) [114] K5-rtTA; P Premature regression of HFs (P14) TetO-Cre; Wls fl/fl - External hair loss and formation of cysts (P60) - HFSCs remain expressing SC makers - CD34 and KRT15 (P60) [91] VC AlphaMed Press 2017 STEM CELLS

8 Veltri, Lang, Lien 29 Table 1. Continued Target gene Target tissues Mutation Induction Phenotypes (stages) References HFSCs K15-CrePR; Wls fl/fl P HFSCs remain expressing SC makers - CD34 and KRT15 (P60) [91] Dermal fibroblasts En1-Cre; Wls fl/fl E No defects in HF initiation (E14.5) [69] [90] Porcupine (loss-of-function) IFE and HFs Sox2-Cre; Porcn fl/1 E6.5 - Reduced density of hair placodes (E14.5) - Partial skin with hair loss (P22) [75] DKK1 (gain-of-function) IFE and HFs K14-Dkk1 E9.5 - Failure of placode formation (E15.5) - Lack of HF formation (P16) - No effect in epidermal differentiation (P28) [91] P Decreased HF matrix proliferation (P8) - Premature HF regression (P14) - HFSCs remain expressing SC makers - CD34 and KRT15 (P24) K5-rtTA; TetO-Dkk1 - Reduced proliferation in shg (P57; 3DPP) P plucking at P54 - Inhibition of hair growth and hair loss (P475) - HFSCs remain expressing SC makers - CD34 and KRT15 (P475) - Hair growth inhibition is reversed after stopping DKK1 expression (P489) P withdrawal for 14d (P489) [68] DKK2 (gain-of-function) IFE and HFs Foxn1-Dkk2 E Reduced density of HFs (E14.5) - New hair growths from existing HFs (P1) Abbreviations: DKK, Dickkopf proteins; DPP, day post-plucking; DPG, day post-grafting; E, embryonic day; HF, hair follicle; HFSC, hair follicle stem cell; IFE, interfollicular epidermis; LEF, lymphoidenhancing factor; P, postnatal day; SC, stem cell; shg, secondary hair germ; TCF, T-cell factor; Wls, Wntless. Using different experimental approaches, evidence for both the hierarchical and the stochastic model have been presented. On the one hand, IFE lineage tracing experiments support the hierarchical model and show the existence of two proliferative cell populations representing slow-cycling SCs and committed progenitors produced in hierarchical manner. Upon wounding, only SCs are capable of responding to proliferative stimuli and contributing to long-term regeneration of the tissue [85]. In accordance with this finding, a recent study further shows that epidermal label-retaining cells (LRCs) and non-lrcs constitute two SC populations of the IFE. Although these two SC populations are molecularly distinct and produce unique differentiated lineages during homeostasis, they are functionally interchangeable and able to replenish each other s territories upon wounding [86]. On the other hand, using live imaging techniques, Rompolas and colleagues support the stochastic model and demonstrate that IFE basal cells are born as uncommitted SCs which have equal potential to proliferate or undergo differentiation [87]. In agreement, a recent study using multicolor lineage tracing shows that the IFE can be maintained without a hierarchy of SCs and TACs [88]. The tools used to mark basal cells and examined epidermal regions at different body sites are likely to account for the differences among these studies. Despite the growing evidence from various studies, the existence of a hierarchy between slow-cycling SCs and committed TACs in the IFE remains as an open question. In the developing epidermis, a high level of Wnt/b-catenin signaling is essential for HF induction. The constitutive attenuation of epidermal Wnt/b-catenin signaling impairs HF formation, but does not impact IFE integrity [73 75, 89, 90]. Notably, K14Cre-driven b-catenin depletion in developing IFE even causes epidermal hyperproliferation [73]. Nevertheless, lineage tracing studies with an inducible Cre recombinase gene driven by Wnt reporter Axin2 (Axin2-Cre) show the presence of Wnt/b-catenin signaling activity in the basal cells of non-hairy epidermis, and that Axin2-marked basal cells are long-term IFE progenitors. When b-catenin is depleted in these cells, the epidermis exhibits severe hypoproliferation [91, 92] (Table 1). The disparate results in epidermal proliferation could be partially explained by the fundamental differences between hairy and non-hairy epidermis because it is thought that epidermal hyperproliferation of hairy skin could partially result from inflammatory response to the HF disintegration [91, 93]. However, such hyperproliferation caused by HF loss is not found in epidermis expressing dominantnegative LEF1 or Wnt inhibitor DKK1 [74, 75]. In addition, given that b-catenin-null SCs are still capable of generating epidermis when chamber grafted [94], it remains unsolved whether Wnt/b-catenin signaling is necessary for the generation and maintenance of IFE. HF Stem Cells In contrast to the epidermis which continuously regenerates, mature HFs progress through cycles of growth (anagen), degeneration (catagen), and rest (telogen) all through life (Fig. 3). During the hair cycle, the upper portion of the HF is permanent while the lower portion undergoes degeneration and then regeneration by the SCs. Stem cells in the HF can be divided into two populations: one population residing in the outer layer of the bulge, known as bulge hair follicle stem VC AlphaMed Press 2017

9 30 Wnt Signaling in Skin Development and Stem Cells Figure 3. The hair cycle. During the resting phase (Telogen), HFSCs residing in the outer layer of bulge remain in quiescence as the inner bulge K61 cells express high levels of inhibitory signals. At the onset of the growth phase (Anagen), stem cell progenies located in shg proliferate and initiate HF regeneration in response to the activating cues produced from the underlying DP. At early anagen, HFSCs begin to proliferate and give rise to ORS, a population of cells migrating downward from the bulge and feeding into the bulb of matrix at the base of the newly formed HF. At the same time, shg develops into matrix, composed of the pool of transient-amplifying cells that rapidly proliferate and then terminally differentiate to form hair shaft and IRS at later stage of anagen. At Mid-Anagen, matrix progenitors in the pre-cortex region terminally differentiate to form the hair shaft. At the end of anagen when the HF enters a destructive phase (Catagen), the lower part of the HF undergoes apoptosis and the epithelial strand regresses upward. Once the DP is drawn upward toward the bulge/shg, the HF re-enters telogen. Abbreviations: DP, dermal papilla; HF, hair follicle; HFSCs, hair follicle stem cells; IFE, interfollicular epidermis; IRS, inner root sheath; ORS, outer root sheath; shg, secondary hair germ; SG, sebaceous gland. cells (HFSCs); the other population located within the secondary hair germ (shg) right below the bulge [83]. During telogen, bulge HFSCs remain in quiescence while SC progenies in the shg are primed, which makes them become the first group of cells getting activated at the anagen onset [95, 96]. At early anagen, bulge HFSCs begin to proliferate and give rise to the outer root sheath (ORS), a population of cells migrating downward from the bulge and feeding into the bulb of the matrix at the base of the newly formed HF, resulting in follicle growth [95, 97]. On the contrary, the shg develops into the matrix, composed of the pool of TACs that rapidly proliferate and then terminally differentiate to form hair shaft and the inner root sheath (IRS) [97, 98]. As soon as the TACs and the DP move far away from the SC niche, bulge HFSCs return to quiescence [99]. Once reaching catagen, the lower portion of ORS and matrix cells die through apoptosis while upper and middle portions of ORS cells migrate upward to form a new bulge and a new HG, respectively [97]. VC AlphaMed Press 2017 Bulge HFSCs were initially identified as slow-cycling LRCs in the postnatal mouse skin [100, 101]. Recent studies with lineage tracing and live-imaging experiments postulate the early specification of HFSCs during skin development [102, 103]. Xu and colleagues discovered that HFSCs are derived from the upper region of hair placode/hg cells which display attenuated Wnt/b-catenin signaling. Elevation of Wnt/b-catenin signaling in embryonic epidermal cells abolishes HFSC specification and suppresses SC marker expression [102]. Furthermore, recent work demonstrated that HFSCs are born from asymmetric divisions of the developing hair bud in which daughter cells differentially display Wnt and Shh signaling [103]. The Wnt-high basal cells maintain slow-cycling and then grow as short-lived progenitors that eventually differentiate; by contrast, the Wnt-low suprabasal cells become SCs and symmetrically expand to form the early HFSC pool [103]. Both studies support the notion that attenuation of Wnt/b-catenin signaling in the early hair bud is a prerequisite for HFSC specification. STEM CELLS

10 Veltri, Lang, Lien 31 In the mature HF, the quiescence and activation of HFSCs are tightly regulated by a balance of BMP and Wnt signals coming from their niche cells [83]. During telogen, the inner bulge K61 cells secrete high levels of BMP6 and FGF18 [97]. Dermal fibroblasts express BMP4 and subcutaneous mature adipocytes express BMP2 [104]. All of these BMPs serve as inhibitory signals to maintain HFSCs in a quiescence state. Toward the end of telogen, the dermal macro-environment reduces BMP expression. By secreting Noggin, the DP turns BMP signaling off in HFSCs, allowing them to transduce Wnt/ b-catenin signaling and thereby promoting anagen onset [95, ]. If the adjacent DPs are ablated, telogen HFs are unable to enter the hair cycle [107, 108], underscoring the necessity of the DP for HFSC activation. In fact, decreased BMP signaling unleashes Wnt signaling activation via upregulation of Wnt ligands and receptors as well as downregulation of Wnt antagonists [109]. Among expressed Wnts, upregulated Wnt7b primes and tips the balance toward Wnt/b-catenin activation and HF growth. Postnatal Wnt7b ablation delays HFSC activation during the telogen-anagen transition [109]. In addition to the DP, adipocyte precursor cells could also indirectly contribute to HFSC activation by modulating DP activity via stimulation of platelet-derived growth factor signaling [110]. At the telogen phase, bulge HFSCs appear to reside in a Wnt-restricted environment as Wnt repressors, such as SFRP1, WIF1, and Dkk3, are highly expressed by HFSCs and the inner bulge K61 niche cells [78, 95, 97, 100, 111, 112]. In agreement, Wnt/b-catenin signaling reporters, TOPGAL or Axin2-LacZ, are inactive in the telogen HFSCs consistent with the absence of nuclear b-catenin [71, 96, 104]. A recent study reports that some Wnt ligands are expressed by HFSCs and that Axin2, a Wnt/b-catenin signaling target gene, is detectable in the outer bulge compartment throughout telogen, implicating the presence of Wnt/b-catenin signaling in telogen HFSCs [113]. However, it is unclear whether this low level of Wnt/b-catenin activity in quiescent HFSCs is required or functional for HFSC maintenance. Telogen HFSCs depleted for b-catenin or Wls are arrested in a quiescent state, and silent b-catenin- or Wls-null HFSCs can be maintained in their niche for a long time without losing the expression of HFSC markers [91, 96, 114]. Only when arrested b-catenin- or Wls-null HFSCs are stimulated by wounding conditions, for example, hair depilation, they begin to exhibit the defects in which they fail to differentiate into HF lineage [96, 114]. Such deficiencies are also found in those developing epidermis losing b-catenin, LEF1-b-catenin interaction or Wls, before HFSCs are specified [73, 74, 93, 115]. Taken together, these findings suggest that the low level of Wnt/b-catenin signaling in telogen HFSCs might not be essential to maintain HFSCs as long as they are kept within their native niche; however, the presence of Wnt/b-catenin signaling is critical for activated HFSCs to retain their potency of HF lineage commitment and differentiation. SIGNALING CROSSTALK OF THE WNT PATHWAYS IN SKIN EPIDERMIS During epidermal development and homeostasis, Wnt signaling pathways function independently or collaboratively with other pathways in order to elicit appropriate cell responses. Some regulatory axes are well studied, and others remain hypothetical and need further investigations. Below we discuss a few selected signaling pathways that cross-interact with Wnt pathways in skin epidermis. Wnt-BMP signaling crosstalk is a regulatory axis that is required for the development of suprabasal keratinocytes and the cyclical regeneration of the HF. During skin development, epidermal Wnts activate mesenchymal responses by transducing a BMP-FGF signaling cascade in the dermis. Such a Wnt- BMP regulatory loop is crucial for the feedback regulation that controls the stratification processes of developing epidermis [64]. Moreover, at the telogen-anagen transition of the hair cycle, the activity of BMP signaling in HFSCs progressively decreases, which permits the activation of Wnt/b-catenin signaling [116, 117]. The mechanism by which BMP inhibition regulates canonical Wnt activation promotes HFSC activation and initiates cyclic regeneration of HFs [104, 109]. In early epidermal development, Shh is expressed in the placode of developing HFs, and Shh signals can be received by both developing epidermis and underlying dermal condensate. The epidermis expressing stabilized b-catenin displays ectopic HF and Shh expression [76, 118]; conversely, in the epidermis lacking b-catenin, Shh is not expressed [73]. These results indicate that Shh signaling serves as a downstream pathway of Wnt/b-catenin signaling to regulate HF induction. On the contrary, Wnt5a, only expressed in the developing dermal condensate, becomes absent in Shh-null embryos, placing Wnt5a as a downstream target of Shh signaling in HF morphogenesis [78]. Moreover, a recent study uncovers that cell fates of the daughter cells in the developing hair bud are determined by their Wnt signaling activities and responses to Shh stimulation. Wnt-low suprabasal cells, responding to Shh, expand symmetrically and become HFSCs, whereas Wnt-high basal cells, producing but not responding to Shh, grow as short-lived progenitors that eventually differentiate [103]. Collectively, these results demonstrate that Wnt-Shh regulatory axis plays multiple roles in HF induction, morphogenesis, and fate choices. Both Wnt and Notch signaling pathways regulate HF maintenance. The induction of new HFs in adult epidermis by stabilized b-catenin can be inhibited by blocking Notch signaling through Jagged1 deletion or treatment with the g-secretase inhibitor [119]. This finding places Notch pathway as a downstream pathway of Wnt/b-catenin signaling to determine HF fate. Furthermore, in a culture system when HFSCs are induced to differentiate toward a hair fate, upregulated Lef1 activates the target gene Jagged1, leading to the co-activation of Wnt/b-catenin and Notch signaling pathways [120]. On the contrary, in the stratified IFE, nuclear b-catenin is only detected in some proliferating basal cells while Notch1 is predominately expressed in differentiated suprabasal layer. Depletion of Notch1 causes upregulation of b-catenin-mediated signaling in multiple layers of hyperproliferative IFE, suggesting Notch1 can repress Wnt/b-catenin signaling pathway to restrict its activation to the basal layer [121]. The synergistic and antagonistic effects between Wnt and Notch signaling seem dependent on the cell fate of skin epidermis. Emerging evidence shows that the Hippo pathway could engage a crosstalk with Wnt/b-catenin signaling to regulate self-renewal of embryonic and intestinal SCs as well as the growth of cancer cells. The transducers of Hippo signaling, VC AlphaMed Press 2017

11 32 Wnt Signaling in Skin Development and Stem Cells YAP/TAZ, act as integral components of the b-catenin destruction complex, and their incorporation orchestrates the Wnt response to regulate SC self-renewal and tissue homeostasis [122, 123]. In cancer cells, downregulation of Hippo signaling correlates with upregulation of b-catenin activity. The proposed mechanism attributed to this correlation is that phosphorylated YAP/TAZ suppresses phosphorylation of Dvl and nuclear translation of b-catenin, thereby inhibiting Wnt/b-catenin signaling [124, 125]. Given that Hippo signaling has been identified as an important regulator of epidermal proliferation [126, 127], the cross-interaction between Hippo and Wnt signaling would be worthwhile further investigation in the regulation of epidermal development and HF regeneration. Several studies have shown that non-canonical Wnt ligands can inhibit canonical Wnt signaling depending on the cellular context. Ectopic Wnt5a expression causes loss of Wnt/ b-catenin signaling and inhibition of HF formation in the developing skin, but gain of Wnt/b-catenin signaling in the meninges [128]. A number of mechanisms for the antagonistic effects of non-canonical Wnt signaling against Wnt/b-catenin signaling have been proposed. For instance, Wnt5a stimulation can lead to increased PKC activity that phosphorylates retinoic acid-related orphan nuclear receptor a (RORa). Phosphorylated RORa binds to b-catenin and forms a transcriptional complex, thereby inhibiting Wnt/b-catenin transcriptional activity [129]. In addition, Wnt5a can also inhibit Wnt3ainduced b-catenin signaling through Ror2 activation [34]. Wnt5a stimulation induces Ror2-Dvl interaction via a CK1- dependent mechanism, and this interaction negatively regulate transcriptional activity of Wnt/b-catenin signaling [130]. Whether similar mechanisms are transduced by non-canonical Wnt ligands during epidermal development and HF regeneration requires further examination. of Wnt signaling in skin epidermis are diverse, it is more and more clear that this diversity is a combined outcome of crosstalk with other signaling inputs in a stage- and contextdependent manner. Multiple Wnt ligands, receptors and differential ligand-receptor compositions add additional layers of complexity to Wnt signaling regulation. Some of Wnt ligands trigger canonical Wnt signaling; others lead to activation of non-canonical Wnt signaling cascades. It remains unclear how cells respond to the stimulation of multiple Wnt ligands in their native microenvironment, and how activated Wnt signaling pathways work collaboratively or antagonistically to orchestrate skin development and HF regeneration. Future investigations should draw more attention to the temporal and spatial signaling cross-interaction required in specific developmental stages and tissue types. It might be worthwhile to investigate which Wnt ligands and receptors are expressed in every cell layers of the epidermis, HFs and their underlying dermal cells. The outputs of Wnt ligand-receptor combinations in individual cell types and their neighboring cells will provide integrative views of signaling crosstalk among Wnt-dependent pathways. ACKNOWLEDGMENTS We thank our colleagues for valuable discussions and acknowledge support from the Fonds de la Recherche Scientifique (FNRS) (MIS Ulysse-F and PDR-T to W.- H.L.), Fonds Joseph Maisin (research grant to W.-H.L.), and Fondation Contre le Cancer (FAF-F/2016/792 to W.-H.L.). A.V. and C.L. are supported by FRIA fellowships from FNRS (1.E to A.V. and 1.E to C.L.). W.-H.L. is an independent investigator of FNRS. CONCLUSION In this review, we summarized the current knowledge of the Wnt-dependent signaling pathways and their functions in skin development and adult epidermal SCs. It is perceivable that Wnt signaling is essential at early time points of skin development for epidermal fate specification, HF induction and morphogenesis, and continuously regulates HFSC activation, lineage commitment, and differentiation. Although the roles AUTHOR CONTRIBUTIONS A.V.: conception and manuscript writing; C.L.: conception and manuscript editing; W.-H.L.: conception, financial support, manuscript writing, and final approval of manuscript. DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST The authors indicated no potential conflicts of interest. REFERENCES 1 Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol 2004;20: Nusse R. Wnt signaling and stem cell control. Cell Res 2008;18: Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell 2012;149: Holland JD, Klaus A, Garratt AN et al. Wnt signaling in stem and cancer stem cells. Curr Opin Cell Biol 2013;25: Clevers H. Wnt/beta-catenin signaling in development and disease. Cell 2006;127: van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development 2009;136: Polakis P. The many ways of Wnt in cancer. Curr Opin Genet Dev 2007;17: VC AlphaMed Press Kohn AD, Moon RT. Wnt and calcium signaling: Beta-catenin-independent pathways. Cell Calcium 2005;38: K uhl M, Sheldahl LC, Park M et al. The Wnt/Ca21 pathway: A new vertebrate Wnt signaling pathway takes shape. Trends Genet 2000;16: Nusse R, Clevers H. Wnt/beta-catenin signaling, disease, and emerging therapeutic modalities. Cell 2017;169: Nelson WJ, Nusse R. Convergence of Wnt, beta-catenin, and cadherin pathways. Science 2004;303: Herr P, Basler K. Porcupine-mediated lipidation is required for Wnt recognition by Wls. Dev Biol 2012;361: van den Heuvel M, Harryman-Samos C, Klingensmith J et al. Mutations in the segment polarity genes wingless and porcupine impair secretion of the wingless protein. EMBO J 1993;12: Kadowaki T, Wilder E, Klingensmith J et al. The segment polarity gene porcupine encodes a putative multitransmembrane protein involved in Wingless processing. Genes Dev 1996;10: B anziger C, Soldini D, Sch utt C et al. Wntless, a conserved membrane protein dedicated to the secretion of Wnt proteins from signaling cells. Cell 2006;125: Bartscherer K, Pelte N, Ingelfinger D et al. Secretion of Wnt ligands requires Evi, a conserved transmembrane protein. Cell 2006; 125: Wehrli M, Dougan ST, Caldwell K et al. arrow encodes an LDL-receptor-related protein essential for Wingless signalling. Nature 2000;407: STEM CELLS

The Beauty of the Skin

The Beauty of the Skin The Beauty of the Skin Rose-Anne Romano, Ph.D Assistant Professor Department of Oral Biology School of Dental Medicine State University of New York at Buffalo The Big Question How do approximately 50 trillion

More information

CHAPTER 6 SUMMARIZING DISCUSSION

CHAPTER 6 SUMMARIZING DISCUSSION CHAPTER 6 SUMMARIZING DISCUSSION More than 20 years ago the founding member of the Wnt gene family, Wnt-1/Int1, was discovered as a proto-oncogene activated in mammary gland tumors by the mouse mammary

More information

Cell Cell Communication

Cell Cell Communication IBS 8102 Cell, Molecular, and Developmental Biology Cell Cell Communication January 29, 2008 Communicate What? Why do cells communicate? To govern or modify each other for the benefit of the organism differentiate

More information

Cell Cell Communication

Cell Cell Communication IBS 8102 Cell, Molecular, and Developmental Biology Cell Cell Communication January 29, 2008 Communicate What? Why do cells communicate? To govern or modify each other for the benefit of the organism differentiate

More information

Signaling Vascular Morphogenesis and Maintenance

Signaling Vascular Morphogenesis and Maintenance Signaling Vascular Morphogenesis and Maintenance Douglas Hanahan Science 277: 48-50, in Perspectives (1997) Blood vessels are constructed by two processes: vasculogenesis, whereby a primitive vascular

More information

ANAT3231: lectures overview

ANAT3231: lectures overview ANAT3231: lectures overview Stem Cell Biology Stem Cell Technology Resources: http://php.med.unsw.edu.au/cell biology/ Essential Cell Biology 3 rd edition Alberts Dr Annemiek Beverdam School of Medical

More information

ANAT3231: lectures overview

ANAT3231: lectures overview ANAT3231: lectures overview Stem Cell Biology Stem Cell Technology Resources: http://php.med.unsw.edu.au/cell biology/ Essential Cell Biology 3 rd edition Alberts Dr Annemiek Beverdam School of Medical

More information

A Genetic Program for Embryonic Development

A Genetic Program for Embryonic Development Concept 18.4: A program of differential gene expression leads to the different cell types in a multicellular organism During embryonic development, a fertilized egg gives rise to many different cell types

More information

Supplementary Information

Supplementary Information Supplementary Information Figure S1: Follicular melanocytes in the wound peripheral area migrate to the epidermis in response to wounding stimuli. Dorsal skin of Trp2-LacZ mice stained with X-gal and analyzed

More information

Vertebrate Limb Patterning

Vertebrate Limb Patterning Vertebrate Limb Patterning What makes limb patterning an interesting/useful developmental system How limbs develop Key events in limb development positioning and specification initiation of outgrowth establishment

More information

Multipotency, Differentiation and Malfunction of Epidermal Stem Cells

Multipotency, Differentiation and Malfunction of Epidermal Stem Cells Cover Multipotency, Differentiation and Malfunction of Epidermal Stem Cells Francisca Peixoto 1 Acknowledgments I would like to thank Salvador Aznar-Benitah for his outstanding supervision. Writing this

More information

Epidermal Stem Cells of the Skin

Epidermal Stem Cells of the Skin I ANRV288-CB22-14 ARI 22 June 2006 19:51 R E V I E W S First published online as a Review in Advance on July 11, 2006 E C N A D V A N Annu. Rev. Cell Dev. Biol. 2006. 22:339 73 The Annual Review of Cell

More information

Glutamate transporter Slc1a3 mediates inter-niche stem cell activation during skin growth

Glutamate transporter Slc1a3 mediates inter-niche stem cell activation during skin growth Article Glutamate transporter Slc1a3 mediates inter-niche stem cell activation during skin growth Bettina Reichenbach 1,, Johanna Classon 1,, Tomomi Aida 2, Kohichi Tanaka 2, Maria Genander 1 & Christian

More information

Integumentary System. Integumentary System

Integumentary System. Integumentary System 1. General aspects a. The integumentary system consists of several organs major organ of the system is the skin other organs are relatively small and they can be considered as specialized structures of

More information

MBios 401/501: Lecture 12.1 Signaling IV. Slide 1

MBios 401/501: Lecture 12.1 Signaling IV. Slide 1 MBios 401/501: Lecture 12.1 Signaling IV Slide 1 Pathways that require regulated proteolysis 1. Notch and Delta 2. Wnt/ b-catenin 3. Hedgehog 4. NFk-B Our last topic on cell signaling are pathways that

More information

CHAPTER VII CONCLUDING REMARKS AND FUTURE DIRECTION. Androgen deprivation therapy is the most used treatment of de novo or recurrent

CHAPTER VII CONCLUDING REMARKS AND FUTURE DIRECTION. Androgen deprivation therapy is the most used treatment of de novo or recurrent CHAPTER VII CONCLUDING REMARKS AND FUTURE DIRECTION Stathmin in Prostate Cancer Development and Progression Androgen deprivation therapy is the most used treatment of de novo or recurrent metastatic PCa.

More information

From crypt stem cell to colorectal cancer

From crypt stem cell to colorectal cancer 19 3 2007 6 Chinese Bulletin of Life Sciences Vol. 19, No. 3 Jun., 2007 1004-0374(2007)03-0321-05 ( 510405) Wnt Notch BMP R735.35; R730.21 A From crypt stem cell to colorectal cancer WEN Bin*, CHEN Weiwen

More information

Principles of Genetics and Molecular Biology

Principles of Genetics and Molecular Biology Cell signaling Dr. Diala Abu-Hassan, DDS, PhD School of Medicine Dr.abuhassand@gmail.com Principles of Genetics and Molecular Biology www.cs.montana.edu Modes of cell signaling Direct interaction of a

More information

Chapter 4 Opener Pearson Education, Inc.

Chapter 4 Opener Pearson Education, Inc. Chapter 4 Opener Introduction The integumentary system is composed of: Skin Hair Nails Sweat glands Oil glands Mammary glands The skin is the most visible organ of the body Clinicians can tell a lot about

More information

Cell Death & Renewal (part 2)

Cell Death & Renewal (part 2) 17 Cell Death & Renewal (part 2) Programmed Cell Death A major signaling pathway that promotes cell survival is initiated by the enzyme PI 3-kinase, which phosphorylates PIP2 to form PIP3, which activates

More information

B. Incorrect! The ectoderm does not produce the dermis. C. Incorrect! The dermis is derived from the mesoderm.

B. Incorrect! The ectoderm does not produce the dermis. C. Incorrect! The dermis is derived from the mesoderm. Human Anatomy - Problem Drill 04: The Integumentary System Question No. 1 of 10 Instructions: (1) Read the problem and answer choices carefully, (2) Work the problems on paper as 1. From the inner cell

More information

Tetrapod Limb Development

Tetrapod Limb Development IBS 8102 Cell, Molecular and Developmental Biology Tetrapod Limb Development February 11, 2008 Tetrapod Limbs Merlin D. Tuttle Vicki Lockard and Paul Barry Father Alejandro Sanchez Anne Fischer Limb Patterning

More information

Neoplasia 18 lecture 6. Dr Heyam Awad MD, FRCPath

Neoplasia 18 lecture 6. Dr Heyam Awad MD, FRCPath Neoplasia 18 lecture 6 Dr Heyam Awad MD, FRCPath ILOS 1. understand the role of TGF beta, contact inhibition and APC in tumorigenesis. 2. implement the above knowledge in understanding histopathology reports.

More information

Wnt/β-catenin s regulatory role in embryonic stem cell pluripotency and differentiation

Wnt/β-catenin s regulatory role in embryonic stem cell pluripotency and differentiation Wnt/β-catenin s regulatory role in embryonic stem cell pluripotency and differentiation Wim de Jonge 3361039 Supervisor: Prof. Dr. S.J.L. van den Heuvel Group: Developmental Biology, Faculty of Science,

More information

BMP Signaling and Its psmad1/5 Target Genes Differentially Regulate Hair Follicle Stem Cell Lineages

BMP Signaling and Its psmad1/5 Target Genes Differentially Regulate Hair Follicle Stem Cell Lineages Article BMP Signaling and Its psmad1/5 Target Genes Differentially Regulate Hair Follicle Stem Cell Lineages Graphical Abstract Authors Maria Genander, Peter J. Cook,..., Rickard Sandberg, Elaine Fuchs

More information

Supporting Information

Supporting Information Supporting Information Plikus et al. 10.1073/pnas.1215935110 SI Text Movies S1, S2, S3, and S4 are time-lapse recordings from individually cultured Period2 Luc vibrissa follicles show that circadian cycles

More information

Cell Biology Lecture 9 Notes Basic Principles of cell signaling and GPCR system

Cell Biology Lecture 9 Notes Basic Principles of cell signaling and GPCR system Cell Biology Lecture 9 Notes Basic Principles of cell signaling and GPCR system Basic Elements of cell signaling: Signal or signaling molecule (ligand, first messenger) o Small molecules (epinephrine,

More information

Skin (Integumentary System) Wheater, Chap. 9

Skin (Integumentary System) Wheater, Chap. 9 Skin (Integumentary System) Wheater, Chap. 9 Skin (Integument) Consists of skin and associated derivatives Largest organ of body (21 ft 2 ; 9 lbs.; has 11 miles of blood vessels) Functions: Protection

More information

Supplementary Figure 1: Signaling centers contain few proliferating cells, express p21, and

Supplementary Figure 1: Signaling centers contain few proliferating cells, express p21, and Supplementary Figure 1: Signaling centers contain few proliferating cells, express p21, and exclude YAP from the nucleus. (a) Schematic diagram of an E10.5 mouse embryo. (b,c) Sections at B and C in (a)

More information

Skin. Kristine Krafts, M.D.

Skin. Kristine Krafts, M.D. Skin Kristine Krafts, M.D. Skin Lecture Objectives Describe the functions of skin. Describe the structure, location and function of the cell types found in epidermis: keratinocytes, melanocytes, Langerhans

More information

Lab 7: Integumentary System Hamilton ANSWERS TO PRE- LAB ASSIGNMENTS

Lab 7: Integumentary System Hamilton ANSWERS TO PRE- LAB ASSIGNMENTS Lab 7: Integumentary System Hamilton ANSWERS TO PRE- LAB ASSIGNMENTS Pre-Lab Activity 1: 1. a. epidermis b. dermis c. hypodermis d. adipose tissue e. hair f. sebaceous gland g. sweat gland 2. a Pre-Lab

More information

Summary and Concluding Remarks

Summary and Concluding Remarks Summary and Concluding Remarks Chapter 6 The intestinal epithelium provides an excellent model system for investigating molecular mechanisms regulating cell lineage establishment, stem cell proliferation,

More information

Skin stem cells: rising to the surface

Skin stem cells: rising to the surface JCB: MINI-REVIEW Skin stem cells: rising to the surface Elaine Fuchs Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY 10065

More information

Building Epithelial Tissues from Skin Stem Cells

Building Epithelial Tissues from Skin Stem Cells Building Epithelial Tissues from Skin Stem Cells E. FUCHS AND J.A. NOWAK Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New

More information

Tetrapod Limb Development

Tetrapod Limb Development Biology 4361 Developmental Biology Tetrapod Limb Development July 29, 2009 Tetrapod Limbs Merlin D. Tuttle Vicki Lockard and Paul Barry Father Alejandro Sanchez Anne Fischer Limb Development - Overview

More information

Stem Cells. Induced Stem Cells

Stem Cells. Induced Stem Cells Induced Stem Cells Stem Cells Mouse and human somatic cells can either be reprogrammed to a pluripotent state or converted to another lineage with a combination of transcription factors suggesting that

More information

CHAPTER 3. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 23: (2005)

CHAPTER 3. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 23: (2005) CHAPTER 3 Expression pattern of Wnt signaling components in the adult intestine Gastroenterology 23:1825-1833 (2005) Chapter 3 Expression pattern of Wnt signaling components in the adult intestine Alex

More information

Biology Developmental Biology Spring Quarter Midterm 1 Version A

Biology Developmental Biology Spring Quarter Midterm 1 Version A Biology 411 - Developmental Biology Spring Quarter 2013 Midterm 1 Version A 75 Total Points Open Book Choose 15 out the 20 questions to answer (5 pts each). Only the first 15 questions that are answered

More information

Anatomy and Physiology I Student Outline The Integumentary System. Integumentary System. Page 1

Anatomy and Physiology I Student Outline The Integumentary System. Integumentary System. Page 1 Anatomy and Physiology I Student Outline The Integumentary System Integumentary System Page 1 Have a very clear understanding of the each particular tissue and their unique functions in each layer of the

More information

Development of the skin and its derivatives

Development of the skin and its derivatives Development of the skin and its derivatives Resources: http://php.med.unsw.edu.au/embryology/ Larsen s Human Embryology The Developing Human: Clinically Oriented Embryology Dr Annemiek Beverdam School

More information

WNT signalling pathways as therapeutic targets in cancer

WNT signalling pathways as therapeutic targets in cancer WNT signalling pathways as therapeutic targets in cancer Jamie N. Anastas 1,2,3,4 and Randall T. Moon 1,2,4 Abstract Since the initial discovery of the oncogenic activity of WNT1 in mouse mammary glands,

More information

Haematopoietic stem cells

Haematopoietic stem cells Haematopoietic stem cells Neil P. Rodrigues, DPhil NIH Centre for Biomedical Research Excellence in Stem Cell Biology Boston University School of Medicine neil.rodrigues@imm.ox.ac.uk Haematopoiesis: An

More information

RAS Genes. The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes.

RAS Genes. The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes. ۱ RAS Genes The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes. Oncogenic ras genes in human cells include H ras, N ras,

More information

Integumentary System and Body Membranes

Integumentary System and Body Membranes Integumentary System and Body Membranes The Skin and its appendages hair, nails, and skin glands Anatomy/Physiology NHS http://www.lab.anhb.uwa.edu.au/mb140/corepages/integumentary/integum.htm I. System

More information

Abnormal Hair Development and Apparent Follicular Transformation to Mammary Gland in the Absence of Hedgehog Signaling

Abnormal Hair Development and Apparent Follicular Transformation to Mammary Gland in the Absence of Hedgehog Signaling Article Abnormal Hair Development and Apparent Follicular Transformation to Mammary Gland in the Absence of Hedgehog Signaling Amel Gritli-Linde, 1, * Kristina Hallberg, 1 Brian D. Harfe, 2 Azadeh Reyahi,

More information

Chapter 4 :Organization & Regulation of Body Systems

Chapter 4 :Organization & Regulation of Body Systems Chapter 4 :Organization & Regulation of Body Systems 4.1 Types of tissues What is a tissue? A collection of cells of the same type that perform a common function There are 4 major tissue types in the body:

More information

Molecular Cell Biology - Problem Drill 19: Cell Signaling Pathways and Gene Expression

Molecular Cell Biology - Problem Drill 19: Cell Signaling Pathways and Gene Expression Molecular Cell Biology - Problem Drill 19: Cell Signaling Pathways and Gene Expression Question No. 1 of 10 1. Which statement about cell signaling is correct? Question #1 (A) Cell signaling involves receiving

More information

T Cell Development. Xuefang Cao, MD, PhD. November 3, 2015

T Cell Development. Xuefang Cao, MD, PhD. November 3, 2015 T Cell Development Xuefang Cao, MD, PhD November 3, 2015 Thymocytes in the cortex of the thymus Early thymocytes development Positive and negative selection Lineage commitment Exit from the thymus and

More information

General Principles of Endocrine Physiology

General Principles of Endocrine Physiology General Principles of Endocrine Physiology By Dr. Isabel S.S. Hwang Department of Physiology Faculty of Medicine University of Hong Kong The major human endocrine glands Endocrine glands and hormones

More information

Page 32 AP Biology: 2013 Exam Review CONCEPT 6 REGULATION

Page 32 AP Biology: 2013 Exam Review CONCEPT 6 REGULATION Page 32 AP Biology: 2013 Exam Review CONCEPT 6 REGULATION 1. Feedback a. Negative feedback mechanisms maintain dynamic homeostasis for a particular condition (variable) by regulating physiological processes,

More information

Wnt signaling. Ramray Bhat.

Wnt signaling. Ramray Bhat. Wnt signaling Ramray Bhat ramray@mrdg.iisc.ernet.in Starting with animal biology and viral infections The discovery of certain laboratory murine strains that were highly susceptible to mammary gland cancer.

More information

TISSUE-SPECIFIC STEM CELLS

TISSUE-SPECIFIC STEM CELLS TISSUE-SPECIFIC STEM CELLS Bone Morphogenetic Protein Signaling Inhibits Hair Follicle Anagen Induction by Restricting Epithelial Stem/Progenitor Cell Activation and Expansion JIWANG ZHANG, a XI C. HE,

More information

Principles of cell signaling Lecture 4

Principles of cell signaling Lecture 4 Principles of cell signaling Lecture 4 Johan Lennartsson Molecular Cell Biology (1BG320), 2014 Johan.Lennartsson@licr.uu.se 1 Receptor tyrosine kinase-induced signal transduction Erk MAP kinase pathway

More information

Epidermis. Integumentary system

Epidermis. Integumentary system Epidermis the doctor mentioned at the begging of the lecture that the slides is from different sources and has information and details that is enough for us so we don t have to go back and read from the

More information

Early Embryonic Development

Early Embryonic Development Early Embryonic Development Maternal effect gene products set the stage by controlling the expression of the first embryonic genes. 1. Transcription factors 2. Receptors 3. Regulatory proteins Maternal

More information

Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment

Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment In multicellular eukaryotes, gene expression regulates development

More information

Cell Communication and Cell Signaling

Cell Communication and Cell Signaling Cell Communication and Cell Signaling Why is cell signaling important? Why is cell signaling important? Allows cells to communicate and coordinate functions/activities of the organism Usually involves

More information

Protein Kinase C Modulates Wnt Signaling In Colon Tumoral Cell Lines

Protein Kinase C Modulates Wnt Signaling In Colon Tumoral Cell Lines Protein Kinase C Modulates Wnt Signaling In Colon Tumoral Cell Lines Dra Martha Robles Flores Department of Biochemistry Facultad de Medicina Universidad Nacional Autónoma de México Tissue anatomy of the

More information

Dr Narmeen S. Ahmad. Lab 1

Dr Narmeen S. Ahmad. Lab 1 Dr Narmeen S. Ahmad Lab 1 1 Tissues are groups of cells with a common structure (form) and function (job). There are (4) types of tissue: 1. Epithelial 2. Connective 3. Muscle 4. Nervous 2 Epithelial cells

More information

Cytological and Histological Study of Adult and Neonate Epidermis in Thick and Thin Skin of Various Anatomical Sites

Cytological and Histological Study of Adult and Neonate Epidermis in Thick and Thin Skin of Various Anatomical Sites Available online on www.ijpqa.com International Journal of Pharmaceutical Quality Assurance 218; 9(2); 174-179 doi: 1.25258/ijpqa.v9i2.13642 ISSN 975 956 Research Article Cytological and Histological Study

More information

Signaling. Dr. Sujata Persad Katz Group Centre for Pharmacy & Health research

Signaling. Dr. Sujata Persad Katz Group Centre for Pharmacy & Health research Signaling Dr. Sujata Persad 3-020 Katz Group Centre for Pharmacy & Health research E-mail:sujata.persad@ualberta.ca 1 Growth Factor Receptors and Other Signaling Pathways What we will cover today: How

More information

Src-INACTIVE / Src-INACTIVE

Src-INACTIVE / Src-INACTIVE Biology 169 -- Exam 1 February 2003 Answer each question, noting carefully the instructions for each. Repeat- Read the instructions for each question before answering!!! Be as specific as possible in each

More information

Axis Formation and Mesoderm Induction

Axis Formation and Mesoderm Induction Developmental Biology Biology 4361 Axis Formation and Mesoderm Induction October 27, 2005 Amphibian anteroposterior specification polarized eggs animal/vegetal pigment yolk v. clear cytoplasm mitochondrial

More information

Lecture 14 - The cell cycle and cell death

Lecture 14 - The cell cycle and cell death 02.17.10 Lecture 14 - The cell cycle and cell death The cell cycle: cells duplicate their contents and divide The cell cycle may be divided into 4 phases The cell cycle triggers essential processes (DNA

More information

G-Protein Signaling. Introduction to intracellular signaling. Dr. SARRAY Sameh, Ph.D

G-Protein Signaling. Introduction to intracellular signaling. Dr. SARRAY Sameh, Ph.D G-Protein Signaling Introduction to intracellular signaling Dr. SARRAY Sameh, Ph.D Cell signaling Cells communicate via extracellular signaling molecules (Hormones, growth factors and neurotransmitters

More information

Regulators of Cell Cycle Progression

Regulators of Cell Cycle Progression Regulators of Cell Cycle Progression Studies of Cdk s and cyclins in genetically modified mice reveal a high level of plasticity, allowing different cyclins and Cdk s to compensate for the loss of one

More information

INTEGUMENTARY 1-Epidermis, 2-Dermis, Structure of thick and thin skin I- Epidermis . Stratum basale

INTEGUMENTARY 1-Epidermis, 2-Dermis, Structure of thick and thin skin I- Epidermis . Stratum basale INTEGUMENTARY The skin (integument, cutis ) and its derivatives constitute the integumentary system. It form the external covering of the body and is the largest organ of the body. The skin consists of

More information

Inner ear development Nervous system development

Inner ear development Nervous system development Upcoming Sessions April 22: Nervous System Development Lecture April 24: Reviews of Axonal Pathfinding in Sensory Systems April 29: Inner Ear Development Lecture May 1: May 6: May 8: Auditory System Pathfinding

More information

WNT2 and WNT7B Cooperative Signaling in Lung Development

WNT2 and WNT7B Cooperative Signaling in Lung Development University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 1-1-2012 WNT2 and WNT7B Cooperative Signaling in Lung Development Mayumi Miller University of Pennsylvania, mayumimiller@gmail.com

More information

Early cell death (FGF) B No RunX transcription factor produced Yes No differentiation

Early cell death (FGF) B No RunX transcription factor produced Yes No differentiation Solution Key - Practice Questions Question 1 a) A recent publication has shown that the fat stem cells (FSC) can act as bone stem cells to repair cavities in the skull, when transplanted into immuno-compromised

More information

The Integumentary System: An Overview

The Integumentary System: An Overview The Integumentary System: An Overview Functions: Protective covering Helps regulate body temperature Retards water loss from deeper tissues Houses sensory receptors Synthesizes biochemicals Excretes small

More information

Biol403 MAP kinase signalling

Biol403 MAP kinase signalling Biol403 MAP kinase signalling The mitogen activated protein kinase (MAPK) pathway is a signalling cascade activated by a diverse range of effectors. The cascade regulates many cellular activities including

More information

Histology of Integumentary System

Histology of Integumentary System Histology of Integumentary System CONTENT / Topics Integumentary System Skin, thick - Major Layers Epidermis Dermis Hair Skin, hairy and Hair Follicle Sebaceous Glands Sebaceous Gland Sweat Glands Merocrine

More information

Bio 111 Study Guide Chapter 11 Cell Communication

Bio 111 Study Guide Chapter 11 Cell Communication Bio 111 Study Guide Chapter 11 Cell Communication BEFORE CLASS: Reading: Read the introduction on p. 210, and for Concept 11.1, read from the first full paragraph on p. 212. Read all of Concept 11.2. Pay

More information

C. elegans Embryonic Development

C. elegans Embryonic Development Autonomous Specification in Tunicate development Autonomous & Conditional Specification in C. elegans Embryonic Development Figure 8.36 Bilateral Symmetry in the Egg of the Tunicate Styela partita Fig.

More information

1. Introduction (Open your text to the image of a cross section of skin) i. Organ of the Integument. Connective Tissues. Epithelial Tissues

1. Introduction (Open your text to the image of a cross section of skin) i. Organ of the Integument. Connective Tissues. Epithelial Tissues Integumentary System 1. Introduction (Open your text to the image of a cross section of skin) A. Integumentary System i. Organ of the Integument a. Tissues Connective Tissues * Tissue / Location Relationships

More information

This section covers the basic knowledge of normal skin structure and function required to help understand how skin diseases occur.

This section covers the basic knowledge of normal skin structure and function required to help understand how skin diseases occur. Background Knowledge Functions of normal skin Background Knowledge This section covers the basic knowledge of normal skin structure and function required to help understand how skin diseases occur. Learning

More information

Roles of Flow Mechanics in Vascular Cell Biology in Health and Disease

Roles of Flow Mechanics in Vascular Cell Biology in Health and Disease Roles of Flow Mechanics in Vascular Cell Biology in Health and Disease Shu Chien Dept. of Bioengineering & Medicine UC, San Diego Presented by Ming-Shaung Ju Dept. of Mech. Eng., NCKU, Tainan Background

More information

Chapter 9. Cellular Signaling

Chapter 9. Cellular Signaling Chapter 9 Cellular Signaling Cellular Messaging Page 215 Cells can signal to each other and interpret the signals they receive from other cells and the environment Signals are most often chemicals The

More information

Principles of Anatomy and Physiology

Principles of Anatomy and Physiology Principles of Anatomy and Physiology 14 th Edition CHAPTER 5 The Integumentary System Introduction The organs of the integumentary system include the skin and its accessory structures including hair, nails,

More information

TISSUE-SPECIFIC STEM CELLS

TISSUE-SPECIFIC STEM CELLS TISSUE-SPECIFIC Concise Review: Mechanisms of Quiescent Hair Follicle Stem Cell Regulation RUI YI Key Words. Self-renewal Adult stem cells Cell signaling Epidermis Transcriptional regulation Department

More information

Branching morphogenesis of the lung: new molecular insights into an old problem

Branching morphogenesis of the lung: new molecular insights into an old problem 86 Review TRENDS in Cell Biology Vol.13 No.2 February 2003 Branching morphogenesis of the lung: new molecular insights into an old problem Pao-Tien Chuang 1 and Andrew P. McMahon 2 1 Cardiovascular Research

More information

number Done by Corrected by Doctor Maha Shomaf

number Done by Corrected by Doctor Maha Shomaf number 19 Done by Waseem Abo-Obeida Corrected by Abdullah Zreiqat Doctor Maha Shomaf Carcinogenesis: the molecular basis of cancer. Non-lethal genetic damage lies at the heart of carcinogenesis and leads

More information

Multiple Roles of Notch Signaling in the Regulation of Epidermal Development

Multiple Roles of Notch Signaling in the Regulation of Epidermal Development Article Multiple Roles of Notch Signaling in the Regulation of Epidermal Development Mariko Moriyama, 1 André-Dante Durham, 2 Hiroyuki Moriyama, 1 Kiyotaka Hasegawa, 1 Shin-Ichi Nishikawa, 3 Freddy Radtke,

More information

Computational Biology I LSM5191

Computational Biology I LSM5191 Computational Biology I LSM5191 Aylwin Ng, D.Phil Lecture 6 Notes: Control Systems in Gene Expression Pulling it all together: coordinated control of transcriptional regulatory molecules Simple Control:

More information

Ch 4. Skin and Body Membranes

Ch 4. Skin and Body Membranes Ch 4 Skin and Body Membranes TITLE HISTOLOGY SLIDES & NOTES ESSENTIAL QUESTION What tissues compose the integumentary system? Stratified Squamous Epithelium Stratified = several layers; Squamous = shape

More information

Integumentary System

Integumentary System Chapter 5 Integumentary System 5-1 Skin: composed of dermis and epidermis Dermis. Gives structural strength. C.T. with many fibers, fibroblasts, macrophages. Some adipocytes and blood vessels. Contains

More information

Tetrapod Limb Development

Tetrapod Limb Development Biology 4361 Developmental Biology Tetrapod Limb Development July 29, 2009 Tetrapod Limbs Merlin D. Tuttle Vicki Lockard and Paul Barry Father Alejandro Sanchez Anne Fischer Limb Development - Overview

More information

Journal Club. 03/04/2012 Lama Nazzal

Journal Club. 03/04/2012 Lama Nazzal Journal Club 03/04/2012 Lama Nazzal NOTCH and the kidneys Is an evolutionarily conserved cell cell communication mechanism. Is a regulator of cell specification, differentiation, and tissue patterning.

More information

Cell signaling. How do cells receive and respond to signals from their surroundings?

Cell signaling. How do cells receive and respond to signals from their surroundings? Cell signaling How do cells receive and respond to signals from their surroundings? Prokaryotes and unicellular eukaryotes are largely independent and autonomous. In multicellular organisms there is a

More information

EMT: Epithelial Mesenchimal Transition

EMT: Epithelial Mesenchimal Transition EMT: Epithelial Mesenchimal Transition A phenotypic change that is characteristic of some developing tissues and certain forms of cancer. This is a multistep, key process in embryonic development and metastasis

More information

Basal cell carcinomas in mice arise from hair follicle stem cells and multiple epithelial progenitor populations

Basal cell carcinomas in mice arise from hair follicle stem cells and multiple epithelial progenitor populations Research article Basal cell carcinomas in mice arise from hair follicle stem cells and multiple epithelial progenitor populations Marina Grachtchouk, 1 Joanna Pero, 1 Steven H. Yang, 1,2 Alexandre N. Ermilov,

More information

Cell Communication. Local and Long Distance Signaling

Cell Communication. Local and Long Distance Signaling Cell Communication Cell to cell communication is essential for multicellular organisms Some universal mechanisms of cellular regulation providing more evidence for the evolutionary relatedness of all life

More information

Skin. Lecture #14. Ref:

Skin. Lecture #14. Ref: Skin Lecture #14 Ref: http://www.ccunix.ccu.edu.tw/~chenmsl/tea/skin_910721.htm Structure of Skin 1. Epidermis 2. Dermis 3. Subcutis 4. Hair follicle 5. Sebaceous gland 6. Sweat gland Skin Largest human

More information

Animal Tissue Culture SQG 3242 Biology of Cultured Cells. Dr. Siti Pauliena Mohd Bohari

Animal Tissue Culture SQG 3242 Biology of Cultured Cells. Dr. Siti Pauliena Mohd Bohari Animal Tissue Culture SQG 3242 Biology of Cultured Cells Dr. Siti Pauliena Mohd Bohari The Culture Environment Changes of Cell s microenvironment needed that favor the spreading, migration, and proliferation

More information

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation Cancer The fundamental defect is unregulated cell division. Properties of Cancerous Cells Altered growth and proliferation Loss of growth factor dependence Loss of contact inhibition Immortalization Alterated

More information

Prof. R. V. Skibbens

Prof. R. V. Skibbens Prof. R. V. Skibbens September 8, 2017 BioScience in the 21 st Century Cell Cycle, Cell Division and intro to Cancer Cell growth and division What are the goals? I Cell Cycle what is this? response to

More information

INTERACTION DRUG BODY

INTERACTION DRUG BODY INTERACTION DRUG BODY What the drug does to the body What the body does to the drug Receptors - intracellular receptors - membrane receptors - Channel receptors - G protein-coupled receptors - Tyrosine-kinase

More information

HHS Public Access Author manuscript Nat Med. Author manuscript; available in PMC 2015 March 13.

HHS Public Access Author manuscript Nat Med. Author manuscript; available in PMC 2015 March 13. Emerging interactions between skin stem cells and their niches Ya-Chieh Hsu 1,2, Lishi Li 1, and Elaine Fuchs 1 1 Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development,

More information